Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS Biol ; 22(6): e3002701, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38913712

RESUMEN

In the highly regulative embryo of the sea urchin Paracentrotus lividus, establishment of the dorsal-ventral (D/V) axis critically depends on the zygotic expression of the TGF-ß nodal in the ventral ectoderm. nodal expression is first induced ubiquitously in the 32-cell embryo and becomes progressively restricted to the presumptive ventral ectoderm by the early blastula stage. This early spatial restriction of nodal expression is independent of Lefty, and instead relies on the activity of Panda, a maternally expressed TGF-ß ligand related to Lefty and Inhibins, which is required maternally for D/V axis specification. However, the mechanism by which Panda restricts the early nodal expression has remained enigmatic and it is not known if Panda works like a BMP ligand by opposing Nodal and antagonizing Smad2/3 signaling, or if it works like Lefty by sequestering an essential component of the Nodal signaling pathway. In this study, we report that Panda functions as an antagonist of the TGF-ß type II receptor ACVRII (Activin receptor type II), which is the only type II receptor for Nodal signaling in the sea urchin and is also a type II receptor for BMP ligands. Inhibiting translation of acvrII mRNA disrupted D/V patterning across all 3 germ layers and caused acvrII morphants to develop with a typical Nodal loss-of-function phenotype. In contrast, embryos overexpressing acvrII displayed strong ectopic Smad1/5/8 signaling at blastula stages and developed as dorsalized larvae, a phenotype very similar to that caused by over activation of BMP signaling. Remarkably, embryos co-injected with acvrII mRNA and panda mRNA did not show ectopic Smad1/5/8 signaling and developed with a largely normal dorsal-ventral polarity. Furthermore, using an axis induction assay, we found that Panda blocks the ability of ACVRII to orient the D/V axis when overexpressed locally. Using co-immunoprecipitation, we showed that Panda physically interacts with ACVRII, as well as with the Nodal co-receptor Cripto, and with TBR3 (Betaglycan), which is a non-signaling receptor for Inhibins in mammals. At the molecular level, we have traced back the antagonistic activity of Panda to the presence of a single proline residue, conserved with all the Lefty factors, in the ACVRII binding motif of Panda, instead of a serine as in most of TGF-ß ligands. Conversion of this proline to a serine converted Panda from an antagonist that opposed Nodal signaling and promoted dorsalization to an agonist that promoted Nodal signaling and triggered ventralization when overexpressed. Finally, using phylogenomics, we analyzed the emergence of the agonist and antagonist form of Panda in the course of evolution. Our data are consistent with the idea that the presence of a serine at that position, like in most TGF-ß, was the ancestral condition and that the initial function of Panda was possibly in promoting and not in antagonizing Nodal signaling. These results highlight the existence of key functional and structural elements conserved between Panda and Lefty, allow to draw an intriguing parallel between sea urchin Panda and mammalian Inhibin α and raise the unexpected possibility that the original function of Panda may have been in activation of the Nodal pathway rather than in its inhibition.


Asunto(s)
Receptores de Activinas Tipo II , Tipificación del Cuerpo , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica , Proteína Nodal , Paracentrotus , Factor de Crecimiento Transformador beta , Animales , Factor de Crecimiento Transformador beta/metabolismo , Tipificación del Cuerpo/genética , Paracentrotus/embriología , Paracentrotus/metabolismo , Paracentrotus/genética , Receptores de Activinas Tipo II/metabolismo , Receptores de Activinas Tipo II/genética , Proteína Nodal/metabolismo , Proteína Nodal/genética , Embrión no Mamífero/metabolismo , Ligandos , Transducción de Señal
2.
Nat Commun ; 14(1): 3970, 2023 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-37407549

RESUMEN

During early development of the sea urchin embryo, activation of ERK signalling in mesodermal precursors is not triggered by extracellular RTK ligands but by a cell-autonomous, RAS-independent mechanism that was not understood. We discovered that in these cells, ERK signalling is activated through the transcriptional activation of a gene encoding a protein related to Kinase Suppressor of Ras, that we named KSR3. KSR3 belongs to a family of catalytically inactive allosteric activators of RAF. Phylogenetic analysis revealed that genes encoding kinase defective KSR3 proteins are present in most non-chordate metazoa but have been lost in flies and nematodes. We show that the structure of KSR3 factors resembles that of several oncogenic human RAF mutants and that KSR3 from echinoderms, cnidarians and hemichordates activate ERK signalling independently of RAS when overexpressed in cultured cells. Finally, we used the sequence of KSR3 factors to identify activating mutations of human B-RAF. These findings reveal key functions for this family of factors as activators of RAF in RAS-independent ERK signalling in invertebrates. They have implications on the evolution of the ERK signalling pathway and suggest a mechanism for its co-option in the course of evolution.


Asunto(s)
Sistema de Señalización de MAP Quinasas , Transducción de Señal , Animales , Humanos , Filogenia , Sistema de Señalización de MAP Quinasas/genética , Proteínas Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-raf/metabolismo
3.
Cell Genom ; 3(4): 100295, 2023 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-37082140

RESUMEN

Sea urchins are emblematic models in developmental biology and display several characteristics that set them apart from other deuterostomes. To uncover the genomic cues that may underlie these specificities, we generated a chromosome-scale genome assembly for the sea urchin Paracentrotus lividus and an extensive gene expression and epigenetic profiles of its embryonic development. We found that, unlike vertebrates, sea urchins retained ancestral chromosomal linkages but underwent very fast intrachromosomal gene order mixing. We identified a burst of gene duplication in the echinoid lineage and showed that some of these expanded genes have been recruited in novel structures (water vascular system, Aristotle's lantern, and skeletogenic micromere lineage). Finally, we identified gene-regulatory modules conserved between sea urchins and chordates. Our results suggest that gene-regulatory networks controlling development can be conserved despite extensive gene order rearrangement.

4.
Matrix Biol ; 114: 35-66, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36343860

RESUMEN

All epithelia have their basal side in contact with a specialized extracellular matrix, the basement membrane (BM). During development, the BM contributes to the shaping of epithelial organs via its mechanical properties. These properties rely on two core components of the BM, collagen type IV and perlecan/HSPG2, which both interact with another core component, laminin, the initiator of BM assembly. While collagen type IV supplies the BM with rigidity to constrain the tissue, perlecan antagonizes this effect. Nevertheless, the number of organs that has been studied is still scarce, and given that epithelial tissues exhibit a wide array of shapes, their forms are bound to be regulated by distinct mechanisms. This is underscored by mounting evidence that BM composition and assembly/biogenesis is tissue-specific. Moreover, previous reports have essentially focused on the mechanical role of the BM in morphogenesis at the tissue scale, but not the cell scale. Here, we took advantage of the robust conservation of core BM proteins and the limited genetic redundancy of the Drosophila model system to address how this matrix shapes the wing imaginal disc, a complex organ comprising a squamous, a cuboidal and a columnar epithelium. With the use of a hypomorphic allele, we show that the depletion of Trol (Drosophila perlecan) affects the morphogenesis of the three epithelia, but particularly that of the squamous one. The planar surface of the squamous epithelium (SE) becomes extremely narrow, due to a function for Trol in the control of the squamous shape of its cells. Furthermore, we find that the lack of Trol impairs the biogenesis of the BM of the SE by modifying the structure of the collagen type IV lattice. Through atomic force microscopy and laser surgery, we demonstrate that Trol provides elasticity to the SE's BM, thereby regulating the mechanical properties of the SE. Moreover, we show that Trol acts via collagen type IV, since the global reduction in the trol mutant context of collagen type IV or the enzyme that cross-links its 7S -but not the enzyme that cross-links its NC1- domain substantially restores the morphogenesis of the SE. In addition, a stronger decrease in collagen type IV achieved by the overexpression of the matrix metalloprotease 2 exclusively in the BM of the SE, significantly rescues the organization of the two other epithelia. Our data thus sustain a model in which Trol counters the rigidity conveyed by collagen type IV to the BM of the SE, via the regulation of the NC1-dependant assembly of its scaffold, allowing the spreading of the squamous cells, spreading which is compulsory for the architecture of the whole organ.


Asunto(s)
Carcinoma de Células Escamosas , Colágeno Tipo IV , Animales , Colágeno Tipo IV/genética , Colágeno Tipo IV/química , Membrana Basal/metabolismo , Matriz Extracelular/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Morfogénesis , Laminina/genética , Laminina/metabolismo , Drosophila/metabolismo , Células Epiteliales/metabolismo , Carcinoma de Células Escamosas/metabolismo
5.
Dev Dyn ; 250(4): 542-561, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33269518

RESUMEN

BACKGROUND: The basement membrane (BM) provides mechanical shaping of tissues during morphogenesis. The Drosophila BM proteoglycan Perlecan is vital for this process in the wing imaginal disc. This function is thought to be fostered by the heparan sulfate chains attached to the domain I of vertebrate Perlecan. However, this domain is not present in Drosophila, and the source of Perlecan for the wing imaginal disc BM remains unclear. Here, we tackle these two issues. RESULTS: In silico analysis shows that Drosophila Perlecan holds a domain I. Moreover, by combining in situ hybridization of Perlecan mRNA and protein staining, together with tissue-specific Perlecan depletion, we find that there is an autonomous and a non-autonomous source for Perlecan deposition in the wing imaginal disc BM. We further show that both sources cooperate for correct distribution of Perlecan in the wing imaginal disc and morphogenesis of this tissue. CONCLUSIONS: These results show that Perlecan is fully conserved in Drosophila, providing a valuable in vivo model system to study its role in BM function. The existence of two different sources for Perlecan incorporation in the wing imaginal disc BM raises the possibility that inter-organ communication mediated at the level of the BM is involved in organogenesis.


Asunto(s)
Membrana Basal/metabolismo , Drosophila/crecimiento & desarrollo , Proteoglicanos de Heparán Sulfato/metabolismo , Discos Imaginales/metabolismo , Alas de Animales/crecimiento & desarrollo , Secuencia de Aminoácidos , Animales , Secuencia de Consenso , Drosophila/genética , Drosophila/metabolismo , Proteoglicanos de Heparán Sulfato/genética , Alas de Animales/metabolismo
6.
PLoS Genet ; 14(9): e1007621, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30222786

RESUMEN

In the sea urchin embryo, specification of the dorsal-ventral axis critically relies on the spatially restricted expression of nodal in the presumptive ventral ectoderm. The ventral restriction of nodal expression requires the activity of the maternal TGF-ß ligand Panda but the mechanism by which Panda restricts nodal expression is unknown. Similarly, what initiates expression of nodal in the ectoderm and what are the mechanisms that link patterning along the primary and secondary axes is not well understood. We report that in Paracentrotus lividus, the activity of the maternally expressed ETS-domain transcription factor Yan/Tel is essential for the spatial restriction of nodal. Inhibiting translation of maternal yan/tel mRNA disrupted dorsal-ventral patterning in all germ layers by causing a massive ectopic expression of nodal starting from cleavage stages, mimicking the phenotype caused by inactivation of the maternal Nodal antagonist Panda. We show that like in the fly or in vertebrates, the activity of sea urchin Yan/Tel is regulated by phosphorylation by MAP kinases. However, unlike in the fly or in vertebrates, phosphorylation by GSK3 plays a central role in the regulation Yan/Tel stability in the sea urchin. We show that GSK3 phosphorylates Yan/Tel in vitro at two different sites including a ß-TRCP ubiquitin ligase degradation motif and a C-terminal Ser/Thr rich cluster and that phosphorylation of Yan/Tel by GSK3 triggers its degradation by a ß-TRCP/proteasome pathway. Finally, we show that, Yan is epistatic to Panda and that the activity of Yan/Tel is required downstream of Panda to restrict nodal expression. Our results identify Yan/Tel as a central regulator of the spatial expression of nodal in Paracentrotus lividus and uncover a key interaction between the gene regulatory networks responsible for patterning the embryo along the dorsal-ventral and animal-vegetal axes.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Proteína Nodal/metabolismo , Paracentrotus/crecimiento & desarrollo , Proteínas Represoras/metabolismo , Transducción de Señal/fisiología , Animales , Animales Modificados Genéticamente , Tipificación del Cuerpo/fisiología , Motivo ETS , Embrión no Mamífero , Glucógeno Sintasa Quinasa 3/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Mutagénesis Sitio-Dirigida , Proteína Nodal/genética , Proteolisis , Proteínas con Repetición de beta-Transducina/metabolismo
7.
Development ; 144(12): 2270-2281, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28507001

RESUMEN

Dorsal-ventral axis formation in the sea urchin embryo relies on the asymmetrical expression of the TGFß Nodal. The p38-MAPK pathway has been proposed to be essential for dorsal-ventral axis formation by acting upstream of nodal expression. Here, we report that, in contrast to previous studies that used pharmacological inhibitors of p38, manipulating the activity of p38 by genetic means has no obvious impact on morphogenesis. Instead, we discovered that p38 inhibitors strongly disrupt specification of all germ layers by blocking signalling from the Nodal receptor and by interfering with the ERK pathway. Strikingly, while expression of a mutant p38 that is resistant to SB203580 did not rescue dorsal-ventral axis formation or skeletogenesis in embryos treated with this inhibitor, expression of mutant Nodal receptors that are resistant to SB203580 fully restored nodal expression in SB203580-treated embryos. Taken together, these results establish that p38 activity is not required for dorsal-ventral axis formation through nodal expression nor for skeletogenesis. Our results prompt a re-evaluation of the conclusions of several recent studies that linked p38 activity to dorsal-ventral axis formation and to patterning of the skeleton.


Asunto(s)
Paracentrotus/embriología , Paracentrotus/enzimología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Secuencia de Aminoácidos , Animales , Tipificación del Cuerpo/efectos de los fármacos , Tipificación del Cuerpo/genética , Tipificación del Cuerpo/fisiología , Regulación del Desarrollo de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Imidazoles/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Morfogénesis/efectos de los fármacos , Morfogénesis/genética , Morfogénesis/fisiología , Mutación , Ligandos de Señalización Nodal/genética , Ligandos de Señalización Nodal/metabolismo , Paracentrotus/genética , Fenotipo , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Homología de Secuencia de Aminoácido , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/genética
8.
Psychopharmacology (Berl) ; 192(3): 373-83, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17279373

RESUMEN

RATIONALE: Prenatal methylazoxymethanol (MAM) administration at gestational day 17 has been shown to induce in adult rats schizophrenia-like behaviours as well as morphological and/or functional abnormalities in structures such as the hippocampus, medial prefrontal cortex (mPFC) and nucleus accumbens (NAcc), consistent with human data. OBJECTIVES: The aim of the present study was to further characterize the neurochemical alterations associated with this neurodevelopmental animal model of schizophrenia. MATERIALS AND METHODS: We performed simultaneous measurements of locomotor activity and extracellular concentrations of glutamate, dopamine and noradrenaline in the mPFC and the NAcc of adult rats prenatally exposed to MAM or saline after acute systemic injection of a noncompetitive NMDA antagonist, MK-801 (0.1 mg/kg s.c.). RESULTS: A significant attenuation of the MK-801-induced increase in glutamate levels associated with a potentiation of the increase in noradrenaline concentrations was found in the mPFC of MAM-exposed rats, whereas no significant change was observed in the NAcc. MAM-exposed rats also exhibited an exaggerated locomotor hyperactivity, in line with the exacerbation of symptoms reported in schizophrenic patients after administration of noncompetitive NMDA antagonists. CONCLUSIONS: Given the importance of the mPFC in regulating the hyperlocomotor effect of NMDA antagonists, our results suggest that the prefrontal neurochemical alterations induced by MK-801 may sustain the exaggerated locomotor response in MAM-exposed rats.


Asunto(s)
Maleato de Dizocilpina/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Corteza Prefrontal/efectos de los fármacos , Esquizofrenia/fisiopatología , Animales , Conducta Animal/efectos de los fármacos , Modelos Animales de Enfermedad , Dopamina/metabolismo , Femenino , Ácido Glutámico/efectos de los fármacos , Ácido Glutámico/metabolismo , Masculino , Acetato de Metilazoximetanol/análogos & derivados , Acetato de Metilazoximetanol/farmacología , Microdiálisis , Actividad Motora/efectos de los fármacos , Norepinefrina/metabolismo , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Corteza Prefrontal/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores
9.
Synapse ; 60(4): 280-7, 2006 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-16786530

RESUMEN

A previous study has demonstrated that disruption of fear extinction-induced long-term potentiation (LTP) in the medial prefrontal cortex (mPFC) is associated with the return of fear responding. Given that immediate posttraining infusion of PD098059, an inhibitor of extracellular signal-regulated kinase (ERK) mitogen-activated protein kinase (MAPK) cascade, into the mPFC also promotes recovery of fear, we investigated whether impairment of mPFC ERK/MAPK cascade also interferes with development of extinction-related LTP in the mPFC in rats. In Experiment 1, extinction training consisting of repetitive presentations of a tone previously associated with eyelid-shock application induced LTP-like changes at hippocampal inputs to the mPFC that were evident for approximately 2 h following fear extinction. Infusion of PD098059 into the mPFC immediately after extinction training abolished training-related prefrontal LTP and impaired retention of extinction memory tested on the following day. In Experiment 2, immunoblotting assays revealed that posttraining infusion of PD098059 into the mPFC produced a significant reduction of mPFC ERK2. These data, along with previous findings, suggest that low levels of ERK2 phosphorylation in the mPFC may interfere with mechanisms of retention of extinction training. The involvement of mPFC LTP in fear extinction is discussed.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Extinción Psicológica/efectos de los fármacos , Flavonoides/farmacología , MAP Quinasa Quinasa 2/antagonistas & inhibidores , MAP Quinasa Quinasa 2/metabolismo , Plasticidad Neuronal/efectos de los fármacos , Corteza Prefrontal/fisiología , Sinapsis/efectos de los fármacos , Animales , Electrodos Implantados , Inhibidores Enzimáticos/administración & dosificación , Flavonoides/administración & dosificación , Inyecciones , Masculino , Memoria/efectos de los fármacos , Fosforilación , Ratas , Ratas Wistar
10.
Neuroreport ; 17(3): 319-22, 2006 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-16462605

RESUMEN

Stress has been reported to disrupt the induction of synaptic plasticity in different fimbria target structures. The aim of the present study was to investigate whether chronic mild stress may also affect synaptic plasticity in the medial prefrontal cortex, a fimbria target structure. Fimbria tetanus (100 Hz) did not produce any changes in medial prefrontal cortex synaptic efficacy in non-stressed rats. Rats exposed to chronic mild stress, however, developed significant long-term potentiation. Treatment with fluoxetine (10 mg/kg, intraperitoneal) suppressed long-term potentiation induction in the chronic mild stress group. These data demonstrate that stress not only inhibits long-term potentiation development, as often demonstrated, but can also facilitate long-term potentiation development in certain brain circuits.


Asunto(s)
Fluoxetina/farmacología , Potenciación a Largo Plazo/efectos de los fármacos , Neuronas/efectos de los fármacos , Corteza Prefrontal/patología , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Estrés Fisiológico/fisiopatología , Animales , Relación Dosis-Respuesta en la Radiación , Estimulación Eléctrica/métodos , Fórnix/efectos de los fármacos , Fórnix/fisiopatología , Fórnix/efectos de la radiación , Masculino , Ratas , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología , Transmisión Sináptica/efectos de la radiación
11.
Synapse ; 57(1): 61-5, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15858833

RESUMEN

Excitatory glutamatergic fibers from limbic structures, such as the hippocampus and the basolateral amygdala, are known to converge on the same neurons in the nucleus accumbens. We have recently shown that ketamine, at a dose (25 mg/kg) that produces psychosis-like behaviors in rats, decreases glutamatergic transmission between the hippocampus and the nucleus accumbens. Here we investigated whether ketamine also affects glutamatergic transmission between the basolateral amygdala and the nucleus accumbens. We also studied the effects of amphetamine (1.5 mg/kg), known to evoke psychosis-like behaviors in rats. We found that each drug produced a long-lasting (at least 30 min) potentiation of synaptic efficacy in the projection from the basolateral amygdala to the nucleus accumbens. However, while this synaptic potentiation developed shortly after ketamine injection (within 4 min), it occurred after a 30-min delay in rats injected with amphetamine. These data reveal, in freely behaving rats, that ketamine has a more rapid and powerful effect on projection targets of the basolateral amygdala than does amphetamine.


Asunto(s)
Anfetamina/farmacología , Amígdala del Cerebelo/fisiología , Estimulantes del Sistema Nervioso Central/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Ketamina/farmacología , Núcleo Accumbens/fisiología , Transmisión Sináptica/efectos de los fármacos , Animales , Electrodos Implantados , Electrofisiología , Cinética , Masculino , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/fisiología , Ratas , Ratas Wistar , Estimulación Química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...