66 research outputs found

    Dissection du programme développemental du noyau paraventriculaire de l'hypothalamus

    Full text link
    Une cascade de facteurs de transcription composée de SIM1, ARNT2, OTP, BRN2 et SIM2 est requise pour la différenciation des cinq types cellulaires qui peuplent le noyau paraventriculaire (PVN) de l’hypothalamus, un régulateur critique de plusieurs processus physiologiques essentiels à la survie. De plus, l’haploinsuffisance de Sim1 est aussi une cause d’hyperphagie isolée chez la souris et chez l’homme. Nous désirons disséquer le programme développemental du PVN, via une approche intégrative, afin d’identifier de nouveaux gènes qui ont le potentiel de réguler l’homéostasie chez l’individu adulte. Premièrement, nous avons utilisé une approche incluant l’analyse du transcriptome du PVN à différents stades du développement de la souris pour identifier de tels gènes. Nous avons comparé les transcriptomes de l’hypothalamus antérieur chez des embryons de souris Sim1+/+ et Sim1-/- à E12.5 issus de la même portée. De cette manière, nous avons identifié 56 gènes agissant en aval de Sim1 dont 5 facteurs de transcription - Irx3, Sax1, Rxrg, Ror et Neurod6. Nous avons également proposé un modèle de développement à deux couches de l’hypothalamus antérieur. Selon ce modèle, les gènes qui occupent un domaine médial dans la zone du manteau caractérisent des cellules qui peupleront le PVN alors que les gènes qui ont une expression latérale identifient des cellules qui donneront plus tard naissance aux structures ventrolatérales de l’hypothalamus. Nous avons aussi démontré que Sim1 est impliqué à la fois dans la différenciation, la migration et la prolifération des neurones qui peuplent le PVN tout comme Otp. Nous avons également isolé par microdissection au laser le PVN et l’hypothalamus médiobasal chez des souris de type sauvage à E14.5 pour en comparer les transcriptomes. Ceci nous a permis d’identifier 34 facteurs de transcription spécifiques au PVN et 76 facteurs spécifiques à l’hypothalamus médiobasal. Ces gènes représentent des régulateurs potentiels du développement hypothalamique. Deuxièmement, nous avons identifié 3 blocs de séquences au sein de la région 5’ d’Otp qui sont conservés chez l’homme, la souris et le poisson. Nous avons construit un transgène qui est composé d’un fragment de 7 kb contenant ces blocs de séquences et d’un gène rapporteur. L’analyse de 4 lignées de souris a montré que ce transgène est uniquement exprimé dans le PVN en développement. Nous avons généré un deuxième transgène dans lequel le fragment de 7 kb est inséré en amont de l’ADNc de Brn2 ou Sim1 et de Gfp. Nous avons obtenu quatre lignées de souris dans lesquels le profil d’expression de Brn2 et de Gfp reproduit celui d’Otp. Nous étudierons le développement du PVN et la prise alimentaire chez ces souris. En parallèle, nous croisons ces lignées avec les souris déficientes en Sim1 pour déterminer si l’expression de Brn2 permet le développement des cellules du PVN en absence de Sim1. En résumé, nous avons généré le premier transgène qui est exprimé spécifiquement dans le PVN. Ce transgène constitue un outil critique pour la dissection du programme développemental de l’hypothalamus. Troisièmement, nous avons caractérisé le développement de l’hypothalamus antérieur chez l’embryon de poulet qui représente un modèle intéressant pour réaliser des études de perte et de gain de fonction au cours du développement de cette structure. Il faut souligner que le modèle de développement à deux couches de l’hypothalamus antérieur semble être conservé chez l’embryon de poulet où il est aussi possible de classer les gènes selon leur profil d’expression médio-latéral et le devenir des régions qu’ils définissent. Finalement, nous croyons que cette approche intégrative nous permettra d’identifier et de caractériser des régulateurs du développement du PVN qui pourront potentiellement être associés à des pathologies chez l’adulte telles que l’obésité ou l’hypertension.A cascade of transcription factors composed of SIM1, ARNT2, OTP, BRN2 and SIM2 is required for the differentiation of the five major cell types populating the paraventricular nucleus (PVN) of the hypothalamus, a critical integrator of several homeostatic processes that are required for the survival of vertebrates. Haploinsufficency of Sim1 also causes isolated hyperphagia in mice and humans. The goal of our study is to dissect the developmental program of the PVN using an integrative approach in order to identify new genes that could potentially be implicated in the regulation of homeostasis in adults. First, we used a comparative approach to analyse the PVN transcriptome at different developmental stages in mice embryos in order to identity new genes implicated in PVN development. We compared gene expression in the anterior hypothalamus of E12.5 Sim1-/- and Sim1+/+ littermate embryos using a microarray approach. We identified 56 genes acting downstream of Sim1 including 5 transcription factors - Irx3, Sax1, Rxrg, Ror and Neurod6. We proposed a model for the development of the anterior hypothalamus. In this model, the genes expressed in the medial domain of the mantle layer characterise cells that will form the PVN and genes expressed in the lateral domain identify cells that will give rise to ventrolateral areas of the hypothalamus. We also showed that Sim1, like Otp, is implicated in the differentiation, migration and proliferation of the neurons populating the PVN. Furthermore, we have isolated by laser captured microdissection the PVN and ARC nucleus in wild type mice at E14.5 and compared their transcriptomes. This technique allowed us to identity 34 transcription factors specific to the PVN and 76 factors specific to the ARC. These genes represent potential regulators of hypothalamic development. Second, we identified 3 blocks of sequence in the 5’ region of Otp that are conserved between human, mouse and fish. We constructed a transgene which included a 7 Kb fragment encompassing these sequences followed by a reporter gene. The analysis of 4 mice strains showed that this transgene is specifically expressed in the prospective PVN. We have generated a second transgene in which the 7 Kb fragment is located upstream of the cDNA encoding Brn2 or Sim1 and Gfp. We obtained 4 mice strains in which the Brn2 and Gfp expression pattern is similar to the Otp expression pattern. These mice will be used to study PVN development and food intake. Also, to determine if Brn2 expression only – without Sim1 gene expression - allows the development of PVN cells, we are presently crossing these mice with Sim1 deficient mice. In conclusion, we have generated the first transgene that is specifically expressed in the PVN. This transgene constitutes a critical tool for dissecting the developmental program of the hypothalamus. Third, we have characterised the development of the anterior hypothalamus of chick embryos which represent an interesting model for loss and gain of function experiments during the development of this brain region. Interestingly, our proposed model for the development of the anterior hypothalamus seems to be conserved in chick embryos. As a matter of fact, it is possible to classify genes according to their medio-lateral expression patterns and the outcome of the regions that they are defining. Finally, we believe that this integrative approach will allow us to identify and characterize factors implicated in PVN development. From a clinical point of view, these factors could potentially be associated with pathologies such as obesity or arterial hypertension

    Genetic mapping of Foxb1-cell lineage shows migration from caudal diencephalon to telencephalon and lateral hypothalamus

    Get PDF
    The hypothalamus is a brain region with vital functions, and alterations in its development can cause human disease. However, we still do not have a complete description of how this complex structure is put together during embryonic and early postnatal stages. Radially oriented, outside-in migration of cells is prevalent in the developing hypothalamus. In spite of this, cell contingents from outside the hypothalamus as well as tangential hypothalamic migrations also have an important role. Here we study migrations in the hypothalamic primordium by genetically labeling the Foxb1 diencephalic lineage. Foxb1 is a transcription factor gene expressed in the neuroepithelium of the developing neural tube with a rostral expression boundary between caudal and rostral diencephalon, and therefore appropriate for marking migrations from caudal levels into the hypothalamus. We have found a large, longitudinally oriented migration stream apparently originating in the thalamic region and following an axonal bundle to end in the anterior portion of the lateral hypothalamic area. Additionally, we have mapped a specific expansion of the neuroepithelium into the rostral diencephalon. The expanded neuroepithelium generates abundant neurons for the medial hypothalamus at the tuberal level. Finally, we have uncovered novel diencephalon-to-telencephalon migrations into septum, piriform cortex and amygdala

    Homeodomain protein Otp affects developmental neuropeptide switching in oxytocin neurons associated with a long-term effect on social behavior

    Get PDF
    No supplementary materials.Proper response to stress and social stimuli depends on orchestrated development of hypothalamic neuronal circuits. Here we address the effects of the developmental transcription factor orthopedia (Otp) on hypothalamic development and function. We show that developmental mutations in the zebrafish paralogous gene otpa but not otpb affect both stress response and social preference. These behavioral phenotypes were associated with developmental alterations in oxytocinergic (OXT) neurons. Thus, otpa and otpb differentially regulate neuropeptide switching in a newly identified subset of OXT neurons that co-express the corticotropin-releasing hormone (CRH). Single-cell analysis revealed that these neurons project mostly to the hindbrain and spinal cord. Ablation of this neuronal subset specifically reduced adult social preference without affecting stress behavior, thereby uncoupling the contribution of a specific OXT cluster to social behavior from the general otpa(-/-) deficits. Our findings reveal a new role for Otp in controlling developmental neuropeptide balance in a discrete OXT circuit whose disrupted development affects social behavior.Israel Science Foundation grants: (1511/16, 957/12, 2137/16).info:eu-repo/semantics/publishedVersio

    Mice Doubly-Deficient in Lysosomal Hexosaminidase A and Neuraminidase 4 Show Epileptic Crises and Rapid Neuronal Loss

    Get PDF
    Tay-Sachs disease is a severe lysosomal disorder caused by mutations in the HexA gene coding for the α-subunit of lysosomal β-hexosaminidase A, which converts GM2 to GM3 ganglioside. Hexa−/− mice, depleted of β-hexosaminidase A, remain asymptomatic to 1 year of age, because they catabolise GM2 ganglioside via a lysosomal sialidase into glycolipid GA2, which is further processed by β-hexosaminidase B to lactosyl-ceramide, thereby bypassing the β-hexosaminidase A defect. Since this bypass is not effective in humans, infantile Tay-Sachs disease is fatal in the first years of life. Previously, we identified a novel ganglioside metabolizing sialidase, Neu4, abundantly expressed in mouse brain neurons. Now we demonstrate that mice with targeted disruption of both Neu4 and Hexa genes (Neu4−/−;Hexa−/−) show epileptic seizures with 40% penetrance correlating with polyspike discharges on the cortical electrodes of the electroencephalogram. Single knockout Hexa−/− or Neu4−/− siblings do not show such symptoms. Further, double-knockout but not single-knockout mice have multiple degenerating neurons in the cortex and hippocampus and multiple layers of cortical neurons accumulating GM2 ganglioside. Together, our data suggest that the Neu4 block exacerbates the disease in Hexa−/− mice, indicating that Neu4 is a modifier gene in the mouse model of Tay-Sachs disease, reducing the disease severity through the metabolic bypass. However, while disease severity in the double mutant is increased, it is not profound suggesting that Neu4 is not the only sialidase contributing to the metabolic bypass in Hexa−/− mice

    Colour removal from beet molasses by ultrafiltration with activated charcoal

    Get PDF
    The feasibility of an activated charcoal/ultrafiltration process for the decolouration of beet molasses, and subsequent regeneration of the exhausted charcoal by thermal and chemical methods, has been examined. Several activated charcoals were assayed prior to the selection of Norit powdered activated charcoal (NPAC). The affinity of NPAC for the adsorption of dark colour compounds was studied at 25 C. A colour reduction of over 98% was achieved at equilibrium using an NPAC concentration of 5 g/L from the beet molasses at pH 3, with no betaine or sucrose co-adsorptions. Crossflow ultrafiltration experiments with NPAC were performed using a 100 kDa TiO2 tubular ceramic membrane, in order to select the optimal operating conditions. Experiments with several ultrafiltration stages for the decolouration of beet molasses, and subsequent regeneration of the exhausted NPAC with sodium hydroxide solutions, were also performed under the conditions identified previously. A high colour reduction in the molasses of over 96.5%, with no adsorption of sucrose, betaine, citric acid or lactic acid, was achieved in the first decolouration stage at pH 3, with an initial NPAC concentration of 5 g/L, a transmembrane pressure of 100 kPa and a feed flowrate of 4.24 L/h. A good NPAC regeneration was obtained, with a loss of its colour removal capacity lower than 10%.Ministerio de Economía y Competitividad (MINECO, Spain) through project CTQ2011-25239 and from the Junta de Castilla y León through project BU175A11-

    Molecular evolution of the LNX gene family

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>LNX (Ligand of Numb Protein-X) proteins typically contain an amino-terminal RING domain adjacent to either two or four PDZ domains - a domain architecture that is unique to the LNX family. LNX proteins function as E3 ubiquitin ligases and their domain organisation suggests that their ubiquitin ligase activity may be targeted to specific substrates or subcellular locations by PDZ domain-mediated interactions. Indeed, numerous interaction partners for LNX proteins have been identified, but the <it>in vivo </it>functions of most family members remain largely unclear.</p> <p>Results</p> <p>To gain insights into their function we examined the phylogenetic origins and evolution of the <it>LNX </it>gene family. We find that a <it>LNX1/LNX2</it>-like gene arose in an early metazoan lineage by gene duplication and fusion events that combined a RING domain with four PDZ domains. These PDZ domains are closely related to the four carboxy-terminal domains from multiple PDZ domain containing protein-1 (MUPP1). Duplication of the <it>LNX1/LNX2</it>-like gene and subsequent loss of PDZ domains appears to have generated a gene encoding a LNX3/LNX4-like protein, with just two PDZ domains. This protein has novel carboxy-terminal sequences that include a potential modular LNX3 homology domain. The two ancestral <it>LNX </it>genes are present in some, but not all, invertebrate lineages. They were, however, maintained in the vertebrate lineage, with further duplication events giving rise to five LNX family members in most mammals. In addition, we identify novel interactions of LNX1 and LNX2 with three known MUPP1 ligands using yeast two-hybrid asssays. This demonstrates conservation of binding specificity between LNX and MUPP1 PDZ domains.</p> <p>Conclusions</p> <p>The <it>LNX </it>gene family has an early metazoan origin with a LNX1/LNX2-like protein likely giving rise to a LNX3/LNX4-like protein through the loss of PDZ domains. The absence of LNX orthologs in some lineages indicates that LNX proteins are not essential in invertebrates. In contrast, the maintenance of both ancestral <it>LNX </it>genes in the vertebrate lineage suggests the acquisition of essential vertebrate specific functions. The revelation that the LNX PDZ domains are phylogenetically related to domains in MUPP1, and have common binding specificities, suggests that LNX and MUPP1 may have similarities in their cellular functions.</p

    Differential requirements for Gli2 and Gli3 in the regional specification of the mouse hypothalamus.

    Get PDF
    Secreted protein Sonic hedgehog (Shh) ventralizes the neural tube by modulating the crucial balance between activating and repressing functions (GliA, GliR) of transcription factors Gli2 and Gli3. This balance—the Shh-Gli code—is species- and context-dependent and has been elucidated for the mouse spinal cord. The hypothalamus, a forebrain region regulating vital functions like homeostasis and hormone secretion, shows dynamic and intricate Shh expression as well as complex regional differentiation. Here we asked if particular combinations of Gli2 and Gli3 and of GliA and GliR functions contribute to the variety of hypothalamic regions, i.e. we wanted to clarify the hypothalamic version of the Shh-Gli code. Based on mouse mutant analysis, we show that: 1) hypothalamic regional heterogeneity is based in part on differentially stringent requirements for Gli2 or Gli3; 2) another source of diversity are differential requirements for Shh of neural vs non-neural origin; 3) Gli2 is indispensable for the specification of a medial progenitor domain generating several essential hypothalamic nuclei plus the pituitary and median eminence; 4) the suppression of Gli3R by neural and non-neural Shh is essential for hypothalamic specification. Finally, we have mapped our results on a recent model which considers the hypothalamus as a transverse region with alar and basal portions. Our data confirm the model and are explained by it
    corecore