Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Medicine (Baltimore) ; 101(35): e30472, 2022 Sep 02.
Article in English | MEDLINE | ID: mdl-36107571

ABSTRACT

BACKGROUND: Localized senile pruritus is a continued health problem for the elderly. This study aimed to evaluate the efficacy and safety of artemether emulsion on localized senile pruritus. METHODS: Sixty patients diagnosed with senile pruritus were randomized into the artemether emulsion (1%) group or emulsion base group in a 1:1 ratio (the artemether group vs the control group). The patients used artemether emulsion or emulsion base for pruritus twice daily for 2 weeks. The pruritus visual analog scale (VAS) and the rate of adverse events were evaluated in week 0 and week 2. RESULTS: The VAS scores in week 2 after treatment decreased significantly compared with those before treatment in both groups (P < .05). After treatment, patients receiving the artemether emulsion had significantly lower mean VAS scores compared to those who received the emulsion base (1.21 ±â€…1.64 vs 3.67 ±â€…2.97, P < .05). When the VAS scores were compared between the 2 groups before treatment, the effective rate of the artemether group was significantly higher than that of the control group (χ2 = 55, P < .05) in week 2 after treatment. Besides, no adverse events occurred in both groups. CONCLUSIONS: Both artemether emulsion and emulsion base were effective in treating localized senile pruritus, and artemether emulsion was superior to emulsion base.


Subject(s)
Pruritus , Aged , Artemether , Emulsions , Humans , Pilot Projects , Pruritus/drug therapy , Pruritus/etiology , Visual Analog Scale
2.
World J Clin Cases ; 10(1): 166-176, 2022 Jan 07.
Article in English | MEDLINE | ID: mdl-35071516

ABSTRACT

BACKGROUND: Tissue resident memory T (TRM) cells have been reported to play a significant role in the pathogenesis and relapse of chronic eczema. AIM: To compare the efficacy and safety of the intralesional injection of 5-fluorouracil (5-FU) and triamcinolone (TA) with those associated with TA alone for the treatment of chronic eczema. METHODS: A total of 168 patients were randomized to 5-FU+TA or TA groups and received a one-time intralesional injection of 5-FU+TA or TA only. Biopsies were collected before and 2 wk after treatment for evaluation of histopathological changes. All patients were followed up monthly for up to 1 year. RESULTS: No serious adverse event was observed in either group. Although the mean atopic dermatitis severity index scores and effective rates were comparable between the two groups after 2 wk of treatment, the relapse rate was significantly lower in the 5-FU+TA group than in the TA group. Histological examination showed significantly fewer CD8+ and CD103+ T cells but not CD4+ T cells in the 5-FU+TA group. CONCLUSION: One-time intralesional injection of 5-FU+TA is effective and safe for chronic eczema treatment and can further reduce the retention of TRM cells in the lesional skin and the relapse rate of chronic eczema.

3.
J Dermatolog Treat ; 32(7): 762-765, 2021 Nov.
Article in English | MEDLINE | ID: mdl-31884836

ABSTRACT

OBJECTIVE: To evaluate the efficacy and safety of artemether emulsion treating patients with mild-to-moderate acne vulgaris. METHODS: A total of 73 (randomized 1:1) patients were externally administered either artemether emulsion (1%) or fusidic acid emulsion (5g: 0.1g) twice daily for 12 weeks. Efficacy and safety evaluations were performed at weeks 0 and 12 by Global acne Grading System (GAGS), the number of acne and papule, as well as the rate of clinical respond. RESULTS: After 12 weeks, patients randomized to the artemether emulsion group received artemether emulsion had significantly lower GAGS scores (5.08 ± 1.99 versus 13.75 ± 4.87, p < .001) compared to patients who received fusidic acid emulsion. Patients in the artemether emulsion group had comparable baseline acne scores (11.11 ± 3.73 versus 10.75 ± 4.66, p = .626) and papule score (16.11 ± 5.58 versus 17.03 ± 6.34, p = .356), but significantly lower acne score (3.00 ± 1.55 versus 9.08 ± 4.90, p < .001) and comparable papule score (2.81 ± 1.61 versus 12.69 ± 5.45, p < .001) compared to the fusidic acid emulsion group at 12 weeks. No major adverse events were noted in either treatment group through 12 weeks. CONCLUSIONS: Artemether emulsion had better effect in improving mild-to-moderate AV compared to fusidic acid emulsion with barely AEs.


Subject(s)
Acne Vulgaris , Acne Vulgaris/drug therapy , Artemether , Emulsions , Humans , Pilot Projects , Treatment Outcome
4.
J Dermatolog Treat ; 30(8): 809-812, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31017492

ABSTRACT

Objective: To assess the efficacy and safety of artemether emulsion in patients with papulopustular rosacea. Methods: A total of 130 (randomized 1:1) were externally administered either artemether emulsion (1%) or metronidazole emulsion (3%) twice daily for 4 weeks with an open-label 8-week follow-up. The primary endpoints included the proportion of patients who achieved clinical effective responses, as well as erythema and papule and pustule score at week 4. Results: Numerically more patients achieved an effective response at week 4 with artemether emulsion (87.1%) than metronidazole emulsion (80.0%) (p > .05). Patients with artemether emulsion had comparable baseline erythema score (2.45 ± 0.67 versus 2.42 ± 0.70, p = .809) and papule and pustule score (2.11 ± 0.96 versus 2.32 ± 0.83, p = .264), but significantly lower papule and pustule score (0.21 ± 0.52 versus 0.42 ± 0.83, p = .001) and comparable erythema score (0.53 ± 0.88 versus 0.62 ± 0.88, p = .999) compared to patients with metronidazole emulsion at week 4. There was a significantly higher proportion of patients with metronidazole emulsion relapse compared to metronidazole emulsion during the open-label 8-week follow-up period (21.6% versus 2.4%, p < .01). Conclusions: Artemether emulsion improved papulopustular rosacea in the metronidazole emulsion group as early as 4 weeks, but its beneficial effect was maintained through the 8-week follow-up period compared to metronidazole emulsion.


Subject(s)
Artemether/therapeutic use , Rosacea/drug therapy , Adult , Artemether/adverse effects , Artemether/chemistry , Drug Administration Schedule , Emulsions/chemistry , Female , Humans , Male , Metronidazole/chemistry , Metronidazole/therapeutic use , Middle Aged , Pilot Projects , Pruritus/etiology , Rosacea/pathology , Severity of Illness Index , Treatment Outcome , Young Adult
5.
Acta Pharmacol Sin ; 40(1): 64-74, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30013035

ABSTRACT

Isoalantolactone (IAL) is a sesquiterpene lactone extracted from roots of Inula helenium L and has shown anti-inflammatory effects. In this study we investigated the therapeutic effects of IAL on acute lung injury (ALI) and elucidated the mechanisms underlying its anti-inflammation potential in vitro and in vivo. Treatment with lipopolysaccharide (LPS, 100 ng/mL) drastically stimulated production of inflammatory mediators such as NO, TNF-α, IL-1ß, and IL-6 in mouse bone marrow-derived macrophages (BMDMs), which was dose-dependently suppressed by pretreatment with IAL (2.5, 5, 10, 20 µM). We further revealed that IAL suppressed LPS-induced NF-κB, ERK, and Akt activation. Moreover, the downregulation of non-degradable K63-linked polyubiquitination of TRAF6, an upstream transcription factor of NF-κB, contributed to the anti-inflammatory effects of IAL. ALI was induced in mice by intratracheal injection of LPS (5 mg/kg). Administration of IAL (20 mg/kg, i.p.) significantly suppressed pulmonary pathological changes, neutrophil infiltration, pulmonary permeability, and pro-inflammatory cytokine expression. Our results demonstrate that IAL is a potential therapeutic reagent against inflammation and ALI.


Subject(s)
Acute Lung Injury/drug therapy , Anti-Inflammatory Agents/therapeutic use , Inflammation/drug therapy , Sesquiterpenes/therapeutic use , TNF Receptor-Associated Factor 6/metabolism , Ubiquitination/drug effects , Acute Lung Injury/chemically induced , Acute Lung Injury/pathology , Animals , Cytokines/metabolism , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins , Lipopolysaccharides , Lung/pathology , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , NF-kappa B p50 Subunit/metabolism , Signal Transduction/drug effects
6.
Acta Pharmacol Sin ; 39(1): 85-96, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29047459

ABSTRACT

Protostemonine (PSN) is the main anti-inflammatory alkaloid extracted from the roots of Stemona sessilifolia (known as "Baibu" in traditional Chinese medicine). Here, we reported the inhibitory effects of PSN on lipopolysaccharide (LPS)-induced macrophage activation in vitro and LPS-induced acute lung injury in mice. Macrophage cell line RAW264.7 cells and mouse bone marrow-derived macrophages (BMDMs) were treated with PSN (1, 3, 10, 30 and 100 µmol/L) for 0.5 h and then challenged with LPS (0.1 µg/mL) for 24 h. Pretreatment with PSN significantly inhibited LPS-induced phosphorylation of MAPKs and AKT, iNOS expression and NO production in the macrophages. C57BL/6 mice were intratracheally injected with LPS (5 mg/kg) to induce acute lung injury (ALI). The mice were subsequently treated with PSN (10 mg/kg, ip) at 4 and 24 h after LPS challenge. PSN administration significantly attenuated LPS-induced inflammatory cell infiltration, reduced pro-inflammatory cytokine (TNF-α, IL-1ß and IL-6) production and eliminated LPS-mediated lung edema. Furthermore, PSN administration significantly inhibited LPS-induced pulmonary MPO activity. Meanwhile, LPS-induced phosphorylation of p38 MAPK, iNOS expression and NO production in the lungs were also suppressed. The results demonstrate that PSN effectively attenuates LPS-induced inflammatory responses in vitro and in vivo; the beneficial effects are associated with the decreased phosphorylation of MAPK and AKT and the reduced expression of pro-inflammatory mediators, such as iNOS, NO and cytokines. These data suggest that PSN may be a potential therapeutic agent in the treatment of ALI.


Subject(s)
Acute Lung Injury/prevention & control , Alkaloids/therapeutic use , Acute Lung Injury/chemically induced , Alkaloids/administration & dosage , Animals , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Lung/pathology , Macrophage Activation , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Peroxidase/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Edema/prevention & control , RAW 264.7 Cells , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Molecules ; 22(3)2017 Mar 13.
Article in English | MEDLINE | ID: mdl-28335409

ABSTRACT

The formyl peptide receptors (FPRs) are G protein-coupled receptors that transduce chemotactic signals in phagocytes and mediate host-defense as well as inflammatory responses including cell adhesion, directed migration, granule release and superoxide production. In recent years, the cellular distribution and biological functions of FPRs have expanded to include additional roles in homeostasis of organ functions and modulation of inflammation. In a prototype, FPRs recognize peptides containing N-formylated methionine such as those produced in bacteria and mitochondria, thereby serving as pattern recognition receptors. The repertoire of FPR ligands, however, has expanded rapidly to include not only N-formyl peptides from microbes but also non-formyl peptides of microbial and host origins, synthetic small molecules and an eicosanoid. How these chemically diverse ligands are recognized by the three human FPRs (FPR1, FPR2 and FPR3) and their murine equivalents is largely unclear. In the absence of crystal structures for the FPRs, site-directed mutagenesis, computer-aided ligand docking and structural simulation have led to the identification of amino acids within FPR1 and FPR2 that interact with several formyl peptides. This review article summarizes the progress made in the understanding of FPR ligand diversity as well as ligand recognition mechanisms used by these receptors.


Subject(s)
Receptors, Formyl Peptide/chemistry , Receptors, Formyl Peptide/metabolism , Amino Acid Motifs , Animals , Binding Sites , Homeostasis , Humans , Ligands , Mice , Models, Molecular , Molecular Docking Simulation , Mutagenesis, Site-Directed , Protein Binding
8.
Acta Pharmacol Sin ; 38(3): 342-350, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28112185

ABSTRACT

Toll-like receptor 4 (TLR4)-mediated signaling plays a critical role in sepsis-induced acute lung injury (ALI). LYRM03 (3-amino-2-hydroxy-4-phenyl-valyl-isoleucine) is a novel derivative of ubenimex, a widely used antineoplastic medicine. We previously found that LYRM03 has anti-inflammatory effects in cecal ligation puncture mouse model. In this study we determined whether LYRM03 attenuated LPS-induced ALI in mice. LPS-induced ALI mouse model was established by challenging the mice with intratracheal injection of LPS (5 mg/kg), which was subsequently treated with LYRM03 (10 mg/kg, ip). LYRM03 administration significantly alleviated LPS-induced lung edema, inflammatory cell (neutrophils and macrophages) infiltration and myeloperoxidase (MPO) activity, decreased pro-inflammatory and chemotactic cytokine (TNF-α, IL-6, IL-1ß, MIP-2) generation and reduced iNOS and COX-2 expression in the lung tissues. In cultured mouse alveolar macrophages in vitro, pretreatment with LYRM03 (100 µmol/L) suppressed LPS-induced macrophage activation by reducing Myd88 expression, increasing IκB stability and inhibiting p38 phosphorylation. These results suggest that LYRM03 effectively attenuates LPS-induced ALI by inhibiting the expression of pro-inflammatory mediators and Myd88-dependent TLR4 signaling pathways in alveolar macrophages. LYRM03 may serve as a potential treatment for sepsis-mediated lung injuries.


Subject(s)
Acute Lung Injury/drug therapy , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Lipopolysaccharides/pharmacology , Oligopeptides/therapeutic use , Toll-Like Receptor 4/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/immunology , Animals , Cytokines/metabolism , Inflammation/drug therapy , Inflammation/immunology , Inflammation/metabolism , Macrophage Activation/drug effects , Male , Mice, Inbred C57BL , Neutrophil Infiltration/drug effects , Signal Transduction
9.
Chem Biol Drug Des ; 87(6): 895-904, 2016 06.
Article in English | MEDLINE | ID: mdl-26804061

ABSTRACT

DMAKO-05((S)-1-((5E,8E)-5,8-bis(hydroxyimino)-1,4-dimethoxy-5,8-dihydronaphthalen-2-yl)-4-methylpent-3-enyl 3-methylbutanoate) is a novel oxime derivative of shikonin, the major component extracted from Chinese herb Lithospermun erythrorhizon. Here, we report that DMAKO-05 had an antitumor activity against mouse melanoma cell line B16F0. Our studies indicated that DMAKO-05 not only inhibited B16F0 proliferation and migration but also led to cell cycle arrest at G1 phase and cell apoptosis, in which DMAKO-05 triggered mitochondrial-mediated apoptosis signal including caspase-9/3 and PARP. In response to DMAKO-05 treatment, the Akt-mediated survival signals were remarkably attenuated in B16F0 cells. Collectively, DMAKO-05 has a strong cytotoxicity in B16F0 cells via inhibiting Akt activation, inducing G1 arrest, and promoting B16F0 cell apoptosis. DMAKO-05 might serve as a potential candidate lead compound for melanoma.


Subject(s)
Antineoplastic Agents, Phytogenic , Cell Proliferation/drug effects , Melanoma , Naphthoquinones , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Animals , Antineoplastic Agents, Phytogenic/chemical synthesis , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , Drug Screening Assays, Antitumor , Melanoma/drug therapy , Melanoma/metabolism , Naphthoquinones/chemical synthesis , Naphthoquinones/chemistry , Naphthoquinones/pharmacology , Rats
10.
Acta Pharmacol Sin ; 35(5): 653-63, 2014 May.
Article in English | MEDLINE | ID: mdl-24658352

ABSTRACT

AIM: The chemerin receptor CMKLR1 is one type of G protein-coupled receptors abundant in monocyte-derived dendritic cells and macrophages, which plays a key role in the entry of a subset of immunodeficiency viruses including HIV/SIV into lymphocytes and macrophages. The aim of this work was to investigate how CMKLR1 was internalized and whether its internalization affected cell signaling in vitro. METHODS: Rat basophilic leukemia RBL-2H3 cells, HEK 293 cells, and HeLa cells were used. CMKLR1 internalization was visualized by confocal microscopy imaging or using a FACScan flow cytometer. Six potential phosphorylation sites (Ser337, Ser343, Thr352, Ser344, Ser347, and Ser350) in CMKLR1 were substituted with alanine using site-directed mutagenesis. Heterologous expression of wild type and mutant CMKLR1 allowed for functional characterization of endocytosis, Ca(2+) flux and extracellular signal-regulated kinase (ERK) phosphorylation. RESULTS: Chemerin and the chemerin-derived nonapeptide (C9) induced dose-dependent loss of cell surface CMKLR1-GFP fusion protein and increased its intracellular accumulation in HEK 293 cells and RBL-2H3 cells stably expressing CMKLR1. Up to 90% of CMKLR1 was internalized after treatment with C9 (1 µmol/L). By using different agents, it was demonstrated that clathrin-independent mechanism was involved in CMKLR1 internalization. Mutations in Ser343 for G protein-coupled receptor kinase phosphorylation and in Ser347 for PKC phosphorylation abrogated CMKLR1 internalization. Loss of CMKLR1 internalization partially enhanced the receptor signaling, as shown by increased Ca(2+) flux and a shorter latency to peak level of ERK phosphorylation. CONCLUSION: CMKLR1 internalization occurs in a clathrin-independent manner, which negatively regulated the receptor-mediated Ca(2+) flux and ERK phosphorylation.


Subject(s)
Clathrin/metabolism , Receptors, Chemokine/metabolism , Signal Transduction/physiology , Animals , Calcium/metabolism , Cell Line , Cell Line, Tumor , Endocytosis/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Green Fluorescent Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , Leukemia, Basophilic, Acute/metabolism , Phosphorylation/physiology , Rats , Receptors, G-Protein-Coupled/metabolism
11.
J Biol Chem ; 289(4): 2295-306, 2014 Jan 24.
Article in English | MEDLINE | ID: mdl-24285541

ABSTRACT

Unlike formyl peptide receptor 1 (FPR1), FPR2/ALX (FPR2) interacts with peptides of diverse sequences but has low affinity for the Escherichia coli-derived chemotactic peptide fMet-Leu-Phe (fMLF). Using computer modeling and site-directed mutagenesis, we investigated the structural requirements for FPR2 to interact with formyl peptides of different length and composition. In calcium flux assay, the N-formyl group of these peptides is necessary for activation of both FPR2 and FPR1, whereas the composition of the C-terminal amino acids appears more important for FPR2 than FPR1. FPR2 interacts better with pentapeptides (fMLFII, fMLFIK) than tetrapeptides (fMLFK, fMLFW) and tripeptide (fMLF) but only weakly with peptides carrying negative charges at the C terminus (e.g. fMLFE). In contrast, FPR1 is less sensitive to negative charges at the C terminus. A CXCR4-based homology model of FPR1 and FPR2 suggested that Asp-281(7.32) is crucial for the interaction of FPR2 with certain formyl peptides as its negative charge may be repulsive with the terminal COO- group of fMLF and negatively charged Glu in fMLFE. Asp-281(7.32) might also form a stable interaction with the positively charged Lys in fMLFK. Site-directed mutagenesis was performed to remove the negative charge at position 281 in FPR2. The D281(7.32)G mutant showed improved affinity for fMLFE and fMLF and reduced affinity for fMLFK compared with wild type FPR2. These results indicate that different structural determinants are used by FPR1 and FPR2 to interact with formyl peptides.


Subject(s)
Molecular Dynamics Simulation , N-Formylmethionine Leucyl-Phenylalanine/analogs & derivatives , Receptors, Formyl Peptide/chemistry , Receptors, Lipoxin/chemistry , Amino Acid Substitution , Humans , Ligands , Mutagenesis, Site-Directed , Mutation, Missense , N-Formylmethionine Leucyl-Phenylalanine/chemistry , N-Formylmethionine Leucyl-Phenylalanine/metabolism , Receptors, CXCR4/chemistry , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Receptors, Formyl Peptide/genetics , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/genetics , Receptors, Lipoxin/metabolism , Structural Homology, Protein , Structure-Activity Relationship
12.
Mol Pharmacol ; 83(2): 389-98, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23160941

ABSTRACT

The evolutionary relationship and functional correlation between human formyl peptide receptors (FPRs) and their mouse counterparts remain incompletely understood. We examined three members of the mouse formyl peptide receptor subfamily (mFprs) and found that they differ in agonist preference and cellular distributions. When stably expressed in transfected rat basophilic leukemia (RBL-2H3) cells, mFpr1 was readily activated by N-formylated peptides derived from Listeria monocytogenes (fMIVTLF), Staphylococcus aureus (fMIFL), and mitochondria (fMMYALF). In contrast, the Escherichia coli-derived fMLF was 1000-fold less potent. The aforementioned peptides were much less efficacious at mFpr2, which responded better to the synthetic hexapeptide WKYMVm, the synthetic agonists Quin-C1 (a substituted quinazolinone), and compound 43 (a nitrosylated pyrazolone derivative). Saturation binding assays showed that mFpr1 and mFpr2 were expressed at similar levels on the cell surface, although their affinity for N-formyl-Met-Leu-Phe-Ile-Ile-Lys-fluorescein isothiocyanate varied by more than 1000-fold [dissociation constant (K(d)) values of 2.8 nM for mFpr1 and 4.8 µM for mFpr2]). Contrary to these receptors, mFpr-rs1 responded poorly to all the previously mentioned peptides that were tested. Fluorescent microscopy revealed an intracellular distribution pattern of mFpr-rs1. On the basis of these results, we conclude that mFpr1 is an ortholog of human FPR1 with certain pharmacologic properties of human FPR2/ALX, whereas mFpr2 has much lower affinity for formyl peptides. The intracellular distribution of mFpr-rs1 suggests an evolutionary correlation with human FPR3.


Subject(s)
N-Formylmethionine Leucyl-Phenylalanine/metabolism , Receptors, Formyl Peptide/metabolism , Animals , Benzamides/pharmacology , Calcium/metabolism , Cell Line, Tumor , Escherichia coli/metabolism , Leukemia, Basophilic, Acute/metabolism , Listeria monocytogenes/metabolism , Mice , Mitochondria/metabolism , Oligopeptides/pharmacology , Protein Binding , Quinazolines/pharmacology , Rats , Staphylococcus aureus/metabolism , Transfection/methods
13.
Neurosci Bull ; 28(3): 209-21, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22622820

ABSTRACT

OBJECTIVE: In this study, the pharmacological kinetics of Buthus martensi Karsch (BmK) AS, a specific modulator of voltage-gated sodium channel site 4, was investigated on Na(v)1.3 expressed in Xenopus oocytes. METHODS: Two-electrode voltage clamp was used to record the whole-cell sodium current. RESULTS: The peak currents of Na(v)1.3 were depressed by BmK AS over a wide range of concentrations (10, 100, and 500 nmol/L). Most remarkably, BmK AS at 100 nmol/L hyperpolarized the voltage-dependence and increased the voltage-sensitivity of steady-state activation/inactivation. In addition, BmK AS was capable of hyperpolarizing not only the fast inactivation but also the slow inactivation, with a greater preference for the latter. Moreover, BmK AS accelerated the time constant and increased the ratio of recovery in Na(v)1.3 at all concentrations. CONCLUSION: This study provides direct evidence that BmK AS facilitates steady-state activation and inhibits slow inactivation by stabilizing both the closed and open states of the Na(v)1.3 channel, which might result from an integrative binding to two receptor sites on the voltage-gated sodium channels. These results may shed light on therapeutics against Na(v)1.3-targeted pathology.


Subject(s)
NAV1.3 Voltage-Gated Sodium Channel/drug effects , NAV1.3 Voltage-Gated Sodium Channel/metabolism , Peptides/pharmacology , Scorpion Venoms/pharmacology , Animals , Dose-Response Relationship, Drug , Kinetics , Membrane Potentials/drug effects , Oocytes/drug effects , Oocytes/metabolism , Xenopus
14.
Neurosci Bull ; 24(5): 283-7, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18839021

ABSTRACT

OBJECTIVE: To examine the effect of deglycosylation on gating properties of rNav1.3. METHODS: rNav1.3 was expressed in Xenopus oocyte, with glycosylation inhibition by using tunicamycin. Two-electrode voltage clamp was employed to record the whole-cell sodium current and data were analyzed by Origin software. Those of glycosylated rNav1.3 were kept as control. RESULTS: Compared with glycosylated ones, the steady-state activation curve of deglycosylated rNav1.3 was positively shifted by about 10 mV, while inactivation curve was negatively shifted by about 8 mV. CONCLUSION: Glycosylation altered the gating properties of rNav1.3 and contributed to the functional diversity.


Subject(s)
Homeostasis/physiology , Ion Channel Gating/physiology , Membrane Potentials/physiology , Nerve Tissue Proteins/physiology , Sodium Channels/physiology , Animals , Electric Conductivity , Electric Stimulation , Gene Transfer Techniques , Glycosylation/drug effects , Homeostasis/drug effects , Ion Channel Gating/drug effects , Membrane Potentials/drug effects , NAV1.3 Voltage-Gated Sodium Channel , Oocytes , Patch-Clamp Techniques , Static Electricity , Tunicamycin/pharmacology , Xenopus
15.
Biophys J ; 94(9): 3706-13, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18199674

ABSTRACT

Martentoxin as a 37-residue peptide was capable of blocking large-conductance Ca(2+)-activated K(+) (BK) channels in adrenal medulla chromaffin cells. This study investigated the pharmacological discrimination of martentoxin on BK channel subtypes. The results showed that the iberiotoxin-insensitive neuronal BK channels (alpha+beta4) could be potently blocked by martentoxin (IC(50) = approximately 80 nM). In contrast, the iberiotoxin-sensitive BK channel consisting of only alpha-subunit was less sensitive to martentoxin. Distinctively, martentoxin inhibited neuronal BK channels (alpha+beta4) with a novel interaction mode. Two possible interaction sites of neuronal BK channels (alpha+beta4) might be responsible for the binding with martentoxin: one for trapping and the other located at the pore region for blocking. In addition, the inhibition of martentoxin on neuronal BK channels (alpha+beta4) depended on cytoplasmic Ca(2+) concentration. On the other hand, in vivo experiments from EEG recordings suggested that neuronal BK channels (alpha+beta4) were the primary target of martentoxin. Therefore, this research not only sheds light on a unique ligand for neuronal BK channels (alpha+beta4), but also highlights a novel model approach for the interaction between K(+) channels and specific-ligands.


Subject(s)
Ion Channel Gating/drug effects , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Models, Biological , Neurons/metabolism , Scorpion Venoms/toxicity , Allosteric Regulation/drug effects , Animals , Brain/drug effects , Calcium/metabolism , Cattle , Cell Line , Electroencephalography/drug effects , Humans , Male , Peptides/toxicity , Rats , Rats, Sprague-Dawley , Substrate Specificity
16.
FEBS Lett ; 580(18): 4508-14, 2006 Aug 07.
Article in English | MEDLINE | ID: mdl-16870180

ABSTRACT

Scorpion toxins have been found lacking effect on Na(+) current of its own sodium channel, whereas the molecular mechanism remains mystery. In this study, the binding affinity of pharmacologically distinct scorpion toxins was found much weaker to scorpion (Buthus martensii) nerve synaptosomes than to spider (Ornithoctonus huwena) ones. The sodium channel cDNA from these two species were further cloned. The deduced proteins contain 1871 and 1987 amino acids respectively. Several key amino acid substitutions, i.e., A1610V, I1611L and S1617K, are found in IVS3-S4 constituting receptor site-3, and for receptor site-4, two residues (Leu-Pro) are inserted near IIS4 of scorpion sodium channel.


Subject(s)
Scorpion Venoms/metabolism , Sodium Channels/genetics , Sodium Channels/metabolism , Adaptation, Physiological/genetics , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites , Cloning, Molecular , Molecular Sequence Data , Mutation , Phylogeny , Scorpions/genetics , Sequence Alignment , Sodium Channels/classification , Spiders/genetics , Synaptosomes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...