Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 258
Filtrar
1.
Nature ; 628(8009): 826-834, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38538787

RESUMEN

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.


Asunto(s)
Tronco Encefálico , Células Ependimogliales , Conducta Alimentaria , Calor , Hipotálamo , Vías Nerviosas , Neuronas , Animales , Femenino , Masculino , Ratones , Proteína Relacionada con Agouti/metabolismo , Núcleo Arqueado del Hipotálamo/metabolismo , Núcleo Arqueado del Hipotálamo/citología , Tronco Encefálico/citología , Tronco Encefálico/fisiología , Dopamina/metabolismo , Ingestión de Alimentos/fisiología , Células Ependimogliales/citología , Células Ependimogliales/fisiología , Conducta Alimentaria/fisiología , Ácido Glutámico/metabolismo , Hipotálamo/citología , Hipotálamo/fisiología , Vías Nerviosas/metabolismo , Neuronas/metabolismo , Núcleos Parabraquiales/citología , Núcleos Parabraquiales/metabolismo , Núcleos Parabraquiales/fisiología , Sensación Térmica/fisiología , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular/líquido cefalorraquídeo , Factor A de Crecimiento Endotelial Vascular/metabolismo
2.
Biochem Biophys Res Commun ; 636(Pt 2): 79-86, 2022 12 25.
Artículo en Inglés | MEDLINE | ID: mdl-36368158

RESUMEN

During mammalian retinal development, the differentiation of multipotent progenitors depends on the coordinated action of a variety of intrinsic factors including non-coding RNAs (ncRNAs). To date, many small open reading frames have been identified in ncRNAs to encode micropeptides that function in diverse biological processes; however, it remains unclear whether they have a role in retinal development. Here we report that the 47-amino acid (AA) mitochondrial micropeptide Stmp1 encoded by the lncRNA 1810058I24Rik is involved in retinal differentiation. As the major protein product of 1810058I24Rik, Stmp1 promotes the differentiation of bipolar, amacrine and Müller cells as 1810058I24Rik does when overexpressed in neonatal murine retinas. Moreover, we have identified the 15-AA N-terminus of Stmp1 as its mitochondrion-targeting sequence as well as 5 conserved AA residues that affect protein stability and/or retinal cell differentiation. Together, our data reveal several novel characteristics of Stmp1 and uncover a role for Stmp1 in retinal cell differentiation perhaps through regulating mitochondrial function.


Asunto(s)
Diferenciación Celular , Péptidos y Proteínas de Señalización Intracelular , Mitocondrias , Proteínas Mitocondriales , Retina , Animales , Ratones , Células Ependimogliales/citología , Mitocondrias/metabolismo , Neuronas/citología , Retina/citología , ARN no Traducido/genética , Proteínas Mitocondriales/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología
3.
Science ; 377(6611): eabl6422, 2022 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-36074851

RESUMEN

Neanderthal brains were similar in size to those of modern humans. We sought to investigate potential differences in neurogenesis during neocortex development. Modern human transketolase-like 1 (TKTL1) differs from Neanderthal TKTL1 by a lysine-to-arginine amino acid substitution. Using overexpression in developing mouse and ferret neocortex, knockout in fetal human neocortical tissue, and genome-edited cerebral organoids, we found that the modern human variant, hTKTL1, but not the Neanderthal variant, increases the abundance of basal radial glia (bRG) but not that of intermediate progenitors (bIPs). bRG generate more neocortical neurons than bIPs. The hTKTL1 effect requires the pentose phosphate pathway and fatty acid synthesis. Inhibition of these metabolic pathways reduces bRG abundance in fetal human neocortical tissue. Our data suggest that neocortical neurogenesis in modern humans differs from that in Neanderthals.


Asunto(s)
Hombre de Neandertal , Neocórtex , Neurogénesis , Transcetolasa , Animales , Células Ependimogliales/citología , Hurones , Humanos , Ratones , Hombre de Neandertal/embriología , Hombre de Neandertal/genética , Neocórtex/embriología , Neurogénesis/genética , Neurogénesis/fisiología , Transcetolasa/genética , Transcetolasa/metabolismo
4.
Proc Natl Acad Sci U S A ; 119(38): e2206147119, 2022 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-36095192

RESUMEN

The neocortex, the center for higher brain function, first emerged in mammals and has become massively expanded and folded in humans, constituting almost half the volume of the human brain. Primary microcephaly, a developmental disorder in which the brain is smaller than normal at birth, results mainly from there being fewer neurons in the neocortex because of defects in neural progenitor cells (NPCs). Outer radial glia (oRGs), NPCs that are abundant in gyrencephalic species but rare in lissencephalic species, are thought to play key roles in the expansion and folding of the neocortex. However, how oRGs expand, whether they are necessary for neocortical folding, and whether defects in oRGs cause microcephaly remain important questions in the study of brain development, evolution, and disease. Here, we show that oRG expansion in mice, ferrets, and human cerebral organoids requires cyclin-dependent kinase 6 (CDK6), the mutation of which causes primary microcephaly via an unknown mechanism. In a mouse model in which increased Hedgehog signaling expands oRGs and intermediate progenitor cells and induces neocortical folding, CDK6 loss selectively decreased oRGs and abolished neocortical folding. Remarkably, this function of CDK6 in oRG expansion did not require its kinase activity, was not shared by the highly similar CDK4 and CDK2, and was disrupted by the mutation causing microcephaly. Therefore, our results indicate that CDK6 is conserved to promote oRG expansion, that oRGs are necessary for neocortical folding, and that defects in oRG expansion may cause primary microcephaly.


Asunto(s)
Quinasa 6 Dependiente de la Ciclina , Células Ependimogliales , Microcefalia , Neocórtex , Animales , Quinasa 6 Dependiente de la Ciclina/genética , Quinasa 6 Dependiente de la Ciclina/metabolismo , Células Ependimogliales/citología , Células Ependimogliales/enzimología , Hurones , Proteínas Hedgehog/metabolismo , Humanos , Ratones , Microcefalia/genética , Neocórtex/anomalías , Neocórtex/enzimología , Células-Madre Neurales/citología , Células-Madre Neurales/enzimología , Organoides/embriología
5.
Exp Eye Res ; 217: 108958, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35085579

RESUMEN

The purpose of this study was to investigate Müller cells during the fetal development of the human eye. Müller cells in eyes of 39 human fetuses (11-38 weeks of gestation, WOG) and 6 infants (5 died of abusive head trauma, AHT, aged 1-9 months) were immunohistochemically stained and investigated for spatial and temporal immunoreaction of nestin, CD44, collagen IX and GFAP, which are stem cell markers or markers of intermediate filaments, respectively, in one of the hitherto largest cohorts of fetal eyes. Müller cells could be detected immunohistochemically as early as 12 WOG by immunohistochemical staining with nestin. Nestin was more strongly expressed in Müller cells of the peripheral retina and a centroperipheral gradient of immunoreaction over time was observed. CD44 was predominantly expressed in fetal eyes of the late second and early third trimester between (23 and 27 WOG) and significantly stronger in the infant eyes. Collagen IX labeling in the central retina was significantly stronger than in more peripheral sectors and increased with fetal age. GFAP staining in Müller cells was seen in the eye of a fetus of 38 WOG who died postnatally and in the infant eyes with and without history of AHT. Additionally, GFAP staining was present in the astrocytes of fetal and infant eyes. All examined markers were expressed by Müller cells at different developmental stages highlighting the plasticity of Müller cells during the development of the human eye. GFAP should be cautiously used as a marker for AHT as it was also expressed in fetal astrocytes and Müller cells in eyes without history of AHT.


Asunto(s)
Colágeno Tipo IX , Células Ependimogliales , Proteína Ácida Fibrilar de la Glía , Receptores de Hialuranos , Nestina , Retina , Colágeno Tipo IX/metabolismo , Células Ependimogliales/citología , Células Ependimogliales/metabolismo , Feto , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Receptores de Hialuranos/metabolismo , Lactante , Nestina/metabolismo , Neuroglía/citología , Neuroglía/metabolismo , Retina/embriología , Retina/metabolismo
6.
J Neuroendocrinol ; 34(1): e13079, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34970803

RESUMEN

In addition to the hypophysiotropic thyrotropin-releasing hormone (TRH)-synthesizing neurons, a glial cell type, the tanycytes, also play a role in the regulation of the hypothalamic-pituitary-thyroid (HPT) axis. Tanycytes modulate the feedback regulation of the axis by regulating the local thyroid hormone availability in the median eminence where the hypophysiotropic axons terminate. Recently, we showed that tanycytes produce diacylglycerol lipase alpha (DAGLα), the synthesizing enzyme of the endocannabinoid 2-arachidonoylglycerol (2-AG) that inhibits the release of TRH from the hypophysiotropic terminals in median eminence explants. To determine the importance of the endocannabinoid production of tanycytes, adult male Rax-CreERT2//DAGLαfl/fl mice were treated with tamoxifen to induce a tanycyte specific decrease of DAGLα expression (T-DAGLα KO). The effect of this genetic manipulation on the activity of the HPT axis was determined. Tanycyte specific decrease of DAGLα expression resulted in an approximately 2-fold increase of TSHß mRNA level that was accompanied by increased levels of circulating free T4. The TRH mRNA level was, however, not influenced by the genetic manipulation. In addition to the effects on the HPT axis, the T-DAGLα KO mice showed increased fat mass ratio and decreased blood glucose levels. These data indicate that when endocannabinoid release of tanycytes is decreased, the disinhibition of the TRH release induces increased TSH synthesis and higher circulating T4 levels. Thus it suggests that in wild-type mice, tanycytes exert a tonic inhibitory effect on the TRH release of hypophysiotropic axons. Furthermore, the endocannabinoid release of tanycytes also influences glucose homeostasis and fat deposition.


Asunto(s)
Células Ependimogliales/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Lipoproteína Lipasa/genética , Glándula Tiroides/metabolismo , Hormona Liberadora de Tirotropina/metabolismo , Animales , Endocannabinoides/farmacología , Células Ependimogliales/citología , Regulación Enzimológica de la Expresión Génica/fisiología , Técnicas de Inactivación de Genes/métodos , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Lipoproteína Lipasa/metabolismo , Masculino , Ratones , Ratones Transgénicos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Glándula Tiroides/efectos de los fármacos , Glándula Tiroides/fisiología
7.
Cells ; 10(11)2021 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-34831460

RESUMEN

During embryonic development, progenitor cells are progressively restricted in their potential to generate different neural cells. A specific progenitor cell type, the radial glial cells, divides symmetrically and then asymmetrically to produce neurons, astrocytes, oligodendrocytes, and NG2-glia in the cerebral cortex. However, the potential of individual progenitors to form glial lineages remains poorly understood. To further investigate the cell progeny of single pallial GFAP-expressing progenitors, we used the in vivo genetic lineage-tracing method, the UbC-(GFAP-PB)-StarTrack. After targeting those progenitors in embryonic mice brains, we tracked their adult glial progeny in lower cortical layers. Clonal analyses revealed the presence of clones containing sibling cells of either a glial cell type (uniform clones) or two different glial cell types (mixed clones). Further, the clonal size and rostro-caudal cell dispersion of sibling cells differed depending on the cell type. We concluded that pallial E14 neural progenitors are a heterogeneous cell population with respect to which glial cell type they produce, as well as the clonal size of their cell progeny.


Asunto(s)
Corteza Cerebral/citología , Células Ependimogliales/citología , Neurogénesis , Envejecimiento/fisiología , Animales , Linaje de la Célula , Células Clonales , Femenino , Ratones Endogámicos C57BL , Células-Madre Neurales/citología , Embarazo
8.
Cells ; 10(10)2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34685774

RESUMEN

The central nervous system of adult zebrafish displays an extraordinary neurogenic and regenerative capacity. In the zebrafish adult brain, this regenerative capacity relies on neural stem cells (NSCs) and the careful management of the NSC pool. However, the mechanisms controlling NSC pool maintenance are not yet fully understood. Recently, Bone Morphogenetic Proteins (BMPs) and their downstream effector Id1 (Inhibitor of differentiation 1) were suggested to act as key players in NSC maintenance under constitutive and regenerative conditions. Here, we further investigated the role of BMP/Id1 signaling in these processes, using different genetic and pharmacological approaches. Our data show that BMPs are mainly expressed by neurons in the adult telencephalon, while id1 is expressed in NSCs, suggesting a neuron-NSC communication via the BMP/Id1 signaling axis. Furthermore, manipulation of BMP signaling by conditionally inducing or repressing BMP signaling via heat-shock, lead to an increase or a decrease of id1 expression in the NSCs, respectively. Induction of id1 was followed by an increase in the number of quiescent NSCs, while knocking down id1 expression caused an increase in NSC proliferation. In agreement, genetic ablation of id1 function lead to increased proliferation of NSCs, followed by depletion of the stem cell pool with concomitant failure to heal injuries in repeatedly injured mutant telencephala. Moreover, pharmacological inhibition of BMP and Notch signaling suggests that the two signaling systems cooperate and converge onto the transcriptional regulator her4.1. Interestingly, brain injury lead to a depletion of NSCs in animals lacking BMP/Id1 signaling despite an intact Notch pathway. Taken together, our data demonstrate how neurons feedback on NSC proliferation and that BMP1/Id1 signaling acts as a safeguard of the NSC pool under regenerative conditions.


Asunto(s)
Envejecimiento/fisiología , Proteínas Morfogenéticas Óseas/metabolismo , Comunicación Celular , Células Ependimogliales/citología , Neuronas/citología , Regeneración/fisiología , Telencéfalo/fisiopatología , Proteínas de Pez Cebra/metabolismo , Animales , Ciclo Celular/genética , Proliferación Celular , Regulación del Desarrollo de la Expresión Génica , Células-Madre Neurales/citología , Receptores Notch/metabolismo , Transducción de Señal , Telencéfalo/lesiones , Telencéfalo/patología , Pez Cebra/genética , Proteínas de Pez Cebra/genética
9.
Mol Cell Endocrinol ; 538: 111449, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34478806

RESUMEN

Ghrelin is a peptide hormone mainly secreted from gastrointestinal tract that acts via the growth hormone secretagogue receptor (GHSR), which is highly expressed in the brain. Strikingly, the accessibility of ghrelin to the brain seems to be limited and restricted to few brain areas. Previous studies in mice have shown that ghrelin can access the brain via the blood-cerebrospinal fluid (CSF) barrier, an interface constituted by the choroid plexus and the hypothalamic tanycytes. Here, we performed a variety of in vivo and in vitro studies to test the hypothesis that the transport of ghrelin across the blood-CSF barrier occurs in a GHSR-dependent manner. In vivo, we found that the uptake of systemically administered fluorescent ghrelin in the choroid plexus epithelial (CPE) cells and in hypothalamic tanycytes depends on the presence of GHSR. Also, we detected lower levels of CSF ghrelin after a systemic ghrelin injection in GHSR-deficient mice, as compared to WT mice. In vitro, the internalization of fluorescent ghrelin was reduced in explants of choroid plexus from GHSR-deficient mice, and unaffected in primary cultures of hypothalamic tanycytes derived from GHSR-deficient mice. Finally, we found that the GHSR mRNA is detected in a pool of CPE cells, but is nearly undetectable in hypothalamic tanycytes with current approaches. Thus, our results suggest that circulating ghrelin crosses the blood-CSF barrier mainly by a mechanism that involves the GHSR, and also possibly via a GHSR-independent mechanism.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Ghrelina/sangre , Ghrelina/líquido cefalorraquídeo , Receptores de Ghrelina/metabolismo , Animales , Células Cultivadas , Plexo Coroideo/metabolismo , Células Ependimogliales/citología , Células Ependimogliales/metabolismo , Ghrelina/genética , Ratones , Cultivo Primario de Células , Transducción de Señal
10.
Cells ; 10(8)2021 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-34440741

RESUMEN

Retinal detachment (RD) is a sight-threatening condition, leading to photoreceptor cell death; however, only a few studies provide insight into its effects on the entire retinal region. We examined the spatiotemporal changes in glial responses in a mouse RD model. In electroretinography, a- and b-waves were reduced in a time-dependent manner. Hematoxylin and eosin staining revealed a gradual decrease in the outer nuclear layer throughout the retinal region. Terminal deoxynucleotidyltransferase dUTP nick end labeling (TUNEL) assay showed that TUNEL-positive photoreceptors increased 5 days after RD and decreased by 14 days. Glial response was evaluated by immunohistochemistry using antibodies against glial fibrillary acidic protein (GFAP, Müller glial marker) and Iba-1 (microglial marker) and osteopontin (OPN, activated microglial marker). GFAP immunoreactivity increased after 7 days in complete RD, and was retained for 14 days. OPN expression increased in microglial cells 3-7 days after RD, and decreased by 14 days in the detached and border regions. Although OPN was not expressed in the intact region, morphologically activated microglial cells were observed. These retinal glial cell responses and photoreceptor degeneration in the border and intact regions suggest that the effects of RD in the border and intact retinal regions need to be understood further.


Asunto(s)
Células Ependimogliales/metabolismo , Microglía/metabolismo , Desprendimiento de Retina/patología , Animales , Proteínas de Unión al Calcio/metabolismo , Modelos Animales de Enfermedad , Electrorretinografía , Células Ependimogliales/citología , Proteína Ácida Fibrilar de la Glía/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/metabolismo , Microglía/citología , Osteopontina/metabolismo , Retina/metabolismo , Retina/patología , Desprendimiento de Retina/metabolismo , Regulación hacia Arriba
11.
J Neurosci ; 41(33): 6969-6986, 2021 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-34266896

RESUMEN

Radial glial progenitor cells (RGCs) in the dorsal telencephalon directly or indirectly produce excitatory projection neurons and macroglia of the neocortex. Recent evidence shows that the pool of RGCs is more heterogeneous than originally thought and that progenitor subpopulations can generate particular neuronal cell types. Using single-cell RNA sequencing, we have studied gene expression patterns of RGCs with different neurogenic behavior at early stages of cortical development. At this early age, some RGCs rapidly produce postmitotic neurons, whereas others self-renew and undergo neurogenic divisions at a later age. We have identified candidate genes that are differentially expressed among these early RGC subpopulations, including the transcription factor Sox9. Using in utero electroporation in embryonic mice of either sex, we demonstrate that elevated Sox9 expression in progenitors affects RGC cell cycle duration and leads to the generation of upper layer cortical neurons. Our data thus reveal molecular differences between progenitor cells with different neurogenic behavior at early stages of corticogenesis and indicates that Sox9 is critical for the maintenance of RGCs to regulate the generation of upper layer neurons.SIGNIFICANCE STATEMENT The existence of heterogeneity in the pool of RGCs and its relationship with the generation of cellular diversity in the cerebral cortex has been an interesting topic of debate for many years. Here we describe the existence of RGCs with reduced neurogenic behavior at early embryonic ages presenting a particular molecular signature. This molecular signature consists of differential expression of some genes including the transcription factor Sox9, which has been found to be a specific regulator of this subpopulation of progenitor cells. Functional experiments perturbing expression levels of Sox9 reveal its instructive role in the regulation of the neurogenic behavior of RGCs and its relationship with the generation of upper layer projection neurons at later ages.


Asunto(s)
Autorrenovación de las Células/genética , Células Ependimogliales/citología , Regulación del Desarrollo de la Expresión Génica/genética , Neocórtex/citología , Proteínas del Tejido Nervioso/fisiología , Neurogénesis/genética , Factor de Transcripción SOX9/fisiología , Animales , Ciclo Celular/genética , Electroporación , Células Ependimogliales/metabolismo , Femenino , Genes Reporteros , Vectores Genéticos/administración & dosificación , Inyecciones Intraventriculares , Ratones , Ratones Endogámicos C57BL , Neocórtex/embriología , Neocórtex/crecimiento & desarrollo , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Neuroglía/citología , Neuronas/citología , Embarazo , Regiones Promotoras Genéticas/genética , Factor de Transcripción SOX9/biosíntesis , Factor de Transcripción SOX9/genética , Análisis de la Célula Individual , Transcripción Genética
12.
Biomolecules ; 11(5)2021 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-34068807

RESUMEN

To investigate the mechanism of vascular endothelial growth factor (VEGF) and brain-derived neurotrophic factor (BDNF) in Müller cell (MC) viability and neuroprotection in diabetic retinopathy (DR), we examined the role of VEGF in MC viability and BDNF production, and the effect of BDNF on MC viability under diabetic conditions. Mouse primary MCs and cells of a rat MC line, rMC1, were used in investigating MC viability and BDNF production under diabetic conditions. VEGF-stimulated BDNF production was confirmed in mice. The mechanism of BDNF-mediated MC viability was examined using siRNA knockdown. Under diabetic conditions, recombinant VEGF (rVEGF) stimulated MC viability and BDNF production in a dose-dependent manner. rBDNF also supported MC viability in a dose-dependent manner. Targeting BDNF receptor tropomyosin receptor kinase B (TRK-B) with siRNA knockdown substantially downregulated the activated (phosphorylated) form of serine/threonine-specific protein kinase (AKT) and extracellular signal-regulated kinase (ERK), classical survival and proliferation mediators. Finally, the loss of MC viability in TrkB siRNA transfected cells under diabetic conditions was rescued by rBDNF. Our results provide direct evidence that VEGF is a positive regulator for BDNF production in diabetes for the first time. This information is essential for developing BDNF-mediated neuroprotection in DR and hypoxic retinal diseases, and for improving anti-VEGF treatment for these blood-retina barrier disorders, in which VEGF is a major therapeutic target for vascular abnormalities.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Retinopatía Diabética/tratamiento farmacológico , Células Ependimogliales/citología , Fármacos Neuroprotectores/farmacología , Receptor trkB/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología , Animales , Supervivencia Celular/fisiología , Células Cultivadas , Retinopatía Diabética/metabolismo , Retinopatía Diabética/patología , Células Ependimogliales/efectos de los fármacos , Células Ependimogliales/metabolismo , Ratones , Ratas , Transducción de Señal , Receptor de Retrovirus Xenotrópico y Politrópico
13.
Nat Neurosci ; 24(8): 1089-1099, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34083786

RESUMEN

Methods to enhance adult neurogenesis by reprogramming glial cells into neurons enable production of new neurons in the adult nervous system. Development of therapeutically viable approaches to induce new neurons is now required to bring this concept to clinical application. Here, we successfully generate new neurons in the cortex and dentate gyrus of the aged adult mouse brain by transiently suppressing polypyrimidine tract binding protein 1 using an antisense oligonucleotide delivered by a single injection into cerebral spinal fluid. Radial glial-like cells and other GFAP-expressing cells convert into new neurons that, over a 2-month period, acquire mature neuronal character in a process mimicking normal neuronal maturation. The new neurons functionally integrate into endogenous circuits and modify mouse behavior. Thus, generation of new neurons in the dentate gyrus of the aging brain can be achieved with a therapeutically feasible approach, thereby opening prospects for production of neurons to replace those lost to neurodegenerative disease.


Asunto(s)
Giro Dentado , Células Ependimogliales , Neurogénesis/fisiología , Neuronas , Proteína de Unión al Tracto de Polipirimidina/antagonistas & inhibidores , Animales , Reprogramación Celular/fisiología , Giro Dentado/citología , Giro Dentado/fisiología , Células Ependimogliales/citología , Células Ependimogliales/fisiología , Ratones , Neuronas/citología , Neuronas/fisiología , Oligonucleótidos Antisentido
14.
Nat Commun ; 12(1): 3100, 2021 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-34035288

RESUMEN

Hippo signaling is an evolutionarily conserved pathway that restricts growth and regeneration predominantly by suppressing the activity of the transcriptional coactivator Yap. Using a high-throughput phenotypic screen, we identified a potent and non-toxic activator of Yap. In vitro kinase assays show that the compound acts as an ATP-competitive inhibitor of Lats kinases-the core enzymes in Hippo signaling. The substance prevents Yap phosphorylation and induces proliferation of supporting cells in the murine inner ear, murine cardiomyocytes, and human Müller glia in retinal organoids. RNA sequencing indicates that the inhibitor reversibly activates the expression of transcriptional Yap targets: upon withdrawal, a subset of supporting-cell progeny exits the cell cycle and upregulates genes characteristic of sensory hair cells. Our results suggest that the pharmacological inhibition of Lats kinases may promote initial stages of the proliferative regeneration of hair cells, a process thought to be permanently suppressed in the adult mammalian inner ear.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proliferación Celular/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Proliferación Celular/genética , Células Ependimogliales/citología , Células Ependimogliales/efectos de los fármacos , Células Ependimogliales/metabolismo , Células HEK293 , Células Ciliadas Auditivas Internas/citología , Células Ciliadas Auditivas Internas/efectos de los fármacos , Células Ciliadas Auditivas Internas/metabolismo , Humanos , Ratones Noqueados , Ratones Transgénicos , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteínas Supresoras de Tumor/metabolismo , Proteínas Señalizadoras YAP
15.
Neurosci Lett ; 756: 135978, 2021 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-34023416

RESUMEN

Zebrafish have a greater capacity for adult neurogenesis and brain regeneration than mammals. In the adult zebrafish optic tectum (OT), neuroepithelial-like stem cells (NE) contribute to adult neurogenesis, whereas radial glia (RG) contribute to neuronal regeneration after the stab wound injury. The molecular mechanisms regulated by acetylated histone play important roles in these events; however, the functions of histone acetyltransferase (HAT) require further elucidation. The aim of this study was to study the proliferation and differentiation of neural stem cells (NSCs) following treatment with C646, a HAT EP300 inhibitor, to identify the functions of HAT in adult neurogenesis and neuronal regeneration. C646 treatment decreased acetylation of histone 3 lysine 9 in the adult OT. Under physiological conditions, C646 promoted NE proliferation and generation of newborn neurons. EP300 inhibition promoted RG proliferation but suppressed the generation of newborn neurons after the injury. EP300 inhibition downregulated the Notch target genes her4 and her6, which was correlated with NE and RG proliferation in the adult OT. EP300 inhibition regulates the proliferation and differentiation of NSCs by inhibiting histone acetylation and Notch target genes expression, suggesting that the functions of HAT in neurogenesis are opposite to those of histone deacetylase.


Asunto(s)
Proliferación Celular/fisiología , Histona Acetiltransferasas/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis/fisiología , Colículos Superiores/metabolismo , Animales , Benzoatos/farmacología , Proliferación Celular/efectos de los fármacos , Células Ependimogliales/citología , Células Ependimogliales/efectos de los fármacos , Células Ependimogliales/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Nitrobencenos/farmacología , Pirazolonas/farmacología , Colículos Superiores/citología , Colículos Superiores/efectos de los fármacos , Pez Cebra
16.
Nat Neurosci ; 24(4): 584-594, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33723434

RESUMEN

The human cortex comprises diverse cell types that emerge from an initially uniform neuroepithelium that gives rise to radial glia, the neural stem cells of the cortex. To characterize the earliest stages of human brain development, we performed single-cell RNA-sequencing across regions of the developing human brain, including the telencephalon, diencephalon, midbrain, hindbrain and cerebellum. We identify nine progenitor populations physically proximal to the telencephalon, suggesting more heterogeneity than previously described, including a highly prevalent mesenchymal-like population that disappears once neurogenesis begins. Comparison of human and mouse progenitor populations at corresponding stages identifies two progenitor clusters that are enriched in the early stages of human cortical development. We also find that organoid systems display low fidelity to neuroepithelial and early radial glia cell types, but improve as neurogenesis progresses. Overall, we provide a comprehensive molecular and spatial atlas of early stages of human brain and cortical development.


Asunto(s)
Corteza Cerebral/embriología , Células Ependimogliales/citología , Células-Madre Neurales/citología , Células Neuroepiteliales/citología , Neurogénesis , Animales , Corteza Cerebral/citología , Humanos , Análisis de la Célula Individual
17.
Mol Med Rep ; 23(4)2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33576465

RESUMEN

During embryonic cortical development, radial glial cells (RGCs) are the major source of neurons, and these also serve as a supportive scaffold to guide neuronal migration. Similar to Vimentin, glial fibrillary acidic protein (GFAP) is one of the major intermediate filament proteins present in glial cells. Previous studies confirmed that prenatal ethanol exposure (PEE) significantly affected the levels of GFAP and increased the disassembly of radial glial fibers. GFAPδ is a variant of GFAP that is specifically expressed in RGCs; however, to the best of our knowledge, there are no reports regarding how PEE influences its expression during cortical development. In the present study, the effects of PEE on the expression and distribution of GFAPδ during early cortical development were assessed. It was found that PEE significantly decreased the expression levels of GFAP and GFAPδ. Using double immunostaining, GFAPδ was identified to be specifically expressed in apical and basal RGCs, and was co­localized with other intermediate filament proteins, such as GFAP, Nestin and Vimentin. Additionally, PEE significantly affected the morphology of radial glial fibers and altered the behavior of RGCs. The loss of GFAPδ accelerated the transformation of RGCs into astrocytes. Using co­immunostaining with Ki67 or phospho­histone H3, GFAPδ+ cells were observed to be proliferative or mitotic cells, and ethanol treatment significantly decreased the proliferative or mitotic activities of GFAPδ+ RGCs. Taken together, the results suggested that PEE altered the expression patterns of GFAPδ and impaired the development of radial glial fibers and RGC behavior. The results of the present study provided evidence that GFAPδ may be a promising target to rescue the damage induced by PEE.


Asunto(s)
Astrocitos/efectos de los fármacos , Encéfalo/efectos de los fármacos , Células Ependimogliales/efectos de los fármacos , Etanol/farmacología , Proteína Ácida Fibrilar de la Glía/metabolismo , Animales , Astrocitos/citología , Astrocitos/metabolismo , Encéfalo/embriología , Encéfalo/metabolismo , Proliferación Celular/efectos de los fármacos , Células Ependimogliales/citología , Células Ependimogliales/metabolismo , Femenino , Exposición Materna , Ratones Endogámicos C57BL , Nestina/metabolismo , Neurogénesis/efectos de los fármacos , Embarazo , Vimentina/metabolismo
18.
Brain Struct Funct ; 226(2): 365-379, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33398432

RESUMEN

The dentate gyrus (DG) is a unique brain structure in that neurons can be generated postnatally and integrated within existing circuitry throughout life. The maturation process of these newly generated neurons (granule cells) is modulated by nicotinic acetylcholine receptors (nAChRs) through a variety of mechanisms such as neural stem pool proliferation, cell survival, signal modulation, and dendritic integration. Disrupted nAChR signaling has been implicated in neuropsychiatric and neurodegenerative disorders, potentially via alterations in DG neurogenesis. GABAergic interneurons are known to express nAChRs, predominantly the α7 subtype, and have been shown to shape development, integration, and circuit reorganization of DG granule cells. Therefore, we examined histological and behavioral effects of knocking out α7 nAChRs in GABAergic neurons. Deletion of α7 nAChRs resulted in a reduction of radial glia-like cells within the subgranular zone of the DG and a concomitant trend towards decreased immature neurons, specifically in male mice, as well as sex-dependent changes in several behaviors, including social recognition and spatial learning. Overall, these findings suggest α7 nAChRs expressed in GABAergic neurons play an important role in regulating the adult neural stem cell pool and behavior in a sex-dependent manner. This provides important insight into the mechanisms by which cholinergic dysfunction contributes to the cognitive and behavioral changes associated with neurodevelopmental and neurodegenerative disorders.


Asunto(s)
Cognición/fisiología , Células Ependimogliales/metabolismo , Neuronas GABAérgicas/metabolismo , Conducta Social , Receptor Nicotínico de Acetilcolina alfa 7/genética , Animales , Conducta Animal/fisiología , Recuento de Células , Giro Dentado/citología , Giro Dentado/metabolismo , Proteína Doblecortina , Células Ependimogliales/citología , Femenino , Neuronas GABAérgicas/citología , Masculino , Ratones , Ratones Noqueados , Neurogénesis/fisiología , Factores Sexuales , Aprendizaje Espacial/fisiología , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo
19.
J Chem Neuroanat ; 113: 101920, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33515665

RESUMEN

Hibernation is a seasonal strategy to conserve energy, characterized by modified thermoregulation, an increase in sleep pressure and drastic metabolic changes. Glial cells such as astrocytes and tanycytes are the brain metabolic sensors, but it remains unknown whether they contribute to seasonal expression of hibernation. The onset of hibernation is controlled by an undefined endogenous circannual rhythm in which adenosine plays a role through the activation of the A1 adenosine receptor (A1AR). Seasonal changes in brain levels of adenosine may contribute to an increase in A1AR sensitivity leading to the onset of hibernation. The primary regulator of extracellular adenosine concentration is adenosine kinase, which is located in astrocytes. Using immunohistochemistry to localize and quantify adenosine kinase in Arctic ground squirrels' brain collected during different seasons, we report lower expression of adenosine kinase in the third ventricle tanycytes in winter compared to summer; a similar change was not seen in astrocytes. Moreover, for the first time, we describe adenosine kinase expression in tanycyte cell bodies in the hypothalamus and in the area postrema, both brain regions involved in energy homeostasis. Next we describe seasonal changes in tanycyte morphology in the hypothalamus. Although still speculative, our findings contribute to a model whereby adenosine kinase in tanycytes regulates seasonal changes in extracellular concentration of adenosine underling the seasonal expression of hibernation.


Asunto(s)
Adenosina Quinasa/metabolismo , Células Ependimogliales/metabolismo , Hibernación/fisiología , Hipotálamo/metabolismo , Animales , Forma de la Célula/fisiología , Células Ependimogliales/citología , Hipotálamo/citología , Sciuridae , Estaciones del Año
20.
Stem Cell Reports ; 16(2): 264-280, 2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33513360

RESUMEN

Organoids (ORGs) are increasingly used as models of cerebral cortical development. Here, we compared transcriptome and cellular phenotypes between telencephalic ORGs and monolayers (MONs) generated in parallel from three biologically distinct induced pluripotent stem cell (iPSC) lines. Multiple readouts revealed increased proliferation in MONs, which was caused by increased integrin signaling. MONs also exhibited altered radial glia (RG) polarity and suppression of Notch signaling, as well as impaired generation of intermediate progenitors, outer RG, and cortical neurons, which were all partially reversed by reaggregation of dissociated cells. Network analyses revealed co-clustering of cell adhesion, Notch-related transcripts and their transcriptional regulators in a module strongly downregulated in MONs. The data suggest that ORGs, with respect to MONs, initiate more efficient Notch signaling in ventricular RG owing to preserved cell adhesion, resulting in subsequent generation of intermediate progenitors and outer RG, in a sequence that recapitulates the cortical ontogenetic process.


Asunto(s)
Adhesión Celular , Corteza Cerebral/metabolismo , Células Ependimogliales/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Neurogénesis , Organoides/metabolismo , Transcriptoma , Diferenciación Celular , Corteza Cerebral/citología , Células Ependimogliales/citología , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Células Madre Pluripotentes Inducidas/citología , Integrinas/metabolismo , Masculino , Neuronas/citología , Neuronas/metabolismo , Técnicas de Cultivo de Órganos/métodos , Organoides/citología , Proteoma , RNA-Seq , Receptores Notch/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...