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










Database
Publication year range
1.
Aging (Albany NY) ; 13(20): 23620-23636, 2021 10 13.
Article in English | MEDLINE | ID: mdl-34644262

ABSTRACT

Amyloid-ß (Aß) accumulating is considered as a causative factor for formation of senile plaque in Alzheimer's disease (AD), but its mechanism is still elusive. The Nicotinamide mononucleotide adenylyltransferase 2 (Nmnat2), a key redox cofactor for energy metabolism, is reduced in AD. Accumulative evidence has shown that the decrease of α-secretase activity, a disintegrin and metalloprotease domain 10 (ADAM10), is responsible for the increase of Aß productions in AD patient's brain. Here, we observe that the activity of α-secretase ADAM10 and levels of Nmnat2 are significantly decreased, meanwhile there is a simultaneous elevation of Aß in Tg2576 mice. Over-expression of Nmnat2 increases the mRNA expression of α-secretase ADAM10 and its activity and inhibits Aß production in N2a/APPswe cells, which can be abolished by Compound C, an AMPK antagonist, suggesting that AMPK is involved in over-expression of Nmnat2 against Aß production. The further assays demonstrate that Nmnat2 activates AMPK by up-regulating the ratio of NAD+/NADH, moreover AMPK agonist AICAR can also increase ADAM10 activity and reduces Aß1-40/1-42. Taken together, Nmnat2 suppresses Aß production and up-regulates ADAM10 in AMPK activity-dependent manner, suggesting that Nmnat2 may serve as a new potential target in arresting AD.


Subject(s)
ADAM10 Protein , AMP-Activated Protein Kinases , Amyloid Precursor Protein Secretases , Amyloid , Membrane Proteins , Nicotinamide-Nucleotide Adenylyltransferase , ADAM10 Protein/genetics , ADAM10 Protein/metabolism , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Amyloid/genetics , Amyloid/metabolism , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , Cell Line , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Up-Regulation/genetics
2.
Oxid Med Cell Longev ; 2018: 9647809, 2018.
Article in English | MEDLINE | ID: mdl-29977458

ABSTRACT

Diaphragm dysfunction is an important clinical problem worldwide. Hydrogen sulfide (H2S) is involved in many physiological and pathological processes in mammals. However, the effect and mechanism of H2S in diaphragm dysfunction have not been fully elucidated. In this study, we detected that the level of H2S was decreased in lipopolysaccharide- (LPS-) treated L6 cells. Treatment with H2S increased the proliferation and viability of LPS-treated L6 cells. We found that H2S decreased reactive oxygen species- (ROS-) induced apoptosis through the mitogen-activated protein kinase (MAPK) signaling pathway in LPS-treated L6 cells. Administration of H2S alleviated LPS-induced inflammation by mediating the toll-like receptor-4 (TLR-4)/nuclear factor-kappa B (NF-κB) signaling pathway in L6 cells. Furthermore, H2S improved diaphragmatic function and structure through the reduction of inflammation and apoptosis in the diaphragm of septic rats. In conclusion, these findings indicate that H2S ameliorates LPS-induced diaphragm dysfunction in rats by reducing apoptosis and inflammation through ROS/MAPK and TLR4/NF-κB signaling pathways. Novel slow-releasing H2S donors can be designed and applied for the treatment of diaphragm dysfunction.


Subject(s)
Apoptosis/drug effects , Diaphragm/drug effects , Hydrogen Sulfide/pharmacology , Inflammation/metabolism , Signal Transduction/drug effects , Animals , Apoptosis/physiology , Diaphragm/metabolism , Lipopolysaccharides/toxicity , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , NF-kappa B/drug effects , NF-kappa B/metabolism , Rats , Reactive Oxygen Species/metabolism , Signal Transduction/physiology , Toll-Like Receptor 4/drug effects , Toll-Like Receptor 4/metabolism
3.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 23(6): 1749-52, 2015 Dec.
Article in Chinese | MEDLINE | ID: mdl-26708904

ABSTRACT

OBJECTIVE: To identify the genotypes of the blood sample whose blood grouping showed discrepancies and study the ABO alleles' molecular characteristics of the involved ancestry. METHODS: Blood samples were preliminary genotyped by PCR-SSP. Complete exon 6 and 7 in the ABO genes were amplified by PCR and the PCR products were directly sequenced and cloning sequenced to identify its genotype. RESULTS: Sequence analysis indicated that 3 samples of the family had an nt905A>G mutation in the B gene compared with ABO*B101. Combined with the serological results, the propositus could be typed as Bx02/O102. CONCLUSION: DNA sequencing analysis is able to identify the serological phenotype samples that forward and reverse group methods were incongruous.


Subject(s)
Alleles , ABO Blood-Group System , Base Sequence , Blood Grouping and Crossmatching , Exons , Genetic Testing , Genotype , Humans , Mutation , Phenotype , Polymerase Chain Reaction
4.
Age (Dordr) ; 36(2): 613-23, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24142524

ABSTRACT

Patients with diabetes in the aging population are at high risk of Alzheimer's disease (AD), and reduction of sirtuin 1 (SIRT1) activity occurs simultaneously with the accumulation of hyperphosphorylated tau in the AD-affected brain. It is not clear, however, whether SIRT1 is a suitable molecular target for the treatment of AD. Here, we employed a rat model of brain insulin resistance with intracerebroventricular injection of streptozotocin (ICV-STZ; 3 mg/kg, twice with an interval of 48 h). The ICV-STZ-treated rats were administrated with resveratrol (RSV; SIRT1-specific activator) or a vehicle via intraperitoneal injection for 8 weeks (30 mg/kg, once per day). In ICV-STZ-treated rats, the levels of phosphorylated tau and phosphorylated extracellular signal-regulated kinases 1 and 2 (ERK1/2) at the hippocampi were increased significantly, whereas SIRT1 activity was decreased without change of its expression level. The capacity of spatial memory was also significantly lower in ICV-STZ-treated rats compared with age-matched control. RSV, a specific activator of SIRT1, which reversed the ICV-STZ-induced decrease in SIRT1 activity, increases in ERK1/2 phosphorylation, tau phosphorylation, and impairment of cognitive capability in rats. In conclusion, SIRT1 protects hippocampus neurons from tau hyperphosphorylation and prevents cognitive impairment induced by ICV-STZ brain insulin resistance with decreased hippocampus ERK1/2 activity.


Subject(s)
Cerebral Ventricles/metabolism , Cognition Disorders/drug therapy , Cognition/drug effects , Hippocampus/drug effects , Sirtuin 1/biosynthesis , Stilbenes/administration & dosage , Aging , Animals , Antioxidants/administration & dosage , Blotting, Western , Cerebral Ventricles/drug effects , Cerebral Ventricles/physiopathology , Cognition Disorders/chemically induced , Cognition Disorders/metabolism , Disease Models, Animal , Fluorometry , Hippocampus/metabolism , Hippocampus/physiopathology , Injections, Intraperitoneal , MAP Kinase Signaling System/drug effects , Male , Phosphorylation/drug effects , Rabbits , Rats , Rats, Sprague-Dawley , Resveratrol , Sirtuin 1/drug effects , Streptozocin/toxicity , Vasodilator Agents , tau Proteins/drug effects , tau Proteins/metabolism
5.
J Alzheimers Dis ; 37(4): 795-808, 2013.
Article in English | MEDLINE | ID: mdl-23948915

ABSTRACT

The intracellular accumulation of hyperphosphorylated tau plays a crucial role in neurodegeneration of Alzheimer's disease (AD), but the mechanism is not fully understood. From the observation that tau hyperphosphorylation renders cells more resistant to chemically-induced cell apoptosis, we have proposed that tau-involved apoptotic abortion may be the trigger of neurodegeneration. Here, we further studied whether this phenomenon is also applicable for the cell death induced by constitutively expressed factors, such as death-associated protein kinase 1 (DAPK1). We found that DAPK1 was upregulated and accumulated in the brain of human tau transgenic mice. Overexpression of DAPK1 in HEK293 and N2a cells decreased cell viability with activation of caspase-3, whereas simultaneous expression of tau antagonized DAPK1-induced apoptotic cell death. Expression of DAPK1 induced tau hyperphosphorylation at Thr231, Ser262, and Ser396 with no effects on protein phosphatase 2A, glycogen synthase kinase-3ß, protein kinase A, calcium/calmodulin dependent protein kinase II, cell division cycle 2, or cyclin dependent protein kinase 5. The phosphorylation level of microtubule affinity-regulating kinase 2 (MARK2) was increased by expression of DAPK1, but simultaneous downregulation of MARK2 did not affect the DAPK1-induced tau hyperphosphorylation. DAPK1 was co-immunoprecipitated with tau proteins both in vivo and in vitro, and expression of the kinase domain-truncated DAPK1 did not induce tau hyperphosphorylation. These data suggest that tau hyperphosphorylation at Thr231, Ser262, and Ser396 by DAPK1 renders the cells more resistant to the kinase-induced apoptotic cell death, providing new insights into the tau-involved apoptotic abortion in the course of chronic neurodegeneration.


Subject(s)
Apoptosis/physiology , Death-Associated Protein Kinases/antagonists & inhibitors , Death-Associated Protein Kinases/physiology , tau Proteins/metabolism , Animals , Brain/enzymology , Brain/metabolism , Brain/pathology , Cell Line, Tumor , Death-Associated Protein Kinases/biosynthesis , HEK293 Cells , Humans , Mice , Mice, Transgenic , Phosphorylation/physiology , Up-Regulation/genetics , tau Proteins/analysis
6.
J Alzheimers Dis ; 36(1): 185-95, 2013.
Article in English | MEDLINE | ID: mdl-23579329

ABSTRACT

The activity of protein phosptase-2A (PP2A) is significantly decreased in the brains of Alzheimer's disease (AD) patients, but the upstream effectors for regulating PP2A activity are not fully understood. Nicotinamide mononucleotide adenylyltransferase 2 (Nmnat2) is a key enzyme involved in energy metabolism and its gene expression level is reduced in AD brain specimens. Whether Nmnat2 can activate PP2A deserves to be explored. Here, we first measured the level of Nmnat2, Tyr307-phosphorylation of PP2A, and tau phosphorylation in Tg2576 mice. We observed that the mRNA and protein levels of Nmnat2 were significantly decreased with a simultaneous elevation of p-Tyr307-PP2A and tau phosphorylation in Tg2576 mice. Further studies in HEK293 cells with stable expression of human tau441 (HEK293/tau) demonstrated that simultaneous inhibition of PP2A by okadaic acid abolished the Nmnat2-induced tau dephosphorylation. Moreover, we further demonstrated that overexpression of Nmnat2 could activate PP2A with attenuation of tau phosphorylation, whereas downregulation of Nmnat2 by shRNA inhibited PP2A with tau hyperphosphorylation at multiple AD-associated sites. Our data provide the first evidence that Nmnat2 affects tau phosphorylation by regulating PP2A activity, suggesting that Nmnat2 may serve as a potential target in arresting AD-like tau pathologies.


Subject(s)
Nicotinamide-Nucleotide Adenylyltransferase/physiology , Protein Phosphatase 2/metabolism , tau Proteins/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Animals , Blotting, Western , Disease Models, Animal , Down-Regulation/physiology , Enzyme Activation , HEK293 Cells , Humans , Mice , Okadaic Acid/pharmacology , Phosphorylation , Protein Phosphatase 2/antagonists & inhibitors , Real-Time Polymerase Chain Reaction
7.
J Alzheimers Dis ; 35(1): 91-105, 2013.
Article in English | MEDLINE | ID: mdl-23340038

ABSTRACT

Hyperphosphorylated tau aggregated into neurofibrillary tangles is a hallmark lesion of Alzheimer's disease (AD) and is linked to synaptic and cognitive impairments. In animal models, cold water stress (CWS) can cause cognitive disorder and tau hyperphosphorylation. Capsaicin (CAP), a specific TRPV1 agonist, is neuroprotective against stress-induced impairment, but the detailed mechanisms are still elusive. Here, we investigated whether CAP mitigates CWS-induced cognitive and AD-like pathological alterations in rats. The animals were administered CAP (10 mg/kg in 0.2 ml, 0.1% ethanol) or a control (0.2 ml normal saline, 0.1% ethanol) by intragastric infusion 1 h before CWS treatment. Our results showed that CAP significantly attenuated CWS-induced spatial memory impairment and suppression of PP-DG long-term potentiation; CAP abolished CWS-induced dendritic regression and enhanced several memory-associated proteins decreased by CWS, such as synapsin I and PSD93; CAP also prevented CWS-induced tau hyperphosphorylation by abolishing inhibition of protein phosphatase 2A. Taken together, this study demonstrated that activation of TRPV1 can mitigate CWS-induced AD-like neuropathological alterations and cognitive impairment and may be a promising target for therapeutic intervention in AD.


Subject(s)
Alzheimer Disease/prevention & control , Capsaicin/therapeutic use , Cognition Disorders/prevention & control , Disease Models, Animal , Stress, Psychological/drug therapy , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Animals , Capsaicin/pharmacology , Cognition Disorders/etiology , Cognition Disorders/pathology , Dendritic Spines/drug effects , Dendritic Spines/pathology , Male , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Rats , Rats, Sprague-Dawley , Stress, Psychological/pathology , Stress, Psychological/psychology
8.
J Neurochem ; 124(3): 388-96, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23157378

ABSTRACT

Hyperhomocysteinemia (Hhcy) may induce memory deficits with ß-amyloid (Aß) accumulation and tau hyperphosphorylation. Simultaneous supplement of folate and vitamin B12 partially restored the plasma homocysteine level and attenuated tau hyperphosphorylation, Aß accumulation and memory impairments induced by Hhcy. However, folate and vitamin B12 treatment have no effects on Hhcy which has the methylenetetrahydrofolate reductase genotype mutation. In this study, we investigated the effects of simultaneous supplement of betaine on Alzheimer-like pathological changes and memory deficits in hyperhomocysteinemic rats after a 2-week induction by vena caudalis injection of homocysteine (Hcy). We found that supplementation of betaine could ameliorate the Hcy-induced memory deficits, enhance long-term potentiation (LTP) and increase dendritic branches numbers and the density of the dendritic spines, with up-regulation of NR1, NR2A, synaptotagmin, synaptophysin, and phosphorylated synapsin I protein levels. Supplementation of betaine also attenuated the Hcy-induced tau hyperphosphorylation at multiple AD-related sites through activation protein phosphatase-2A (PP2A) with decreased inhibitory demethylated PP2A(C) at Leu309 and phosphorylated PP2A(C) at Tyr307. In addition, supplementation of betaine also decreased Aß production with decreased presenilin-1 protein levels. Our data suggest that betaine could be a promising candidate for arresting Hcy-induced AD-like pathological changes and memory deficits.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Betaine/toxicity , Homocysteine/toxicity , Hyperhomocysteinemia/drug therapy , Memory Disorders/drug therapy , Alzheimer Disease/blood , Animals , Disease Models, Animal , Homocysteine/blood , Hyperhomocysteinemia/chemically induced , Lipotropic Agents/pharmacology , Male , Memory Disorders/chemically induced , Rats , Rats, Sprague-Dawley
9.
Neuromolecular Med ; 14(4): 338-48, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22798221

ABSTRACT

The hyperphosphorylated tau is a major protein component of neurofibrillary tangle, which is one of hallmarks of Alzheimer's disease (AD). While the level of methylglyoxal (MG) is significantly increased in the AD brains, the role of MG in tau phosphorylation is still not reported. Here, we found that MG could induce tau hyperphosphorylation at multiple AD-related sites in neuroblastoma 2a cells under maintaining normal cell viability. MG treatment increased the level of advanced glycation end products (AGEs) and the receptor of AGEs (RAGE). Glycogen synthesis kinase-3ß (GSK-3ß) and p38 MAPK were activated, whereas the level and activity of JNK, Erk1/2, cdk5, and PP2A were not altered after MG treatment. Simultaneous inhibition of GSK-3ß or p38 attenuated the MG-induced tau hyperphosphorylation. Aminoguanidine, a blocker of AGEs formation, could effectively reverse the MG-induced tau hyperphosphorylation. These data suggest that MG induces AD-like tau hyperphosphorylation through AGEs formation involving RAGE up-regulation and GSK-3ß activation and p38 activation is also partially involved in MG-induced tau hyperphosphorylation. Thus, targeting MG may be a promising therapeutic strategy to prevent AD-like tau hyperphosphorylation.


Subject(s)
Glycation End Products, Advanced/metabolism , Pyruvaldehyde/pharmacology , tau Proteins/metabolism , Alzheimer Disease/metabolism , Animals , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Enzyme Activation/drug effects , Glycation End Products, Advanced/antagonists & inhibitors , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Guanidines/pharmacology , Mice , Neuroblastoma/pathology , Phosphorylation/drug effects , Protein Kinases/metabolism , Protein Processing, Post-Translational/drug effects , Receptor for Advanced Glycation End Products , Receptors, Immunologic/biosynthesis , Receptors, Immunologic/genetics , Up-Regulation/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...