Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Dev Biol ; 495: 21-34, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36587799

RESUMO

Septate junctions (SJs) evolved as cell-cell junctions that regulate the paracellular barrier and integrity of epithelia in invertebrates. Multiple morphological variants of SJs exist specific to different epithelia and/or phyla but the biological significance of varied SJ morphology is unclear because the knowledge of the SJ associated proteins and their functions in non-insect invertebrates remains largely unknown. Here we report cell-specific expression of nine candidate SJ genes in the early life stages of the sea urchin Strongylocentrotus purpuratus. By use of in situ RNA hybridization and single cell RNA-seq we found that the expression of selected genes encoding putatively SJ associated transmembrane and cytoplasmic scaffold molecules was dynamically regulated during epithelial development in the embryos and larvae with different epithelia expressing different cohorts of SJ genes. We focused a functional analysis on SpMesh, a homolog of the Drosophila smooth SJ component Mesh, which was highly enriched in the endodermal epithelium of the mid- and hindgut. Functional perturbation of SpMesh by both CRISPR/Cas9 mutagenesis and vivo morpholino-mediated knockdown shows that loss of SpMesh does not disrupt the formation of the gut epithelium during gastrulation. However, loss of SpMesh resulted in a severely reduced gut-paracellular barrier as quantitated by increased permeability to 3-5 â€‹kDa FITC-dextran. Together, these studies provide a first look at the molecular SJ physiology during the development of a marine organism and suggest a shared role for Mesh-homologous proteins in forming an intestinal barrier in invertebrates. Results have implications for consideration of the traits underlying species-specific sensitivity of marine larvae to climate driven ocean change.


Assuntos
Proteínas de Drosophila , Strongylocentrotus purpuratus , Animais , Strongylocentrotus purpuratus/genética , Strongylocentrotus purpuratus/metabolismo , Junções Íntimas/genética , Junções Íntimas/metabolismo , Epitélio/metabolismo , Junções Intercelulares/metabolismo , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Ouriços-do-Mar/genética , Ouriços-do-Mar/metabolismo , Larva/genética , Larva/metabolismo
2.
Dev Biol ; 472: 98-114, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33484703

RESUMO

microRNAs (miRNAs) play a critical role in a variety of biological processes, including embryogenesis and the physiological functions of cells. Evolutionarily conserved microRNA-31 (miR-31) has been found to be involved in cancer, bone formation, and lymphatic development. We previously discovered that, in the sea urchin, miR-31 knockdown (KD) embryos have shortened dorsoventral connecting rods, mispatterned skeletogenic primary mesenchyme cells (PMCs) and shifted and expanded Vegf3 expression domain. Vegf3 itself does not contain miR-31 binding sites; however, we identified its upstream regulators Eve and Wnt1 to be directly suppressed by miR-31. Removal of miR-31's suppression of Eve and Wnt1 resulted in skeletal and PMC patterning defects, similar to miR-31 KD phenotypes. Additionally, removal of miR-31's suppression of Eve and Wnt1 results in an expansion and anterior shift in expression of Veg1 ectodermal genes, including Vegf3 in the blastulae. This indicates that miR-31 indirectly regulates Vegf3 expression through directly suppressing Eve and Wnt1. Furthermore, removing miR-31 suppression of Eve is sufficient to cause skeletogenic defects, revealing a novel regulatory role of Eve in skeletogenesis and PMC patterning. Overall, this study provides a proposed molecular mechanism of miR-31's regulation of skeletogenesis and PMC patterning through its cross-regulation of a Wnt signaling ligand and a transcription factor of the endodermal and ectodermal gene regulatory network.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/metabolismo , MicroRNAs/metabolismo , Desenvolvimento Musculoesquelético/genética , Strongylocentrotus purpuratus/embriologia , Strongylocentrotus purpuratus/genética , Proteína Wnt1/metabolismo , Animais , Animais Geneticamente Modificados , Padronização Corporal/genética , Desenvolvimento Embrionário/genética , Feminino , Técnicas de Silenciamento de Genes , Redes Reguladoras de Genes , Masculino , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/genética , Fenótipo , Transdução de Sinais/genética , Strongylocentrotus purpuratus/metabolismo , Fatores de Transcrição/metabolismo
3.
Development ; 144(7): 1201-1210, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28235822

RESUMO

Stem cells in animals often exhibit a slow cell cycle and/or low transcriptional activity referred to as quiescence. Here, we report that the translational activity in the primordial germ cells (PGCs) of the sea urchin embryo (Strongylocentrotus purpuratus) is quiescent. We measured new protein synthesis with O-propargyl-puromycin and L-homopropargylglycine Click-iT technologies, and determined that these cells synthesize protein at only 6% the level of their adjacent somatic cells. Knockdown of translation of the RNA-binding protein Nanos2 by morpholino antisense oligonucleotides, or knockout of the Nanos2 gene by CRISPR/Cas9 resulted in a significant, but partial, increase (47%) in general translation specifically in the PGCs. We found that the mRNA of the translation factor eEF1A is excluded from the PGCs in a Nanos2-dependent manner, a consequence of a Nanos/Pumilio response element (PRE) in its 3'UTR. In addition to eEF1A, the cytoplasmic pH of the PGCs appears to repress translation and simply increasing the pH also significantly restores translation selectively in the PGCs. We conclude that the PGCs of this sea urchin institute parallel pathways to quiesce translation thoroughly but transiently.


Assuntos
Ciclo Celular , Células Germinativas/citologia , Biossíntese de Proteínas , Strongylocentrotus purpuratus/citologia , Strongylocentrotus purpuratus/metabolismo , Animais , Sequência de Bases , Blástula/citologia , Blástula/metabolismo , Sistemas CRISPR-Cas/genética , Ciclo Celular/genética , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas/metabolismo , Concentração de Íons de Hidrogênio , Mitocôndrias/metabolismo , Fator 1 de Elongação de Peptídeos/genética , Fator 1 de Elongação de Peptídeos/metabolismo , Biossíntese de Proteínas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Strongylocentrotus purpuratus/genética
4.
Exp Cell Res ; 359(1): 205-214, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28782554

RESUMO

The sea urchin larval embryo elaborates two calcitic endoskeletal elements called spicules. Spicules are synthesized by the primary mesenchyme cells (PMCs) and begin to form at early gastrula stage. It is known that the calcium comprising the spicules comes from the seawater and we wish to further consider the mode of calcium transport from the extracellular seawater to the PMCs and then onto the forming spicules. We used PMC in vitro cultures, calcein, fluorescently labeled dextran, and fluorescently labeled Wheat Germ Agglutinin (WGA) to track calcium transport from the seawater into PMCs and spicules and to determine how molecules from the surface of PMCs interact with the incoming calcium. Labeling of PMC endocytic vesicles and forming spicules by both calcein and fluorescently tagged dextran indicate that calcium is taken up from the seawater by endocytosis and directly incorporated into spicules. Calcein labeling studies also indicate that calcium from the extracellular seawater begins to be incorporated into spicules within 30min of uptake. In addition, we demonstrate that fluorescently labeled WGA and calcein are taken up by many of the same endocytic vesicles and are incorporated into growing spicules. These findings suggest that PMC specific surface molecules accompany calcium ions as they enter PMCs via endocytosis and are incorporated together in the growing spicule. Using anti-spicule matrix protein antibodies, we pinpoint a subset of spicule matrix proteins that may accompany calcium ions from the surface of the PMCs until they are incorporated into spicules. Msp130 is identified as one of these spicule matrix proteins.


Assuntos
Endocitose , Mesoderma/citologia , Osteogênese , Strongylocentrotus purpuratus/citologia , Strongylocentrotus purpuratus/crescimento & desenvolvimento , Animais , Cálcio/metabolismo , Células Cultivadas , Dextranos/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Espaço Extracelular/metabolismo , Fluoresceínas/metabolismo , Cinética , Larva/citologia , Larva/metabolismo , Mesoderma/metabolismo , Strongylocentrotus purpuratus/metabolismo , Aglutininas do Germe de Trigo/metabolismo
5.
Proc Natl Acad Sci U S A ; 112(15): 4696-701, 2015 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-25825763

RESUMO

Energy is required to maintain physiological homeostasis in response to environmental change. Although responses to environmental stressors frequently are assumed to involve high metabolic costs, the biochemical bases of actual energy demands are rarely quantified. We studied the impact of a near-future scenario of ocean acidification [800 µatm partial pressure of CO2 (pCO2)] during the development and growth of an important model organism in developmental and environmental biology, the sea urchin Strongylocentrotus purpuratus. Size, metabolic rate, biochemical content, and gene expression were not different in larvae growing under control and seawater acidification treatments. Measurements limited to those levels of biological analysis did not reveal the biochemical mechanisms of response to ocean acidification that occurred at the cellular level. In vivo rates of protein synthesis and ion transport increased ∼50% under acidification. Importantly, the in vivo physiological increases in ion transport were not predicted from total enzyme activity or gene expression. Under acidification, the increased rates of protein synthesis and ion transport that were sustained in growing larvae collectively accounted for the majority of available ATP (84%). In contrast, embryos and prefeeding and unfed larvae in control treatments allocated on average only 40% of ATP to these same two processes. Understanding the biochemical strategies for accommodating increases in metabolic energy demand and their biological limitations can serve as a quantitative basis for assessing sublethal effects of global change. Variation in the ability to allocate ATP differentially among essential functions may be a key basis of resilience to ocean acidification and other compounding environmental stressors.


Assuntos
Metabolismo Energético , Expressão Gênica , Strongylocentrotus purpuratus/genética , Strongylocentrotus purpuratus/metabolismo , Ácidos/química , Fatores Etários , Análise de Variância , Animais , Dióxido de Carbono/metabolismo , Feminino , Concentração de Íons de Hidrogênio , Larva/genética , Larva/crescimento & desenvolvimento , Larva/metabolismo , Masculino , Oceanos e Mares , Proteínas/genética , Proteínas/metabolismo , Água do Mar/química , ATPase Trocadora de Sódio-Potássio/genética , ATPase Trocadora de Sódio-Potássio/metabolismo , Strongylocentrotus purpuratus/crescimento & desenvolvimento , Fatores de Tempo
6.
Differentiation ; 95: 31-43, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28188999

RESUMO

The small GTPase Arf6 is a conserved protein that is expressed in all metazoans. Arf6 remodels cytoskeletal actin and mediates membrane protein trafficking between the plasma membrane in its active form and endosomal compartments in its inactive form. While a rich knowledge exists for the cellular functions of Arf6, relatively little is known about its physiological role in development. This study examines the function of Arf6 in mediating cellular morphogenesis in early development. We dissect the function of Arf6 with a loss-of-function morpholino and constitutively active Arf6-Q67L construct. We focus on the two cell types that undergo active directed migration: the primary mesenchyme cells (PMCs) that give rise to the sea urchin skeleton and endodermal cells that form the gut. Our results indicate that Arf6 plays an important role in skeleton formation and PMC migration, in part due to its ability to remodel actin. We also found that embryos injected with Arf6 morpholino have gastrulation defects and embryos injected with constitutively active Arf6 have endodermal cells detached from the gut epithelium with decreased junctional cadherin staining, indicating that Arf6 may mediate the recycling of cadherin. Thus, Arf6 impacts cells that undergo coordinated movement to form embryonic structures in the developing embryo.


Assuntos
Fatores de Ribosilação do ADP/metabolismo , Morfogênese , Strongylocentrotus purpuratus/metabolismo , Fatores de Ribosilação do ADP/genética , Animais , Caderinas/metabolismo , Endoderma/citologia , Endoderma/metabolismo , Mesoderma/citologia , Mesoderma/metabolismo , Strongylocentrotus purpuratus/embriologia
7.
Biochemistry ; 55(31): 4410-21, 2016 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-27426695

RESUMO

In the purple sea urchin Strongylocentrotus purpuratus, the formation and mineralization of fracture-resistant skeletal elements such as the embryonic spicule require the combinatorial participation of numerous spicule matrix proteins such as the SpSM30A-F isoforms. However, because of limited abundance, it has been difficult to pursue extensive biochemical studies of the SpSM30 proteins and deduce their role in spicule formation and mineralization. To circumvent these problems, we expressed a model recombinant spicule matrix protein, rSpSM30B/C, which possesses the key sequence attributes of isoforms "B" and "C". Our findings indicate that rSpSM30B/C is expressed in insect cells as a single polypeptide containing variations in glycosylation that create microheterogeneity in rSpSM30B/C molecular masses. These post-translational modifications incorporate O- and N-glycans and anionic mono- and bisialylated and mono- and bisulfated monosaccharides on the protein molecules and enhance its aggregation propensity. Bioinformatics and biophysical experiments confirm that rSpSM30B/C is an intrinsically disordered, aggregation-prone protein that forms porous protein hydrogels that control the in vitro mineralization process in three ways: (1) increase the time interval for prenucleation cluster formation and transiently stabilize an ACC polymorph, (2) promote and organize single-crystal calcite nanoparticles, and (3) promote faceted growth and create surface texturing of calcite crystals. These features are also common to mollusk shell nacre proteins, and we conclude that rSpSM30B/C is a spiculogenesis protein that exhibits traits found in other calcium carbonate mineral modification proteins.


Assuntos
Proteínas do Citoesqueleto/química , Proteínas do Citoesqueleto/metabolismo , Strongylocentrotus purpuratus/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Carbonato de Cálcio/química , Carbonato de Cálcio/metabolismo , Proteínas do Citoesqueleto/genética , Glicosilação , Hidrogéis , Microscopia de Força Atômica , Microscopia Eletrônica de Varredura , Minerais/química , Minerais/metabolismo , Modelos Moleculares , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Multimerização Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Strongylocentrotus purpuratus/química , Strongylocentrotus purpuratus/genética
8.
BMC Evol Biol ; 16(1): 117, 2016 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-27230062

RESUMO

BACKGROUND: Digestive cells are present in all metazoans and provide the energy necessary for the whole organism. Pancreatic exocrine cells are a unique vertebrate cell type involved in extracellular digestion of a wide range of nutrients. Although the organization and regulation of this cell type is intensively studied in vertebrates, its evolutionary history is still unknown. In order to understand which are the elements that define the pancreatic exocrine phenotype, we have analyzed the expression of genes that contribute to specification and function of this cell-type in an early branching deuterostome, the sea urchin Strongylocentrotus purpuratus. RESULTS: We defined the spatial and temporal expression of sea urchin orthologs of pancreatic exocrine genes and described a unique population of cells clustered in the upper stomach of the sea urchin embryo where exocrine markers are co-expressed. We used a combination of perturbation analysis, drug and feeding experiments and found that in these cells of the sea urchin embryo gene expression and gene regulatory interactions resemble that of bona fide pancreatic exocrine cells. We show that the sea urchin Ptf1a, a key transcriptional activator of digestive enzymes in pancreatic exocrine cells, can substitute for its vertebrate ortholog in activating downstream genes. CONCLUSIONS: Collectively, our study is the first to show with molecular tools that defining features of a vertebrate cell-type, the pancreatic exocrine cell, are shared by a non-vertebrate deuterostome. Our results indicate that the functional cell-type unit of the vertebrate pancreas may evolutionarily predate the emergence of the pancreas as a discrete organ. From an evolutionary perspective, these results encourage to further explore the homologs of other vertebrate cell-types in traditional or newly emerging deuterostome systems.


Assuntos
Evolução Biológica , Estômago/citologia , Strongylocentrotus purpuratus/citologia , Animais , Diferenciação Celular , Linhagem da Célula , Digestão/genética , Digestão/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Genes Reguladores , Células HEK293 , Células HeLa , Humanos , Larva/citologia , Larva/metabolismo , Pâncreas/citologia , Ratos , Strongylocentrotus purpuratus/crescimento & desenvolvimento , Strongylocentrotus purpuratus/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fatores de Transcrição/fisiologia , Vertebrados/anatomia & histologia , Vertebrados/metabolismo
9.
Proc Biol Sci ; 283(1826): 20152978, 2016 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-26962139

RESUMO

Pou domain transcription factor Pou4f2 is essential for the development of retinal ganglion cells (RGCs) in the vertebrate retina. A distant orthologue of Pou4f2 exists in the genome of the sea urchin (class Echinoidea) Strongylocentrotus purpuratus (SpPou4f1/2), yet the photosensory structure of sea urchins is strikingly different from that of the mammalian retina. Sea urchins have no obvious eyes, but have photoreceptors clustered around their tube feet disc. The mechanisms that are associated with the development and function of photoreception in sea urchins are largely unexplored. As an initial approach to better understand the sea urchin photosensory structure and relate it to the mammalian retina, we asked whether SpPou4f1/2 could support RGC development in the absence of Pou4f2. To answer this question, we replaced genomic Pou4f2 with an SpPou4f1/2 cDNA. In Pou4f2-null mice, retinas expressing SpPou4f1/2 were outwardly identical to those of wild-type mice. SpPou4f1/2 retinas exhibited dark-adapted electroretinogram scotopic threshold responses, indicating functionally active RGCs. During retinal development, SpPou4f1/2 activated RGC-specific genes and in S. purpuratus, SpPou4f2 was expressed in photoreceptor cells of tube feet in a pattern distinct from Opsin4 and Pax6. Our results suggest that SpPou4f1/2 and Pou4f2 share conserved components of a gene network for photosensory development and they maintain their conserved intrinsic functions despite vast morphological differences in mouse and sea urchin photosensory structures.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Camundongos/genética , Células Ganglionares da Retina/metabolismo , Strongylocentrotus purpuratus/genética , Fator de Transcrição Brn-3B/genética , Animais , Embrião de Mamíferos/embriologia , Embrião não Mamífero/embriologia , Proteínas de Homeodomínio/metabolismo , Camundongos/crescimento & desenvolvimento , Camundongos/metabolismo , Células Ganglionares da Retina/citologia , Strongylocentrotus purpuratus/metabolismo , Fator de Transcrição Brn-3B/metabolismo
10.
PLoS Biol ; 11(1): e1001467, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23335859

RESUMO

Patterning the neuroectoderm along the anterior-posterior (AP) axis is a critical event in the early development of deuterostome embryos. However, the mechanisms that regulate the specification and patterning of the neuroectoderm are incompletely understood. Remarkably, the anterior neuroectoderm (ANE) of the deuterostome sea urchin embryo expresses many of the same transcription factors and secreted modulators of Wnt signaling, as does the early vertebrate ANE (forebrain/eye field). Moreover, as is the case in vertebrate embryos, confining the ANE to the anterior end of the embryo requires a Wnt/ß-catenin-dependent signaling mechanism. Here we use morpholino- or dominant negative-mediated interference to demonstrate that the early sea urchin embryo integrates information not only from Wnt/ß-catenin but also from Wnt/Fzl5/8-JNK and Fzl1/2/7-PKC pathways to provide precise spatiotemporal control of neuroectoderm patterning along its AP axis. Together, through the Wnt1 and Wnt8 ligands, they orchestrate a progressive posterior-to-anterior wave of re-specification that restricts the initial, ubiquitous, maternally specified, ANE regulatory state to the most anterior blastomeres. There, the Wnt receptor antagonist, Dkk1, protects this state through a negative feedback mechanism. Because these different Wnt pathways converge on the same cell fate specification process, our data suggest they may function as integrated components of an interactive Wnt signaling network. Our findings provide strong support for the idea that the sea urchin ANE regulatory state and the mechanisms that position and define its borders represent an ancient regulatory patterning system that was present in the common echinoderm/vertebrate ancestor.


Assuntos
Padronização Corporal/genética , Placa Neural/embriologia , Strongylocentrotus purpuratus/embriologia , Proteínas Wnt/metabolismo , Animais , Blastômeros/metabolismo , Padronização Corporal/fisiologia , Diferenciação Celular/genética , Regulação da Expressão Gênica no Desenvolvimento , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Morfolinos/genética , Placa Neural/metabolismo , RNA Mensageiro/genética , Strongylocentrotus purpuratus/genética , Strongylocentrotus purpuratus/metabolismo , Fatores de Transcrição/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo
11.
Proteomics ; 15(23-24): 4080-95, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26227301

RESUMO

Fertilization triggers a dynamic symphony of molecular transformations induced by a rapid rise in intracellular calcium. Most prominent are surface alterations, metabolic activation, cytoskeletal reorganization, and cell-cycle reentry. While the activation process appears to be broadly evolutionarily conserved, and protein phosphorylation is known to play a key role, the signaling networks mediating the response to fertilization are not well described. To address this gap, we performed a time course phosphoproteomic analysis of egg activation in the sea urchin Strongylocentrotus purpuratus, a system that offers biochemical tractability coupled with exquisite synchronicity. By coupling large-scale phosphopeptide enrichment with unbiased quantitative MS, we identified striking changes in global phosphoprotein patterns at 2- and 5-min postfertilization as compared to unfertilized eggs. Overall, we mapped 8796 distinct phosphosite modifications on 2833 phosphoproteins, of which 15% were differentially regulated in early egg activation. Activated kinases were identified by phosphosite mapping, while enrichment analyses revealed conserved signaling cascades not previously associated with egg activation. This work represents the most comprehensive study of signaling associated with egg activation to date, suggesting novel mechanisms that can be experimentally tested and providing a valuable resource for the broader research community. All MS data have been deposited in the ProteomeXchange with identifier PXD002239 (http://proteomecentral.proteomexchange.org/dataset/PXD002239).


Assuntos
Proteômica , Ouriços-do-Mar/metabolismo , Strongylocentrotus purpuratus/metabolismo , Animais , Cálcio/metabolismo
12.
J Biol Chem ; 289(25): 17427-44, 2014 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-24794867

RESUMO

Cilia and flagella are conserved, motile, and sensory cell organelles involved in signal transduction and human disease. Their scaffold consists of a 9-fold array of remarkably stable doublet microtubules (DMTs), along which motor proteins transmit force for ciliary motility and intraflagellar transport. DMTs possess Ribbons of three to four hyper-stable protofilaments whose location, organization, and specialized functions have been elusive. We performed a comprehensive analysis of the distribution and structural arrangements of Ribbon proteins from sea urchin sperm flagella, using quantitative immunobiochemistry, proteomics, immuno-cryo-electron microscopy, and tomography. Isolated Ribbons contain acetylated α-tubulin, ß-tubulin, conserved protein Rib45, >95% of the axonemal tektins, and >95% of the calcium-binding proteins, Rib74 and Rib85.5, whose human homologues are related to the cause of juvenile myoclonic epilepsy. DMTs contain only one type of Ribbon, corresponding to protofilaments A11-12-13-1 of the A-tubule. Rib74 and Rib85.5 are associated with the Ribbon in the lumen of the A-tubule. Ribbons contain a single ∼5-nm wide filament, composed of equimolar tektins A, B, and C, which interact with the nexin-dynein regulatory complex. A summary of findings is presented, and the functions of Ribbon proteins are discussed in terms of the assembly and stability of DMTs, ciliary motility, and other microtubule systems.


Assuntos
Proteínas de Ligação ao Cálcio/química , Proteínas dos Microtúbulos/química , Microtúbulos/química , Complexos Multiproteicos/química , Cauda do Espermatozoide/química , Strongylocentrotus purpuratus/química , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Cílios/química , Cílios/genética , Cílios/metabolismo , Humanos , Masculino , Proteínas dos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Microtúbulos/ultraestrutura , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Motilidade dos Espermatozoides/fisiologia , Cauda do Espermatozoide/metabolismo , Cauda do Espermatozoide/ultraestrutura , Strongylocentrotus purpuratus/metabolismo , Strongylocentrotus purpuratus/ultraestrutura
13.
Proc Natl Acad Sci U S A ; 109(16): 6088-93, 2012 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-22492931

RESUMO

Crystalline biominerals do not resemble faceted crystals. Current explanations for this property involve formation via amorphous phases. Using X-ray absorption near-edge structure (XANES) spectroscopy and photoelectron emission microscopy (PEEM), here we examine forming spicules in embryos of Strongylocentrotus purpuratus sea urchins, and observe a sequence of three mineral phases: hydrated amorphous calcium carbonate (ACC · H(2)O) → dehydrated amorphous calcium carbonate (ACC) → calcite. Unexpectedly, we find ACC · H(2)O-rich nanoparticles that persist after the surrounding mineral has dehydrated and crystallized. Protein matrix components occluded within the mineral must inhibit ACC · H(2)O dehydration. We devised an in vitro, also using XANES-PEEM, assay to identify spicule proteins that may play a role in stabilizing various mineral phases, and found that the most abundant occluded matrix protein in the sea urchin spicules, SM50, stabilizes ACC · H(2)O in vitro.


Assuntos
Materiais Biocompatíveis/química , Calcificação Fisiológica , Carbonato de Cálcio/química , Transição de Fase , Animais , Materiais Biocompatíveis/metabolismo , Carbonato de Cálcio/metabolismo , Cristalização , Embrião não Mamífero/química , Embrião não Mamífero/metabolismo , Embrião não Mamífero/ultraestrutura , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Microscopia Eletrônica/métodos , Minerais/química , Minerais/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Strongylocentrotus purpuratus/química , Strongylocentrotus purpuratus/embriologia , Strongylocentrotus purpuratus/metabolismo , Água/química , Espectroscopia por Absorção de Raios X/métodos
14.
Dev Biol ; 375(1): 92-104, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23261933

RESUMO

Specification of the non-skeletogenic mesoderm (NSM) in sea urchin embryos depends on Delta signaling. Signal reception leads to expression of regulatory genes that later contribute to the aboral NSM regulatory state. In oral NSM, this is replaced by a distinct oral regulatory state in consequence of Nodal signaling. Through regulome wide analysis we identify the homeobox gene not as an immediate Nodal target. not expression in NSM causes extinction of the aboral regulatory state in the oral NSM, and expression of a new suite of regulatory genes. All NSM specific regulatory genes are henceforth expressed exclusively, in oral or aboral domains, presaging the mesodermal cell types that will emerge. We have analyzed the regulatory linkages within the aboral NSM gene regulatory network. A linchpin of this network is gataE which as we show is a direct Gcm target and part of a feedback loop locking down the aboral regulatory state.


Assuntos
Embrião não Mamífero/metabolismo , Mesoderma/embriologia , Strongylocentrotus purpuratus/embriologia , Strongylocentrotus purpuratus/genética , Animais , Técnicas de Cultura Embrionária , Fatores de Transcrição GATA/genética , Fatores de Transcrição GATA/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Morfolinos/genética , Boca/embriologia , Proteína Nodal/genética , Proteína Nodal/metabolismo , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Oligonucleotídeos Antissenso/genética , Transdução de Sinais , Strongylocentrotus purpuratus/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
15.
Biochim Biophys Acta ; 1830(11): 5305-15, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23928041

RESUMO

BACKGROUND: Sea urchin sperm motility is regulated by Speract, a sperm-activating peptide (SAP) secreted from the outer egg coat. Upon binding to its receptor in the sperm flagellum, Speract induces a series of ionic and metabolic changes in Strongylocentrotus purpuratus spermatozoa that regulate their motility. Among these events, protein phosphorylation is one of the most relevant and evidence indicates that some proteins of the Speract signaling cascade localize in low density detergent-insoluble membranes (LD-DIM). METHODS: LD-DIM-derived proteins from immotile, motile or Speract-stimulated S. purpuratus sperm were resolved in 2-D gels and the PKA and PKC substrates detected with specific antibodies were identified by LC-MS/MS. RESULTS: Differential PKA and PKC substrate phosphorylation levels among the LD-DIM isolated from sperm in different motility conditions were found and identified by mass spectrometry as: ATP synthase, creatine kinase, NADH dehydrogenase (ubiquinone) flavoprotein 2, succinyl-CoA ligase and the voltage-dependent anion channel 2 (VDAC2), which are mitochondrial proteins, as well as, the cAMP-dependent protein kinase type II regulatory (PKA RII) subunit, Tubulin ß chain and Actin Cy I changed their phosphorylation state. CONCLUSIONS: Some mitochondrial proteins regulated by PKA or PKC may influence sea urchin sperm motility. GENERAL SIGNIFICANCE: The fact that a high percentage (66%) of the PKA or PKC substrates identified in LD-DIM are mitochondrial proteins suggests that the phosphorylation of these proteins modulates sea urchin sperm motility via Speract stimulation by providing sufficient energy to sperm physiology. Those mitochondrial proteins are indeed PKA- or PKC-substrates in the sea urchin spermatozoa.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas Mitocondriais/metabolismo , Proteína Quinase C/metabolismo , Motilidade dos Espermatozoides/fisiologia , Espermatozoides/fisiologia , Strongylocentrotus purpuratus/fisiologia , Animais , Proteínas Quinases Dependentes de AMP Cíclico/química , Detergentes/química , Masculino , Proteínas Mitocondriais/química , Oligopeptídeos/metabolismo , Fosforilação/fisiologia , Proteína Quinase C/química , Ouriços-do-Mar , Transdução de Sinais , Cauda do Espermatozoide/metabolismo , Cauda do Espermatozoide/fisiologia , Espermatozoides/química , Espermatozoides/metabolismo , Strongylocentrotus purpuratus/química , Strongylocentrotus purpuratus/metabolismo
16.
Development ; 138(11): 2217-22, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21525076

RESUMO

Vasa is a broadly conserved ATP-dependent RNA helicase that functions in the germ line of organisms from cnidarians to mammals. Curiously, Vasa is also present in the somatic cells of many animals and functions as a regulator of multipotent cells. Here, we report a mitotic function of Vasa revealed in the sea urchin embryo. We found that Vasa protein is present in all blastomeres of the early embryo and that its abundance oscillates with the cell cycle. Vasa associates with the spindle and the separating sister chromatids at metaphase, and then quickly disappears after telophase. Inhibition of Vasa protein synthesis interferes with proper chromosome segregation, arrests cells at M-phase, and delays overall cell cycle progression. Cdk activity is necessary for the proper localization of Vasa, implying that Vasa is involved in the cyclin-dependent cell cycle network, and Vasa is required for the efficient translation of cyclinB mRNA. Our results suggest an evolutionarily conserved role of Vasa that is independent of its function in germ line determination.


Assuntos
Ciclo Celular/fisiologia , RNA Helicases DEAD-box/metabolismo , Mitose , Ouriços-do-Mar/embriologia , Animais , Blastômeros/citologia , Cromátides/metabolismo , Segregação de Cromossomos , Ciclina B/genética , Quinases Ciclina-Dependentes/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Lytechinus/embriologia , Lytechinus/genética , Lytechinus/metabolismo , RNA Mensageiro , Ouriços-do-Mar/genética , Ouriços-do-Mar/metabolismo , Fuso Acromático/metabolismo , Strongylocentrotus purpuratus/embriologia , Strongylocentrotus purpuratus/genética , Strongylocentrotus purpuratus/metabolismo
17.
Dev Dyn ; 242(2): 155-63, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23172739

RESUMO

BACKGROUND: Meiosis is a unique mechanism in gamete production and a fundamental process shared by all sexually reproducing eukaryotes. Meiosis requires several specialized and highly conserved genes whose expression can also identify the germ cells undergoing gametogenic differentiation. Sea urchins are echinoderms, which form a phylogenetic sister group of chordates. Sea urchin embryos undergo a feeding, planktonic larval phase in which they construct an adult rudiment prior to metamorphosis. Although a series of conserved meiosis genes (e.g., dmc1, msh5, rad21, rad51, and sycp1) is expressed in sea urchin oocytes, we sought to determine when in development meiosis would first be initiated. RESULTS: We surveyed the expression of several meiotic genes and their corresponding proteins in the sea urchin Strongylocentrotus purpuratus. Surprisingly, meiotic genes are highly expressed not only in ovaries but beginning in larvae. Both RNA and protein localizations strongly suggest that meiotic gene expression initiates in tissues that will eventually give rise to the adult rudiment of the late larva. CONCLUSIONS: These results demonstrate that broad expression of the molecules associated with meiotic differentiation initiates prior to metamorphosis and may have additional functions in these cells, or mechanisms repressing their function, until later in development when gametogenesis begins.


Assuntos
Proteínas de Ciclo Celular/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Meiose/genética , Strongylocentrotus purpuratus/metabolismo , Animais , Sequência de Bases , Clonagem Molecular , Primers do DNA/genética , Perfilação da Expressão Gênica , Hibridização in Situ Fluorescente , Larva/crescimento & desenvolvimento , Larva/metabolismo , Meiose/fisiologia , Dados de Sequência Molecular , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Strongylocentrotus purpuratus/crescimento & desenvolvimento
18.
Dev Biol ; 369(2): 377-85, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22771578

RESUMO

The Nodal signaling pathway is known from earlier work to be an essential mediator of oral ectoderm specification in the sea urchin embryo, and indirectly, of aboral ectoderm specification as well. Following expression of the Nodal ligand in the future oral ectoderm during cleavage, a sequence of regulatory gene activations occur within this territory which depend directly or indirectly on nodal gene expression. Here we describe additional regulatory genes that contribute to the oral ectoderm regulatory state during specification in Strongylocentrotus purpuratus, and show how their spatial expression changes dynamically during development. By means of system wide perturbation analyses we have significantly improved current knowledge of the epistatic relations among the regulatory genes of the oral ectoderm. From these studies there emerge diverse circuitries relating downstream regulatory genes directly and indirectly to Nodal signaling. A key intermediary regulator, the role of which had not previously been discerned, is the not gene. In addition to activating several genes earlier described as targets of Nodal signaling, the not gene product acts to repress other oral ectoderm genes, contributing crucially to the bilateral spatial organization of the embryonic oral ectoderm.


Assuntos
Ligantes da Sinalização Nodal/metabolismo , Strongylocentrotus purpuratus/embriologia , Strongylocentrotus purpuratus/genética , Animais , Sequência de Bases , Padronização Corporal/genética , Padronização Corporal/fisiologia , Primers do DNA/genética , Ectoderma/embriologia , Ectoderma/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Redes Reguladoras de Genes , Modelos Biológicos , Boca/embriologia , Boca/metabolismo , Ligantes da Sinalização Nodal/genética , Transdução de Sinais , Strongylocentrotus purpuratus/metabolismo
19.
J Biol Chem ; 287(4): 2308-15, 2012 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-22117077

RESUMO

Nicotinic acid adenine dinucleotide phosphate (NAADP) is a messenger that regulates calcium release from intracellular acidic stores. Recent studies have identified two-pore channels (TPCs) as endolysosomal channels that are regulated by NAADP; however, the nature of the NAADP receptor binding site is unknown. To further study NAADP binding sites, we have synthesized and characterized [(32)P-5-azido]nicotinic acid adenine dinucleotide phosphate ([(32)P-5N(3)]NAADP) as a photoaffinity probe. Photolysis of sea urchin egg homogenates preincubated with [(32)P-5N(3)]NAADP resulted in specific labeling of 45-, 40-, and 30-kDa proteins, which was prevented by inclusion of nanomolar concentrations of unlabeled NAADP or 5N(3)-NAADP, but not by micromolar concentrations of structurally related nucleotides such as NAD, nicotinic acid adenine dinucleotide, nicotinamide mononucleotide, nicotinic acid, or nicotinamide. [(32)P-5N(3)]NAADP binding was saturable and displayed high affinity (K(d) ∼10 nM) in both binding and photolabeling experiments. [(32)P-5N(3)]NAADP photolabeling was irreversible in a high K(+) buffer, a hallmark feature of NAADP binding in the egg system. The proteins photolabeled by [(32)P-5N(3)]NAADP have molecular masses smaller than the sea urchin TPCs, and antibodies to TPCs do not detect any immunoreactivity that comigrates with either the 45-kDa or the 40-kDa photolabeled proteins. Interestingly, antibodies to TPC1 and TPC3 were able to immunoprecipitate a small fraction of the 45- and 40-kDa photolabeled proteins, suggesting that these proteins associate with TPCs. These data suggest that high affinity NAADP binding sites are distinct from TPCs.


Assuntos
Canais de Cálcio/metabolismo , NADP/análogos & derivados , Óvulo/metabolismo , Strongylocentrotus purpuratus/metabolismo , Animais , Sítios de Ligação , NADP/metabolismo , Marcadores de Fotoafinidade/química , Ligação Proteica
20.
J Biol Chem ; 287(52): 43876-83, 2012 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-23124201

RESUMO

In this study, we cloned, expressed and functionally characterized Stronglycentrotus purpuratus (Sp) ATP-binding cassette (ABC) transporters. This screen identified three multidrug resistance (MDR) transporters with functional homology to the major types of MDR transporters found in humans. When overexpressed in embryos, the apical transporters Sp-ABCB1a, ABCB4a, and ABCG2a can account for as much as 87% of the observed efflux activity, providing a robust assay for their substrate selectivity. Using this assay, we found that sea urchin MDR transporters export canonical MDR susbtrates such as calcein-AM, bodipy-verapamil, bodipy-vinblastine, and mitoxantrone. In addition, we characterized the impact of nonconservative substitutions in the primary sequences of drug binding domains of sea urchin versus murine ABCB1 by mutation of Sp-ABCB1a and treatment of embryos with stereoisomeric cyclic peptide inhibitors (QZ59 compounds). The results indicated that two substitutions in transmembrane helix 6 reverse stereoselectivity of Sp-ABCB1a for QZ59 enantiomers compared with mouse ABCB1a. This suggests that subtle changes in the primary sequence of transporter drug binding domains could fine-tune substrate specificity through evolution.


Assuntos
Proteínas de Membrana Transportadoras/metabolismo , Strongylocentrotus purpuratus/metabolismo , Animais , Embrião não Mamífero/metabolismo , Proteínas de Membrana Transportadoras/genética , Camundongos , Mutação , Peptídeos Cíclicos/farmacologia , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Strongylocentrotus purpuratus/genética , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA