Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 4.189
1.
Nat Commun ; 15(1): 3443, 2024 Apr 24.
Article En | MEDLINE | ID: mdl-38658557

The hypothalamus contains a remarkable diversity of neurons that orchestrate behavioural and metabolic outputs in a highly plastic manner. Neuronal diversity is key to enabling hypothalamic functions and, according to the neuroscience dogma, it is predetermined during embryonic life. Here, by combining lineage tracing of hypothalamic pro-opiomelanocortin (Pomc) neurons with single-cell profiling approaches in adult male mice, we uncovered subpopulations of 'Ghost' neurons endowed with atypical molecular and functional identity. Compared to 'classical' Pomc neurons, Ghost neurons exhibit negligible Pomc expression and are 'invisible' to available neuroanatomical approaches and promoter-based reporter mice for studying Pomc biology. Ghost neuron numbers augment in diet-induced obese mice, independent of neurogenesis or cell death, but weight loss can reverse this shift. Our work challenges the notion of fixed, developmentally programmed neuronal identities in the mature hypothalamus and highlight the ability of specialised neurons to reversibly adapt their functional identity to adult-onset obesogenic stimuli.


Hypothalamus , Neurons , Obesity , Pro-Opiomelanocortin , Single-Cell Analysis , Animals , Pro-Opiomelanocortin/metabolism , Pro-Opiomelanocortin/genetics , Neurons/metabolism , Obesity/metabolism , Obesity/pathology , Male , Mice , Hypothalamus/metabolism , Hypothalamus/cytology , Disease Models, Animal , Diet, High-Fat , Mice, Inbred C57BL , Mice, Transgenic , Neurogenesis , Mice, Obese
2.
Nature ; 628(8009): 826-834, 2024 Apr.
Article En | MEDLINE | ID: mdl-38538787

Empirical evidence suggests that heat exposure reduces food intake. However, the neurocircuit architecture and the signalling mechanisms that form an associative interface between sensory and metabolic modalities remain unknown, despite primary thermoceptive neurons in the pontine parabrachial nucleus becoming well characterized1. Tanycytes are a specialized cell type along the wall of the third ventricle2 that bidirectionally transport hormones and signalling molecules between the brain's parenchyma and ventricular system3-8. Here we show that tanycytes are activated upon acute thermal challenge and are necessary to reduce food intake afterwards. Virus-mediated gene manipulation and circuit mapping showed that thermosensing glutamatergic neurons of the parabrachial nucleus innervate tanycytes either directly or through second-order hypothalamic neurons. Heat-dependent Fos expression in tanycytes suggested their ability to produce signalling molecules, including vascular endothelial growth factor A (VEGFA). Instead of discharging VEGFA into the cerebrospinal fluid for a systemic effect, VEGFA was released along the parenchymal processes of tanycytes in the arcuate nucleus. VEGFA then increased the spike threshold of Flt1-expressing dopamine and agouti-related peptide (Agrp)-containing neurons, thus priming net anorexigenic output. Indeed, both acute heat and the chemogenetic activation of glutamatergic parabrachial neurons at thermoneutrality reduced food intake for hours, in a manner that is sensitive to both Vegfa loss-of-function and blockage of vesicle-associated membrane protein 2 (VAMP2)-dependent exocytosis from tanycytes. Overall, we define a multimodal neurocircuit in which tanycytes link parabrachial sensory relay to the long-term enforcement of a metabolic code.


Brain Stem , Ependymoglial Cells , Feeding Behavior , Hot Temperature , Hypothalamus , Neural Pathways , Neurons , Animals , Female , Male , Mice , Agouti-Related Protein/metabolism , Arcuate Nucleus of Hypothalamus/metabolism , Arcuate Nucleus of Hypothalamus/cytology , Brain Stem/cytology , Brain Stem/physiology , Dopamine/metabolism , Eating/physiology , Ependymoglial Cells/cytology , Ependymoglial Cells/physiology , Feeding Behavior/physiology , Glutamic Acid/metabolism , Hypothalamus/cytology , Hypothalamus/physiology , Neural Pathways/metabolism , Neurons/metabolism , Parabrachial Nucleus/cytology , Parabrachial Nucleus/metabolism , Parabrachial Nucleus/physiology , Thermosensing/physiology , Time Factors , Vascular Endothelial Growth Factor A/cerebrospinal fluid , Vascular Endothelial Growth Factor A/metabolism
3.
Nature ; 626(7998): 347-356, 2024 Feb.
Article En | MEDLINE | ID: mdl-38267576

To survive in a complex social group, one needs to know who to approach and, more importantly, who to avoid. In mice, a single defeat causes the losing mouse to stay away from the winner for weeks1. Here through a series of functional manipulation and recording experiments, we identify oxytocin neurons in the retrochiasmatic supraoptic nucleus (SOROXT) and oxytocin-receptor-expressing cells in the anterior subdivision of the ventromedial hypothalamus, ventrolateral part (aVMHvlOXTR) as a key circuit motif for defeat-induced social avoidance. Before defeat, aVMHvlOXTR cells minimally respond to aggressor cues. During defeat, aVMHvlOXTR cells are highly activated and, with the help of an exclusive oxytocin supply from the SOR, potentiate their responses to aggressor cues. After defeat, strong aggressor-induced aVMHvlOXTR cell activation drives the animal to avoid the aggressor and minimizes future defeat. Our study uncovers a neural process that supports rapid social learning caused by defeat and highlights the importance of the brain oxytocin system in social plasticity.


Aggression , Avoidance Learning , Hypothalamus , Neural Pathways , Neurons , Oxytocin , Social Learning , Animals , Mice , Aggression/physiology , Avoidance Learning/physiology , Cues , Fear/physiology , Hypothalamus/cytology , Hypothalamus/metabolism , Neural Pathways/physiology , Neurons/metabolism , Oxytocin/metabolism , Receptors, Oxytocin/metabolism , Social Behavior , Social Learning/physiology , Supraoptic Nucleus/cytology , Supraoptic Nucleus/metabolism , Ventromedial Hypothalamic Nucleus/cytology , Ventromedial Hypothalamic Nucleus/metabolism , Neuronal Plasticity
4.
Nature ; 624(7991): 333-342, 2023 Dec.
Article En | MEDLINE | ID: mdl-38092915

The function of the mammalian brain relies upon the specification and spatial positioning of diversely specialized cell types. Yet, the molecular identities of the cell types and their positions within individual anatomical structures remain incompletely known. To construct a comprehensive atlas of cell types in each brain structure, we paired high-throughput single-nucleus RNA sequencing with Slide-seq1,2-a recently developed spatial transcriptomics method with near-cellular resolution-across the entire mouse brain. Integration of these datasets revealed the cell type composition of each neuroanatomical structure. Cell type diversity was found to be remarkably high in the midbrain, hindbrain and hypothalamus, with most clusters requiring a combination of at least three discrete gene expression markers to uniquely define them. Using these data, we developed a framework for genetically accessing each cell type, comprehensively characterized neuropeptide and neurotransmitter signalling, elucidated region-specific specializations in activity-regulated gene expression and ascertained the heritability enrichment of neurological and psychiatric phenotypes. These data, available as an online resource ( www.BrainCellData.org ), should find diverse applications across neuroscience, including the construction of new genetic tools and the prioritization of specific cell types and circuits in the study of brain diseases.


Brain , Gene Expression Profiling , Animals , Mice , Brain/anatomy & histology , Brain/cytology , Brain/metabolism , Gene Expression Profiling/methods , High-Throughput Nucleotide Sequencing , Hypothalamus/cytology , Hypothalamus/metabolism , Mesencephalon/cytology , Mesencephalon/metabolism , Neuropeptides/metabolism , Neurotransmitter Agents/metabolism , Phenotype , Rhombencephalon/cytology , Rhombencephalon/metabolism , Single-Cell Gene Expression Analysis , Transcriptome/genetics
5.
Nature ; 624(7991): 355-365, 2023 Dec.
Article En | MEDLINE | ID: mdl-38092919

Single-cell analyses parse the brain's billions of neurons into thousands of 'cell-type' clusters residing in different brain structures1. Many cell types mediate their functions through targeted long-distance projections allowing interactions between specific cell types. Here we used epi-retro-seq2 to link single-cell epigenomes and cell types to long-distance projections for 33,034 neurons dissected from 32 different regions projecting to 24 different targets (225 source-to-target combinations) across the whole mouse brain. We highlight uses of these data for interrogating principles relating projection types to transcriptomics and epigenomics, and for addressing hypotheses about cell types and connections related to genetics. We provide an overall synthesis with 926 statistical comparisons of discriminability of neurons projecting to each target for every source. We integrate this dataset into the larger BRAIN Initiative Cell Census Network atlas, composed of millions of neurons, to link projection cell types to consensus clusters. Integration with spatial transcriptomics further assigns projection-enriched clusters to smaller source regions than the original dissections. We exemplify this by presenting in-depth analyses of projection neurons from the hypothalamus, thalamus, hindbrain, amygdala and midbrain to provide insights into properties of those cell types, including differentially expressed genes, their associated cis-regulatory elements and transcription-factor-binding motifs, and neurotransmitter use.


Brain , Epigenomics , Neural Pathways , Neurons , Animals , Mice , Amygdala , Brain/cytology , Brain/metabolism , Consensus Sequence , Datasets as Topic , Gene Expression Profiling , Hypothalamus/cytology , Mesencephalon/cytology , Neural Pathways/cytology , Neurons/metabolism , Neurotransmitter Agents/metabolism , Regulatory Sequences, Nucleic Acid , Rhombencephalon/cytology , Single-Cell Analysis , Thalamus/cytology , Transcription Factors/metabolism
6.
Science ; 382(6669): 399-404, 2023 10 27.
Article En | MEDLINE | ID: mdl-37883550

Sexual, parental, and aggressive behaviors are central to the reproductive success of individuals and species survival and thus are supported by hardwired neural circuits. The reproductive behavior control column (RBCC), which comprises the medial preoptic nucleus (MPN), the ventrolateral part of the ventromedial hypothalamus (VMHvl), and the ventral premammillary nucleus (PMv), is essential for all social behaviors. The RBCC integrates diverse hormonal and metabolic cues and adjusts an animal's physical activity, hence the chance of social encounters. The RBCC further engages the mesolimbic dopamine system to maintain social interest and reinforces cues and actions that are time-locked with social behaviors. We propose that the RBCC and brainstem form a dual-control system for generating moment-to-moment social actions. This Review summarizes recent progress regarding the identities of RBCC cells and their pathways that drive different aspects of social behaviors.


Hypothalamus , Social Behavior , Animals , Aggression/physiology , Hypothalamus/cytology , Hypothalamus/physiology , Sexual Behavior/physiology , Male , Female , Maternal Behavior/physiology , Paternal Behavior/physiology , Consummatory Behavior
7.
Science ; 382(6669): 405-412, 2023 10 27.
Article En | MEDLINE | ID: mdl-37883555

Neural substrates of wakefulness, rapid eye movement sleep (REMS), and non-REMS (NREMS) in the mammalian hypothalamus overlap both anatomically and functionally with cellular networks that support physiological and behavioral homeostasis. Here, we review the roles of sleep neurons of the hypothalamus in the homeostatic control of thermoregulation or goal-oriented behaviors during wakefulness. We address how hypothalamic circuits involved in opposing behaviors such as core body temperature and sleep compute conflicting information and provide a coherent vigilance state. Finally, we highlight some of the key unresolved questions and challenges, and the promise of a more granular view of the cellular and molecular diversity underlying the integrative role of the hypothalamus in physiological and behavioral homeostasis.


Hypothalamus , Neurons , Sleep, REM , Sleep, Slow-Wave , Wakefulness , Animals , Body Temperature Regulation , Electroencephalography , Hypothalamus/cytology , Hypothalamus/physiology , Sleep, REM/physiology , Wakefulness/physiology , Humans , Neurons/physiology , Sleep, Slow-Wave/physiology
8.
Nature ; 621(7980): 788-795, 2023 Sep.
Article En | MEDLINE | ID: mdl-37730989

Oxytocin is a neuropeptide that is important for maternal physiology and childcare, including parturition and milk ejection during nursing1-6. Suckling triggers the release of oxytocin, but other sensory cues-specifically, infant cries-can increase the levels of oxytocin in new human mothers7, which indicates that cries can activate hypothalamic oxytocin neurons. Here we describe a neural circuit that routes auditory information about infant vocalizations to mouse oxytocin neurons. We performed in vivo electrophysiological recordings and photometry from identified oxytocin neurons in awake maternal mice that were presented with pup calls. We found that oxytocin neurons responded to pup vocalizations, but not to pure tones, through input from the posterior intralaminar thalamus, and that repetitive thalamic stimulation induced lasting disinhibition of oxytocin neurons. This circuit gates central oxytocin release and maternal behaviour in response to calls, providing a mechanism for the integration of sensory cues from the offspring in maternal endocrine networks to ensure modulation of brain state for efficient parenting.


Maternal Behavior , Neural Pathways , Neurons , Oxytocin , Vocalization, Animal , Animals , Female , Mice , Cues , Hypothalamus/cytology , Hypothalamus/physiology , Maternal Behavior/physiology , Neurons/metabolism , Oxytocin/metabolism , Photometry , Thalamic Nuclei/physiology , Vocalization, Animal/physiology , Wakefulness
9.
Cell ; 186(6): 1195-1211.e19, 2023 03 16.
Article En | MEDLINE | ID: mdl-36796363

Social interactions require awareness and understanding of the behavior of others. Mirror neurons, cells representing an action by self and others, have been proposed to be integral to the cognitive substrates that enable such awareness and understanding. Mirror neurons of the primate neocortex represent skilled motor tasks, but it is unclear if they are critical for the actions they embody, enable social behaviors, or exist in non-cortical regions. We demonstrate that the activity of individual VMHvlPR neurons in the mouse hypothalamus represents aggression performed by self and others. We used a genetically encoded mirror-TRAP strategy to functionally interrogate these aggression-mirroring neurons. We find that their activity is essential for fighting and that forced activation of these cells triggers aggressive displays by mice, even toward their mirror image. Together, we have discovered a mirroring center in an evolutionarily ancient region that provides a subcortical cognitive substrate essential for a social behavior.


Aggression , Hypothalamus , Mirror Neurons , Animals , Mice , Aggression/physiology , Hypothalamus/cytology , Social Behavior
10.
Nature ; 612(7940): 512-518, 2022 12.
Article En | MEDLINE | ID: mdl-36477539

Progress has been made in the elucidation of sleep and wakefulness regulation at the neurocircuit level1,2. However, the intracellular signalling pathways that regulate sleep and the neuron groups in which these intracellular mechanisms work remain largely unknown. Here, using a forward genetics approach in mice, we identify histone deacetylase 4 (HDAC4) as a sleep-regulating molecule. Haploinsufficiency of Hdac4, a substrate of salt-inducible kinase 3 (SIK3)3, increased sleep. By contrast, mice that lacked SIK3 or its upstream kinase LKB1 in neurons or with a Hdac4S245A mutation that confers resistance to phosphorylation by SIK3 showed decreased sleep. These findings indicate that LKB1-SIK3-HDAC4 constitute a signalling cascade that regulates sleep and wakefulness. We also performed targeted manipulation of SIK3 and HDAC4 in specific neurons and brain regions. This showed that SIK3 signalling in excitatory neurons located in the cerebral cortex and the hypothalamus positively regulates EEG delta power during non-rapid eye movement sleep (NREMS) and NREMS amount, respectively. A subset of transcripts biased towards synaptic functions was commonly regulated in cortical glutamatergic neurons through the expression of a gain-of-function allele of Sik3 and through sleep deprivation. These findings suggest that NREMS quantity and depth are regulated by distinct groups of excitatory neurons through common intracellular signals. This study provides a basis for linking intracellular events and circuit-level mechanisms that control NREMS.


Neurons , Sleep Duration , Sleep , Wakefulness , Animals , Mice , Electroencephalography , Neurons/metabolism , Neurons/physiology , Sleep/genetics , Sleep/physiology , Sleep Deprivation/genetics , Wakefulness/genetics , Wakefulness/physiology , Signal Transduction , Delta Rhythm , Cerebral Cortex/cytology , Cerebral Cortex/physiology , Hypothalamus/cytology , Hypothalamus/physiology , Glutamic Acid/metabolism , Sleep, Slow-Wave/genetics , Sleep, Slow-Wave/physiology
11.
Mol Metab ; 66: 101604, 2022 12.
Article En | MEDLINE | ID: mdl-36184065

OBJECTIVE: Insulin-like peptide 5 (INSL5) signalling, through its cognate receptor relaxin/insulin-like family peptide receptor 4 (RXFP4), has been reported to be orexigenic, and the high fat diet (HFD) preference observed in wildtype mice is altered in Rxfp4 knock-out mice. In this study, we used a new Rxfp4-Cre mouse model to investigate the mechanisms underlying these observations. METHODS: We generated transgenic Rxfp4-Cre mice and investigated central expression of Rxfp4 by RT-qPCR, RNAscope and intraparenchymal infusion of INSL5. Rxfp4-expressing cells were chemogenetically manipulated in global Cre-reporter mice using designer receptors exclusively activated by designer drugs (DREADDs) or after stereotactic injection of a Cre-dependent AAV-DIO-Dq-DREADD targeting a population located in the ventromedial hypothalamus (RXFP4VMH). Food intake and feeding motivation were assessed in the presence and absence of a DREADD agonist. Rxfp4-expressing cells in the hypothalamus were characterised by single-cell RNA-sequencing (scRNAseq) and the connectivity of RXFP4VMH cells was investigated using viral tracing. RESULTS: Rxfp4-Cre mice displayed Cre-reporter expression in the hypothalamus. Active expression of Rxfp4 in the adult mouse brain was confirmed by RT-qPCR and RNAscope. Functional receptor expression was supported by cyclic AMP-responses to INSL5 application in ex vivo brain slices and increased HFD and highly palatable liquid meal (HPM), but not chow, intake after intra-VMH INSL5 infusion. scRNAseq of hypothalamic RXFP4 neurons defined a cluster expressing VMH markers, alongside known appetite-modulating neuropeptide receptors (Mc4r, Cckar and Nmur2). Viral tracing demonstrated RXFP4VMH neural projections to nuclei implicated in hedonic feeding behaviour. Whole body chemogenetic inhibition (Di-DREADD) of Rxfp4-expressing cells, mimicking physiological INSL5-RXFP4 Gi-signalling, increased intake of the HFD and HPM, but not chow, whilst activation (Dq-DREADD), either at whole body level or specifically within the VMH, reduced HFD and HPM intake and motivation to work for the HPM. CONCLUSION: These findings identify RXFP4VMH neurons as regulators of food intake and preference, and hypothalamic RXFP4 signalling as a target for feeding behaviour manipulation.


Eating , Neurons , Receptors, G-Protein-Coupled , Animals , Mice , Hypothalamus/cytology , Hypothalamus/metabolism , Neurons/metabolism , Receptors, G-Protein-Coupled/metabolism
12.
Nature ; 608(7924): 741-749, 2022 08.
Article En | MEDLINE | ID: mdl-35922505

Mating and aggression are innate social behaviours that are controlled by subcortical circuits in the extended amygdala and hypothalamus1-4. The bed nucleus of the stria terminalis (BNSTpr) is a node that receives input encoding sex-specific olfactory cues from the medial amygdala5,6, and which in turn projects to hypothalamic nuclei that control mating7-9 (medial preoptic area (MPOA)) and aggression9-14 (ventromedial hypothalamus, ventrolateral subdivision (VMHvl)), respectively15. Previous studies have demonstrated that male aromatase-positive BNSTpr neurons are required for mounting and attack, and may identify conspecific sex according to their overall level of activity16. However, neural representations in BNSTpr, their function and their transformations in the hypothalamus have not been characterized. Here we performed calcium imaging17,18 of male BNSTprEsr1 neurons during social behaviours. We identify distinct populations of female- versus male-tuned neurons in BNSTpr, with the former outnumbering the latter by around two to one, similar to the medial amygdala and MPOA but opposite to VMHvl, in which male-tuned neurons predominate6,9,19. Chemogenetic silencing of BNSTprEsr1 neurons while imaging MPOAEsr1 or VMHvlEsr1 neurons in behaving animals showed, unexpectedly, that the male-dominant sex-tuning bias in VMHvl was inverted to female-dominant whereas a switch from sniff- to mount-selective neurons during mating was attenuated in MPOA. Our data also indicate that BNSTprEsr1 neurons are not essential for conspecific sex identification. Rather, they control the transition from appetitive to consummatory phases of male social behaviours by shaping sex- and behaviour-specific neural representations in the hypothalamus.


Sexual Behavior, Animal , Social Behavior , Aggression/physiology , Amygdala/cytology , Amygdala/physiology , Animals , Calcium/analysis , Calcium/metabolism , Female , Hypothalamus/cytology , Hypothalamus/physiology , Male , Neurons/physiology , Preoptic Area/cytology , Preoptic Area/physiology , Sex Characteristics , Sexual Behavior, Animal/physiology
13.
Nature ; 608(7921): 146-152, 2022 08.
Article En | MEDLINE | ID: mdl-35831500

Social affiliation emerges from individual-level behavioural rules that are driven by conspecific signals1-5. Long-distance attraction and short-distance repulsion, for example, are rules that jointly set a preferred interanimal distance in swarms6-8. However, little is known about their perceptual mechanisms and executive neural circuits3. Here we trace the neuronal response to self-like biological motion9,10, a visual trigger for affiliation in developing zebrafish2,11. Unbiased activity mapping and targeted volumetric two-photon calcium imaging revealed 21 activity hotspots distributed throughout the brain as well as clustered biological-motion-tuned neurons in a multimodal, socially activated nucleus of the dorsal thalamus. Individual dorsal thalamus neurons encode local acceleration of visual stimuli mimicking typical fish kinetics but are insensitive to global or continuous motion. Electron microscopic reconstruction of dorsal thalamus neurons revealed synaptic input from the optic tectum and projections into hypothalamic areas with conserved social function12-14. Ablation of the optic tectum or dorsal thalamus selectively disrupted social attraction without affecting short-distance repulsion. This tectothalamic pathway thus serves visual recognition of conspecifics, and dissociates neuronal control of attraction from repulsion during social affiliation, revealing a circuit underpinning collective behaviour.


Crowding , Neurons , Social Behavior , Superior Colliculi , Thalamus , Visual Pathways , Zebrafish , Animals , Brain Mapping , Calcium/analysis , Hypothalamus/cytology , Hypothalamus/physiology , Locomotion , Microscopy, Electron , Neurons/cytology , Neurons/physiology , Neurons/ultrastructure , Pattern Recognition, Visual , Photic Stimulation , Superior Colliculi/cytology , Superior Colliculi/physiology , Thalamus/cytology , Thalamus/physiology , Visual Pathways/cytology , Visual Pathways/physiology , Visual Pathways/ultrastructure , Zebrafish/physiology
14.
Nature ; 608(7922): 374-380, 2022 08.
Article En | MEDLINE | ID: mdl-35831501

Food and water are rewarding in part because they satisfy our internal needs1,2. Dopaminergic neurons in the ventral tegmental area (VTA) are activated by gustatory rewards3-5, but how animals learn to associate these oral cues with the delayed physiological effects of ingestion is unknown. Here we show that individual dopaminergic neurons in the VTA respond to detection of nutrients or water at specific stages of ingestion. A major subset of dopaminergic neurons tracks changes in systemic hydration that occur tens of minutes after thirsty mice drink water, whereas different dopaminergic neurons respond to nutrients in the gastrointestinal tract. We show that information about fluid balance is transmitted to the VTA by a hypothalamic pathway and then re-routed to downstream circuits that track the oral, gastrointestinal and post-absorptive stages of ingestion. To investigate the function of these signals, we used a paradigm in which a fluid's oral and post-absorptive effects can be independently manipulated and temporally separated. We show that mice rapidly learn to prefer one fluid over another based solely on its rehydrating ability and that this post-ingestive learning is prevented if dopaminergic neurons in the VTA are selectively silenced after consumption. These findings reveal that the midbrain dopamine system contains subsystems that track different modalities and stages of ingestion, on timescales from seconds to tens of minutes, and that this information is used to drive learning about the consequences of ingestion.


Dopamine , Dopaminergic Neurons , Hypothalamus , Neural Pathways , Nutrients , Organism Hydration Status , Ventral Tegmental Area , Animals , Cues , Digestion , Dopamine/metabolism , Dopaminergic Neurons/physiology , Eating , Gastrointestinal Tract/metabolism , Hypothalamus/cytology , Hypothalamus/physiology , Mesencephalon/cytology , Mesencephalon/physiology , Mice , Nutrients/metabolism , Organism Hydration Status/drug effects , Reward , Time Factors , Ventral Tegmental Area/cytology , Ventral Tegmental Area/physiology , Water/metabolism , Water/pharmacology , Water-Electrolyte Balance
15.
Biochem Biophys Res Commun ; 599: 134-141, 2022 04 09.
Article En | MEDLINE | ID: mdl-35182939

Metabolic abnormalities are tightly connected to the perturbation of normal brain functions, thereby causing multiple neurodegenerative diseases. The hypothalamus is the master unit that controls the whole-body energy homeostasis. Thus, altered metabolic activity in the hypothalamus could be a crucial clue to better understand the development of metabolic disorders during aging. The current study aimed to investigate the changes in hypothalamic metabolites according to the aging process using gas chromatography-mass spectrometry. We identified that multiple metabolites and neurotransmitters were effectively reduced in the hypothalamus of aged mice. In addition, we observed increased levels of genes linked to the production and utilization of monocarboxylates in the aged hypothalamus, indicating the initiation of metabolic activity to produce alternative nutrient sources. Lastly, we found a reduced number of astrocytes in the hypothalamus of aged mice, suggesting that reduced nutrient availability in the hypothalamus might be associated with the decreased activity of astrocytes during aging. Collectively, the present study suggests that the deterioration of metabolic activities in the hypothalamus might be a primary cause and/or outcome of metabolic diseases associated with the aging process.


Aging/metabolism , Hypothalamus/metabolism , Metabolome/physiology , Animals , Astrocytes/metabolism , Blood/metabolism , Gene Expression Regulation , Glial Fibrillary Acidic Protein/immunology , Hypothalamus/cytology , Hypothalamus/physiology , Immunohistochemistry/methods , Male , Mice, Inbred C57BL
16.
Reprod Toxicol ; 108: 1-9, 2022 03.
Article En | MEDLINE | ID: mdl-34974146

Tamoxifen, a selective non-steroidal estrogen receptor modulator, is the standard adjuvant endocrine treatment for breast cancer. Since information on the risk of using tamoxifen during pregnancy is still scarce, this study evaluated whether the in utero and lactational treatment with this drug could compromise reproductive and behavioural parameters in male offspring. Pregnant Wistar rats were exposed to three doses of tamoxifen (0.12; 0.6; 3 µg/kg), by gavage, from gestational day 15 to lactational day 20. Tamoxifen exposure did not alter the anogenital distance in the male offspring; however, there was a significant increase in the body weight in the 0.12 µg/kg dose and a decrease in the 0.6 µg/kg dose. The male offspring treated with the highest dose exhibited a delay in the onset of puberty, evidenced by an increase in the age of preputial separation. Regarding sperm parameters, there was an increase in the sperm count in the cauda epididymis in the intermediate and highest dose groups, in addition to an increase in the number of static sperm and a decrease in the progressive sperm in the same groups. Moreover, an increase in the number of hyperplasia of the epithelial clear cells was observed in the epididymis. In conclusion, the present study demonstrated that maternal exposure to tamoxifen compromised the installation of puberty of the male offspring and the maturation of the epididymis, affecting sperm storage and motility in the adult life.


Behavior, Animal/drug effects , Prenatal Exposure Delayed Effects/chemically induced , Selective Estrogen Receptor Modulators/toxicity , Spermatozoa/drug effects , Tamoxifen/toxicity , Animals , Epididymis/drug effects , Epididymis/growth & development , Female , Hypothalamus/cytology , Lactation , Male , Maternal-Fetal Exchange , Neurons/drug effects , Neurons/metabolism , Pregnancy , Rats, Wistar , Receptors, Androgen/metabolism , Sexual Maturation/drug effects , Sperm Count , Sperm Motility/drug effects , Spermatozoa/physiology
17.
Mol Cell Endocrinol ; 539: 111480, 2022 01 01.
Article En | MEDLINE | ID: mdl-34624438

Bisphenol A (BPA) is an endocrine disrupting chemical that promotes obesity. It acts on the hypothalamus by increasing expression of the orexigenic neuropeptides, Npy and Agrp. Exactly how BPA dysregulates energy homeostasis is not completely clear. Since microRNAs (miRNA) have emerged as crucial weight regulators, the question of whether BPA could alter hypothalamic miRNA profiles was examined. Treatment of the mHypoA-59 cell line with 100 µM BPA altered a specific subset of miRNAs, and the most upregulated was miR-708-5p. BPA was found to increase the levels of miR-708-5p, and its parent gene Odz4, through the ER stress-related protein Chop. Overexpression of an miR-708-5p mimic resulted in a reduction of neuronatin, a proteolipid whose loss of expression is associated with obesity, and an increase in orexigenic Npy expression, thus potentially increasing feeding through converging regulatory pathways. Therefore, hypothalamic exposure to BPA can increase miR-708-5p that controls neuropeptides directly linked to obesity.


Benzhydryl Compounds/adverse effects , Endoplasmic Reticulum Stress/drug effects , Hypothalamus/cytology , Membrane Proteins/genetics , MicroRNAs/genetics , Nerve Tissue Proteins/genetics , Neuropeptide Y/genetics , Phenols/adverse effects , Animals , Cells, Cultured , Female , Gene Expression Regulation/drug effects , Hypothalamus/drug effects , Hypothalamus/metabolism , Male , Mice , Models, Biological , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Transcription Factor CHOP/metabolism , Up-Regulation
18.
Endocrinology ; 163(1)2022 01 01.
Article En | MEDLINE | ID: mdl-34919671

MicroRNAs (miRNAs) expressed in the hypothalamus are capable of regulating energy balance and peripheral metabolism by inhibiting translation of target messenger RNAs (mRNAs). Hypothalamic insulin resistance is known to precede that in the periphery, thus a critical unanswered question is whether central insulin resistance creates a specific hypothalamic miRNA signature that can be identified and targeted. Here we show that miR-1983, a unique miRNA, is upregulated in vitro in 2 insulin-resistant immortalized hypothalamic neuronal neuropeptide Y-expressing models, and in vivo in hyperinsulinemic mice, with a concomitant decrease of insulin receptor ß subunit protein, a target of miR-1983. Importantly, we demonstrate that miR-1983 is detectable in human blood serum and that its levels significantly correlate with blood insulin and the homeostatic model assessment of insulin resistance. Levels of miR-1983 are normalized with metformin exposure in mouse hypothalamic neuronal cell culture. Our findings provide evidence for miR-1983 as a unique biomarker of cellular insulin resistance, and a potential therapeutic target for prevention of human metabolic disease.


Hypothalamus/metabolism , Insulin/pharmacology , Metformin/pharmacology , MicroRNAs/genetics , Receptor, Insulin/genetics , Adult , Animals , Cell Line , Cells, Cultured , Female , Gene Expression Profiling/methods , Gene Expression Regulation/drug effects , Humans , Hypoglycemic Agents/pharmacology , Hypothalamus/cytology , Insulin/blood , Insulin/metabolism , Insulin Resistance/genetics , Male , Mice , MicroRNAs/blood , Middle Aged , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Obesity/blood , Obesity/genetics , Obesity/metabolism , Receptor, Insulin/metabolism
19.
Nat Commun ; 12(1): 6749, 2021 11 19.
Article En | MEDLINE | ID: mdl-34799566

The hypothalamus regulates metabolic homeostasis by influencing behavior and endocrine systems. Given its role governing key traits, such as body weight and reproductive timing, understanding the genetic regulation of hypothalamic development and function could yield insights into disease pathogenesis. However, given its inaccessibility, studying human hypothalamic gene regulation has proven challenging. To address this gap, we generate a high-resolution chromatin architecture atlas of an established embryonic stem cell derived hypothalamic-like neuron model across three stages of in vitro differentiation. We profile accessible chromatin and identify physical contacts between gene promoters and putative cis-regulatory elements to characterize global regulatory landscape changes during hypothalamic differentiation. Next, we integrate these data with GWAS loci for various complex traits, identifying multiple candidate effector genes. Our results reveal common target genes for these traits, potentially affecting core developmental pathways. Our atlas will enable future efforts to determine hypothalamic mechanisms influencing disease susceptibility.


Gene Expression Regulation, Developmental , Gene Regulatory Networks , Human Embryonic Stem Cells/physiology , Hypothalamus/embryology , Neurons/physiology , Cell Differentiation/genetics , Cell Line , Chromosome Mapping , Genome-Wide Association Study , Humans , Hypothalamus/cytology , Multifactorial Inheritance , RNA-Seq , Regulatory Elements, Transcriptional/genetics
20.
Nature ; 599(7885): 436-441, 2021 11.
Article En | MEDLINE | ID: mdl-34732894

The state of somatic energy stores in metazoans is communicated to the brain, which regulates key aspects of behaviour, growth, nutrient partitioning and development1. The central melanocortin system acts through melanocortin 4 receptor (MC4R) to control appetite, food intake and energy expenditure2. Here we present evidence that MC3R regulates the timing of sexual maturation, the rate of linear growth and the accrual of lean mass, which are all energy-sensitive processes. We found that humans who carry loss-of-function mutations in MC3R, including a rare homozygote individual, have a later onset of puberty. Consistent with previous findings in mice, they also had reduced linear growth, lean mass and circulating levels of IGF1. Mice lacking Mc3r had delayed sexual maturation and an insensitivity of reproductive cycle length to nutritional perturbation. The expression of Mc3r is enriched in hypothalamic neurons that control reproduction and growth, and expression increases during postnatal development in a manner that is consistent with a role in the regulation of sexual maturation. These findings suggest a bifurcating model of nutrient sensing by the central melanocortin pathway with signalling through MC4R controlling the acquisition and retention of calories, whereas signalling through MC3R primarily regulates the disposition of calories into growth, lean mass and the timing of sexual maturation.


Child Development/physiology , Nutritional Status/physiology , Puberty/physiology , Receptor, Melanocortin, Type 3/metabolism , Sexual Maturation/physiology , Adolescent , Aged, 80 and over , Animals , Child , Estrous Cycle/genetics , Estrous Cycle/physiology , Female , Homozygote , Humans , Hypothalamus/cytology , Hypothalamus/physiology , Insulin-Like Growth Factor I/metabolism , Male , Melanocortins/metabolism , Menarche/genetics , Menarche/physiology , Mice , Phenotype , Puberty/genetics , Receptor, Melanocortin, Type 3/deficiency , Receptor, Melanocortin, Type 3/genetics , Sexual Maturation/genetics , Time Factors , Weight Gain
...