Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 196
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
EBioMedicine ; 65: 103272, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33691246

RESUMO

BACKGROUND: Despite the fundamental clinical significance of general anaesthesia, the cortical mechanism underlying anaesthetic-induced loss of consciousness (aLOC) remains elusive. METHODS: Here, we measured the dynamics of two major cortical neurotransmitters, gamma-aminobutyric acid (GABA) and glutamate, through in vivo two-photon imaging and genetically encoded neurotransmitter sensors in a cell type-specific manner in the primary visual (V1) cortex. FINDINGS: We found a general decrease in cortical GABA transmission during aLOC. However, the glutamate transmission varies among different cortical cell types, where in it is almost preserved on pyramidal cells and is significantly reduced on inhibitory interneurons. Cortical interneurons expressing vasoactive intestinal peptide (VIP) and parvalbumin (PV) specialize in disinhibitory and inhibitory effects, respectively. During aLOC, VIP neuronal activity was delayed, and PV neuronal activity was dramatically inhibited and highly synchronized. INTERPRETATION: These data reveal that aLOC resembles a cortical state with a disrupted excitatory-inhibitory network and suggest that a functional inhibitory network is indispensable in the maintenance of consciousness. FUNDING: This work was supported by the grants of the National Natural Science Foundation of China (grant nos. 81620108012 and 82030038 to H.D. and grant nos. 31922029, 61890951, and 61890950 to J.H.).


Assuntos
Anestésicos Inalatórios/farmacologia , Ácido Glutâmico/metabolismo , Isoflurano/farmacologia , Transmissão Sináptica/efeitos dos fármacos , Ácido gama-Aminobutírico/metabolismo , Animais , Cálcio/metabolismo , Eletroencefalografia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência por Excitação Multifotônica , Parvalbuminas/metabolismo , Células Piramidais/citologia , Células Piramidais/metabolismo , Inconsciência , Peptídeo Intestinal Vasoativo/genética , Peptídeo Intestinal Vasoativo/metabolismo
2.
J Nutr ; 151(1): 235-244, 2021 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-33245133

RESUMO

BACKGROUND: Both iron deficiency and overload may adversely affect neurodevelopment. OBJECTIVES: The study assessed how changes in early-life iron status affect iron homeostasis and cytoarchitecture of hippocampal neurons in a piglet model. METHODS: On postnatal day (PD) 1, 30 Hampshire × Yorkshire crossbreed piglets (n = 15/sex) were stratified by sex and litter and randomly assigned to experimental groups receiving low (L-Fe), adequate (A-Fe), or high (H-Fe) levels of iron supplement during the pre- (PD1-21) and postweaning periods (PD22-35). Pigs in the L-Fe, A-Fe, and H-Fe groups orally received 0, 1, and 30 mg Fe · kg weight-1 · d-1 preweaning and were fed a diet containing 30, 125, and 1000 mg Fe/kg postweaning, respectively. Heme indexes were analyzed weekly, and gene and protein expressions of iron regulatory proteins in duodenal mucosa, liver, and hippocampus were analyzed through qRT-PCR and western blot, respectively, on PD35. Hippocampal neurons stained using the Golgi-Cox method were traced and their dendritic arbors reconstructed in 3-D using Neurolucida. Dendritic complexity was quantified using Sholl and branch order analyses. RESULTS: Pigs in the L-Fe group developed iron deficiency anemia (hemoglobin = 8.2 g/dL, hematocrit = 20.1%) on PD35 and became stunted during week 5 with lower final body weight than H-Fe group pigs (6.6 compared with 9.6 kg, P < 0.05). In comparison with A-Fe, H-Fe increased hippocampal ferritin expression by 38% and L-Fe decreased its expression by 52% (P < 0.05), suggesting altered hippocampal iron stores. Pigs in the H-Fe group had greater dendritic complexity in CA1/3 pyramidal neurons than L-Fe group pigs as shown by more dendritic intersections with Sholl rings (P ≤ 0.04) and a greater number of dendrites (P ≤ 0.016). CONCLUSIONS: In piglets, the developing hippocampus is susceptible to perturbations by dietary iron, with deficiency and overload differentially affecting dendritic arborization.


Assuntos
Anemia Ferropriva , Dendritos , Hipocampo , Ferro da Dieta , Células Piramidais , Suínos , Animais , Feminino , Masculino , Anemia Ferropriva/veterinária , Dendritos/fisiologia , Relação Dose-Resposta a Droga , Duodeno , Regulação da Expressão Gênica/efeitos dos fármacos , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Ferro da Dieta/administração & dosagem , Células Piramidais/citologia , Células Piramidais/efeitos dos fármacos
3.
Sci Rep ; 10(1): 10857, 2020 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-32616804

RESUMO

PHRF1 is involved in transforming growth factor ß (TGF-ß) signaling to constrain the formation of acute promyelocytic leukemia (APL) in mouse APL models. PHRF1 also participates in modulating non-homologous end-joining. However, the role of PHRF1 in mammalian dendrite architecture and synaptic plasticity is unclear. Here, we investigated the role of PHRF1 in dendritic formation in the murine hippocampus using Camk2a promoter driven-iCre recombinase to conduct a PHRF1 conditional knockout, namely PHRF1Δ/Δ, in the forebrain region. PHRF1Δ/Δ mice developed normally, but exhibited anxiety-like behaviors and displayed defective spatial memory. Alterations of dendritic complexity in apical and basal dendrites of pyramidal neurons were noticed in PHRF1Δ/Δ mutants. Furthermore, electrical stimulation in the hippocampal CA1 region after the TGF-ß1 treatment showed a reduced synaptic plasticity in PHRF1Δ/Δ mice. Immunoblotting analysis indicated that PHRF1 ablation affected the TGF-ß signaling. Collectively, our results demonstrate that PHRF1 is important for the dendritic architecture and required for spatial memory formation in the hippocampus.


Assuntos
Dendritos/química , Hipocampo/metabolismo , Proteínas de Membrana/fisiologia , Células Piramidais/metabolismo , Domínios RING Finger/fisiologia , Memória Espacial/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Dendritos/fisiologia , Feminino , Masculino , Camundongos , Camundongos Knockout , Plasticidade Neuronal , Células Piramidais/citologia , Transdução de Sinais , Fator de Crescimento Transformador beta/genética
4.
Int J Mol Sci ; 21(9)2020 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-32349283

RESUMO

The brain-derived neurotrophic factor (BDNF) plays crucial roles in both the developing and mature brain. Moreover, alterations in BDNF levels are correlated with the cognitive impairment observed in several neurological diseases. Among the different therapeutic strategies developed to improve endogenous BDNF levels is the administration of the BDNF-inducing drug Fingolimod, an agonist of the sphingosine-1-phosphate receptor. Fingolimod treatment was shown to rescue diverse symptoms associated with several neurological conditions (i.e., Alzheimer disease, Rett syndrome). However, the cellular mechanisms through which Fingolimod mediates its BDNF-dependent therapeutic effects remain unclear. We show that Fingolimod regulates the dendritic architecture, dendritic spine density and morphology of healthy mature primary hippocampal neurons. Moreover, the application of Fingolimod upregulates the expression of activity-related proteins c-Fos and pERK1/2 in these cells. Importantly, we show that BDNF release is required for these actions of Fingolimod. As alterations in neuronal structure underlie cognitive impairment, we tested whether Fingolimod application might prevent the abnormalities in neuronal structure typical of two neurodevelopmental disorders, namely Rett syndrome and Cdk5 deficiency disorder. We found a significant rescue in the neurite architecture of developing cortical neurons from Mecp2 and Cdkl5 mutant mice. Our study provides insights into understanding the BDNF-dependent therapeutic actions of Fingolimod.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Espinhas Dendríticas/metabolismo , Cloridrato de Fingolimode/farmacologia , Células Piramidais/efeitos dos fármacos , Células Piramidais/metabolismo , Animais , Biomarcadores , Imunofluorescência , Expressão Gênica , Regulação da Expressão Gênica , Genes fos , Imunossupressores/farmacologia , Camundongos , Células Piramidais/citologia , Síndrome de Rett/etiologia , Síndrome de Rett/metabolismo
5.
Cell Rep ; 30(13): 4490-4504.e4, 2020 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-32234482

RESUMO

Neural stem cells (NSCs) in the prenatal neocortex progressively generate different subtypes of glutamatergic projection neurons. Following that, NSCs have a major switch in their progenitor properties and produce γ-aminobutyric acid (GABAergic) interneurons for the olfactory bulb (OB), cortical oligodendrocytes, and astrocytes. Herein, we provide evidence for the molecular mechanism that underlies this switch in the state of neocortical NSCs. We show that, at around E16.5, mouse neocortical NSCs start to generate GSX2-expressing (GSX2+) intermediate progenitor cells (IPCs). In vivo lineage-tracing study revealed that GSX2+ IPC population gives rise not only to OB interneurons but also to cortical oligodendrocytes and astrocytes, suggesting that they are a tri-potential population. We demonstrated that Sonic hedgehog signaling is both necessary and sufficient for the generation of GSX2+ IPCs by reducing GLI3R protein levels. Using single-cell RNA sequencing, we identify the transcriptional profile of GSX2+ IPCs and the process of the lineage switch of cortical NSCs.


Assuntos
Linhagem da Célula , Proteínas Hedgehog/metabolismo , Neocórtex/citologia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Transdução de Sinais , Animais , Astrócitos/metabolismo , Biomarcadores/metabolismo , Embrião de Mamíferos/metabolismo , Proteínas de Homeodomínio/metabolismo , Interneurônios/citologia , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/metabolismo , Neurogênese , Neuroglia/citologia , Neuroglia/metabolismo , Bulbo Olfatório/citologia , Oligodendroglia/metabolismo , Células Piramidais/citologia , Células Piramidais/metabolismo , Reprodutibilidade dos Testes , Proteína Gli3 com Dedos de Zinco/metabolismo
6.
Dev Biol ; 461(1): 86-95, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-31982375

RESUMO

One of the main obstacles for studying the molecular and cellular mechanisms underlying human neurodevelopment in vivo is the scarcity of experimental models. The discovery that neurons can be generated from human induced pluripotent stem cells (hiPSCs) paves the way for novel approaches that are stem cell-based. Here, we developed a technique to follow the development of transplanted hiPSC-derived neuronal precursors in the cortex of mice over time. Using post-mortem immunohistochemistry we quantified the differentiation and maturation of dendritic patterns of the human neurons over a total of six months. In addition, entirely hiPSC-derived neuronal parenchyma was followed over eight months using two-photon in vivo imaging through a cranial window. We found that transplanted hiPSC-derived neuronal precursors exhibit a "protracted" human developmental programme in different cortical areas. This offers novel possibilities for the sequential in vivo study of human cortical development and its alteration, followed in "real time".


Assuntos
Células-Tronco Pluripotentes Induzidas/transplante , Córtex Motor/embriologia , Neurogênese/fisiologia , Células Piramidais/transplante , Animais , Encéfalo/embriologia , Células Cultivadas , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Córtex Motor/citologia , Células Piramidais/citologia , Transplante Heterólogo
7.
Behav Brain Res ; 379: 112355, 2020 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-31730784

RESUMO

Ovarian steroids modulate the neuronal structure and function during the estrous cycle, contrasting peak effects during the proestrus cycle and low effects during the metestrus cycle. An ovariectomy (OVX) decreases gonadal hormones and tests the effects of substitutive therapies. We studied female rats with a normal estrous cycle and we also studied the effects of systemic progesterone (P4, 4.0 mg/kg) or its reduced metabolite allopregnanolone (ALLO, 4.0 mg/kg, both for 10 days) in females who had had an OVX 16.5 weeks prior to the study (long-term OVX) with the novel object recognition test (NORT) for associative memory. The dendritic shape and spine density in Golgi-impregnated basal dendrites (stratum oriens) of hippocampal pyramidal neurons was also studied. Proestrus females had a better performance than metestrus or OVX females in short-term memory (tested 1 h after the acquisition phase). Proestrus and metestrus females showed better results than OVX females for long-term memory (24 h after the initial phase). Both P4 and ALLO recovered the cognitive impairment induced by long-term OVX. Also, proestrus females had a higher density of dendritic spines than metestrus females, OVX reduced the density of spines when compared to intact females, whereas both P4 and ALLO treatments increased the dendritic spine density, number of dendritic branches along the dendritic length, and branching order compared to vehicle. These data add the dendrites of the stratum oriens as an additional site for naturally occurring changes in spine density during the estrous cycle and evidence the actions of progestins in both behavioral recovery and the structural dendritic rearrangement of hippocampal pyramidal neurons in long-term OVX female rats.


Assuntos
Região CA1 Hipocampal , Região CA2 Hipocampal , Disfunção Cognitiva , Espinhas Dendríticas , Ciclo Estral/metabolismo , Aprendizagem , Ovariectomia/efeitos adversos , Pregnanolona/metabolismo , Pregnanolona/farmacologia , Progesterona/metabolismo , Progesterona/farmacologia , Células Piramidais , Animais , Aprendizagem por Associação/efeitos dos fármacos , Aprendizagem por Associação/fisiologia , Comportamento Animal/fisiologia , Região CA1 Hipocampal/citologia , Região CA1 Hipocampal/efeitos dos fármacos , Região CA1 Hipocampal/metabolismo , Região CA2 Hipocampal/citologia , Região CA2 Hipocampal/efeitos dos fármacos , Região CA2 Hipocampal/metabolismo , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/metabolismo , Espinhas Dendríticas/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Aprendizagem/efeitos dos fármacos , Aprendizagem/fisiologia , Memória de Longo Prazo/efeitos dos fármacos , Memória de Longo Prazo/fisiologia , Memória de Curto Prazo/efeitos dos fármacos , Memória de Curto Prazo/fisiologia , Pregnanolona/administração & dosagem , Progesterona/administração & dosagem , Células Piramidais/citologia , Células Piramidais/efeitos dos fármacos , Ratos Wistar , Reconhecimento Psicológico/fisiologia
8.
Nat Methods ; 17(1): 107-113, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31686040

RESUMO

Spontaneous and sensory-evoked activity propagates across varying spatial scales in the mammalian cortex, but technical challenges have limited conceptual links between the function of local neuronal circuits and brain-wide network dynamics. We present a method for simultaneous cellular-resolution two-photon calcium imaging of a local microcircuit and mesoscopic widefield calcium imaging of the entire cortical mantle in awake mice. Our multi-scale approach involves a microscope with an orthogonal axis design where the mesoscopic objective is oriented above the brain and the two-photon objective is oriented horizontally, with imaging performed through a microprism. We also introduce a viral transduction method for robust and widespread gene delivery in the mouse brain. These approaches allow us to identify the behavioral state-dependent functional connectivity of pyramidal neurons and vasoactive intestinal peptide-expressing interneurons with long-range cortical networks. Our imaging system provides a powerful strategy for investigating cortical architecture across a wide range of spatial scales.


Assuntos
Encéfalo/fisiologia , Cálcio/metabolismo , Córtex Cerebral/fisiologia , Rede Nervosa/fisiologia , Neuroimagem/métodos , Neurônios/fisiologia , Fótons , Animais , Comportamento Animal , Encéfalo/citologia , Córtex Cerebral/citologia , Interneurônios/citologia , Interneurônios/fisiologia , Camundongos , Neurônios/citologia , Células Piramidais/citologia , Células Piramidais/fisiologia , Peptídeo Intestinal Vasoativo/metabolismo
9.
Neuron ; 104(5): 972-986.e6, 2019 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-31761708

RESUMO

How neural circuits develop in the human brain has remained almost impossible to study at the neuronal level. Here, we investigate human cortical neuron development, plasticity, and function using a mouse/human chimera model in which xenotransplanted human cortical pyramidal neurons integrate as single cells into the mouse cortex. Combined neuronal tracing, electrophysiology, and in vivo structural and functional imaging of the transplanted cells reveal a coordinated developmental roadmap recapitulating key milestones of human cortical neuron development. The human neurons display a prolonged developmental timeline, indicating the neuron-intrinsic retention of juvenile properties as an important component of human brain neoteny. Following maturation, human neurons in the visual cortex display tuned, decorrelated responses to visual stimuli, like mouse neurons, demonstrating their capacity for physiological synaptic integration in host cortical circuits. These findings provide new insights into human neuronal development and open novel avenues for the study of human neuronal function and disease. VIDEO ABSTRACT.


Assuntos
Neurogênese/fisiologia , Células Piramidais/citologia , Células Piramidais/fisiologia , Células Piramidais/transplante , Animais , Diferenciação Celular/fisiologia , Xenoenxertos , Humanos , Camundongos , Córtex Visual/citologia , Córtex Visual/fisiologia
10.
Neuron ; 103(6): 1086-1095.e5, 2019 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-31488328

RESUMO

Astrocytes are particularly promising candidates for reprogramming into neurons, as they maintain some of the original patterning information from their radial glial ancestors. However, to which extent the position of astrocytes influences the fate of reprogrammed neurons remains unknown. To elucidate this, we performed stab wound injury covering an entire neocortical column, including the gray matter (GM) and white matter (WM), and targeted local reactive astrocytes via injecting FLEx switch (Cre-On) adeno-associated viral (AAV) vectors into mGFAP-Cre mice. Single proneural factors were not sufficient for adequate reprogramming, although their combination with the nuclear receptor-related 1 protein (Nurr1) improved reprogramming efficiency. Nurr1 and Neurogenin 2 (Ngn2) resulted in high-efficiency reprogramming of targeted astrocytes into neurons that develop lamina-specific hallmarks, including the appropriate long-distance axonal projections. Surprisingly, in the WM, we did not observe any reprogrammed neurons, thereby unveiling a crucial role of region- and layer-specific differences in astrocyte reprogramming.


Assuntos
Astrócitos/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Técnicas de Reprogramação Celular/métodos , Córtex Cerebral/citologia , Córtex Cerebral/lesões , Proteínas do Tecido Nervoso/genética , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Células Piramidais/metabolismo , Animais , Astrócitos/citologia , Lesões Encefálicas Traumáticas , Dependovirus , Vetores Genéticos , Gliose , Substância Cinzenta/citologia , Camundongos , Neurônios/citologia , Neurônios/metabolismo , Células Piramidais/citologia , Substância Branca/citologia , Ferimentos Perfurantes
11.
EMBO Rep ; 20(11): e47732, 2019 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-31486213

RESUMO

Crosstalk between the actin and microtubule cytoskeletons underlies cellular morphogenesis. Interactions between actin filaments and microtubules are particularly important for establishing the complex polarized morphology of neurons. Here, we characterized the neuronal function of growth arrest-specific 2-like 1 (Gas2L1), a protein that can directly bind to actin, microtubules and microtubule plus-end-tracking end binding proteins. We found that Gas2L1 promotes axon branching, but restricts axon elongation in cultured rat hippocampal neurons. Using pull-down experiments and in vitro reconstitution assays, in which purified Gas2L1 was combined with actin and dynamic microtubules, we demonstrated that Gas2L1 is autoinhibited. This autoinhibition is relieved by simultaneous binding to actin filaments and microtubules. In neurons, Gas2L1 primarily localizes to the actin cytoskeleton and functions as an actin stabilizer. The microtubule-binding tail region of Gas2L1 directs its actin-stabilizing activity towards the axon. We propose that Gas2L1 acts as an actin regulator, the function of which is spatially modulated by microtubules.


Assuntos
Actinas/metabolismo , Axônios/metabolismo , Proteínas dos Microfilamentos/metabolismo , Microtúbulos/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Animais , Biomarcadores , Células COS , Chlorocebus aethiops , Feminino , Células HEK293 , Hipocampo/metabolismo , Humanos , Masculino , Imagem Molecular , Neuritos/metabolismo , Ligação Proteica , Estabilidade Proteica , Transporte Proteico , Células Piramidais/citologia , Células Piramidais/metabolismo , Ratos
12.
Int J Mol Sci ; 20(10)2019 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-31109056

RESUMO

Developmental actions of estradiol in the hypothalamus are well characterized. This hormone generates sex differences in the development of hypothalamic neuronal circuits controlling neuroendocrine events, feeding, growth, reproduction and behavior. In vitro, estradiol promotes sexually dimorphic effects on hypothalamic neuritogenesis. Previous studies have shown that developmental actions of the phytoestrogen genistein result in permanent sexually dimorphic effects in some behaviors and neural circuits in vivo. In the present study, we have explored if genistein, like estradiol, affects neuritogenesis in primary hypothalamic neurons and investigated the estrogen receptors implicated in this action. Hypothalamic neuronal cultures, obtained from male or female embryonic day 14 (E14) CD1 mice, were treated with genistein (0.1 µM, 0.5 µM or 1 µM) or vehicle. Under basal conditions, female neurons had longer primary neurites, higher number of secondary neurites and higher neuritic arborization compared to male neurons. The treatment with genistein increased neuritic arborization and the number of primary neurites and decreased the number of secondary neurites in female neurons, but not in male neurons. In contrast, genistein resulted in a significant increase in primary neuritic length in male neurons, but not in female neurons. The use of selective estrogen receptor antagonists suggests that estrogen receptor α, estrogen receptor ß and G-protein-coupled estrogen receptors are involved in the neuritogenic action of genistein. In summary, these findings indicate that genistein exerts sexually dimorphic actions on the development of hypothalamic neurons, altering the normal pattern of sex differences in neuritogenesis.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Genisteína/farmacologia , Fitoestrógenos/farmacologia , Células Piramidais/citologia , Células Piramidais/efeitos dos fármacos , Caracteres Sexuais , Animais , Biomarcadores , Feminino , Masculino , Camundongos , Neuritos/efeitos dos fármacos , Neuritos/metabolismo , Neurogênese/efeitos dos fármacos , Células Piramidais/metabolismo , Receptores de Estrogênio/antagonistas & inibidores , Receptores de Estrogênio/metabolismo
13.
Sci Signal ; 12(582)2019 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-31113852

RESUMO

Astrocytes are the major glial subtype in the brain and mediate numerous functions ranging from metabolic support to gliotransmitter release through signaling mechanisms controlled by Ca2+ Despite intense interest, the Ca2+ influx pathways in astrocytes remain obscure, hindering mechanistic insights into how Ca2+ signaling is coupled to downstream astrocyte-mediated effector functions. Here, we identified store-operated Ca2+ release-activated Ca2+ (CRAC) channels encoded by Orai1 and STIM1 as a major route of Ca2+ entry for driving sustained and oscillatory Ca2+ signals in astrocytes after stimulation of metabotropic purinergic and protease-activated receptors. Using synaptopHluorin as an optical reporter, we showed that the opening of astrocyte CRAC channels stimulated vesicular exocytosis to mediate the release of gliotransmitters, including ATP. Furthermore, slice electrophysiological recordings showed that activation of astrocytes by protease-activated receptors stimulated interneurons in the CA1 hippocampus to increase inhibitory postsynaptic currents on CA1 pyramidal cells. These results reveal a central role for CRAC channels as regulators of astrocyte Ca2+ signaling, gliotransmitter release, and astrocyte-mediated tonic inhibition of CA1 pyramidal neurons.


Assuntos
Astrócitos/fisiologia , Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Neurônios GABAérgicos/fisiologia , Proteína ORAI1/metabolismo , Molécula 1 de Interação Estromal/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Astrócitos/citologia , Astrócitos/metabolismo , Região CA1 Hipocampal/citologia , Região CA1 Hipocampal/metabolismo , Canais de Cálcio Ativados pela Liberação de Cálcio/genética , Canais de Cálcio Ativados pela Liberação de Cálcio/metabolismo , Células Cultivadas , Exocitose/fisiologia , Feminino , Neurônios GABAérgicos/citologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteína ORAI1/genética , Células Piramidais/citologia , Células Piramidais/fisiologia , Molécula 1 de Interação Estromal/genética , Transmissão Sináptica/fisiologia
14.
J Neurosci ; 39(25): 4874-4888, 2019 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-30992373

RESUMO

Surgical ovariectomy has been shown to reduce spine density in hippocampal CA1 pyramidal cells of rodents, and this reduction is reversed by 17ß-estradiol (E2) treatment in a model of human estrogen replacement therapy. Here, we report reduction of spine density in apical dendrites of layer 5 pyramidal neurons of several neocortical regions that is reversed by subsequent E2 treatment in ovariectomized (OVX) female Thy1M-EGFP mice. We also found that OVX-associated reduction of spine density in somatosensory cortex was accompanied by a reduction in miniature EPSC (mEPSC) frequency (but not mIPSC frequency), indicating a change in functional synapses. OVX-associated spine loss in somatosensory cortex was also rescued by an agonist of the G-protein-linked estrogen receptor (GPER) but not by agonists of the classic estrogen receptors ERα/ERß, whereas the opposite selectivity was found in area CA1. Acute treatment of neocortical slices with E2 also rescued the OVX-associated reduction in mEPSC frequency, which could be mimicked by a GPER agonist and abolished by a GPER antagonist. Time-lapse in vivo two-photon imaging showed that OVX-associated reduction in spine density is achieved by both an increase in spine loss rate and a decrease in spine gain rate and that subsequent rescue by E2 reversed both of these processes. Crucially, the spines added after E2 rescue were no more likely to reappear at or nearby the sites of pre-OVX spines than those in control mice treated with vehicle. Thus, a model of estrogen replacement therapy, although restoring spine density and dynamics, does not entirely restore functional connectivity.SIGNIFICANCE STATEMENT Estrogen replacement therapy following menopause or surgical removal of the ovaries is a widespread medical practice, yet little is known about the consequences of such treatment for cells in the brain. Here, we show that estrogen replacement reverses some of the effects of surgical removal of the ovaries on the structure and function of brain cells in the mouse. Yet, importantly, the fine wiring of the brain is not returned to the presurgery state by estrogen treatment, suggesting lasting functional consequences.


Assuntos
Espinhas Dendríticas/efeitos dos fármacos , Estradiol/farmacologia , Neocórtex/efeitos dos fármacos , Células Piramidais/efeitos dos fármacos , Animais , Espinhas Dendríticas/metabolismo , Estrogênios/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Feminino , Camundongos , Potenciais Pós-Sinápticos em Miniatura/efeitos dos fármacos , Neocórtex/citologia , Neocórtex/metabolismo , Ovariectomia , Células Piramidais/citologia , Células Piramidais/metabolismo
15.
Neuron ; 101(6): 1150-1165.e8, 2019 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-30713030

RESUMO

Diverse computations in the neocortex are aided by specialized GABAergic interneurons (INs), which selectively target other INs. However, much less is known about how these canonical disinhibitory circuit motifs contribute to network operations supporting spatial navigation and learning in the hippocampus. Using chronic two-photon calcium imaging in mice performing random foraging or goal-oriented learning tasks, we found that vasoactive intestinal polypeptide-expressing (VIP+), disinhibitory INs in hippocampal area CA1 form functional subpopulations defined by their modulation by behavioral states and task demands. Optogenetic manipulations of VIP+ INs and computational modeling further showed that VIP+ disinhibition is necessary for goal-directed learning and related reorganization of hippocampal pyramidal cell population dynamics. Our results demonstrate that disinhibitory circuits in the hippocampus play an active role in supporting spatial learning. VIDEO ABSTRACT.


Assuntos
Região CA1 Hipocampal/citologia , Interneurônios/fisiologia , Inibição Neural/fisiologia , Células Piramidais/fisiologia , Aprendizagem Espacial/fisiologia , Animais , Comportamento Apetitivo/fisiologia , Região CA1 Hipocampal/fisiologia , Objetivos , Hipocampo/citologia , Hipocampo/fisiologia , Interneurônios/citologia , Interneurônios/metabolismo , Camundongos , Neocórtex/citologia , Neocórtex/fisiologia , Optogenética , Células Piramidais/citologia , Peptídeo Intestinal Vasoativo/metabolismo
16.
Horm Behav ; 107: 26-34, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30465772

RESUMO

The endocrine disruptor, Bisphenol-A (BPA), alters many behavioral and neural parameters in rodents. BPA administration to gonadally intact adolescent rats increases anxiety, impairs spatial memory, and decreases dendritic spine density when measured in adulthood. Since BPA's action seems to be mediated through gonadal steroid receptors, the current experiments were done in ovariectomized (OVX) female rats to examine the effects on behavior and spine density of adolescent BPA exposure under controlled hormone conditions. OVX (postnatal day, PND, 21) female Sprague-Dawley rats (n = 66) received subcutaneous injections of BPA (40 µg/kg/bodyweight), 17ß-Estradiol (E2, 50 µg/kg/bodyweight), or saline during adolescence (PND 38-49). Following the last injection brains were processed for Golgi impregnation (Exp1), behavioral and spine density in adolescence (Exp2), or in adulthood (Exp3). In Exp1, E2 increased spine density in CA1 pyramidal cells and BPA decreased spine density in granule cells of the dentate gyrus (DG). In Exp2, BPA impaired spatial memory on the object placement (OP) task, E2 increased spine density in CA1, BPA decreased spine density in the DG and the medial prefrontal cortex (mPFC). When measured in adulthood (Exp3), BPA impaired OP and object recognition (OR) performance, E2 increased spine density in CA1, and BPA decreased spine density in CA1, the mPFC and the DG. Results provide novel data on the effects of adolescent BPA in an OVX model and are compared to data in intact animals and within the context of understanding the importance of the profound neuronal alterations occurring during adolescent development.


Assuntos
Compostos Benzidrílicos/toxicidade , Espinhas Dendríticas/efeitos dos fármacos , Disruptores Endócrinos/toxicidade , Memória/efeitos dos fármacos , Fenóis/toxicidade , Maturidade Sexual/efeitos dos fármacos , Fatores Etários , Animais , Contagem de Células , Espinhas Dendríticas/fisiologia , Giro Denteado/efeitos dos fármacos , Exposição Ambiental/efeitos adversos , Estradiol/farmacologia , Feminino , Hipocampo/efeitos dos fármacos , Ovariectomia , Córtex Pré-Frontal/efeitos dos fármacos , Células Piramidais/citologia , Células Piramidais/efeitos dos fármacos , Células Piramidais/fisiologia , Ratos , Ratos Sprague-Dawley
17.
PLoS One ; 13(10): e0204688, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30307948

RESUMO

Human neurodegenerative disorders affect specific types of cortical neurons. Efficient protocols for the generation of such neurons for cell replacement, disease modeling and drug screening are highly warranted. Current methods for the production of cortical neurons from human embryonic stem (ES) cells are often time-consuming and inefficient, and the functional properties of the generated cells have been incompletely characterized. Here we have used transcription factor (TF) programming with the aim to induce rapid differentiation of human ES cells to layer-specific cortical neurons (hES-iNs). Three different combinations of TFs, NEUROGENIN 2 (NGN2) only, NGN2 plus Forebrain Embryonic Zinc Finger-Like Protein 2 (FEZF2), and NGN2 plus Special AT-Rich Sequence-Binding Protein 2 (SATB2), were delivered to human ES cells by lentiviral vectors. We observed only subtle differences between the TF combinations, which all gave rise to the formation of pyramidal-shaped cells, morphologically resembling adult human cortical neurons expressing cortical projection neuron (PN) markers and with mature electrophysiological properties. Using ex vivo transplantation to human organotypic cultures, we found that the hES-iNs could integrate into adult human cortical networks. We obtained no evidence that the hES-iNs had acquired a distinct cortical layer phenotype. Instead, our single-cell data showed that the hES-iNs, similar to fetal human cortical neurons, expressed both upper and deep layer cortical neuronal markers. Taken together, our findings provide evidence that TF programming can direct human ES cells towards cortical neurons but that the generated cells are transcriptionally profiled to generate both upper and deep layer cortical neurons. Therefore, most likely additional cues will be needed if these cells should adopt a specific cortical layer and area identity.


Assuntos
Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Embrionárias Humanas/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Fatores de Transcrição/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Biomarcadores/metabolismo , Diferenciação Celular , Linhagem Celular , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Vetores Genéticos , Células-Tronco Embrionárias Humanas/transplante , Humanos , Técnicas In Vitro , Proteínas de Ligação à Região de Interação com a Matriz/genética , Proteínas de Ligação à Região de Interação com a Matriz/metabolismo , Camundongos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurogênese , Técnicas de Cultura de Órgãos , Células Piramidais/citologia , Células Piramidais/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Fatores de Transcrição/genética
18.
PLoS Biol ; 16(6): e2006387, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29912866

RESUMO

Understanding any brain circuit will require a categorization of its constituent neurons. In hippocampal area CA1, at least 23 classes of GABAergic neuron have been proposed to date. However, this list may be incomplete; additionally, it is unclear whether discrete classes are sufficient to describe the diversity of cortical inhibitory neurons or whether continuous modes of variability are also required. We studied the transcriptomes of 3,663 CA1 inhibitory cells, revealing 10 major GABAergic groups that divided into 49 fine-scale clusters. All previously described and several novel cell classes were identified, with three previously described classes unexpectedly found to be identical. A division into discrete classes, however, was not sufficient to describe the diversity of these cells, as continuous variation also occurred between and within classes. Latent factor analysis revealed that a single continuous variable could predict the expression levels of several genes, which correlated similarly with it across multiple cell types. Analysis of the genes correlating with this variable suggested it reflects a range from metabolically highly active faster-spiking cells that proximally target pyramidal cells to slower-spiking cells targeting distal dendrites or interneurons. These results elucidate the complexity of inhibitory neurons in one of the simplest cortical structures and show that characterizing these cells requires continuous modes of variation as well as discrete cell classes.


Assuntos
Região CA1 Hipocampal/citologia , Região CA1 Hipocampal/metabolismo , Neurônios GABAérgicos/classificação , Neurônios GABAérgicos/metabolismo , Potenciais de Ação , Algoritmos , Animais , Quimiocinas CXC/genética , Dendritos/metabolismo , Neurônios GABAérgicos/citologia , Interneurônios/citologia , Interneurônios/metabolismo , Camundongos , Camundongos Transgênicos , Modelos Neurológicos , Células Piramidais/citologia , Células Piramidais/metabolismo , Proteína Serina-Treonina Quinase 2 de Interação com Receptor , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Análise de Sequência de RNA , Análise de Célula Única , Transmissão Sináptica , Transcriptoma , Peptídeo Intestinal Vasoativo/genética
19.
PLoS One ; 13(4): e0193859, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29617377

RESUMO

Inositol 1,4,5-trisphosphate 3-kinase A (IP3K-A) regulates the level of the inositol polyphosphates, inositol trisphosphate (IP3) and inositol tetrakisphosphate to modulate cellular signaling and intracellular calcium homeostasis in the central nervous system. IP3K-A binds to F-actin in an activity-dependent manner and accumulates in dendritic spines, where it is involved in the regulation of synaptic plasticity. IP3K-A knockout mice exhibit deficits in some forms of hippocampus-dependent learning and synaptic plasticity, such as long-term potentiation in the dentate gyrus synapses of the hippocampus. In the present study, to further elucidate the role of IP3K-A in the brain, we developed a transgenic (Tg) mouse line in which IP3K-A is conditionally overexpressed approximately 3-fold in the excitatory neurons of forebrain regions, including the hippocampus. The Tg mice showed an increase in both presynaptic release probability of evoked responses, along with bigger synaptic vesicle pools, and miniature excitatory postsynaptic current amplitude, although the spine density or the expression levels of the postsynaptic density-related proteins NR2B, synaptotagmin 1, and PSD-95 were not affected. Hippocampal-dependent learning and memory tasks, including novel object recognition and radial arm maze tasks, were partially impaired in Tg mice. Furthermore, (R,S)-3,5-dihydroxyphenylglycine-induced metabotropic glutamate receptor long-term depression was inhibited in Tg mice and this inhibition was dependent on protein kinase C but not on the IP3 receptor. Long-term potentiation and depression dependent on N-methyl-d-aspartate receptor were marginally affected in Tg mice. In summary, this study shows that overexpressed IP3K-A plays a role in some forms of hippocampus-dependent learning and memory tasks as well as in synaptic transmission and plasticity by regulating both presynaptic and postsynaptic functions.


Assuntos
Região CA1 Hipocampal/citologia , Depressão Sináptica de Longo Prazo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Células Piramidais/citologia , Receptores de Glutamato Metabotrópico/metabolismo , Transmissão Sináptica , Animais , Região CA1 Hipocampal/fisiologia , Masculino , Aprendizagem em Labirinto , Memória , Camundongos , Camundongos Transgênicos , Plasticidade Neuronal , Fosfotransferases (Aceptor do Grupo Álcool)/análise , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Prosencéfalo/citologia , Prosencéfalo/fisiologia , Células Piramidais/metabolismo , Regulação para Cima
20.
Neuroscience ; 372: 114-125, 2018 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-29306053

RESUMO

Morphological screening of mouse brains with known behavioral deficits can give great insight into the relationship between brain regions and their behavior. Oxytocin- and CD38-deficient mice have previously been shown to have behavioral phenotypes, such as restrictions in social memory, social interactions, and maternal behavior. CD38 is reported as an autism spectrum disorder (ASD) candidate gene and its behavioral phenotypes may be linked to ASD. To address whether these behavioral phenotypes relate to brain pathology and neuronal morphology, here we investigate the morphological changes in the CD38-deficient mice brains, with focus on the pathology and neuronal morphology of the cortex and hippocampus, using Nissl staining, immunohistochemistry, and Golgi staining. No difference was found in terms of cortical layer thickness. However, we found abnormalities in the number of neurons and neuronal morphology in the visual cortex and dentate gyrus (DG). In particular, there were arborisation differences between CD38-/- and CD38+/+ mice in the apical dendrites of the visual cortex and hippocampal CA1 pyramidal neurons. The data suggest that CD38 is implicated in appropriate development of brain regions important for social behavior.


Assuntos
ADP-Ribosil Ciclase 1/metabolismo , Dendritos/metabolismo , Hipocampo/citologia , Hipocampo/metabolismo , Glicoproteínas de Membrana/metabolismo , Córtex Visual/citologia , Córtex Visual/metabolismo , ADP-Ribosil Ciclase 1/genética , Animais , Contagem de Células , Dendritos/patologia , Hipocampo/patologia , Proteínas de Homeodomínio/metabolismo , Imuno-Histoquímica , Glicoproteínas de Membrana/genética , Camundongos Endogâmicos ICR , Camundongos Knockout , Microscopia Confocal , Proteínas Nucleares/metabolismo , Tamanho do Órgão , Células Piramidais/citologia , Células Piramidais/metabolismo , Células Piramidais/patologia , Proteínas Repressoras/metabolismo , Coloração pela Prata , Córtex Visual/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA