Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
Int J Mol Sci ; 24(3)2023 Jan 20.
Article in English | MEDLINE | ID: mdl-36768379

ABSTRACT

Alzheimer's disease (AD) is the most common neurodegenerative disease and the blood-brain barrier dysfunction has been suggested as a key pathological feature of the disease. Our research group successfully established a synthetic protocol for oleracones, a novel series of flavonoids isolated from the plant extract of Portulaca oleracea L. (PO). PO extract was reported to have anti-inflammatory and antioxidant effects, enhancing cognitive function. Thus, we investigated the effects and mechanism of oleracones on cognition using AD model transgenic mice (Tg; APPswe/PSEN1dE9). Oleracone F treatment significantly improved memory dysfunction in Tg mice. Oleracone F decreased the number, burden, and immunoreactivity of amyloid plaques and amyloid precursor protein (APP) protein levels in the brains of Tg mice compared to wild-type mice. Oleracone F also alleviated inflammation observed in Tg mice brains. In vitro studies in human microvascular endothelial cells (HBMVECs) demonstrated that oleracones D, E, and F blocked the elevations in VCAM-1 protein induced by tumor necrosis factor-α (TNF-α), hindering leukocyte adhesion to HBMVECs. Taken together, our results suggest that oleracones ameliorated cognitive impairment by blocking TNF-α-induced increases in VCAM-1, thereby reducing leukocyte infiltration to the brain and modulating brain inflammation.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Neurodegenerative Diseases , Mice , Humans , Animals , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism , Tumor Necrosis Factor-alpha/metabolism , Endothelial Cells/metabolism , Neurodegenerative Diseases/metabolism , Alzheimer Disease/metabolism , Brain/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Mice, Transgenic , Cognitive Dysfunction/metabolism , Amyloid beta-Peptides/metabolism
2.
Neuromolecular Med ; 25(2): 193-204, 2023 06.
Article in English | MEDLINE | ID: mdl-35948857

ABSTRACT

Intercellular adhesion molecule 1 (ICAM1) is a vessel adhesion protein induced during brain vascular inflammation, which could be closely linked with the development of Alzheimer's disease (AD). This study investigated the effect of ICAM1 on amyloid-degrading enzymes (ADEs) in endothelial cells and their potential involvement in inflammation and AD progression. TNF-α treatment increased ICAM1 in human brain microvascular endothelial cells (HBMVECs) but decreased the neprilysin (NEP) protein level. Knock-down of ICAM1 using siRNA enhanced NEP, which increased the degradation of amyloid-ß. In the brains of 4-month-old AD transgenic mice (APPswe/PSEN1dE9), there were significantly higher levels of ICAM1 expression and amyloid deposits but lower levels of NEP and insulin-degrading enzymes (IDE), demonstrating an inverse correlation of ICAM1 with NEP and IDE expression. Further studies demonstrated significantly increased GFAP protein levels in the brain, specifically localized near blood vessels, of both TNF-α-injected and 4-month-old AD transgenic mice. Taken together, the induction of ICAM1 in endothelial cells suppresses NEP expression, accelerating the accumulation of amyloid-ß in blood vessels. It also enhances leukocyte adhesion to blood vessels stimulating the migration of leukocytes into the brain, subsequently triggering brain inflammation.


Subject(s)
Alzheimer Disease , Insulysin , Mice , Animals , Humans , Infant , Alzheimer Disease/genetics , Neprilysin/genetics , Neprilysin/metabolism , Neprilysin/pharmacology , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Intercellular Adhesion Molecule-1/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Endothelial Cells/metabolism , Amyloid beta-Peptides/metabolism , Mice, Transgenic , Insulysin/genetics , Insulysin/metabolism , Insulysin/pharmacology , Brain/metabolism
3.
Cells ; 11(16)2022 08 19.
Article in English | MEDLINE | ID: mdl-36010661

ABSTRACT

There is currently no effective treatment against Alzheimer's disease (AD), although many strategies have been applied to reduce beta-amyloid (Aß) levels. Here, we investigated 2,4-diacetylphloroglucinol (DAPG) effects on Aß levels and mechanisms of action. DAPG was the most effective phloroglucinol derivative for reducing Aß levels, without being toxic, in various models including HEK293 cells overexpressing Swedish mutant amyloid precursor protein (APP) (293sw), primary astrocytes isolated from APPsw/PS1dE9 transgenic mice, and after intrahippocampal injection of DAPG in APPsw/PS1dE9 transgenic mice. DAPG-mediated Aß reduction was associated with increased soluble APPα (sAPPα) levels mediated by a disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) but not ADAM17. ADAM10 inhibition in DAPG-treated cells prevented the effects on sAPPα but only partly on intracellular and secreted Aß. To identify regulators of sAPPα and Aß secretion, various inhibitors of intracellular trafficking were administered with DAPG. Brefeldin A (BFA) reversed DAPG-mediated changes in Aß secretion in 293sw cells, whereas golgicide A (GCA) and BFA were effective in primary astrocytes, indicating a cell type-specific regulation of the trafficking. Moreover, GCA or BFA effects on sAPPα, but not Aß, levels in primary astrocytes resembled those of ADAM10 inhibition, indicating at least partly independent trafficking pathways for sAPPα and Aß. In conclusion, DAPG might be a promising drug candidate against AD regulating ADAM10 and intracellular trafficking, but optimizing DAPG ability to cross the BBB will be needed.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , ADAM10 Protein/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Animals , HEK293 Cells , Humans , Membrane Proteins/metabolism , Mice , Models, Animal , Phloroglucinol/analogs & derivatives
4.
Tissue Eng Regen Med ; 19(5): 1063-1075, 2022 10.
Article in English | MEDLINE | ID: mdl-35857260

ABSTRACT

BACKGROUND: Mesenchymal stem cells (MSCs) are considered a potential tool for regenerating damaged tissues due to their great multipotency into various cell types. Here, we attempted to find the appropriate conditions for neuronal differentiation of tonsil-derived MSCs (TMSCs) and expand the potential application of TMSCs for treating neurological diseases. METHODS: The TMSCs were differentiated in DMEM/F-12 (Dulbecco's Modified Eagle Medium/Nutrient Mixture F-12) supplemented with various neurotrophic factors for 7-28 days to determine the optimal neuronal differentiation condition for the TMSCs. The morphologies as well as the levels of the neural markers and neurotransmitters were assessed to determine neuronal differentiation potentials and the neuronal lineages of the differentiated TMSCs. RESULTS: Our initial study demonstrated that DMEM/F12 supplemented with 50 ng/mL basic fibroblast growth factor with 10 µM forskolin was the optimal condition for neuronal differentiation for the TMSCs. TMSCs had higher protein expression of neuronal markers, including neuron-specific enolase (NSE), GAP43, postsynaptic density protein 95 (PSD95), and synaptosomal-associated protein of 25 kDa (SNAP25) compared to the undifferentiated TMSCs. Immunofluorescence staining also validated the increased mature neuron markers, NeuN and synaptophysin, in the differentiated TMSCs. The expression of glial fibrillar acidic protein and ionized calcium-binding adaptor molecule 1 the markers of astrocytes and microglia, were also slightly increased. Additionally, the differentiated TMSCs released a significantly higher level of acetylcholine, the cholinergic neurotransmitter, as analyzed by the liquid chromatography-tandem mass spectrometry and showed an enhanced choline acetyltransferase immunoreactivity compared to the undifferentiated cells. CONCLUSION: Our study suggests that the optimized condition favors the TMSCs to differentiate into cholinergic neuron-like phenotype, which could be used as a possible therapeutic tool in treating certain neurological disorders such as Alzheimer's disease.


Subject(s)
Mesenchymal Stem Cells , Palatine Tonsil , Acetylcholine/metabolism , Calcium/metabolism , Choline O-Acetyltransferase/metabolism , Cholinergic Agents/metabolism , Colforsin/metabolism , Disks Large Homolog 4 Protein/metabolism , Fibroblast Growth Factor 2/metabolism , Mesenchymal Stem Cells/metabolism , Nerve Growth Factors/metabolism , Phosphopyruvate Hydratase/metabolism , Synaptophysin/metabolism
5.
Antioxidants (Basel) ; 11(2)2022 Jan 20.
Article in English | MEDLINE | ID: mdl-35204080

ABSTRACT

The blood-brain barrier (BBB), which consists mainly of brain microvascular endothelial cells and astrocytes connected by tight junctions (TJs) and adhesion molecules (AMs), maintains the homeostatic balance between brain parenchyma and extracellular fluid. Accumulating evidence shows that BBB dysfunction is a common feature of neurodegenerative diseases, including stroke, traumatic brain injury, and Alzheimer's disease. Among the various pathological pathways of BBB dysfunction, reactive oxygen species (ROS) are known to play a key role in inducing BBB disruption mediated via TJ modification, AM induction, cytoskeletal reorganization, and matrix metalloproteinase activation. Thus, antioxidants have been suggested to exert beneficial effects on BBB dysfunction-associated brain diseases. In this review, we summarized the sources of ROS production in multiple cells that constitute or surround the BBB, such as BBB endothelial cells, astrocytes, microglia, and neutrophils. We also reviewed various pathological mechanisms by which BBB disruption is caused by ROS in these cells. Finally, we summarized the effects of various natural polyphenols on BBB dysfunction to suggest a therapeutic strategy for BBB disruption-related brain diseases.

6.
Cells ; 11(3)2022 02 03.
Article in English | MEDLINE | ID: mdl-35159343

ABSTRACT

Our previous study found that the level of CCN1 increases as osteogenic differentiation progresses in tonsil-derived mesenchymal stem cells (TMSCs). This study investigated how CCN1 is regulated through HDAC inhibition in TMSCs and their relationship with osteogenesis. Valproic acid (VPA) (1-5 mM), a well-known histone deacetylase (HDAC) inhibitor, strongly inhibited TMSC proliferation without altering MSC-specific surface markers, CD14, 34, 45, 73, 90 and 105. However, CD146 expression increased at 5 mM VPA. VPA increased osteogenic differentiation of TMSCs but decreased adipogenesis and chondrogenesis, as evidenced by the cell-specific staining of differentiation. The former was validated by the increased osteocalcin (OCN). The changes in CCN1 by VPA was biphasic; it increased until 48 h and decreased thereafter. Knockdown of CCN1 by using siRNA inhibited the osteogenic effect of VPA. VPA had no effect on CCN1 mRNA expression, but inhibition of protein synthesis by cycloheximide showed that VPA slowed down the CCN1 protein degradation. Moreover, overexpression of HDAC1 completely inhibited VPA-induced CCN1. Our results indicate that VPA inhibits the HDAC1, inducing CCN1 protein stability rather than gene expression, thereby promoting osteogenic differentiation of TMSCs. These findings present the noble implication of VPA as an inhibitor of HDAC1 to facilitate CCN1-induced osteogenic differentiation of MSCs.


Subject(s)
Mesenchymal Stem Cells , Osteogenesis , Cysteine-Rich Protein 61/metabolism , Histone Deacetylase Inhibitors/pharmacology , Mesenchymal Stem Cells/metabolism , Palatine Tonsil , Protein Stability , Valproic Acid/metabolism , Valproic Acid/pharmacology
7.
Biomol Ther (Seoul) ; 28(3): 230-239, 2020 May 01.
Article in English | MEDLINE | ID: mdl-32028757

ABSTRACT

Previous studies have shown disrupted synaptic plasticity and neural activity in depression. Such alteration is strongly associated with disrupted synaptic structures. Chronic stress has been known to induce changes in dendritic structure in the basolateral amygdala (BLA) and medial prefrontal cortex (mPFC), but antidepressant effect on structure of these brain areas has been unclear. Here, the effects of imipramine on dendritic spine density and morphology in BLA and mPFC subregions of stressed mice were examined. Chronic restraint stress caused depressive-like behaviors such as enhanced social avoidance and despair level coincident with differential changes in dendritic spine structure. Chronic stress enhanced dendritic spine density in the lateral nucleus of BLA with no significant change in the basal nucleus of BLA, and altered the proportion of stubby or mushroom spines in both subregions. Conversely, in the apical and basal mPFC, chronic stress caused a significant reduction in spine density. The proportion of stubby or mushroom spines in these subregions overall reduced while the proportion of thin spines increased after repeated stress. Interestingly, most of these structural alterations by chronic stress were reversed by imipramine. In addition, structural changes caused by stress and blocking the changes by imipramine were corelated well with altered activation and expression of synaptic plasticity-promoting molecules such as phospho-CREB, phospho-CAMKII, and PSD-95. Collectively, our data suggest that imipramine modulates stress-induced changes in synaptic structure and synaptic plasticity-promoting molecules in a coordinated manner although structural and molecular alterations induced by stress are distinct in the BLA and mPFC.

8.
Cytotherapy ; 20(8): 1013-1027, 2018 08.
Article in English | MEDLINE | ID: mdl-30072298

ABSTRACT

BACKGROUND AND AIMS: Osteoporosis, which is a disease characterized by weakening of the bone, affects a large portion of the senior population. The current therapeutic options for osteoporosis have side effects, and there is no effective treatment for severe osteoporosis. Thus, we urgently need new treatment strategies, such as topical therapies and/or safe and effective stem cell therapies. METHODS: We investigated the therapeutic potential of directly injecting human tonsil-derived mesenchymal stem cells (TMSC) into the right proximal tibias of ovariectomized postmenopausal osteoporosis model mice. Injections were given once (1×) or twice (2×) during the 3-month experimental period. At the end of the experiment, micro-computed tomographic images revealed some improvement in the proximal tibias and more significant improvement in the femoral heads of treated mice. RESULTS: Osteogenic effect was qualitatively and quantitatively more pronounced in TMSC/2×-treated mice. Furthermore, TMSC/2× mice exhibited significant recovery of the serum osteocalcin level, which is pathologically elevated in osteoporosis, and increased serum alkaline phosphatase, which indicates bone formation. TMSC therapy was generally well tolerated and caused no apparent toxicity in the experimental mice. Moreover, TMSC therapy reduced visceral fat. CONCLUSION: Our results demonstrate that double injection of TMSC directly into the proximal tibia triggers recovery of osteoporosis, and thus could be a potential therapeutic approach for severe bone loss.


Subject(s)
Infusions, Intraosseous , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Osteoporosis/therapy , Palatine Tonsil/cytology , Animals , Bone Density/physiology , Child , Female , Heterografts , Humans , Infusions, Intraosseous/methods , Injections , Male , Mice , Mice, Inbred ICR , Osteoporosis/diagnosis , Osteoporosis/pathology , Postmenopause/physiology , Remission Induction , Tibia/diagnostic imaging
9.
Biochem Biophys Res Commun ; 497(1): 416-423, 2018 02 26.
Article in English | MEDLINE | ID: mdl-29448109

ABSTRACT

Our previous study showed that the level of glutamate carboxypeptidase II (GCPII) protein is regulated by valproic acid, a histone deacetylase (HDAC) inhibitor, through acetylation of lysine residue in the GCPII protein in human astrocytes, U-87MG. The present study further investigated which HDAC subtype is involved in the acetylation of GCPII. The results revealed that GCPII interacted with HDAC1 but not with HDAC2, HDAC3, HDAC4, HDAC5, and HDAC6. Overexpression of catalytic domain (1-56 aa)-deleted HDAC1, which poorly binds to GCPII, enhanced lysine acetylation in GCPII and increased the level of GCPII protein when compared with that of the wild-type HDAC1. Further experiments showed that HDAC1 regulated the stability of GCPII protein. These data suggest that acetylation of GCPII is facilitated by HDAC1, and the acetylated GCPII is more stable than the non-acetylated GCPII. Additional experiments using siRNA HDAC1 and by HDAC1 overexpression confirmed the role of HDAC1 in regulating the stability of GCPII protein. Further, database search of acetylation and ubiquitination sites showed four candidate lysine sites in human GCPII protein that can be both acetylated and ubiquitinylated (K207, K479, K491, and K699). Mutation (lysine residues to arginine (R)) analysis showed that in the presence of cycloheximide K479R- and K491R-hGCPII mutants were less ubiquitinylated and degraded, and decrease in the level of GCPII protein by HDAC1 was significantly blocked by K479R mutants. These data suggest that K479 is a possible site of acetylation or ubiquitination. Furthermore, the results also demonstrate that the stability of GCPII protein is regulated by HDAC1 through acetylation at the lysine 479 residue.


Subject(s)
Astrocytes/metabolism , Histone Deacetylase 1/metabolism , Lysine/metabolism , Acetylation , Antigens, Surface , Cell Line , Enzyme Stability , Glutamate Carboxypeptidase II , Humans , Lysine/chemistry , Substrate Specificity
10.
Cornea ; 36(9): 1116-1123, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28644233

ABSTRACT

PURPOSE: Vatalanib is a small-molecule tyrosine kinase inhibitor. We investigated the effects of vatalanib on the proliferation and migration of cultured human pterygial fibroblasts (HPFs). METHODS: Pterygium tissues were obtained after pterygium excision surgery and subjected to primary culture. HPFs were treated with vatalanib at various concentrations. Mitomycin C (MMC) was used as a positive control. Cell proliferation and migration assays were used to investigate the effects of vatalanib. Cell death was measured using flow cytometry analysis. Western blot analysis was performed to identify signaling molecules associated with the response to vatalanib. RESULTS: Vatalanib inhibited both proliferation and migration of HPFs in a dose-dependent manner. Cell proliferation was significantly suppressed by vatalanib (10 and 100 µM) and MMC (0.004% and 0.04%) treatments. Migration assays revealed significant HPF delay when treated with vatalanib (1, 10, and 100 µM) and MMC (0.004% and 0.04%) compared with that in a negative control. Cell death analysis showed that high concentrations of vatalanib (100 µM) and MMC (0.004% and 0.04%) decreased cell numbers. Western blot analysis of vatalanib-treated cells showed vascular endothelial growth factor and transforming growth factor-ß significantly reduced, but there was no alteration in p53 protein levels in HPFs. CONCLUSIONS: These results indicate that vatalanib significantly suppressed the proliferation and migration of HPFs by decreasing vascular endothelial growth factor and transforming growth factor-ß. Vatalanib showed less toxicity than that of MMC. Based on these results, vatalanib may potentially serve as a new adjuvant treatment after pterygium excision surgery.


Subject(s)
Fibroblasts/drug effects , Phthalazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pterygium/drug therapy , Pyridines/pharmacology , Blotting, Western , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Humans , Transforming Growth Factor beta/antagonists & inhibitors , Vascular Endothelial Growth Factor A/antagonists & inhibitors
11.
Biochem Biophys Res Commun ; 478(3): 1355-62, 2016 09 23.
Article in English | MEDLINE | ID: mdl-27565733

ABSTRACT

Intercellular adhesion molecule1 (ICAM1) is involved in adhesion and transmigration of leukocytes across endothelium, promoting brain inflammation and leading to brain diseases. Here, we studied the mechanism that regulates ICAM expression in response to proinflammatory cytokine tumor necrosis factor alpha (TNF-α). ICAM1 mRNA and protein levels in human brain microvascular endothelial cells dramatically increased after TNF-α treatment. TNF-α also upregulated histone demethylases KDM1B and KDM7A responsible for demethylation of H3K9me2, a well-known repression marker. Knockdown of KDM7A by a small interfering RNA reduced the ICAM1 protein level and leukocyte adhesion without an effect on ICAM1 mRNA expression. In contrast, a KDM1B knockdown did not affect TNF-α-induced ICAM1 expression. Thus, KDM7A-mediated ICAM1 protein upregulation is likely related to protein stability, not a histone-mediated epigenetic mechanism. Experiments with cycloheximide supported the role of KDM7A in ICAM1 protein stabilization. Further experiments suggest that KDM7A regulates ICAM1 protein stability via a lysosome-dependent pathway. Lysosome inhibitors increased the TNF-α-induced ICAM1 protein level and restored KDM7A knockdown-induced downregulation of ICAM1. In contrast, the KDM7A knockdown had no effect on proteasome-mediated ICAM1 degradation. We also found that the transcription factor EB protein level reduced in response to TNF-α but increased by the KDM7A knockdown. Immunocytochemical analysis revealed weak lysosome formation with high ICAM1 immunoreactivity after TNF-α treatment, but KDM7A knockdown reversed this response, resulting in strong lysosome formation with ICAM1 protein clustering in lysosomes. Taken together, our results show that KDM7A mediates TNF-α-induced ICAM1 protein upregulation and is mediated by induction of KDM7A, which regulates the TFEB-mediated lysosomal activity.


Subject(s)
Intercellular Adhesion Molecule-1/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Lysosomes/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Cells, Cultured , Humans , Lysosomes/drug effects , Protein Stability/drug effects , RNA, Small Interfering/metabolism , Up-Regulation/drug effects
12.
FEBS J ; 283(18): 3438-56, 2016 09.
Article in English | MEDLINE | ID: mdl-27444540

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE) is an inflammatory disease in the murine central nervous system (CNS) and recapitulates the clinical and pathological features of human multiple sclerosis (MS). Glutamate carboxipeptidase II (GCPII), an enzyme expressed exclusively on astrocytes, is known to affect the disease progression of various neurological disorders by producing glutamate. Despite several findings indicating possible link between glutamate and MS/EAE, however, the involvement of astrocyte or GCPII on glutamate excitotoxicity has not received much attention in MS/EAE. When we examined GCPII expression during EAE progression in this study, we observed significantly elevated GCPII expression in peak stage of disease localized mainly in astrocytes. Intrigued by these results, we tried a potent GCPII inhibitor, 2-phosphonomethyl pentanedioic acid (2-PMPA), on EAE mice and noticed markedly attenuated EAE clinical signs along with significantly inhibited infiltration of inflammatory cells into CNS. Furthermore, 2-PMPA dampened the function of Th1 cell lineage and down-regulated mGluR1 expression in both periphery and CNS contributing to glutamate-mediated immune regulation. Our observations identify a sequence of events triggering EAE through GCPII overexpression, which may offer a novel therapeutic approach to the treatment of MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/enzymology , Glutamate Carboxypeptidase II/antagonists & inhibitors , Organophosphorus Compounds/pharmacology , Amino Acids/pharmacology , Animals , Astrocytes/drug effects , Astrocytes/enzymology , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/immunology , Excitatory Amino Acid Antagonists/pharmacology , Female , Glutamate Carboxypeptidase II/metabolism , Glutamic Acid/metabolism , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Protease Inhibitors/pharmacology , Rats, Inbred Lew , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/metabolism , Spinal Cord/drug effects , Spinal Cord/enzymology , Spinal Cord/immunology , Th1 Cells/drug effects , Th1 Cells/immunology , Xanthenes/pharmacology
13.
Psychiatry Investig ; 13(1): 127-34, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26766955

ABSTRACT

OBJECTIVE: Biological markers for Alzheimer's disease (AD) will help clinicians make objective diagnoses early during the course of dementia. Previous studies have suggested that cell cycle dysregulation begins earlier than the onset of clinical manifestations in AD. METHODS: We examined the lymphocyte expression of cell cycle proteins in AD patients, dementia controls (DC), and normal controls (NC). One-hundred seventeen subjects (36 AD, 31 DC, and 50 NC) were recruited. The cell cycle proteins CDK2, CDK4, CDK6, cyclin B, and cyclin D were measured in peripheral lymphocytes. Cell cycle protein expression in the three groups was compared after adjusting for age and sex. RESULTS: The levels of cell cycle proteins CDK2, CDK4, CDK6, cyclin B, and cyclin D were significantly higher in AD patients than in the NC subjects. The DC group manifested intermediate levels of cell cycle proteins compared with the AD patients and the NC subjects. The present study indicates that cell cycle proteins are upregulated in the peripheral lymphocytes of AD patients. CONCLUSION: Cell cycle dysregulation in peripheral lymphocytes may present a promising starting point for identifying peripheral biomarkers of AD.

14.
Tissue Eng Regen Med ; 13(1): 66-69, 2016 Feb.
Article in English | MEDLINE | ID: mdl-30603386

ABSTRACT

T-vectors are widely used for cloning the polymerase chain reaction (PCR) products. However, the low conversion efficiency of a plasmid into the linear T-vector usually results in non-recombinants. Here, we designed a new plasmid pNBQ-T to easily select the recombinant colonies harboring PCR products. pNBQ-T plasmid, which contains a DsRed indicator gene between two Nt.BspQI restriction cassettes, each of which contains palindromic sequences susceptible to Nt.BspQI nickase (5'-GCTCTTCT^GAAGAGC-3') at each end. Thus, this plasmid can be easily converted into T-vectors by a nickase (quadruple nicking), which results in two double strand breaks with 3'-thymidine overhangs. DsRed indicator gene, which is inserted between the restriction sites, helps identifying the PCR recombinants. Using this pNBQ-T plasmid the insertion efficiency of a PCR product was examined. We successfully identified white colony of the recombinants with the inserted myostatin promoter gene: the cloning efficiency was 93%. Therefore, this simple method utilizing pNBQ-T plasmid will serve as a useful and efficient technique for preparation of home-made T-vectors.

15.
PLoS One ; 10(9): e0138590, 2015.
Article in English | MEDLINE | ID: mdl-26375285

ABSTRACT

The green tea component (-)-epigallocatechin-3-gallate (EGCG) has been shown to sensitize many different types of cancer cells to anticancer drug-induced apoptosis, although it protects against non-cancerous primary cells against toxicity from certain conditions such as exposure to arsenic (As) or ultraviolet irradiation. Here, we found that EGCG promotes As-induced toxicity of primary-cultured bovine aortic endothelial cells (BAEC) at doses in which treatment with each chemical alone had no such effect. Increased cell toxicity was accompanied by an increased condensed chromatin pattern and fragmented nuclei, cleaved poly(ADP-ribose) polymerase (PARP), activity of the pro-apoptotic enzymes caspases 3, 8 and 9, and Bax translocation into mitochondria, suggesting the involvement of an apoptotic signaling pathway. Fluorescence activated cell sorting analysis revealed that compared with EGCG or As alone, combined EGCG and As (EGCG/As) treatment significantly induced production of reactive oxygen species (ROS), which was accompanied by decreased catalase activity and increased lipid peroxidation. Pretreatment with N-acetyl-L-cysteine or catalase reversed EGCG/As-induced caspase activation and EC toxicity. EGCG/As also increased the phosphorylation of c-Jun N-terminal kinase (JNK), which was not reversed by catalase. However, pretreatment with the JNK inhibitor SP600125 reversed all of the observed effects of EGCG/As, suggesting that JNK may be the most upstream protein examined in this study. Finally, we also found that all the observed effects by EGCG/As are true for other types of EC tested. In conclusion, this is firstly to show that EGCG sensitizes non-cancerous EC to As-induced toxicity through ROS-mediated apoptosis, which was attributed at least in part to a JNK-activated decrease in catalase activity.


Subject(s)
Aorta/pathology , Arsenites/pharmacology , Catalase/metabolism , Catechin/analogs & derivatives , Endothelium, Vascular/pathology , JNK Mitogen-Activated Protein Kinases/metabolism , Tea/chemistry , Animals , Antioxidants/pharmacology , Aorta/drug effects , Aorta/enzymology , Blotting, Western , Catechin/pharmacology , Cattle , Cells, Cultured , Drug Resistance/drug effects , Drug Synergism , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Humans , Immunoenzyme Techniques , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondria/pathology , Oxidation-Reduction , Phosphorylation/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Teratogens/pharmacology
16.
Neurosci Lett ; 580: 163-8, 2014 Sep 19.
Article in English | MEDLINE | ID: mdl-25116759

ABSTRACT

Depression is one of the most prevalent mental illnesses, and causes a constant feeling of sadness and lose of interest, which often leads to suicide. Evidence suggests that depression is associated with aberrant MEK/ERK signaling. However, studies on MEK/ERK signaling in depression have only been done in a few brain regions, such as the hippocampus and mesolimbic reward pathways. Recent studies also implicate the involvement of the prefrontal cortex in depression. Thus, we examined the changes in MEK/ERK signaling in subregions of the prefrontal cortex of C57BL/6 mice by immunohistochemistry using phospho-MEK1/2 (Ser 217/221) and ERK1/2 (Thr202/Tyr204) antibodies. Mice were subjected to 21 consecutive days of restraint stress (for 2h daily), and depression-like behavior was evaluated using a sociability test and tail suspension test. The antidepressant, imipramine (20mg/kg) was injected intraperitoneally 30min before restraint stress exposure. Chronic/repeated restraint stress produced depressive-like behavior, such as increased social avoidance in the social interaction test, and enhanced immobility time in the tail suspension test. This depressive behavior was ameliorated by imipramine. The behavioral changes well corresponded to a decrease in MEK/ERK immunoreactivity in the medial orbital (MO) cortex and dorsal endopiriform nuclei (DEn), which was averted by imipramine, but not in cingulate, prelimbic, infralimbic, and motor cortex. These results suggest that MEK/ERK signaling is disrupted in the DEn and MO subregions of the prefrontal cortex in the depressive phenotype, and that blocking a decrease in activated MEK/ERK is inherent to the antidepressant imipramine response.


Subject(s)
Depression/enzymology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Piriform Cortex/enzymology , Prefrontal Cortex/enzymology , Restraint, Physical , Stress, Psychological/enzymology , Animals , Antidepressive Agents/therapeutic use , Chronic Disease , Depression/psychology , Imipramine/therapeutic use , Male , Mice, Inbred C57BL , Phosphorylation , Stress, Psychological/drug therapy , Stress, Psychological/psychology
17.
Biochem Biophys Res Commun ; 450(1): 372-7, 2014 Jul 18.
Article in English | MEDLINE | ID: mdl-24939622

ABSTRACT

Glutamate carboxypeptidase II (GCPII) is known to be implicated in brain diseases such as schizophrenia and bipolar disorder, and dramatically increases in prostate cancer. Here, we investigated the regulation of GCPII expression in astrocytes and examined whether GCPII is epigenetically regulated through histone modification. In this study, valproic acid (VPA), a drug used for bipolar disorder and epilepsy and a known histone deacetylase (HDAC) inhibitor was used. We found that acute exposure of VPA for 4-6h increased the GCPII protein level in human astrocyte U87MG cells but did not have a similar effect after 12-24h exposure. Real-time polymerase chain reaction analysis revealed that VPA did not affect the GCPII mRNA expression. In contrast, decrease in GCPII protein level by cycloheximide treatment was blocked by VPA, indicating that VPA increases GCPII protein stability. Treatment with MG132, a proteasome inhibitor, suggested that the VPA-induced increase of GCPII protein level is dependent on the ubiquitin/proteasome pathway. In addition, immunoprecipitation analysis revealed that VPA increased the acetylation of GCPII protein at the lysine residues and facilitated a decrease of the poly-ubiquitinated GCPII level. Similarly, M344, a specific HDAC 1/6 inhibitor, also increased the GCPII protein level. In contrast, treatment with C646, a histone acetyltransferase inhibitor of p300/CBP, significantly reduced the level of GCPII protein. Taken together, this study demonstrated that the increase in GCPII induced by VPA is not due to the classical epigenetic mechanism, but via enhanced acetylation of lysine residues in GCPII.


Subject(s)
Antigens, Surface/metabolism , Astrocytes/enzymology , Glutamate Carboxypeptidase II/metabolism , Valproic Acid/pharmacology , Acetylation/drug effects , Astrocytes/drug effects , Cell Line , Enzyme Stability , Humans
18.
FEBS J ; 280(24): 6691-701, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24200051

ABSTRACT

Calpains are involved in calcium-induced neuronal cell toxicity, which is associated with the pathophysiology of Alzheimer's disease (AD). The activity of calpains is regulated by the inhibitor calpastatin, and increased activity of calpains and decreased calpastastin are often found in AD. Histone deacetylase (HDAC) inhibitors are implicated in AD treatment through the improvement of learning and memory but the underlying mechanism is yet to be understood. Here, using SH-SY5Y neuroblastoma cells and a calcium ionophore ionomycin, we examined whether and how HDAC inhibitor trichostatin A (TSA) inhibits calcium-induced neuronal cell death. TSA increased both the mRNA and protein levels of calpastatin, with no alterations in those of calpain 1 and calpain 2. Furthermore, TSA-stimulated increase of calpastatin was accompanied by a significant attenuation of ionomycin-induced autolysis of calpain 1, but not of calpain 2, and calpain-dependent 150 kDa αII spectrin cleavage. Under these conditions, however, caspase activity was unaltered. Moreover, ectopic expression of small interfering RNA of calpastatin reversed the inhibitory effect of TSA on ionomycin-induced calpain 1 autolysis and αII spectrin cleavage. Chromatin immunoprecipitation assay revealed the increased levels of acetylation at lysine 5 of histone H4 (H4K5-Ac), H3K9-Ac and H3K14-Ac within the calpastatin promoter region in TSA-treated cells relative to control cells. Finally, TSA significantly decreased ionomycin-induced cell toxicity. This study demonstrates that TSA attenuates calcium-induced neuronal cell death by the inhibition of calpain activity which is mediated in part by increased calpastatin expression via histone hyperacetylation within the calpastatin promoter region. Our study provides a novel mechanism for the neuroprotective effect of HDAC inhibitors on AD.


Subject(s)
Apoptosis/drug effects , Calcium-Binding Proteins/genetics , Calcium/pharmacology , Calpain/metabolism , Histones/metabolism , Hydroxamic Acids/pharmacology , Neuroblastoma/pathology , Acetylation/drug effects , Blotting, Western , Calcium-Binding Proteins/antagonists & inhibitors , Calcium-Binding Proteins/metabolism , Calpain/genetics , Cell Proliferation/drug effects , Chromatin Immunoprecipitation , Epigenomics , Histone Deacetylase Inhibitors/pharmacology , Humans , Ionomycin/pharmacology , Luciferases/metabolism , Neuroblastoma/genetics , Neuroblastoma/metabolism , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
19.
Biochem Biophys Res Commun ; 438(4): 765-71, 2013 Sep 06.
Article in English | MEDLINE | ID: mdl-23891752

ABSTRACT

We recently reported that glutamate carboxypeptidase II (GCPII) has a new physiological function degrading amyloid-ß (Aß), distinct from its own hydrolysis activity in N-acetyl-L-aspartyl-L-glutamate (NAAG); however, its underlying mechanism remains undiscovered. Using site-directed mutagenesis and S1 pocket-specific chemical inhibitor (compound 2), which was developed for the present study based on in sillico computational modeling, we discovered that the Aß degradation occurs through S1 pocket but not through S1' pocket responsible for NAAG hydrolysis. Treatment with compound 2 prevented GCPII from Aß degradation without any impairment in NAAG hydrolysis. Likewise, 2-PMPA (specific GCPII inhibitor developed targeting S1' pocket) completely blocked the NAAG hydrolysis without any effect on Aß degradation. Pre-incubation with NAAG and Aß did not affect Aß degradation and NAAG hydrolysis, respectively. These data suggest that GCPII has two distinctive binding sites for two different substrates and that Aß degradation occurs through binding to S1 pocket of GCPII.


Subject(s)
Alzheimer Disease/enzymology , Amyloid beta-Peptides/metabolism , Glutamate Carboxypeptidase II/metabolism , Proteolysis , Alzheimer Disease/metabolism , Animals , Binding Sites/drug effects , Cell Line, Tumor , Dipeptides/metabolism , Enzyme Inhibitors/pharmacology , Glutamate Carboxypeptidase II/antagonists & inhibitors , Glutamate Carboxypeptidase II/chemistry , Glutamate Carboxypeptidase II/genetics , Glutamic Acid/metabolism , Humans , Mice , Mice, Transgenic , Molecular Docking Simulation , Mutagenesis, Site-Directed , Organophosphorus Compounds/pharmacology , Proteolysis/drug effects
20.
Eur Neurol ; 70(1-2): 1-5, 2013.
Article in English | MEDLINE | ID: mdl-23636001

ABSTRACT

BACKGROUND: There is still debate over the utility of carotid intima-media thickness (C-IMT) or carotid plaque in predicting future cardiovascular events and death. Additionally, the importance of plasma homocysteine levels was raised as a predictor of cardiovascular events and death. METHODS: 1,391 subjects were recruited from the Ansan Geriatric cohort. We used B-mode carotid ultrasonography to assess C-IMT and plaque, measuring average maximal IMT and average mean IMT through 6-8 measurements of far-wall IMT in both common carotid arteries. We evaluated the presence of plaque in carotid segments. Multivariable Cox regression analysis was used to predict both cardiovascular and all-cause mortality. RESULTS: During a mean follow-up of 62.4 ± 12.4 months, 71 subjects (5.12%) died and 23 (1.66%) died of cardiovascular causes. Multivariable Cox regression analysis found the predictors of cardiovascular mortality to be average maximal IMT (HR = 3.709; 95% CI: 1.202-11.446) and plasma homocysteine (HR = 1.057; 95% CI: 1.012-1.103). All-cause mortality was independently associated with C-IMT (average maximal and average mean IMT) and plasma homocysteine. CONCLUSIONS: C-IMT and plasma homocysteine levels were found to predict cardiovascular and all-cause mortality independently of the presence of carotid plaque and other cardiovascular risk factors.


Subject(s)
Cardiovascular Diseases/blood , Cardiovascular Diseases/mortality , Cardiovascular Diseases/pathology , Carotid Intima-Media Thickness , Homocysteine/blood , Aged , Carotid Arteries/diagnostic imaging , Cause of Death , Cohort Studies , Female , Humans , Male , Proportional Hazards Models , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL