Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Neurotrauma ; 40(5-6): 561-577, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36262047

RESUMO

Chronic mental health problems are common among military veterans who sustained blast-related traumatic brain injuries. The reasons for this association remain unexplained. Male rats exposed to repetitive low-level blast overpressure (BOP) exposures exhibit chronic cognitive and post-traumatic stress disorder (PTSD)-related traits that develop in a delayed fashion. We examined blast-induced alterations on the transcriptome in four brain areas (anterior cortex, hippocampus, amygdala, and cerebellum) across the time frame over which the PTSD-related behavioral phenotype develops. When analyzed at 6 weeks or 12 months after blast exposure, relatively few differentially expressed genes (DEGs) were found. However, longitudinal analysis of amygdala, hippocampus, and anterior cortex between 6 weeks and 12 months revealed blast-specific DEG patterns. Six DEGs (hyaluronan and proteoglycan link protein 1 [Hapln1], glutamate metabotropic receptor 2 [Grm2], purinergic receptor P2y12 [P2ry12], C-C chemokine receptor type 5 [Ccr5], phenazine biosynthesis-like protein domain containing 1 [Pbld1], and cadherin related 23 [Cdh23]) were found altered in all three brain regions in blast-exposed animals. Pathway enrichment analysis using all DEGs or those uniquely changed revealed different transcription patterns in blast versus sham. In particular, the amygdala in blast-exposed animals had a unique set of enriched pathways related to stress responses, oxidative phosphorylation, and mitochondrial dysfunction. Upstream analysis implicated tumor necrosis factor (TNF)α signaling in blast-related effects in amygdala and anterior cortex. Eukaryotic initiating factor eIF4E (EIF4e), an upstream regulator of P2ry12 and Ccr5, was predicted to be activated in the amygdala. Quantitative polymerase chain reaction (qPCR) validated longitudinal changes in two TNFα regulated genes (cathepsin B [Ctsb], Hapln1), P2ry12, and Grm2. These studies have implications for understanding how blast injury damages the brain and implicates inflammation as a potential therapeutic target.


Assuntos
Traumatismos por Explosões , Lesões Encefálicas Traumáticas , Ratos , Masculino , Animais , Doenças Neuroinflamatórias , Fator de Iniciação 4E em Eucariotos/metabolismo , Explosões , Lesões Encefálicas Traumáticas/metabolismo , Traumatismos por Explosões/patologia , Tonsila do Cerebelo/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
2.
PLoS Genet ; 17(5): e1009506, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33956822

RESUMO

Identifying the molecular underpinnings of the neural specializations that underlie human cognitive and behavioral traits has long been of considerable interest. Much research on human-specific changes in gene expression and epigenetic marks has focused on the prefrontal cortex, a brain structure distinguished by its role in executive functions. The cerebellum shows expansion in great apes and is gaining increasing attention for its role in motor skills and cognitive processing, including language. However, relatively few molecular studies of the cerebellum in a comparative evolutionary context have been conducted. Here, we identify human-specific methylation in the lateral cerebellum relative to the dorsolateral prefrontal cortex, in a comparative study with chimpanzees (Pan troglodytes) and rhesus macaques (Macaca mulatta). Specifically, we profiled genome-wide methylation levels in the three species for each of the two brain structures and identified human-specific differentially methylated genomic regions unique to each structure. We further identified which differentially methylated regions (DMRs) overlap likely regulatory elements and determined whether associated genes show corresponding species differences in gene expression. We found greater human-specific methylation in the cerebellum than the dorsolateral prefrontal cortex, with differentially methylated regions overlapping genes involved in several conditions or processes relevant to human neurobiology, including synaptic plasticity, lipid metabolism, neuroinflammation and neurodegeneration, and neurodevelopment, including developmental disorders. Moreover, our results show some overlap with those of previous studies focused on the neocortex, indicating that such results may be common to multiple brain structures. These findings further our understanding of the cerebellum in human brain evolution.


Assuntos
Cerebelo/metabolismo , Metilação de DNA , Epigênese Genética , Proteínas ADAM , Animais , Autoantígenos , Proteínas de Transporte , Chade , Ilhas de CpG , Feminino , Regulação da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Macaca mulatta/genética , Masculino , Proteínas dos Microfilamentos , Proteínas do Tecido Nervoso , Pan troglodytes/genética , Fosfoinositídeo Fosfolipase C , Proteínas Serina-Treonina Quinases , Proteínas , Proteínas Associadas SAP90-PSD95 , Especificidade da Espécie , Sítio de Iniciação de Transcrição
3.
Front Aging ; 2: 726714, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35822016

RESUMO

Elephants are large-brained, social mammals with a long lifespan. Studies of elephants can provide insight into the aging process, which may be relevant to understanding diseases that affect elderly humans because of their shared characteristics that have arisen through independent evolution. Elephants become sexually mature at 12 to 14 years of age and are known to live into, and past, their 7th decade of life. Because of their relatively long lifespans, elephants may have evolved mechanisms to counter age-associated morbidities, such as cancer and cognitive decline. Elephants rely heavily on their memory, and engage in multiple levels of competitive and collaborative relationships because they live in a fission-fusion system. Female matrilineal relatives and dependent offspring form tight family units led by an older-aged matriarch, who serves as the primary repository for social and ecological knowledge in the herd. Similar to humans, elephants demonstrate a dependence on social bonds, memory, and cognition to navigate their environment, behaviors that might be associated with specializations of brain anatomy. Compared with other mammals, the elephant hippocampus is proportionally smaller, whereas the temporal lobe is disproportionately large and expands laterally. The elephant cerebellum is also relatively enlarged, and the cerebral cortex is highly convoluted with numerous gyral folds, more than in humans. Last, an interesting characteristic unique to elephants is the presence of at least 20 copies of the TP53 tumor suppressor gene. Humans have only a single copy. TP53 encodes for the p53 protein, which is known to orchestrate cellular response to DNA damage. The effects of these multiple copies of TP53 are still being investigated, but it may be to protect elephants against multiple age-related diseases. For these reasons, among others, studies of elephants would be highly informative for aging research. Elephants present an underappreciated opportunity to explore further common principles of aging in a large-brained mammal with extended longevity. Such research can contribute to contextualizing our knowledge of age-associated morbidities in humans.

4.
Oxid Med Cell Longev ; 2020: 9834742, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32733640

RESUMO

Copper, a transition metal with essential cellular functions, exerts neurotoxic effects when present in excess by promoting production of reactive oxygen species (ROS). The aim of the present study was to investigate potential benefits of flavonoid quercetin against copper-induced toxicity. Results obtained with MTT assay indicate that the effects of quercetin are determined by the severity of the toxic insult. In moderately injured P19 neuronal cells, concomitant treatment with 150 µM quercetin improved viability by preventing ROS formation, caspase-3 activation, and chromatin condensation. Western blot analysis revealed that quercetin reduced copper-induced increase in p53 upregulated modulator of apoptosis (PUMA) expression and promoted upregulation of nucleoside diphosphate kinase NME1. Levels of p53 and Bax proteins were not affected by both copper and quercetin. UO126 and wortmannin, inhibitors of ERK1/2 and PI3K/Akt signalling pathways, respectively, prevented neuroprotective effects of quercetin. In severely injured neurons, 30 µM quercetin exerted strong prooxidative action and exacerbated cytotoxic effects of copper, whereas 150 µM quercetin failed to affect neuronal survival. These results demonstrate the dual nature of quercetin action in copper-related neurodegeneration. Hence, they are relevant in the context of considering quercetin as a possible therapeutic for neuroprotection and imply that detailed pharmacological and toxicological studies must be carried out for natural compounds capable of acting both as antioxidants and prooxidants.


Assuntos
Antioxidantes/uso terapêutico , Cobre/efeitos adversos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Quercetina/uso terapêutico , Animais , Antioxidantes/farmacologia , Humanos , Sistema de Sinalização das MAP Quinases , Quercetina/farmacologia , Transdução de Sinais
5.
J Alzheimers Dis ; 75(3): 1029-1047, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32390629

RESUMO

BACKGROUND: Neuroinflammation plays an important role in Alzheimer's disease (AD). During this process, activated microglia release pro-inflammatory cytokines such as interleukin (IL)-1α, IL-1ß, IL-6, and tumor necrosis factor α (TNFα) that participate in neuron damage, but also anti-inflammatory cytokines (such as IL-10), which maintain homeostasis of immune response. Previous studies showed the association of IL-1α -889C/T (rs1800587), IL-1ß-1473G/C (rs1143623), IL-6 -174C/G (rs1800795), IL-10 -1082G/A (rs1800896), and TNFα -308A/G (rs1800629) polymorphisms with AD. OBJECTIVE: We aimed to investigate whether people with certain IL-1α, IL-1ß, IL-6, IL-10, and TNFα genotypes in these polymorphisms are more prone to develop AD-related pathology, reflected by pathological levels of cerebrospinal fluid (CSF) AD biomarkers including amyloid-ß1-42, total tau (t-tau), tau phosphorylated at Thr 181 (p-tau181), Ser 199 (p-tau199), and Thr 231 (p-tau231), and visinin-like protein 1 (VILIP-1). METHODS: The study included 115 AD patients, 53 patients with mild cognitive impairment, and 11 healthy controls. The polymorphisms were determined using real-time polymerase chain reaction. Levels of CSF biomarkers were determined by enzyme-linked immunosorbent assay. RESULTS: A significant increase in p-tau CSF levels was found in patients with the AA IL-10 -1082G/A and GG TNFα -308A/G genotypes, and in carriers of a G allele in IL-1ß -1473C/G and IL-6 -174C/G polymorphisms. t-tau levels were increased in carriers of a G allele in IL-1ß -1473C/G polymorphism. An increase in VILIP-1 levels was observed in patients with CG and GG IL-1ß -1473C/G, GC IL-6 -174C/G, and GG TNFα -308A/G genotype. CONCLUSION: These results suggest that persons carrying certain genotypes in IL10 (-1082G/A), IL1ß (1473C/G), IL6 (-174C/G), and TNFIα (-308A/G) could be more vulnerable to development of neuroinflammation, and consequently of AD.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Interleucina-10/genética , Interleucina-1beta/genética , Interleucina-6/genética , Fator de Necrose Tumoral alfa/genética , Idoso , Doença de Alzheimer/complicações , Peptídeos beta-Amiloides/líquido cefalorraquidiano , Biomarcadores/líquido cefalorraquidiano , Encefalite/complicações , Encefalite/genética , Feminino , Predisposição Genética para Doença , Genótipo , Humanos , Interleucina-10/líquido cefalorraquidiano , Interleucina-1beta/líquido cefalorraquidiano , Interleucina-6/líquido cefalorraquidiano , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Fator de Necrose Tumoral alfa/líquido cefalorraquidiano , Proteínas tau/líquido cefalorraquidiano
6.
Science ; 366(6464): 454-460, 2019 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-31624095

RESUMO

We present histological and molecular analyses of the developing human cerebellum from 30 days after conception to 9 months after birth. Differences in developmental patterns between humans and mice include spatiotemporal expansion of both ventricular and rhombic lip primary progenitor zones to include subventricular zones containing basal progenitors. The human rhombic lip persists longer through cerebellar development than in the mouse and undergoes morphological changes to form a progenitor pool in the posterior lobule, which is not seen in other organisms, not even in the nonhuman primate the macaque. Disruptions in human rhombic lip development are associated with posterior cerebellar vermis hypoplasia and Dandy-Walker malformation. The presence of these species-specific neural progenitor populations refines our insight into human cerebellar developmental disorders.


Assuntos
Cerebelo/embriologia , Cerebelo/crescimento & desenvolvimento , Células-Tronco/citologia , Animais , Síndrome de Dandy-Walker , Humanos , Camundongos , Malformações do Sistema Nervoso , Análise Espaço-Temporal , Especificidade da Espécie , Transcriptoma
7.
Cereb Cortex ; 29(12): 4932-4947, 2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-30877788

RESUMO

Preterm-born children suffer from neurological and behavioral disorders. Herein, we hypothesized that premature birth and non-maternal care of preterm newborns might disrupt neurobehavioral function, hippocampal dendritic arborization, and dendritic spine density. Additionally, we assessed whether 17ß-estradiol (E2) replacement or the TrkB receptor agonist, 7,8-dihydroxyflavone (DHF), would reverse compromised dendritic development and cognitive function in preterm newborns. These hypotheses were tested by comparing preterm (E28.5) rabbit kits cared and gavage-fed by laboratory personnel and term-kits reared and breast-fed by their mother doe at an equivalent postconceptional age. Neurobehavioral tests showed that both premature-birth and formula-feeding with non-maternal care led to increased anxiety behavior, poor social interaction, and lack of novelty preference compared with term-kits. Dendritic branching and number of total or mushroom dendritic spines were reduced in the CA1 field of preterm-kits compared with term controls. While CDC42 and Rac1/2/3 expression levels were lower, RhoA-activity was higher in preterm-kits compared with term controls. Both E2 and DHF treatment reversed prematurity-induced reduction in spine density, reduced total RhoA-GTPase levels, and enhanced cognitive function. Hence, prematurity and non-maternal care result in cognitive deficits, and reduced dendritic arbors and spines in CA1. E2 replacement or DHF treatment might reverse changes in dendritic spines and improve neurodevelopment in premature infants.


Assuntos
Cognição/fisiologia , Espinhas Dendríticas/patologia , Estradiol/farmacologia , Hipocampo/patologia , Nascimento Prematuro/fisiopatologia , Receptor trkB/agonistas , Animais , Cognição/efeitos dos fármacos , Espinhas Dendríticas/efeitos dos fármacos , Estrogênios/farmacologia , Feminino , Flavonas/farmacologia , Hipocampo/efeitos dos fármacos , Privação Materna , Gravidez , Nascimento Prematuro/patologia , Coelhos , Receptor trkB/efeitos dos fármacos
8.
J Neurosci Methods ; 319: 60-68, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30278184

RESUMO

BACKGROUND: Early stages of Alzheimer's disease (AD) are characterized by high phosphorylation of microtubule-associated protein tau, which may result from the downregulation of protein phosphatases. NEW METHOD: In order to model phosphatase downregulation and analyze its effect on tau aggregation in vitro, we treated neuroblastoma SH-SY5Y cells with okadaic acid (OA), a protein phosphatase inhibitor, and examined high molecular weight phospho-tau species. RESULTS AND COMPARISON WITH EXISTING METHODS: OA treatment led to the appearance of heat-stable protein species with apparent molecular weight around 100 kDa, which were immunoreactive to anti-tau antibodies against phosphorylated Ser202 and Ser396. As these high molecular weight tau-immunoreactive proteins (HMW-TIPs) corresponded to the predicted size of two tau monomers, we considered the possibility that they represent phosphorylation-induced tau oligomers. We attempted to dissociate HMW-TIPs by urea and guanidine, as well as by alkaline phosphatase treatment, but HMW-TIPs were stable under all conditions tested. These characteristics resemble properties of certain sodium dodecyl sulfate (SDS)-resistant tau oligomers from AD brains. The absence of HMW-TIPs detection by anti-total tau antibodies Tau46, CP27 and Tau13 may be a consequence of epitope masking and protein truncation. Alternatively, HMW-TIPs may represent previously unreported phosphoproteins cross-reacting with tau. CONCLUSIONS: Taken together, our data provide a novel characterization of an OA-based cell culture model in which OA induces the appearance of HMW-TIPs. These findings have implications for further studies of tau under the conditions of protein phosphatase downregulation, aiming to explain mechanisms involved in early events leading to AD.


Assuntos
Doença de Alzheimer/enzimologia , Inibidores Enzimáticos/administração & dosagem , Modelos Biológicos , Ácido Okadáico/administração & dosagem , Fosfoproteínas Fosfatases/metabolismo , Proteínas tau/metabolismo , Anticorpos , Linhagem Celular Tumoral , Humanos , Fosfoproteínas Fosfatases/antagonistas & inibidores , Fosforilação , Ensaio de Radioimunoprecipitação , Proteínas tau/imunologia
9.
J Comp Neurol ; 527(10): 1654-1674, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-30552685

RESUMO

Interlaminar astrocytes (ILA) in the cerebral cortex possess a soma in layer I and extend an interlaminar process that runs perpendicular to the pia into deeper cortical layers. We examined cerebral cortex from 46 species that encompassed most orders of therian mammalians, including 22 primate species. We described two distinct cell types with interlaminar processes that have been referred to as ILA, that we termed pial ILA and supial ILA. ILA subtypes differ in somatic morphology, position in layer I, and presence across species. We further described rudimentary ILA that have short GFAP+ processes that do not exit layer I, and "typical" ILA with longer GFAP+ processes that exit layer I. Pial ILA were present in all mammalian species analyzed, with typical ILA observed in Primates, Scandentia, Chiroptera, Carnivora, Artiodactyla, Hyracoidea, and Proboscidea. Subpial ILA were absent in Marsupialia, and typical subpial ILA were only found in Primate. We focused on the properties of pial ILA by investigating the molecular properties of pial ILA and confirming their astrocytic nature. We found that while the density of pial ILA somata only varied slightly, the complexity of ILA processes varied greatly across species. Primates, specifically bonobo, chimpanzee, orangutan, and human, exhibited pial ILA with the highest complexity. We showed that interlaminar processes contact neurons, pia, and capillaries, suggesting a potential role for ILA in the blood-brain barrier and facilitating communication among cortical neurons, astrocytes, capillaries, meninges, and cerebrospinal fluid.


Assuntos
Astrócitos/citologia , Córtex Cerebral/citologia , Animais , Mamíferos
10.
Prog Neurobiol ; 168: 104-127, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29733887

RESUMO

Alzheimer's disease (AD), the most common progressive neurodegenerative disorder, is characterized by severe cognitive decline and personality changes as a result of synaptic and neuronal loss. The defining clinicopathological hallmarks of the disease are deposits of amyloid precursor protein (APP)-derived amyloid-ß peptides (Aß) in the brain parenchyma, and intracellular aggregates of truncated and hyperphosphorylated tau protein in neurofibrillary tangles (NFT). At the cellular and molecular levels, many intertwined pathological mechanisms that relate Aß and tau pathology with a transcription factor p53 have been revealed. p53 is activated in response to various stressors that threaten genomic stability. Depending on damage severity, it promotes neuronal death or survival, predominantly via transcription-dependent mechanisms that affect expression of apoptosis-related target genes. Levels of p53 are enhanced in the AD brain and maintain sustained tau hyperphosphorylation, whereas intracellular Aß directly contributes to p53 pool and promotes downstream p53 effects. The review summarizes the role of p53 in neuronal function, discusses the interactions of p53, tau, and Aß in the normal brain and during the progression of AD pathology, and considers the impact of the most prominent hereditary risk factors of AD on p53/tau/Aß interactions. A better understanding of this intricate interplay would provide deeper insight into AD pathology and might offer some novel therapeutic targets for the improvement of treatment options. In this regard, drugs and natural compounds targeting the p53 pathway are of growing interest in neuroprotection as they may represent promising therapeutic approaches in the prevention of oxidative stress-dependent pathological processes underlying AD.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas tau/metabolismo , Animais , Humanos
11.
Acta Neuropathol Commun ; 5(1): 80, 2017 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-29126430

RESUMO

Blast-related traumatic brain injury (TBI) has been a common cause of injury in the recent conflicts in Iraq and Afghanistan. Blast waves can damage blood vessels, neurons, and glial cells within the brain. Acutely, depending on the blast energy, blast wave duration, and number of exposures, blast waves disrupt the blood-brain barrier, triggering microglial activation and neuroinflammation. Recently, there has been much interest in the role that ongoing neuroinflammation may play in the chronic effects of TBI. Here, we investigated whether chronic neuroinflammation is present in a rat model of repetitive low-energy blast exposure. Six weeks after three 74.5-kPa blast exposures, and in the absence of hemorrhage, no significant alteration in the level of microglia activation was found. At 6 weeks after blast exposure, plasma levels of fractalkine, interleukin-1ß, lipopolysaccharide-inducible CXC chemokine, macrophage inflammatory protein 1α, and vascular endothelial growth factor were decreased. However, no differences in cytokine levels were detected between blast-exposed and control rats at 40 weeks. In brain, isolated changes were seen in levels of selected cytokines at 6 weeks following blast exposure, but none of these changes was found in both hemispheres or at 40 weeks after blast exposure. Notably, one animal with a focal hemorrhagic tear showed chronic microglial activation around the lesion 16 weeks post-blast exposure. These findings suggest that focal hemorrhage can trigger chronic focal neuroinflammation following blast-induced TBI, but that in the absence of hemorrhage, chronic neuroinflammation is not a general feature of low-level blast injury.


Assuntos
Lesões Encefálicas Traumáticas/complicações , Citocinas/metabolismo , Encefalite/etiologia , Hemorragias Intracranianas/complicações , Hemorragias Intracranianas/etiologia , Animais , Lesões Encefálicas Traumáticas/etiologia , Lesões Encefálicas Traumáticas/patologia , Córtex Cerebral/patologia , Quimiocina CCL3/metabolismo , Quimiocina CCL5/metabolismo , Modelos Animais de Doenças , Feminino , Hipocampo/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/metabolismo , Microglia/patologia , Mutação/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
12.
Sci Rep ; 6: 30267, 2016 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-27443835

RESUMO

Presenilin 1 (Psen1) is important for vascular brain development and is known to influence cellular stress responses. To understand the role of Psen1 in endothelial stress responses, we investigated the effects of serum withdrawal on wild type (wt) and Psen1-/- embryonic brain endothelial cells. Serum starvation induced apoptosis in Psen1-/- cells but did not affect wt cells. PI3K/AKT signaling was reduced in serum-starved Psen1-/- cells, and this was associated with elevated levels of phospho-p38 consistent with decreased pro-survival AKT signaling in the absence of Psen1. Fibroblast growth factor (FGF1 and FGF2), but not vascular endothelial growth factor (VEGF) rescued Psen1-/- cells from serum starvation induced apoptosis. Inhibition of FGF signaling induced apoptosis in wt cells under serum withdrawal, while blocking γ-secretase activity had no effect. In the absence of serum, FGF2 immunoreactivity was distributed diffusely in cytoplasmic and nuclear vesicles of wt and Psen1-/- cells, as levels of FGF2 in nuclear and cytosolic fractions were not significantly different. Thus, sensitivity of Psen1-/- cells to serum starvation is not due to lack of FGF synthesis but likely to effects of Psen1 on FGF release onto the cell surface and impaired activation of the PI3K/AKT survival pathway.


Assuntos
Encéfalo/metabolismo , Fator 1 de Crescimento de Fibroblastos/genética , Fator 2 de Crescimento de Fibroblastos/genética , Presenilina-1/genética , Animais , Apoptose/genética , Encéfalo/citologia , Sobrevivência Celular , Meios de Cultura Livres de Soro , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Camundongos , Fosfatidilinositol 3-Quinases/genética , Presenilina-1/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/genética
13.
J Comput Neurosci ; 38(2): 263-83, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25527184

RESUMO

Layer 3 (L3) pyramidal neurons in the lateral prefrontal cortex (LPFC) of rhesus monkeys exhibit dendritic regression, spine loss and increased action potential (AP) firing rates during normal aging. The relationship between these structural and functional alterations, if any, is unknown. To address this issue, morphological and electrophysiological properties of L3 LPFC pyramidal neurons from young and aged rhesus monkeys were characterized using in vitro whole-cell patch-clamp recordings and high-resolution digital reconstruction of neurons. Consistent with our previous studies, aged neurons exhibited significantly reduced dendritic arbor length and spine density, as well as increased input resistance and firing rates. Computational models using the digital reconstructions with Hodgkin-Huxley and AMPA channels allowed us to assess relationships between demonstrated age-related changes and to predict physiological changes that have not yet been tested empirically. For example, the models predict that in both backpropagating APs and excitatory postsynaptic currents (EPSCs), attenuation is lower in aged versus young neurons. Importantly, when identical densities of passive parameters and voltage- and calcium-gated conductances were used in young and aged model neurons, neither input resistance nor firing rates differed between the two age groups. Tuning passive parameters for each model predicted significantly higher membrane resistance (R m ) in aged versus young neurons. This R m increase alone did not account for increased firing rates in aged models, but coupling these R m values with subtle differences in morphology and membrane capacitance did. The predicted differences in passive parameters (or parameters with similar effects) are mathematically plausible, but must be tested empirically.


Assuntos
Potenciais de Ação/fisiologia , Envelhecimento/fisiologia , Dendritos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Modelos Neurológicos , Células Piramidais/fisiologia , Animais , Eletrofisiologia/métodos , Processamento de Imagem Assistida por Computador/métodos , Macaca mulatta , Técnicas de Patch-Clamp/métodos , Córtex Pré-Frontal/citologia , Receptores de AMPA/fisiologia , Canais de Sódio Disparados por Voltagem/fisiologia
14.
Acta Neuropathol Commun ; 2: 67, 2014 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-24938728

RESUMO

BACKGROUND: Blast-related traumatic brain injury (TBI) is a common cause of injury in the military operations in Iraq and Afghanistan. How the primary blast wave affects the brain is not well understood. The aim of the present study was to examine whether blast exposure affects the cerebral vasculature in a rodent model. We analyzed the brains of rats exposed to single or multiple (three) 74.5 kPa blast exposures, conditions that mimic a mild TBI. Rats were sacrificed 24 hours or between 6 and 10 months after exposure. Blast-induced cerebral vascular pathology was examined by a combination of light microscopy, immunohistochemistry, and electron microscopy. RESULTS: We describe a selective vascular pathology that is present acutely at 24 hours after injury. The vascular pathology is found at the margins of focal shear-related injuries that, as we previously showed, typically follow the patterns of penetrating cortical vessels. However, changes in the microvasculature extend beyond the margins of such lesions. Electron microscopy revealed that microvascular pathology is found in regions of the brain with an otherwise normal neuropil. This initial injury leads to chronic changes in the microvasculature that are still evident many months after the initial blast exposure. CONCLUSIONS: These studies suggest that vascular pathology may be a central mechanism in the induction of chronic blast-related injury.


Assuntos
Traumatismos por Explosões/complicações , Lesões Encefálicas/etiologia , Lesões Encefálicas/patologia , Córtex Cerebral/patologia , Hemorragia Cerebral/etiologia , Vasculite do Sistema Nervoso Central/etiologia , Animais , Córtex Cerebral/metabolismo , Córtex Cerebral/ultraestrutura , Hemorragia Cerebral/patologia , Modelos Animais de Doenças , Proteína Glial Fibrilar Ácida/metabolismo , Gliose/etiologia , Masculino , Microscopia Eletrônica , Microvasos/metabolismo , Microvasos/patologia , Microvasos/ultraestrutura , Ratos , Ratos Long-Evans , Fatores de Tempo , Vasculite do Sistema Nervoso Central/patologia
15.
J Neurooncol ; 117(2): 269-77, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24549750

RESUMO

The insular cortex is located deep within the Sylvian fissure between multi-functional and structurally-compressed cerebral structures, and has been suggested to play an important role in both basic sensorimotor and complex social-emotional functions. Such structural and functional complexity presents a challenge for neurosurgeons to remove tumors within the insula safely. It has therefore not yet been documented how neurosurgical resection of insular gliomas would impact social-emotional functions. In this study, we examined empathy, a high-level social-emotional function, in four patients with localized insular gliomas pre- and post-operatively. The patients completed an empathy-for others pain task in which they viewed another person's hand or foot in painful or non-painful situations and made judgments about either pain (explicit empathy) or laterality of the hand or foot (implicit empathy). They also completed questionnaires assessing general emotional processing and personality. Deficits in both explicit and implicit empathetic pain processing were found in patients before the operations. However, the operations significantly improved their empathetic ability after surgery, accompanied by unchanged personality traits. These results confirmed previous findings that the insula plays a critical role for empathetic pain perception. Importantly, the current results suggest that surgical resection is not only a suitable treatment for insular gliomas for clinical consideration, but also effective in improving high-level functions such as empathetic pain perception.


Assuntos
Neoplasias Encefálicas/cirurgia , Córtex Cerebral/cirurgia , Empatia , Glioma/cirurgia , Procedimentos Neurocirúrgicos/efeitos adversos , Adulto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Inquéritos e Questionários
16.
J Comp Neurol ; 522(10): 2319-35, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24415002

RESUMO

Alzheimer's disease (AD) is a complex and slowly progressing dementing disorder that results in neuronal and synaptic loss, deposition in brain of aberrantly folded proteins, and impairment of spatial and episodic memory. Most studies of mouse models of AD have employed analyses of cognitive status and assessment of amyloid burden, gliosis, and molecular pathology during disease progression. Here we sought to understand the behavioral, cellular, ultrastructural, and molecular changes that occur at a pathological stage equivalent to the early stages of human AD. We studied the TgCRND8 mouse, a model of aggressive AD amyloidosis, at an early stage of plaque pathology (3 months of age) in comparison to their wildtype littermates and assessed changes in cognition, neuron and spine structure, and expression of synaptic glutamate receptor proteins. We found that, at this age, TgCRND8 mice display substantial plaque deposition in the neocortex and hippocampus and impairment on cued and contextual memory tasks. Of particular interest, we also observed a significant decrease in the number of neurons in the hippocampus. Furthermore, analysis of CA1 neurons revealed significant changes in apical and basal dendritic spine types, as well as altered expression of GluN1 and GluA2 receptors. This change in molecular architecture within the hippocampus may reflect a rising representation of inherently less stable thin spine populations, which can cause cognitive decline. These changes, taken together with toxic insults from amyloid-ß protein, may underlie the observed neuronal loss.


Assuntos
Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Medo , Transtornos da Memória/patologia , Transtornos da Memória/fisiopatologia , Plasticidade Neuronal/fisiologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Amiloidose/metabolismo , Amiloidose/patologia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Contagem de Células , Sinais (Psicologia) , Dendritos/metabolismo , Dendritos/patologia , Espinhas Dendríticas/metabolismo , Espinhas Dendríticas/patologia , Modelos Animais de Doenças , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Masculino , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Placa Amiloide/metabolismo , Placa Amiloide/patologia
17.
BMC Med ; 10: 127, 2012 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-23098093

RESUMO

Since the first description of the case of Auguste Deter, presented in Tübingen in 1906 by Alois Alzheimer, there has been an exponential increase in our knowledge of the neuropathological, cellular, and molecular foundation of Alzheimer's disease (AD). The concept of AD pathogenesis has evolved from a static, binary view discriminating cognitive normality from dementia, towards a dynamic view that considers AD pathology as a long-lasting morbid process that takes place progressively over years, or even decades, before the first symptoms become apparent, and thus operating in a continuum between the two aforementioned extreme states. Several biomarkers have been proposed to predict AD-related cognitive decline, initially in cases with mild cognitive impairment, and more recently in cognitively intact individuals. These early markers define at-risk individuals thought to be in the preclinical phase of AD. However, the clinical relevance of this preclinical phase remains controversial. The fate of such individuals, who are cognitively intact, but positive for some early AD biomarkers, is currently uncertain at best. In this report, we advocate the point of view that although most of these preclinical cases will evolve to clinically overt AD, some appear to have efficient compensatory mechanisms and virtually never develop dementia. We critically review the currently available early AD markers, discuss their clinical relevance, and propose a novel classification of preclinical AD, designating these non-progressing cases as 'stable asymptomatic cerebral amyloidosis'.


Assuntos
Doença de Alzheimer/diagnóstico , Doença de Alzheimer/patologia , Doenças Assintomáticas , Biomarcadores , Angiopatia Amiloide Cerebral/diagnóstico , Demência/diagnóstico , Humanos , Medição de Risco
18.
J Neurosci ; 32(40): 13644-60, 2012 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-23035077

RESUMO

Whole-cell patch-clamp recordings and high-resolution 3D morphometric analyses of layer 3 pyramidal neurons in in vitro slices of monkey primary visual cortex (V1) and dorsolateral granular prefrontal cortex (dlPFC) revealed that neurons in these two brain areas possess highly distinctive structural and functional properties. Area V1 pyramidal neurons are much smaller than dlPFC neurons, with significantly less extensive dendritic arbors and far fewer dendritic spines. Relative to dlPFC neurons, V1 neurons have a significantly higher input resistance, depolarized resting membrane potential, and higher action potential (AP) firing rates. Most V1 neurons exhibit both phasic and regular-spiking tonic AP firing patterns, while dlPFC neurons exhibit only tonic firing. Spontaneous postsynaptic currents are lower in amplitude and have faster kinetics in V1 than in dlPFC neurons, but are no different in frequency. Three-dimensional reconstructions of V1 and dlPFC neurons were incorporated into computational models containing Hodgkin-Huxley and AMPA receptor and GABA(A) receptor gated channels. Morphology alone largely accounted for observed passive physiological properties, but led to AP firing rates that differed more than observed empirically, and to synaptic responses that opposed empirical results. Accordingly, modeling predicts that active channel conductances differ between V1 and dlPFC neurons. The unique features of V1 and dlPFC neurons are likely fundamental determinants of area-specific network behavior. The compact electrotonic arbor and increased excitability of V1 neurons support the rapid signal integration required for early processing of visual information. The greater connectivity and dendritic complexity of dlPFC neurons likely support higher level cognitive functions including working memory and planning.


Assuntos
Neurônios/fisiologia , Córtex Pré-Frontal/fisiologia , Células Piramidais/fisiologia , Córtex Visual/fisiologia , Potenciais de Ação , Animais , Espinhas Dendríticas/fisiologia , Espinhas Dendríticas/ultraestrutura , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Técnicas In Vitro , Potenciais Pós-Sinápticos Inibidores/fisiologia , Macaca mulatta , Masculino , Microscopia Confocal , Modelos Neurológicos , Neurônios/ultraestrutura , Especificidade de Órgãos , Técnicas de Patch-Clamp , Córtex Pré-Frontal/citologia , Transmissão Sináptica , Córtex Visual/citologia
19.
Dement Geriatr Cogn Disord ; 33(6): 385-92, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22814030

RESUMO

AIM: The aim of this study was standardization and validation of the Mini-Mental State Examination (MMSE) in the general Croatian aging population. METHODS: Three-hundred and forty-four participants underwent the MMSE test, 217 cognitively healthy subjects without neurological and psychiatric disorders and 127 patients with mild cognitive impairment (MCI) or dementia. RESULTS: The optimal cutoff point for screening of the general Croatian population (cognitively healthy vs. MCI and dementia) is 26/27; in the Croatian population aged ≥65 years, the cutoff point is 24/25, whereas for screening of highly educated persons (≥14 years of education) aged ≥65 years a higher cutoff point should be used (26/27). CONCLUSIONS: MMSE results when standardized and validated in a certain population might better contribute to recognition of the individuals at risk that should be directed to dementia outpatient clinics.


Assuntos
Disfunção Cognitiva/diagnóstico , Demência/diagnóstico , Entrevista Psiquiátrica Padronizada/estatística & dados numéricos , Idoso , Doença de Alzheimer/diagnóstico , Estudos de Casos e Controles , Croácia , Escolaridade , Feminino , Humanos , Masculino , Programas de Rastreamento , Pessoa de Meia-Idade , Psicometria/instrumentação , Reprodutibilidade dos Testes
20.
Cereb Cortex ; 20(11): 2560-7, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20139149

RESUMO

We have recently reported in male rats that medial prefrontal cortex (mPFC) neurons that project to the basolateral nucleus of the amygdala (BLA) are resilient to stress-induced dendritic remodeling. The present study investigated whether this also occurs in female rats. This pathway was identified using the retrograde tracer Fast Blue injected into the BLA of ovariectomized female rats with estrogen replacement (OVX + E) and without (OVX + veh). Animals were exposed for 10 days either to 2-h immobilization stress or to home cage rest, after which layer III mPFC neurons that were either retrogradely labeled by Fast Blue or unlabeled were filled with Lucifer Yellow and analyzed for apical dendritic length and spine density. No dendritic remodeling occurred in unlabeled neurons from OVX + veh or OVX + E animals. In BLA-projecting neurons, however, stress had no effect on length in OVX + veh animals, but stressed OVX + E females showed greater dendritic length than controls at intermediate branches. Stress also caused an increase in spine density in all neurons in OVX + veh animals and a spine density increase in BLA-projecting neurons in OVX + E females. Estrogen also increased spine density on BLA-projecting neurons in unstressed animals. These data demonstrate both independent effects of estrogen on pyramidal cell morphology and effects that are interactive with stress, with the BLA-projecting neurons being sensitive to both kinds of effects.


Assuntos
Tonsila do Cerebelo/fisiopatologia , Estrogênios/farmacologia , Estresse Psicológico/metabolismo , Tonsila do Cerebelo/citologia , Tonsila do Cerebelo/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Estrogênios/fisiologia , Feminino , Vias Neurais/citologia , Vias Neurais/efeitos dos fármacos , Vias Neurais/fisiopatologia , Córtex Pré-Frontal/citologia , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/fisiopatologia , Ratos , Ratos Sprague-Dawley , Estresse Psicológico/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA