Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Nat Commun ; 14(1): 6381, 2023 10 11.
Article in English | MEDLINE | ID: mdl-37821426

ABSTRACT

Circadian clocks generate rhythms of arousal, but the underlying molecular and cellular mechanisms remain unclear. In Drosophila, the clock output molecule WIDE AWAKE (WAKE) labels rhythmic neural networks and cyclically regulates sleep and arousal. Here, we show, in a male mouse model, that mWAKE/ANKFN1 labels a subpopulation of dorsomedial hypothalamus (DMH) neurons involved in rhythmic arousal and acts in the DMH to reduce arousal at night. In vivo Ca2+ imaging reveals elevated DMHmWAKE activity during wakefulness and rapid eye movement (REM) sleep, while patch-clamp recordings show that DMHmWAKE neurons fire more frequently at night. Chemogenetic manipulations demonstrate that DMHmWAKE neurons are necessary and sufficient for arousal. Single-cell profiling coupled with optogenetic activation experiments suggest that GABAergic DMHmWAKE neurons promote arousal. Surprisingly, our data suggest that mWAKE acts as a clock-dependent brake on arousal during the night, when mice are normally active. mWAKE levels peak at night under clock control, and loss of mWAKE leads to hyperarousal and greater DMHmWAKE neuronal excitability specifically at night. These results suggest that the clock does not solely promote arousal during an animal's active period, but instead uses opposing processes to produce appropriate levels of arousal in a time-dependent manner.


Subject(s)
Circadian Clocks , Sleep , Mice , Animals , Male , Arousal/physiology , Neurons/physiology , Hypothalamus/physiology , Circadian Rhythm/physiology
2.
Elife ; 122023 02 10.
Article in English | MEDLINE | ID: mdl-36718990

ABSTRACT

The tuberal hypothalamus controls life-supporting homeostatic processes, but despite its fundamental role, the cells and signalling pathways that specify this unique region of the central nervous system in embryogenesis are poorly characterised. Here, we combine experimental and bioinformatic approaches in the embryonic chick to show that the tuberal hypothalamus is progressively generated from hypothalamic floor plate-like cells. Fate-mapping studies show that a stream of tuberal progenitors develops in the anterior-ventral neural tube as a wave of neuroepithelial-derived BMP signalling sweeps from anterior to posterior through the hypothalamic floor plate. As later-specified posterior tuberal progenitors are generated, early specified anterior tuberal progenitors become progressively more distant from these BMP signals and differentiate into tuberal neurogenic cells. Gain- and loss-of-function experiments in vivo and ex vivo show that BMP signalling initiates tuberal progenitor specification, but must be eliminated for these to progress to anterior neurogenic progenitors. scRNA-Seq profiling shows that tuberal progenitors that are specified after the major period of anterior tuberal specification begin to upregulate genes that characterise radial glial cells. This study provides an integrated account of the development of the tuberal hypothalamus.


Subject(s)
Hypothalamus , Neurogenesis , Animals , Hypothalamus/metabolism , Neurogenesis/physiology , Signal Transduction , Chickens
3.
Cell Rep ; 38(3): 110251, 2022 01 18.
Article in English | MEDLINE | ID: mdl-35045288

ABSTRACT

The hypothalamus regulates many innate behaviors, but its development remains poorly understood. Here, we used single-cell RNA sequencing (RNA-seq) and hybridization chain reaction (HCR) to profile multiple stages of early hypothalamic development in the chick. Hypothalamic neuroepithelial cells are initially induced from prethalamic-like cells. Two distinct hypothalamic progenitor populations then emerge and give rise to tuberal and mammillary/paraventricular hypothalamic cells. At later stages, the regional organization of the chick and mouse hypothalamus is highly similar. We identify selective markers for major subdivisions of the developing chick hypothalamus and many previously uncharacterized candidate regulators of hypothalamic induction, regionalization, and neurogenesis. As proof of concept for the power of the dataset, we demonstrate that prethalamus-derived follistatin inhibits hypothalamic induction. This study clarifies the organization of the nascent hypothalamus and identifies molecular mechanisms that may control its induction and subsequent development.


Subject(s)
Hypothalamus/embryology , Neural Stem Cells/cytology , Neurogenesis/physiology , Animals , Chick Embryo , RNA-Seq , Single-Cell Analysis
4.
Sci Adv ; 7(22)2021 05.
Article in English | MEDLINE | ID: mdl-34049878

ABSTRACT

Hypothalamic tanycytes, radial glial cells that share many features with neuronal progenitors, can generate small numbers of neurons in the postnatal hypothalamus, but the identity of these neurons and the molecular mechanisms that control tanycyte-derived neurogenesis are unknown. In this study, we show that tanycyte-specific disruption of the NFI family of transcription factors (Nfia/b/x) robustly stimulates tanycyte proliferation and tanycyte-derived neurogenesis. Single-cell RNA sequencing (scRNA-seq) and single-cell assay for transposase-accessible chromatin sequencing (scATAC-seq) analysis reveals that NFI (nuclear factor I) factors repress Sonic hedgehog (Shh) and Wnt signaling in tanycytes and modulation of these pathways blocks proliferation and tanycyte-derived neurogenesis in Nfia/b/x-deficient mice. Nfia/b/x-deficient tanycytes give rise to multiple mediobasal hypothalamic neuronal subtypes that can mature, fire action potentials, receive synaptic inputs, and selectively respond to changes in internal states. These findings identify molecular mechanisms that control tanycyte-derived neurogenesis, which can potentially be targeted to selectively remodel the hypothalamic neural circuitry that controls homeostatic physiological processes.


Subject(s)
Ependymoglial Cells , Hedgehog Proteins , Animals , Ependymoglial Cells/metabolism , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Hypothalamus/metabolism , Mammals/metabolism , Mice , Neurogenesis/genetics , Neurons/metabolism
5.
Elife ; 102021 02 01.
Article in English | MEDLINE | ID: mdl-33522480

ABSTRACT

The ubiquitous presence of inhibitory interneurons in the thalamus of primates contrasts with the sparsity of interneurons reported in mice. Here, we identify a larger than expected complexity and distribution of interneurons across the mouse thalamus, where all thalamic interneurons can be traced back to two developmental programmes: one specified in the midbrain and the other in the forebrain. Interneurons migrate to functionally distinct thalamocortical nuclei depending on their origin: the abundant, midbrain-derived class populates the first and higher order sensory thalamus while the rarer, forebrain-generated class is restricted to some higher order associative regions. We also observe that markers for the midbrain-born class are abundantly expressed throughout the thalamus of the New World monkey marmoset. These data therefore reveal that, despite the broad variability in interneuron density across mammalian species, the blueprint of the ontogenetic organisation of thalamic interneurons of larger-brained mammals exists and can be studied in mice.


Subject(s)
Cell Lineage , Interneurons , Thalamus/growth & development , Animals , Callithrix , Cell Movement , Female , GABAergic Neurons , Gene Expression Regulation, Developmental , In Situ Hybridization , Male , Mesencephalon/growth & development , Mice , Mice, Transgenic , Prosencephalon/growth & development , Thalamus/cytology
6.
Commun Biol ; 4(1): 95, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33479483

ABSTRACT

GABAergic neurons of the hypothalamus regulate many innate behaviors, but little is known about the mechanisms that control their development. We previously identified hypothalamic neurons that express the LIM homeodomain transcription factor Lhx6, a master regulator of cortical interneuron development, as sleep-promoting. In contrast to telencephalic interneurons, hypothalamic Lhx6 neurons do not undergo long-distance tangential migration and do not express cortical interneuronal markers such as Pvalb. Here, we show that Lhx6 is necessary for the survival of hypothalamic neurons. Dlx1/2, Nkx2-2, and Nkx2-1 are each required for specification of spatially distinct subsets of hypothalamic Lhx6 neurons, and that Nkx2-2+/Lhx6+ neurons of the zona incerta are responsive to sleep pressure. We further identify multiple neuropeptides that are enriched in spatially segregated subsets of hypothalamic Lhx6 neurons, and that are distinct from those seen in cortical neurons. These findings identify common and divergent molecular mechanisms by which Lhx6 controls the development of GABAergic neurons in the hypothalamus.


Subject(s)
Cell Differentiation , GABAergic Neurons/physiology , Gene Regulatory Networks , Hypothalamus/cytology , LIM-Homeodomain Proteins/metabolism , Nerve Tissue Proteins/metabolism , Transcription Factors/metabolism , Animals , Cell Survival , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/metabolism , Hypothalamus/metabolism , Mice , Nuclear Proteins , Sleep/physiology
7.
Nat Commun ; 11(1): 4360, 2020 08 31.
Article in English | MEDLINE | ID: mdl-32868762

ABSTRACT

The hypothalamus is a central regulator of many innate behaviors essential for survival, but the molecular mechanisms controlling hypothalamic patterning and cell fate specification are poorly understood. To identify genes that control hypothalamic development, we have used single-cell RNA sequencing (scRNA-Seq) to profile mouse hypothalamic gene expression across 12 developmental time points between embryonic day 10 and postnatal day 45. This identified genes that delineated clear developmental trajectories for all major hypothalamic cell types, and readily distinguished major regional subdivisions of the developing hypothalamus. By using our developmental dataset, we were able to rapidly annotate previously unidentified clusters from existing scRNA-Seq datasets collected during development and to identify the developmental origins of major neuronal populations of the ventromedial hypothalamus. We further show that our approach can rapidly and comprehensively characterize mutants that have altered hypothalamic patterning, identifying Nkx2.1 as a negative regulator of prethalamic identity. These data serve as a resource for further studies of hypothalamic development, physiology, and dysfunction.


Subject(s)
Cell Differentiation , Hypothalamus , Neurons/metabolism , Thyroid Nuclear Factor 1/metabolism , Animals , Base Sequence , Body Patterning , Gene Expression Regulation, Developmental , Hypothalamus/cytology , Hypothalamus/embryology , Hypothalamus/growth & development , Hypothalamus/metabolism , Mice , Mutation , Single-Cell Analysis , Thyroid Nuclear Factor 1/genetics
8.
PLoS Comput Biol ; 15(8): e1007040, 2019 08.
Article in English | MEDLINE | ID: mdl-31469823

ABSTRACT

Single-cell RNA-sequencing (scRNA-seq) provides new opportunities to gain a mechanistic understanding of many biological processes. Current approaches for single cell clustering are often sensitive to the input parameters and have difficulty dealing with cell types with different densities. Here, we present Panoramic View (PanoView), an iterative method integrated with a novel density-based clustering, Ordering Local Maximum by Convex hull (OLMC), that uses a heuristic approach to estimate the required parameters based on the input data structures. In each iteration, PanoView will identify the most confident cell clusters and repeat the clustering with the remaining cells in a new PCA space. Without adjusting any parameter in PanoView, we demonstrated that PanoView was able to detect major and rare cell types simultaneously and outperformed other existing methods in both simulated datasets and published single-cell RNA-sequencing datasets. Finally, we conducted scRNA-Seq analysis of embryonic mouse hypothalamus, and PanoView was able to reveal known cell types and several rare cell subpopulations.


Subject(s)
Algorithms , Sequence Analysis, RNA/statistics & numerical data , Animals , Cluster Analysis , Computational Biology , Computer Simulation , Databases, Nucleic Acid/statistics & numerical data , Hypothalamus/cytology , Hypothalamus/embryology , Hypothalamus/metabolism , Mice , Single-Cell Analysis/statistics & numerical data
9.
Dev Biol ; 442(2): 236-248, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30063881

ABSTRACT

The hypothalamus is a small, but anatomically and functionally complex region of the brain whose development is poorly understood. In this study, we have explored its development by studying the canonical Wnt signaling pathway, generating gain and loss of function mutations of beta-catenin (Ctnnb1) in both hypothalamic and prethalamic neuroepithelium. Deletion of Ctnnb1 resulted in an anteriorized and hypoplastic hypothalamus. Posterior structures were lost or reduced, and anterior structures were expanded. In contrast, overexpression of a constitutively active mutant form of Ctnnb1 resulted in severe hyperplasia of prethalamus and hypothalamus, and expanded expression of a subset of posterior and premamillary hypothalamic markers. Moderate defects in differentiation of Arx-positive GABAergic neural precursors were observed in both prethalamus and hypothalamus of Ctnnb1 loss of function mutants, while in gain of function mutants, their differentiation was completely suppressed, although markers of prethalamic progenitors were preserved. Multiple other region-specific markers, including several specific posterior hypothalamic structures, were also suppressed in Ctnnb1 gain of function mutations. Severe, region-specific defects in hypothalamic nucleogenesis were also observed in both gain and loss of function mutations of Ctnnb1. Finally, both gain and loss of function of Ctnnb1 also produced severe, non-cell autonomous disruptions of pituitary development. These findings demonstrate a central and multifaceted role for canonical Wnt signaling in regulating growth, patterning, differentiation and nucleogenesis in multiple diencephalic regions.


Subject(s)
Hypothalamus/embryology , Hypothalamus/metabolism , Wnt Signaling Pathway/physiology , Animals , Body Patterning/physiology , Cell Differentiation/physiology , Female , Hypothalamus/cytology , Male , Mice , Mice, Transgenic , Pregnancy , beta Catenin/genetics , beta Catenin/metabolism
10.
Prog Neurobiol ; 170: 53-66, 2018 11.
Article in English | MEDLINE | ID: mdl-29631023

ABSTRACT

Over the past two decades, evidence has accumulated that neurogenesis can occur in both the juvenile and adult mammalian hypothalamus. Levels of hypothalamic neurogenesis can be regulated by dietary, environmental and hormonal signals. Since the hypothalamus has a central role in controlling a broad range of homeostatic physiological processes, these findings may have far ranging behavioral and medical implications. However, many questions in the field remain unresolved, including the cells of origin of newborn hypothalamic neurons and the extent to which these cells actually regulate hypothalamic-controlled behaviors. In this manuscript, we conduct a critical review of the literature on postnatal hypothalamic neurogenesis in mammals, lay out the main outstanding controversies in the field, and discuss how best to advance our knowledge of this fascinating but still poorly understood process.


Subject(s)
Hypothalamus/physiology , Neurogenesis/physiology , Animals , Humans
11.
Dev Biol ; 439(2): 102-111, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29679559

ABSTRACT

Although the hypothalamus functions as a master homeostat for many behaviors, little is known about the transcriptional networks that control its development. To investigate this question, we analyzed mice deficient for the Forkhead domain transcription factor Foxd1. Foxd1 is selectively expressed in neuroepithelial cells of the prethalamus and hypothalamus prior to the onset of neurogenesis, and is later restricted to neural progenitors of the prethalamus and anterior hypothalamus. During early stages of neurogenesis, we observed that Foxd1-deficient mice showed reduced expression of Six3 and Vax1 in anterior hypothalamus, but overall patterning of the prethalamus and hypothalamus is unaffected. After neurogenesis is complete, however, a progressive reduction and eventual loss of expression of molecular markers of the suprachiasmatic, paraventricular and periventricular hypothalamic is observed. These findings demonstrate that Foxd1 acts in hypothalamic progenitors to allow sustained expression of a subset of genes selectively expressed in mature neurons of the anterior hypothalamus.


Subject(s)
Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Animals , Anterior Hypothalamic Nucleus/metabolism , Anterior Hypothalamic Nucleus/physiology , Body Patterning/genetics , Cell Differentiation/genetics , Eye Proteins/genetics , Eye Proteins/metabolism , Forkhead Transcription Factors/physiology , Gene Expression Regulation, Developmental/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Hypothalamus/metabolism , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurogenesis/physiology , Neurons/metabolism , Neuropeptides/genetics , Neuropeptides/metabolism , Stem Cells/metabolism , Stem Cells/physiology , Transcription Factors/metabolism , Homeobox Protein SIX3
12.
Sleep ; 39(3): 613-24, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26612390

ABSTRACT

STUDY OBJECTIVES: Optimal sleep is ensured by the interaction of circadian and homeostatic processes. Although synaptic plasticity seems to contribute to both processes, the specific players involved are not well understood. The EphA4 tyrosine kinase receptor is a cell adhesion protein regulating synaptic plasticity. We investigated the role of EphA4 in sleep regulation using electrocorticography in mice lacking EphA4 and gene expression measurements. METHODS: EphA4 knockout (KO) mice, Clock(Δ19/Δ19) mutant mice and littermates, C57BL/6J and CD-1 mice, and Sprague-Dawley rats were studied under a 12 h light: 12 h dark cycle, under undisturbed conditions or 6 h sleep deprivation (SLD), and submitted to a 48 h electrophysiological recording and/or brain sampling at different time of day. RESULTS: EphA4 KO mice showed less rapid eye movement sleep (REMS), enhanced duration of individual bouts of wakefulness and nonrapid eye movement sleep (NREMS) during the light period, and a blunted daily rhythm of NREMS sigma activity. The NREMS delta activity response to SLD was unchanged in EphA4 KO mice. However, SLD increased EphA4 expression in the thalamic/hypothalamic region in C57BL/6J mice. We further show the presence of E-boxes in the promoter region of EphA4, a lower expression of EphA4 in Clock mutant mice, a rhythmic expression of EphA4 ligands in several brain areas, expression of EphA4 in the suprachiasmatic nuclei of the hypothalamus (SCN), and finally an unchanged number of cells expressing Vip, Grp and Avp in the SCN of EphA4 KO mice. CONCLUSIONS: Our results suggest that EphA4 is involved in circadian sleep regulation.


Subject(s)
Circadian Rhythm/physiology , Receptor, EphA4/metabolism , Sleep Deprivation/physiopathology , Sleep/physiology , Animals , CLOCK Proteins/genetics , Circadian Rhythm/genetics , Darkness , Electrocorticography , Electrophysiological Phenomena , Homeostasis , Light , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuronal Plasticity , Promoter Regions, Genetic/genetics , Rats , Rats, Sprague-Dawley , Receptor, EphA4/biosynthesis , Receptor, EphA4/deficiency , Receptor, EphA4/genetics , Sleep/genetics , Sleep Deprivation/genetics , Sleep, REM/genetics , Sleep, REM/physiology , Suprachiasmatic Nucleus/metabolism , Thalamus/metabolism , Time Factors , Wakefulness/genetics , Wakefulness/physiology
13.
Wiley Interdiscip Rev Dev Biol ; 4(5): 445-68, 2015.
Article in English | MEDLINE | ID: mdl-25820448

ABSTRACT

Owing to its complex structure and highly diverse cell populations, the study of hypothalamic development has historically lagged behind that of other brain regions. However, in recent years, a greatly expanded understanding of hypothalamic gene expression during development has opened up new avenues of investigation. In this review, we synthesize existing work to present a holistic picture of hypothalamic development from early induction and patterning through nuclear specification and differentiation, with a particular emphasis on determination of cell fate. We will also touch on special topics in the field including the prosomere model, adult neurogenesis, and integration of migratory cells originating outside the hypothalamic neuroepithelium, and how these topics relate to our broader theme.


Subject(s)
Body Patterning , Cell Differentiation , Gene Expression Regulation, Developmental , Hypothalamus/embryology , Animals , Humans , Hypothalamus/metabolism , Signal Transduction
14.
J Neurosci ; 34(50): 16809-20, 2014 Dec 10.
Article in English | MEDLINE | ID: mdl-25505333

ABSTRACT

Hypothalamic tanycytes, a radial glial-like ependymal cell population that expresses numerous genes selectively enriched in embryonic hypothalamic progenitors and adult neural stem cells, have recently been observed to serve as a source of adult-born neurons in the mammalian brain. The genetic mechanisms that regulate the specification and maintenance of tanycyte identity are unknown, but are critical for understanding how these cells can act as adult neural progenitor cells. We observe that LIM (Lin-11, Isl-1, Mec-3)-homeodomain gene Lhx2 is selectively expressed in hypothalamic progenitor cells and tanycytes. To test the function of Lhx2 in tanycyte development, we used an intersectional genetic strategy to conditionally delete Lhx2 in posteroventral hypothalamic neuroepithelium, both embryonically and postnatally. We observed that tanycyte development was severely disrupted when Lhx2 function was ablated during embryonic development. Lhx2-deficient tanycytes lost expression of tanycyte-specific genes, such as Rax, while also displaying ectopic expression of genes specific to cuboid ependymal cells, such as Rarres2. Ultrastructural analysis revealed that mutant tanycytes exhibited a hybrid identity, retaining radial morphology while becoming multiciliated. In contrast, postnatal loss of function of Lhx2 resulted only in loss of expression of tanycyte-specific genes. Using chromatin immunoprecipitation, we further showed that Lhx2 directly regulated expression of Rax, an essential homeodomain factor for tanycyte development. This study identifies Lhx2 as a key intrinsic regulator of tanycyte differentiation, sustaining Rax-dependent activation of tanycyte-specific genes while also inhibiting expression of ependymal cell-specific genes. These findings provide key insights into the transcriptional regulatory network specifying this still poorly characterized cell type.


Subject(s)
Cell Differentiation/physiology , Ependymoglial Cells/physiology , Hypothalamus/cytology , Hypothalamus/physiology , LIM-Homeodomain Proteins/physiology , Neurogenesis/physiology , Transcription Factors/physiology , Animals , Female , Male , Mice , Mice, Transgenic
15.
PLoS One ; 9(4): e90381, 2014.
Article in English | MEDLINE | ID: mdl-24699247

ABSTRACT

To study gene function in neural progenitors and radial glia of the retina and hypothalamus, we developed a Rax-CreERT2 mouse line in which a tamoxifen-inducible Cre recombinase is inserted into the endogenous Rax locus. By crossing Rax-CreER(T2) with the Cre-dependent Ai9 reporter line, we demonstrate that tamoxifen-induced Cre activity recapitulates the endogenous Rax mRNA expression pattern. During embryonic development, Cre recombinase activity in Rax-CreER(T2) is confined to retinal and hypothalamic progenitor cells, as well as progenitor cells of the posterior pituitary. At postnatal time points, selective Cre recombinase activity is seen in radial glial-like cell types in these organs--specifically Müller glia and tanycytes--as well as pituicytes. We anticipate that this line will prove useful for cell lineage analysis and investigation of gene function in the developing and mature retina, hypothalamus and pituitary.


Subject(s)
Eye Proteins/physiology , Gene Deletion , Homeodomain Proteins/physiology , Hypothalamus/metabolism , Integrases/metabolism , Neuroglia/metabolism , Receptors, Estrogen/physiology , Retina/metabolism , Stem Cells/metabolism , Transcription Factors/physiology , Animals , Antineoplastic Agents, Hormonal/pharmacology , Blotting, Southern , Cell Lineage , Female , Hypothalamus/cytology , Hypothalamus/drug effects , Immunoenzyme Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuroglia/cytology , Neuroglia/drug effects , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Recombination, Genetic , Retina/cytology , Retina/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/cytology , Stem Cells/drug effects , Tamoxifen/pharmacology
16.
J Comp Neurol ; 522(4): 876-99, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23939786

ABSTRACT

The wall of the ventral third ventricle is composed of two distinct cell populations: tanycytes and ependymal cells. Tanycytes regulate many aspects of hypothalamic physiology, but little is known about the transcriptional network that regulates their development and function. We observed that the retina and anterior neural fold homeobox transcription factor (Rax) is selectively expressed in hypothalamic tanycytes, and showed a complementary pattern of expression to markers of hypothalamic ependymal cells, such as Rarres2 (retinoic acid receptor responder [tazarotene induced] 2). To determine whether Rax controls tanycyte differentiation and function, we generated Rax haploinsufficient mice and examined their cellular and molecular phenotype in adulthood. These mice appeared grossly normal, but careful examination revealed a thinning of the third ventricular wall and reduction of both tanycyte and ependymal markers. These experiments show that Rax is required for hypothalamic tanycyte and ependymal cell differentiation. Rax haploinsufficiency also resulted in the ectopic presence of ependymal cells in the α2 tanycytic zone, where few ependymal cells are normally found, suggesting that Rax is selectively required for α2 tanycyte differentiation. These changes in the ventricular wall were associated with reduced diffusion of Evans Blue tracer from the ventricle to the hypothalamic parenchyma, with no apparent repercussion on the gross anatomical or behavioral phenotype of these mice. In conclusion, we have provided evidence that Rax is required for the normal differentiation and patterning of hypothalamic tanycytes and ependymal cells, as well as for maintenance of the cerebrospinal fluid-hypothalamus barrier.


Subject(s)
Cell Differentiation/physiology , Ependymoglial Cells/physiology , Eye Proteins/physiology , Homeodomain Proteins/physiology , Hypothalamus/cytology , Transcription Factors/physiology , Animals , Chemokines , Chemotactic Factors/metabolism , Evans Blue , Eye Proteins/genetics , Female , Gene Expression Regulation/genetics , Genotype , Homeodomain Proteins/genetics , Hypothalamus/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neuroglia/metabolism , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/metabolism , Third Ventricle/metabolism , Transcription Factors/genetics
17.
J Vis Exp ; (81): e50716, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24300415

ABSTRACT

The functional characterization of adult-born neurons remains a significant challenge. Approaches to inhibit adult neurogenesis via invasive viral delivery or transgenic animals have potential confounds that make interpretation of results from these studies difficult. New radiological tools are emerging, however, that allow one to noninvasively investigate the function of select groups of adult-born neurons through accurate and precise anatomical targeting in small animals. Focal ionizing radiation inhibits the birth and differentiation of new neurons, and allows targeting of specific neural progenitor regions. In order to illuminate the potential functional role that adult hypothalamic neurogenesis plays in the regulation of physiological processes, we developed a noninvasive focal irradiation technique to selectively inhibit the birth of adult-born neurons in the hypothalamic median eminence. We describe a method for Computer tomography-guided focal irradiation (CFIR) delivery to enable precise and accurate anatomical targeting in small animals. CFIR uses three-dimensional volumetric image guidance for localization and targeting of the radiation dose, minimizes radiation exposure to nontargeted brain regions, and allows for conformal dose distribution with sharp beam boundaries. This protocol allows one to ask questions regarding the function of adult-born neurons, but also opens areas to questions in areas of radiobiology, tumor biology, and immunology. These radiological tools will facilitate the translation of discoveries at the bench to the bedside.


Subject(s)
Hypothalamus/cytology , Hypothalamus/radiation effects , Neurogenesis/radiation effects , Neurons/cytology , Neurons/radiation effects , Tomography, X-Ray Computed/methods , Animals , Female , Histones/metabolism , Immunohistochemistry , Mice , Mice, Inbred C57BL , Neural Stem Cells/cytology , Neural Stem Cells/radiation effects , Tomography, X-Ray Computed/instrumentation
18.
PLoS One ; 8(11): e80781, 2013.
Article in English | MEDLINE | ID: mdl-24312242

ABSTRACT

During development, prenatal and postnatal factors program homeostatic set points to regulate food intake and body weight in the adult. Combinations of genetic and environmental factors contribute to the development of neural circuitry that regulates whole-body energy homeostasis. Brain-derived neurotrophic factor (Bdnf) and its receptor, Tyrosine kinase receptor B (TrkB), are strong candidates for mediating the reshaping of hypothalamic neural circuitry, given their well-characterized role in the central regulation of feeding and body weight. Here, we employ a chemical-genetic approach using the TrkB(F616A/F616A) knock-in mouse model to define the critical developmental period in which TrkB inhibition contributes to increased adult fat mass. Surprisingly, transient TrkB inhibition in embryos, preweaning pups, and adults all resulted in long-lasting increases in body weight and fat content. Moreover, sex-specific differences in the effects of TrkB inhibition on both body weight and hypothalamic gene expression were observed at multiple developmental stages. Our results highlight both the importance of the Bdnf/TrkB pathway in maintaining normal body weight throughout life and the role of sex-specific differences in the organization of hypothalamic neural circuitry that regulates body weight.


Subject(s)
Body Weight/drug effects , Gene Expression Regulation, Developmental/drug effects , Hypothalamus/drug effects , Hypothalamus/metabolism , Protein Kinase Inhibitors/pharmacology , Receptor, trkB/antagonists & inhibitors , Sex Characteristics , Animals , Body Composition/drug effects , Body Weight/genetics , Female , Male , Mice , Mice, Transgenic , Receptor, trkB/genetics
19.
Am J Physiol Regul Integr Comp Physiol ; 304(12): R1085-95, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23576617

ABSTRACT

Disruption of finely coordinated neuropeptide signals in the hypothalamus can result in altered food intake and body weight. We identified neuron-derived neurotrophic factor (NENF) as a novel secreted protein through a large-scale screen aimed at identifying novel secreted hypothalamic proteins that regulate food intake. We observed robust Nenf expression in hypothalamic nuclei known to regulate food intake, and its expression was altered under the diet-induced obese (DIO) condition relative to the fed state. Hypothalamic Nenf mRNA was regulated by brain-derived neurotrophic factor (BDNF) signaling, itself an important regulator of appetite. Delivery of purified recombinant BDNF into the lateral cerebral ventricle decreased hypothalamic Nenf expression, while pharmacological inhibition of trkB signaling increased Nenf mRNA expression. Furthermore, recombinant NENF administered via an intracerebroventricular cannula decreased food intake and body weight and increased hypothalamic Pomc and Mc4r mRNA expression. Importantly, the appetite-suppressing effect of NENF was abrogated in obese mice fed a high-fat diet, demonstrating a diet-dependent modulation of NENF function. We propose the existence of a regulatory circuit involving BDNF, NENF, and melanocortin signaling. Our study validates the power of using an integrated experimental and bioinformatic approach to identify novel CNS-derived proteins with appetite-modulating function and reveals NENF as an important central modulator of food intake.


Subject(s)
Appetite/physiology , Hypothalamus/metabolism , Nerve Tissue Proteins/metabolism , Obesity/physiopathology , Signal Transduction/physiology , Animals , Appetite/drug effects , Brain-Derived Neurotrophic Factor/administration & dosage , Brain-Derived Neurotrophic Factor/pharmacology , Diet, High-Fat/adverse effects , Disease Models, Animal , Energy Metabolism/drug effects , Energy Metabolism/physiology , Injections, Intraventricular , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/administration & dosage , Nerve Tissue Proteins/pharmacology , Obesity/chemically induced , Obesity/metabolism , Pro-Opiomelanocortin/metabolism , Receptor, Melanocortin, Type 4/metabolism , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Signal Transduction/drug effects
20.
Int J Dev Neurosci ; 30(8): 615-21, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22867732

ABSTRACT

Adult neurogenesis represents a striking example of structural plasticity in the mature brain. Research on adult mammalian neurogenesis today focuses almost exclusively on two areas: the subgranular zone (SGZ) in the dentate gyrus of the hippocampus, and the subventricular zone (SVZ) of the lateral ventricles. Numerous studies, however, have also reported adult neurogenesis in the hypothalamus, a brain structure that serves as a central homeostatic regulator of numerous physiological and behavioral functions, such as feeding, metabolism, body temperature, thirst, fatigue, aggression, sleep, circadian rhythms, and sexual behavior. Recent studies on hypothalamic neurogenesis have identified a progenitor population within a dedicated hypothalamic neurogenic zone. Furthermore, adult born hypothalamic neurons appear to play a role in the regulation of metabolism, weight, and energy balance. It remains to be seen what other functional roles adult hypothalamic neurogenesis may play. This review summarizes studies on the identification and characterization of neural stem/progenitor cells in the mammalian hypothalamus, in what contexts these stem/progenitor cells engage in neurogenesis, and potential functions of postnatally generated hypothalamic neurons.


Subject(s)
Hypothalamus/cytology , Hypothalamus/physiology , Neurogenesis/physiology , Neurons/physiology , Animals , Humans , Mammals , Neural Stem Cells/physiology
SELECTION OF CITATIONS
SEARCH DETAIL