Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Dev Dyn ; 2023 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-38071599

RESUMEN

BACKGROUND: Gastrulation is one of the most important events in our lives (Barresi and Gilbert, 2020, Developmental Biology, 12th ed.). The molecular mechanisms of gastrulation in multicellular organisms are not yet fully understood, since many molecular, physical, and chemical factors are involved in the event. RESULTS: Here, we found that one of muscle components, Troponin-I (TnI), is expressed in future gut cells, which are not muscular cells at all, and regulates gastrulation in embryos of a sea urchin, Hemicentrotus pulcherrimus. When we block the function of TnI, the invagination was inhibited in spite that the gut-cell specifier gene is normally expressed. In addition, blocking myosin activity also induced incomplete gastrulation. CONCLUSION: These results strongly suggested that TnI regulates nonmuscular actin-myosin interactions during sea urchin gastrulation. So far, Troponin system is treated as specific only for muscle components, especially for striated muscle, but our data clearly show that TnI is involved in nonmuscular event. It is also reported that recent sensitive gene expression analysis revealed that Troponin genes are expressed in nonmuscular tissues in mammals (Ono et al., Sci Data, 2017;4:170105). These evidences propose the new evolutionary and functional scenario of the involvement of Troponin system in nonmuscular cell behaviors using actin-myosin system in bilaterians including human being.

2.
Front Cell Dev Biol ; 11: 1240767, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37655161

RESUMEN

Acetylcholine, a vital neurotransmitter, plays a multifarious role in the brain and peripheral nervous system of various organisms. Previous research has demonstrated the proximity of cholinergic neurons to serotonergic neurons in the apical organ of sea urchin embryos. While several transcription factors have been identified as playing a role in the development of serotonergic neurons in this region of a sea urchin, Hemicentrotus pulcherrimus, comparatively little is known about the specific transcription factors and their spatiotemporal expression patterns that regulate the development of cholinergic neurons. In this study, we establish the requirement of the transcription factor Rx for the development of cholinergic neurons in the apical organ of the species. Furthermore, we investigate the role of the RNA-binding protein Musashi1, known to be involved in neurogenesis, including cholinergic neurons in other organisms, and demonstrate that it is a downstream factor of Rx, and that choline acetyltransferase expression is suppressed in Musashi1 downregulated embryos. Our research also highlights the intricate network formed by neurons and other cells in and around the apical organ of sea urchin larvae through axons and dendrites, providing possibility for a systematic and complexed neural pattern like those of the brain in other organisms.

3.
PLoS Genet ; 18(2): e1010033, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35143488

RESUMEN

To survive, organisms need to precisely respond to various environmental factors, such as light and gravity. Among these, light is so important for most life on Earth that light-response systems have become extraordinarily developed during evolution, especially in multicellular animals. A combination of photoreceptors, nervous system components, and effectors allows these animals to respond to light stimuli. In most macroscopic animals, muscles function as effectors responding to light, and in some microscopic aquatic animals, cilia play a role. It is likely that the cilia-based response was the first to develop and that it has been substituted by the muscle-based response along with increases in body size. However, although the function of muscle appears prominent, it is poorly understood whether ciliary responses to light are present and/or functional, especially in deuterostomes, because it is possible that these responses are too subtle to be observed, unlike muscle responses. Here, we show that planktonic sea urchin larvae reverse their swimming direction due to the inhibitory effect of light on the cholinergic neuron signaling>forward swimming pathway. We found that strong photoirradiation of larvae that stay on the surface of seawater immediately drives the larvae away from the surface due to backward swimming. When Opsin2, which is expressed in mesenchymal cells in larval arms, is knocked down, the larvae do not show backward swimming under photoirradiation. Although Opsin2-expressing cells are not neuronal cells, immunohistochemical analysis revealed that they directly attach to cholinergic neurons, which are thought to regulate forward swimming. These data indicate that light, through Opsin2, inhibits the activity of cholinergic signaling, which normally promotes larval forward swimming, and that the light-dependent ciliary response is present in deuterostomes. These findings shed light on how light-responsive tissues/organelles have been conserved and diversified during evolution.


Asunto(s)
Movimiento/fisiología , Células Fotorreceptoras/metabolismo , Erizos de Mar/metabolismo , Animales , Cilios/metabolismo , Larva/metabolismo , Luz , Locomoción/fisiología , Músculos/fisiología , Opsinas/genética , Opsinas/metabolismo , Plancton
4.
Dev Growth Differ ; 64(1): 59-66, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34923630

RESUMEN

Echinoderms, including sea urchins and starfish, have played important roles in cell, developmental and evolutionary biology research for more than a century. However, since most of them take a long time to mature sexually and their breeding seasons are limited, it has been difficult to obtain subsequent generations in the laboratory, resulting in them not being recognized as model organisms in recent genetics research. To overcome this issue, we maintained and obtained gametes from several nonmodel sea urchins in Japan and finally identified Temnopleurus reevesii as a suitable model for sea urchin genetics. Genomic and transcriptomic information was obtained for this model, and the DNA database TrBase was made publicly available. In this review, we describe how we found this species useful for biological research and show an example of CRISPR/Cas9 based knockout sea urchin.


Asunto(s)
Genoma , Erizos de Mar , Animales , Japón , Erizos de Mar/genética
5.
Exp Anim ; 70(3): 378-386, 2021 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-33828019

RESUMEN

Sea urchin embryos have been one of model organisms to investigate cellular behaviors because of their simple cell composition and transparent body. They also give us an opportunity to investigate molecular functions of human proteins of interest that are conserved in sea urchin. Here we report that human disease-associated extracellular matrix orthologues ECM3 and QBRICK are necessary for mesenchymal cell migration during sea urchin embryogenesis. Immunofluorescence has visualized the colocalization of QBRICK and ECM3 on both apical and basal surface of ectoderm. On the basal surface, QBRICK and ECM3 constitute together a mesh-like fibrillar structure along the blastocoel wall. When the expression of ECM3 was knocked down by antisense-morpholino oligonucleotides, the ECM3-QBRICK fibrillar structure completely disappeared. When QBRICK was knocked down, the ECM3 was still present, but the basally localized fibers became fragmented. The ingression and migration of primary mesenchymal cells were not critically affected, but their migration at later stages was severely affected in both knock-down embryos. As a consequence of impaired primary mesenchymal cell migration, improper spicule formation was observed. These results indicate that ECM3 and QBRICK are components of extracellular matrix, which play important role in primary mesenchymal cell migration, and that sea urchin is a useful experimental animal model to investigate human disease-associated extracellular matrix proteins.


Asunto(s)
Embrión no Mamífero/fisiología , Desarrollo Embrionario/fisiología , Proteínas de la Matriz Extracelular/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Hemicentrotus/fisiología , Células Madre Mesenquimatosas/fisiología , Animales , Movimiento Celular , Proteínas de la Matriz Extracelular/metabolismo , Hemicentrotus/genética , Hemicentrotus/crecimiento & desarrollo
6.
BMC Biol ; 19(1): 64, 2021 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-33820528

RESUMEN

BACKGROUND: Light is essential for various biological activities. In particular, visual information through eyes or eyespots is very important for most of animals, and thus, the functions and developmental mechanisms of visual systems have been well studied to date. In addition, light-dependent non-visual systems expressing photoreceptor Opsins have been used to study the effects of light on diverse animal behaviors. However, it remains unclear how light-dependent systems were acquired and diversified during deuterostome evolution due to an almost complete lack of knowledge on the light-response signaling pathway in Ambulacraria, one of the major groups of deuterostomes and a sister group of chordates. RESULTS: Here, we show that sea urchin larvae utilize light for digestive tract activity. We found that photoirradiation of larvae induces pyloric opening even without addition of food stimuli. Micro-surgical and knockdown experiments revealed that this stimulating light is received and mediated by Go(/RGR)-Opsin (Opsin3.2 in sea urchin genomes) cells around the anterior neuroectoderm. Furthermore, we found that the anterior neuroectodermal serotoninergic neurons near Go-Opsin-expressing cells are essential for mediating light stimuli-induced nitric oxide (NO) release at the pylorus. Our results demonstrate that the light>Go-Opsin>serotonin>NO pathway functions in pyloric opening during larval stages. CONCLUSIONS: The results shown here will lead us to understand how light-dependent systems of pyloric opening functioning via neurotransmitters were acquired and established during animal evolution. Based on the similarity of nervous system patterns and the gut proportions among Ambulacraria, we suggest the light>pyloric opening pathway may be conserved in the clade, although the light signaling pathway has so far not been reported in other members of the group. In light of brain-gut interactions previously found in vertebrates, we speculate that one primitive function of anterior neuroectodermal neurons (brain neurons) may have been to regulate the function of the digestive tract in the common ancestor of deuterostomes. Given that food consumption and nutrient absorption are essential for animals, the acquirement and development of brain-based sophisticated gut regulatory system might have been important for deuterostome evolution.


Asunto(s)
Luz , Píloro/efectos de la radiación , Erizos de Mar/efectos de la radiación , Animales , Larva/metabolismo , Larva/efectos de la radiación , Píloro/metabolismo , Erizos de Mar/metabolismo
7.
Curr Biol ; 30(10): R427-R429, 2020 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-32428469

RESUMEN

Yaguchi et al. establish a homozygous knock-out sea urchin line by applying the CRISPR-Cas9 system to a new model species, Temnopleurus reevesii, whose breeding cycle takes about half a year.


Asunto(s)
Sistemas CRISPR-Cas , Técnicas de Inactivación de Genes/métodos , Sintasas Poliquetidas/genética , Erizos de Mar/genética , Animales , Secuencia de Bases , ADN/química , ADN/genética , Femenino , Regulación Enzimológica de la Expresión Génica , Homocigoto , Masculino , Mutación
8.
Genesis ; 57(6): e23302, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31025827

RESUMEN

The specification of anterior neuroectoderm is controlled by a highly conserved molecular mechanism in bilaterians. A forkhead family gene, foxQ2, is known to be one of the pivotal regulators, which is zygotically expressed in this region during embryogenesis of a broad range of bilaterians. However, what controls the expression of this essential factor has remained unclear to date. To reveal the regulatory mechanism of foxQ2, we performed cis-regulatory analysis of two foxQ2 genes, foxQ2a and foxQ2b, in a sea urchin Hemicentrotus pulcherrimus. In sea urchin embryos, foxQ2 is initially expressed in the entire animal hemisphere and subsequently shows narrower expression restricted to the anterior pole region. In this study, as a first step to understand the foxQ2 regulation, we focused on the later restricted expression and analyzed the upstream cis-regulatory sequences of foxQ2a and foxQ2b by using the constructs fused to short half-life green fluorescent protein. Based on deletion and mutation analyses of both foxQ2, we identified each of the five regulatory sequences, which were 4-9 bp long. Neither of the regulatory sequences contains any motifs for ectopic activation or spatial repression, suggesting that later mRNA localization is regulated in situ. We also suggest that the three amino acid loop extension-class homeobox gene Meis is involved in the maintenance of foxQ2b, the expression of which is dominant during embryogenesis.


Asunto(s)
Erizos de Mar/embriología , Erizos de Mar/genética , Animales , Tipificación del Cuerpo/fisiología , Embrión no Mamífero/metabolismo , Desarrollo Embrionario/genética , Desarrollo Embrionario/fisiología , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Placa Neural/embriología , Placa Neural/metabolismo , ARN Mensajero/genética , Proteínas Wnt/metabolismo
9.
Proc Natl Acad Sci U S A ; 116(12): 5607-5612, 2019 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-30833398

RESUMEN

Although morphologies are diverse, the common pattern in bilaterians is for passage of food in the gut to be controlled by nerves and endodermally derived neuron-like cells. In vertebrates, nitric oxide (NO) derived from enteric nerves controls relaxation of the pyloric sphincter. Here, we show that in the larvae of sea urchins, there are endoderm-derived neuronal nitric oxide synthase (nNOS)-positive cells expressing pan-neural marker, Synaptotagmin-B (SynB), in sphincters and that NO regulates the relaxation of the pyloric sphincter. Our results indicate that NO-dependent pylorus regulation is a shared feature within the deuterostomes, and we speculate that it was a characteristic of stem deuterostomes.


Asunto(s)
Óxido Nítrico Sintasa de Tipo I/metabolismo , Óxido Nítrico/metabolismo , Píloro/fisiología , Animales , Evolución Biológica , Evolución Molecular , Larva/fisiología , Neuronas/metabolismo , Píloro/metabolismo , Erizos de Mar/fisiología , Sinaptotagminas
10.
Methods Cell Biol ; 150: 173-188, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30777175

RESUMEN

Methods for microinjection into sea urchin eggs have become relatively easier because of the technical improvements by a number of researchers in the past decades. However, the size and the characteristics, such as the elasticity and toughness, of the eggs and embryos differ in species, so that we still need to modify the details of methods to adapt to each target. In this section, I list microinjection methods for three species: Hemicentrotus pulcherrimus, which has relatively tough eggs, Temnopleurus reevesii, which has slightly weak eggs, and Strongylocentrotus purpuratus, which is the most used species in sea urchin biology. In addition, I describe the methods for co-injection of morpholino anti-sense oligonucleotides and mRNAs, as well as the method for microinjection into blastomeres.


Asunto(s)
Blastómeros/citología , Microinyecciones/métodos , Oocitos/citología , Erizos de Mar/citología , Animales , Embrión no Mamífero/citología
11.
Dev Biol ; 444(1): 1-8, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30266259

RESUMEN

Precise body axis formation is an essential step in the development of multicellular organisms, for most of which the molecular gradient and/or specifically biased localization of cell-fate determinants in eggs play important roles. In sea urchins, however, any biased proteins and mRNAs have not yet been identified in the egg except for vegetal cortex molecules, suggesting that sea urchin development is mostly regulated by uniformly distributed maternal molecules with contributions to axis formation that are not well characterized. Here, we describe that the maternal Meis transcription factor regulates anterior-posterior axis formation through maintenance of the most anterior territory in embryos of a sea urchin, Hemicentrotus pulcherrimus. Loss-of-function experiments revealed that Meis is intrinsically required for maintenance of the anterior neuroectoderm specifier foxQ2 after hatching and, consequently, the morphant lost anterior neuroectoderm characteristics. In addition, the expression patterns of univin and VEGF, the lateral ectoderm markers, and the mesenchyme-cell pattern shifted toward the anterior side in Meis morphants more than they did in control embryos, indicating that Meis contributes to the precise anteroposterior patterning by regulating the anterior neuroectodermal fate.


Asunto(s)
Tipificación del Cuerpo/genética , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/fisiología , Animales , Diferenciación Celular , Ectodermo/metabolismo , Embrión no Mamífero/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Hemicentrotus/embriología , Hemicentrotus/genética , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/metabolismo , Placa Neural/metabolismo , Neurogénesis/genética , ARN Mensajero/metabolismo , Erizos de Mar/embriología , Erizos de Mar/genética , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo , Proteínas Wnt/metabolismo
12.
Sci Rep ; 7(1): 10751, 2017 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-28883641

RESUMEN

Through their coordinated alignment and beating, motile cilia generate directional fluid flow and organismal movement. While the mechanisms used by multiciliated epithelial tissues to achieve this coordination have been widely studied, much less is known about regulation of monociliated tissues such as those found in the vertebrate node and swimming planktonic larvae. Here, we show that a calcium sensor protein associated with outer arm dynein, calaxin, is a critical regulator for the coordinated movements of monocilia. Knockdown of calaxin gene in sea urchin embryos results in uncoordinated ciliary beating and defective directional movement of the embryos, but no apparent abnormality in axoneme ultrastructure. Examination of the beating cycle of individual calaxin-deficient cilia revealed a marked effect on the waveform and spatial range of ciliary bending. These findings indicate that calaxin-mediated regulation of ciliary beating is responsible for proper basal body orientation and ciliary alignment in fields of monociliated cells.


