Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Comp Neurol ; 531(17): 1828-1845, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37814509

RESUMEN

The blood-brain barrier (BBB) is a physical interface between the blood and the brain parenchyma, playing key roles in brain homeostasis. In mammals, the BBB is established thanks to tight junctions between cerebral endothelial cells, involving claudin, occludin, and zonula occludens proteins. Estrogens have been documented to modulate BBB permeability. Interestingly, in the brain of zebrafish, the estrogen-synthesizing activity is strong due to the high expression of Aromatase B protein, encoded by the cyp19a1b gene, in radial glial cells (neural stem cells). Given the roles of estrogens in BBB function, we investigated their impact on the expression of genes involved in BBB tight junctions. We treated zebrafish embryos and adult males with 17ß-estradiol and observed an increased cerebral expression of tight junction and claudin 5 genes in adult males only. In females, treatment with the nuclear estrogen receptor antagonist (ICI182,780 ) had no impact. Interestingly, telencephalic injuries performed in males decreased tight junction gene expression that was partially reversed with 17ß-estradiol. This was further confirmed by extravasation experiments of Evans blue showing that estrogenic treatment limits BBB leakage. We also highlighted the intimate links between endothelial cells and neural stem cells, suggesting that cholesterol and peripheral steroids could be taken up by endothelial cells and used as precursors for estrogen synthesis by neural stem cells. Together, our results show that zebrafish provides an alternative model to further investigate the role of steroids on the expression of genes involved in BBB integrity, both in constitutive and regenerative physiological conditions. The link we described between capillaries endothelial cells and steroidogenic neural cells encourages the use of this model in understanding the mechanisms by which peripheral steroids get into neural tissue and modulate neurogenic activity.


Asunto(s)
Barrera Hematoencefálica , Pez Cebra , Animales , Femenino , Masculino , Barrera Hematoencefálica/metabolismo , Claudina-5/genética , Claudina-5/metabolismo , Células Endoteliales/metabolismo , Estradiol/farmacología , Estrógenos/farmacología , Estrógenos/metabolismo , Expresión Génica , Mamíferos , Proteínas de Uniones Estrechas/genética , Proteínas de Uniones Estrechas/metabolismo , Pez Cebra/metabolismo , Proteína de la Zonula Occludens-1/metabolismo
2.
Environ Sci Pollut Res Int ; 29(26): 39578-39592, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35106724

RESUMEN

The present study was conducted to provide new insights into the mechanisms that may be responsible for cadmium (Cd)-induced toxicity in zebrafish larvae as well as the role of the trace element zinc (Zn) in reversing Cd harmful effects. For this purpose, zebrafish eggs were exposed to Cd or/and Zn for 96 h. The effects on morphological aspect; mortality rate; Cd, Zn, and metallothionein (MT) levels; oxidative stress biomarkers; as well as molecular expression of some genes involved in Zn metabolism (Zn-MT, ZIP10, and ZnT1) and in antioxidant defense system (Cu/Zn-SOD, CAT and GPx) were examined. Our results showed that Cd toxicity was exerted, initially, by an interference with Zn metabolism. Thus, Cd was able to modify the expression of the corresponding genes so as to ensure its intracellular accumulation at the expense of Zn, causing its depletion. An oxidative stress was then generated, representing the second mode of Cd action which resulted in developmental anomalies and subsequently mortality. Interestingly, significant corrections have been noted following Zn supplementation based, essentially, on its ability to interact with the toxic metal. The increases of Zn bioavailability, the improvement of the oxidative status, as well as changes in Zn transporter expression profile are part of the protection mechanisms. The decrease of Cd-induced MTs after Zn supplement, both at the protein and the mRNA level, suggests that the protection provided by Zn is ensured through mechanisms not involving MT expression but which rather depend on the oxidative status.


Asunto(s)
Cadmio , Pez Cebra , Animales , Cadmio/metabolismo , Homeostasis , Metalotioneína/genética , Metalotioneína/metabolismo , Estrés Oxidativo , Pez Cebra/metabolismo , Zinc/metabolismo
3.
Gen Comp Endocrinol ; 288: 113345, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31812531

RESUMEN

Natural and synthetic estrogens and progestins are widely used in human and veterinary medicine and are detected in waste and surface waters. Our previous studies have clearly shown that a number of these substances targets the brain to induce the estrogen-regulated brain aromatase expression but the consequences on brain development remain virtually unexplored. The aim of the present study was therefore to investigate the effect of estradiol (E2), progesterone (P4) and norethindrone (NOR), a 19-nortestosterone progestin, on zebrafish larval neurogenesis. We first demonstrated using real-time quantitative PCR that nuclear estrogen and progesterone receptor brain expression is impacted by E2, P4 and NOR. We brought evidence that brain proliferative and apoptotic activities were differentially affected depending on the steroidal hormone studied, the concentration of steroids and the region investigated. Our findings demonstrate for the first time that steroid compounds released in aquatic environment have the capacity to disrupt key cellular events involved in brain development in zebrafish embryos further questioning the short- and long-term consequences of this disruption on the physiology and behavior of organisms.


Asunto(s)
Congéneres del Estradiol/farmacología , Estrógenos/farmacología , Sistema Nervioso/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Congéneres de la Progesterona/farmacología , Progesterona/farmacología , Pez Cebra/embriología , Animales , Embrión no Mamífero , Desarrollo Embrionario/efectos de los fármacos , Disruptores Endocrinos/farmacología , Estradiol/farmacología , Estrógenos/análogos & derivados , Estrógenos/síntesis química , Humanos , Ligandos , Nandrolona/farmacología , Sistema Nervioso/embriología , Células Neuroendocrinas/efectos de los fármacos , Células Neuroendocrinas/fisiología , Noretindrona/farmacología , Progesterona/análogos & derivados , Progesterona/síntesis química , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/agonistas , Receptores de Progesterona/metabolismo , Pez Cebra/crecimiento & desarrollo
4.
J Neuroendocrinol ; 31(9): e12731, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31066122

RESUMEN

Glyphosate is found in a large array of non-selective herbicides such as Roundup® (Monsanto, Creve Coeur, MO, USA) and is by far the most widely used herbicide. Recent work in rodent models suggests that glyphosate-based herbicides during development can affect neuronal communication and result in altered behaviours, albeit through undefined mechanisms of action. To our knowledge, no study has investigated the effects glyphosate or its formulation in herbicide on maternal behaviour and physiology. In the present study, relatively low doses of glyphosate (5 mg kg-1  d-1 ), Roundup® (5 mg kg-1  d-1 glyphosate equivalent), or vehicle were administered by ingestion to Sprague-Dawley rats from gestational day (GD) 10 to postpartum day (PD)22. The treatments significantly altered licking behaviour toward pups between PD2 and PD6. We also show in the dams at PD22 that Roundup exposure affected the maturation of doublecortin-immunoreactive new neurones in the dorsal dentate gyrus of the hippocampus of the mother. In addition, the expression of synaptophysin was up-regulated by glyphosate in the dorsal and ventral dentate gyrus and CA3 regions of the hippocampus, and down-regulated in the cingulate gyrus. Although a direct effect of glyphosate alone or its formulation on the central nervous system is currently not clear, we show that gut microbiota is significantly altered by the exposure to the pesticides, with significant alteration of the phyla Bacteroidetes and Firmicutes. This is the first study to provide evidence that glyphosate alone or in formulation (Roundup) differentially affects maternal behaviour and modulates neuroplasticity and gut microbiota in the mother.


Asunto(s)
Microbioma Gastrointestinal/efectos de los fármacos , Glicina/análogos & derivados , Herbicidas/toxicidad , Hipocampo/efectos de los fármacos , Conducta Materna/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Periodo Periparto/efectos de los fármacos , Animales , Proliferación Celular/efectos de los fármacos , Proteína Doblecortina , Femenino , Glicina/toxicidad , Hipocampo/fisiología , Conducta Materna/fisiología , Neurogénesis/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/fisiología , Ratas Sprague-Dawley , Glifosato
5.
J Comp Neurol ; 526(4): 569-582, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29124763

RESUMEN

The reparative ability of the central nervous system varies widely in the animal kingdom. In the mammalian brain, the regenerative mechanisms are very limited and newly formed neurons do not survive longer, probably due to a non-suitable local environment. On the opposite, fish can repair the brain after injury, with fast and complete recovery of damaged area. The brain of zebrafish, a teleost fish widely used as vertebrate model, also possesses high regenerative properties after injury. Taking advantage of this relevant model, the aim of the present study was to investigate the role of brain-derived neurotrophic factor (BDNF) in the regenerative ability of adult brain, after stab wound telencephalic injury. BDNF is involved in many brain functions and plays key roles in the repair process after traumatic brain lesions. It has been reported that BDNF strengthens the proliferative activity of neuronal precursor cells, facilitates the neuronal migration toward injured areas, and shows survival properties due to its anti-apoptotic effects. BDNF mRNA levels, assessed by quantitative PCR and in situ hybridization at 1, 4, 7, and 15 days after the lesion, were increased in the damaged telencephalon, mostly suddenly after the lesion. Double staining using in situ hybridization and immunocytochemistry revealed that BDNF mRNA was restricted to cells identified as mature neurons. BDNF mRNA expressing neurons mostly increased in the area around the lesion, showing a peak 1 day after the lesion. Taken together, these results highlight the role of BDNF in brain repair processes and reinforce the value of zebrafish for the study of regenerative neurogenesis.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Telencéfalo/lesiones , Telencéfalo/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Modelos Animales de Enfermedad , Lateralidad Funcional , Masculino , Regeneración Nerviosa/fisiología , Neuronas/metabolismo , Neuronas/patología , ARN Mensajero/metabolismo , Telencéfalo/patología , Heridas Punzantes/metabolismo , Heridas Punzantes/patología , Pez Cebra
6.
J Comp Neurol ; 525(3): 478-497, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-27414756

RESUMEN

The epigenetic mark 5-hydroxymethylcytosine (5hmC) is a cytosine modification that is abundant in the central nervous system of mammals and which results from 5-methylcytosine oxidation by TET enzymes. Such a mark is suggested to play key roles in the regulation of chromatin structure and gene expression. However, its precise functions still remain poorly understood and information about its distribution in non-mammalian species is still lacking. Here, the distribution of 5hmC was investigated in the brain of adult zebrafish, African claw frog, and mouse in a comparative manner. We show that zebrafish neurons are endowed with high levels of 5hmC, whereas quiescent or proliferative neural progenitors show low to undetectable levels of the modified cytosine. In the brain of larval and juvenile Xenopus, 5hmC is also detected in neurons, while ventricular proliferative cells do not display this epigenetic mark. Similarly, 5hmC is enriched in neurons compared to neural progenitors of the ventricular zone in the mouse developing cortex. Interestingly, 5hmC colocalized with the methylated DNA binding protein MeCP2 and with the active chromatin histone modification H3K4me2 in mouse neurons. Taken together, our results show an evolutionarily conserved cerebral distribution of 5hmC between fish and tetrapods and reinforce the idea that 5hmC fulfills major functions in the control of chromatin activity in vertebrate neurons. J. Comp. Neurol. 525:478-497, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
5-Metilcitosina/análogos & derivados , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Neuronas/metabolismo , 5-Metilcitosina/metabolismo , Animales , Animales Modificados Genéticamente , Encéfalo/citología , Dermoscopía , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Inmunohistoquímica , Hibridación in Situ , Masculino , Ratones , Microscopía Confocal , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis/fisiología , Neuroglía/citología , Neuroglía/metabolismo , Neuronas/citología , Mucosa Olfatoria/citología , Mucosa Olfatoria/crecimiento & desarrollo , Mucosa Olfatoria/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Xenopus , Pez Cebra
7.
Biol Aujourdhui ; 210(3): 153-166, 2016.
Artículo en Francés | MEDLINE | ID: mdl-27813476

RESUMEN

In response to various types of vascular stress, the smooth muscle cells of the vessel wall (VSMCs) change phenotype and acquire the capacity to react to abnormal signals. This phenomenon favors the involvement of these cells in the development of major vascular diseases, such as atherosclerosis, and some complications of angioplasty, such as restenosis. The cyclic adenosine monophosphate (cAMP) pathway plays a key role in the integration of stimuli from the immediate environment and in the development of cellular responses. The temporal and spatial subcellular compartmentalization of cAMP ensures that the signals transmitted are specific. This compartmentalization is dependent on the diversity of (1) proteins directly or indirectly regulating the synthesis, degradation or release of cAMP; (2) intracellular effectors of cAMP; (3) isoforms of all these proteins with unique biochemical properties and unique patterns of regulation and (4) the scaffolding proteins on which the macromolecular complexes are built. This review illustrates the ways in which changes in the profile of adenylyl cyclases (ACs) may play critical roles in signal integration, the response of muscle cells and pathological vascular remodeling. It also illustrates the relevance of the renewed consideration of ACs as potentially interesting treatment targets.


Asunto(s)
Adenilil Ciclasas/fisiología , Transdiferenciación Celular , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/fisiología , Remodelación Vascular/fisiología , Animales , AMP Cíclico/metabolismo , AMP Cíclico/fisiología , Humanos , Terapia Molecular Dirigida/tendencias , Transducción de Señal
8.
PLoS One ; 11(6): e0158057, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27336917

RESUMEN

Brain-derived neurotrophic factor (BDNF), a member of the neurotrophin family, has emerged as an active mediator in many essential functions in the central nervous system of mammals. BDNF plays significant roles in neurogenesis, neuronal maturation and/or synaptic plasticity and is involved in cognitive functions such as learning and memory. Despite the vast literature present in mammals, studies devoted to BDNF in the brain of other animal models are scarse. Zebrafish is a teleost fish widely known for developmental genetic studies and is emerging as model for translational neuroscience research. In addition, its brain shows many sites of adult neurogenesis allowing higher regenerative properties after traumatic injuries. To add further knowledge on neurotrophic factors in vertebrate brain models, we decided to determine the distribution of bdnf mRNAs in the larval and adult zebrafish brain and to characterize the phenotype of cells expressing bdnf mRNAs by means of double staining studies. Our results showed that bdnf mRNAs were widely expressed in the brain of 7 days old larvae and throughout the whole brain of mature female and male zebrafish. In adults, bdnf mRNAs were mainly observed in the dorsal telencephalon, preoptic area, dorsal thalamus, posterior tuberculum, hypothalamus, synencephalon, optic tectum and medulla oblongata. By combining immunohistochemistry with in situ hybridization, we showed that bdnf mRNAs were never expressed by radial glial cells or proliferating cells. By contrast, bdnf transcripts were expressed in cells with neuronal phenotype in all brain regions investigated. Our results provide the first demonstration that the brain of zebrafish expresses bdnf mRNAs in neurons and open new fields of research on the role of the BDNF factor in brain mechanisms in normal and brain repairs situations.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/genética , Encéfalo/metabolismo , Expresión Génica , Pez Cebra/genética , Animales , Biomarcadores , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proliferación Celular , Inmunohistoquímica , Hibridación in Situ , Larva , Neuroglía/metabolismo , Neuronas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
9.
J Appl Toxicol ; 36(6): 863-71, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26857037

RESUMEN

The present study was conducted to assess the effects of Cd exposure on estrogen signaling in the zebrafish brain, as well as the potential protective role of Zn against Cd-induced toxicity. For this purpose, the effects on transcriptional activation of the estrogen receptors (ERs), aromatase B (Aro-B) protein expression and molecular expression of related genes were examined in vivo using wild-type and transgenic zebrafish embryos. For in vitro studies, an ER-negative glial cell line (U251MG) transfected with different zebrafish ER subtypes (ERα, ERß1 and ERß2) was also used. Embryos were exposed either to estradiol (E2 ), Cd, E2 +Cd or E2 +Cd+Zn for 72 h and cells were exposed to the same treatments for 30 h. Our results show that E2 treatment promoted the transcriptional activation of ERs and increased Aro-B expression, at both the protein and mRNA levels. Although exposure to Cd, does not affect the studied parameters when administered alone, it significantly abolished the E2 -stimulated transcriptional response of the reporter gene for the three ER subtypes in U251-MG cells, and clearly inhibited the E2 induction of Aro-B in radial glial cells of zebrafish embryos. These inhibitory effects were accompanied by a significant downregulation of the expression of esr1, esr2a, esr2b and cyp19a1b genes compared to the E2 -treated group used as a positive control. Zn administration during simultaneous exposure to E2 and Cd strongly stimulated zebrafish ERs transactivation and increased Aro-B protein expression, whereas mRNA levels of the three ERs as well as the cyp19a1b remained unchanged in comparison with Cd-treated embryos. In conclusion, our results clearly demonstrate that Cd acts as a potent anti-estrogen in vivo and in vitro, and that Cd-induced E2 antagonism can be reversed, at the protein level, by Zn supplement. Copyright © 2016 John Wiley & Sons, Ltd.


Asunto(s)
Encéfalo/efectos de los fármacos , Intoxicación por Cadmio/prevención & control , Cadmio/toxicidad , Embrión no Mamífero/efectos de los fármacos , Contaminantes Químicos del Agua/toxicidad , Pez Cebra , Zinc/uso terapéutico , Animales , Animales Modificados Genéticamente , Aromatasa/genética , Aromatasa/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Cadmio/química , Intoxicación por Cadmio/embriología , Intoxicación por Cadmio/metabolismo , Intoxicación por Cadmio/veterinaria , Línea Celular , Embrión no Mamífero/metabolismo , Embrión no Mamífero/patología , Antagonistas de Estrógenos/química , Antagonistas de Estrógenos/toxicidad , Estrógenos/agonistas , Estrógenos/química , Estrógenos/metabolismo , Enfermedades de los Peces/embriología , Enfermedades de los Peces/metabolismo , Enfermedades de los Peces/patología , Enfermedades de los Peces/prevención & control , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Genes Reporteros/efectos de los fármacos , Humanos , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Neuroglía/patología , Receptores de Estrógenos/antagonistas & inhibidores , Receptores de Estrógenos/química , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal/efectos de los fármacos , Contaminantes Químicos del Agua/antagonistas & inhibidores , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/agonistas , Proteínas de Pez Cebra/antagonistas & inhibidores , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Cigoto/efectos de los fármacos , Cigoto/metabolismo , Cigoto/patología
10.
J Steroid Biochem Mol Biol ; 160: 27-36, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26151741

RESUMEN

Estrogens are known as steroid hormones affecting the brain in many different ways and a wealth of data now document effects on neurogenesis. Estrogens are provided by the periphery but can also be locally produced within the brain itself due to local aromatization of circulating androgens. Adult neurogenesis is described in all vertebrate species examined so far, but comparative investigations have brought to light differences between vertebrate groups. In teleost fishes, the neurogenic activity is spectacular and adult stem cells maintain their mitogenic activity in many proliferative areas within the brain. Fish are also quite unique because brain aromatase expression is limited to radial glia cells, the progenitor cells of adult fish brain. The zebrafish has emerged as an interesting vertebrate model to elucidate the cellular and molecular mechanisms of adult neurogenesis, and notably its modulation by steroids. The main objective of this review is to summarize data related to the functional link between estrogens production in the brain and neurogenesis in fish. First, we will demonstrate that the brain of zebrafish is an endogenous source of steroids and is directly targeted by local and/or peripheral steroids. Then, we will present data demonstrating the progenitor nature of radial glial cells in the brain of adult fish. Next, we will emphasize the role of estrogens in constitutive neurogenesis and its potential contribution to the regenerative neurogenesis. Finally, the negative impacts on neurogenesis of synthetic hormones used in contraceptive pills production and released in the aquatic environment will be discussed.


Asunto(s)
Encéfalo/fisiología , Estrógenos/metabolismo , Neurogénesis , Neurotransmisores/metabolismo , Pez Cebra/fisiología , Animales , Aromatasa/metabolismo , Encéfalo/efectos de los fármacos , Disruptores Endocrinos/efectos adversos , Neurogénesis/efectos de los fármacos , Neuroglía/citología , Neuroglía/efectos de los fármacos , Neuroglía/fisiología , Células Madre/citología , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Proteínas de Pez Cebra/metabolismo
11.
Oncotarget ; 6(41): 43216-29, 2015 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-26646450

RESUMEN

The Notch1 and Notch4 signaling pathways regulate endothelial cell homeostasis. Inflammatory cytokines induce the expression of endothelial adhesion molecules, including VCAM1, partly by downregulating Notch4 signaling. We investigated the role of endothelial Notch1 in this IL-1ß-mediated process. Brief treatment with IL-1ß upregulated endothelial VCAM1 and Notch ligand Jagged1. IL-1ß decreased Notch1 mRNA levels, but levels of the active Notch1ICD protein remained constant. IL-1ß-mediated VCAM1 induction was downregulated in endothelial cells subjected to pretreatment with a pharmacological inhibitor of the γ-secretase, which activates Notch receptors, producing NotchICD. It was also downregulated in cells in which Notch1 and/or Jagged1 were silenced.Conversely, the forced expression of Notch1ICD in naïve endothelial cells upregulated VCAM1 per se and amplified IL-1ß-mediated VCAM1 induction. Jagged1 levels increased and Notch4 signaling was downregulated in parallel. Finally, Notch1ICD and Jagged1 expression was upregulated in the endothelium of the liver in a model of chronic liver inflammation.In conclusion, we describe here a cell-autonomous, pro-inflammatory endothelial Notch1-Jagged1 circuit (i) triggering the expression of VCAM1 even in the absence of inflammatory cytokines and (ii) enhancing the effects of IL-1ß. Thus, IL-1ß regulates Notch1 and Notch4 activity in opposite directions, consistent with a selective targeting of Notch1 in inflamed endothelium.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Células Endoteliales/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Interleucina-1beta/metabolismo , Proteínas de la Membrana/metabolismo , Receptor Notch1/metabolismo , Transducción de Señal/fisiología , Molécula 1 de Adhesión Celular Vascular/biosíntesis , Animales , Western Blotting , Línea Celular , Separación Celular , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica/fisiología , Técnicas de Silenciamiento del Gen , Humanos , Inmunohistoquímica , Inflamación/metabolismo , Proteína Jagged-1 , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , ARN Interferente Pequeño , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Serrate-Jagged , Transfección
12.
PLoS One ; 10(10): e0141043, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26506093

RESUMEN

Ghrelin is a gut-brain peptide hormone, which binds to the growth hormone secretagogue receptor (GHS-R) to regulate a wide variety of biological processes in fish. Despite these prominent physiological roles, no studies have reported the anatomical distribution of preproghrelin transcripts using in situ hybridization in a non-mammalian vertebrate, and its mapping within the different encephalic areas remains unknown. Similarly, no information is available on the possible 24-h variations in the expression of preproghrelin and its receptor in any vertebrate species. The first aim of this study was to investigate the anatomical distribution of ghrelin and GHS-R1a ghrelin receptor subtype in brain and gastrointestinal tract of goldfish (Carassius auratus) using immunohistochemistry and in situ hybridization. Our second aim was to characterize possible daily variations of preproghrelin and ghs-r1 mRNA expression in central and peripheral tissues using real-time reverse transcription-quantitative PCR. Results show ghrelin expression and immunoreactivity in the gastrointestinal tract, with the most abundant signal observed in the mucosal epithelium. These are in agreement with previous findings on mucosal cells as the primary synthesizing site of ghrelin in goldfish. Ghrelin receptor was observed mainly in the hypothalamus with low expression in telencephalon, pineal and cerebellum, and in the same gastrointestinal areas as ghrelin. Daily rhythms in mRNA expression were found for preproghrelin and ghs-r1 in hypothalamus and pituitary with the acrophase occurring at nighttime. Preproghrelin, but not ghs-r1a, displayed a similar daily expression rhythm in the gastrointestinal tract with an amplitude 3-fold higher than the rest of tissues. Together, these results described for the first time in fish the mapping of preproghrelin and ghrelin receptor ghs-r1a in brain and gastrointestinal tract of goldfish, and provide the first evidence for a daily regulation of both genes expression in such locations, suggesting a possible connection between the ghrelinergic and circadian systems in teleosts.


Asunto(s)
Ritmo Circadiano/genética , Ghrelina/biosíntesis , Receptores de Ghrelina/biosíntesis , Receptores de Ghrelina/metabolismo , Animales , Encéfalo/metabolismo , Tracto Gastrointestinal/metabolismo , Regulación de la Expresión Génica , Ghrelina/genética , Ghrelina/metabolismo , Carpa Dorada/genética , Hipófisis/metabolismo , Receptores de Ghrelina/genética
13.
Chronobiol Int ; 32(7): 889-902, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26171989

RESUMEN

The functional organization of the circadian system and the location of the main circadian oscillators vary through phylogeny. Present study investigates by in situ hybridization the anatomical location of the clock gene gPer1b in forebrain and midbrain, pituitary, and in two peripheral locations, the anterior intestine and liver, in a teleost fish, the goldfish (Carassius auratus). Moreover, the daily expression profiles of this gene were also studied by quantitative Real Time-PCR. Goldfish were maintained under a 12L-12D photoperiod and fed daily at 2 h after lights were switched on. A wide distribution of gPer1b mRNA in goldfish brain and pituitary was found in telencephalon, some hypothalamic nuclei (including the homologous to mammalian SCN), habenular nucleus, optic tectum, cerebellum and torus longitudinalis. Moreover, gPer1b expression was observed, for the first time in teleosts, in the pituitary, liver and anterior intestine. Day/night differences in gper1b mRNA abundance were found by in situ hybridization, with higher signal at nighttime that correlates with the results obtained by RT-PCR, where a rhythmic gPer1b expression was found in all tissues with acrophases at the end of the night. Amplitudes of gper1b rhythms vary among tissues, being higher in liver and intestine than in the brain, maybe because different cues entrain clocks in these locations. These results support the existence of functional clocks in many central and peripheral locations in goldfish coordinated, ticking at the same time.


Asunto(s)
Relojes Biológicos , Encéfalo/metabolismo , Ritmo Circadiano , Carpa Dorada/metabolismo , Mucosa Intestinal/metabolismo , Hígado/metabolismo , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Animales , Clonación Molecular , Femenino , Regulación de la Expresión Génica , Carpa Dorada/genética , Hibridación in Situ , Fotoperiodo , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Tiempo
14.
Eur J Neurosci ; 38(9): 3292-301, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23981075

RESUMEN

In non-mammalian vertebrates, serotonin (5-HT)-producing neurons exist in the paraventricular organ (PVO), a diencephalic structure containing cerebrospinal fluid (CSF)-contacting neurons exhibiting 5-HT or dopamine (DA) immunoreactivity. Because the brain of the adult teleost is known for its neurogenic activity supported, for a large part, by radial glial progenitors, this study addresses the origin of newborn 5-HT neurons in the hypothalamus of adult zebrafish. In this species, the PVO exhibits numerous radial glial cells (RGCs) whose somata are located at a certain distance from the ventricle. To study relationships between RGCs and 5-HT CSF-contacting neurons, we performed 5-HT immunohistochemistry in transgenic tg(cyp19a1b-GFP) zebrafish in which RGCs are labelled with GFP under the control of the cyp19a1b promoter. We show that the somata of the 5-HT neurons are located closer to the ventricle than those of RGCs. RGCs extend towards the ventricle cytoplasmic processes that form a continuous barrier along the ventricular surface. In turn, 5-HT neurons contact the CSF via processes that cross this barrier through small pores. Further experiments using proliferating cell nuclear antigen or 5-bromo-2'-deoxyuridine indicate that RGCs proliferate and give birth to 5-HT neurons migrating centripetally instead of centrifugally as in other brain regions. Furthermore, treatment of adult zebrafish with tryptophan hydroxylase inhibitor causes a significant decrease in the number of proliferating cells in the PVO, but not in the mediobasal hypothalamus. These data point to the PVO as an intriguing region in which 5-HT appears to promote genesis of 5-HT neurons that accumulate along the brain ventricles and contact the CSF.


Asunto(s)
Células Ependimogliales/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis , Núcleo Hipotalámico Paraventricular/metabolismo , Neuronas Serotoninérgicas/metabolismo , Serotonina/metabolismo , Animales , Proliferación Celular , Células Ependimogliales/citología , Células-Madre Neurales/citología , Núcleo Hipotalámico Paraventricular/citología , Neuronas Serotoninérgicas/citología , Pez Cebra
15.
PLoS One ; 8(7): e70177, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23894610

RESUMEN

This study, conducted in the brain of a perciform fish, the European sea bass, aimed at raising antibodies against the precursor of the kisspeptins in order to map the kiss systems and to correlate the expression of kisspeptins, kiss1 and kiss2, with that of kisspeptin receptors (kiss-R1 and kiss-R2). Specific antibodies could be raised against the preprokiss2, but not the preoprokiss1. The data indicate that kiss2 neurons are mainly located in the hypothalamus and project widely to the subpallium and pallium, the preoptic region, the thalamus, the pretectal area, the optic tectum, the torus semicircularis, the mediobasal medial and caudal hypothalamus, and the neurohypophysis. These results were compared to the expression of kiss-R1 and kiss-R2 messengers, indicating a very good correlation between the wide distribution of Kiss2-positive fibers and that of kiss-R2 expressing cells. The expression of kiss-R1 messengers was more limited to the habenula, the ventral telencephalon and the proximal pars distalis of the pituitary. Attempts to characterize the phenotype of the numerous cells expressing kiss-R2 showed that neurons expressing tyrosine hydroxylase, neuropeptide Y and neuronal nitric oxide synthase are targets for kisspeptins, while GnRH1 neurons did not appear to express kiss-R1 or kiss-R2 messengers. In addition, a striking result was that all somatostatin-positive neurons expressed-kissR2. These data show that kisspeptins are likely to regulate a wide range of neuronal systems in the brain of teleosts.


Asunto(s)
Lubina/metabolismo , Encéfalo/metabolismo , Proteínas de Peces/metabolismo , Kisspeptinas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Lubina/genética , Química Encefálica , Femenino , Proteínas de Peces/análisis , Proteínas de Peces/genética , Kisspeptinas/genética , Masculino , Neuronas/metabolismo , ARN Mensajero/análisis , Receptores Acoplados a Proteínas G/análisis , Receptores Acoplados a Proteínas G/genética
16.
Horm Behav ; 63(2): 193-207, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22521210

RESUMEN

The brain of the adult teleost fish exhibits intense neurogenic activity and an outstanding capability for brain repair. Remarkably, the brain estrogen-synthesizing enzyme, aromatase B, is strongly expressed, particularly in adult fishes, in radial glial cells, which act as progenitors. Using zebrafish, we tested the hypothesis that estrogens affect adult neurogenesis and brain regeneration by modulating the neurogenic activity of radial glial cells. To investigate this, the estrogenic environment was modified through inhibition of aromatase activity, blockade of nuclear estrogen receptors, or estrogenic treatments. Estrogens significantly decreased cell proliferation and migration at the olfactory bulbs/telencephalon junction and in the mediobasal hypothalamus. It also appears that cell survival is reduced at the olfactory bulbs/telencephalon junction. We also developed a model of telencephalic lesion to assess the role of aromatase and estrogens in brain repair. Proliferation increased rapidly immediately after the lesion in the parenchyma of the injured telencephalon, while proliferation at the ventricular surface appeared after 48 h and peaked at 7 days. At this time, most proliferative cells express Sox2, however, none of these Sox2 positive cells correspond to aromatase B-positive radial glial cells. Interestingly, aromatase B expression was significantly reduced 48 h and 7 days after the injury, but surprisingly, at 72 h after lesion, aromatase B expression appeared de novo expressed in parenchyma cells, suggesting a role for this ectopic expression of aromatase in brain repair mechanisms. Altogether these data suggest that estrogens modulate adult, but not reparative neurogenesis, in zebrafish.


Asunto(s)
Células Madre Adultas/efectos de los fármacos , Lesiones Encefálicas/fisiopatología , Estradiol/farmacología , Neurogénesis/efectos de los fármacos , Cicatrización de Heridas/efectos de los fármacos , Pez Cebra , Células Madre Adultas/fisiología , Factores de Edad , Animales , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Masculino , Modelos Biológicos , Regeneración Nerviosa/efectos de los fármacos , Regeneración Nerviosa/fisiología , Prosencéfalo/efectos de los fármacos , Prosencéfalo/fisiología , Cicatrización de Heridas/fisiología
17.
J Comp Neurol ; 521(4): 933-48, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22886357

RESUMEN

Kisspeptins are now considered key players in the neuroendocrine control of puberty and reproduction, at least in mammals. Most teleosts have two kiss genes, kiss1 and kiss2, but their sites of expression are still poorly documented. As a first step in investigating the role of kisspeptins in the European sea bass, a perciform fish, we studied the distribution of kiss1 and kiss2-expressing cells in the brain of males and females undergoing their first sexual maturation. Animals were examined at early and late in the reproductive season. We also examined the putative expression of estrogen receptors in kiss-expressing cells and, finally, we investigated whether kisspeptins are expressed in the pituitary gland. We show that kiss1-expressing cells were consistently detected in the habenula and, in mature males and females, in the rostral mediobasal hypothalamus. In both sexes, kiss2-expressing cells were consistently detected at the level of the preoptic area, but the main kiss2 mRNA-positive population was observed in the dorsal hypothalamus, above and under the lateral recess. No obvious sexual differences in kiss1 and kiss2 mRNA expression were detected. Additional studies based on confocal imaging clearly showed that most kiss1 mRNA-containing cells of the mediobasal hypothalamus strongly express ERα and slightly express ERß2. At the pituitary level, both sexes exhibited kiss1 mRNA expression in most FSHß-positive cells and never in LHß-positive cells.


Asunto(s)
Lubina/metabolismo , Encéfalo/metabolismo , Kisspeptinas/biosíntesis , Hipófisis/metabolismo , Animales , Western Blotting , Ensayo de Inmunoadsorción Enzimática , Femenino , Inmunohistoquímica , Hibridación Fluorescente in Situ , Kisspeptinas/análisis , Masculino , ARN Mensajero/análisis , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
18.
PLoS One ; 7(9): e44750, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22957105

RESUMEN

The cyp19a1 gene that encodes aromatase, the only enzyme permitting conversion of C19 aromatizable androgens into estrogens, is present as a single copy in the genome of most vertebrate species, except in teleosts in which it has been duplicated. This study aimed at investigating the brain expression of a cyp19a1 gene expressed in both gonad and brain of Japanese eel, a basal teleost. By means of immunohistochemistry and in situ hybridization, we show that cyp19a1 is expressed only in radial glial cells of the brain and in pituitary cells. Treatments with salmon pituitary homogenates (female) or human chorionic gonadotrophin (male), known to turn on steroid production in immature eels, strongly stimulated cyp19a1 messenger and protein expression in radial glial cells and pituitary cells. Using double staining studies, we also showed that aromatase-expressing radial glial cells exhibit proliferative activity in both the brain and the pituitary. Altogether, these data indicate that brain and pituitary expression of Japanese eel cyp19a1 exhibits characteristics similar to those reported for the brain specific cyp19a1b gene in teleosts having duplicated cyp19a1 genes. This supports the hypothesis that, despite the fact that eels also underwent the teleost specific genome duplication, they have a single cyp19a1 expressed in both brain and gonad. Such data also suggest that the intriguing features of brain aromatase expression in teleost fishes were not gained after the whole genome duplication and may reflect properties of the cyp19a1 gene of ancestral Actinopterygians.


Asunto(s)
Aromatasa/biosíntesis , Anguilas/fisiología , Regulación Enzimológica de la Expresión Génica , Neuroglía/enzimología , Animales , Aromatasa/química , Encéfalo/metabolismo , Gonadotropina Coriónica/metabolismo , Evolución Molecular , Femenino , Peces , Gonadotropinas/metabolismo , Humanos , Inmunohistoquímica/métodos , Hibridación in Situ , Masculino , Neuroglía/citología , Hipófisis , Salmón
19.
J Biol Chem ; 287(30): 24978-89, 2012 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-22613711

RESUMEN

Vascular smooth muscle cell (VSMC) trans-differentiation, or their switch from a contractile/quiescent to a secretory/inflammatory/migratory state, is known to play an important role in pathological vascular remodeling including atherosclerosis and postangioplasty restenosis. Several reports have established the Notch pathway as tightly regulating VSMC response to various stress factors through growth, migration, apoptosis, and de-differentiation. More recently, we showed that alterations of the Notch pathway also govern VSMC acquisition of the inflammatory state, one of the major events accelerating atherosclerosis. We also evidenced that the inflammatory context of atherosclerosis triggers a de novo expression of adenylyl cyclase isoform 8 (AC8), associated with the properties developed by trans-differentiated VSMCs. As an initial approach to understanding the regulation of AC8 expression, we examined the role of the Notch pathway. Here we show that inhibiting the Notch pathway enhances the effect of IL1ß on AC8 expression, amplifies its deleterious effects on the VSMC trans-differentiated phenotype, and decreases Notch target genes Hrt1 and Hrt3. Conversely, Notch activation resulted in blocking AC8 expression and up-regulated Hrt1 and Hrt3 expression. Furthermore, overexpressing Hrt1 and Hrt3 significantly decreased IL1ß-induced AC8 expression. In agreement with these in vitro findings, the in vivo rat carotid balloon-injury model of restenosis evidenced that AC8 de novo expression coincided with down-regulation of the Notch3 pathway. These results, demonstrating that the Notch pathway attenuates IL1ß-mediated AC8 up-regulation in trans-differentiated VSMCs, suggest that AC8 expression, besides being induced by the proinflammatory cytokine IL1ß, is also dependent on down-regulation of the Notch pathway occurring in an inflammatory context.


Asunto(s)
Adenilil Ciclasas/biosíntesis , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Interleucina-1beta/farmacología , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Receptores Notch/metabolismo , Transducción de Señal/efectos de los fármacos , Adenilil Ciclasas/genética , Animales , Traumatismos de las Arterias Carótidas/genética , Traumatismos de las Arterias Carótidas/metabolismo , Traumatismos de las Arterias Carótidas/patología , Transdiferenciación Celular , Modelos Animales de Enfermedad , Regulación Enzimológica de la Expresión Génica/genética , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Interleucina-1beta/metabolismo , Masculino , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Ratas , Ratas Wistar , Receptores Notch/genética , Proteínas Represoras/biosíntesis , Proteínas Represoras/genética , Transducción de Señal/genética , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
20.
PLoS One ; 6(11): e28375, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22140581

RESUMEN

In rodents, there is increasing evidence that nuclear progesterone receptors are transiently expressed in many regions of the developing brain, notably outside the hypothalamus. This suggests that progesterone and/or its metabolites could be involved in functions not related to reproduction, particularly in neurodevelopment. In this context, the adult fish brain is of particular interest, as it exhibits constant growth and high neurogenic activity that is supported by radial glia progenitors. However, although synthesis of neuroprogestagens has been documented recently in the brain of zebrafish, information on the presence of progesterone receptors is very limited. In zebrafish, a single nuclear progesterone receptor (pgr) has been cloned and characterized. Here, we demonstrate that this pgr is widely distributed in all regions of the zebrafish brain. Interestingly, we show that Pgr is strongly expressed in radial glial cells and more weakly in neurons. Finally, we present evidence, based on quantitative PCR and immunohistochemistry, that nuclear progesterone receptor mRNA and proteins are upregulated by estrogens in the brain of adult zebrafish. These data document for the first time the finding that radial glial cells are preferential targets for peripheral progestagens and/or neuroprogestagens. Given the crucial roles of radial glial cells in adult neurogenesis, the potential effects of progestagens on their activity and the fate of daughter cells require thorough investigation.


Asunto(s)
Encéfalo/metabolismo , Estrógenos/farmacología , Neuronas/metabolismo , Receptores de Progesterona/genética , Células Madre/metabolismo , Regulación hacia Arriba/genética , Pez Cebra/metabolismo , Animales , Encéfalo/citología , Encéfalo/crecimiento & desarrollo , Estradiol/farmacología , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Neuroglía/citología , Neuronas/citología , Neuronas/efectos de los fármacos , Área Preóptica/citología , Área Preóptica/efectos de los fármacos , Área Preóptica/metabolismo , Transporte de Proteínas/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Progesterona/metabolismo , Células Madre/citología , Células Madre/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...