Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Bioengineering (Basel) ; 11(6)2024 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-38927787

RESUMO

BACKGROUND: Acute coronary syndrome (ACS) is a severe cardiovascular disease with globally rising incidence and mortality rates. Traditional risk assessment tools are widely used but are limited due to the complexity of the data. METHODS: This study introduces a gated Transformer model utilizing machine learning to analyze electronic health records (EHRs) for an enhanced prediction of major adverse cardiovascular events (MACEs) in ACS patients. The model's efficacy was evaluated using metrics such as area under the curve (AUC), precision-recall (PR), and F1-scores. Additionally, a patient management platform was developed to facilitate personalized treatment strategies. RESULTS: Incorporating a gating mechanism substantially improved the Transformer model's performance, especially in identifying true-positive cases. The TabTransformer+Gate model demonstrated an AUC of 0.836, a 14% increase in average precision (AP), and a 6.2% enhancement in accuracy, significantly outperforming other deep learning approaches. The patient management platform enabled healthcare professionals to effectively assess patient risks and tailor treatments, improving patient outcomes and quality of life. CONCLUSION: The integration of a gating mechanism within the Transformer model markedly increases the accuracy of MACE risk predictions in ACS patients, optimizes personalized treatment, and presents a novel approach for advancing clinical practice and research.

2.
CNS Neurosci Ther ; 30(3): e14432, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-37641913

RESUMO

AIMS: The autophagy-lysosomal pathway is important for maintaining cellular proteostasis, while dysfunction of this pathway has been suggested to drive the aberrant intraneuronal accumulation of tau protein, leading to synaptic damage and cognitive impairment. Previous studies have demonstrated that the activation of transient receptor potential vanilloid 1 (TRPV1) by capsaicin has a positive impact on cognition and AD-related biomarkers. However, the effect and mechanism of TPRV1 activation on neuronal tau homeostasis remain elusive. METHODS: A mouse model of tauopathy was established by overexpressing full-length human tau in the CA3 area. Mice were fed capsaicin diet (0.0125%) or normal diet for 9 weeks. The cognitive ability, synaptic function, tau phosphorylation levels, and autophagy markers were detected. In vitro, capsaicin-induced alterations in cellular autophagy and tau degradation were characterized using two cell models. Besides, various inhibitors were applied to validate the role of TRPV1-mediated autophagy enhancement in tau clearance. RESULTS: We observed that TRPV1 activation by capsaicin effectively mitigates hippocampal tau accumulation-induced synaptic damages, gliosis, and cognitive impairment in vivo. Capsaicin promotes the degradation of abnormally accumulated tau through enhancing autophagic function in neurons, which is dependent on TRPV1-mediated activation of AMP-activated protein kinase (AMPK) and subsequent inhibition of the mammalian target of rapamycin (mTOR). Blocking AMPK activation abolishes capsaicin-induced autophagy enhancement and tau degradation in neurons. CONCLUSION: Our findings reveal that capsaicin-induced TRPV1 activation confers neuroprotection by restoring neuronal tau homeostasis via modulating cellular autophagy and provides additional evidence to support the potential of TRPV1 as a therapeutic target for tauopathies.


Assuntos
Antineoplásicos , Disfunção Cognitiva , Animais , Humanos , Camundongos , Proteínas Quinases Ativadas por AMP/metabolismo , Autofagia , Capsaicina/farmacologia , Disfunção Cognitiva/tratamento farmacológico , Mamíferos/metabolismo , Proteínas tau/metabolismo , Canais de Cátion TRPV/metabolismo
3.
IEEE J Biomed Health Inform ; 28(3): 1656-1667, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38117618

RESUMO

Type 2 diabetes (T2D) is a worldwide chronic disease that is difficult to cure and causes a heavy social burden. Early prediction of T2D can effectively identify high-risk populations and facilitate earlier implementation of appropriate preventive interventions. Various early prediction models for T2D have been proposed. However, these methods do not consider the following factors: 1) health examination records (HER) containing health information before diagnosis; 2) rating information containing clinical knowledge; and 3) local and global information of time-series features. These diagnostically relevant factors need to be considered. It is challenging due to irregular and multivariate time series. In this paper, we propose the multi-feature map integrated attention model (MFMAM) for early diabetes prediction using HER. Specifically, HER is converted into the multi-feature map to capture local and global volatility, as well as the sequence order of high-dimensional features. We concatenate rating indicators to introduce clinical knowledge. In addition, considering missing and temporal patterns, we utilize missing and time embedding to learn the complex transition of health status. We adopt attention mechanisms to capture essential features (channels) and time points (spatial). To evaluate the proposed model, we conducted experiments on real-world long-term HER. The results demonstrated that MFMAM outperformed baseline models on tasks of varying sequence lengths and prediction windows. Moreover, we applied our designs to baseline models, and their performance was considerably improved. The proposed model contributes to the short-term and long-term early prediction of T2D in individuals with varying information richness.


Assuntos
Diabetes Mellitus Tipo 2 , Humanos , Diabetes Mellitus Tipo 2/diagnóstico , Fatores de Risco , Doença Crônica
4.
Arch Pharm Res ; 46(11-12): 882-896, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37804415

RESUMO

Breast cancer is one of the major malignancies in women, and most related deaths are due to recurrence, drug resistance, and metastasis. The expression of the mouse double minute 2 (MDM2) oncogene is upregulated in breast cancer; however, its regulatory mechanism has yet to be fully elucidated. Herein, we identified the tumor suppressor death-associated protein kinase 1 (DAPK1) as a novel MDM2 regulator by unbiased peptide library screening. DAPK1 is directly bound to MDM2 and phosphorylates it at Thr419. DAPK1-mediated MDM2 phosphorylation promoted its protein degradation via the ubiquitin-proteasome pathway, resulting in upregulated p53 expression. DAPK1 overexpression, but not its kinase activity-deficient form, decreased colony formation and increased doxorubicin-induced cell death; however, DAPK1 knockdown produced the opposite effects in human breast cancer cells. In a xenograft tumorigenesis assay, DAPK1 overexpression significantly reduced tumor formation, whereas inhibition of DAPK1 kinase activity reduced its antitumorigenic effect. Finally, DAPK1 expression was negatively correlated with MDM2 levels in human breast cancer tissues. Thus, these results suggest that DAPK1-mediated MDM2 phosphorylation and its protein degradation may contribute to its antitumorigenic function in breast cancer.


Assuntos
Neoplasias da Mama , Proteína Supressora de Tumor p53 , Animais , Feminino , Humanos , Camundongos , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Proteínas Quinases Associadas com Morte Celular/metabolismo , Fosforilação , Estabilidade Proteica , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína Supressora de Tumor p53/metabolismo
5.
Clin Exp Med ; 23(6): 1917-1931, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36788153

RESUMO

RNA modification is manifested as chemically altered nucleotides, widely exists in diverse natural RNAs, and is closely related to RNA structure and function. Currently, mRNA-based vaccines have received great attention and rapid development as novel and mighty fighters against various diseases including cancer. The achievement of RNA vaccines in clinical application is largely attributed to some methodological innovations including the incorporation of modified nucleotides into the synthetic RNA. The selection of optimal RNA modifications aimed at reducing the instability and immunogenicity of RNA molecules is a very critical task to improve the efficacy and safety of mRNA vaccines. This review summarizes the functions of RNA modifications and their application in mRNA vaccines, highlights recent advances of mRNA vaccines in cancer immunotherapy, and provides perspectives for future development of mRNA vaccines in the context of personalized tumor therapy.


Assuntos
Vacinas Anticâncer , Neoplasias , Humanos , RNA Mensageiro/genética , RNA/química , Neoplasias/terapia , Neoplasias/tratamento farmacológico , Nucleotídeos
6.
ACS Chem Neurosci ; 13(24): 3554-3566, 2022 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-36454178

RESUMO

Dysregulation of microRNAs has been implicated in diverse diseases, including Alzheimer's disease (AD). MiR-191-5p in plasma/serum has been identified as a novel and promising noninvasive diagnostic biomarker for AD. However, whether miR-191-5p is involved in AD pathogenesis is largely unknown, and its levels in human AD brains are undetermined. Herein, we demonstrated that miR-191-5p downregulated tau phosphorylation at multiple AD-related sites and promoted neurite outgrowth using immunoblotting, immunofluorescence, and neurite outgrowth assays. Moreover, immunoblotting and enzyme-linked immunosorbent assays indicated that miR-191-5p decreased amyloid precursor protein phosphorylation levels and beta-amyloid (Aß) generation. Furthermore, miR-191-5p reduced ceramide-induced neuronal cell death analyzed by trypan blue staining, the in situ cell death detection kit, and Annexin V-FITC/PI flow cytometry. Next, we verified that death-associated protein kinase 1 (DAPK1) was a direct target of miR-191-5p through the dual luciferase reporter assay and confirmed that the effects of miR-191-5p were antagonized by restoration of DAPK1 expression. Finally, the hippocampal miR-191-5p level was found to be decreased in humans with AD compared with controls and was inversely correlated with the DAPK1 expression level. Collectively, these findings suggest that miR-191-5p might exert inhibitory effects on tau phosphorylation, Aß secretion, and neuronal cell death by directly targeting DAPK1, providing an attractive therapeutic option for AD.


Assuntos
Doença de Alzheimer , Proteínas Quinases Associadas com Morte Celular , MicroRNAs , Humanos , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Morte Celular , Proteínas Quinases Associadas com Morte Celular/genética , Proteínas Quinases Associadas com Morte Celular/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Fosforilação
7.
Int J Mol Sci ; 23(14)2022 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-35887339

RESUMO

The neuropathology of Alzheimer's disease (AD) is characterized by intracellular aggregation of hyperphosphorylated tau and extracellular accumulation of beta-amyloid (Aß). Death-associated protein kinase 1 (DAPK1), as a novel therapeutic target, shows promise for the treatment of human AD, but the regulatory mechanisms of DAPK1 expression in AD remain unclear. In this study, we identified miR-143-3p as a promising candidate for targeting DAPK1. miR-143-3p directly bound to the 3' untranslated region of human DAPK1 mRNA and inhibited its translation. miR-143-3p decreased tau phosphorylation and promoted neurite outgrowth and microtubule assembly. Moreover, miR-143-3p attenuated amyloid precursor protein (APP) phosphorylation and reduced the generation of Aß40 and Aß42. Furthermore, restoring DAPK1 expression with miR-143-3p antagonized the effects of miR-143-3p in attenuating tau hyperphosphorylation and Aß production. In addition, the miR-143-3p levels were downregulated and correlated inversely with the expression of DAPK1 in the hippocampus of AD patients. Our results suggest that miR-143-3p might play critical roles in regulating both aberrant tau phosphorylation and amyloidogenic processing of APP by targeting DAPK1 and thus offer a potential novel therapeutic strategy for AD.


Assuntos
Doença de Alzheimer , MicroRNAs , Regiões 3' não Traduzidas , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Proteínas Quinases Associadas com Morte Celular/metabolismo , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Fosforilação , Proteínas tau/genética , Proteínas tau/metabolismo
8.
Int J Mol Sci ; 23(12)2022 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-35742817

RESUMO

Glutamate excitotoxicity induces neuronal cell death during epileptic seizures. Death-associated protein kinase 1 (DAPK1) expression is highly increased in the brains of epilepsy patients; however, the underlying mechanisms by which DAPK1 influences neuronal injury and its therapeutic effect on glutamate excitotoxicity have not been determined. We assessed multiple electroencephalograms and seizure grades and performed biochemical and cell death analyses with cellular and animal models. We applied small molecules and peptides and knocked out and mutated genes to evaluate the therapeutic efficacy of kainic acid (KA), an analog of glutamate-induced neuronal damage. KA administration increased DAPK1 activity by promoting its phosphorylation by activated extracellular signal-regulated kinase (ERK). DAPK1 activation increased seizure severity and neuronal cell death in mice. Selective ERK antagonist treatment, DAPK1 gene ablation, and uncoupling of DAPK1 and ERK peptides led to potent anti-seizure and anti-apoptotic effects in vitro and in vivo. Moreover, a DAPK1 phosphorylation-deficient mutant alleviated glutamate-induced neuronal apoptosis. These results provide novel insight into the pathogenesis of epilepsy and indicate that targeting DAPK1 may be a potential therapeutic strategy for treating epilepsy.


Assuntos
Epilepsia , Ácido Glutâmico , Animais , Proteínas Quinases Associadas com Morte Celular/metabolismo , Epilepsia/genética , MAP Quinases Reguladas por Sinal Extracelular , Ácido Glutâmico/metabolismo , Ácido Glutâmico/toxicidade , Humanos , Ácido Caínico/toxicidade , Camundongos , Convulsões/induzido quimicamente
9.
Transl Neurodegener ; 11(1): 27, 2022 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-35527277

RESUMO

BACKGROUND: Intracellular accumulation of the microtubule-associated protein tau and its hyperphosphorylated forms is a key neuropathological feature of Alzheimer's disease (AD). Melatonin has been shown to prevent tau hyperphosphorylation in cellular and animal models. However, the molecular mechanisms by which melatonin attenuates tau hyperphosphorylation and tau-related pathologies are not fully understood. METHODS: Immunofluorescence, immunoblotting analysis and thioflavin-S staining were employed to examine the effects of early and late treatment of melatonin on tau-related pathology in hTau mice, in which nonmutated human tau is overexpressed on a mouse tau knockout background. High-throughput microRNA (miRNA) sequencing, quantitative RT-PCR, luciferase reporter assay and immunoblotting analysis were performed to determine the molecular mechanism. RESULTS: We found that both early and late treatment of melatonin efficiently decreased the phosphorylation of soluble and insoluble tau at sites related to AD. Moreover, melatonin significantly reduced the number of neurofibrillary tangles (NFTs) and attenuated neuronal loss in the cortex and hippocampus. Furthermore, using miRNA microarray analysis, we found that miR-504-3p expression was upregulated by melatonin in the hTau mice. The administration of miR-504-3p mimics dramatically decreased tau phosphorylation by targeting p39, an activator of the well-known tau kinase cyclin-dependent kinase 5 (CDK5). Compared with miR-504-3p mimics alone, co-treatment with miR-504-3p mimics and p39 failed to reduce tau hyperphosphorylation. CONCLUSIONS: Our results suggest for the first time that melatonin alleviates tau-related pathologies through upregulation of miR-504-3p expression by targeting the p39/CDK5 axis and provide novel insights into AD treatment strategies.


Assuntos
Doença de Alzheimer , Quinase 5 Dependente de Ciclina , Melatonina , MicroRNAs , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/genética , Animais , Quinase 5 Dependente de Ciclina/genética , Melatonina/farmacologia , Camundongos , MicroRNAs/genética , Emaranhados Neurofibrilares/metabolismo
10.
Int J Biol Sci ; 18(2): 693-706, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35002518

RESUMO

The aggregation of amyloid-ß (Aß) peptides into oligomers and fibrils is a key pathological feature of Alzheimer's disease (AD). An increasing amount of evidence suggests that oligomeric Aß might be the major culprit responsible for various neuropathological changes in AD. Death-associated protein kinase 1 (DAPK1) is abnormally elevated in brains of AD patients and plays an important role in modulating tau homeostasis by regulating prolyl isomerase Pin1 phosphorylation. However, it remains elusive whether and how Aß species influence the function of DAPK1, and whether this may further affect the function and phosphorylation of tau in neurons. Herein, we demonstrated that Aß aggregates (both oligomers and fibrils) prepared from synthetic Aß42 peptides were able to upregulate DAPK1 protein levels and thereby its function through heat shock protein 90 (HSP90)-mediated protein stabilization. DAPK1 activation not only caused neuronal apoptosis, but also phosphorylated Pin1 at the Ser71 residue, leading to tau accumulation and phosphorylation at multiple AD-related sites in primary neurons. Both DAPK1 knockout (KO) and the application of a specific DAPK1 inhibitor could effectively protect primary neurons against Aß aggregate-induced cell death and tau dysregulation, corroborating the critical role of DAPK1 in mediating Aß aggregation-induced neuronal damage. Our study suggests a mechanistic link between Aß oligomerization and tau hyperphosphorylation mediated by DAPK1, and supports the role of DAPK1 as a promising target for early intervention in AD.


Assuntos
Doença de Alzheimer/enzimologia , Peptídeos beta-Amiloides/metabolismo , Encéfalo/enzimologia , Proteínas Quinases Associadas com Morte Celular/metabolismo , Neurônios/enzimologia , Fragmentos de Peptídeos/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/genética , Animais , Apoptose/genética , Encéfalo/patologia , Proteínas Quinases Associadas com Morte Celular/deficiência , Proteínas Quinases Associadas com Morte Celular/genética , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peptidilprolil Isomerase de Interação com NIMA/metabolismo , Neurônios/patologia , Fragmentos de Peptídeos/genética , Fosforilação , Proteínas tau/genética , Proteínas tau/metabolismo
11.
Int J Biol Sci ; 17(9): 2356-2366, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34239362

RESUMO

Epilepsy is a chronic encephalopathy and one of the most common neurological disorders. Death-associated protein kinase 1 (DAPK1) expression has been shown to be upregulated in the brains of human epilepsy patients compared with those of normal subjects. However, little is known about the impact of DAPK1 on epileptic seizure conditions. In this study, we aim to clarify whether and how DAPK1 is regulated in epilepsy and whether targeting DAPK1 expression or activity has a protective effect against epilepsy using seizure animal models. Here, we found that cortical and hippocampal DAPK1 activity but not DAPK1 expression was increased immediately after convulsive pentylenetetrazol (PTZ) exposure in mice. However, DAPK1 overexpression was found after chronic low-dose PTZ insults during the kindling paradigm. The suppression of DAPK1 expression by genetic knockout significantly reduced PTZ-induced seizure phenotypes and the development of kindled seizures. Moreover, pharmacological inhibition of DAPK1 activity exerted rapid antiepileptic effects in both acute and chronic epilepsy mouse models. Mechanistically, PTZ stimulated the phosphorylation of NR2B through DAPK1 activation. Combined together, these results suggest that DAPK1 regulation is a novel mechanism for the control of both acute and chronic epilepsy and provide new therapeutic strategies for the treatment of human epilepsy.


Assuntos
Proteínas Quinases Associadas com Morte Celular/antagonistas & inibidores , Proteínas Quinases Associadas com Morte Celular/metabolismo , Epilepsia/tratamento farmacológico , Convulsões/tratamento farmacológico , Animais , Modelos Animais de Doenças , Hipocampo/metabolismo , Excitação Neurológica/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pentilenotetrazol/farmacologia , Receptores de N-Metil-D-Aspartato/metabolismo , Convulsões/induzido quimicamente
12.
J Pineal Res ; 69(2): e12665, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32358852

RESUMO

Death-associated protein kinase 1 (DAPK1) is upregulated in the brains of human Alzheimer's disease (AD) patients compared with normal subjects, and aberrant DAPK1 regulation is implicated in the development of AD. However, little is known about whether and how DAPK1 function is regulated in AD. Here, we identified melatonin as a critical regulator of DAPK1 levels and function. Melatonin significantly decreases DAPK1 expression in a post-transcriptional manner in neuronal cell lines and mouse primary cortical neurons. Moreover, melatonin directly binds to DAPK1 and promotes its ubiquitination, resulting in increased DAPK1 protein degradation through a proteasome-dependent pathway. Furthermore, in tau-overexpressing mouse brain slices, melatonin treatment and the inhibition of DAPK1 kinase activity synergistically decrease tau phosphorylation at multiple sites related to AD. In addition, melatonin and DAPK1 inhibitor dramatically accelerate neurite outgrowth and increase the assembly of microtubules. Mechanistically, melatonin-mediated DAPK1 degradation increases the activity of Pin1, a prolyl isomerase known to play a protective role against tau hyperphosphorylation and tau-related pathologies. Finally, elevated DAPK1 expression shows a strong correlation with the decrease in melatonin levels in human AD brains. Combined, these results suggest that DAPK1 regulation by melatonin is a novel mechanism that controls tau phosphorylation and function and offers new therapeutic options for treating human AD.


Assuntos
Doença de Alzheimer/enzimologia , Encéfalo/enzimologia , Proteínas Quinases Associadas com Morte Celular/biossíntese , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Melatonina/farmacologia , Doença de Alzheimer/patologia , Animais , Encéfalo/patologia , Células HeLa , Humanos , Melatonina/metabolismo , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...