Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Development ; 150(4)2023 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-36789950

RESUMO

We show that the zebrafish maternal-effect mutation too much information (tmi) corresponds to zebrafish prc1-like (prc1l), which encodes a member of the MAP65/Ase1/PRC1 family of microtubule-associated proteins. Embryos from tmi homozygous mutant mothers display cytokinesis defects in meiotic and mitotic divisions in the early embryo, indicating that Prc1l has a role in midbody formation during cell division at the egg-to-embryo transition. Unexpectedly, maternal Prc1l function is also essential for the reorganization of vegetal pole microtubules required for the segregation of dorsal determinants. Whereas Prc1 is widely regarded to crosslink microtubules in an antiparallel conformation, our studies provide evidence for an additional function of Prc1l in the bundling of parallel microtubules in the vegetal cortex of the early embryo during cortical rotation and prior to mitotic cycling. These findings highlight common yet distinct aspects of microtubule reorganization that occur during the egg-to-embryo transition, driven by maternal product for the midbody component Prc1l and required for embryonic cell division and pattern formation.


Assuntos
Citocinese , Proteínas Associadas aos Microtúbulos , Peixe-Zebra , Animais , Divisão Celular , Citocinese/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo
2.
Development ; 148(19)2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34463742

RESUMO

In animals, early development is dependent on a pool of maternal factors, both RNA and proteins, which are required for basic cellular processes and cell differentiation until zygotic genome activation. The role of the majority of these maternally expressed factors is not fully understood. By exploiting the biallelic editing ability of CRISPR-Cas9, we identify and characterize maternal-effect genes in a single generation, using a maternal crispant technique. We validated the ability to generate biallelic mutations in the germ line by creating maternal crispants that phenocopied previously characterized maternal-effect genes: birc5b, tmi and mid1ip1. Additionally, by targeting maternally expressed genes of unknown function in zebrafish, we identified two maternal-effect zebrafish genes, kpna7 and fhdc3. The genetic identity of these maternal crispants was confirmed by sequencing haploid progeny from F0 females, which allowed the analysis of newly induced lesions in the maternal germ line. Our studies show that maternal crispants allow for the effective identification and primary characterization of maternal-effect genes in a single generation, facilitating the reverse genetics analysis of maternal factors that drive embryonic development.


Assuntos
Sistemas CRISPR-Cas , Edição de Genes/métodos , Herança Materna , Proteínas de Peixe-Zebra/genética , Animais , Óvulo/citologia , Óvulo/metabolismo , Peixe-Zebra
3.
Development ; 147(24)2020 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-33246928

RESUMO

Heart regeneration in regeneration-competent organisms can be accomplished through the remodeling of gene expression in response to cardiac injury. This dynamic transcriptional response relies on the activities of tissue regeneration enhancer elements (TREEs); however, the mechanisms underlying TREEs are poorly understood. We dissected a cardiac regeneration enhancer in zebrafish to elucidate the mechanisms governing spatiotemporal gene expression during heart regeneration. Cardiac lepb regeneration enhancer (cLEN) exhibits dynamic, regeneration-dependent activity in the heart. We found that multiple injury-activated regulatory elements are distributed throughout the enhancer region. This analysis also revealed that cardiac regeneration enhancers are not only activated by injury, but surprisingly, they are also actively repressed in the absence of injury. Our data identified a short (22 bp) DNA element containing a key repressive element. Comparative analysis across Danio species indicated that the repressive element is conserved in closely related species. The repression mechanism is not operational during embryogenesis and emerges when the heart begins to mature. Incorporating both activation and repression components into the mechanism of tissue regeneration constitutes a new paradigm that might be extrapolated to other regeneration scenarios.


Assuntos
Elementos Facilitadores Genéticos , Traumatismos Cardíacos/genética , Coração/crescimento & desenvolvimento , Regeneração/genética , Animais , Regulação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento/genética , Traumatismos Cardíacos/patologia , Traumatismos Cardíacos/reabilitação , Humanos , Organogênese/genética , Regeneração/fisiologia , Cicatrização/genética , Cicatrização/fisiologia , Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento
4.
Genesis ; 59(10): e23452, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34617657

RESUMO

In many animal species, germ cell specification requires the inheritance of germ plasm, a biomolecular condensate containing maternally derived RNAs and proteins. Most studies of germ plasm composition and function have been performed in widely evolutionarily divergent model organisms, such as Caenorhabditis elegans, Drosophila, Xenopus laevis, and Danio rerio (zebrafish). In zebrafish, 12 RNAs localize to germ plasm at the furrows of the early embryo. Here, we tested for the presence of these RNAs in three additional species within the Danionin clade: Danio kyathit, Danio albolineatus, and Devario aequipinnatus. By visualizing nanos RNA, we find that germ plasm segregation patterns during early embryogenesis are conserved across these species. Ten additional germ plasm RNAs exhibit localization at the furrows of early embryos in all three non-zebrafish Danionin species, consistent with germ plasm localization. One component of zebrafish germ plasm, ca15b, lacked specific localization in embryos of the more distantly related D. aequipinnatus. Our findings show that within a subset of closely related Danionin species, the vast majority of germ plasm RNA components are conserved. At the same time, the lack of ca15b localization in D. aequipinnatus germ plasm highlights the potential for the divergence of germ plasm composition across a restricted phylogenetic space.


Assuntos
Desenvolvimento Embrionário/genética , Evolução Molecular , RNA/genética , Peixe-Zebra/genética , Animais , Caenorhabditis elegans/genética , Sequência Conservada/genética , Drosophila/genética , Embrião não Mamífero , Células Germinativas/crescimento & desenvolvimento , Células Germinativas/metabolismo , Filogenia , RNA/isolamento & purificação , Xenopus laevis/genética
5.
Development ; 145(10)2018 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-29724756

RESUMO

During the early embryonic cell cycles, zebrafish germ plasm ribonucleoparticles (RNPs) gradually multimerize and become recruited to the forming furrows. RNPs multimerization occurs prior to and during furrow initiation, as forming aggregates move outward through their association with the tips of growing interphase astral microtubules. Germ plasm RNPs are also associated with short cortical F-actin. We show that, in embryos mutant for the cytoskeletal regulator mid1ip1l, germ plasm RNPs fail to become recruited to the furrow, accumulating instead at the periphery of the blastodisc. RNP aggregates are associated with zones of mid1ip1l-dependent cyclical local cortical F-actin network enrichments, as well as contractions at both the cortex and the contractile ring. F-actin inhibition in wild-type embryos mimics the RNP peripheral accumulation defect of mid1ip1l mutants. Our studies suggest that a common mechanism underlies distinct steps of germ plasm RNP segregation. At the cortex, this process attenuates microtubule-dependent outward RNP movement to retain RNPs in the blastodisc cortex and allow their recruitment to the furrows. F-actin network contraction likely also facilitates higher-order germ plasm RNP multimerization.


Assuntos
Actinas/metabolismo , Citoplasma/metabolismo , Proteínas do Citoesqueleto/metabolismo , Embrião não Mamífero/embriologia , Ribonucleoproteínas/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Citoesqueleto de Actina/metabolismo , Actinas/antagonistas & inibidores , Animais , Blastodisco/metabolismo , Membrana Celular/metabolismo , Proteínas do Citoesqueleto/genética , Embrião não Mamífero/metabolismo , Células Germinativas/metabolismo , Microtúbulos/metabolismo , Multimerização Proteica/fisiologia , Transporte Proteico/genética , Proteínas de Peixe-Zebra/genética
6.
Development ; 145(10)2018 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-29632136

RESUMO

Zebrafish germ plasm ribonucleoparticles (RNPs) become recruited to furrows of early zebrafish embryos through their association with astral microtubules ends. During the initiation of cytokinesis, microtubules are remodeled into a furrow microtubule array (FMA), which is thought to be analogous to the mammalian midbody involved in membrane abscission. During furrow maturation, RNPs and FMA tubules transition from their original distribution along the furrow to enrichments at the furrow distal ends, which facilitates germ plasm mass compaction. We show that nebel mutants exhibit reduced furrow-associated slow calcium waves (SCWs), caused at least in part by defective enrichment of calcium stores. RNP and FMA distal enrichment mirrors the medial-to-distal polarity of SCWs, and inhibition of calcium release or downstream mediators such as Calmodulin affects RNP and FMA distal enrichment. Blastomeres with reduced or lacking SCWs, such as early blastomeres in nebel mutants and wild-type blastomeres at later stages, exhibit medially bundling microtubules similar to midbodies in other cell types. Our data indicate that SCWs provide medial-to-distal directionality along the furrow to facilitate germ plasm RNP enrichment at the furrow ends.


Assuntos
Sinalização do Cálcio/fisiologia , Citoplasma/metabolismo , Microtúbulos/metabolismo , Ribonucleoproteínas/metabolismo , Peixe-Zebra/embriologia , Actinas/metabolismo , Animais , Blastômeros/metabolismo , Padronização Corporal/genética , Padronização Corporal/fisiologia , Cálcio/metabolismo , Calmodulina/metabolismo , Fase de Clivagem do Zigoto/fisiologia , Citocinese/fisiologia
7.
Dev Dyn ; 248(4): 306-318, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30741457

RESUMO

BACKGROUND: In zebrafish and many other organisms, specification of primordial germ cells (PGCs) requires the transmission of maternally-derived germ plasm. Zebrafish germ plasm ribonucleoparticles (RNPs) aggregate along the cleavage furrows during the first several cell cycles, segregate asymmetrically during the cleavage stages, and undergo cytoplasmic dispersal in the late blastula. RESULTS: For all tested germ plasm RNAs [carbonic anhydrase 15b (ca15b), deleted in azoospermia-like (dazl), dead end (dnd), nanos 3 (nos3), regulator of G-protein signaling14a (rgs14a), and vasa/DEAD box polypeptide 4 (vasa/ddx4)], RNPs are homotypic (containing a single RNA type), with RNPs packing tightly yet remaining distinct within germ plasm aggregates. Homotypic clustering of RNAs within RNPs is observed before aggregation in the cortex and is maintained through germ plasm recruitment, asymmetric segregation and RNP dispersal. We also identify a step of germ plasm fragmentation during the cleavage stages that precedes RNP dispersal. CONCLUSIONS: Our findings suggest that germ plasm aggregates act as subcellular compartments that temporarily collect and carry single RNA-type RNPs from fertilization until their cytoplasmic dispersal in PGCs at the end of the blastula period, and describe a previously unknown fragmentation step that allows for an increase in the pool of germ plasm-carrying cells, presumably PGCs. Developmental Dynamics 248:306-318, 2019. © 2019 Wiley Periodicals, Inc.


Assuntos
Embrião não Mamífero , RNA/metabolismo , Animais , Blástula , Citoplasma/metabolismo , Embrião não Mamífero/ultraestrutura , Células Germinativas/ultraestrutura , Peixe-Zebra/embriologia , Peixe-Zebra/genética
8.
Development ; 143(9): 1585-99, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-26965374

RESUMO

Embryos from females homozygous for a recessive maternal-effect mutation in the gene aura exhibit defects including reduced cortical integrity, defective cortical granule (CG) release upon egg activation, failure to complete cytokinesis, and abnormal cell wound healing. We show that the cytokinesis defects are associated with aberrant cytoskeletal reorganization during furrow maturation, including abnormal F-actin enrichment and microtubule reorganization. Cortical F-actin prior to furrow formation fails to exhibit a normal transition into F-actin-rich arcs, and drug inhibition is consistent with aura function promoting F-actin polymerization and/or stabilization. In mutants, components of exocytic and endocytic vesicles, such as Vamp2, Clathrin and Dynamin, are sequestered in unreleased CGs, indicating a need for CG recycling in the normal redistribution of these factors. However, the exocytic targeting factor Rab11 is recruited to the furrow plane normally at the tip of bundling microtubules, suggesting an alternative anchoring mechanism independent of membrane recycling. A positional cloning approach indicates that the mutation in aura is associated with a truncation of Mid1 interacting protein 1 like (Mid1ip1l), previously identified as an interactor of the X-linked Opitz G/BBB syndrome gene product Mid1. A Cas9/CRISPR-induced mutant allele in mid1ip1l fails to complement the originally isolated aura maternal-effect mutation, confirming gene assignment. Mid1ip1l protein localizes to cortical F-actin aggregates, consistent with a direct role in cytoskeletal regulation. Our studies indicate that maternally provided aura (mid1ip1l) acts during the reorganization of the cytoskeleton at the egg-to-embryo transition and highlight the importance of cytoskeletal dynamics and membrane recycling during this developmental period.


Assuntos
Citocinese/genética , Proteínas do Citoesqueleto/genética , Citoesqueleto/metabolismo , Microtúbulos/metabolismo , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/embriologia , Actinas/metabolismo , Animais , Clatrina/metabolismo , Grânulos Citoplasmáticos/metabolismo , Proteínas do Citoesqueleto/metabolismo , Dinaminas/metabolismo , Proteína 2 Associada à Membrana da Vesícula/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo
9.
Adv Exp Med Biol ; 953: 117-171, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27975272

RESUMO

The pattern of the earliest cell divisions in a vertebrate embryo lays the groundwork for later developmental events such as gastrulation, organogenesis, and overall body plan establishment. Understanding these early cleavage patterns and the mechanisms that create them is thus crucial for the study of vertebrate development. This chapter describes the early cleavage stages for species representing ray-finned fish, amphibians, birds, reptiles, mammals, and proto-vertebrate ascidians and summarizes current understanding of the mechanisms that govern these patterns. The nearly universal influence of cell shape on orientation and positioning of spindles and cleavage furrows and the mechanisms that mediate this influence are discussed. We discuss in particular models of aster and spindle centering and orientation in large embryonic blastomeres that rely on asymmetric internal pulling forces generated by the cleavage furrow for the previous cell cycle. Also explored are mechanisms that integrate cell division given the limited supply of cellular building blocks in the egg and several-fold changes of cell size during early development, as well as cytoskeletal specializations specific to early blastomeres including processes leading to blastomere cohesion. Finally, we discuss evolutionary conclusions beginning to emerge from the contemporary analysis of the phylogenetic distributions of cleavage patterns. In sum, this chapter seeks to summarize our current understanding of vertebrate early embryonic cleavage patterns and their control and evolution.


Assuntos
Evolução Biológica , Divisão Celular/genética , Desenvolvimento Embrionário/genética , Vertebrados/embriologia , Animais , Ciclo Celular/genética , Fase de Clivagem do Zigoto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Mamíferos , Fuso Acromático/genética , Vertebrados/genética
10.
PLoS Genet ; 10(6): e1004422, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24967891

RESUMO

Maternal homozygosity for three independent mutant hecate alleles results in embryos with reduced expression of dorsal organizer genes and defects in the formation of dorsoanterior structures. A positional cloning approach identified all hecate mutations as stop codons affecting the same gene, revealing that hecate encodes the Glutamate receptor interacting protein 2a (Grip2a), a protein containing multiple PDZ domains known to interact with membrane-associated factors including components of the Wnt signaling pathway. We find that grip2a mRNA is localized to the vegetal pole of the oocyte and early embryo, and that during egg activation this mRNA shifts to an off-center vegetal position corresponding to the previously proposed teleost cortical rotation. hecate mutants show defects in the alignment and bundling of microtubules at the vegetal cortex, which result in defects in the asymmetric movement of wnt8a mRNA as well as anchoring of the kinesin-associated cargo adaptor Syntabulin. We also find that, although short-range shifts in vegetal signals are affected in hecate mutant embryos, these mutants exhibit normal long-range, animally directed translocation of cortically injected dorsal beads that occurs in lateral regions of the yolk cortex. Furthermore, we show that such animally-directed movement along the lateral cortex is not restricted to a single arc corresponding to the prospective dorsal region, but occur in multiple meridional arcs even in opposite regions of the embryo. Together, our results reveal a role for Grip2a function in the reorganization and bundling of microtubules at the vegetal cortex to mediate a symmetry-breaking short-range shift corresponding to the teleost cortical rotation. The slight asymmetry achieved by this directed process is subsequently amplified by a general cortical animally-directed transport mechanism that is neither dependent on hecate function nor restricted to the prospective dorsal axis.


Assuntos
Padronização Corporal/genética , Proteínas de Transporte/genética , Desenvolvimento Embrionário/genética , Proteínas de Xenopus/genética , Peixe-Zebra/genética , Alelos , Animais , Proteínas de Transporte/biossíntese , Proteínas do Citoesqueleto/genética , Citoesqueleto/genética , Embrião não Mamífero , Regulação da Expressão Gênica no Desenvolvimento , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas Associadas aos Microtúbulos/genética , Microtúbulos/genética , Oócitos/crescimento & desenvolvimento , Oócitos/metabolismo , Domínios PDZ/genética , Fenótipo , RNA Mensageiro/biossíntese , Proteínas Wnt/genética , Xenopus , Proteínas de Xenopus/biossíntese , Peixe-Zebra/embriologia , Proteínas de Peixe-Zebra/genética
11.
PLoS Genet ; 9(4): e1003448, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23637620

RESUMO

Microtubule-microfilament interactions are important for cytokinesis and subcellular localization of proteins and mRNAs. In the early zebrafish embryo, astral microtubule-microfilament interactions also facilitate a stereotypic segregation pattern of germ plasm ribonucleoparticles (GP RNPs), which is critical for their eventual selective inheritance by germ cells. The precise mechanisms and molecular mediators for both cytoskeletal interactions and GP RNPs segregation are the focus of intense research. Here, we report the molecular identification of a zebrafish maternal-effect mutation motley as Birc5b, a homolog of the mammalian Chromosomal Passenger Complex (CPC) component Survivin. The meiosis and mitosis defects in motley/birc5b mutant embryos are consistent with failed CPC function, and additional defects in astral microtubule remodeling contribute to failures in the initiation of cytokinesis furrow ingression. Unexpectedly, the motley/birc5b mutation also disrupts cortical microfilaments and GP RNP aggregation during early cell divisions. Birc5b localizes to the tips of astral microtubules along with polymerizing cortical F-actin and the GP RNPs. Mutant Birc5b co-localizes with cortical F-actin and GP RNPs, but fails to associate with astral microtubule tips, leading to disorganized microfilaments and GP RNP aggregation defects. Thus, maternal Birc5b localizes to astral microtubule tips and associates with cortical F-actin and GP RNPs, potentially linking the two cytoskeletons to mediate microtubule-microfilament reorganization and GP RNP aggregation during early embryonic cell cycles in zebrafish. In addition to the known mitotic function of CPC components, our analyses reveal a non-canonical role for an evolutionarily conserved CPC protein in microfilament reorganization and germ plasm aggregation.


Assuntos
Citoesqueleto de Actina , Peixe-Zebra , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Animais , Citoesqueleto/metabolismo , Microtúbulos/metabolismo , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
12.
Dev Dyn ; 244(10): 1300-12, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26249657

RESUMO

BACKGROUND: Whole genome duplication is a useful genetic tool because it allows immediate and complete genetic homozygosity in gynogenetic offspring. A whole genome duplication method in zebrafish, Heat Shock, involves a heat pulse in the period 13-15 min postfertilization (mpf) to inhibit cytokinesis of the first mitotic cycle. However, Heat Shock produces a relatively low yield of gynogenotes. RESULTS: A heat pulse at a later time point during the first cell cycle (22 mpf, HS2) results in a high (>80%) frequency of embryos exhibiting a precise one-cell division stall during the second cell cycle, inducing whole genome duplication. Coupled with haploid production, HS2 generates viable gynogenetic diploids with yields up to 4 times higher than those achieved through standard Heat Shock. The cell cycle delay also causes blastomere cleavage pattern variations, supporting a role for cytokinesis in spindle orientation during the following cell cycle. CONCLUSIONS: Our studies provide a new tool for whole genome duplication, induced gynogenesis, and cleavage pattern alteration in zebrafish, based on a time period before the initiation of cell division that is sensitive to temperature-mediated interference with centrosome duplication. Targeting of this period may also facilitate genetic and developmental manipulations in other organisms.


Assuntos
Centrossomo/fisiologia , Fase de Clivagem do Zigoto , Embriologia/métodos , Técnicas Genéticas , Ploidias , Animais , Ciclo Celular , Homozigoto , Temperatura Alta , Mutação , Peixe-Zebra
13.
Dev Dyn ; 242(1): 44-52, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23074011

RESUMO

In animals, females deposit gene products into developing oocytes, which drive early cellular events in embryos immediately after fertilization. As maternal gene products are present before fertilization, the functional manipulation of maternal genes is often challenging to implement, requiring gene expression or targeting during oogenesis. Maternal expression can be achieved through transgenesis, but transgenic approaches are time consuming and subject to undesired epigenetic effects. Here, we have implemented in vitro culturing of experimentally manipulated immature oocytes to study maternal gene contribution to early embryonic development in the zebrafish. We demonstrate phenotypic rescue of a maternal-effect mutation by expressing wild-type product in cultured oocytes. We also generate loss-of-function phenotypes in embryos through either the expression of a dominant-negative transcript or injection of translation-blocking morpholino oligonucleotides. Finally, we demonstrate subcellular localization during the early cell divisions immediately after fertilization of an exogenously provided maternal product fused to a fluorescent protein. These manipulations extend the potential to carry out genetic and imaging studies of zebrafish maternal genes during the egg-to-embryo transition.


Assuntos
Técnicas de Cultura de Células/métodos , Técnicas de Transferência de Genes , Oócitos/metabolismo , RNA Mensageiro Estocado/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Animais , Primers do DNA/genética , Feminino , Fertilização in vitro , Microinjeções , Microscopia Confocal , Morfolinos/administração & dosagem , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
Dev Biol ; 353(2): 275-89, 2011 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-21396359

RESUMO

Early animal embryonic development requires maternal products that drive developmental processes prior to the activation of the zygotic genome at the mid-blastula transition. During and after this transition, maternal products may continue to act within incipient zygotic developmental programs. Mechanisms that control maternally-inherited products to spatially and temporally restrict developmental responses remain poorly understood, but necessarily depend on posttranscriptional regulation. We report the functional analysis and molecular identification of the zebrafish maternal-effect gene mission impossible (mis). Our studies suggest requirements for maternally-derived mis function in events that occur during gastrulation, including cell movement and the activation of some endodermal target genes. Cell transplantation experiments show that the cell movement defect is cell autonomous. Within the endoderm induction pathway, mis is not required for the activation of early zygotic genes, but is essential to implement nodal activity downstream of casanova/sox 32 but upstream of sox17 expression. Activation of nodal signaling in blastoderm explants shows that the requirement for mis function in endoderm gene induction is independent of the underlying yolk cell. Positional cloning of mis, including genetic rescue and complementation analysis, shows that it encodes the DEAH-box RNA helicase Dhx16, shown in other systems to act in RNA regulatory processes such as splicing and translational control. Analysis of a previously identified insertional dhx16 mutation shows that the zygotic component of this gene is also essential for embryonic viability. Our studies provide a striking example of the interweaving of maternal and zygotic genetic functions during the egg-to-embryo transition. Maternal RNA helicases have long been known to be involved in the development of the animal germ line, but our findings add to growing evidence that these factors may also control specific gene expression programs in somatic tissues.


Assuntos
RNA Helicases/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Movimento Celular/genética , Primers do DNA/genética , Endoderma/embriologia , Endoderma/metabolismo , Feminino , Gastrulação/genética , Regulação da Expressão Gênica no Desenvolvimento , Modelos Biológicos , Dados de Sequência Molecular , Mutação , Ligantes da Sinalização Nodal/genética , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Peixe-Zebra/metabolismo , Zigoto/metabolismo
15.
Adv Exp Med Biol ; 953: E1, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28299712
16.
PLoS Genet ; 5(6): e1000518, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19543364

RESUMO

Females homozygous for a mutation in cellular island (cei) produce embryos with defects in cytokinesis during early development. Analysis of the cytoskeletal events associated with furrow formation reveal that these defects include a general delay in furrow initiation as well as a complete failure to form furrow-associated structures in distal regions of the blastodisc. A linkage mapping-based candidate gene approach, including transgenic rescue, shows that cei encodes the zebrafish Aurora B kinase homologue. Genetic complementation analysis between the cei mutation and aurB zygotic lethal mutations corroborate gene assignment and reveal a complex nature of the maternal-effect cei allele, which appears to preferentially affect a function important for cytokinesis in the early blastomeres. Surprisingly, in cei mutant embryos a short yet otherwise normal furrow forms in the center of the blastodisc. Furrow formation is absent throughout the width of the blastodisc in cei mutant embryos additionally mutant for futile cycle, which lack a spindle apparatus, showing that the residual furrow signal present in cei mutants is derived from the mitotic spindle. Our analysis suggests that partially redundant signals derived from the spindle and astral apparatus mediate furrow formation in medial and distal regions of the early embryonic blastomeres, respectively, possibly as a spatial specialization to achieve furrow formation in these large cells. In addition, our data also suggest a role for Cei/AurB function in the reorganization of the furrow-associated microtubules in both early cleavage- and somite-stage embryos. In accordance with the requirement for cei/aurB in furrow induction in the early cleavage embryo, germ plasm recruitment to the forming furrow is also affected in embryos lacking normal cei/aurB function.


Assuntos
Citocinese , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo , Animais , Aurora Quinases , Padronização Corporal , Embrião não Mamífero/citologia , Embrião não Mamífero/enzimologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Proteínas Serina-Treonina Quinases/genética , Especificidade da Espécie , Fuso Acromático/enzimologia , Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento , Proteínas de Peixe-Zebra/genética
17.
Dev Biol ; 341(2): 444-58, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20226780

RESUMO

Chordin is the prototype of a group of cysteine-rich domain-containing proteins that bind and modulate signaling of various TGFbeta-like ligands. Chordin-like 1 and 2 (CHL1 and 2) are two members of this group that have been described in human, mouse, and chick. However, in vivo roles for CHL1 and 2 in early development are unknown due to lack of loss-of-function analysis. Here we identify and characterize zebrafish, Danio rerio, CHL (Chl). The chl gene is on a region of chromosome 21 syntenic with the area of murine chromosome 7 bearing the CHL2 gene. Inability to identify a separate zebrafish gene corresponding to the mammalian CHL1 gene suggests that Chl may serve roles in zebrafish distributed between CHL1 and CHL2 in other species. Chl is a maternal factor that is also zygotically expressed later in development and has spatiotemporal expression patterns that differ from but overlap those of zebrafish chordin (Chd), suggesting differences but also possible overlap in developmental roles of the two proteins. Chl, like Chd, dorsalizes embryos upon overexpression and is cleaved by BMP1, which antagonizes this activity. Loss-of-function experiments demonstrate that Chl serves as a BMP antagonist with functions that overlap and are redundant with those of Chd in forming the dorsoventral axis.


Assuntos
Padronização Corporal , Glicoproteínas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo , Sequência de Aminoácidos , Animais , Proteína Morfogenética Óssea 1/metabolismo , Embrião não Mamífero/metabolismo , Glicoproteínas/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Dados de Sequência Molecular , Alinhamento de Sequência , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
18.
Front Cell Dev Biol ; 9: 730332, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34604230

RESUMO

The differentiation of primordial germ cells (PGCs) occurs during early embryonic development and is critical for the survival and fitness of sexually reproducing species. Here, we review the two main mechanisms of PGC specification, induction, and preformation, in the context of four model vertebrate species: mouse, axolotl, Xenopus frogs, and zebrafish. We additionally discuss some notable molecular characteristics shared across PGC specification pathways, including the shared expression of products from three conserved germline gene families, DAZ (Deleted in Azoospermia) genes, nanos-related genes, and DEAD-box RNA helicases. Then, we summarize the current state of knowledge of the distribution of germ cell determination systems across kingdom Animalia, with particular attention to vertebrate species, but include several categories of invertebrates - ranging from the "proto-vertebrate" cephalochordates to arthropods, cnidarians, and ctenophores. We also briefly highlight ongoing investigations and potential lines of inquiry that aim to understand the evolutionary relationships between these modes of specification.

19.
Methods Mol Biol ; 2218: 219-244, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33606235

RESUMO

Zebrafish embryos, with their large size (>0.5 mm) and accessibility, are valuable tools for investigating core cellular processes. Many of those processes, such as cell division, asymmetric inheritance of cellular components, and structural dynamics involved in cell motility and morphology rely on cytoskeletal rearrangements and associated macromolecules. In addition to the protein-rich cytoskeleton, the early embryo is packed with maternally deposited RNA, which serves essential roles in establishing cell polarity, cell fate, and cell organization. Here, we present methods for visualizing endogenous RNA along with cytoskeletal structures, including microtubules and filamentous actin (F-actin) in the context of an intact vertebrate embryo. Each of the four protocols described herein (embryo fixation, RNA probe design/synthesis, double fluorescent in situ hybridization with tubulin immunofluorescence, and fluorescent in situ hybridization with phalloidin labeling of F-actin) are intended for optimal preservation and visualization of both the cytoskeleton and RNAs of interest. These methods can also be modified and applied to a broad range of other uses.


Assuntos
Citoesqueleto/metabolismo , Embrião não Mamífero/metabolismo , Hibridização in Situ Fluorescente/métodos , RNA/metabolismo , Peixe-Zebra/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Animais , Divisão Celular/fisiologia , Proteínas do Citoesqueleto/metabolismo , Microtúbulos/metabolismo , Proteínas de Peixe-Zebra/metabolismo
20.
J Vis Exp ; (178)2021 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-35001909

RESUMO

Early development depends on a pool of maternal factors incorporated into the mature oocyte during oogenesis that perform all cellular functions necessary for development until zygotic genome activation. Typically, genetic targeting of these maternal factors requires an additional generation to identify maternal-effect phenotypes, hindering the ability to determine the role of maternally-expressed genes during development. The discovery of the biallelic editing capabilities of CRISPR-Cas9 has allowed screening of embryonic phenotypes in somatic tissues of injected embryos or "crispants," augmenting the understanding of the role zygotically-expressed genes play in developmental programs. This article describes a protocol that is an extension of the crispant method. In this method, the biallelic editing of germ cells allows for the isolation of a maternal-effect phenotype in a single generation, or "maternal crispants." Multiplexing guide RNAs to a single target promotes the efficient production of maternal crispants, while sequence analysis of maternal crispant haploids provides a simple method to corroborate genetic lesions that produce a maternal-effect phenotype. The use of maternal crispants supports the rapid identification of essential maternally-expressed genes, thus facilitating the understanding of early development.


Assuntos
RNA Guia de Cinetoplastídeos , Zigoto , Sistemas CRISPR-Cas , Genoma , Células Germinativas , Oogênese , RNA Guia de Cinetoplastídeos/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA