Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Res Sq ; 2024 Feb 09.
Article in English | MEDLINE | ID: mdl-38405925

ABSTRACT

Astrocytes safeguard the homeostasis of the central nervous system1,2. Despite their prominent morphological plasticity under conditions that challenge the brain's adaptive capacity3-5, the classification of astrocytes, and relating their molecular make-up to spatially devolved neuronal operations that specify behavior or metabolism, remained mostly futile6,7. Although it seems unexpected in the era of single-cell biology, the lack of a major advance in stratifying astrocytes under physiological conditions rests on the incompatibility of 'neurocentric' algorithms that rely on stable developmental endpoints, lifelong transcriptional, neurotransmitter, and neuropeptide signatures for classification6-8 with the dynamic functional states, anatomic allocation, and allostatic plasticity of astrocytes1. Simplistically, therefore, astrocytes are still grouped as 'resting' vs. 'reactive', the latter referring to pathological states marked by various inducible genes3,9,10. Here, we introduced a machine learning-based feature recognition algorithm that benefits from the cumulative power of published single-cell RNA-seq data on astrocytes as a reference map to stepwise eliminate pleiotropic and inducible cellular features. For the healthy hypothalamus, this walk-back approach revealed gene regulatory networks (GRNs) that specified subsets of astrocytes, and could be used as landmarking tools for their anatomical assignment. The core molecular censuses retained by astrocyte subsets were sufficient to stratify them by allostatic competence, chiefly their signaling and metabolic interplay with neurons. Particularly, we found differentially expressed mitochondrial genes in insulin-sensing astrocytes and demonstrated their reciprocal signaling with neurons that work antagonistically within the food intake circuitry. As a proof-of-concept, we showed that disrupting Mfn2 expression in astrocytes reduced their ability to support dynamic circuit reorganization, a time-locked feature of satiety in the hypothalamus, thus leading to obesity in mice. Overall, our results suggest that astrocytes in the healthy brain are fundamentally more heterogeneous than previously thought and topologically mirror the specificity of local neurocircuits.

2.
J Med Chem ; 66(21): 14853-14865, 2023 11 09.
Article in English | MEDLINE | ID: mdl-37857356

ABSTRACT

Oxytocin (OT) and vasopressin (VP) are related neuropeptides that regulate many biological processes. In humans, OT and VP act via four G protein-coupled receptors, OTR, V1aR, V1bR, and V2R (VPRs), which are associated with several disorders. To investigate the therapeutic potential of these receptors, particularly in the receptor-dense areas of the brain, molecular probes with a high temporal and spatial resolution are required. Such a spatiotemporal resolution can be achieved by incorporating photochromic moieties into OT and VP. Here, we report the design, synthesis, and (photo)pharmacological characterization of 12 OT- and VP-derived photoprobes using different modification strategies. Despite OT's and VP's sensitivity toward structural changes, we identified two photoprobes with good potency and photoswitch window for investigating the OTR and V1bR. These photoprobes should be of high value for producing cutting-edge photocontrollable peptide probes for the study of dynamic and kinetic receptor activation processes in specific regions of the brain.


Subject(s)
Neuropeptides , Oxytocin , Humans , Oxytocin/pharmacology , Receptors, Oxytocin , Vasopressins/pharmacology , Receptors, G-Protein-Coupled
3.
J Neuroendocrinol ; 35(9): e13320, 2023 09.
Article in English | MEDLINE | ID: mdl-37497857

ABSTRACT

Obesity is a key medical challenge of our time. The increasing number of children born to overweight or obese women is alarming. During pregnancy, the circulation of the mother and her fetus interact to maintain the uninterrupted availability of essential nutrients for fetal organ development. In doing so, the mother's dietary preference determines the amount and composition of nutrients reaching the fetus. In particular, the availability of polyunsaturated fatty acids (PUFAs), chiefly their ω-3 and ω-6 subclasses, can change when pregnant women choose a specific diet. Here, we provide a succinct overview of PUFA biochemistry, including exchange routes between ω-3 and ω-6 PUFAs, the phenotypes, and probable neurodevelopmental disease associations of offspring born to mothers consuming specific PUFAs, and their mechanistic study in experimental models to typify signaling pathways, transcriptional, and epigenetic mechanisms by which PUFAs can imprint long-lasting modifications to brain structure and function. We emphasize that the ratio, rather than the amount of individual ω-3 or ω-6 PUFAs, might underpin physiologically correct cellular differentiation programs, be these for neurons or glia, during pregnancy. Thereupon, the PUFA-driven programming of the brain is contextualized for childhood obesity, metabolic, and endocrine illnesses.


Subject(s)
Fatty Acids, Omega-3 , Pediatric Obesity , Humans , Child , Female , Pregnancy , Fatty Acids, Omega-6/metabolism , Fatty Acids, Omega-6/pharmacology , Fatty Acids, Omega-3/metabolism , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Unsaturated/pharmacology , Brain/metabolism , Fetus/metabolism
4.
Acta Physiol (Oxf) ; 238(3): e13973, 2023 07.
Article in English | MEDLINE | ID: mdl-37029761

ABSTRACT

AIM: This study mapped the spatiotemporal positions and connectivity of Onecut3+ neuronal populations in the developing and adult mouse brain. METHODS: We generated fluorescent reporter mice to chart Onecut3+ neurons for brain-wide analysis. Moreover, we crossed Onecut3-iCre and Mapt-mGFP (Tau-mGFP) mice to visualize axonal projections. A dual Cre/Flp-dependent AAV construct in Onecut3-iCre cross-bred with Slc17a6-FLPo mice was used in an intersectional strategy to map the connectivity of glutamatergic lateral hypothalamic neurons in the adult mouse. RESULTS: We first found that Onecut3 marks a hitherto undescribed Slc17a6+ /Vglut2+ neuronal cohort in the lateral hypothalamus, with the majority expressing thyrotropin-releasing hormone. In the adult, Onecut3+ /Vglut2+ neurons of the lateral hypothalamus had both intra- and extrahypothalamic efferents, particularly to the septal complex and habenula, where they targeted other cohorts of Onecut3+ neurons and additionally to the neocortex and hippocampus. This arrangement suggests that intrinsic reinforcement loops could exist for Onecut3+ neurons to coordinate their activity along the brain's midline axis. CONCLUSION: We present both a toolbox to manipulate novel subtypes of hypothalamic neurons and an anatomical arrangement by which extrahypothalamic targets can be simultaneously entrained.


Subject(s)
Hypothalamic Area, Lateral , Neurons , Mice , Animals , Mice, Transgenic , Neurons/physiology , Hypothalamus , Brain
5.
Neurochem Res ; 48(4): 1242-1253, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36482034

ABSTRACT

Cannabis legalization prompted the dilemma if plant-derived recreational drugs can have therapeutic potential and, consequently, how to address their regulation and safe distribution. In parallel, the steady worldwide decriminalization of cannabis and the enhanced content of its main psychoactive compound Δ9-tetrahydrocannabinol (THC), exposes populations to increasing amounts of cannabis and THC across all ages. While adverse effects of cannabis during critical stages of fetal neurodevelopment are investigated, these studies center on neurons alone. Thus, a gap of knowledge exists on how intercellular interactions between neighboring cell types, particularly astrocytes and neurons, could modify THC action. Here, we combine transcriptome analysis, transgenic models, high resolution microscopy and live cell imaging to demonstrate that hippocampal astrocytes accumulate in the strata radiatum and lacunosum moleculare of the CA1 subfield, containing particularly sensitive neurons to stressors, upon long term postnatal THC exposure in vivo. As this altered distribution is not dependent on cell proliferation, we propose that resident astrocytes accumulate in select areas to protect pyramidal neurons and their neurite extensions from pathological damage. Indeed, we could recapitulate the neuroprotective effect of astrocytes in vitro, as their physical presence significantly reduced the death of primary hippocampal neurons upon THC exposure (> 5 µM). Even so, astrocytes are also affected by a reduced metabolic readiness to stressors, as reflected by a downregulation of mitochondrial proteins. Thus, we find that astrocytes exert protective functions on local neurons during THC exposure, even though their mitochondrial electron transport chain is disrupted.


Subject(s)
Astrocytes , Dronabinol , Astrocytes/metabolism , Dronabinol/toxicity , Neurons/metabolism , Hippocampus/metabolism , Pyramidal Cells/metabolism , Cannabinoid Receptor Agonists
6.
Eur J Neurosci ; 55(3): 725-732, 2022 02.
Article in English | MEDLINE | ID: mdl-34978111

ABSTRACT

Experimental investigation of early postnatal brain development in infant mice (

Subject(s)
Brain , Imaging, Three-Dimensional , Animals , Head , Humans , Mice , Printing, Three-Dimensional , Software
8.
Cannabis Cannabinoid Res ; 6(2): 119-136, 2021 04.
Article in English | MEDLINE | ID: mdl-33912677

ABSTRACT

Introduction: In mammals, sn-1-diacylglycerol lipases (DAGL) generate 2-arachidonoylglycerol (2-AG) that, as the major endocannabinoid, modulates synaptic neurotransmission by acting on CB1 cannabinoid receptors (CB1R). Even though the insect genome codes for inaE, which is a DAGL ortholog (dDAGL), its products and their functions remain unknown particularly because insects lack chordate-type cannabinoid receptors. Materials and Methods: Gain-of-function and loss-of-function genetic manipulations were carried out in Drosophila melanogaster, including the generation of both dDAGL-deficient and mammalian CB1R-overexpressing flies. Neuroanatomy, dietary manipulations coupled with targeted mass spectrometry determination of arachidonic acid and 2-linoleoyl glycerol (2-LG) production, behavioral assays, and signal transduction profiling for Akt and Erk kinases were employed. Findings from Drosophilae were validated by a CB1R-binding assay for 2-LG in mammalian cortical homogenates with functionality confirmed in neurons using high-throughput real-time imaging in vitro. Results: In this study, we show that dDAGL is primarily expressed in the brain and nerve cord of Drosophila during larval development and in adult with 2-LG being its chief product as defined by dietary precursor availability. Overexpression of the human CB1R in the ventral nerve cord compromised the mobility of adult Drosophilae. The causality of 2-LG signaling to CB1R-induced behavioral impairments was shown by inaE inactivation normalizing defunct motor coordination. The 2-LG-induced activation of transgenic CB1Rs affected both Akt and Erk kinase cascades by paradoxical signaling. Data from Drosophila models were substantiated by showing 2-LG-mediated displacement of [3H]CP 55,940 in mouse cortical homogenates and reduced neurite extension and growth cone collapsing responses in cultured mouse neurons. Conclusions: Overall, these results suggest that 2-LG is an endocannabinoid-like signal lipid produced by dDAGL in Drosophila.


Subject(s)
Drosophila melanogaster , Lipoprotein Lipase , Animals , Drosophila melanogaster/genetics , Gain of Function Mutation , Glycerol , Lipoprotein Lipase/genetics , Mice , Receptors, Cannabinoid , Signal Transduction/genetics
9.
JCI Insight ; 5(23)2020 12 03.
Article in English | MEDLINE | ID: mdl-33141759

ABSTRACT

Ongoing societal changes in views on the medical and recreational roles of cannabis increased the use of concentrated plant extracts with a Δ9-tetrahydrocannabinol (THC) content of more than 90%. Even though prenatal THC exposure is widely considered adverse for neuronal development, equivalent experimental data for young age cohorts are largely lacking. Here, we administered plant-derived THC (1 or 5 mg/kg) to mice daily during P5-P16 and P5-P35 and monitored its effects on hippocampal neuronal survival and specification by high-resolution imaging and iTRAQ proteomics, respectively. We found that THC indiscriminately affects pyramidal cells and both cannabinoid receptor 1+ (CB1R)+ and CB1R- interneurons by P16. THC particularly disrupted the expression of mitochondrial proteins (complexes I-IV), a change that had persisted even 4 months after the end of drug exposure. This was reflected by a THC-induced loss of membrane integrity occluding mitochondrial respiration and could be partially or completely rescued by pH stabilization, antioxidants, bypassed glycolysis, and targeting either mitochondrial soluble adenylyl cyclase or the mitochondrial voltage-dependent anion channel. Overall, THC exposure during infancy induces significant and long-lasting reorganization of neuronal circuits through mechanisms that, in large part, render cellular bioenergetics insufficient to sustain key developmental processes in otherwise healthy neurons.


Subject(s)
Dronabinol/adverse effects , Neurogenesis/drug effects , Animals , Animals, Newborn , Cell Death/drug effects , Female , Hippocampus/drug effects , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects
11.
Nature ; 582(7811): 246-252, 2020 06.
Article in English | MEDLINE | ID: mdl-32499648

ABSTRACT

A wealth of specialized neuroendocrine command systems intercalated within the hypothalamus control the most fundamental physiological needs in vertebrates1,2. Nevertheless, we lack a developmental blueprint that integrates the molecular determinants of neuronal and glial diversity along temporal and spatial scales of hypothalamus development3. Here we combine single-cell RNA sequencing of 51,199 mouse cells of ectodermal origin, gene regulatory network (GRN) screens in conjunction with genome-wide association study-based disease phenotyping, and genetic lineage reconstruction to show that nine glial and thirty-three neuronal subtypes are generated by mid-gestation under the control of distinct GRNs. Combinatorial molecular codes that arise from neurotransmitters, neuropeptides and transcription factors are minimally required to decode the taxonomical hierarchy of hypothalamic neurons. The differentiation of γ-aminobutyric acid (GABA) and dopamine neurons, but not glutamate neurons, relies on quasi-stable intermediate states, with a pool of GABA progenitors giving rise to dopamine cells4. We found an unexpected abundance of chemotropic proliferation and guidance cues that are commonly implicated in dorsal (cortical) patterning5 in the hypothalamus. In particular, loss of SLIT-ROBO signalling impaired both the production and positioning of periventricular dopamine neurons. Overall, we identify molecular principles that shape the developmental architecture of the hypothalamus and show how neuronal heterogeneity is transformed into a multimodal neural unit to provide virtually infinite adaptive potential throughout life.


Subject(s)
Gene Expression Regulation, Developmental , Hypothalamus/cytology , Hypothalamus/embryology , Morphogenesis , Animals , Cell Differentiation , Cell Lineage , Dopamine/metabolism , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Ectoderm/cytology , Ectoderm/metabolism , Female , GABAergic Neurons/cytology , GABAergic Neurons/metabolism , Gene Regulatory Networks , Genome-Wide Association Study , Glutamic Acid/metabolism , Hypothalamus/metabolism , Male , Mice , Morphogenesis/genetics , Nerve Tissue Proteins/metabolism , Neuroglia/cytology , Neuroglia/metabolism , Neuropeptides/metabolism , Neurotransmitter Agents/metabolism , Receptors, Immunologic/metabolism , Regulon/genetics , Signal Transduction , Transcription Factors/metabolism , gamma-Aminobutyric Acid/metabolism , Roundabout Proteins
12.
Mol Psychiatry ; 25(1): 22-36, 2020 01.
Article in English | MEDLINE | ID: mdl-31735910

ABSTRACT

The evolution of human diets led to preferences toward polyunsaturated fatty acid (PUFA) content with 'Western' diets enriched in ω-6 PUFAs. Mounting evidence points to ω-6 PUFA excess limiting metabolic and cognitive processes that define longevity in humans. When chosen during pregnancy, ω-6 PUFA-enriched 'Western' diets can reprogram maternal bodily metabolism with maternal nutrient supply precipitating the body-wide imprinting of molecular and cellular adaptations at the level of long-range intercellular signaling networks in the unborn fetus. Even though unfavorable neurological outcomes are amongst the most common complications of intrauterine ω-6 PUFA excess, cellular underpinnings of life-long modifications to brain architecture remain unknown. Here, we show that nutritional ω-6 PUFA-derived endocannabinoids desensitize CB1 cannabinoid receptors, thus inducing epigenetic repression of transcriptional regulatory networks controlling neuronal differentiation. We found that cortical neurons lose their positional identity and axonal selectivity when mouse fetuses are exposed to excess ω-6 PUFAs in utero. Conversion of ω-6 PUFAs into endocannabinoids disrupted the temporal precision of signaling at neuronal CB1 cannabinoid receptors, chiefly deregulating Stat3-dependent transcriptional cascades otherwise required to execute neuronal differentiation programs. Global proteomics identified the immunoglobulin family of cell adhesion molecules (IgCAMs) as direct substrates, with DNA methylation and chromatin accessibility profiling uncovering epigenetic reprogramming at >1400 sites in neurons after prolonged cannabinoid exposure. We found anxiety and depression-like behavioral traits to manifest in adult offspring, which is consistent with genetic models of reduced IgCAM expression, to suggest causality for cortical wiring defects. Overall, our data uncover a regulatory mechanism whose disruption by maternal food choices could limit an offspring's brain function for life.


Subject(s)
Brain/drug effects , Diet, Western/adverse effects , Epigenesis, Genetic/drug effects , Animals , Anxiety , Brain/metabolism , DNA Methylation/drug effects , Depression , Diet , Dietary Supplements , Endocannabinoids/metabolism , Epigenesis, Genetic/genetics , Epigenomics/methods , Fatty Acids, Omega-3/metabolism , Fatty Acids, Omega-6/metabolism , Fatty Acids, Unsaturated/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism , Pregnancy , Receptor, Cannabinoid, CB1/drug effects
13.
EMBO J ; 39(1): e100882, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31750562

ABSTRACT

Maternal drug abuse during pregnancy is a rapidly escalating societal problem. Psychostimulants, including amphetamine, cocaine, and methamphetamine, are amongst the illicit drugs most commonly consumed by pregnant women. Neuropharmacology concepts posit that psychostimulants affect monoamine signaling in the nervous system by their affinities to neurotransmitter reuptake and vesicular transporters to heighten neurotransmitter availability extracellularly. Exacerbated dopamine signaling is particularly considered as a key determinant of psychostimulant action. Much less is known about possible adverse effects of these drugs on peripheral organs, and if in utero exposure induces lifelong pathologies. Here, we addressed this question by combining human RNA-seq data with cellular and mouse models of neuroendocrine development. We show that episodic maternal exposure to psychostimulants during pregnancy coincident with the intrauterine specification of pancreatic ß cells permanently impairs their ability of insulin production, leading to glucose intolerance in adult female but not male offspring. We link psychostimulant action specifically to serotonin signaling and implicate the sex-specific epigenetic reprogramming of serotonin-related gene regulatory networks upstream from the transcription factor Pet1/Fev as determinants of reduced insulin production.


Subject(s)
Diabetes Mellitus, Type 2/etiology , Glucose Intolerance/etiology , Glucose/metabolism , Homeostasis/drug effects , Islets of Langerhans/pathology , Methamphetamine/toxicity , Prenatal Exposure Delayed Effects/chemically induced , Animals , Central Nervous System Stimulants/toxicity , DNA Methylation , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Female , Gene Expression Profiling , Gene Expression Regulation , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , Glucose Intolerance/pathology , Humans , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , Maternal Exposure/adverse effects , Mice , Pregnancy , Prenatal Exposure Delayed Effects/genetics , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/pathology
14.
JCI Insight ; 4(4)2019 02 21.
Article in English | MEDLINE | ID: mdl-30830860

ABSTRACT

GPR55, a lipid-sensing receptor, is implicated in cell cycle control, malignant cell mobilization, and tissue invasion in cancer. However, a physiological role for GPR55 is virtually unknown for any tissue type. Here, we localize GPR55 to self-renewing ductal epithelial cells and their terminally differentiated progeny in both human and mouse salivary glands. Moreover, we find GPR55 expression downregulated in salivary gland mucoepidermoid carcinomas and GPR55 reinstatement by antitumor irradiation, suggesting that GPR55 controls renegade proliferation. Indeed, GPR55 antagonism increases cell proliferation and function determination in quasiphysiological systems. In addition, Gpr55-/- mice present ~50% enlarged submandibular glands with many more granulated ducts, as well as disordered endoplasmic reticuli and with glycoprotein content. Next, we hypothesized that GPR55 could also modulate salivation and glycoprotein content by entraining differentiated excretory progeny. Accordingly, GPR55 activation facilitated glycoprotein release by itself, inducing low-amplitude Ca2+ oscillations, as well as enhancing acetylcholine-induced Ca2+ responses. Topical application of GPR55 agonists, which are ineffective in Gpr55-/- mice, into adult rodent submandibular glands increased salivation and saliva glycoprotein content. Overall, we propose that GPR55 signaling in epithelial cells ensures both the life-long renewal of ductal cells and the continuous availability of saliva and glycoproteins for oral health and food intake.


Subject(s)
Adult Stem Cells/physiology , Carcinoma, Mucoepidermoid/pathology , Cell Differentiation/physiology , Receptors, Cannabinoid/metabolism , Salivary Gland Neoplasms/pathology , Salivation/physiology , Adult , Adult Stem Cells/drug effects , Aged , Aged, 80 and over , Animals , Cannabinoid Receptor Agonists/pharmacology , Cannabinoid Receptor Antagonists/pharmacology , Carcinoma, Mucoepidermoid/radiotherapy , Cell Differentiation/drug effects , Cell Self Renewal/drug effects , Cell Self Renewal/physiology , Down-Regulation , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Glycoproteins/metabolism , Humans , Male , Mice , Mice, Knockout , Middle Aged , Receptors, Cannabinoid/genetics , Saliva/chemistry , Saliva/metabolism , Salivary Gland Neoplasms/radiotherapy , Salivation/drug effects , Submandibular Gland/drug effects , Submandibular Gland/metabolism , Submandibular Gland/pathology
15.
Behav Brain Res ; 362: 173-180, 2019 04 19.
Article in English | MEDLINE | ID: mdl-30659847

ABSTRACT

Spatial reference memory is known to be modulated by the dopaminergic system involving different brain regions. Here, we sought to identify the contribution of D1 (D1R) and D2 (D2R)-like dopamine receptor signaling on learning and memory in a food rewarded hole-board task by intracerebroventricular infusing D1R- and D2R- like receptor agonists (SKF-81297 and Sumanirole) and antagonists (SCH 23390 and Remoxipride) once 30 min prior to daily training sessions. D1R agonism induced persistent enhancement of performance, whereas D1R antagonism impaired reference memory formation. D2R agonist and antagonist exerted no effects. Phase specific comparisons revealed an enhancement of spatial acquisition in the presence of the D1R but not D2R agonism on acquisition, but not during retention. Since task difficulty might skew dopamine-induced improvements in learning and memory, we tested the D1R agonist in the hole-board task with increased difficulty. Drug treated animals performed significantly better during all training phases, with results better resolved than in the easy task. Additionally, proteomic analysis of the prefrontal cortex revealed ninety six proteins to be regulated by D1R agonism, from which 35 were correlated with behavioral performance. Obtained targets were grouped by function, showing synaptic transmission, synaptic remodeling, and dendritic spine morphology as the major functional classes affected. In sum, we find that activation of D1R signaling during spatial acquisition and retention improved reference memory index, depended on the task difficulty, and altered the proteome landscape of the prefrontal cortex indicative of massive organizational synaptic restructuring.


Subject(s)
Receptors, Dopamine D1/agonists , Receptors, Dopamine D2/agonists , Spatial Learning/physiology , Spatial Memory/physiology , Animals , Benzazepines/pharmacology , Dendritic Spines/drug effects , Dendritic Spines/metabolism , Dopamine/metabolism , Male , Rats, Sprague-Dawley , Receptors, Dopamine D1/physiology , Receptors, Dopamine D2/physiology , Signal Transduction/drug effects , Spatial Learning/drug effects , Spatial Memory/drug effects , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
16.
EMBO J ; 37(21)2018 11 02.
Article in English | MEDLINE | ID: mdl-30209240

ABSTRACT

Stress-induced cortical alertness is maintained by a heightened excitability of noradrenergic neurons innervating, notably, the prefrontal cortex. However, neither the signaling axis linking hypothalamic activation to delayed and lasting noradrenergic excitability nor the molecular cascade gating noradrenaline synthesis is defined. Here, we show that hypothalamic corticotropin-releasing hormone-releasing neurons innervate ependymal cells of the 3rd ventricle to induce ciliary neurotrophic factor (CNTF) release for transport through the brain's aqueductal system. CNTF binding to its cognate receptors on norepinephrinergic neurons in the locus coeruleus then initiates sequential phosphorylation of extracellular signal-regulated kinase 1 and tyrosine hydroxylase with the Ca2+-sensor secretagogin ensuring activity dependence in both rodent and human brains. Both CNTF and secretagogin ablation occlude stress-induced cortical norepinephrine synthesis, ensuing neuronal excitation and behavioral stereotypes. Cumulatively, we identify a multimodal pathway that is rate-limited by CNTF volume transmission and poised to directly convert hypothalamic activation into long-lasting cortical excitability following acute stress.


Subject(s)
Adrenergic Neurons/metabolism , Ciliary Neurotrophic Factor/metabolism , Hypothalamus/metabolism , Locus Coeruleus/metabolism , Stress, Physiological , Adrenergic Neurons/pathology , Animals , Ciliary Neurotrophic Factor/genetics , Hypothalamus/pathology , Locus Coeruleus/pathology , Mice , Mice, Knockout , Rats
17.
Brain Struct Funct ; 223(3): 1501-1518, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29168008

ABSTRACT

The function, regulation and cellular distribution of GABAA receptor subunits have been extensively documented in the adult rodent brain and are linked to numerous neurological disorders. However, there is a surprising lack of knowledge on the cellular (sub-) distribution of GABAA receptor subunits and of their expressional regulation in developing healthy and diseased foetal human brains. To propose a role for GABAA receptor subunits in neurodevelopmental disorders, we studied the developing hippocampus of normal and Down syndrome foetuses. Among the α1-3 and γ2 subunits probed, we find significantly altered expression profiles of the α1, α3 and γ2 subunits in developing Down syndrome hippocampi, with the α3 subunit being most affected. α3 subunits were selectively down-regulated in all hippocampal subfields and developmental periods tested in Down syndrome foetuses, presenting a developmental mismatch by their adult-like distribution in early foetal development. We hypothesized that increased levels of the amyloid precursor protein (APP), and particularly its neurotoxic ß-amyloid (1-42) fragment, could disrupt α3 gene expression, likely by facilitating premature neuronal differentiation. Indeed, we find increased APP content in the hippocampi of the Down foetuses. In a corresponding cellular model, soluble ß-amyloid (1-42) administered to cultured SH-SY5Y neuroblastoma cells, augmented by retinoic acid-induced differentiation towards a neuronal phenotype, displayed a reduction in α3 subunit levels. In sum, this study charts a comprehensive regional and subcellular map of key GABAA receptor subunits in identified neuronal populations in the hippocampus of healthy and Down syndrome foetuses and associates increased ß-amyloid load with discordant down-regulation of α3 subunits.


Subject(s)
Down Syndrome/pathology , Gene Expression Regulation, Developmental/physiology , Hippocampus/metabolism , Hippocampus/pathology , Protein Subunits/metabolism , Receptors, GABA-A/metabolism , Adolescent , Adult , Age Factors , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/pharmacology , Animals , Animals, Newborn , Case-Control Studies , Doublecortin Domain Proteins , Down Syndrome/genetics , Embryo, Mammalian , Female , Fetus , Gene Expression Regulation, Developmental/drug effects , Gestational Age , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Hippocampus/growth & development , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Middle Aged , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuroblastoma/pathology , Neurons/drug effects , Neurons/metabolism , Neuropeptides/metabolism , POU Domain Factors/genetics , POU Domain Factors/metabolism , Peptide Fragments/pharmacology , Protein Subunits/genetics , Tretinoin/pharmacology , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism , Young Adult
18.
Proc Natl Acad Sci U S A ; 114(10): E2006-E2015, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28223495

ABSTRACT

The rostral migratory stream (RMS) is viewed as a glia-enriched conduit of forward-migrating neuroblasts in which chemorepulsive signals control the pace of forward migration. Here we demonstrate the existence of a scaffold of neurons that receive synaptic inputs within the rat, mouse, and human fetal RMS equivalents. These neurons express secretagogin, a Ca2+-sensor protein, to execute an annexin V-dependent externalization of matrix metalloprotease-2 (MMP-2) for reconfiguring the extracellular matrix locally. Mouse genetics combined with pharmacological probing in vivo and in vitro demonstrate that MMP-2 externalization occurs on demand and that its loss slows neuroblast migration. Loss of function is particularly remarkable upon injury to the olfactory bulb. Cumulatively, we identify a signaling cascade that provokes structural remodeling of the RMS through recruitment of MMP-2 by a previously unrecognized neuronal constituent. Given the life-long presence of secretagogin-containing neurons in human, this mechanism might be exploited for therapeutic benefit in rescue strategies.


Subject(s)
Calcium/metabolism , Matrix Metalloproteinase 2/genetics , Neuroglia/metabolism , Neurons/metabolism , Olfactory Bulb/metabolism , Secretagogins/genetics , Animals , Annexin A5/genetics , Annexin A5/metabolism , Cell Movement , Fetus , Gene Expression Regulation , Humans , Male , Matrix Metalloproteinase 2/metabolism , Mice , Microtomy , Neuroglia/ultrastructure , Neurons/ultrastructure , Olfactory Bulb/cytology , Primary Cell Culture , Rats , Rats, Wistar , Secretagogins/metabolism , Synapses/metabolism , Synapses/ultrastructure , Tissue Culture Techniques
19.
Cereb Cortex ; 27(4): 2453-2468, 2017 04 01.
Article in English | MEDLINE | ID: mdl-27102657

ABSTRACT

Although extensively studied postnatally, the functional differentiation of cholecystokinin (CCK)-containing interneurons en route towards the cerebral cortex during fetal development is incompletely understood. Here, we used CCKBAC/DsRed mice encoding a CCK promoter-driven red fluorescent protein to analyze the temporal dynamics of DsRed expression, neuronal identity, and positioning through high-resolution developmental neuroanatomy. Additionally, we developed a dual reporter mouse line (CCKBAC/DsRed::GAD67gfp/+) to differentiate CCK-containing interneurons from DsRed+ principal cells during prenatal development. We show that DsRed is upregulated in interneurons once they exit their proliferative niche in the ganglionic eminence and remains stably expressed throughout their long-distance migration towards the cerebrum, particularly in the hippocampus. DsRed+ interneurons, including a cohort coexpressing calretinin, accumulated at the palliosubpallial boundary by embryonic day 12.5. Pioneer DsRed+ interneurons already reached deep hippocampal layers by embryonic day 14.5 and were morphologically differentiated by birth. Furthermore, we probed migrating interneurons entering and traversing the cortical plate, as well as stationary cells in the hippocampus by patch-clamp electrophysiology to show the first signs of Na+ and K+ channel activity by embryonic day 12.5 and reliable adult-like excitability by embryonic day 18.5. Cumulatively, this study defines key positional, molecular, and biophysical properties of CCK+ interneurons in the prenatal brain.


Subject(s)
Cell Differentiation/physiology , Cerebral Cortex/cytology , Cholecystokinin/metabolism , Interneurons/cytology , Neurogenesis/physiology , Animals , Cell Movement , Cerebral Cortex/embryology , Cerebral Cortex/metabolism , Immunohistochemistry , In Situ Hybridization , Interneurons/metabolism , Mice , Mice, Transgenic , Microscopy, Confocal , Patch-Clamp Techniques
20.
Nat Neurosci ; 20(2): 176-188, 2017 02.
Article in English | MEDLINE | ID: mdl-27991900

ABSTRACT

The hypothalamus contains the highest diversity of neurons in the brain. Many of these neurons can co-release neurotransmitters and neuropeptides in a use-dependent manner. Investigators have hitherto relied on candidate protein-based tools to correlate behavioral, endocrine and gender traits with hypothalamic neuron identity. Here we map neuronal identities in the hypothalamus by single-cell RNA sequencing. We distinguished 62 neuronal subtypes producing glutamatergic, dopaminergic or GABAergic markers for synaptic neurotransmission and harboring the ability to engage in task-dependent neurotransmitter switching. We identified dopamine neurons that uniquely coexpress the Onecut3 and Nmur2 genes, and placed these in the periventricular nucleus with many synaptic afferents arising from neuromedin S+ neurons of the suprachiasmatic nucleus. These neuroendocrine dopamine cells may contribute to the dopaminergic inhibition of prolactin secretion diurnally, as their neuromedin S+ inputs originate from neurons expressing Per2 and Per3 and their tyrosine hydroxylase phosphorylation is regulated in a circadian fashion. Overall, our catalog of neuronal subclasses provides new understanding of hypothalamic organization and function.


Subject(s)
Dopamine/metabolism , Dopaminergic Neurons/metabolism , Hypothalamus/metabolism , Neuropeptides/metabolism , Tyrosine 3-Monooxygenase/metabolism , Animals , Immunohistochemistry/methods , Mice, Inbred C57BL , Mice, Transgenic , Neurotransmitter Agents/physiology , Suprachiasmatic Nucleus/metabolism , Synaptic Transmission/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...