Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Cell ; 186(16): 3350-3367.e19, 2023 08 03.
Article in English | MEDLINE | ID: mdl-37421950

ABSTRACT

Synucleinopathies are characterized by the accumulation of α-synuclein (α-Syn) aggregates in the brain. Positron emission tomography (PET) imaging of synucleinopathies requires radiopharmaceuticals that selectively bind α-Syn deposits. We report the identification of a brain permeable and rapid washout PET tracer [18F]-F0502B, which shows high binding affinity for α-Syn, but not for Aß or Tau fibrils, and preferential binding to α-Syn aggregates in the brain sections. Employing several cycles of counter screenings with in vitro fibrils, intraneuronal aggregates, and neurodegenerative disease brain sections from several mice models and human subjects, [18F]-F0502B images α-Syn deposits in the brains of mouse and non-human primate PD models. We further determined the atomic structure of the α-Syn fibril-F0502B complex by cryo-EM and revealed parallel diagonal stacking of F0502B on the fibril surface through an intense noncovalent bonding network via inter-ligand interactions. Therefore, [18F]-F0502B is a promising lead compound for imaging aggregated α-Syn in synucleinopathies.


Subject(s)
Neurodegenerative Diseases , Synucleinopathies , Animals , Humans , alpha-Synuclein/metabolism , Synucleinopathies/diagnostic imaging , Synucleinopathies/metabolism , Neurodegenerative Diseases/metabolism , Positron-Emission Tomography , Brain/diagnostic imaging , Brain/metabolism
2.
Chem Biodivers ; : e202401063, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38924351

ABSTRACT

In the process of searching for anti-breast cancer agents, five sesquiterpene lactones (1-5), including two previously undescribed ones, yjaponica B-C (1-2), were isolated from the herb of Youngia japonica. Their structures were elucidated by spectroscopic data analyses and Marfey's method. Cytotoxic activities of all compounds against A549, U87, and 4T1 cell lines were tested using the CCK8 assay. The result showed that compound 3 possessed the highest cytotoxic activity against 4T1 cells with an IC50 value of 10.60 µM. Furthermore, compound 3 distinctly induced apoptosis, inhibited immigration, and blocked the cell cycle of 4T1 cells. In addition, compound 3 induced the production of reactive oxygen species. Further anticancer mechanism studies showed that compound 3 significantly upregulated expression of the cleaved caspase 3 and PARP, whereas it downregulated the expression of Bcl-2, cyclin D1, cyclin A2, CDK4, and CDK2. Taken together, our results demonstrate that compound 3 has a high potential of being used as a leading compound for the discovery of new anti-breast cancer agent.

3.
J Asian Nat Prod Res ; : 1-8, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38963349

ABSTRACT

The preliminary study revealed that the ethyl acetate eluate of Youngia japonica (YJ-E) could inhibit the expression of key proteins of p-p65, p-IκBα, p-IKKα/ß, and p-AKT in LPS stimulated BV2 cell. Further phytochemical study led to the isolation of eight compounds from YJ-E, including one new sesquiterpene lactone. Their structures were elucidated by several spectroscopic data, and comparing the NMR data of known compound. In addition, all of the isolates were evaluated for the anti-inflammatory effect. As a result, compounds 3 and 4 distinctly attenuated the expressions of p-IκBα, p-p65, and p-AKT in LPS stimulated BV2 cell, respectively.

4.
Int J Environ Health Res ; 34(3): 1602-1614, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37394938

ABSTRACT

Ambient fine particulate matter (PM2.5) is a threat to public health. The P2 X 7purinergic receptor (P2X7R) is a modulator that responds to inflammation. Yet the role of P2X7R in the mediation of PM2.5-induced pulmonary cytotoxicity is rarely investigated. In this study, the expression of P2X7R and its effect on cell viability, oxidative damage, apoptosis, mitochondrial dysfunction and underlying mechanism following PM2.5 treatment in rat alveolar macrophages (NR8383) were analyzed. The outcome indicated that PM2.5 exposure significantly increased the expression of P2X7R, while P2X7R antagonist oATP markedly alleviate the production of reactive oxygen species (ROS), Nitrite Oxidation (NO), mitochondrial membrane potential, apoptosis rate, and release of inflammatory cytokines. In contrast, P2X7 agonist BzATP showed opposite effect in PM2.5-treated NR8383 cells. Therefore, these results demonstrated that P2X7R participated in PM2.5-induced pulmonary toxicity, while the blockade of P2X7R is a promising therapeutic approach of treating PM2.5-induced lung diseases.


Subject(s)
Particulate Matter , Receptors, Purinergic P2X7 , Rats , Animals , Receptors, Purinergic P2X7/metabolism , Particulate Matter/toxicity , Lung , Reactive Oxygen Species/metabolism , Oxidative Stress
5.
Mol Psychiatry ; 27(8): 3396-3409, 2022 08.
Article in English | MEDLINE | ID: mdl-35546632

ABSTRACT

Diabetes is a risk factor for Alzheimer's disease (AD), which is also called type 3 diabetes with insulin reduction and insulin resistance in AD patient brains. However, the molecular mechanism coupling diabetes to AD onset remains incompletely understood. Here we show that inflammation, associated with obesity and diabetes elicited by high-fat diet (HFD), activates neuronal C/EBPß/AEP signaling that drives AD pathologies and cognitive disorders. HFD stimulates diabetes and insulin resistance in neuronal Thy1-C/EBPß transgenic (Tg) mice, accompanied with prominent mouse Aß accumulation and hyperphosphorylated Tau aggregation in the brain, triggering cognitive deficits. These effects are profoundly diminished when AEP is deleted from C/EBPß Tg mice. Chronic treatment with inflammatory lipopolysaccharide (LPS) facilitates AD pathologies and cognitive disorders in C/EBPß Tg but not in wild-type mice, and these deleterious effects were substantially alleviated in C/EBPß Tg/AEP -/- mice. Remarkably, the anti-inflammatory drug aspirin strongly attenuates HFD-induced diabetes and AD pathologies in neuronal C/EBPß Tg mice. Therefore, our findings demonstrate that inflammation-activated neuronal C/EBPß/AEP signaling couples diabetes to AD.


Subject(s)
Alzheimer Disease , Diabetes Mellitus , Insulin Resistance , Animals , Mice , Alzheimer Disease/metabolism , Diet, High-Fat/adverse effects , Insulin Resistance/physiology , Mice, Transgenic , Inflammation/metabolism , Brain/metabolism , CCAAT-Enhancer-Binding Protein-beta/metabolism , Diabetes Mellitus/metabolism , Disease Models, Animal
6.
Gut ; 71(11): 2233-2252, 2022 11.
Article in English | MEDLINE | ID: mdl-35017199

ABSTRACT

OBJECTIVE: This study is to investigate the role of gut dysbiosis in triggering inflammation in the brain and its contribution to Alzheimer's disease (AD) pathogenesis. DESIGN: We analysed the gut microbiota composition of 3×Tg mice in an age-dependent manner. We generated germ-free 3×Tg mice and recolonisation of germ-free 3×Tg mice with fecal samples from both patients with AD and age-matched healthy donors. RESULTS: Microbial 16S rRNA sequencing revealed Bacteroides enrichment. We found a prominent reduction of cerebral amyloid-ß plaques and neurofibrillary tangles pathology in germ-free 3×Tg mice as compared with specific-pathogen-free mice. And hippocampal RNAseq showed that inflammatory pathway and insulin/IGF-1 signalling in 3×Tg mice brain are aberrantly altered in the absence of gut microbiota. Poly-unsaturated fatty acid metabolites identified by metabolomic analysis, and their oxidative enzymes were selectively elevated, corresponding with microglia activation and inflammation. AD patients' gut microbiome exacerbated AD pathologies in 3×Tg mice, associated with C/EBPß/asparagine endopeptidase pathway activation and cognitive dysfunctions compared with healthy donors' microbiota transplants. CONCLUSIONS: These findings support that a complex gut microbiome is required for behavioural defects, microglia activation and AD pathologies, the gut microbiome contributes to pathologies in an AD mouse model and that dysbiosis of the human microbiome might be a risk factor for AD.


Subject(s)
Alzheimer Disease , Gastrointestinal Microbiome , Insulins , Alzheimer Disease/metabolism , Animals , Cognition , Disease Models, Animal , Dysbiosis , Fatty Acids, Unsaturated , Gastrointestinal Microbiome/physiology , Humans , Inflammation/metabolism , Insulin-Like Growth Factor I , Mice , Neuroinflammatory Diseases , Plaque, Amyloid/pathology , RNA, Ribosomal, 16S
7.
Biochem Biophys Res Commun ; 636(Pt 2): 62-70, 2022 12 25.
Article in English | MEDLINE | ID: mdl-36356543

ABSTRACT

Epicardial adipose tissue (EAT) is a metabolically active organ which generates inflammatory cytokines. Thickness of EAT is associated with onset and development of heart failure with preserved ejection fraction (HFpEF). However, it is still unclear the specific mechanisms and pharmacological targets on EAT induced inflammation in HFpEF. A two-hit protocol with western diet and Nω-nitrol-arginine methyl ester (L-NAME) was used to establish HFpEF mouse model. In HFpEF mice, inflammatory biomarkers, such as tumor necrosis factor (TNF)-α, interleukin (IL)-1ß and von willebrand factor (vWF) elevated in myocardium compared to control. Inflammatory cell infiltration in myocardium was increased. In HFpEF mice, inflammasome-mediated pyroptosis pathway was activated in the EAT. Suppression of pyroptosis-related protein gasdermin D (GSDMD) in cultured EAT could lower cardiomyocyte inflammation and autophagy. Furthermore, spironolactone and rosuvastatin, the two-hit anti-inflammatory agents, reduced NLR family pyrin domain containing 3 (NLRP3)/GSDMD pyroptosis in EAT and autophagy in myocardium of HFpEF mouse. The combination treatment also enhanced exercise tolerance and appeased inflammatory injuries in HFpEF mice. CONCLUSION: Pyroptosis signaling is involved in EAT-myocardium axis in mouse model of HFpEF. Targeting adipocyte-derived inflammation in EAT bears potential to treatment HFpEF.


Subject(s)
Heart Failure , Pyroptosis , Mice , Animals , Heart Failure/metabolism , Stroke Volume , Inflammasomes/metabolism , Myocardium/metabolism , Adipose Tissue/metabolism , Inflammation/pathology , Disease Models, Animal , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
8.
Mol Psychiatry ; 26(7): 2943-2963, 2021 07.
Article in English | MEDLINE | ID: mdl-32782380

ABSTRACT

Neurotrophins promote neuronal survival and synaptic plasticity via activating the tropomyosin receptor kinases. BDNF and its high-affinity receptor TrkB are reduced in Alzheimer's disease (AD), contributing to progressive cognitive decline. However, how the signaling mediates AD pathologies remains incompletely understood. Here we show that the TrkB receptor binds and phosphorylates APP, reducing amyloid-ß production, which are abrogated by δ-secretase cleavage of TrkB in AD. Remarkably, BDNF stimulates TrkB to phosphorylate APP Y687 residue that accumulates APP in the TGN (Trans-Golgi Network) and diminishes its amyloidogenic cleavage. Delta-secretase cleaves TrkB at N365 and N486/489 residues and abolishes its neurotrophic activity, decreasing p-APP Y687 and altering its subcellular trafficking. Notably, both TrkB and APP are robustly cleaved by δ-secretase in AD brains, accompanied by mitigated TrkB signaling and reduced p-Y687. Blockade of TrkB cleavage attenuates AD pathologies in 5xFAD mice, rescuing the learning and memory. Viral expression of TrkB 1-486 fragment in the hippocampus of APP/PS1 mice facilitates amyloid pathology and mitigates cognitive functions. Hence, δ-secretase cleaves TrkB and blunts its phosphorylation of APP, facilitating AD pathogenesis.


Subject(s)
Alzheimer Disease , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Membrane Glycoproteins , Mice , Phosphorylation , Protein-Tyrosine Kinases , Receptor, trkB/metabolism
9.
Mol Psychiatry ; 26(2): 568-585, 2021 02.
Article in English | MEDLINE | ID: mdl-32086435

ABSTRACT

Parkinson's disease (PD) is characterized by dopaminergic neuronal loss and the presence of intra-neuronal Lewy body (LB) inclusions with aggregated α-synuclein (α-Syn) as the major component. MAOB, a crucial monoamine oxidase for dopamine metabolism, triggers oxidative stress in dopaminergic neurons and α-Syn aggregation. However, the key molecular mechanism that mediates PD pathogenesis remains elusive. Here we show that C/EBPß acts as an age-dependent transcription factor for both α-Syn and MAOB, and initiates the PD pathologies by upregulating these two pivotal players, in addition to escalating δ-secretase activity to cleave α-Syn and promotes its neurotoxicity. Overexpression of C/EBPß in human wild-type α-Syn transgenic mice facilitates PD pathologies and elicits motor disorders associated with augmentation of δ-secretase, α-Syn, and MAOB. In contrast, depletion of C/EBPß from human α-Syn Tg mice abolishes rotenone-elicited PD pathologies and motor impairments via downregulating the expression of these key factors. Hence, our study supports that C/EBPß/δ-secretase signaling mediates PD pathogenesis via regulating the expression and cleavage of α-Syn and MAOB.


Subject(s)
Parkinson Disease , alpha-Synuclein , Amyloid Precursor Protein Secretases , Animals , CCAAT-Enhancer-Binding Protein-beta/genetics , Dopaminergic Neurons , Mice , Parkinson Disease/genetics , alpha-Synuclein/genetics
10.
Mol Psychiatry ; 26(10): 6002-6022, 2021 10.
Article in English | MEDLINE | ID: mdl-33339957

ABSTRACT

The apolipoprotein E ε4 (APOE4) allele is a major genetic risk factor for Alzheimer's disease (AD), and its protein product, ApoE4, exerts its deleterious effects mainly by influencing amyloid-ß (Aß) and Tau (neurofibrillary tangles, NFTs) deposition in the brain. However, the molecular mechanism dictating its expression during ageing and in AD remains incompletely clear. Here we show that C/EBPß acts as a pivotal transcription factor for APOE and mediates its mRNA levels in an age-dependent manner. C/EBPß binds the promoter of APOE and escalates its expression in the brain. Knockout of C/EBPß in AD mouse models diminishes ApoE expression and Aß pathologies, whereas overexpression of C/EBPß accelerates AD pathologies, which can be attenuated by anti-ApoE monoclonal antibody or deletion of ApoE via its specific shRNA. Remarkably, C/EBPß selectively promotes more ApoE4 expression versus ApoE3 in human neurons, correlating with higher activation of C/EBPß in human AD brains with ApoE4/4 compared to ApoE3/3. Therefore, our data support that C/EBPß is a crucial transcription factor for temporally regulating APOE gene expression, modulating ApoE4's role in AD pathogenesis.


Subject(s)
Alzheimer Disease , Apolipoprotein E4 , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Animals , Apolipoprotein E4/genetics , Apolipoproteins E , Brain/metabolism , CCAAT-Enhancer-Binding Protein-beta/genetics , Mice , Mice, Knockout
11.
Brain ; 144(6): 1833-1852, 2021 07 28.
Article in English | MEDLINE | ID: mdl-33880508

ABSTRACT

Amyloid-ß precursor protein (APP) is sequentially cleaved by secretases and generates amyloid-ß, the major components in senile plaques in Alzheimer's disease. APP is upregulated in human Alzheimer's disease brains. However, the molecular mechanism of how APP contributes to Alzheimer's disease pathogenesis remains incompletely understood. Here we show that truncated APP C586-695 fragment generated by δ-secretase directly binds to CCAAT/enhancer-binding protein beta (CEBPB), an inflammatory transcription factor, and enhances its transcriptional activity, escalating Alzheimer's disease-related gene expression and pathogenesis. The APP C586-695 fragment, but not full-length APP, strongly associates with CEBPB and elicits its nuclear translocation and augments the transcriptional activities on APP itself, MAPT (microtubule-associated protein tau), δ-secretase and inflammatory cytokine mRNA expression, finally triggering Alzheimer's disease pathology and cognitive disorder in a viral overexpression mouse model. Blockade of δ-secretase cleavage of APP by mutating the cleavage sites reduces its stimulatory effect on CEBPB, alleviating amyloid pathology and cognitive dysfunctions. Clearance of APP C586-695 from 5xFAD mice by antibody administration mitigates Alzheimer's disease pathologies and restores cognitive functions. Thus, in addition to the sequestration of amyloid-ß, APP implicates in Alzheimer's disease pathology by activating CEBPB upon δ-secretase cleavage.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/metabolism , CCAAT-Enhancer-Binding Protein-beta/metabolism , Gene Expression Regulation/physiology , Aged , Animals , Cysteine Endopeptidases/metabolism , Female , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged
12.
Proc Natl Acad Sci U S A ; 115(15): 3954-3959, 2018 04 10.
Article in English | MEDLINE | ID: mdl-29581300

ABSTRACT

Amyloid beta (Aß) is a major pathological marker in Alzheimer's disease (AD), which is principally regulated by the rate-limiting ß-secretase (i.e., BACE1) cleavage of amyloid precursor protein (APP). However, how BACE1 activity is posttranslationally regulated remains incompletely understood. Here, we show that BACE1 is predominantly SUMOylated at K501 residue, which escalates its protease activity and stability and subsequently increases Aß production, leading to cognitive defect seen in the AD mouse model. Compared with a non-SUMOylated K501R mutant, injection of wild-type BACE1 significantly increases Aß production and triggers cognitive dysfunction. Furthermore, overexpression of wild-type BACE1, but not non-SUMOylated K501R mutant, facilitates senile plaque formation and aggravates the cognitive deficit seen in the APP/PS1 AD mouse model. Together, our data strongly suggest that K501 SUMOylation on BACE1 plays a critical role in mediating its stability and enzymatic activity.


Subject(s)
Alzheimer Disease/enzymology , Amyloid Precursor Protein Secretases/chemistry , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/chemistry , Aspartic Acid Endopeptidases/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/psychology , Amino Acid Motifs , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Animals , Aspartic Acid Endopeptidases/genetics , Cognition , Disease Models, Animal , Enzyme Stability , Humans , Mice , Mice, Transgenic , Sumoylation
13.
J Cell Biochem ; 120(6): 8935-8948, 2019 06.
Article in English | MEDLINE | ID: mdl-28419530

ABSTRACT

This study aimed to elucidate the precise mechanisms underlying the protective effects of phosphoseryl-tRNA kinase (PSTK) against cisplatin-induced podocyte injury. PSTK overexpression and knockdown vectors were generated and transfected into murine podocyte cells-5. PSTK levels were measured, and transcriptome sequencing was conducted. Differential expression analysis was performed to identify messenger RNAs (mRNAs) that were positively and negatively correlated with PSTK. We selected 10 candidate genes identified via real-time quantitative polymerase chain reaction and Western blot analysis for further analysis. As expected, PSTK levels were significantly higher in PSTK-overexpressing podocytes and significantly lower in PSTK-knockdown podocytes. PSTK overexpression resulted in the upregulation of 122 genes and downregulation of 372 genes in podocytes. On the other hand, PSTK knockdown resulted in the upregulation of 231 genes and downregulation of 445 genes. Furthermore, the analysis revealed that 11 genes were positively correlated with PSTK, whereas 20 genes were negatively correlated with PSTK. The obtained PSTK-regulated genes were primarily involved in molecular function, biological process, and cellular component, as well as the angiogenesis pathway. The Wnt family member 10A levels were significantly higher after PSTK overexpression, but were significantly lower after PSTK knockdown. In addition, Na+/K+ ATPase subunit α-2 and matrix metalloproteinase 9 levels were significantly downregulated after PSTK overexpression, but significantly upregulated upon PSTK knockdown. Cell proliferation was significantly increased upon PSTK overexpression, but significantly decreased upon PSTK suppression. The results of this study not only identified several significant PSTK-regulated genes for further validation, but also provided insights into the mechanisms underlying the protective effects of PSTK on podocytes.


Subject(s)
Cisplatin/adverse effects , Gene Expression Profiling/methods , Phosphorylase Kinase/genetics , Podocytes/cytology , Animals , Cell Line , Cell Proliferation/drug effects , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Humans , Matrix Metalloproteinase 9/genetics , Mice , Nerve Tissue Proteins/genetics , Phosphorylase Kinase/metabolism , Podocytes/drug effects , Podocytes/metabolism , Sodium-Potassium-Exchanging ATPase/genetics , Wnt Proteins/genetics
14.
Proc Natl Acad Sci U S A ; 111(46): 16586-91, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25378699

ABSTRACT

Intracellular accumulation of the abnormally modified tau is hallmark pathology of Alzheimer's disease (AD), but the mechanism leading to tau aggregation is not fully characterized. Here, we studied the effects of tau SUMOylation on its phosphorylation, ubiquitination, and degradation. We show that tau SUMOylation induces tau hyperphosphorylation at multiple AD-associated sites, whereas site-specific mutagenesis of tau at K340R (the SUMOylation site) or simultaneous inhibition of tau SUMOylation by ginkgolic acid abolishes the effect of small ubiquitin-like modifier protein 1 (SUMO-1). Conversely, tau hyperphosphorylation promotes its SUMOylation; the latter in turn inhibits tau degradation with reduction of solubility and ubiquitination of tau proteins. Furthermore, the enhanced SUMO-immunoreactivity, costained with the hyperphosphorylated tau, is detected in cerebral cortex of the AD brains, and ß-amyloid exposure of rat primary hippocampal neurons induces a dose-dependent SUMOylation of the hyperphosphorylated tau. Our findings suggest that tau SUMOylation reciprocally stimulates its phosphorylation and inhibits the ubiquitination-mediated tau degradation, which provides a new insight into the AD-like tau accumulation.


Subject(s)
Alzheimer Disease/metabolism , Cerebral Cortex/metabolism , Hippocampus/metabolism , Nerve Tissue Proteins/metabolism , Point Mutation , Protein Processing, Post-Translational , SUMO-1 Protein/metabolism , tau Proteins/metabolism , Alzheimer Disease/pathology , Amino Acid Substitution , Amyloid beta-Peptides/pharmacology , Androstadienes/pharmacology , Animals , Cerebral Cortex/pathology , Female , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , HEK293 Cells , Humans , Indoles/pharmacology , Male , Maleimides/pharmacology , Middle Aged , Mutagenesis, Site-Directed , Mutation, Missense , Nerve Tissue Proteins/genetics , Peptide Fragments/pharmacology , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/metabolism , SUMO-1 Protein/genetics , Salicylates/pharmacology , Solubility , Sumoylation , Ubiquitination , Wortmannin , tau Proteins/genetics
15.
STAR Protoc ; 5(1): 102788, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38117656

ABSTRACT

Alpha-synuclein (α-Syn) positron emission tomography (PET) imaging is a valuable approach for diagnosing and monitoring synucleinopathies-related diseases, such as Parkinson disease. Here, we present a protocol for screening potential α-Syn PET tracers using in vitro and ex vivo approaches. We describe steps for employing recombinant pre-formed fibrils and conducting screening procedures on neuronal models, mouse models, and patients' brain tissue sections to assess the specificity and selectivity of the candidate compounds. For complete details on the use and execution of this protocol, please refer to Xiang et al. (2023).1.


Subject(s)
Parkinson Disease , alpha-Synuclein , Mice , Animals , Humans , Parkinson Disease/diagnostic imaging , Positron-Emission Tomography
16.
Sci Rep ; 14(1): 5386, 2024 03 05.
Article in English | MEDLINE | ID: mdl-38443672

ABSTRACT

Systemic inflammation and reciprocal organ interactions are associated with the pathophysiology of heart failure with preserved ejection fraction (HFpEF). However, the clinical value, especially the diagnositc prediction power of inflammation and extra-cardiac organ dysfunction for HfpEF is not explored. In this cross-sectional study, 1808 hospitalized patients from January 2014 to June 2022 in ChiHFpEF cohort were totally enrolled according to inclusion and exclusion criteria. A diagnostic model with markers from routine blood test as well as liver and renal dysfunction for HFpEF was developed using data from ChiHFpEF-cohort by logistic regression and assessed by receiver operating characteristic curve (ROC) and Brier score. Then, the model was validated by the tenfold cross-validation and presented as nomogram and a web-based online risk calculator as well. Multivariate and LASSO regression analysis revealed that age, hemoglobin, neutrophil to lymphocyte ratio, AST/ALT ratio, creatinine, uric acid, atrial fibrillation, and pulmonary hypertension were associated with HFpEF. The predictive model exhibited reasonably accurate discrimination (ROC, 0.753, 95% CI 0.732-0.772) and calibration (Brier score was 0.200). Subsequent internal validation showed good discrimination and calibration (AUC = 0.750, Brier score was 0.202). In additoin to participating in pathophysiology of HFpEF, inflammation and multi-organ interactions have diagnostic prediction value for HFpEF. Screening and optimizing biomarkers of inflammation and multi-organ interactions stand for a new field to improve noninvasive diagnostic tool for HFpEF.


Subject(s)
Heart Failure , Humans , Heart Failure/diagnosis , Cross-Sectional Studies , Stroke Volume , Inflammation , Liver
17.
Front Pharmacol ; 15: 1372110, 2024.
Article in English | MEDLINE | ID: mdl-38694913

ABSTRACT

Prevalent neurological disorders such as Alzheimer's disease, Parkinson's disease, and stroke are increasingly becoming a global burden as society ages. It is well-known that degeneration and loss of neurons are the fundamental underlying processes, but there are still no effective therapies for these neurological diseases. In recent years, plenty of studies have focused on the pharmacology and feasibility of natural products as new strategies for the development of drugs that target neurological disorders. Antrodia camphorata has become one of the most promising candidates, and the crude extracts and some active metabolites of it have been reported to play various pharmacological activities to alleviate neurological symptoms at cellular and molecular levels. This review highlights the current evidence of Antrodia camphorata against neurological disorders, including safety evaluation, metabolism, blood-brain barrier penetration, neuroprotective activities, and the potential on regulating the gut-microbiome-brain axis. Furthermore, potential strategies to resolve problematic issues identified in previous studies are also discussed. We aim to provide an overview for the ongoing development and utilization of Antrodia camphorata in cerebral neuropathology.

18.
Fitoterapia ; 174: 105869, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38378132

ABSTRACT

Fourteen sesquiterpenes, including one undescribed sesquiterpene lactone, were isolated from Youngia japonica, and their structures were identified by NMR, HRESIMS, ECD and calculated ECD. Cytotoxic activities of all isolates against A549, HeLa, and 4 T1 cell lines were detected by CCK8 assay. Among them, 2 showed obvious cytotoxic activity against A549 cells. Subsequently, the production of ROS, and apoptosis of A549 cells treated with 2 were evaluated. The result showed that 2 distinctly increased the ROS level, and induced the apoptosis of A549 cells. Further anticancer mechanism studies showed that 2 increased the expression of cleaved caspase 3. Taken together, our results demonstrated that 2 might become potential leading compounds for the treatment of lung cancer.


Subject(s)
Antineoplastic Agents , Asteraceae , Sesquiterpenes , Humans , Cell Line, Tumor , Molecular Structure , Reactive Oxygen Species , Antineoplastic Agents/pharmacology , Apoptosis , Sesquiterpenes/pharmacology , Sesquiterpenes/chemistry
19.
Life Sci ; 346: 122618, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38614306

ABSTRACT

AIMS: This study was designed to investigate the role of growth arrest and DNA damage-inducible ß (GADD45B) in modulating fear memory acquisition and elucidate its underlying mechanisms. MAIN METHODS: Adeno-associated virus (AAV) that knockdown or overexpression GADD45B were injected into ventral hippocampal CA1 (vCA1) by stereotactic, and verified by fluorescence and Western blot. The contextual fear conditioning paradigm was employed to examine the involvement of GADD45B in modulating aversive memory acquisition. The Y-maze and novel location recognition (NLR) tests were used to examine non-aversive cognition. The synaptic plasticity and electrophysiological properties of neurons were measured by slice patch clamp. KEY FINDINGS: Knockdown of GADD45B in the vCA1 significantly enhanced fear memory acquisition, accompanied by an upregulation of long-term potentiation (LTP) expression and intrinsic excitability of vCA1 pyramidal neurons (PNs). Conversely, overexpression of GADD45B produced the opposite effects. Notably, silencing the activity of vCA1 neurons abolished the impact of GADD45B knockdown on fear memory development. Moreover, mice with vCA1 GADD45B overexpression exhibited impaired spatial cognition, whereas mice with GADD45B knockdown did not display such impairment. SIGNIFICANCE: These results provided compelling evidence for the crucial involvement of GADD45B in the formation of aversive memory and spatial cognition.


Subject(s)
CA1 Region, Hippocampal , Fear , GADD45 Proteins , Mice, Inbred C57BL , Animals , Male , Fear/physiology , Mice , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/physiology , Cognition/physiology , Memory/physiology , Long-Term Potentiation/physiology , Maze Learning/physiology , Neuronal Plasticity/physiology , Antigens, Differentiation/metabolism , Antigens, Differentiation/genetics , Gene Knockdown Techniques
20.
J Neurochem ; 126(2): 234-42, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23646926

ABSTRACT

Glycogen synthase kinase-3 beta (GSK-3ß) dysfunction may play an essential role in the pathogenesis of psychiatric, metabolic, neurodegenerative diseases, in which oxidative stress exists concurrently. Some studies have shown that GSK-3ß activity is up-regulated under oxidative stress. This study evaluated how oxidative stress regulates GSK-3ß activity in human embryonic kidney 293 (HEK293)/Tau cells treated with hydrogen peroxide (H2O2). Here, we show that H2O2 induced an obvious increase of GSK-3ß activity. Surprisingly, H2O2 dramatically increased phosphorylation of GSK-3ß at Ser9, an inactive form of GSK-3ß,while there were no changes of phosphorylation of GSK-3ß at Tyr216. Moreover, H2O2 led to a transient [Ca²âº](i) elevation, and simultaneously increased the truncation of GSK-3ß into two fragments of 40 kDa and 30 kDa, whereas inhibition of calpain decreased the truncation and recovered the activity of GSK-3ß. Furthermore, tau was hyperphosphorylated at Ser396, Ser404, and Thr231, three most common GSK-3ß targeted sites after 100 µM H2O2 administration in HEK293/Tau cells, whereas inhibition of calpain blocked the tau phosphorylation. In addition, we found that there were no obvious changes of Cyclin-dependent kinase 5 (CDK5) expression (responsible for tau phosphorylation) and of p35 cleavage, the regulatory subunit of CDK5 in H2O2-treated HEK293/Tau cells. In conclusion, Ca²âº-dependent calpain activation leads to GSK-3ß truncation, which counteracts the inhibitory effect of Ser9 phosphorylation, up-regulates GSK-3ß activity, and phosphorylates tau in H2O2-treated HEK293/Tau cells.


Subject(s)
Calpain/pharmacology , Glycogen Synthase Kinase 3/drug effects , Glycogen Synthase Kinase 3/metabolism , Hydrogen Peroxide/pharmacology , Serine/metabolism , Up-Regulation/drug effects , Calcium/metabolism , Cell Survival/drug effects , Cell Survival/genetics , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Glycoproteins/pharmacology , HEK293 Cells , Humans , Intracellular Fluid/drug effects , Intracellular Fluid/metabolism , Malondialdehyde/metabolism , Mutation/genetics , Oncogene Protein v-akt/metabolism , Oxidative Stress/drug effects , Phosphorylation/drug effects , Signal Transduction/drug effects , Superoxide Dismutase/metabolism , Transfection , tau Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL