Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 168(1-2): 86-100.e15, 2017 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-27916275

RESUMEN

Type 1 diabetes is characterized by the destruction of pancreatic ß cells, and generating new insulin-producing cells from other cell types is a major aim of regenerative medicine. One promising approach is transdifferentiation of developmentally related pancreatic cell types, including glucagon-producing α cells. In a genetic model, loss of the master regulatory transcription factor Arx is sufficient to induce the conversion of α cells to functional ß-like cells. Here, we identify artemisinins as small molecules that functionally repress Arx by causing its translocation to the cytoplasm. We show that the protein gephyrin is the mammalian target of these antimalarial drugs and that the mechanism of action of these molecules depends on the enhancement of GABAA receptor signaling. Our results in zebrafish, rodents, and primary human pancreatic islets identify gephyrin as a druggable target for the regeneration of pancreatic ß cell mass from α cells.


Asunto(s)
Artemisininas/farmacología , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Modelos Animales de Enfermedad , Receptores de GABA-A/metabolismo , Transducción de Señal , Animales , Arteméter , Artemisininas/administración & dosificación , Proteínas Portadoras/metabolismo , Transdiferenciación Celular/efectos de los fármacos , Células Cultivadas , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus Tipo 1/patología , Perfilación de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Humanos , Insulina/genética , Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Ratones , Estabilidad Proteica/efectos de los fármacos , Ratas , Análisis de la Célula Individual , Factores de Transcripción/metabolismo , Pez Cebra , Ácido gamma-Aminobutírico/metabolismo
2.
Annu Rev Neurosci ; 42: 1-26, 2019 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-30735460

RESUMEN

Peripheral endocrine output relies on either direct or feed-forward multi-order command from the hypothalamus. Efficient coding of endocrine responses is made possible by the many neuronal cell types that coexist in intercalated hypothalamic nuclei and communicate through extensive synaptic connectivity. Although general anatomical and neurochemical features of hypothalamic neurons were described during the past decades, they have yet to be reconciled with recently discovered molecular classifiers and neurogenetic function determination. By interrogating magnocellular as well as parvocellular dopamine, GABA, glutamate, and phenotypically mixed neurons, we integrate available information at the molecular, cellular, network, and endocrine output levels to propose a framework for the comprehensive classification of hypothalamic neurons. Simultaneously, we single out putative neuronal subclasses for which future research can fill in existing gaps of knowledge to rationalize cellular diversity through function-determinant molecular marks in the hypothalamus.


Asunto(s)
Hipotálamo/citología , Neuronas/clasificación , Animales , Conectoma , Humanos , Hormonas Hipotalámicas/análisis , Red Nerviosa/ultraestructura , Neuronas/citología , Neuronas/metabolismo , Neurotransmisores/análisis , Hormonas Peptídicas/análisis , Análisis de la Célula Individual
3.
Nature ; 582(7811): 246-252, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32499648

RESUMEN

A wealth of specialized neuroendocrine command systems intercalated within the hypothalamus control the most fundamental physiological needs in vertebrates1,2. Nevertheless, we lack a developmental blueprint that integrates the molecular determinants of neuronal and glial diversity along temporal and spatial scales of hypothalamus development3. Here we combine single-cell RNA sequencing of 51,199 mouse cells of ectodermal origin, gene regulatory network (GRN) screens in conjunction with genome-wide association study-based disease phenotyping, and genetic lineage reconstruction to show that nine glial and thirty-three neuronal subtypes are generated by mid-gestation under the control of distinct GRNs. Combinatorial molecular codes that arise from neurotransmitters, neuropeptides and transcription factors are minimally required to decode the taxonomical hierarchy of hypothalamic neurons. The differentiation of γ-aminobutyric acid (GABA) and dopamine neurons, but not glutamate neurons, relies on quasi-stable intermediate states, with a pool of GABA progenitors giving rise to dopamine cells4. We found an unexpected abundance of chemotropic proliferation and guidance cues that are commonly implicated in dorsal (cortical) patterning5 in the hypothalamus. In particular, loss of SLIT-ROBO signalling impaired both the production and positioning of periventricular dopamine neurons. Overall, we identify molecular principles that shape the developmental architecture of the hypothalamus and show how neuronal heterogeneity is transformed into a multimodal neural unit to provide virtually infinite adaptive potential throughout life.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Hipotálamo/citología , Hipotálamo/embriología , Morfogénesis , Animales , Diferenciación Celular , Linaje de la Célula , Dopamina/metabolismo , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/metabolismo , Ectodermo/citología , Ectodermo/metabolismo , Femenino , Neuronas GABAérgicas/citología , Neuronas GABAérgicas/metabolismo , Redes Reguladoras de Genes , Estudio de Asociación del Genoma Completo , Ácido Glutámico/metabolismo , Hipotálamo/metabolismo , Masculino , Ratones , Morfogénesis/genética , Proteínas del Tejido Nervioso/metabolismo , Neuroglía/citología , Neuroglía/metabolismo , Neuropéptidos/metabolismo , Neurotransmisores/metabolismo , Receptores Inmunológicos/metabolismo , Regulón/genética , Transducción de Señal , Factores de Transcripción/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Proteínas Roundabout
4.
Proc Natl Acad Sci U S A ; 120(31): e2222095120, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37487094

RESUMEN

The locus coeruleus (LC) is a small nucleus in the pons from which ascending and descending projections innervate major parts of the central nervous system. Its major transmitter is norepinephrine (NE). This system is evolutionarily conserved, including in humans, and its functions are associated with wakefulness and related to disorders, such as depression. Here, we performed single-cell ribonucleic acid-sequencing (RNA-seq) to subdivide neurons in the LC (24 clusters in total) into 3 NE, 17 glutamate, and 5 γ-aminobutyric acid (GABA) subtypes, and to chart their neuropeptide, cotransmitter, and receptor profiles. We found that NE neurons expressed at least 19 neuropeptide transcripts, notably galanin (Gal) but not Npy, and >30 neuropeptide receptors. Among the galanin receptors, Galr1 was expressed in ~19% of NE neurons, as was also confirmed by in situ hybridization. Unexpectedly, Galr1 was highly expressed in GABA neurons surrounding the NE ensemble. Patch-clamp electrophysiology and cell-type-specific Ca2+-imaging using GCaMP6s revealed that a GalR1 agonist inhibits up to ~35% of NE neurons. This effect is direct and does not rely on feed-forward GABA inhibition. Our results define a role for the galanin system in NE functions, and a conceptual framework for the action of many other peptides and their receptors.


Asunto(s)
Galanina , Hormonas Peptídicas , Humanos , Animales , Ratones , Locus Coeruleus , Neuronas , Ácido Glutámico , Norepinefrina
5.
Proc Natl Acad Sci U S A ; 119(7)2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35135875

RESUMEN

The L-type voltage-gated Ca2+ channel gene CACNA1C is a risk gene for various psychiatric conditions, including schizophrenia and bipolar disorder. However, the cellular mechanism by which CACNA1C contributes to psychiatric disorders has not been elucidated. Here, we report that the embryonic deletion of Cacna1c in neurons destined for the cerebral cortex using an Emx1-Cre strategy disturbs spontaneous Ca2+ activity and causes abnormal brain development and anxiety. By combining computational modeling with electrophysiological membrane potential manipulation, we found that neural network activity was driven by intrinsic spontaneous Ca2+ activity in distinct progenitor cells expressing marginally increased levels of voltage-gated Ca2+ channels. MRI examination of the Cacna1c knockout mouse brains revealed volumetric differences in the neocortex, hippocampus, and periaqueductal gray. These results suggest that Cacna1c acts as a molecular switch and that its disruption during embryogenesis can perturb Ca2+ handling and neural development, which may increase susceptibility to psychiatric disease.


Asunto(s)
Trastornos de Ansiedad/metabolismo , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Canales de Calcio Tipo L/metabolismo , Calcio/metabolismo , Animales , Relojes Biológicos , Canales de Calcio Tipo L/genética , Regulación del Desarrollo de la Expresión Génica , Predisposición Genética a la Enfermedad , Ratones , Ratones Noqueados , Células-Madre Neurales
6.
EMBO J ; 39(1): e100882, 2020 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-31750562

RESUMEN

Maternal drug abuse during pregnancy is a rapidly escalating societal problem. Psychostimulants, including amphetamine, cocaine, and methamphetamine, are amongst the illicit drugs most commonly consumed by pregnant women. Neuropharmacology concepts posit that psychostimulants affect monoamine signaling in the nervous system by their affinities to neurotransmitter reuptake and vesicular transporters to heighten neurotransmitter availability extracellularly. Exacerbated dopamine signaling is particularly considered as a key determinant of psychostimulant action. Much less is known about possible adverse effects of these drugs on peripheral organs, and if in utero exposure induces lifelong pathologies. Here, we addressed this question by combining human RNA-seq data with cellular and mouse models of neuroendocrine development. We show that episodic maternal exposure to psychostimulants during pregnancy coincident with the intrauterine specification of pancreatic ß cells permanently impairs their ability of insulin production, leading to glucose intolerance in adult female but not male offspring. We link psychostimulant action specifically to serotonin signaling and implicate the sex-specific epigenetic reprogramming of serotonin-related gene regulatory networks upstream from the transcription factor Pet1/Fev as determinants of reduced insulin production.


Asunto(s)
Diabetes Mellitus Tipo 2/etiología , Intolerancia a la Glucosa/etiología , Glucosa/metabolismo , Homeostasis/efectos de los fármacos , Islotes Pancreáticos/patología , Metanfetamina/toxicidad , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Animales , Estimulantes del Sistema Nervioso Central/toxicidad , Metilación de ADN , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Intolerancia a la Glucosa/genética , Intolerancia a la Glucosa/metabolismo , Intolerancia a la Glucosa/patología , Humanos , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Masculino , Exposición Materna/efectos adversos , Ratones , Embarazo , Efectos Tardíos de la Exposición Prenatal/genética , Efectos Tardíos de la Exposición Prenatal/metabolismo , Efectos Tardíos de la Exposición Prenatal/patología
7.
Proc Natl Acad Sci U S A ; 118(7)2021 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-33558223

RESUMEN

The perception of and response to danger is critical for an individual's survival and is encoded by subcortical neurocircuits. The amygdaloid complex is the primary neuronal site that initiates bodily reactions upon external threat with local-circuit interneurons scaling output to effector pathways. Here, we categorize central amygdala neurons that express secretagogin (Scgn), a Ca2+-sensor protein, as a subset of protein kinase Cδ (PKCδ)+ interneurons, likely "off cells." Chemogenetic inactivation of Scgn+/PKCδ+ cells augmented conditioned response to perceived danger in vivo. While Ca2+-sensor proteins are typically implicated in shaping neurotransmitter release presynaptically, Scgn instead localized to postsynaptic compartments. Characterizing its role in the postsynapse, we found that Scgn regulates the cell-surface availability of NMDA receptor 2B subunits (GluN2B) with its genetic deletion leading to reduced cell membrane delivery of GluN2B, at least in vitro. Conclusively, we describe a select cell population, which gates danger avoidance behavior with secretagogin being both a selective marker and regulatory protein in their excitatory postsynaptic machinery.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Interneuronas/metabolismo , Proteína Quinasa C-delta/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Secretagoginas/metabolismo , Amígdala del Cerebelo/citología , Amígdala del Cerebelo/fisiología , Animales , Reacción de Prevención , Línea Celular Tumoral , Células Cultivadas , Miedo , Femenino , Humanos , Interneuronas/fisiología , Masculino , Transporte de Proteínas , Ratas , Ratas Wistar , Secretagoginas/genética , Potenciales Sinápticos
8.
Mol Psychiatry ; 2022 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-35581295

RESUMEN

Immune activation is one of the most common complications during pregnancy, predominantly evoked by viral infections. Nevertheless, how immune activation affects mother-offspring relationships postpartum remains unknown. Here, by using the polyinosinic-polycytidylic acid (Poly I:C) model of gestational infection we show that viral-like immune activation at mid-gestation persistently changes hypothalamic neurocircuit parameters in mouse dams and, consequently, is adverse to parenting behavior. Poly I:C-exposed dams favor non-pup-directed exploratory behavior at the expense of pup retrieval. These behavioral deficits are underlain by dendrite pruning and lesser immediate early gene activation in Galanin (Gal)+ neurons with dam-specific transcriptional signatures that reside in the medial preoptic area (mPOA). Reduced activation of an exclusively inhibitory contingent of these distal-projecting Gal+ neurons allows for increased feed-forward inhibition onto putative dopaminergic neurons in the ventral tegmental area (VTA) in Poly I:C-exposed dams. Notably, destabilized VTA output specifically accompanies post-pup retrieval epochs. We suggest that gestational immunogenic insults bias both threat processing and reward perception, manifesting as disfavored infant caregiving.

9.
Int J Mol Sci ; 24(3)2023 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-36768453

RESUMEN

4-methylumbelliferone (4MU) is a well-known hyaluronic acid synthesis inhibitor and an approved drug for the treatment of cholestasis. In animal models, 4MU decreases inflammation, reduces fibrosis, and lowers body weight, serum cholesterol, and insulin resistance. It also inhibits tumor progression and metastasis. The broad spectrum of effects suggests multiple and yet unknown targets of 4MU. Aiming at 4MU target deconvolution, we have analyzed publicly available data bases, including: 1. Small molecule library Bio Assay screening (PubChemBioAssay); 2. GO pathway databases screening; 3. Protein Atlas Database. We also performed comparative liver transcriptome analysis of mice on normal diet and mice fed with 4MU for two weeks. Potential targets of 4MU public data base analysis fall into two big groups, enzymes and transcription factors (TFs), including 13 members of the nuclear receptor superfamily regulating lipid and carbohydrate metabolism. Transcriptome analysis revealed changes in the expression of genes involved in bile acid metabolism, gluconeogenesis, and immune response. It was found that 4MU feeding decreased the accumulation of the glycogen granules in the liver. Thus, 4MU has multiple targets and can regulate cell metabolism by modulating signaling via nuclear receptors.


Asunto(s)
Himecromona , Transcriptoma , Ratones , Animales , Himecromona/farmacología , Hígado/metabolismo , Inflamación/metabolismo , Transducción de Señal , Metabolismo de los Lípidos
10.
EMBO J ; 37(21)2018 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-30209240

RESUMEN

Stress-induced cortical alertness is maintained by a heightened excitability of noradrenergic neurons innervating, notably, the prefrontal cortex. However, neither the signaling axis linking hypothalamic activation to delayed and lasting noradrenergic excitability nor the molecular cascade gating noradrenaline synthesis is defined. Here, we show that hypothalamic corticotropin-releasing hormone-releasing neurons innervate ependymal cells of the 3rd ventricle to induce ciliary neurotrophic factor (CNTF) release for transport through the brain's aqueductal system. CNTF binding to its cognate receptors on norepinephrinergic neurons in the locus coeruleus then initiates sequential phosphorylation of extracellular signal-regulated kinase 1 and tyrosine hydroxylase with the Ca2+-sensor secretagogin ensuring activity dependence in both rodent and human brains. Both CNTF and secretagogin ablation occlude stress-induced cortical norepinephrine synthesis, ensuing neuronal excitation and behavioral stereotypes. Cumulatively, we identify a multimodal pathway that is rate-limited by CNTF volume transmission and poised to directly convert hypothalamic activation into long-lasting cortical excitability following acute stress.


Asunto(s)
Neuronas Adrenérgicas/metabolismo , Factor Neurotrófico Ciliar/metabolismo , Hipotálamo/metabolismo , Locus Coeruleus/metabolismo , Estrés Fisiológico , Neuronas Adrenérgicas/patología , Animales , Factor Neurotrófico Ciliar/genética , Hipotálamo/patología , Locus Coeruleus/patología , Ratones , Ratones Noqueados , Ratas
11.
Proc Natl Acad Sci U S A ; 116(51): 25958-25967, 2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31796600

RESUMEN

Psychostimulant use is an ever-increasing socioeconomic burden, including a dramatic rise during pregnancy. Nevertheless, brain-wide effects of psychostimulant exposure are incompletely understood. Here, we performed Fos-CreERT2-based activity mapping, correlated for pregnant mouse dams and their fetuses with amphetamine, nicotine, and caffeine applied acutely during midgestation. While light-sheet microscopy-assisted intact tissue imaging revealed drug- and age-specific neuronal activation, the indusium griseum (IG) appeared indiscriminately affected. By using GAD67gfp/+ mice we subdivided the IG into a dorsolateral domain populated by γ-aminobutyric acidergic interneurons and a ventromedial segment containing glutamatergic neurons, many showing drug-induced activation and sequentially expressing Pou3f3/Brn1 and secretagogin (Scgn) during differentiation. We then combined Patch-seq and circuit mapping to show that the ventromedial IG is a quasi-continuum of glutamatergic neurons (IG-Vglut1+) reminiscent of dentate granule cells in both rodents and humans, whose dendrites emanate perpendicularly toward while their axons course parallel with the superior longitudinal fissure. IG-Vglut1+ neurons receive VGLUT1+ and VGLUT2+ excitatory afferents that topologically segregate along their somatodendritic axis. In turn, their efferents terminate in the olfactory bulb, thus being integral to a multisynaptic circuit that could feed information antiparallel to the olfactory-cortical pathway. In IG-Vglut1+ neurons, prenatal psychostimulant exposure delayed the onset of Scgn expression. Genetic ablation of Scgn was then found to sensitize adult mice toward methamphetamine-induced epilepsy. Overall, our study identifies brain-wide targets of the most common psychostimulants, among which Scgn+/Vglut1+ neurons of the IG link limbic and olfactory circuits.


Asunto(s)
Mapeo Encefálico , Encéfalo/metabolismo , Regulación de la Expresión Génica , Lóbulo Límbico/metabolismo , Animales , Axones/metabolismo , Encéfalo/diagnóstico por imagen , Dendritas/metabolismo , Femenino , Glutamato Descarboxilasa/genética , Humanos , Interneuronas/metabolismo , Lóbulo Límbico/anatomía & histología , Lóbulo Límbico/efectos de los fármacos , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Bulbo Olfatorio/metabolismo , Factores del Dominio POU/genética , Factores del Dominio POU/metabolismo , Secretagoginas/genética , Secretagoginas/metabolismo , Proteína 1 de Transporte Vesicular de Glutamato/genética , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo , Proteína 2 de Transporte Vesicular de Glutamato/genética , Proteína 2 de Transporte Vesicular de Glutamato/metabolismo , Ácido gamma-Aminobutírico/metabolismo
12.
EMBO J ; 36(14): 2107-2125, 2017 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-28637794

RESUMEN

Ca2+-sensor proteins are generally implicated in insulin release through SNARE interactions. Here, secretagogin, whose expression in human pancreatic islets correlates with their insulin content and the incidence of type 2 diabetes, is shown to orchestrate an unexpectedly distinct mechanism. Single-cell RNA-seq reveals retained expression of the TRP family members in ß-cells from diabetic donors. Amongst these, pharmacological probing identifies Ca2+-permeable transient receptor potential vanilloid type 1 channels (TRPV1) as potent inducers of secretagogin expression through recruitment of Sp1 transcription factors. Accordingly, agonist stimulation of TRPV1s fails to rescue insulin release from pancreatic islets of glucose intolerant secretagogin knock-out(-/-) mice. However, instead of merely impinging on the SNARE machinery, reduced insulin availability in secretagogin-/- mice is due to ß-cell loss, which is underpinned by the collapse of protein folding and deregulation of secretagogin-dependent USP9X deubiquitinase activity. Therefore, and considering the desensitization of TRPV1s in diabetic pancreata, a TRPV1-to-secretagogin regulatory axis seems critical to maintain the structural integrity and signal competence of ß-cells.


Asunto(s)
Regulación de la Expresión Génica , Células Secretoras de Insulina/fisiología , Proteínas/metabolismo , Secretagoginas/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Supervivencia Celular , Perfilación de la Expresión Génica , Humanos , Ratones , Ratones Noqueados , Secretagoginas/deficiencia , Análisis de la Célula Individual
13.
EMBO J ; 34(1): 36-54, 2015 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-25430741

RESUMEN

A hierarchical hormonal cascade along the hypothalamic-pituitary-adrenal axis orchestrates bodily responses to stress. Although corticotropin-releasing hormone (CRH), produced by parvocellular neurons of the hypothalamic paraventricular nucleus (PVN) and released into the portal circulation at the median eminence, is known to prime downstream hormone release, the molecular mechanism regulating phasic CRH release remains poorly understood. Here, we find a cohort of parvocellular cells interspersed with magnocellular PVN neurons expressing secretagogin. Single-cell transcriptome analysis combined with protein interactome profiling identifies secretagogin neurons as a distinct CRH-releasing neuron population reliant on secretagogin's Ca(2+) sensor properties and protein interactions with the vesicular traffic and exocytosis release machineries to liberate this key hypothalamic releasing hormone. Pharmacological tools combined with RNA interference demonstrate that secretagogin's loss of function occludes adrenocorticotropic hormone release from the pituitary and lowers peripheral corticosterone levels in response to acute stress. Cumulatively, these data define a novel secretagogin neuronal locus and molecular axis underpinning stress responsiveness.


Asunto(s)
Corticosterona/metabolismo , Hormona Liberadora de Corticotropina/metabolismo , Neuronas/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Secretagoginas/metabolismo , Estrés Fisiológico/fisiología , Animales , Corticosterona/genética , Hormona Liberadora de Corticotropina/genética , Masculino , Ratones , Neuronas/citología , Núcleo Hipotalámico Paraventricular/citología , Hipófisis/citología , Hipófisis/metabolismo , Interferencia de ARN , Secretagoginas/genética , Transcriptoma/fisiología
14.
J Neurosci ; 37(34): 8166-8179, 2017 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-28733354

RESUMEN

Temporal lobe epilepsy (TLE) is the most frequent form of focal epilepsies and is generally associated with malfunctioning of the hippocampal formation. Recently, a preferential loss of parvalbumin (PV) neurons has been observed in the subiculum of TLE patients and in animal models of TLE. To demonstrate a possible causative role of defunct PV neurons in the generation of TLE, we permanently inhibited GABA release selectively from PV neurons of the ventral subiculum by injecting a viral vector expressing tetanus toxin light chain in male mice. Subsequently, mice were subjected to telemetric EEG recording and video monitoring. Eighty-eight percent of the mice presented clusters of spike-wave discharges (C-SWDs; 40.0 ± 9.07/month), and 64% showed spontaneous recurrent seizures (SRSs; 5.3 ± 0.83/month). Mice injected with a control vector presented with neither C-SWDs nor SRSs. No neurodegeneration was observed due to vector injection or SRS. Interestingly, mice that presented with only C-SWDs but no SRSs, developed SRSs upon injection of a subconvulsive dose of pentylenetetrazole after 6 weeks. The initial frequency of SRSs declined by ∼30% after 5 weeks. In contrast to permanent silencing of PV neurons, transient inhibition of GABA release from PV neurons through the designer receptor hM4Di selectively expressed in PV-containing neurons transiently reduced the seizure threshold of the mice but induced neither acute nor recurrent seizures. Our data demonstrate a critical role for perisomatic inhibition mediated by PV-containing interneurons, suggesting that their sustained silencing could be causally involved in the development of TLE.SIGNIFICANCE STATEMENT Development of temporal lobe epilepsy (TLE) generally takes years after an initial insult during which maladaptation of hippocampal circuitries takes place. In human TLE and in animal models of TLE, parvalbumin neurons are selectively lost in the subiculum, the major output area of the hippocampus. The present experiments demonstrate that specific and sustained inhibition of GABA release from parvalbumin-expressing interneurons (mostly basket cells) in sector CA1/subiculum is sufficient to induce hyperexcitability and spontaneous recurrent seizures in mice. As in patients with nonlesional TLE, these mice developed epilepsy without signs of neurodegeneration. The experiments highlight the importance of the potent inhibitory action mediated by parvalbumin cells in the hippocampus and identify a potential mechanism in the development of TLE.


Asunto(s)
Hipocampo/fisiopatología , Interneuronas/fisiología , Parvalbúminas/antagonistas & inhibidores , Parvalbúminas/fisiología , Convulsiones/fisiopatología , Animales , Electroencefalografía/métodos , Hipocampo/efectos de los fármacos , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Potenciales Postsinápticos Inhibidores/fisiología , Interneuronas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Técnicas de Cultivo de Órganos , Pentilenotetrazol/toxicidad , Convulsiones/inducido químicamente
15.
Pflugers Arch ; 468(2): 305-19, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26530828

RESUMEN

Specialized Ca(2+)-dependent ion channels ubiquitously couple intracellular Ca(2+) signals to a change in cell polarization. The existing physiological evidence suggests that Ca(2+)-activated Cl(-) channels (CaCCs) are functional in taste cells. Because Ano1 and Ano2 encode channel proteins that form CaCCs in a variety of cells, we analyzed their expression in mouse taste cells. Transcripts for Ano1 and Ano2 were detected in circumvallate (CV) papillae, and their expression in taste cells was confirmed using immunohistochemistry. When dialyzed with CsCl, taste cells of the type III exhibited no ion currents dependent on cytosolic Ca(2+). Large Ca(2+)-gated currents mediated by TRPM5 were elicited in type II cells by Ca(2+) uncaging. When TRPM5 was inhibited by triphenylphosphine oxide (TPPO), ionomycin stimulated a small but resolvable inward current that was eliminated by anion channel blockers, including T16Ainh-A01 (T16), a specific Ano1 antagonist. This suggests that CaCCs, including Ano1-like channels, are functional in type II cells. In type I cells, CaCCs were prominently active, blockable with the CaCC antagonist CaCCinh-A01 but insensitive to T16. By profiling Ano1 and Ano2 expressions in individual taste cells, we revealed Ano1 transcripts in type II cells only, while Ano2 transcripts were detected in both type I and type II cells. P2Y agonists stimulated Ca(2+)-gated Cl(-) currents in type I cells. Thus, CaCCs, possibly formed by Ano2, serve as effectors downstream of P2Y receptors in type I cells. While the role for TRPM5 in taste transduction is well established, the physiological significance of expression of CaCCs in type II cells remains to be elucidated.


Asunto(s)
Canales de Cloruro/metabolismo , Papilas Gustativas/metabolismo , Potenciales de Acción , Animales , Anoctamina-1 , Anoctaminas , Células CHO , Calcio/metabolismo , Células Cultivadas , Canales de Cloruro/antagonistas & inhibidores , Canales de Cloruro/genética , Cricetinae , Cricetulus , Células HEK293 , Humanos , Ratones , Antagonistas del Receptor Purinérgico P2Y/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Purinérgicos P2Y/metabolismo , Canales Catiónicos TRPM/antagonistas & inhibidores , Canales Catiónicos TRPM/metabolismo , Papilas Gustativas/efectos de los fármacos , Papilas Gustativas/fisiología
16.
J Cell Sci ; 125(Pt 22): 5514-23, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22956545

RESUMEN

Afferent output in type II taste cells is mediated by ATP liberated through ion channels. It is widely accepted that pannexin 1 (Panx1) channels are responsible for ATP release in diverse cell types, including taste cells. While biophysical evidence implicates slow deactivation of ion channels following ATP release in taste cells, recombinant Panx1 activates and deactivates rapidly. This inconsistency could indicate that the cellular context specifies Panx1 functioning. We cloned Panx1 from murine taste tissue, and heterologously expressed it in three different cell lines: HEK-293, CHO and neuroblastoma SK-N-SH cells. In all three cell lines, Panx1 transfection yielded outwardly rectifying anion channels that exhibited fast gating and negligible permeability to anions exceeding 250 Da. Despite expression of Panx1, the host cells did not liberate ATP upon stimulation, making it unclear whether Panx1 is involved in taste-related ATP secretion. This issue was addressed using mice with genetic ablation of the Panx1 gene. The ATP-biosensor assay revealed that, in taste cells devoid of Panx1, ATP secretion was robust and apparently unchanged compared with the control. Our data suggest that Panx1 alone forms a channel that has insufficient permeability to ATP. Perhaps, a distinct subunit and/or a regulatory circuit that is absent in taste cells is required to enable a high ATP-permeability mode of a native Panx1-based channel.


Asunto(s)
Adenosina Trifosfato/farmacología , Conexinas/metabolismo , Mamíferos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Papilas Gustativas/metabolismo , Animales , Aniones/metabolismo , Células CHO , Carbenoxolona/farmacología , Conexinas/deficiencia , Cricetinae , Cricetulus , Células HEK293 , Humanos , Activación del Canal Iónico/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/deficiencia , Permeabilidad/efectos de los fármacos , Papilas Gustativas/efectos de los fármacos
17.
Res Sq ; 2024 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-38405925

RESUMEN

Astrocytes safeguard the homeostasis of the central nervous system1,2. Despite their prominent morphological plasticity under conditions that challenge the brain's adaptive capacity3-5, the classification of astrocytes, and relating their molecular make-up to spatially devolved neuronal operations that specify behavior or metabolism, remained mostly futile6,7. Although it seems unexpected in the era of single-cell biology, the lack of a major advance in stratifying astrocytes under physiological conditions rests on the incompatibility of 'neurocentric' algorithms that rely on stable developmental endpoints, lifelong transcriptional, neurotransmitter, and neuropeptide signatures for classification6-8 with the dynamic functional states, anatomic allocation, and allostatic plasticity of astrocytes1. Simplistically, therefore, astrocytes are still grouped as 'resting' vs. 'reactive', the latter referring to pathological states marked by various inducible genes3,9,10. Here, we introduced a machine learning-based feature recognition algorithm that benefits from the cumulative power of published single-cell RNA-seq data on astrocytes as a reference map to stepwise eliminate pleiotropic and inducible cellular features. For the healthy hypothalamus, this walk-back approach revealed gene regulatory networks (GRNs) that specified subsets of astrocytes, and could be used as landmarking tools for their anatomical assignment. The core molecular censuses retained by astrocyte subsets were sufficient to stratify them by allostatic competence, chiefly their signaling and metabolic interplay with neurons. Particularly, we found differentially expressed mitochondrial genes in insulin-sensing astrocytes and demonstrated their reciprocal signaling with neurons that work antagonistically within the food intake circuitry. As a proof-of-concept, we showed that disrupting Mfn2 expression in astrocytes reduced their ability to support dynamic circuit reorganization, a time-locked feature of satiety in the hypothalamus, thus leading to obesity in mice. Overall, our results suggest that astrocytes in the healthy brain are fundamentally more heterogeneous than previously thought and topologically mirror the specificity of local neurocircuits.

18.
ACS Appl Bio Mater ; 7(4): 2325-2337, 2024 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-38483087

RESUMEN

This article addresses the entire life cycle of the all-green fibrous materials based on poly(3-hydroxybutyrate) (PHB) containing a natural biocompatible additive Hemin (Hmi): from preparation, service life, and the end of life upon in-soil biodegradation. Fibrous PHB/Hmi materials with a highly developed surface and interconnected porosity were prepared by electrospinning (ES) from Hmi-containing feed solutions. Structural organization of the PHB/Hmi materials (porosity, uniform structure, diameter of fibers, surface area, distribution of Hmi within the PHB matrix, phase composition, etc.) is shown to be governed by the ES conditions: the presence of even minor amounts of Hmi in the PHB/Hmi (below 5 wt %) serves as a powerful tool for the control over their structure, performance, and biodegradation. Service characteristics of the PHB/Hmi materials (wettability, prolonged release of Hmi, antibacterial activity, breathability, and mechanical properties) were studied by different physicochemical methods (scanning electron microscopy, Fourier transform infrared spectroscopy, energy-dispersive X-ray spectroscopy, differential scanning calorimetry, contact angle measurements, antibacterial tests, etc.). The effect of the structural organization of the PHB/Hmi materials on their in-soil biodegradation at the end of life was analyzed, and key factors providing efficient biodegradation of the PHB/Hmi materials at all stages (from adaptation to mineralization) are highlighted (high surface area and porosity, thin fibers, release of Hmi, etc.). The proposed approach allows for target-oriented preparation and structural design of the functional PHB/Hmi nonwovens when their structural supramolecular organization with a highly developed surface area controls both their service properties as efficient antibacterial materials and in-soil biodegradation upon the end of life.


Asunto(s)
Materiales Biocompatibles , Hemina , Animales , Materiales Biocompatibles/química , Polihidroxibutiratos , Hidroxibutiratos/química , Antibacterianos/química , Estadios del Ciclo de Vida , Muerte , Suelo
19.
Sci Rep ; 14(1): 2797, 2024 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-38307876

RESUMEN

Hepatic fibrosis remains a significant clinical challenge due to ineffective treatments. 4-methylumbelliferone (4MU), a hyaluronic acid (HA) synthesis inhibitor, has proven safe in phase one clinical trials. In this study, we aimed to ameliorate liver fibrosis by inhibiting HA synthesis. We compared two groups of mice with CCl4-induced fibrosis, treated with 4-methylumbelliferone (4MU) and hyaluronan synthase 2 (HAS2) targeting siRNA (siHAS2). The administration of 4MU and siHAS2 significantly reduced collagen and HA deposition, as well as biochemical markers of hepatic damage induced by repeated CCl4 injections. The transcriptomic analysis revealed converging pathways associated with downstream HA signalling. 4MU- and siHAS2-treated fibrotic livers shared 405 upregulated and 628 downregulated genes. These genes were associated with xenobiotic and cholesterol metabolism, mitosis, endoplasmic reticulum stress, RNA processing, and myeloid cell migration. The functional annotation of differentially expressed genes (DEGs) in siHAS2-treated mice revealed attenuation of extracellular matrix-associated pathways. In comparison, in the 4MU-treated group, DEGs were related to lipid and bile metabolism pathways and cell cycle. These findings confirm that HAS2 is an important pharmacological target for suppressing hepatic fibrosis using siRNA.


Asunto(s)
Ácido Hialurónico , Himecromona , Animales , Ratones , Perfilación de la Expresión Génica , Hialuronano Sintasas/genética , Hialuronano Sintasas/metabolismo , Ácido Hialurónico/metabolismo , Himecromona/farmacología , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/genética , ARN Interferente Pequeño
20.
Heliyon ; 10(6): e27538, 2024 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-38509939

RESUMEN

The plasmonic sensors based on silver nanoparticles are limited in application due to their relatively fast degradation in the ambient atmosphere. The technology of ion-beam modification for the creation of monocrystalline silver nanoparticles (NPs) with stable plasmonic properties will expand the application of silver nanostructures. In the present study, highly-stable monocrystalline NPs were formed on the basis of a thin silver film by low-energy ion irradiation. Combined with lithography, this technique allows the creation of nanoparticle ensembles in variant forms. The characterization of the nanoparticles formed by ion-beam modification showed long-term outstanding for Ag nanoparticles stability of their plasmonic properties due to their monocrystalline structure. According to optical spectroscopy data, the reliable plasmonic properties in the ambient atmosphere are preserved for up to 39 days. The mapping of crystal violet dye via surface-enhanced Raman spectroscopy (SERS) revealed a strong amplification factor sustaining at least thrice as long as the one of similarly sized polycrystalline silver NPs formed by annealing. The plasmonic properties sustain more than a month of storage in the ambient atmosphere. Thus, ion-beam modification of silver film makes it possible to fabricate NPs with stable plasmonic properties and form clusters of NPs for sensor technology and SERS applications.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA