Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
1.
PLoS One ; 16(4): e0250663, 2021.
Article in English | MEDLINE | ID: mdl-33905439

ABSTRACT

In a disease-state-dependent manner, the histamine-resistant itch in dry skin-based skin diseases such as atopic dermatitis (AD) and xerosis is mainly due to hyperinnervation in the epidermis. Semaphorin 3A (Sema3A) is a nerve repulsion factor expressed in keratinocytes and it suppresses nerve fiber elongation in the epidermis. Our previous studies have shown that Sema3A ointment inhibits epidermal hyperinnervation and scratching behavior and improves dermatitis scores in AD model mice. Therefore, we consider Sema3A as a key therapeutic target for improving histamine-resistant itch in AD and xerosis. This study was designed to screen a library of herbal plant extracts to discover compounds with potential to induce Sema3A in normal human epidermal keratinocytes (NHEKs) using a reporter gene assay, so that positive samples were found. Among the positive samples, only the extract of S. baicalensis was found to consistently increase Sema3A levels in cultured NHEKs in assays using quantitative real-time PCR and ELISA. In evaluation of reconstituted human epidermis models, the level of Sema3A protein in culture supernatants significantly increased by application of the extract of S. baicalensis. In addition, we investigated which components in the extract of S. baicalensis contributed to Sema3A induction and found that baicalin and baicalein markedly increased the relative luciferase activity, and that baicalein had higher induction activity than baicalin. Thus, these findings suggest that S. baicalensis extract and its compounds, baicalin and baicalein, may be promising candidates for improving histamine-resistant itch via the induction of Sema3A expression in epidermal keratinocytes.


Subject(s)
Plant Extracts/chemistry , Scutellaria baicalensis/chemistry , Semaphorin-3A/metabolism , Cell Line , Flavanones/genetics , Flavanones/metabolism , Flavonoids/genetics , Flavonoids/metabolism , Genes, Reporter , Humans , Keratinocytes/cytology , Keratinocytes/drug effects , Keratinocytes/metabolism , Models, Biological , Plant Extracts/pharmacology , RNA, Messenger/metabolism , Scutellaria baicalensis/metabolism , Semaphorin-3A/genetics
2.
J Med Food ; 22(3): 257-263, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30543483

ABSTRACT

Compound K (CK) is a metabolite of a saponin in Panax ginseng, formed from ginsenoside, a triterpenoid glycoside, by human intestinal bacteria. Lactobacillus paracasei A221 isolated from fermented food can hydrolyze (deglycosylate) the main ginsenoside, ginsenoside Rb1, and generate CK. However, the pharmacokinetics of L. paracasei A221 fermented ginseng (FG) and nonfermented ginseng (NFG) have not been investigated so far. The aim of this study was to investigate the pharmacokinetics of CK after oral administration of single doses of FG and NFG in healthy Japanese adults. An open-label, randomized, single-dose, two-period, crossover study was conducted in 12 Japanese healthy volunteers (five men and seven women, aged 40-60 years). All subjects were equally allocated into two groups and administered tablets containing FG or NFG. Until 24 h after the administration, blood samples were sequentially collected, plasma concentrations of CK were measured, and the pharmacokinetic parameters were calculated. We also expected restoration of decreased testosterone level as one of the beneficial effects of FG and measured plasma total testosterone concentrations in male volunteers. The means of Tmax, Cmax, and area under the concentration-time curve (AUC) were significantly different between the two groups. In the FG group, AUC0-12h (ng h/mL) and AUC0-24h (ng h/mL) were, respectively, 58.3- and 17.5-fold higher than those in the NFG group. Moreover, mean testosterone concentration in the FG group significantly increased 24 h after administration. These results showed that the main ginsenoside metabolite of ginseng, CK, produced by L. paracasei A221 has potential utility in health maintenance in healthy middle-aged and old Japanese adults.


Subject(s)
Ginsenosides/pharmacokinetics , Lacticaseibacillus paracasei/metabolism , Panax/microbiology , Adult , Cross-Over Studies , Female , Fermentation , Ginsenosides/administration & dosage , Ginsenosides/blood , Humans , Japan , Male , Middle Aged , Panax/chemistry , Testosterone/blood
3.
J Pharm Sci ; 107(11): 2883-2890, 2018 11.
Article in English | MEDLINE | ID: mdl-30055224

ABSTRACT

Heparinoid is commonly used for the treatment of superficial thrombophlebitis, a condition wherein inflammation and clotting occurs in the veins below the skin surface. However, stratum corneum is a major barrier that limits the delivery of hydrophilic heparinoid, in and across the skin. The aim of the present study was to develop a nonirritant topical formulation for heparinoid incorporating chemical penetration enhancers and investigate the delivery of heparinoid across the human epidermis using in vitro vertical Franz diffusion cells. The developed oil-in-water nanoemulsions (NEs; NE-1 and NE-2) delivered higher amount of heparinoid (91.58 ± 25.75 µg/sq.cm and 62.67 ± 5.66 µg/sq.cm, respectively) after 72 h compared with the other developed formulations, which in turn also delivered significantly higher amount compared with commercial formulations: cream (1.78 ± 0.07 µg/sq.cm), ointment (9.95 ± 4.41 µg/sq.cm), and gel (0 µg/sq.cm) (p <0.05). Transmission electron microscopy, polarizing light microscopy, and dynamic light scattering studies were performed to characterize the microstructure of these NEs with chemical enhancers. NE-1 was tested to be nonirritant with cell viability greater than 50% and a minimal release of IL-1α by using the "in vitro Epiderm tissue" model. Our results demonstrate that NE formulations represent a potential strategy for providing a localized therapy for the treatment of superficial thrombophlebitis.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacokinetics , Epidermis/metabolism , Heparinoids/administration & dosage , Heparinoids/pharmacokinetics , Pharmaceutical Vehicles/chemistry , Skin Absorption , Administration, Cutaneous , Drug Compounding , Emulsions/chemistry , Humans , Permeability , Solubility , Thermodynamics
4.
PLoS One ; 11(10): e0164799, 2016.
Article in English | MEDLINE | ID: mdl-27736988

ABSTRACT

Aloe has been used as a folk medicine because it has several important therapeutic properties. These include wound and burn healing, and Aloe is now used in a variety of commercially available topical medications for wound healing and skin care. However, its effects on epidermal keratinocytes remain largely unclear. Our data indicated that both Aloe vera gel (AVG) and Cape aloe extract (CAE) significantly improved wound healing in human primary epidermal keratinocytes (HPEKs) and a human skin equivalent model. In addition, flow cytometry analysis revealed that cell surface expressions of ß1-, α6-, ß4-integrin, and E-cadherin increased in HPEKs treated with AVG and CAE. These increases may contribute to cell migration and wound healing. Treatment with Aloe also resulted in significant changes in cell-cycle progression and in increases in cell number. Aloe increased gene expression of differentiation markers in HPEKs, suggesting roles for AVG and CAE in the improvement of keratinocyte function. Furthermore, human skin epidermal equivalents developed from HPEKs with medium containing Aloe were thicker than control equivalents, indicating the effectiveness of Aloe on enhancing epidermal development. Based on these results, both AVG and CAE have benefits in wound healing and in treatment of rough skin.


Subject(s)
Aloe/chemistry , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Plant Extracts/pharmacology , Aloe/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Line , Cell Movement/drug effects , Cornified Envelope Proline-Rich Proteins/genetics , Cornified Envelope Proline-Rich Proteins/metabolism , Humans , Integrin alpha6/genetics , Integrin alpha6/metabolism , Integrin beta1/genetics , Integrin beta1/metabolism , Integrin beta4/genetics , Integrin beta4/metabolism , Keratinocytes/cytology , Keratinocytes/drug effects , Keratinocytes/metabolism , Microscopy, Fluorescence , Models, Biological , Plant Extracts/chemistry , Wound Healing
5.
Neurol Res ; 37(6): 525-30, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25591424

ABSTRACT

OBJECTIVES: The (+)-isomer of indeloxazine AS1069562 has multiple pharmacological actions, such as serotonin (5-HIT) and norepinephrine (NE) reuptake inhibition and analgesic effects in animal models of neuropathic pain. Here, we investigated the analgesic effects of AS1069562 in rat models of inflammatory and noninflammatory nociceptive pain. METHODS: Adjuvant-induced arthritis (AIA) and bradykinin-induced knee joint pain were used as rat models of inflammatory pain. The chronic phase of monoiodoacetate-induced arthritis (MIA) was used as a rat model of noninflammatory pain. Analgesic effects were evaluated by weight-bearing deficit in the AIA and MIA models and by pain response in the bradykinin-induced knee joint pain model. RESULTS: In the AIA model and the bradykinin-induced knee joint pain model, AS1069562 significantly ameliorated the pain-related behavior of weight-bearing deficit and the pain response, respectively. AS1069562 also significantly improved the pain-related behavior of weight-bearing deficit in the chronic phase of the MIA model. Further, following monoiodoacetate injection, repeated administration of AS1069562 or duloxetine significantly improved weight-bearing deficit in the MIA model. Interestingly, the analgesic effect of AS1069562 was sustained for 24 hours after the last administration, although the plasma concentration of AS1069562 was reduced to undetectable levels. In contrast, the analgesic effect of duloxetine did not continue after treatment discontinuation. DISCUSSION: AS1069562 exerts analgesic effects on inflammatory and noninflammatory nociceptive pain in rat models of arthritis pain, and repeated administration of AS1069562 exerts a more persistent analgesic effect on arthritis pain than duloxetine. These findings suggest that AS1069562 has an attractive analgesic profile for the treatment of nociceptive pain.


Subject(s)
Analgesics/pharmacology , Arthralgia/drug therapy , Arthritis, Experimental/drug therapy , Morpholines/pharmacology , Nociceptive Pain/drug therapy , Analgesics/blood , Animals , Arthralgia/physiopathology , Arthritis, Experimental/physiopathology , Bradykinin , Chronic Disease , Disease Models, Animal , Duloxetine Hydrochloride/pharmacology , Female , Male , Morpholines/blood , Nociceptive Pain/physiopathology , Pain Measurement , Rats, Inbred Lew , Rats, Sprague-Dawley , Time Factors
6.
Eur Neuropsychopharmacol ; 24(10): 1698-708, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25108314

ABSTRACT

We recently identified ASP5736, (N-(diaminomethylene)-1-(3,5-difluoropyridin-4-yl)-4-fluoroisoquinoline-7-carboxamide (2E)-but-2-enedioate), a novel antagonist of 5-HT5A receptor, and here describe the in vitro and in vivo characterization of this compound. ASP5736 exhibited a high affinity for the human 5-HT5A receptor (Ki = 3.6 ± 0.66 nM) and antagonized 5-carboxamidotryptamine (5-CT)-induced Ca(2+) influx in human cells stably expressing the 5-HT5A receptor with approximately 200-fold selectivity over other receptors, including other 5-HT receptor subtypes, enzymes, and channels except human 5-HT2c receptor (Ki = 286.8 nM) and 5-HT7 receptor (Ki = 122.9 nM). Further, ASP5736 dose-dependently antagonized the 5-CT-induced decrease in cAMP levels in HEK293 cells stably expressing the 5-HT5A receptor. We then evaluated the effects of ASP5736 on cognitive impairments in several animal models of schizophrenia. Working memory deficit in MK-801-treated mice and visual learning deficit in neonatally phencyclidine (PCP)-treated mice were both ameliorated by ASP5736. In addition, ASP5736 also attenuated MK-801- and methamphetamine (MAP)-induced hyperactivity in mice without causing sedation, catalepsy, or plasma prolactin increase. The addition of olanzapine did not affect ASP5736-induced cognitive enhancement, and neither the sedative nor cataleptogenic effects of olanzapine were worsened by ASP5736. These results collectively suggest that ASP5736 is a novel and potent 5-HT5A receptor antagonist that not only ameliorates positive-like symptoms but also cognitive impairments in animal models of schizophrenia, without adverse effects. Present studies also indicate that ASP5736 holds potential to satisfy currently unmet medical needs for the treatment of schizophrenia by either mono-therapy or co-administered with commercially available antipsychotics.


Subject(s)
Antipsychotic Agents/pharmacology , Guanidines/pharmacology , Isoquinolines/pharmacology , Schizophrenia/drug therapy , Serotonin Antagonists/pharmacology , Animals , Antipsychotic Agents/chemistry , Antipsychotic Agents/pharmacokinetics , Calcium/metabolism , Catalepsy/drug therapy , Catalepsy/physiopathology , Cognition Disorders/drug therapy , Cognition Disorders/physiopathology , Cyclic AMP/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Guanidines/chemistry , Guanidines/pharmacokinetics , HEK293 Cells , Humans , Isoquinolines/chemistry , Isoquinolines/pharmacokinetics , Male , Memory Disorders/drug therapy , Memory Disorders/physiopathology , Mice , Mice, Inbred ICR , Motor Activity/drug effects , Motor Activity/physiology , Receptor, Serotonin, 5-HT2C/metabolism , Receptors, Serotonin/genetics , Receptors, Serotonin/metabolism , Schizophrenia/physiopathology , Serotonin Antagonists/chemistry , Serotonin Antagonists/pharmacokinetics
7.
Eur J Pharmacol ; 733: 54-61, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24704374

ABSTRACT

The (+)-isomer of indeloxazine AS1069562 exerts multiple pharmacological actions including the inhibition of serotonin (5-HT) and norepinephrine reuptake and analgesia in experimental animal pain models. Here, we evaluated the antinociceptive effects of AS1069562 and the antidepressants duloxetine and amitriptyline in mouse models of prostaglandin-induced spinal hypersensitivity. Prostaglandin E2 (PGE2) and F2α (PGF2α) were intrathecally administered to induce spinal hypersensitivity, causing tactile allodynia in mice. Allodynia induced by PGF2α but not by PGE2 was suppressed by desensitization of C-fibers with systemic pretreatment with resiniferatoxin. C-fiber hyperexcitability might therefore play a role in allodynia induced by PGF2α but not PGE2. In the PGE2-induced allodynia model, AS1069562 and duloxetine significantly suppressed allodynia, whereas amitriptyline did not. In the PGF2α-induced allodynia model, AS1069562 and amitriptyline significantly ameliorated allodynia, whereas duloxetine did not. To demonstrate the broad effects of AS1069562 compared to duloxetine, additional studies were conducted to elucidate other target mechanisms of AS1069562 beyond 5-HT and norepinephrine reuptake inhibition. AS1069562 exhibited affinity for both 5-HT1A and 5-HT3 receptors, and the analgesic effect of AS1069562 on PGF2α-induced allodynia was significantly blocked by the 5-HT1A receptor antagonist (S)-WAY100135 and the 5-HT3 receptor agonist SR57227. Taken together, these results indicate that AS1069562 inhibits both C-fiber- and non-C-fiber-dependent prostaglandin-induced allodynia, while duloxetine inhibits only non-C-fiber-triggered allodynia, and amitriptyline inhibits only C-fiber-triggered allodynia. These broad antinociceptive effects of AS1069562 may be due not only to 5-HT and norepinephrine reuptake inhibition but also to its effects on 5-HT receptors such as 5-HT1A and 5-HT3 receptors.


Subject(s)
Amitriptyline/therapeutic use , Analgesics/therapeutic use , Antidepressive Agents/therapeutic use , Hyperalgesia/drug therapy , Morpholines/therapeutic use , Prostaglandins/pharmacology , Spinal Cord/drug effects , Thiophenes/therapeutic use , Amitriptyline/administration & dosage , Amitriptyline/chemistry , Analgesics/administration & dosage , Analgesics/chemistry , Animals , Antidepressive Agents/administration & dosage , Antidepressive Agents/chemistry , Disease Models, Animal , Dose-Response Relationship, Drug , Duloxetine Hydrochloride , Hyperalgesia/etiology , Hyperalgesia/metabolism , Injections, Spinal , Male , Mice, Inbred ICR , Morpholines/administration & dosage , Morpholines/chemistry , Neuralgia/complications , Neuralgia/drug therapy , Neuralgia/metabolism , Spinal Cord/metabolism , Stereoisomerism , Thiophenes/administration & dosage , Thiophenes/chemistry
8.
J Pharmacol Exp Ther ; 348(3): 372-82, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24338505

ABSTRACT

AS1069562 [(R)-2-[(1H-inden-7-yloxy)methyl]morpholine monobenzenesulfonate] is the (+)-isomer of indeloxazine, which had been used clinically for the treatment of cerebrovascular diseases with multiple pharmacological actions, including serotonin (5-HT) and norepinephrine (NE) reuptake inhibition. Here we investigated the analgesic effects of AS1069562 in a rat model of chronic constriction injury (CCI)-induced neuropathic pain and the spinal monoamine turnover. These effects were compared with those of the antidepressants duloxetine and amitriptyline. AS1069562 significantly elevated extracellular 5-HT and NE levels in the rat spinal dorsal horn, although its 5-HT and NE reuptake inhibition was much weaker than that of duloxetine in vitro. In addition, AS1069562 increased the ratio of the contents of both 5-HT and NE to their metabolites in rat spinal cord, whereas duloxetine slightly increased only the ratio of the content of 5-HT to its metabolite. In CCI rats, AS1069562 and duloxetine significantly ameliorated mechanical allodynia, whereas amitriptyline did not. AS1069562 and amitriptyline significantly ameliorated thermal hyperalgesia, and duloxetine tended to ameliorate it. Furthermore, AS1069562, duloxetine, and amitriptyline significantly improved spontaneous pain-associated behavior. In a gastric emptying study, AS1069562 affected gastric emptying at the same dose that exerted analgesia in CCI rats. On the other hand, duloxetine and amitriptyline significantly reduced gastric emptying at lower doses than those that exerted analgesic effects. These results indicate that AS1069562 broadly improved various types of neuropathic pain-related behavior in CCI rats with unique characteristics in spinal monoamine turnover, suggesting that AS1069562 may have potential as a treatment option for patients with neuropathic pain, with a different profile from currently available antidepressants.


Subject(s)
Analgesics/therapeutic use , Biogenic Monoamines/metabolism , Morpholines/therapeutic use , Neuralgia/drug therapy , Spinal Cord/drug effects , Amitriptyline/pharmacology , Analgesics/pharmacokinetics , Analgesics/pharmacology , Animals , Dopamine/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Duloxetine Hydrochloride , Gastric Emptying/drug effects , HEK293 Cells , Humans , Male , Morpholines/pharmacokinetics , Morpholines/pharmacology , Motor Activity/drug effects , Neuralgia/physiopathology , Norepinephrine/metabolism , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Protein Binding , Rats , Rats, Sprague-Dawley , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Spinal Cord/metabolism , Stereoisomerism , Thiophenes/pharmacokinetics , Thiophenes/pharmacology
9.
Neuropharmacology ; 79: 10-6, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24211301

ABSTRACT

AS1069562 is the (+)-isomer of indeloxazine, which had been clinically used as a cerebral activator for the treatment of cerebrovascular diseases with serotonin and norepinephrine reuptake inhibition (SNRI) and neuroprotection. Here, we compared the analgesic effects of repeated treatment with AS1069562 and duloxetine, a selective SNRI, on pain-related behavior in a rat model of streptozotocin (STZ)-induced diabetic neuropathy. Further, we also evaluated the effects on the expression of neurotrophic factors and nerve conduction velocity. AS1069562 and duloxetine by single daily administration for 4 weeks significantly improved mechanical allodynia in STZ-induced diabetic rats and did not affect plasma glucose level or body weight. Interestingly, the analgesic effect of AS1069562 continued after a consecutive 1-week treatment discontinuation, although the plasma concentration of AS1069562 was reduced to undetectable levels. In contrast, the efficacy of duloxetine disappeared after treatment discontinuation. Expression analysis demonstrated that AS1069562 significantly restored decreased insulin-like growth factor 1 and fibroblast growth factor 2 mRNA levels in dorsal root ganglion and spinal cord, respectively, whereas duloxetine did not affect the expression levels of neurotrophic factors. In addition, AS1069562 reversed the slowing of nerve conduction velocity. The results of this study indicate that the analgesic effect of repeated dosing of AS1069562 but not duloxetine is persistent even after a 1-week drug discontinuation in STZ-induced diabetic rats. Restoration of neurotrophic factors may be involved in the curative-like pharmacological effect of this agent. Thus, AS1069562 may potentially offer a better treatment option for patients with painful diabetic neuropathy than duloxetine via different mechanisms.


Subject(s)
Analgesics/pharmacology , Diabetes Mellitus, Experimental , Diabetic Neuropathies/drug therapy , Morpholines/pharmacology , Neurotransmitter Uptake Inhibitors/pharmacology , Thiophenes/pharmacology , Animals , Diabetes Mellitus, Experimental/physiopathology , Diabetic Neuropathies/physiopathology , Duloxetine Hydrochloride , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Ganglia, Spinal/drug effects , Ganglia, Spinal/physiopathology , Hyperalgesia/drug therapy , Hyperalgesia/physiopathology , Insulin-Like Growth Factor I/metabolism , Male , Morpholines/blood , Morpholines/pharmacokinetics , Neural Conduction/drug effects , Neurotransmitter Uptake Inhibitors/blood , Neurotransmitter Uptake Inhibitors/pharmacokinetics , RNA, Messenger/metabolism , Random Allocation , Rats , Rats, Sprague-Dawley , Spinal Cord/drug effects , Spinal Cord/physiopathology , Streptozocin , Time Factors
10.
Neuropharmacology ; 79: 412-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24373902

ABSTRACT

γ-Secretase is the enzyme responsible for the intramembranous proteolysis of various substrates, such as amyloid precursor protein (APP) and Notch. Amyloid-ß peptide 42 (Aß42) is produced through the sequential proteolytic cleavage of APP by ß- and γ-secretase and causes the synaptic dysfunction associated with memory impairment in Alzheimer's disease. Here, we identified a novel cyclohexylamine-derived γ-secretase modulator, {(1R*,2S*,3R*)-3-[(cyclohexylmethyl)(3,3-dimethylbutyl)amino]-2-[4-(trifluoromethyl)phenyl]cyclohexyl}acetic acid (AS2715348), that may inhibit this pathological response. AS2715348 was seen to reduce both cell-free and cellular production of Aß42 without increasing levels of APP ß-carboxyl terminal fragment or inhibiting Notch signaling. Additionally, the compound increased Aß38 production, suggesting a shift of the cleavage site in APP. The inhibitory potency of AS2715348 on endogenous Aß42 production was similar across human, mouse, and rat cells. Oral administration with AS2715348 at 1 mg/kg and greater significantly reduced brain Aß42 levels in rats, and no Notch-related toxicity was observed after 28-day treatment at 100 mg/kg. Further, AS2715348 significantly ameliorated cognitive deficits in APP-transgenic Tg2576 mice. Finally, AS2715348 significantly reduced brain Aß42 levels in cynomolgus monkeys. These findings collectively show the promise for AS2715348 as a potential disease-modifying drug for Alzheimer's disease.


Subject(s)
Acetates/pharmacology , Alzheimer Disease/drug therapy , Amyloid Precursor Protein Secretases/metabolism , Brain/drug effects , Cyclohexylamines/pharmacology , Neuroprotective Agents/pharmacology , Acetates/adverse effects , Acetates/pharmacokinetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , Cell Line, Tumor , Cognition/drug effects , Cyclohexylamines/adverse effects , Cyclohexylamines/pharmacokinetics , Disease Models, Animal , Female , Humans , Macaca fascicularis , Male , Mice , Mice, Transgenic , Molecular Structure , Neuroprotective Agents/adverse effects , Neuroprotective Agents/pharmacokinetics , Nootropic Agents/adverse effects , Nootropic Agents/chemistry , Nootropic Agents/pharmacology , Peptide Fragments/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Notch/metabolism
11.
J Anesth ; 27(6): 939-41, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23760511

ABSTRACT

Voltage-dependent sodium channels (VDSCs) are crucial for pain generation. Here, to develop a new behavioral index of pain induced by spinal VDSC activation, we examined whether intrathecal veratridine injection produced nociceptive behavior. Intrathecal injection of the VDSC opener veratridine in mice dose-dependently induced nociceptive responses, with response times subsequently reduced by administration of morphine or pregabalin. Systemic administration of lidocaine and mexiletine, but not amitriptyline, also decreased this response time. Taken together, these results demonstrated that response time of nociceptive behavior induced by intrathecal veratridine injection is a quantitative index of pain triggered by spinal VDSC activation.


Subject(s)
Nociceptive Pain/chemically induced , Pain Management/methods , Pain Measurement/methods , Spine/drug effects , Voltage-Gated Sodium Channel Agonists/pharmacology , Voltage-Gated Sodium Channels/metabolism , Amitriptyline/pharmacology , Animals , Injections, Spinal , Lidocaine/pharmacology , Male , Mexiletine/pharmacology , Mice , Mice, Inbred ICR , Morphine/pharmacology , Nociceptive Pain/drug therapy , Pregabalin , Spine/metabolism , gamma-Aminobutyric Acid/analogs & derivatives , gamma-Aminobutyric Acid/pharmacology
12.
Eur J Pharmacol ; 703(1-3): 53-61, 2013 Mar 05.
Article in English | MEDLINE | ID: mdl-23276665

ABSTRACT

Alzheimer's disease is characterized by a progressive decline in cognitive function and involves ß-amyloid (Aß) in its pathogenesis. To characterize cognitive deficits associated with Aß accumulation, we analyzed PS1/APP mice overexpressing mutant presenilin-1 (PS1, M146L; line 6.2) and amyloid precursor protein (APP, K670N/M671L; line Tg2576), a mouse model of Alzheimer's disease with accelerated Aß production. Age-dependent changes in working and spatial memory behaviors were investigated using Y-maze and Morris water maze tasks, respectively, in female PS1/APP mice at ages of 2, 4, 6, and 12 months. Significant deficits in working and spatial memory were observed from 4 and 6 months of age, respectively. Acute single-dose administrations of memantine, a low-to-moderate-affinity N-methyl-d-aspartate (NMDA) antagonist, showed improvements in working memory deficits at 4 months of age, whereas donepezil, an acetylcholinesterase (AChE) inhibitor, did not. However, both drugs improved spatial memory dysfunction at 6 months of age at therapeutically relevant doses. No age-related dramatic changes were observed in expression levels of several proteins relating to memory dysfunction and also the mechanisms of donepezil and memantine in the cerebral cortex of PS1/APP mice until 6 months of age. Taken together, these results suggest dysfunctions in cholinergic and/or glutamatergic transmissions may be involved in the cognitive deficits associated with Aß toxicity. Since donepezil and memantine have been widely used for treating patients of Alzheimer's disease, these results also suggest that cognitive deficits in PS1/APP mice assessed in the Y-maze and Morris water maze tasks are a useful animal model for evaluating novel Alzheimer's disease therapeutics.


Subject(s)
Alzheimer Disease/drug therapy , Cholinesterase Inhibitors/therapeutic use , Indans/therapeutic use , Memantine/therapeutic use , Memory Disorders/drug therapy , Nootropic Agents/therapeutic use , Piperidines/therapeutic use , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Animals , Brain/metabolism , Cholinesterase Inhibitors/blood , Cholinesterase Inhibitors/pharmacology , Cyclic AMP Response Element-Binding Protein/metabolism , Disease Models, Animal , Donepezil , Female , Indans/blood , Indans/pharmacology , Maze Learning , Memantine/blood , Memantine/pharmacology , Memory/drug effects , Memory Disorders/metabolism , Memory Disorders/physiopathology , Mice , Mice, Transgenic , Nootropic Agents/blood , Nootropic Agents/pharmacology , Piperidines/blood , Piperidines/pharmacology , Receptors, AMPA/metabolism
13.
J Neurochem ; 125(3): 465-72, 2013 May.
Article in English | MEDLINE | ID: mdl-23240999

ABSTRACT

Given that amyloid-ß 42 (Aß42) is believed to be a culprit in Alzheimer's disease (AD), reducing Aß42 production should be a potential therapeutic approach. γ-Secretase modulators (GSMs) cause selective reduction of Aß42 or both reduction of Aß42 and Aß40 without affecting total Aß through shifting the γ-cleavage position in amyloid precursor protein. We recently reported on GSM-2, one of the second-generation GSMs, that selectively reduced brain Aß42 level and significantly ameliorated cognitive deficits in plaque-free 5.5-month-old Tg2576 AD model mice. Here, we investigated the effects of GSM-2 on 10-, 14-, and 18-month-old mice which had age-dependent increase in amyloid plaques. Eight-day treatment with GSM-2 significantly ameliorated cognitive deficits measured by Y-maze task in the mice of any age. However, GSM-2 reduced brain soluble Aß42 only in 10-month-old mice. In contrast, GSM-2 markedly reduced newly synthesized soluble Aß42 in both 10- and 18-month-old mice with similar efficacy when measured using the stable isotope-labeling technique, suggesting that nascent Aß42 plays a more significant role than plaque-associated soluble Aß42 in the cognitive deterioration of Tg2576 mice. These findings further indicate the potential utility of approach to reducing Aß42 synthesis in AD therapeutic regimens.


Subject(s)
Alzheimer Disease/complications , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Cognition Disorders/metabolism , Enzyme Inhibitors/therapeutic use , Peptide Fragments/metabolism , Acetates/pharmacology , Acetates/therapeutic use , Age Factors , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/immunology , Amyloid beta-Protein Precursor/genetics , Animals , Antibodies/therapeutic use , Chromatography, Liquid , Cognition Disorders/drug therapy , Cognition Disorders/etiology , Disease Models, Animal , Enzyme Inhibitors/chemistry , Enzyme-Linked Immunosorbent Assay , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Mass Spectrometry , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Peptide Fragments/immunology , Piperidines/pharmacology , Piperidines/therapeutic use
14.
Neurol Res ; 34(5): 469-77, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22642793

ABSTRACT

OBJECTIVES: FK1706, a non-immunosuppressive immunophilin ligand, potentiated nerve growth factor-induced neurite outgrowth, putatively mediated via FKBP-52 and the Ras/Raf/MAPK signaling pathway. It also improved mechanical allodynia accompanied by the recovery of intraepidermal nerve fiber density in a painful diabetic neuropathy in rats. The aim of this study was to demonstrate the gene expression profiling in dorsal root ganglion in streptozotocin-induced diabetic rats related to pain and anti-allodynia effects of FK1706 administration to elucidate the putative mechanisms of its neurotrophic activity in vivo. Here, we analyzed gene expression of the dorsal root ganglia using microarray together with behavioral measurement of mechanical allodynia in diabetic rats to try to capture the global fingerprint of changes in gene expression associated with FK1706 administration. METHODS: The withdrawal threshold of streptozotocin-induced diabetic rats was measured by an electronic von Frey system. The gene expression of the ganglia from L4 to L6 obtained from streptozotocin-treated rats with or without chronic administration of FK1706 was analyzed using an Affymetrix GeneChip to extract interesting genes in the development of mechanical allodynia in diabetes and anti-allodynia effect of FK1706. RESULTS: Daily oral administration of FK1706 improved mechanical allodynia without decreasing plasma glucose levels. From gene expression analysis, the expression of thioredoxin interacting protein gene was sustained to increased change, whereas those of collagen I alpha1, II alpha1 and IX alpha1 genes were decreased from 2 to 4 weeks after streptozotocin injection. While no changes occurred after 1 week of commencing of FK1706 administration (2 weeks after streptozotocin injection), changes in expression more than 1.5-fold were observed for genes such as Ckm, Actn3, Atp2a1, Bglap, Acta1, Myl1, Tnnc2, and Mylpf at 2 weeks of FK1706 administration (3 weeks after streptozotocin injection). The genes RGD1564519, Hbb, LOC689064, Arpc4 and S100a9 were upregulated in comparison with streptozotocin-injected control group at 3 weeks of FK1706 administration; on the other hand, those of Actn3, Atp2a1 were downregulated by FK1706. DISCUSSION: FK1706 ameliorates mechanical allodynia with accompanying increases in gene expressions possibly related to neurite outgrowth, development, differentiation, and nociceptive sensitivity.


Subject(s)
Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/pathology , Ganglia, Spinal/drug effects , Gene Expression Regulation/drug effects , Hyperalgesia/etiology , Immunophilins/metabolism , Animals , Diabetes Mellitus, Experimental/drug therapy , Disease Models, Animal , Gene Expression Profiling , Hyperalgesia/drug therapy , Male , Oligonucleotide Array Sequence Analysis , Pain Threshold/drug effects , Rats , Rats, Sprague-Dawley , Tacrolimus/analogs & derivatives , Tacrolimus/pharmacology , Tacrolimus/therapeutic use , Thioredoxins/metabolism , Time Factors
15.
Eur J Pharmacol ; 685(1-3): 59-69, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22542656

ABSTRACT

Hypofunction of brain N-methyl-d-aspartate (NMDA) receptors has been implicated in psychiatric disorders such as schizophrenia and Alzheimer's disease. Inhibition of glycine transporter-1 (GlyT1) is expected to increase glycine, a co-agonist of the NMDA receptor and, consequently, to facilitate NMDA receptor function. We have identified ASP2535 (4-[3-isopropyl-5-(6-phenyl-3-pyridyl)-4H-1,2,4-triazol-4-yl]-2,1,3-benzoxadiazole) as a novel GlyT1 inhibitor, and here describe our in vitro and in vivo characterization of this compound. ASP2535 potently inhibited rat GlyT1 (IC(50)=92 nM) with 50-fold selectivity over rat glycine transporter-2 (GlyT2). It showed minimal affinity for many other receptors except for µ-opioid receptors (IC(50)=1.83 µM). Oral administration of ASP2535 dose-dependently inhibited ex vivo [(3)H]-glycine uptake in mouse cortical homogenate, suggesting good brain permeability. This profile was confirmed by pharmacokinetic analysis. We then evaluated the effect of ASP2535 on animal models of cognitive impairment in schizophrenia and Alzheimer's disease. Working memory deficit in MK-801-treated mice and visual learning deficit in neonatally phencyclidine (PCP)-treated mice were both attenuated by ASP2535 (0.3-3mg/kg, p.o. and 0.3-1mg/kg, p.o., respectively). ASP2535 (1-3mg/kg, p.o.) also improved the PCP-induced deficit in prepulse inhibition in rats. Moreover, the working memory deficit in scopolamine-treated mice and the spatial learning deficit in aged rats were both attenuated by ASP2535 (0.1-3mg/kg, p.o. and 0.1mg/kg, p.o., respectively). These studies provide compelling evidence that ASP2535 is a novel and centrally-active GlyT1 inhibitor that can improve cognitive impairment in animal models of schizophrenia and Alzheimer's disease, suggesting that ASP2535 may satisfy currently unmet medical needs for the treatment of these diseases.


Subject(s)
Alzheimer Disease/drug therapy , Cognition Disorders/drug therapy , Glycine Plasma Membrane Transport Proteins/antagonists & inhibitors , Oxadiazoles/pharmacology , Schizophrenia/drug therapy , Triazoles/pharmacology , Administration, Oral , Alzheimer Disease/physiopathology , Animals , Brain/metabolism , Cognition Disorders/etiology , Cognition Disorders/physiopathology , Disease Models, Animal , Dizocilpine Maleate/toxicity , Dose-Response Relationship, Drug , Female , Humans , Inhibitory Concentration 50 , Male , Memory Disorders/drug therapy , Memory Disorders/physiopathology , Mice , Oxadiazoles/administration & dosage , Oxadiazoles/pharmacokinetics , Permeability , Rats , Rats, Wistar , Schizophrenia/physiopathology , Triazoles/administration & dosage , Triazoles/pharmacokinetics
16.
J Neurosci ; 32(6): 2037-50, 2012 Feb 08.
Article in English | MEDLINE | ID: mdl-22323718

ABSTRACT

γ-Secretase inhibitors (GSIs) reduce amyloid-ß (Aß) peptides but inevitably increase the ß-C-terminal fragment (ß-CTF) of amyloid precursor protein (APP), potentially having undesirable effects on synapses. In contrast, γ-secretase modulators (GSMs) reduce Aß42 without increasing ß-CTF. Although the Aß-lowering effects of these compounds have been extensively studied, little effort has been made to investigate their effects on cognition. Here, we compared the effects of two GSIs--(2S)-2-hydroxy-3-methyl-N-[(2S)-1-{[(1S)-3-methyl-2-oxo-2,3,4,5-tetrahydro-1H-3-benzazepin-1-yl]amino}-1-oxopropan-2-yl]butanamide (LY450139, semagacestat) and (2R)-2-[[(4-chlorophenyl)sulfonyl][[2-fluoro-4-(1,2,4-oxazol-3-yl)phenyl]methyl]amino-5,5,5-trifluoropentanamide (BMS-708163)--and a second-generation GSM [{(2S,4R)-1-[(4R)-1,1,1-trifluoro-7-methyloctan-4-yl]-2-[4-(trifluoromethyl)phenyl]piperidin-4-yl}acetic acid (GSM-2)] on spatial working memory in APP-transgenic (Tg2576) and nontransgenic mice using the Y-maze task. While acute dosing with either GSI ameliorated memory deficits in 5.5-month-old Tg2576 mice, these effects disappeared after 8 d subchronic dosing. Subchronic dosing with either GSI rather impaired normal cognition in 3-month-old Tg2576 mice, with no inhibition on the processing of other γ-secretase substrates, such as Notch, N-cadherin, or EphA4, in the brain. LY450139 also impaired normal cognition in wild-type mice; however, the potency was 10-fold lower than that in Tg2576 mice, indicating an APP-dependent mechanism likely with ß-CTF accumulation. Immunofluorescence studies revealed that the ß-CTF accumulation was localized in the presynaptic terminals of the hippocampal stratum lucidum and dentate hilus, implying an effect on presynaptic function in the mossy fibers. In contrast, both acute and subchronic dosing with GSM-2 significantly ameliorated memory deficits in Tg2576 mice and did not affect normal cognition in wild-type mice. We demonstrated a clear difference between GSI and GSM in effects on functional consequences, providing new insights into strategies for developing these drugs against Alzheimer's disease.


Subject(s)
Alanine/analogs & derivatives , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/physiology , Amyloid beta-Protein Precursor/physiology , Azepines/pharmacology , Cognition/drug effects , Cognition/physiology , Protease Inhibitors/pharmacology , Alanine/pharmacology , Amyloid beta-Protein Precursor/genetics , Animals , Cell Line, Tumor , Female , Humans , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Transgenic
17.
Biol Pharm Bull ; 34(7): 1105-8, 2011.
Article in English | MEDLINE | ID: mdl-21720020

ABSTRACT

Transient receptor potential vanilloid 1 (TRPV1) is primarily expressed in central and peripheral terminals of non-myelinated primary afferent neurons. We previously showed that AS1928370, a novel TRPV1 antagonist that can prevent ligand-induced activation but not proton-induced activation, ameliorates neuropathic pain in rats without hyperthermic effect. In this study, we investigated its analgesic profile in mice. AS1928370 showed good oral bioavailability and high penetration into the brain and spinal cord in mice. The mean plasma-to-brain and plasma-to-spinal cord ratios were 4.3 and 3.5, respectively. Pretreatment with AS1928370 significantly suppressed both capsaicin-induced acute pain and withdrawal response in hot plate test at 10-30 mg/kg per os (p.o.). At lower oral doses (0.3-1.0 mg/kg), AS1928370 improved mechanical allodynia in mice undergoing spinal nerve ligation. Intrathecal administration of AS1928370 (30 µg/body) also significantly suppressed mechanical allodynia. In addition, AS1928370 showed no effect on locomotor activity up to 30 mg/kg p.o. These results suggest that spinal TRPV1 has an important role in the transmission of neuropathic pain and that the central nervous system (CNS) penetrant TRPV1 receptor antagonist AS1928370 is a promising candidate for treating neuropathic pain.


Subject(s)
Benzamides/pharmacology , Hyperalgesia/prevention & control , Neuralgia/prevention & control , Quinolones/pharmacology , TRPV Cation Channels/antagonists & inhibitors , Animals , Benzamides/administration & dosage , Benzamides/pharmacokinetics , Capsaicin/pharmacology , Disease Models, Animal , Injections, Spinal , Mice , Neuralgia/chemically induced , Quinolones/administration & dosage , Quinolones/pharmacokinetics , Tissue Distribution
18.
Life Sci ; 88(17-18): 761-5, 2011 Apr 25.
Article in English | MEDLINE | ID: mdl-21356217

ABSTRACT

AIMS: Clinical use of olanzapine has been suggested to be associated with weight gain and adiposity in schizophrenic patients. While studies in experimental animals have noted weight gain in olanzapine-treated female rats, these findings have yet to be replicated in males. This study investigated the effect of chronic subcutaneous infusion of olanzapine in male rats via a recently developed electrical microinfusion pump. MAIN METHODS: An electrical microinfusion pump was subcutaneously implanted in male Sprague-Dawley rats who were then chronically administered olanzapine. Plasma olanzapine concentration and body weight were monitored, and fat pads were weighed after six weeks' olanzapine treatment. KEY FINDINGS: Plasma olanzapine concentration plateaued within 4h of commencement of chronic olanzapine 1.5mg/animal/day infusion and remained constant until day 21. Six-week infusion of olanzapine at 1.5 but not 1mg/animal/day induced significant adiposity in subcutaneous, epididymal, and retroperitoneal fat. Body weight and food intake values did not differ between olanzapine- and vehicle-treated rats throughout the experiment. SIGNIFICANCE: The present study demonstrated that chronic infusion of olanzapine induced adiposity in male rats without inducing weight gain or hyperphagia, even with sufficient plasma concentration. This report is the first to provide information about adiposity-inducible plasma concentration of olanzapine in male rats.


Subject(s)
Adipose Tissue/drug effects , Antipsychotic Agents/pharmacology , Benzodiazepines/pharmacology , Animals , Antipsychotic Agents/administration & dosage , Antipsychotic Agents/pharmacokinetics , Benzodiazepines/administration & dosage , Benzodiazepines/pharmacokinetics , Body Weight/drug effects , Eating/drug effects , Infusion Pumps, Implantable , Infusions, Subcutaneous , Male , Olanzapine , Rats , Rats, Sprague-Dawley
19.
Behav Brain Res ; 216(2): 561-8, 2011 Jan 20.
Article in English | MEDLINE | ID: mdl-20816897

ABSTRACT

Body weight gain is one of the most serious side effects associated with clinical use of antipsychotics. However, the mechanisms by which antipsychotics induce body weight gain are unknown, and no reliable animal models of antipsychotics-induced weight gain have been established. The present studies were designed to establish male rat models of weight gain induced by chronic and acute treatment with antipsychotics. Six-week chronic treatment with olanzapine (5, 7.5, and 10mg/kg/day) in male Sprague-Dawley rats fed a daily diet resembling a human macronutrient diet, significantly increased body weight gain and weight of fatty tissues. In contrast, ziprasidone (1.25, 2.5, and 5mg/kg/day) administration caused no observable adverse effects. We then investigated feeding behavior with acute antipsychotic treatment in male rats using an automated food measurement apparatus. Rats were allowed restricted access to normal laboratory chow (4h/day). With acute olanzapine (0.5, 1, and 2mg/kg, i.p.) treatment in the light phase, food intake volume and duration were significantly increased, while treatment with ziprasidone (0.3, 1, and 3mg/kg, i.p.) did not increase food intake volume or meal time duration. Findings from the present studies showed that chronic treatment with olanzapine in male rats induced body weight gain, and acute injection induced hyperphagia, suggesting that hyperphagia may be involved in the weight gain and obesity-inducing properties of chronically administered olanzapine. These animal models may provide useful experimental platforms for analysis of the mechanism of hyperphagia and evaluating the potential risk of novel antipsychotics to induce weight gain in humans.


Subject(s)
Antipsychotic Agents/adverse effects , Benzodiazepines/adverse effects , Disease Models, Animal , Hyperphagia/chemically induced , Piperazines/adverse effects , Thiazoles/adverse effects , Weight Gain/drug effects , Animals , Antipsychotic Agents/administration & dosage , Benzodiazepines/administration & dosage , Dose-Response Relationship, Drug , Feeding Behavior/drug effects , Male , Olanzapine , Piperazines/administration & dosage , Rats , Rats, Sprague-Dawley , Thiazoles/administration & dosage
20.
J Pharmacol Exp Ther ; 336(3): 743-50, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21098091

ABSTRACT

Transient receptor potential vanilloid 1 (TRPV1) is activated by a variety of stimulations, such as endogenous ligands and low pH, and is believed to play a role in pain transmission. TRPV1 antagonists have been reported to be effective in several animal pain models; however, some compounds induce hyperthermia in animals and humans. We discovered the novel TRPV1 antagonist (R)-N-(1-methyl-2-oxo-1,2,3,4-tetrahydro-7-quinolyl)-2-[(2-methylpyrrolidin-1-yl)methyl]biphenyl-4-carboxamide (AS1928370) in our laboratory. AS1928370 bound to the resiniferatoxin-binding site on TRPV1 and inhibited capsaicin-mediated inward currents with an IC50 value of 32.5 nM. Although AS1928370 inhibited the capsaicin-induced Ca²(+) flux in human and rat TRPV1-expressing cells, the inhibitory effect on proton-induced Ca²(+) flux was extremely small. In addition, AS1928370 showed no inhibitory effects on transient receptor potential vanilloid 4, transient receptor potential ankyrin 1, and transient receptor potential melastatin 8 in concentrations up to 10 µM. AS1928370 improved capsaicin-induced secondary hyperalgesia and mechanical allodynia in an L5/L6 spinal nerve ligation model in rats with respective ED50 values of 0.17 and 0.26 mg/kg p.o. Furthermore, AS1928370 alleviated inflammatory pain in a complete Freund's adjuvant model at 10 mg/kg p.o. AS1928370 had no effect on rectal body temperature up to 10 mg/kg p.o., although a significant hypothermic effect was noted at 30 mg/kg p.o. In addition, AS1928370 showed no significant effect on motor coordination. These results suggest that blockage of the TRPV1 receptor without affecting the proton-mediated TRPV1 activation is a promising approach to treating neuropathic pain because of the potential wide safety margin against hyperthermic effects. As such, compounds such as ASP1928370 may have potential as new analgesic agents for treating neuropathic pain.


Subject(s)
Analgesics/therapeutic use , Benzamides/therapeutic use , Fever , Neuralgia/drug therapy , Pain Measurement/drug effects , Quinolones/therapeutic use , TRPV Cation Channels/antagonists & inhibitors , Analgesics/pharmacology , Animals , Benzamides/chemistry , Benzamides/pharmacology , Capsaicin/pharmacology , Capsaicin/therapeutic use , Fever/chemically induced , HEK293 Cells , Humans , Male , Neuralgia/physiopathology , Pain/drug therapy , Pain/physiopathology , Pain Measurement/methods , Protein Binding/physiology , Quinolones/chemistry , Quinolones/pharmacology , Rats , Rats, Sprague-Dawley , TRPV Cation Channels/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...