Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
EMBO Rep ; 25(1): 351-377, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38177913

ABSTRACT

Hypothalamic Adult Neurogenesis (hAN) has been implicated in regulating energy homeostasis. Adult-generated neurons and adult Neural Stem Cells (aNSCs) in the hypothalamus control food intake and body weight. Conversely, diet-induced obesity (DIO) by high fat diets (HFD) exerts adverse influence on hAN. However, the effects of anti-obesity compounds on hAN are not known. To address this, we administered a lipidized analogue of an anti-obesity neuropeptide, Prolactin Releasing Peptide (PrRP), so-called LiPR, to mice. In the HFD context, LiPR rescued the survival of adult-born hypothalamic neurons and increased the number of aNSCs by reducing their activation. LiPR also rescued the reduction of immature hippocampal neurons and modulated calcium dynamics in iPSC-derived human neurons. In addition, some of these neurogenic effects were exerted by another anti-obesity compound, Liraglutide. These results show for the first time that anti-obesity neuropeptides influence adult neurogenesis and suggest that the neurogenic process can serve as a target of anti-obesity pharmacotherapy.


Subject(s)
Neuropeptides , Obesity , Mice , Humans , Animals , Prolactin-Releasing Hormone/pharmacology , Prolactin-Releasing Hormone/therapeutic use , Obesity/drug therapy , Body Weight , Neurogenesis , Hypothalamus
2.
Development ; 148(12)2021 06 15.
Article in English | MEDLINE | ID: mdl-34170290

ABSTRACT

The third 'Symposium for the Next Generation of Stem Cell Research' (SY-Stem) was held virtually on 3-5 March 2021, having been cancelled in 2020 due to the COVID-19 pandemic. As in previous years, the meeting highlighted the work of early career researchers, ranging from postgraduate students to young group leaders working in developmental and stem cell biology. Here, we summarize the excellent work presented at the Symposium, which covered topics ranging from pluripotency, species-specific aspects of development and emerging technologies, through to organoids, single-cell technology and clinical applications.


Subject(s)
Congresses as Topic/organization & administration , Inventions/trends , Stem Cell Research , Animals , COVID-19/epidemiology , Cell Differentiation , Congresses as Topic/history , Congresses as Topic/trends , History, 21st Century , Humans , Internet , Inventions/history , Online Systems , Pandemics , Single-Cell Analysis/methods , Single-Cell Analysis/trends , Stem Cell Research/history , Stem Cells/physiology , Tissue Culture Techniques/methods , Tissue Culture Techniques/trends
3.
Nature ; 545(7653): 229-233, 2017 05 11.
Article in English | MEDLINE | ID: mdl-28445466

ABSTRACT

Human pluripotent stem cells (hPS cells) can self-renew indefinitely, making them an attractive source for regenerative therapies. This expansion potential has been linked with the acquisition of large copy number variants that provide mutated cells with a growth advantage in culture. The nature, extent and functional effects of other acquired genome sequence mutations in cultured hPS cells are not known. Here we sequence the protein-coding genes (exomes) of 140 independent human embryonic stem cell (hES cell) lines, including 26 lines prepared for potential clinical use. We then apply computational strategies for identifying mutations present in a subset of cells in each hES cell line. Although such mosaic mutations were generally rare, we identified five unrelated hES cell lines that carried six mutations in the TP53 gene that encodes the tumour suppressor P53. The TP53 mutations we observed are dominant negative and are the mutations most commonly seen in human cancers. We found that the TP53 mutant allelic fraction increased with passage number under standard culture conditions, suggesting that the P53 mutations confer selective advantage. We then mined published RNA sequencing data from 117 hPS cell lines, and observed another nine TP53 mutations, all resulting in coding changes in the DNA-binding domain of P53. In three lines, the allelic fraction exceeded 50%, suggesting additional selective advantage resulting from the loss of heterozygosity at the TP53 locus. As the acquisition and expansion of cancer-associated mutations in hPS cells may go unnoticed during most applications, we suggest that careful genetic characterization of hPS cells and their differentiated derivatives be carried out before clinical use.


Subject(s)
Genes, Dominant/genetics , Genes, p53 , Human Embryonic Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Mutation/genetics , Selection, Genetic , Tumor Suppressor Protein p53/genetics , Alleles , Cell Count , Cell Differentiation/genetics , Cell Division/genetics , Cell Line , DNA/metabolism , DNA Mutational Analysis , Exome/genetics , Human Embryonic Stem Cells/cytology , Humans , Loss of Heterozygosity/genetics , Mosaicism , Neoplasms/genetics , Protein Domains , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/metabolism
4.
Neuroendocrinology ; 110(1-2): 92-104, 2020.
Article in English | MEDLINE | ID: mdl-31104058

ABSTRACT

In outbred mice, susceptibility or resistance to diet-induced obesity is associated with rapid changes in hypothalamic proopiomelanocortin (POMC) levels. Here, we evaluated 3 hypotheses that potentially explain the development of the different obesity phenotypes in outbred Swiss mice. First, rapid and differential changes in the gut microbiota in obesity-prone (OP) and obesity-resistant (OR) mice fed on a high-fat diet (HFD) might cause differential efficiencies in fatty acid harvesting leading to changes in systemic fatty acid concentrations that in turn affect POMC expression and processing. Second, independently of the gut microbiota, OP mice might have increased blood fatty acid levels after the introduction of a HFD, which could affect POMC expression and processing. Third, fatty acids might act directly in the hypothalamus to differentially regulate POMC expression and/or processing in OP and OR mice. We evaluated OP and OR male Swiss mice using 16S rRNA sequencing for the determination of gut microbiota; gas chromatography for blood lipid determination; and immunoblot and real-time polymerase chain reaction for protein and transcript determination and indirect calorimetry. Some experiments were performed with human pluripotent stem cells differentiated into hypothalamic neurons. We did not find evidence supporting the first 2 hypotheses. However, we found that in OP but not in OR mice, palmitate induces a rapid increase in hypothalamic POMC, which is followed by increased expression of proprotein convertase subtilisin/kexin type 1 PC1/3. Lentiviral inhibition of hypothalamic PC1/3 increased caloric intake and body mass in both OP and OR mice. In human stem cell-derived hypothalamic cells, we found that palmitate potently suppressed the production of POMC-derived peptides. Palmitate directly regulates PC1/3 in OP mice and likely has a functional impact on POMC processing.


Subject(s)
Gastrointestinal Microbiome , Hypothalamus/metabolism , Inflammation/metabolism , Neurons/metabolism , Obesity/metabolism , Palmitates/pharmacology , Pro-Opiomelanocortin/metabolism , Animals , Diet, High-Fat , Disease Models, Animal , Humans , Linoleic Acid/pharmacology , Male , Mice , Obesity/blood , Obesity/etiology , Pluripotent Stem Cells , RNA, Ribosomal, 16S
5.
Development ; 142(4): 633-43, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25670790

ABSTRACT

Hypothalamic neurons orchestrate many essential physiological and behavioral processes via secreted neuropeptides, and are relevant to human diseases such as obesity, narcolepsy and infertility. We report the differentiation of human pluripotent stem cells into many of the major types of neuropeptidergic hypothalamic neurons, including those producing pro-opiolemelanocortin, agouti-related peptide, hypocretin/orexin, melanin-concentrating hormone, oxytocin, arginine vasopressin, corticotropin-releasing hormone (CRH) or thyrotropin-releasing hormone. Hypothalamic neurons can be generated using a 'self-patterning' strategy that yields a broad array of cell types, or via a more reproducible directed differentiation approach. Stem cell-derived human hypothalamic neurons share characteristic morphological properties and gene expression patterns with their counterparts in vivo, and are able to integrate into the mouse brain. These neurons could form the basis of cellular models, chemical screens or cellular therapies to study and treat common human diseases.


Subject(s)
Hypothalamus/cytology , Neurons/cytology , Pluripotent Stem Cells/cytology , Agouti-Related Protein/metabolism , Arginine Vasopressin/metabolism , Humans , Hypothalamic Hormones/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Melanins/metabolism , Neuropeptides/metabolism , Orexins , Oxytocin/metabolism , Pituitary Hormones/metabolism , Pro-Opiomelanocortin/metabolism , Thyrotropin-Releasing Hormone/metabolism
6.
Development ; 142(6): 1113-24, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25725064

ABSTRACT

Loss of neurons that express the neuropeptide hypocretin (Hcrt) has been implicated in narcolepsy, a debilitating disorder characterized by excessive daytime sleepiness and cataplexy. Cell replacement therapy, using Hcrt-expressing neurons generated in vitro, is a potentially useful therapeutic approach, but factors sufficient to specify Hcrt neurons are unknown. Using zebrafish as a high-throughput system to screen for factors that can specify Hcrt neurons in vivo, we identified the LIM homeobox transcription factor Lhx9 as necessary and sufficient to specify Hcrt neurons. We found that Lhx9 can directly induce hcrt expression and we identified two potential Lhx9 binding sites in the zebrafish hcrt promoter. Akin to its function in zebrafish, we found that Lhx9 is sufficient to specify Hcrt-expressing neurons in the developing mouse hypothalamus. Our results elucidate an evolutionarily conserved role for Lhx9 in Hcrt neuron specification that improves our understanding of Hcrt neuron development.


Subject(s)
Cell Separation/methods , Gene Expression Regulation/physiology , Hypothalamus/embryology , Intracellular Signaling Peptides and Proteins/metabolism , Neurons/metabolism , Neuropeptides/metabolism , Transcription Factors/metabolism , Zebrafish Proteins/metabolism , Animals , Animals, Genetically Modified , Cloning, Molecular , DNA Primers/genetics , Electrophoretic Mobility Shift Assay , Gene Knockdown Techniques , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , High-Throughput Screening Assays/methods , Hypothalamus/metabolism , Immunohistochemistry , Mice , Microarray Analysis , Orexins , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Zebrafish , Zebrafish Proteins/genetics
7.
Cells ; 13(13)2024 Jul 06.
Article in English | MEDLINE | ID: mdl-38995007

ABSTRACT

Primary cilia are finger-like sensory organelles that extend from the bodies of most cell types and have a distinct lipid and protein composition from the plasma membrane. This partitioning is maintained by a diffusion barrier that restricts the entry of non-ciliary proteins, and allows the selective entry of proteins harboring a ciliary targeting sequence (CTS). However, CTSs are not stereotyped and previously reported sequences are insufficient to drive efficient ciliary localisation across diverse cell types. Here, we describe a short peptide sequence that efficiently targets transmembrane proteins to primary cilia in all tested cell types, including human neurons. We generate human-induced pluripotent stem cell (hiPSC) lines stably expressing a transmembrane construct bearing an extracellular HaloTag and intracellular fluorescent protein, which enables the bright, specific labeling of primary cilia in neurons and other cell types to facilitate studies of cilia in health and disease. We demonstrate the utility of this resource by developing an image analysis pipeline for the automated measurement of primary cilia to detect changes in their length associated with altered signaling or disease state.


Subject(s)
Cilia , Induced Pluripotent Stem Cells , Membrane Proteins , Cilia/metabolism , Humans , Membrane Proteins/metabolism , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Animals , Neurons/metabolism , Amino Acid Sequence , Cell Line , Protein Transport
8.
bioRxiv ; 2024 Jan 29.
Article in English | MEDLINE | ID: mdl-38352495

ABSTRACT

Gracia-Diaz and colleagues analysed high-density DNA microarray and whole genome sequencing (WGS) data from the KOLF2.1J 'reference' human induced pluripotent stem cell (hiPSC) line1, and report the presence of five high-confidence heterozygous copy number variants (CNVs) at least 100kbp in length2. Since three of these CNVs span coding genes, some of which have been associated with neurodevelopmental disease, the authors raise the concern that these CNVs may compromise the utility of KOLF2.1J for neurological disease modelling. We appreciate their thorough analysis and thoughtful interpretation, and agree that potential users of this line should be made aware of all cases where KOLF2.1J differs from the reference genome. However, we believe that the benefits from the widespread use of KOLF2.1J outweigh the potential risks that might arise from the identified CNVs.

9.
Curr Opin Cell Biol ; 18(6): 704-9, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17046226

ABSTRACT

Neural stem cells (NSCs) are primary progenitors that give rise to neurons and glia in the embryonic, neonatal and adult brain. In recent years, we have learned three important things about these cells. First, NSCs correspond to cells previously thought to be committed glial cells. Second, embryonic and adult NSCs are lineally related: they transform from neuroepithelial cells into radial glia, then into cells with astroglial characteristics. Third, NSCs divide asymmetrically and often amplify the number of progeny they generate via symmetrically dividing intermediate progenitors. These advances challenge our traditional perceptions of glia and stem cells, and provide the foundation for understanding the molecular basis of mammalian NSC behavior.


Subject(s)
Cell Differentiation/physiology , Central Nervous System/embryology , Neuroglia/metabolism , Neurons/metabolism , Stem Cells/metabolism , Animals , Cell Lineage/physiology , Cell Movement/physiology , Central Nervous System/cytology , Central Nervous System/metabolism , Gene Expression Regulation, Developmental/physiology , Humans , Neuroglia/cytology , Neurons/cytology , Stem Cells/cytology , Transcription Factors/metabolism
10.
Cereb Cortex ; 22(2): 245-50, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21653703

ABSTRACT

Human anterior cingulate and frontoinsular cortices participate in healthy social-emotional processing. These regions feature 2 related layer 5 neuronal morphotypes, the von Economo neurons and fork cells. In this paper, we review the historical accounts of these neurons and provide a German-to-English translation of von Economo's seminal paper describing the neurons which have come to bear his name. We close with a brief discussion regarding the functional and clinical relevance of these neurons and their home regions.


Subject(s)
Frontal Lobe/cytology , Gyrus Cinguli/cytology , Neurons/physiology , Humans
11.
Trends Cell Biol ; 33(11): 979-990, 2023 11.
Article in English | MEDLINE | ID: mdl-37302961

ABSTRACT

Primary cilia protrude from most vertebrate cell bodies and act as specialized 'signalling antennae' that can substantially lengthen or retract in minutes to hours in response to specific stimuli. Here, we review the conditions and mechanisms responsible for regulating primary cilia length (PCL) in mammalian nonsensory neurons, and propose four models of how they could affect ciliary signalling and alter cell state and suggest experiments to distinguish between them. These models include (i) the passive indicator model, where changes in PCL have no consequence; (ii) the rheostat model, in which a longer cilium enhances signalling; (iii) the local concentration model, where ciliary shortening increases the local protein concentration to facilitate signalling; and (iv) the altered composition model where changes in PCL skew signalling.


Subject(s)
Cilia , Signal Transduction , Humans , Animals , Cilia/metabolism , Vertebrates , Neurons , Mammals
12.
Curr Protoc ; 3(6): e786, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37272700

ABSTRACT

Neurons in the hypothalamus orchestrate homeostatic physiological processes and behaviors essential for life. Human pluripotent stem cells (hPSCs) can be differentiated into many types of hypothalamic neurons, progenitors, and glia. This updated unit includes published studies and protocols with new advances in the differentiation, maturation, and interrogation by transcriptomic profiling and calcium imaging of human hypothalamic cell populations. Specifically, new methods to freeze and thaw hypothalamic progenitors after they have been patterned and before substantial neurogenesis has occurred are provided that will facilitate experimental flexibility and planning. Also included are updated recipes and protocols for neuronal maturation, with details on the equipment and methods for examining their transcriptomic response and cell-autonomous properties in culture in the presence of synaptic blockers. Together, these protocols facilitate the adoption and use of this model system for fundamental biological discovery and therapeutic translation to human diseases such as obesity, diabetes, sleep disorders, infertility, and chronic stress. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: hPSC maintenance Basic Protocol 2: Hypothalamic neuron differentiation Support Protocol 1: Cortical neuron (control) differentiation Basic Protocol 3: Neuronal maturation Support Protocol 2: Cryopreservation and thawing of neuronal progenitors Support Protocol 3: Quality control: Confirmation of hypothalamic patterning and neurogenesis Support Protocol 4: Bulk RNA sequencing of hypothalamic cultures Basic Protocol 4: Calcium imaging of hypothalamic neurons using Fura-2 AM Alternate Protocol: Calcium imaging of green fluorescent hypothalamic neurons using Rhod-3 AM.


Subject(s)
Neurons , Transcriptome , Humans , Neurons/physiology , Cell Differentiation/physiology , Hypothalamus/diagnostic imaging , Neurogenesis/genetics , Calcium, Dietary
13.
JCI Insight ; 8(8)2023 04 24.
Article in English | MEDLINE | ID: mdl-37092553

ABSTRACT

Makorin ring finger protein 3 (MKRN3) was identified as an inhibitor of puberty initiation with the report of loss-of-function mutations in association with central precocious puberty. Consistent with this inhibitory role, a prepubertal decrease in Mkrn3 expression was observed in the mouse hypothalamus. Here, we investigated the mechanisms of action of MKRN3 in the central regulation of puberty onset. We showed that MKRN3 deletion in hypothalamic neurons derived from human induced pluripotent stem cells was associated with significant changes in expression of genes controlling hypothalamic development and plasticity. Mkrn3 deletion in a mouse model led to early puberty onset in female mice. We found that Mkrn3 deletion increased the number of dendritic spines in the arcuate nucleus but did not alter the morphology of GnRH neurons during postnatal development. In addition, we identified neurokinin B (NKB) as an Mkrn3 target. Using proteomics, we identified insulin-like growth factor 2 mRNA-binding protein 1 (IGF2BP1) as another target of MKRN3. Interactome analysis revealed that IGF2BP1 interacted with MKRN3, along with several members of the polyadenylate-binding protein family. Our data show that one of the mechanisms by which MKRN3 inhibits pubertal initiation is through regulation of prepubertal hypothalamic development and plasticity, as well as through effects on NKB and IGF2BP1.


Subject(s)
Induced Pluripotent Stem Cells , Puberty, Precocious , Humans , Female , Mice , Animals , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Induced Pluripotent Stem Cells/metabolism , Hypothalamus/metabolism , Puberty , Gonadotropin-Releasing Hormone/metabolism , Puberty, Precocious/genetics , Puberty, Precocious/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
14.
Cell Stem Cell ; 29(3): 472-486.e7, 2022 03 03.
Article in English | MEDLINE | ID: mdl-35176222

ABSTRACT

Despite their widespread use in research, there has not yet been a systematic genomic analysis of human embryonic stem cell (hESC) lines at a single-nucleotide resolution. We therefore performed whole-genome sequencing (WGS) of 143 hESC lines and annotated their single-nucleotide and structural genetic variants. We found that while a substantial fraction of hESC lines contained large deleterious structural variants, finer-scale structural and single-nucleotide variants (SNVs) that are ascertainable only through WGS analyses were present in hESC genomes and human blood-derived genomes at similar frequencies. Moreover, WGS allowed us to identify SNVs associated with cancer and other diseases that could alter cellular phenotypes and compromise the safety of hESC-derived cellular products transplanted into humans. As a resource to enable reproducible hESC research and safer translation, we provide a user-friendly WGS data portal and a data-driven scheme for cell line maintenance and selection.


Subject(s)
Human Embryonic Stem Cells , Genetic Variation , Genome, Human/genetics , Humans , Nucleotides , Whole Genome Sequencing
15.
Cell Stem Cell ; 29(12): 1624-1636, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36459966

ABSTRACT

It is well established that human pluripotent stem cells (hPSCs) can acquire genetic and epigenetic changes during culture in vitro. Given the increasing use of hPSCs in research and therapy and the vast expansion in the number of hPSC lines available for researchers, the International Society for Stem Cell Research has recognized the need to reassess quality control standards for ensuring the genetic integrity of hPSCs. Here, we summarize current knowledge of the nature of recurrent genetic and epigenetic variants in hPSC culture, the methods for their detection, and what is known concerning their effects on cell behavior in vitro or in vivo. We argue that the potential consequences of low-level contamination of cell therapy products with cells bearing oncogenic variants are essentially unknown at present. We highlight the key challenges facing the field with particular reference to safety assessment of hPSC-derived cellular therapeutics.


Subject(s)
Epigenomics , Pluripotent Stem Cells , Humans , Stem Cell Research , Oncogenes , Epigenesis, Genetic
16.
Front Neurosci ; 15: 709825, 2021.
Article in English | MEDLINE | ID: mdl-34720852

ABSTRACT

When mice are exposed to external warmth, nitric oxide synthase (NOS1) neurons in the median and medial preoptic (MnPO/MPO) hypothalamus induce sleep and concomitant body cooling. However, how these neurons regulate baseline sleep and body temperature is unknown. Using calcium photometry, we show that NOS1 neurons in MnPO/MPO are predominantly NREM and REM active, especially at the boundary of wake to NREM transitions, and in the later parts of REM bouts, with lower activity during wakefulness. In addition to releasing nitric oxide, NOS1 neurons in MnPO/MPO can release GABA, glutamate and peptides. We expressed tetanus-toxin light-chain in MnPO/MPO NOS1 cells to reduce vesicular release of transmitters. This induced changes in sleep structure: over 24 h, mice had less NREM sleep in their dark (active) phase, and more NREM sleep in their light (sleep) phase. REM sleep episodes in the dark phase were longer, and there were fewer REM transitions between other vigilance states. REM sleep had less theta power. Mice with synaptically blocked MnPO/MPO NOS1 neurons were also warmer than control mice at the dark-light transition (ZT0), as well as during the dark phase siesta (ZT16-20), where there is usually a body temperature dip. Also, at this siesta point of cooled body temperature, mice usually have more NREM, but mice with synaptically blocked MnPO/MPO NOS1 cells showed reduced NREM sleep at this time. Overall, MnPO/MPO NOS1 neurons promote both NREM and REM sleep and contribute to chronically lowering body temperature, particularly at transitions where the mice normally enter NREM sleep.

17.
Nat Commun ; 12(1): 6746, 2021 11 19.
Article in English | MEDLINE | ID: mdl-34799581

ABSTRACT

DNA replication follows a strict spatiotemporal program that intersects with chromatin structure but has a poorly understood genetic basis. To systematically identify genetic regulators of replication timing, we exploited inter-individual variation in human pluripotent stem cells from 349 individuals. We show that the human genome's replication program is broadly encoded in DNA and identify 1,617 cis-acting replication timing quantitative trait loci (rtQTLs) - sequence determinants of replication initiation. rtQTLs function individually, or in combinations of proximal and distal regulators, and are enriched at sites of histone H3 trimethylation of lysines 4, 9, and 36 together with histone hyperacetylation. H3 trimethylation marks are individually repressive yet synergistically associate with early replication. We identify pluripotency-related transcription factors and boundary elements as positive and negative regulators of replication timing, respectively. Taken together, human replication timing is controlled by a multi-layered mechanism with dozens of effectors working combinatorially and following principles analogous to transcription regulation.


Subject(s)
DNA Replication Timing , Genome, Human , Pluripotent Stem Cells/metabolism , Acetylation , Biological Variation, Population/genetics , DNA Methylation , Datasets as Topic , Female , Gene Expression Regulation , Histone Code/genetics , Histones/metabolism , Humans , Male , Quantitative Trait Loci , Transcription Factors/metabolism , Whole Genome Sequencing
18.
Nat Genet ; 53(3): 304-312, 2021 03.
Article in English | MEDLINE | ID: mdl-33664506

ABSTRACT

Studying the function of common genetic variants in primary human tissues and during development is challenging. To address this, we use an efficient multiplexing strategy to differentiate 215 human induced pluripotent stem cell (iPSC) lines toward a midbrain neural fate, including dopaminergic neurons, and use single-cell RNA sequencing (scRNA-seq) to profile over 1 million cells across three differentiation time points. The proportion of neurons produced by each cell line is highly reproducible and is predictable by robust molecular markers expressed in pluripotent cells. Expression quantitative trait loci (eQTL) were characterized at different stages of neuronal development and in response to rotenone-induced oxidative stress. Of these, 1,284 eQTL colocalize with known neurological trait risk loci, and 46% are not found in the Genotype-Tissue Expression (GTEx) catalog. Our study illustrates how coupling scRNA-seq with long-term iPSC differentiation enables mechanistic studies of human trait-associated genetic variants in otherwise inaccessible cell states.


Subject(s)
Dopaminergic Neurons/cytology , Dopaminergic Neurons/physiology , Induced Pluripotent Stem Cells/cytology , Quantitative Trait Loci , Transcriptome , Cell Differentiation/genetics , Genetic Predisposition to Disease , Humans , Induced Pluripotent Stem Cells/physiology , Neurogenesis/genetics , Oxidative Stress/drug effects , Receptor, Fibroblast Growth Factor, Type 1/genetics , Rotenone/toxicity , Sequence Analysis, RNA , Single-Cell Analysis
19.
Cell Rep ; 37(2): 109800, 2021 10 12.
Article in English | MEDLINE | ID: mdl-34644574

ABSTRACT

Hypothalamic pro-opiomelanocortin (POMC) neurons are known to trigger satiety. However, these neuronal cells encompass heterogeneous subpopulations that release γ-aminobutyric acid (GABA), glutamate, or both neurotransmitters, whose functions are poorly defined. Using conditional mutagenesis and chemogenetics, we show that blockade of the energy sensor mechanistic target of rapamycin complex 1 (mTORC1) in POMC neurons causes hyperphagia by mimicking a cellular negative energy state. This is associated with decreased POMC-derived anorexigenic α-melanocyte-stimulating hormone and recruitment of POMC/GABAergic neurotransmission, which is restrained by cannabinoid type 1 receptor signaling. Electrophysiology and optogenetic studies further reveal that pharmacological blockade of mTORC1 simultaneously activates POMC/GABAergic neurons and inhibits POMC/glutamatergic ones, implying that the functional specificity of these subpopulations relies on mTORC1 activity. Finally, POMC neurons with different neurotransmitter profiles possess specific molecular signatures and spatial distribution. Altogether, these findings suggest that mTORC1 orchestrates the activity of distinct POMC neurons subpopulations to regulate feeding behavior.


Subject(s)
Appetite Regulation , Feeding Behavior , GABAergic Neurons/metabolism , Glutamic Acid/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Neural Inhibition , Paraventricular Hypothalamic Nucleus/metabolism , Pro-Opiomelanocortin/metabolism , Animals , Male , Mechanistic Target of Rapamycin Complex 1/genetics , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Pro-Opiomelanocortin/genetics , Signal Transduction
20.
Trends Neurosci ; 31(8): 392-400, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18603310

ABSTRACT

In adult rodents, subventricular zone (SVZ) astrocytes (B cells) function as primary progenitors in the generation of new neurons that migrate to the olfactory bulb (OB), where they differentiate into multiple types of interneurons. It has been generally considered that individual adult SVZ stem cells are capable of generating different types of neurons and glial cells. However, recent studies indicate that these adult SVZ primary progenitors are heterogeneous and predetermined to generate specific types of neurons. Surprisingly, OB interneurons are generated by stem cells not only in the walls of the lateral ventricle facing the striatum but also in the rostral migratory stream and walls of the lateral ventricle facing the cortex and the septum. SVZ B cells in different locations within this extensive germinal region generate different kinds of interneurons. General physiological characteristics of major classes of OB interneurons have begun to emerge, but the functional contribution of each subtype remains unknown. The mosaic organization of the SVZ offers a unique opportunity to understand the origin of interneuron diversity and how this assortment of neurons contributes to plasticity of postnatal olfactory circuits.


Subject(s)
Cell Differentiation/physiology , Interneurons/cytology , Olfactory Bulb/cytology , Olfactory Pathways/cytology , Stem Cells/cytology , Animals , Cell Movement/physiology , Cell Proliferation , Cerebral Ventricles/cytology , Humans , Interneurons/physiology , Olfactory Bulb/growth & development , Olfactory Bulb/metabolism , Olfactory Pathways/growth & development , Olfactory Pathways/metabolism , Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL