Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 129
Filter
1.
Sci Rep ; 14(1): 9700, 2024 04 27.
Article in English | MEDLINE | ID: mdl-38678148

ABSTRACT

Ocular abnormal angiogenesis and edema are featured in several ocular diseases. S1P signaling via S1P1 likely is part of the negative feedback mechanism necessary to maintain vascular health. In this study, we conducted pharmacological experiments to determine whether ASP4058, a sphingosine 1-phosphate receptor 1/5 (S1P1/5) agonist, is useful in abnormal vascular pathology in the eye. First, human retinal microvascular endothelial cells (HRMECs) were examined using vascular endothelial growth factor (VEGF)-induced cell proliferation and hyperpermeability. ASP4058 showed high affinity and inhibited VEGF-induced proliferation and hyperpermeability of HRMECs. Furthermore, S1P1 expression and localization changes were examined in the murine laser-induced choroidal neovascularization (CNV) model, a mouse model of exudative age-related macular degeneration, and the efficacy of ASP4058 was verified. In the CNV model mice, S1P1 tended to decrease in expression immediately after laser irradiation and colocalized with endothelial cells and Müller glial cells. Oral administration of ASP4058 also suppressed vascular hyperpermeability and CNV, and the effect was comparable to that of the intravitreal administration of aflibercept, an anti-VEGF drug. Next, efficacy was also examined in a retinal vein occlusion (RVO) model in which retinal vascular permeability was increased. ASP4058 dose-dependently suppressed the intraretinal edema. In addition, it suppressed the expansion of the perfusion area observed in the RVO model. ASP4058 also suppressed the production of VEGF in the eye. Collectively, ASP4058 can be a potential therapeutic agent that normalizes abnormal vascular pathology, such as age-related macular degeneration and RVO, through its direct action on endothelial cells.


Subject(s)
Choroidal Neovascularization , Disease Models, Animal , Animals , Humans , Choroidal Neovascularization/drug therapy , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Mice , Sphingosine-1-Phosphate Receptors/metabolism , Sphingosine-1-Phosphate Receptors/agonists , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Vascular Endothelial Growth Factor A/metabolism , Cell Proliferation/drug effects , Mice, Inbred C57BL , Receptors, Lysosphingolipid/agonists , Receptors, Lysosphingolipid/metabolism , Male
2.
Biochem Biophys Res Commun ; 711: 149897, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38608433

ABSTRACT

PURPOSE: Dipeptidyl peptidase-4 (DPP-4) inhibitors are oral hypoglycemic drugs and are used for type II diabetes. Previous studies showed that DPP-4 expression is observed in several tumor types and DPP-4 inhibitors suppress the tumor progression on murine tumor models. In this study, we evaluated the role of DPP-4 and the antitumor effect of a DPP-4 inhibitor, linagliptin, on glioblastoma (GBM). METHODS: We analyzed DPP-4 expression in glioma patients by the public database. We also analyzed DPP-4 expression in GBM cells and the murine GBM model. Then, we evaluated the cell viability, cell proliferation, cell migration, and expression of some proteins on GBM cells with linagliptin. Furthermore, we evaluated the antitumor effect of linagliptin in the murine GBM model. RESULTS: The upregulation of DPP-4 expression were observed in human GBM tissue and murine GBM model. In addition, DPP-4 expression levels were found to positively correlate with the grade of glioma patients. Linagliptin suppressed cell viability, cell proliferation, and cell migration in GBM cells. Linagliptin changed the expression of phosphorylated NF-kB, cell cycle, and cell adhesion-related proteins. Furthermore, oral administration of linagliptin decreases the tumor progression in the murine GBM model. CONCLUSION: Inhibition of DPP-4 by linagliptin showed the antitumor effect on GBM cells and the murine GBM model. The antitumor effects of linagliptin is suggested to be based on the changes in the expression of several proteins related to cell cycle and cell adhesion via the regulation of phosphorylated NF-kB. This study suggested that DPP-4 inhibitors could be a new therapeutic strategy for GBM.


Subject(s)
Brain Neoplasms , Cell Movement , Cell Proliferation , Dipeptidyl Peptidase 4 , Dipeptidyl-Peptidase IV Inhibitors , Disease Progression , Glioblastoma , Linagliptin , Glioblastoma/drug therapy , Glioblastoma/pathology , Glioblastoma/metabolism , Linagliptin/pharmacology , Linagliptin/therapeutic use , Animals , Humans , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Dipeptidyl Peptidase 4/metabolism , Mice , Cell Proliferation/drug effects , Cell Movement/drug effects , Cell Line, Tumor , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Male , Cell Survival/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
3.
J Pharmacol Sci ; 155(2): 44-51, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38677785

ABSTRACT

Subretinal hemorrhages result in poor vision and visual field defects. During hemorrhage, several potentially toxic substances are released from iron-based hemoglobin and hemin, inducing cellular damage, the detailed mechanisms of which remain unknown. We examined the effects of excess intracellular iron on retinal pigment epithelial (RPE) cells. A Fe2+ probe, SiRhoNox-1 was used to investigate Fe2+ accumulation after treatment with hemoglobin or hemin in the human RPE cell line ARPE-19. We also evaluated the production of reactive oxygen species (ROS) and lipid peroxidation. Furthermore, the protective effect of-an iron chelator, 2,2'-bipyridyl (BP), and ferrostatin-1 (Fer-1) on the cell damage, was evaluated. Fe2+ accumulation increased in the hemoglobin- or hemin-treated groups, as well as intracellular ROS production and lipid peroxidation. In contrast, BP treatment suppressed RPE cell death, ROS production, and lipid peroxidation. Pretreatment with Fer-1 ameliorated cell death in a concentration-dependent manner and suppressed ROS production and lipid peroxidation. Taken together, these findings indicate that hemoglobin and hemin, as well as subretinal hemorrhage, may induce RPE cell damage and visual dysfunction via intracellular iron accumulation.


Subject(s)
Hemin , Hemoglobins , Iron , Retinal Pigment Epithelium , Humans , Cell Death/drug effects , Cell Line , Cyclohexylamines/pharmacology , Hemin/pharmacology , Hemoglobins/metabolism , Iron/metabolism , Iron Chelating Agents/pharmacology , Lipid Peroxidation/drug effects , Phenylenediamines/pharmacology , Reactive Oxygen Species/metabolism , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/pathology
4.
BMC Complement Med Ther ; 24(1): 3, 2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38167061

ABSTRACT

BACKGROUND: Blue light exposure is known to induce reactive oxygen species (ROS) production and increased endoplasmic reticulum stress, leading to apoptosis of photoreceptors. Maqui berry (Aristotelia chilensis) is a fruit enriched in anthocyanins, known for beneficial biological activities such as antioxidation. In this study, we investigated the effects of Maqui berry extract (MBE) and its constituents on the subcellular damage induced by blue light irradiation in mouse retina-derived 661W cells. METHODS: We evaluated the effects of MBE and its main delphinidins, delphinidin 3-O-sambubioside-5-O-glucoside (D3S5G) and delphinidin 3,5-O-diglucoside (D3G5G), on blue light-induced damage on retinal cell line 661W cells. We investigated cell death, the production of ROS, and changes in organelle morphology using fluorescence microscopy. The signaling pathway linked to stress response was evaluated by immunoblotting in the whole cell lysates or nuclear fractions. We also examined the effects of MBE and delphinidins against rotenone-induced mitochondrial dysfunction. RESULTS: Blue light-induced cell death, increased intracellular ROS generation and mitochondrial fragmentation, decreased ATP-production coupled respiration, caused lysosomal membrane permeabilization, and increased ATF4 protein level. Treatment with MBE and its main constituents, delphinidin 3-O-sambubioside-5-O-glucoside and delphinidin 3,5-O-diglucoside, prevented these defects. Furthermore, MBE and delphinidins also protected 661W cells from rotenone-induced cell death. CONCLUSIONS: Maqui berry may be a useful protective agent for photoreceptors against the oxidative damage induced by exposure to blue light.


Subject(s)
Anthocyanins , Elaeocarpaceae , Animals , Mice , Anthocyanins/pharmacology , Reactive Oxygen Species/metabolism , Fruit , Blue Light , Rotenone , Elaeocarpaceae/metabolism , Glucosides , Organelles/metabolism
5.
J Pharmacol Sci ; 154(2): 52-60, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38246728

ABSTRACT

Many glaucoma treatments focus on lowering intraocular pressure (IOP), with novel drugs continuing to be developed. One widely used model involves raising IOP by applying a laser to the trabecular iris angle (TIA) of cynomolgus monkeys to damage the trabecular meshwork. This model, however, presents challenges such as varying IOP values, potential trabecular meshwork damage, and risk of animal distress. This study investigated whether animals with naturally high IOP (>25 mmHg) could be used to effectively evaluate IOP-lowering drugs, thereby possibly replacing laser-induced models. Relationships between TIA size, IOP, and pupil diameter were also examined. Three representative IOP-lowering drugs (latanoprost, timolol, ripasudil) were administered, followed by multiple IOP measurements and assessment of corneal thickness, TIA, and pupil diameter via anterior segment optical coherence tomography (AS-OCT). There was a positive correlation was noted between IOP and corneal thickness before instillation, and a negative correlation between IOP and TIA before instillation. Our findings suggest animals with naturally high IOP could be beneficial for glaucoma research and development as a viable replacement for the laser-induced model and that measuring TIA using AS-OCT along with IOP yields a more detailed evaluation.


Subject(s)
Glaucoma , Intraocular Pressure , Animals , Macaca fascicularis , Timolol/pharmacology , Trabecular Meshwork
6.
Biochem Biophys Res Commun ; 691: 149341, 2024 Jan 08.
Article in English | MEDLINE | ID: mdl-38039836

ABSTRACT

Myocardial infarction (MI) induces structural and electrical cardiac remodeling in response to ischemic insult, causing lethal arrhythmias and sudden death. Progranulin (PGRN) is a glycoprotein mainly expressed in macrophages that modulates the immune responses. In this study, we investigated the direct influence of PGRN knockout (Grn-/-) macrophages on post-MI pathophysiology. An MI mouse model was established by ligating the left coronary artery for RNA sequencing and electrocardiographic analysis. Bone marrow-derived macrophages (BMDMs) were injected into mice and supernatant was collected for the measurement of reactive oxygen species (ROS) levels and extracellular flux analysis. Administration of Grn-/- BMDMs prolonged the QT intervals in the MI mouse model. Moreover, genes highly expressed in macrophages were upregulated in Grn-/- heart after MI. Post-hypoxic supernatant of Grn-/- BMDMs increased the oxygen-glucose deprivation-induced cardiomyocyte death. Grn-/- BMDMs exhibited increased ROS production, oxygen consumption, and extracellular acidification under hypoxia and inflammatory conditions. These findings suggest that PGRN deficiency causes cardiotoxicity via secretory components of macrophages that exhibit metabolic abnormalities under hypoxia.


Subject(s)
Cardiotoxicity , Myocardial Infarction , Mice , Animals , Progranulins/metabolism , Cardiotoxicity/metabolism , Reactive Oxygen Species/metabolism , Macrophages/metabolism , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , Disease Models, Animal , Hypoxia/genetics , Hypoxia/metabolism
7.
FASEB Bioadv ; 5(10): 395-411, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37810172

ABSTRACT

Myocardial infarction (MI) is a lethal disease that causes irreversible cardiomyocyte death and subsequent cardiovascular remodeling. We have previously shown that the administration of recombinant progranulin (PGRN) protects against myocardial ischemia and reperfusion injury. However, the post-MI role of PGRN remains unclear. In the present study, we investigated the effects of PGRN deficiency on cardiac remodeling after MI. Wild-type and PGRN-knockout mice were subjected to MI by ligation of the left coronary artery for histological, electrophysiological, and protein expression analysis. Cardiac macrophage subpopulations were analyzed by flow cytometry. Bone marrow-derived macrophages (BMDMs) were acquired and treated with LPS + IFN-γ and IL-4 to evaluate mRNA levels and phagocytic ability. PGRN expression was gradually increased in the whole heart at 1, 3, and 7 days after MI. Macrophages abundantly expressed PGRN at the border areas at 3 days post-MI. PGRN-knockout mice showed higher mortality, increased LV fibrosis, and severe arrhythmia following MI. PGRN deficiency increased the levels of CD206 and MerTK expression and macrophage infiltration in the infarcted myocardium, which was attributed to a larger subpopulation of cardiac CCR2+ Ly6Clow CD11b+ macrophages. PGRN-deficient BMDMs exhibited higher TGF-ß, IL-4R, and lower IL-1ß, IL-10 and increased acute phagocytosis following stimulation of LPS and IFN-γ. PGRN deficiency reduced survival and increased cardiac fibrosis following MI with the induction of abnormal subpopulation of cardiac macrophages early after MI, thereby providing insight into the relationship between properly initiating cardiac repair and macrophage polarization after MI.

8.
Biol Pharm Bull ; 46(8): 1032-1040, 2023.
Article in English | MEDLINE | ID: mdl-37532554

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder characterized by motor symptoms and neuropathological features, such as loss of dopaminergic neurons in the substantia nigra pars compacta and accumulation of alpha-synuclein (α-Syn). Progranulin (PGRN) is a secreted growth factor that exhibits anti-inflammatory properties and regulates lysosomal function. Although autophagy-lysosome pathway is the main degradative pathway for α-Syn, the molecular mechanistic relationship between PD and PGRN remains unclear. In this study, we investigated the role of PGRN in PD pathology. PGRN protein expression in striatum was increased in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD model mice. Intracerebroventricular (i.c.v.) administration of PGRN ameliorated the decrease in expression of tyrosine hydroxylase, a dopaminergic neuron marker, in MPTP-treated mice. Furthermore, i.c.v. administration of PGRN ameliorated 6-hydroxydopamine-induced motor deficits. In SH-SY5Y human neuroblastoma cells, 1-methyl-4-phenylpyridinium ion (MPP+), an active metabolite of MPTP, increased α-Syn expression. In contrast, PGRN ameliorated MPP+-induced increase in α-Syn expression. Although PGRN decreased the levels of autophagy-related proteins Sequestosome-1 (p62) and MAP1LC3 (LC3)-II, PGRN did not influence the phosphorylation of AMP-activated protein kinase and mechanistic target of rapamycin, which are also proteins that regulate autophagy. Immunostaining analysis showed that PGRN ameliorated MPP+-induced increase of LC3 puncta, indicator of autophagosome, and co-localization of LC3 and α-Syn. The DALGreen assay showed that PGRN ameliorated MPP+-induced decreasing trend of autolysosomes. These results suggest that PGRN participates in α-Syn degradation via acceleration of the autophagy-lysosome pathway and is a potential therapeutic target for PD.


Subject(s)
Neuroblastoma , Parkinson Disease , Animals , Humans , Mice , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/adverse effects , alpha-Synuclein/metabolism , Disease Models, Animal , Dopaminergic Neurons/metabolism , Lysosomes/metabolism , Mice, Inbred C57BL , Neuroblastoma/metabolism , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Progranulins/metabolism
9.
Arterioscler Thromb Vasc Biol ; 43(6): 927-942, 2023 06.
Article in English | MEDLINE | ID: mdl-37078291

ABSTRACT

BACKGROUND: Endothelial cell activation is tightly controlled by the balance between VEGF (vascular endothelial cell growth factor) and Notch signaling pathway. VEGF destabilizes blood vessels and promotes neovascularization, which are common features of sight-threatening ocular vascular disorders. Here, we show that BCL6B (B-cell CLL/lymphoma 6 member B protein), also known as BAZF, ZBTB28, and ZNF62, plays a pivotal role in the development of retinal edema and neovascularization. METHODS: The pathophysiological physiological role of BCL6B was investigated in cellular and animal models mimicking 2 pathological conditions: retinal vein occlusion and choroidal neovascularization. An in vitro experimental system was used in which human retinal microvascular endothelial cells were supplemented with VEGF. Choroidal neovascularization cynomolgus monkey model was generated to investigate the involvement of BCL6B in the pathogenesis. Mice lacking BCL6B or treated with BCL6B-targeting small-interfering ribose nucleic acid were examined for histological and molecular phenotypes. RESULTS: In retinal endothelial cells, the BCL6B expression level was increased by VEGF. BCL6B-deficient endothelial cells showed Notch signal activation and attenuated cord formation via blockage of the VEGF-VEGFR2 signaling pathway. Optical coherence tomography images showed that choroidal neovascularization lesions were decreased by BCL6B-targeting small-interfering ribose nucleic acid. Although BCL6B mRNA expression was significantly increased in the retina, BCL6B-targeting small-interfering ribose nucleic acid suppressed ocular edema in the neuroretina. The increase in proangiogenic cytokines and breakdown of the inner blood-retinal barrier were abrogated in BCL6B knockout (KO) mice via Notch transcriptional activation by CBF1 (C promotor-binding factor 1) and its activator, the NICD (notch intracellular domain). Immunostaining showed that Müller cell activation, a source of VEGF, was diminished in BCL6B-KO retinas. CONCLUSIONS: These data indicate that BCL6B may be a novel therapeutic target for ocular vascular diseases characterized by ocular neovascularization and edema.


Subject(s)
Choroidal Neovascularization , Nucleic Acids , Retinal Neovascularization , Vascular Diseases , Animals , Humans , Mice , Choroidal Neovascularization/genetics , Choroidal Neovascularization/metabolism , Endothelial Cells/metabolism , Macaca fascicularis/metabolism , Nucleic Acids/metabolism , Nucleic Acids/therapeutic use , Retinal Neovascularization/genetics , Retinal Neovascularization/metabolism , Ribose/metabolism , Ribose/therapeutic use , Vascular Diseases/pathology , Vascular Endothelial Growth Factor A/metabolism
10.
Biol Pharm Bull ; 46(3): 473-481, 2023.
Article in English | MEDLINE | ID: mdl-36858576

ABSTRACT

Macular edema causes vision loss in patients with retinal vein occlusion (RVO) and diabetic macular edema (DME). The intravitreal injection of anti-vascular endothelial growth factor (VEGF) agents is used for treatment; however, this therapy is invasive, and recurrence occurs in some cases. The establishment of a non-invasive treatment would help to solve these problems. Here, we focused on arctigenin, a lignan polyphenol found in burdock sprout, and has effects on inflammatory and microcirculatory when taken orally. We hypothesized that oral intake of arctigenin could be effective against retinal edema in RVO and DME. In this study, the degree of retinal edema by measuring the total retinal thickness using optical coherence tomography (OCT) and the thickness of the inner nuclear layer (INL) by hematoxylin-eosin (H&E) staining were investigated. Oral administration of arctigenin ameliorated retinal edema in an RVO murine model by inhibiting the decrease in occludin and vascular endothelial (VE)-cadherin. Moreover, in retinas with edema, arctigenin suppressed the induction of VEGF, tumor necrosis factor α (TNFα), and matrix metallopeptidase 9 (MMP9). Next, the effects of arctigenin on barrier function were assessed in human retinal microvascular endothelial cells (HRMECs) by measuring the trans-endothelial electrical resistance (TEER) and conducting fluorescein isothiocyanate (FITC)-dextran permeability assays. Arctigenin showed a protective effect against VEGF-induced barrier dysfunction. In addition, arctigenin inhibited the TNFα-mediated activation of the nuclear factor-kappaB (NF-κB)/p38 mitogen-activated protein kinase (MAPK) pathway. These results suggested that oral administration of arctigenin has beneficial effects on retinal edema by inhibiting vascular hyperpermeability in endothelial cells.


Subject(s)
Diabetic Retinopathy , Lignans , Macular Edema , Papilledema , Retinal Vein Occlusion , Humans , Animals , Mice , Endothelial Cells , Microcirculation , Tumor Necrosis Factor-alpha , Vascular Endothelial Growth Factor A
11.
J Endocrinol ; 257(3)2023 06 01.
Article in English | MEDLINE | ID: mdl-36988989

ABSTRACT

VGF nerve growth factor inducible (VGF) is a secreted polypeptide involved in metabolic regulation. VGF-derived peptides have been reported to regulate insulin secretion in the plasma of patients with type 2 diabetes and model mice. However, the protective effects of VGF on pancreatic ß-cells in diabetic model are not well understood. In this study, we aimed to elucidate the ß-cell protective effect of VGF on a streptozotocin (STZ)-induced diabetic model using VGF-overexpressing (OE) mice and also examined the therapeutic effect by a small molecule, SUN N8075 which is an inducer of VGF. VGF-OE mice improved blood glucose levels and maintained ß-cell mass compared to wild-type (WT) mice on STZ-induced diabetic model. In addition, VGF-OE mice showed better glucose tolerance than WT mice. In culture, AQEE-30, a VGF-derived peptide, suppressed STZ-induced ß-cell death in vitro and attenuated the decrease in the phosphorylation of Akt and GSK3ß. Furthermore, SUN N8075 suppressed the blood glucose levels and increased VGF expression in the pancreatic islet. SUN N8075 also protected STZ-induced ß-cell death in vitro. These findings indicate that VGF plays a hypoglycemic role in response to blood glucose levels in diabetes and protects ß-cells from STZ-induced cell death. Therefore, VGF and its inducer have the therapeutic potential by preserving ß-cells in diabetes.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Mice , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/metabolism , Aniline Compounds/pharmacology , Piperazines/metabolism , Piperazines/pharmacology , Streptozocin , Insulin/metabolism , Insulin-Secreting Cells/metabolism
12.
Biol Pharm Bull ; 46(2): 177-186, 2023.
Article in English | MEDLINE | ID: mdl-36724946

ABSTRACT

Huntington's disease (HD) is a hereditary neurodegenerative disease that involves an expansion of the CAG repeats of the Huntingtin (HTT) gene, but the disease onset and progression do not necessarily correspond to the extent of CAG repeats. Decreased mitochondrial complex II activity has also been reported to be closely associated with disease pathogenesis. Here, we examined the mechanism of cell death induced by 3-nitropropionic acid (3-NP), a mitochondrial complex II inhibitor, using striatal cells (STHdhQ111 cells) derived from HD model mice with mutant HTT carrying the CAG repeat extended. Treatment with 3-NP (5 mM) enhanced cell death in STHdhQ111 compared to STHdhQ7 cells with normal HTT. Ferrostatin-1, an inhibitor of ferroptosis, and deferoxamine, an iron chelator, markedly inhibited 3-NP-induced cell death in both the STHdh cell lines. On the other hands, cell death was not abrogated by a broad-spectrum caspase inhibitor, Z-VAD-FMK, indicating that this cell death was caspase-independent. Cell death caused by 3-NP is suggested to be due to ferroptosis. Furthermore, 3-NP-induced cell death was markedly inhibited by GSK2795039, a reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 2 (NOX2) inhibitor, suggesting that cell death is mainly mediated by intracellular superoxide anion (O2-) production through NOX2. Furthermore, a mitochondria-targeted superoxide dismutase mimetic (Mito-TEMPO), partially inhibited 3-NP-induced cell death, suggesting that O2- production in the mitochondria is partially responsible for cell death. These results indicate that 3-NP-induced cell death in the STHdhQ111 cells is caspase-independent, non-apoptotic, and that ferroptotic cell death is mainly induced via NOX2 activation.


Subject(s)
Huntington Disease , Neurodegenerative Diseases , Mice , Animals , Reactive Oxygen Species/metabolism , Mice, Transgenic , Nuclear Proteins/genetics , Nerve Tissue Proteins/metabolism , Caspases/metabolism , Huntington Disease/chemically induced , Huntington Disease/genetics , Huntington Disease/metabolism
13.
Exp Eye Res ; 228: 109405, 2023 03.
Article in English | MEDLINE | ID: mdl-36773739

ABSTRACT

This study aimed to determine the role of transient receptor potential vanilloid 4 (TRPV4), a calcium (Ca2+)-permeable cation channel, in the pathophysiology of retinal vascular disease. The retinal vein occlusion (RVO) murine model was created by irradiating retinal veins using lasers. TRPV4 expression and localization were evaluated in RVO mice retinas. In addition, we examined the effects of TRPV4 antagonists (RQ-00317310, HC-067047, GSK2193874, and GSK2798745) on retinal edema, blood flow, and ischemic areas in RVO mice. Furthermore, changes in the retinal expression of tumor necrosis factor (TNF)-α and aquaporin4 (AQP4) by RQ-00317310 were analyzed using Western blot. We also assessed the barrier integrity of epithelial cell monolayers using trans-endothelial electrical resistance (TEER) in Human Retinal Microvascular Endothelial Cells (HRMECs). The expression of TRPV4 was significantly increased and co-localized with glutamine synthetase (GS), a Müller glial marker, in the ganglion cell layer (GCL) of the RVO mice. Moreover, RQ-00317310 administration ameliorated the development of retinal edema and ischemia in RVO mice. In addition, the up regulation of TNF-α and down-regulation of AQP4 were lessened by the treatment with RQ-00317310. Treatment with GSK1016790A, a TRPV4 agonist, increased vascular permeability, while RQ-00317310 treatment decreased vascular endothelial growth factor (VEGF)- or TRPV4-induced retinal vascular hyperpermeability in HRMECs. These findings suggest that TRPV4 plays a role in the development of retinal edema and ischemia. Thus, TRPV4 could be a new therapeutic target against the pathological symptoms of retinal vascular diseases.


Subject(s)
Papilledema , Retinal Diseases , Transient Receptor Potential Channels , Vascular Diseases , Mice , Humans , Animals , Capillary Permeability , TRPV Cation Channels/metabolism , Transient Receptor Potential Channels/metabolism , Transient Receptor Potential Channels/pharmacology , Endothelial Cells/metabolism , Papilledema/metabolism , Vascular Endothelial Growth Factor A/metabolism , Retinal Diseases/metabolism , Vascular Diseases/metabolism
14.
Exp Eye Res ; 227: 109382, 2023 02.
Article in English | MEDLINE | ID: mdl-36634838

ABSTRACT

Intraocular pressure (IOP) is the most important risk factor for the onset and progression of glaucoma. IOP reduction has been proven effective in the treatment of glaucoma. IOP is controlled by the production and outflow of the aqueous humor (AH), and the trabecular meshwork (TM) is the main pathway for AH drainage from the eye. However, there are few conventional IOP-lowering treatments that target TM, and there is a need for such treatments. In this study, we screened for the expression level of fibronectin as an indicator and identified an activin receptor-like kinase (ALK) 5 inhibitor. Western blot analysis showed that SB431542, an ALK 5 inhibitor, reduced fibronectin and α-SMA expression. Moreover, a single dose of the ALK5 inhibitor SB431542 reduced IOP in mice, and the IOP-lowering effect of the ALK5 inhibitor was greater than that of a Rho-associated coiled-coil-containing protein kinase inhibitor (Y-27632). Repeated dosing with ALK5 inhibitor eye drops (once daily) enhanced the murine IOP-lowering effect. Furthermore, ALK5 inhibition decreased the expression of extracellular matrix (ECM) mRNA and suppressed ECM production. These findings suggest that ALK5 inhibitors may contribute to the development of new treatments for glaucoma that target the TM.


Subject(s)
Glaucoma , Trabecular Meshwork , Mice , Animals , Trabecular Meshwork/metabolism , Intraocular Pressure , Fibronectins/metabolism , Aqueous Humor/metabolism , Glaucoma/drug therapy , Glaucoma/metabolism , Receptor Protein-Tyrosine Kinases/metabolism
15.
J Control Release ; 353: 216-228, 2023 01.
Article in English | MEDLINE | ID: mdl-36410615

ABSTRACT

The production of reactive oxygen species (ROS) during and after the onset of an ischemic stroke induces neuronal cell death and severely damages brain function. Therefore, reducing ROS by administrating antioxidant compounds is a promising approach to improving ischemic symptoms. Alpha-mangostin (α-M) is an antioxidant compound extracted from the pericarp of the mangosteen fruit. Reportedly, α-M decreases neuronal toxicity in primary rat cerebral cortical neurons. In this study, we investigated the neuroprotective activity of α-M in both in vitro and in vivo assays. Pretreatment with α-M inhibited excessive cellular ROS production after oxygen-glucose deprivation/reoxygenation (OGD/R) in vitro using an SH-SY5Y (human neuroblastoma) cell line. In addition, α-M maintained mitochondrial membrane potential and suppressed mitochondrial-specific ROS production induced by OGD/R. Meanwhile, the low bioavailability of α-M due to its poor water solubility has been an insuperable obstruction impeding extensive investigations of the biological functions of α-M and its medical applications. To overcome this problem, we synthesized a cyclodextrin-based nanoparticle (CDNP) that is known to increase the loading efficiency and binding constant of α-M, compared with cyclodextrins themselves. This nano-formulated α-M (α-M/CDNP) was optimized for an in vivo ischemic stroke model. Our results indicated that α-M/CDNP (25 mg/kg/injection) reduced infarct volume and improved neurological behavior (p = 0.036 and p = 0.046, respectively). These in vivo results suggest that α-M appears to cross the blood-brain barrier (BBB) with the help of a nano-formulation with CDNP. Combining an in vitro BBB model and a physicochemical binding assay between α-M and albumin, it is speculated that α-M released from CDNP would interact with albumin during its prolonged circulation in the blood, and the resultant α-M/albumin complex may cross the BBB through the absorptive-mediated transcytosis pathway. These findings suggest the potential clinical application of α-M in ischemic stroke treatment.


Subject(s)
Brain Ischemia , Cyclodextrins , Ischemic Stroke , Neuroblastoma , Neuroprotective Agents , Reperfusion Injury , Rats , Humans , Animals , Reactive Oxygen Species/metabolism , Antioxidants/pharmacology , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Oxygen/therapeutic use , Glucose/metabolism , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/pharmacology , Reperfusion Injury/metabolism , Apoptosis
16.
Cell Mol Neurobiol ; 43(2): 879-892, 2023 Mar.
Article in English | MEDLINE | ID: mdl-35435536

ABSTRACT

Sodium-glucose transporter 2 (SGLT2) inhibitors are antidiabetic drugs affecting SGLT2. Recent studies have shown various cancers expressing SGLT2, and SGLT2 inhibitors attenuating tumor proliferation. We evaluated the antitumor activities of canagliflozin, a SGLT2 inhibitor, on glioblastoma (GBM). Three GBM cell lines, U251MG (human), U87MG (human), and GL261 (murine), were used. We assessed the expression of SGLT2 of GBM through immunoblotting, specimen-use, cell viability assays, and glucose uptake assay with canagliflozin. Then, we assessed phosphorylation of AMP-activated protein kinase (AMPK), p70 S6 kinase, and S6 ribosomal protein by immunoblotting. Concentrations of 5, 10, 20, and 40 µM canagliflozin were used in these tests. We also evaluated cell viability and immunoblotting using U251MG with siRNA knockdown of SGLT2. Furthermore, we divided the mice into vehicle group and canagliflozin group. The canagliflozin group was administrated with 100 mg/kg of canagliflozin orally for 10 days starting from the third days post-GBM transplant. The brains were removed and the tumor volume was evaluated using sections. SGLT2 was expressed in GBM cell and GBM allograft mouse. Canagliflozin administration at 40 µM significantly inhibited cell proliferation and glucose uptake into the cell. Additionally, canagliflozin at 40 µM significantly increased the phosphorylation of AMPK and suppressed that of p70 S6 kinase and S6 ribosomal protein. Similar results of cell viability assays and immunoblotting were obtained using siRNA SGLT2. Furthermore, although less effective than in vitro, the canagliflozin group significantly suppressed tumor growth in GBM-transplanted mice. This suggests that canagliflozin can be used as a potential treatment for GBM.


Subject(s)
Glioblastoma , Sodium-Glucose Transporter 2 Inhibitors , Humans , Mice , Animals , Canagliflozin/pharmacology , Canagliflozin/therapeutic use , AMP-Activated Protein Kinases/metabolism , Glioblastoma/drug therapy , Sodium-Glucose Transporter 2/genetics , Sodium-Glucose Transporter 2/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Cell Proliferation , Glucose/metabolism , Ribosomal Proteins/metabolism
17.
Mol Vis ; 29: 188-196, 2023.
Article in English | MEDLINE | ID: mdl-38222457

ABSTRACT

Purpose: To investigate the therapeutic effects of eye drops, namely, timolol maleate, a ß-adrenergic receptor antagonist, and latanoprost, a prostaglandin F2α analog, on retinal edema in a murine retinal vein occlusion (RVO) model. Methods: An RVO model was established using laser-induced RVO in mice, which were administered timolol maleate and latanoprost eye drops several times after venous occlusion. Subsequently, the thickness of the inner nuclear layer (INL) and the expression levels of such genes as Vegf and Atf4, which are stress markers of the endoplasmic reticulum, were examined. Primary human cultured retinal microvascular endothelial cells (HRMECs) were treated with timolol under hypoxic conditions, after which the gene expression pattern was investigated. Importantly, an integrated stress response inhibitor (ISRIB) was used in the RVO model, he known ISRIB, which suppresses the expression of ATF4 in retinal edema. Results: Increased INL thickness was suppressed by timolol eye drops, as were the expressions of Vegf and Atf4, in the RVO model. However, latanoprost eye drops did not induce any change in INL thickness. In HRMECs, hypoxic stress and serum deprivation increased the Vegf and Atf4 expressions; in response, treatment with timolol suppressed the Vegf expression. Furthermore, the ISRIB decreased the Vegf expression pattern and edema formation, which are associated with RVO. Conclusions: These results indicate that timolol eye drops may be a potential option for RVO treatment.


Subject(s)
Papilledema , Retinal Vein Occlusion , Male , Humans , Mice , Animals , Timolol/pharmacology , Timolol/therapeutic use , Timolol/metabolism , Retinal Vein Occlusion/complications , Retinal Vein Occlusion/drug therapy , Retinal Vein Occlusion/metabolism , Ophthalmic Solutions/therapeutic use , Latanoprost/pharmacology , Latanoprost/metabolism , Latanoprost/therapeutic use , Papilledema/drug therapy , Endothelial Cells/metabolism , Vascular Endothelial Growth Factor A/metabolism , Edema/complications
18.
J Pharmacol Sci ; 150(4): 279-288, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36344051

ABSTRACT

The corneal epithelium is located at the outermost layer of the ocular surface and continuously exposed to environmental factors, such as ultraviolet (UV) radiation from sunlight. UV irradiation causes excessive production of reactive oxygen species (ROS) in cells, which results in oxidative damage to membrane-bound organelles such as mitochondria, eventually leading to cell death. Crocetin, a natural carotenoid found in plants, has various biological properties including antioxidant activity. In this study, we investigated the effects of crocetin on UV-A-induced cell injury in the corneal epithelium. Using an in vitro system with the human corneal epithelial cell-transformed (HCE-T) cell line, pretreatment with 10 µM crocetin suppressed the reduction of cell viability induced by UV-A exposure. Crocetin ameliorated the decrease in oxygen consumption rates and the mitochondrial fragmentation that occurred following UV-A irradiation. Crocetin inhibited both ROS production and the activation of the apoptosis pathway; it also preserved the defects of epithelial cell polarity and barrier function in UV-A-irradiated HCE-T cells. The reduction in apical Mucin-16 expression was partially recovered in the presence of crocetin. Taking these findings together, we conclude that crocetin has a protective effect against UV-A irradiation-induced mitochondrial injury in corneal epithelial cells.


Subject(s)
Epithelial Cells , Ultraviolet Rays , Humans , Reactive Oxygen Species/metabolism , Ultraviolet Rays/adverse effects , Epithelial Cells/metabolism , Oxidative Stress , T-Lymphocytes/metabolism
19.
J Neurooncol ; 160(2): 375-388, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36308592

ABSTRACT

PURPOSE: The alkylating agent temozolomide (TMZ) has a significant impact on the prognosis of glioblastoma (GBM) patients. Therefore, maximizing TMZ efficacy is important for GBM treatment. Many reports have shown that glutamate signaling promotes GBM progression via glutamate receptors, including N-methyl-D-aspartate receptors (NMDARs). Although NMDARs promote cell migration and invasion of GBM cells, their role in TMZ resistance remains unclear. Therefore, we focused on NMDAR signaling and investigated its effects on TMZ resistance. METHODS: We investigated the effect of NMDAR signaling on O6-methylguanine DNA methyltransferase (MGMT), a DNA repair enzyme that induces chemoresistance to TMZ, using quantitative real-time polymerase chain reaction and western blotting in human GBM T98G cells. In addition, we used memantine (MEM), an NMDAR antagonist, to investigate the cytotoxic effect of TMZ/MEM combination and its detailed mechanism. RESULTS: Activation of NMDAR by N-methyl-D-aspartate (NMDA) elevated MGMT expression and suppressed the effect of TMZ in T98G cells. In contrast, knockdown of NMDAR by NMDAR1 shRNA decreased MGMT expression and enhanced the effect of TMZ in T98G cells. The cytotoxic effect of TMZ was enhanced by MEM in T98G cells. Inhibition of NMDAR by MEM decreased MGMT expression and increased DNA alkylation by TMZ. CONCLUSION: NMDAR signaling induced chemoresistance of TMZ via the upregulation of MGMT expression in GBM cells. Furthermore, MEM inhibited TMZ-induced MGMT upregulation and increased the cytotoxic effect of TMZ on MGMT-positive cells. This study demonstrates that the combination of TMZ and MEM could be a new therapeutic strategy for MGMT-positive GBM. Overview of this study. NMDAR signaling controls the expression of MGMT and the cytotoxic effect of TMZ.


Subject(s)
Antineoplastic Agents , Glioblastoma , Humans , Temozolomide/pharmacology , Temozolomide/therapeutic use , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, N-Methyl-D-Aspartate/therapeutic use , Drug Resistance, Neoplasm , Up-Regulation , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , DNA Modification Methylases/metabolism , O(6)-Methylguanine-DNA Methyltransferase/genetics , DNA Repair Enzymes/metabolism , Antineoplastic Agents/therapeutic use , DNA/pharmacology , DNA/therapeutic use , Cell Line, Tumor , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
20.
Invest Ophthalmol Vis Sci ; 63(8): 14, 2022 07 08.
Article in English | MEDLINE | ID: mdl-35822950

ABSTRACT

Purpose: Semaphorin 3A (Sema3A) is a promising therapeutic target for macular edema in age-related macular degeneration, diabetic retinopathy, and retinal vein occlusion (RVO). Anti-vascular endothelial growth factors (anti-VEGFs) are the current standard of care for many retinal diseases. This study investigated the Sema3A neutralizing antibody BI-X and/or anti-VEGF therapy (aflibercept) in an RVO mouse model. Treatment efficacy was examined and grouped by timing subsequent to the RVO mouse model induction: efficacy against the onset of intraretinal edema 1 day postinduction and protective effects at 7 days postinduction. Methods: We examined the changes in expression of Sema3A in the retina of an RVO mouse model. In addition, changes in expression of tumor necrosis factor (TNF)-α and semaphorin-related proteins (neuropilin-1 and plexin A1) in the retina upon treatment were analyzed by Western blotting. The effects of BI-X and/or aflibercept were evaluated using measures of retinal edema, blood flow, and thinning of the inner nuclear layer. Results: Induction of vein occlusion in the RVO mouse model significantly increased Sema3A expression in the retina, particularly in the inner nuclear layer. BI-X was effective as a monotherapy and in combination with anti-VEGF therapy, demonstrating a beneficial effect on intraretinal edema and retinal blood flow. Moreover, in the RVO mouse model, BI-X monotherapy normalized the changes in expression of TNF-α and semaphorin-related proteins. Conclusions: These findings support targeting Sema3A to treat intraretinal edema and retinal ischemia.


Subject(s)
Macular Edema , Retinal Diseases , Retinal Vein Occlusion , Angiogenesis Inhibitors/therapeutic use , Animals , Antibodies, Neutralizing/pharmacology , Antibodies, Neutralizing/therapeutic use , Disease Models, Animal , Intravitreal Injections , Macular Edema/drug therapy , Male , Mice , Retina/pathology , Retinal Diseases/pathology , Retinal Vein Occlusion/metabolism , Semaphorin-3A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...