Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.487
Filtrar
1.
Sci Rep ; 14(1): 14990, 2024 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-38951511

RESUMEN

The unfolded protein response (UPR) maintains proteostasis upon endoplasmic reticulum (ER) stress, and is initiated by a range of physiological and pathological processes. While there have been advances in developing fluorescent reporters for monitoring individual signaling pathways of the UPR, this approach may not capture a cell's overall UPR activity. Here we describe a novel sensor of UPR activity, sUPRa, which is designed to report the global UPR. sUPRa displays excellent response characteristics, outperforms reporters of individual UPR pathways in terms of sensitivity and kinetics, and responds to a range of different ER stress stimuli. Furthermore, sUPRa's dual promoter and fluorescent protein design ensures that both UPR-active and inactive cells are detected, and controls for reporter copy number. Using sUPRa, we reveal UPR activation in layer 2/3 pyramidal neurons of mouse cerebral cortex following a period of sleep deprivation. sUPRa affords new opportunities for quantifying physiological UPR activity with cellular resolution.


Asunto(s)
Estrés del Retículo Endoplásmico , Respuesta de Proteína Desplegada , Animales , Ratones , Genes Reporteros , Humanos , Células Piramidales/metabolismo , Transducción de Señal , Proteínas Luminiscentes/metabolismo , Proteínas Luminiscentes/genética
2.
Int J Mol Sci ; 25(13)2024 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-38999971

RESUMEN

Major burdens for patients suffering from stroke are cognitive co-morbidities and epileptogenesis. Neural network disinhibition and deficient inhibitive pulses for fast network activities may result from impaired presynaptic release of the inhibitory neurotransmitter GABA. To test this hypothesis, a cortical photothrombotic stroke was induced in Sprague Dawley rats, and inhibitory currents were recorded seven days later in the peri-infarct blood-brain barrier disrupted (BBBd) hippocampus via patch-clamp electrophysiology in CA1 pyramidal cells (PC). Miniature inhibitory postsynaptic current (mIPSC) frequency was reduced to about half, and mIPSCs decayed faster in the BBBd hippocampus. Furthermore, the paired-pulse ratio of evoked GABA release was increased at 100 Hz, and train stimulations with 100 Hz revealed that the readily releasable pool (RRP), usually assumed to correspond to the number of tightly docked presynaptic vesicles, is reduced by about half in the BBBd hippocampus. These pathophysiologic changes are likely to contribute significantly to disturbed fast oscillatory activity, like cognition-associated gamma oscillations or sharp wave ripples and epileptogenesis in the BBBd hippocampus.


Asunto(s)
Barrera Hematoencefálica , Hipocampo , Potenciales Postsinápticos Inhibidores , Ratas Sprague-Dawley , Ácido gamma-Aminobutírico , Animales , Barrera Hematoencefálica/metabolismo , Ratas , Ácido gamma-Aminobutírico/metabolismo , Hipocampo/metabolismo , Masculino , Células Piramidales/metabolismo , Vesículas Sinápticas/metabolismo , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/fisiopatología , Transmisión Sináptica
3.
Nat Commun ; 15(1): 5609, 2024 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-38965228

RESUMEN

Epilepsy affects 1% of the general population and 30% of patients are resistant to antiepileptic drugs. Although optogenetics is an efficient antiepileptic strategy, the difficulty of illuminating deep brain areas poses translational challenges. Thus, the search of alternative light sources is strongly needed. Here, we develop pH-sensitive inhibitory luminopsin (pHIL), a closed-loop chemo-optogenetic nanomachine composed of a luciferase-based light generator, a fluorescent sensor of intracellular pH (E2GFP), and an optogenetic actuator (halorhodopsin) for silencing neuronal activity. Stimulated by coelenterazine, pHIL experiences bioluminescence resonance energy transfer between luciferase and E2GFP which, under conditions of acidic pH, activates halorhodopsin. In primary neurons, pHIL senses the intracellular pH drop associated with hyperactivity and optogenetically aborts paroxysmal activity elicited by the administration of convulsants. The expression of pHIL in hippocampal pyramidal neurons is effective in decreasing duration and increasing latency of pilocarpine-induced tonic-clonic seizures upon in vivo coelenterazine administration, without affecting higher brain functions. The same treatment is effective in markedly decreasing seizure manifestations in a murine model of genetic epilepsy. The results indicate that pHIL represents a potentially promising closed-loop chemo-optogenetic strategy to treat drug-refractory epilepsy.


Asunto(s)
Epilepsia , Neuronas , Optogenética , Animales , Concentración de Iones de Hidrógeno , Ratones , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Epilepsia/fisiopatología , Epilepsia/metabolismo , Epilepsia/tratamiento farmacológico , Humanos , Convulsiones/tratamiento farmacológico , Convulsiones/fisiopatología , Convulsiones/metabolismo , Halorrodopsinas/metabolismo , Halorrodopsinas/genética , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos , Masculino , Luciferasas/metabolismo , Luciferasas/genética , Células Piramidales/metabolismo , Células Piramidales/efectos de los fármacos , Imidazoles/farmacología , Pilocarpina/farmacología , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Células HEK293 , Pirazinas
4.
Brain Res Bull ; 214: 111008, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38866373

RESUMEN

The infralimbic (IL) cortex dysfunction has been implicated in major depressive disorder (MDD), yet the precise cellular and molecular mechanisms remain poorly understood. In this study, we investigated the role of layer V pyramidal neurons in a mouse model of MDD induced by repeated lipopolysaccharide (LPS) administration. Our results demonstrate that three days of systemic LPS administration induced depressive-like behavior and upregulated mRNA levels of interleukin-1ß (IL-1ß), tumor necrosis factor-alpha (TNF-α), and transforming growth factor-ß (TGF-ß) in the IL cortex. Electrophysiological recordings revealed a significant decrease in the intrinsic excitability of layer V pyramidal neurons in the IL following systemic LPS exposure. Importantly, chemogenetic activation of IL pyramidal neurons ameliorated LPS-induced depressive-like behavior. Additionally, LPS administration significantly increased microglial activity in the IL, as evidenced by a greater number of Ionized calcium binding adaptor molecule-1 (IBA-1)-positive cells. Morphometric analysis further unveiled enlarged soma, decreased branch numbers, and shorter branch lengths of microglial cells in the IL cortex following LPS exposure. Moreover, the activation of pyramidal neurons by clozapine-N-oxide increased the microglia branch length but did not change branch number or cytosolic area. These results collectively suggest that targeted activation of pyramidal neurons in the IL cortex mitigates microglial response and ameliorates depressive-like behaviors induced by systemic LPS administration. Therefore, our findings offer potential therapeutic targets for the development of interventions aimed at alleviating depressive symptoms by modulating IL cortical circuitry and microglial activity.


Asunto(s)
Lipopolisacáridos , Microglía , Células Piramidales , Animales , Células Piramidales/efectos de los fármacos , Células Piramidales/metabolismo , Lipopolisacáridos/farmacología , Ratones , Masculino , Microglía/efectos de los fármacos , Microglía/metabolismo , Ratones Endogámicos C57BL , Depresión/inducido químicamente , Depresión/metabolismo , Depresión/tratamiento farmacológico , Clozapina/farmacología , Clozapina/análogos & derivados , Modelos Animales de Enfermedad , Trastorno Depresivo Mayor/metabolismo
5.
Science ; 384(6700): eadn0886, 2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38843332

RESUMEN

In addition to their intrinsic rewarding properties, opioids can also evoke aversive reactions that protect against misuse. Cellular mechanisms that govern the interplay between opioid reward and aversion are poorly understood. We used whole-brain activity mapping in mice to show that neurons in the dorsal peduncular nucleus (DPn) are highly responsive to the opioid oxycodone. Connectomic profiling revealed that DPn neurons innervate the parabrachial nucleus (PBn). Spatial and single-nuclei transcriptomics resolved a population of PBn-projecting pyramidal neurons in the DPn that express µ-opioid receptors (µORs). Disrupting µOR signaling in the DPn switched oxycodone from rewarding to aversive and exacerbated the severity of opioid withdrawal. These findings identify the DPn as a key substrate for the abuse liability of opioids.


Asunto(s)
Analgésicos Opioides , Reacción de Prevención , Trastornos Relacionados con Opioides , Oxicodona , Núcleos Parabraquiales , Corteza Prefrontal , Receptores Opioides mu , Recompensa , Animales , Masculino , Ratones , Analgésicos Opioides/farmacología , Conectoma , Ratones Endogámicos C57BL , Neuronas/metabolismo , Neuronas/fisiología , Trastornos Relacionados con Opioides/metabolismo , Oxicodona/farmacología , Núcleos Parabraquiales/metabolismo , Corteza Prefrontal/metabolismo , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/fisiología , Células Piramidales/metabolismo , Receptores Opioides mu/metabolismo , Receptores Opioides mu/genética , Síndrome de Abstinencia a Sustancias/metabolismo , Transcriptoma
6.
Mol Autism ; 15(1): 28, 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38877552

RESUMEN

BACKGROUND: Mutations in the X-linked gene cyclin-dependent kinase-like 5 (CDKL5) cause a severe neurological disorder characterised by early-onset epileptic seizures, autism and intellectual disability (ID). Impaired hippocampal function has been implicated in other models of monogenic forms of autism spectrum disorders and ID and is often linked to epilepsy and behavioural abnormalities. Many individuals with CDKL5 deficiency disorder (CDD) have null mutations and complete loss of CDKL5 protein, therefore in the current study we used a Cdkl5-/y rat model to elucidate the impact of CDKL5 loss on cellular excitability and synaptic function of CA1 pyramidal cells (PCs). We hypothesised abnormal pre and/or post synaptic function and plasticity would be observed in the hippocampus of Cdkl5-/y rats. METHODS: To allow cross-species comparisons of phenotypes associated with the loss of CDKL5, we generated a loss of function mutation in exon 8 of the rat Cdkl5 gene and assessed the impact of the loss of CDLK5 using a combination of extracellular and whole-cell electrophysiological recordings, biochemistry, and histology. RESULTS: Our results indicate that CA1 hippocampal long-term potentiation (LTP) is enhanced in slices prepared from juvenile, but not adult, Cdkl5-/y rats. Enhanced LTP does not result from changes in NMDA receptor function or subunit expression as these remain unaltered throughout development. Furthermore, Ca2+ permeable AMPA receptor mediated currents are unchanged in Cdkl5-/y rats. We observe reduced mEPSC frequency accompanied by increased spine density in basal dendrites of CA1 PCs, however we find no evidence supporting an increase in silent synapses when assessed using a minimal stimulation protocol in slices. Additionally, we found no change in paired-pulse ratio, consistent with normal release probability at Schaffer collateral to CA1 PC synapses. CONCLUSIONS: Our data indicate a role for CDKL5 in hippocampal synaptic function and raise the possibility that altered intracellular signalling rather than synaptic deficits contribute to the altered plasticity. LIMITATIONS: This study has focussed on the electrophysiological and anatomical properties of hippocampal CA1 PCs across early postnatal development. Studies involving other brain regions, older animals and behavioural phenotypes associated with the loss of CDKL5 are needed to understand the pathophysiology of CDD.


Asunto(s)
Modelos Animales de Enfermedad , Potenciación a Largo Plazo , Proteínas Serina-Treonina Quinasas , Receptores AMPA , Receptores de N-Metil-D-Aspartato , Espasmos Infantiles , Animales , Masculino , Ratas , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/patología , Región CA1 Hipocampal/fisiopatología , Síndromes Epilépticos/genética , Síndromes Epilépticos/metabolismo , Potenciales Postsinápticos Excitadores , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Enfermedades Genéticas Ligadas al Cromosoma X/metabolismo , Enfermedades Genéticas Ligadas al Cromosoma X/fisiopatología , Hipocampo/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Células Piramidales/metabolismo , Células Piramidales/patología , Receptores AMPA/metabolismo , Receptores AMPA/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Espasmos Infantiles/genética , Espasmos Infantiles/metabolismo , Sinapsis/metabolismo
7.
Proc Natl Acad Sci U S A ; 121(24): e2311570121, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38830095

RESUMEN

Even a transient period of hearing loss during the developmental critical period can induce long-lasting deficits in temporal and spectral perception. These perceptual deficits correlate with speech perception in humans. In gerbils, these hearing loss-induced perceptual deficits are correlated with a reduction of both ionotropic GABAA and metabotropic GABAB receptor-mediated synaptic inhibition in auditory cortex, but most research on critical period plasticity has focused on GABAA receptors. Therefore, we developed viral vectors to express proteins that would upregulate gerbil postsynaptic inhibitory receptor subunits (GABAA, Gabra1; GABAB, Gabbr1b) in pyramidal neurons, and an enzyme that mediates GABA synthesis (GAD65) presynaptically in parvalbumin-expressing interneurons. A transient period of developmental hearing loss during the auditory critical period significantly impaired perceptual performance on two auditory tasks: amplitude modulation depth detection and spectral modulation depth detection. We then tested the capacity of each vector to restore perceptual performance on these auditory tasks. While both GABA receptor vectors increased the amplitude of cortical inhibitory postsynaptic potentials, only viral expression of postsynaptic GABAB receptors improved perceptual thresholds to control levels. Similarly, presynaptic GAD65 expression improved perceptual performance on spectral modulation detection. These findings suggest that recovering performance on auditory perceptual tasks depends on GABAB receptor-dependent transmission at the auditory cortex parvalbumin to pyramidal synapse and point to potential therapeutic targets for developmental sensory disorders.


Asunto(s)
Corteza Auditiva , Gerbillinae , Pérdida Auditiva , Animales , Corteza Auditiva/metabolismo , Corteza Auditiva/fisiopatología , Pérdida Auditiva/genética , Pérdida Auditiva/fisiopatología , Receptores de GABA-B/metabolismo , Receptores de GABA-B/genética , Glutamato Descarboxilasa/metabolismo , Glutamato Descarboxilasa/genética , Receptores de GABA-A/metabolismo , Receptores de GABA-A/genética , Parvalbúminas/metabolismo , Parvalbúminas/genética , Percepción Auditiva/fisiología , Células Piramidales/metabolismo , Células Piramidales/fisiología , Vectores Genéticos/genética
8.
eNeuro ; 11(7)2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38871458

RESUMEN

Glutamatergic synapses exhibit significant molecular diversity, but circuit-specific mechanisms that underlie synaptic regulation are not well characterized. Prior reports show that Rho-guanine nucleotide exchange factor (RhoGEF) Tiam1 regulates perforant path→dentate gyrus granule neuron synapses. In the present study, we report Tiam1's homolog Tiam2 is implicated in glutamatergic neurotransmission in CA1 pyramidal neurons. We find that Tiam2 regulates evoked excitatory glutamatergic currents via a postsynaptic mechanism mediated by the catalytic Dbl-homology domain. Overall, we present evidence for RhoGEF Tiam2's role in glutamatergic synapse function at Schaffer collateral→CA1 pyramidal neuron synapses.


Asunto(s)
Región CA1 Hipocampal , Potenciales Postsinápticos Excitadores , Ácido Glutámico , Factores de Intercambio de Guanina Nucleótido , Células Piramidales , Transmisión Sináptica , Animales , Células Piramidales/fisiología , Células Piramidales/efectos de los fármacos , Células Piramidales/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Región CA1 Hipocampal/fisiología , Región CA1 Hipocampal/efectos de los fármacos , Región CA1 Hipocampal/metabolismo , Transmisión Sináptica/fisiología , Transmisión Sináptica/efectos de los fármacos , Ácido Glutámico/metabolismo , Masculino , Potenciales Postsinápticos Excitadores/fisiología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Ratones Endogámicos C57BL , Femenino , Sinapsis/fisiología , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Ratones
9.
eNeuro ; 11(7)2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38886063

RESUMEN

Persistent activity in excitatory pyramidal cells (PYRs) is a putative mechanism for maintaining memory traces during working memory. We have recently demonstrated persistent interruption of firing in fast-spiking parvalbumin-expressing interneurons (PV-INs), a phenomenon that could serve as a substrate for persistent activity in PYRs through disinhibition lasting hundreds of milliseconds. Here, we find that hippocampal CA1 PV-INs exhibit type 2 excitability, like striatal and neocortical PV-INs. Modeling and mathematical analysis showed that the slowly inactivating potassium current KV1 contributes to type 2 excitability, enables the multiple firing regimes observed experimentally in PV-INs, and provides a mechanism for robust persistent interruption of firing. Using a fast/slow separation of times scales approach with the KV1 inactivation variable as a bifurcation parameter shows that the initial inhibitory stimulus stops repetitive firing by moving the membrane potential trajectory onto a coexisting stable fixed point corresponding to a nonspiking quiescent state. As KV1 inactivation decays, the trajectory follows the branch of stable fixed points until it crosses a subcritical Hopf bifurcation (HB) and then spirals out into repetitive firing. In a model describing entorhinal cortical PV-INs without KV1, interruption of firing could be achieved by taking advantage of the bistability inherent in type 2 excitability based on a subcritical HB, but the interruption was not robust to noise. Persistent interruption of firing is therefore broadly applicable to PV-INs in different brain regions but is only made robust to noise in the presence of a slow variable, KV1 inactivation.


Asunto(s)
Interneuronas , Modelos Neurológicos , Parvalbúminas , Parvalbúminas/metabolismo , Interneuronas/fisiología , Interneuronas/metabolismo , Animales , Potenciales de Acción/fisiología , Región CA1 Hipocampal/fisiología , Región CA1 Hipocampal/metabolismo , Inhibición Neural/fisiología , Células Piramidales/fisiología , Células Piramidales/metabolismo , Canales de Potasio de la Superfamilia Shaker/metabolismo , Corteza Entorrinal/fisiología , Corteza Entorrinal/metabolismo , Masculino
10.
Neurobiol Dis ; 198: 106558, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38852754

RESUMEN

Periventricular nodular heterotopia (PNH), the most common brain malformation diagnosed in adulthood, is characterized by the presence of neuronal nodules along the ventricular walls. PNH is mainly associated with mutations in the FLNA gene - encoding an actin-binding protein - and patients often develop epilepsy. However, the molecular mechanisms underlying the neuronal failure still remain elusive. It has been hypothesized that dysfunctional cortical circuitry, rather than ectopic neurons, may explain the clinical manifestations. To address this issue, we depleted FLNA from cortical pyramidal neurons of a conditional Flnaflox/flox mice by timed in utero electroporation of Cre recombinase. We found that FLNA regulates dendritogenesis and spinogenesis thus promoting an appropriate excitatory/inhibitory inputs balance. We demonstrated that FLNA modulates RAC1 and cofilin activity through its interaction with the Rho-GTPase Activating Protein 24 (ARHGAP24). Collectively, we disclose an uncharacterized role of FLNA and provide strong support for neural circuit dysfunction being a consequence of FLNA mutations.


Asunto(s)
Corteza Cerebral , Filaminas , Proteína de Unión al GTP rac1 , Animales , Ratones , Factores Despolimerizantes de la Actina/metabolismo , Corteza Cerebral/metabolismo , Filaminas/metabolismo , Filaminas/genética , Proteínas Activadoras de GTPasa/metabolismo , Proteínas Activadoras de GTPasa/genética , Ratones Transgénicos , Neurogénesis/fisiología , Neuronas/metabolismo , Neuropéptidos/metabolismo , Neuropéptidos/genética , Heterotopia Nodular Periventricular/genética , Heterotopia Nodular Periventricular/metabolismo , Heterotopia Nodular Periventricular/patología , Células Piramidales/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rac1/genética
11.
Mol Brain ; 17(1): 33, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38840181

RESUMEN

Loss-of-function mutations in the progranulin (GRN) gene are an autosomal dominant cause of Frontotemporal Dementia (FTD). These mutations typically result in haploinsufficiency of the progranulin protein. Grn+/- mice provide a model for progranulin haploinsufficiency and develop FTD-like behavioral abnormalities by 9-10 months of age. In previous work, we demonstrated that Grn+/- mice develop a low dominance phenotype in the tube test that is associated with reduced dendritic arborization of layer II/III pyramidal neurons in the prelimbic region of the medial prefrontal cortex (mPFC), a region key for social dominance behavior in the tube test assay. In this study, we investigated whether progranulin haploinsufficiency induced changes in dendritic spine density and morphology. Individual layer II/III pyramidal neurons in the prelimbic mPFC of 9-10 month old wild-type or Grn+/- mice were targeted for iontophoretic microinjection of fluorescent dye, followed by high-resolution confocal microscopy and 3D reconstruction for morphometry analysis. Dendritic spine density in Grn+/- mice was comparable to wild-type littermates, but the apical dendrites in Grn+/- mice had a shift in the proportion of spine types, with fewer stubby spines and more thin spines. Additionally, apical dendrites of Grn+/- mice had longer spines and smaller thin spine head diameter in comparison to wild-type littermates. These changes in spine morphology may contribute to altered circuit-level activity and social dominance deficits in Grn+/- mice.


Asunto(s)
Espinas Dendríticas , Haploinsuficiencia , Corteza Prefrontal , Progranulinas , Animales , Espinas Dendríticas/metabolismo , Corteza Prefrontal/patología , Corteza Prefrontal/metabolismo , Progranulinas/deficiencia , Progranulinas/genética , Ratones , Células Piramidales/metabolismo , Células Piramidales/patología , Masculino , Ratones Endogámicos C57BL
12.
Proc Natl Acad Sci U S A ; 121(27): e2314702121, 2024 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-38916997

RESUMEN

Enlargement of the cerebrospinal fluid (CSF)-filled brain ventricles (cerebral ventriculomegaly), the cardinal feature of congenital hydrocephalus (CH), is increasingly recognized among patients with autism spectrum disorders (ASD). KATNAL2, a member of Katanin family microtubule-severing ATPases, is a known ASD risk gene, but its roles in human brain development remain unclear. Here, we show that nonsense truncation of Katnal2 (Katnal2Δ17) in mice results in classic ciliopathy phenotypes, including impaired spermatogenesis and cerebral ventriculomegaly. In both humans and mice, KATNAL2 is highly expressed in ciliated radial glia of the fetal ventricular-subventricular zone as well as in their postnatal ependymal and neuronal progeny. The ventriculomegaly observed in Katnal2Δ17 mice is associated with disrupted primary cilia and ependymal planar cell polarity that results in impaired cilia-generated CSF flow. Further, prefrontal pyramidal neurons in ventriculomegalic Katnal2Δ17 mice exhibit decreased excitatory drive and reduced high-frequency firing. Consistent with these findings in mice, we identified rare, damaging heterozygous germline variants in KATNAL2 in five unrelated patients with neurosurgically treated CH and comorbid ASD or other neurodevelopmental disorders. Mice engineered with the orthologous ASD-associated KATNAL2 F244L missense variant recapitulated the ventriculomegaly found in human patients. Together, these data suggest KATNAL2 pathogenic variants alter intraventricular CSF homeostasis and parenchymal neuronal connectivity by disrupting microtubule dynamics in fetal radial glia and their postnatal ependymal and neuronal descendants. The results identify a molecular mechanism underlying the development of ventriculomegaly in a genetic subset of patients with ASD and may explain persistence of neurodevelopmental phenotypes in some patients with CH despite neurosurgical CSF shunting.


Asunto(s)
Cilios , Hidrocefalia , Microtúbulos , Animales , Femenino , Humanos , Masculino , Ratones , ATPasas Asociadas con Actividades Celulares Diversas/genética , ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/patología , Trastorno del Espectro Autista/metabolismo , Cilios/metabolismo , Cilios/patología , Epéndimo/metabolismo , Epéndimo/patología , Hidrocefalia/genética , Hidrocefalia/patología , Hidrocefalia/metabolismo , Katanina/metabolismo , Katanina/genética , Microtúbulos/metabolismo , Neuronas/metabolismo , Células Piramidales/metabolismo , Células Piramidales/patología
13.
Commun Biol ; 7(1): 678, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38831002

RESUMEN

Deciphering the functional organization of large biological networks is a major challenge for current mathematical methods. A common approach is to decompose networks into largely independent functional modules, but inferring these modules and their organization from network activity is difficult, given the uncertainties and incompleteness of measurements. Typically, some parts of the overall functional organization, such as intermediate processing steps, are latent. We show that the hidden structure can be determined from the statistical moments of observable network components alone, as long as the functional relevance of the network components lies in their mean values and the mean of each latent variable maps onto a scaled expectation of a binary variable. Whether the function of biological networks permits a hierarchical modularization can be falsified by a correlation-based statistical test that we derive. We apply the test to gene regulatory networks, dendrites of pyramidal neurons, and networks of spiking neurons.


Asunto(s)
Redes Reguladoras de Genes , Humanos , Animales , Células Piramidales/fisiología , Células Piramidales/metabolismo
14.
Philos Trans R Soc Lond B Biol Sci ; 379(1906): 20230222, 2024 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-38853550

RESUMEN

N-methyl-d-aspartate receptors (NMDARs) play a pivotal role in synaptic plasticity. While the functional role of post-synaptic NMDARs is well established, pre-synaptic NMDAR (pre-NMDAR) function is largely unexplored. Different pre-NMDAR subunit populations are documented at synapses, suggesting that subunit composition influences neuronal transmission. Here, we used electrophysiological recordings at Schaffer collateral-CA1 synapses partnered with Ca2+ imaging and glutamate uncaging at boutons of CA3 pyramidal neurones to reveal two populations of pre-NMDARs that contain either the GluN2A or GluN2B subunit. Activation of the GluN2B population decreases action potential-evoked Ca2+ influx via modulation of small-conductance Ca2+-activated K+ channels, while activation of the GluN2A population does the opposite. Critically, the level of functional expression of the subunits is subject to homeostatic regulation, bidirectionally affecting short-term facilitation, thus providing a capacity for a fine adjustment of information transfer. This article is part of a discussion meeting issue 'Long-term potentiation: 50 years on'.


Asunto(s)
Potenciales de Acción , Calcio , Receptores de N-Metil-D-Aspartato , Canales de Potasio de Pequeña Conductancia Activados por el Calcio , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo , Potenciales de Acción/fisiología , Calcio/metabolismo , Ratas , Sinapsis/fisiología , Sinapsis/metabolismo , Plasticidad Neuronal/fisiología , Células Piramidales/fisiología , Células Piramidales/metabolismo
15.
Int J Mol Sci ; 25(11)2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38892448

RESUMEN

Canonical transient receptor potential channel 3 (TRPC3) is the most abundant TRPC channel in the brain and is highly expressed in all subfields of the hippocampus. Previous studies have suggested that TRPC3 channels may be involved in the hyperexcitability of hippocampal pyramidal neurons and seizures. Genetic ablation of TRPC3 channel expression reduced the intensity of pilocarpine-induced status epilepticus (SE). However, the underlying cellular mechanisms remain unexplored and the contribution of TRPC3 channels to SE-induced neurodegeneration is not determined. In this study, we investigated the contribution of TRPC3 channels to the electrophysiological properties of hippocampal pyramidal neurons and hippocampal synaptic plasticity, and the contribution of TRPC3 channels to seizure-induced neuronal cell death. We found that genetic ablation of TRPC3 expression did not alter basic electrophysiological properties of hippocampal pyramidal neurons and had a complex impact on epileptiform bursting in CA3. However, TRPC3 channels contribute significantly to long-term potentiation in CA1 and SE-induced neurodegeneration. Our results provided further support for therapeutic potential of TRPC3 inhibitors and raised new questions that need to be answered by future studies.


Asunto(s)
Muerte Celular , Hipocampo , Células Piramidales , Convulsiones , Canales Catiónicos TRPC , Animales , Canales Catiónicos TRPC/metabolismo , Canales Catiónicos TRPC/genética , Ratones , Células Piramidales/metabolismo , Células Piramidales/patología , Hipocampo/metabolismo , Hipocampo/patología , Convulsiones/metabolismo , Convulsiones/patología , Estado Epiléptico/metabolismo , Estado Epiléptico/patología , Estado Epiléptico/inducido químicamente , Masculino , Neuronas/metabolismo , Pilocarpina , Potenciación a Largo Plazo , Ratones Noqueados , Ratones Endogámicos C57BL , Plasticidad Neuronal
16.
Cell Rep ; 43(6): 114295, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38796850

RESUMEN

Anxiety plays a key role in guiding behavior in response to potential threats. Anxiety is mediated by the activation of pyramidal neurons in the ventral hippocampus (vH), whose activity is controlled by GABAergic inhibitory interneurons. However, how different vH interneurons might contribute to anxiety-related processes is unclear. Here, we investigate the role of vH parvalbumin (PV)-expressing interneurons while mice transition from safe to more anxiogenic compartments of the elevated plus maze (EPM). We find that vH PV interneurons increase their activity in anxiogenic EPM compartments concomitant with dynamic changes in inhibitory interactions between PV interneurons and pyramidal neurons. By optogenetically inhibiting PV interneurons, we induce an increase in the activity of vH pyramidal neurons and persistent anxiety. Collectively, our results suggest that vH inhibitory microcircuits may act as a trigger for enduring anxiety states.


Asunto(s)
Ansiedad , Hipocampo , Interneuronas , Parvalbúminas , Células Piramidales , Animales , Interneuronas/metabolismo , Parvalbúminas/metabolismo , Ansiedad/metabolismo , Hipocampo/metabolismo , Ratones , Células Piramidales/metabolismo , Masculino , Ratones Endogámicos C57BL , Optogenética
17.
Cell Rep ; 43(6): 114277, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38805397

RESUMEN

Affective empathy enables social mammals to learn and transfer emotion to conspecifics, but an understanding of the neural circuitry and genetics underlying affective empathy is still very limited. Here, using the naive observational fear between cagemates as a paradigm similar to human affective empathy and chemo/optogenetic neuroactivity manipulation in mouse brain, we investigate the roles of multiple brain regions in mouse affective empathy. Remarkably, two neural circuits originating from the ventral hippocampus, previously unknown to function in empathy, are revealed to regulate naive observational fear. One is from ventral hippocampal pyramidal neurons to lateral septum GABAergic neurons, and the other is from ventral hippocampus pyramidal neurons to nucleus accumbens dopamine-receptor-expressing neurons. Furthermore, we identify the naive observational-fear-encoding neurons in the ventral hippocampus. Our findings highlight the potentially diverse regulatory pathways of empathy in social animals, shedding light on the mechanisms underlying empathy circuity and its disorders.


Asunto(s)
Empatía , Hipocampo , Animales , Empatía/fisiología , Hipocampo/fisiología , Hipocampo/metabolismo , Ratones , Masculino , Miedo/fisiología , Ratones Endogámicos C57BL , Neuronas GABAérgicas/metabolismo , Neuronas GABAérgicas/fisiología , Células Piramidales/fisiología , Células Piramidales/metabolismo , Vías Nerviosas/fisiología , Núcleo Accumbens/fisiología
18.
Sci Rep ; 14(1): 11713, 2024 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-38778177

RESUMEN

The development of neurons is regulated by several spatiotemporally changing factors, which are crucial to give the ability of neurons to form functional networks. While external physical stimuli may impact the early developmental stages of neurons, the medium and long-term consequences of these influences have yet to be thoroughly examined. Using an animal model, this study focuses on the morphological and transcriptome changes of the hippocampus that may occur as a consequence of fetal ultrasound examination. We selectively labeled CA1 neurons of the hippocampus with in-utero electroporation to analyze their morphological features. Furthermore, certain samples also went through RNA sequencing after repetitive ultrasound exposure. US exposure significantly changed several morphological properties of the basal dendritic tree. A notable increase was also observed in the density of spines on the basal dendrites, accompanied by various alterations in individual spine morphology. Transcriptome analysis revealed several up or downregulated genes, which may explain the molecular background of these alterations. Our results suggest that US-derived changes in the dendritic trees of CA1 pyramidal cells might be connected to modification of the transcriptome of the hippocampus and may lead to an increased dendritic input.


Asunto(s)
Región CA1 Hipocampal , Dendritas , Transcriptoma , Animales , Región CA1 Hipocampal/metabolismo , Dendritas/metabolismo , Femenino , Embarazo , Células Piramidales/metabolismo , Ratones , Hipocampo/metabolismo , Perfilación de la Expresión Génica , Espinas Dendríticas/metabolismo , Ultrasonografía Prenatal
19.
Sci Rep ; 14(1): 10054, 2024 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-38698053

RESUMEN

ß-Thalassaemia is one of the most common genetic diseases worldwide. During the past few decades, life expectancy of patients has increased significantly owing to advance in medical treatments. Cognitive impairment, once has been neglected, has gradually become more documented. Cognitive impairment in ß-thalassaemia patients is associated with natural history of the disease and socioeconomic factors. Herein, to determined effect of ß-thalassaemia intrinsic factors, 22-month-old ß-thalassaemia mouse was used as a model to assess cognitive impairment and to investigate any aberrant brain pathology in ß-thalassaemia. Open field test showed that ß-thalassaemia mice had decreased motor function. However, no difference of neuronal degeneration in primary motor cortex, layer 2/3 area was found. Interestingly, impaired learning and memory function accessed by a Morris water maze test was observed and correlated with a reduced number of living pyramidal neurons in hippocampus at the CA3 region in ß-thalassaemia mice. Cognitive impairment in ß-thalassaemia mice was significantly correlated with several intrinsic ß-thalassaemic factors including iron overload, anaemia, damaged red blood cells (RBCs), phosphatidylserine (PS)-exposed RBC large extracellular vesicles (EVs) and PS-exposed medium EVs. This highlights the importance of blood transfusion and iron chelation in ß-thalassaemia patients. In addition, to improve patients' quality of life, assessment of cognitive functions should become part of routine follow-up.


Asunto(s)
Disfunción Cognitiva , Modelos Animales de Enfermedad , Hipocampo , Talasemia beta , Animales , Talasemia beta/patología , Talasemia beta/complicaciones , Talasemia beta/genética , Disfunción Cognitiva/etiología , Disfunción Cognitiva/patología , Ratones , Hipocampo/patología , Hipocampo/metabolismo , Masculino , Neuronas/metabolismo , Neuronas/patología , Sobrecarga de Hierro/patología , Sobrecarga de Hierro/metabolismo , Sobrecarga de Hierro/complicaciones , Vesículas Extracelulares/metabolismo , Eritrocitos/metabolismo , Eritrocitos/patología , Células Piramidales/metabolismo , Células Piramidales/patología , Aprendizaje por Laberinto
20.
Commun Biol ; 7(1): 642, 2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38802535

RESUMEN

Alterations in the experience-dependent and autonomous elaboration of neural circuits are assumed to underlie autism spectrum disorder (ASD), though it is unclear what synaptic traits are responsible. Here, utilizing a valproic acid-induced ASD marmoset model, which shares common molecular features with idiopathic ASD, we investigate changes in the structural dynamics of tuft dendrites of upper-layer pyramidal neurons and adjacent axons in the dorsomedial prefrontal cortex through two-photon microscopy. In model marmosets, dendritic spine turnover is upregulated, and spines are generated in clusters and survived more often than in control marmosets. Presynaptic boutons in local axons, but not in commissural long-range axons, demonstrate hyperdynamic turnover in model marmosets, suggesting alterations in projection-specific plasticity. Intriguingly, nasal oxytocin administration attenuates clustered spine emergence in model marmosets. Enhanced clustered spine generation, possibly unique to certain presynaptic partners, may be associated with ASD and be a potential therapeutic target.


Asunto(s)
Callithrix , Modelos Animales de Enfermedad , Plasticidad Neuronal , Oxitocina , Animales , Oxitocina/metabolismo , Masculino , Sinapsis/metabolismo , Espinas Dendríticas/metabolismo , Espinas Dendríticas/patología , Espinas Dendríticas/efectos de los fármacos , Trastorno del Espectro Autista/metabolismo , Trastorno Autístico/metabolismo , Trastorno Autístico/patología , Corteza Prefrontal/metabolismo , Corteza Prefrontal/patología , Corteza Prefrontal/efectos de los fármacos , Células Piramidales/metabolismo , Células Piramidales/patología , Ácido Valproico/farmacología , Terminales Presinápticos/metabolismo , Femenino , Axones/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...