Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Cell ; 137(5): 961-71, 2009 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-19490899

RESUMO

It has been proposed that two amino acid substitutions in the transcription factor FOXP2 have been positively selected during human evolution due to effects on aspects of speech and language. Here, we introduce these substitutions into the endogenous Foxp2 gene of mice. Although these mice are generally healthy, they have qualitatively different ultrasonic vocalizations, decreased exploratory behavior and decreased dopamine concentrations in the brain suggesting that the humanized Foxp2 allele affects basal ganglia. In the striatum, a part of the basal ganglia affected in humans with a speech deficit due to a nonfunctional FOXP2 allele, we find that medium spiny neurons have increased dendrite lengths and increased synaptic plasticity. Since mice carrying one nonfunctional Foxp2 allele show opposite effects, this suggests that alterations in cortico-basal ganglia circuits might have been important for the evolution of speech and language in humans.


Assuntos
Substituição de Aminoácidos , Gânglios da Base/metabolismo , Evolução Biológica , Fatores de Transcrição Forkhead/metabolismo , Vocalização Animal , Animais , Dendritos/metabolismo , Dopamina/metabolismo , Expressão Gênica , Heterozigoto , Humanos , Idioma , Depressão Sináptica de Longo Prazo , Camundongos , Vias Neurais , Plasticidade Neuronal , Fala
2.
Acta Neuropathol ; 130(4): 501-10, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26298468

RESUMO

Alzheimer's disease (AD) is a neurodegenerative disorder characterized by fibrillary aggregates of Aß peptide and tau protein. The distribution of these pathological hallmarks throughout the brain is not random; it follows a predictive pattern that is used for pathological staging. However, most etiopathogenetic concepts, irrespective of whether they focus on Aß or tau pathology, leave a key question unanswered: what is the explanation for the different vulnerabilities of brain regions in AD? The pattern of regional progression of neurofibrillary degeneration in AD to some extent inversely recapitulates ontogenetic and phylogenetic brain development. Accordingly, degeneration preferentially affects brain areas that have recently been acquired or restructured during anthropoid evolution, which means that the involvement of a neurodevelopmental mechanism is highly likely. Since evolutionary expansion of the neocortex is based on a substantial extension of the mitotic activity of progenitor cells, we propose a conceptual link between neurogenesis in anthropoid primates and a higher risk of accumulating mitotic errors that give rise to genomic aberrations commonly referred to as DNA content variation (DCV). If increased rates of DCV make neurons more vulnerable to AD-related pathology, one might expect there to be a higher rate of DCV in areas that are affected very early during the course of AD, as compared to areas which are hardly affected or are affected only during the most advanced stages. Therefore, in the present study, we comparatively analyzed the DCV in five different cortical areas that are affected during the early stage (entorhinal cortex), the intermediate stage (temporal, frontal, and parietal association cortex), and the late stage (primary sensory occipital cortex) of AD in both normal elderly subjects and AD patients. On average, we observed about 10 % neuronal mosaic DCV in the normal elderly and a two- to threefold increase in DCV in AD patients. We were able to demonstrate, moreover, that the neuronal DCV in the cerebral cortex of the normal elderly as well as the increased neuronal DCV in AD patients are not randomly distributed but instead show systematic regional differences which correspond to differences in vulnerability. These findings provide additional evidence that mosaic genomic heterogeneity may play a key role in AD pathology.


Assuntos
Envelhecimento/genética , Envelhecimento/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Córtex Cerebral/metabolismo , Neurônios/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/patologia , Córtex Cerebral/patologia , DNA/metabolismo , Feminino , Variação Genética , Humanos , Masculino , Pessoa de Meia-Idade , Neurônios/patologia
3.
Neuropathol Appl Neurobiol ; 40(7): 815-32, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24964035

RESUMO

AIMS: Neurodegeneration in Alzheimer's disease (AD) is characterized by pathological protein aggregates and inadequate activation of cell cycle regulating proteins. Recently, Smad proteins were identified to control the expression of AD relevant proteins such as APP, CDK4 and CDK inhibitors, both critical regulators of cell cycle activation. This might indicate a central role for Smads in AD pathology where they show a substantial deficiency and disturbed subcellular distribution in neurones. Still, the mechanisms driving relocation and decrease of neuronal Smad in AD are not well understood. However, Pin1, a peptidyl-prolyl-cis/trans-isomerase, which allows isomerization of tau protein, was recently identified also controlling the fate of Smads. Here we analyse a possible role of Pin1 for Smad disturbances in AD. METHODS: Multiple immunofluorescence labelling and confocal laser-scanning microscopy were performed to examine the localization of Smad and Pin1 in human control and AD hippocampi. Ectopic Pin1 expression in neuronal cell cultures combined with Western blot analysis and immunoprecipitation allowed studying Smad level and subcellular distribution. Luciferase reporter assays, electromobility shift, RNAi-technique and qRT-PCR revealed a potential transcriptional impact of Smad on Pin1 promoter. RESULTS: We report on a colocalization of phosphorylated Smad in AD with Pin1. Pin1 does not only affect Smad phosphorylation and stability but also regulates subcellular localization of Smad2 and supports its binding to phosphorylated tau protein. Smads, in turn, exert a negative feed-back regulation on Pin1. CONCLUSION: Our data suggest both Smad proteins and Pin1 to be elements of a vicious circle with potential pathogenetic significance in AD.


Assuntos
Doença de Alzheimer/metabolismo , Peptidilprolil Isomerase/metabolismo , Proteínas Smad/metabolismo , Proteínas tau/metabolismo , Idoso , Idoso de 80 Anos ou mais , Feminino , Hipocampo/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Peptidilprolil Isomerase de Interação com NIMA , Fosforilação , Proteólise
4.
Am J Pathol ; 177(1): 15-20, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20472889

RESUMO

Aneuploidy, an abnormal number of copies of a genomic region, might be a significant source for neuronal complexity, intercellular diversity, and evolution. Genomic instability associated with aneuploidy, however, can also lead to developmental abnormalities and decreased cellular fitness. Here we show that neurons with a more-than-diploid content of DNA are increased in preclinical stages of Alzheimer's disease (AD) and are selectively affected by cell death during progression of the disease. Present findings show that neuronal hyperploidy in AD is associated with a decreased viability. Hyperploidy of neurons thus represents a direct molecular signature of cells prone to death in AD and indicates that a failure of neuronal differentiation is a critical pathogenetic event in AD.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Morte Celular/genética , Neurônios/patologia , Neurônios/fisiologia , Poliploidia , Idoso , Idoso de 80 Anos ou mais , Animais , Encéfalo/citologia , Encéfalo/patologia , Encéfalo/fisiologia , Diferenciação Celular/fisiologia , Humanos , Neurônios/citologia
5.
J Neural Transm (Vienna) ; 118(5): 765-72, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21533606

RESUMO

Cerebral amyloid angiopathy is manifested as accumulation of amyloid ß (Aß) peptide in the wall of meningeal and cerebral arteries, arterioles and capillaries and is frequently found postmortem in sporadic Alzheimer's disease (sAD) patients. It is difficult to assess when and how cerebral amyloid angiopathy develops and progresses in humans in vivo, which is why animal AD models are used. Streptozotocin-intracerebroventricularly (STZ-icv) treated rats have been recently proposed as the model of sAD which develops insulin resistant brain state preceding Aß pathology development. Vascular Aß deposits in the brain of STZ-icv-treated rats (3 months old at the time of icv treatment) were visualized by Thioflavine-S staining, Congo red staining and Aß immunohistochemistry. Thioflavine-S and Congo red staining revealed diffuse congophilic deposits in the wall of meningeal and cortical blood vessels both 6 and 9 months after the STZ-icv treatment. Preliminary Aß1-42 and Aß1-16 immunohistochemistry experiments showed positive staining in blood vessels 3 and 9 months after the STZ-icv treatment, respectively. Results suggest that cerebral amyloid angiopathy observed 6 and 9 months after the STZ-icv treatment seems to be a continuation and progression of the amyloid pathology observed already 3 months following the STZ-icv treatment in this non-transgenic sAD animal model.


Assuntos
Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/complicações , Antibióticos Antineoplásicos/toxicidade , Angiopatia Amiloide Cerebral/etiologia , Estreptozocina/toxicidade , Peptídeos beta-Amiloides/metabolismo , Animais , Benzotiazóis , Angiopatia Amiloide Cerebral/patologia , Modelos Animais de Doenças , Progressão da Doença , Estudos Longitudinais , Masculino , Fragmentos de Peptídeos/metabolismo , Ratos , Ratos Wistar , Coloração e Rotulagem/métodos , Tiazóis
6.
Cell Rep ; 35(4): 109035, 2021 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-33910019

RESUMO

Several neurodegenerative diseases present Tau accumulation as the main pathological marker. Tau post-translational modifications such as phosphorylation and acetylation are increased in neurodegeneration. Here, we show that Tau hyper-acetylation at residue 174 increases its own nuclear presence and is the result of DNA damage signaling or the lack of SIRT6, both causative of neurodegeneration. Tau-K174ac is deacetylated in the nucleus by SIRT6. However, lack of SIRT6 or chronic DNA damage results in nuclear Tau-K174ac accumulation. Once there, it induces global changes in gene expression, affecting protein translation, synthesis, and energy production. Concomitantly, Alzheimer's disease (AD) case subjects show increased nucleolin and a decrease in SIRT6 levels. AD case subjects present increased levels of nuclear Tau, particularly Tau-K174ac. Our results suggest that increased Tau-K174ac in AD case subjects is the result of DNA damage signaling and SIRT6 depletion. We propose that Tau-K174ac toxicity is due to its increased stability, nuclear accumulation, and nucleolar dysfunction.


Assuntos
Doença de Alzheimer/genética , Biossíntese de Proteínas/genética , Sirtuínas/metabolismo , Proteínas tau/metabolismo , Humanos , Sirtuínas/genética
7.
Neuroscience ; 449: 202-213, 2020 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-32926955

RESUMO

Recently, circular RNAs (circRNAs) have been revealed to be an important non-coding element of the transcriptome. The brain contains the most abundant and widespread expression of circRNA. There are also indications that the circular transcriptome undergoes dynamic changes as a result of brain ageing. Diminished cognitive function with increased age reflects the dysregulation of synaptic function and ineffective neurotransmission through alterations of the synaptic proteome. Here, we present changes in the circular transcriptome in ageing synapses using a mouse model. Specifically, we observed an accumulation of uniquely expressed circular transcripts in the synaptosomes of aged mice compared to young mice. Individual circRNA expression patterns were characterized by an increased abundance in the synaptosomes of young or aged mice, whereas the opposite expression was observed for the parental gene linear transcripts. These changes in expression were validated by RT-qPCR. We provide the first comprehensive survey of the circular transcriptome in mammalian synapses, thereby paving the way for future studies. Additionally, we present 16 genes that express solely circRNAs, without linear RNAs co-expression, exclusively in young and aged synaptosomes, suggesting a synaptic gene network that functions along canonical splicing activity.


Assuntos
Sinaptossomos , Transcriptoma , Animais , Encéfalo , Redes Reguladoras de Genes , RNA/genética , RNA Circular
8.
Biochim Biophys Acta ; 1772(4): 413-21, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17236750

RESUMO

Higher cerebral functions are based upon a dynamic organization of neuronal networks. To form synaptic connections and to continuously re-shape them in a process of ongoing structural adaptation, neurons must permanently withdraw from the cell cycle. In other words, synaptic plasticity can only occur on the expense of the ability to proliferate. Previously, we have put forward a hypothesis, coined "Dr. Jekyll and Mr. Hyde concept" that differentiated neurons after having withdrawn from the cell cycle are able to use those molecular mechanisms primarily developed to control proliferation alternatively to control synaptic plasticity [T. Arendt, Synaptic plasticity and cell cycle activation in neurons are alternative effector pathways The Dr. Jekyll and Mr. Hyde Theory of Alzheimer's disease or The yin and yang of Neuroplasticity. Progr. Neurobiol. 71 (2003) 83-248]. The existence of these alternative effector pathways within a neuron might put it on the risk to erroneously convert signals derived from plastic synaptic changes into cell cycle activation which subsequently leads to cell death. Here we add further evidence to this hypothesis demonstrating a tight association of the origin recognition complex (ORC) with neurofibrillar pathology in AD. The ORC is a critical "guard" of DNA replication and point of convergence of numerous functionally redundant signaling pathways involved in cell cycle progression and transcriptional silencing of apoptotic programmes. ORC subunits in the mammalian brain and their homologes in Drosophila, however, have further been implicated in the regulation of structural neuronal plasticity and cognitive function. We propose that the abnormal subcellular distribution and segregation of ORC proteins in AD might compromise their physiological function in gene silencing and plasticity. This might result in cell cycle activation, DNA-replication and de-repression of apoptotic programmes. ORC subunits might, thus, provide a direct molecular link between synaptic plasticity, DNA replication and cell death.


Assuntos
Doença de Alzheimer/patologia , Encéfalo/patologia , Ciclo Celular , Plasticidade Neuronal/fisiologia , Sinapses/patologia , Doença de Alzheimer/fisiopatologia , Encéfalo/fisiopatologia , Humanos , Modelos Neurológicos , Complexo de Reconhecimento de Origem/análise , Sinapses/fisiologia
9.
Neurobiol Aging ; 51: 19-30, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28033505

RESUMO

Impaired cholinergic neurotransmission associated with cognitive dysfunction occurs in various mental disorders of different etiologies including Alzheimer's disease and postalcoholic dementia and others. To address the question whether there exists a common endophenotype with a defined genetic and/or epigenetic signature causing mental dysfunction in these disorders, we investigated 2 generations of offspring born to alcohol-treated mothers. Here, we show that memory impairment and reduced synthesis of acetylcholine occurs in both F1 (exposed to ethanol in utero) and F2 generation (never been exposed to ethanol). Effects in the F2 generation are most likely consequences of transgenerationally transmitted epigenetic modifications in stem cells induced by alcohol. This clearly documents the role of ancestral history of drug abuse on the brain development of subsequent generations. The results further suggest an epigenetic trait for an anticholinergic endophenotype associated with cognitive dysfunction which might be relevant to our understanding of mental impairment in neurodegenerative disorders such as Alzheimer's disease and related disorders.


Assuntos
Acetilcolina/biossíntese , Transtornos Cognitivos/genética , Endofenótipos , Etanol/efeitos adversos , Transtornos da Memória/genética , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/genética , Doença de Alzheimer/genética , Animais , Epigênese Genética , Feminino , Transtornos do Espectro Alcoólico Fetal/genética , Troca Materno-Fetal , Gravidez , Ratos
10.
Brain Res ; 1099(1): 189-98, 2006 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-16781686

RESUMO

The extracellular deposition of amyloid-beta peptide (Abeta) in brain parenchyma is one of the characteristic features of Alzheimer's disease and is suggested to induce reactive and degenerative changes in neuronal cell bodies, axons and dendritic processes. In particular, within and in close proximity to amyloid plaques, distinctive morphological alterations have been observed, including changes in neurite trajectory and decreases in dendritic diameter and in spine density. Apart from these plaque-associated focal aberrations, little is known regarding modifications of the global dendritic morphology including the detailed and comparative quantitative analysis of apical and basal arbors. The objective of the present study was to investigate the effects of amyloid plaque deposition and elevated soluble Abeta on neuronal morphology in mutant human amyloid precursor protein (hAPP) transgenic mice (line Tg2576; [K. Hsiao, P. Chapman, S. Nilsen, C. Eckman, Y. Harigaya, S. Younkin, F. Yang, G. Cole, Correlative memory deficits, Abeta elevation, and amyloid plaques in transgenic mice, Science 274 (1996) 99-102]). Retrogradelly labeled callosal-projecting pyramidal cells in the primary somatosensory cortex were three-dimensionally analyzed. Although basal dendrites remained unaffected, analysis of apical trees revealed a number of unambiguous morphological changes. Thus, in TG2576 mice, the apical arbors were shortened in total length and less branched. Furthermore, the diameter of proximal dendritic segments was increased whereas that of distal segments was reduced. Analysis of spine numbers and distribution on basal and apical trees demonstrated a significant reduction in spine densities along the whole course of dendrites. The findings suggest that Abeta-related pathology induces morphological aberrations in basal and apical arbors to different degrees which are unrelated to direct plaque-associated changes.


Assuntos
Precursor de Proteína beta-Amiloide/genética , Dendritos , Mutação , Córtex Somatossensorial/patologia , Animais , Dendritos/classificação , Dendritos/genética , Dendritos/patologia , Espinhas Dendríticas/patologia , Espinhas Dendríticas/ultraestrutura , Humanos , Camundongos , Camundongos Transgênicos , Córtex Somatossensorial/ultraestrutura
11.
Int J Dev Neurosci ; 24(2-3): 133-40, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16384682

RESUMO

The current study addresses the morphoregulatory effects of human amyloid precursor protein expression on neocortical pyramidal cells in vivo. For this purpose, a transgenic mouse line was used that expresses wild-type human amyloid precursor protein (APP) at levels similar to endogenous mouse APP. This strain does not develop Alzheimer's disease-related pathology which allowed to study effects of APP or APP cleavage products but excluded the influence of amyloid deposits. Commissural projecting pyramidal neurons of layers II/III within the primary somatosensory cortex were retrogradely labelled by injection of biotinylated dextran amine into the corpus callosum. In transgenic mice, computer-aided morphometric analysis revealed an increase in the surface area of proximal and intermediate basal dendritic segments resulting from an enlarged diameter. On the other hand, the length of the same segments was reduced. Both basal and apical dendrites were characterized by a higher dendritic density within the proximal and intermediate fields. Although the total spatial extension of basal and apical dendrites remained unchanged, a moderate withdrawal of arbors is suggested. The results implicate a physiological function for APP in regulatory mechanisms of neuronal morphogenesis.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Dendritos/metabolismo , Expressão Gênica/fisiologia , Neocórtex/citologia , Células Piramidais/citologia , Precursor de Proteína beta-Amiloide/genética , Animais , Western Blotting/métodos , Tamanho Celular , Humanos , Camundongos , Camundongos Transgênicos , Fenótipo
12.
Int J Dev Neurosci ; 24(2-3): 177-86, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16386398

RESUMO

In the brain the expression of transforming growth factor beta1 (TGF-beta1) is involved both in neuroprotective and neurodegenerative processes. Recently, we have established a transgenic mouse model with inducible neuron-specific expression of TGF-beta1 based on the tetracycline-regulated gene expression system. A long-term expression of TGF-beta1 results in persisting perivascular thioflavin-positive depositions, which did not disappear even though the transgene synthesis was repressed completely by administration of doxycycline. Formation and composition of these depositions are hardly elucidated. The aim of this study was to identify TGF-beta1 responding genes potentially participating in forming these depositions. To address this problem we have compared the cortical mRNA expression pattern of TGF-beta1 expressing mice with mice impeded to express the transgenic protein using oligonucleotide microarray analysis. Differential gene expression was further characterized by quantitative real-time reverse transcription-polymerase chain reaction including animals, where the long-lasting TGF-beta1 expression was repressed. While no change of amyloid precursor protein RNA expression level was detected, various genes strongly involved in calcium homeostasis, tissue mineralization or vascular calcification were identified differentially expressed. It is suggested, that these genes might contribute to the perivascular depositions in the TGF-beta1 expressing mice.


Assuntos
Córtex Cerebral/patologia , Regulação da Expressão Gênica/genética , Doenças Neurodegenerativas/metabolismo , Neurônios/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Células Cultivadas , Córtex Cerebral/metabolismo , Embrião de Mamíferos , Expressão Gênica/genética , Hibridização Genética/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Análise em Microsséries/métodos , Biologia Molecular/métodos , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/fisiopatologia , Neurônios/patologia , Análise de Sequência com Séries de Oligonucleotídeos/métodos , RNA Mensageiro/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta1
13.
Acta Neuropathol Commun ; 3: 10, 2015 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-25853173

RESUMO

Alzheimer's disease (AD) is a degenerative disorder where the distribution of pathology throughout the brain is not random but follows a predictive pattern used for pathological staging. While the involvement of defined functional systems is fairly well established for more advanced stages, the initial sites of degeneration are still ill defined. The prevailing concept suggests an origin within the transentorhinal and entorhinal cortex (EC) from where pathology spreads to other areas. Still, this concept has been challenged recently suggesting a potential origin of degeneration in nonthalamic subcortical nuclei giving rise to cortical innervation such as locus coeruleus (LC) and nucleus basalis of Meynert (NbM). To contribute to the identification of the early site of degeneration, here, we address the question whether cortical or subcortical degeneration occurs more early and develops more quickly during progression of AD. To this end, we stereologically assessed neurone counts in the NbM, LC and EC layer-II in the same AD patients ranging from preclinical stages to severe dementia. In all three areas, neurone loss becomes detectable already at preclinical stages and is clearly manifest at prodromal AD/MCI. At more advanced AD, cell loss is most pronounced in the NbM > LC > layer-II EC. During early AD, however, the extent of cell loss is fairly balanced between all three areas without clear indications for a preference of one area. We can thus not rule out that there is more than one way of spreading from its site of origin or that degeneration even occurs independently at several sites in parallel.


Assuntos
Doença de Alzheimer/patologia , Núcleo Basal de Meynert/patologia , Córtex Entorrinal/citologia , Locus Cerúleo/citologia , Degeneração Neural/patologia , Neurônios/patologia , Idoso , Idoso de 80 Anos ou mais , Progressão da Doença , Córtex Entorrinal/patologia , Feminino , Humanos , Locus Cerúleo/patologia , Masculino , Fatores de Tempo
14.
J Neuropathol Exp Neurol ; 61(8): 678-88, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12152783

RESUMO

Regular use of non-steroidal anti-inflammatory drugs (NSAIDs) seems to reduce the progression of several diseases, including colon cancer, lung cancer, breast cancer and Alzheimer disease (AD). Several studies have shown that NSAIDs can modulate cell cycle progression, especially in the G0/G1 phase. The main target of most NSAIDs is the enzyme cyclo-oxygenase (COX), which occurs in 2 isoforms, COX-1 and COX-2. In AD and non-demented control brain, COX-2 is expressed in neuronal cells. In this study the expression of COX-2, cyclin D1, and cyclin E was investigated at the immunohistochemical level in AD and non-demented control temporal cortex. COX-2, cyclin D1, and cyclin E expression was detected in pyramidal neurons in both AD and control patients. The number of COX-2-immunoreactive neurons positively correlated with the number of cyclin E- and cyclin D1-immunoreactive neurons. Moreover, immunostaining of sequential tissue sections and double immunofluorescence labeling revealed co-expression of COX-2 and cyclin D1 and E in neuronal cells. In addition, an inverse correlation was observed between the neuronal expression of COX-2 and cyclin E and the Braak score for amyloid beta deposits. Our findings suggest a relationship between the neuronal expression of COX-2 and cell cycle markers, which may be involved early in AD pathology.


Assuntos
Doença de Alzheimer/metabolismo , Ciclina D1/metabolismo , Ciclina E/metabolismo , Isoenzimas/metabolismo , Prostaglandina-Endoperóxido Sintases/metabolismo , Células Piramidais/metabolismo , Lobo Temporal/metabolismo , Idoso , Idoso de 80 Anos ou mais , Ciclo-Oxigenase 2 , Feminino , Humanos , Masculino , Proteínas de Membrana , Pessoa de Meia-Idade , Neurônios/metabolismo , Valores de Referência , Distribuição Tecidual
15.
Aging Cell ; 11(4): 628-33, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22510449

RESUMO

The human brain has been proposed to represent a genetic mosaic, containing a small but constant number of neurons with an amount of DNA exceeding the diploid level that appear to be generated through various chromosome segregation defects initially. While a portion of these cells apparently die during development, neurons with abnormal chromosomal copy number have been identified in the mature brain. This genomic alteration might to lead to chromosomal instability affecting neuronal viability and could thus contribute to age-related mental disorders. Changes in the frequency of neurons with such structural genomic variation in the adult and aging brain, however, are unknown. Here, we quantified the frequency of neurons with a more than diploid DNA content in the cerebral cortex of normal human brain and analyzed its changes between the fourth and ninth decades of life. We applied a protocol of slide-based cytometry optimized for DNA quantification of single identified neurons, which allowed to analyze the DNA content of about 500 000 neurons for each brain. On average, 11.5% of cortical neurons showed DNA content above the diploid level. The frequency of neurons with this genomic alteration was highest at younger age and declined with age. Our results indicate that the genomic variation associated with DNA content exceeding the diploid level might compromise viability of these neurons in the aging brain and might thus contribute to susceptibilities for age-related CNS disorders. Alternatively, a potential selection bias of "healthy aging brains" needs to be considered, assuming that DNA content variation above a certain threshold associates with Alzheimer's disease.


Assuntos
Envelhecimento/metabolismo , Envelhecimento/patologia , Encéfalo/citologia , Encéfalo/metabolismo , DNA/metabolismo , Neurônios/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Envelhecimento/genética , Aneuploidia , Contagem de Células , Senescência Celular/genética , Senescência Celular/fisiologia , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Instabilidade Cromossômica , DNA/genética , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
16.
Neurobiol Aging ; 33(12): 2827-40, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22418736

RESUMO

Alzheimer's disease (AD) is a neurodegenerative disorder characterized by deregulation of neuronal cell cycle and differentiation control eventually resulting in cell death. During brain development, neuronal differentiation is regulated by Smad proteins, which are elements of the canonical transforming growth factor ß (TGF-ß) signaling pathway, linking receptor activation to gene expression. In the normal adult brain, Smad proteins are constitutively phosphorylated and predominantly localized in neuronal nuclei. Under neurodegenerative conditions such as AD, the subcellular localization of their phosphorylated forms is heavily disturbed, raising the question of whether a nuclear Smad deficiency in neurons might contribute to a loss of neuronal differentiation control and subsequent cell cycle re-entry. Here, we show by luciferase reporter assays, electromobility shift, and RNA interference (RNAi) technique a direct binding of Smad proteins to the CDK4 promoter inducing transcriptional inhibition of cell cycle-dependent kinase 4 (Cdk4). Mimicking the neuronal deficiency of Smad proteins observed in AD in cell culture by RNAi results in elevation of Cdk4 and retardation of neurite outgrowth. The results identify Smad proteins as direct transcriptional regulators of Cdk4 and add further evidence to a Smad-dependent deregulation of Cdk4 in AD, giving rise to neuronal dedifferentiation and cell death.


Assuntos
Doença de Alzheimer/patologia , Encéfalo/metabolismo , Quinase 4 Dependente de Ciclina/metabolismo , Regulação da Expressão Gênica/fisiologia , Biossíntese de Proteínas/fisiologia , Proteínas Smad/metabolismo , Idoso , Idoso de 80 Anos ou mais , Análise de Variância , Animais , Bucladesina/farmacologia , Morte Celular/efeitos dos fármacos , Morte Celular/genética , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Quinase 4 Dependente de Ciclina/genética , Ensaio de Desvio de Mobilidade Eletroforética , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Luciferases/genética , Luciferases/metabolismo , Masculino , Camundongos , Neuritos/efeitos dos fármacos , Neuroblastoma/patologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Regiões Promotoras Genéticas/fisiologia , Ligação Proteica/fisiologia , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , RNA Interferente Pequeno/farmacologia , Proteínas Smad/genética , Transfecção
17.
PLoS One ; 6(1): e14530, 2011 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-21267079

RESUMO

Abnormal phosphorylation and aggregation of tau protein are hallmarks of a variety of neurological disorders, including Alzheimer's disease (AD). Increased tau phosphorylation is assumed to represent an early event in pathogenesis and a pivotal aspect for aggregation and formation of neurofibrillary tangles. However, the regulation of tau phosphorylation in vivo and the causes for its increased stage of phosphorylation in AD are still not well understood, a fact that is primarily based on the lack of adequate animal models. Recently we described the reversible formation of highly phosphorylated tau protein in hibernating European ground squirrels. Hence, mammalian hibernation represents a model system very well suited to study molecular mechanisms of both tau phosphorylation and dephosphorylation under in vivo physiological conditions. Here, we analysed the extent and kinetics of hibernation-state dependent tau phosphorylation in various brain regions of three species of hibernating mammals: arctic ground squirrels, Syrian hamsters and black bears. Overall, tau protein was highly phosphorylated in torpor states and phosphorylation levels decreased after arousal in all species. Differences between brain regions, hibernation-states and phosphosites were observed with respect to degree and kinetics of tau phosphorylation. Furthermore, we tested the phosphate net turnover of tau protein to analyse potential alterations in kinase and/or phosphatase activities during hibernation. Our results demonstrate that the hibernation-state dependent phosphorylation of tau protein is specifically regulated but involves, in addition, passive, temperature driven regulatory mechanisms. By determining the activity-state profile for key enzymes of tau phosphorylation we could identify kinases potentially involved in the differentially regulated, reversible tau phosphorylation that occurs during hibernation. We show that in black bears hibernation is associated with conformational changes of highly phosphorylated tau protein that are typically related to neuropathological alterations. The particular hibernation characteristics of black bears with a continuous torpor period and an only slightly decreased body temperature, therefore, potentially reflects the limitations of this adaptive reaction pattern and, thus, might indicate a transitional state of a physiological process.


Assuntos
Metabolismo Basal , Hibernação , Proteínas tau/metabolismo , Animais , Temperatura Corporal , Encéfalo/metabolismo , Cricetinae , Cinética , Mesocricetus , Fosforilação , Sciuridae , Ursidae
18.
Int J Dev Neurosci ; 27(5): 501-10, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19375497

RESUMO

Smad2 and Smad3 are central molecules of the TGFbeta and activin receptor complex mediated intracellular signaling pathway. They function as important transcription factors playing essential roles in brain development. Interestingly they are also known to be involved in the pathogenesis of various neurological disorders (including Alzheimer's disease). Due to structural differences in the N-terminal Mad homology domain 1, Smad2 and Smad3 differ in their ability to bind DNA directly. A splice form of Smad2 lacking exon3, Smad2(Deltaexon3), assumes features of Smad3, in that it can directly bind to DNA resulting in a functional hybrid of Smad2 and Smad3 properties. There is very little information available on the expression of Smad2 isoforms in the brain. We report here that Smad2(Deltaexon3) is the most abundant of the two Smad2 isoforms in mouse brain and that Smad expression pattern alters during development and aging. Neuronal expression of Smad2(Deltaexon3) was confirmed by a single-cell PCR approach. Moreover, Smad2(Deltaexon3) predominates in the nuclear fraction of neurons, suggesting special function during brain differentiation. Our data indicate that there may be a specific role for Smad2(Deltaexon3) in neurons.


Assuntos
Envelhecimento/genética , Encéfalo/fisiologia , Neurônios/fisiologia , Proteína Smad2/genética , Processamento Alternativo , Animais , Encéfalo/crescimento & desenvolvimento , Células Cultivadas , Expressão Gênica , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Isoformas de Proteínas , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Frações Subcelulares/fisiologia
19.
Eur J Neurosci ; 22(1): 50-64, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16029195

RESUMO

Various chronic neurological diseases are associated with increased expression of transforming growth factor-beta1 (TGF-beta1) in the brain. TGF-beta1 has both neuroprotective and neurodegenerative functions, depending on conditions such as duration and the local and temporal pattern of its expression. Previous transgenic approaches did not enable control for these dynamic aspects. To overcome these limitations, we established a transgenic mouse model with inducible neuron-specific expression of TGF-beta1 based on the tetracycline-regulated gene expression system. TGF-beta1 expression was restricted to the brain where it was particularly pronounced in the neocortex, hippocampus and striatum. Transgene expression was highly sensitive to the presence of doxycycline and completely silenced within 6 days after doxycycline application. After long-term expression, perivascular thioflavin-positive depositions, formed by amyloid fibrils, developed. These depositions persisted even after prolonged silencing of the transgene, indicating an irreversible process. Similarly, strong perivascular apolipoprotein E (ApoE) depositions were found after TGF-beta1 expression and these remained despite TGF-beta1 removal. These in vivo observations suggests that the continuous presence of TGF-beta1 as initial trigger is not necessary for the persistence and development of chronic lesions. Neuroprotective effects were observed after short-term expression of TGF-beta1. Death of striatal neurons induced by 3-nitropropionic acid was markedly reduced after induced TGF-beta1 expression.


Assuntos
Encéfalo/metabolismo , Degeneração Neural/metabolismo , Doenças Neurodegenerativas/metabolismo , Neurônios/metabolismo , Fator de Crescimento Transformador beta/biossíntese , Animais , Antibacterianos/farmacologia , Apolipoproteínas E/genética , Benzotiazóis , Encéfalo/patologia , Encéfalo/fisiopatologia , Doxiciclina/farmacologia , Regulação da Expressão Gênica/genética , Inativação Gênica/fisiologia , Camundongos , Camundongos Transgênicos , Biologia Molecular/métodos , Degeneração Neural/induzido quimicamente , Degeneração Neural/genética , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/fisiopatologia , Neurônios/patologia , Fármacos Neuroprotetores/metabolismo , Neurotoxinas/farmacologia , Nitrocompostos , Placa Amiloide/genética , Placa Amiloide/metabolismo , Propionatos/farmacologia , Tiazóis/metabolismo , Transfecção/métodos , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta1 , Transgenes/genética
20.
Exp Neurol ; 188(2): 309-15, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15246831

RESUMO

A specialized form of extracellular matrix (ECM) termed perineuronal nets (PNs) consisting of large aggregating chondroitin sulfate proteoglycans (CSPGs), with hyaluronan and tenascin as main components, surrounds subpopulations of neurons. The glycosaminoglycan components of perineuronal nets form highly charged structures in the direct microenvironment of neurons and thus might be involved in local ion homeostasis. The polyanionic character suggests that perineuronal nets also potentially contribute to reduce the local oxidative potential in the neuronal microenvironment by scavenging and binding redox-active iron, thus providing some neuroprotection to net-associated neurons. Here, we show that neurons ensheathed by a perineuronal net in the human cerebral cortex are less frequently affected by lipofuscin accumulation than neurons without a net both in normal-aged brain and Alzheimer's disease (AD). As lipofuscin is an intralysosomal pigment composed of cross-linked proteins and lipids generated by iron-catalyzed oxidative processes, the present results suggest a neuroprotective function of perineuronal nets against oxidative stress, potentially involved in neurodegeneration.


Assuntos
Doença de Alzheimer/metabolismo , Matriz Extracelular/metabolismo , Lipofuscina/metabolismo , Neurônios/metabolismo , Estresse Oxidativo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/patologia , Contagem de Células , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Matriz Extracelular/patologia , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Neurônios/patologia , Células Piramidais/metabolismo , Células Piramidais/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA