Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
1.
PLoS Genet ; 20(7): e1011343, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39052672

RESUMEN

Maternally-loaded factors in the egg accumulate during oogenesis and are essential for the acquisition of oocyte and egg developmental competence to ensure the production of viable embryos. However, their molecular nature and functional importance remain poorly understood. Here, we present a collection of 9 recessive maternal-effect mutants identified in a zebrafish forward genetic screen that reveal unique molecular insights into the mechanisms controlling the vertebrate oocyte-to-embryo transition. Four genes, over easy, p33bjta, poached and black caviar, were found to control initial steps in yolk globule sizing and protein cleavage during oocyte maturation that act independently of nuclear maturation. The krang, kazukuram, p28tabj, and spotty genes play distinct roles in egg activation, including cortical granule biology, cytoplasmic segregation, the regulation of microtubule organizing center assembly and microtubule nucleation, and establishing the basic body plan. Furthermore, we cloned two of the mutant genes, identifying the over easy gene as a subunit of the Adaptor Protein complex 5, Ap5m1, which implicates it in regulating intracellular trafficking and yolk vesicle formation. The novel maternal protein Krang/Kiaa0513, highly conserved in metazoans, was discovered and linked to the function of cortical granules during egg activation. These mutant genes represent novel genetic entry points to decipher the molecular mechanisms functioning in the oocyte-to-embryo transition, fertility, and human disease. Additionally, our genetic adult screen not only contributes to the existing knowledge in the field but also sets the basis for future investigations. Thus, the identified maternal genes represent key players in the coordination and execution of events prior to fertilization.


Asunto(s)
Oocitos , Oogénesis , Proteínas de Pez Cebra , Pez Cebra , Animales , Pez Cebra/genética , Oocitos/metabolismo , Oocitos/crecimiento & desarrollo , Oogénesis/genética , Femenino , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Regulación del Desarrollo de la Expresión Génica , Herencia Materna/genética , Mutación , Embrión no Mamífero , Desarrollo Embrionario/genética
3.
J Bone Miner Res ; 38(9): 1364-1385, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37329499

RESUMEN

Fibrodysplasia ossificans progressiva (FOP) is a rare human genetic condition characterized by altered skeletal development and extraskeletal bone formation. All cases of FOP are caused by mutations in the type I bone morphogenetic protein (BMP) receptor gene ACVR1 that result in overactivation of the BMP signaling pathway. Activation of the wild-type ACVR1 kinase requires assembly of a tetrameric type I and II BMP receptor complex followed by phosphorylation of the ACVR1 GS domain by type II BMP receptors. Previous studies showed that the FOP-mutant ACVR1-R206H required type II BMP receptors and presumptive glycine/serine-rich (GS) domain phosphorylation for overactive signaling. Structural modeling of the ACVR1-R206H mutant kinase domain supports the idea that FOP mutations alter the conformation of the GS domain, but it is unclear how this leads to overactive signaling. Here we show, using a developing zebrafish embryo BMP signaling assay, that the FOP-mutant receptors ACVR1-R206H and -G328R have reduced requirements for GS domain phosphorylatable sites to signal compared to wild-type ACVR1. Further, ligand-independent and ligand-dependent signaling through the FOP-mutant ACVR1 receptors have distinct GS domain phosphorylatable site requirements. ACVR1-G328R showed increased GS domain serine/threonine requirements for ligand-independent signaling compared to ACVR1-R206H, whereas it exhibited reduced serine/threonine requirements for ligand-dependent signaling. Remarkably, while ACVR1-R206H does not require the type I BMP receptor partner, Bmpr1, to signal, a ligand-dependent GS domain mutant of ACVR1-R206H could signal independently of Bmpr1 only when Bmp7 ligand was overexpressed. Of note, unlike human ACVR1-R206H, the zebrafish paralog Acvr1l-R203H does not show increased signaling activity. However, in domain-swapping studies, the human kinase domain, but not the human GS domain, was sufficient to confer overactive signaling to the Acvr1l-R203H receptor. Together these results reflect the importance of GS domain activation and kinase domain functions in regulating ACVR1 signaling and identify mechanisms of reduced regulatory constraints conferred by FOP mutations. © 2023 American Society for Bone and Mineral Research (ASBMR).


Asunto(s)
Miositis Osificante , Animales , Humanos , Receptores de Activinas Tipo I/genética , Receptores de Activinas Tipo I/metabolismo , Receptores de Proteínas Morfogenéticas Óseas/genética , Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Ligandos , Mutación/genética , Miositis Osificante/genética , Miositis Osificante/metabolismo , Transducción de Señal/genética , Pez Cebra/metabolismo
4.
BMC Biol ; 21(1): 16, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36726183

RESUMEN

BACKGROUND: Proteins of the TGFß family, which are largely studied as homodimers, are also known to form heterodimers with biological activity distinct from their component homodimers. For instance, heterodimers of bone morphogenetic proteins, including BMP2/BMP7, BMP2/BMP6, and BMP9/BMP10, among others, have illustrated the importance of these heterodimeric proteins within the context of TGFß signaling. RESULTS: In this study, we have determined that mature GDF5 can be combined with mature BMP2 or BMP4 to form BMP2/GDF5 and BMP4/GDF5 heterodimer. Intriguingly, this combination of a BMP2 or BMP4 monomer, which exhibit high affinity to heparan sulfate characteristic to the BMP class, with a GDF5 monomer with low heparan sulfate affinity produces a heterodimer with an intermediate affinity. Using heparin affinity chromatography to purify the heterodimeric proteins, we then determined that both the BMP2/GDF5 and BMP4/GDF5 heterodimers consistently signaled potently across an array of cellular and in vivo systems, while the activities of their homodimeric counterparts were more context dependent. These differences were likely driven by an increase in the combined affinities for the type 1 receptors, Alk3 and Alk6. Furthermore, the X-ray crystal structure of BMP2/GDF5 heterodimer was determined, highlighting the formation of two asymmetric type 1 receptor binding sites that are both unique relative to the homodimers. CONCLUSIONS: Ultimately, this method of heterodimer production yielded a signaling molecule with unique properties relative to the homodimeric ligands, including high affinity to multiple type 1 and moderate heparan binding affinity.


Asunto(s)
Proteína Morfogenética Ósea 2 , Proteínas Morfogenéticas Óseas , Proteínas Morfogenéticas Óseas/metabolismo , Proteína Morfogenética Ósea 2/genética , Proteína Morfogenética Ósea 2/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Unión Proteica , Proteínas Portadoras/metabolismo , Heparitina Sulfato
5.
Curr Top Dev Biol ; 150: 149-209, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35817502

RESUMEN

Body axis formation in vertebrate development entails the remarkable feat of patterning a myriad of specialized cell types and organ progenitors from a field of unpatterned, multipotent cells. This feat is achieved largely by secreted cell-cell signaling molecules, enabling cells at different positions within the embryo to adopt distinct fates. During patterning of the vertebrate embryonic axes, a multitude of cell fates is induced by a surprisingly small set of signaling pathways: Wnt, Nodal, Bone Morphogenetic Protein (BMP), and Fibroblast Growth Factor (FGF) signaling. These signals function as morphogens, specifying multiple cell fates in a concentration-dependent mechanism, and must therefore be distributed non-uniformly throughout the embryo. A primary signaling center that sets up spatial asymmetries in these signaling pathways to break the symmetry of the vertebrate embryo is known as the dorsal organizer. Discovered nearly a century ago by Hilde Mangold and Hans Spemann in the newt, the organizer has the remarkable ability to induce a secondary body axis when grafted ectopically into a host embryo. Here, we review the cell-cell signaling pathways that control the establishment of the dorsal organizer and its inductive functions in the zebrafish Danio rerio, a vertebrate model highly amenable to genetic manipulation. The organizer's remarkable inductive abilities continue to provide a fascinating source of scientific inquiry in the field of developmental biology.


Asunto(s)
Tipificación del Cuerpo , Pez Cebra , Animales , Tipificación del Cuerpo/genética , Proteínas Morfogenéticas Óseas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Transducción de Señal , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
6.
Front Cell Dev Biol ; 10: 826892, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35733854

RESUMEN

Oogenesis produces functional eggs and is essential for fertility, embryonic development, and reproduction. The zebrafish ovary is an excellent model to study oogenesis in vertebrates, and recent studies have identified multiple regulators in oocyte development through forward genetic screens, as well as reverse genetics by CRISPR mutagenesis. However, many developmental steps in oogenesis, in zebrafish and other species, remain poorly understood, and their underlying mechanisms are unknown. Here, we take a genomic approach to systematically uncover biological activities throughout oogenesis. We performed transcriptomic analysis on five stages of oogenesis, from the onset of oocyte differentiation through Stage III, which precedes oocyte maturation. These transcriptomes revealed thousands of differentially expressed genes across stages of oogenesis. We analyzed trends of gene expression dynamics along oogenesis, as well as their expression in pair-wise comparisons between stages. We determined their functionally enriched terms, identifying uniquely characteristic biological activities in each stage. These data identified two prominent developmental phases in oocyte differentiation and traced the accumulation of maternally deposited embryonic regulator transcripts in the developing oocyte. Our analysis provides the first molecular description for oogenesis in zebrafish, which we deposit online as a resource for the community. Further, the presence of multiple gene paralogs in zebrafish, and the exclusive curation by many bioinformatic tools of the single paralogs present in humans, challenge zebrafish genomic analyses. We offer an approach for converting zebrafish gene name nomenclature to the human nomenclature for supporting genomic analyses generally in zebrafish. Altogether, our work provides a valuable resource as a first step to uncover oogenesis mechanisms and candidate regulators and track accumulating transcripts of maternal regulators of embryonic development.

7.
Dev Biol ; 484: 1-11, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35065906

RESUMEN

The Balbiani body (Bb) is the first marker of polarity in vertebrate oocytes. The Bb is a conserved structure found in diverse animals including insects, fish, amphibians, and mammals. During early zebrafish oogenesis, the Bb assembles as a transient aggregate of mRNA, proteins, and membrane-bound organelles at the presumptive vegetal side of the oocyte. As the early oocyte develops, the Bb appears to grow slowly, until at the end of stage I of oogenesis it disassembles and deposits its cargo of localized mRNAs and proteins. In fish and frogs, this cargo includes the germ plasm as well as gene products required to specify dorsal tissues of the future embryo. We demonstrate that the Bb is a stable, solid structure that forms a size exclusion barrier similar to other biological hydrogels. Despite its central role in oocyte polarity, little is known about the mechanism behind the Bb's action. Analysis of the few known protein components of the Bb is insufficient to explain how the Bb assembles, translocates, and disassembles. We isolated Bbs from zebrafish oocytes and performed mass spectrometry to define the Bb proteome. We successfully identified 77 proteins associated with the Bb sample, including known Bb proteins and novel RNA-binding proteins. In particular, we identified Cirbpa and Cirbpb, which have both an RNA-binding domain and a predicted self-aggregation domain. In stage I oocytes, Cirbpa and Cirbpb localize to the Bb rather than the nucleus (as in somatic cells), indicating that they may have a specialized function in the germ line. Both the RNA-binding domain and the self-aggregation domain are sufficient to localize to the Bb, suggesting that Cirbpa and Cirbpb interact with more than just their mRNA targets within the Bb. We propose that Cirbp proteins crosslink mRNA cargo and proteinaceous components of the Bb as it grows. Beyond Cirbpa and Cirbpb, our proteomics dataset presents many candidates for further study, making it a valuable resource for building a comprehensive mechanism for Bb function at a protein level.


Asunto(s)
Proteínas de Pez Cebra , Pez Cebra , Animales , Polaridad Celular/genética , Mamíferos/metabolismo , Oocitos/metabolismo , Oogénesis/genética , Orgánulos/metabolismo , Proteómica , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
8.
Development ; 148(24)2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34913466

RESUMEN

In the 1990s, labs on both sides of the Atlantic performed the largest genetic mutagenesis screen at that time using an emerging model organism: the zebrafish. Led by Christiane Nüsslein-Volhard in Tübingen, Germany, and Wolfgang Driever in Boston, USA, these colossal screens culminated in 1996 with the publication of 37 articles in a special issue of Development, which remains the journal's largest issue to this day. To celebrate the anniversary of the zebrafish issue and reflect on the 25 years since its publication, five zebrafish researchers share what the issue means to them, how it has contributed to their career and its impact on the zebrafish community.


Asunto(s)
Modelos Animales , Mutagénesis/genética , Pez Cebra/genética , Animales , Humanos
9.
Front Cell Dev Biol ; 9: 753642, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34820378

RESUMEN

Maternal factors which accumulate and establish oocyte polarity during the early stages of oogenesis play key roles in embryonic development, as well as germ cell formation. However, vertebrate oogenesis, especially early stages of oogenesis, is not well understood due to the difficulty of accessing these oocytes and the lack of analytical methods. Here, we report on a microinjection method for analyzing zebrafish early-stage oocytes and some artifacts to be aware of when performing oocyte injections or analyzing oocytes. Using this method, we successfully injected mRNAs encoding fluorescent-tagged proteins into early-stage oocytes and observed subcellular localization in the live oocytes. This method is expected to advance the functional analysis of genes involved in oogenesis.

10.
PLoS Comput Biol ; 17(9): e1009422, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34591841

RESUMEN

Numerous stages of organismal development rely on the cellular interpretation of gradients of secreted morphogens including members of the Bone Morphogenetic Protein (BMP) family through transmembrane receptors. Early gradients of BMPs drive dorsal/ventral patterning throughout the animal kingdom in both vertebrates and invertebrates. Growing evidence in Drosophila, zebrafish, murine and other systems suggests that BMP ligand heterodimers are the primary BMP signaling ligand, even in systems in which mixtures of BMP homodimers and heterodimers are present. Signaling by heterodimers occurs through a hetero-tetrameric receptor complex comprising of two distinct type one BMP receptors and two type II receptors. To understand the system dynamics and determine whether kinetic assembly of heterodimer-heterotetramer BMP complexes is favored, as compared to other plausible BMP ligand-receptor configurations, we developed a kinetic model for BMP tetramer formation based on current measurements for binding rates and affinities. We find that contrary to a common hypothesis, heterodimer-heterotetramer formation is not kinetically favored over the formation of homodimer-tetramer complexes under physiological conditions of receptor and ligand concentrations and therefore other mechanisms, potentially including differential kinase activities of the formed heterotetramer complexes, must be the cause of heterodimer-heterotetramer signaling primacy. Further, although BMP complex assembly favors homodimer and homomeric complex formation over a wide range of parameters, ignoring these signals and instead relying on the heterodimer improves the range of morphogen interpretation in a broad set of conditions, suggesting a performance advantage for heterodimer signaling in patterning multiple cell types in a gradient.


Asunto(s)
Proteínas Morfogenéticas Óseas/química , Proteínas Morfogenéticas Óseas/metabolismo , Modelos Biológicos , Animales , Fenómenos Biofísicos , Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Biología Computacional , Simulación por Computador , Ligandos , Modelos Moleculares , Morfogénesis , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Transducción de Señal
11.
Development ; 148(7)2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33795238

RESUMEN

Pattern formation by bone morphogenetic proteins (BMPs) demonstrates remarkable plasticity and utility in several contexts, such as early embryonic development, tissue patterning and the maintenance of stem cell niches. BMPs pattern tissues over many temporal and spatial scales: BMP gradients as short as 1-2 cell diameters maintain the stem cell niche of the Drosophila germarium over a 24-h cycle, and BMP gradients of several hundred microns establish dorsal-ventral tissue specification in Drosophila, zebrafish and Xenopus embryos in timescales between 30 min and several hours. The mechanisms that shape BMP signaling gradients are also incredibly diverse. Although ligand diffusion plays a dominant role in forming the gradient, a cast of diffusible and non-diffusible regulators modulate gradient formation and confer robustness, including scale invariance and adaptability to perturbations in gene expression and growth. In this Review, we document the diverse ways that BMP gradients are formed and refined, and we identify the core principles that they share to achieve reliable performance.


Asunto(s)
Tipificación del Cuerpo/fisiología , Desarrollo Óseo , Proteínas Morfogenéticas Óseas/metabolismo , Desarrollo Embrionario , Animales , Tipificación del Cuerpo/genética , Proteínas Morfogenéticas Óseas/genética , Huesos , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Embrión no Mamífero/metabolismo , Regulación del Desarrollo de la Expresión Génica , Transducción de Señal , Xenopus laevis/embriología , Xenopus laevis/metabolismo , Pez Cebra/embriología , Pez Cebra/metabolismo , Proteínas de Pez Cebra
12.
Proc Natl Acad Sci U S A ; 118(15)2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33827919

RESUMEN

Heterodimeric TGF-ß ligands outperform homodimers in a variety of developmental, cell culture, and therapeutic contexts; however, the mechanisms underlying this increased potency remain uncharacterized. Here, we use dorsal-ventral axial patterning of the zebrafish embryo to interrogate the BMP2/7 heterodimer signaling mechanism. We demonstrate that differential interactions with BMP antagonists do not account for the reduced signaling ability of homodimers. Instead, we find that while overexpressed BMP2 homodimers can signal, they require two nonredundant type I receptors, one from the Acvr1 subfamily and one from the Bmpr1 subfamily. This implies that all BMP signaling within the zebrafish gastrula, even BMP2 homodimer signaling, requires Acvr1. This is particularly surprising as BMP2 homodimers do not bind Acvr1 in vitro. Furthermore, we find that the roles of the two type I receptors are subfunctionalized within the heterodimer signaling complex, with the kinase activity of Acvr1 being essential, while that of Bmpr1 is not. These results suggest that the potency of the Bmp2/7 heterodimer arises from the ability to recruit both Acvr1 and Bmpr1 into the same signaling complex.


Asunto(s)
Proteína Morfogenética Ósea 2/metabolismo , Proteína Morfogenética Ósea 7/metabolismo , Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Transducción de Señal , Proteínas de Pez Cebra/metabolismo , Receptores de Activinas Tipo I/metabolismo , Animales , Proteína Morfogenética Ósea 2/genética , Proteína Morfogenética Ósea 7/genética , Receptores de Proteínas Morfogenéticas Óseas/genética , Gástrula/metabolismo , Mutación , Unión Proteica , Multimerización de Proteína , Pez Cebra , Proteínas de Pez Cebra/genética
13.
PLoS Biol ; 19(1): e3001059, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33481775

RESUMEN

Bone Morphogenetic Protein (BMP) patterns the dorsal-ventral (DV) embryonic axis in all vertebrates, but it is unknown how cells along the DV axis interpret and translate the gradient of BMP signaling into differential gene activation that will give rise to distinct cell fates. To determine the mechanism of BMP morphogen interpretation in the zebrafish gastrula, we identified 57 genes that are directly activated by BMP signaling. By using Seurat analysis of single-cell RNA sequencing (scRNA-seq) data, we found that these genes are expressed in at least 3 distinct DV domains of the embryo. We distinguished between 3 models of BMP signal interpretation in which cells activate distinct gene expression through interpretation of thresholds of (1) the BMP signaling gradient slope; (2) the BMP signal duration; or (3) the level of BMP signal activation. We tested these 3 models using quantitative measurements of phosphorylated Smad5 (pSmad5) and by examining the spatial relationship between BMP signaling and activation of different target genes at single-cell resolution across the embryo. We found that BMP signaling gradient slope or BMP exposure duration did not account for the differential target gene expression domains. Instead, we show that cells respond to 3 distinct levels of BMP signaling activity to activate and position target gene expression. Together, we demonstrate that distinct pSmad5 threshold levels activate spatially distinct target genes to pattern the DV axis.


Asunto(s)
Tipificación del Cuerpo/genética , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Animales , Animales Modificados Genéticamente , Embrión no Mamífero , Gástrula/metabolismo , Regulación del Desarrollo de la Expresión Génica , Fosforilación , Transducción de Señal/genética , Proteína Smad5/metabolismo , Distribución Tisular/genética , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
14.
Elife ; 92020 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-32897189

RESUMEN

Fibrodysplasia ossificans progressiva (FOP) is a rare human genetic disorder characterized by altered skeletal development and extraskeletal ossification. All cases of FOP are caused by activating mutations in the type I BMP/TGFß cell surface receptor ACVR1, which over-activates signaling through phospho-Smad1/5 (pSmad1/5). To investigate the mechanism by which FOP-ACVR1 enhances pSmad1/5 activation, we used zebrafish embryonic dorsoventral (DV) patterning as an assay for BMP signaling. We determined that the FOP mutants ACVR1-R206H and -G328R do not require their ligand binding domain to over-activate BMP signaling in DV patterning. However, intact ACVR1-R206H has the ability to respond to both Bmp7 and Activin A ligands. Additionally, BMPR1, a type I BMP receptor normally required for BMP-mediated patterning of the embryo, is dispensable for both ligand-independent signaling pathway activation and ligand-responsive signaling hyperactivation by ACVR1-R206H. These results demonstrate that FOP-ACVR1 is not constrained by the same receptor/ligand partner requirements as WT-ACVR1.


Asunto(s)
Receptores de Activinas Tipo I/genética , Proteínas de Peces/genética , Miositis Osificante/genética , Pez Cebra/genética , Receptores de Activinas Tipo I/metabolismo , Animales , Modelos Animales de Enfermedad , Embrión no Mamífero/metabolismo , Proteínas de Peces/metabolismo , Miositis Osificante/metabolismo , Pez Cebra/embriología , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo
15.
Cell Rep ; 32(7): 108039, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32814043

RESUMEN

A fundamental question in developmental biology is how morphogens, such as bone morphogenetic protein (BMP), form precise signaling gradients to impart positional and functional identity to the cells of the early embryo. We combine rigorous mutant analyses with quantitative immunofluorescence to determine that the proteases Bmp1a and Tolloid spatially restrict the BMP antagonist Chordin in dorsoventral (DV) axial patterning of the early zebrafish gastrula. We show that maternally deposited Bmp1a plays an unexpected and non-redundant role in establishing the BMP signaling gradient, while the Bmp1a/Tolloid antagonist Sizzled is surprisingly dispensable. Combining computational modeling and in vivo analyses with an immobile Chordin construct, we demonstrate that long-range Chordin diffusion is not necessary for BMP gradient formation and DV patterning. Our data do not support a counter-gradient of Chordin and instead favor a Chordin sink, established by Bmp1a and Tolloid, as the primary mechanism that drives BMP gradient formation.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Glicoproteínas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Animales , Transducción de Señal , Pez Cebra
16.
Curr Top Dev Biol ; 140: 341-389, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32591080

RESUMEN

Axis specification of the zebrafish embryo begins during oogenesis and relies on proper formation of well-defined cytoplasmic domains within the oocyte. Upon fertilization, maternally-regulated cytoplasmic flow and repositioning of dorsal determinants establish the coordinate system that will build the structure and developmental body plan of the embryo. Failure of specific genes that regulate the embryonic coordinate system leads to catastrophic loss of body structures. Here, we review the genetic principles of axis formation and discuss how maternal factors orchestrate axis patterning during zebrafish early embryogenesis. We focus on the molecular identity and functional contribution of genes controlling critical aspects of oogenesis, egg activation, blastula, and gastrula stages. We examine how polarized cytoplasmic domains form in the oocyte, which set off downstream events such as animal-vegetal polarity and germ line development. After gametes interact and form the zygote, cytoplasmic segregation drives the animal-directed reorganization of maternal determinants through calcium- and cell cycle-dependent signals. We also summarize how maternal genes control dorsoventral, anterior-posterior, mesendodermal, and left-right cell fate specification and how signaling pathways pattern these axes and tissues during early development to instruct the three-dimensional body plan. Advances in reverse genetics and phenotyping approaches in the zebrafish model are revealing positional patterning signatures at the single-cell level, thus enhancing our understanding of genotype-phenotype interactions in axis formation. Our emphasis is on the genetic interrogation of novel and specific maternal regulatory mechanisms of axis specification in the zebrafish.


Asunto(s)
Tipificación del Cuerpo/genética , Embrión no Mamífero/metabolismo , Regulación del Desarrollo de la Expresión Génica , Oocitos/metabolismo , Pez Cebra/genética , Cigoto/metabolismo , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Embrión no Mamífero/citología , Embrión no Mamífero/embriología , Cinesinas/genética , Cinesinas/metabolismo , Herencia Materna/genética , Oocitos/citología , Pez Cebra/embriología , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Cigoto/citología
17.
PLoS Genet ; 16(4): e1008652, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32267837

RESUMEN

Forward genetic screens remain at the forefront of biology as an unbiased approach for discovering and elucidating gene function at the organismal and molecular level. Past mutagenesis screens targeting maternal-effect genes identified a broad spectrum of phenotypes ranging from defects in oocyte development to embryonic patterning. However, earlier vertebrate screens did not reach saturation, anticipated classes of phenotypes were not uncovered, and technological limitations made it difficult to pinpoint the causal gene. In this study, we performed a chemically-induced maternal-effect mutagenesis screen in zebrafish and identified eight distinct mutants specifically affecting the cleavage stage of development and one cleavage stage mutant that is also male sterile. The cleavage-stage phenotypes fell into three separate classes: developmental arrest proximal to the mid blastula transition (MBT), irregular cleavage, and cytokinesis mutants. We mapped each mutation to narrow genetic intervals and determined the molecular basis for two of the developmental arrest mutants, and a mutation causing male sterility and a maternal-effect mutant phenotype. One developmental arrest mutant gene encodes a maternal specific Stem Loop Binding Protein, which is required to maintain maternal histone levels. The other developmental arrest mutant encodes a maternal-specific subunit of the Minichromosome Maintenance Protein Complex, which is essential for maintaining normal chromosome integrity in the early blastomeres. Finally, we identify a hypomorphic allele of Polo-like kinase-1 (Plk-1), which results in a male sterile and maternal-effect phenotype. Collectively, these mutants expand our molecular-genetic understanding of the maternal regulation of early embryonic development in vertebrates.


Asunto(s)
División Celular/genética , Desarrollo Embrionario/genética , Herencia Materna/genética , Mutación , Pez Cebra/embriología , Pez Cebra/genética , Alelos , Animales , Blástula/citología , Blástula/embriología , Blástula/metabolismo , Tipificación del Cuerpo/genética , Núcleo Celular , Citocinesis/genética , Femenino , Infertilidad Masculina/genética , Masculino , Mutagénesis , Fenotipo , Proteínas de Pez Cebra/genética
18.
J Math Biol ; 80(1-2): 505-520, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31773243

RESUMEN

Bone Morphogenetic Proteins (BMPs) play an important role in dorsal-ventral (DV) patterning of the early zebrafish embryo. BMP signaling is regulated by a network of extracellular and intracellular factors that impact the range and signaling of BMP ligands. Recent advances in understanding the mechanism of pattern formation support a source-sink mechanism, however it is not clear how the source-sink mechanism shapes patterns in 3D, nor how sensitive the pattern is to biophysical rates and boundary conditions along both the anteroposterior (AP) and DV axes of the embryo. We propose a new three-dimensional growing Partial Differential Equation (PDE)-based model to simulate the BMP patterning process during the blastula stage. This model provides a starting point to elucidate how different mechanisms and components work together in 3D to create and maintain the BMP gradient in the embryo. We also show how the 3D model fits the BMP signaling gradient data at multiple time points along both axes. Furthermore, sensitivity analysis of the model suggests that the spatiotemporal patterns of Chordin and BMP ligand gene expression are dominant drivers of shape in 3D and more work is needed to quantify the spatiotemporal profiles of gene and protein expression to further refine the models.


Asunto(s)
Blástula/embriología , Tipificación del Cuerpo/fisiología , Proteínas Morfogenéticas Óseas/metabolismo , Modelos Biológicos , Proteínas de Pez Cebra/metabolismo , Animales , Glicoproteínas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Transducción de Señal/fisiología , Análisis Espacio-Temporal , Pez Cebra/embriología , Pez Cebra/genética
19.
Curr Top Dev Biol ; 135: 1-34, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31155356

RESUMEN

The fate of future generations depends on a high-quality germ line. For a female to successfully produce offspring, her oocytes must be successfully specified and their contents meticulously organized. Germ cells are specified by two general mechanisms: inductive and inherited. In the inductive mechanism, the primordial germ cells (PGCs) are induced by signals from the surrounding cells. In the inherited mechanism, PGCs are specified by passing localized germ plasm material from the oocyte to the future germ cells. The Balbiani body, a conserved oocyte aggregate, facilitates the organization of the oocyte into a polarized cell with discrete cytoplasmic domains, including localizing the germ plasm. In the mouse, the Balbiani body is implicated in oocyte survival, while in frogs and zebrafish the Balbiani body carries specific mRNAs to the vegetal pole. These asymmetric mRNAs form the foundation of the functionally polarized oocyte and play important roles in axial patterning and germ plasm formation of the embryo.


Asunto(s)
Polaridad Celular/genética , Puffs Cromosómicos/genética , Oocitos/citología , Vertebrados/genética , Animales , Humanos , Patrón de Herencia/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo
20.
Methods Mol Biol ; 1920: 295-302, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30737698

RESUMEN

Proteomic characterization of isolated organelles can provide insight into the functional components of the structure and novel targets for further testing. Germplasm in developing oocytes is difficult to isolate for protein identification because not all types of germplasm are stable outside of the cytoplasm. In zebrafish, the Balbiani body forms a proteinaceous aggregate that contains the germplasm and we found is stable outside of the oocyte. Here we present a manual isolation protocol that collects intact Balbiani bodies from stage I zebrafish oocytes. We lysed oocytes by passing them through a syringe, and then used a fine injection needle to wick up Balbiani bodies by capillary action with minimal buffer solution. Using this protocol we collected sufficient material for proteomic analysis of the zebrafish Balbiani body.


Asunto(s)
Fraccionamiento Celular , Oocitos/metabolismo , Orgánulos/metabolismo , Proteoma , Proteómica , Pez Cebra/metabolismo , Animales , Biomarcadores , Fraccionamiento Celular/métodos , Técnica del Anticuerpo Fluorescente/métodos , Oogénesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA