Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Artigo em Inglês | MEDLINE | ID: mdl-36554305

RESUMO

The effects of 5-methyltetrahydrofolate (5-MTHF) on a rat model of Alzheimer's disease (AD) induced by D-galactose (D-gal) and aluminum chloride (AlCl3) were investigated. Wistar rats were given an i.p. injection of 60 mg/kg D-gal and 10 mg/kg AlCl3 to induce AD and three doses of 1 mg/kg, 5 mg/kg or 10 mg/kg 5-MTHF by oral gavage. A positive control group was treated with 1 mg/kg donepezil by gavage. Morris water maze performance showed that 5 and 10 mg/kg 5-MTHF significantly decreased escape latency and increased the number of platform crossings and time spent in the target quadrant for AD rats. The administration of 10 mg/kg 5-MTHF decreased the brain content of amyloid ß-protein 1-42 (Aß1-42) and phosphorylated Tau protein (p-Tau) and decreased acetylcholinesterase and nitric oxide synthase activities. Superoxide dismutase activity, vascular endothelial growth factor level and glutamate concentration were increased, and malondialdehyde, endothelin-1, interleukin-6, tumor necrosis factor-alpha and nitric oxide decreased. The administration of 10 mg/kg 5-MTHF also increased the expression of disintegrin and metallopeptidase domain 10 mRNA and decreased the expression of ß-site amyloid precursor protein cleavage enzyme 1 mRNA. In summary, 5-MTHF alleviates memory impairment in a D-gal- and AlCl3-exposed rat model of AD. The inhibition of Aß1-42 and p-Tau release, reduced oxidative stress, the regulation of amyloid precursor protein processing and the release of excitatory amino acids and cytokines may be responsible.


Assuntos
Doença de Alzheimer , Animais , Ratos , Acetilcolinesterase/metabolismo , Cloreto de Alumínio/toxicidade , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/toxicidade , Precursor de Proteína beta-Amiloide/efeitos adversos , Precursor de Proteína beta-Amiloide/metabolismo , Modelos Animais de Doenças , Galactose/toxicidade , Hipocampo/metabolismo , Aprendizagem em Labirinto/fisiologia , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/tratamento farmacológico , Estresse Oxidativo , Ratos Wistar , RNA Mensageiro/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
2.
Neurobiol Aging ; 105: 137-147, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34062489

RESUMO

Alzheimer's disease (AD) is the prevailing form of dementia. Protein degradation and antioxidant pathways have a critical role in preventing the accumulation of protein aggregation; thus, failure of proteostasis in neurons along with redox imbalance mark AD. Herein, we exploited an AD Drosophila model expressing human amyloid precursor (hAPP) and beta-secretase 1 (hBACE1) proteins, to better understand the role of proteostatic or antioxidant pathways in AD. Ubiquitous expression of hAPP, hBACE1 in flies caused more severe degenerative phenotypes versus neuronal targeted expression; it also, suppressed proteasome activity, increased oxidative stress and significantly enhanced stress-sensitivity. Overexpression of Prosß5 proteasomal subunit or Nrf2 transcription factor in AD Drosophila flies partially restored proteasomal activity but did not rescue hAPP, hBACE1 induced neurodegeneration. On the other hand, expression of autophagy-related Atg8a in AD flies decelerated neurodegeneration, increased stress-resistance, and improved flies' health-/lifespan. Overall, our data suggest that the noxious effects of amyloid-beta aggregates can be alleviated by enhanced autophagy, thus dietary or pharmacological interventions that target autophagy should be considered in AD therapeutic approaches.


Assuntos
Doença de Alzheimer/etiologia , Doença de Alzheimer/terapia , Autofagia/genética , Autofagia/fisiologia , Drosophila , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/efeitos adversos , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Bromisoval , Modelos Animais de Doenças , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiologia , Combinação de Medicamentos , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Degeneração Neural/genética , Degeneração Neural/patologia , Complexo de Endopeptidases do Proteassoma/metabolismo
3.
Exp Mol Pathol ; 120: 104622, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33684392

RESUMO

Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by neuronal and synaptic loss. The cytoplasmic tail of amyloid precursor protein (APP) undergoes sequential cleavage at a specific intracellular caspase site to generate the cytoplasmic terminal 31 (CT31) fragment. The APP-CT31 fragment is a potent inducer of apoptosis. The cytotoxicity of APP-CT31 in SH-SY5Y cells was evaluated by the lactate dehydrogenase (LDH) assay. TUNEL staining was used to detect apoptotic signals in SH-SY5Y cells and primary cortical neurons. The expression of apoptosis-related proteins, such as p53, PUMA (p53 up-regulated modulator of apoptosis), and cleaved was investigated by immunofluorescence analysis and Western blotting. In this study, we investigated the neuroprotective effect of neuregulin 1 (NRG1) against cytotoxicity induced by APP-CT31. Our data showed that CT31 induced cytotoxicity and apoptosis in SH-SY5Y cells and primary cortical neurons. NRG1 attenuated the neurotoxicity induced by the expression of APP-CT31. We also showed that APP-CT31 altered the expression of p53 and cleaved caspase 3. However, treatment with NRG1 rescued the APP-CT31-induced upregulation of p53 and cleaved caspase 3 expression. The protective effect of NRG1 was abrogated by inhibition of the ErbB4 receptor and Akt. These results indicate an important role of ErbB4/Akt signaling in NRG1-mediated neuroprotection, suggesting that endogenous NRG1/ErbB4 signaling represents a valuable therapeutic target in AD.


Assuntos
Precursor de Proteína beta-Amiloide/efeitos adversos , Neuregulina-1/metabolismo , Neuroblastoma/prevenção & controle , Fármacos Neuroprotetores/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor ErbB-4/metabolismo , Apoptose , Proliferação de Células , Humanos , Neuregulina-1/genética , Neuroblastoma/etiologia , Neuroblastoma/patologia , Domínios Proteicos , Proteínas Proto-Oncogênicas c-akt/genética , Receptor ErbB-4/genética , Células Tumorais Cultivadas
4.
Int J Med Mushrooms ; 22(2): 145-159, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32479003

RESUMO

One of the major causes of Alzheimer's disease (AD) is oxidative stress, which accelerates ß-amyloid peptide (AP) plaque and neurofibrillary tangle accumulation in the brain. Pleurotus eryngii is known to be rich in antioxidants, including ergothioneine, adenosine, and polyphenol, which can reduce oxidative stress-related aging. The aim of this study was to investigate the proximate and functional composition of P. eryngii, and evaluate the cognitive effects of low (LPE), medium (MPE), and high (HPE) P. eryngii dosages in an Aß-induced Alzheimer's disease C57BL/6J mouse model. Mice fed P. eryngii for six weeks showed no adverse effects on body weight gain, food intake efficiency, serum biochemical parameters, and liver and kidney histopathological features. The relative brain weight was significantly lower in Aß-injected mice (p < 0.05). Further, P. eryngii was shown to delay brain atrophy. Reference memory behavioral tasks showed that LPE, MPE, and HPE significantly decreased escape latency (49-85%) and distance (53-69%, p < 0.05). Probe and T-maze tasks showed that P. eryngii potently ameliorated memory deficit in mice. An AD pathology index analysis showed that P. eryngii significantly decreased levels of brain phosphorylated τ-protein, Aß plaque deposition, malondialdehyde, and protein carbonyl (p < 0.05). P. eryngii may therefore promote memory and learning capacity in an Aß-induced AD mouse model.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/metabolismo , Antioxidantes/farmacologia , Carpóforos/química , Deficiências da Aprendizagem/tratamento farmacológico , Transtornos da Memória/tratamento farmacológico , Pleurotus/química , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/efeitos adversos , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Antioxidantes/química , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Modelos Animais de Doenças , Humanos , Deficiências da Aprendizagem/patologia , Transtornos da Memória/patologia , Camundongos , Camundongos Endogâmicos C57BL , Estresse Oxidativo , Pós/química , Pós/farmacologia
5.
Alzheimers Dement ; 16(2): 273-282, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31677937

RESUMO

INTRODUCTION: The levels and distribution of amyloid deposits in the brain does not correlate well with Alzheimer's disease (AD) progression. Therefore, it is likely that amyloid precursor protein and its proteolytic fragments other than amyloid b (Ab) contribute to the onset of AD. METHODS: We developed a sensitive assay adapted to the detection of C99, the direct precursor of b-amyloid. Three postmortem groups were studied: control with normal and stable cognition; patients with moderate AD, and individuals with severe AD. The amount of C99 and Aß was quantified and correlated with the severity of AD. RESULTS: C99 accumulates in vulnerable neurons, and its levels correlate with the degree of cognitive impairment in patients suffering from AD. In contrast, Aß levels are increased in both vulnerable and resistant brain areas. DISCUSSION: These results raise the possibility that C99, rather than Aß plaques, is responsible for the death of nerve cells in AD.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Progressão da Doença , Neurônios/metabolismo , Fragmentos de Peptídeos/efeitos adversos , Precursor de Proteína beta-Amiloide/efeitos adversos , Animais , Autopsia , Encéfalo/metabolismo , Humanos
6.
Artigo em Inglês | MEDLINE | ID: mdl-31152860

RESUMO

Neuronal apoptosis and impaired hippocampal neurogenesis are major players in cognitive/memory dysfunctions including Alzheimer's disease (AD). Interferon beta (IFNß) is a cytokine with anti-apoptotic and neuroprotective properties on the central nervous system (CNS) cells which specifically affects neural progenitor cells (NPCs) even in the adult brain. In this study, we examined the effect of IFNß on memory impairment as well as hippocampal neurogenesis and apoptosis in a rat model of AD. AD model was induced by lentiviral-mediated overexpression of mutant APP in the hippocampus of adult rats. Intranasal (IN) administration of IFNß (0.5 µg/kg and 1 µg/kg doses) was started from day 23 after virus injection and continued every other day to the final day of experiments. The expression levels of APP, neurogenesis (Nestin, Ki67, DCX, and Reelin) and apoptosis (Bax/Bcl-2 ratio, cleaved-caspase-3 and seladin-1) markers were evaluated by immunohistochemistry, real-time PCR, immunofluorescence and western blotting. Moreover, thioflavin T and Nissl stainings were used to assess Aß plaque levels and neuronal degeneration in the hippocampus, respectively. Our results showed that IFNß treatment reduced APP expression and Aß plaque formation, and concomitantly ameliorated spatial learning and memory deficits examined in Y-maze and Morris water maze tests. Moreover, in parallel with reducing apoptosis and neural loss in the hippocampal subfields, IFNß decreased ectopic neurogenesis in the CA1 and CA3 regions of the AD rat hippocampus. However, IFNß increased neurogenesis in the dentate gyrus neurogenic niche. Our findings suggest that IFNß exerts neuroprotective effects at least partly by inhibition of apoptosis and modulation of neurogenesis. Taken together, IFNß can be a promising therapeutic approach to improve cognitive performance in AD-like neurodegenerative context.


Assuntos
Doença de Alzheimer/prevenção & controle , Apoptose/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Interferon beta/farmacologia , Transtornos da Memória/prevenção & controle , Neurogênese/efeitos dos fármacos , Administração Intranasal , Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Doença de Alzheimer/psicologia , Precursor de Proteína beta-Amiloide/efeitos adversos , Precursor de Proteína beta-Amiloide/biossíntese , Precursor de Proteína beta-Amiloide/genética , Animais , Proteína Duplacortina , Vetores Genéticos , Interferon beta/administração & dosagem , Lentivirus , Masculino , Mutação , Placa Amiloide/patologia , Ratos , Proteína Reelina
7.
Eur Rev Med Pharmacol Sci ; 23(5): 2182-2187, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30915764

RESUMO

OBJECTIVE: To explore the effect of neurotrophin-3 (NT-3) messenger ribonucleic acid (mRNA) in the hippocampus on infection-induced memory impairment of neonatal rats. MATERIALS AND METHODS: 80 female Sprague-Dawley (SD) rats in the neonatal stage were selected to establish memory impairment model by bacterial meningitis infection. Rats were randomly divided into experimental group (n=40) and control group (n=40). Rats in experimental group were injected with ß-amyloid precursor protein 319-335 peptide APP17p into brain tissue to up-regulate the expression of NT-3, and the rats in control group didn't receive treatment. Behavioral changes of rats were observed in Morris water maze and passive avoidance experiment. Apoptosis of nerve cells was detected by terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick end labeling (TUNEL) method and Fluoro-Jade B method. NT-3 mRNA expression level was measured via reverse transcription polymerase chain reaction (RT-PCR). RESULTS: NT-3 expression level in experimental group was higher than that in control group (p<0.05). Apoptosis rate of nerve cells in experimental group was lower than that in control group, but the learning and memory ability of rats in experimental group was better than that in control group (p<0.05). CONCLUSIONS: Reduced NT-3 expression level may be correlated with the occurrence of meningitis because NT-3 can suppress nerve cell apoptosis and ameliorate learning and memory impairment to a certain extent to exert neuroprotective effects.


Assuntos
Precursor de Proteína beta-Amiloide/efeitos adversos , Transtornos da Memória/genética , Meningites Bacterianas/psicologia , Neurotrofina 3/genética , Fragmentos de Peptídeos/efeitos adversos , Regulação para Cima , Animais , Animais Recém-Nascidos , Aprendizagem da Esquiva , Modelos Animais de Doenças , Feminino , Masculino , Aprendizagem em Labirinto , Transtornos da Memória/induzido quimicamente , Neurotrofina 3/metabolismo , Ratos , Ratos Sprague-Dawley
8.
Nature ; 564(7736): 415-419, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30546139

RESUMO

We previously reported1 the presence of amyloid-ß protein (Aß) deposits in individuals with Creutzfeldt-Jakob disease (CJD) who had been treated during childhood with human cadaveric pituitary-derived growth hormone (c-hGH) contaminated with prions. The marked deposition of parenchymal and vascular Aß in these relatively young individuals with treatment-induced (iatrogenic) CJD (iCJD), in contrast to other prion-disease patients and population controls, allied with the ability of Alzheimer's disease brain homogenates to seed Aß deposition in laboratory animals, led us to argue that the implicated c-hGH batches might have been contaminated with Aß seeds as well as with prions. However, this was necessarily an association, and not an experimental, study in humans and causality could not be concluded. Given the public health importance of our hypothesis, we proceeded to identify and biochemically analyse archived vials of c-hGH. Here we show that certain c-hGH batches to which patients with iCJD and Aß pathology were exposed have substantial levels of Aß40, Aß42 and tau proteins, and that this material can seed the formation of Aß plaques and cerebral Aß-amyloid angiopathy in intracerebrally inoculated mice expressing a mutant, humanized amyloid precursor protein. These results confirm the presence of Aß seeds in archived c-hGH vials and are consistent with the hypothesized iatrogenic human transmission of Aß pathology. This experimental confirmation has implications for both the prevention and the treatment of Alzheimer's disease, and should prompt a review of the risk of iatrogenic transmission of Aß seeds by medical and surgical procedures long recognized to pose a risk of accidental prion transmission2,3.


Assuntos
Doença de Alzheimer/induzido quimicamente , Peptídeos beta-Amiloides/metabolismo , Cadáver , Síndrome de Creutzfeldt-Jakob/induzido quimicamente , Contaminação de Medicamentos , Hormônio do Crescimento/farmacologia , Doença Iatrogênica , Doença de Alzheimer/etiologia , Peptídeos beta-Amiloides/análise , Precursor de Proteína beta-Amiloide/administração & dosagem , Precursor de Proteína beta-Amiloide/efeitos adversos , Animais , Estudos de Casos e Controles , Síndrome de Creutzfeldt-Jakob/etiologia , Modelos Animais de Doenças , Transmissão de Doença Infecciosa/prevenção & controle , Transmissão de Doença Infecciosa/estatística & dados numéricos , Contaminação de Medicamentos/prevenção & controle , Contaminação de Medicamentos/estatística & dados numéricos , Feminino , Hormônio do Crescimento/administração & dosagem , Humanos , Masculino , Camundongos , Modelos Biológicos , Príons/metabolismo , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacologia , Reprodutibilidade dos Testes , Proteínas tau/análise , Proteínas tau/metabolismo
9.
Oncologist ; 23(11): 1273-1281, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30108157

RESUMO

BACKGROUND: ß-amyloid precursor protein (APP), a potential target for Alzheimer's disease treatment, has recently been shown to take part in carcinogenesis. Increased APP promotes migration, survival, and proliferation in breast cancer cell lines. We examined the clinical value of APP in breast cancers. A comprehensive examination of clinicopathological features related to APP expression in a large cohort of breast cancers and the corresponding metastatic lymph nodes was performed. APP expression and its prognostic impact in different breast cancer subtypes were examined. RESULTS: APP was highly expressed in nonluminal breast cancers and correlated with features associated with nonluminal breast cancers (including higher grade, the presence of necrosis, and higher proliferative index, growth factor receptor, and basal marker expression). Multivariate Cox hazard analysis demonstrated that APP was an independent adverse prognostic factor of disease-free survival (DFS; hazard ratio [HR], 2.090; p = .013; 95% confidence interval [CI], 1.165-3.748) and breast cancer-specific survival (BCSS; HR, 2.631; p = .002; 95% CI, 1.408-4.915) in the nonluminal group. The independent prognostic impact was also seen in triple negative breast cancers. Interestingly, a higher expression of APP was found in nodal metastasis compared with primary tumor. Such APP upregulation was correlated with further distal metastasis and poorer outcome (DFS: log-rank, 12.848; p < .001; BCSS: log-rank, 13.947; p < .001). CONCLUSION: Our findings provided evidence of oncogenic roles of APP in clinical breast cancers. Patients with positive APP expression, particularly those with APP upregulation in lymph node metastases, may require vigilant monitoring of their disease and more aggressive therapy. IMPLICATIONS FOR PRACTICE: ß-amyloid precursor protein (APP), a potential target for Alzheimer's disease, has recently been implicated in oncogenesis. Here, evidence of its roles in clinical breast cancers is provided. Positive APP expression was found to be an independent prognostic factor in nonluminal cancers, particularly triple negative breast cancers (TNBCs). Interestingly, a higher APP in nodal metastases was associated with distal metastases. TNBCs are heterogeneous and currently have no available target therapy. APP could have therapeutic potential and be used to define the more aggressive cases in TNBCs. Current prognostic analysis is based on primary tumor. The present data suggest that investigation of nodal metastases could provide additional prognostic value.


Assuntos
Agressão/fisiologia , Precursor de Proteína beta-Amiloide/efeitos adversos , Neoplasias da Mama/complicações , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias da Mama/psicologia , Feminino , Humanos , Pessoa de Meia-Idade , Prognóstico , Adulto Jovem
10.
Molecules ; 23(4)2018 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-29642617

RESUMO

Oxidative stress, which is caused by Amyloid-ß deposition in brain, plays an important role in Alzheimer's disease. In this study, we found that lignans from Schisandra chinensis rattan stems (rsSCH-L) could reduce the escape latency and the distance travelled by the Aß1-42 injected rats while the crossing platform time was enhanced in the Morris water maze test. Further research demonstrated that lignans from rsSCH-L attenuated Aß1-42-induced neuronal cell injury by increasing the content of SOD and GSH-Px and decreasing the levels of LDH, ROS, and MDA. Moreover, rsSCH-L also inhibited the apoptosis of primary neuronal cells. The mechanisms of the apoptosis were related with the downregulation of caspase-3, caspase-8, Bax, and upregulation of Bcl-2. Taken together, the results show that rsSCH-L can improve cognitive ability in vivo. Meanwhile rsSCH-L exhibit a neuroprotective environment against oxidative stress and apoptosis in vitro. Therefore, rsSCH-L may be a potential therapeutic agent for this neurodegenerative disease.


Assuntos
Precursor de Proteína beta-Amiloide/efeitos adversos , Lignanas/administração & dosagem , Transtornos da Memória/tratamento farmacológico , Fármacos Neuroprotetores/administração & dosagem , Schisandra/química , Precursor de Proteína beta-Amiloide/administração & dosagem , Animais , Apoptose/efeitos dos fármacos , Modelos Animais de Doenças , Lignanas/química , Lignanas/farmacologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Transtornos da Memória/metabolismo , Neurônios/citologia , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/farmacologia , Caules de Planta/química , Ratos , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/metabolismo
11.
J Alzheimers Dis ; 27(2): 385-91, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21841249

RESUMO

An autosomal dominant mutation in the BRI2/ITM2B gene causes Familial Danish Dementia (FDD). We have generated a mouse model of FDD, called FDDKI, genetically congruous to the human disease. These mice carry one mutant and one wild type Bri2/Itm2b allele, like FDD patients. Analysis of FDDKI mice and samples from human patients has shown that the Danish mutation causes loss of Bri2 protein. FDDKI mice show synaptic plasticity and memory impairments. BRI2 is a physiological interactor of amyloid-ß protein precursor (AßPP), a gene associated with Alzheimer's disease, which inhibits processing of AßPP. AßPP/Bri2 complexes are reduced in synaptic membranes of FDDKI mice. Consequently, AßPP metabolites derived from processing of AßPP by ß-, α-, and γ-secretases are increased in Danish dementia mice. AßPP haplodeficiency prevents memory and synaptic dysfunctions, consistent with a role for AßPP-metabolites in the pathogenesis of memory and synaptic deficits. This genetic suppression provides compelling evidence that AßPP and BRI2 functionally interact. Here, we have investigated whether AßPP processing is altered in FDD patients' brain samples. We find that the levels of several AßPP metabolites, including Aß, are significantly increased in the brain sample derived from an FDD patient. Our data are consistent with the findings in FDDKI mice, and support the hypothesis that the neurological effects of the Danish form of BRI2 are caused by toxic AßPP metabolites, suggesting that Familial Danish and Alzheimer's dementias share common pathogenic mechanisms.


Assuntos
Precursor de Proteína beta-Amiloide/biossíntese , Catarata/metabolismo , Ataxia Cerebelar/metabolismo , Surdez/metabolismo , Demência/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Doença de Alzheimer/etiologia , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/efeitos adversos , Precursor de Proteína beta-Amiloide/genética , Química Encefálica/genética , Catarata/etiologia , Catarata/genética , Ataxia Cerebelar/etiologia , Ataxia Cerebelar/genética , Surdez/etiologia , Surdez/genética , Demência/etiologia , Demência/genética , Humanos , Proteínas de Membrana/efeitos adversos , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Processamento de Proteína Pós-Traducional/genética , Regulação para Cima/genética
12.
Trends Pharmacol Sci ; 27(12): 619-25, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17056129

RESUMO

In recent years, the most common pharmacological treatment for Alzheimer's disease (AD) has been acetylcholinesterase (AChE) inhibition. However, this single-target approach has limited effectiveness and there is evidence that a multitarget approach might be more effective. Huperzine A (HupA), a novel alkaloid isolated from a Chinese herb, has neuroprotective effects that go beyond the inhibition of AChE. Recent data have demonstrated that HupA can ameliorate the learning and memory deficiency in animal models and AD patients. Its potentially beneficial actions include modification of beta-amyloid peptide processing, reduction of oxidative stress, neuronal protection against apoptosis, and regulation of the expression and secretion of nerve growth factor (NGF) and NGF signaling.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Precursor de Proteína beta-Amiloide/efeitos dos fármacos , Fármacos Neuroprotetores/uso terapêutico , Sesquiterpenos/uso terapêutico , Alcaloides , Doença de Alzheimer/fisiopatologia , Precursor de Proteína beta-Amiloide/efeitos adversos , Animais , Apoptose/efeitos dos fármacos , Inibidores da Colinesterase/uso terapêutico , Humanos
13.
Curr Drug Targets ; 5(6): 553-8, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15270201

RESUMO

A growing wealth of evidence indicates that the key pathological event in Alzheimer's disease is the conversion of the normal soluble amyloid-beta peptide into beta-sheet-rich oligomeric structures which have a neurotoxic activity and ability to form insoluble amyloid deposits that accumulate in the brain. Beta-sheet breakers constitute a new class of drugs that are designed to specifically bind amyloid-beta peptide and block and/or reverse this abnormal conformational change. In this article we review this approach, describe diverse compounds reported to have this activity and summarize the data supporting the view that beta-sheet breakers could be serious candidates to combat this devastating disease.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/antagonistas & inibidores , Fragmentos de Peptídeos/antagonistas & inibidores , Fragmentos de Peptídeos/uso terapêutico , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/efeitos adversos , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/efeitos adversos , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Expressão Gênica/genética , Humanos , Modelos Biológicos , Fragmentos de Peptídeos/efeitos adversos , Fragmentos de Peptídeos/metabolismo
14.
Eur J Neurosci ; 19(9): 2356-64, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15128389

RESUMO

The 24-residue peptide Humanin (HN) protects neuronal cells from insults of various Alzheimer's disease (AD) genes and Abeta by forming a homodimer. We have previously shown that P3A, S7A, C8A, L9A, L12A, T13A, S14A and P19A mutations nullify the neuroprotective function of HN [Yamagishi, Y., Hashimoto, Y., Niikura, T. & Nishimoto, I. (2003) Peptides, 24, 585-595]. Here we examined whether any of these 'null' mutants could function as dominant-negative mutants. Homodimerization-defective mutants, P3A-, L12A-, S14A- and P19A-HN, specifically blocked neuroprotection by HN, but not by activity-dependent neurotrophic factor. Furthermore, insertion of S7A, the mutation that blocks the homodimerization of HN, but not insertion of G5A abolished the antagonizing function of L12A-HN. While L12A-HN and G5A/L12A-HN actually inhibited HN homodimerization, S7A/L12A-HN had no effect. These data indicate that P3A-, L12A-, S14A- and P19A-HN function as HN antagonists by forming an inactive dimer with HN. This study provides a novel insight into the understanding of the in vivo function of HN, as well as into the development of clinically applicable HN neutralizers.


Assuntos
Mutação , Fármacos Neuroprotetores/farmacologia , Proteínas/farmacologia , Peptídeos beta-Amiloides/toxicidade , Precursor de Proteína beta-Amiloide/efeitos adversos , Precursor de Proteína beta-Amiloide/genética , Animais , Western Blotting/métodos , Contagem de Células/métodos , Morte Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Córtex Cerebral/citologia , Meios de Cultivo Condicionados/farmacologia , Dimerização , Relação Dose-Resposta a Droga , Interações Medicamentosas , Células Híbridas , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Proteínas do Tecido Nervoso/farmacologia , Neuroblastoma , Neurônios , Neuropeptídeos , Oligopeptídeos , Fragmentos de Peptídeos/toxicidade , Peptídeos/farmacologia , Proteínas/química , Proteínas/genética , Ratos , Transfecção/métodos
15.
Curr Opin Neurol ; 15(5): 525-31, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12351995

RESUMO

PURPOSE OF REVIEW: Sporadic inclusion-body myositis (s-IBM) and hereditary inclusion body myopathies are progressive muscle diseases that lead to severe disability. We discuss recent advances in illuminating their pathogenic mechanism(s). RECENT FINDINGS: We emphasize how different etiologies might lead to the strikingly similar pathology and possibly similar pathogenic cascade. Our basic hypothesis is that over-expression of amyloid-beta precursor protein within aging muscle fibers is an early upstream event causing the subsequent pathogenic cascade. On the basis of our research, several processes seem to be important in relation to the still speculative pathogenesis: (a) increased transcription and accumulation of amyloid-beta precursor protein, and accumulation of its proteolytic fragment Abeta; (b) accumulations of phosphorylated tau and other Alzheimer-related proteins; (c) accumulation of cholesterol and low-density lipoprotein receptors, the cholesterol accumulation possibly due to its abnormal trafficking; (d) oxidative stress; and (e) a milieu of muscle cellular aging in which these changes occur. We discuss unfolded and/or misfolded proteins as a possible mechanism in formation of the inclusion bodies and their consequences. The remarkable pathologic similarities between s-IBM muscle and Alzheimer disease brain are discussed. SUMMARY: Unfolding knowledge of the various pathogenetic aspects of the s-IBMs and hereditary inclusion body myopathies may lead to new therapeutic avenues.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Miosite de Corpos de Inclusão/etiologia , Miosite de Corpos de Inclusão/fisiopatologia , Envelhecimento/genética , Envelhecimento/patologia , Doença de Alzheimer/etiologia , Doença de Alzheimer/fisiopatologia , Precursor de Proteína beta-Amiloide/efeitos adversos , Colesterol/efeitos adversos , Humanos , Miosite de Corpos de Inclusão/genética , Miosite de Corpos de Inclusão/metabolismo , Estresse Oxidativo , Fenótipo , Dobramento de Proteína , Receptores de Lipoproteínas/metabolismo , Regulação para Cima , Proteínas tau/efeitos adversos
16.
J Neurosci ; 22(9): 3445-53, 2002 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-11978821

RESUMO

In the brain of Alzheimer's disease (AD) patients, neurotoxic amyloid peptides accumulate and are deposited as senile plaques. A major therapeutic strategy aims to decrease production of amyloid peptides by inhibition of gamma-secretase. Presenilins are polytopic transmembrane proteins that are essential for gamma-secretase activity during development and in amyloid production. By loxP/Cre-recombinase-mediated deletion, we generated mice with postnatal, neuron-specific presenilin-1 (PS1) deficiency, denoted PS1(n-/-), that were viable and fertile, with normal brain morphology. In adult PS1(n-/-) mice, levels of endogenous brain amyloid peptides were strongly decreased, concomitant with accumulation of amyloid precursor protein (APP) C-terminal fragments. In the cross of APP[V717I]xPS1 (n-/-) double transgenic mice, the neuronal absence of PS1 effectively prevented amyloid pathology, even in mice that were 18 months old. This contrasted sharply with APP[V717I] single transgenic mice that all develop amyloid pathology at the age of 10-12 months. In APP[V717I]xPS1 (n-/-) mice, long-term potentiation (LTP) was practically rescued at the end of the 2 hr observation period, again contrasting sharply with the strongly impaired LTP in APP[V717I] mice. The findings demonstrate the critical involvement of amyloid peptides in defective LTP in APP transgenic mice. Although these data open perspectives for therapy of AD by gamma-secretase inhibition, the neuronal absence of PS1 failed to rescue the cognitive defect, assessed by the object recognition test, of the parent APP[V717I] transgenic mice. This points to potentially detrimental effects of accumulating APP C99 fragments and demands further study of the consequences of inhibition of gamma-secretase activity. In addition, our data highlight the complex functional relation of APP and PS1 to cognition and neuronal plasticity in adult and aging brain.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Transtornos Cognitivos/fisiopatologia , Hipocampo/metabolismo , Proteínas de Membrana/deficiência , Neurônios/metabolismo , Placa Amiloide/metabolismo , Doença de Alzheimer/etiologia , Secretases da Proteína Precursora do Amiloide , Precursor de Proteína beta-Amiloide/efeitos adversos , Precursor de Proteína beta-Amiloide/genética , Animais , Ácido Aspártico Endopeptidases , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Encéfalo/anatomia & histologia , Encéfalo/metabolismo , Transtornos Cognitivos/genética , Transtornos Cognitivos/patologia , Cruzamentos Genéticos , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Estimulação Elétrica , Endopeptidases/metabolismo , Hipocampo/patologia , Técnicas In Vitro , Peptídeos e Proteínas de Sinalização Intracelular , Potenciação de Longa Duração/fisiologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Plasticidade Neuronal/genética , Neurônios/citologia , Neurônios/efeitos dos fármacos , Fragmentos de Peptídeos/metabolismo , Fenótipo , Placa Amiloide/patologia , Presenilina-1 , RNA Mensageiro/metabolismo , Reconhecimento Psicológico , Proteínas Repressoras/metabolismo
18.
Dement Geriatr Cogn Disord ; 12(1): 40-51, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11125240

RESUMO

The membrane-bound glycoprotein, amyloid precursor protein (APP), plays a central role in Alzheimer's disease (AD). The present paper investigates the neuritogenic-neurotoxic properties of this protein and relates them to possible aetiopathological mechanisms in AD. Marked differences in neuritic differentiation were detected when comparing untransfected tetraploid mouse neuroblastoma cells (or vector only cells) with transfected cells overexpressing APP(751). Transfected cells developed neurites quicker, and whereas all transfected cells differentiated, the degree of differentiation of untransfected cells was more variable. Fully differentiated transfected and untransfected cells had marked differences in neuritic morphology. Transfected cells had more neurites per cell, these being shorter and more branched than neurites on untransfected cells. The precocious differentiation of transfected cells was not maintained with neuritic process disintegration and cell death occurring from the seventh day onwards. Untransfected cells continued to extend their neuritic processes for up to five weeks. Membrane-associated forms of APP were responsible for these effects, rather than secreted APP or the beta/A4-peptide. Combined data from Western blot and immunocytochemical procedures showed a prominent accumulation of APP-C-terminal fragments in the perinuclear region, neuritic varicosities and growth cones of transfected cells, suggesting their involvement in the neuritogenic-neurotoxic process. Similar neuritogenic-neurotoxic mechanisms occurring in vivo, in association with compensatory synaptoplastic responses in the aged brain, may be part of the pathological process in AD.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/metabolismo , Glicoproteínas de Membrana/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Neurotoxinas/metabolismo , Doença de Alzheimer/etiologia , Precursor de Proteína beta-Amiloide/efeitos adversos , Precursor de Proteína beta-Amiloide/genética , Animais , Western Blotting , Diferenciação Celular , Divisão Celular , Linhagem Celular , Imuno-Histoquímica , Técnicas In Vitro , Glicoproteínas de Membrana/efeitos adversos , Camundongos , Neuroblastoma/genética , Neurotoxinas/efeitos adversos , Transfecção
19.
Acta Neuropathol ; 92(2): 130-7, 1996 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8841658

RESUMO

Abnormal processing of beta-amyloid precursor protein (APP) and precipitation of amyloidogenic beta A4 peptide have been implicated in the pathogenesis of Alzheimer's disease (AD). In an attempt to generate an in vivo model of APP-associated pathology, we have introduced recombinant retroviral vectors harboring normal and mutant cDNAs for human neuron-specific APP 695 into fetal rat brain transplants. A minor population of recombinant neural cells with expression of APP was identified immunohistochemically in the grafts. The expression was maintained during an observation period of 9 months. Neurons with strong immunoreactivity for human APP exhibited markedly swollen perikarya and showed signs of cell degeneration. At 6 months post-transplantation, recombinant grafts developed APP-positive neuropil deposits with morphological features of neuritic plaques, but without beta A4 peptide immunoreactivity. No difference was observed between wild-type APP and the mutant APP construct derived from a family with autosomal dominant AD. These observations indicate that long-term neuronal overexpression of human APP has the potential to generate APP plaques in the rodent brain.


Assuntos
Precursor de Proteína beta-Amiloide/efeitos adversos , Transplante de Tecido Encefálico/patologia , Emaranhados Neurofibrilares/patologia , Precursor de Proteína beta-Amiloide/biossíntese , Precursor de Proteína beta-Amiloide/genética , Animais , Vetores Genéticos/metabolismo , Humanos , Imuno-Histoquímica , Mutação , Ratos , Ratos Endogâmicos F344 , Retroviridae/metabolismo , Técnicas Estereotáxicas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...