Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
1.
Exp Mol Med ; 56(3): 686-699, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38480902

ABSTRACT

Cancer cells often exhibit resistance to apoptotic cell death, but they may be vulnerable to other types of cell death. Elucidating additional mechanisms that govern cancer cell death is crucial for developing new therapies. Our research identified cyclic AMP-responsive element-binding protein 3 (CREB3) as a crucial regulator and initiator of a unique cell death mechanism known as karyoptosis. This process is characterized by nuclear shrinkage, deformation, and the loss of nuclear components following nuclear membrane rupture. We found that the N-terminal domain (aa 1-230) of full-length CREB3 (CREB3-FL), which is anchored to the nuclear inner membrane (INM), interacts with lamins and chromatin DNA. This interaction maintains a balance between the outward force exerted by tightly packed DNA and the inward constraining force, thereby preserving INM integrity. Under endoplasmic reticulum (ER) stress, aberrant cleavage of CREB3-FL at the INM leads to abnormal accumulation of the cleaved form of CREB3 (CREB3-CF). This accumulation disrupts the attachment of CREB3-FL to the INM, resulting in sudden rupture of the nuclear membrane and the onset of karyoptosis. Proteomic studies revealed that CREB3-CF overexpression induces a DNA damage response akin to that caused by UVB irradiation, which is associated with cellular senescence in cancer cells. These findings demonstrated that the dysregulation of CREB3-FL cleavage is a key factor in karyoptotic cell death. Consequently, these findings suggest new therapeutic strategies in cancer treatment that exploit the process of karyoptosis.


Subject(s)
Cyclic AMP Response Element-Binding Protein , Nuclear Envelope , Proteomics , Apoptosis , DNA , Nuclear Envelope/metabolism , Humans , Cell Line, Tumor , Cyclic AMP Response Element-Binding Protein/metabolism
2.
Cell Rep ; 43(2): 113685, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38261513

ABSTRACT

Tumor necrosis factor α (TNF-α) is a major pro-inflammatory cytokine, important in many diseases, that sensitizes nociceptors through its action on a variety of ion channels, including voltage-gated sodium (NaV) channels. We show here that TNF-α acutely upregulates sensory neuron excitability and current density of threshold channel NaV1.7. Using electrophysiological recordings and live imaging, we demonstrate that this effect on NaV1.7 is mediated by p38 MAPK and identify serine 110 in the channel's N terminus as the phospho-acceptor site, which triggers NaV1.7 channel insertion into the somatic membrane. We also show that the N terminus of NaV1.7 is sufficient to mediate this effect. Although acute TNF-α treatment increases NaV1.7-carrying vesicle accumulation at axonal endings, we did not observe increased channel insertion into the axonal membrane. These results identify molecular determinants of TNF-α-mediated regulation of NaV1.7 in sensory neurons and demonstrate compartment-specific effects of TNF-α on channel insertion in the neuronal plasma membrane.


Subject(s)
Sensory Receptor Cells , Tumor Necrosis Factor-alpha , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Sensory Receptor Cells/metabolism , Axons/metabolism , Nociceptors/metabolism , Cell Membrane/metabolism
3.
Biomol Ther (Seoul) ; 31(2): 168-175, 2023 Mar 01.
Article in English | MEDLINE | ID: mdl-36779240

ABSTRACT

Tramadol is an opioid analog used to treat chronic and acute pain. Intradermal injections of tramadol at hundreds of millimoles have been shown to produce a local anesthetic effect. We used the whole-cell patch-clamp technique in this study to investigate whether tramadol blocks the sodium current in HEK293 cells, which stably express the pain threshold sodium channel Nav1.7 or the cardiac sodium channel Nav1.5. The half-maximal inhibitory concentration of tramadol was 0.73 mM for Nav1.7 and 0.43 mM for Nav1.5 at a holding potential of -100 mV. The blocking effects of tramadol were completely reversible. Tramadol shifted the steady-state inactivation curves of Nav1.7 and Nav1.5 toward hyperpolarization. Tramadol also slowed the recovery rate from the inactivation of Nav1.7 and Nav1.5 and induced stronger use-dependent inhibition. Because the mean plasma concentration of tramadol upon oral administration is lower than its mean blocking concentration of sodium channels in this study, it is unlikely that tramadol in plasma will have an analgesic effect by blocking Nav1.7 or show cardiotoxicity by blocking Nav1.5. However, tramadol could act as a local anesthetic when used at a concentration of several hundred millimoles by intradermal injection and as an antiarrhythmic when injected intravenously at a similar dose, as does lidocaine.

4.
Arch Pharm Res ; 46(1): 44-58, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36607545

ABSTRACT

E2F 1, 2, and 3a, (refer to as E2Fs) are a subfamily of E2F transcription factor family that play essential roles in cell-cycle progression, DNA replication, DNA repair, apoptosis, and differentiation. Although the transcriptional regulation of E2Fs has focused on pocket protein retinoblastoma protein complex, recent studies indicate that post-translational modification and stability regulation of E2Fs play key roles in diverse cellular processes. In this study, we found that FBXO1, a component of S-phase kinase-associated protein 1 (SKP1)-cullin 1-F-box protein (SCF) complex, is an E2Fs binding partner. Furthermore, FBXO1 to E2Fs binding induced K48 ubiquitination and subsequent proteasomal degradation of E2Fs. Binding domain analysis indicated that the Arg (R)/Ile (I) and R/Val (V) motifs, which are located in the dimerization domain of E2Fs, of E2F 1 and 3a and E2F2, respectively, acted as degron motifs (DMs) for FBXO1. Notably, RI/AA or RV/AA mutation in the DMs reduced FBXO1-mediated ubiquitination and prolonged the half-lives of E2Fs. Importantly, the stabilities of E2Fs were affected by phosphorylation of threonine residues located near RI and RV residues of DMs. Phosphorylation prediction database analysis and specific inhibitor analysis revealed that MEK/ERK signaling molecules play key roles in FBXO1/E2Fs' interaction and modulate E2F protein turnover. Moreover, both elevated E2Fs protein levels by knockdown of FBXO1 and decreased E2Fs protein levels by sh-E2F3a delayed G1/S cell cycle transition, resulting in inhibition of cancer cell proliferation. These results demonstrated that FBXO1-E2Fs axis-mediated precise E2Fs stability regulation plays a key role in cell proliferation via G1/S cell cycle transition.


Subject(s)
Mitogen-Activated Protein Kinase Kinases , Neoplasms , E2F Transcription Factors/metabolism , Cell Cycle , Cell Proliferation , Cell Cycle Proteins
5.
Mol Pain ; 19: 17448069221150138, 2023.
Article in English | MEDLINE | ID: mdl-36550597

ABSTRACT

Polysorbate 80 is a non-ionic detergent derived from polyethoxylated sorbitan and oleic acid. It is widely used in pharmaceuticals, foods, and cosmetics as an emulsifier. Nav1.7 is a peripheral sodium channel that is highly expressed in sympathetic and sensory neurons, and it plays a critical role in determining the threshold of action potentials (APs). We found that 10 µg/mL polysorbate 80 either abolished APs or increased the threshold of the APs of dorsal root ganglions. We thus investigated whether polysorbate 80 inhibits Nav1.7 sodium current using a whole-cell patch-clamp recording technique. Polysorbate 80 decreased the Nav1.7 current in a concentration-dependent manner with a half-maximal inhibitory concentration (IC50) of 250.4 µg/mL at a holding potential of -120 mV. However, the IC50 was 1.1 µg/mL at a holding potential of -90 mV and was estimated to be 0.9 µg/mL at the resting potentials of neurons, where most channels are inactivated. The activation rate and the voltage dependency of activation of Nav1.7 were not changed by polysorbate 80. However, polysorbate 80 caused hyperpolarizing shifts in the voltage dependency of the steady-state fast inactivation curve. The blocking of Nav1.7 currents by polysorbate 80 was not reversible at a holding potential of -90 mV but was completely reversible at -120 mV, where the channels were mostly in the closed state. Polysorbate 80 also slowed recovery from inactivation and induced robust use-dependent inhibition, indicating that it is likely to bind to and stabilize the inactivated state. Our results indicate that polysorbate 80 inhibits Nav1.7 current in concentration-, state-, and use-dependent manners when used even below commercial concentrations. This suggests that polysorbate 80 may be helpful in pain medicine as an excipient. In addition, in vitro experiments using polysorbate 80 with neurons should be conducted with caution.


Subject(s)
Neurons , Polysorbates , Polysorbates/pharmacology , Polysorbates/metabolism , Neurons/metabolism , Sodium Channels/metabolism , Membrane Potentials/physiology , Action Potentials , NAV1.7 Voltage-Gated Sodium Channel/metabolism , NAV1.8 Voltage-Gated Sodium Channel/metabolism
6.
Biochem Biophys Res Commun ; 642: 66-74, 2023 01 29.
Article in English | MEDLINE | ID: mdl-36566564

ABSTRACT

p90 Ribosomal S6 kinase 2 (RSK2), a member of mitogen-activated protein kinase regulating cell proliferation and transformation induced by tumor promoters, such as epidermal growth factor, plays a vital role as a signaling hub to modulate cell proliferation, transformation, cell cycle transition, and chromatin remodeling by tumor promoter stimulation such as epidermal growth factor. On the other hand, the RSK2-mediated signaling networks that regulate cancer cell proliferation are unclear. In this study, SKOV3, an ovarian cancer cell that exhibits chemoresistant properties, and TOV-112D cells showed different sensitivities to colony growth in soft agar. Based on the protein profile shown in a previous report, RSK2 knockdown preferentially and significantly suppressed cell proliferation and colony growth. Moreover, RSK2 interacted with AKTs (AKT 1-3) via the N-terminal kinase domain (NTKD) of RSK2, resulting in the phosphorylation of RSK2. The AKT-mediated phosphorylation consensus sequence, RxRxxS/T, on RSK2 NTKD (Thr115) was well conserved in different species. In particular, an in vitro kinase assay showed that NTKD deleted and Thr115Ala mutants of RSK2 abolished AKT1-mediated phosphorylation. In the physiological assay of RSK2 phosphorylation at Thr115 on cell proliferation, AKT1-mediated RSK2 phosphorylation at Thr115 played an essential role in cell proliferation. The re-introduction of RSK2-T115A to RSK2-/- MEF attenuated the EGF-induced G1/S cell cycle transition compared to RSK2-wt introducing RSK2-/- MEFs. This attenuation was observed by EGF stimulations and insulin-like growth factor-1. Overall, these results show that novel wiring of the AKT/RSKs signaling axis plays an important role in cancer cell proliferation by modulating the G1/S cell cycle transition.


Subject(s)
Epidermal Growth Factor , Ovarian Neoplasms , Female , Humans , Epidermal Growth Factor/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Cell Proliferation , Signal Transduction , Phosphorylation , Cell Cycle , Carcinogens
7.
J Korean Phys Soc ; 80(8): 817-851, 2022.
Article in English | MEDLINE | ID: mdl-35261432

ABSTRACT

Nonthermal biocompatible plasma (NBP) sources operating in atmospheric pressure environments and their characteristics can be used for plasma bioscience, medicine, and hygiene applications, especially for COVID-19 and citizen. This review surveyed the various NBP sources, including a plasma jet, micro-DBD (dielectric barrier discharge) and nanosecond discharged plasma. The electron temperatures and the plasma densities, which are produced using dielectric barrier discharged electrode systems, can be characterized as 0.7 ~ 1.8 eV and (3-5) × 1014-15 cm-3, respectively. Herein, we introduce a general schematic view of the plasma ultraviolet photolysis of water molecules for reactive oxygen and nitrogen species (RONS) generation inside biological cells or living tissues, which would be synergistically important with RONS diffusive propagation into cells or tissues. Of the RONS, the hydroxyl radical [OH] and hydrogen peroxide H2O2 species would mainly result in apoptotic cell death with other RONS in plasma bioscience and medicines. The diseased biological protein, cancer, and mutated cells could be treated by using a NBP or plasma activated water (PAW) resulting in their apoptosis for a new paradigm of plasma medicine.

8.
Exp Mol Med ; 54(1): 35-46, 2022 01.
Article in English | MEDLINE | ID: mdl-35022544

ABSTRACT

Extracellular signal-regulated kinase 3 (ERK3) is an atypical member of the mitogen-activated protein kinase (MAPK) family, members of which play essential roles in diverse cellular processes during carcinogenesis, including cell proliferation, differentiation, migration, and invasion. Unlike other MAPKs, ERK3 is an unstable protein with a short half-life. Although deubiquitination of ERK3 has been suggested to regulate the activity, its ubiquitination has not been described in the literature. Here, we report that FBXW7 (F-box and WD repeat domain-containing 7) acts as a ubiquitination E3 ligase for ERK3. Mammalian two-hybrid assay and immunoprecipitation results demonstrated that ERK3 is a novel binding partner of FBXW7. Furthermore, complex formation between ERK3 and the S-phase kinase-associated protein 1 (SKP1)-cullin 1-F-box protein (SCF) E3 ligase resulted in the destabilization of ERK3 via a ubiquitination-mediated proteasomal degradation pathway, and FBXW7 depletion restored ERK3 protein levels by inhibiting this ubiquitination. The interaction between ERK3 and FBXW7 was driven by binding between the C34D of ERK3, especially at Thr417 and Thr421, and the WD40 domain of FBXW7. A double mutant of ERK3 (Thr417 and Thr421 to alanine) abrogated FBXW7-mediated ubiquitination. Importantly, ERK3 knockdown inhibited the proliferation of lung cancer cells by regulating the G1/S-phase transition of the cell cycle. These results show that FBXW7-mediated ERK3 destabilization suppresses lung cancer cell proliferation in vitro.


Subject(s)
Lung Neoplasms , Mitogen-Activated Protein Kinase 6 , Animals , Cell Proliferation , F-Box-WD Repeat-Containing Protein 7/genetics , F-Box-WD Repeat-Containing Protein 7/metabolism , Lung Neoplasms/genetics , Mammals/metabolism , Mitogen-Activated Protein Kinase 6/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
9.
Arch Pharm Res ; 44(12): 1091-1108, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34750753

ABSTRACT

Resolution to chemoresistance is a major challenge in patients with advanced-stage malignancies. Thus, identification of action points and elucidation of molecular mechanisms for chemoresist human cancer are necessary to overcome this challenge. In this study, we provide important evidence that kaempferol targeting RSKs might be a strategy to reduce the oxaliplatin-resistant colon cancer cells. We found that MAPK and PI3K-AKT signaling were increased in oxaliplatin (Ox)-resistant HCT116 (HCT116-OxR) cells compared to Ox-sensitive HCT116 (HCT116-OxS) cells. Comparison of cell sensitivities using SP600125 (JNK inhibitor), SB206580 (p38 kinase inhibitor), or MK-2206 (AKT inhibitor) revealed that cell proliferation inhibition was strongly observed in HT29 cells compared to that in HCT116 cells in both OxS and OxR cells. Interestingly, SP600125, SB206580, and MK-2206 treatment showed higher cell proliferation inhibition in OxS cells than that in OxR cells in both HCT116 and HT29 cells, except following treatments with 10 µM of SP600125, and 30 µM of SB206580. In comparison to magnolin and aschantin, kaempferol showed the strongest inhibitory effect on cell proliferation in both HCT116 and HT29 cells. Importantly, HCT116- and HT29-OxR cells showed higher sensitivities to cell proliferation inhibition than those of HCT116- and HT29-OxS cells, resulting in the accumulation of cells at the G2/M-phases of the cell cycle. Finally, we showed that AP-1 transactivation activity was markedly decreased by kaempferol in HCT116- and HT29-OxR cells compared to the activity levels in HCT116- and HT29-OxS cells. Taken together, the results demonstrate that kaempferol-mediated AP-1 inhibition might be an important signaling mechanism to resolve the chemoresistance of Ox-resistant colon cancer cells.


Subject(s)
Colonic Neoplasms/drug therapy , Kaempferols/pharmacology , Oxaliplatin/pharmacology , Transcription Factor AP-1/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Benzodioxoles/pharmacology , Cell Cycle/drug effects , Cell Proliferation/drug effects , Colonic Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , HCT116 Cells , HEK293 Cells , HT29 Cells , Humans , Lignans/pharmacology , Signal Transduction/drug effects
10.
Neuroreport ; 32(16): 1299-1306, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34605450

ABSTRACT

OBJECTIVES: Iloperidone is an atypical antipsychotic drug that is widely used for the treatment of schizophrenia. hERG 3.1, alternatively spliced form of hERG 1A, is considered a potential target for an antipsychotic drug. The present study was designed to study the effects of iloperidone on hERG 1A/3.1 heterotetrameric channels. METHODS: The interactions of iloperidone with hERG 1A/3.1 heterotetrameric channels stably expressed in HEK cells were investigated using the whole-cell patch-clamp technique and western blot analysis. RESULTS: Iloperidone inhibited the hERG 1A/3.1 tail currents at -50 mV in a concentration-dependent manner with an IC50 value of 0.44 µM. The block of hERG 1A/3.1 currents by iloperidone was voltage-dependent and increased over a range of voltage for channel activation. However, the block by iloperidone was voltage-independent at more depolarized potentials where the channels were fully activated. A fast application of iloperidone inhibited the hERG 1A/3.1 current elicited by a 5-s depolarizing pulse to +60 mV to fully inactivate the hERG 1A/3.1 currents. Iloperidone also induced a rapid and reversible inhibition of hERG 1A/3.1 tail currents during repolarization. However, iloperidone had no effect on either hERG 1A or hERG 1A/3.1 channel trafficking to the cell membrane. CONCLUSIONS: Our results indicated that iloperidone concentration-dependently inhibited hERG 1A/3.1 currents by preferentially interacting with the open states of channels, but not by the disruption of membrane trafficking or surface membrane expression of hERG 1A and hERG 1A/3.1 channel proteins.


Subject(s)
Ether-A-Go-Go Potassium Channels/metabolism , Isoxazoles/pharmacology , Piperidines/pharmacology , Potassium Channel Blockers/pharmacology , Biotinylation/drug effects , HEK293 Cells , Humans
11.
Neurosci Lett ; 741: 135446, 2021 01 10.
Article in English | MEDLINE | ID: mdl-33166641

ABSTRACT

Peripheral neuropathy is associated with enhanced activity of primary afferents which is often manifested as pain. Voltage-gated sodium channels (VGSCs) are critical for the initiation and propagation of action potentials and are thus essential for the transmission of the noxious stimuli from the periphery. Human peripheral sensory neurons express multiple VGSCs, including Nav1.7, Nav1.8, and Nav1.9 that are almost exclusively expressed in the peripheral nervous system. Distinct biophysical properties of Nav1.7, Nav1.8, and Nav1.9 underlie their differential contributions to finely tuned neuronal firing of nociceptors, and mutations in these channels have been associated with several inherited human pain disorders. Functional characterization of these mutations has provided additional insights into the role of these channels in electrogenesis in nociceptive neurons and pain sensation. Peripheral tissue damage activates an inflammatory response and triggers generation and release of inflammatory mediators, which can act through diverse signaling cascades to modulate expression and activity of ion channels including VGSCs, contributing to the development and maintenance of pathological pain conditions. In this review, we discuss signaling pathways that are activated by pro-nociceptive inflammatory mediators that regulate peripheral sodium channels, with a specific focus on direct phosphorylation of these channels by multiple protein kinases.


Subject(s)
Cytokines/metabolism , Neurons/metabolism , Peripheral Nervous System Diseases/metabolism , Protein Kinases/metabolism , Voltage-Gated Sodium Channels/metabolism , Action Potentials , Animals , Humans , Inflammation/complications , Inflammation/metabolism , Mice , Pain/complications , Pain/metabolism , Signal Transduction
12.
Korean J Physiol Pharmacol ; 24(6): 545-553, 2020 Nov 01.
Article in English | MEDLINE | ID: mdl-33093275

ABSTRACT

Aripiprazole is a quinolinone derivative approved as an atypical antipsychotic drug for the treatment of schizophrenia and bipolar disorder. It acts as with partial agonist activities at the dopamine D2 receptors. Although it is known to be relatively safe for patients with cardiac ailments, less is known about the effect of aripiprazole on voltage-gated ion channels such as transient A-type K+ channels, which are important for the repolarization of cardiac and neuronal action potentials. Here, we investigated the effects of aripiprazole on Kv1.4 currents expressed in HEK293 cells using a whole-cell patch-clamp technique. Aripiprazole blocked Kv1.4 channels in a concentration-dependent manner with an IC50 value of 4.4 µM and a Hill coefficient of 2.5. Aripiprazole also accelerated the activation (time-to-peak) and inactivation kinetics. Aripiprazole induced a voltage-dependent (δ = 0.17) inhibition, which was use-dependent with successive pulses on Kv1.4 currents without altering the time course of recovery from inactivation. Dehydroaripiprazole, an active metabolite of aripiprazole, inhibited Kv1.4 with an IC50 value of 6.3 µM (p < 0.05 compared with aripiprazole) with a Hill coefficient of 2.0. Furthermore, aripiprazole inhibited Kv4.3 currents to a similar extent in a concentration-dependent manner with an IC50 value of 4.9 µM and a Hill coefficient of 2.3. Thus, our results indicate that aripiprazole blocked Kv1.4 by preferentially binding to the open state of the channels.

13.
Eur J Pharmacol ; 885: 173532, 2020 Oct 15.
Article in English | MEDLINE | ID: mdl-32882214

ABSTRACT

Quetiapine, an atypical antipsychotic drug, is used for the treatment of schizophrenia and acute mania. Although a previous report showed that quetiapine blocked hERG potassium current, quetiapine has been considered relatively safe in terms of cardiovascular side effects. In the present study, we used the whole-cell patch-clamp technique to investigate the effect that quetiapine and its major metabolite norquetiapine can exert on human cardiac sodium channels (hNav1.5). The half-maximal inhibitory concentrations of quetiapine and norquetiapine at a holding potential of -90 mV near the resting potential of cardiomyocytes were 30 and 6 µM, respectively. Norquetiapine as well as quetiapine was preferentially bound in the inactivated state of the hNav1.5 channel. Norquetiapine slowed the recovery from inactivation of hNav1.5 and consequently induced strong use-dependent inhibition. Our results indicate that norquetiapine blocks hNav1.5 current in concentration-, state- and use-dependent manners, suggesting that the blockade of hNav1.5 current by norquetiapine may shorten the cardiac action potential duration and reduce the risk of QT interval prolongation induced by the inhibition of hERG potassium currents.


Subject(s)
Dibenzothiazepines/pharmacology , Heart/drug effects , Myocardium/metabolism , NAV1.5 Voltage-Gated Sodium Channel/drug effects , Sodium Channel Blockers/pharmacology , Action Potentials/drug effects , HEK293 Cells , Humans , Long QT Syndrome/prevention & control , Membrane Potentials/drug effects , Patch-Clamp Techniques , Quetiapine Fumarate/pharmacology
14.
ACS Omega ; 5(25): 15202-15209, 2020 Jun 30.
Article in English | MEDLINE | ID: mdl-32637793

ABSTRACT

Controlling the size and rigidity of calcium carbonate became possible. HCCs were developed and manufactured by the in situ reaction of carbon dioxide and calcium oxide, which were already preflocculated together with GCC using ionic polymers before the reaction. HCC is deformable under pressure during the papermaking process, and its degree of rigidity can be controlled by adjusting the fraction of calcium oxide. The size of HCC can be further controlled by adjusting shearing force. The more the fraction of calcium oxide, the more rigid the HCC and the smaller the diameter of the HCC. When used in papermaking, HCC increased the tensile strength and bulk of paper simultaneously without lowering other essential paper properties, and its deformable nature under pressure improved paper smoothness. Saving chemical pulp by 10% by replacing it with HCC, which is 3-4 times less expensive than the chemical pulp, was demonstrated successfully without lowering the essential properties of paper. Implementation of HCC in the paper mill may result in saving chemical pulp, drying energy, and production cost. The paper mill may utilize the carbon dioxide from the mill stack after purification for HCC preparation.

15.
Eur J Pharmacol ; 854: 92-100, 2019 Jul 05.
Article in English | MEDLINE | ID: mdl-30954564

ABSTRACT

Cariprazine is a novel atypical antipsychotic drug that is widely used for the treatment of schizophrenia and bipolar mania/mixed disorder. We used the whole-cell patch-clamp technique to investigate the effects of cariprazine on hERG channels that are stably expressed in HEK cells. Cariprazine inhibited the hERG 1A and hERG 1A/3.1 tail currents at -50 mV in a concentration-dependent manner with IC50 values of 4.1 and 12.2 µM, respectively. The block of hERG 1A currents by cariprazine was voltage-dependent, and increased over a range of voltage for channel activation. Cariprazine shifted the steady-state inactivation curve of the hERG 1A currents in a hyperpolarizing direction and produced a use-dependent block. A fast application of cariprazine inhibited the hERG 1A currents elicited by a 5 s depolarizing pulse to +60 mV to fully inactivate the hERG 1A currents. During a repolarizing pulse wherein the hERG 1A current was deactivated slowly, cariprazine rapidly and reversibly blocked the open state of the hERG 1A current. However, cariprazine did not affect hERG 1A and hERG 1A/3.1 channel trafficking to the cell membrane. Our results indicated that cariprazine concentration-dependently inhibited hERG 1A and hERG 1A/3.1 currents by preferentially interacting with the open states of the hERG 1A channel, but not by the disruption of hERG 1A and hERG 1A/3.1 channel protein trafficking. Our study examined cariprazine's mechanism of action provides a biophysical profile that is necessary to assess the potential therapeutic effects of this drug.


Subject(s)
Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Piperazines/pharmacology , Potassium Channel Blockers/pharmacology , Dose-Response Relationship, Drug , Electrophysiological Phenomena/drug effects , Ether-A-Go-Go Potassium Channels/chemistry , Ether-A-Go-Go Potassium Channels/metabolism , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Protein Multimerization/drug effects , Protein Structure, Quaternary
16.
Biomaterials ; 156: 258-273, 2018 02.
Article in English | MEDLINE | ID: mdl-29222974

ABSTRACT

Plasma, formed by ionization of gas molecules or atoms, is the most abundant form of matter and consists of highly reactive physicochemical species. In the physics and chemistry fields, plasma has been extensively studied; however, the exact action mechanisms of plasma on biological systems, including cells and humans, are not well known. Recent evidence suggests that cold atmospheric plasma (CAP), which refers to plasma used in the biomedical field, may regulate diverse cellular processes, including neural differentiation. However, the mechanism by which these physicochemical signals, elicited by reactive oxygen and nitrogen species (RONS), are transmitted to biological system remains elusive. In this study, we elucidated the physicochemical and biological (PCB) connection between the CAP cascade and Trk/Ras/ERK signaling pathway, which resulted in neural differentiation. Excited atomic oxygen in the plasma phase led to the formation of RONS in the PCB network, which then interacted with reactive atoms in the extracellular liquid phase to form nitric oxide (NO). Production of large amounts of superoxide radical (O2-) in the mitochondria of cells exposed to CAP demonstrated that extracellular NO induced the reversible inhibition of mitochondrial complex IV. We also demonstrated that cytosolic hydrogen peroxide, formed by O2- dismutation, act as an intracellular messenger to specifically activate the Trk/Ras/ERK signaling pathway. This study is the first to elucidate the mechanism linking physicochemical signals from the CAP cascade to the intracellular neural differentiation signaling pathway, providing physical, chemical and biological insights into the development of therapeutic techniques to treat neurological diseases.


Subject(s)
Cell Differentiation/drug effects , MAP Kinase Signaling System/drug effects , Neurons/cytology , Plasma Gases/pharmacology , Reactive Nitrogen Species/metabolism , Receptors, Nerve Growth Factor/metabolism , ras Proteins/metabolism , Animals , Catecholamines/metabolism , Cell Line, Tumor , Cytosol/metabolism , Hydrogen Peroxide/metabolism , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurons/drug effects , Neurons/ultrastructure , Nitric Oxide/metabolism , Zebrafish
17.
Neurosci Lett ; 664: 66-73, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29133173

ABSTRACT

Quetiapine is an atypical antipsychotic drug that is widely used for the treatment of schizophrenia. It is mainly metabolized by a cytochrome P450 system in the liver. Norquetiapine is a major active metabolite in humans with a pharmacological profile that differs distinctly from that of quetiapine. We used the whole-cell patch-clamp technique to investigate the effects of norquetiapine on hERG channels that are stably expressed in HEK cells. Quetiapine and norquetiapine inhibited the hERG tail currents at -50mV in a concentration-dependent manner with IC50 values of 8.3 and 10.8µM, respectively, which suggested equal potency. The block of hERG currents by norquetiapine was voltage-dependent with a steep increase over a range of voltages for channel activation. However, at more depolarized potentials where the channels were fully activated, the block by norquetiapine was voltage-independent. The steady-state inactivation curve of the hERG currents was shifted to the hyperpolarizing direction in the presence of norquetiapine. Norquetiapine did not produce a use-dependent block. A fast application of norquetiapine inhibited the hERG current elicited by a 5s depolarizing pulse to +60mV, which fully inactivated the hERG currents, suggesting an inactivated-state block. During a repolarizing pulse wherein the hERG current was slowly deactivated, albeit remaining in an open state, a fast application of norquetiapine rapidly and reversibly inhibited the open state of the hERG current. Our results indicated that quetiapine and norquetiapine had equal potency in inhibiting hERG tail currents. Norquetiapine inhibited the hERG current by preferentially interacting with the open and/or inactivated states of the channels.


Subject(s)
Cloning, Molecular , Dibenzothiazepines/pharmacology , ERG1 Potassium Channel/antagonists & inhibitors , ERG1 Potassium Channel/physiology , Quetiapine Fumarate/pharmacology , Antidepressive Agents/metabolism , Antidepressive Agents/pharmacology , Dibenzothiazepines/metabolism , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Quetiapine Fumarate/metabolism
18.
J Cell Physiol ; 233(1): 549-558, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28322444

ABSTRACT

The potassium ion channel Kv3.1b is a member of a family of voltage-gated ion channels that are glycosylated in their mature form. In the present study, we demonstrate the impact of N-glycosylation at specific asparagine residues on the trafficking of the Kv3.1b protein. Large quantities of asparagine 229 (N229)-glycosylated Kv3.1b reached the plasma membrane, whereas N220-glycosylated and unglycosylated Kv3.1b were mainly retained in the endoplasmic reticulum (ER). These ER-retained Kv3.1b proteins were susceptible to degradation, when co-expressed with calnexin, whereas Kv3.1b pools located at the plasma membrane were resistant. Mass spectrometry analysis revealed a complex type Hex3 HexNAc4 Fuc1 glycan as the major glycan component of the N229-glycosylated Kv3.1b protein, as opposed to a high-mannose type Man8 GlcNAc2 glycan for N220-glycosylated Kv3.1b. Taken together, these results suggest that trafficking-dependent roles of the Kv3.1b potassium channel are dependent on N229 site-specific glycosylation and N-glycan structure, and operate through a mechanism whereby specific N-glycan structures regulate cell surface expression.


Subject(s)
Nerve Tissue Proteins/metabolism , Protein Processing, Post-Translational , Shaw Potassium Channels/metabolism , Animals , Asparagine , COS Cells , Cell Membrane/metabolism , Chlorocebus aethiops , Endoplasmic Reticulum/metabolism , Glycosylation , Mutation , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Protein Conformation , Protein Transport , Rats , Shaw Potassium Channels/chemistry , Shaw Potassium Channels/genetics , Structure-Activity Relationship , Transfection
19.
Sci Rep ; 7(1): 13978, 2017 10 25.
Article in English | MEDLINE | ID: mdl-29070885

ABSTRACT

Odorants are non-nutrients. However, they exist abundantly in foods, wines, and teas, and thus can be ingested along with the other nutrients during a meal. Here, we have focused on the chemical-recognition ability of these ORs and hypothesized that the odorants ingested during a meal may play a physiological role by activating the gut-expressed ORs. Using a human-derived enteroendocrine L cell line, we discovered the geraniol- and citronellal-mediated stimulation of glucagon-like peptide-1 (GLP-1) secretion and elucidated the corresponding cellular downstream signaling pathways. The geraniol-stimulated GLP-1 secretion event in the enteroendocrine cell line was mediated by the olfactory-type G protein, the activation of adenylyl cyclase, increased intracellular cAMP levels, and extracellular calcium influx. TaqMan qPCR demonstrated that two ORs corresponding to geraniol and citronellal were expressed in the human enteroendocrine cell line and in mouse intestinal specimen. In a type 2 diabetes mellitus mouse model (db/db), oral administration of geraniol improved glucose homeostasis by increasing plasma GLP-1 and insulin levels. This insulinotropic action of geraniol was GLP-1 receptor-mediated, and also was glucose-dependent. This study demonstrates that odor compounds can be recognized by gut-expressed ORs during meal ingestion and therefore, participate in the glucose homeostasis by inducing the secretion of gut-peptides.


Subject(s)
Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 2/complications , Enteroendocrine Cells/metabolism , Glucagon-Like Peptide 1/metabolism , Hyperglycemia/prevention & control , Intestinal Mucosa/metabolism , Receptors, Odorant/metabolism , Animals , Blood Glucose/metabolism , Hyperglycemia/etiology , Hyperglycemia/metabolism , Male , Mice , Mice, Inbred C57BL , Receptors, Odorant/genetics
20.
Naunyn Schmiedebergs Arch Pharmacol ; 390(6): 633-642, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28265686

ABSTRACT

Risperidone and one of its active metabolites, paliperidone, are widely used for the treatment of schizophrenia. We used a patch-clamp study to investigate the effects of paliperidone on hERG potassium channels expressed in HEK cells. Western blot analyses were used to study the effects of risperidone and paliperidone on hERG and hERG 3.1 isoform channel trafficking. Risperidone and paliperidone inhibited the hERG tail currents in a concentration-dependent manner with IC50 values of 0.16 and 0.57 µM, respectively. The block of hERG currents by paliperidone was voltage-dependent, increasing over a range of voltages for channel activation. A fast application of paliperidone inhibited the hERG current elicited by a 5-s depolarizing pulse to +60 mV to fully inactivate the hERG currents, suggesting an inactivated state block. A fast application of paliperidone during repolarization reversibly inhibited the hERG tail currents in a concentration-dependent manner with a IC50 value of 1.26 µM. Kinetic analysis of paliperidone interaction with the open state of the hERG channels showed that the rate constants of association (k +1) and dissociation (k -1) for paliperidone were 0.45 µM-1 s-1 and 1.07 s-1, respectively. Paliperidone shifted the steady-state inactivation curve of the hERG currents in a hyperpolarizing direction and also produced a use-dependent block. Risperidone and paliperidone had no effect on hERG and hERG 3.1 channel trafficking to the cell membrane. Our results indicated that paliperidone inhibited the hERG current by preferentially interacting with the open and inactivated states of the channel, but not by disruption of hERG channel protein trafficking.


Subject(s)
Antipsychotic Agents/pharmacology , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Paliperidone Palmitate/pharmacology , Risperidone/pharmacology , Antipsychotic Agents/administration & dosage , Cell Membrane/drug effects , Cell Membrane/metabolism , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Inhibitory Concentration 50 , Paliperidone Palmitate/administration & dosage , Patch-Clamp Techniques , Risperidone/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...