Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Med Food ; 26(7): 462-469, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37358589

RESUMO

Diabetes is highly linked to the occurrence of Alzheimer disease (AD), which is characterized by beta amyloid peptide (Aß) and hyperphosphorylation of tau (p-tau), and neuron damage particularly in hippocampus. Type 2 diabetes (T2D) is featured by insulin resistance, and phosphorylation of Ser307-IRS-1 is regarded as a resistance marker. Inhibitors of dipeptidyl peptidase-4 (DPP-4) are effective tools for treating T2D. Previously, we reported subfractions of Abelmoschus esculentus (AE, okra) (F1 rich in quercetin glycosides; F2 composed of polysaccharide) attenuated DPP-4 and its downstream signals of insulin resistance, thus preventing Aß-induced neuron damage. Since autophagy could be protective, we now explore if AE works to modulate neuron autophagy by regulating DPP-4 and insulin resistance and, thus, improves the hippocampal function and behavior. We demonstrated that AE subfractions attenuate Aß-induced insulin resistance and the expression of p-tau and normalize the autophagy and survival of hippocampal neurons. The action of AE may be attributed to the downregulation of DPP-4, which plays a critical role in mediating insulin resistance and hinders neuron autophagy. The in vivo findings reveal that the hippocampal insulin resistance appears to link with loss of memory, reduction of curiosity, and depression, whereas treatment with AE significantly improves the insulin sensitivity and hippocampal function. Noteworthy, even at only 5 µg/mL, F2 seems to exhibit a meaningful effect. In conclusion, we suggest that AE attenuates insulin resistance and recovers neuron autophagy which are regulated by DPP-4, thus preventing the damage to the hippocampus, improving recognition and emotion. AE may be an effective adjuvant or supplement to prevent insulin resistance-associated pathogenesis of AD if these results can be confirmed in human clinical trials.


Assuntos
Abelmoschus , Doença de Alzheimer , Diabetes Mellitus Tipo 2 , Resistência à Insulina , Humanos , Diabetes Mellitus Tipo 2/tratamento farmacológico , Dipeptidil Peptidase 4 , Doença de Alzheimer/tratamento farmacológico , Autofagia , Hipocampo , Neurônios
2.
PLoS One ; 17(3): e0265444, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35290413

RESUMO

Nonalcoholic fatty liver disease (NAFLD) is recognized as the liver component of metabolic syndrome. The regulation of hepatic lipid should be emphasized to prevent accompanying illness. As AMP-activated protein kinase (AMPK) and sterol regulatory element binding protein (SREBP) regulate lipid metabolism, CD36 and fatty acid synthase (FAS) promote lipid uptake and lipogenesis respectively, while acetyl-CoA carboxylase (ACC) is an indicator of negative feedback. The increase of IRS-1 phosphorylation at the residue ser307 (p-ser307-IRS-1) and decrease of p-ser473-Akt (p-Akt) are viewed as the insulin resistance markers, and our previous reports suggested dipeptidyl peptidase-4 (DPP-4) mediates insulin resistance, the crucial factor of metabolic syndrome. Abelmoschus esculentus (AE) fruit is well-known for its antidiabetic utility. We had isolated several AE subfractions by successive steps, and found that F1 and F2 were especially valid in suppressing DPP-4 signaling. Since little is known if AE works on NAFLD, now we first attempt to investigate whether AE is useful to attenuate hepatic lipogenesis and lipid uptake in liver cells, along with improving the metabolic targets. We demonstrated that AE subfractions attenuated the hepatic lipid accumulation induced by free fatty acids. Treatment of AE alleviated FAS and returned the level of p-ser79-ACC (p-ACC). Although F1 was more effective on AMPK, F2 seemed more stable to attenuate SREBP-1. Moreover, as fatty acids stimulated the expression of CD36, F2 showed a superior effect to down-regulate the lipid uptake. Both AE subfractions reduced the generation of ROS, decreased the level of p-ser307-IRS-1, and restored the expression of p-Akt. Moreover, treatment of DPP-4 inhibitor linagliptin revealed that, AE could prevent the hepatic lipogenesis, oxidative burden, and the related insulin resistance via downregulating DPP-4. In conclusion, the present investigation revealed that AE, especially F2, is potential to be developed as adjuvant to prevent NAFLD.


Assuntos
Abelmoschus , Resistência à Insulina , Síndrome Metabólica , Hepatopatia Gordurosa não Alcoólica , Proteínas Quinases Ativadas por AMP/metabolismo , Abelmoschus/química , Acetil-CoA Carboxilase/metabolismo , Ácidos Graxos não Esterificados/metabolismo , Metabolismo dos Lipídeos , Lipogênese , Fígado/metabolismo , Síndrome Metabólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Hepatopatia Gordurosa não Alcoólica/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo
3.
Integr Cancer Ther ; 20: 15347354211044833, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34477012

RESUMO

BACKGROUND: Subjects with head and neck cancer (HNC) often experience post-treatment side effects, particularly radiation-induced oral mucositis (RIOM). This study aimed to explore the association of Chinese herbal medicine use with the sequent risk of RIOM among them. METHODS: This cohort study used a nationwide health insurance database to identify subjects newly diagnosed with HNC, aged 20 to 60 years, who received treatment between 2000 and 2007. Among them, a total of 561 cases received CHM after HNC onset (CHM users); the remaining 2395 cases were non-CHM users. All patients were followed to the end of 2012 to identify any treatment for RIOM as the end point. Cox proportional hazards regression was used to compute the adjusted hazard ratio (aHR) of RIOM by CHM use. RESULTS: During the follow-up period, 183 CHM users and 989 non-CHM users developed RIOM at incidence rates of 40.98 and 57.91 per 1000 person-years, respectively. CHM users had a lower RIOM risk than the non-CHM users (aHR: 0.68; 95% Confidence Interval: 0.58-0.80). The most potent effect was observed in those taking CHM for more than 1 year. Use of Baizhi, Danshen, Shao-Yao-Gan-Cao-Tang, Gan-Lu-Yin, Huangqin, Shu-Jing-Huo-Xue-Tang, and Xin-Yi-Qing-Fei-Tang, was significantly related to a lower risk of RIOM. CONCLUSION: Findings of this study indicated that adding CHM to conventional clinical care could be helpful in protecting those with HNC against the onset of RIOM. Further clinical and mechanistic studies are warranted.


Assuntos
Medicamentos de Ervas Chinesas , Neoplasias de Cabeça e Pescoço , Lesões por Radiação/tratamento farmacológico , Estomatite , Estudos de Coortes , Medicamentos de Ervas Chinesas/uso terapêutico , Neoplasias de Cabeça e Pescoço/radioterapia , Humanos , Incidência , Medicina Tradicional Chinesa , Estomatite/tratamento farmacológico , Estomatite/etiologia , Taiwan
4.
BMC Complement Med Ther ; 20(1): 370, 2020 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-33267804

RESUMO

BACKGROUND: Insulin resistance could be associated with the development of Alzheimer disease (AD). The neuropathological hallmarks of AD are beta amyloid (Aß) produced from sequential cleavage initiated by ß-secretase and degraded by insulin degradation enzyme (IDE), as well as hyperphosphorylation of tau (p-tau). Insulin action involves the cascades of insulin receptor substrates (IRS) and phosphatidylinositol 3-kinase (PI3K), while phosphorylation of IRS-1 at ser307 (p-ser307IRS-1) hinders the response. Our previous report suggested dipeptidyl peptidase-4 (DPP-4) is crucial to insulin resistance, and the subfractions of Abelmoschus esculentus (AE), F1 and F2, attenuate the signaling. Here we aim to investigate whether AE works to reduce Aß generation via regulating DPP4 and insulin resistance. METHODS: The subfractions F1 and F2 were prepared according to a succession of procedures. F1 was composed by quercetin glycosides and triterpene ester, and F2 contained a large amount of polysaccharides. The in vitro insulin resistance model was established by SK-N-MC cell line treated with palmitate. MTT was used to define the dose range, and thereby Western blot, ELISA, and the activity assay were used to detect the putative markers. One-way ANOVA was performed for the statistical analysis. RESULTS: Treatment of palmitate induced the level of p-ser307IRS-1. Both F1 and F2 effectively decrease p-ser307IRS-1, and recover the expression of p-PI3K. However, the expression of total IRS plunged with 25 µg/mL of F1, while descended steadily with 5 µg/mL of F2. As palmitate increased the levels of Aß40 and Aß42, both AE subfractions were effective to reduce Aß generation of and ß-secretase activity, but IDE was not altered in any treatment conditions. The expression of DPP4 was also accompanied with insulin resistance signals. Inhibition of DPP4 attenuated the activity of ß-secretase and production of Aß. Moreover, the present data revealed that both AE subfractions significantly decrease the level of p-Tau. CONCLUSIONS: In conclusion, we demonstrated that AE would be a potential adjuvant to prevent insulin resistance and the associated pathogenesis of AD, and F2 seems more feasible to be developed.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Dipeptidil Peptidase 4/metabolismo , Resistência à Insulina , Extratos Vegetais/farmacologia , Proteínas tau/metabolismo , Abelmoschus , Doença de Alzheimer/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Frutas , Humanos , Taiwan
5.
Cells ; 9(6)2020 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-32471175

RESUMO

Amyloid ß (Aß) is a peptide fragment of the amyloid precursor protein that triggers the progression of Alzheimer's Disease (AD). It is believed that Aß contributes to neurodegeneration in several ways, including mitochondria dysfunction, oxidative stress and brain insulin resistance. Therefore, protecting neurons from Aß-induced neurotoxicity is an effective strategy for attenuating AD pathogenesis. Recently, applications of stem cell-based therapies have demonstrated the ability to reduce the progression and outcome of neurodegenerative diseases. Particularly, Nanog is recognized as a stem cell-related pluripotency factor that enhances self-renewing capacities and helps reduce the senescent phenotypes of aged neuronal cells. However, whether the upregulation of Nanog can be an effective approach to alleviate Aß-induced neurotoxicity and senescence is not yet understood. In the present study, we transiently overexpressed Nanog-both in vitro and in vivo-and investigated the protective effects and underlying mechanisms against Aß. We found that overexpression of Nanog is responsible for attenuating Aß-triggered neuronal insulin resistance, which restores cell survival through reducing intracellular mitochondrial superoxide accumulation and cellular senescence. In addition, upregulation of Nanog expression appears to increase secretion of neurotrophic factors through activation of the Nrf2 antioxidant defense pathway. Furthermore, improvement of memory and learning were also observed in rat model of Aß neurotoxicity mediated by upregulation of Nanog in the brain. Taken together, our study suggests a potential role for Nanog in attenuating the neurotoxic effects of Aß, which in turn, suggests that strategies to enhance Nanog expression may be used as a novel intervention for reducing Aß neurotoxicity in the AD brain.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Resistência à Insulina , Proteína Homeobox Nanog/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Estresse Oxidativo/efeitos dos fármacos , Células-Tronco Pluripotentes/metabolismo , Animais , Apoptose/efeitos dos fármacos , Encéfalo/patologia , Linhagem Celular Tumoral , Senescência Celular/efeitos dos fármacos , Transtornos Cognitivos/complicações , Transtornos Cognitivos/patologia , Humanos , Insulina/metabolismo , Masculino , Transtornos da Memória/complicações , Transtornos da Memória/patologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Neurônios/efeitos dos fármacos , Neuroproteção/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Superóxidos/metabolismo , Regulação para Cima/efeitos dos fármacos , Proteínas tau/metabolismo
6.
PLoS One ; 14(6): e0217400, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31237881

RESUMO

The association of Alzheimer disease (AD) and Diabetes (DM) is less clear. Accumulation of beta amyloid (Aß) and presence of hyperphosphorylated tau (p-tau) are hallmarks of AD, spreading in the region where insulin receptors are also found. Aß exerts neuron toxicity, and could disturb insulin signaling of phosphatidylinositol 3-kinase (PI3K), glycogen synthase kinase (GSK)-3ß and AMP-activated protein kinase (AMPK), but increase IRS-1-Ser307 phosphorylation which is viewed as insulin resistance marker. Previously we reported dipeptidyl peptidase-4 (DPP-4) mediate insulin resistance signals, and Abelmoschus esculentus (AE) subfractions F1 (rich in quercetin glucosides and triterpene ester) and F2 (containing large amount of polysaccharides) attenuate DPP-4-mediated apoptosis. In the present study, we aim to investigate if Aß induce neuron death by regulating DPP-4 and insulin resistance signals, and the putative effect of F1 and F2. By MTT, microscopy, and Western blotting, we demonstrate treatment of appropriate doses of AE subfractions prevent Aß-induced neuron apoptosis. F1 attenuate Aß-induced caspase 3 expression especially at 25 µg/mL, while F2 attenuate caspase 3 activation even at the low dose of 1 µg/mL. Both AE subfractions decrease Aß-enhanced DPP-4, but increase Aß-reduced p-AMPK and p-PI3K. The activity analysis reveals that F2 is more valid than F1 to reduce DPP-4 activity. The inhibition of DPP-4 demonstrates it plays the pivotal role in Aß-induced neuron apoptosis. Moreover, although both F1 and F2 are effective to inhibit p-IRS-1-Ser307, F2 takes advantage to reduce p-Tau while F1 is superior to enhance p-GSK-3ß. This implies AE subfractions act on different targets, and could be developed respectively. In conclusion, we demonstrate AE is potential to prevent Aß-induced neuron damage by regulating DPP-4 and the insulin resistance cascades. AE could be an adjuvant to protect neuron degenerative disease related to Aß and insulin resistance.


Assuntos
Abelmoschus/química , Peptídeos beta-Amiloides/metabolismo , Apoptose/efeitos dos fármacos , Dipeptidil Peptidase 4/metabolismo , Resistência à Insulina , Neurônios/metabolismo , Extratos Vegetais/farmacologia , Transdução de Sinais/efeitos dos fármacos , Proteínas Quinases Ativadas por AMP/metabolismo , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Linhagem Celular , Diabetes Mellitus/tratamento farmacológico , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patologia , Humanos , Neurônios/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Extratos Vegetais/química
7.
Int J Mol Sci ; 19(9)2018 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-30149534

RESUMO

Huntington's disease (HD) is a progressive and fatal neurodegenerative disease caused by CAG repeat expansion in the coding region of huntingtin (HTT) protein. The accumulation of mutant HTT (mHTT) contributes to neurotoxicity by causing autophagy defects and oxidative stress that ultimately lead to neuronal death. Interestingly, epidemiologic studies have demonstrated that the prevalence of type-2 diabetes, a metabolic disease mainly caused by defective insulin signaling, is higher in patients with HD than in healthy controls. Although the precise mechanisms of mHTT-mediated toxicity remain unclear, the blockade of brain insulin signaling may initiate or exacerbate mHTT-induced neurodegeneration. In this study, we used an in vitro HD model to investigate whether neuronal insulin signaling is involved in mHTT-mediated neurotoxicity. Our results demonstrated that mHTT overexpression significantly impairs insulin signaling and causes apoptosis in neuronal cells. However, treatment with liraglutide, a GLP-1 analogue, markedly restores insulin sensitivity and enhances cell viability. This neuroprotective effect may be attributed to the contribution of the upregulated expression of genes associated with endogenous antioxidant pathways to oxidative stress reduction. In addition, liraglutide stimulates autophagy through AMPK activation, which attenuates the accumulation of HTT aggregates within neuronal cells. Our findings collectively suggest that liraglutide can rescue impaired insulin signaling caused by mHTT and that GLP-1 may potentially reduce mHTT-induced neurotoxicity in the pathogenesis of HD.


Assuntos
Proteína Huntingtina/genética , Insulina/metabolismo , Liraglutida/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fármacos Neuroprotetores/farmacologia , Transdução de Sinais/efeitos dos fármacos , Linhagem Celular Tumoral , Peptídeo 1 Semelhante ao Glucagon/análogos & derivados , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Humanos , Proteína Huntingtina/metabolismo , Hipoglicemiantes/farmacologia , Imuno-Histoquímica
8.
Metab Brain Dis ; 32(6): 1999-2007, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28840430

RESUMO

Statins or HMG-CoA reductase inhibitors have been shown to be effective at lowering cholesterol levels, and the application of these molecules has gradually emerged as an attractive therapeutic strategy for neurodegenerative diseases. Epidemiological studies suggest that statin use is associated with a decreased incidence of Alzheimer's disease (AD). Thus, statins may play a beneficial role in reducing amyloid ß (Aß) toxicity, the most relevant pathological feature and pathogenesis of AD. However, the precise mechanisms involved in statin-inhibited Aß toxicity remain unclear. In the present study, we report that mevastatin significantly protects against Aß-induced neurotoxicity in SK-N-MC neuronal cells by restoring impaired insulin signaling. This protection appears to be associated with the activation of AMP-activated protein kinase (AMPK), which has long been known to increase insulin sensitivity. Our results also indicate that high levels of cholesterol likely underlie Aß-induced neurotoxicity and that activation of AMPK by mevastatin alleviates insulin resistance. Signaling through the insulin receptor substrate-1/Akt pathway appears to lead to cell survival. These findings demonstrate that mevastatin plays a potential therapeutic role in targeting Aß-mediated neurotoxicity. The molecule presents a novel therapeutic strategy for further studies in AD prevention and therapeutics.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Peptídeos beta-Amiloides/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Lovastatina/análogos & derivados , Neurônios/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Lovastatina/farmacologia , Neurônios/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos
9.
J Alzheimers Dis ; 50(4): 1083-98, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26890744

RESUMO

Deficiency of insulin signaling has been linked to diabetes and ageing-related neurodegenerative diseases such as Alzheimer's disease (AD). In this regard, brains exhibit defective insulin receptor substrate-1 (IRS-1) and hence result in alteration of insulin signaling in progression of AD, the most common cause of dementia. Consequently, dysregulation of insulin signaling plays an important role in amyloid-ß (Aß)-induced neurotoxicity. As the derivation of induced pluripotent stem cells (iPSC) involves cell reprogramming, it may provide a means for regaining the control of ageing-associated dysfunction and neurodegeneration via affecting insulin-related signaling. To this, we found that an embryonic stem cell (ESC)-specific microRNA, miR-302, silences phosphatase and tensin homolog (PTEN) to activate Akt signaling, which subsequently stimulates nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) elevation and hence inhibits Aß-induced neurotoxicity. miR-302 is predominantly expressed in iPSCs and is known to regulate several important biological processes of anti-oxidative stress, anti-apoptosis, and anti-aging through activating Akt signaling. In addition, we also found that miR-302-mediated Akt signaling further stimulates Nanog expression to suppress Aß-induced p-Ser307 IRS-1 expression and thus enhances tyrosine phosphorylation and p-Ser 473-Akt/p-Ser 9-GSK3ß formation. Furthermore, our in vivo studies revealed that the mRNA expression levels of both Nanog and miR-302-encoding LARP7 genes were significantly reduced in AD patients' blood cells, providing a novel diagnosis marker for AD. Taken together, our findings demonstrated that miR-302 is able to inhibit Aß-induced cytotoxicity via activating Akt signaling to upregulate Nrf2 and Nanog expressions, leading to a marked restoration of insulin signaling in AD neurons.


Assuntos
Peptídeos beta-Amiloides/toxicidade , MicroRNAs/metabolismo , Neurônios/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Idoso , Idoso de 80 Anos ou mais , Linhagem Celular Tumoral , Sobrevivência Celular/fisiologia , Feminino , Glicogênio Sintase Quinase 3 beta/metabolismo , Heme Oxigenase-1/metabolismo , Humanos , Insulina/metabolismo , Masculino , Potencial da Membrana Mitocondrial/fisiologia , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo/fisiologia , PTEN Fosfo-Hidrolase/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Ribonucleoproteínas/metabolismo
10.
Exp Neurol ; 275 Pt 1: 84-98, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26515689

RESUMO

Dementia with Lewy bodies (DLB) is a neurodegenerative disorder caused by abnormal accumulation of Lewy bodies, which are intracellular deposits composed primarily of aggregated α-synuclein (αSyn). Although αSyn has been strongly implicated to induce neurotoxicity, overexpression of wild-type αSyn is shown to be insufficient to trigger formation of protein aggregates by itself. Therefore, investigating the possible mechanism underlying αSyn aggregation is essential to understand the pathogenesis of DLB. Previous studies have demonstrated that amyloid ß (Aß), the primary cause of Alzheimer's disease (AD), may promote the formation of αSyn inclusion bodies. However, it remains unclear how Aß contributes to the deposition and neurotoxicity of αSyn. In the present study, we investigated the cytotoxic effects of Aß in αSyn-overexpressed neuronal cells. Our results showed that Aß inhibits autophagy and enhances αSyn aggregation in αSyn-overexpressed cells. Moreover, Aß also reduced sirtuin 1 (Sirt1) and its downstream signaling, resulting in increased intracellular ROS accumulation and mitochondrial dysfunction. Our in vitro and in vivo studies support that Aß-inhibition of AMP-activated protein kinase (AMPK) signaling is involved in the neurotoxic effects of αSyn. Taken together, our findings suggest that Aß plays a synergistic role in αSyn aggregation and cytotoxicity, which may provide a novel understanding for exploring the underlying molecular mechanism of DLB.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Peptídeos beta-Amiloides/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , alfa-Sinucleína/metabolismo , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/fisiologia , Humanos , Doença por Corpos de Lewy/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Potencial da Membrana Mitocondrial/fisiologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Neurônios/metabolismo , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , alfa-Sinucleína/genética
11.
Chem Biol Interact ; 240: 12-21, 2015 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-26271894

RESUMO

Amyloid ß (Aß) peptides are identified in cause of neurodegenerative diseases such as Alzheimer's disease (AD). Previous evidence suggests Aß-induced neurotoxicity is linked to the stimulation of reactive oxygen species (ROS) production. The accumulation of Aß-induced ROS leads to increased mitochondrial dysfunction and triggers apoptotic cell death. This suggests antioxidant therapies may be beneficial for preventing ROS-related diseases such as AD. Recently, hydrogen-rich water (HRW) has been proven effective in treating oxidative stress-induced disorders because of its ROS-scavenging abilities. However, the precise molecular mechanisms whereby HRW prevents neuronal death are still unclear. In the present study, we evaluated the putative pathways by which HRW protects against Aß-induced cytotoxicity. Our results indicated that HRW directly counteracts oxidative damage by neutralizing excessive ROS, leading to the alleviation of Aß-induced cell death. In addition, HRW also stimulated AMP-activated protein kinase (AMPK) in a sirtuin 1 (Sirt1)-dependent pathway, which upregulates forkhead box protein O3a (FoxO3a) downstream antioxidant response and diminishes Aß-induced mitochondrial potential loss and oxidative stress. Taken together, our findings suggest that HRW may have potential therapeutic value to inhibit Aß-induced neurotoxicity.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Peptídeos beta-Amiloides/toxicidade , Fatores de Transcrição Forkhead/genética , Hidrogênio/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Sirtuína 1/genética , Água , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/metabolismo , Sequestradores de Radicais Livres/farmacologia , Humanos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Sirtuína 1/metabolismo , Água/química , Água/farmacologia
12.
Int J Mol Sci ; 16(5): 10426-42, 2015 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-25961951

RESUMO

Humic acid (HA) is a possible etiological factor associated with for several vascular diseases. It is known that vascular risk factors can directly increase the susceptibility to Alzheimer's disease (AD), which is a neurodegenerative disorder due to accumulation of amyloid ß (Aß) peptide in the brain. However, the role that HA contributes to Aß-induced cytotoxicity has not been demonstrated. In the present study, we demonstrate that HA exhibits a synergistic effect enhancing Aß-induced cytotoxicity in cultured human SK-N-MC neuronal cells. Furthermore, this deterioration was mediated through the activation of endoplasmic reticulum (ER) stress by stimulating PERK and eIF2α phosphorylation. We also observed HA and Aß-induced cytotoxicity is associated with mitochondrial dysfunction caused by down-regulation of the Sirt1/PGC1α pathway, while in contrast, treating the cells with the ER stress inhibitor Salubrinal, or over-expression of Sirt1 significantly reduced loss of cell viability by HA and Aß. Our findings suggest a new mechanism by which HA can deteriorate Aß-induced cytotoxicity through modulation of ER stress, which may provide significant insights into the pathogenesis of AD co-occurring with vascular injury.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Estresse do Retículo Endoplasmático , Substâncias Húmicas/toxicidade , Neurônios/efeitos dos fármacos , Linhagem Celular Tumoral , Cinamatos/farmacologia , Humanos , Neurônios/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Sirtuína 1/metabolismo , Tioureia/análogos & derivados , Tioureia/farmacologia , Fatores de Transcrição/metabolismo , eIF-2 Quinase/metabolismo
13.
CNS Neurosci Ther ; 21(7): 549-57, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26010513

RESUMO

AIM: It is now clear that insulin signaling has important roles in regulation of neuronal functions in the brain. Dysregulation of brain insulin signaling has been linked to neurodegenerative disease, particularly Alzheimer's disease (AD). In this regard, there is evidence that improvement of neuronal insulin signaling has neuroprotective activity against amyloid ß (Aß)-induced neurotoxicity for patients with AD. Linagliptin is an inhibitor of dipeptidylpeptidase-4 (DPP-4), which improves impaired insulin secretion and insulin downstream signaling in the in peripheral tissues. However, whether the protective effects of linagliptin involved in Aß-mediated neurotoxicity have not yet been investigated. METHODS: In the present study, we evaluated the mechanisms by which linagliptin protects against Aß-induced impaired insulin signaling and cytotoxicity in cultured SK-N-MC human neuronal cells. RESULTS: Our results showed that Aß impairs insulin signaling and causes cell death. However, linagliptin significantly protected against Aß-induced cytotoxicity, and prevented the activation of glycogen synthase kinase 3ß (GSK3ß) and tau hyperphosphorylation by restoring insulin downstream signaling. Furthermore, linagliptin alleviated Aß-induced mitochondrial dysfunction and intracellular ROS generation, which may be due to the activation of 5' AMP-activated protein kinase (AMPK)-Sirt1 signaling. This upregulation of Sirt1 expression was also observed in diabetic patients with AD coadministration of linagliptin. CONCLUSIONS: Taken together, our findings suggest linagliptin can restore the impaired insulin signaling caused by Aß in neuronal cells, suggesting DPP-4 inhibitors may have therapeutic potential for reducing Aß-induced impairment of insulin signaling and neurotoxicity in AD pathogenesis.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Peptídeos beta-Amiloides/toxicidade , Inibidores da Dipeptidil Peptidase IV/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Linagliptina/farmacologia , Fragmentos de Peptídeos/toxicidade , Proteínas Quinases Ativadas por AMP/genética , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/complicações , Doença de Alzheimer/tratamento farmacológico , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cromonas/farmacologia , Diabetes Mellitus/tratamento farmacológico , Diabetes Mellitus/fisiopatologia , Inibidores Enzimáticos/farmacologia , Feminino , Peptídeo 1 Semelhante ao Glucagon/genética , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/metabolismo , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Morfolinas/farmacologia , Neuroblastoma/patologia , RNA Mensageiro/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Sirtuína 1/metabolismo , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA