Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Métodos Terapéuticos y Terapias MTCI
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Environ Sci Pollut Res Int ; 26(21): 21629-21640, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31129896

RESUMEN

Pesticides are capable of increasing risks to the early development of nontarget organisms through oxidative stress. The supplementation of antioxidants could help to modulate the toxic effects of pesticides, but much remains to be understood in the interactions between pesticides and antioxidants in amphibians. In the present study, the embryotoxicity of a widely used pyrethroid, lambda-cyhalothrin (LCT), and the potential effect of α-tocopherol (TOC) on embryos of Xenopus tropicalis were evaluated. Exposure to LCT did not affect the hatch rate, survival, or body length of the embryos. However, environmentally relevant concentrations of LCT could induce significant malformations on the larvae. Exposure to LCT led to a concentration-dependent induction of oxidative stress and cytotoxicity that subsequently resulted in embryotoxicity. During the early developmental stages, vitamin E could work as a powerful protective antioxidant. The LCT-induced overproduction of reactive oxygen species and increased enzymatic activities were fully inhibited by treatment with 1 µg/L TOC. However, only supplementation with 100 µg/L TOC provided partial protection against the morphological changes caused by LCT. The results from the present study suggest that antioxidant vitamin E possesses protective potential against pyrethroid-induced embryotoxicity in amphibian embryos through the prevention of oxidative stress.


Asunto(s)
Antioxidantes/metabolismo , Insecticidas/toxicidad , Nitrilos/toxicidad , Piretrinas/toxicidad , Vitamina E/metabolismo , Xenopus/embriología , Animales , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/fisiología , Estrés Oxidativo/efectos de los fármacos , Plaguicidas/toxicidad
2.
FASEB J ; 31(8): 3622-3635, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28432198

RESUMEN

Periconception maternal folic acid (vitamin B9) supplementation can reduce the prevalence of neural tube defects (NTDs), although just how folates benefit the developing embryo and promote closing of the neural tube and other morphologic processes during development remains unknown. Folate contributes to a 1-carbon metabolism, which is essential for purine biosynthesis and methionine recycling and affects methylation of DNA, histones, and nonhistone proteins. Herein, we used animal models and cultured mammalian cells to demonstrate that disruption of the methylation pathway mediated by folate compromises normal neural tube closure (NTC) and ciliogenesis. We demonstrate that the embryos with NTD failed to adequately methylate septin2, a key regulator of cilium structure and function. We report that methylation of septin2 affected its GTP binding activity and formation of the septin2-6-7 complex. We propose that folic acid promotes normal NTC in some embryos by regulating the methylation of septin2, which is critical for normal cilium formation during early embryonic development.-Toriyama, M., Toriyama, M., Wallingford, J. B., Finnell, R. H. Folate-dependent methylation of septins governs ciliogenesis during neural tube closure.


Asunto(s)
Cilios/fisiología , Embrión de Mamíferos/metabolismo , Embrión no Mamífero/metabolismo , Ácido Fólico/metabolismo , Tubo Neural/fisiología , Septinas/metabolismo , Animales , Dactinomicina/análogos & derivados , Desarrollo Embrionario/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Células HEK293 , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Metilación , Ratones , Defectos del Tubo Neural/etiología , Plásmidos , Transducción de Señal , Xenopus/embriología
3.
Cancer Sci ; 107(6): 803-11, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27019404

RESUMEN

Cancer tissues have biological characteristics similar to those observed in embryos during development. Many types of cancer cells acquire pro-invasive ability through epithelial-mesenchymal transition (EMT). Similar processes (gastrulation and migration of cranial neural crest cells [CNCC]) are observed in the early stages of embryonic development in Xenopus during which cells that originate from epithelial sheets through EMT migrate to their final destinations. The present study examined Xenopus embryonic tissues to identify anti-cancer compounds that prevent cancer invasion. From the initial test of known anti-cancer drugs, AMD3100 (an inhibitor of CXCR4) and paclitaxel (a cytoskeletal drug targeting microtubules) effectively prevented migration during gastrulation or CNCC development. Blind-screening of 100 synthesized chemical compounds was performed, and nine candidates that inhibited migration of these embryonic tissues without embryonic lethality were selected. Of these, C-157 (an analog of podophyllotoxin) and D-572 (which is an indole alkaroid) prevented cancer cell invasion through disruption of interphase microtubules. In addition, these compounds affected progression of mitotic phase and induced apoptosis of SAS oral cancer cells. SAS tumors were reduced in size after intratumoral injection of C-157, and peritoneal dissemination of melanoma cells and intracranial invasion of glioma cells were inhibited by C-157 and D-572. When the other analogues of these chemicals were compared, those with subtle effect on embryos were not tumor suppressive. These results suggest that a novel chemical-screening approach based on Xenopus embryos is an effective method for isolating anti-cancer drugs and, in particular, targeting cancer cell invasion and proliferation.


Asunto(s)
Antineoplásicos/análisis , Antineoplásicos/farmacología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Embrión no Mamífero/citología , Embrión no Mamífero/efectos de los fármacos , Xenopus/embriología , Animales , Antineoplásicos/toxicidad , Benzodioxoles/análisis , Benzodioxoles/farmacología , Benzodioxoles/toxicidad , Benzofuranos/análisis , Benzofuranos/farmacología , Benzofuranos/toxicidad , Carbolinas/análisis , Carbolinas/farmacología , Carbolinas/toxicidad , Línea Celular Tumoral , Pérdida del Embrión , Femenino , Gastrulación/efectos de los fármacos , Glioma/patología , Alcaloides Indólicos/análisis , Alcaloides Indólicos/farmacología , Alcaloides Indólicos/toxicidad , Melanoma Experimental/patología , Ratones , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Invasividad Neoplásica/prevención & control , Paclitaxel/farmacología , Podofilotoxina/análogos & derivados , Ratas , Receptores CXCR4/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Development ; 142(19): 3416-28, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26443638

RESUMEN

V1 interneurons are inhibitory neurons that play an essential role in vertebrate locomotion. The molecular mechanisms underlying their genesis remain, however, largely undefined. Here, we show that the transcription factor Prdm12 is selectively expressed in p1 progenitors of the hindbrain and spinal cord in the frog embryo, and that a similar restricted expression profile is observed in the nerve cord of other vertebrates as well as of the cephalochordate amphioxus. Using frog, chick and mice, we analyzed the regulation of Prdm12 and found that its expression in the caudal neural tube is dependent on retinoic acid and Pax6, and that it is restricted to p1 progenitors, due to the repressive action of Dbx1 and Nkx6-1/2 expressed in the adjacent p0 and p2 domains. Functional studies in the frog, including genome-wide identification of its targets by RNA-seq and ChIP-Seq, reveal that vertebrate Prdm12 proteins act as a general determinant of V1 cell fate, at least in part, by directly repressing Dbx1 and Nkx6 genes. This probably occurs by recruiting the methyltransferase G9a, an activity that is not displayed by the amphioxus Prdm12 protein. Together, these findings indicate that Prdm12 promotes V1 interneurons through cross-repressive interactions with Dbx1 and Nkx6 genes, and suggest that this function might have only been acquired after the split of the vertebrate and cephalochordate lineages.


Asunto(s)
Proteínas Portadoras/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Morfogénesis/fisiología , Proteínas del Tejido Nervioso/metabolismo , Células de Renshaw/fisiología , Xenopus/embriología , Animales , Secuencia de Bases , Embrión de Pollo , Inmunoprecipitación de Cromatina , Biología Computacional , Cartilla de ADN/genética , ADN Complementario/genética , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas de Homeodominio/metabolismo , Inmunohistoquímica , Hibridación in Situ , Ratones , Datos de Secuencia Molecular , Rombencéfalo/metabolismo , Análisis de Secuencia de ARN , Especificidad de la Especie , Médula Espinal/metabolismo
5.
Adv Drug Deliv Rev ; 69-70: 225-46, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24576445

RESUMEN

Many rare human inherited diseases remain untreatable despite the fact that the disease causing genes are known and adequate mouse disease models have been developed. In vivo phenotypic drug screening relies on isolating drug candidates by their ability to produce a desired therapeutic phenotype in whole organisms. Embryos of zebrafish and Xenopus frogs are abundant, small and free-living. They can be easily arrayed in multi-well dishes and treated with small organic molecules. With the development of novel genome modification tools, such a zinc-finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and CRISPR/Cas, it is now possible to efficiently engineer non-mammalian models of inherited human diseases. Here, we will review the rapid progress made in adapting these novel genome editing tools to Xenopus. The advantages of Xenopus embryos as in vivo models to study human inherited diseases will be presented and their utility for drug discovery screening will be discussed. Being a tetrapod, Xenopus complements zebrafish as an indispensable non-mammalian animal model for the study of human disease pathologies and the discovery of novel therapeutics for inherited diseases.


Asunto(s)
Descubrimiento de Drogas/métodos , Ingeniería Genética/métodos , Fenotipo , Xenopus/embriología , Animales , Descubrimiento de Drogas/tendencias , Evaluación Preclínica de Medicamentos/métodos , Evaluación Preclínica de Medicamentos/tendencias , Ingeniería Genética/tendencias , Humanos , Estadios del Ciclo de Vida/fisiología
6.
Phytochemistry ; 96: 318-29, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24011802

RESUMEN

An ethyl acetate extract of Curcuma kwangsiensis S.G. Lee & C.F. Liang (Zingiberaceae) rhizomes (100 µg/ml) enhanced the GABA-induced chloride current (IGABA) through GABAA receptors of the α1ß2γ2S subtype by 79.0±7.0%. Potentiation of IGABA was measured using the two-microelectrode voltage-clamp technique and Xenopus laevis oocytes. HPLC-based activity profiling of the crude extract led to the identification of 11 structurally related labdane diterpenoids, including four new compounds. Structure elucidation was achieved by comprehensive analysis of on-line (LC-PDA-ESI-TOF-MS) and off-line (microprobe 1D and 2D NMR) spectroscopic data. The absolute configuration of the compounds was established by comparison of experimental and calculated ECD spectra. Labdane diterpenes represent a new class of plant secondary metabolites eliciting positive GABAA receptor modulation. The highest efficiency was observed for zerumin A (maximum potentiation of IGABA by 309.4±35.6%, and EC50 of 24.9±8.8 µM).


Asunto(s)
Curcuma/química , Diterpenos/aislamiento & purificación , Diterpenos/farmacología , Medicamentos Herbarios Chinos/aislamiento & purificación , Medicamentos Herbarios Chinos/farmacología , Receptores de GABA-A/efectos de los fármacos , Animales , Cromatografía Líquida de Alta Presión , Diterpenos/química , Medicamentos Herbarios Chinos/química , Resonancia Magnética Nuclear Biomolecular , Oocitos/metabolismo , Rizoma/química , Xenopus/embriología
7.
Dev Biol ; 373(1): 39-52, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23064029

RESUMEN

The Dmrt (doublesex and mab-3 related transcription factor) genes encode a large family of evolutionarily conserved transcription factors whose function in sex specific differentiation has been well studied in all animal lineages. In vertebrates, their function is not restricted to the developing gonads. For example, Xenopus Dmrt4 is essential for neurogenesis in the olfactory system. Here we have isolated and characterized Xenopus Dmrt5 and found that it is coexpressed with Dmrt4 in the developing olfactory placodes. As Dmrt4, Dmrt5 is positively regulated in the ectoderm by neural inducers and negatively by proneural factors. Both Dmrt5 and Dmrt4 genes are also activated by the combined action of the transcription factor Otx2, broadly transcribed in the head ectoderm and of Notch signaling, activated in the anterior neural ridge. As for Dmrt4, knockdown of Dmrt5 impairs neurogenesis in the embryonic olfactory system and in neuralized animal caps. Conversely, its overexpression promotes neuronal differentiation in animal caps, a property that requires the conserved C-terminal DMA and DMB domains. We also found that the sea anenome Dmrt4/5 related gene NvDmrtb also induces neurogenesis in Xenopus animal caps and that conversely, its knockdown in Nematostella reduces elav-1 positive neurons. Together, our data identify Dmrt5 as a novel important regulator of neurogenesis whose function overlaps with that of Dmrt4 during Xenopus olfactory system development. They also suggest that Dmrt may have had a role in neurogenesis in the last common ancestor of cnidarians and bilaterians.


Asunto(s)
Neurogénesis/fisiología , Mucosa Olfatoria/embriología , Factores de Transcripción/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus/embriología , Animales , Células COS , Chlorocebus aethiops , Cartilla de ADN/genética , ADN Complementario/genética , Ensayo de Cambio de Movilidad Electroforética , Técnicas de Silenciamiento del Gen , Etiquetado Corte-Fin in Situ , Factores de Transcripción Otx/metabolismo , Plásmidos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Anémonas de Mar/genética , Especificidad de la Especie , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Xenopus/genética , Proteínas de Xenopus/genética , Proteínas de Xenopus/fisiología
8.
Exp Hematol ; 39(7): 784-94, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21627978

RESUMEN

OBJECTIVE: Most patients with erythropoietic protoporphyria have deficient ferrochelatase (FECH) activity due to changes in FECH DNA. We evaluated seven patients with erythropoietic protoporphyria phenotype in whom abnormalities of FECH DNA were not found by conventional analysis. The major focus was mitoferrin-1 (MFRN1), the mitochondrial transporter of Fe used for heme formation by FECH and for 2Fe2S cluster synthesis, which is critical to FECH activity/stability. MATERIALS AND METHODS: Four patients had a deletion in ALAS2 that causes enzyme gain-of-function, resulting in increased formation of protoporphyrin; one had a heterozygous major deletion in FECH DNA. All had an abnormal transcript of MFRN1 in messenger RNA extracted from blood leukocytes and/or liver tissue. The abnormal transcript contained an insert of intron 2 that had a stop codon. The consequences of abnormal MFRN1 expression were examined using zebrafish and yeast MFRN-deficient strains and cultured lymphoblasts from the patients. RESULTS: Abnormal human MFRN1 complementary DNA showed loss-of-function in zebrafish and yeast mutants, whereas normal human MFRN1 complementary DNA rescued both. Using cultured lymphoblasts, quantitative reverse transcription polymerase chain reaction showed increased formation of abnormal transcript that was accompanied by decreased formation of normal transcript and reduced FECH activity in patients compared to normal lines. A positive correlation coefficient (0.75) was found between FECH activity and normal MFRN1 messenger RNA in lymphoblasts. However, no obvious cause for increased formation of abnormal transcript was identified in MFRN1 exons and splice junctions. CONCLUSIONS: Abnormal MFRN1 expression can contribute to erythropoietic protoporphyria phenotype in some patients, probably by causing a reduction in FECH activity.


Asunto(s)
Proteínas de Transporte de Catión/genética , Ferroquelatasa/genética , Expresión Génica , Proteínas Mitocondriales/genética , Protoporfiria Eritropoyética/genética , 5-Aminolevulinato Sintetasa/genética , 5-Aminolevulinato Sintetasa/metabolismo , Adolescente , Adulto , Anciano , Animales , Secuencia de Bases , Células COS , Proteínas de Transporte de Catión/metabolismo , Niño , Chlorocebus aethiops , Embrión no Mamífero/embriología , Embrión no Mamífero/metabolismo , Femenino , Ferroquelatasa/metabolismo , Prueba de Complementación Genética , Humanos , Células K562 , Masculino , Persona de Mediana Edad , Proteínas Mitocondriales/metabolismo , Datos de Secuencia Molecular , Mutación , Protoporfiria Eritropoyética/metabolismo , Protoporfiria Eritropoyética/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Xenopus/embriología , Xenopus/genética , Levaduras/genética , Levaduras/crecimiento & desarrollo , Adulto Joven
9.
Dev Biol ; 329(2): 258-68, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19268445

RESUMEN

The Iroquois (Irx) genes encode homeoproteins conserved during evolution. Vertebrate genomes contain six Irx genes organized in two clusters, IrxA (which harbors Irx1, Irx2 and Irx4) and IrxB (which harbors Irx3, Irx5 and Irx6). To determine the precise role of these genes during development and their putative redundancies, we conducted a comparative expression analysis and a comprehensive loss-of-function study of all the early expressed Irx genes (Irx1-5) using specific morpholinos in Xenopus. We found that the five Irx genes display largely overlapping expression patterns and contribute to neural patterning. All Irx genes are required for proper formation of posterior forebrain, midbrain, hindbrain and, to a lesser an extent, spinal cord. Nevertheless, Irx1 and Irx3 seem to have a predominant role during regionalization of the neural plate. In addition, we find that the common anterior limit of Irx gene expression, which will correspond to the future border between the prethalamus and thalamus, is defined by mutual repression between Fezf and Irx proteins. This mutual repression is likely direct. Finally, we show that Arx, another anteriorly expressed repressor, also contribute to delineate the anterior border of Irx expression.


Asunto(s)
Tipificación del Cuerpo , Proteínas de Homeodominio/genética , Proteínas Represoras/genética , Tálamo/embriología , Factores de Transcripción/genética , Proteínas de Xenopus/genética , Xenopus/embriología , Animales , Secuencia de Bases , Cartilla de ADN , Hibridación in Situ , Xenopus/genética
10.
Development ; 134(23): 4297-306, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17993468

RESUMEN

Partitioning-defective 1 (PAR1) and atypical protein kinase C (aPKC) are conserved serine/threonine protein kinases implicated in the establishment of cell polarity in many species from yeast to humans. Here we investigate the roles of these protein kinases in cell fate determination in Xenopus epidermis. Early asymmetric cell divisions at blastula and gastrula stages give rise to the superficial (apical) and the deep (basal) cell layers of epidermal ectoderm. These two layers consist of cells with different intrinsic developmental potential, including superficial epidermal cells and deep ciliated cells. Our gain- and loss-of-function studies demonstrate that aPKC inhibits ciliated cell differentiation in Xenopus ectoderm and promotes superficial cell fates. We find that the crucial molecular substrate for aPKC is PAR1, which is localized in a complementary domain in superficial ectoderm cells. We show that PAR1 acts downstream of aPKC and is sufficient to stimulate ciliated cell differentiation and inhibit superficial epidermal cell fates. Our results suggest that aPKC and PAR1 function sequentially in a conserved molecular pathway that links apical-basal cell polarity to Notch signaling and cell fate determination. The observed patterning mechanism may operate in a wide range of epithelial tissues in many species.


Asunto(s)
Cilios/enzimología , Ectodermo/enzimología , Embrión no Mamífero/fisiología , MicroARNs/genética , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Xenopus/embriología , Animales , Ectodermo/fisiología , Hibridación in Situ , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
Dev Growth Differ ; 48(9): 575-85, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17118012

RESUMEN

Using a subtracted Xenopus cDNA library based on the differential sensitivity of anterior and posterior genes to retinoic acid, we isolated a novel Xenopus nuclear GTP-binding protein (XGB). XGB is expressed prominently in the optic primordia at the tailbud stage. The N-terminal region of XGB contains a set of GTP-binding protein motifs, and the C-terminal region contains two putative nuclear localization signals and two coiled regions. A GFP-XGB fusion protein was expressed in the nucleus of NIH3T3 cells where it bound to subnuclear structures. Truncated C-terminal constructs of XGB containing both nuclear localization signal(s) and coiled region(s) suppressed eye formation, whereas neither the N-terminal construct nor constructs with a mutated GTP-binding protein motif affected eye formation. Expression of Pax6 and Rx1 genes, which are crucial for eye development, was reduced in embryos overexpressing the C-terminal constructs of XGB. Suppression of Pax6 and Rx1 at earlier developmental stages as well as perturbation of eye formation at later stages was counteracted by co-expression of wild-type XGB. We conclude that XGB plays a role in the formation of optic primordia through activation of at least two eye field transcription factors.


Asunto(s)
Ojo/embriología , Proteínas de Unión al GTP/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus/embriología , Secuencia de Aminoácidos , Animales , ADN Complementario , Embrión no Mamífero , Desarrollo Embrionario/genética , Ojo/metabolismo , Proteínas de Unión al GTP/genética , Regulación del Desarrollo de la Expresión Génica , Genes Homeobox , Datos de Secuencia Molecular , Xenopus/genética , Proteínas de Xenopus/genética
12.
Gene ; 367: 135-41, 2006 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-16314051

RESUMEN

We have characterized a cDNA encoding a Xenopus laevis apyrase (XAPY) that is expressed during embryogenesis. XAPY is highly homologous to two recently described mammalian apyrases, human SCAN-1 and rat Ca2+-NDPase, and to a lesser extent the salivary apyrase of the blood-feeding arthropod Cimex lectularis. RT-PCR analysis shows that Xapy is expressed at all the developmental stages tested, from oocytes through to tadpoles. Xapy transcripts are widely distributed in the embryo, but from late neurulae through to late tailbud stages they are highly enriched in the cement gland, an adhesive organ in the epidermis of the head. When expressed in HEK 293 cells, XAPY is largely retained in the endoplasmic reticulum, although some is also secreted. XAPY conditioned media hydrolyses UDP and UTP, confirming that it is a functional apyrase.


Asunto(s)
Apirasa/metabolismo , Regulación del Desarrollo de la Expresión Génica , Nucleotidasas/metabolismo , Xenopus/genética , Secuencia de Aminoácidos , Animales , Apirasa/química , Apirasa/genética , Secuencia de Bases , Chinches/enzimología , Línea Celular , Codón , Codón Iniciador , Secuencia Conservada , Medios de Cultivo Condicionados/farmacología , ADN Complementario/genética , Embrión no Mamífero , Retículo Endoplásmico/enzimología , Retículo Endoplásmico/metabolismo , Fluoresceína , Técnica del Anticuerpo Fluorescente Indirecta , Colorantes Fluorescentes , Humanos , Hibridación in Situ , Metamorfosis Biológica , Microscopía Fluorescente , Datos de Secuencia Molecular , Nucleotidasas/química , Nucleotidasas/genética , Estructura Terciaria de Proteína , ARN Mensajero/análisis , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ARN , Homología de Secuencia de Aminoácido , Distribución Tisular , Transcripción Genética , Uridina Difosfato/metabolismo , Uridina Trifosfato/metabolismo , Xenopus/embriología , Xenopus/metabolismo
13.
Cell ; 115(5): 603-13, 2003 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-14651851

RESUMEN

Gastrulation generates mesoderm and endoderm from embryonic epiblast; soon after, the neural plate is established within the epiblast-both events require FGF signaling. We describe a zinc finger transcriptional activator, Churchill (ChCh), which acts as a switch between different roles of FGF. FGF induces ChCh slowly; this activates Smad-interacting-protein-1 (Sip1), which blocks further induction of the mesoderm markers brachyury and Tbx6L by FGF. ChCh is first expressed as cells stop migrating through the primitive streak, and we show that it regulates cell ingression. We propose a simple mechanism by which FGF sensitizes cells to BMP signals. These results reveal that neural induction requires cessation of mesoderm formation at the midline in addition to the decision between epidermis and neural plate.


Asunto(s)
Proteínas Aviares/aislamiento & purificación , Sistema Nervioso Central/embriología , Proteínas Fetales , Gástrula/metabolismo , Factores de Transcripción/aislamiento & purificación , Activación Transcripcional/genética , Secuencia de Aminoácidos/genética , Animales , Proteínas Aviares/genética , Secuencia de Bases/genética , Diferenciación Celular/genética , Movimiento Celular/genética , Sistema Nervioso Central/citología , Sistema Nervioso Central/metabolismo , Embrión de Pollo , ADN Complementario/análisis , ADN Complementario/genética , Embrión no Mamífero/citología , Embrión no Mamífero/embriología , Embrión no Mamífero/metabolismo , Inducción Embrionaria/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Gástrula/citología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Mesodermo/metabolismo , Datos de Secuencia Molecular , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Proteínas de Dominio T Box/metabolismo , Transactivadores , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Xenopus/embriología , Proteínas de Xenopus , Dedos de Zinc/genética
14.
Dev Growth Differ ; 45(2): 143-52, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12752502

RESUMEN

The processes of development and differentiation of the pancreas, an endoderm-derived vital organ that consists of both endocrine and exocrine cells, are highly conserved across most vertebrates. Recently, an in vitro system has been reported to induce embryonic pancreas using multipotent Xenopus ectodermal cells treated with activin and retinoic acid. In this study, this system was first modified to eliminate the mesoderm-derived pronephros. It was found that pronephros, which appeared with the use of low concentrations of activin, was eliminated at higher concentrations (400 ng/mL), while pancreas developed at a high frequency. Using this modified system, subtractive hybridization screening for novel pancreatic genes was done to better understand the molecular mechanisms of pancreas formation. Four novel genes were identified and characterized that were also found to be specifically expressed in the developing pancreas: carboxyl ester lipase, pancreatic elastase2, placental protein11 and protein disulfide isomerase A2 precursor. This in vitro pancreas-induction system may provide a useful model for analysis of the molecular mechanisms that function during pancreas development.


Asunto(s)
Embrión no Mamífero/fisiología , Regulación del Desarrollo de la Expresión Génica , Páncreas/embriología , Xenopus/embriología , Xenopus/genética , Activinas/farmacología , Secuencia de Aminoácidos , Animales , Células Cultivadas , ADN Complementario/genética , Ectodermo/citología , Embrión no Mamífero/citología , Enzimas/química , Enzimas/genética , Femenino , Humanos , Hibridación in Situ/métodos , Islotes Pancreáticos/embriología , Masculino , Datos de Secuencia Molecular , Morfogénesis , Técnicas de Cultivo de Órganos , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Tretinoina/farmacología
15.
J Nat Prod ; 66(5): 630-3, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12762796

RESUMEN

Four new euphane-type triterpenes, kansenone (1), kansenonol (3), 11-oxo-kansenonol (4), kansenol (5), and a new tirucallane-type triterpene, epi-kansenone (2), were isolated from a 60% EtOH extract of Euphorbia kansui, together with alpha-euphol. Their structures were elucidated on the basis of extensive analysis of their 1D and 2D NMR spectral data. This appears to be the first report of the natural occurrence of euphane/tirucallane-type triterpenes with a ketone at C-7. In vitro treatment of cultured individual Xenopus laevis cells at the blastular stage with 1-4 significantly arrested cleavage of the cells (10 microg/mL of each compound resulted in >50% cleavage arrest).


Asunto(s)
Antineoplásicos Fitogénicos/aislamiento & purificación , Medicamentos Herbarios Chinos/química , Euphorbia/química , Plantas Medicinales/química , Triterpenos/aislamiento & purificación , Animales , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/farmacología , División Celular/efectos de los fármacos , Estructura Molecular , Resonancia Magnética Nuclear Biomolecular , Raíces de Plantas/química , Triterpenos/química , Triterpenos/farmacología , Xenopus/embriología
16.
J Nat Prod ; 65(9): 1246-51, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12350140

RESUMEN

Twelve polycyclic diterpenes have been isolated from the roots of Euphorbia kansui. Nine were assigned with an ingenol skeleton, 20-O-(2'E,4'E-decadienoyl)ingenol (1), 20-O-(2'E,4'Z-decadienoyl)ingenol (2), 3-O-(2'E,4'Z-decadienoyl)ingenol (3), 3-O-(2'E,4'E-decadienoyl)ingenol (4), 3-O-(2'E,4'Z-decadienoyl)-5-O-acetylingenol (5), 3-O-(2'E,4'Z-decadienoyl)-20-O-acetylingenol (6), 3-O-(2'E,4'E-decadienoyl)-20-O-acetylingenol (7), 20-O-(decanoyl)ingenol (8), and 5-O-(2'E,4'E-decadienoyl)ingenol (9), and three with a jatrophane skeleton, kansuinins A (12), B (11), and C (10). Compounds 1, 2, 5, 9, and 12 are new compounds, while 4 and 7 were assigned with new geometric configurations. Their structures were elucidated by spectroscopic and chemical analysis. In vitro treatment of cultured individual Xenopus cells at the blastular stage with 1-9 arrested cleavage significantly (0.5 microg/mL of each compound resulted in >75% cleavage arrest). Of the three jatrophane diterpenes (10-12), only kansuinin B (11) showed any activity, resulting in 87% cleavage arrest at 50 microg/mL.


Asunto(s)
Diterpenos/aislamiento & purificación , Euphorbia/química , Plantas Medicinales/química , Receptores Fc/biosíntesis , Animales , División Celular/efectos de los fármacos , Células Cultivadas/efectos de los fármacos , Cromatografía Líquida de Alta Presión , Diterpenos/química , Diterpenos/farmacología , Técnicas In Vitro , Medicina Tradicional China , Conformación Molecular , Estructura Molecular , Resonancia Magnética Nuclear Biomolecular , Raíces de Plantas/química , Estereoisomerismo , Regulación hacia Arriba , Xenopus/embriología , Xenopus/crecimiento & desarrollo
17.
Biochem Cell Biol ; 79(2): 113-21, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11310558

RESUMEN

We report the isolation and characterization of the Xenopus homolog to human T1 ANT (adenine nucleotide translocase). The 1290-nucleotide sequence contains initiation and termination signals, and encodes a conceptual protein of 298 amino acids. The sequence shares high amino acid identity with the mammalian adenine translocases. The transcript is present in unfertilized eggs, and it is expressed at higher levels during formation of the antero-posterior dorsal axis in embryos. Although low levels are expressed constitutively except in endodermal cells, adenine nucleotide translocase (ANT) expression is dynamically regulated during neurulation. At this stage, expression in ectoderm rapidly diminishes as the neural folds form, and then ANT expression increases slightly in mesoderm. At the culmination of neurulation, the neural tube briefly expresses ANT, and thereafter its expression predominates in the somitic mesoderm and also the chordoneural hinge. In addition, ANT expression is particularly high in the prosencephalon, the mesencephalon, the branchial arches, eye, and the otic vesicle. Treatment of embryos with retinoic acid has the effect of diminishing constitutive expression of ANT, but microinjection studies demonstrate that immediate and local repression cannot be induced in dorsal structures.


Asunto(s)
Ectodermo/enzimología , Regulación del Desarrollo de la Expresión Génica/fisiología , Mesodermo/enzimología , Translocasas Mitocondriales de ADP y ATP/genética , Secuencia de Aminoácidos/genética , Animales , ADN Complementario/aislamiento & purificación , Gástrula/enzimología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Mamíferos/genética , Mamíferos/metabolismo , Microinyecciones/instrumentación , Translocasas Mitocondriales de ADP y ATP/biosíntesis , Translocasas Mitocondriales de ADP y ATP/efectos de los fármacos , Datos de Secuencia Molecular , Prosencéfalo/enzimología , ARN Mensajero/análisis , Homología de Secuencia de Aminoácido , Tretinoina/farmacología , Xenopus/embriología , Xenopus/metabolismo
18.
Mech Dev ; 101(1-2): 91-103, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11231062

RESUMEN

Retinoic acid (RA) metabolizing enzymes play important roles in RA signaling during vertebrate embryogenesis. We have previously reported on a RA degrading enzyme, XCYP26, which appears to be critical for the anteroposterior patterning of the central nervous system (EMBO J. 17 (1998) 7361). Here, we report on the sequence, expression and function of its counterpart, XRALDH2, a RA generating enzyme in Xenopus. During gastrulation and neurulation, XRALDH2 and XCYP26 show non-overlapping, complementary expression domains. Upon misexpression, XRALDH2 is found to reduce the forebrain territory and to posteriorize the molecular identity of midbrain and individual hindbrain rhombomeres in Xenopus embryos. Furthermore, ectopic XRALDH2, in combination with its substrate, all-trans-retinal (ATR), can mimic the RA phenotype to result in microcephalic embryos. Taken together, our data support the notion that XRALDH2 plays an important role in RA homeostasis by the creation of a critical RA concentration gradient along the anteroposterior axis of early embryos, which is essential for proper patterning of the central nervous system in Xenopus.


Asunto(s)
Aldehído Oxidorreductasas/genética , Aldehído Oxidorreductasas/metabolismo , Sistema Nervioso Central/embriología , Xenopus/embriología , Familia de Aldehído Deshidrogenasa 1 , Aldehído Oxidasa , Aldehído Oxidorreductasas/química , Secuencia de Aminoácidos , Animales , Encéfalo/metabolismo , ADN Complementario/metabolismo , Regulación hacia Abajo , Gástrula/metabolismo , Humanos , Hibridación in Situ , Mesencéfalo/embriología , Datos de Secuencia Molecular , Sistema Nervioso/metabolismo , Sistemas de Lectura Abierta , Fenotipo , Estructura Terciaria de Proteína , Retinal-Deshidrogenasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rombencéfalo/embriología , Homología de Secuencia de Aminoácido , Factores de Tiempo , Distribución Tisular , Tretinoina/farmacología , Proteínas de Xenopus
19.
J Biol Chem ; 276(17): 13600-5, 2001 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-11278406

RESUMEN

KCNQ1 inactivation bears electrophysiological characteristics different from classical N- and C-type inactivation in Shaker-like potassium channels. However, the molecular site of KCNQ1 inactivation has not yet been determined. KCNQ2 channels do not exert a fast inactivation in contrast to KCNQ1 channels. By expressing functional chimeras between KCNQ1 and KCNQ2 in Xenopus oocytes, we mapped the region of this inactivation to transmembrane domain S5 and the pore loop H5 and finally narrowed down the site to positions Gly(272) and Val(307) in KCNQ1. Exchanging these two amino acids individually with the analogous KCNQ2 residue abolished inactivation. Furthermore, a KCNQ1-like inactivation was introduced into KCNQ2 by mutagenesis in the corresponding region, confirming its relevance for the inactivation process. As KCNQ1 inactivation involves the regions S5 and H5, it exhibits a geography distinct from N- or C-type inactivation. Native cardiac I(Ks) channels comprising KCNQ1 and accessory MinK subunits do not inactivate because of the functional interaction of KCNQ1 with MinK. Mutations in KCNQ1 can lead to long QT1 syndrome, an inherited form of arrhythmia. The long QT1 mutant KCNQ1(L273F) displays a pronounced KCNQ1 inactivation. Here we show that when expressing mutant I(Ks) channels formed from KCNQ1(L273F) and MinK, MinK association no longer eliminates KCNQ1 inactivation. This results in smaller repolarizing currents in the heart and therefore represents a novel mechanism leading to long QT syndrome.


Asunto(s)
Canales de Potasio con Entrada de Voltaje , Canales de Potasio/química , Secuencia de Aminoácidos , Animales , Arritmias Cardíacas/genética , Electrofisiología , Glicina/química , Humanos , Canales de Potasio KCNQ , Canal de Potasio KCNQ1 , Canal de Potasio KCNQ2 , Síndrome de QT Prolongado/genética , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación , Oocitos/metabolismo , Técnicas de Placa-Clamp , Mutación Puntual , Canales de Potasio/genética , Canales de Potasio/metabolismo , Estructura Terciaria de Proteína , ARN Complementario/metabolismo , Homología de Secuencia de Aminoácido , Factores de Tiempo , Valina/química , Xenopus/embriología
20.
Mol Cell Biol ; 21(4): 1360-9, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11158321

RESUMEN

Polycomb group (PcG) proteins form multimeric protein complexes which are involved in the heritable stable repression of genes. Previously, we identified two distinct human PcG protein complexes. The EED-EZH protein complex contains the EED and EZH2 PcG proteins, and the HPC-HPH PcG complex contains the HPC, HPH, BMI1, and RING1 PcG proteins. Here we show that YY1, a homolog of the Drosophila PcG protein pleiohomeotic (Pho), interacts specificially with the human PcG protein EED but not with proteins of the HPC-HPH PcG complex. Since YY1 and Pho are DNA-binding proteins, the interaction between YY1 and EED provides a direct link between the chromatin-associated EED-EZH PcG complex and the DNA of target genes. To study the functional significance of the interaction, we expressed the Xenopus homologs of EED and YY1 in Xenopus embryos. Both Xeed and XYY1 induce an ectopic neural axis but do not induce mesodermal tissues. In contrast, members of the HPC-HPH PcG complex do not induce neural tissue. The exclusive, direct neuralizing activity of both the Xeed and XYY1 proteins underlines the significance of the interaction between the two proteins. Our data also indicate a role for chromatin-associated proteins, such as PcG proteins, in Xenopus neural induction.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Tejido Nervioso/embriología , Proteínas Represoras/fisiología , Factores de Transcripción/fisiología , Proteínas de Xenopus , Xenopus/embriología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Cartilla de ADN/genética , Proteínas de Unión al ADN/genética , Drosophila/genética , Proteínas de Drosophila , Factores de Unión al ADN Específico de las Células Eritroides , Humanos , Datos de Secuencia Molecular , Defectos del Tubo Neural/embriología , Defectos del Tubo Neural/genética , Fenotipo , Complejo Represivo Polycomb 2 , Proteínas del Grupo Polycomb , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Represoras/genética , Homología de Secuencia de Aminoácido , Factores de Transcripción/genética , Técnicas del Sistema de Dos Híbridos , Xenopus/genética , Factor de Transcripción YY1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA