Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.284
Filtrar
1.
Nature ; 628(8009): 818-825, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38658687

RESUMO

Timothy syndrome (TS) is a severe, multisystem disorder characterized by autism, epilepsy, long-QT syndrome and other neuropsychiatric conditions1. TS type 1 (TS1) is caused by a gain-of-function variant in the alternatively spliced and developmentally enriched CACNA1C exon 8A, as opposed to its counterpart exon 8. We previously uncovered several phenotypes in neurons derived from patients with TS1, including delayed channel inactivation, prolonged depolarization-induced calcium rise, impaired interneuron migration, activity-dependent dendrite retraction and an unanticipated persistent expression of exon 8A2-6. We reasoned that switching CACNA1C exon utilization from 8A to 8 would represent a potential therapeutic strategy. Here we developed antisense oligonucleotides (ASOs) to effectively decrease the inclusion of exon 8A in human cells both in vitro and, following transplantation, in vivo. We discovered that the ASO-mediated switch from exon 8A to 8 robustly rescued defects in patient-derived cortical organoids and migration in forebrain assembloids. Leveraging a transplantation platform previously developed7, we found that a single intrathecal ASO administration rescued calcium changes and in vivo dendrite retraction of patient neurons, suggesting that suppression of CACNA1C exon 8A expression is a potential treatment for TS1. Broadly, these experiments illustrate how a multilevel, in vivo and in vitro stem cell model-based approach can identify strategies to reverse disease-relevant neural pathophysiology.


Assuntos
Transtorno Autístico , Canais de Cálcio Tipo L , Movimento Celular , Éxons , Síndrome do QT Longo , Neurônios , Oligonucleotídeos Antissenso , Sindactilia , Humanos , Oligonucleotídeos Antissenso/uso terapêutico , Oligonucleotídeos Antissenso/farmacologia , Oligonucleotídeos Antissenso/administração & dosagem , Canais de Cálcio Tipo L/metabolismo , Canais de Cálcio Tipo L/genética , Transtorno Autístico/genética , Transtorno Autístico/terapia , Transtorno Autístico/tratamento farmacológico , Neurônios/metabolismo , Neurônios/efeitos dos fármacos , Éxons/genética , Sindactilia/genética , Sindactilia/terapia , Animais , Síndrome do QT Longo/genética , Síndrome do QT Longo/tratamento farmacológico , Síndrome do QT Longo/terapia , Camundongos , Movimento Celular/efeitos dos fármacos , Cálcio/metabolismo , Organoides/metabolismo , Prosencéfalo/metabolismo , Prosencéfalo/citologia , Processamento Alternativo/genética , Masculino , Dendritos/metabolismo , Dendritos/efeitos dos fármacos , Feminino
2.
Dev Neurobiol ; 84(2): 47-58, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38466218

RESUMO

In sexually dimorphic zebra finches (Taeniopygia guttata), only males learn to sing their father's song, whereas females learn to recognize the songs of their father or mate but cannot sing themselves. Memory of learned songs is behaviorally expressed in females by preferring familiar songs over unfamiliar ones. Auditory association regions such as the caudomedial mesopallium (CMM; or caudal mesopallium) have been shown to be key nodes in a network that supports preferences for learned songs in adult females. However, much less is known about how song preferences develop during the sensitive period of learning in juvenile female zebra finches. In this study, we used blood-oxygen level-dependent (BOLD) functional magnetic resonance imaging (fMRI) to trace the development of a memory-based preference for the father's song in female zebra finches. Using BOLD fMRI, we found that only in adult female zebra finches with a preference for learned song over novel conspecific song, neural selectivity for the father's song was localized in the thalamus (dorsolateral nucleus of the medial thalamus; part of the anterior forebrain pathway, AFP) and in CMM. These brain regions also showed a selective response in juvenile female zebra finches, although activation was less prominent. These data reveal that neural responses in CMM, and perhaps also in the AFP, are shaped during development to support behavioral preferences for learned songs.


Assuntos
Tentilhões , Vocalização Animal , Masculino , Animais , Feminino , Vocalização Animal/fisiologia , alfa-Fetoproteínas/metabolismo , Tentilhões/metabolismo , Estimulação Acústica/métodos , Percepção Auditiva/fisiologia , Prosencéfalo/metabolismo , Imageamento por Ressonância Magnética/métodos
3.
J Alzheimers Dis ; 98(1): 301-318, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38427475

RESUMO

Background: Alzheimer's disease (AD) is characterized by disrupted proteostasis and macroautophagy (hereafter "autophagy"). The pharmacological agent suramin has known autophagy modulation properties with potential efficacy in mitigating AD neuronal pathology. Objective: In the present work, we investigate the impact of forebrain neuron exposure to suramin on the Akt/mTOR signaling pathway, a major regulator of autophagy, in comparison with rapamycin and chloroquine. We further investigate the effect of suramin on several AD-related biomarkers in sporadic AD (sAD)-derived forebrain neurons. Methods: Neurons differentiated from ReNcell neural progenitors were used to assess the impact of suramin on the Akt/mTOR signaling pathway relative to the autophagy inducer rapamycin and autophagy inhibitor chloroquine. Mature forebrain neurons were differentiated from induced pluripotent stem cells (iPSCs) sourced from a late-onset sAD patient and treated with 100µM suramin for 72 h, followed by assessments for amyloid-ß, phosphorylated tau, oxidative/nitrosative stress, and synaptic puncta density. Results: Suramin treatment of sAD-derived neurons partially ameliorated the increased p-Tau(S199)/Tau ratio, and fully remediated the increased glutathione to oxidized nitric oxide ratio, observed in untreated sAD-derived neurons relative to healthy controls. These positive results may be due in part to the distinct increases in Akt/mTOR pathway mediator p-p70S6K noted with suramin treatment of both ReNcell-derived and iPSC-derived neurons. Longer term neuronal markers, such as synaptic puncta density, were unaffected by suramin treatment. Conclusions: These findings provide initial evidence supporting the potential of suramin to reduce the degree of dysregulation in sAD-derived forebrain neurons in part via the modulation of autophagy.


Assuntos
Doença de Alzheimer , Células-Tronco Pluripotentes Induzidas , Humanos , Doença de Alzheimer/patologia , Suramina/farmacologia , Suramina/metabolismo , Proteínas tau/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Peptídeos beta-Amiloides/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Prosencéfalo/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Neurônios/metabolismo , Sirolimo/farmacologia , Cloroquina/metabolismo , Cloroquina/farmacologia
4.
Brain Struct Funct ; 229(3): 759-773, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38411929

RESUMO

Malformation during cortical development can disrupt the balance of excitatory and inhibitory neural circuits, contributing to various psychiatric and developmental disorders. One of the critical factors of cortical neural networks is the fine regulation of neurogenesis through mechanical cues, such as shear stress and substrate stiffness. Piezo1, a mechanically-activated channel, serves as a transducer for these mechanical cues, regulating embryogenesis. However, specific cell-type expression patterns of this channel during cortical development have not yet been characterized. In the present study, we conducted an RNAscope experiment to visualize the location of Piezo1 transcripts with embryonic neuronal/glial lineage cell markers. Our analysis covered coronal sections of the mouse forebrain on embryonic day 12.5 (E12.5), E14.5, E16.5, and E18.5. In addition, applying Yoda1, a specific Piezo1 agonist, evoked distinct calcium elevation in piriform cortices of E16.5 and E18.5 embryonic slices. Furthermore, pharmacological activation or inhibition of this channel significantly modulated the migration of neurosphere-derived cells in vitro. These findings contribute valuable insights to the field of mechanobiology and provide an understanding of the intricate processes underlying embryonic brain development.


Assuntos
Canais Iônicos , Neurogênese , Animais , Camundongos , Canais Iônicos/genética , Canais Iônicos/metabolismo , Mecanotransdução Celular/fisiologia , Neurogênese/genética , Prosencéfalo/metabolismo
5.
PLoS One ; 19(1): e0296790, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38227598

RESUMO

SpinoCerebellar Ataxia type 7 (SCA7) is an inherited disorder caused by CAG triplet repeats encoding polyglutamine expansion in the ATXN7 protein, which is part of the transcriptional coactivator complex SAGA. The mutation primarily causes neurodegeneration in the cerebellum and retina, as well as several forebrain structures. The SCA7140Q/5Q knock-in mouse model recapitulates key disease features, including loss of vision and motor performance. To characterize the temporal progression of brain degeneration of this model, we performed a longitudinal study spanning from early to late symptomatic stages using high-resolution magnetic resonance imaging (MRI) and in vivo 1H-magnetic resonance spectroscopy (1H-MRS). Compared to wild-type mouse littermates, MRI analysis of SCA7 mice shows progressive atrophy of defined brain structures, with the striatum, thalamus and cortex being the first and most severely affected. The volume loss of these structures coincided with increased motor impairments in SCA7 mice, suggesting an alteration of the sensory-motor network, as observed in SCA7 patients. MRI also reveals atrophy of the hippocampus and anterior commissure at mid-symptomatic stage and the midbrain and brain stem at late stage. 1H-MRS of hippocampus, a brain region previously shown to be dysfunctional in patients, reveals early and progressive metabolic alterations in SCA7 mice. Interestingly, abnormal glutamine accumulation precedes the hippocampal atrophy and the reduction in myo-inositol and total N-acetyl-aspartate concentrations, two markers of glial and neuronal damage, respectively. Together, our results indicate that non-cerebellar alterations and glial and neuronal metabolic impairments may play a crucial role in the development of SCA7 mouse pathology, particularly at early stages of the disease. Degenerative features of forebrain structures in SCA7 mice correspond to current observations made in patients. Our study thus provides potential biomarkers that could be used for the evaluation of future therapeutic trials using the SCA7140Q/5Q model.


Assuntos
Ataxias Espinocerebelares , Humanos , Camundongos , Animais , Estudos Longitudinais , Ataxias Espinocerebelares/diagnóstico por imagem , Ataxias Espinocerebelares/genética , Ataxias Espinocerebelares/patologia , Ataxina-7/genética , Imageamento por Ressonância Magnética , Prosencéfalo/metabolismo , Espectroscopia de Ressonância Magnética , Atrofia/patologia
6.
Biochem Biophys Res Commun ; 681: 212-217, 2023 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-37783119

RESUMO

Fibroblast growth factors (Fgfs) play crucial roles in various developmental processes including brain development. We previously identified Fgf22 in zebrafish and found that fgf22 is involved in midbrain patterning during embryogenesis. Here, we investigated the role of Fgf22 in the formation of the zebrafish forebrain. We found that fgf22 was essential for determining the ventral properties of the telencephalon and diencephalon but not for cell proliferation. In addition, the knockdown of fgf22 inhibited the generation of glutamatergic neurons, γ-aminobutyric acid (GABA)ergic interneurons and astrocytes. Recently, Fgf signaling has received much attention because of its importance in the pathogenesis of multiple sclerosis, in which oligodendrocytes and myelin are destroyed. However, the effects of each Fgf on oligodendrocytes remain largely unknown. Therefore, we also investigated the role of Fgf22 in oligodendrocyte development and explored whether there is a difference between Fgf22 and other Fgfs. Knockdown of fgf22 promoted the generation of oligodendrocytes. Conversely, overexpression of fgf22 inhibited the generation of oligodendrocytes. Furthermore, the forebrain phenotypes of fgfr2b knockdown zebrafish were remarkably similar to those of fgf22 knockdown zebrafish. This establishes the Fgf22-Fgfr2b axis as a key ligand‒receptor partnership in neurogenesis and gliogenesis in the forebrain. Our results indicate that Fgf22 has a unique function in suppressing oligodendrocyte differentiation through Fgfr2b without affecting cell proliferation.


Assuntos
Receptor Tipo 2 de Fator de Crescimento de Fibroblastos , Peixe-Zebra , Animais , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Neurogênese/genética , Prosencéfalo/metabolismo , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Peixe-Zebra/genética
7.
eNeuro ; 10(10)2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37816595

RESUMO

Lysosomes are acidic organelles that traffic throughout neurons delivering catabolic enzymes to distal regions of the cell and maintaining degradative demands. Loss of function mutations in the gene GBA encoding the lysosomal enzyme glucocerebrosidase (GCase) cause the lysosomal storage disorder Gaucher's disease (GD) and are the most common genetic risk factor for synucleinopathies like Parkinson's disease (PD) and dementia with Lewy bodies (DLB). GCase degrades the membrane lipid glucosylceramide (GlcCer) and mutations in GBA, or inhibiting its activity, results in the accumulation of GlcCer and disturbs the composition of the lysosomal membrane. The lysosomal membrane serves as the platform to which intracellular trafficking complexes are recruited and activated. Here, we investigated whether lysosomal trafficking in axons was altered by inhibition of GCase with the pharmacological agent Conduritol B Epoxide (CBE). Using live cell imaging in human male induced pluripotent human stem cell (iPSC)-derived forebrain neurons, we demonstrated that lysosomal transport was similar in both control and CBE-treated neurons. Furthermore, we tested whether lysosomal rupture, a process implicated in various neurodegenerative disorders, was affected by inhibition of GCase. Using L-leucyl-L-leucine methyl ester (LLoME) to induce lysosomal membrane damage and immunocytochemical staining for markers of lysosomal rupture, we found no difference in susceptibility to rupture between control and CBE-treated neurons. These results suggest the loss of GCase activity does not contribute to neurodegenerative disease by disrupting either lysosomal transport or rupture.


Assuntos
Células-Tronco Pluripotentes Induzidas , Doenças Neurodegenerativas , Masculino , Humanos , Glucosilceramidase/genética , Glucosilceramidase/metabolismo , Doenças Neurodegenerativas/metabolismo , Transporte Axonal , Neurônios/metabolismo , Prosencéfalo/metabolismo , Lisossomos/metabolismo , alfa-Sinucleína/metabolismo
8.
Hum Mol Genet ; 32(23): 3276-3298, 2023 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-37688574

RESUMO

Cyclin-dependent kinase-like 5 (CDKL5) is a serine-threonine kinase enriched in the forebrain to regulate neuronal development and function. Patients with CDKL5 deficiency disorder (CDD), a severe neurodevelopmental condition caused by mutations of CDKL5 gene, present early-onset epilepsy as the most prominent feature. However, spontaneous seizures have not been reported in mouse models of CDD, raising vital questions on the human-mouse differences and the roles of CDKL5 in early postnatal brains. Here, we firstly measured electroencephalographic (EEG) activities via a wireless telemetry system coupled with video-recording in neonatal mice. We found that mice lacking CDKL5 exhibited spontaneous epileptic EEG discharges, accompanied with increased burst activities and ictal behaviors, specifically at postnatal day 12 (P12). Intriguingly, those epileptic spikes disappeared after P14. We next performed an unbiased transcriptome profiling in the dorsal hippocampus and motor cortex of Cdkl5 null mice at different developmental timepoints, uncovering a set of age-dependent and brain region-specific alterations of gene expression in parallel with the transient display of epileptic activities. Finally, we validated multiple differentially expressed genes, such as glycine receptor alpha 2 and cholecystokinin, at the transcript or protein levels, supporting the relevance of these genes to CDKL5-regulated excitability. Our findings reveal early-onset neuronal hyperexcitability in mouse model of CDD, providing new insights into CDD etiology and potential molecular targets to ameliorate intractable neonatal epilepsy.


Assuntos
Epilepsia Resistente a Medicamentos , Epilepsia , Espasmos Infantis , Humanos , Animais , Camundongos , Transcriptoma/genética , Espasmos Infantis/genética , Espasmos Infantis/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Epilepsia/genética , Prosencéfalo/metabolismo , Camundongos Knockout
9.
Nature ; 622(7982): 359-366, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37758944

RESUMO

The assembly of cortical circuits involves the generation and migration of interneurons from the ventral to the dorsal forebrain1-3, which has been challenging to study at inaccessible stages of late gestation and early postnatal human development4. Autism spectrum disorder and other neurodevelopmental disorders (NDDs) have been associated with abnormal cortical interneuron development5, but which of these NDD genes affect interneuron generation and migration, and how they mediate these effects remains unknown. We previously developed a platform to study interneuron development and migration in subpallial organoids and forebrain assembloids6. Here we integrate assembloids with CRISPR screening to investigate the involvement of 425 NDD genes in human interneuron development. The first screen aimed at interneuron generation revealed 13 candidate genes, including CSDE1 and SMAD4. We subsequently conducted an interneuron migration screen in more than 1,000 forebrain assembloids that identified 33 candidate genes, including cytoskeleton-related genes and the endoplasmic reticulum-related gene LNPK. We discovered that, during interneuron migration, the endoplasmic reticulum is displaced along the leading neuronal branch before nuclear translocation. LNPK deletion interfered with this endoplasmic reticulum displacement and resulted in abnormal migration. These results highlight the power of this CRISPR-assembloid platform to systematically map NDD genes onto human development and reveal disease mechanisms.


Assuntos
Sistemas CRISPR-Cas , Edição de Genes , Transtornos do Neurodesenvolvimento , Feminino , Humanos , Recém-Nascido , Gravidez , Movimento Celular/genética , Sistemas CRISPR-Cas/genética , Interneurônios/citologia , Interneurônios/metabolismo , Interneurônios/patologia , Transtornos do Neurodesenvolvimento/genética , Transtornos do Neurodesenvolvimento/patologia , Organoides/citologia , Organoides/embriologia , Organoides/crescimento & desenvolvimento , Organoides/metabolismo , Organoides/patologia , Retículo Endoplasmático/metabolismo , Prosencéfalo/citologia , Prosencéfalo/embriologia , Prosencéfalo/crescimento & desenvolvimento , Prosencéfalo/metabolismo , Prosencéfalo/patologia , Transporte Ativo do Núcleo Celular
10.
J Neurosci ; 43(41): 6854-6871, 2023 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-37640551

RESUMO

Neural progenitor cells in the developing dorsal forebrain generate excitatory neurons followed by oligodendrocytes (OLs) and astrocytes. However, the specific mechanisms that regulate the timing of this neuron-glia switch are not fully understood. In this study, we show that the proper balance of Notch signaling in dorsal forebrain progenitors is required to generate oligodendrocytes during late stages of embryonic development. Using ex vivo and in utero approaches in mouse embryos of both sexes, we found that Notch inhibition reduced the number of oligodendrocyte lineage cells in the dorsal pallium. However, Notch overactivation also prevented oligodendrogenesis and maintained a progenitor state. These results point toward a dual role for Notch signaling in both promoting and inhibiting oligodendrogenesis, which must be fine-tuned to generate oligodendrocyte lineage cells at the right time and in the right numbers. We further identified the canonical Notch downstream factors HES1 and HES5 as negative regulators in this process. CRISPR (clustered regularly interspaced short palindromic repeat)/Cas9-mediated knockdown of Hes1 and Hes5 caused increased expression of the pro-oligodendrocyte factor ASCL1 and led to precocious oligodendrogenesis. Conversely, combining Notch overactivation with ASCL1 overexpression robustly promoted oligodendrogenesis, indicating a separate mechanism of Notch that operates synergistically with ASCL1 to specify an oligodendrocyte fate. We propose a model in which Notch signaling works together with ASCL1 to specify progenitors toward the oligodendrocyte lineage but also maintains a progenitor state through Hes-dependent repression of Ascl1 so that oligodendrocytes are not made too early, thus contributing to the precise timing of the neuron-glia switch.SIGNIFICANCE STATEMENT Neural progenitors make oligodendrocytes after neurogenesis starts to wind down, but the mechanisms that control the timing of this switch are poorly understood. In this study, we identify Notch signaling as a critical pathway that regulates the balance between progenitor maintenance and oligodendrogenesis. Notch signaling is required for the oligodendrocyte fate, but elevated Notch signaling prevents oligodendrogenesis and maintains a progenitor state. We provide evidence that these opposing functions are controlled by different mechanisms. Before the switch, Notch signaling through Hes factors represses oligodendrogenesis. Later, Notch signaling through an unknown mechanism promotes oligodendrogenesis synergistically with the transcription factor ASCL1. Our study underscores the complexity of Notch and reveals its importance in regulating the timing and numbers of oligodendrocyte production.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos , Neurônios , Masculino , Feminino , Camundongos , Animais , Diferenciação Celular/fisiologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Neurônios/metabolismo , Prosencéfalo/metabolismo , Oligodendroglia/metabolismo , Receptores Notch/metabolismo
11.
Int J Mol Sci ; 24(15)2023 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-37569457

RESUMO

Forebrain ischemia-reperfusion (IR) injury causes neurological impairments due to decreased cerebral autoregulation, hypoperfusion, and edema in the hours to days following the restoration of spontaneous circulation. This study aimed to examine the protective and/or therapeutic effects of cerebrolysin (CBL) in managing forebrain IR injury and any probable underlying mechanisms. To study the contribution of reperfusion to forebrain injury, we developed a transient dual carotid artery ligation (tDCAL/IR) mouse model. Five equal groups of six BLC57 mice were created: Group 1: control group (no surgery was performed); Group 2: sham surgery (surgery was performed without IR); Group 3: tDCAL/IR (surgery with IR via permanently ligating the left CA and temporarily closing the right CA for 30 min, followed by reperfusion for 72 h); Group 4: CBL + tDCAL/IR (CBL was given intravenously at a 60 mg/kg BW dose 30 min before IR); and Group 5: tDCAL/IR + CBL (CBL was administered i.v. at 60 mg/kg BW three hours after IR). At 72 h following IR, the mice were euthanized. CBL administration 3 h after IR improved neurological functional recovery, enhanced anti-inflammatory and antioxidant activities, alleviated apoptotic neuronal death, and inhibited reactive microglial and astrocyte activation, resulting in neuroprotection after IR injury in the tDCAL/IR + CBL mice group as compared to the other groups. Furthermore, CBL reduced the TLRs/NF-kB/cytokines while activating the Keap1/Nrf2/antioxidant signaling pathway. These results indicate that CBL may improve neurologic function in mice following IR.


Assuntos
Antioxidantes , Traumatismo por Reperfusão , Camundongos , Animais , Antioxidantes/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Transdução de Sinais , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/metabolismo , Modelos Animais de Doenças , Prosencéfalo/metabolismo , Estresse Oxidativo
12.
Nat Neurosci ; 26(9): 1505-1515, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37563294

RESUMO

Idiopathic autism spectrum disorder (ASD) is highly heterogeneous, and it remains unclear how convergent biological processes in affected individuals may give rise to symptoms. Here, using cortical organoids and single-cell transcriptomics, we modeled alterations in the forebrain development between boys with idiopathic ASD and their unaffected fathers in 13 families. Transcriptomic changes suggest that ASD pathogenesis in macrocephalic and normocephalic probands involves an opposite disruption of the balance between excitatory neurons of the dorsal cortical plate and other lineages such as early-generated neurons from the putative preplate. The imbalance stemmed from divergent expression of transcription factors driving cell fate during early cortical development. While we did not find genomic variants in probands that explained the observed transcriptomic alterations, a significant overlap between altered transcripts and reported ASD risk genes affected by rare variants suggests a degree of gene convergence between rare forms of ASD and the developmental transcriptome in idiopathic ASD.


Assuntos
Transtorno do Espectro Autista , Transtorno Autístico , Masculino , Humanos , Transtorno Autístico/genética , Transtorno do Espectro Autista/patologia , Neurônios/metabolismo , Neurogênese , Prosencéfalo/metabolismo , Organoides/metabolismo
13.
Front Endocrinol (Lausanne) ; 14: 1212104, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37396176

RESUMO

AXL is a receptor tyrosine kinase commonly associated with a variety of human cancers. Along with its ligand Gas6 (growth arrest-specific protein 6), AXL is emerging as an important regulator of neuroendocrine development and function. AXL signaling in response to Gas6 binding impacts neuroendocrine structure and function at the level of the brain, pituitary, and gonads. During development, AXL has been identified as an upstream inhibitor of gonadotropin receptor hormone (GnRH) production and also plays a key role in the migration of GnRH neurons from the olfactory placode to the forebrain. AXL is implicated in reproductive diseases including some forms of idiopathic hypogonadotropic hypogonadism and evidence suggests that AXL is required for normal spermatogenesis. Here, we highlight research describing AXL/Gas6 signaling mechanisms with a focus on the molecular pathways related to neuroendocrine function in health and disease. In doing so, we aim to present a concise account of known AXL/Gas6 signaling mechanisms to identify current knowledge gaps and inspire future research.


Assuntos
Receptor Tirosina Quinase Axl , Proteínas Proto-Oncogênicas , Humanos , Masculino , Hormônio Liberador de Gonadotropina/metabolismo , Eixo Hipotalâmico-Hipofisário-Gonadal , Prosencéfalo/metabolismo , Proteínas Proto-Oncogênicas/metabolismo
14.
Proc Natl Acad Sci U S A ; 120(28): e2218830120, 2023 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-37399414

RESUMO

The cholinergic system of the basal forebrain plays an integral part in behaviors ranging from attention to learning, partly by altering the impact of noise in neural populations. The circuit computations underlying cholinergic actions are confounded by recent findings that forebrain cholinergic neurons corelease both acetylcholine (ACh) and GABA. We have identified that corelease of ACh and GABA by cholinergic inputs to the claustrum, a structure implicated in the control of attention, has opposing effects on the electrical activity of claustrum neurons that project to cortical vs. subcortical targets. These actions differentially alter neuronal gain and dynamic range in the two types of neurons. In model networks, the differential effects of ACh and GABA toggle network efficiency and the impact of noise on population dynamics between two different projection subcircuits. Such cholinergic switching between subcircuits provides a potential logic for neurotransmitter corelease in implementing behaviorally relevant computations.


Assuntos
Acetilcolina , Colinérgicos , Acetilcolina/metabolismo , Prosencéfalo/metabolismo , Neurônios Colinérgicos/metabolismo , Ácido gama-Aminobutírico/metabolismo , Lógica
15.
Int J Mol Sci ; 24(12)2023 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-37373050

RESUMO

Neuroinflammation is one of the postulated mechanisms for Pb neurotoxicity. However, the exact molecular mechanisms responsible for its pro-inflammatory effect are not fully elucidated. In this study, we examined the role of glial cells in neuroinflammation induced by Pb exposure. We investigated how microglia, a type of glial cell, responded to the changes caused by perinatal exposure to Pb by measuring the expression of Iba1 at the mRNA and protein levels. To assess the state of microglia, we analyzed the mRNA levels of specific markers associated with the cytotoxic M1 phenotype (Il1b, Il6, and Tnfa) and the cytoprotective M2 phenotype (Arg1, Chi3l1, Mrc1, Fcgr1a, Sphk1, and Tgfb1). Additionally, we measured the concentration of pro-inflammatory cytokines (IL-1ß, IL-6, and TNF-α). To assess the reactivity and functionality status of astrocytes, we analyzed the GFAP (mRNA expression and protein concentration) as well as glutamine synthase (GS) protein level and activity. Using an electron microscope, we assessed ultrastructural abnormalities in the examined brain structures (forebrain cortex, cerebellum, and hippocampus). In addition, we measured the mRNA levels of Cxcl1 and Cxcl2, and their receptor, Cxcr2. Our data showed that perinatal exposure to Pb at low doses affected both microglia and astrocyte cells' status (their mobilization, activation, function, and changes in gene expression profile) in a brain-structure-specific manner. The results suggest that both microglia and astrocytes represent a potential target for Pb neurotoxicity, thus being key mediators of neuroinflammation and further neuropathology evoked by Pb poisoning during perinatal brain development.


Assuntos
Astrócitos , Microglia , Gravidez , Feminino , Humanos , Astrócitos/metabolismo , Microglia/metabolismo , Chumbo/metabolismo , Doenças Neuroinflamatórias , Citocinas/metabolismo , Prosencéfalo/metabolismo , RNA Mensageiro/metabolismo
16.
Neurobiol Aging ; 129: 15-27, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37257405

RESUMO

Brain-derived estrogen is an endogenous neuroprotective agent, whether and how might this protective function with aging, especially postmenopausal drops in circulating estrogen, remain unclear. We herein subjected 6, 14, and 18 Mon female rats to mimic natural aging, and found that estrogen synthesis is more active in the healthy aged brain, as evidenced by the highest levels of mRNA and protein expression of aromatase, the key enzyme of E2 biosynthesis, among the three groups. Aromatase knockout in forebrain neurons (FBN-Aro-/-) impaired hippocampal and cortical neurons, and cognitive function in 18 Mon rats, compared to wild-type controls. Furthermore, estrogen nuclear receptors (ERα/ß) displayed opposite changes, with a significant ERα decrease and ERß increase, while membrane receptor GPR30 expressed stably in hippocampus during aging. Intriguingly, GPR30, but not ERα and ERß, was decreased by FBN-Aro-/-. The results indicate that GPR30 is more sensitive to brain local E2 synthesis. Our findings provide evidence of a critical role for brain-derived estrogen in maintaining healthy brain function in older individuals, possibly involving GPR30.


Assuntos
Aromatase , Cognição , Estrogênios , Animais , Feminino , Ratos , Aromatase/metabolismo , Estradiol/farmacologia , Estradiol/metabolismo , Receptor alfa de Estrogênio/genética , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Prosencéfalo/metabolismo , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
17.
Proc Natl Acad Sci U S A ; 120(23): e2300722120, 2023 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-37252963

RESUMO

Mental health disorders often arise as a combination of environmental and genetic factors. The FKBP5 gene, encoding the GR co-chaperone FKBP51, has been uncovered as a key genetic risk factor for stress-related illness. However, the exact cell type and region-specific mechanisms by which FKBP51 contributes to stress resilience or susceptibility processes remain to be unravelled. FKBP51 functionality is known to interact with the environmental risk factors age and sex, but so far data on behavioral, structural, and molecular consequences of these interactions are still largely unknown. Here we report the cell type- and sex-specific contribution of FKBP51 to stress susceptibility and resilience mechanisms under the high-risk environmental conditions of an older age, by using two conditional knockout models within glutamatergic (Fkbp5Nex) and GABAergic (Fkbp5Dlx) neurons of the forebrain. Specific manipulation of Fkbp51 in these two cell types led to opposing effects on behavior, brain structure and gene expression profiles in a highly sex-dependent fashion. The results emphasize the role of FKBP51 as a key player in stress-related illness and the need for more targeted and sex-specific treatment strategies.


Assuntos
Transtornos Mentais , Masculino , Feminino , Humanos , Transtornos Mentais/genética , Neurônios GABAérgicos/metabolismo , Prosencéfalo/metabolismo , Proteínas de Ligação a Tacrolimo/genética , Proteínas de Ligação a Tacrolimo/metabolismo
18.
J Comp Neurol ; 531(11): 1126-1146, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37071579

RESUMO

Islet-1 (Isl1) is one of the most conserved transcription factors in the evolution of vertebrates, due to its continuing involvement in such important functions as the differentiation of motoneurons, among other essential roles in cell fate in the forebrain. Although its functions are thought to be similar in all vertebrates, the knowledge about the conservation of its expression pattern in the central nervous system goes as far as teleosts, leaving the basal groups of actinopterygian fishes overlooked, despite their important phylogenetic position. In order to assess the extent of its conservation among vertebrates, we studied its expression pattern in the central nervous system of selected nonteleost actinopterygian fishes. By means of immunohistochemical techniques, we analyzed the Isl1 expression in the brain, spinal cord, and sensory ganglia of the cranial nerves of young adult specimens of the cladistian species Polypterus senegalus and Erpetoichthys calabaricus, the chondrostean Acipenser ruthenus, and the holostean Lepisosteus oculatus. We also detected the presence of the transcription factor Orthopedia and the enzymes tyrosine hydroxylase (TH) and choline acetyltransferase (ChAT) to better locate all the immunoreactive structures in the different brain areas and to reveal the possible coexpression with Isl1. Numerous conserved features in the expression pattern of Isl1 were observed in these groups of fishes, such as populations of cells in the subpallial nuclei, preoptic area, subparaventricular and tuberal hypothalamic regions, prethalamus, epiphysis, cranial motor nuclei and sensory ganglia of the cranial nerves, and the ventral horn of the spinal cord. Double labeling of TH and Isl1 was observed in cells of the preoptic area, the subparaventricular and tuberal hypothalamic regions, and the prethalamus, while virtually all motoneurons in the hindbrain and the spinal cord coexpressed ChAT and Isl1. Altogether, these results show the high degree of conservation of the expression pattern of the transcription factor Isl1, not only among fish, but in the subsequent evolution of vertebrates.


Assuntos
Encéfalo , Sistema Nervoso Central , Animais , Filogenia , Sistema Nervoso Central/metabolismo , Encéfalo/metabolismo , Peixes/metabolismo , Colina O-Acetiltransferase/metabolismo , Prosencéfalo/metabolismo , Colinérgicos/metabolismo , Fatores de Transcrição/metabolismo
19.
Neurobiol Aging ; 125: 98-108, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36889122

RESUMO

Hyperphosphorylated tau in the locus coeruleus (LC) is ubiquitous in prodromal Alzheimer's disease (AD), and LC neurons degenerate as AD progresses. Hyperphosphorylated tau alters firing rates in other brain regions, but its effects on LC neurons are unknown. We assessed single unit LC activity in anesthetized wild-type (WT) and TgF344-AD rats at 6 months, which represents a prodromal stage when LC neurons are the only cells containing hyperphosphorylated tau in TgF344-AD animals, and at 15 months when amyloid-ß (Aß) and tau pathology are both abundant in the forebrain. At baseline, LC neurons from TgF344-AD rats were hypoactive at both ages compared to WT littermates but showed elevated spontaneous bursting properties. Differences in footshock-evoked LC firing depended on age, with 6-month TgF344-AD rats demonstrating aspects of hyperactivity, and 15-month transgenic rats showing hypoactivity. Early LC hyperactivity is consistent with appearance of prodromal neuropsychiatric symptoms and is followed by LC hypoactivity which contributes to cognitive impairment. These results support further investigation into disease stage-dependent noradrenergic interventions for AD.


Assuntos
Doença de Alzheimer , Ratos , Animais , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Locus Cerúleo/patologia , Ratos Transgênicos , Peptídeos beta-Amiloides , Prosencéfalo/metabolismo , Modelos Animais de Doenças , Proteínas tau/metabolismo
20.
J Comp Neurol ; 531(7): 702-719, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36855269

RESUMO

The hippocampus (HP) receives neurochemically diverse inputs from the raphe nuclei, including glutamatergic axons characterized by the expression of the vesicular glutamate transporter type 3 (VGLUT3). These raphe-HP VGLUT3 projections have been suggested to play a critical role in HP functions, yet a complete anatomical overview of raphe VGLUT3 projections to the forebrain, and in particular to the HP, is lacking. Using anterograde viral tracing, we describe largely nonoverlapping VGLUT3-positive projections from the dorsal raphe (DR) and median raphe (MnR) to the forebrain, with the HP receiving inputs from the MnR. A limited subset of forebrain regions such as the amygdaloid complex, claustrum, and hypothalamus receives projections from both the DR and MnR that remain largely segregated. This highly complementary anatomical pattern suggests contrasting roles for DR and MnR VGLUT3 neurons. To further analyze the topography of VGLUT3 raphe projections to the HP, we used retrograde tracing and found that HP-projecting VGLUT3-positive neurons (VGLUT3HP ) distribute over several raphe subregions (including the MnR, paramedian raphe, and B9 cell group) and lack co-expression of serotonergic markers. Strikingly, double retrograde tracing experiments unraveled two parallel streams of VGLUT3-positive projections targeting the dorsal and ventral poles of the HP. These results demonstrate highly organized and segregated VGLUT3-positive projections to the HP, suggesting independent modulation of HP functions such as spatial memory and emotion-related behavior.


Assuntos
Axônios , Neurônios , Axônios/metabolismo , Núcleo Dorsal da Rafe , Hipocampo , Neurônios/metabolismo , Prosencéfalo/metabolismo , Proteínas Vesiculares de Transporte de Glutamato/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...