Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
Gan To Kagaku Ryoho ; 50(9): 993-996, 2023 Sep.
Article in Japanese | MEDLINE | ID: mdl-37800295

ABSTRACT

The implantation of a totally implantable central venous(CV)access port is considered a risk factor for venous thromboembolism( VTE). In the treatment of catheter-related thrombosis(CRT), both European and American guidelines recommend anticoagulation therapy with catheters in place. We experienced 2 cases of upper extremity deep vein thrombosis (UEDVT)after the implantation of CV access ports through the left subclavian vein for adjuvant chemotherapy in patients with resected breast cancer. Both patients were successfully treated with direct oral anticoagulants(DOAC) while the port remained in place with a careful follow-up that included monitoring of serum D-dimer levels. The administration of DOAC to CRT that develops in patients undergoing postoperative adjuvant chemotherapy for breast cancer may be relatively safe, with a low potential for adverse events such as bleeding.


Subject(s)
Breast Neoplasms , Central Venous Catheters , Upper Extremity Deep Vein Thrombosis , Venous Thromboembolism , Venous Thrombosis , Humans , Female , Venous Thrombosis/drug therapy , Venous Thrombosis/etiology , Upper Extremity Deep Vein Thrombosis/drug therapy , Upper Extremity Deep Vein Thrombosis/etiology , Central Venous Catheters/adverse effects , Venous Thromboembolism/drug therapy , Breast Neoplasms/drug therapy , Anticoagulants/adverse effects
2.
Gan To Kagaku Ryoho ; 50(12): 1339-1341, 2023 Dec.
Article in Japanese | MEDLINE | ID: mdl-38247077

ABSTRACT

An 86-year-old woman was referred to our hospital after an incidental CT scan of the trunk revealed a mass in the left breast and enlarged axillary lymph nodes. A core needle biopsy(CNB)from a 2 cm mass in the left breast revealed invasive ductal carcinoma, weakly positive result for ER, negative result for PgR, and negative result for HER2. She also had multiple enlarged left supraclavicular lymph nodes and was T2N3cM0, Stage ⅢC on pretreatment evaluation. She was given the S-1 oral drug of choice, starting with 80 mg/day/4-week dosing with a 2-week rest. Eight months after the start of S-1, a partial mastectomy and sentinel lymph node biopsy were performed. Pathological findings showed a pathological complete response(ypTis/ypN0)with only a 2 mm non-invasive carcinoma remnant in the left mammary gland. S-1 is weakly recommended as primary chemotherapy for HER2-negative metastatic recurrent breast cancer, but there are no reports to date of complete response in resection cases. S-1 may be administered to patients with locally advanced breast cancer who cannot tolerate standard drug therapy and may be converted to resection after a successful response.


Subject(s)
Breast Neoplasms , Lymphadenopathy , Aged , Female , Humans , Aged, 80 and over , Breast Neoplasms/drug therapy , Breast Neoplasms/surgery , Mastectomy , Neoadjuvant Therapy , Axilla , Pathologic Complete Response
3.
Neurosci Bull ; 38(2): 135-148, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34542799

ABSTRACT

The family of voltage-gated potassium Kv2 channels consists of the Kv2.1 and Kv2.2 subtypes. Kv2.1 is constitutively highly phosphorylated in neurons and its function relies on its phosphorylation state. Whether the function of Kv2.2 is also dependent on its phosphorylation state remains unknown. Here, we investigated whether Kv2.2 channels can be phosphorylated by protein kinase C (PKC) and examined the effects of PKC-induced phosphorylation on their activity and function. Activation of PKC inhibited Kv2.2 currents and altered their steady-state activation in HEK293 cells. Point mutations and specific antibodies against phosphorylated S481 or S488 demonstrated the importance of these residues for the PKC-dependent modulation of Kv2.2. In layer II pyramidal neurons in cortical slices, activation of PKC similarly regulated native Kv2.2 channels and simultaneously reduced the frequency of action potentials. In conclusion, this study provides the first evidence to our knowledge that PKC-induced phosphorylation of the Kv2.2 channel controls the excitability of cortical pyramidal neurons.


Subject(s)
Protein Kinase C , Pyramidal Cells/enzymology , Shab Potassium Channels , Action Potentials , HEK293 Cells , Humans , Protein Kinase C/metabolism , Shab Potassium Channels/genetics
4.
Neuroscience Bulletin ; (6): 135-148, 2022.
Article in English | WPRIM (Western Pacific) | ID: wpr-922667

ABSTRACT

The family of voltage-gated potassium Kv2 channels consists of the Kv2.1 and Kv2.2 subtypes. Kv2.1 is constitutively highly phosphorylated in neurons and its function relies on its phosphorylation state. Whether the function of Kv2.2 is also dependent on its phosphorylation state remains unknown. Here, we investigated whether Kv2.2 channels can be phosphorylated by protein kinase C (PKC) and examined the effects of PKC-induced phosphorylation on their activity and function. Activation of PKC inhibited Kv2.2 currents and altered their steady-state activation in HEK293 cells. Point mutations and specific antibodies against phosphorylated S481 or S488 demonstrated the importance of these residues for the PKC-dependent modulation of Kv2.2. In layer II pyramidal neurons in cortical slices, activation of PKC similarly regulated native Kv2.2 channels and simultaneously reduced the frequency of action potentials. In conclusion, this study provides the first evidence to our knowledge that PKC-induced phosphorylation of the Kv2.2 channel controls the excitability of cortical pyramidal neurons.


Subject(s)
Humans , Action Potentials , HEK293 Cells , Protein Kinase C/metabolism , Pyramidal Cells/enzymology , Shab Potassium Channels/genetics
5.
Neurobiol Dis ; 156: 105407, 2021 08.
Article in English | MEDLINE | ID: mdl-34058347

ABSTRACT

Stroke is one of the leading causes of death worldwide, with intracerebral hemorrhage (ICH) being the most lethal subtype. Neuritin (Nrn) is a neurotropic factor that has been reported to have neuroprotective effects in acute brain and spinal cord injury. However, whether Nrn has a protective role in ICH has not been investigated. In this study, ICH was induced in C57BL/6 J mice by injection of collagenase VII, while the overexpression of Nrn in the striatum was induced by an adeno-associated virus serotype 9 (AAV9) vector. We found that compared with GFP-ICH mice, Nrn-ICH mice showed improved performance in the corner, cylinder and forelimb tests after ICH, and showed less weight loss and more rapid weight recovery. Overexpression of Nrn reduced brain lesions, edema, neuronal death and white matter and synaptic integrity dysfunction caused by ICH. Western blot results showed that phosphorylated PERK and ATF4 were significantly inhibited, while phosphorylation of Akt/mammalian target of rapamycin was increased in the Nrn-ICH group, compared with the GFP-ICH group. Whole cell recording from motor neurons indicated that overexpression of Nrn reversed the decrease of spontaneous excitatory postsynaptic currents (sEPSCs) and action potential frequencies induced by ICH. These data show that Nrn improves neurological deficits in mice with ICH by reducing brain lesions and edema, inhibiting neuronal death, and possibly by increasing neuronal connections.


Subject(s)
Brain/metabolism , Cerebral Hemorrhage/metabolism , Nerve Tissue Proteins/biosynthesis , Recovery of Function/physiology , Adenine/administration & dosage , Adenine/analogs & derivatives , Animals , Brain/drug effects , Brain/pathology , Cerebral Hemorrhage/pathology , Dependovirus/genetics , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Furans/administration & dosage , GPI-Linked Proteins/biosynthesis , GPI-Linked Proteins/genetics , Indoles/administration & dosage , Injections, Intraventricular , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Organ Culture Techniques , Pyridines/administration & dosage , Pyrimidines/administration & dosage , Recovery of Function/drug effects
6.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-885162

ABSTRACT

Objective:To explore the associations of urinary retinol binding protein (RBP) and β 2-microglobulin (β 2-MG) with urinary albumin to creatinine ratio (UACR) and renal function in hospitalized patients with type 2 diabetes mellitus (T2DM). Methods:A total of 1 030 Chinese patients with T2DM were included in this study. The subjects were divided into the UACR normal group (<30 mg/g), microalbuminuria group (30-300 mg/g) and macroalbuminuria group (>300 mg/g). Patients with normal UACR were further divided into two groups according to the estimated glomerular filtration rate (eGFR): the eGFR low group (<90 ml·min -1·1.73m -2) and the normal eGFR group (≥90 ml·min -1·1.73m -2). Urine RBP and β 2-MG levels among the groups were compared. Multiple linear regression analyses were applied to evaluate risk factors of urine RBP and β 2-MG. Results:In all patients ( n=1 030), urine RBP and β 2-MG increased gradually with the increase of UACR across the three groups, the proportions of abnormal urine RBP (>0.7 mg/L) and β 2-MG (>370 μg/L) in these groups were 3.8%, 8.5%, 39.0% ( P<0.001), and 12.9%, 26.7%, 46.8% ( P<0.001), respectively. In the UACR normal group ( n=788), 12.2% of the patients were with eGFR<90 ml·min -1·1.73m -2. The proportion of abnormal β 2-MG (>370 μg/L) was higher in the eGFR low group than that in the eGFR normal group (29.2% vs. 10.7%, P<0.001). Multivariate linear stepwise regression analyses were performed using natural logarithm of urine RBP or β 2-MG as dependent variable, and showed that urine RBP was independently associated with UACR ( β=0.0005, P<0.001), serum creatinine ( β=0.006, P<0.001) and glycosylated hemoglobin A1c ( β=0.050, P=0.001), and β 2-MG was independently correlated with UACR ( β=0.000 4, P<0.001), serum creatinine ( β=0.011, P<0.001), systolic blood pressure ( β=0.005, P=0.031) and fasting blood-glucose ( β=0.027, P=0.046). Conclusions:Urine RBP and β 2-MG are positively associated with high UACR and impaired renal function in T2DM patients, and these changes could occur before UACR and eGFR turned out to be abnormal. It is recommended that urine RBP and β 2-MG be detected as early as possible to identify diabetic kidney disease in patients with normal UACR and eGFR.

7.
Chemosphere ; 252: 126640, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32443282

ABSTRACT

This work focuses on the bioaccumulation and toxic effects of di-(2-ethylhexyl) phthalate (DEHP) in the leafy vegetable Shanghaiqing (SHQ) (Brassica chinensis L.). The accumulated DEHP amount in the edible part and roots of SHQ increased as the DEHP concentration in the soil increased. DEHP accumulation was higher in the roots than in the edible part of the plant. The root concentration factors and bioaccumulation factors for DEHP in SHQ were 0.13-2.49 and 0.03-2.00, respectively. The DEHP translocation factors were below 1.0, indicating that DEHP preferentially accumulated in plant roots. The DEHP risk index in the edible part of SHQ in relation to the human body and in terms of dietary exposure risk assessment was also below 1.0, indicating a low health risk. High DEHP concentrations caused 1) inhibition of SHQ growth, 2) an increase in SHQ chlorophyll and malondialdehyde contents and 3) a decrease in soluble sugar and vitamin contents. Low DEHP concentrations stimulated total superoxide dismutase, peroxidase and catalase activities, while high DEHP levels showed an inhibitory effect. DEHP presence in soil affected not only SHQ growth but also quality. Our results provide the data needed for the proper assessment of food safety and the ecological impact of DEHP contamination in agricultural soils.


Subject(s)
Brassica/metabolism , Diethylhexyl Phthalate/metabolism , Soil Pollutants/metabolism , Agriculture , Brassica/growth & development , Diethylhexyl Phthalate/toxicity , Malondialdehyde , Phthalic Acids , Plant Leaves/chemistry , Soil , Soil Pollutants/analysis , Superoxide Dismutase , Vegetables/drug effects
8.
Mol Imaging Biol ; 21(3): 529-537, 2019 06.
Article in English | MEDLINE | ID: mdl-30112727

ABSTRACT

PURPOSE: Although a preparation method for F-18-labeled proteins that used a cell-free translation system and 4-[18F]fluoro-L-proline instead of L-proline has been reported, its introduction depends on amino acid sequences of target proteins. The purpose of the study was to propose site-specific labeling method of F-18 by using cell-free translation systems supplemented with an engineered orthogonal aminoacyl-tRNA synthetase derived from Methanocaldococcus jannaschii (pCNF-RS)/suppressor tRNA (tRNACUAopt) pair, O-2-[18F]fluoroethyl-L-tyrosine ([18F]FET), and template DNA inserted with an amber codon. PROCEDURES: [18F]FET was prepared from the corresponding precursor and determined whether [18F]FET could be incorporated into an affibody molecule for human epidermal growth factor receptor type 2 (HER2; ZHER2:342) as the 21st amino acid used with the pCNF-RS-tRNACUAopt pair and template DNA inserted with an amber codon in a cell-free translation system. Using SKOV-3 cells, we performed an in vitro binding assay of [18F]FET-ZHER2:342. Furthermore, in vivo positron emission tomography (PET) imaging in SKOV-3 xenograft-bearing mice was performed after the intravenous administration of [18F]FET-ZHER2:342. RESULTS: [18F]FET was successfully incorporated into proteins by using commercially available cell-free protein synthesis reagents with a pCNF-RS-tRNACUAopt pair and template DNA of the desired proteins inserted with an amber codon. The mean radiochemical yield (non-decay-corrected) of [18F]FET-ZHER2:342 was 6.5 ± 4.1 %. An in vitro cell binding assay revealed that SKOV-3 cells-bound [18F]FET-ZHER2:342 expressed HER2. The in vivo PET imaging in SKOV-3 xenograft-bearing mice revealed that [18F]FET-ZHER2:342 accumulated in SKOV-3 xenografts. CONCLUSION: The method proposed in this study might be useful for preparing proteins with F-18 and molecular imaging in the preclinical development.


Subject(s)
Fluorine Radioisotopes/chemistry , Protein Biosynthesis , Proteins/metabolism , Recombinant Fusion Proteins/metabolism , Staining and Labeling , Tyrosine/analogs & derivatives , Amino Acid Sequence , Amino Acyl-tRNA Synthetases/metabolism , Animals , Cell Line, Tumor , Cell-Free System , Female , HEK293 Cells , Humans , Interleukin-8/metabolism , Mice, SCID , Proteins/chemistry , RNA, Transfer/metabolism , Tyrosine/chemistry , Xenograft Model Antitumor Assays
9.
J Neurochem ; 147(1): 40-57, 2018 10.
Article in English | MEDLINE | ID: mdl-29920676

ABSTRACT

Neuritin is a neurotrophic factor that is activated by neural activity and neurotrophins. Its major function is to promote neurite growth and branching; however, the underlying mechanisms are not fully understood. To address this issue, this study investigated the effects of neuritin on neurite and spine growth and intracellular Ca2+ concentration in rat cerebellar granule neurons (CGNs). Incubation of CGNs for 24 h with neuritin increased neurite length and spine density; this effect was mimicked by insulin and abolished by inhibiting insulin receptor (IR) or mitogen-activated protein kinase kinase/extracellular signal-regulated kinase (ERK) activity. Calcium imaging and western blot analysis revealed that neuritin enhanced the increase in intracellular Ca2+ level induced by high K+ , and stimulated the cell surface expression of CaV 1.2 and CaV 1.3 α subunits of the L-type calcium channel, which was suppressed by inhibition of IR or mitogen-activated protein kinase kinase/ERK. Treatment with inhibitors of L-type calcium channels, calmodulin, and calcineurin (CaN) abrogated the effects of neuritin on neurite length and spine density. A similar result was obtained by silencing nuclear factor of activated T cells c4, which is known to be activated by neuritin in CGNs. These results indicate that IR and ERK signaling as well as the Ca2+ /CaN/nuclear factor of activated T cells c4 axis mediate the effects of neuritin on neurite and spine growth in CGNs. OPEN PRACTICES: Open Science: This manuscript was awarded with the Open Materials Badge. For more information see: https://cos.io/our-services/open-science-badges/ Cover Image for this issue: doi: 10.1111/jnc.14195.


Subject(s)
Calcium Channels, L-Type/drug effects , Calcium Signaling/drug effects , Cerebellum/cytology , Dendritic Spines/drug effects , Neurites/drug effects , Neuropeptides/pharmacology , Animals , Calcium Channels/metabolism , Calcium Channels, L-Type/metabolism , Cerebellum/drug effects , Cerebellum/growth & development , Cytoplasmic Granules/drug effects , Female , GPI-Linked Proteins/pharmacology , Gene Silencing , Humans , Insulin/pharmacology , MAP Kinase Signaling System/drug effects , NFATC Transcription Factors/antagonists & inhibitors , NFATC Transcription Factors/genetics , Rats , Rats, Sprague-Dawley , Receptor, Insulin/antagonists & inhibitors
10.
Tohoku J Exp Med ; 245(1): 13-19, 2018 05.
Article in English | MEDLINE | ID: mdl-29731479

ABSTRACT

Positron emission mammography (PEM) has higher detection sensitivity for breast cancer compared with whole-body positron emission tomography (PET) due to higher spatial resolution. We have developed a new PEM device with high resolution over a wide field of view. This PEM device comprises novel scintillation crystals, praseodymium-doped lutetium aluminum garnet (Pr:LuAG). In the present study, the clinical use of the newly developed PEM for the detection of small breast cancer was compared with that of the conventional PET-computed tomography (PET/CT). Eighty-two patients with breast cancer less than 20 mm (UICC T1) participated in this study, including 23 patients with T1a or T1b breast cancer (less than 10 mm). Histologically-proved lesions were examined by PET/CT and PEM on the same day after injection of [18F]fluoro-2-deoxy-2-fluoro-D-glucose ([18F]FDG), a marker of glycolytic activity. The newly developed PEM showed better sensitivity of cancer detection compared with PET/CT especially in case of the small T1a or T1b lesions. Moreover, when the conventional PET/CT and new PEM were combined, the detection sensitivity with [18F]FDG molecular imaging for T1 (N = 82) and T1a plus T1b breast cancer (N = 23) were 90% and 70%, respectively. The uptake of [18F]FDG was proportional to the histological malignancy of breast cancer. Using the newly-developed PEM with [18F]FDG, we are able to identify and characterize exactly the small breast tumors less than 10 mm in combination with the conventional PET/CT. These data indicate that PEM and PET/CT are synergic and complementary for the detection of small breast cancer.


Subject(s)
Breast Neoplasms/diagnostic imaging , Breast Neoplasms/diagnosis , Mammography , Positron-Emission Tomography , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/diagnosis , Carcinoma, Ductal, Breast/diagnostic imaging , Female , Fluorodeoxyglucose F18/pharmacokinetics , Humans , Image Processing, Computer-Assisted , Middle Aged , Positron Emission Tomography Computed Tomography
11.
Neurosci Bull ; 34(5): 759-768, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29671217

ABSTRACT

Cyproheptadine (CPH), a first-generation antihistamine, enhances the delayed rectifier outward K+ current (IK) in mouse cortical neurons through a sigma-1 receptor-mediated protein kinase A pathway. In this study, we aimed to determine the effects of CPH on neuronal excitability in current-clamped pyramidal neurons in mouse medial prefrontal cortex slices. CPH (10 µmol/L) significantly reduced the current density required to generate action potentials (APs) and increased the instantaneous frequency evoked by a depolarizing current. CPH also depolarized the resting membrane potential (RMP), decreased the delay time to elicit an AP, and reduced the spike threshold potential. This effect of CPH was mimicked by a sigma-1 receptor agonist and eliminated by an antagonist. Application of tetraethylammonium (TEA) to block IK channels hyperpolarized the RMP and reduced the instantaneous frequency of APs. TEA eliminated the effects of CPH on AP frequency and delay time, but had no effect on spike threshold or RMP. The current-voltage relationship showed that CPH increased the membrane depolarization in response to positive current pulses and hyperpolarization in response to negative current pulses, suggesting that other types of membrane ion channels might also be affected by CPH. These results suggest that CPH increases the excitability of medial prefrontal cortex neurons by regulating TEA-sensitive IK channels as well as other TEA-insensitive K+ channels, probably ID and inward-rectifier Kir channels. This effect of CPH may explain its apparent clinical efficacy as an antidepressant and antipsychotic.


Subject(s)
Cyproheptadine/pharmacology , Histamine H1 Antagonists/pharmacology , Prefrontal Cortex/drug effects , Pyramidal Cells/drug effects , Animals , Female , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice, Inbred C57BL , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Potassium Channels/metabolism , Prefrontal Cortex/physiology , Pyramidal Cells/physiology , Receptors, sigma/agonists , Receptors, sigma/metabolism , Tetraethylammonium/pharmacology , Tissue Culture Techniques
12.
Acta Pharmacol Sin ; 39(9): 1414-1420, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29595190

ABSTRACT

Neuritin is a member of the neurotrophic factor family, which is activated by neural activity and neurotrophins, and promotes neurite growth and branching. It has shown to play an important role in neuronal plasticity and regeneration. It is also involved in other biological processes such as angiogenesis, tumorigenesis and immunomodulation. Thus far, however, the primary mechanisms of neuritin, including whether or not it acts through a receptor or which downstream signals might be activated following binding, are not fully understood. Recent evidence suggests that neuritin may be a potential therapeutic target in several neurodegenerative diseases. This review focuses on the recent advances in studies regarding the newly identified functions of neuritin and the signaling pathways related to these functions. We also discuss current hot topics and difficulties in neuritin research.


Subject(s)
Neuropeptides/physiology , Signal Transduction/physiology , Animals , GPI-Linked Proteins/physiology , Humans , Mental Disorders/etiology , Mental Disorders/physiopathology , Neurogenesis/physiology , Neuronal Plasticity/physiology , Synapses/physiology
13.
Neuroscience Bulletin ; (6): 759-768, 2018.
Article in English | WPRIM (Western Pacific) | ID: wpr-777023

ABSTRACT

Cyproheptadine (CPH), a first-generation antihistamine, enhances the delayed rectifier outward K current (I) in mouse cortical neurons through a sigma-1 receptor-mediated protein kinase A pathway. In this study, we aimed to determine the effects of CPH on neuronal excitability in current-clamped pyramidal neurons in mouse medial prefrontal cortex slices. CPH (10 µmol/L) significantly reduced the current density required to generate action potentials (APs) and increased the instantaneous frequency evoked by a depolarizing current. CPH also depolarized the resting membrane potential (RMP), decreased the delay time to elicit an AP, and reduced the spike threshold potential. This effect of CPH was mimicked by a sigma-1 receptor agonist and eliminated by an antagonist. Application of tetraethylammonium (TEA) to block I channels hyperpolarized the RMP and reduced the instantaneous frequency of APs. TEA eliminated the effects of CPH on AP frequency and delay time, but had no effect on spike threshold or RMP. The current-voltage relationship showed that CPH increased the membrane depolarization in response to positive current pulses and hyperpolarization in response to negative current pulses, suggesting that other types of membrane ion channels might also be affected by CPH. These results suggest that CPH increases the excitability of medial prefrontal cortex neurons by regulating TEA-sensitive I channels as well as other TEA-insensitive K channels, probably I and inward-rectifier Kir channels. This effect of CPH may explain its apparent clinical efficacy as an antidepressant and antipsychotic.


Subject(s)
Animals , Female , Cyproheptadine , Pharmacology , Histamine H1 Antagonists , Pharmacology , Membrane Potentials , Physiology , Mice, Inbred C57BL , Patch-Clamp Techniques , Potassium Channel Blockers , Pharmacology , Potassium Channels , Metabolism , Prefrontal Cortex , Physiology , Pyramidal Cells , Physiology , Receptors, sigma , Metabolism , Tetraethylammonium , Pharmacology , Tissue Culture Techniques
14.
Cereb Cortex ; 27(7): 3842-3855, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28475719

ABSTRACT

Neuritin is a neurotrophic factor involved in neural development and synaptic plasticity. However, its role in modulating synaptic transmission remains unclear. Here, we investigated the effects of neuritin on miniature excitatory postsynaptic currents (mEPSCs) and glutamate release in the medial prefrontal cortex (mPFC) in mice. Incubation of mPFC slices with neuritin for 45 min significantly increased mEPSC frequency and glutamate release as measured by high-performance liquid chromatography, which was mimicked by insulin and abrogated by an insulin receptor (IR) inhibitor. Neuritin-induced upregulation of synaptic transmission was correlated with activation of ERK, and inhibition of mitogen-activated protein kinases/extracellular signal-regulated kinases (MEK/ERK) activity attenuated the neuritin-induced increase in mEPSC frequency and glutamate release. T-type calcium channel inhibitors but not the L-type inhibitor abolished the inward calcium current and the effects of neuritin on mEPSC frequency and glutamate release. Western blotting of membrane proteins showed that neuritin promoted surface expression of CaV3.3 α-subunit, which was also eliminated by inhibition of IR or MEK/ERK activity. The effects of neuritin on mEPSC frequency, glutamate release, and CaV3.3 α-subunit expression were inhibited by an intracellular protein-transport inhibitor. These results confirm involvement of the IR and ERK signaling pathway, and provide novel insights into the mechanisms of neuritin function in synaptic transmission.


Subject(s)
Calcium Channels, T-Type/metabolism , Gene Expression Regulation/drug effects , Neurons/drug effects , Neuropeptides/pharmacology , Prefrontal Cortex/cytology , Synaptic Transmission/drug effects , Action Potentials/drug effects , Animals , Calcium Channel Blockers/pharmacology , Dose-Response Relationship, Drug , Electric Stimulation , Enzyme Inhibitors/pharmacology , Female , GPI-Linked Proteins/metabolism , GPI-Linked Proteins/pharmacology , Glutamic Acid/metabolism , In Vitro Techniques , Mice , Mice, Inbred C57BL , Neuronal Plasticity , Neuropeptides/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Silver Staining , Synaptic Transmission/physiology , Time Factors , Transduction, Genetic
15.
Pharmacol Ther ; 178: 148-156, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28457804

ABSTRACT

We previously reported on brain H1 receptor occupancy measurements of antihistamines in human brain using [11C]doxepin and positron emission tomography (PET). We proposed the use of brain H1 receptor occupancy to classify antihistamines objectively into three categories of sedating, less-sedating, and non-sedating antihistamines according to their sedative effects. Non-sedating antihistamines are recommended for the treatment of allergies such as pollinosis and atopic dermatitis because of their low penetration into the central nervous system. Physicians and pharmacists are responsible for fully educating patients about the risks of sedating antihistamines from pharmacological points of view. If a sedating antihistamine must be prescribed, its sedative effects should be thoroughly considered before choosing the drug. Non-sedating antihistamines should be preferentially used whenever possible as most antihistamines are equally efficacious, while adverse effects of sedating antihistamines can be serious. This review summarizes the pharmacological properties of clinically useful non-sedating antihistamines from the perspective of histamine function in the CNS.


Subject(s)
Histamine H1 Antagonists, Non-Sedating/pharmacology , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , Animals , Brain/diagnostic imaging , Brain/metabolism , Gene Expression , Genes, MDR , Histamine/metabolism , Humans , Positron-Emission Tomography
16.
Sci Rep ; 7: 44521, 2017 03 17.
Article in English | MEDLINE | ID: mdl-28303965

ABSTRACT

Mounting evidence suggests that exposure to radiofrequency electromagnetic radiation (RF-EMR) can influence learning and memory in rodents. In this study, we examined the effects of single exposure to 1.8 GHz RF-EMR for 30 min on subsequent recognition memory in mice, using the novel object recognition task (NORT). RF-EMR exposure at an intensity of >2.2 W/kg specific absorption rate (SAR) power density induced a significant density-dependent increase in NORT index with no corresponding changes in spontaneous locomotor activity. RF-EMR exposure increased dendritic-spine density and length in hippocampal and prefrontal cortical neurons, as shown by Golgi staining. Whole-cell recordings in acute hippocampal and medial prefrontal cortical slices showed that RF-EMR exposure significantly altered the resting membrane potential and action potential frequency, and reduced the action potential half-width, threshold, and onset delay in pyramidal neurons. These results demonstrate that exposure to 1.8 GHz RF-EMR for 30 min can significantly increase recognition memory in mice, and can change dendritic-spine morphology and neuronal excitability in the hippocampus and prefrontal cortex. The SAR in this study (3.3 W/kg) was outside the range encountered in normal daily life, and its relevance as a potential therapeutic approach for disorders associated with recognition memory deficits remains to be clarified.


Subject(s)
Electromagnetic Fields/adverse effects , Electromagnetic Radiation , Pattern Recognition, Visual/radiation effects , Pyramidal Cells/radiation effects , Action Potentials/radiation effects , Animals , Dendritic Spines/pathology , Dendritic Spines/radiation effects , Hippocampus/physiopathology , Hippocampus/radiation effects , Memory , Memory Disorders/etiology , Memory Disorders/physiopathology , Mice , Pyramidal Cells/pathology , Radio Waves/adverse effects
17.
Sheng Li Xue Bao ; 69(1): 109-121, 2017 Feb 25.
Article in Chinese | MEDLINE | ID: mdl-28217814

ABSTRACT

Growth differentiation factor-15 (GDF-15) is a member of the transforming growth factor beta superfamily. GDF-15 expression is dramatically upregulated during acute brain injury, cancer, cardiovascular disease, and inflammation, suggesting its potential value as a disease biomarker. It has been suggested that GDF-15 has neurotropic effects in the nervous system. Our studies showed that GDF-15 modulated the expression of neuronal K+ and Ca2+ ion channels and increased the release of excitatory transmitter in the medial prefrontal cortex of mice. GDF-15 is also involved in the complex modulation of cancer and cardiovascular disease. Here, we reviewed studies involving the modulation of GDF-15 expression and its mechanisms, the primary pathological and physiological functions of GDF-15 in neurological and cardiovascular systems, and its role in cancer progression. The biological effects and the values of GDF-15 in basic research and clinical applications were also addressed.


Subject(s)
Cardiovascular Diseases/physiopathology , Growth Differentiation Factor 15/metabolism , Neoplasms/physiopathology , Nervous System/metabolism , Animals , Brain Injuries/physiopathology , Calcium Channels/metabolism , Disease Progression , Humans , Inflammation , Mice , Potassium Channels/metabolism , Prefrontal Cortex/metabolism , Transforming Growth Factor beta , Up-Regulation
18.
Acta Physiologica Sinica ; (6): 109-121, 2017.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-348293

ABSTRACT

Growth differentiation factor-15 (GDF-15) is a member of the transforming growth factor beta superfamily. GDF-15 expression is dramatically upregulated during acute brain injury, cancer, cardiovascular disease, and inflammation, suggesting its potential value as a disease biomarker. It has been suggested that GDF-15 has neurotropic effects in the nervous system. Our studies showed that GDF-15 modulated the expression of neuronal Kand Caion channels and increased the release of excitatory transmitter in the medial prefrontal cortex of mice. GDF-15 is also involved in the complex modulation of cancer and cardiovascular disease. Here, we reviewed studies involving the modulation of GDF-15 expression and its mechanisms, the primary pathological and physiological functions of GDF-15 in neurological and cardiovascular systems, and its role in cancer progression. The biological effects and the values of GDF-15 in basic research and clinical applications were also addressed.


Subject(s)
Animals , Humans , Mice , Brain Injuries , Calcium Channels , Metabolism , Cardiovascular Diseases , Disease Progression , Growth Differentiation Factor 15 , Metabolism , Inflammation , Neoplasms , Nervous System , Metabolism , Potassium Channels , Metabolism , Prefrontal Cortex , Metabolism , Transforming Growth Factor beta , Up-Regulation
19.
Hypertension ; 68(3): 785-95, 2016 09.
Article in English | MEDLINE | ID: mdl-27432863

ABSTRACT

Aldosterone, which plays a key role in maintaining water and electrolyte balance, is produced by zona glomerulosa cells of the adrenal cortex. Autonomous overproduction of aldosterone from zona glomerulosa cells causes primary hyperaldosteronism. Recent clinical studies have highlighted the pathological role of the KCNJ5 potassium channel in primary hyperaldosteronism. Our objective was to determine whether small-conductance Ca(2+)-activated potassium (SK) channels may also regulate aldosterone secretion in human adrenocortical cells. We found that apamin, the prototypic inhibitor of SK channels, decreased membrane voltage, raised intracellular Ca(2+) and dose dependently increased aldosterone secretion from human adrenocortical H295R cells. By contrast, 1-Ethyl-2-benzimidazolinone, an agonist of SK channels, antagonized apamin's action and decreased aldosterone secretion. Commensurate with an increase in aldosterone production, apamin increased mRNA expression of steroidogenic acute regulatory protein and aldosterone synthase that control the early and late rate-limiting steps in aldosterone biosynthesis, respectively. In addition, apamin increased angiotensin II-stimulated aldosterone secretion, whereas 1-Ethyl-2-benzimidazolinone suppressed both angiotensin II- and high K(+)-stimulated production of aldosterone in H295R cells. These findings were supported by apamin-modulation of basal and angiotensin II-stimulated aldosterone secretion from acutely prepared slices of human adrenals. We conclude that SK channel activity negatively regulates aldosterone secretion in human adrenocortical cells. Genetic association studies are necessary to determine whether mutations in SK channel subtype 2 genes may also drive aldosterone excess in primary hyperaldosteronism.


Subject(s)
Adrenal Cortex/cytology , Aldosterone/metabolism , Calcium Channel Agonists/pharmacology , Hyperaldosteronism/physiopathology , Potassium Channels/metabolism , Adrenal Cortex/metabolism , Adult , Aged , Analysis of Variance , Angiotensin II/administration & dosage , Apamin/administration & dosage , Cells, Cultured/drug effects , Female , Humans , Hyperaldosteronism/metabolism , Male , Middle Aged , Potassium Channels/drug effects , RNA, Messenger/metabolism , Sampling Studies
20.
Sci Rep ; 6: 28653, 2016 06 29.
Article in English | MEDLINE | ID: mdl-27353765

ABSTRACT

Growth differentiation factor-15 (GDF-15) has been implicated in ischemic brain injury and synapse development, but its involvement in modulating neuronal excitability and synaptic transmission remain poorly understood. In this study, we investigated the effects of GDF-15 on non-evoked miniature excitatory post-synaptic currents (mEPSCs) and neurotransmitter release in the medial prefrontal cortex (mPFC) in mice. Incubation of mPFC slices with GDF-15 for 60 min significantly increased the frequency of mEPSCs without effect on their amplitude. GDF-15 also significantly elevated presynaptic glutamate release, as shown by HPLC. These effects were blocked by dual TGF-ß type I receptor (TßRI) and TGF-ß type II receptor (TßRII) antagonists, but not by a TßRI antagonist alone. Meanwhile, GDF-15 enhanced pERK level, and inhibition of MAPK/ERK activity attenuated the GDF-15-induced increases in mEPSC and glutamate release. Blocking T-type calcium channels reduced the GDF-15 induced up-regulation of synaptic transmission. Membrane-protein extraction and use of an intracellular protein-transport inhibitor showed that GDF-15 promoted CaV3.1 and CaV3.3 α-subunit expression by trafficking to the membrane. These results confirm previous findings in cerebellar granule neurons, in which GDF-15 induces its neurobiological effects via TßRII and activation of the ERK pathway, providing novel insights into the mechanism of GDF-15 function in cortical neurons.


Subject(s)
Calcium Channels, T-Type/biosynthesis , Glutamic Acid/metabolism , Growth Differentiation Factor 15/metabolism , MAP Kinase Signaling System/physiology , Prefrontal Cortex/metabolism , Synaptic Transmission/physiology , Animals , Female , Mice , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...