Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
J Pharmacol Sci ; 155(2): 52-62, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38677786

ABSTRACT

The ubiquitin-proteasome system (UPS) is a major proteolytic system that plays an important role in the regulation of various cell processes, such as cell cycle, stress response, and transcriptional regulation, especially in neurons, and dysfunction of UPS is considered to be a cause of neuronal cell death in neurodegenerative diseases. However, the mechanism of neuronal cell death caused by UPS dysfunction has not yet been fully elucidated. In this study, we investigated the mechanism of neuronal cell death induced by proteasome inhibitors using human neuroblastoma SH-SY5Y cells. Z-Leu-D-Leu-Leu-al (MG132), a proteasome inhibitor, induced apoptosis in SH-SY5Y cells in a concentration- and time-dependent manner. Antioxidants N-acetylcysteine and EUK-8 attenuated MG132-induced apoptosis. Apocynin and diphenyleneiodonium, inhibitors of NADPH oxidase (NOX), an enzyme that produces superoxide anions, also attenuated MG132-induced apoptosis. It was also found that MG132 treatment increased the expression of NOX5, a NOX family member, and that siRNA-mediated silencing of NOX5 and BAPTA-AM, which inhibits NOX5 by chelating calcium, suppressed MG132-induced apoptosis and production of reactive oxygen species in SH-SY5Y cells. These results suggest that MG132 induces apoptosis in SH-SY5Y cells through the production of superoxide anion by NOX5.


Subject(s)
Apoptosis , Leupeptins , NADPH Oxidase 5 , NADPH Oxidases , Neuroblastoma , Proteasome Inhibitors , Superoxides , Humans , Apoptosis/drug effects , Apoptosis/genetics , Proteasome Inhibitors/pharmacology , Superoxides/metabolism , Cell Line, Tumor , Neuroblastoma/pathology , Neuroblastoma/metabolism , Leupeptins/pharmacology , NADPH Oxidases/metabolism , NADPH Oxidases/genetics , NADPH Oxidase 5/genetics , NADPH Oxidase 5/metabolism , Antioxidants/pharmacology , Dose-Response Relationship, Drug , Acetylcysteine/pharmacology , Neurons/metabolism , Neurons/drug effects
2.
J Pharmacol Sci ; 153(2): 73-83, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37640472

ABSTRACT

Schwann cells and oligodendrocytes secrete proteins that promote neuron survival, but their role in amyotrophic lateral sclerosis (ALS) is unclear. To address this question, we evaluated the effect of molecules secreted by Schwann cells on reactive oxygen species (ROS)-induced motor neuronal cell death. We observed that in motor neuron cell line NSC-34 cultures, the conditioned medium (CM) from Schwann cell line YST-1 (YST-1 CM) cultures had a protective effect against hydrogen peroxide-induced cell death. However, this protective effect of YST-1 CM was abolished by removing peroxiredoxin 1-4 (PRDX1-4) from the CM. We found that the expression of PRDX1 mRNA was markedly downregulated in the lumbar spinal cord of the superoxide dismutase 1 (SOD1)G93A mouse model of ALS. We also found that transient transfection of YST-1 cells with G93A SOD1 resulted in reduced PRDX1 mRNA expression. Additionally, in the mutant transfected cells, YST-1 CM showed decreased neuroprotective effect against hydrogen peroxide-induced NSC-34 cell death compared to those transfected with WT SOD1. Our results suggest that Schwann cells protect motor neurons from oxidative stress by secreting PRDX1 and that the reduction of PRDX secreted from Schwann cells contributes to increased ROS and associated motor neuronal death in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Hydrogen Peroxide , Animals , Mice , Hydrogen Peroxide/toxicity , Amyotrophic Lateral Sclerosis/genetics , Reactive Oxygen Species , Superoxide Dismutase-1/genetics , Motor Neurons , Cell Death , Schwann Cells , Cell Line , Peroxiredoxins/genetics
3.
Neurochem Res ; 48(12): 3571-3584, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37556038

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a severe neurodegenerative disease with selective degeneration of motor neurons. It has been reported that an increase in the levels of inflammatory cytokines and glial cells such as reactive astrocytes is closely involved in the pathological progression of ALS. Recently, the levels of neuropathic cytotoxic (A1) astrocytes among reactive astrocytes have reportedly increased in the central nervous system of ALS mice, which induce motor neuron degeneration through the production of inflammatory cytokines and secretion of neuropathic factors. Hence, elucidating the induction mechanism of A1 astrocytes in ALS is important to understand the mechanism of disease progression in ALS. In this study, we observed that the expression of peroxiredoxin 6 (PRDX6), a member of the peroxiredoxin family, was markedly upregulated in astrocytes of the lumbar spinal cord of SOD1G93A mice model for ALS. Additionally, when PRDX6 was transiently transfected into the mouse astrocyte cell line C8-D1A and human astrocytoma cell line U-251 MG, the mRNA expression of complement C3 (a marker for A1 astrocyte phenotype) and inflammatory cytokines was increased. Furthermore, the mRNA expression of C3 and inflammatory cytokine was increased in C8-D1A and U-251 MG cells stably expressing PRDX6, and the increased mRNA expression was significantly suppressed by MJ33 (lithium[1-hexadecoxy-3-(2,2,2-trifluoroethoxy) propan-2-yl] methyl phosphate), an inhibitor of the phospholipase A2 activity of PRDX6. Our results suggest that the expression of PRDX6 in astrocytes plays an important role in the induction of A1 astrocytes and expression of inflammatory cytokines in the ALS mice model.


Subject(s)
Amyotrophic Lateral Sclerosis , Neurodegenerative Diseases , Neurotoxicity Syndromes , Mice , Humans , Animals , Amyotrophic Lateral Sclerosis/metabolism , Astrocytes/metabolism , Peroxiredoxin VI/genetics , Peroxiredoxin VI/metabolism , Neurodegenerative Diseases/metabolism , Mice, Transgenic , Spinal Cord/metabolism , Cytokines/metabolism , Disease Models, Animal , Neurotoxicity Syndromes/metabolism , RNA, Messenger/metabolism , Superoxide Dismutase-1/genetics , Superoxide Dismutase/metabolism
4.
J Pharmacol Sci ; 148(1): 41-50, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34924128

ABSTRACT

We previously reported that dopamine (DA) attenuated lipopolysaccharide (LPS)-induced expression of proinflammatory cytokines through the formation of DA quinone (DAQ) in murine microglial cell line BV-2 and primary murine microglial cells. To reveal whether DA inhibits the expression of proinflammatory cytokines of microglial cells through the formation of DAQ in the central nervous system (CNS), in this study, we examined the effect of DAQ on LPS-induced mRNA expression of proinflammatory cytokines in C57BL/6 mouse brain under two experimental conditions: 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) administration and l-dopa/carbidopa administration. Acute MPTP administration reduced the number of tyrosine hydroxylase-positive cells in the substantia nigra, and decreased the level of quinoprotein, an indicator of DAQ formation, in the striatum. Real-time RT-PCR analysis revealed that intraperitoneal administration of LPS increased the mRNA levels of proinflammatory cytokines, including tumor-necrosis factor-α and interleukin-1ß, in the striatum. These increases were enhanced in MPTP-treated mice. On the other hand, l-dopa/carbidopa administration increased the level of quinoprotein, attenuated the LPS-induced mRNA expression of proinflammatory cytokines, and reduced the LPS-induced increase in the number of microglial cells in the striatum. These results suggest that DA attenuate the expression of proinflammatory cytokines in microglia through the formation of DAQ in the CNS.


Subject(s)
Corpus Striatum/metabolism , Cytokines/genetics , Cytokines/metabolism , Dopamine/analogs & derivatives , Dopamine/pharmacology , Gene Expression/drug effects , Gene Expression/genetics , Inflammation Mediators/metabolism , Microglia/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Cell Line , Depression, Chemical , Dopamine/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Male , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
5.
J Neurosci Res ; 99(2): 621-637, 2021 02.
Article in English | MEDLINE | ID: mdl-32954502

ABSTRACT

Oxidative stress has been implicated in a variety of neurodegenerative disorders, such as Alzheimer's and Parkinson's disease. Astrocytes play a significant role in maintaining survival of neurons by supplying antioxidants such as glutathione (GSH) to neurons. Recently, we found that noradrenaline increased the intracellular GSH concentration in astrocytes via ß3 -adrenoceptor stimulation. These observations suggest that noradrenaline protects neurons from oxidative stress-induced death by increasing the supply of GSH from astrocytes to neurons via the stimulation of ß3 -adrenoceptor in astrocytes. In the present study, we examined the protective effect of noradrenaline against H2 O2 -induced neurotoxicity using two different mixed cultures: the mixed culture of human astrocytoma U-251 MG cells and human neuroblastoma SH-SY5Y cells, and the mouse primary cerebrum mixed culture of neurons and astrocytes. H2 O2 -induced neuronal cell death was significantly attenuated by pretreatment with noradrenaline in both mixed cultures but not in single culture of SH-SY5Y cells or in mouse cerebrum neuron-rich culture. The neuroprotective effect of noradrenaline was inhibited by SR59230A, a selective ß3 -adrenoceptor antagonist, and CL316243, a selective ß3 -adrenoceptor agonist, mimicked the neuroprotective effect of noradrenaline. DL-buthionine-[S,R]-sulfoximine, a GSH synthesis inhibitor, negated the neuroprotective effect of noradrenaline in both mixed cultures. MK571, which inhibits the export of GSH from astrocytes mediated by multidrug resistance-associated protein 1, also prevented the neuroprotective effect of noradrenaline. These results suggest that noradrenaline protects neurons against H2 O2 -induced death by increasing the supply of GSH from astrocytes via ß3 -adrenoceptor stimulation.


Subject(s)
Astrocytes/drug effects , Glutathione/metabolism , Neurons/drug effects , Neuroprotective Agents/pharmacology , Norepinephrine/pharmacology , Receptors, Adrenergic, beta-3/physiology , Adrenergic beta-3 Receptor Agonists/pharmacology , Adrenergic beta-3 Receptor Antagonists/pharmacology , Animals , Astrocytes/metabolism , Astrocytoma , Brain/cytology , Buthionine Sulfoximine/pharmacology , Cell Line, Tumor , Coculture Techniques , Dioxoles/pharmacology , Humans , Hydrogen Peroxide/toxicity , Mice , Mice, Inbred C57BL , Neuroblastoma , Oxidative Stress , Propanolamines/pharmacology , Propionates/pharmacology , Quinolines/pharmacology
6.
Front Aging Neurosci ; 12: 58, 2020.
Article in English | MEDLINE | ID: mdl-32296325

ABSTRACT

Age-associated loss of retinal ganglion cells (RGCs) causes visual deficits, but there is not yet any therapeutic agent to prevent the loss of these cells. Herein, we report that apelin, an endogenous peptide ligand of APJ receptor, is protective against the age-related loss of RGCs in mice. The mRNA expression of apelin was reduced in the retina of old mice compared with that in young mice, whereas retinal APJ expression increased with age. Immunofluorescence staining showed that APJ was present in RGCs and their surrounding cells expressed apelin. In addition, both functional and histological analyses demonstrated that apelin deficiency accelerated the loss of RGCs associated with age in mice. These results suggest that endogenous apelin plays a protective role against the degeneration of RGCs and that the apelinergic axis may be a new target for preventing age-related visual impairment.

7.
Neurochem Res ; 45(4): 752-759, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31894462

ABSTRACT

Glutamate excitotoxicity via N-methyl-D-aspartate (NMDA) receptors is thought to be a factor involved in the loss of retinal neuronal cells, including retinal ganglion cells, in retinal diseases such as diabetic retinopathy and acute angle closure glaucoma. Herein we report the protective effect of systemic administration of ML233, an apelin receptor agonist, against retinal neuronal cell death induced by the intravitreal injection of NMDA into mice. Intraperitoneal administration of ML233 prevented the NMDA-induced reduction in the amplitude of scotopic threshold responses (STR), which mainly reflect the activity of the retinal ganglion cells. Immunohistochemical staining showed that ML233 inhibited the NMDA-induced loss of retinal ganglion cells and amacrine cells. In addition, ML233 suppressed the breakdown of spectrin αII, a neuronal cytoskeleton protein cleaved by calpain activation, in the retina after intravitreal injection of NMDA. Intraperitoneal administration of ML233 increased the phosphorylation of Akt, a potent anti-apoptotic protein in neurons, in the retina. Furthermore, oral administration of ML233 protected against the decrease in the STR amplitudes and the loss of retinal ganglion cells caused by NMDA. These results suggest that systemic administration of ML233 protected retinal neurons from NMDA receptor-mediated excitotoxicity and that drugs activating the apelin receptor may be a new candidate for preventing the progression of these retinal diseases.


Subject(s)
Apelin Receptors/agonists , Imines/pharmacology , Mesylates/pharmacology , N-Methylaspartate/toxicity , Retinal Diseases/prevention & control , Retinal Neurons/drug effects , Administration, Oral , Animals , Imines/administration & dosage , Injections, Intraperitoneal , Intravitreal Injections , Male , Mesylates/administration & dosage , Mice, Inbred C57BL , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Retinal Diseases/metabolism , Retinal Neurons/metabolism
8.
Eur J Pharmacol ; 866: 172826, 2020 Jan 05.
Article in English | MEDLINE | ID: mdl-31790652

ABSTRACT

Many reports have indicated that dopamine has immunomodulatory effects on peripheral immune cells. The purpose of this study was to reveal the immunomodulatory effect of dopamine on the expression of proinflammatory cytokines in microglial cells, which are the immune cells of the central nervous system. In murine microglial cell line BV-2 cells, pretreatment with dopamine for 24 h attenuated the lipopolysaccharide (LPS)-induced expression of proinflammatory cytokines such as tumor-necrosis factor-α, interleukin-1ß, and interleukin-6. Neither (5R)-8-chloro-3-methyl-5-phenyl-1,2,4,5-tetrahydro-3-benzazepin-7-ol; hydrochloride (SCH-23390) nor sulpiride, which are dopamine D1-like and D2-like receptor antagonists, respectively, affected the attenuation of LPS-induced expression of cytokines by dopamine. In addition, pretreatment with neither (-)-(6aR,12bR)-4,6,6a,7,8,12b-Hexahydro-7-methylindolo[4,3-a]phenanthridin (CY208-243) nor bromocriptine, dopamine D1-like and D2-like receptor agonists, respectively, was effective in doing so. However, N-acetylcysteine (NAC), which inhibits dopamine oxidation to dopamine quinone, did inhibit this attenuated expression. Dopamine increased the level of quinoproteins, and this increase was inhibited by NAC. Western blot and immunocytochemical analyses revealed that dopamine inhibited LPS-induced nuclear translocation of nuclear factor-kappa B (NF-κB) p65. Dopamine also attenuated the expression of cytokines and the nuclear translocation of NF-κB p65 induced by LPS in mouse microglial cells in primary culture. These results suggest that dopamine attenuated LPS-induced expression of cytokines by inhibiting the nuclear translocation of NF-κB p65 through the formation of dopamine quinone in microglial cells.


Subject(s)
Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cytokines/metabolism , Dopamine/analogs & derivatives , Dopamine/pharmacology , Microglia/drug effects , Transcription Factor RelA/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , Cell Line , Dopamine/biosynthesis , Gene Expression Regulation/drug effects , Lipopolysaccharides/pharmacology , Mice , Microglia/cytology , Microglia/metabolism
9.
Sci Rep ; 7(1): 15062, 2017 11 08.
Article in English | MEDLINE | ID: mdl-29118394

ABSTRACT

Pathological retinal angiogenesis is caused by the progression of ischemic retinal diseases and can result in retinal detachment and irreversible blindness. This neovascularization is initiated from the retinal veins and their associated capillaries and involves the overgrowth of vascular endothelial cells. Since expression of the apelin receptor (APJ) is restricted to the veins and proliferative endothelial cells during physiological retinal angiogenesis, in the present study, we investigated the effect of APJ inhibition on pathological retinal angiogenesis in a mouse model of oxygen-induced retinopathy (OIR). In vitro experiments revealed that ML221, an APJ antagonist, suppressed cultured-endothelial cell proliferation in a dose-dependent manner. Intraperitoneal administration of ML221 inhibited pathological angiogenesis but enhanced the recovery of normal vessels into the ischemic regions in the retina of the OIR model mice. ML221 did not affect the expression levels of vascular endothelial growth factor (VEGF) and its receptor (VEGFR2) in the retina. APJ was highly expressed in the endothelial cells within abnormal vessels but was only detected in small amounts in morphologically normal vessels. These results suggest that APJ inhibitors selectively prevent pathological retinal angiogenesis and that the drugs targeting APJ may be new a candidate for treating ischemic retinopathy.


Subject(s)
Apelin Receptors/antagonists & inhibitors , Nitrobenzoates/pharmacology , Pyrans/pharmacology , Retinal Diseases/prevention & control , Retinal Neovascularization/prevention & control , Animals , Apelin/genetics , Apelin/metabolism , Apelin Receptors/genetics , Apelin Receptors/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Endothelial Cells/cytology , Endothelial Cells/drug effects , Gene Expression/drug effects , Ischemia/genetics , Ischemia/metabolism , Ischemia/prevention & control , Mice , Retinal Diseases/genetics , Retinal Diseases/metabolism , Retinal Neovascularization/genetics , Retinal Neovascularization/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
10.
J Pharmacol Sci ; 133(1): 34-41, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28087150

ABSTRACT

Glutamate excitotoxicity mediated by N-methyl-d-aspartate (NMDA) receptors is an important cause of retinal ganglion cell death in glaucoma. To elucidate whether apelin protects against retinal neuronal cell death, we examined protective effects of exogenous and endogenous apelin on neuronal cell death induced by intravitreal injection of NMDA in the retinas of mice. An intravitreal injection of NMDA induced neuronal cell death in both the retinal ganglion cell layer and inner nuclear layer, and reduced the amplitudes of scotopic threshold response (STR) in electroretinography studies. Both cell death and STR amplitudes decrease induced by NMDA were prevented by a co-injection of [Pyr1]-apelin-13, and were facilitated by apelin deficiency. The neuroprotective effects of [Pyr1]-apelin-13 were blocked by an apelin receptor APJ antagonist, and by inhibitors of Akt and extracellular signal-regulated kinase 1/2 signaling pathways. Additionally, an intravitreal injection of tumor necrosis factor-α (TNF-α) neutralizing antibody prevented NMDA-induced retinal neuronal cell death, and exogenous and endogenous apelin suppressed NMDA-induced upregulation of TNF-α in the retina. These results suggest that apelin protects neuronal cells against NMDA-induced death via an APJ receptor in the retina, and that apelin may have beneficial effects in the treatment of glaucoma.


Subject(s)
Cell Death/drug effects , Intercellular Signaling Peptides and Proteins/pharmacology , MAP Kinase Signaling System/drug effects , N-Methylaspartate/toxicity , Proto-Oncogene Proteins c-akt/metabolism , Receptors, G-Protein-Coupled/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Apelin Receptors , Intravitreal Injections , Male , Mice , N-Methylaspartate/administration & dosage , N-Methylaspartate/antagonists & inhibitors , Neurons/drug effects , Neuroprotective Agents/pharmacology , Night Vision/drug effects , Retina/drug effects , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology
11.
J Pharmacol Sci ; 130(2): 51-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26908040

ABSTRACT

Dopamine (DA) has been suggested to modulate functions of glial cells including microglial cells. To reveal the regulatory role of DA in microglial function, in the present study, we investigated the effect of DA on lipopolysaccharide (LPS)-induced nitric oxide (NO) production in murine microglial cell line BV-2. Pretreatment with DA for 24 h concentration-dependently attenuated LPS-induced NO production in BV-2 cells. The inhibitory effect of DA on LPS-induced NO production was not inhibited by SCH-23390 and sulpiride, D1-like and D2-like DA receptor antagonists, respectively. In addition, pretreatment with (-)-(6aR,12bR)-4,6,6a,7,8,12b-Hexahydro-7-methylindolo[4,3-a]phenanthridin (CY 208-243) and bromocriptine, D1-like and D2-like DA receptor agonists, respectively, did not affect the LPS-induced NO production. N-Acetylcysteine, which inhibits DA oxidation, completely inhibited the effect of DA. Tyrosinase, which catalyzes the oxidation of DA to DA quionone (DAQ), accelerated the inhibitory effect of DA on LPS-induced NO production. These results suggest that DA attenuates LPS-induced NO production through the formation of DAQ in BV-2 cells.


Subject(s)
Dopamine/analogs & derivatives , Dopamine/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Microglia/metabolism , Nitric Oxide/metabolism , Acetylcysteine/pharmacology , Animals , Cells, Cultured , Dopamine/metabolism , Dopamine Antagonists , Drug Synergism , Lipopolysaccharides/pharmacology , Mice , Monophenol Monooxygenase/pharmacology , Oxidation-Reduction/drug effects
12.
Eur J Pharmacol ; 772: 51-61, 2016 Feb 05.
Article in English | MEDLINE | ID: mdl-26724392

ABSTRACT

Glutathione (GSH) plays a critical role in protecting cells from oxidative damage. Since neurons rely on the supply of GSH from astrocytes to maintain optimal intracellular GSH concentrations, the GSH concentration of astrocytes is important for the survival of neighboring neurons against oxidative stress. The neurotransmitter noradrenaline is known to modulate the functions of astrocytes and has been suggested to have neuroprotective properties in neurodegenerative diseases. To elucidate the mechanisms underlying the neuroprotective properties of noradrenaline, in this study, we investigated the effect of noradrenaline on the concentrations of intracellular GSH in human U-251 malignant glioma (MG; astrocytoma) cells. Treatment of the cells with noradrenaline for 24h concentration-dependently increased their intracellular GSH concentration. This increase was inhibited by a non-selective ß-adrenoceptor antagonist propranolol and by a selective ß3-adrenoceptor antagonist SR59230A, but not by a non-selective α-adrenoceptor antagonist phenoxybenzamine, or by a selective ß1-adrenoceptor antagonist atenolol or by a selective ß2-adrenoceptor antagonist butoxamine. In addition, the selective ß3-adrenoceptor agonist CL316243 increased the intracellular GSH in U-251 MG cells. Treatment of the cells with noradrenaline (10µM) for 24h increased the protein level of the catalytic subunit of glutamate-cysteine ligase (GCLc), the rate-limiting enzyme of GSH synthesis; and this increase was inhibited by SR59230A. These results thus suggest that noradrenaline increased the GSH concentration in astrocytes by inducing GCLc protein in them via ß3-adrenoceptor stimulation.


Subject(s)
Astrocytoma/pathology , Glutamate-Cysteine Ligase/metabolism , Glutathione/metabolism , Intracellular Space/drug effects , Intracellular Space/metabolism , Norepinephrine/pharmacology , Receptors, Adrenergic, beta-3/metabolism , Adrenergic beta-3 Receptor Antagonists/pharmacology , Cell Line, Tumor , Gene Expression Regulation, Enzymologic/drug effects , Humans
13.
Bioconjug Chem ; 26(8): 1782-90, 2015 Aug 19.
Article in English | MEDLINE | ID: mdl-26252905

ABSTRACT

We have been investigating the potential use of cell-penetrating peptide-linked polymers as a novel penetration enhancer. Since previous in vivo studies demonstrated that poly(N-vinylacetamide-co-acrylic acid) bearing D-octaarginine, a typical cell-penetrating peptide, enhanced membrane permeation of biomolecules, its potential as an in vitro transfection tool was evaluated in this study. A plasmid DNA encoding green fluorescent protein (pGFP-C1), ß-galactosidase, and bovine serum albumin (BSA) were used as model biomolecules. Anionic pGFP-C1 interacted electrostatically with cationic d-octaarginine-linked polymers. When the ratio of mass concentration of polymers to that of pGFP-C1 reached 2.5, complexes whose size and zeta potential were approximately 200 nm and 15 mV, respectively, were obtained. GFP expression was observed in cells incubated with complexes prepared under conditions in which the polymer/pDNA concentration ratio exceeded 2.5. The expression level elevated with an increase in the concentration ratio, but physicochemical properties of the complexes remained unchanged. Results suggested that free polymers contributed to pGFP-C1 internalization. Another cell study demonstrated that ß-galactosidase premixed with polymers was taken up into cells in its active tetrameric form. Similar electrostatic interaction-driven complex formation was observed for BSA charged negatively in neutral solution. However, it appeared that the internalization processes of BSA differed from those of pGFP-C1. A mass concentration-dependent increase in internalized BSA was observed, irrespective of the polymer/protein concentration ratio. Due to frail interactions, polymers that were released from the complexes and subsequently immobilized on cell membranes might also contribute to membrane permeation of BSA.


Subject(s)
Green Fluorescent Proteins/metabolism , Oligopeptides/chemistry , Plasmids/administration & dosage , Polymers/chemistry , Serum Albumin, Bovine/metabolism , beta-Galactosidase/metabolism , Animals , Cattle , Cell Membrane Permeability , Drug Carriers/chemistry , Green Fluorescent Proteins/genetics , HeLa Cells , Humans , Serum Albumin, Bovine/genetics , Transfection , beta-Galactosidase/genetics
14.
Pediatr Surg Int ; 30(5): 499-502, 2014 May.
Article in English | MEDLINE | ID: mdl-24626878

ABSTRACT

PURPOSE: A prospective study was conducted to confirm the safety and efficacy of the selective sac extraction method (SSEM) of inguinal hernia repairs in children. METHODS: Primary endpoints of the study were the incidence of any complication related to the SSEM, or hernia recurrence. Secondary endpoints included the success rate of the SSEM, length of incision at the end of operation, and duration of operation. The incidence of contralateral manifestation of hernia was also examined. RESULTS: Between October 2009 and December 2011, a total of 317 repairs, 145 male repairs and 172 female repairs, were performed by applying the SSEM. There were three operative conversions, and the success rate of the SSEM was 99% in both male and female patients. The length of incision ranged from 4.0 to 12.5 mm (median 6.0 mm) and was ≤7.0 mm in 93% repairs. The incisional length for male repairs ranged from 4.0 to 12.5 mm (median 6.0 mm) and was ≤7.0 mm in 86% repairs, while it ranged from 4.0 to 9.0 mm (median 5.5 mm) in female repairs and was ≤6.5 mm in 96% repairs. The duration of the operation for unilateral repair ranged from 9 to 66 min (median 21 min). Eighty percent of repairs were examined 6-44 months (median 12 months) after the operation. There was one (0.4%) recurrence among 250 repairs and two (1.7%) cases of testicular dislocation among 115 male repairs. Contralateral hernia presented in 19 (9.5%) of 199 patients with unilateral hernia who underwent the follow-up. CONCLUSIONS: The feasibility of the SSEM was reconfirmed, and it was revealed that the complication and recurrence rates were low and acceptable. The SSEM is safe and effective, and should be a standard method for repairing inguinal hernia in children.


Subject(s)
Hernia, Inguinal/surgery , Minimally Invasive Surgical Procedures/methods , Postoperative Complications/epidemiology , Adolescent , Child , Child, Preschool , Feasibility Studies , Female , Follow-Up Studies , Hernia, Inguinal/mortality , Humans , Incidence , Infant , Japan/epidemiology , Male , Minimally Invasive Surgical Procedures/adverse effects , Prospective Studies , Recurrence , Treatment Outcome , Wound Healing
15.
Angiogenesis ; 16(3): 723-34, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23640575

ABSTRACT

The recruitment of mural cells such as pericytes to patent vessels with an endothelial lumen is a key factor for the maturation of blood vessels and the prevention of hemorrhage in pathological angiogenesis. To date, our understanding of the specific trigger underlying the transition from cell growth to the maturation phase remains incomplete. Since rapid endothelial cell growth causes pericyte loss, we hypothesized that suppression of endothelial growth factors would both promote pericyte recruitment, in addition to inhibiting pathological angiogenesis. Here, we demonstrate that targeted knockdown of apelin in endothelial cells using siRNA induced the expression of monocyte chemoattractant protein-1 (MCP-1) through activation of Smad3, via suppression of the PI3K/Akt pathway. The conditioned medium of endothelial cells treated with apelin siRNA enhanced the migration of vascular smooth muscle cells, through MCP-1 and its receptor pathway. Moreover, in vivo delivery of siRNA targeting apelin, which causes exuberant endothelial cell proliferation and pathological angiogenesis through its receptor APJ, led to increased pericyte coverage and suppressed pathological angiogenesis in an oxygen-induced retinopathy model. These data demonstrate that apelin is not only a potent endothelial growth factor, but also restricts pericyte recruitment, establishing a new connection between endothelial cell proliferation signaling and a trigger of mural recruitment.


Subject(s)
Chemokine CCL2/metabolism , Endothelial Cells/cytology , Intercellular Signaling Peptides and Proteins/metabolism , Neovascularization, Pathologic/physiopathology , Retinal Vessels/physiopathology , Adipokines , Analysis of Variance , Animals , Apelin , Apelin Receptors , Blotting, Western , Culture Media, Conditioned/pharmacology , Endothelial Cells/metabolism , Gene Knockdown Techniques , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/genetics , Mice , Muscle, Smooth, Vascular/metabolism , RNA, Small Interfering/pharmacology , Real-Time Polymerase Chain Reaction , Receptors, G-Protein-Coupled/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Smad3 Protein/metabolism , Tumor Cells, Cultured
16.
Surg Today ; 43(7): 800-5, 2013 Jul.
Article in English | MEDLINE | ID: mdl-22825653

ABSTRACT

We herein report a case of cystic-type congenital biliary dilatation (CBD) in whom an extremely rare anomalous duplication of the common bile duct and pancreaticobiliary maljunction were diagnosed intraoperatively by meticulous surgical manipulations via conventional open surgery. By performing a dissection at the outer epicholedochal layer of the cyst, a thin cord-like structure shown to be the distal part of the common bile duct was identified. A further exploration revealed that the most distal (extra- and intrapancreatic) part of the common bile duct was duplicated, and each branch of the duct was connected to the main and accessory pancreatic ducts. The experience with our case and a literature review showed that extrahepatic bile duct duplication is generally associated with pancreaticobiliary maljunction and CBD. We conclude that an extremely careful exploration with delicate and meticulous surgical manipulation is essential to identify these morphological anomalies and prevent intraoperative and postoperative complications of CBD, such as pancreatic duct injury or pancreatitis.


Subject(s)
Abnormalities, Multiple , Bile Ducts, Extrahepatic/abnormalities , Bile Ducts, Extrahepatic/surgery , Biliary Tract/pathology , Choledochal Cyst/surgery , Pancreatic Ducts/abnormalities , Pancreatic Ducts/surgery , Biliary Tract Surgical Procedures , Choledochal Cyst/diagnosis , Dilatation, Pathologic/congenital , Female , Humans , Infant , Intraoperative Complications/prevention & control , Intraoperative Period , Postoperative Complications/prevention & control
17.
Masui ; 61(9): 925-30; discussion 930-1, 2012 Sep.
Article in Japanese | MEDLINE | ID: mdl-23012829

ABSTRACT

In this review article, we discussed the pathogenesis, pathophysiology, diagnosis and treatment of acute appendicitis in children. Indications for early surgery, the operative methods of laparoscopic appendectomy and the treatment outcome are also presented.


Subject(s)
Appendicitis/therapy , Emergency Medical Services , Acute Disease , Adolescent , Anesthesia/methods , Appendectomy/methods , Appendicitis/diagnosis , Appendicitis/etiology , Appendicitis/physiopathology , Child , Child, Preschool , Humans , Laparoscopy/methods , Tomography, X-Ray Computed , Treatment Outcome , Ultrasonography, Doppler, Color
18.
Eur J Pharm Biopharm ; 81(1): 64-73, 2012 May.
Article in English | MEDLINE | ID: mdl-22306700

ABSTRACT

We are investigating a new class of penetration enhancers that enable poorly membrane-permeable molecules physically mixed with them to effectively penetrate cell membranes without their concomitant cellular uptake. Since we previously revealed that poly(N-vinylacetamide-co-acrylic acid) modified with d-octaarginine, which is a typical cell-penetrating peptide, significantly enhanced the nasal absorption of insulin, we examined the performance of the polymers on cell membranes. When Caco-2 cells were incubated with 5(6)-carboxyfluorescein (CF) for 30 min, approximately 0.1% of applied CF was internalized into the cells. This poor membrane permeability was dramatically enhanced by d-octaarginine-linked polymers; a 25-fold increase in the cellular uptake of CF was observed when the polymer concentration was adjusted to 0.2mg/mL. None of the individual components, for example, d-octaarginine, had any influence on CF uptake, demonstrating that only d-octaarginine anchored chemically to the polymeric platform enhanced the membrane permeation of CF. The polymer-induced CF uptake was consistently high even when the incubation time was extended to 120 min. Confocal laser scanning microphotographs of cells incubated with d-octaarginine-linked polymers bearing rhodamine red demonstrated that the cell outline was stained with red fluorescence. The polymer-induced CF uptake was significantly suppressed by 5-(N-ethyl-N-isopropyl)amiloride, which is an inhibitor of macropinocytosis. Results indicated that d-octaarginine-linked polymers remained on the cell membrane and poorly membrane-permeable CF was continuously internalized into cells mainly via macropinocytosis repeated for the individual peptidyl branches in the polymer backbone.


Subject(s)
Acetamides/chemistry , Cell Membrane Permeability/drug effects , Fluoresceins/pharmacokinetics , Oligopeptides/chemistry , Polyvinyls/chemistry , Acrylates/chemistry , Amiloride/analogs & derivatives , Amiloride/pharmacology , Caco-2 Cells , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell-Penetrating Peptides/chemistry , Fluorescent Dyes/chemistry , Humans , Microscopy, Confocal , Pinocytosis/drug effects , Rhodamines/chemistry , Time Factors
19.
PLoS One ; 6(8): e23968, 2011.
Article in English | MEDLINE | ID: mdl-21887354

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by the selective loss of motor neurons. Recent studies have implicated that chronic hypoxia and insufficient vascular endothelial growth factor (VEGF)-dependent neuroprotection may lead to the degeneration of motor neurons in ALS. Expression of apelin, an endogenous ligand for the G protein-coupled receptor APJ, is regulated by hypoxia. In addition, recent reports suggest that apelin protects neurons against glutamate-induced excitotoxicity. Here, we examined whether apelin is an endogenous neuroprotective factor using SOD1(G93A) mouse model of ALS. In mouse CNS tissues, the highest expressions of both apelin and APJ mRNAs were detected in spinal cord. APJ immunoreactivity was observed in neuronal cell bodies located in gray matter of spinal cord. Although apelin mRNA expression in the spinal cord of wild-type mice was not changed from 4 to 18 weeks age, that of SOD1(G93A) mice was reduced along with the paralytic phenotype. In addition, double mutant apelin-deficient and SOD1(G93A) displayed the disease phenotypes earlier than SOD1(G93A) littermates. Immunohistochemical observation revealed that the number of motor neurons was decreased and microglia were activated in the spinal cord of the double mutant mice, indicating that apelin deficiency pathologically accelerated the progression of ALS. Furthermore, we showed that apelin enhanced the protective effect of VEGF on H(2)O(2)-induced neuronal death in primary neurons. These results suggest that apelin/APJ system in the spinal cord has a neuroprotective effect against the pathogenesis of ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Intercellular Signaling Peptides and Proteins/deficiency , Age Factors , Animals , Apelin , Disease Progression , Intercellular Signaling Peptides and Proteins/analysis , Intercellular Signaling Peptides and Proteins/genetics , Mice , Motor Neurons/pathology , Neuroprotective Agents , RNA, Messenger/analysis , Spinal Cord/chemistry , Tissue Distribution
20.
Arterioscler Thromb Vasc Biol ; 30(11): 2182-7, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20705920

ABSTRACT

OBJECTIVE: To investigate the role of endogenous apelin in pathological retinal angiogenesis. METHODS AND RESULTS: The progression of ischemic retinal diseases, such as diabetic retinopathy, is closely associated with pathological retinal angiogenesis, mainly induced by vascular endothelial growth factor (VEGF) and erythropoietin. Although antiangiogenic therapies using anti-VEGF drugs are effective in treating retinal neovascularization, they show a transient efficacy and cause general adverse effects. New therapeutic target molecules are needed to resolve these issues. It was recently demonstrated that the apelin/APJ system, a newly deorphanized G protein-coupled receptor system, is involved in physiological retinal vascularization. Retinal angiography and mRNA expression were examined during hypoxia-induced retinal angiogenesis in a mouse model of oxygen-induced retinopathy. Compared with age-matched control mice, retinal apelin expression was dramatically increased during the hypoxic phase in oxygen-induced retinopathy model mice. APJ was colocalized in proliferative cells, which were probably endothelial cells of the ectopic vessels in the vitreous body. Apelin deficiency hardly induced hypoxia-induced retinal angiogenesis despite the upregulation of VEGF and erythropoietin mRNA in oxygen-induced retinopathy model mice. Apelin small and interfering RNA suppressed the proliferation of endothelial cells independent of the VEGF/VEGF receptor 2 signaling pathway. CONCLUSIONS: These results suggest that apelin is a prerequisite factor for hypoxia-induced retinal angiogenesis.


Subject(s)
Carrier Proteins/physiology , Hypoxia/complications , Retinal Neovascularization/physiopathology , Adipokines , Animals , Apelin , Carrier Proteins/adverse effects , Disease Models, Animal , Intercellular Signaling Peptides and Proteins , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/physiopathology , Retinal Neovascularization/etiology
SELECTION OF CITATIONS
SEARCH DETAIL
...