Asunto(s)
Cilios/fisiología , Dineínas/metabolismo , Erizos de Mar/fisiología , Animales , Axonema/ultraestructura , Cuerpos Basales , Cilios/genética , Cilios/metabolismo , Dineínas/genética , Embrión no Mamífero/fisiología , Movimiento , Orientación Espacial , Erizos de Mar/embriología , Erizos de Mar/genética
13.
Sci Rep ; 7: 43563, 2017 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-28272398

RESUMEN

The troponin complex, composed of Troponin-I, Troponin-T and Troponin-C, is an essential mediator of the contraction of striated muscle downstream of calcium signaling in almost all bilaterians. However, in echinoderms and hemichordates, collectively termed Ambulacraria, the components of the troponin complex have never been isolated, thus suggesting that these organisms lost the troponin system during evolution. Here, by analyzing genomic information from sea urchins, we identify the troponin-I gene and isolate its complete mRNA sequence. Using this information, we reveal that the larval muscles express this gene and its translated product and that the protein is definitely a functional molecule expressed in sea urchin larvae by showing that Troponin-I morphants are unable to swallow algae. We conclude that muscular contraction in all bilaterians universally depends on a regulatory system mediated by Troponin-I, which emerged in the common ancestor of bilaterians.


Asunto(s)
Equinodermos/metabolismo , Músculo Estriado/metabolismo , Troponina I/metabolismo , Animales , Biomarcadores , Calmodulina/genética , Calmodulina/metabolismo , Equinodermos/clasificación , Equinodermos/genética , Expresión Génica , Inmunohistoquímica , Larva , Filogenia , Erizos de Mar/genética , Erizos de Mar/metabolismo , Troponina C/genética , Troponina C/metabolismo , Troponina I/química , Troponina I/genética
14.
PLoS Genet ; 12(4): e1006001, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27101101

RESUMEN

When early canonical Wnt is experimentally inhibited, sea urchin embryos embody the concept of a Default Model in vivo because most of the ectodermal cell fates are specified as anterior neuroectoderm. Using this model, we describe here how the combination of orthogonally functioning anteroposterior Wnt and dorsoventral Nodal signals and their targeting transcription factors, FoxQ2 and Homeobrain, regulates the precise patterning of normal neuroectoderm, of which serotonergic neurons are differentiated only at the dorsal/lateral edge. Loss-of-function experiments revealed that ventral Nodal is required for suppressing the serotonergic neural fate in the ventral side of the neuroectoderm through the maintenance of foxQ2 and the repression of homeobrain expression. In addition, non-canonical Wnt suppressed homeobrain in the anterior end of the neuroectoderm, where serotonergic neurons are not differentiated. Canonical Wnt, however, suppresses foxQ2 to promote neural differentiation. Therefore, the three-dimensionally complex patterning of the neuroectoderm is created by cooperative signals, which are essential for the formation of primary and secondary body axes during embryogenesis.


Asunto(s)
Tipificación del Cuerpo/fisiología , Embrión no Mamífero/embriología , Hemicentrotus/embriología , Placa Neural/embriología , Proteína Nodal/metabolismo , Proteínas Wnt/metabolismo , Animales , Tipificación del Cuerpo/genética , Factores de Transcripción Forkhead/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/metabolismo
15.
Dev Growth Differ ; 57(3): 242-50, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25754419

RESUMEN

Sea urchins are model non-chordate deuterostomes, and studying the nervous system of their embryos can aid in the understanding of the universal mechanisms of neurogenesis. However, despite the long history of sea urchin embryology research, the molecular mechanisms of their neurogenesis have not been well investigated, in part because neurons appear relatively late during embryogenesis. In this study, we used the species Temnopleurus reevesii as a new sea urchin model and investigated the detail of its development and neurogenesis during early embryogenesis. We found that the embryos of T. reevesii were tolerant of high temperatures and could be cultured successfully at 15-30°C during early embryogenesis. At 30°C, the embryos developed rapidly enough that the neurons appeared at just after 24 h. This is faster than the development of other model urchins, such as Hemicentrotus pulcherrimus or Strongylocentrotus purpuratus. In addition, the body of the embryo was highly transparent, allowing the details of the neural network to be easily captured by ordinary epifluorescent and confocal microscopy without any additional treatments. Because of its rapid development and high transparency during embryogenesis, T. reevesii may be a suitable sea urchin model for studying neurogenesis. Moreover, the males and females are easily distinguishable, and the style of early cleavages is intriguingly unusual, suggesting that this sea urchin might be a good candidate for addressing not only neurology but also cell and developmental biology.


Asunto(s)
Técnicas de Cultivo de Embriones/métodos , Modelos Animales , Neurogénesis/fisiología , Erizos de Mar/embriología , Animales , Femenino , Inmunohistoquímica , Masculino , Temperatura
16.
Sci Rep ; 4: 6852, 2014 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-25358387

RESUMEN

bicaudal-C (bicC) mRNA encodes a protein containing RNA-binding domains that is reported to be maternally present with deflection in the oocytes/eggs of some species. The translated protein plays a critical role in the regulation of cell fate specification along the body axis during early embryogenesis in flies and frogs. However, it is unclear how it functions in eggs in which bicC mRNA is uniformly distributed, for instance, sea urchin eggs. Here, we show the function of BicC in the formation of neurogenic ectoderm of the sea urchin embryo. Loss-of-function experiments reveal that BicC is required for serotonergic neurogenesis and for expression of ankAT-1 gene, which is essential for the formation of apical tuft cilia in the neurogenic ectoderm of the sea urchin embryo. In contrast, the expression of FoxQ2, the neurogenic ectoderm specification transcription factor, is invariant in BicC morphants. Because FoxQ2 is an upstream factor of serotonergic neurogenesis and ankAT-1 expression, these data indicate that BicC functions in regulating the events that are coordinated by FoxQ2 during sea urchin embryogenesis.


Asunto(s)
Placa Neural/embriología , Placa Neural/metabolismo , Proteínas de Unión al ARN/genética , Erizos de Mar/embriología , Erizos de Mar/genética , Animales , Desarrollo Embrionario/genética , Endodermo/embriología , Endodermo/metabolismo , Expresión Génica , ARN Mensajero/genética
17.
Methods Mol Biol ; 1128: 147-60, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24567212

RESUMEN

Imaging is a critical tool in neuroscience, and our understanding of the structure and function of sea urchin nervous systems owes much to this approach. In particular, studies of neural development have been facilitated by methods that enable the accurate identification of specific types of neurons. Here we describe methods that have been successfully employed to study neural development in sea urchin embryos. Altering gene expression in part of an embryo is facilitated by injection of reagents into individual blastomeres, which enables studies of cell autonomous effects and single embryo rescue experiments. The simultaneous localization of an in situ RNA hybridization probe and a cell type specific antigen has enabled studies of gene expression in specific types of neurons. Fixatives and antibodies can be capricious; thus, we provide data on preservation of antigens with commonly used fixatives and buffers.


Asunto(s)
Erizos de Mar/embriología , Animales , Blastómeros/fisiología , Embrión no Mamífero/citología , Desarrollo Embrionario , Larva/citología , Sistema Nervioso/citología , Sistema Nervioso/embriología , Erizos de Mar/citología , Técnicas de Cultivo de Tejidos , Fijación del Tejido
18.
Cytoskeleton (Hoboken) ; 70(8): 453-70, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23907936

RESUMEN

An apical tuft, which is observed in a wide range of embryos/larvae of marine invertebrates, is composed of a group of cilia that are longer and less motile than the abundant lateral cilia covering the rest of the embryonic surface. Although the apical tuft has been thought to function as a sensory organ, its molecular composition and roles are poorly understood. Here, we identified a glutathione transferase theta (GSTT) as an abundant and specific component of the apical tuft in sea urchin embryos. The expression of GSTT mRNA increases and becomes limited to the animal plate of the mesenchyme blastula, gastrula, and prism larva. Electron microscopy and tandem mass spectrometry demonstrated that the apical tuft contains almost every axonemal component for ciliary motility. Low concentrations of an inhibitor of glutathione transferase bromosulphophthalein (BSP) induce bending of apical tuft, suggesting that GSTT regulates motility of apical tuft cilia. Embryos treated with BSP swim with normal velocity and trajectories but show less efficiency of changing direction when they collide with an object. These results suggest that GSTT in the apical tuft plays an important role in the mechanical reception for the motility regulation of lateral motile cilia in sea urchin embryos.


Asunto(s)
Cilios/metabolismo , Glutatión Transferasa/metabolismo , Erizos de Mar/metabolismo , Erizos de Mar/fisiología , Animales , Cilios/fisiología , Embrión no Mamífero/metabolismo
19.
Dev Biol ; 363(1): 74-83, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22210002

RESUMEN

Serotonergic neurons differentiate in the neurogenic animal plate ectoderm of the sea urchin embryo. The regulatory mechanisms that control the specification or differentiation of these neurons in the sea urchin embryo are not yet understood, although, after the genome was sequenced, many genes encoding transcription factors expressed in this region were identified. Here, we report that zinc finger homeobox (zfhx1/z81) is expressed in serotonergic neural precursor cells, using double in situ hybridization screening with a serotonergic neural marker, tryptophan 5-hydroxylase (tph) encoding a serotonin synthase that is required for the differentiation of serotonergic neurons. zfhx1/z81 begins to be expressed at gastrula stage in individual cells in the anterior neuroectoderm, some of which also express delta. zfhx1/z81 expression gradually disappears as neural differentiation begins with tph expression. When the translation of Zfhx1/Z81 is blocked by morpholino injection, embryos express neither tph nor the neural marker synaptotagminB in cells of the animal plate, and serotonergic neurons do not differentiate. In contrast, Zfhx1/Z81 morphants do express fez, another neural precursor marker, which appears to function in the initial phase of specification/differentiation of serotonergic neurons. In addition, zfhx1/z81 is one of the targets suppressed in the animal plate by anti-neural signals such as Nodal as well as Delta-Notch. We conclude that Zfhx1/Z81 functions during the specification of individual anterior neural precursors and promotes the expression of tph and synaptotagminB, required for the differentiation of serotonergic neurons.


Asunto(s)
Diferenciación Celular/genética , Genes Homeobox/genética , Hemicentrotus/genética , Neuronas Serotoninérgicas/metabolismo , Dedos de Zinc/genética , Secuencia de Aminoácidos , Animales , Embrión no Mamífero/citología , Embrión no Mamífero/embriología , Embrión no Mamífero/metabolismo , Gástrula/embriología , Gástrula/metabolismo , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Hemicentrotus/embriología , Proteínas de Homeodominio/genética , Hibridación in Situ/métodos , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Modelos Genéticos , Datos de Secuencia Molecular , Proteína Nodal/genética , Receptores Notch/genética , Homología de Secuencia de Aminoácido , Neuronas Serotoninérgicas/citología , Transducción de Señal/genética , Sinaptotagminas/genética , Triptófano Hidroxilasa/genética
20.
Development ; 138(19): 4233-43, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21852402

RESUMEN

Partitioning ectoderm precisely into neurogenic and non-neurogenic regions is an essential step for neurogenesis of almost all bilaterian embryos. Although it is widely accepted that antagonism between BMP and its inhibitors primarily sets up the border between these two types of ectoderm, it is unclear how such extracellular, diffusible molecules create a sharp and precise border at the single-cell level. Here, we show that Fez, a zinc finger protein, functions as an intracellular factor attenuating BMP signaling specifically within the neurogenic region at the anterior end of sea urchin embryos, termed the animal plate. When Fez function is blocked, the size of this neurogenic ectoderm becomes smaller than normal. However, this reduction is rescued in Fez morphants simply by blocking BMP2/4 translation, indicating that Fez maintains the size of the animal plate by attenuating BMP2/4 function. Consistent with this, the gradient of BMP activity along the aboral side of the animal plate, as measured by pSmad1/5/8 levels, drops significantly in cells expressing Fez and this steep decline requires Fez function. Our data reveal that this neurogenic ectoderm produces an intrinsic system that attenuates BMP signaling to ensure the establishment of a stable, well-defined neural territory, the animal plate.


Asunto(s)
Proteínas Morfogenéticas Óseas/química , Regulación del Desarrollo de la Expresión Génica , Proteínas Smad/metabolismo , Factores de Transcripción/fisiología , Dedos de Zinc , Animales , Blástula/metabolismo , Tipificación del Cuerpo/genética , Linaje de la Célula , Ectodermo/metabolismo , Embrión no Mamífero/metabolismo , Inmunohistoquímica/métodos , Modelos Biológicos , Hibridación de Ácido Nucleico , Análisis de Secuencia por Matrices de Oligonucleótidos , Erizos de Mar , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA