Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oral Dis ; 29(1): 21-28, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34698406

RESUMO

Periodontitis is a chronic non-communicable disease caused by a dysbiotic microbiota. Pathogens can spread to the bloodstream, colonize other tissues or organs, and favor the onset of other pathologies, such as Alzheimer's disease (AD). Pathogens could permanently or transiently colonize the brain and induce an immune response. Thus, we analyzed the evidence combining oral bacteria's detection in the brain, both in animals and humans affected with AD. This systematic review was carried out following the PRISMA guideline. Studies that detected oral bacteria at the brain level were selected. The search was carried out in the Medline, Latindex, SciELO, and Cochrane Library databases. SYRCLE tool and Newcastle-Ottawa Scale were used for the risk of bias assessment. 23 studies were selected according to the eligibility criteria. Infection with oral pathogens in animals was related to developing neuropathological characteristics of AD and bacteria detection in the brain. In patients with AD, oral bacteria were detected in brain tissues, and increased levels of pro-inflammatory cytokines were also detected. There is evidence of a microbiological susceptibility to develop AD when the most dysbiosis-associated oral bacteria are present. The presence of bacteria in the brain is related to AD's pathological characteristics, suggesting an etiological oral-brain axis.


Assuntos
Doença de Alzheimer , Microbiota , Periodontite , Animais , Humanos , Periodontite/microbiologia , Bactérias , Encéfalo , Disbiose/complicações
2.
Front Aging Neurosci ; 12: 255, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32903806

RESUMO

Nicotinamide (vitamin B3) is a key component in the cellular production of Nicotinamide Adenine Dinucleotide (NAD) and has long been associated with neuronal development, survival and death. Numerous data suggest that nicotinamide may offer therapeutic benefits in neurodegenerative disorders, including Alzheimer's Disease (AD). Beyond its effect in NAD+ stores, nicotinamide is an inhibitor of Poly [ADP-ribose] polymerase 1 (PARP-1), an enzyme with multiple cellular functions, including regulation of cell death, energy/metabolism and inflammatory response. PARP-1 functions as a DNA repair enzyme but under intense DNA damage depletes the cell of NAD+ and ATP and leads to a non-apoptotic type of cell death called Parthanatos, which has been associated with the pathogenesis of neurodegenerative diseases. Moreover, NAD+ availability might potentially improve mitochondrial function, which is severely impaired in AD. PARP-1 inhibition may also exert a protective effect against neurodegeneration by its action to diminish neuroinflammation and microglial activation which are also implicated in the pathogenesis of AD. Here we discuss the evidence supporting the use of nicotinamide as adjunctive therapy for the treatment of early stages of AD based on the inhibitory effect of nicotinamide on PARP-1 activity. The data support evaluating nicotinamide as an adjunctive treatment for AD at early stages of the disease not only to increase NAD+ stores but as a PARP-1 inhibitor, raising the hypothesis that other PARP-1 inhibitors - drugs that are already approved for breast cancer treatment - might be explored for the treatment of AD.

3.
Free Radic Biol Med ; 117: 132-144, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29309895

RESUMO

The transcription factor STAT3 has a crucial role in the development and maintenance of the nervous system. In this work, we treated astrocytes with oligomers of the amyloid beta peptide (AßOs), which display potent synaptotoxic activity, and studied the effects of mediators released by AßOs-treated astrocytes on the nuclear location of neuronal serine-727-phosphorylated STAT3 (pSerSTAT3). Treatment of mixed neuron-astrocyte cultures with 0.5µMAßOs induced in neurons a significant decrease of nuclear pSerSTAT3, but not of phosphotyrosine-705 STAT3, the other form of STAT3 phosphorylation. This decrease did not occur in astrocyte-poor neuronal cultures revealing a pivotal role for astrocytes in this response. To test if mediators released by astrocytes in response to AßOs induce pSerSTAT3 nuclear depletion, we used conditioned medium derived from AßOs-treated astrocyte cultures. Treatment of astrocyte-poor neuronal cultures with this medium caused pSerSTAT3 nuclear depletion but did not modify overall STAT3 levels. Extracellular catalase prevented the pSerSTAT3 nuclear depletion caused by astrocyte-conditioned medium, indicating that reactive oxygen species (ROS) mediate this response. This conditioned medium also increased neuronal oxidative tone, leading to a ryanodine-sensitive intracellular calcium signal that proved to be essential for pSerSTAT3 nuclear depletion. In addition, this depletion decreased BCL2 and Survivin transcription and significantly increased BAX/BCL2 ratio. This is the first description that ROS generated by AßOs-treated astrocytes and neuronal calcium signals jointly regulate pSerSTAT3 nuclear distribution in neurons. We propose that astrocytes release ROS in response to AßOs, which by increasing neuronal oxidative tone, generate calcium signals that cause pSerSTAT3 nuclear depletion and loss of STAT3 protective transcriptional activity.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Astrócitos/metabolismo , Neurônios/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Núcleo Celular/metabolismo , Sobrevivência Celular , Regulação da Expressão Gênica/fisiologia , Fosforilação , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Serina/metabolismo
4.
Antioxid Redox Signal ; 29(12): 1125-1146, 2018 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-29357673

RESUMO

AIMS: Previous studies indicate that hippocampal synaptic plasticity and spatial memory processes entail calcium release from intracellular stores mediated by ryanodine receptor (RyR) channels. In particular, RyR-mediated Ca2+ release is central for the dendritic spine remodeling induced by brain-derived neurotrophic factor (BDNF), a neurotrophin that stimulates complex signaling pathways leading to memory-associated protein synthesis and structural plasticity. To examine if upregulation of ryanodine receptor type-2 (RyR2) channels and the spine remodeling induced by BDNF entail reactive oxygen species (ROS) generation, and to test if RyR2 downregulation affects BDNF-induced spine remodeling and spatial memory. RESULTS: Downregulation of RyR2 expression (short hairpin RNA [shRNA]) in primary hippocampal neurons, or inhibition of nitric oxide synthase (NOS) or NADPH oxidase, prevented agonist-mediated RyR-mediated Ca2+ release, whereas BDNF promoted cytoplasmic ROS generation. RyR2 downregulation or inhibitors of N-methyl-d-aspartate (NMDA) receptors, or NOS or of NADPH oxidase type-2 (NOX2) prevented RyR2 upregulation and the spine remodeling induced by BDNF, as did incubation with the antioxidant agent N-acetyl l-cysteine. In addition, intrahippocampal injection of RyR2-directed antisense oligodeoxynucleotides, which caused significant RyR2 downregulation, caused conspicuous defects in a memorized spatial memory task. INNOVATION: The present novel results emphasize the key role of redox-sensitive Ca2+ release mediated by RyR2 channels for hippocampal structural plasticity and spatial memory. CONCLUSION: Based on these combined results, we propose (i) that BDNF-induced RyR2-mediated Ca2+ release and ROS generation via NOS/NOX2 are strictly required for the dendritic spine remodeling and the RyR2 upregulation induced by BDNF, and (ii) that RyR2 channel expression is crucial for spatial memory processes. Antioxid. Redox Signal. 29, 1125-1146.


Assuntos
Cálcio/metabolismo , Hipocampo/metabolismo , Plasticidade Neuronal , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Memória Espacial , Animais , Células Cultivadas , Hipocampo/citologia , Oxirredução , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo
5.
Front Mol Neurosci ; 10: 115, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28487634

RESUMO

Amyloid ß peptide oligomers (AßOs), toxic aggregates with pivotal roles in Alzheimer's disease, trigger persistent and low magnitude Ca2+ signals in neurons. We reported previously that these Ca2+ signals, which arise from Ca2+ entry and subsequent amplification by Ca2+ release through ryanodine receptor (RyR) channels, promote mitochondrial network fragmentation and reduce RyR2 expression. Here, we examined if AßOs, by inducing redox sensitive RyR-mediated Ca2+ release, stimulate mitochondrial Ca2+-uptake, ROS generation and mitochondrial fragmentation, and also investigated the effects of the antioxidant N-acetyl cysteine (NAC) and the mitochondrial antioxidant EUK-134 on AßOs-induced mitochondrial dysfunction. In addition, we studied the contribution of the RyR2 isoform to AßOs-induced Ca2+ release, mitochondrial Ca2+ uptake and fragmentation. We show here that inhibition of NADPH oxidase type-2 prevented the emergence of RyR-mediated cytoplasmic Ca2+ signals induced by AßOs in primary hippocampal neurons. Treatment with AßOs promoted mitochondrial Ca2+ uptake and increased mitochondrial superoxide and hydrogen peroxide levels; ryanodine, at concentrations that suppress RyR activity, prevented these responses. The antioxidants NAC and EUK-134 impeded the mitochondrial ROS increase induced by AßOs. Additionally, EUK-134 prevented the mitochondrial fragmentation induced by AßOs, as previously reported for NAC and ryanodine. These findings show that both antioxidants, NAC and EUK-134, prevented the Ca2+-mediated noxious effects of AßOs on mitochondrial function. Our results also indicate that Ca2+ release mediated by the RyR2 isoform causes the deleterious effects of AßOs on mitochondrial function. Knockdown of RyR2 with antisense oligonucleotides reduced by about 50% RyR2 mRNA and protein levels in primary hippocampal neurons, decreased by 40% Ca2+ release induced by the RyR agonist 4-chloro-m-cresol, and significantly reduced the cytoplasmic and mitochondrial Ca2+ signals and the mitochondrial fragmentation induced by AßOs. Based on our results, we propose that AßOs-induced Ca2+ entry and ROS generation jointly stimulate RyR2 activity, causing mitochondrial Ca2+ overload and fragmentation in a feed forward injurious cycle. The present novel findings highlight the specific participation of RyR2-mediated Ca2+ release on AßOs-induced mitochondrial malfunction.

6.
Front Mol Neurosci ; 7: 13, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24653672

RESUMO

Mounting evidence indicates that iron accumulation impairs brain function. We have reported previously that addition of sub-lethal concentrations of iron to primary hippocampal neurons produces Ca(2) (+) signals and promotes cytoplasmic generation of reactive oxygen species. These Ca(2) (+) signals, which emerge within seconds after iron addition, arise mostly from Ca(2) (+) release through the redox-sensitive ryanodine receptor (RyR) channels present in the endoplasmic reticulum. We have reported also that addition of synaptotoxic amyloid-ß oligomers to primary hippocampal neurons stimulates RyR-mediated Ca(2) (+) release, generating long-lasting Ca(2) (+) signals that activate Ca(2) (+)-sensitive cellular effectors and promote the disruption of the mitochondrial network. Here, we describe that 24 h incubation of primary hippocampal neurons with iron enhanced agonist-induced RyR-mediated Ca(2) (+) release and promoted mitochondrial network fragmentation in 43% of neurons, a response significantly prevented by RyR inhibition and by the antioxidant agent N-acetyl-L-cysteine. Stimulation of RyR-mediated Ca(2) (+) release by a RyR agonist promoted mitochondrial Ca(2) (+) uptake in control neurons and in iron-treated neurons that displayed non-fragmented mitochondria, but not in neurons with fragmented mitochondria. Yet, the global cytoplasmic Ca(2) (+) increase induced by the Ca(2) (+) ionophore ionomycin prompted significant mitochondrial Ca(2) (+) uptake in neurons with fragmented mitochondria, indicating that fragmentation did not prevent mitochondrial Ca(2) (+) uptake but presumably decreased the functional coupling between RyR-mediated Ca(2) (+) release and the mitochondrial Ca(2) (+) uniporter. Taken together, our results indicate that stimulation of redox-sensitive RyR-mediated Ca(2) (+) release by iron causes significant neuronal mitochondrial fragmentation, which presumably contributes to the impairment of neuronal function produced by iron accumulation.

7.
Antioxid Redox Signal ; 21(6): 892-914, 2014 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-24410659

RESUMO

SIGNIFICANCE: Memory is an essential human cognitive function. Consequently, to unravel the cellular and molecular mechanisms responsible for the synaptic plasticity events underlying memory formation, storage and loss represents a major challenge of present-day neuroscience. RECENT ADVANCES: This review article first describes the wide-ranging functions played by intracellular Ca2+ signals in the activity-dependent synaptic plasticity processes underlying hippocampal spatial memory, and next, it focuses on how the endoplasmic reticulum Ca2+ release channels, the ryanodine receptors, and the inositol 1,4,5-trisphosphate receptors contribute to these processes. We present a detailed examination of recent evidence supporting the key role played by Ca2+ release channels in synaptic plasticity, including structural plasticity, and the formation/consolidation of spatial memory in the hippocampus. CRITICAL ISSUES: Changes in cellular oxidative state particularly affect the function of Ca2+ release channels and alter hippocampal synaptic plasticity and the associated memory processes. Emphasis is placed in this review on how defective Ca2+ release, presumably due to increased levels of reactive oxygen species, may cause the hippocampal functional defects that are associated to aging and Alzheimer's disease (AD). FUTURE DIRECTIONS: Additional studies should examine the precise molecular mechanisms by which Ca2+ release channels contribute to hippocampal synaptic plasticity and spatial memory formation/consolidation. Future studies should test whether redox-modified Ca2+ release channels contribute toward generating the intracellular Ca2+ signals required for sustained synaptic plasticity and hippocampal spatial memory, and whether loss of redox balance and oxidative stress, by altering Ca2+ release channel function, presumably contribute to the abnormal memory processes that occur during aging and AD.


Assuntos
Canais de Cálcio/metabolismo , Cálcio/metabolismo , Hipocampo/metabolismo , Plasticidade Neuronal , Memória Espacial , Envelhecimento , Doença de Alzheimer/metabolismo , Animais , Sinalização do Cálcio , Retículo Endoplasmático/metabolismo , Humanos , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Potenciação de Longa Duração , Depressão Sináptica de Longo Prazo , Neurônios/metabolismo , Oxirredução , Espécies Reativas de Oxigênio/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo
9.
J Neurochem ; 126(2): 191-202, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23668663

RESUMO

Alzheimer's disease (AD) is the most common form of dementia in the elderly. Memory loss in AD is increasingly attributed to soluble oligomers of the amyloid-ß peptide (AßOs), toxins that accumulate in AD brains and target particular synapses. Glutamate receptors appear to be centrally involved in synaptic targeting by AßOs. Once bound to neurons, AßOs dysregulate the activity and reduce the surface expression of both N-methyl-D-aspartate (NMDA) and 2-amino-3-(3-hydroxy-5-methyl-isoxazol-4-yl)propanoic acid (AMPA) types of glutamate receptors, impairing signaling pathways involved in synaptic plasticity. In the extracellular milieu, AßOs promote accumulation of the excitatory amino acids, glutamate and D-serine. This leads to overactivation of glutamate receptors, triggering abnormal calcium signals with noxious impacts on neurons. Here, we review key findings linking AßOs to deregulated glutamate neurotransmission and implicating this as a primary mechanism of synapse failure in AD. We also discuss strategies to counteract the impact of AßOs on excitatory neurotransmission. In particular, we review evidence showing that inducing neuronal hyperpolarization via activation of inhibitory GABA(A) receptors prevents AßO-induced excitotoxicity, suggesting that this could comprise a possible therapeutic approach in AD.


Assuntos
Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Sinapses/metabolismo , Transmissão Sináptica/fisiologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Humanos , Potenciação de Longa Duração/fisiologia , Receptores de Glutamato/metabolismo
10.
J Biol Chem ; 287(10): 7436-45, 2012 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-22235132

RESUMO

Cognitive decline in Alzheimer disease (AD) is increasingly attributed to the neuronal impact of soluble oligomers of the amyloid-ß peptide (AßOs). Current knowledge on the molecular and cellular mechanisms underlying the toxicity of AßOs stems largely from rodent-derived cell/tissue culture experiments or from transgenic models of AD, which do not necessarily recapitulate the complexity of the human disease. Here, we used DNA microarray and RT-PCR to investigate changes in transcription in adult human cortical slices exposed to sublethal doses of AßOs. The results revealed a set of 27 genes that showed consistent differential expression upon exposure of slices from three different donors to AßOs. Functional classification of differentially expressed genes revealed that AßOs impact pathways important for neuronal physiology and known to be dysregulated in AD, including vesicle trafficking, cell adhesion, actin cytoskeleton dynamics, and insulin signaling. Most genes (70%) were down-regulated by AßO treatment, suggesting a predominantly inhibitory effect on the corresponding pathways. Significantly, AßOs induced down-regulation of synaptophysin, a presynaptic vesicle membrane protein, suggesting a mechanism by which oligomers cause synapse failure. The results provide insight into early mechanisms of pathogenesis of AD and suggest that the neuronal pathways affected by AßOs may be targets for the development of novel diagnostic or therapeutic approaches.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Regulação da Expressão Gênica , Proteínas do Tecido Nervoso/biossíntese , Adulto , Doença de Alzheimer/diagnóstico , Doença de Alzheimer/patologia , Doença de Alzheimer/terapia , Encéfalo/patologia , Feminino , Perfilação da Expressão Gênica , Humanos , Masculino , Análise de Sequência com Séries de Oligonucleotídeos
11.
Proc Natl Acad Sci U S A ; 108(7): 3029-34, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21282625

RESUMO

Ryanodine receptors (RyR) amplify activity-dependent calcium influx via calcium-induced calcium release. Calcium signals trigger postsynaptic pathways in hippocampal neurons that underlie synaptic plasticity, learning, and memory. Recent evidence supports a role of the RyR2 and RyR3 isoforms in these processes. Along with calcium signals, brain-derived neurotrophic factor (BDNF) is a key signaling molecule for hippocampal synaptic plasticity and spatial memory. Upon binding to specific TrkB receptors, BDNF initiates complex signaling pathways that modify synaptic structure and function. Here, we show that BDNF-induced remodeling of hippocampal dendritic spines required functional RyR. Additionally, incubation with BDNF enhanced the expression of RyR2, RyR3, and PKMζ, an atypical protein kinase C isoform with key roles in hippocampal memory consolidation. Consistent with their increased RyR protein content, BDNF-treated neurons generated larger RyR-mediated calcium signals than controls. Selective inhibition of RyR-mediated calcium release with inhibitory ryanodine concentrations prevented the PKMζ, RyR2, and RyR3 protein content enhancement induced by BDNF. Intrahippocampal injection of BDNF or training rats in a spatial memory task enhanced PKMζ, RyR2, RyR3, and BDNF hippocampal protein content, while injection of ryanodine at concentrations that stimulate RyR-mediated calcium release improved spatial memory learning and enhanced memory consolidation. We propose that RyR-generated calcium signals are key features of the complex neuronal plasticity processes induced by BDNF, which include increased expression of RyR2, RyR3, and PKMζ and the spine remodeling required for spatial memory formation.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Hipocampo/citologia , Memória/fisiologia , Plasticidade Neuronal/fisiologia , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Percepção Espacial/fisiologia , Sinapses/fisiologia , Análise de Variância , Animais , Cálcio/metabolismo , Hipocampo/metabolismo , Imuno-Histoquímica , Masculino , Aprendizagem em Labirinto/fisiologia , Ratos , Ratos Sprague-Dawley , Rianodina/administração & dosagem , Transdução de Sinais/fisiologia
12.
Antioxid Redox Signal ; 14(7): 1209-23, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20712397

RESUMO

Soluble amyloid ß-peptide oligomers (AßOs), increasingly recognized as causative agents of Alzheimer's disease (AD), disrupt neuronal Ca(2+) homeostasis and synaptic function. Here, we report that AßOs at sublethal concentrations generate prolonged Ca(2+) signals in primary hippocampal neurons; incubation in Ca(2+)-free solutions, inhibition of ryanodine receptors (RyRs) or N-methyl-d-aspartate receptors (NMDARs), or preincubation with N-acetyl-l-cysteine abolished these signals. AßOs decreased (6 h) RyR2 and RyR3 mRNA and RyR2 protein, and promoted mitochondrial fragmentation after 24 h. NMDAR inhibition abolished the RyR2 decrease, whereas RyR inhibition prevented significantly the RyR2 protein decrease and mitochondrial fragmentation induced by AßOs. Incubation with AßOs (6 h) eliminated the RyR2 increase induced by brain-derived nerve factor (BDNF) and the dendritic spine remodeling induced within minutes by BDNF or the RyR agonist caffeine. Addition of BDNF to neurons incubated with AßOs for 24 h, which had RyR2 similar to and slightly higher RyR3 protein content than those of controls, induced dendritic spine growth but at slower rates than in controls. These combined effects of sublethal AßOs concentrations (which include redox-sensitive stimulation of RyR-mediated Ca(2+) release, decreased RyR2 protein expression, mitochondrial fragmentation, and prevention of RyR-mediated spine remodeling) may contribute to impairing the synaptic plasticity in AD.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Cálcio/metabolismo , Espinhas Dendríticas/metabolismo , Hipocampo/citologia , Mitocôndrias/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Forma Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Citocromos c/metabolismo , Regulação para Baixo , Hipocampo/metabolismo , Humanos , Ratos , Ratos Sprague-Dawley , Rianodina/farmacologia , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Transcrição Gênica
13.
Int J Biochem Cell Biol ; 41(6): 1361-70, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19130896

RESUMO

Aggregates of the amyloid-beta peptide (Abeta) play a central role in the pathogenesis of Alzheimer's disease (AD). Identification of proteins that physiologically bind Abeta and modulate its aggregation and neurotoxicity could lead to the development of novel disease-modifying approaches in AD. By screening a phage display peptide library for high affinity ligands of aggregated Abeta(1-42), we isolated a peptide homologous to a highly conserved amino acid sequence present in the N-terminus of apolipoprotein A-I (apoA-I). We show that purified human apoA-I and Abeta form non-covalent complexes and that interaction with apoA-I affects the morphology of amyloid aggregates formed by Abeta. Significantly, Abeta/apoA-I complexes were also detected in cerebrospinal fluid from AD patients. Interestingly, apoA-I and apoA-I-containing reconstituted high density lipoprotein particles protect hippocampal neuronal cultures from Abeta-induced oxidative stress and neurodegeneration. These results suggest that human apoA-I modulates Abeta aggregation and Abeta-induced neuronal damage and that the Abeta-binding domain in apoA-I may constitute a novel framework for the design of inhibitors of Abeta toxicity.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Apolipoproteína A-I/metabolismo , Fragmentos de Peptídeos/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Sequência de Aminoácidos , Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/toxicidade , Animais , Apolipoproteína A-I/genética , Eletroforese em Gel de Poliacrilamida , Feminino , Humanos , Neurônios/metabolismo , Neurônios/patologia , Estresse Oxidativo , Fragmentos de Peptídeos/antagonistas & inibidores , Fragmentos de Peptídeos/toxicidade , Gravidez , Ratos , Ratos Sprague-Dawley
14.
Biol Res ; 41(2): 129-36, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18949129

RESUMO

We have previously characterized a number of small molecule organic compounds that prevent the aggregation of the beta-amyloid peptide and its neurotoxicity in hippocampal neuronal cultures. We have now evaluated the effects of such compounds on amyloid precursor protein (APP) accumulation in the CTb immortalized cell line derived from the cerebral cortex of a trisomy 16 mouse, an animal model of Down's syndrome. Compared to a non-trisomic cortical cell line (CNh), CTb cells overexpress APP and exhibit slightly elevated resting intracellular Ca2+ levels ([Ca2+] inverted exclamation mark). Here, we show that the compounds 2,4-dinitrophenol, 3-nitrophenol and 4-anisidine decreased intracellular accumulation of APP in CTb cells. Those compounds were non-toxic to the cells, and slightly increased the basal [Ca2+] inverted exclamation mark. Results indicate that the compounds tested can be leads for the development of drugs to decrease intracellular vesicular accumulation of APP in trisomic cells.


Assuntos
2,4-Dinitrofenol/farmacologia , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/antagonistas & inibidores , Compostos de Anilina/farmacologia , Síndrome de Down/metabolismo , Nitrofenóis/farmacologia , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Linhagem Celular , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Modelos Animais de Doenças , Camundongos
15.
Neuropharmacology ; 49(8): 1140-8, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16150468

RESUMO

The beta-amyloid peptide (Abeta) is centrally related to the pathogenesis of Alzheimer's disease (AD) and is potently neurotoxic to central nervous system neurons. The neurotoxicity of Abeta has been partially related to the over activation of glutamatergic transmission and excitotoxicity. Taurine is a naturally occurring beta-amino acid present in the mammalian brain. Due to its safety and tolerability, taurine has been clinically used in humans in the treatment of a number of non-neurological disorders. Here, we show that micromolar doses of taurine block the neurotoxicity of Abeta to rat hippocampal and cortical neurons in culture. Moreover, taurine also rescues central neurons from the excitotoxicity induced by high concentrations of extracellular glutamate. Neuroprotection by taurine is abrogated by picrotoxin, a GABA(A) receptor antagonist. GABA and muscimol, an agonist of the GABA(A) receptor, also block neuronal death induced by Abeta in rat hippocampal and cortical neurons. These results suggest that activation of GABA(A) receptors protects neurons against Abeta toxicity in AD-affected regions of the mammalian brain and that taurine should be investigated as a novel therapeutic tool in the treatment of AD and of other neurological disorders in which excitotoxicity plays a relevant role.


Assuntos
Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/toxicidade , Córtex Cerebral/citologia , Agonistas GABAérgicos/farmacologia , Agonistas de Receptores de GABA-A , Hipocampo/efeitos dos fármacos , Muscimol/farmacologia , Neurônios/efeitos dos fármacos , Taurina/farmacologia , Animais , Sobrevivência Celular , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
16.
FASEB J ; 18(3): 511-8, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15003996

RESUMO

Alzheimer's disease (AD) and several other neurological disorders have been linked to the overactivation of glutamatergic transmission and excitotoxicity as a common pathway of neuronal injury. The beta-amyloid peptide (Abeta) is centrally related to the pathogenesis of AD, and previous reports have demonstrated that the blockade of glutamate receptors prevents Abeta-induced neuronal death. We show that taurine, a beta-amino acid found at high concentrations in the brain, protects chick retinal neurons in culture against the neurotoxicity of Abeta and glutamate receptor agonists. The protective effect of taurine is not mediated by interaction with glutamate receptors, as demonstrated by binding studies using radiolabeled glutamate receptor ligands. The neuroprotective action of taurine is blocked by picrotoxin, an antagonist of GABA(A) receptors. GABA and the GABA(A) receptor agonists phenobarbital and melatonin also protect neurons against Abeta-induced neurotoxicity. These results suggest that activation of GABA receptors decreases neuronal vulnerability to excitotoxic damage and that pharmacological manipulation of the excitatory and inhibitory neurotransmitter tonus may protect neurons against a variety of insults. GABAergic transmission may represent a promising target for the treatment of AD and other neurological disorders in which excitotoxicity plays a relevant role.


Assuntos
Precursor de Proteína beta-Amiloide/toxicidade , Proteínas do Olho/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Receptores de GABA/efeitos dos fármacos , Receptores de Glutamato/efeitos dos fármacos , Taurina/farmacologia , Doença de Alzheimer/tratamento farmacológico , Animais , Células Cultivadas , Senescência Celular , Embrião de Galinha , Maleato de Dizocilpina/farmacologia , Agonistas de Aminoácidos Excitatórios/farmacologia , Proteínas do Olho/fisiologia , Ácido Glutâmico/farmacologia , Ácido Caínico/toxicidade , N-Metilaspartato/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Picrotoxina/farmacologia , Receptores de AMPA/efeitos dos fármacos , Receptores de GABA/fisiologia , Receptores de GABA-A/efeitos dos fármacos , Receptores de GABA-A/fisiologia , Receptores de Glutamato/fisiologia , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Retina/citologia , Retina/embriologia , Taurina/uso terapêutico , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/farmacologia
17.
Neurotox Res ; 5(5): 323-7, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14715451

RESUMO

The beta-amyloid peptide (Abeta) is centrally related to the pathogenesis of Alzheimer's disease (AD). Previous studies have suggested that the neurotoxicity of Abeta may be related to the overactivation of glutamatergic transmission and excitotoxicity, and that blockade of glutamate receptors prevents Abeta-induced cell death. Here, we show that melatonin, a pineal hormone, protects chick retinal neurons in culture against the neurotoxicity of Abeta and glutamate. Right-angle light scattering and thioflavin T fluorescence measurements, as well as light microscopy analysis, indicated that, under our experimental conditions, melatonin had no effect on the aggregation of Abeta. Interestingly, the neuroprotective action of melatonin against the toxicity of Abeta was significantly decreased in the presence of picrotoxin, an antagonist of GABA(A)-like receptors. By itself, picrotoxin had no effect. These results suggest that the neuroprotective effects of melatonin against Abeta neurotoxicity could be at least in part related to a decrease in the excitatory tonus, mediated by activation of GABA receptors and the resulting hyper-polarization of the neurons. Thus, selective pharmacological manipulation of neuronal excitatory/inhibitory tonus could be a potentially interesting new approach in the treatment of AD.


Assuntos
Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/toxicidade , Antioxidantes/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Ácido Glutâmico/toxicidade , Melatonina/farmacologia , Fragmentos de Peptídeos/antagonistas & inibidores , Fragmentos de Peptídeos/toxicidade , Receptores de GABA/efeitos dos fármacos , Animais , Benzotiazóis , Embrião de Galinha , Corantes Fluorescentes , Antagonistas GABAérgicos/farmacologia , Luz , Neurônios/efeitos dos fármacos , Picrotoxina/farmacologia , Retina/citologia , Retina/efeitos dos fármacos , Espalhamento de Radiação , Tiazóis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...