Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Cell ; 165(4): 921-35, 2016 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-27114033

RESUMEN

Microglia maintain homeostasis in the brain, but whether aberrant microglial activation can cause neurodegeneration remains controversial. Here, we use transcriptome profiling to demonstrate that deficiency in frontotemporal dementia (FTD) gene progranulin (Grn) leads to an age-dependent, progressive upregulation of lysosomal and innate immunity genes, increased complement production, and enhanced synaptic pruning in microglia. During aging, Grn(-/-) mice show profound microglia infiltration and preferential elimination of inhibitory synapses in the ventral thalamus, which lead to hyperexcitability in the thalamocortical circuits and obsessive-compulsive disorder (OCD)-like grooming behaviors. Remarkably, deleting C1qa gene significantly reduces synaptic pruning by Grn(-/-) microglia and mitigates neurodegeneration, behavioral phenotypes, and premature mortality in Grn(-/-) mice. Together, our results uncover a previously unrecognized role of progranulin in suppressing aberrant microglia activation during aging. These results represent an important conceptual advance that complement activation and microglia-mediated synaptic pruning are major drivers, rather than consequences, of neurodegeneration caused by progranulin deficiency.


Asunto(s)
Envejecimiento/metabolismo , Encéfalo/metabolismo , Activación de Complemento , Complemento C1q/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Microglía/metabolismo , Envejecimiento/inmunología , Animales , Líquido Cefalorraquídeo , Complemento C1q/genética , Demencia Frontotemporal/genética , Demencia Frontotemporal/metabolismo , Granulinas , Humanos , Inmunidad Innata , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Péptidos y Proteínas de Señalización Intercelular/genética , Lisosomas/metabolismo , Redes y Vías Metabólicas , Ratones , Trastorno Obsesivo Compulsivo/genética , Trastorno Obsesivo Compulsivo/metabolismo , Progranulinas , Sinapsis/metabolismo , Tálamo/metabolismo
2.
J Immunol ; 210(2): 119-125, 2023 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-36596217

RESUMEN

The complement field has recently experienced a strong resurgence of interest because of the unexpected discovery of new complement functions extending complement's role beyond immunity and pathogen clearance, a growing list of diseases in which complement plays a role, and the proliferation of complement therapeutics. Importantly, although the majority of complement components in the circulation are generated by the liver and activated extracellularly, complement activation unexpectedly also occurs intracellularly across a broad range of cells. Such cell-autonomous complement activation can engage intracellular complement receptors, which then drive noncanonical cell-specific effector functions. Thus, much remains to be discovered about complement biology. In this brief review, we focus on novel noncanonical activities of complement in its "classic areas of operation" (kidney and brain biology, infection, and autoimmunity), with an outlook on the next generation of complement-targeted therapeutics.


Asunto(s)
Activación de Complemento , Proteínas del Sistema Complemento
3.
Neurobiol Dis ; 181: 106094, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-36990364

RESUMEN

Generalized epilepsy affects 24 million people globally; at least 25% of cases remain medically refractory. The thalamus, with widespread connections throughout the brain, plays a critical role in generalized epilepsy. The intrinsic properties of thalamic neurons and the synaptic connections between populations of neurons in the nucleus reticularis thalami and thalamocortical relay nuclei help generate different firing patterns that influence brain states. In particular, transitions from tonic firing to highly synchronized burst firing mode in thalamic neurons can cause seizures that rapidly generalize and cause altered awareness and unconsciousness. Here, we review the most recent advances in our understanding of how thalamic activity is regulated and discuss the gaps in our understanding of the mechanisms of generalized epilepsy syndromes. Elucidating the role of the thalamus in generalized epilepsy syndromes may lead to new opportunities to better treat pharmaco-resistant generalized epilepsy by thalamic modulation and dietary therapy.


Asunto(s)
Epilepsia Tipo Ausencia , Epilepsia Generalizada , Epilepsia Generalizada/terapia , Humanos , Convulsiones , Tálamo
4.
Neurobiol Dis ; 186: 106263, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37591465

RESUMEN

The R47H variant of triggering receptor expressed on myeloid cells 2 (TREM2) increases the risk of Alzheimer's disease (AD). To investigate potential mechanisms, we analyzed knockin mice expressing human TREM2-R47H from one mutant mouse Trem2 allele. TREM2-R47H mice showed increased seizure activity in response to an acute excitotoxin challenge, compared to wildtype controls or knockin mice expressing the common variant of human TREM2. TREM2-R47H also increased spontaneous thalamocortical epileptiform activity in App knockin mice expressing amyloid precursor proteins bearing autosomal dominant AD mutations and a humanized amyloid-ß sequence. In mice with or without such App modifications, TREM2-R47H increased the density of putative synapses in cortical regions without amyloid plaques. TREM2-R47H did not affect synaptic density in hippocampal regions with or without plaques. We conclude that TREM2-R47H increases AD-related network hyperexcitability and that it may do so, at least in part, by causing an imbalance in synaptic densities across brain regions.


Asunto(s)
Enfermedad de Alzheimer , Humanos , Animales , Ratones , Enfermedad de Alzheimer/genética , Alelos , Convulsiones , Péptidos beta-Amiloides , Modelos Animales de Enfermedad , Placa Amiloide , Sinapsis , Glicoproteínas de Membrana/genética , Receptores Inmunológicos/genética
5.
Epilepsia ; 64(10): e214-e221, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37501613

RESUMEN

The solute carrier family 6 member 1 (SLC6A1) gene encodes GAT-1, a γ-aminobutyric acid transporter expressed on astrocytes and inhibitory neurons. Mutations in SLC6A1 are associated with epilepsy and developmental disorders, including motor and social impairments, but variant-specific animal models are needed to elucidate mechanisms. Here, we report electrocorticographic (ECoG) recordings and clinical data from a patient with a variant in SLC6A1 that encodes GAT-1 with a serine-to-leucine substitution at amino acid 295 (S295L), who was diagnosed with childhood absence epilepsy. Next, we show that mice bearing the S295L mutation (GAT-1S295L/+ ) have spike-and-wave discharges with motor arrest consistent with absence-type seizures, similar to GAT-1+/- mice. GAT-1S295L/+ and GAT-1+/- mice follow the same pattern of pharmacosensitivity, being bidirectionally modulated by ethosuximide (200 mg/kg ip) and the GAT-1 antagonist NO-711 (10 mg/kg ip). By contrast, GAT-1-/- mice were insensitive to both ethosuximide and NO-711 at the doses tested. In conclusion, ECoG findings in GAT-1S295L/+ mice phenocopy GAT-1 haploinsufficiency and provide a useful preclinical model for drug screening and gene therapy investigations.


Asunto(s)
Epilepsia Tipo Ausencia , Etosuximida , Humanos , Ratones , Animales , Niño , Etosuximida/uso terapéutico , Haploinsuficiencia/genética , Ácidos Nipecóticos/uso terapéutico , Epilepsia Tipo Ausencia/tratamiento farmacológico , Proteínas Transportadoras de GABA en la Membrana Plasmática/genética , Proteínas Transportadoras de GABA en la Membrana Plasmática/metabolismo
6.
J Neurosci ; 38(22): 5209-5219, 2018 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-29712783

RESUMEN

Persistent neural activity, the substrate of working memory, is thought to emerge from synaptic reverberation within recurrent networks. However, reverberation models do not robustly explain the fundamental dynamics of persistent activity, including high-spiking irregularity, large intertrial variability, and state transitions. While cellular bistability may contribute to persistent activity, its rigidity appears incompatible with persistent activity labile characteristics. Here, we unravel in a cellular model a form of spike-mediated conditional bistability that is robust and generic. and provides a rich repertoire of mnemonic computations. Under asynchronous synaptic inputs of the awakened state, conditional bistability generates spiking/bursting episodes, accounting for the irregularity, variability, and state transitions characterizing persistent activity. This mechanism has likely been overlooked because of the subthreshold input it requires, and we predict how to assess it experimentally. Our results suggest a reexamination of the role of intrinsic properties in the collective network dynamics responsible for flexible working memory.SIGNIFICANCE STATEMENT This study unravels a novel form of intrinsic neuronal property: conditional bistability. We show that, thanks to its conditional character, conditional bistability favors the emergence of flexible and robust forms of persistent activity in PFC neural networks, in opposition to previously studied classical forms of absolute bistability. Specifically, we demonstrate for the first time that conditional bistability (1) is a generic biophysical spike-dependent mechanism of layer V pyramidal neurons in the PFC and that (2) it accounts for essential neurodynamical features for the organization and flexibility of PFC persistent activity (the large irregularity and intertrial variability of the discharge and its organization under discrete stable states), which remain unexplained in a robust fashion by current models.


Asunto(s)
Algoritmos , Memoria a Corto Plazo/fisiología , Neuronas/fisiología , Potenciales de Acción/fisiología , Canales de Calcio/fisiología , Simulación por Computador , Humanos , Modelos Neurológicos , Red Nerviosa/fisiología , Redes Neurales de la Computación , Células Piramidales/fisiología , Sinapsis , Vigilia/fisiología , Sustancia Blanca/fisiología
7.
Brain ; 139(Pt 2): 468-80, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26685158

RESUMEN

Ischaemic stroke is the leading cause of severe long-term disability yet lacks drug therapies that promote the repair phase of recovery. This repair phase of stroke occurs days to months after stroke onset and involves brain remapping and plasticity within the peri-infarct zone. Elucidating mechanisms that promote this plasticity is critical for the development of new therapeutics with a broad treatment window. Inhibiting tonic (extrasynaptic) GABA signalling during the repair phase was reported to enhance functional recovery in mice suggesting that GABA plays an important function in modulating brain repair. While tonic GABA appears to suppress brain repair after stroke, less is known about the role of phasic (synaptic) GABA during the repair phase. We observed an increase in postsynaptic phasic GABA signalling in mice within the peri-infarct cortex specific to layer 5; we found increased numbers of α1 receptor subunit-containing GABAergic synapses detected using array tomography, and an associated increased efficacy of spontaneous and miniature inhibitory postsynaptic currents in pyramidal neurons. Furthermore, we demonstrate that enhancing phasic GABA signalling using zolpidem, a Food and Drug Administration (FDA)-approved GABA-positive allosteric modulator, during the repair phase improved behavioural recovery. These data identify potentiation of phasic GABA signalling as a novel therapeutic strategy, indicate zolpidem's potential to improve recovery, and underscore the necessity to distinguish the role of tonic and phasic GABA signalling in stroke recovery.


Asunto(s)
Sistemas de Liberación de Medicamentos , Agonistas de Receptores de GABA-A/administración & dosificación , Inhibición Neural/fisiología , Piridinas/administración & dosificación , Receptores de GABA-A/fisiología , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Sistemas de Liberación de Medicamentos/tendencias , Masculino , Ratones , Ratones Endogámicos C57BL , Neocórtex/efectos de los fármacos , Neocórtex/fisiología , Inhibición Neural/efectos de los fármacos , Técnicas de Cultivo de Órganos , Recuperación de la Función/efectos de los fármacos , Recuperación de la Función/fisiología , Accidente Cerebrovascular/patología , Accidente Cerebrovascular/fisiopatología , Zolpidem
9.
Nature ; 477(7363): 171-8, 2011 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-21796121

RESUMEN

Severe behavioural deficits in psychiatric diseases such as autism and schizophrenia have been hypothesized to arise from elevations in the cellular balance of excitation and inhibition (E/I balance) within neural microcircuitry. This hypothesis could unify diverse streams of pathophysiological and genetic evidence, but has not been susceptible to direct testing. Here we design and use several novel optogenetic tools to causally investigate the cellular E/I balance hypothesis in freely moving mammals, and explore the associated circuit physiology. Elevation, but not reduction, of cellular E/I balance within the mouse medial prefrontal cortex was found to elicit a profound impairment in cellular information processing, associated with specific behavioural impairments and increased high-frequency power in the 30-80 Hz range, which have both been observed in clinical conditions in humans. Consistent with the E/I balance hypothesis, compensatory elevation of inhibitory cell excitability partially rescued social deficits caused by E/I balance elevation. These results provide support for the elevated cellular E/I balance hypothesis of severe neuropsychiatric disease-related symptoms.


Asunto(s)
Modelos Neurológicos , Inhibición Neural/fisiología , Neuronas/metabolismo , Corteza Prefrontal/fisiología , Corteza Prefrontal/fisiopatología , Conducta Social , Animales , Trastorno Autístico/fisiopatología , Modelos Animales de Enfermedad , Células HEK293 , Hipocampo/citología , Humanos , Aprendizaje , Trastornos Mentales/fisiopatología , Ratones , Actividad Motora , Opsinas/metabolismo , Esquizofrenia/fisiopatología
10.
J Exp Med ; 220(2)2023 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-36520518

RESUMEN

Microglia are critical regulators of brain development that engulf synaptic proteins during postnatal synapse remodeling. However, the mechanisms through which microglia sense the brain environment are not well defined. Here, we characterized the regulatory program downstream of interleukin-33 (IL-33), a cytokine that promotes microglial synapse remodeling. Exposing the developing brain to a supraphysiological dose of IL-33 altered the microglial enhancer landscape and increased binding of stimulus-dependent transcription factors including AP-1/FOS. This induced a gene expression program enriched for the expression of pattern recognition receptors, including the scavenger receptor MARCO. CNS-specific deletion of IL-33 led to increased excitatory/inhibitory synaptic balance, spontaneous absence-like epileptiform activity in juvenile mice, and increased seizure susceptibility in response to chemoconvulsants. We found that MARCO promoted synapse engulfment, and Marco-deficient animals had excess thalamic excitatory synapses and increased seizure susceptibility. Taken together, these data define coordinated epigenetic and functional changes in microglia and uncover pattern recognition receptors as potential regulators of postnatal synaptic refinement.


Asunto(s)
Interleucina-33 , Microglía , Animales , Ratones , Microglía/metabolismo , Interleucina-33/metabolismo , Sinapsis/metabolismo , Encéfalo/metabolismo , Convulsiones/metabolismo , Ratones Endogámicos C57BL
11.
J Comp Neurol ; 530(7): 998-1019, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34633669

RESUMEN

While cortical injuries, such as traumatic brain injury (TBI) and neocortical stroke, acutely disrupt the neocortex, most of their consequent disabilities reflect secondary injuries that develop over time. Thalamic neuroinflammation has been proposed to be a biomarker of cortical injury and of the long-term cognitive and neurological deficits that follow. However, the extent to which thalamic neuroinflammation depends on the type of cortical injury or its location remains unknown. Using two mouse models of focal neocortical injury that do not directly damage subcortical structures-controlled cortical impact and photothrombotic ischemic stroke-we found that chronic neuroinflammation in the thalamic region mirrors the functional connections with the injured cortex, and that sensory corticothalamic regions may be more likely to sustain long-term damage than nonsensory circuits. Currently, heterogeneous clinical outcomes complicate treatment. Understanding how thalamic inflammation depends on the injury site can aid in predicting features of subsequent deficits and lead to more effective, customized therapies.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Accidente Cerebrovascular , Animales , Lesiones Traumáticas del Encéfalo/complicaciones , Modelos Animales de Enfermedad , Ratones , Microglía , Enfermedades Neuroinflamatorias , Accidente Cerebrovascular/complicaciones , Tálamo
12.
Sci Transl Med ; 14(652): eabj4310, 2022 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-35857628

RESUMEN

Inflammatory processes induced by brain injury are important for recovery; however, when uncontrolled, inflammation can be deleterious, likely explaining why most anti-inflammatory treatments have failed to improve neurological outcomes after brain injury in clinical trials. In the thalamus, chronic activation of glial cells, a proxy of inflammation, has been suggested as an indicator of increased seizure risk and cognitive deficits that develop after cortical injury. Furthermore, lesions in the thalamus, more than other brain regions, have been reported in patients with viral infections associated with neurological deficits, such as SARS-CoV-2. However, the extent to which thalamic inflammation is a driver or by-product of neurological deficits remains unknown. Here, we found that thalamic inflammation in mice was sufficient to phenocopy the cellular and circuit hyperexcitability, enhanced seizure risk, and disruptions in cortical rhythms that develop after cortical injury. In our model, down-regulation of the GABA transporter GAT-3 in thalamic astrocytes mediated this neurological dysfunction. In addition, GAT-3 was decreased in regions of thalamic reactive astrocytes in mouse models of cortical injury. Enhancing GAT-3 in thalamic astrocytes prevented seizure risk, restored cortical states, and was protective against severe chemoconvulsant-induced seizures and mortality in a mouse model of traumatic brain injury, emphasizing the potential of therapeutically targeting this pathway. Together, our results identified a potential therapeutic target for reducing negative outcomes after brain injury.


Asunto(s)
Lesiones Encefálicas , COVID-19 , Animales , Astrocitos/metabolismo , Modelos Animales de Enfermedad , Proteínas Transportadoras de GABA en la Membrana Plasmática/metabolismo , Inflamación/patología , Ratones , Polímeros , Roedores/metabolismo , SARS-CoV-2 , Convulsiones , Tálamo/metabolismo , Tálamo/patología
13.
J Neurosci ; 30(15): 5465-79, 2010 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-20392967

RESUMEN

Focal cortical injuries result in death of cortical neurons and their efferents and ultimately in death or damage of thalamocortical relay (TCR) neurons that project to the affected cortical area. Neurons of the inhibitory reticular thalamic nucleus (nRT) receive excitatory inputs from corticothalamic and thalamocortical axons and are thus denervated by such injuries, yet nRT cells generally survive these insults to a greater degree than TCR cells. nRT cells inhibit TCR cells, regulate thalamocortical transmission, and generate cerebral rhythms including those involved in thalamocortical epilepsies. The survival and reorganization of nRT after cortical injury would determine recovery of thalamocortical circuits after injury. However, the physiological properties and connectivity of the survivors remain unknown. To study possible alterations in nRT neurons, we used the rat photothrombosis model of cortical stroke. Using in vitro patch-clamp recordings at various times after the photothrombotic injury, we show that localized strokes in the somatosensory cortex induce long-term reductions in intrinsic excitability and evoked synaptic excitation of nRT cells by the end of the first week after the injury. We find that nRT neurons in injured rats show (1) decreased membrane input resistance, (2) reduced low-threshold calcium burst responses, and (3) weaker evoked excitatory synaptic responses. Such alterations in nRT cellular excitability could lead to loss of nRT-mediated inhibition in relay nuclei, increased output of surviving TCR cells, and enhanced thalamocortical excitation, which may facilitate recovery of thalamic and cortical sensory circuits. In addition, such changes could be maladaptive, leading to injury-induced epilepsy.


Asunto(s)
Infarto Encefálico/fisiopatología , Corteza Cerebral/fisiopatología , Neuronas/fisiología , Sinapsis/fisiología , Transmisión Sináptica/fisiología , Núcleos Talámicos/fisiopatología , Animales , Infarto Encefálico/patología , Calcio/metabolismo , Canales de Calcio Tipo T/metabolismo , Muerte Celular , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Impedancia Eléctrica , Potenciales Evocados , Técnicas In Vitro , Potenciales de la Membrana/fisiología , Vías Nerviosas/fisiopatología , Plasticidad Neuronal/fisiología , Neuronas/patología , Ratas , Ratas Sprague-Dawley , Accidente Cerebrovascular/fisiopatología , Sinapsis/patología , Núcleos Talámicos/patología , Factores de Tiempo
14.
Elife ; 102021 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-33843585

RESUMEN

Visual perception in natural environments depends on the ability to focus on salient stimuli while ignoring distractions. This kind of selective visual attention is associated with gamma activity in the visual cortex. While the nucleus reticularis thalami (nRT) has been implicated in selective attention, its role in modulating gamma activity in the visual cortex remains unknown. Here, we show that somatostatin- (SST) but not parvalbumin-expressing (PV) neurons in the visual sector of the nRT preferentially project to the dorsal lateral geniculate nucleus (dLGN), and modulate visual information transmission and gamma activity in primary visual cortex (V1). These findings pinpoint the SST neurons in nRT as powerful modulators of the visual information encoding accuracy in V1 and represent a novel circuit through which the nRT can influence representation of visual information.


Asunto(s)
Ritmo Gamma/fisiología , Neuronas/fisiología , Núcleos Talámicos/fisiología , Corteza Visual/fisiología , Percepción Visual/fisiología , Animales , Femenino , Masculino , Ratones , Somatostatina/metabolismo
15.
Science ; 373(6560): eabj2685, 2021 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-34516796

RESUMEN

Although traumatic brain injury (TBI) acutely disrupts the cortex, most TBI-related disabilities reflect secondary injuries that accrue over time. The thalamus is a likely site of secondary damage because of its reciprocal connections with the cortex. Using a mouse model of mild TBI (mTBI), we found a chronic increase in C1q expression specifically in the corticothalamic system. Increased C1q expression colocalized with neuron loss and chronic inflammation and correlated with disruption in sleep spindles and emergence of epileptic activities. Blocking C1q counteracted these outcomes, suggesting that C1q is a disease modifier in mTBI. Single-nucleus RNA sequencing demonstrated that microglia are a source of thalamic C1q. The corticothalamic circuit could thus be a new target for treating TBI-related disabilities.


Asunto(s)
Lesiones Encefálicas/complicaciones , Complemento C1q/fisiología , Fases del Sueño , Trastornos del Sueño-Vigilia/etiología , Trastornos del Sueño-Vigilia/fisiopatología , Tálamo/fisiopatología , Animales , Lesiones Encefálicas/fisiopatología , Complemento C1q/genética , Modelos Animales de Enfermedad , Epilepsia/fisiopatología , Ratones , Microglía/metabolismo , Tálamo/metabolismo
16.
Mol Neurodegener ; 15(1): 53, 2020 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-32921309

RESUMEN

BACKGROUND: Alzheimer's disease (AD) is the most frequent and costly neurodegenerative disorder. Although diverse lines of evidence suggest that the amyloid precursor protein (APP) is involved in its causation, the precise mechanisms remain unknown and no treatments are available to prevent or halt the disease. A favorite hypothesis has been that APP contributes to AD pathogenesis through the cerebral accumulation of the amyloid-ß peptide (Aß), which is derived from APP through sequential proteolytic cleavage by BACE1 and γ-secretase. However, inhibitors of these enzymes have failed in clinical trials despite clear evidence for target engagement. METHODS: To further elucidate the roles of APP and its metabolites in AD pathogenesis, we analyzed transgenic mice overexpressing wildtype human APP (hAPP) or hAPP carrying mutations that cause autosomal dominant familial AD (FAD), as well as App knock-in mice that do not overexpress hAPP but have two mouse App alleles with FAD mutations and a humanized Aß sequence. RESULTS: Although these lines of mice had marked differences in cortical and hippocampal levels of APP, APP C-terminal fragments, soluble Aß, Aß oligomers and age-dependent amyloid deposition, they all developed cognitive deficits as well as non-convulsive epileptiform activity, a type of network dysfunction that also occurs in a substantive proportion of humans with AD. Pharmacological inhibition of BACE1 effectively reduced levels of amyloidogenic APP C-terminal fragments (C99), soluble Aß, Aß oligomers, and amyloid deposits in transgenic mice expressing FAD-mutant hAPP, but did not improve their network dysfunction and behavioral abnormalities, even when initiated at early stages before amyloid deposits were detectable. CONCLUSIONS: hAPP transgenic and App knock-in mice develop similar pathophysiological alterations. APP and its metabolites contribute to AD-related functional alterations through complex combinatorial mechanisms that may be difficult to block with BACE inhibitors and, possibly, also with other anti-Aß treatments.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Conducta Animal/fisiología , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Red Nerviosa/metabolismo , Red Nerviosa/patología
17.
Elife ; 92020 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-32452758

RESUMEN

​Maf (c-Maf) and Mafb transcription factors (TFs) have compensatory roles in repressing somatostatin (SST+) interneuron (IN) production in medial ganglionic eminence (MGE) secondary progenitors in mice. Maf and Mafb conditional deletion (cDKO) decreases the survival of MGE-derived cortical interneurons (CINs) and changes their physiological properties. Herein, we show that (1) Mef2c and Snap25 are positively regulated by Maf and Mafb to drive IN morphological maturation; (2) Maf and Mafb promote Mef2c expression which specifies parvalbumin (PV+) INs; (3) Elmo1, Igfbp4 and Mef2c are candidate markers of immature PV+ hippocampal INs (HIN). Furthermore, Maf/Mafb neonatal cDKOs have decreased CINs and increased HINs, that express Pnoc, an HIN specific marker. Our findings not only elucidate key gene targets of Maf and Mafb that control IN development, but also identify for the first time TFs that differentially regulate CIN vs. HIN production.


Asunto(s)
Regulación de la Expresión Génica , Interneuronas/metabolismo , Factor de Transcripción MafB/fisiología , Proteínas Proto-Oncogénicas c-maf/fisiología , Animales , Femenino , Factores de Transcripción MEF2/metabolismo , Ratones , Enfermedades del Sistema Nervioso/etiología , Embarazo , Precursores de Proteínas/genética , Receptores CXCR4/metabolismo , Receptores Opioides/genética , Análisis de la Célula Individual , Proteína 25 Asociada a Sinaptosomas/metabolismo , Transcriptoma
18.
J Physiol ; 587(Pt 13): 3189-205, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19433575

RESUMEN

Synaptic plasticity is classically considered as the neuronal substrate for learning and memory. However, activity-dependent changes in neuronal intrinsic excitability have been reported in several learning-related brain regions, suggesting that intrinsic plasticity could also participate to information storage. Compared to synaptic plasticity, there has been little exploration of the properties of induction and expression of intrinsic plasticity in an intact brain. Here, by the means of in vivo intracellular recordings in the rat we have examined how the intrinsic excitability of layer V motor cortex pyramidal neurones is altered following brief periods of repeated firing. Changes in membrane excitability were assessed by modifications in the discharge frequency versus injected current (F-I) curves. Most (approximately 64%) conditioned neurones exhibited a long-lasting intrinsic plasticity, which was expressed either by selective changes in the current threshold or in the slope of the F-I curve, or by concomitant changes in both parameters. These modifications in the neuronal input-output relationship led to a global increase or decrease in intrinsic excitability. Passive electrical membrane properties were unaffected by the intracellular conditioning, indicating that intrinsic plasticity resulted from modifications of voltage-gated ion channels. These results demonstrate that neocortical pyramidal neurones can express in vivo a bidirectional use-dependent intrinsic plasticity, modifying their sensitivity to weak inputs and/or the gain of their input-output function. These multiple forms of experience-dependent intrinsic changes, which expand the computational abilities of individual neurones, could shape new network dynamics and thus might participate in the formation of mnemonic motor engrams.


Asunto(s)
Plasticidad Neuronal/fisiología , Células Piramidales/fisiología , Animales , Condicionamiento Psicológico/fisiología , Estimulación Eléctrica , Fenómenos Electrofisiológicos , Memoria/fisiología , Modelos Neurológicos , Corteza Motora/citología , Corteza Motora/fisiología , Neuronas Motoras/fisiología , Neocórtex/citología , Neocórtex/fisiología , Ratas , Ratas Sprague-Dawley
19.
Epilepsy Curr ; 24(3): 203-205, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38898906
20.
Cell Rep ; 26(5): 1157-1173.e5, 2019 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-30699346

RESUMEN

Mafb and c-Maf transcription factor (TF) expression is enriched in medial ganglionic eminence (MGE) lineages, beginning in late-secondary progenitors and continuing into mature parvalbumin (PV+) and somatostatin (SST+) interneurons. However, the functions of Maf TFs in MGE development remain to be elucidated. Herein, Mafb and c-Maf were conditionally deleted, alone and together, in the MGE and its lineages. Analyses of Maf mutant mice revealed redundant functions of Mafb and c-Maf in secondary MGE progenitors, where they repress the generation of SST+ cortical and hippocampal interneurons. By contrast, Mafb and c-Maf have distinct roles in postnatal cortical interneuron (CIN) morphological maturation, synaptogenesis, and cortical circuit integration. Thus, Mafb and c-Maf have redundant and opposing functions at different steps in CIN development.


Asunto(s)
Linaje de la Célula , Corteza Cerebral/metabolismo , Interneuronas/metabolismo , Factor de Transcripción MafB/metabolismo , Proteínas Proto-Oncogénicas c-maf/metabolismo , Potenciales de Acción , Animales , Animales Recién Nacidos , Apoptosis , Membrana Celular/metabolismo , Movimiento Celular , Proliferación Celular , Hipocampo/metabolismo , Eminencia Media/metabolismo , Ratones Noqueados , Neuritas/metabolismo , Neurogénesis , Parvalbúminas/metabolismo , Somatostatina/metabolismo , Sinapsis/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA