Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Anesth Analg ; 120(3): 671-677, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25695583

ABSTRACT

BACKGROUND: The regulator of G-protein signaling protein type 4 (RGS4) accelerates the guanosine triphosphatase activity of G(αi) and G(αo), resulting in the inactivation of G-protein-coupled receptor signaling. An opioid receptor (OR), a G(αi)-coupled receptor, plays an important role in pain modulation in the central nervous system. In this study, we examined whether (1) spinal RGS4 affected nociceptive responses in the formalin pain test, (2) this RGS4-mediated effect was involved in OR activation, and (3) the µ-OR agonist-induced antinociceptive effect was modified by RGS4 modulation. METHODS: Formalin (1%, 20 µL) was injected subcutaneously into the right hindpaws of male 129S4/SvJae×C57BL/6J (RGS4(+/+) or RGS4(-/-)) mice, and the licking responses were counted for 40 minutes. The time periods (seconds) spent licking the injected paw during 0 to 10 minutes (early phase) and 10 to 40 minutes (late phase) were measured as indicators of acute nociception and inflammatory pain response, respectively. An RGS4 inhibitor, CCG50014, and/or a µ-OR agonist, [D-Ala², N-MePhe4, Gly-ol]-enkephalin (DAMGO), were intrathecally injected 5 minutes before the formalin injection. A nonselective OR antagonist, naloxone, was intraperitoneally injected 30 minutes before the CCG50014 injection. RESULTS: Mice that received the formalin injection exhibited typical biphasic nociceptive behaviors. The nociceptive responses in RGS4-knockout mice were significantly decreased during the late phase but not during the early phase. Similarly, intrathecally administered CCG50014 (10, 30, or 100 nmol) attenuated the nociceptive responses during the late phase in a dose-dependent manner. The antinociceptive effect of the RGS4 inhibitor was totally blocked by naloxone (5 mg/kg). In contrast, intrathecal injection of DAMGO achieved a dose-dependent reduction of the nociceptive responses at the early and late phases. This analgesic effect of DAMGO was significantly enhanced by the genetic depletion of RGS4 or by coadministration of CCG50014 (10 nmol). CONCLUSIONS: These findings demonstrated that spinal RGS4 inhibited the endogenous or exogenous OR-mediated antinociceptive effect in the formalin pain test. Thus, the inhibition of RGS4 activity can enhance OR agonist-induced analgesia. The enhancement of OR agonist-induced analgesia by coadministration of the RGS4 inhibitor suggests a new therapeutic strategy for the management of inflammatory pain.


Subject(s)
Analgesics, Opioid/pharmacology , Analgesics/administration & dosage , Behavior, Animal/drug effects , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Formaldehyde , Nociception/drug effects , Nociceptive Pain/prevention & control , RGS Proteins/antagonists & inhibitors , Spinal Cord/drug effects , Thiazolidinediones/administration & dosage , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Synergism , Injections, Spinal , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Narcotic Antagonists/pharmacology , Nociceptive Pain/genetics , Nociceptive Pain/metabolism , Nociceptive Pain/physiopathology , Nociceptive Pain/psychology , Pain Measurement , RGS Proteins/deficiency , RGS Proteins/genetics , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism , Spinal Cord/metabolism , Spinal Cord/physiopathology , Time Factors
2.
Nat Cell Biol ; 9(11): 1303-10, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17952061

ABSTRACT

Mitochondrial proteins function as essential regulators in apoptosis. Here, we show that mitochondrial adenylate kinase 2 (AK2) mediates mitochondrial apoptosis through the formation of an AK2-FADD-caspase-10 (AFAC10) complex. Downregulation of AK2 attenuates etoposide- or staurosporine-induced apoptosis in human cells, but not that induced by tumour-necrosis-factor-related apoptosis-inducing ligand (TRAIL) or Fas ligand (FasL). During intrinsic apoptosis, AK2 translocates to the cytoplasm, whereas this event is diminished in Apaf-1 knockdown cells and prevented by Bcl-2 or Bcl-X(L). Addition of purified AK2 protein to cell extracts first induces activation of caspase-10 via FADD and subsequently caspase-3 activation, but does not affect caspase-8. AFAC10 complexes are detected in cells undergoing intrinsic cell death and AK2 promotes the association of caspase-10 with FADD. In contrast, AFAC10 complexes are not detected in several etoposide-resistant human tumour cell lines. Taken together, these results suggest that, acting in concert with FADD and caspase-10, AK2 mediates a novel intrinsic apoptotic pathway that may be involved in tumorigenesis.


Subject(s)
Adenylate Kinase/physiology , Apoptosis/physiology , Caspase 10/metabolism , Fas-Associated Death Domain Protein/metabolism , Isoenzymes/physiology , Adenylate Kinase/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cells, Cultured , HeLa Cells , Humans , Isoenzymes/pharmacology , Multienzyme Complexes/metabolism , Subcellular Fractions/metabolism
3.
Exp Neurobiol ; 31(2): 116-130, 2022 Apr 30.
Article in English | MEDLINE | ID: mdl-35674000

ABSTRACT

Absence seizures are caused by abnormal synchronized oscillations in the thalamocortical (TC) circuit, which result in widespread spike-and-wave discharges (SWDs) on electroencephalography (EEG) as well as impairment of consciousness. Thalamic reticular nucleus (TRN) and TC neurons are known to interact dynamically to generate TC circuitry oscillations during SWDs. Clinical studies have suggested the association of Plcß1 with early-onset epilepsy, including absence seizures. However, the brain regions and circuit mechanisms related to the generation of absence seizures with Plcß1 deficiency are unknown. In this study, we found that loss of Plcß1 in mice caused spontaneous complex-type seizures, including convulsive and absence seizures. Importantly, TRN-specific deletion of Plcß1 led to the development of only spontaneous SWDs, and no other types of seizures were observed. Ex vivo slice patch recording demonstrated that the number of spikes, an intrinsic TRN neuronal property, was significantly reduced in both tonic and burst firing modes in the absence of Plcß1 . We conclude that the loss of Plcß1 in the TRN leads to decreased excitability and impairs normal inhibitory neuronal function, thereby disrupting feedforward inhibition of the TC circuitry, which is sufficient to cause hypersynchrony of the TC system and eventually leads to spontaneous absence seizures. Our study not only provides a novel mechanism for the induction of SWDs in Plcß1 -deficient patients but also offers guidance for the development of diagnostic and therapeutic tools for absence epilepsy.

4.
Biochem Biophys Rep ; 25: 100871, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33367116

ABSTRACT

Human prostaglandin E2 receptor 4 (EP4) is one of the four subtypes of prostaglandin E2 (PGE2) receptors and belongs to the rhodopsin-type G protein-coupled receptor (GPCR) family. Particularly, EP4 is expressed in various cancer cells and is involved in cancer-cell proliferation by a G protein signaling cascade. To prepare an active form of EP4 for biochemical characterization and pharmaceutical application, this study designed a recombinant protein comprising human EP4 fused to the P9 protein (a major envelope protein of phi6 phage) and overexpressed the P9-EP4 fusion protein in the membrane fraction of E. coli. The solubilized P9-EP4 with sarkosyl (a strong anionic detergent) was purified by affinity chromatography. The purified protein was stabilized with amphiphilic polymers derived from poly-γ-glutamate. The polymer-stabilized P9-EP4 showed specific interaction with the alpha subunits of Gs or Gi proteins, and a high content of α-helical structure by a circular dichroism spectroscopy. Furthermore, the polymer-stabilized P9-EP4 showed strong heat resistance compared with P9-EP4 in detergents. The functional preparation of EP4 and its stabilization with amphiphilic polymers could facilitate both the biochemical characterization and pharmacological applications targeting EP4.

5.
Neuroscience ; 455: 177-194, 2021 02 10.
Article in English | MEDLINE | ID: mdl-33359660

ABSTRACT

The analgesic effect of alpha-2 adrenergic receptor (α2AR) agonists, which relieve chronic neuropathic pain, is highly variable among individuals. Here, we used a mouse model of spared nerve injury (SNI) to show that treatment time after the establishment of neuropathic pain was important for the variability in the analgesic efficacy of α2AR agonists, which was related to the activity of regulator of G-protein signaling protein 4 (RGS4). Intrathecal treatment with α2AR agonists, clonidine (0.1-1 nmol) or dexmedetomidine (0.3-1 nmol), relieved mechanical allodynia and thermal hyperalgesia on postoperative day (POD) 14, but their efficacy was weaker on POD28 and absent on POD56. The RGS4 level of plasma membrane was increased on POD56 compared to that on POD14. Moreover, in RGS4-deficient or RGS4 inhibitor (CCG50014)-treated mice, the analgesic effect of the α2AR agonists was conserved even on POD56. The increased plasma membrane RGS4 expression and the reduced level of active Gαi after clonidine injection on POD56 were completely restored by CCG50014. Higher doses of clonidine (10 nmol) and dexmedetomidine (3 nmol) relieved neuropathic pain on POD56 but were accompanied with serious side effects. Whereas, the coadministration of CCG50014 with clonidine (1 nmol) or dexmedetomidine (1 nmol) did not cause side effects. These findings demonstrated that SNI-induced increase in plasma membrane RGS4 expression was associated with low efficacy of α2AR agonists in a model of persistent, chronic neuropathic pain. Furthermore, α2AR agonist administration together with RGS4-targeted intervention represents a novel strategy for the treatment of neuropathic pain to overcome dose-limiting side effects.


Subject(s)
Adrenergic alpha-2 Receptor Agonists , Analgesics , Hyperalgesia , Neuralgia , Receptors, Adrenergic, alpha-2 , Adrenergic Agonists , Adrenergic alpha-2 Receptor Agonists/pharmacology , Adrenergic alpha-Agonists , Analgesics/pharmacology , Animals , Clonidine/pharmacology , Hyperalgesia/drug therapy , Mice , Neuralgia/drug therapy
6.
Prog Neurobiol ; 204: 102110, 2021 09.
Article in English | MEDLINE | ID: mdl-34166773

ABSTRACT

Mitochondrial dysfunction is associated with neuronal damage in Huntington's disease (HD), but the precise mechanism of mitochondria-dependent pathogenesis is not understood yet. Herein, we found that colocalization of XIAP and p53 was prominent in the cytosolic compartments of normal subjects but reduced in HD patients and HD transgenic animal models. Overexpression of mutant Huntingtin (mHTT) reduced XIAP levels and elevated mitochondrial localization of p53 in striatal cells in vitro and in vivo. Interestingly, XIAP interacted directly with the C-terminal domain of p53 and decreased its stability via autophagy. Overexpression of XIAP prevented mitochondrially targeted-p53 (Mito-p53)-induced mitochondrial oxidative stress and striatal cell death, whereas, knockdown of XIAP exacerbated Mito-p53-induced neuronal damage in vitro. In vivo transduction of AAV-shRNA XIAP in the dorsal striatum induced rapid onset of disease and reduced the lifespan of HD transgenic (N171-82Q) mice compared to WT littermate mice. XIAP dysfunction led to ultrastructural changes of the mitochondrial cristae and nucleus morphology in striatal cells. Knockdown of XIAP exacerbated neuropathology and motor dysfunctions in N171-82Q mice. In contrast, XIAP overexpression improved neuropathology and motor behaviors in both AAV-mHTT-transduced mice and N171-82Q mice. Our data provides a molecular and pathological mechanism that deregulation of XIAP triggers mitochondria dysfunction and other neuropathological processes via the neurotoxic effect of p53 in HD. Together, the XIAP-p53 pathway is a novel pathological marker and can be a therapeutic target for improving the symptoms in HD.


Subject(s)
Huntington Disease , Animals , Corpus Striatum , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Tumor Suppressor Protein p53/genetics , X-Linked Inhibitor of Apoptosis Protein/genetics
7.
Biochemistry ; 49(7): 1435-47, 2010 Feb 23.
Article in English | MEDLINE | ID: mdl-20070122

ABSTRACT

The death effector domain (DED) of the mammalian apoptosis mediator, Fas-associated death domain protein (FADD), induces Escherichia coli cell death under aerobic culture conditions, yet the mechanisms by which FADD-DED induces cell death are not fully understood. Oxidative stress has been implicated as one of the mechanisms. Using a proteomic approach and validation by coexpression analysis, we illustrate that overexpression of FADD-DED in E. coli invokes protein expression changes that facilitate conversion of pro-oxidant NADH into antioxidant NADPH. Typically, isocitrate dehydrogenase, phosphoenolpyruvate carboxykinase, and pyruvate kinase are downregulated and malate dehydrogenase is upregulated. We reasoned that such a change in E. coli cells is an active response to reduce the size of the NADH pool, thereby decreasing the level of ROS generation. From the coexpression studies, we observed that DNA binding protein Hns, which induces growth arrest when overexpressed heterologously, alleviated the cell killing effect of FADD-DED. FADD-DED was expressed as a noncovalently linked multimeric protein in the membrane of E. coli. Exogenous treatment of E. coli cells with FADD-DED in the presence of a membrane component induced cell death, which was accompanied by a shift of the redox balance and a decrease in the cellular ATP level. Cell death was blocked by prior expression of thioredoxin. Localization of FADD-DED to the membrane may shift the cells into a state that stimulates and fuels ROS generation. The cell death mechanism mediated by ROS may mimic antibiotic-mediated bacterial cell death or Bax-mediated apoptosis in mammalian cells. Our results provide a common mechanistic feature of ROS-involved cell death throughout prokaryotes and eukaryotes.


Subject(s)
Apoptosis , Cell Membrane/metabolism , Escherichia coli/cytology , Escherichia coli/metabolism , Fas-Associated Death Domain Protein/metabolism , Reactive Oxygen Species/metabolism , fas Receptor/metabolism , Apoptosis/genetics , Cell Membrane/genetics , Down-Regulation/genetics , Energy Metabolism/genetics , Escherichia coli/genetics , Fas-Associated Death Domain Protein/biosynthesis , Fas-Associated Death Domain Protein/genetics , Fas-Associated Death Domain Protein/physiology , Humans , Protein Interaction Domains and Motifs/genetics , Protein Structure, Tertiary/genetics , Reactive Oxygen Species/pharmacology , Up-Regulation/genetics , fas Receptor/biosynthesis , fas Receptor/genetics , fas Receptor/physiology
8.
Biochem Biophys Res Commun ; 394(3): 515-21, 2010 Apr 09.
Article in English | MEDLINE | ID: mdl-20206602

ABSTRACT

This study reports the crystal structures of Bcl-xl wild type and three Bcl-xl mutants (Y101A, F105A, and R139A) with amino acid substitutions in the hydrophobic groove of the Bcl-xl BH3 domain. An additional 12 ordered residues were observed in a highly flexible loop between the alpha1 and alpha2 helices, and were recognized as an important deamidation site for the regulation of apoptosis. The autophagy-effector protein, Beclin 1, contains a novel BH3 domain (residues 101-125), which binds to the surface cleft of Bcl-xl, as confirmed by nuclear magnetic resonance (NMR) spectroscopy and analytical gel-filtration results. Gossypol, a potent inhibitor of Bcl-xl, had a K(d) value of 0.9 microM. In addition, the structural and biochemical analysis of five Bcl-xl substitution mutants will provide structural insights into the design and development of anti-cancer drugs.


Subject(s)
Apoptosis Regulatory Proteins/chemistry , Gossypol/chemistry , bcl-X Protein/antagonists & inhibitors , bcl-X Protein/chemistry , Amino Acid Substitution , Animals , Antineoplastic Agents/chemistry , Beclin-1 , Chromatography, Gel , Crystallography, X-Ray , Drug Design , Mice , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , bcl-X Protein/genetics
9.
PLoS Pathog ; 4(2): e25, 2008 Feb 08.
Article in English | MEDLINE | ID: mdl-18248095

ABSTRACT

All gammaherpesviruses express homologues of antiapoptotic B-cell lymphoma-2 (BCL-2) to counter the clearance of infected cells by host antiviral defense machineries. To gain insights into the action mechanisms of these viral BCL-2 proteins, we carried out structural and biochemical analyses on the interactions of M11, a viral BCL-2 of murine gamma-herpesvirus 68, with a fragment of proautophagic Beclin1 and BCL-2 homology 3 (BH3) domain-containing peptides derived from an array of proapoptotic BCL-2 family proteins. Mainly through hydrophobic interactions, M11 bound the BH3-like domain of Beclin1 with a dissociation constant of 40 nanomole, a markedly tighter affinity compared to the 1.7 micromolar binding affinity between cellular BCL-2 and Beclin1. Consistently, M11 inhibited autophagy more efficiently than BCL-2 in NIH3T3 cells. M11 also interacted tightly with a BH3 domain peptide of BAK and those of the upstream BH3-only proteins BIM, BID, BMF, PUMA, and Noxa, but weakly with that of BAX. These results collectively suggest that M11 potently inhibits Beclin1 in addition to broadly neutralizing the proapoptotic BCL-2 family in a similar but distinctive way from cellular BCL-2, and that the Beclin1-mediated autophagy may be a main target of the virus.


Subject(s)
Apoptosis/physiology , Autophagy/physiology , Proteins/metabolism , Rhadinovirus/physiology , Viral Proteins/metabolism , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins , Beclin-1 , Gene Expression Regulation, Viral , Genes, Tumor Suppressor , Host-Pathogen Interactions , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Mice , Molecular Sequence Data , Proteins/chemistry , Proteins/genetics , Rhadinovirus/pathogenicity , Trans-Activators/genetics , Trans-Activators/metabolism , Viral Proteins/chemistry , Viral Proteins/genetics
10.
Cell Mol Neurobiol ; 30(5): 807-15, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20198420

ABSTRACT

We have screened new drugs with a view to developing effective drugs against glutamate-induced excitotoxicity. In the present work, we show effects of a new drug, 2-cyclopropylimino-3-methyl-1,3-thiazoline hydrochloride against glutamate-induced excitotoxicity in primary rat glial cultures. Pretreatment of glial cells with 2-cyclopropylimino-3-methyl-1,3-thiazoline hydrochloride for 2 h significantly protected glial cells against glutamate-induced excitotoxicity in a time- and dose-dependent manner with an optimum concentration of 100 microM. The drug significantly reduced production of proinflammatory cytokines, tumor necrosis factor-alpha, and interlukin-1beta in glutamate-induced excitotoxicity. The drug also prevented glutamate-induced intracellular Ca2+ influx and reduced the subsequent overproduction of nitric oxide and reactive oxygen species. Furthermore, the drug preserved the mitochondrial potential and inhibited the overproduction of cytochrome c. In addition, the drug effectively attenuated the protein level changes of beta-catenin and glycogen synthase kinase-3beta. These results suggest that 2-cyclopropylimino-3-methyl-1,3-thiazoline hydrochloride effectively protected primary cultures of rat glial cells against glutamate-induced excitotoxicity.


Subject(s)
Glutamic Acid/toxicity , Neuroglia/drug effects , Neuroglia/metabolism , Thiazoles/pharmacology , Animals , Calcium/metabolism , Cells, Cultured , Cytochromes c/metabolism , Cytokines/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Inflammation Mediators/metabolism , Membrane Potential, Mitochondrial/drug effects , Neuroglia/enzymology , Nitrites/metabolism , Protective Agents/chemistry , Protective Agents/pharmacology , Protein Processing, Post-Translational/drug effects , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Thiazoles/chemistry , beta Catenin/metabolism
11.
PLoS One ; 15(10): e0240608, 2020.
Article in English | MEDLINE | ID: mdl-33048999

ABSTRACT

Fibrillar aggregates of amyloid-ß (Aß) are the main component of plaques lining the cerebrovasculature in cerebral amyloid angiopathy. As the predominant Aß isoform in vascular deposits, Aß40 is a valuable target in cerebral amyloid angiopathy research. However, the slow process of Aß40 aggregation in vitro is a bottleneck in the search for Aß-targeting molecules. In this study, we sought a method to accelerate the aggregation of Aß40 in vitro, to improve experimental screening procedures. We evaluated the aggregating ability of bicine, a biological buffer, using various in vitro methods. Our data suggest that bicine promotes the aggregation of Aß40 with high speed and reproducibility, yielding a mixture of aggregates with significant ß-sheet-rich fibril formation and toxicity.


Subject(s)
Amyloid beta-Peptides/metabolism , Cerebral Amyloid Angiopathy/pathology , Glycine/analogs & derivatives , Peptide Fragments/metabolism , Protein Aggregates/drug effects , Amyloid beta-Peptides/toxicity , Animals , Cell Line , Cell Survival , Glycine/pharmacology , Humans , Mice , Neurons , Peptide Fragments/toxicity , Protein Conformation, beta-Strand/drug effects
12.
Biochem Biophys Res Commun ; 386(1): 65-70, 2009 Aug 14.
Article in English | MEDLINE | ID: mdl-19497306

ABSTRACT

Regulator of G protein signaling 11 (RGS11) is the least characterized member of the R7 family of Ggamma-like GGL domain-containing RGS proteins. All R7-RGS proteins of a variety of cell types are found in Gbeta5-containing complexes that exhibit a number of unique functional properties. However, presence of Gbeta5 reduced the affinity of R7-RGS7 for Galpha subunits, also only RGS7 bound to Muscarinic M3-Receptor, but the Gbeta5-RGS7 dimer did not, making it difficult to study differential interaction of R7-RGS proteins. Here, we report the successful purification of functionally intact, Gbeta5-free recombinant RGS11 (rRGS11), obtained by expressing N- and C-terminally truncated form of RGS11 in Escherichia coli BL 21 (DE3), that differentially interact with R7BP and Galpha(oa). rRGS11 was capable of interacting with Galpha(oa) and R7BP (RGS7 family binding protein) with equilibrium dissociation constants (K(D)) of 904 (+/- 208) nM, and 308 (+/- 97) nM, respectively. It also induced several-fold increase in the GTPase activity of Galpha(oa). The binding of rRGS11 was differential with a binding preference for R7BP over Galpha(oa) implying extended roles of R7BP. In addition, we identified a novel interaction between Galpha(oa) and R7BP with a K(D) of 592 (+/- 150) nM. The production of stable and functional rRGS11 would provide chances to discover more functions of RGS11 yet to be identified.


Subject(s)
Carrier Proteins/metabolism , RGS Proteins/metabolism , Cloning, Molecular , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein beta Subunits/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Protein Folding , RGS Proteins/chemistry , RGS Proteins/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
13.
Biochem Biophys Res Commun ; 380(4): 797-801, 2009 Mar 20.
Article in English | MEDLINE | ID: mdl-19338755

ABSTRACT

MenD (2-succinyl-5-enolpyruvyl-6-hydroxy-3-cyclohexadiene-1-carboxylate) synthase belongs to the superfamily of thiamin diphosphate-dependent decarboxylases, which converts isochorismate and 2-oxoglutarate to SHCHC, pyruvate, and carbon dioxide. Here, we report the first crystal structure of apo-MenD from Escherichia coli determined in tetragonal crystal form. The subunit displays the typical three-domain structure observed for ThDP-dependent enzymes. Analytical gel filtration shows that EcMenD behaves as a dimer as well as a tetramer. Circular dichroism and isothermal calorimetry results confirm EcMenD dependency on ThDP, which concomitantly helps to stabilize with better configuration.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli/enzymology , Nucleotidyltransferases/chemistry , Pyruvate Oxidase/chemistry , Dimerization , Enzyme Stability , Protein Conformation , Protein Subunits/genetics , X-Ray Diffraction
14.
Peptides ; 29(7): 1102-8, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18406495

ABSTRACT

The structure-activity relations and mechanism of action of parasin I, a 19-amino acid histone H2A-derived antimicrobial peptide, were investigated. Parasin I formed an amphipathic alpha-helical structure (residues 9-17) flanked by two random coil regions (residues 1-8 and 18-19) in helix-promoting environments. Deletion of the lysine residue at the N-terminal [Pa(2-19)] resulted in loss of antimicrobial activity, but did not affect the alpha-helical content of the peptide. The antimicrobial activity was recovered when the lysine residue was substituted with another basic residue, arginine ([R(1)]Pa), but not with polar, neutral, or acidic residues. Progressive deletions from the C-terminal [Pa(1-17), Pa(1-15)] slightly increased the antimicrobial activity (1-4 microg/ml) without affecting the alpha-helical content of the peptide. However, further deletion [Pa(1-14)] resulted in nearly complete loss of antimicrobial activity and alpha-helical structure. Confocal microscopic analysis and membrane permeabilization assays showed that parasin I and its analogs with comparable antimicrobial activities localized to the cell membrane and subsequently permeabilized the outer and cytoplasmic membranes. Pa(1-14) also localized to the cell membrane, but lost membrane-permeabilizing activity, whereas Pa(2-19) showed poor membrane-binding and -permeabilizing activities. The results indicate that the basic residue at the N-terminal is essential for the membrane-binding activity of parasin I, and among the membrane-binding parasin I analogs, the alpha-helical structure is necessary for the membrane-permeabilizing activity.


Subject(s)
Anti-Infective Agents/chemistry , Antimicrobial Cationic Peptides/chemistry , Histones/chemistry , Peptide Fragments/chemistry , Amino Acid Sequence , Amino Acid Substitution , Anti-Infective Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Cell Membrane Permeability , Circular Dichroism , Fungi/drug effects , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Histones/pharmacology , Histones/physiology , Hydrophobic and Hydrophilic Interactions , Microbial Sensitivity Tests , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Peptide Fragments/chemical synthesis , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Peptide Fragments/physiology , Protein Structure, Secondary , Proteins/chemistry , Proteins/pharmacology , Proteins/physiology , Sequence Deletion , Structure-Activity Relationship
15.
Mol Cells ; 26(2): 165-70, 2008 Aug 31.
Article in English | MEDLINE | ID: mdl-18596409

ABSTRACT

Procaspase-8 is activated by forming a death-inducing signaling complex (DISC) with the Fas-associated death domain (FADD) and the Fas receptor, but the mechanism of its activation is not well understood. Procaspase-8 devoid of the death effector domain at its N-terminus (delta nprocaspase-8) was reported to be activated by kosmotropic salts, but it has not been induced to form a DISC in vitro because it cannot interact with FADD. Here, we report the production of full-length procaspase-8 and show that it is activated by adding the Fas death domain (Fas-DD) and the FADD forming the cytoplasmic part of the DISC (cDISC). Furthermore, mutations known to affect DISC formation in vivo were shown to have the same effect on procaspase-8 activation in vitro. An antibody that induces Fas-DD association enhanced procaspase-8 activation, suggesting that the Fas ligand is not required for low-level activation of procaspase-8, but that Fas receptor clustering is needed for high-level activation of procaspase-8 leading to cell death. In vitro activation of procaspase-8 by forming a cDISC will be invaluable for investigating activation of ligand-mediated apoptosis and the numerous interactions affecting procaspase-8 activation.


Subject(s)
Caspase 8/metabolism , Death Domain Receptor Signaling Adaptor Proteins/biosynthesis , Enzyme Activation , Fas-Associated Death Domain Protein/physiology , Humans , Protein Folding
16.
Mol Cells ; 41(5): 454-464, 2018 May 31.
Article in English | MEDLINE | ID: mdl-29754475

ABSTRACT

Crosstalk between G-protein signaling and glutamatergic transmission within the brain reward circuits is critical for long-term emotional effects (depression and anxiety), cravings, and negative withdrawal symptoms associated with opioid addiction. A previous study showed that Regulator of G-protein signaling 4 (RGS4) may be implicated in opiate action in the nucleus accumbens (NAc). However, the mechanism of the NAc-specific RGS4 actions that induce the behavioral responses to opiates remains largely unknown. The present study used a short hairpin RNA (shRNA)-mediated knock-down of RGS4 in the NAc of the mouse brain to investigate the relationship between the activation of ionotropic glutamate receptors and RGS4 in the NAc during morphine reward. Additionally, the shRNA-mediated RGS4 knock-down was implemented in NAc/striatal primary-cultured neurons to investigate the role that striatal neurons have in the morphine-induced activation of ionotropic glutamate receptors. The results of this study show that the NAc-specific knockdown of RGS4 significantly increased the behaviors associated with morphine and did so by phosphorylation of the GluR1 (Ser831) and NR2A (Tyr1325) glutamate receptors in the NAc. Furthermore, the knock-down of RGS4 enhanced the phosphorylation of the GluR1 and NR2A glutamate receptors in the primary NAc/striatal neurons during spontaneous morphine withdrawal. These findings show a novel molecular mechanism of RGS4 in glutamatergic transmission that underlies the negative symptoms associated with morphine administration.


Subject(s)
Glutamic Acid/physiology , Morphine/pharmacology , Nerve Tissue Proteins/physiology , Nucleus Accumbens/physiology , RGS Proteins/physiology , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Reward , Animals , Cells, Cultured , Corpus Striatum/cytology , Exploratory Behavior/drug effects , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nucleus Accumbens/cytology , Nucleus Accumbens/drug effects , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , RGS Proteins/antagonists & inhibitors , RGS Proteins/genetics , RNA Interference , RNA, Small Interfering/genetics
17.
ACS Cent Sci ; 4(7): 885-893, 2018 Jul 25.
Article in English | MEDLINE | ID: mdl-30062117

ABSTRACT

LK-3, an amphipathic dimeric peptide linked by two disulfide bonds, and related isomeric bundles were synthesized, and their cell penetrating abilities were investigated. The measurements using size exclusion chromatography and dynamic light scattering show that LK-3 and its isomers form cell penetrating oligomers. Calculations, performed for various types of peptide isomers, elucidate a strong correlation between the amphipathic character of dimers and cell penetration ability. The results suggest that the amphipathicities of LK-3 and related bundle dimers are responsible for their oligomerization propensities which in turn determine their cell penetrating abilities. The observations made in this study provide detailed information about the mechanism of cell uptake of LK-3 and suggest a plausible insight of the early stage of nanoparticle formation of the cell penetrating amphipathic peptides.

18.
Sci Rep ; 8(1): 13752, 2018 09 13.
Article in English | MEDLINE | ID: mdl-30213954

ABSTRACT

Established fear memory becomes vulnerable to disruption after memory retrieval and extinction; this labile state is critical for inhibiting the return of fear memory. However, the labile state has a very narrow time window after retrieval, and underlying molecular mechanisms are not well known. To that end, we isolated the hippocampus immediately after fear memory retrieval and performed proteomics. We identified Neurobeachin (NBEA), an autism-related regulator of synaptic protein trafficking, to be upregulated after contextual fear memory retrieval. NBEA protein expression was rapid and transient after fear memory retrieval at the synapse. Nbea mRNA was enriched at the synapses, and the rapid induction of NBEA expression was blocked by inhibition of the mammalian target of rapamycin (mTOR)-dependent signaling pathway. Mice with cornu ammonis 1 (CA1)-specific Nbea shRNA knockdown showed normal fear acquisition and contextual fear memory but impaired extinction, suggesting an important role of Nbea in fear memory extinction processes. Consistently, Nbea heterozygotes showed normal fear acquisition and fear memory recall but showed impairment in extinction. Our data suggest that NBEA is necessary either for induction of memory lability or for the physiological process of memory extinction.


Subject(s)
Carrier Proteins/genetics , Fear/physiology , Memory/physiology , Nerve Tissue Proteins/genetics , Animals , Autistic Disorder/genetics , Autistic Disorder/pathology , CA1 Region, Hippocampal/physiology , Carrier Proteins/chemistry , Carrier Proteins/physiology , Chromosome Pairing/genetics , Chromosome Pairing/physiology , Heterozygote , Hippocampus/physiology , Humans , Membrane Proteins , Mice , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/physiology , Protein Transport/genetics , Proteomics , TOR Serine-Threonine Kinases/genetics
19.
Oncol Lett ; 13(5): 3681-3687, 2017 May.
Article in English | MEDLINE | ID: mdl-28521470

ABSTRACT

Human ribosomal protein S3 (hRpS3) is a component of the 40S ribosomal subunit that associated in protein synthesis. hRpS3 has additional ribosomal functions such as DNA repair, transcription, metastasis, and apoptosis via interaction with numerous signaling molecules and has different modifications. Cyclin-dependent kinases (CDKs) are heterodimeric serine/threonine protein kinases that regulate cell cycle progression. Among its members, the Cdk1-cyclin B complex is known to control cell progression in the G2/M phase, while Cdk2-cyclin E/A complexes function in G1/S and S/G2 transition. In our previous study, we observed interaction between hRpS3 and Cdk1. The present study investigated the interaction between hRpS3 and Cdk2. Cdk2 phosphorylated hRps3 at amino acid residues S6 and T221 during the S-phase. Furthermore, hRpS3 knockdown delayed cell cycle progression by modulating the expression of cell cycle-related proteins, including cyclin B1 and cyclin E1. These findings suggest that hRpS3 is involved in Cdk2-mediated cell cycle regulation.

20.
Phytochemistry ; 67(9): 870-5, 2006 May.
Article in English | MEDLINE | ID: mdl-16603211

ABSTRACT

Kachri fruit, Cucumis trigonus Roxburghi, contains high protease activity and has been used as meat tenderizer in the Indian subcontinent. A 67 kDa serine protease from Kachri fruit was purified by DEAE-Sepharose and CM-Sepharose chromatography, whose optimum activity was at pH 11 and 70 degrees C. Its activity was strongly inhibited by PMSF, but not by EDTA, pepstatin, or cysteine protease inhibitors. The substrate specificity of the purified protease towards synthetic peptides was comparable to cucumisin, the first characterized subtilisin class plant protease from the sarcocarp of melon fruit (Cucumis melo). These characteristics, along with the N-terminal amino acid sequence, indicated that the isolated protease from Cucumis trigonus Roxburghi is a cucumisin homologue, which belongs to the serine protease family.


Subject(s)
Cucumis/enzymology , Serine Endopeptidases/chemistry , Serine Endopeptidases/isolation & purification , Amino Acid Sequence , Cucumis/chemistry , Fruit/chemistry , Hydrogen-Ion Concentration , Molecular Sequence Data , Plant Extracts/chemistry , Subtilisins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL