Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.656
Filtrar
1.
Cereb Cortex ; 34(4)2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38572735

RESUMEN

Many studies indicate a broad role of various classes of GABAergic interneurons in the processes related to learning. However, little is known about how the learning process affects intrinsic excitability of specific classes of interneurons in the neocortex. To determine this, we employed a simple model of conditional learning in mice where vibrissae stimulation was used as a conditioned stimulus and a tail shock as an unconditioned one. In vitro whole-cell patch-clamp recordings showed an increase in intrinsic excitability of low-threshold spiking somatostatin-expressing interneurons (SST-INs) in layer 4 (L4) of the somatosensory (barrel) cortex after the conditioning paradigm. In contrast, pseudoconditioning reduced intrinsic excitability of SST-LTS, parvalbumin-expressing interneurons (PV-INs), and vasoactive intestinal polypeptide-expressing interneurons (VIP-INs) with accommodating pattern in L4 of the barrel cortex. In general, increased intrinsic excitability was accompanied by narrowing of action potentials (APs), whereas decreased intrinsic excitability coincided with AP broadening. Altogether, these results show that both conditioning and pseudoconditioning lead to plastic changes in intrinsic excitability of GABAergic interneurons in a cell-specific manner. In this way, changes in intrinsic excitability can be perceived as a common mechanism of learning-induced plasticity in the GABAergic system.


Asunto(s)
Neocórtex , Ratones , Animales , Neocórtex/metabolismo , Interneuronas/fisiología , Aprendizaje/fisiología , Condicionamiento Clásico/fisiología , Parvalbúminas/metabolismo
2.
Methods Mol Biol ; 2794: 187-200, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38630230

RESUMEN

In utero electroporation (IUE) enables labeling and manipulating specific types of cells by introducing DNA plasmids with desired promoters. After the surgery, mouse brains are fixed at any stage and analyzed after staining using specific antibodies. Here, we describe the flow of the IUE experiment from the preparation to microscopic observations.


Asunto(s)
Electroporación , Neocórtex , Animales , Ratones , Diferenciación Celular , Manejo de Especímenes , Anticuerpos
3.
Cereb Cortex ; 34(4)2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38610088

RESUMEN

The axons of neocortical pyramidal neurons are frequently myelinated. Heterogeneity in the topography of axonal myelination in the cerebral cortex has been attributed to a combination of electrophysiological activity, axonal morphology, and neuronal-glial interactions. Previously, we showed that axonal segment length and caliber are critical local determinants of fast-spiking interneuron myelination. However, the factors that determine the myelination of individual axonal segments along neocortical pyramidal neurons remain largely unexplored. Here, we used structured illumination microscopy to examine the extent to which axonal morphology is predictive of the topography of myelination along neocortical pyramidal neurons. We identified critical thresholds for axonal caliber and interbranch distance that are necessary, but not sufficient, for myelination of pyramidal cell axons in mouse primary somatosensory cortex (S1). Specifically, we found that pyramidal neuron axonal segments with a caliber < 0.24 µm or interbranch distance < 18.10 µm are rarely myelinated. Moreover, we further confirmed that these findings in mice are similar for human neocortical pyramidal cell myelination (caliber < 0.25 µm, interbranch distance < 19.00 µm), suggesting that this mechanism is evolutionarily conserved. Taken together, our findings suggest that axonal morphology is a critical correlate of the topography and cell-type specificity of neocortical myelination.


Asunto(s)
Neocórtex , Células Piramidales , Humanos , Animales , Ratones , Axones , Vaina de Mielina , Interneuronas
4.
J Theor Biol ; 588: 111818, 2024 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-38621583

RESUMEN

The standard consolidation theory states that short-term memories located in the hippocampus enable the consolidation of long-term memories in the neocortex. In other words, the neocortex slowly learns long-term memories with a transient support of the hippocampus that quickly learns unstable memories. However, it is not clear yet what could be the neurobiological mechanisms underlying these differences in learning rates and memory time-scales. Here, we propose a novel modeling approach of the standard consolidation theory, that focuses on its potential neurobiological mechanisms. In addition to synaptic plasticity and spike frequency adaptation, our model incorporates adult neurogenesis in the dentate gyrus as well as the difference in size between the neocortex and the hippocampus, that we associate with distance-dependent synaptic plasticity. We also take into account the interconnected spatial structure of the involved brain areas, by incorporating the above neurobiological mechanisms in a coupled neural field framework, where each area is represented by a separate neural field with intra- and inter-area connections. To our knowledge, this is the first attempt to apply neural fields to this process. Using numerical simulations and mathematical analysis, we explore the short-term and long-term dynamics of the model upon alternance of phases of hippocampal replay and retrieval cue of an external input. This external input is encodable as a memory pattern in the form of a multiple bump attractor pattern in the individual neural fields. In the model, hippocampal memory patterns become encoded first, before neocortical ones, because of the smaller distances between the bumps of the hippocampal memory patterns. As a result, retrieval of the input pattern in the neocortex at short time-scales necessitates the additional input delivered by the memory pattern of the hippocampus. Neocortical memory patterns progressively consolidate at longer times, up to a point where their retrieval does not need the support of the hippocampus anymore. At longer times, perturbation of the hippocampal neural fields by neurogenesis erases the hippocampus pattern, leading to a final state where the memory pattern is exclusively evoked in the neocortex. Therefore, the dynamics of our model successfully reproduces the main features of the standard consolidation theory. This suggests that neurogenesis in the hippocampus and distance-dependent synaptic plasticity coupled to synaptic depression and spike frequency adaptation, are indeed critical neurobiological processes in memory consolidation.


Asunto(s)
Hipocampo , Consolidación de la Memoria , Modelos Neurológicos , Plasticidad Neuronal , Plasticidad Neuronal/fisiología , Humanos , Hipocampo/fisiología , Consolidación de la Memoria/fisiología , Neocórtex/fisiología , Animales , Neurogénesis/fisiología
5.
eNeuro ; 11(5)2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38653560

RESUMEN

Forebrain neurons deprived of activity become hyperactive when activity is restored. Rebound activity has been linked to spontaneous seizures in vivo following prolonged activity blockade. Here, we measured the time course of rebound activity and the contributing circuit mechanisms using calcium imaging, synaptic staining, and whole-cell patch clamp in organotypic slice cultures of mouse neocortex. Calcium imaging revealed hypersynchronous activity increasing in intensity with longer periods of deprivation. While activity partially recovered 3 d after slices were released from 5 d of deprivation, they were less able to recover after 10 d of deprivation. However, even after the longer period of deprivation, activity patterns eventually returned to baseline levels. The degree of deprivation-induced rebound was age-dependent, with the greatest effects occurring when silencing began in the second week. Pharmacological blockade of NMDA receptors indicated that hypersynchronous rebound activity did not require activation of Hebbian plasticity. In single-neuron recordings, input resistance roughly doubled with a concomitant increase in intrinsic excitability. Synaptic imaging of pre- and postsynaptic proteins revealed dramatic reductions in the number of presumptive synapses with a larger effect on inhibitory than excitatory synapses. Putative excitatory synapses colocalizing PSD-95 and Bassoon declined by 39 and 56% following 5 and 10 d of deprivation, but presumptive inhibitory synapses colocalizing gephyrin and VGAT declined by 55 and 73%, respectively. The results suggest that with prolonged deprivation, a progressive reduction in synapse number is accompanied by a shift in the balance between excitation and inhibition and increased cellular excitability.


Asunto(s)
Homólogo 4 de la Proteína Discs Large , Neocórtex , Animales , Neocórtex/fisiología , Homólogo 4 de la Proteína Discs Large/metabolismo , Neuronas/fisiología , Neuronas/metabolismo , Técnicas de Cultivo de Órganos , Sinapsis/fisiología , Técnicas de Placa-Clamp , Ratones , Ratones Endogámicos C57BL , Femenino , Calcio/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Factores de Tiempo , Proteínas del Tejido Nervioso
6.
Int J Mol Sci ; 25(7)2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38612801

RESUMEN

The Piezo1 mechanosensitive ion channel is abundant on several elements of the central nervous system including astrocytes. It has been already demonstrated that activation of these channels is able to elicit calcium waves on astrocytes, which contributes to the release of gliotransmitters. Astrocyte- and N-methyl-D-aspartate (NMDA) receptor-dependent slow inward currents (SICs) are hallmarks of astrocyte-neuron communication. These currents are triggered by glutamate released as gliotransmitter, which in turn activates neuronal NMDA receptors responsible for this inward current having slower kinetics than any synaptic events. In this project, we aimed to investigate whether Piezo1 activation and inhibition is able to alter spontaneous SIC activity of murine neocortical pyramidal neurons. When the Piezo1 opener Yoda1 was applied, the SIC frequency and the charge transfer by these events in a minute time was significantly increased. These changes were prevented by treating the preparations with the NMDA receptor inhibitor D-AP5. Furthermore, Yoda1 did not alter the spontaneous EPSC frequency and amplitude when SICs were absent. The Piezo1 inhibitor Dooku1 effectively reverted the actions of Yoda1 and decreased the rise time of SICs when applied alone. In conclusion, activation of Piezo1 channels is able to alter astrocyte-neuron communication. Via enhancement of SIC activity, astrocytic Piezo1 channels have the capacity to determine neuronal excitability.


Asunto(s)
Astrocitos , Neocórtex , Animales , Ratones , Receptores de N-Metil-D-Aspartato , Neuronas , Ácido Glutámico , Canales Iónicos
7.
Nature ; 629(8011): 384-392, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38600385

RESUMEN

Debate remains around the anatomical origins of specific brain cell subtypes and lineage relationships within the human forebrain1-7. Thus, direct observation in the mature human brain is critical for a complete understanding of its structural organization and cellular origins. Here we utilize brain mosaic variation within specific cell types as distinct indicators for clonal dynamics, denoted as cell-type-specific mosaic variant barcode analysis. From four hemispheres and two different human neurotypical donors, we identified 287 and 780 mosaic variants, respectively, that were used to deconvolve clonal dynamics. Clonal spread and allele fractions within the brain reveal that local hippocampal excitatory neurons are more lineage-restricted than resident neocortical excitatory neurons or resident basal ganglia GABAergic inhibitory neurons. Furthermore, simultaneous genome transcriptome analysis at both a cell-type-specific and a single-cell level suggests a dorsal neocortical origin for a subgroup of DLX1+ inhibitory neurons that disperse radially from an origin shared with excitatory neurons. Finally, the distribution of mosaic variants across 17 locations within one parietal lobe reveals that restriction of clonal spread in the anterior-posterior axis precedes restriction in the dorsal-ventral axis for both excitatory and inhibitory neurons. Thus, cell-type-resolved somatic mosaicism can uncover lineage relationships governing the development of the human forebrain.


Asunto(s)
Linaje de la Célula , Neuronas GABAérgicas , Proteínas de Homeodominio , Mosaicismo , Prosencéfalo , Factores de Transcripción , Humanos , Prosencéfalo/citología , Neuronas GABAérgicas/citología , Neuronas GABAérgicas/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Linaje de la Célula/genética , Masculino , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Neuronas/citología , Neuronas/metabolismo , Femenino , Hipocampo/citología , Células Clonales/citología , Células Clonales/metabolismo , Análisis de la Célula Individual , Lóbulo Parietal/citología , Alelos , Neocórtex/citología , Transcriptoma
8.
Nature ; 629(8011): 402-409, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38632412

RESUMEN

Throughout life, neuronal networks in the mammalian neocortex maintain a balance of excitation and inhibition, which is essential for neuronal computation1,2. Deviations from a balanced state have been linked to neurodevelopmental disorders, and severe disruptions result in epilepsy3-5. To maintain balance, neuronal microcircuits composed of excitatory and inhibitory neurons sense alterations in neural activity and adjust neuronal connectivity and function. Here we identify a signalling pathway in the adult mouse neocortex that is activated in response to increased neuronal network activity. Overactivation of excitatory neurons is signalled to the network through an increase in the levels of BMP2, a growth factor that is well known for its role as a morphogen in embryonic development. BMP2 acts on parvalbumin-expressing (PV) interneurons through the transcription factor SMAD1, which controls an array of glutamatergic synapse proteins and components of perineuronal nets. PV-interneuron-specific disruption of BMP2-SMAD1 signalling is accompanied by a loss of glutamatergic innervation in PV cells, underdeveloped perineuronal nets and decreased excitability. Ultimately, this impairment of the functional recruitment of PV interneurons disrupts the cortical excitation-inhibition balance, with mice exhibiting spontaneous epileptic seizures. Our findings suggest that developmental morphogen signalling is repurposed to stabilize cortical networks in the adult mammalian brain.


Asunto(s)
Proteína Morfogenética Ósea 2 , Interneuronas , Neocórtex , Parvalbúminas , Transducción de Señal , Proteína Smad1 , Animales , Proteína Smad1/metabolismo , Ratones , Interneuronas/metabolismo , Neocórtex/metabolismo , Neocórtex/citología , Parvalbúminas/metabolismo , Proteína Morfogenética Ósea 2/metabolismo , Masculino , Femenino , Neuronas/metabolismo , Inhibición Neural , Epilepsia/metabolismo , Epilepsia/fisiopatología , Sinapsis/metabolismo , Red Nerviosa/metabolismo
9.
Proc Natl Acad Sci U S A ; 121(16): e2317783121, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38588430

RESUMEN

GABAergic inhibitory interneurons, originating from the embryonic ventral forebrain territories, traverse a convoluted migratory path to reach the neocortex. These interneuron precursors undergo sequential phases of tangential and radial migration before settling into specific laminae during differentiation. Here, we show that the developmental trajectory of FoxG1 expression is dynamically controlled in these interneuron precursors at critical junctures of migration. By utilizing mouse genetic strategies, we elucidate the pivotal role of precise changes in FoxG1 expression levels during interneuron specification and migration. Our findings underscore the gene dosage-dependent function of FoxG1, aligning with clinical observations of FOXG1 haploinsufficiency and duplication in syndromic forms of autism spectrum disorders. In conclusion, our results reveal the finely tuned developmental clock governing cortical interneuron development, driven by temporal dynamics and the dose-dependent actions of FoxG1.


Asunto(s)
Corteza Cerebral , Neocórtex , Ratones , Animales , Corteza Cerebral/metabolismo , Movimiento Celular/fisiología , Neurogénesis/fisiología , Interneuronas/fisiología , Biomarcadores/metabolismo , Neuronas GABAérgicas/fisiología
10.
Nat Commun ; 15(1): 3468, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38658571

RESUMEN

Metabolism has recently emerged as a major target of genes implicated in the evolutionary expansion of human neocortex. One such gene is the human-specific gene ARHGAP11B. During human neocortex development, ARHGAP11B increases the abundance of basal radial glia, key progenitors for neocortex expansion, by stimulating glutaminolysis (glutamine-to-glutamate-to-alpha-ketoglutarate) in mitochondria. Here we show that the ape-specific protein GLUD2 (glutamate dehydrogenase 2), which also operates in mitochondria and converts glutamate-to-αKG, enhances ARHGAP11B's ability to increase basal radial glia abundance. ARHGAP11B + GLUD2 double-transgenic bRG show increased production of aspartate, a metabolite essential for cell proliferation, from glutamate via alpha-ketoglutarate and the TCA cycle. Hence, during human evolution, a human-specific gene exploited the existence of another gene that emerged during ape evolution, to increase, via concerted changes in metabolism, progenitor abundance and neocortex size.


Asunto(s)
Proteínas Activadoras de GTPasa , Glutamato Deshidrogenasa , Neocórtex , Neocórtex/metabolismo , Neocórtex/embriología , Neocórtex/crecimiento & desarrollo , Neocórtex/citología , Humanos , Animales , Glutamato Deshidrogenasa/metabolismo , Glutamato Deshidrogenasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Proteínas Activadoras de GTPasa/genética , Ácidos Cetoglutáricos/metabolismo , Neuroglía/metabolismo , Ácido Glutámico/metabolismo , Mitocondrias/metabolismo , Mitocondrias/genética , Ratones , Ciclo del Ácido Cítrico/genética , Femenino
11.
Cell Rep ; 43(4): 114056, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38581678

RESUMEN

Little is known of the brain mechanisms that mediate sex-specific autism symptoms. Here, we demonstrate that deletion of the autism spectrum disorder (ASD)-risk gene, Pten, in neocortical pyramidal neurons (NSEPten knockout [KO]) results in robust cortical circuit hyperexcitability selectively in female mice observed as prolonged spontaneous persistent activity states. Circuit hyperexcitability in females is mediated by metabotropic glutamate receptor 5 (mGluR5) and estrogen receptor α (ERα) signaling to mitogen-activated protein kinases (Erk1/2) and de novo protein synthesis. Pten KO layer 5 neurons have a female-specific increase in mGluR5 and mGluR5-dependent protein synthesis. Furthermore, mGluR5-ERα complexes are generally elevated in female cortices, and genetic reduction of ERα rescues enhanced circuit excitability, protein synthesis, and neuron size selectively in NSEPten KO females. Female NSEPten KO mice display deficits in sensory processing and social behaviors as well as mGluR5-dependent seizures. These results reveal mechanisms by which sex and a high-confidence ASD-risk gene interact to affect brain function and behavior.


Asunto(s)
Trastorno Autístico , Modelos Animales de Enfermedad , Receptor alfa de Estrógeno , Ratones Noqueados , Neocórtex , Fosfohidrolasa PTEN , Receptor del Glutamato Metabotropico 5 , Animales , Femenino , Masculino , Ratones , Trastorno Autístico/metabolismo , Trastorno Autístico/fisiopatología , Trastorno Autístico/genética , Trastorno Autístico/patología , Receptor alfa de Estrógeno/metabolismo , Ratones Endogámicos C57BL , Neocórtex/metabolismo , Neocórtex/patología , Fosfohidrolasa PTEN/metabolismo , Fosfohidrolasa PTEN/genética , Células Piramidales/metabolismo , Receptor del Glutamato Metabotropico 5/metabolismo , Conducta Social
12.
Genes (Basel) ; 15(4)2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38674342

RESUMEN

Hypophosphatasia is a rare inherited metabolic disorder caused by the deficiency of tissue-nonspecific alkaline phosphatase. More severe and early onset cases present symptoms of muscle weakness, diminished motor coordination, and epileptic seizures. These neurological manifestations are poorly characterized. Thus, it is urgent to discover novel differentially expressed genes for investigating the genetic mechanisms underlying the neurological manifestations of hypophosphatasia. RNA-sequencing data offer a high-resolution and highly accurate transcript profile. In this study, we apply an empirical Bayes model to RNA-sequencing data acquired from the spinal cord and neocortex tissues of a mouse model, individually, to more accurately estimate the genetic effects without bias. More importantly, we further develop two integration methods, weighted gene approach and weighted Z method, to incorporate two RNA-sequencing data into a model for enhancing the effects of genetic markers in the diagnostics of hypophosphatasia disease. The simulation and real data analysis have demonstrated the effectiveness of our proposed integration methods, which can maximize genetic signals identified from the spinal cord and neocortex tissues, minimize the prediction error, and largely improve the prediction accuracy in risk prediction.


Asunto(s)
Fosfatasa Alcalina , Teorema de Bayes , Hipofosfatasia , Hipofosfatasia/genética , Animales , Ratones , Fosfatasa Alcalina/genética , Análisis de Secuencia de ARN/métodos , Médula Espinal/metabolismo , Médula Espinal/patología , Humanos , Modelos Animales de Enfermedad , Neocórtex/metabolismo , Neocórtex/patología
13.
eNeuro ; 11(5)2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38637152

RESUMEN

Canonically, action potentials of most mammalian neurons initiate at the axon initial segment (AIS) and propagate bidirectionally: orthodromically along the distal axon and retrogradely into the soma and dendrites. Under some circumstances, action potentials may initiate ectopically, at sites distal to the AIS, and propagate antidromically along the axon. These "ectopic action potentials" (EAPs) have been observed in experimental models of seizures and chronic pain, and more rarely in nonpathological forebrain neurons. Here we report that a large majority of parvalbumin-expressing (PV+) interneurons in the upper layers of mouse neocortex, from both orbitofrontal and primary somatosensory areas, fire EAPs after sufficient activation of their somata. Somatostatin-expressing interneurons also fire EAPs, though less robustly. Ectopic firing in PV+ cells occurs in varying temporal patterns and can persist for several seconds. PV+ cells evoke strong synaptic inhibition in pyramidal neurons and interneurons and play critical roles in cortical function. Our results suggest that ectopic spiking of PV+ interneurons is common and may contribute to both normal and pathological network functions of the neocortex.


Asunto(s)
Potenciales de Acción , Interneuronas , Ratones Transgénicos , Neocórtex , Parvalbúminas , Animales , Parvalbúminas/metabolismo , Interneuronas/fisiología , Interneuronas/metabolismo , Neocórtex/fisiología , Potenciales de Acción/fisiología , Masculino , Ratones , Femenino , Ratones Endogámicos C57BL , Células Piramidales/fisiología , Somatostatina/metabolismo
14.
Chemosphere ; 358: 142124, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38677614

RESUMEN

Metformin, the most commonly prescribed drug for the treatment of diabetes, is increasingly used during pregnancy to address various disorders such as diabetes, obesity, preeclampsia, and metabolic diseases. However, its impact on neocortex development remains unclear. Here, we investigated the direct effects of metformin on neocortex development, focusing on ERK and p35/CDK5 regulation. Using a pregnant rat model, we found that metformin treatment during pregnancy induces small for gestational age (SGA) and reduces relative cortical thickness in embryos and neonates. Additionally, we discovered that metformin inhibits neural progenitor cell proliferation in the sub-ventricular zone (SVZ)/ventricular zone (VZ) of the developing neocortex, a process possibly mediated by ERK inactivation. Furthermore, metformin induces neuronal apoptosis in the SVZ/VZ area of the developing neocortex. Moreover, metformin retards neuronal migration, cortical lamination, and differentiation, potentially through p35/CDK5 inhibition in the developing neocortex. Remarkably, compensating for p35 through in utero electroporation partially rescues metformin-impaired neuronal migration and development. In summary, our study reveals that metformin disrupts neocortex development by inhibiting neuronal progenitor proliferation, neuronal migration, cortical layering, and cortical neuron maturation, likely via ERK and p35/CDK5 inhibition. Consequently, our findings advocate for caution in metformin usage during pregnancy, given its potential adverse effects on fetal brain development.


Asunto(s)
Proliferación Celular , Quinasa 5 Dependiente de la Ciclina , Metformina , Neocórtex , Metformina/farmacología , Animales , Femenino , Embarazo , Neocórtex/efectos de los fármacos , Quinasa 5 Dependiente de la Ciclina/metabolismo , Ratas , Proliferación Celular/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Neuronas/efectos de los fármacos , Ratas Sprague-Dawley , Diferenciación Celular/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Apoptosis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
15.
Cell Rep Methods ; 4(3): 100738, 2024 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-38508188

RESUMEN

Spatially resolved epigenomic profiling is critical for understanding biology in the mammalian brain. Single-cell spatial epigenomic assays were developed recently for this purpose, but they remain costly and labor intensive for examining brain tissues across substantial dimensions and surveying a collection of brain samples. Here, we demonstrate an approach, epigenomic tomography, that maps spatial epigenomes of mouse brain at the scale of centimeters. We individually profiled neuronal and glial fractions of mouse neocortex slices with 0.5 mm thickness. Tri-methylation of histone 3 at lysine 27 (H3K27me3) or acetylation of histone 3 at lysine 27 (H3K27ac) features across these slices were grouped into clusters based on their spatial variation patterns to form epigenomic brain maps. As a proof of principle, our approach reveals striking dynamics in the frontal cortex due to kainic-acid-induced seizure, linked with transmembrane ion transporters, exocytosis of synaptic vesicles, and secretion of neurotransmitters. Epigenomic tomography provides a powerful and cost-effective tool for characterizing brain disorders based on the spatial epigenome.


Asunto(s)
Cromatina , Neocórtex , Ratones , Animales , Histonas/genética , Epigenómica/métodos , Lisina , Neocórtex/metabolismo , Mamíferos/metabolismo
16.
EMBO J ; 43(8): 1388-1419, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38514807

RESUMEN

Neocortex expansion during evolution is linked to higher numbers of neurons, which are thought to result from increased proliferative capacity and neurogenic potential of basal progenitor cells during development. Here, we show that EREG, encoding the growth factor EPIREGULIN, is expressed in the human developing neocortex and in gorilla cerebral organoids, but not in the mouse neocortex. Addition of EPIREGULIN to the mouse neocortex increases proliferation of basal progenitor cells, whereas EREG ablation in human cortical organoids reduces proliferation in the subventricular zone. Treatment of cortical organoids with EPIREGULIN promotes a further increase in proliferation of gorilla but not of human basal progenitor cells. EPIREGULIN competes with the epidermal growth factor (EGF) to promote proliferation, and inhibition of the EGF receptor abrogates the EPIREGULIN-mediated increase in basal progenitor cells. Finally, we identify putative cis-regulatory elements that may contribute to the observed inter-species differences in EREG expression. Our findings suggest that species-specific regulation of EPIREGULIN expression may contribute to the increased neocortex size of primates by providing a tunable pro-proliferative signal to basal progenitor cells in the subventricular zone.


Asunto(s)
Epirregulina , Neocórtex , Animales , Humanos , Ratones , Proliferación Celular , Epirregulina/genética , Epirregulina/metabolismo , Gorilla gorilla/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Neocórtex/citología , Neocórtex/metabolismo , Primates/fisiología
17.
J Alzheimers Dis ; 98(4): 1391-1401, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38552111

RESUMEN

Background: Deposits of amyloid-ß (Aß) appear early in Alzheimer's disease (AD). Objective: The aim of the present study was to compare the presence of cortical and subcortical Aß in early AD using positron emission tomography (PET). Methods: Eight cognitively unimpaired (CU) subjects, 8 with mild cognitive impairment (MCI) and 8 with mild AD were examined with PET and [11C]AZD2184. A data driven cut-point for Aß positivity was defined by Gaussian mixture model of isocortex binding potential (BPND) values. Results: Sixteen subjects (3 CU, 5 MCI and 8 AD) were Aß-positive. BPND was lower in subcortical and allocortical regions compared to isocortex. Fifteen of the 16 Aß-positive subjects displayed Aß binding in striatum, 14 in thalamus and 10 in allocortical regions. Conclusions: Aß deposits appear to be widespread in early AD. It cannot be excluded that deposits appear simultaneously throughout the whole brain which has implications for improved diagnostics and disease monitoring.


Asunto(s)
Enfermedad de Alzheimer , Aminopiridinas , Benzotiazoles , Disfunción Cognitiva , Neocórtex , Humanos , Enfermedad de Alzheimer/metabolismo , Radioisótopos de Carbono , Péptidos beta-Amiloides/metabolismo , Disfunción Cognitiva/diagnóstico por imagen , Disfunción Cognitiva/metabolismo , Tomografía de Emisión de Positrones/métodos , Neocórtex/metabolismo
18.
Epilepsy Res ; 202: 107355, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38555654

RESUMEN

BACKGROUND: The hyperpolarization-activated cyclic nucleotide-gated cation channel (HCN1) is predominantly located in key regions associated with epilepsy, such as the neocortex and hippocampus. Under normal physiological conditions, HCN1 plays a crucial role in the excitatory and inhibitory regulation of neuronal networks. In temporal lobe epilepsy, the expression of HCN1 is decreased in the hippocampi of both animal models and patients. However, whether HCN1 expression changes during epileptogenesis preceding spontaneous seizures remains unclear. OBJECTIVE: The aim of this study was to determine whether the expression of HCN1 is altered during the epileptic prodromal phase, thereby providing evidence for its role in epileptogenesis. METHODS: We utilized a cobalt wire-induced rat epilepsy model to observe changes in HCN1 during epileptogenesis and epilepsy. Additionally, we also compared HCN1 alterations in epileptogenic tissues between cobalt wire- and pilocarpine-induced epilepsy rat models. Long-term video EEG recordings were used to confirm seizures development. Transcriptional changes, translation, and distribution of HCN1 were assessed using high-throughput transcriptome sequencing, total protein extraction, membrane and cytoplasmic protein fractionation, western blotting, immunohistochemistry, and immunofluorescence techniques. RESULTS: In the cobalt wire-induced rat epilepsy model during the epileptogenesis phase, total HCN1 mRNA and protein levels were downregulated. Specifically, the membrane expression of HCN1 was decreased, whereas cytoplasmic HCN1 expression showed no significant change. The distribution of HCN1 in the distal dendrites of neurons decreased. During the epilepsy period, similar HCN1 alterations were observed in the neocortex of rats with cobalt wire-induced epilepsy and hippocampus of rats with lithium pilocarpine-induced epilepsy, including downregulation of mRNA levels, decreased total protein expression, decreased membrane expression, and decreased distal dendrite expression. CONCLUSIONS: Alterations in HCN1 expression and distribution are involved in epileptogenesis beyond their association with seizure occurrence. Similarities in HCN1 alterations observed in epileptogenesis-related tissues from different models suggest a shared pathophysiological pathway in epileptogenesis involving HCN1 dysregulation. Therefore, the upregulation of HCN1 expression in neurons, maintenance of the HCN1 membrane, and distal dendrite distribution in neurons may represent promising disease-modifying strategies in epilepsy.


Asunto(s)
Modelos Animales de Enfermedad , Epilepsia , Hipocampo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Canales de Potasio , Ratas Sprague-Dawley , Animales , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Masculino , Epilepsia/metabolismo , Epilepsia/inducido químicamente , Epilepsia/genética , Epilepsia/fisiopatología , Ratas , Hipocampo/metabolismo , Canales de Potasio/metabolismo , Canales de Potasio/genética , Pilocarpina/toxicidad , Cobalto/farmacología , Electroencefalografía , Neuronas/metabolismo , Neocórtex/metabolismo
19.
Nat Cell Biol ; 26(5): 698-709, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38548890

RESUMEN

The human neocortex has undergone strong evolutionary expansion, largely due to an increased progenitor population, the basal radial glial cells. These cells are responsible for the production of a diversity of cell types, but the successive cell fate decisions taken by individual progenitors remain unknown. Here we developed a semi-automated live/fixed correlative imaging method to map basal radial glial cell division modes in early fetal tissue and cerebral organoids. Through the live analysis of hundreds of dividing progenitors, we show that basal radial glial cells undergo abundant symmetric amplifying divisions, and frequent self-consuming direct neurogenic divisions, bypassing intermediate progenitors. These direct neurogenic divisions are more abundant in the upper part of the subventricular zone. We furthermore demonstrate asymmetric Notch activation in the self-renewing daughter cells, independently of basal fibre inheritance. Our results reveal a remarkable conservation of fate decisions in cerebral organoids, supporting their value as models of early human neurogenesis.


Asunto(s)
Linaje de la Célula , Neocórtex , Células-Madre Neurales , Neurogénesis , Organoides , Humanos , Neocórtex/citología , Neocórtex/embriología , Neocórtex/metabolismo , Organoides/citología , Organoides/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Diferenciación Celular , Células Ependimogliales/citología , Células Ependimogliales/metabolismo , Receptores Notch/metabolismo , Receptores Notch/genética , División Celular , Proliferación Celular
20.
Sleep ; 47(5)2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38452190

RESUMEN

STUDY OBJECTIVES: Sleep supports systems memory consolidation through the precise temporal coordination of specific oscillatory events during slow-wave sleep, i.e. the neocortical slow oscillations (SOs), thalamic spindles, and hippocampal ripples. Beneficial effects of sleep on memory are also observed in infants, although the contributing regions, especially hippocampus and frontal cortex, are immature. Here, we examined in rats the development of these oscillatory events and their coupling during early life. METHODS: EEG and hippocampal local field potentials were recorded during sleep in male rats at postnatal days (PD)26 and 32, roughly corresponding to early (1-2 years) and late (9-10 years) human childhood, and in a group of adult rats (14-18 weeks, corresponding to ~22-29 years in humans). RESULTS: SO and spindle amplitudes generally increased from PD26 to PD32. In parallel, frontocortical EEG spindles increased in density and frequency, while changes in hippocampal ripples remained nonsignificant. The proportion of SOs co-occurring with spindles also increased from PD26 to PD32. Whereas parietal cortical spindles were phase-locked to the depolarizing SO-upstate already at PD26, over frontal cortex SO-spindle phase-locking emerged not until PD32. Co-occurrence of hippocampal ripples with spindles was higher during childhood than in adult rats, but significant phase-locking of ripples to the excitable spindle troughs was observed only in adult rats. CONCLUSIONS: Results indicate a protracted development of synchronized thalamocortical processing specifically in frontocortical networks (i.e. frontal SO-spindle coupling). However, synchronization within thalamocortical networks generally precedes synchronization of thalamocortical with hippocampal processing as reflected by the delayed occurrence of spindle-ripple phase-coupling.


Asunto(s)
Electroencefalografía , Hipocampo , Animales , Ratas , Masculino , Hipocampo/fisiología , Tálamo/fisiología , Neocórtex/fisiología , Sueño/fisiología , Sueño de Onda Lenta/fisiología , Ondas Encefálicas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA