Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Biochim Biophys Acta Mol Basis Dis ; : 167441, 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39069011

ABSTRACT

As we previously revealed, major vault protein (MVP) is a virus-induced host factor, and its expression is crucial for innate immune responses. Nevertheless, the function of MVP in adaptive immunity is poorly known. Here, we demonstrate that Mvp knockout mice had attenuated antibody responses and reduced survival after rechallenge with homologous influenza A virus (IAV) relative to wild-type mice. Analysis of B cell populations showed that MVP promoted germinal center (GC) responses to develop optimal antiviral humoral immunity. Although Mvp-deficient T cells and dendritic cells (DCs) were not intrinsically damaged, MVP promoted activating effector T cells and T follicular helper responses and regulated specific DC subsets. These findings suggest that MVP directs an effective adaptive immune response against IAV by directly engaging in GC reactions or indirectly augmenting cellular immunity via innate immune pathways.

2.
ACS Chem Neurosci ; 13(24): 3554-3566, 2022 12 21.
Article in English | MEDLINE | ID: mdl-36454178

ABSTRACT

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.


Subject(s)
Alzheimer Disease , Death-Associated Protein Kinases , MicroRNAs , Humans , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Cell Death , Death-Associated Protein Kinases/genetics , Death-Associated Protein Kinases/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Phosphorylation
3.
Int J Mol Sci ; 23(14)2022 Jul 20.
Article in English | MEDLINE | ID: mdl-35887339

ABSTRACT

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.


Subject(s)
Alzheimer Disease , MicroRNAs , 3' Untranslated Regions , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Death-Associated Protein Kinases/metabolism , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Phosphorylation , tau Proteins/genetics , tau Proteins/metabolism
4.
Transl Neurodegener ; 11(1): 27, 2022 05 09.
Article in English | MEDLINE | ID: mdl-35527277

ABSTRACT

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.


Subject(s)
Alzheimer Disease , Cyclin-Dependent Kinase 5 , Melatonin , MicroRNAs , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Animals , Cyclin-Dependent Kinase 5/genetics , Melatonin/pharmacology , Mice , MicroRNAs/genetics , Neurofibrillary Tangles/metabolism
5.
Int J Biol Sci ; 18(2): 693-706, 2022.
Article in English | MEDLINE | ID: mdl-35002518

ABSTRACT

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.


Subject(s)
Alzheimer Disease/enzymology , Amyloid beta-Peptides/metabolism , Brain/enzymology , Death-Associated Protein Kinases/metabolism , Neurons/enzymology , Peptide Fragments/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Animals , Apoptosis/genetics , Brain/pathology , Death-Associated Protein Kinases/deficiency , Death-Associated Protein Kinases/genetics , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Knockout , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Neurons/pathology , Peptide Fragments/genetics , Phosphorylation , tau Proteins/genetics , tau Proteins/metabolism
6.
Int J Biol Sci ; 17(9): 2356-2366, 2021.
Article in English | MEDLINE | ID: mdl-34239362

ABSTRACT

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.


Subject(s)
Death-Associated Protein Kinases/antagonists & inhibitors , Death-Associated Protein Kinases/metabolism , Epilepsy/drug therapy , Seizures/drug therapy , Animals , Disease Models, Animal , Hippocampus/metabolism , Kindling, Neurologic/drug effects , Male , Mice , Mice, Inbred C57BL , Pentylenetetrazole/pharmacology , Receptors, N-Methyl-D-Aspartate/metabolism , Seizures/chemically induced
7.
J Immunol ; 207(2): 577-589, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34145061

ABSTRACT

The IL family of cytokines participates in immune response and regulation. We previously found that soluble IL-6 receptor plays an important role in the host antiviral response. In this study, we detected the IL-6-IL-27 complex in serum and throat swab samples from patients infected with influenza A virus. A plasmid expressing the IL-6-IL-27 complex was constructed to explore its biological function. The results indicated that the IL-6-IL-27 complex has a stronger antiviral effect than the individual subunits of IL-6, IL-27A, and EBV-induced gene 3. Furthermore, the activity of the IL-6-IL-27 complex is mainly mediated by the IL-27A subunit and the IL-27 receptor α. The IL-6-IL-27 complex can positively regulate virus-triggered expression of IFN and IFN-stimulated genes by interacting with adaptor protein mitochondrial antiviral signaling protein, potentiating the ubiquitination of TNF receptor-associated factors 3 and 6 and NF-κB nuclear translocation. The secreted IL-6-IL-27 complex can induce the phosphorylation of STAT1 and STAT3 and shows antiviral activity. Our results demonstrate a previously unrecognized mechanism by which IL-6, IL-27A, and EBV-induced gene 3 form a large complex both intracellularly and extracellularly, and this complex acts in the host antiviral response.


Subject(s)
Antiviral Agents/immunology , Immunity/immunology , Interleukin-6/immunology , Interleukins/immunology , A549 Cells , Cell Line , Cell Line, Tumor , Cytokines/immunology , HEK293 Cells , Humans , Influenza A virus/immunology , Interferons/immunology , NF-kappa B/immunology , Phosphorylation/immunology , STAT1 Transcription Factor/immunology , STAT3 Transcription Factor/immunology , Signal Transduction/immunology
8.
J Pineal Res ; 69(2): e12665, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32358852

ABSTRACT

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.


Subject(s)
Alzheimer Disease/enzymology , Brain/enzymology , Death-Associated Protein Kinases/biosynthesis , Gene Expression Regulation, Enzymologic/drug effects , Melatonin/pharmacology , Alzheimer Disease/pathology , Animals , Brain/pathology , HeLa Cells , Humans , Melatonin/metabolism , Mice
9.
R Soc Open Sci ; 5(8): 172027, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30224986

ABSTRACT

In experimental investigations on axial symmetry, over-ventilated CH4/air diffusion combustion in a packed bed is executed to study the height, shape and stability of the flame. The combustor is a quartz tube packed with alumina pellets in which a cylindrical fuel stream is surrounded by a coflow air nozzle. The results show that the bed length and pellet diameter have a significant influence on the flame properties. In general, the flame above the pellet surface has axial symmetry, and its shape and colour are similar to those of a conventional diffusion flame when the bed length is smaller. The colour of the flame front varies with the bed length. The changed colour indicates an increased flame front temperature and that the combustion regime above the bed surface may change from non-premixed combustion to partially premixed combustion or even premixed combustion owing to the mix and dispersion effect in the packed bed. In addition, multiple flame behaviours, such as an inclined flame front, isolated reaction zone and oscillatory motion followed by a pulsating sound with a few hertz in a packed bed, are observed experimentally. The possible reasons for these phenomena are discussed.

SELECTION OF CITATIONS
SEARCH DETAIL