Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 212
Filtrar
1.
J Neuroinflammation ; 17(1): 8, 2020 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-31906973

RESUMO

BACKGROUND: Cognitive impairment in schizophrenia, aging, and Alzheimer's disease is associated with spine and synapse loss from the dorsolateral prefrontal cortex (dlPFC) layer III. Complement cascade signaling is critical in driving spine loss and disease pathogenesis. Complement signaling is initiated by C1q, which tags synapses for elimination. C1q is thought to be expressed predominately by microglia, but its expression in primate dlPFC has never been examined. The current study assayed C1q levels in aging primate dlPFC and rat medial PFC (mPFC) and used immunoelectron microscopy (immunoEM), immunoblotting, and co-immunoprecipitation (co-IP) to reveal the precise anatomical distribution and interactions of C1q. METHODS: Age-related changes in C1q levels in rhesus macaque dlPFC and rat mPFC were examined using immunoblotting. High-spatial resolution immunoEM was used to interrogate the subcellular localization of C1q in aged macaque layer III dlPFC and aged rat layer III mPFC. co-IP techniques quantified protein-protein interactions for C1q and proteins associated with excitatory and inhibitory synapses in macaque dlPFC. RESULTS: C1q levels were markedly increased in the aged macaque dlPFC. Ultrastructural localization found the expected C1q localization in glia, including those ensheathing synapses, but also revealed extensive localization within neurons. C1q was found near synapses, within terminals and in spines, but was also observed in dendrites, often near abnormal mitochondria. Similar analyses in aging rat mPFC corroborated the findings in rhesus macaques. C1q protein increasingly associated with PSD95 with age in macaque, consistent with its synaptic localization as evidenced by EM. CONCLUSIONS: These findings reveal novel, intra-neuronal distribution patterns for C1q in the aging primate cortex, including evidence of C1q in dendrites. They suggest that age-related changes in the dlPFC may increase C1q expression and synaptic tagging for glial phagocytosis, a possible mechanism for age-related degeneration.


Assuntos
Envelhecimento/metabolismo , Complemento C1q/análise , Complemento C1q/metabolismo , Neurônios/metabolismo , Córtex Pré-Frontal/química , Córtex Pré-Frontal/metabolismo , Animais , Macaca mulatta , Neurônios/ultraestrutura , Córtex Pré-Frontal/ultraestrutura , Ratos , Ratos Sprague-Dawley
2.
Dev Sci ; 22(6): e12834, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30964952

RESUMO

Biologically embedded experiences alter developmental trajectories in ways that can influence health, learning, and/or behavior. These systematic differences in experiences may contribute to different biological outcomes as individuals grow and develop, including at the neural level. Previous studies of biologically embedded experiences on neurodevelopment have focused on large-scale institutional or economic factors (e.g. socioeconomic status [SES]) and psychosocial factors (e.g. caregiving behavior). Less attention has focused on how the quality of the immediate home settings, such as the physical home environment (PHYS), influences neurodevelopment. Moreover, no study has investigated these effects in adolescents, who undergo significant physical maturation and neurodevelopment that may influence how they respond to their physical environments. The goal of the current study was to examine whether PHYS quality is biologically embedded in the developing adolescent brain as evidenced by cognitive achievement and cortical development in 56 (48% female) healthy adolescents (14-18 years (M = 16.83 years, SD = 1.17). Using in-home assessments of the physical home environment, anatomical brain scans, and indices of academic achievement, we found that adolescents who have more physical problems in the home (e.g. structural hazards, crowding, excessive noise, poorly lit) have thinner prefrontal cortices, which was associated with lower levels of reading achievement, independent of SES and psychosocial factors. By conducting home visits to assess physical characteristics of adolescents' home, we highlight a typically overlooked aspect of the home environment that has relevance for adolescents' cognitive and brain development.


Assuntos
Desenvolvimento do Adolescente/fisiologia , Encéfalo/crescimento & desenvolvimento , Meio Ambiente , Córtex Pré-Frontal/ultraestrutura , Classe Social , Sucesso Acadêmico , Adolescente , Encéfalo/fisiologia , Criança , Feminino , Humanos , Masculino , Neuroimagem , Córtex Pré-Frontal/crescimento & desenvolvimento , Leitura
3.
Cereb Cortex ; 28(3): 974-987, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28108498

RESUMO

The newly evolved circuits in layer III of primate dorsolateral prefrontal cortex (dlPFC) generate the neural representations that subserve working memory. These circuits are weakened by increased cAMP-K+ channel signaling, and are a focus of pathology in schizophrenia, aging, and Alzheimer's disease. Cognitive deficits in these disorders are increasingly associated with insults to mGluR3 metabotropic glutamate receptors, while reductions in mGluR2 appear protective. This has been perplexing, as mGluR3 has been considered glial receptors, and mGluR2 and mGluR3 have been thought to have similar functions, reducing glutamate transmission. We have discovered that, in addition to their astrocytic expression, mGluR3 is concentrated postsynaptically in spine synapses of layer III dlPFC, positioned to strengthen connectivity by inhibiting postsynaptic cAMP-K+ channel actions. In contrast, mGluR2 is principally presynaptic as expected, with only a minor postsynaptic component. Functionally, increase in the endogenous mGluR3 agonist, N-acetylaspartylglutamate, markedly enhanced dlPFC Delay cell firing during a working memory task via inhibition of cAMP signaling, while the mGluR2 positive allosteric modulator, BINA, produced an inverted-U dose-response on dlPFC Delay cell firing and working memory performance. These data illuminate why insults to mGluR3 would erode cognitive abilities, and support mGluR3 as a novel therapeutic target for higher cognitive disorders.


Assuntos
Memória de Curto Prazo/fisiologia , Neurônios/citologia , Densidade Pós-Sináptica/metabolismo , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/fisiologia , Receptores de Glutamato Metabotrópico/metabolismo , Potenciais de Ação/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Fármacos Atuantes sobre Aminoácidos Excitatórios/farmacologia , Movimentos Oculares/efeitos dos fármacos , Movimentos Oculares/fisiologia , Feminino , Processamento de Imagem Assistida por Computador , Macaca mulatta , Imageamento por Ressonância Magnética , Masculino , Memória de Curto Prazo/efeitos dos fármacos , Neurônios/metabolismo , Densidade Pós-Sináptica/ultraestrutura , Córtex Pré-Frontal/diagnóstico por imagem , Córtex Pré-Frontal/ultraestrutura , Ratos , Receptores de Glutamato Metabotrópico/ultraestrutura , Aprendizagem Espacial/efeitos dos fármacos , Frações Subcelulares/efeitos dos fármacos
4.
J Neurosci ; 37(9): 2292-2304, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28137966

RESUMO

In chronic pain, the medial prefrontal cortex (mPFC) is deactivated and mPFC-dependent tasks such as attention and working memory are impaired. We investigated the mechanisms of mPFC deactivation in the rat spared nerve injury (SNI) model of neuropathic pain. Patch-clamp recordings in acute slices showed that, 1 week after the nerve injury, cholinergic modulation of layer 5 (L5) pyramidal neurons was severely impaired. In cells from sham-operated animals, focal application of acetylcholine induced a left shift of the input/output curve and persistent firing. Both of these effects were almost completely abolished in cells from SNI-operated rats. The cause of this impairment was an ∼60% reduction of an M1-coupled, pirenzepine-sensitive depolarizing current, which appeared to be, at least in part, the consequence of M1 receptor internalization. Although no changes were detected in total M1 protein or transcript, both the fraction of the M1 receptor in the synaptic plasma membrane and the biotinylated M1 protein associated with the total plasma membrane were decreased in L5 mPFC of SNI rats. The loss of excitatory cholinergic modulation may play a critical role in mPFC deactivation in neuropathic pain and underlie the mPFC-specific cognitive deficits that are comorbid with neuropathic pain.SIGNIFICANCE STATEMENT The medial prefrontal cortex (mPFC) undergoes major reorganization in chronic pain. Deactivation of mPFC output is causally correlated with both the cognitive and the sensory component of neuropathic pain. Here, we show that cholinergic excitation of commissural layer 5 mPFC pyramidal neurons is abolished in neuropathic pain rats due to a severe reduction of a muscarinic depolarizing current and M1 receptor internalization. Therefore, in neuropathic pain rats, the acetylcholine (ACh)-dependent increase in neuronal excitability is reduced dramatically and the ACh-induced persisting firing, which is critical for working memory, is abolished. We propose that the blunted cholinergic excitability contributes to the functional mPFC deactivation that is causal for the pain phenotype and represents a cellular mechanism for the attention and memory impairments comorbid with chronic pain.


Assuntos
Acetilcolina/metabolismo , Limiar da Dor/fisiologia , Córtex Pré-Frontal/metabolismo , Receptor Muscarínico M1/metabolismo , Ciática/patologia , Acetilcolina/farmacologia , Potenciais de Ação/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Antagonistas de Aminoácidos Excitatórios/farmacologia , Antagonistas GABAérgicos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Hiperalgesia/fisiopatologia , Masculino , Picrotoxina/farmacologia , Córtex Pré-Frontal/patologia , Córtex Pré-Frontal/ultraestrutura , Células Piramidais/efeitos dos fármacos , Células Piramidais/fisiologia , Quinoxalinas/farmacologia , Ratos , Ratos Sprague-Dawley , Receptor Muscarínico M1/genética , Ciática/fisiopatologia , Frações Subcelulares/metabolismo , Frações Subcelulares/patologia , Transmissão Sináptica/efeitos dos fármacos , Valina/análogos & derivados , Valina/farmacologia
5.
Acta Neuropathol ; 135(2): 213-226, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29273900

RESUMO

In addition to motor neurone degeneration, up to 50% of amyotrophic lateral sclerosis (ALS) patients present with cognitive decline. Understanding the neurobiological changes underlying these cognitive deficits is critical, as cognitively impaired patients exhibit a shorter survival time from symptom onset. Given the pathogenic role of synapse loss in other neurodegenerative diseases in which cognitive decline is apparent, such as Alzheimer's disease, we aimed to assess synaptic integrity in the ALS brain. Here, we have applied a unique combination of high-resolution imaging of post-mortem tissue with neuropathology, genetic screening and cognitive profiling of ALS cases. Analyses of more than 1 million synapses using two complimentary high-resolution techniques (electron microscopy and array tomography) revealed a loss of synapses from the prefrontal cortex of ALS patients. Importantly, synapse loss was significantly greater in cognitively impaired cases and was not due to cortical atrophy, nor associated with dementia-associated neuropathology. Interestingly, we found a trend between pTDP-43 pathology and synapse loss in the frontal cortex and discovered pTDP-43 puncta at a subset of synapses in the ALS brains. From these data, we postulate that synapse loss in the prefrontal cortex represents an underlying neurobiological substrate of cognitive decline in ALS.


Assuntos
Esclerose Lateral Amiotrófica/patologia , Esclerose Lateral Amiotrófica/psicologia , Disfunção Cognitiva/patologia , Córtex Pré-Frontal/patologia , Sinapses/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/metabolismo , Atrofia , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/genética , Disfunção Cognitiva/metabolismo , Estudos de Coortes , Proteínas de Ligação a DNA/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fosforilação , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/ultraestrutura , Sinapses/metabolismo , Sinapses/ultraestrutura
6.
Cereb Cortex ; 27(5): 2871-2884, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-27178192

RESUMO

Adverse early-life experience such as depriving the relationship between parents and children induces permanent phenotypic changes, and impairs the cognitive functions associated with the prefrontal cortex (PFC). However, the underlying mechanism remains unclear. In this work, we used rat neonatal maternal separation (NMS) model to illuminate whether and how NMS in early life affects cognitive functions, and what the underlying cellular and molecular mechanism is. We showed that rat pups separated from their dam 3 h daily during the first 3 postnatal weeks alters medial prefrontal cortex (mPFC) myelination and impairs mPFC-dependent behaviors. Myelination appears necessary for mPFC-dependent behaviors, as blockade of oligodendrocytes (OLs) differentiation or lysolecithin-induced demyelination, impairs mPFC functions. We further demonstrate that histone deacetylases 1/2 (HDAC1/2) are drastically reduced in NMS rats. Inhibition of HDAC1/2 promotes Wnt activation, which negatively regulates OLs development. Conversely, selective inhibition of Wnt signaling by XAV939 partly rescue myelination arrestment and behavior deficiency caused by NMS. These findings indicate that NMS impairs mPFC cognitive functions, at least in part, through modulation of oligodendrogenesis and myelination. Understanding the mechanism of NMS on mPFC-dependent behaviors is critical for developing pharmacological and psychological interventions for child neglect and abuse.


Assuntos
Transtornos Cognitivos/etiologia , Doenças Desmielinizantes/patologia , Privação Materna , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/patologia , Proteínas Wnt/metabolismo , Via de Sinalização Wnt/fisiologia , Animais , Animais Recém-Nascidos , Ansiedade/etiologia , Transtornos Cognitivos/patologia , Doenças Desmielinizantes/etiologia , Inibidores Enzimáticos/farmacologia , Comportamento Exploratório/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Compostos Heterocíclicos com 3 Anéis/farmacologia , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Lipopolissacarídeos/toxicidade , Aprendizagem em Labirinto/efeitos dos fármacos , Proteína Básica da Mielina/metabolismo , Neurogênese/efeitos dos fármacos , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/ultraestrutura , Ratos , Ratos Sprague-Dawley , Ácido Valproico/farmacologia , Proteínas Wnt/genética , Via de Sinalização Wnt/efeitos dos fármacos
7.
Bull Exp Biol Med ; 164(3): 376-381, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29313234

RESUMO

We performed ultrastructural study of cerebral cortex mitochondria in rats with different tolerance to oxygen deficiency (low resistant and highly resistant specimens). Low resistant rats were characterized by the prevalence of mitochondria with lightened matrix due to the nondense packing of cristae. By contrast, mitochondria of highly resistant animals had the dense packing of cristae. The structure of mitochondria underwent adaptive changes at 14-10% O2 in the inspired air. Under these conditions, structural characteristics of the cerebral cortex in hypoxia-sensitive rats resembled those in resistant animals. The decrease in O2 concentration to 8% was accompanied by ultrastructural signs of mitochondrial damage, which correlated with de-energization of the cell and dysfunction of adaptive signaling systems. Ultrastructural features of cerebral cortex mitochondria in animals with low and high tolerance to acute oxygen deficiency confirm the hypothesis that they are associated with two different "functionaland-metabolic portraits".


Assuntos
Adaptação Fisiológica , Doença da Altitude/patologia , Mitocôndrias/ultraestrutura , Oxigênio/farmacologia , Córtex Pré-Frontal/ultraestrutura , Doença da Altitude/fisiopatologia , Animais , Animais não Endogâmicos , Modelos Animais de Doenças , Microtomia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/patologia , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/patologia , Córtex Pré-Frontal/fisiopatologia , Ratos , Técnicas de Cultura de Tecidos
8.
J Proteome Res ; 16(12): 4481-4494, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-28949146

RESUMO

Schizophrenia is a chronic and incurable neuropsychiatric disorder that affects about one percent of the world population. The proteomic characterization of the synaptosome fraction of the orbitofrontal cortex is useful for providing valuable information about the molecular mechanisms of synaptic functions in these patients. Quantitative analyses of synaptic proteins were made with eight paranoid schizophrenia patients and a pool of eight healthy controls free of mental diseases. Label-free and iTRAQ labeling identified a total of 2018 protein groups. Statistical analyses revealed 12 and 55 significantly dysregulated proteins by iTRAQ and label-free, respectively. Quantitative proteome analyses showed an imbalance in the calcium signaling pathway and proteins such as reticulon-1 and cytochrome c, related to endoplasmic reticulum stress and programmed cell death. Also, it was found that there is a significant increase in limbic-system-associated membrane protein and α-calcium/calmodulin-dependent protein kinase II, associated with the regulation of human behavior. Our data contribute to a better understanding about apoptosis as a possible pathophysiological mechanism of this disease as well as neural systems supporting social behavior in schizophrenia. This study also is a joint effort of the Chr 15 C-HPP team and the Human Brain Proteome Project of B/D-HPP. All MS proteomics data are deposited in the ProteomeXchange Repository under PXD006798.


Assuntos
Córtex Pré-Frontal/química , Proteoma/análise , Proteômica/métodos , Esquizofrenia/patologia , Sinaptossomos/química , Estudos de Casos e Controles , Humanos , Espectrometria de Massas , Redes e Vias Metabólicas , Córtex Pré-Frontal/ultraestrutura
9.
Int J Neuropsychopharmacol ; 20(11): 886-895, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-29016795

RESUMO

Background: Leonurine is an active alkaloid that is extracted from Traditional Chinese Medicine Herba leonuri. Emerging evidence indicates that leonurine produces neuroprotective effects in ischemic stroke, Parkinson's disease, and Alzheimer's disease. However, the effect of leonurine in neuropsychiatric disorders, especially in major depression, remains unknown. Methods: We used the chronic mild stress mouse model to explore the antidepressant effects of leonurine and the potential mechanisms. Behavioral tests including sucrose preference test, forced swimming test, and tail suspension test were taken to evaluate depression symptoms. Moreover, the contents of monoamine neurotransmitters in hippocampus and prefrontal cortex were measured by high-performance liquid chromatography. Neuronal morphology was detected by transmission electron microscopy. Results: Administration of leonurine (60 mg/kg) for 4 weeks significantly alleviated depression-like behaviors of chronic mild stress mice, including increased sucrose preference and reduced immobility time in forced swimming test and tail suspension test. We further found that leonurine (60 mg/kg) effectively restored the levels of 5-hydroxytryptamine, noradrenaline, and dopamine in the hippocampus and prefrontal cortex of chronic mild stress mice, accompanied by amelioration of hippocampal neuronal damage. Furthermore, leonurine (60 mg/kg) significantly inhibited the production of proinflammatory cytokine interleukin-1ß, interleukin-6 and TNF-α, and suppressed the nuclear factor kappa B signaling pathway. Conclusions: These findings demonstrate that leonurine exerts antidepressant-like effects, which may be mediated, at least in part, by improving monoamine neurotransmitters and inhibiting neuroinflammation. Our study provides insight into the potential of leonurine in depression therapy.


Assuntos
Antidepressivos/uso terapêutico , Depressão/tratamento farmacológico , Depressão/etiologia , Encefalite/tratamento farmacológico , Ácido Gálico/análogos & derivados , Estresse Psicológico/complicações , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Citocinas/genética , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Encefalite/etiologia , Fluoxetina/uso terapêutico , Preferências Alimentares/efeitos dos fármacos , Ácido Gálico/uso terapêutico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas dos Microfilamentos/metabolismo , Microglia/efeitos dos fármacos , Microglia/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/patologia , Neurônios/ultraestrutura , Neurotransmissores/metabolismo , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/patologia , Córtex Pré-Frontal/ultraestrutura , Estresse Psicológico/patologia , Natação/psicologia
10.
Brain Behav Immun ; 64: 232-243, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28412142

RESUMO

Early social isolation (SI) produces a variety of emotional, behavioral and cognitive abnormalities. Conversely, environmental enrichment (EE), a complicated social and physical construct, offers beneficial effects on brain plasticity and development. However, whether or not exclusive physical EE is sufficient to reverse the adverse consequences of early SI remains unclear. Here we reported that 1month-old solitary mice housed in the EE for 8weeks corrected spatial cognitive dysfunction, but did not ameliorate social interaction deficits and increased anxiety-like behavior. Pathological analyses revealed that the enriched environment decreased cellular apoptosis, synaptic protein loss, myelination defect and microglial activation in the hippocampus, but not medial prefrontal cortex (mPFC) of mice housed singly. Moreover, increased nuclear factor-kappaB and interleukin-1ß levels, and downregulation of brain-derived neurotrophic factor signaling pathway were normalized in the hippocampus rather than mPFC of these animals. Our results revealed a brain region-specific effectiveness of physical EE in remediating brain impairment of adolescent SI mice, with a complete reversal of hippocampus-dependent cognitive dysfunctions, but without mitigation of mPFC associated anxiety and social interaction defects. This finding emphasizes the irreplaceable role of social life for the early brain development.


Assuntos
Meio Ambiente , Hipocampo/metabolismo , Córtex Pré-Frontal/metabolismo , Isolamento Social , Animais , Ansiedade , Apoptose , Comportamento Animal , Encefalite/metabolismo , Hipocampo/patologia , Hipocampo/ultraestrutura , Camundongos , Bainha de Mielina/patologia , Córtex Pré-Frontal/patologia , Córtex Pré-Frontal/ultraestrutura , Aprendizagem Espacial , Memória Espacial
11.
Proc Natl Acad Sci U S A ; 111(32): 11852-7, 2014 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-25071187

RESUMO

Understanding the mechanisms by which long-term memories are formed and stored in the brain represents a central aim of neuroscience. Prevailing theory suggests that long-term memory encoding involves early plasticity within hippocampal circuits, whereas reorganization of the neocortex is thought to occur weeks to months later to subserve remote memory storage. Here we report that long-term memory encoding can elicit early transcriptional, structural, and functional remodeling of the neocortex. Parallel studies using genome-wide RNA sequencing, ultrastructural imaging, and whole-cell recording in wild-type mice suggest that contextual fear conditioning initiates a transcriptional program in the medial prefrontal cortex (mPFC) that is accompanied by rapid expansion of the synaptic active zone and postsynaptic density, enhanced dendritic spine plasticity, and increased synaptic efficacy. To address the real-time contribution of the mPFC to long-term memory encoding, we performed temporally precise optogenetic inhibition of excitatory mPFC neurons during contextual fear conditioning. Using this approach, we found that real-time inhibition of the mPFC inhibited activation of the entorhinal-hippocampal circuit and impaired the formation of long-term associative memory. These findings suggest that encoding of long-term episodic memory is associated with early remodeling of neocortical circuits, identify the prefrontal cortex as a critical regulator of encoding-induced hippocampal activation and long-term memory formation, and have important implications for understanding memory processing in healthy and diseased brain states.


Assuntos
Memória Episódica , Neocórtex/fisiologia , Animais , Condicionamento Psicológico/fisiologia , Córtex Entorrinal/fisiologia , Medo/fisiologia , Hipocampo/fisiologia , Masculino , Memória de Longo Prazo/fisiologia , Camundongos , Microscopia Eletrônica de Transmissão , Potenciais Pós-Sinápticos em Miniatura/fisiologia , Neocórtex/ultraestrutura , Plasticidade Neuronal/fisiologia , Optogenética , Córtex Pré-Frontal/fisiologia , Córtex Pré-Frontal/ultraestrutura , Transcriptoma
12.
J Neurosci ; 35(34): 11976-87, 2015 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-26311778

RESUMO

The primate amygdala projects to posterior orbitofrontal cortex (pOFC) directly and possibly indirectly through a pathway to the magnocellular mediodorsal thalamic nucleus (MDmc), which may convey signals about the significance of stimuli. However, because MDmc receives input from structures in addition to the amygdala and MDmc projects to areas in addition to pOFC, it is unknown whether amygdalar pathways in MDmc innervate pOFC-bound neurons. We addressed this issue using double- or triple-labeling approaches to identify pathways and key cellular and molecular features in rhesus monkeys. We found that amygdalar terminations innervated labeled neurons in MDmc that project to pOFC. Projection neurons in MDmc directed to pOFC included comparatively fewer "core" parvalbumin neurons that project focally to the middle cortical layers and more "matrix" calbindin neurons that project expansively to the upper cortical layers. In addition, a small and hitherto unknown pathway originated from MDmc calretinin neurons and projected to pOFC. Further, whereas projection neurons directed to MDmc and to pOFC were intermingled in the amygdala, none projected to both structures. Larger amygdalar neurons projected to MDmc and expressed the vesicular glutamate transporter 2 (VGLUT2), which is found in highly efficient "driver" pathways. In contrast, smaller amygdalar neurons directed to pOFC expressed VGLUT1 found in modulatory pathways. The indirect pathway from the amygdala to pOFC via MDmc may provide information about the emotional significance of events and, along with a parallel direct pathway, ensures transfer of signals to all layers of pOFC. SIGNIFICANCE STATEMENT: The amygdala-the brain's center for emotions-is strongly linked with the orbital cortex, a region associated with social interactions. This study provides evidence that a robust pathway from the amygdala reaches neurons in the thalamus that link directly with the orbital cortex, forming a tight tripartite network. The dual pathways from the amygdala to the orbital cortex and to the thalamus are distinct by morphology, neurochemistry, and function. This tightly linked network suggests the presence of fool-proof avenues for emotions to influence high-order cortical areas associated with affective reasoning. Specific nodes of this tripartite network are disrupted in psychiatric diseases, divorcing areas that integrate emotions and thoughts for decisions and flexible behavior.


Assuntos
Tonsila do Cerebelo/fisiologia , Emoções/fisiologia , Lobo Frontal/fisiologia , Núcleo Mediodorsal do Tálamo/fisiologia , Córtex Pré-Frontal/fisiologia , Tonsila do Cerebelo/ultraestrutura , Animais , Feminino , Lobo Frontal/ultraestrutura , Macaca mulatta , Masculino , Núcleo Mediodorsal do Tálamo/ultraestrutura , Vias Neurais/fisiologia , Vias Neurais/ultraestrutura , Córtex Pré-Frontal/ultraestrutura
13.
Ann Neurol ; 78(2): 211-21, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25921485

RESUMO

OBJECTIVE: The proteinaceous inclusions in TDP-43 proteinopathies such as frontotemporal lobar degeneration (FTLD)-TDP are made of high-molecular-weight aggregates of TDP-43. These aggregates have not been classified as amyloids, as prior amyloid staining results were not conclusive. Here we used a specific TDP-43 amyloid oligomer antibody called TDP-O to determine the presence and abundance of TDP-43 oligomers among different subtypes of FTLD-TDP as well as in hippocampal sclerosis (HS), which represents a non-FTLD pathology with TDP-43 inclusions. METHODS: Postmortem tissue from the hippocampus and anterior orbital gyrus from 54 prospectively assessed and diagnosed subjects was used for immunostaining with TDP-O. Electron microscopy was used to assess the subcellular locations of TDP-O-decorated structures. RESULTS: TDP-43 inclusions staining with TDP-O were present in FTLD-TDP and were most conspicuous for FTLD-TDP type C, the subtype seen in most patients with semantic variant primary progressive aphasia. TDP-O immunoreactivity was absent in the hippocampus of HS patients despite abundant TDP-43 inclusions. Ultrastructurally, TDP-43 oligomers resided in granular or tubular structures, frequently in close proximity to, but not within, neuronal lysosomes. INTERPRETATION: TDP-43 forms amyloid oligomers in the human brain, which may cause neurotoxicity in a manner similar to other amyloid oligomers. Oligomer formation may contribute to the conformational heterogeneity of TDP-43 aggregates and mark the different properties of TDP-43 inclusions between FTLD-TDP and HS.


Assuntos
Amiloide/metabolismo , Esclerose Lateral Amiotrófica/metabolismo , Proteínas de Ligação a DNA/metabolismo , Degeneração Lobar Frontotemporal/metabolismo , Hipocampo/metabolismo , Neurônios/metabolismo , Córtex Pré-Frontal/metabolismo , Idoso , Biopolímeros/metabolismo , Proteínas de Ligação a DNA/ultraestrutura , Feminino , Hipocampo/patologia , Hipocampo/ultraestrutura , Humanos , Imuno-Histoquímica , Masculino , Microscopia Eletrônica , Pessoa de Meia-Idade , Neurônios/ultraestrutura , Córtex Pré-Frontal/ultraestrutura , Esclerose
14.
Environ Res ; 146: 404-17, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26829765

RESUMO

Millions of urban children are chronically exposed to high concentrations of air pollutants, i.e., fine particulate matter (PM2.5) and ozone, associated with increased risk for Alzheimer's disease. Compared with children living with clear air those in Mexico City (MC) exhibit systemic, brain and intrathecal inflammation, low CSF Aß42, breakdown of the BBB, attention and short-term memory deficits, prefrontal white matter hyperintensities, damage to epithelial and endothelial barriers, tight junction and neural autoantibodies, and Alzheimer and Parkinson's hallmarks. The prefrontal white matter is a target of air pollution. We examined by light and electron microscopy the prefrontal white matter of MC dogs (n: 15, age 3.17±0.74 years), children and teens (n: 34, age: 12.64±4.2 years) versus controls. Major findings in MC residents included leaking capillaries and small arterioles with extravascular lipids and erythrocytes, lipofuscin in pericytes, smooth muscle and endothelial cells (EC), thickening of cerebrovascular basement membranes with small deposits of amyloid, patchy absence of the perivascular glial sheet, enlarged Virchow-Robin spaces and nanosize particles (20-48nm) in EC, basement membranes, axons and dendrites. Tight junctions, a key component of the neurovascular unit (NVU) were abnormal in MC versus control dogs (χ(2)<0.0001), and white matter perivascular damage was significantly worse in MC dogs (p=0.002). The integrity of the NVU, an interactive network of vascular, glial and neuronal cells is compromised in MC young residents. Characterizing the early NVU damage and identifying biomarkers of neurovascular dysfunction may provide a fresh insight into Alzheimer pathogenesis and open opportunities for pediatric neuroprotection.


Assuntos
Poluição do Ar/efeitos adversos , Córtex Pré-Frontal/patologia , Substância Branca/patologia , Adolescente , Doença de Alzheimer/induzido quimicamente , Animais , Criança , Pré-Escolar , Cães , Feminino , Humanos , Lactente , Masculino , México , Microscopia Eletrônica de Transmissão , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/ultraestrutura , População Urbana , Substância Branca/efeitos dos fármacos , Substância Branca/ultraestrutura
15.
J Neurosci ; 34(24): 8106-18, 2014 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-24920616

RESUMO

The primate amygdala sends dense projections to posterior orbitofrontal cortex (pOFC) in pathways that are critical for processing emotional content, but the synaptic mechanisms are not understood. We addressed this issue by investigating pathways in rhesus monkeys (Macaca mulatta) from the amygdala to pOFC at the level of the system and synapse. Terminations from the amygdala were denser and larger in pOFC compared with the anterior cingulate cortex, which is also strongly connected with the amygdala. Axons from the amygdala terminated most densely in the upper layers of pOFC through large terminals. Most of these terminals innervated spines of presumed excitatory neurons and many were frequently multisynaptic and perforated, suggesting high synaptic efficacy. These amygdalar synapses in pOFC exceeded in size and specialization even thalamocortical terminals from the prefrontal-related thalamic mediodorsal nucleus to the middle cortical layers, which are thought to be highly efficient drivers of cortical neurons. Pathway terminals in the upper layers impinge on the apical dendrites of neurons in other layers, suggesting that the robust amygdalar projections may also activate neurons in layer 5 that project back to the amygdala and beyond to autonomic structures. Among inhibitory neurons, the amygdalar pathway innervated preferentially the neurochemical classes of calbindin and calretinin neurons in the upper layers of pOFC, which are synaptically suited to suppress noise and enhance signals. These features provide a circuit mechanism for flexibly shifting focus and adjusting emotional drive in processes disrupted in psychiatric disorders, such as phobias and obsessive-compulsive disorder.


Assuntos
Vias Aferentes/fisiologia , Tonsila do Cerebelo/fisiologia , Córtex Pré-Frontal/fisiologia , Tonsila do Cerebelo/citologia , Tonsila do Cerebelo/metabolismo , Tonsila do Cerebelo/ultraestrutura , Análise de Variância , Animais , Biotina/análogos & derivados , Biotina/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Dextranos/metabolismo , Feminino , Isoquinolinas/metabolismo , Macaca mulatta , Masculino , Núcleo Mediodorsal do Tálamo/fisiologia , Microscopia Confocal , Microscopia Imunoeletrônica , Modelos Neurológicos , Neurônios/metabolismo , Neurônios/ultraestrutura , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/ultraestrutura , Terminações Pré-Sinápticas/metabolismo , Terminações Pré-Sinápticas/ultraestrutura , Sinapses/metabolismo , Sinapses/ultraestrutura
16.
Neurobiol Dis ; 73: 60-9, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25281318

RESUMO

Current concepts suggest that exposure to THC during adolescence may act as a risk factor for the development of psychiatric disorders later in life. However, the molecular underpinnings of this vulnerability are still poorly understood. To analyze this, we investigated whether and how THC exposure in female rats interferes with different maturational events occurring in the prefrontal cortex during adolescence through biochemical, pharmacological and electrophysiological means. We found that the endocannabinoid system undergoes maturational processes during adolescence and that THC exposure disrupts them, leading to impairment of both endocannabinoid signaling and endocannabinoid-mediated LTD in the adult prefrontal cortex. THC also altered the maturational fluctuations of NMDA subunits, leading to larger amounts of gluN2B at adulthood. Adult animals exposed to THC during adolescence also showed increased AMPA gluA1 with no changes in gluA2 subunits. Finally, adolescent THC exposure altered cognition at adulthood. All these effects seem to be triggered by the disruption of the physiological role played by the endocannabinoid system during adolescence. Indeed, blockade of CB1 receptors from early to late adolescence seems to prevent the occurrence of pruning at glutamatergic synapses. These results suggest that vulnerability of adolescent female rats to long-lasting THC adverse effects might partly reside in disruption of the pivotal role played by the endocannabinoid system in the prefrontal cortex maturation.


Assuntos
Agonistas de Receptores de Canabinoides/farmacologia , Deficiências do Desenvolvimento/induzido quimicamente , Dronabinol/farmacologia , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/fisiologia , Fatores Etários , Animais , Cicloexanóis/farmacocinética , Maleato de Dizocilpina/farmacocinética , Estradiol/sangue , Ciclo Estral/efeitos dos fármacos , Antagonistas de Aminoácidos Excitatórios/farmacocinética , Feminino , Técnicas In Vitro , Neuritos/efeitos dos fármacos , Piperidinas/farmacologia , Córtex Pré-Frontal/diagnóstico por imagem , Córtex Pré-Frontal/ultraestrutura , Pirazóis/farmacologia , Cintilografia , Ratos , Ratos Sprague-Dawley , Receptores de Glutamato/metabolismo , Potenciais Sinápticos/efeitos dos fármacos , Trítio/farmacocinética
17.
Metab Brain Dis ; 30(6): 1531-6, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26307418

RESUMO

Diabetes mellitus (DM) is a risk factor for Alzheimer's disease (AD), and several individuals with AD are diabetic. Most non-transgenic animal models of AD make use of oral treatment with aluminium chloride (AlCl(3)) to induce brain lesions pathognomonic of the disease. Moreover, streptozotocin (STZ) can induce pathological features of either AD or DM depending on the mode of treatment. In the present study, we characterised prefrontal microanatomy and antioxidant defence system in a rat model of AD confounded by DM, with the objective of assessing the suitability of this model in the study of sporadic AD with DM co-morbidity. Adult Wistar rats were randomly assigned to receive either intraperitoneal STZ (30 mg/kg/day for 3 days; to induce DM), oral AlCl(3) (500 mg/kg/day for 4 weeks; to induce some brain lesions characteristic of AD); or both STZ and AlCl(3) (to induce AD with DM co-morbidity). Untreated rats served as controls. During treatment, blood glucose levels and body weights were evaluated repeatedly in all rats. At euthanasia, prefrontal cortex was homogenized in phosphate buffer solution and the supernatants assayed for some antioxidant enzymes (catalase, CAT; superoxide dismutase, SOD; and reduced glutathione, GSH). Moreover, following perfusion-fixation of the brain, frontal lobes were processed by the haematoxylin and eosin (H&E) or Congo red technique. Our findings showed that in rats co-administered AlCl(3) and STZ (AD + DM rats), prefrontal levels of GSH reduced significantly (p < 0.05), while reductions in SOD and CAT were not significant (p > 0.05) compared with the controls. Moreover, in this model of AD with DM co-morbidity, extensive neuronal cell loss was observed in the prefrontal cortex, but Congophilic deposits were not present. The neurodegenerative lesions and antioxidant deficits characteristic of this AlCl(3) + STZ (AD + DM) rat model were more pronounced than similar lesions associated with mono-treatment with either STZ (DM) or AlCl(3) (AD) alone; and this makes the AlCl(3) + STZ model a suitable option for the study of neurodegenerative diseases (such as AD) with DM co-morbidity.


Assuntos
Compostos de Alumínio , Doença de Alzheimer/patologia , Antioxidantes/metabolismo , Cloretos , Diabetes Mellitus Experimental/patologia , Doenças Neurodegenerativas/induzido quimicamente , Doenças Neurodegenerativas/patologia , Córtex Pré-Frontal/patologia , Estreptozocina , Cloreto de Alumínio , Doença de Alzheimer/induzido quimicamente , Animais , Glicemia/metabolismo , Peso Corporal/efeitos dos fármacos , Diabetes Mellitus Experimental/complicações , Masculino , Doenças Neurodegenerativas/complicações , Córtex Pré-Frontal/ultraestrutura , Ratos , Ratos Wistar
18.
Morfologiia ; 148(6): 9-17, 2015.
Artigo em Russo | MEDLINE | ID: mdl-27141578

RESUMO

Recently, a large number of physiological studies on stress and hibernation had described an unusual morphological phenomenon of the rapid disappearance and reapperance of apical dendrites of pyramidal neurons of the hippocampus, prefrontal cortex and other parts of the brain. In this article an attempt is maid to explain this phenomenon on the basis of morphological analysis of natural elastic properties of neuroplasm and structural kinetics of partially preserved processes of the living isolated neurons. The neuroplasm displacement with its bidirectional flow was identified in the processes. A new physiological phenomenon is described--the isometric retraction of nerve cell processes, during which the neuroplasm fluxes were directed to the opposite sides, leading to abrupt thinning of middle parts of the processes and to a thickening of both ends. It is suggested that the extremely attenuated processes can reach the submicroscopic sizes, becoming invisible in the light microscope. The repeated reversible "disappearance" and "appearance" of the processes was demonstrated supravitally in the culture of neurons and of C-1300 neuroblastoma cells. Reduction of the diameter of the processes to a limit of their visibility was demonstrated by the example of their natural stretching. The same effect was observed in the areas between the reversible varicosities of the processes. These areas became extremely thin, and then invisible. Becoming thinner, the processes were capable of sharp extension. A review of the available literature and our own data allow to conclude that the phenomenon of the disappearance of the apical dendrites was due to their isometric retraction, which lead to the emergence of "invisible processes".


Assuntos
Dendritos/ultraestrutura , Hibernação/fisiologia , Hipocampo/ultraestrutura , Córtex Pré-Frontal/ultraestrutura , Células Piramidais/ultraestrutura , Estresse Fisiológico/fisiologia , Animais , Axônios/fisiologia , Axônios/ultraestrutura , Dendritos/fisiologia , Hipocampo/fisiologia , Microscopia Eletrônica , Modelos Neurológicos , Córtex Pré-Frontal/fisiologia , Células Piramidais/fisiologia
19.
Eur J Neurosci ; 40(8): 3202-14, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25131562

RESUMO

Endocannabinoids acting at the cannabinoid type 1 receptor (CB1R) are known to regulate attention, cognition and mood. Previous studies have shown that, in the rat medial prefrontal cortex (mPFC), CB1R agonists increase norepinephrine release, an effect that may be attributed, in part, to CB1Rs localised to noradrenergic axon terminals. The present study was aimed at further characterising functional interactions between CB1R and adrenergic receptor (AR) systems in the mPFC using in vitro intracellular electrophysiology and high-resolution neuroanatomical techniques. Whole-cell patch-clamp recordings of layer V/VI cortical pyramidal neurons in rats revealed that both acute and chronic treatment with the synthetic CB1R agonist WIN 55,212-2 blocked elevations in cortical pyramidal cell excitability and increases in input resistance evoked by the α2-adrenergic receptor (α2-AR) agonist clonidine, suggesting a desensitisation of α2-ARs. These CB1R-α2-AR interactions were further shown to be both action potential- and gamma-aminobutyric acid-independent. To better define sites of cannabinoid-AR interactions, we localised α2A-adrenergic receptors (α2A-ARs) in a genetically modified mouse that expressed a hemoagglutinin (HA) tag downstream of the α2A-AR promoter. Light and electron microscopy indicated that HA-α2A-AR was distributed in axon terminals and somatodendritic processes especially in layer V of the mPFC. Triple-labeling immunocytochemistry revealed that α2A-AR and CB1R were localised to processes that contained dopamine-ß-hydroxylase, a marker of norepinephrine. Furthermore, HA-α2A-AR was localised to processes that were directly apposed to CB1R. These findings suggest multiple sites of interaction between cortical cannabinoid-adrenergic systems that may contribute to understanding the effect of cannabinoids on executive functions and mood.


Assuntos
Córtex Pré-Frontal/fisiologia , Células Piramidais/fisiologia , Receptor CB1 de Canabinoide/fisiologia , Receptores Adrenérgicos alfa 2/fisiologia , Potenciais de Ação/efeitos dos fármacos , Agonistas de Receptores Adrenérgicos alfa 2/farmacologia , Animais , Benzoxazinas/farmacologia , Clonidina/farmacologia , Técnicas de Introdução de Genes , Masculino , Camundongos , Morfolinas/farmacologia , Naftalenos/farmacologia , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/ultraestrutura , Células Piramidais/efeitos dos fármacos , Células Piramidais/ultraestrutura , Ratos , Ratos Sprague-Dawley , Receptor CB1 de Canabinoide/agonistas , Receptores Adrenérgicos alfa 2/análise , Receptores Adrenérgicos alfa 2/genética
20.
BMC Neurosci ; 15: 105, 2014 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-25194917

RESUMO

BACKGROUND: Hypocretins/orexins (Hcrt/Ox) are hypothalamic neuropeptides involved in sleep-wakefulness regulation. Deficiency in Hcrt/Ox neurotransmission results in the sleep disorder narcolepsy, which is characterized by an inability to maintain wakefulness. The Hcrt/Ox neurons are maximally active during wakefulness and project widely to the ventral tegmental area (VTA). A dopamine-containing nucleus projecting extensively to the cerebral cortex, the VTA enhances wakefulness. In the present study, we used retrograde tracing from the medial prefrontal cortex (mPFC) to examine whether Hcrt1/OxA neurons target VTA neurons that could sustain behavioral wakefulness through their projections to mPFC. RESULTS: The retrograde tracer Fluorogold (FG) was injected into mPFC and, after an optimal survival period, sections through the VTA were processed for dual immunolabeling of anti-FG and either anti-Hcrt1/OxA or anti-TH antisera. Most VTA neurons projecting to the mPFC were located in the parabrachial nucleus of the ipsilateral VTA and were non-dopaminergic. Only axonal profiles showed Hcrt1/OxA-immunoreactivity in VTA. Hcrt1/OxA reactivity was observed in axonal boutons and many unmyelinated axons. The Hcrt1/OxA immunoreactivity was found filling axons but it was also observed in parts of the cytoplasm and dense-core vesicles. Hcrt1/OxA-labeled boutons frequently apposed FG-immunolabeled dendrites. However, Hcrt1/OxA-labeled boutons rarely established synapses, which, when they were established, were mainly asymmetric (excitatory-type), with either FG-labeled or unlabeled dendrites. CONCLUSIONS: Our results provide ultrastructural evidence that Hcrt1/OxA neurons may exert a direct synaptic influence on mesocortical neurons that would facilitate arousal and wakefulness. The paucity of synapses, however, suggest that the activity of VTA neurons with cortical projections might also be modulated by Hcrt1/OxA non-synaptic actions. In addition, Hcrt1/OxA could modulate the postsynaptic excitatory responses of VTA neurons with cortical projections to a co-released excitatory transmitter from Hcrt1/OxA axons. Our observation of Hcrt1/OxA targeting of mesocortical neurons supports Hcrt1/OxA wakefulness enhancement in the VTA and could help explain the characteristic hypersomnia present in narcoleptic patients.


Assuntos
Axônios/ultraestrutura , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neurônios/ultraestrutura , Neuropeptídeos/metabolismo , Córtex Pré-Frontal/ultraestrutura , Sinapses/ultraestrutura , Área Tegmentar Ventral/ultraestrutura , Animais , Axônios/metabolismo , Contagem de Células , Imunofluorescência , Imuno-Histoquímica , Masculino , Microscopia Confocal , Microscopia Eletrônica , Vias Neurais/metabolismo , Vias Neurais/ultraestrutura , Técnicas de Rastreamento Neuroanatômico , Neurônios/metabolismo , Orexinas , Córtex Pré-Frontal/metabolismo , Ratos Sprague-Dawley , Estilbamidinas , Sinapses/metabolismo , Tirosina 3-Mono-Oxigenase , Área Tegmentar Ventral/irrigação sanguínea , Área Tegmentar Ventral/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA