Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 369
Filter
1.
J Neurophysiol ; 132(3): 943-952, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39108212

ABSTRACT

Cotransmission, meaning the release of multiple neurotransmitters from one synapse, allows for increased diversity of signaling in the brain. Dopamine (DA) and γ-aminobutyric acid (GABA) are known to coexpress in many regions such as the olfactory bulb and the ventral tegmental area. Tuberoinfundibular dopaminergic neurons (TIDA) in the arcuate nucleus of the hypothalamus (Arc) project to the median eminence (ME) and regulate prolactin release from the pituitary, and prior work suggests dopaminergic Arc neurons also cotransmit GABA. However, the extent of cotransmission, and the projection patterns of these neurons have not been fully revealed. Here, we used a genetic intersectional reporter expression approach to selectively label cells that express both tyrosine hydroxylase (TH) and vesicular GABA transporter (VGAT). Through this approach, we identified cells capable of both DA and GABA cotransmission in the Arc, periventricular (Pe), paraventricular (Pa), ventromedial, and the dorsolateral hypothalamic nuclei, in addition to a novel population in the caudate putamen. The highest density of labeled cells was in the Arc, 6.68% of DAPI-labeled cells at Bregma -2.06 mm, and in the Pe, 2.83% of DAPI-labeled cells at Bregma -1.94 mm. Next, we evaluated the projections of these DA/GABA cells by injecting an mCherry virus that fluoresces in DA/GABA cells. We observed a cotransmitting DA/GABA population, with projections within the Arc, and to the Pa and ME. These data suggest DA/GABA Arc neurons are involved in prolactin release as a subset of TIDA neurons. Further investigation will elucidate the interactions of dopamine and GABA in the hypothalamus.NEW & NOTEWORTHY Cotransmitting dopaminergic (DA) and γ-aminobutyric acid (GABA)ergic (DA/GABA) neurons contribute to the complexity of neural circuits. Using a new genetic technique, we characterized the locations, density, and projections of hypothalamic DA/GABA neurons. DA/GABA cells are mostly in the arcuate nucleus (Arc), from which they project locally within the arcuate, to the median eminence (ME), and to the paraventricular (Pa) nucleus. There is also a small and previously unreported group of DA/GABA cells in the caudate putamen.


Subject(s)
Arcuate Nucleus of Hypothalamus , Dopaminergic Neurons , GABAergic Neurons , Median Eminence , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Arcuate Nucleus of Hypothalamus/cytology , GABAergic Neurons/metabolism , GABAergic Neurons/physiology , Median Eminence/metabolism , Median Eminence/cytology , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/physiology , Male , Mice , Tyrosine 3-Monooxygenase/metabolism , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism , Female , Neural Pathways/metabolism , Neural Pathways/physiology
2.
Ann Neurol ; 91(2): 282-288, 2022 02.
Article in English | MEDLINE | ID: mdl-34981555

ABSTRACT

Narcolepsy type 1 (NT1) is a chronic sleep disorder correlated with loss of hypocretin(orexin). In NT1 post-mortem brains, we observed 88% reduction in corticotropin-releasing hormone (CRH)-positive neurons in the paraventricular nucleus (PVN) and significantly less CRH-positive fibers in the median eminence, whereas CRH-neurons in the locus coeruleus and thalamus, and other PVN neuronal populations were spared: that is, vasopressin, oxytocin, tyrosine hydroxylase, and thyrotropin releasing hormone-expressing neurons. Other hypothalamic cell groups, that is, the suprachiasmatic, ventrolateral preoptic, infundibular, and supraoptic nuclei and nucleus basalis of Meynert, were unaffected. The surprising selective decrease in CRH-neurons provide novel targets for diagnostics and therapeutic interventions. ANN NEUROL 2022;91:282-288.


Subject(s)
Corticotropin-Releasing Hormone/metabolism , Hypothalamus/metabolism , Hypothalamus/pathology , Narcolepsy/pathology , Neurons/pathology , Aged , Aged, 80 and over , Cell Count , Female , Humans , Hypothalamus/diagnostic imaging , Immunohistochemistry , Locus Coeruleus/cytology , Locus Coeruleus/diagnostic imaging , Locus Coeruleus/metabolism , Male , Median Eminence/cytology , Median Eminence/diagnostic imaging , Median Eminence/metabolism , Middle Aged , Narcolepsy/diagnostic imaging , Paraventricular Hypothalamic Nucleus/cytology , Paraventricular Hypothalamic Nucleus/diagnostic imaging , Paraventricular Hypothalamic Nucleus/metabolism
3.
Science ; 375(6579): eabk2346, 2022 01 28.
Article in English | MEDLINE | ID: mdl-35084970

ABSTRACT

The human cortex contains inhibitory interneurons derived from the medial ganglionic eminence (MGE), a germinal zone in the embryonic ventral forebrain. How this germinal zone generates sufficient interneurons for the human brain remains unclear. We found that the human MGE (hMGE) contains nests of proliferative neuroblasts with ultrastructural and transcriptomic features that distinguish them from other progenitors in the hMGE. When dissociated hMGE cells are transplanted into the neonatal mouse brain, they reform into nests containing proliferating neuroblasts that generate young neurons that migrate extensively into the mouse forebrain and mature into different subtypes of functional interneurons. Together, these results indicate that the nest organization and sustained proliferation of neuroblasts in the hMGE provide a mechanism for the extended production of interneurons for the human forebrain.


Subject(s)
Interneurons/physiology , Median Eminence/embryology , Neural Stem Cells/physiology , Neurogenesis , Prosencephalon/cytology , Animals , Animals, Newborn , Cell Movement , Cell Proliferation , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Cerebral Cortex/growth & development , GABAergic Neurons/cytology , GABAergic Neurons/physiology , Gene Expression Profiling , Gestational Age , Humans , Interneurons/cytology , Median Eminence/cytology , Median Eminence/growth & development , Mice , Neural Stem Cells/transplantation , Prosencephalon/embryology , Prosencephalon/growth & development , Transplantation, Heterologous
4.
Neural Plast ; 2021: 4894881, 2021.
Article in English | MEDLINE | ID: mdl-34659396

ABSTRACT

Stem cells have the potential as a regenerative therapy for cerebral ischemia by improving functional outcomes. However, cell transplantation has some limitations, including a low rate of the grafted cell survival. There is still a major challenge of promoting the harmonious symbiosis between grafted cells and the host. Acupuncture can effectively improve the functional outcome after cerebral ischemia. The present study evaluated the therapeutic effects and explored the mechanism of combined medial ganglionic eminence (MGE) neural progenitors differentiated from human embryonic stem cells (hESCs) with electroacupuncture (EA) in a bilateral common carotid artery occlusion (2VO) rat model. The results showed that EA could promote the survival of the grafted MGE neural progenitors differentiated from hESCs and alleviate learning and memory impairment in rats with cerebral ischemia. This may have partially resulted from inhibited expression of TNF-α and IL-1ß and increased vascular endothelial growth factor (VEGF) expression and blood vessel density in the hippocampus. Our findings indicated that EA could promote the survival of the grafted MGE neural progenitors and enhance transplantation therapy's efficacy by promoting angiogenesis and inhibiting inflammation.


Subject(s)
Brain Ischemia/therapy , Electroacupuncture/methods , Inflammation Mediators/antagonists & inhibitors , Median Eminence/transplantation , Neovascularization, Physiologic/physiology , Stem Cell Transplantation/methods , Animals , Brain Ischemia/metabolism , Brain Ischemia/pathology , Cell Survival/physiology , Cells, Cultured , Embryonic Stem Cells/physiology , Embryonic Stem Cells/transplantation , Hippocampus/cytology , Hippocampus/physiology , Humans , Inflammation Mediators/metabolism , Male , Maze Learning/physiology , Median Eminence/cytology , Median Eminence/physiology , Neural Stem Cells/physiology , Neural Stem Cells/transplantation , Rats , Rats, Sprague-Dawley
5.
STAR Protoc ; 2(3): 100787, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34485946

ABSTRACT

The hypothalamic magnocellular neuroendocrine cells (MNCs) project to the posterior pituitary (PPi), regulating reproduction and fluid homeostasis. It has been challenging to selectively label and manipulate MNCs, as they are intermingled with parvocellular neuroendocrine cells projecting to the median eminence. Here, we provide a step-by-step protocol for specifically targeting the MNCs by infusing retrograde viral tracers into the PPi. When combined with optogenetics, chemogenetics, and transgenic animals, this approach allows cell-type-specific manipulation of MNCs in multiple sites for functional dissection. For complete details on the use and execution of this protocol, please refer to Zhang et al. (2021) and Tang et al. (2020).


Subject(s)
Hypothalamus/cytology , Neuroendocrine Cells , Optogenetics/methods , Pituitary Gland, Posterior/cytology , Animals , Animals, Genetically Modified , Male , Median Eminence/cytology , Nerve Net/cytology , Nerve Net/physiology , Neuroendocrine Cells/cytology , Neuroendocrine Cells/physiology , Rats , Rats, Sprague-Dawley
6.
Cell Rep ; 36(2): 109362, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34260928

ABSTRACT

The mediobasal hypothalamus (MBH; arcuate nucleus of the hypothalamus [ARH] and median eminence [ME]) is a key nutrient sensing site for the production of the complex homeostatic feedback responses required for the maintenance of energy balance. Here, we show that refeeding after an overnight fast rapidly triggers proliferation and differentiation of oligodendrocyte progenitors, leading to the production of new oligodendrocytes in the ME specifically. During this nutritional paradigm, ME perineuronal nets (PNNs), emerging regulators of ARH metabolic functions, are rapidly remodeled, and this process requires myelin regulatory factor (Myrf) in oligodendrocyte progenitors. In genetically obese ob/ob mice, nutritional regulations of ME oligodendrocyte differentiation and PNN remodeling are blunted, and enzymatic digestion of local PNN increases food intake and weight gain. We conclude that MBH PNNs are required for the maintenance of energy balance in lean mice and are remodeled in the adult ME by the nutritional control of oligodendrocyte differentiation.


Subject(s)
Cell Differentiation , Median Eminence/cytology , Nerve Net/physiology , Nutritional Physiological Phenomena , Oligodendroglia/cytology , Adult , Animals , Cell Lineage , Cell Proliferation , Humans , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice, Inbred C57BL , Oligodendroglia/ultrastructure , Single-Cell Analysis , Transcriptome/genetics
7.
Nat Commun ; 12(1): 2288, 2021 04 16.
Article in English | MEDLINE | ID: mdl-33863883

ABSTRACT

Hypothalamic tanycytes in median eminence (ME) are emerging as a crucial cell population that regulates endocrine output, energy balance and the diffusion of blood-born molecules. Tanycytes have recently been considered as potential somatic stem cells in the adult mammalian brain, but their regenerative and tumorigenic capacities are largely unknown. Here we found that Rax+ tanycytes in ME of mice are largely quiescent but quickly enter the cell cycle upon neural injury for self-renewal and regeneration. Mechanistically, Igf1r signaling in tanycytes is required for tissue repair under injury conditions. Furthermore, Braf oncogenic activation is sufficient to transform Rax+ tanycytes into actively dividing tumor cells that eventually develop into a papillary craniopharyngioma-like tumor. Together, these findings uncover the regenerative and tumorigenic potential of tanycytes. Our study offers insights into the properties of tanycytes, which may help to manipulate tanycyte biology for regulating hypothalamic function and investigate the pathogenesis of clinically relevant tumors.


Subject(s)
Craniopharyngioma/pathology , Ependymoglial Cells/physiology , Median Eminence/physiology , Neoplasms, Experimental/pathology , Regeneration , Animals , Carcinogenesis/pathology , Cell Self Renewal/physiology , Craniopharyngioma/chemically induced , Craniopharyngioma/genetics , Eye Proteins/metabolism , Female , Homeodomain Proteins/metabolism , Median Eminence/cytology , Mice , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/genetics , Proto-Oncogene Proteins B-raf/genetics , RNA-Seq , Receptor, IGF Type 1/metabolism , Signal Transduction , Single-Cell Analysis , Transcription Factors/metabolism
8.
J Neurosci ; 41(14): 3105-3119, 2021 04 07.
Article in English | MEDLINE | ID: mdl-33637558

ABSTRACT

Interneurons contribute to the complexity of neural circuits and maintenance of normal brain function. Rodent interneurons originate in embryonic ganglionic eminences, but developmental origins in other species are less understood. Here, we show that transcription factor expression patterns in porcine embryonic subpallium are similar to rodents, delineating a distinct medial ganglionic eminence (MGE) progenitor domain. On the basis of Nkx2.1, Lhx6, and Dlx2 expression, in vitro differentiation into neurons expressing GABA, and robust migratory capacity in explant assays, we propose that cortical and hippocampal interneurons originate from a porcine MGE region. Following xenotransplantation into adult male and female rat hippocampus, we further demonstrate that porcine MGE progenitors, like those from rodents, migrate and differentiate into morphologically distinct interneurons expressing GABA. Our findings reveal that basic rules for interneuron development are conserved across species, and that porcine embryonic MGE progenitors could serve as a valuable source for interneuron-based xenotransplantation therapies.SIGNIFICANCE STATEMENT Here we demonstrate that porcine medial ganglionic eminence, like rodents, exhibit a distinct transcriptional and interneuron-specific antibody profile, in vitro migratory capacity and are amenable to xenotransplantation. This is the first comprehensive examination of embryonic interneuron origins in the pig; and because a rich neurodevelopmental literature on embryonic mouse medial ganglionic eminence exists (with some additional characterizations in other species, e.g., monkey and human), our work allows direct neurodevelopmental comparisons with this literature.


Subject(s)
Ganglia/embryology , Ganglia/transplantation , Interneurons/transplantation , Median Eminence/embryology , Median Eminence/transplantation , Transplantation, Heterologous/methods , Animals , Female , Ganglia/cytology , Male , Median Eminence/cytology , Rats , Rats, Sprague-Dawley , Swine , Tissue Culture Techniques/methods
9.
Stem Cell Reports ; 14(6): 1076-1092, 2020 06 09.
Article in English | MEDLINE | ID: mdl-32413277

ABSTRACT

The subventricular zone (SVZ) of the lateral ventricles and the subgranular zone (SGZ) of the dentate gyrus in the hippocampus are known as neurogenic niches. We show that the median eminence (ME) of the hypothalamus comprises BrdU+ newly proliferating cells co-expressing NG2 (oligodendrocyte progenitors) and RIP (pre-myelinating oligodendrocytes), suggesting their differentiation toward mature oligodendrocytes (OLs). ME cells can generate neurospheres (NS) in vitro, which differentiate mostly to OLs compared with SVZ-NS that typically generate neurons. Interestingly, this population of oligodendrocyte progenitors is increased in the ME from experimental autoimmune encephalomyelitis (EAE)-affected mice. Notably, the thrombospondin 1 (TSP1) expressed by astrocytes, acts as negative regulator of oligodendrogenesis in vitro and is downregulated in the ME of EAE mice. Importantly, transplanted ME-NS preferentially differentiate to MBP+ OLs compared with SVZ-NS in Shiverer mice. Hence, discovering the ME as a new site for myelin-producing cells has a great importance for advising future therapy for demyelinating diseases and spinal cord injury.


Subject(s)
Median Eminence/cytology , Neural Stem Cells/cytology , Oligodendroglia/cytology , Stem Cell Niche , Animals , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Mice , Mice, Inbred C57BL , Neural Stem Cells/metabolism , Neurogenesis , Oligodendroglia/metabolism , Thrombospondin 1/genetics , Thrombospondin 1/metabolism
10.
Proc Natl Acad Sci U S A ; 117(11): 6189-6195, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32123116

ABSTRACT

Neurofibromatosis 1 (NF1) is caused by mutations in the NF1 gene, which encodes the protein, neurofibromin, an inhibitor of Ras activity. Cortical GABAergic interneurons (CINs) are implicated in NF1 pathology, but the cellular and molecular changes to CINs are unknown. We deleted mouse Nf1 from the medial ganglionic eminence, which gives rise to both oligodendrocytes and CINs that express somatostatin and parvalbumin. Nf1 loss led to a persistence of immature oligodendrocytes that prevented later-generated oligodendrocytes from occupying the cortex. Moreover, molecular and cellular properties of parvalbumin (PV)-positive CINs were altered by the loss of Nf1, without changes in somatostatin (SST)-positive CINs. We discovered that loss of Nf1 results in a dose-dependent decrease in Lhx6 expression, the transcription factor necessary to establish SST+ and PV+ CINs, which was rescued by the MEK inhibitor SL327, revealing a mechanism whereby a neurofibromin/Ras/MEK pathway regulates a critical CIN developmental milestone.


Subject(s)
Cerebral Cortex/pathology , GABAergic Neurons/pathology , Interneurons/pathology , LIM-Homeodomain Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurofibromatosis 1/pathology , Neurofibromin 1/genetics , Transcription Factors/metabolism , Aminoacetonitrile/administration & dosage , Aminoacetonitrile/analogs & derivatives , Animals , Cells, Cultured , Cerebral Cortex/cytology , Disease Models, Animal , Embryo, Mammalian , Female , GABAergic Neurons/metabolism , Humans , Interneurons/metabolism , MAP Kinase Signaling System/drug effects , Median Eminence/cytology , Mice , Mice, Knockout , Neurofibromatosis 1/genetics , Neurofibromin 1/metabolism , Neuroglia/cytology , Parvalbumins/metabolism , Primary Cell Culture , Somatostatin/metabolism , ras GTPase-Activating Proteins/metabolism
11.
J Neurosci ; 40(11): 2215-2227, 2020 03 11.
Article in English | MEDLINE | ID: mdl-31988060

ABSTRACT

Manipulations that enhance GABAergic inhibition have been associated with improved behavioral phenotypes in autism models, suggesting that autism may be treated by correcting underlying deficits of inhibition. Interneuron transplantation is a method for increasing recipient synaptic inhibition, and it has been considered a prospective therapy for conditions marked by deficient inhibition, including neuropsychiatric disorders. It is unknown, however, whether interneuron transplantation may be therapeutically effective only for conditions marked by reduced inhibition, and it is also unclear whether transplantation improves behavioral phenotypes solely by normalizing underlying circuit defects. To address these questions, we studied the effects of interneuron transplantation in male and female mice lacking the autism-associated gene, Pten, in GABAergic interneurons. Pten mutant mice exhibit social behavior deficits, elevated synaptic inhibition in prefrontal cortex, abnormal baseline and social interaction-evoked electroencephalogram (EEG) signals, and an altered composition of cortical interneuron subtypes. Transplantation of wild-type embryonic interneurons from the medial ganglionic eminence into the prefrontal cortex of neonatal Pten mutants rescued social behavior despite exacerbating excessive levels of synaptic inhibition. Furthermore, transplantation did not normalize recipient EEG signals measured during baseline states. Interneuron transplantation can thus correct behavioral deficits even when those deficits are associated with elevated synaptic inhibition. Moreover, transplantation does not exert therapeutic effects solely by restoring wild-type circuit states. Our findings indicate that interneuron transplantation could offer a novel cell-based approach to autism treatment while challenging assumptions that effective therapies must reverse underlying circuit defects.SIGNIFICANCE STATEMENT Imbalances between neural excitation and inhibition are hypothesized to contribute to the pathophysiology of autism. Interneuron transplantation is a method for altering recipient inhibition, and it has been considered a prospective therapy for neuropsychiatric disorders, including autism. Here we examined the behavioral and physiological effects of interneuron transplantation in a mouse genetic model of autism. They demonstrate that transplantation rescues recipient social interaction deficits without correcting a common measure of recipient inhibition, or circuit-level physiological measures. These findings demonstrate that interneuron transplantation can exert therapeutic behavioral effects without necessarily restoring wild-type circuit states, while highlighting the potential of interneuron transplantation as an autism therapy.


Subject(s)
Autistic Disorder/surgery , Brain Tissue Transplantation , Fetal Tissue Transplantation , GABAergic Neurons/physiology , Interneurons/transplantation , Neural Inhibition/physiology , PTEN Phosphohydrolase/deficiency , Social Behavior , Animals , Autistic Disorder/physiopathology , Autistic Disorder/psychology , Disease Models, Animal , Electroencephalography , Exploratory Behavior , Female , Male , Maze Learning , Median Eminence/cytology , Median Eminence/embryology , Mice , Mice, Knockout , PTEN Phosphohydrolase/physiology , Patch-Clamp Techniques , Phenotype , Prefrontal Cortex/physiopathology , Random Allocation , Synapses/physiology
12.
J Cell Biol ; 219(1)2020 01 06.
Article in English | MEDLINE | ID: mdl-31676717

ABSTRACT

Although the role of transcription factors in fate specification of cortical interneurons is well established, how these interact with extracellular signals to regulate interneuron development is poorly understood. Here we show that the activin receptor ALK4 is a key regulator of the specification of somatostatin interneurons. Mice lacking ALK4 in GABAergic neurons of the medial ganglionic eminence (MGE) showed marked deficits in distinct subpopulations of somatostatin interneurons from early postnatal stages of cortical development. Specific losses were observed among distinct subtypes of somatostatin+/Reelin+ double-positive cells, including Hpse+ layer IV cells targeting parvalbumin+ interneurons, leading to quantitative alterations in the inhibitory circuitry of this layer. Activin-mediated ALK4 signaling in MGE cells induced interaction of Smad2 with SATB1, a transcription factor critical for somatostatin interneuron development, and promoted SATB1 nuclear translocation and repositioning within the somatostatin gene promoter. These results indicate that intrinsic transcriptional programs interact with extracellular signals present in the environment of MGE cells to regulate cortical interneuron specification.


Subject(s)
Activin Receptors, Type I/physiology , Cerebral Cortex/cytology , GABAergic Neurons/cytology , Interneurons/cytology , Median Eminence/cytology , Neurogenesis , Somatostatin/metabolism , Animals , Cell Differentiation , Cell Lineage , Cerebral Cortex/metabolism , Female , GABAergic Neurons/metabolism , Interneurons/metabolism , Male , Median Eminence/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Parvalbumins/metabolism , Reelin Protein , Signal Transduction
13.
Anat Histol Embryol ; 48(5): 415-420, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31241795

ABSTRACT

We examined the distribution of the orexin-like peptides in the pituitary and median eminence of the flat-tailed house gecko (Hemidactylus platyurus) using immunohistochemistry. Orexin-B-like, but not orexin-A-like, immunoreactivity was detected in the pituitary, specifically in the pars intermedia, and these cells corresponded to alpha-melanocyte-stimulating hormone (αMSH)-producing cells. Orexin-B and αMSH secreted from pars intermedia may modulate secretion of adenohypophyseal cells in the pars distalis. In the median eminence, orexin-B-immunoreactive puncta and fibres were observed, and these structures corresponded to gonadotropin-releasing hormone (GnRH)-immunoreactive puncta and fibres. Orexin-B secreted from GnRH-containing neurons in the hypothalamus may affect thyrotropin-releasing hormone-containing neurons resulting in modulation of αMSH secretion of melanotrophs in the pars intermedia.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Lizards , Orexins/metabolism , Pituitary Gland , alpha-MSH/metabolism , Animals , Immunohistochemistry , Median Eminence/cytology , Median Eminence/metabolism , Neurons/metabolism , Pituitary Gland/cytology , Pituitary Gland/metabolism
14.
Stem Cell Reports ; 12(5): 934-949, 2019 05 14.
Article in English | MEDLINE | ID: mdl-31056478

ABSTRACT

Medial ganglionic eminence (MGE)-like cells yielded from human pluripotent stem cells (hPSCs) hold great potentials for cell therapies of related neurological disorders. However, cues that orchestrate the maintenance versus differentiation of human MGE progenitors, and ways for large-scale expansion of these cells have not been investigated. Here, we report that WNT/CTNNB1 signaling plays an essential role in maintaining MGE-like cells derived from hPSCs. Ablation of CTNNB1 in MGE cells led to precocious cell-cycle exit and advanced neuronal differentiation. Activation of WNT signaling through genetic or chemical approach was sufficient to maintain MGE cells in an expandable manner with authentic neuronal differentiation potencies through activation of endogenous NOTCH signaling. Our findings reveal that WNT/NOTCH signaling cascade is a key player in governing the maintenance versus terminal differentiation of MGE progenitors in humans. Large-scale expansion of functional MGE progenitors for cell therapies can therefore be achieved by modifying WNT/NOTCH pathway.


Subject(s)
Cell Proliferation/physiology , Median Eminence/cytology , Pluripotent Stem Cells/cytology , Receptors, Notch/metabolism , Wnt Signaling Pathway/physiology , Cell Cycle/genetics , Cell Cycle/physiology , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Cell Proliferation/genetics , GABAergic Neurons/cytology , GABAergic Neurons/metabolism , Gene Expression Profiling/methods , Gene Regulatory Networks , Humans , Interneurons/cytology , Interneurons/metabolism , Pluripotent Stem Cells/metabolism , Receptors, Notch/genetics , Wnt Signaling Pathway/genetics , beta Catenin/genetics , beta Catenin/metabolism
15.
J Neuroendocrinol ; 31(9): e12723, 2019 09.
Article in English | MEDLINE | ID: mdl-31034718

ABSTRACT

Although the melanin-concentrating hormone (MCH) and its coding mRNA are predominantly found in the tuberal hypothalamus, there is detectable synthesis of MCH in the preoptic hypothalamus exclusively in lactating dams, suggesting a participation of MCH in the alterations that take place after parturition. Also implicated in the dam physiology is oxytocin, a neurohormone released from the posterior pituitary that is necessary for milk ejection. Because the projection fields from oxytocin-immunoreactive (-IR) neurones and the mediobasal preoptic hypothalamus overlap and MCH-IR neurones are found in proximity to oxytocin neurones, we investigated the spatial relationship between MCH and oxytocin fibres. Accordingly, we employed multiple immunohistochemistry labelling for MCH and oxytocin for light and electron microscopy techniques, in addition to i.v. tracer injection combined with in situ hybridisation to identify MCH neurones that project to neurosecretory areas. As described for other strains, lactating Long-Evans dams also display immunoreactivity for MCH in the preoptic hypothalamus on days 12 and 19 of lactation. The appearance of these neurones is contemporaneous with an increase in MCH-IR fibres in both the internal layer of the median eminence and the posterior pituitary. In both regions, MCH- and oxytocin-IR fibres were found in great proximity, although there was no evidence for synaptic interaction between these two populations at the ultrastructural level. The tracer injection revealed that only mediobasal preoptic MCH neurones project to the posterior pituitary, suggesting a neuroendocrine-modulatory role for this population. When taken together, the results obtained in the present study indicate that neuroplasticity events at the mediobasal preoptic hypothalamus that occur during late lactation may be part of a neuroendocrinology control loop involving both MCH and oxytocin.


Subject(s)
Hypothalamic Hormones/metabolism , Median Eminence/cytology , Median Eminence/metabolism , Melanins/metabolism , Pituitary Gland/cytology , Pituitary Gland/metabolism , Pituitary Hormones/metabolism , Animals , Female , Lactation/metabolism , Oxytocin/metabolism , Protein Precursors/metabolism , RNA, Messenger/metabolism , Rats, Long-Evans
16.
J Neurosci ; 39(1): 177-192, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30377227

ABSTRACT

The CCCTC-binding factor (CTCF) is a central regulator of chromatin topology recently linked to neurodevelopmental disorders such as intellectual disability, autism, and schizophrenia. The aim of this study was to identify novel roles of CTCF in the developing mouse brain. We provide evidence that CTCF is required for the expression of the LIM homeodomain factor LHX6 involved in fate determination of cortical interneurons (CINs) that originate in the medial ganglionic eminence (MGE). Conditional Ctcf ablation in the MGE of mice of either sex leads to delayed tangential migration, abnormal distribution of CIN in the neocortex, a marked reduction of CINs expressing parvalbumin and somatostatin (Sst), and an increased number of MGE-derived cells expressing Lhx8 and other markers of basal forebrain projection neurons. Likewise, Ctcf-null MGE cells transplanted into the cortex of wild-type hosts generate fewer Sst-expressing CINs and exhibit lamination defects that are efficiently rescued upon reexpression of LHX6. Collectively, these data indicate that CTCF regulates the dichotomy between Lhx6 and Lhx8 to achieve correct specification and migration of MGE-derived CINs.SIGNIFICANCE STATEMENT This work provides evidence that CCCTC-binding factor (CTCF) controls an early fate decision point in the generation of cortical interneurons mediated at least in part by Lhx6. Importantly, the abnormalities described could reflect early molecular and cellular events that contribute to human neurological disorders previously linked to CTCF, including schizophrenia, autism, and intellectual disability.


Subject(s)
CCCTC-Binding Factor/physiology , Cerebral Cortex/physiology , Interneurons/physiology , Median Eminence/physiology , Animals , CCCTC-Binding Factor/genetics , Cell Count , Cell Movement/genetics , Cell Movement/physiology , Cerebral Cortex/cytology , Female , LIM-Homeodomain Proteins/biosynthesis , LIM-Homeodomain Proteins/genetics , Male , Median Eminence/cytology , Mice , Mice, Inbred C57BL , Neocortex/cytology , Neocortex/physiology , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Parvalbumins/metabolism , Somatostatin/metabolism , Telencephalon/cytology , Telencephalon/growth & development , Transcription Factors/biosynthesis , Transcription Factors/genetics , gamma-Aminobutyric Acid/physiology
17.
Mol Neurobiol ; 56(6): 4440-4454, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30334186

ABSTRACT

The development of inhibitory circuits depends on the action of a network of transcription factors and epigenetic regulators that are critical for interneuron specification and differentiation. Although the identity of many of these transcription factors is well established, much less is known about the specific contribution of the chromatin-modifying enzymes that sculpt the interneuron epigenome. Here, we generated a mouse model in which the lysine acetyltransferase CBP is specifically removed from neural progenitors at the median ganglionic eminence (MGE), the structure where the most abundant types of cortical interneurons are born. Ablation of CBP interfered with the development of MGE-derived interneurons in both sexes, causing a reduction in the number of functionally mature interneurons in the adult forebrain. Genetic fate mapping experiments not only demonstrated that CBP ablation impacts on different interneuron classes, but also unveiled a compensatory increment of interneurons that escaped recombination and cushion the excitatory-inhibitory imbalance. Consistent with having a reduced number of interneurons, CBP-deficient mice exhibited a high incidence of spontaneous epileptic seizures, and alterations in brain rhythms and enhanced low gamma activity during status epilepticus. These perturbations led to abnormal behavior including hyperlocomotion, increased anxiety and cognitive impairments. Overall, our study demonstrates that CBP is essential for interneuron development and the proper functioning of inhibitory circuitry in vivo.


Subject(s)
Cell Differentiation/genetics , Epigenesis, Genetic , Interneurons/cytology , Median Eminence/cytology , Membrane Proteins/metabolism , Phosphoproteins/metabolism , Action Potentials , Animals , Anxiety/complications , Anxiety/physiopathology , Behavior, Animal , Chromosome Mapping , Cognition Disorders/complications , Cognition Disorders/physiopathology , Epilepsy/complications , Epilepsy/pathology , Epilepsy/physiopathology , Female , Hippocampus/metabolism , Interneurons/metabolism , Kainic Acid , Learning , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Parvalbumins/metabolism , Somatostatin/metabolism , Thyroid Nuclear Factor 1/metabolism
18.
Cereb Cortex ; 29(6): 2653-2667, 2019 06 01.
Article in English | MEDLINE | ID: mdl-29878134

ABSTRACT

Immature neurons generated by the subpallial MGE tangentially migrate to the cortex where they become parvalbumin-expressing (PV+) and somatostatin (SST+) interneurons. Here, we show that the Sp9 transcription factor controls the development of MGE-derived cortical interneurons. SP9 is expressed in the MGE subventricular zone and in MGE-derived migrating interneurons. Sp9 null and conditional mutant mice have approximately 50% reduction of MGE-derived cortical interneurons, an ectopic aggregation of MGE-derived neurons in the embryonic ventral telencephalon, and an increased ratio of SST+/PV+ cortical interneurons. RNA-Seq and SP9 ChIP-Seq reveal that SP9 regulates MGE-derived cortical interneuron development through controlling the expression of key transcription factors Arx, Lhx6, Lhx8, Nkx2-1, and Zeb2 involved in interneuron development, as well as genes implicated in regulating interneuron migration Ackr3, Epha3, and St18. Thus, Sp9 has a central transcriptional role in MGE-derived cortical interneuron development.


Subject(s)
Cerebral Cortex/cytology , Interneurons/cytology , Median Eminence/embryology , Neurogenesis/physiology , RNA-Binding Proteins/metabolism , Animals , Cell Movement/physiology , Cerebral Cortex/embryology , Interneurons/metabolism , Median Eminence/cytology , Mice , Transcription Factors/metabolism
19.
Nat Commun ; 9(1): 4595, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30389944

ABSTRACT

Diverse γ-aminobutyric acid (GABA)-ergic interneurons provide different modes of inhibition to support circuit operation in the neocortex. However, the cellular and molecular mechanisms underlying the systematic generation of assorted neocortical interneurons remain largely unclear. Here we show that NKX2.1-expressing radial glial progenitors (RGPs) in the mouse embryonic ventral telencephalon divide progressively to generate distinct groups of interneurons, which occupy the neocortex in a time-dependent, early inside-out and late outside-in, manner. Notably, the late-born chandelier cells, one of the morphologically and physiologically highly distinguishable GABAergic interneurons, arise reliably from continuously dividing RGPs that produce non-chandelier cells initially. Selective removal of Partition defective 3, an evolutionarily conserved cell polarity protein, impairs RGP asymmetric cell division, resulting in premature depletion of RGPs towards the late embryonic stages and a consequent loss of chandelier cells. These results suggest that consecutive asymmetric divisions of multipotent RGPs generate diverse neocortical interneurons in a progressive manner.


Subject(s)
Cell Division , Neocortex/cytology , Neural Stem Cells/cytology , Neurogenesis , Adaptor Proteins, Signal Transducing , Asymmetric Cell Division , Cell Adhesion Molecules/metabolism , Cell Cycle Proteins , Interneurons/cytology , Median Eminence/cytology , Neuroglia/cytology , Neuroglia/metabolism , Preoptic Area/cytology , Staining and Labeling , Thyroid Nuclear Factor 1/metabolism
20.
Endocrinology ; 159(11): 3723-3736, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30272161

ABSTRACT

The pulsatile release of GnRH and LH secretion is essential for fertility in all mammals. Pulses of LH occur approximately every hour in follicular-phase females and every 2 to 3 hours in luteal-phase females and males. Many studies over the last 50 years have sought to identify the nature and mechanism of the "GnRH pulse generator" responsible for pulsatile LH release. This review examines the characteristics of pulsatile hormone release and summarizes investigations that have led to our present understanding of the GnRH pulse generator. There is presently little compelling evidence for an intrinsic mechanism of pulse generation involving interactions between GnRH neuron cell bodies. Rather, data support the presence of an extrinsic pulse generator located within the arcuate nucleus, and attention has focused on the kisspeptin neurons and their projections to GnRH neuron dendrons concentrated around the median eminence. Sufficient evidence has been gathered in rodents to conclude that a subpopulation of arcuate kisspeptin neurons is, indeed, the GnRH pulse generator. Findings in other species are generally compatible with this view and suggest that arcuate/infundibular kisspeptin neurons represent the mammalian GnRH pulse generator. With hindsight, it is likely that past arcuate nucleus multiunit activity recordings have been from kisspeptin neurons. Despite advances in identifying the cells forming the pulse generator, almost nothing is known about their mechanisms of synchronicity and the afferent hormonal and transmitter modulation required to establish the normal patterns of LH pulsatility in mammals.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Gonadotropin-Releasing Hormone/metabolism , Luteinizing Hormone/metabolism , Neurons/metabolism , Animals , Arcuate Nucleus of Hypothalamus/cytology , Female , Follicular Phase/metabolism , Humans , Kisspeptins , Luteal Phase/metabolism , Male , Median Eminence/cytology , Median Eminence/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL