Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
BMC Biol ; 19(1): 257, 2021 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-34863182

RESUMO

BACKGROUND: The evolutionary history of cell types provides insights into how morphological and functional complexity arose during animal evolution. Photoreceptor cell types are particularly broadly distributed throughout Bilateria; however, their evolutionary relationship is so far unresolved. Previous studies indicate that ciliary photoreceptors are homologous at least within chordates, and here, we present evidence that a related form of this cell type is also present in echinoderm larvae. RESULTS: Larvae of the purple sea urchin Strongylocentrotus purpuratus have photoreceptors that are positioned bilaterally in the oral/anterior apical neurogenic ectoderm. Here, we show that these photoreceptors express the transcription factor Rx, which is commonly expressed in ciliary photoreceptors, together with an atypical opsin of the GO family, opsin3.2, which localizes in particular to the cilia on the cell surface of photoreceptors. We show that these ciliary photoreceptors express the neuronal marker synaptotagmin and are located in proximity to pigment cells. Furthermore, we systematically identified additional transcription factors expressed in these larval photoreceptors and found that a majority are orthologous to transcription factors expressed in vertebrate ciliary photoreceptors, including Otx, Six3, Tbx2/3, and Rx. Based on the developmental expression of rx, these photoreceptors derive from the anterior apical neurogenic ectoderm. However, genes typically involved in eye development in bilateria, including pax6, six1/2, eya, and dac, are not expressed in sea urchin larval photoreceptors but are instead co-expressed in the hydropore canal. CONCLUSIONS: Based on transcription factor expression, location, and developmental origin, we conclude that the sea urchin larval photoreceptors constitute a cell type that is likely homologous to the ciliary photoreceptors present in chordates.


Assuntos
Células Fotorreceptoras , Ouriços-do-Mar , Animais , Ectoderma/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Larva , Células Fotorreceptoras/metabolismo , Ouriços-do-Mar/genética , Ouriços-do-Mar/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
2.
Development ; 144(19): 3602-3611, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28851710

RESUMO

We have examined regulation of neurogenesis by Delta/Notch signaling in sea urchin embryos. At gastrulation, neural progenitors enter S phase coincident with expression of Sp-SoxC. We used a BAC containing GFP knocked into the Sp-SoxC locus to label neural progenitors. Live imaging and immunolocalizations indicate that Sp-SoxC-expressing cells divide to produce pairs of adjacent cells expressing GFP. Over an interval of about 6 h, one cell fragments, undergoes apoptosis and expresses high levels of activated Caspase3. A Notch reporter indicates that Notch signaling is activated in cells adjacent to cells expressing Sp-SoxC. Inhibition of γ-secretase, injection of Sp-Delta morpholinos or CRISPR/Cas9-induced mutation of Sp-Delta results in supernumerary neural progenitors and neurons. Interfering with Notch signaling increases neural progenitor recruitment and pairs of neural progenitors. Thus, Notch signaling restricts the number of neural progenitors recruited and regulates the fate of progeny of the asymmetric division. We propose a model in which localized signaling converts ectodermal and ciliary band cells to neural progenitors that divide asymmetrically to produce a neural precursor and an apoptotic cell.


Assuntos
Divisão Celular Assimétrica , Embrião não Mamífero/metabolismo , Células-Tronco Neurais/citologia , Neurogênese , Receptores Notch/metabolismo , Ouriços-do-Mar/embriologia , Ouriços-do-Mar/metabolismo , Transdução de Sinais , Animais , Apoptose , Padronização Corporal , Ectoderma/citologia , Ectoderma/metabolismo , Embrião não Mamífero/citologia , Proteínas de Fluorescência Verde/metabolismo , Mitose , Modelos Biológicos , Células-Tronco Neurais/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Fatores de Transcrição SOX/metabolismo
3.
Development ; 143(2): 286-97, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26511925

RESUMO

A single origin to the diverse mechanisms of metazoan neurogenesis is suggested by the involvement of common signaling components and similar classes of transcription factors. However, in many forms we lack details of where neurons arise, patterns of cell division, and specific differentiation pathway components. The sea urchin larval nervous system is composed of an apical organ, which develops from neuroepithelium and functions as a central nervous system, and peripheral neurons, which differentiate in the ciliary band and project axons to the apical organ. To reveal developmental mechanisms of neurogenesis in this basal deuterostome, we developed antibodies to SoxC, SoxB2, ELAV and Brn1/2/4 and used neurons that develop at specific locations to establish a timeline for neurogenesis. Neural progenitors express, in turn, SoxB2, SoxC, and Brn1/2/4, before projecting neurites and expressing ELAV and SynB. Using pulse-chase labeling of cells with a thymidine analog to identify cells in S-phase, we establish that neurons identified by location are in their last mitotic cycle at the time of hatching, and S-phase is coincident with expression of SoxC. The number of cells expressing SoxC and differentiating as neurons is reduced in embryos injected with antisense morpholino oligonucleotides to SoxC, SoxB2 or Six3. Injection of RNA encoding SoxC into eggs does not enhance neurogenesis. In addition, inhibition of FGF receptors (SU5402) or a morpholino to FGFR1 reduces expression of SoxC. These data indicate that there are common features of neurogenesis in deuterostomes, and that sea urchins employ developmental mechanisms that are distinct from other ambulacraria.


Assuntos
Embrião não Mamífero/citologia , Larva/citologia , Neurogênese/fisiologia , Ouriços-do-Mar/citologia , Animais , Regulação da Expressão Gênica no Desenvolvimento/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Larva/metabolismo , Neurogênese/genética , Neurônios/citologia , Neurônios/metabolismo , Ouriços-do-Mar/metabolismo
4.
RNA ; 23(6): 952-967, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28325844

RESUMO

Proteins of the Sm and Sm-like (LSm) families, referred to collectively as (L)Sm proteins, are found in all three domains of life and are known to promote a variety of RNA processes such as base-pair formation, unwinding, RNA degradation, and RNA stabilization. In eukaryotes, (L)Sm proteins have been studied, inter alia, for their role in pre-mRNA splicing. In many organisms, the LSm proteins form two distinct complexes, one consisting of LSm1-7 that is involved in mRNA degradation in the cytoplasm, and the other consisting of LSm2-8 that binds spliceosomal U6 snRNA in the nucleus. We recently characterized the splicing proteins from the red alga Cyanidioschyzon merolae and found that it has only seven LSm proteins. The identities of CmLSm2-CmLSm7 were unambiguous, but the seventh protein was similar to LSm1 and LSm8. Here, we use in vitro binding measurements, microscopy, and affinity purification-mass spectrometry to demonstrate a canonical splicing function for the C. merolae LSm complex and experimentally validate our bioinformatic predictions of a reduced spliceosome in this organism. Copurification of Pat1 and its associated mRNA degradation proteins with the LSm proteins, along with evidence of a cytoplasmic fraction of CmLSm complexes, argues that this complex is involved in both splicing and cytoplasmic mRNA degradation. Intriguingly, the Pat1 complex also copurifies with all four snRNAs, suggesting the possibility of a spliceosome-associated pre-mRNA degradation complex in the nucleus.


Assuntos
Precursores de RNA/genética , Splicing de RNA , RNA Mensageiro/genética , Proteínas de Ligação a RNA/metabolismo , Rodófitas/genética , Rodófitas/metabolismo , Sequência de Aminoácidos , Sequência de Bases , Biologia Computacional/métodos , Imunoprecipitação , Modelos Moleculares , Conformação de Ácido Nucleico , Filogenia , Ligação Proteica , Conformação Proteica , Transporte Proteico , Precursores de RNA/química , Estabilidade de RNA , RNA Mensageiro/química , RNA Nuclear Pequeno/química , RNA Nuclear Pequeno/genética , Proteínas de Ligação a RNA/química , Espectrometria de Massas em Tandem
5.
Development ; 141(5): 1075-84, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24550115

RESUMO

Apical constriction typically accompanies inward folding of an epithelial sheet. In recent years there has been progress in understanding mechanisms of apical constriction and their contribution to morphogenetic processes. Sea urchin embryos form a specialized region of ectoderm, the ciliary band, which is a strip of epithelium, three to five cells wide, encircling the oral ectoderm and functioning in larval swimming and feeding. Ciliary band cells exhibit distinctive apical-basal elongation, have narrow apices bearing a cilium, and are planar polarized, so that cilia beat away from the mouth. Here, we show that filamentous actin and phosphorylated myosin light chain are uniquely distributed in ciliary band cells. Inhibition of myosin phosphorylation or actin polymerization perturbs this distribution and blocks apical constriction. During ciliary band formation, Sp-Ephrin and Sp-Eph expression overlap in the presumptive ciliary band. Knockdown of Sp-Eph or Sp-Ephrin, or treatment with an Eph kinase inhibitor interferes with actomyosin networks, accumulation of phosphorylated FAK (pY(397)FAK), and apical constriction. The cytoplasmic domain of Sp-Eph, fused to GST and containing a single amino acid substitution reported as kinase dead, will pull down pY(397)FAK from embryo lysates. As well, pY(397)FAK colocalizes with Sp-Eph in a JNK-dependent, planar polarized manner on latitudinal apical junctions of the ciliary band and this polarization is dissociable from apical constriction. We propose that Sp-Eph and pY(397)FAK function together in an apical complex that is necessary for remodeling actomyosin to produce centripetal forces causing apical constriction. Morphogenesis of ciliary band cells is a unique example of apical constriction in which receptor-mediated cell shape change produces a strip of specialized tissue without an accompanying folding of epithelium.


Assuntos
Actomiosina/metabolismo , Efrinas/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Receptores da Família Eph/metabolismo , Strongylocentrotus purpuratus/embriologia , Animais , Polaridade Celular/genética , Polaridade Celular/fisiologia , Embrião não Mamífero/metabolismo , Efrinas/genética , Feminino , Proteína-Tirosina Quinases de Adesão Focal/genética , Masculino , Morfogênese/genética , Morfogênese/fisiologia , Receptores da Família Eph/genética , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
6.
Genesis ; 52(3): 208-21, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25368883

RESUMO

Summary:Urchin embryos continue to prove useful as a means of studying embryonic signaling and gene regulatory networks, which together control early development. Recent progress in understanding the molecular mechanisms underlying the patterning of ectoderm has renewed interest in urchin neurogenesis. We have employed an emerging model of neurogenesis that appears to be broadly shared by metazoans as a framework for this review. We use the model to provide context and summarize what is known about neurogenesis in urchin embryos. We review morphological features of the differentiation phase of neurogenesis and summarize current understanding of neural specification and regulation of proneural networks. Delta-Notch signaling is a common feature of metazoan neurogenesis that produces committed progenitors and it appears to be a critical phase of neurogenesis in urchin embryos. Descriptions of the differentiation phase of neurogenesis indicate a stereotypic sequence of neural differentiation and patterns of axonal growth. Features of neural differentiation are consistent with localized signals guiding growth cones with trophic, adhesive, and tropic cues. Urchins are a facile, postgenomic model with the potential of revealing many shared and derived features of deuterostome neurogenesis.


Assuntos
Neurogênese/fisiologia , Ouriços-do-Mar/embriologia , Animais , Embrião não Mamífero/inervação , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Modelos Biológicos , Neurogênese/genética , Especificidade de Órgãos , Receptores Notch/genética , Receptores Notch/metabolismo , Ouriços-do-Mar/genética
7.
Dev Biol ; 377(1): 236-44, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23506838

RESUMO

The structure and development of the larval nervous systems of all classes of echinoderms have been described and details of embryonic signaling mechanisms patterning neurogenesis have been revealed experimentally in sea urchins. Several features of neuroanatomy and neural development indicate that echinoids are the most derived group. Here we describe the development and organization of the nervous system of a cidaroid, Eucidaris tribuloides. The cidaroids are one of two major clades of echinoids, and are considered to have features of anatomy and development that represent the common ancestor to all echinoids. The embryos of E. tribuloides lack a thickened animal plate and serotonergic neurons arise laterally, associated with the ciliary band. Although lacking a discrete apical organ, plutei have serotonergic neurons associated with the pre-oral ciliary band joined by a few diffusely arranged connecting axons. Chordin and Hnf6, early markers for oral ectoderm and ciliary band, are expressed in similar patterns to euechinoids. However, an animal pole domain marker, Nk2.1, is expressed in a broader region of anterior ectoderm than in euechinoids. Six3, a proneural marker that is restricted to the animal plate of euechinoids, is expressed laterally in the preoral ciliary band at the same location as the serotonergic neurons. We conclude that the organization and development of the larval nervous system of E. tribuloides retains features shared with other echinoderm larvae, but not with euechinoids. These data support a model in which several distinctive features of euechinoid neural organization are derived, having arisen after the divergence of the two clades of echinoids about 265 million years ago. We hypothesize that differences in the developmental mechanisms that restrict neurogenesis to the animal pole forms the basis for the distinctive neuroanatomy of euechinoids.


Assuntos
Evolução Biológica , Sistema Nervoso/crescimento & desenvolvimento , Ouriços-do-Mar/crescimento & desenvolvimento , Animais , Biomarcadores/metabolismo , Ectoderma/citologia , Ectoderma/embriologia , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Larva/citologia , Larva/crescimento & desenvolvimento , Larva/metabolismo , Sistema Nervoso/citologia , Sistema Nervoso/embriologia , Neurogênese , Filogenia , Ouriços-do-Mar/citologia , Ouriços-do-Mar/embriologia , Neurônios Serotoninérgicos/citologia , Neurônios Serotoninérgicos/metabolismo , Serotonina/metabolismo , Sinaptotagminas/metabolismo , Via de Sinalização Wnt
8.
Development ; 138(17): 3613-23, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21828090

RESUMO

Recent studies of the sea urchin embryo have elucidated the mechanisms that localize and pattern its nervous system. These studies have revealed the presence of two overlapping regions of neurogenic potential at the beginning of embryogenesis, each of which becomes progressively restricted by separate, yet linked, signals, including Wnt and subsequently Nodal and BMP. These signals act to specify and localize the embryonic neural fields - the anterior neuroectoderm and the more posterior ciliary band neuroectoderm - during development. Here, we review these conserved nervous system patterning signals and consider how the relationships between them might have changed during deuterostome evolution.


Assuntos
Padronização Corporal/fisiologia , Sistema Nervoso/metabolismo , Ouriços-do-Mar/enzimologia , Ouriços-do-Mar/metabolismo , Animais , Padronização Corporal/genética , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Proteína Nodal/genética , Proteína Nodal/metabolismo , Ouriços-do-Mar/crescimento & desenvolvimento , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
9.
Langmuir ; 28(6): 3239-47, 2012 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-22250577

RESUMO

Ligands on the nanoparticle surface provide steric stabilization, resulting in good dispersion stability. However, because of their highly dynamic nature, they can be replaced irreversibly in buffers and biological medium, leading to poor colloidal stability. To overcome this, we report a simple and effective cross-linking methodology to transfer oleate-stabilized upconverting NaYF(4) core/shell nanoparticles (UCNPs) from hydrophobic to aqueous phase, with long-term dispersion stability in buffers and biological medium. Amphiphilic poly(maleic anhydride-alt-1-octadecene) (PMAO) modified with and without poly(ethylene glycol) (PEG) was used to intercalate with the surface oleates, enabling the transfer of the UCNPs to water. The PMAO units on the phase transferred UCNPs were then successfully cross-linked using bis(hexamethylene)triamine (BHMT). The primary advantage of cross-linking of PMAO by BHMT is that it improves the stability of the UCNPs in water, physiological saline buffers, and biological growth media and in a wide range of pH values when compared to un-cross-linked PMAO. The cross-linked PMAO-BHMT coated UCNPs were found to be stable in water for more than 2 months and in physiological saline buffers for weeks, substantiating the effectiveness of cross-linking in providing high dispersion stability. The PMAO-BHMT cross-linked UCNPs were extensively characterized using various techniques providing supporting evidence for the cross-linking process. These UCNPs were found to be stable in serum supplemented growth medium (37 °C) for more than 2 days. Utilizing this, we demonstrate the uptake of cross-linked UCNPs by LNCaP cells (human prostate cancer cell line), showing their utility as biolabels.


Assuntos
Fluoretos/química , Nanopartículas/química , Nanotecnologia/métodos , Polímeros/química , Ítrio/química , Soluções Tampão , Meios de Cultura/química
10.
Nat Struct Mol Biol ; 14(1): 76-84, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17187076

RESUMO

The ability of pathogenic bacteria to recognize host glycans is often essential to their virulence. Here we report structure-function studies of previously uncharacterized glycogen-binding modules in the surface-anchored pullulanases from Streptococcus pneumoniae (SpuA) and Streptococcus pyogenes (PulA). Multivalent binding to glycogen leads to a strong interaction with alveolar type II cells in mouse lung tissue. X-ray crystal structures of the binding modules reveal a novel fusion of tandem modules into single, bivalent functional domains. In addition to indicating a structural basis for multivalent attachment, the structure of the SpuA modules in complex with carbohydrate provides insight into the molecular basis for glycogen specificity. This report provides the first evidence that intracellular lung glycogen may be a novel target of pathogenic streptococci and thus provides a rationale for the identification of the streptococcal alpha-glucan-metabolizing machinery as virulence factors.


Assuntos
Glicogênio/metabolismo , Glicosídeo Hidrolases/metabolismo , Alvéolos Pulmonares/metabolismo , Streptococcus pneumoniae/patogenicidade , Streptococcus pyogenes/patogenicidade , Fatores de Virulência/química , Fatores de Virulência/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Cristalografia por Raios X , Glucanos/metabolismo , Glicosídeo Hidrolases/química , Glicosídeo Hidrolases/genética , Ligantes , Camundongos , Dados de Sequência Molecular , Alvéolos Pulmonares/citologia , Streptococcus pneumoniae/enzimologia , Streptococcus pneumoniae/metabolismo , Streptococcus pyogenes/enzimologia , Streptococcus pyogenes/genética , Fatores de Virulência/genética
11.
Curr Top Dev Biol ; 146: 149-182, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35152982

RESUMO

Larvae of sea urchins have a population of conspicuous pigmented cells embedded in the outer surface epithelium. Pigment cells are a distinct mesodermal lineage that gives rise to a key component of the larval immune system. During cleavage, signaling from adjacent cells influences a small crescent of cells to initiate a network of genetic interactions that prepare the cells for morphogenesis and specializes them as immunocytes. The cells become active during gastrulation, detach from the epithelium, migrate through the blastocoel, and insert into the ectoderm where they complete their differentiation. Studies of pigment cell development have helped establish how cellular signaling controls networks of genetic interactions that bring about morphogenesis and differentiation. This review summarizes studies of pigment cell development and concludes that pigment cells are an excellent experimental model. Pigment cells provide several opportunities to further test and refine our understanding of the molecular basis of cellular development.


Assuntos
Mesoderma , Ouriços-do-Mar , Animais , Diferenciação Celular , Ectoderma , Gastrulação , Ouriços-do-Mar/genética
12.
Dev Biol ; 347(1): 71-81, 2010 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-20709054

RESUMO

The ciliary band is a distinct region of embryonic ectoderm that is specified between oral and aboral ectoderm. Flask-shaped ciliary cells and neurons differentiate in this region and they are patterned to form an integrated tissue that functions as the principal swimming and feeding organ of the larva. TGFß signaling, which is known to mediate oral and aboral patterning of the ectoderm, has been implicated in ciliary band formation. We have used morpholino knockdown and ectopic expression of RNA to alter TGFß signaling at the level of ligands, receptors, and signal transduction components and assessed the differentiation and patterning of the ciliary band cells and associated neurons. We propose that the primary effects of these signals are to position the ciliary cells, which in turn support neural differentiation. We show that Nodal signaling, which is known to be localized by Lefty, positions the oral margin of the ciliary band. Signaling from BMP through Alk3/6, affects the position of the oral and aboral margins of the ciliary band. Since both Nodal and BMP signaling produce ectoderm that does not support neurogenesis, we propose that formation of a ciliary band requires protection from these signals. Expression of BMP2/4 and Nodal suppress neural differentiation. However, the response to receptor knockdown or dominant-negative forms of signal transduction components indicate signaling is not acting directly on unspecified ectoderm cells to prevent their differentiation as neurons. Instead, it produces a restricted field of ciliary band cells that supports neurogenesis. We propose a model that incorporates spatially regulated control of Nodal and BMP signaling to determine the position and differentiation of the ciliary band, and subsequent neural patterning.


Assuntos
Padronização Corporal , Cílios/metabolismo , Embrião não Mamífero/citologia , Neurônios/metabolismo , Ouriços-do-Mar/embriologia , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Ectoderma/citologia , Ectoderma/metabolismo , Embrião não Mamífero/metabolismo , Larva/citologia , Larva/metabolismo , Modelos Biológicos , Sistema Nervoso/citologia , Sistema Nervoso/embriologia , Sistema Nervoso/metabolismo , Neurônios/citologia , Proteína Nodal/metabolismo , Ouriços-do-Mar/citologia , Ouriços-do-Mar/metabolismo
13.
J Biol Chem ; 284(38): 26161-73, 2009 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-19608744

RESUMO

The presence of a fucose utilization operon in the Streptococcus pneumoniae genome and its established importance in virulence indicates a reliance of this bacterium on the harvesting of host fucose-containing glycans. The identities of these glycans, however, and how they are harvested is presently unknown. The biochemical and high resolution x-ray crystallographic analysis of two family 98 glycoside hydrolases (GH98s) from distinctive forms of the fucose utilization operon that originate from different S. pneumoniae strains reveal that one enzyme, the predominant type among pneumococcal isolates, has a unique endo-beta-galactosidase activity on the LewisY antigen. Altered active site topography in the other species of GH98 enzyme tune its endo-beta-galactosidase activity to the blood group A and B antigens. Despite their different specificities, these enzymes, and by extension all family 98 glycoside hydrolases, use an inverting catalytic mechanism. Many bacterial and viral pathogens exploit host carbohydrate antigens for adherence as a precursor to colonization or infection. However, this is the first evidence of bacterial endoglycosidase enzymes that are known to play a role in virulence and are specific for distinct host carbohydrate antigens. The strain-specific distribution of two distinct types of GH98 enzymes further suggests that S. pneumoniae strains may specialize to exploit host-specific antigens that vary from host to host, a factor that may feature in whether a strain is capable of colonizing a host or establishing an invasive infection.


Assuntos
Proteínas de Bactérias/química , Glicosídeo Hidrolases/química , Antígenos do Grupo Sanguíneo de Lewis/química , Streptococcus pneumoniae/enzimologia , Proteínas de Bactérias/metabolismo , Glicosídeo Hidrolases/metabolismo , Humanos , Antígenos do Grupo Sanguíneo de Lewis/metabolismo , Óperon , Infecções Pneumocócicas/enzimologia , Especificidade da Espécie , Streptococcus pneumoniae/patogenicidade , Especificidade por Substrato/fisiologia
14.
Dev Genes Evol ; 220(7-8): 221-34, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21069538

RESUMO

The transcription factor COE (collier/olfactory-1/early B cell factor) is an unusual basic helix-loop-helix transcription factor as it lacks a basic domain and is maintained as a single copy gene in the genomes of all currently analysed non-vertebrate Metazoan genomes. Given the unique features of the COE gene, its proposed ancestral role in the specification of chemosensory neurons and the wealth of functional data from vertebrates and Drosophila, the evolutionary history of the COE gene can be readily investigated. We have examined the ways in which COE expression has diversified among the Metazoa by analysing its expression from representatives of four disparate invertebrate phyla: Ctenophora (Mnemiopsis leidyi); Mollusca (Haliotis asinina); Annelida (Capitella teleta and Chaetopterus) and Echinodermata (Strongylocentrotus purpuratus). In addition, we have studied COE function with knockdown experiments in S. purpuratus, which indicate that COE is likely to be involved in repressing serotonergic cell fate in the apical ganglion of dipleurula larvae. These analyses suggest that COE has played an important role in the evolution of ectodermally derived tissues (likely primarily nervous tissues) and mesodermally derived tissues. Our results provide a broad evolutionary foundation from which further studies aimed at the functional characterisation and evolution of COE can be investigated.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Evolução Molecular , Invertebrados/crescimento & desenvolvimento , Invertebrados/genética , Mesoderma/crescimento & desenvolvimento , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Invertebrados/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Vertebrados/genética , Vertebrados/crescimento & desenvolvimento , Vertebrados/metabolismo
15.
Dev Genes Evol ; 219(3): 159-66, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19238430

RESUMO

Comparative features of the development of the larval nervous system of ophiuroids have the potential for resolving aspects of echinoderm evolution. In Amphiura filiformis serotonergic neural progenitors appear in the animal plate of late gastrulae. The serotonergic progenitors increase in number and become displaced to the aboral ectoderm side of the developing ciliary band. The ciliary band neurons appear as irregularly spaced neural progenitors on the oral side of the ciliary band lateral to the mouth. These cells extend neurites along the axis of the ciliary band, which meet at the center of the ventral transverse ciliary band. The larval nervous system begins as a U-shaped tract of axons that surrounds the oral field and tracts of axons and neurons in the ciliary bands of the larval arms are added. In addition, the larval nervous system has an extensive pre-oral neuropil, rings of nerves surrounding the anus and pyloric sphincters, and a plexus of axons that surround the esophagus. The nervous system of the juvenile develops beneath the oral ectoderm. The components of the adult nervous system: five segments of radial nerve, commissures that form the nerve ring, and podial nerves all appear as the juvenile develops. The larval nervous system begins to fragment and degenerate as the juvenile grows. The complete description of neural development of an ophiuroid reveals that the four classes so far investigated are consistent with phylogenies based on adult features and comparisons of neural organization help rationalize conflicting hypotheses of the evolution of larval forms in echinoderms.


Assuntos
Equinodermos/crescimento & desenvolvimento , Neurogênese , Animais , Equinodermos/anatomia & histologia , Larva/crescimento & desenvolvimento , Sistema Nervoso/crescimento & desenvolvimento , Filogenia
16.
Biomed Microdevices ; 11(6): 1317-30, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19763834

RESUMO

Single cell research has the potential to revolutionize experimental methods in biomedical sciences and contribute to clinical practices. Recent studies suggest analysis of single cells reveals novel features of intracellular processes, cell-to-cell interactions and cell structure. The methods of single cell analysis require mechanical resolution and accuracy that is not possible using conventional techniques. Robotic instruments and novel microdevices can achieve higher throughput and repeatability; however, the development of such instrumentation is a formidable task. A void exists in the state-of-the-art for automated analysis of single cells. With the increase in interest in single cell analyses in stem cell and cancer research the ability to facilitate higher throughput and repeatable procedures is necessary. In this paper, a high-throughput, single cell microarray-based robotic instrument, called the RoboSCell, is described. The proposed instrument employs a partially transparent single cell microarray (SCM) integrated with a robotic biomanipulator for in vitro analyses of live single cells trapped at the array sites. Cells, labeled with immunomagnetic particles, are captured at the array sites by channeling magnetic fields through encapsulated permalloy channels in the SCM. The RoboSCell is capable of systematically scanning the captured cells temporarily immobilized at the array sites and using optical methods to repeatedly measure extracellular and intracellular characteristics over time. The instrument's capabilities are demonstrated by arraying human T lymphocytes and measuring the uptake dynamics of calcein acetoxymethylester--all in a fully automated fashion.


Assuntos
Robótica , Análise Serial de Tecidos/instrumentação , Contagem de Células , Sobrevivência Celular , Fluorescência , Humanos , Células Jurkat
17.
Methods Cell Biol ; 151: 49-54, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30948029

RESUMO

Studies using sea urchins have contributed substantially to our understanding of how a fertilized egg is transformed during embryonic development. This brief review provides a personal perspective of the remarkable advances that have occurred over the past 45 years in our understanding of how urchin embryos work.


Assuntos
Biologia do Desenvolvimento/história , Desenvolvimento Embrionário/genética , Ouriços-do-Mar/crescimento & desenvolvimento , Animais , Embrião não Mamífero , História do Século XX , História do Século XXI , Ouriços-do-Mar/genética
18.
Methods Cell Biol ; 151: 519-526, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30948030

RESUMO

Fluorescent calcium sensors provide a means of detecting and analyzing cytoplasmic calcium levels in embryos and larvae. Conventional RNA injection of eggs results in expression of protein sensors throughout larval tissues. Larvae are immobilized for wide field or confocal recordings and video records reveal recurrent fluctuations in cytoplasmic calcium levels in several cell types. Neurons can be identified by location and form, and continuous records made of their activity. Confocal image stacks are registered and Z-axis, fluorescence intensity profiles of individual neurons generated to provide time/activity plots. These optogenetic methods enable analysis in intact larvae of the activity of identified neurons or effectors, such as muscles or ciliary band cells.


Assuntos
Técnicas Biossensoriais/métodos , Microscopia Confocal/métodos , Neurônios/efeitos dos fármacos , Óvulo/efeitos dos fármacos , Cálcio/metabolismo , Citoplasma/efeitos dos fármacos , Corantes Fluorescentes/química , Óvulo/crescimento & desenvolvimento , RNA/administração & dosagem , RNA/química
19.
BMC Dev Biol ; 8: 56, 2008 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-18495033

RESUMO

BACKGROUND: As in other vertebrates, avian hindbrain neural crest migrates in streams to specific branchial arches. Signalling from Eph receptors and ephrins has been proposed to provide a molecular mechanism that guides the cells restricting them to streams. In mice and frogs, cranial neural crest express a combination of Eph receptors and ephrins that appear to exclude cells from adjacent tissues by forward and reverse signalling. The objective of this study was to provide comparative data on the distribution and function of Eph receptors and ephrins in avian embryos. RESULTS: To distinguish neural crest from bordering ectoderm and head mesenchyme, we have co-labelled embryos for Eph or ephrin RNA and a neural crest marker protein. Throughout their migration avian cranial neural crest cells express EphA3, EphA4, EphA7, EphB1, and EphB3 and move along pathways bordered by non-neural crest cells expressing ephrin-B1. In addition, avian cranial neural crest cells express ephrin-B2 and migrate along pathways bordered by non-neural crest cells expressing EphB2. Thus, the distribution of avian Eph receptors and ephrins differs from those reported in other vertebrates. In stripe assays when explanted cranial neural crest were given the choice between FN or FN plus clustered ephrin-B1 or EphB2 fusion protein, the cells strongly localize to lanes containing only FN. This preference is mitigated in the presence of soluble ephrin-B1 or EphB2 fusion protein. CONCLUSION: These findings show that avian cranial neural crest use Eph and ephrin receptors as other vertebrates in guiding migration. However, the Eph receptors are expressed in different combinations by neural crest destined for each branchial arch and ephrin-B1 and ephrin-B2 appear to have opposite roles to those reported to guide cranial neural crest migration in mice. Unlike many of the signalling, specification, and effector pathways of neural crest, the roles of Eph receptors and ephrins have not been rigorously conserved. This suggests diversification of receptor and ligand expression is less constrained, possibly by promiscuous binding and use of common downstream pathways.


Assuntos
Embrião de Galinha/embriologia , Efrina-B1/genética , Efrina-B2/genética , Crista Neural/embriologia , Receptor EphB2/genética , Rombencéfalo/embriologia , Animais , Padronização Corporal/genética , Células Cultivadas , Regulação da Expressão Gênica no Desenvolvimento , Hibridização In Situ , Camundongos , Plasmídeos
20.
BMC Cell Biol ; 9: 27, 2008 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-18495034

RESUMO

BACKGROUND: The tissue distributions and functions of Eph receptors and their ephrin ligands have been well studied, however less is known about their evolutionary history. We have undertaken a phylogenetic analysis of Eph receptors and ephrins from a number of invertebrate and vertebrate species. RESULTS: Our findings indicate that Eph receptors form three major clades: one comprised of non-chordate and cephalochordate Eph receptors, a second comprised of urochordate Eph receptors, and a third comprised of vertebrate Eph receptors. Ephrins, on the other hand, fall into either a clade made up of the non-chordate and cephalochordate ephrins plus the urochordate and vertebrate ephrin-Bs or a clade made up of the urochordate and vertebrate ephrin-As. CONCLUSION: We have concluded that Eph receptors and ephrins diverged into A and B-types at different points in their evolutionary history, such that primitive chordates likely possessed an ancestral ephrin-A and an ancestral ephrin-B, but only a single Eph receptor. Furthermore, ephrin-As appear to have arisen in the common ancestor of urochordates and vertebrates, whereas ephrin-Bs have a more ancient bilaterian origin. Ancestral ephrin-B-like ligands had transmembrane domains; as GPI anchors appear to have arisen or been lost at least 3 times.


Assuntos
Crista Neural/metabolismo , Receptores da Família Eph/genética , Animais , Sítios de Ligação , Comunicação Celular , Efrinas/genética , Evolução Molecular , Humanos , Ligantes , Funções Verossimilhança , Crista Neural/embriologia , Filogenia , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Receptor Cross-Talk , Alinhamento de Sequência , Análise de Sequência , Urocordados , Vertebrados
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA