Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 294(16): 6306-6317, 2019 04 19.
Article in English | MEDLINE | ID: mdl-30814253

ABSTRACT

Pathological aggregation of the transactive response DNA-binding protein of 43 kDa (TDP-43) is associated with several neurodegenerative disorders, including ALS, frontotemporal dementia, chronic traumatic encephalopathy, and Alzheimer's disease. TDP-43 aggregation appears to be largely driven by its low-complexity domain (LCD), which also has a high propensity to undergo liquid-liquid phase separation (LLPS). However, the mechanism of TDP-43 LCD pathological aggregation and, most importantly, the relationship between the aggregation process and LLPS remains largely unknown. Here, we show that amyloid formation by the LCD is controlled by electrostatic repulsion. We also demonstrate that the liquid droplet environment strongly accelerates LCD fibrillation and that its aggregation under LLPS conditions involves several distinct events, culminating in rapid assembly of fibrillar aggregates that emanate from within mature liquid droplets. These combined results strongly suggest that LLPS may play a major role in pathological TDP-43 aggregation, contributing to pathogenesis in neurodegenerative diseases.


Subject(s)
Amyloid/chemistry , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/isolation & purification , Protein Aggregation, Pathological , DNA-Binding Proteins/metabolism , Humans , Protein Domains
2.
Food Funct ; 9(1): 234-242, 2018 Jan 24.
Article in English | MEDLINE | ID: mdl-29168878

ABSTRACT

Quercetin and fisetin, known as catechol-containing flavonoids, could positively affect the absorption of catechins due to their strong affinity for catechol-O-methyl transferase (COMT), which can methylate and cause the excretion of catechins. The current study examined the effect of quercetin and fisetin on the absorption of epi-catechins (ECs) by using a Caco-2 cell line and an in vivo model. The intestinal transport of total catechins by Caco-2 cells was enhanced from 1.3- to 1.6-fold and 1.4- to 1.7-fold by adding quercetin and fisetin, respectively, compared to the control. It was even higher in the treatment with a mixture of quercetin and fisetin. While EC had the highest value of intestinal transport (169% of the control) in 10% quercetin treatment, EGC (235%), EGCG (244%), and ECG (242%) were significantly transported in the treatment with a 5% mixture of quercetin and fisetin (p < 0.05). In an in vivo pharmacokinetic study, the values of the area under the plasma concentration-time curve (AUC, ng h mL-1) were also higher in rats orally administered EGCG with 10% quercetin (365.5 ± 25.5) or 10% fisetin (825.3 ± 46.7) than in those administered EGCG only (111.3 ± 13.1). Methylated quercetin and methylated fisetin were determined to be m/z 317.24 and m/z 301.25 [M + H]+ with their own product ions, respectively. The results indicate that quercetin or fisetin is superior to ECs for methylation by COMT.


Subject(s)
Catechin/blood , Flavonoids/administration & dosage , Intestine, Small/drug effects , Plant Extracts/blood , Quercetin/administration & dosage , Animals , Caco-2 Cells , Camellia sinensis/chemistry , Catechin/pharmacokinetics , Flavonoids/chemistry , Flavonols , Humans , Intestine, Small/metabolism , Male , Methylation , Plant Extracts/pharmacokinetics , Quercetin/chemistry , Rats , Rats, Sprague-Dawley
3.
Food Funct ; 8(10): 3664-3674, 2017 Oct 18.
Article in English | MEDLINE | ID: mdl-28914949

ABSTRACT

The impacts of onion peel (OP) and Dendropanax morbifera (DM), as excipient foods rich in flavonols, on the digestive recovery, intestinal absorption, and pharmacokinetics of GT epicatechins were studied via an in vitro digestion model system with Caco-2 cells and an in vivo study. The digestive stability of total epicatechins recovered from GT upon the addition of 2% DM was up to 1.12 times higher than that observed with OP. The combined effects of OP and DM, which were observed with 2% OP + DM in a ratio of 1 : 4 (w : w), significantly increased (by a factor of 1.31) the digestive recovery of total epicatechins (p < 0.05). Remarkable cellular uptakes of EC (185.36%) and ECG (188.08%) were found with 4% OP + DM (4 : 1, w : w), and those of EGC (112.30%) and EGCG (136.27%) were obtained with 2% OP + DM (4 : 1, w : w) and 1% OP + DM (1 : 1, w : w), respectively. The peak plasma concentrations of total epicatechins from GT, GT + 5% OP, GT + 5% DM, and GT + 2% OP + 2% DM were 1044.78 ± 609.10, 2267.18 ± 3734.38, 1270.35 ± 547.59, and 714.53 ± 499.27 ng mL-1, respectively. The Cmax value of total epicatechins in rats orally administrated with GT with 5% OP was found to be approximately twice of that obtained with GT alone. The co-ingestion of GT with flavonol-rich excipient foods possibly enhances the absorption of epicatechins because flavonols act as not only enhancers of digestive stability but also modulators of the biotransformation of epicatechins. The results obtained from the current study suggest that the absorption of GT catechins can vary depending upon the kinds and doses of excipient foods co-ingested.


Subject(s)
Araliaceae/chemistry , Catechin/chemistry , Catechin/pharmacokinetics , Flavonoids/chemistry , Onions/chemistry , Plant Extracts/chemistry , Tea/chemistry , Animals , Biological Availability , Caco-2 Cells , Catechin/administration & dosage , Excipients/chemistry , Humans , Male , Plant Extracts/pharmacokinetics , Rats, Sprague-Dawley
4.
Proc Natl Acad Sci U S A ; 113(48): 13851-13856, 2016 11 29.
Article in English | MEDLINE | ID: mdl-27849581

ABSTRACT

Recombinant C-terminally truncated prion protein PrP23-144 (which corresponds to the Y145Stop PrP variant associated with a Gerstmann-Sträussler-Scheinker-like prion disease) spontaneously forms amyloid fibrils with a parallel in-register ß-sheet architecture and ß-sheet core mapping to residues ∼112-139. Here we report that mice (both tga20 and wild type) inoculated with a murine (moPrP23-144) version of these fibrils develop clinical prion disease with a 100% attack rate. Remarkably, even though fibrils in the inoculum lack the entire C-terminal domain of PrP, brains of clinically sick mice accumulate longer proteinase K-resistant (PrPres) fragments of ∼17-32 kDa, similar to those observed in classical scrapie strains. Shorter, Gerstmann-Sträussler-Scheinker-like PrPres fragments are also present. The evidence that moPrP23-144 amyloid fibrils generated in the absence of any cofactors are bona fide prions provides a strong support for the protein-only hypothesis of prion diseases in its pure form, arguing against the notion that nonproteinaceous cofactors are obligatory structural components of all infectious prions. Furthermore, our finding that a relatively short ß-sheet core of PrP23-144 fibrils (residues ∼112-139) with a parallel in-register organization of ß-strands is capable of seeding the conversion of full-length prion protein to the infectious form has important implications for the ongoing debate regarding structural aspects of prion protein conversion and molecular architecture of mammalian prions.


Subject(s)
Amyloid/chemistry , Gerstmann-Straussler-Scheinker Disease/genetics , Prion Diseases/genetics , Prion Proteins/chemistry , Amyloid/adverse effects , Amyloid/genetics , Animals , Gerstmann-Straussler-Scheinker Disease/etiology , Gerstmann-Straussler-Scheinker Disease/pathology , Humans , Mice , Prion Diseases/etiology , Prion Diseases/pathology , Prion Proteins/adverse effects , Prion Proteins/genetics , Protein Conformation, beta-Strand/genetics , Scrapie/genetics , Scrapie/pathology , Spectroscopy, Fourier Transform Infrared
5.
Neurobiol Dis ; 91: 124-131, 2016 07.
Article in English | MEDLINE | ID: mdl-26949218

ABSTRACT

The pathogenic process in Alzheimer's disease (AD) appears to be closely linked to the neurotoxic action of amyloid-ß (Aß) oligomers. Recent studies have shown that these oligomers bind with high affinity to the membrane-anchored cellular prion protein (PrP(C)). It has also been proposed that this binding might mediate some of the toxic effects of the oligomers. Here, we show that the soluble (membrane anchor-free) recombinant human prion protein (rPrP) and its N-terminal fragment N1 block Aß oligomers-induced inhibition of long-term potentiation (LTP) in hippocampal slices, an important surrogate marker of cognitive deficit associated with AD. rPrP and N1 are also strikingly potent inhibitors of Aß cytotoxicity in primary hippocampal neurons. Furthermore, experiments using hippocampal slices and neurons from wild-type and PrP(C) null mice (as well as rat neurons in which PrP(C) expression was greatly reduced by gene silencing) indicate that, in contrast to the impairment of synaptic plasticity by Aß oligomers, the cytotoxic effects of these oligomers, and the inhibition of these effects by rPrP and N1, are independent of the presence of endogenous PrP(C). This suggests fundamentally different mechanisms by which soluble rPrP and its fragments inhibit these two toxic responses to Aß. Overall, these findings provide strong support to recent suggestions that PrP-based compounds may offer new avenues for pharmacological intervention in AD.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Long-Term Potentiation/drug effects , Neuronal Plasticity/physiology , Prion Proteins/pharmacology , Alzheimer Disease/metabolism , Alzheimer Disease/therapy , Animals , Cognition Disorders/drug therapy , Cognition Disorders/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Long-Term Potentiation/physiology , Neurons/drug effects , Neurons/metabolism , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Prion Proteins/metabolism , Rats, Sprague-Dawley
6.
Dermatol Surg ; 42(1): 63-7, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26671204

ABSTRACT

BACKGROUND: Facial aging is the result of intrinsic and extrinsic factors that lead to gradual reduction of dermal extracellular components and skin elasticity and wrinkle formation. A novel stent-shaped biodegradable and biocompatible scaffold device braided with absorbable polydioxanone (PDO) multifilaments was recently marketed for tissue suturing and augmentation. OBJECTIVE: To explore tissue regeneration profiles following implantation of the stent-shaped hollow scaffold in rats and mini-pigs. MATERIALS AND METHODS: The scaffold device was implanted under the panniculus carnosus of rat dorsal skin and in the subcutaneous layer of mini-pig dorsal skin. Tissue samples were harvested and histologically evaluated after 3 days and 1, 2, 4, and 12 weeks for rats and after 1, 2, 4, 8, and 12 weeks for mini-pigs. RESULTS: Type III collagen was slowly replaced by Type I collagen in the scaffold. Cells from the surrounding tissue infiltrated the hollow space of the scaffold, which induced de novo tissue regeneration in this space. CONCLUSION: The novel stent-shaped scaffold used here may be useful for stimulated tissue remodeling of aged skin, collagen synthesis, and partial restoration of dermal matrix components. The cosmetic purpose of this novel soft tissue augmentation device should be clinically investigated in long-term studies.


Subject(s)
Biocompatible Materials , Guided Tissue Regeneration/instrumentation , Polydioxanone , Tissue Scaffolds , Animals , Collagen/metabolism , Female , Guided Tissue Regeneration/methods , Rats, Sprague-Dawley , Skin Aging/physiology , Swine , Swine, Miniature
7.
ACS Chem Neurosci ; 6(12): 1972-80, 2015 Dec 16.
Article in English | MEDLINE | ID: mdl-26466138

ABSTRACT

A growing number of observations indicate that soluble amyloid-ß (Aß) oligomers play a major role in Alzheimer's disease. Recent studies strongly suggest that at least some of the neurotoxic effects of these oligomers are mediated by cellular, membrane-anchored prion protein and that Aß neurotoxicity can be inhibited by soluble recombinant prion protein (rPrP) and its fragments. However, the mechanism by which rPrP interacts with Aß oligomers and prevents their toxicity is largely unknown, and studies in this regard are hindered by the large structural heterogeneity of Aß oligomers. To overcome this difficulty, here we used photoinduced cross-linking of unmodified proteins (PICUP) to isolate well-defined oligomers of Aß42 and characterize these species with regard to their cytotoxicity and interaction with rPrP, as well the mechanism by which rPrP inhibits Aß42 cytotoxicity. Our data shows that the addition of rPrP to the assembling Aß42 results in a shift in oligomer size distribution, decreasing the population of toxic tetramers and higher order oligomers and increasing the population of nontoxic (and possibly neuroprotective) monomers. Isolated oligomeric species of Aß42 are cytotoxic to primary neurons and cause permeation of model lipid bilayers. These toxic effects, which are oligomer size-dependent, can be inhibited by the addition of rPrP, and our data suggest potential mechanisms of this inhibitory action. This insight should help in current efforts to develop PrP-based therapeutics for Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/metabolism , Prions/chemistry , Prions/metabolism , Amyloid beta-Peptides/chemistry , Animals , Embryo, Mammalian , Fluoresceins/metabolism , Hippocampus/cytology , Humans , L-Lactate Dehydrogenase/metabolism , Microscopy, Atomic Force , Molecular Weight , Neurons/drug effects , Neurons/metabolism , Protein Binding/drug effects , Rats , Rats, Sprague-Dawley , Surface Plasmon Resonance
8.
ACS Appl Mater Interfaces ; 5(7): 2400-9, 2013 Apr 10.
Article in English | MEDLINE | ID: mdl-23510254

ABSTRACT

Striving to replace the well known silicon nanocrystals embedded in oxides with solution-processable charge-trapping materials has been debated because of large scale and cost effective demands. Herein, a silicon quantum dot-polystyrene (SiQD-PS) nanocomposite (NC) was synthesized by post-functionalization of hydrogen-terminated silicon quantum dots (H-SiQDs) with styrene using a thermally induced surface-initiated polymerization approach. The NC contains two miscible components: PS and SiQD@PS which, respectively, are polystyrene and polystyrene chains-capped SiQDs. Spin-coated films of the nanocomposite on various substrate were thermally annealed at different temperatures and subsequently used to construct metal-insulator-semiconductor (MIS) devices and thin film field-effect transistors (TFTs) having a structure of p-Si++/SiO2/NC/pentacene/Au source-drain. Capacitance-voltage (C-V) curves obtained from the MIS devices exhibit a well-defined counterclockwise hysteresis with negative fat band shifts, which was stable over a wide range of curing temperatures (50-250 °C). The positive charge trapping capability of the NC originates from the spherical potential well structure of the SiQD@PS component while the strong chemical bonding between SiQDs and polystyrene chains accounts for the thermal stability of the charge trapping property. The transfer curve of the transistor was controllably shifted to the negative direction by varying applied gate voltage. Thereby, this newly synthesized and solution processable SiQD-PS nanocomposite is applicable as charge trapping materials for TFT based memory devices.

9.
J Vet Sci ; 14(1): 21-6, 2013.
Article in English | MEDLINE | ID: mdl-23388435

ABSTRACT

Chronic wasting disease (CWD) is classified as a transmissible spongiform encephalopathy or prion disease that affects cervids. CWD has been reported in 15 US states, two Canadian provinces, and in imported elk on several farms in Korea. This study was conducted to examine the molecular biological and pathogenic characteristics of a CWD-associated prion isolated in Korea. The epidemiological origin of this pathogen was also determined. Homozygous TgElk mice were infected with a CWD-affected elk brain pool prepared from the brain of an imported Canadian elk. We measured the incubation time of the pathogen, neuropathological changes by immunohistochemical staining, the pattern(s) of scrapie prion protein (PrPSc) deposition, and PrPSc protein profiles by Western blotting. We found that TgElk mice infected with brain homogenate from the elk suffering from CWD showed incubation times, vacuolar degeneration, and PrPSc accumulation similar to those previously reported in the literature. Our results suggest that homozygous TgElk mice efficiently transmit CWD with short incubation times and that this animal can serve a valuable research model and reliable in vivo diagnostic tool.


Subject(s)
Brain/pathology , Deer , Wasting Disease, Chronic/pathology , Animals , Female , Genotype , Mice , Mice, Transgenic , Prions , Republic of Korea/epidemiology , Wasting Disease, Chronic/epidemiology , Wasting Disease, Chronic/transmission
10.
Pharm Biol ; 50(9): 1157-67, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22881141

ABSTRACT

CONTEXT: Botulinum neurotoxins (BoNTs) are popularly used to treat various diseases and for cosmetic purposes. They act by blocking neurotransmission through specific cleavage of soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) proteins. Recently, several polyphenols were shown to interfere with SNARE complex formation by wedging into the hydrophobic core interface, thereby leading to reduced neuroexocytosis. OBJECTIVE: In order to find industrially-viable plant extract that functions like BoNT, 71 methanol extracts of flowers were screened and BoNT-like activity of selected extract was evaluated. MATERIALS AND METHODS: After evaluating the inhibitory effect of 71 flower methanol extracts on SNARE complex formation, seven candidates were selected and they were subjected to SNARE-driven membrane fusion assay. Neurotransmitter release from neuronal PC12 cells and SNARE complex formation inside the cell was also evaluated. Finally, the effect of one selected extract on muscle contraction and digit abduction score was determined. RESULTS: The extract of Potentilla chinensis Ser. (Rosaceae)(Chinese cinquefoil) flower inhibited neurotransmitter release from neuronal PC12 cells by approximately 90% at a concentration of 10 µg/mL. The extract inhibited neuroexocytosis by interfering with SNARE complex formation inside cells. It reduced muscle contraction of phrenic nerve-hemidiaphragm by approximately 70% in 60 min, which is comparable to the action of the Ca²âº-channel blocker verapamil and BoNT type A. DISCUSSION AND CONCLUSION: While BoNT blocks neuroexocytosis by cleaving SNARE proteins, the Potentilla chinensis extract exhibited the same activity by inhibiting SNARE complex formation. The extract paralyzed muscle as efficiently as BoNT, suggesting the potential versatility in cosmetics and therapeutics.


Subject(s)
Membrane Fusion/drug effects , Muscle Contraction/drug effects , Neuromuscular Agents/pharmacology , Neurons/drug effects , Plant Extracts/pharmacology , Potentilla/chemistry , SNARE Proteins/antagonists & inhibitors , Animals , Botulinum Toxins/adverse effects , Botulinum Toxins/pharmacology , Drug Discovery , Exocytosis/drug effects , Female , Flowers/chemistry , Lower Extremity , Mice , Mice, Inbred ICR , Muscle, Skeletal/drug effects , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Neuromuscular Agents/adverse effects , Neurons/metabolism , Norepinephrine/metabolism , PC12 Cells , Plant Extracts/adverse effects , Rats , SNARE Proteins/metabolism , Synaptic Transmission/drug effects
11.
J Biol Chem ; 287(40): 33104-8, 2012 Sep 28.
Article in English | MEDLINE | ID: mdl-22915585

ABSTRACT

The pathogenesis of Alzheimer disease appears to be strongly linked to the aggregation of amyloid-ß (Aß) peptide and, especially, formation of soluble Aß1-42 oligomers. It was recently demonstrated that the cellular prion protein, PrP(C), binds with high affinity to these oligomers, acting as a putative receptor that mediates at least some of their neurotoxic effects. Here we show that the soluble (i.e. glycophosphatidylinositol anchor-free) prion protein and its N-terminal fragment have a strong effect on the aggregation pathway of Aß1-42, inhibiting its assembly into amyloid fibrils. Furthermore, the prion protein prevents formation of spherical oligomers that normally occur during Aß fibrillogenesis, acting as a potent inhibitor of Aß1-42 toxicity as assessed in experiments with neuronal cell culture. These findings may provide a molecular level foundation to explain the reported protective action of the physiologically released N-terminal N1 fragment of PrP(C) against Aß neurotoxicity. They also suggest a novel approach to pharmacological intervention in Alzheimer disease.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid beta-Protein Precursor/chemistry , Alzheimer Disease/metabolism , Cell Line, Tumor , Humans , Microscopy, Atomic Force/methods , Neuroblastoma/metabolism , Peptide Fragments/chemistry , Peptides/chemistry , Prions/chemistry , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary
12.
Arch Virol ; 157(11): 2095-104, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22791111

ABSTRACT

The severe acute respiratory syndrome coronavirus (SARS-CoV) RNA genome is replicated by a virus-encoded RNA replicase, the key component of which is the nonstructural protein 12 (nsp12). In this report, we describe the biochemical properties of a full-length recombinant SARS-CoV nsp12 RNA-dependent RNA polymerase (RdRp) capable of copying viral RNA templates. The purified SARS-CoV nsp12 showed both primer-dependent and primer-independent RNA synthesis activities using homopolymeric RNA templates. The RdRp activity was strictly dependent on Mn(2+). The nsp12 preferentially copied homopolymeric pyrimidine RNA templates in the absence of an added oligonucleotide primer. It was also able to initiate de novo RNA synthesis from the 3'-ends of both the plus- and minus-strand genome of SARS-CoV, using the 3'-terminal 36- and 37-nt RNA, respectively. The in vitro RdRp assay system established with a full-length nsp12 will be useful for understanding the mechanisms of coronavirus replication and for the development of anti-SARS-CoV agents.


Subject(s)
RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/metabolism , Severe acute respiratory syndrome-related coronavirus/enzymology , 3' Untranslated Regions , Cations, Divalent/metabolism , Coenzymes/metabolism , DNA Primers/genetics , Genome, Viral , Manganese/metabolism , RNA-Dependent RNA Polymerase/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Severe acute respiratory syndrome-related coronavirus/genetics
13.
Biochem J ; 445(2): 183-92, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22551201

ABSTRACT

The citrullination of enolase by PAD (peptidylarginine deiminase) has emerged as an important post-translational modification in human disorders; however, the physiological function of citrullination remains unknown. In the present study, we report that citrullination diversely regulates the biological functions of ENO1 (α-enolase) and NSE (neuron-specific enolase). We developed three mouse IgG1 monoclonal antibodies with specificity to the following: (i) citrullination of Arg9 of ENO1 [ENO1Cit9; anti-CE1 (citrullinated enolase 1) antibody]; (ii) citrullination of Arg9 in ENO1 and NSE (ENO1Cit9/NSECit9; anti-CE1/2 antibody); and (iii) citrullination of Arg429 of NSE (NSECit429; anti-CE2 antibody). Regardless of the total protein expression level, the levels of ENO1Cit9 and NSECit429 were elevated, and their immunoreactivities were also increased in cortical neuronal cells or around blood vessels in the frontal cortex of patients with sporadic Creutzfeldt-Jakob disease and Alzheimer's disease compared with controls. In a time- and dose-dependent manner, PAD negatively regulated enolase activity via citrullination, and enolase in diseased patients was more inactive than in controls. Interestingly, the citrullination of enolase effectively promoted its proteolytic degradation by Ca2+-dependent calpain-1, and leupeptin (calpain inhibitor I) abrogated this degradation. Surprisingly, using an affinity assay, the citrullination of enolase enhanced its plasminogen-binding affinity, which was blocked by the lysine analogue ϵ-aminocaproic acid. These findings suggest that PAD-mediated citrullination regulates the diverse physiological activities of enolase and that CE may be a candidate diagnostic/prognostic factor for degenerative diseases.


Subject(s)
Alzheimer Disease/metabolism , Biomarkers, Tumor/metabolism , Carrier Proteins/metabolism , Citrulline/metabolism , Creutzfeldt-Jakob Syndrome/metabolism , DNA-Binding Proteins/metabolism , Hydrolases/metabolism , Phosphopyruvate Hydratase/metabolism , Tumor Suppressor Proteins/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Aminocaproic Acid/pharmacology , Animals , Antibodies, Monoclonal/immunology , Biomarkers, Tumor/immunology , Blotting, Western , Brain/metabolism , Carrier Proteins/immunology , Case-Control Studies , Creutzfeldt-Jakob Syndrome/pathology , DNA-Binding Proteins/immunology , Female , Frontal Lobe/metabolism , Humans , Immunoenzyme Techniques , Male , Mice , Mice, Inbred BALB C , Peptide Fragments/immunology , Phosphopyruvate Hydratase/immunology , Plasminogen/metabolism , Protein Processing, Post-Translational , Protein-Arginine Deiminases , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Tumor Suppressor Proteins/immunology
14.
FASEB J ; 25(12): 4174-83, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21852538

ABSTRACT

Presenilin 1 (PS1) is a component of the γ-secretase complex that cleaves a variety of type I membrane proteins, including the ß-amyloid precursor protein (ß-APP), Notch, and neuronal (N)- and epithelial (E)-cadherins. N-cadherin is an essential adhesion molecule that forms a complex with, and is cleaved by, PS1/γ-secretase and ß-catenin in the plasma membrane. The purpose of this study was to determine whether calsenilin, a presenilin-interacting protein, has a functional role in PS1/γ-secretase-mediated N-cadherin ε-cleavage using Western blot analysis, RT-PCR, immunoprecipitation, subcellular fractionation, biotinylation, and a luciferase reporter assay in SH-SY5Y neuroblastoma cells. Here, we demonstrate that the expression of calsenilin leads to a disruption of PS1/γ-secretase-mediated ε-cleavage of N-cadherin, which results in the significant accumulation of N-cadherin C-terminal fragment 1 (Ncad/CTF1), the reduction of cytoplasmic Ncad/CTF2 release, and a deceleration of PS1-CTF delivery to the cell surface. Interestingly, we also found that the expression of calsenilin is associated with the redistribution of ß-catenin from the cell surface to a cytoplasmic pool, as well as with the negative regulation of genes that are targets of T-cell factor/ß-catenin nuclear signaling. Taken together, our findings suggest that calsenilin is a novel negative regulator of N-cadherin processing that plays an important role in ß-catenin signaling.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Antigens, CD/metabolism , Cadherins/metabolism , Kv Channel-Interacting Proteins/metabolism , Presenilin-1/metabolism , Repressor Proteins/metabolism , beta Catenin/metabolism , Active Transport, Cell Nucleus , Antigens, CD/chemistry , Cadherins/chemistry , Cell Line, Tumor , Cell Membrane/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Kv Channel-Interacting Proteins/genetics , Models, Neurological , Neuroblastoma/genetics , Neuroblastoma/metabolism , Protein Structure, Tertiary , Proteolysis , Repressor Proteins/genetics , Signal Transduction , beta Catenin/genetics
15.
J Am Chem Soc ; 133(20): 7764-85, 2011 May 25.
Article in English | MEDLINE | ID: mdl-21542608

ABSTRACT

A new kind of organic-inorganic hybrid polymer, poly(tetraphenyl)silole siloxane, was invented and synthesized for realization of its unique charge trap properties. The organic portions consisting of (tetraphenyl)silole rings were responsible for negative charge trapping, while the Si-O-Si inorganic linkages provided the intrachain energy barrier for controlling electron transport. The polysilole siloxane dielectric thin films were fabricated by spin-coating and curing of the polymers, followed by characterization with spectroscopic ellipsometry (SE), near edge X-ray absorption fine structure spectroscopy (NEXAFS), and photoemission spectroscopy (PES). The abrupt increase in density and decrease in thickness of the thin film at a curing temperature of 100 °C was attributed to a thermodynamically preferred state in the nanoscopic arrangement of the polymer chains; this was due to cofacial π-π interactions in a skewed manner between peripheral phenyl groups of the (tetraphenyl)silole rings of the adjacent polymer chains. Using the NEXAFS spectrum to assess high electron affinity, the LUMO energy level of the dielectric thin film cured at 150 °C was positioned 1 eV above the Fermi energy level (E(F)). The electron trapping of the dielectric thin films was confirmed from the positive flat band shift (ΔV(FB)) in the capacitance-voltage (C-V) measurements performed within the metal-insulator-semiconductor (MIS) device structure, which strongly verified the polymer design concept. From the simple kinetics model of the electron transport, it was proposed that the flat band shift (ΔV(FB)) or trap density of the negative charges (|ρ|) was logarithmically proportional to the decay constant (ß) for the electron-tunneling process. When a phenyl group of a silole ring in a polymer chain was inserted into the two available phenyl groups of another silole ring in another polymer chain, the electron transfer between the groups was enhanced, decreasing the trap density of the negative charges (|ρ|). For the thermodynamically preferred state generating the high refractive index, the distance between the two phenyl groups of the adjacent polymer chains was estimated to be in the range of 0.27-0.36 nm.

16.
Antiviral Res ; 91(1): 1-10, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21549154

ABSTRACT

The programmed -1 ribosomal frameshifting (-1 PRF) utilized by eukaryotic RNA viruses plays a crucial role for the controlled, limited synthesis of viral RNA replicase polyproteins required for genome replication. The viral RNA replicase polyproteins of severe acute respiratory syndrome coronavirus (SARS-CoV) are encoded by the two overlapping open reading frames 1a and 1b, which are connected by a -1 PRF signal. We evaluated the antiviral effects of antisense peptide nucleic acids (PNAs) targeting a highly conserved RNA sequence on the - PRF signal. The ribosomal frameshifting was inhibited by the PNA, which bound sequence-specifically a pseudoknot structure in the -1 PRF signal, in cell lines as assessed using a dual luciferase-based reporter plasmid containing the -1 PRF signal. Treatment of cells, which were transfected with a SARS-CoV-replicon expressing firefly luciferase, with the PNA fused to a cell-penetrating peptide (CPP) resulted in suppression of the replication of the SARS-CoV replicon, with a 50% inhibitory concentration of 4.4µM. There was no induction of type I interferon responses by PNA treatment, suggesting that the effect of PNA is not due to innate immune responses. Our results demonstrate that -1 PRF, critical for SARS-CoV viral replication, can be inhibited by CPP-PNA, providing an effective antisense strategy for blocking -1 PRF signals.


Subject(s)
Frameshifting, Ribosomal , Oligoribonucleotides, Antisense/genetics , Oligoribonucleotides, Antisense/metabolism , Severe acute respiratory syndrome-related coronavirus/genetics , Virus Replication , Animals , Antiviral Agents/pharmacology , Cell Line , Cell-Penetrating Peptides/genetics , Cricetinae , Electrophoretic Mobility Shift Assay , Genes, Overlapping/genetics , Genes, Reporter , Genome, Viral/genetics , Humans , Interferon Type I/biosynthesis , Luciferases, Firefly , Open Reading Frames , RNA, Viral/chemistry , RNA, Viral/genetics , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Severe acute respiratory syndrome-related coronavirus/physiology
17.
Neurochem Res ; 36(5): 879-93, 2011 May.
Article in English | MEDLINE | ID: mdl-21305389

ABSTRACT

We conducted experiments in Drosophila to investigate the consequences of altered acetylcholinesterase (AChE) activity in the nervous system. In ace hypomorphic mutant larvae, the amount of ace mRNA and the activity of AChE both in vivo and in vitro were significantly reduced compared with those of controls. Reduced Ace in Drosophila larvae resulted in significant down-regulation of branch length and the number of boutons in Type 1 glutamatergic neuromuscular junctions (NMJs). These defects in ace hypomorphic mutant larvae were suppressed when Musca domestica AChE was transgenically expressed. Because AChE inhibitors are utilized for medications for Alzheimer's disease, we investigated whether pharmacological inhibition of AChE activity induced any synaptic defects. We found that controls exposed to a sublethal dose of DDVP phenocopied the synaptic structural defects of the ace hypomorphic mutant. These results suggest that down-regulation of AChE activity, regardless of whether it is due to genetic or pharmacological manipulations, results in altered synaptic architecture. Our study suggests that exposure to AChE inhibitors for 6-12 months may induce altered synaptic architectures in human brains with Alzheimer's diseases, similar to those reported here. These changes may underlie or contribute to the loss of efficacy of AChE inhibitors after prolonged treatment.


Subject(s)
Acetylcholinesterase/drug effects , Cholinesterase Inhibitors/pharmacology , Drosophila/genetics , Neuronal Plasticity/drug effects , Pharmacogenetics , Synapses/drug effects , Acetylcholinesterase/chemistry , Acetylcholinesterase/genetics , Acetylcholinesterase/metabolism , Amino Acid Sequence , Animals , Animals, Genetically Modified , Base Sequence , DNA Primers , Drosophila/enzymology , Immunohistochemistry , Models, Molecular , Molecular Sequence Data , Phylogeny , Protein Conformation , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid
18.
J Neuropathol Exp Neurol ; 70(2): 116-24, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21343880

ABSTRACT

Peptidylarginine deiminase (PAD) and citrullinated proteins have emerged as key molecules in various human diseases, but detailed subcellular localizations of PAD2 and citrullinated proteins are poorly mapped in brain under normal and pathologic conditions. We performed subcellular fractionation and electron microscopic analysis using brains of normal and scrapie-infected mice. Peptidylarginine deiminase 2 was abundantly present in cytosol and weakly in microsomal and mitochondrial fractions and expression in these fractions was higher in brains of scrapie-infected mice. Despite relatively low PAD2 expression, in microsomal and mitochondrial fractions, citrullinated proteins were present at high levels in these fractions in scrapie-infected brains. Surprisingly, increased PAD2 expression and accumulated citrullinated proteins were also found in nuclear fractions in scrapie-infected brains. By electron microscopy, PAD2 and citrullinated proteins in scrapie-infected brains were widely distributed in most cellular compartments including mitochondria, endoplasmic reticulum, glial filaments, nuclei, and Golgi apparatus in astrocytes and hippocampal neurons. Taken together, we report for the first time the nuclear localization of PAD2 and the detailed subcellular localization of PAD2 and of citrullinated proteins in scrapie-infected brains. Our findings suggest that different subcellular compartmentalization of PAD2 and citrullinated proteins may have different physiological roles in normal and neurodegenerative conditions.


Subject(s)
Brain Chemistry/physiology , Citrulline/metabolism , Hydrolases/metabolism , Nerve Tissue Proteins/metabolism , Scrapie/metabolism , Subcellular Fractions/metabolism , Animals , Astrocytes/enzymology , Astrocytes/metabolism , Blotting, Western , Cell Line, Tumor , Cell Membrane/enzymology , Cell Membrane/metabolism , Cells, Cultured , Hippocampus/cytology , Hippocampus/enzymology , Hippocampus/metabolism , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Microscopy, Immunoelectron , Neurons/enzymology , Neurons/metabolism , Nuclear Proteins/metabolism , Protein-Arginine Deiminase Type 2 , Protein-Arginine Deiminases , Scrapie/pathology , Subcellular Fractions/enzymology , Transfection
19.
Biochem Biophys Res Commun ; 405(2): 180-5, 2011 Feb 11.
Article in English | MEDLINE | ID: mdl-21216226

ABSTRACT

Calsenilin, a neuronal calcium binding protein that has been shown to have multiple functions in the cell, interacts with presenilin 1 (PS1) and presenilin 2 (PS2), represses gene transcription and binds to A-type voltage-gated potassium channels. In addition, increased levels of calsenilin are observed in the brains of Alzheimer's disease and epilepsy patients. The present study was designed to investigate the molecular mechanism of calsenilin degradation pathways in cultured cells. Here, we demonstrate that inhibition of the ubiquitin-proteasomal pathway (UPP) but not lysosomal pathway markedly increased the expression levels of calsenilin. Immunofluorescence analysis revealed that following proteasomal inhibition calsenilin accumulated in the endoplasmic reticulum (ER) and Golgi, while lysosomal inhibition had no effect on calsenilin localization. In addition, we found the change of subcellular localization of PS1 from diffuse pattern to punctuate staining pattern in the ER and perinuclear region in the presence of calsenilin. These findings suggest that calsenilin degradation is primarily mediated by the UPP and that impairment in the UPP may contribute to the involvement of calsenilin in disease-associated neurodegeneration.


Subject(s)
Kv Channel-Interacting Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Repressor Proteins/metabolism , Ubiquitin/metabolism , Ubiquitination , Cell Line, Tumor , Cysteine Proteinase Inhibitors/pharmacology , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Humans , Leupeptins/pharmacology , Lysosomes/drug effects , Lysosomes/metabolism , Neurodegenerative Diseases/metabolism , Proteasome Inhibitors
20.
Hum Mol Genet ; 19(22): 4474-89, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20829230

ABSTRACT

We have established a Drosophila model of Gerstmann-Sträussler-Scheinker (GSS) syndrome by expressing mouse prion protein (PrP) having leucine substitution at residue 101 (MoPrP(P101L)). Flies expressing MoPrP(P101L), but not wild-type MoPrP (MoPrP(3F4)), showed severe defects in climbing ability and early death. Expressed MoPrP(P101L) in Drosophila was differentially glycosylated, localized at the synaptic terminals and mainly present as deposits in adult brains. We found that behavioral defects and early death of MoPrP(P101L) flies were not due to Caspase 3-dependent programmed cell death signaling. In addition, we found that Type 1 glutamatergic synaptic boutons in larval neuromuscular junctions of MoPrP(P101L) flies showed significantly increased numbers of satellite synaptic boutons. Furthermore, the amount of Bruchpilot and Discs large in MoPrP(P101L) flies was significantly reduced. Brains from scrapie-infected mice showed significantly decreased ELKS, an active zone matrix marker compared with those of age-matched control mice. Thus, altered active zone structures at the molecular level may be involved in the pathogenesis of GSS syndrome in Drosophila and scrapie-infected mice.


Subject(s)
Disease Models, Animal , Drosophila , Gerstmann-Straussler-Scheinker Disease/genetics , Prions/genetics , Animals , Brain/metabolism , Brain/pathology , Female , Gerstmann-Straussler-Scheinker Disease/metabolism , Gerstmann-Straussler-Scheinker Disease/pathology , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Prion Proteins , Prions/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...