Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Mol Psychiatry ; 26(10): 5790-5796, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-32203153

RESUMO

Alzheimer's disease (AD) is a neurodegenerative disorder of unknown cause with complex genetic and environmental traits. While AD is extremely prevalent in human elderly, it hardly occurs in non-primate mammals and even non-human-primates develop only an incomplete form of the disease. This specificity of AD to human clearly implies a phylogenetic aspect. Still, the evolutionary dimension of AD pathomechanism remains difficult to prove and has not been established so far. To analyze the evolutionary age and dynamics of AD-associated-genes, we established the AD-associated genome-wide RNA-profile comprising both protein-coding and non-protein-coding transcripts. We than applied a systematic analysis on the conservation of splice-sites as a measure of gene-structure based on multiple alignments across vertebrates of homologs of AD-associated-genes. Here, we show that nearly all AD-associated-genes are evolutionarily old and did not originate later in evolution than not-AD-associated-genes. However, the gene-structures of loci, that exhibit AD-associated changes in their expression, evolve faster than the genome at large. While protein-coding-loci exhibit an enhanced rate of small changes in gene structure, non-coding loci show even much larger changes. The accelerated evolution of AD-associated-genes indicates a more rapid functional adaptation of these genes. In particular AD-associated non-coding-genes play an important, as yet largely unexplored, role in AD. This phylogenetic trait indicates that recent adaptive evolution of human brain is causally involved in basic principles of neurodegeneration. It highlights the necessity for a paradigmatic change of our disease-concepts and to reconsider the appropriateness of current animal-models to develop disease-modifying strategies that can be translated to human.


Assuntos
Doença de Alzheimer , Doença de Alzheimer/genética , Animais , Encéfalo , Genoma , Estudo de Associação Genômica Ampla , Filogenia
2.
Genomics ; 112(2): 1162-1166, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31255695

RESUMO

Circular RNAs (circRNAs) have recently attracted significant interest in the realm of science and the evolution of species. Given the lack of information available on circRNAs due to various barriers related to sequencing techniques and bioinformatics tools, little regarding their function is known. It has been predicted that circRNAs contribute to gene expression regulation, but aside from a few specific cases, this contention has yet to be proven. Although the role of circRNAs in evolution remains elusive, from the few studies that have shown circRNA conservation in mammalian species, tissue specificity in brain regions, and the abundance of circRNAs in the brains of various species, the concept is becoming more likely with much gravitas. The proposed functional role of circRNAs being gene regulators is of great interest and would provide a basis to further understand not only the functional capabilities of organisms, but also the evolution of mammalian species.


Assuntos
Encéfalo/metabolismo , Evolução Molecular , RNA Circular/genética , Transcriptoma , Animais , Sequência Conservada , Humanos , RNA Circular/química , RNA Circular/metabolismo
3.
Genomics ; 112(6): 5265-5274, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32966858

RESUMO

The circular transcriptome of human glial cells is an area of neuroscience that has not been thoroughly elucidated. Circular RNAs (circRNAs) have the potential to facilitate the understanding of vast, complex and unknown mechanisms derived from the human transcriptome, including elements of the human brain that are not known and the evolution of the human brain, the complexities of which are not well understood. Moreover, the glial cells have been determined to contribute to human brain evolution. This study presents the first comprehensive analysis of the human brain glia circRNA transcriptome, that is, astrocytes, microglia and oligodendrocytes. After stringent criteria applied to the detection of circRNAs, it was found that the circular transcriptomes of these glia are unique from one another, and hence might be indicative of distinct roles for circRNAs within the brain. This study found 265, 239 and 442 circRNAs comprising the unique circular transcriptome of astrocytes, microglia and oligodendrocytes, respectively. The most abundant circRNAs in these glial cell types are expressed by parent genes co-expressing linear RNAs in low abundance, suggesting spliceosome activity favorable to the back-splicing mechanism instead of canonical splicing activity.


Assuntos
Neuroglia/metabolismo , RNA Circular/metabolismo , Astrócitos/metabolismo , Ontologia Genética , Humanos , Microglia/metabolismo , Oligodendroglia/metabolismo , RNA-Seq , Transcriptoma
4.
J Neurochem ; 143(4): 396-408, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28397252

RESUMO

Alzheimer's disease (AD) is a neurodegenerative disorder of higher age that specifically occurs in human. Its clinical phase, characterized by a decline in physiological, psychological, and social functioning, is preceded by a long clinically silent phase of at least several decades that might perhaps even start very early in life. Overall, key functional abilities in AD patients decline in reverse order of the development of these abilities during normal childhood and adolescence. Early symptoms of AD, thus, typically affect mental functions that have been acquired only during very recent hominid evolution and as such are specific to human. Neurofibrillar degeneration, a typical neuropathological lesion of the disease and one of the most robust pathological correlates of cognitive impairment, is rarely seen in non-primate mammals and even non-human primates hardly develop a pathology comparable to those seen in AD patients. Neurofibrillar degeneration is not randomly distributed throughout the AD brain. It preferentially affects brain areas that become increasingly predominant during the evolutionary process of encephalization. During progression of the disease, it affects cortical areas in a stereotypic sequence that inversely recapitulates ontogenetic brain development. The specific distribution of cortical pathology in AD, moreover, appears to be determined by the modular organization of the cerebral cortex which basically is a structural reflection of its ontogeny. Here, we summarize recent evidence that phylogenetic and ontogenetic dimensions of brain structure and function provide the key to our understanding of AD. More recent molecular biological studies of the potential pathogenetic role of a genomic mosaic in the brains of patients with AD might even provide arguments for a developmental origin of AD. This article is part of a series "Beyond Amyloid".


Assuntos
Doença de Alzheimer/diagnóstico , Encéfalo/patologia , Progressão da Doença , Transtornos do Neurodesenvolvimento/diagnóstico , Doença de Alzheimer/fisiopatologia , Animais , Encéfalo/fisiopatologia , Humanos , Transtornos do Neurodesenvolvimento/fisiopatologia , Filogenia
5.
J Cell Mol Med ; 18(7): 1444-59, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24779367

RESUMO

Members of the transforming growth factor (TGF)-ß family govern a wide range of mechanisms in brain development and in the adult, in particular neuronal/glial differentiation and survival, but also cell cycle regulation and neural stem cell maintenance. This clearly created some discrepancies in the field with some studies favouring neuronal differentiation/survival of progenitors and others favouring cell cycle exit and neural stem cell quiescence/maintenance. Here, we provide a unifying hypothesis claiming that through its regulation of neural progenitor cell (NPC) proliferation, TGF-ß signalling might be responsible for (i) maintaining stem cells in a quiescent stage, and (ii) promoting survival of newly generated neurons and their functional differentiation. Therefore, we performed a detailed histological analysis of TGF-ß1 signalling in the hippocampal neural stem cell niche of a transgenic mouse that was previously generated to express TGF-ß1 under a tetracycline regulatable Ca-Calmodulin kinase promoter. We also analysed NPC proliferation, quiescence, neuronal survival and differentiation in relation to elevated levels of TGF-ß1 in vitro and in vivo conditions. Finally, we performed a gene expression profiling to identify the targets of TGF-ß1 signalling in adult NPCs. The results demonstrate that TGF-ß1 promotes stem cell quiescence on one side, but also neuronal survival on the other side. Thus, considering the elevated levels of TGF-ß1 in ageing and neurodegenerative diseases, TGF-ß1 signalling presents a molecular target for future interventions in such conditions.


Assuntos
Diferenciação Celular , Hipocampo/citologia , Neurogênese/fisiologia , Neurônios/citologia , Nicho de Células-Tronco , Células-Tronco/citologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Biomarcadores/metabolismo , Western Blotting , Proliferação de Células , Células Cultivadas , Senescência Celular , Proteína Duplacortina , Eletrofisiologia , Feminino , Perfilação da Expressão Gênica , Hipocampo/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , Ratos , Ratos Endogâmicos F344 , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/metabolismo , Fator de Crescimento Transformador beta/genética
6.
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
7.
Acta Neuropathol Commun ; 10(1): 175, 2022 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-36451207

RESUMO

The possible role of somatic copy number variations (CNVs) in Alzheimer's disease (AD) aetiology has been controversial. Although cytogenetic studies suggested increased CNV loads in AD brains, a recent single-cell whole-genome sequencing (scWGS) experiment, studying frontal cortex brain samples, found no such evidence. Here we readdressed this issue using low-coverage scWGS on pyramidal neurons dissected via both laser capture microdissection (LCM) and fluorescence activated cell sorting (FACS) across five brain regions: entorhinal cortex, temporal cortex, hippocampal CA1, hippocampal CA3, and the cerebellum. Among reliably detected somatic CNVs identified in 1301 cells obtained from the brains of 13 AD patients and 7 healthy controls, deletions were more frequent compared to duplications. Interestingly, we observed slightly higher frequencies of CNV events in cells from AD compared to similar numbers of cells from controls (4.1% vs. 1.4%, or 0.9% vs. 0.7%, using different filtering approaches), although the differences were not statistically significant. On the technical aspects, we observed that LCM-isolated cells show higher within-cell read depth variation compared to cells isolated with FACS. To reduce within-cell read depth variation, we proposed a principal component analysis-based denoising approach that significantly improves signal-to-noise ratios. Lastly, we showed that LCM-isolated neurons in AD harbour slightly more read depth variability than neurons of controls, which might be related to the reported hyperploid profiles of some AD-affected neurons.


Assuntos
Doença de Alzheimer , Humanos , Doença de Alzheimer/genética , Variações do Número de Cópias de DNA , Neurônios , Córtex Entorrinal , Encéfalo
8.
Sci Rep ; 11(1): 20200, 2021 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-34642398

RESUMO

Alzheimer's disease (AD) is a progressive neurodegenerative disorder with typical neuropathological hallmarks, such as neuritic plaques and neurofibrillary tangles, preferentially found at layers III and V. The distribution of both hallmarks provides the basis for the staging of AD, following a hierarchical pattern throughout the cerebral cortex. To unravel the background of this layer-specific vulnerability, we evaluated differential gene expression of supragranular and infragranular layers and subcortical white matter in both healthy controls and AD patients. We identified AD-associated layer-specific differences involving protein-coding and non-coding sequences, most of those present in the subcortical white matter, thus indicating a critical role for long axons and oligodendrocytes in AD pathomechanism. In addition, GO analysis identified networks containing synaptic vesicle transport, vesicle exocytosis and regulation of neurotransmitter levels. Numerous AD-associated layer-specifically expressed genes were previously reported to undergo layer-specific switches in recent hominid brain evolution between layers V and III, i.e., those layers that are most vulnerable to AD pathology. Against the background of our previous finding of accelerated evolution of AD-specific gene expression, here we suggest a critical role in AD pathomechanism for this phylogenetic layer-specific adaptation of gene expression, which is most prominently seen in the white matter compartment.


Assuntos
Doença de Alzheimer/genética , Perfilação da Expressão Gênica/métodos , Redes Reguladoras de Genes , RNA não Traduzido/genética , Substância Branca/química , Idoso , Idoso de 80 Anos ou mais , Axônios/química , Estudos de Casos e Controles , Evolução Molecular , Feminino , Regulação da Expressão Gênica , Humanos , Masculino , Oligodendroglia/química , Especificidade de Órgãos , Análise de Sequência de RNA
9.
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
10.
Hippocampus ; 20(8): 971-9, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19714567

RESUMO

Besides its role in Alzheimer's disease, the amyloid precursor protein (APP) is implicated in several physiological functions in neuronal tissue such as cell survival, neurite outgrowth, synaptic formation, and neuronal plasticity. The present study analyzed effects of human wild-type APP (hAPP) overexpression on adult hippocampal neurogenesis in transgenic mice. Mice were housed under either standard or enriched conditions, the latter to boost neurogenetic activity. Different aspects of neurogenesis including proliferation, survival, and differentiation were assessed by employing the BrdU-incorporation method and, in parallel, immunohistochemistry for the neuronal and glial markers NeuN and S100b, respectively. Overexpression of hAPP caused a significant decrease in cell proliferation under standard housing conditions. The relative increase in the proliferation rate following housing in enriched environment was not different to that observed in wild-type mice. Overexpression of hAPP, on the other hand, promoted the survival of newly generated cells, but just under conditions of standard housing. Findings further suggest that overexpression of hAPP suppresses the phenotypic shift toward neuronal differentiation under conditions of enriched environment. In summary, the results reveal a dual effect of APP on adult hippocampal neurogenesis, comprising antiproliferative and prosurvival activities.


Assuntos
Precursor de Proteína beta-Amiloide/genética , Hipocampo/citologia , Neurogênese/genética , Análise de Variância , Animais , Animais Geneticamente Modificados , Bromodesoxiuridina/metabolismo , Diferenciação Celular/genética , Proliferação de Células , Sobrevivência Celular/genética , Meio Ambiente , Humanos , Fator de Crescimento Derivado de Plaquetas/genética
11.
Comp Hepatol ; 9: 8, 2010 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-20942944

RESUMO

BACKGROUND: Proliferation of oval cells, the bipotent precursor cells of the liver, requires impeded proliferation and loss of hepatocytes as well as a specific micro-environment, provided by adjacent sinusoidal cells of liver. Despite their immense importance for triggering the oval cell response, cells of hepatic sinusoids are rarely investigated. To elucidate the response of sinusoidal liver cells we have employed a choline-deficient, ethionine-supplemented (CDE) diet, a common method for inducing an oval cell response in rodent liver. We have utilised selected expression markers commonly used in the past for phenotypic discrimination of oval cells and sinusoidal cells: cytokeratin, E-cadherin and M2-pyruvate kinase for oval cells; and glial fibrillary acidic protein (GFAP) was used for hepatic stellate cells (HSCs). RESULTS: CDE diet leads to an activation of all cells of the hepatic sinusoid in the mouse liver. Beside oval cells, also HSCs and Kupffer cells proliferate. The entire fraction of proliferating cells in mouse liver as well as endothelial cells and cholangiocytes express M2-pyruvate kinase. Concomitantly, GFAP, long considered a unique marker of quiescent HSCs was upregulated in activated HSCs and expressed also in cholangiocytes and oval cells. CONCLUSIONS: Our results point to an important role of all types of sinusoidal cells in regeneration from CDE induced liver damage and call for utmost caution in using traditional marker for identifying specific cell types. Thus, M2-pyruvate kinase should no longer be used for estimating the oval cell response in mouse liver. CDE diet leads to activation of GFAP positive HSCs in the pericentral zone of liver lobulus. In the periportal zone the detection of GFAP in biliary cells and oval cells, calls other cell types as progenitors of hepatocytes into question under CDE diet conditions.

12.
Front Genet ; 11: 370, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32411177

RESUMO

Recent evidence indicates that genomic individuality of neurons, characterized by DNA-content variation, is a common if not universal phenomenon in the human brain that occurs naturally but can also show aberrancies that have been linked to the pathomechanism of Alzheimer's disease and related neurodegenerative disorders. Etiologically, this genomic mosaic has been suggested to arise from defects of cell cycle regulation that may occur either during brain development or in the mature brain after terminal differentiation of neurons. Here, we aim to draw attention towards another mechanism that can give rise to genomic individuality of neurons, with far-reaching consequences. This mechanism has its origin in the transcriptome rather than in replication defects of the genome, i.e., somatic gene recombination of RNA. We continue to develop the concept that somatic gene recombination of RNA provides a physiological process that, through integration of intronless mRNA/ncRNA into the genome, allows a particular functional state at the level of the individual neuron to be indexed. By insertion of defined RNAs in a somatic recombination process, the presence of specific mRNA transcripts within a definite temporal context can be "frozen" and can serve as an index that can be recalled at any later point in time. This allows information related to a specific neuronal state of differentiation and/or activity relevant to a memory trace to be fixed. We suggest that this process is used throughout the lifetime of each neuron and might have both advantageous and deleterious consequences.

13.
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
14.
Neurobiol Dis ; 35(2): 258-63, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19450683

RESUMO

Alzheimer's disease (AD) is a chronic neurodegenerative disorder associated with extracellular accumulation of Abeta peptide that derives from the amyloid precursor protein (APP). While amyloidogenic processing of APP has received most attention, the physiological function of APP and the sequelae of potentially impaired APP function are less understood. APP is a transmembrane glycoprotein being widely expressed in neurons in both central and peripheral nervous system. Its physiological function has been associated with neuronal survival, neurite outgrowth and neuronal plasticity. The aim of the present study was to determine whether FAD-linked mutations of APP, known to be associated with early onset of the disease, might impair its synaptotrophic function, potentially contributing to synaptic deficiencies seen in AD. We performed a quantitative electron microscopy study on synapses in well characterized expression-matched transgenic mice lines expressing either wildtype or FAD-mutated hAPP. Using serial electron microscopic sections, we comparatively analyzed by stereological methods the number and sizes of synaptic contacts and the number of synaptic vesicles in the neocortex. We could clearly show a synaptotrophic effect in mice overexpressing wildtype hAPP evidenced by a significant increase in the number of synapses and the number of vesicles per synapse. This effect was abolished when FAD-mutated APP(Sw,Ind) was expressed instead of wildtype APP. The present study demonstrates a synaptotrophic effect of APP which is lost in the presence of a FAD-mutation. This failure could either be due to a synaptotoxic effect of Abeta potentially counteracting the synaptotrophic effect of APP. Alternatively, the FAD-mutation might impair the physiological function of the extracellular domain of APP and its fragments which might be required for the synaptotrophic effect. This suggests that not only "too much Abeta" but also "too less functional intact APP" might be relevant for synaptic pathology and degeneration in AD.


Assuntos
Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/metabolismo , Neocórtex/metabolismo , Receptores de Superfície Celular/metabolismo , Sinapses/metabolismo , Vesículas Sinápticas/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Animais , Western Blotting , Humanos , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica , Mutação , Neocórtex/ultraestrutura , Neurônios/metabolismo , Neurônios/ultraestrutura , Nexinas de Proteases , Receptores de Superfície Celular/genética , Sinapses/ultraestrutura , Vesículas Sinápticas/ultraestrutura
15.
J Neurosci Res ; 87(8): 1813-22, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19170186

RESUMO

The physiological role of the amyloid precursor protein (APP) and its proteolytic fragments in the brain is associated with neuronal survival, neurite outgrowth, synaptic formation, and neuronal plasticity. However, malregulation of APP processing leads to disordered balance of fragments, which may results in opposite, degenerative neuronal effects. In the present study, we analyzed in vivo effects of the expression of wild-type or mutated human APP on afferent deprivation-induced changes of dendritic morphology. After vibrissectomy, expression of wild-type human APP prevented diameter shrinkage of dendritic segments as well as dendritic rarefaction of apical arbors. In contrast, mutant human APP expression exacerbated degenerative changes of deprived barrel neurons. Degradation of apical arbors was especially pronounced. Results demonstrate for the first time opposite effects of the expression of wild-type and mutated human APP on deprivation-induced dendritic restructuring in vivo.


Assuntos
Precursor de Proteína beta-Amiloide/genética , Transtornos Cognitivos/patologia , Dendritos/patologia , Degeneração Neural/patologia , Privação Sensorial/fisiologia , Córtex Somatossensorial/patologia , Vias Aferentes/fisiopatologia , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/metabolismo , Animais , Diferenciação Celular/genética , Transtornos Cognitivos/genética , Transtornos Cognitivos/fisiopatologia , Dendritos/metabolismo , Denervação , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Transgênicos , Degeneração Neural/genética , Degeneração Neural/fisiopatologia , Plasticidade Neuronal/genética , Células Receptoras Sensoriais/fisiologia , Córtex Somatossensorial/metabolismo , Córtex Somatossensorial/fisiopatologia , Nervo Trigêmeo/fisiopatologia , Vibrissas/lesões , Vibrissas/fisiologia
16.
Neuroscientist ; 24(3): 294-307, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-28770651

RESUMO

Multiple system atrophy (MSA) is a complex, multifactorial, debilitating neurodegenerative disease that is often misdiagnosed and misunderstood. MSA has two subclasses, MSA-P and MSA-C, defined by the dominance of parkinsonism or cerebellar dysfunction in the earlier stages of disease, coupled with dysautonomia. This distinction between subclasses becomes largely redundant as the disease progresses. Aggregation of α-synuclein is a clinical marker used to confirm MSA diagnoses, which can only be performed postmortem. Transcriptome profiling provides in-depth information about the diseased state and can contribute to further understanding of MSA, enabling easier and more rapid diagnosis as well as contributing to improving the quality of life of people with MSA. Currently, there is no method of diagnosing MSA with certainty, and there is no cure for this disease. This review provides an update on current advances in investigations of molecular pathology of MSA with particular focus on perturbation of individual gene expression and MSA transcriptome.


Assuntos
Atrofia de Múltiplos Sistemas/metabolismo , Transcriptoma , Animais , Humanos , Atrofia de Múltiplos Sistemas/genética , Atrofia de Múltiplos Sistemas/terapia
17.
Brain Pathol ; 28(4): 495-506, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-28557134

RESUMO

Hereditary cerebral hemorrhage with amyloidosis-Dutch type (HCHWA-D) is an early onset hereditary form of cerebral amyloid angiopathy (CAA) pathology, caused by the E22Q mutation in the amyloid ß (Aß) peptide. Transforming growth factor ß1 (TGFß1) is a key player in vascular fibrosis and in the formation of angiopathic vessels in transgenic mice. Therefore, we investigated whether the TGFß pathway is involved in HCHWA-D pathogenesis in human postmortem brain tissue from frontal and occipital lobes. Components of the TGFß pathway were analyzed with quantitative RT-PCR. TGFß1 and TGFß Receptor 2 (TGFBR2) gene expression levels were significantly increased in HCHWA-D in comparison to the controls, in both frontal and occipital lobes. TGFß-induced pro-fibrotic target genes were also upregulated. We further assessed pathway activation by detecting phospho-SMAD2/3 (pSMAD2/3), a direct TGFß down-stream signaling mediator, using immunohistochemistry. We found abnormal pSMAD2/3 granular deposits specifically on HCHWA-D angiopathic frontal and occipital vessels. We graded pSMAD2/3 accumulation in angiopathic vessels and found a positive correlation with the CAA load independent of the brain area. We also observed pSMAD2/3 granules in a halo surrounding occipital vessels, which was specific for HCHWA-D. The result of this study indicates an upregulation of TGFß1 in HCHWA-D, as was found previously in AD with CAA pathology. We discuss the possible origins and implications of the TGFß pathway deregulation in the microvasculature in HCHWA-D. These findings identify the TGFß pathway as a potential biomarker of disease progression and a possible target of therapeutic intervention in HCHWA-D.


Assuntos
Angiopatia Amiloide Cerebral Familiar/metabolismo , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Idoso , Idoso de 80 Anos ou mais , Angiopatia Amiloide Cerebral Familiar/patologia , Feminino , Lobo Frontal/irrigação sanguínea , Lobo Frontal/metabolismo , Lobo Frontal/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Lobo Occipital/irrigação sanguínea , Lobo Occipital/metabolismo , Lobo Occipital/patologia , Fosforilação , Transdução de Sinais , Regulação para Cima
18.
J Mol Histol ; 38(4): 359-68, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17605082

RESUMO

Oval cells are liver-specific bipotent stem cells which accumulate in injured liver when proliferation of mature hepatocytes and/or cholangiocytes is impaired. They represent an intermediary cell type with phenotypical characteristics of both, hepatocytes and cholangiocytes. Oval cells express specific cell surface proteins allowing their identification in situ. Most of these cell surface proteins, however, are recognized by antibodies in mouse liver tissue that are not commercially available or work only on frozen sections. We show herein the unequivocal identification of oval cells in paraffin-embedded mouse liver samples based on strong E-cadherin expression different from that of hepatocytes and bile duct cells. By comparing the pattern of E-cadherin expression with that of both, A6-antigen and CD44, we suggest a tight control of E-cadherin expression depending on the differentiation stage of the progenitor cells. In human cirrhotic liver samples E-cadherin expression was found as a common feature of both, typical and atypical reactions, and, thus, can also serve as an indication of the progenitor cell compartment activation.


Assuntos
Caderinas/metabolismo , Fígado/citologia , Células-Tronco/citologia , Animais , Biomarcadores/metabolismo , Humanos , Imuno-Histoquímica , Queratinas/metabolismo , Cirrose Hepática/patologia , Camundongos , Inclusão em Parafina , Ratos
19.
Neuroreport ; 18(12): 1247-50, 2007 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-17632276

RESUMO

Alzheimer's disease is characterized by severe neuronal disintegration supposed to be partly associated with amyloid pathology. Recently, we described morphological alterations of pyramidal cell structure in transgenic mice expressing wild-type or mutant human amyloid precursor protein (hAPP) (strains B6-Py8.9 and Tg2576), which are unrelated to direct plaque-associated changes. In this study, we focused on the pattern of cortical afferent connections in these transgenic mice. The quantity of cholinergic afferents is increased in both transgenic lines. Glutamatergic intra- and interhemispheric afferents are augmented in B6-Py8.9 mice but decreased in Tg2576 mice. Furthermore, perisomatic inhibition of pyramidal neurons was found to be reduced in Tg2576 mice. Findings suggest different effects of wild-type and mutant hAPP on neuronal connectivity.


Assuntos
Vias Aferentes/fisiopatologia , Doença de Alzheimer/fisiopatologia , Precursor de Proteína beta-Amiloide/metabolismo , Córtex Cerebral/fisiopatologia , Transmissão Sináptica/genética , Acetilcolina/metabolismo , Vias Aferentes/metabolismo , Vias Aferentes/patologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Animais , Núcleo Basal de Meynert/metabolismo , Núcleo Basal de Meynert/patologia , Núcleo Basal de Meynert/fisiopatologia , Biomarcadores/metabolismo , Biotina/análogos & derivados , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Fibras Colinérgicas/metabolismo , Fibras Colinérgicas/patologia , Dextranos , Modelos Animais de Doenças , Enzimas/metabolismo , Potenciais Pós-Sinápticos Excitadores/genética , Ácido Glutâmico/metabolismo , Humanos , Potenciais Pós-Sinápticos Inibidores/genética , Camundongos , Camundongos Transgênicos , Mutação/genética , Rede Nervosa/metabolismo , Rede Nervosa/patologia , Rede Nervosa/fisiopatologia , Norepinefrina/metabolismo , Terminações Pré-Sinápticas/metabolismo , Terminações Pré-Sinápticas/patologia , Células Piramidais/metabolismo , Células Piramidais/patologia
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA