Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Clin Kidney J ; 16(12): 2614-2625, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38045997

ABSTRACT

Background: Upacicalcet is a novel small-molecule calcimimetic agent developed for intravenous injection. Here, we evaluated the long-term efficacy and safety of upacicalcet treatment via intraindividual dose adjustment in haemodialysis patients with secondary hyperparathyroidism (SHPT). Methods: A phase 2, multicentre, open-label, single-arm study was conducted. Upacicalcet was administered for 52 weeks; the starting dose was 50 µg thrice a week, and then adjusted to 25, 50, 100, 150, 200, 250, or 300 µg, according to the dose-adjustment method set in the protocol. The primary endpoint was the percentage of patients with serum intact parathyroid hormone (iPTH) level achieving a target range of 60-240 pg/mL (target achievement rate) at week 18. Results: A total of 58 patients were administered upacicalcet. The target achievement rate of serum iPTH level at week 18 was 57.9%, which increased to 80.8% at week 52. The serum-corrected calcium (cCa) level decreased immediately after upacicalcet administration, but no further decrease was observed. Adverse events were observed in 94.8% of patients, and adverse drug reactions (ADRs) occurred in 20.7% of patients. The most common ADR was decreased adjusted calcium in eight patients; dizziness occurred as a serious ADR in one patient. The serum cCa level of patients who interrupted upacicalcet treatment at a serum cCa level of <7.5 mg/dL recovered to ≥7.5 mg/dL immediately after the interruption. Conclusions: In haemodialysis patients with SHPT, upacicalcet doses of 25-300 µg for 52 weeks were found to be highly effective and well-tolerated, with minor safety concerns.

2.
Biosci Biotechnol Biochem ; 81(9): 1699-1705, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28743229

ABSTRACT

Type 2 diabetes mellitus (T2DM) is a common global health problem. Prevention of this disease is an important task, and functional food supplements are considered an effective method. We found potent pancreatic α-amylase inhibition in Astilbe thunbergii root extract (AT) and confirmed that AT treatment in a T2DM rat model reduces post-starch administration blood glucose levels. Activity-guided isolation revealed procyanidin (AT-P) as the α-amylase inhibitory component with IC50 = 1.7 µg/mL against porcine pancreatic α-amylase. Structure analysis of AT-P revealed it is a B-type procyanidin comprised of four types of flavan-3-ols, some with a galloyl group, and catechin attached as the terminal unit. The abundant AT-P content and its comparable α-amylase inhibition to acarbose, the anti-diabetic medicine, suggest that AT is a promising food supplement for diabetes prevention.


Subject(s)
Biflavonoids/pharmacology , Catechin/pharmacology , Diabetes Mellitus, Type 2/complications , Glycoside Hydrolase Inhibitors/pharmacology , Hyperglycemia/complications , Hyperglycemia/drug therapy , Proanthocyanidins/pharmacology , Saxifragaceae/chemistry , Animals , Biflavonoids/isolation & purification , Biflavonoids/therapeutic use , Catechin/isolation & purification , Catechin/therapeutic use , Disease Models, Animal , Glucagon-Like Peptide-1 Receptor/blood , Glycoside Hydrolase Inhibitors/isolation & purification , Glycoside Hydrolase Inhibitors/therapeutic use , Hyperglycemia/blood , Male , Proanthocyanidins/isolation & purification , Proanthocyanidins/therapeutic use , Rats
3.
Biosci Biotechnol Biochem ; 80(11): 2087-2092, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27477520

ABSTRACT

Dipeptidyl peptidase-IV (DPP-IV) is a protease responsible for the degradation of the incretin hormone. A number of DPP-IV inhibitors have been approved for use in the treatment of type 2 diabetes. While these inhibitors are effective for this treatment, methods for the prevention of this disease are also required as diabetes patient numbers are currently increasing rapidly worldwide. We screened the DPP-IV inhibitory activities of edible plant extracts with the intention of using these extracts in a functional food supplement for the prevention of diabetes. Rose (Rosa gallica) bud extract powder was a promising material with high inhibitory activity. In this study, seven ellagitannins were isolated as active compounds through activity-guided fractionations, and their DPP-IV inhibitory activities were measured. Among them, rugosin A and B showed the highest inhibitory activities and rugosin B was shown as the major contributing compound in rose bud extract powder.

4.
Bioorg Med Chem ; 23(13): 3317-21, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25943853

ABSTRACT

Hypoglycemic effect is an efficient means to modulate elevated blood glucose levels in patients with diabetes. We found that the extract of lotus plumule (the germ of Nelumbo nucifera Gaertn. seed) showed potent glucose uptake enhancement activity against L6 myotubes, which results in a hypoglycemic effect. This activity was further investigated, and an active constituent was identified as a single bioactive compound, higenamine 4'-O-ß-d-glucoside. Mechanistic studies employing phosphatidylinositol 3-kinase (PI3K) inhibitor, AMP-activated protein kinase (AMPK) inhibitor, or adrenergic receptor antagonist showed that the compound induced its activity through ß2-adrenergic receptor. Patients with type II diabetes mellitus frequently develop insulin resistance. Owing to the differences between the mechanism of action of insulin and of the isolated compound, the compound or lotus plumule itself may have the possibility of modulating blood glucose levels in insulin-resistant patients effectively.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Alkaloids/chemistry , Glucose/metabolism , Glucosides/pharmacology , Hypoglycemic Agents/pharmacology , Nelumbo/chemistry , Receptors, Adrenergic, beta-2/metabolism , Tetrahydroisoquinolines/chemistry , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Adrenergic Antagonists/pharmacology , Adrenergic beta-Agonists/chemistry , Adrenergic beta-Agonists/isolation & purification , Animals , Cell Line , Chromones/pharmacology , Gene Expression Regulation , Glucosides/chemistry , Glucosides/isolation & purification , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/isolation & purification , Mice , Morpholines/pharmacology , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Plant Extracts/chemistry , Propranolol/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Receptors, Adrenergic, beta-2/genetics , Seeds/chemistry , Signal Transduction
5.
J Biol Chem ; 282(26): 19052-61, 2007 Jun 29.
Article in English | MEDLINE | ID: mdl-17412698

ABSTRACT

Sex-determining region Y-box (SOX) 6 negatively regulates glucose-stimulated insulin secretion from beta-cells and is a down-regulated transcription factor in the pancreatic islet cells of hyperinsulinemic obese mice. To determine the contribution of SOX6 to insulin resistance, we analyzed the effects of SOX6 on cell proliferation. Small interfering RNA-mediated attenuation of SOX6 expression stimulated the proliferation of insulinoma INS-1E and NIH-3T3 cells, whereas retroviral overexpression resulted in inhibition of cell growth. Quantitative real time-PCR analysis revealed that the levels of cyclin D1 transcripts were markedly decreased by SOX6 overexpression. Luciferase-reporter assay with beta-catenin showed that SOX6 suppresses cyclin D1 promoter activities. In vitro binding experiments showed that the LZ/Q domain of SOX6 physically interacts with armadillo repeats 1-4 of beta-catenin. Furthermore, chromatin immunoprecipitation assay revealed that increased SOX6 expression significantly reduced the levels of acetylated histones H3 and H4 at the cyclin D1 promoter. By using a histone deacetylase (HDAC) inhibitor and co-immunoprecipitation analysis, we showed that SOX6 suppressed cyclin D1 activities by interacting withbeta-catenin and HDAC1. The data presented suggest that SOX6 may be an important factor in obesity-related insulin resistance.


Subject(s)
Cyclin D1/metabolism , DNA-Binding Proteins/metabolism , High Mobility Group Proteins/metabolism , Histone Deacetylases/metabolism , Insulin-Secreting Cells/physiology , Transcription Factors/metabolism , beta Catenin/metabolism , Animals , Cell Division/physiology , Cell Line, Tumor , Cyclin D1/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Down-Regulation/physiology , High Mobility Group Proteins/chemistry , High Mobility Group Proteins/genetics , Histone Deacetylase 1 , Histones/metabolism , Humans , Hyperinsulinism/metabolism , Hyperinsulinism/physiopathology , Insulin Resistance/physiology , Insulin-Secreting Cells/cytology , Insulinoma , Kidney/cytology , Leucine Zippers/physiology , Mice , NIH 3T3 Cells , Obesity/metabolism , Obesity/physiopathology , Pancreatic Neoplasms , Promoter Regions, Genetic/physiology , Protein Structure, Tertiary , Rats , SOXD Transcription Factors , Transcription Factors/chemistry , Transcription Factors/genetics , Transduction, Genetic
6.
J Biol Chem ; 281(29): 20213-20, 2006 Jul 21.
Article in English | MEDLINE | ID: mdl-16707486

ABSTRACT

Pigment epithelium-derived factor (PEDF) is the most potent inhibitor of angiogenesis, suggesting that loss of PEDF contributes to proliferative diabetic retinopathy. However, the role of PEDF against retinal vascular hyperpermeability remains to be elucidated. We investigated here whether and how PEDF could inhibit the advanced glycation end product (AGE) signaling to vascular hyperpermeability. Intravenous administration of AGEs to normal rats not only increased retinal vascular permeability by stimulating vascular endothelial growth factor (VEGF) expression but also decreased retinal PEDF levels. Simultaneous treatments with PEDF inhibited the AGE-elicited VEGF-mediated permeability by down-regulating mRNA levels of p22(phox) and gp91(phox), membrane components of NADPH oxidase, and subsequently decreasing retinal levels of an oxidative stress marker, 8-hydroxydeoxyguanosine. PEDF also inhibited the AGE-induced vascular hyperpermeability evaluated by transendothelial electrical resistance by suppressing VEGF expression. Furthermore, PEDF decreased reactive oxygen species (ROS) generation in AGE-exposed endothelial cells by suppressing NADPH oxidase activity via down-regulation of mRNA levels of p22(PHOX) and gp91(PHOX). This led to blockade of the AGE-elicited Ras activation and NF-kappaB-dependent VEGF gene induction in endothelial cells. These results indicate that the central mechanism for PEDF inhibition of the AGE signaling to vascular permeability is by suppression of NADPH oxidase-mediated ROS generation and subsequent VEGF expression. Substitution of PEDF may offer a promising strategy for halting the development of diabetic retinopathy.


Subject(s)
Capillary Permeability/physiology , Eye Proteins/physiology , Glycation End Products, Advanced/antagonists & inhibitors , Nerve Growth Factors/physiology , Reactive Oxygen Species/metabolism , Retinal Vessels/physiology , Serpins/physiology , Vascular Endothelial Growth Factor A/genetics , Adult , Animals , Diabetic Retinopathy/physiopathology , Electrophysiology , Female , Humans , Male , Middle Aged , Rats , Reference Values , Signal Transduction/physiology
7.
J Bone Miner Res ; 20(9): 1647-58, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16059636

ABSTRACT

UNLABELLED: The impact of AGEs on human MSCs was studied. AGEs inhibited the proliferation of MSCs, induced apoptosis, and prevented cognate differentiation into adipose tissue, cartilage, and bone, suggesting a deleterious effect of AGEs in the pathogenesis of musculoskeletal disorders in aged and diabetic patients. INTRODUCTION: Advanced glycation end-products (AGEs) are accumulated on long-lived proteins of various tissues in advanced age and diabetes mellitus and have been implicated in chronic complication, including musculoskeletal disorders. Human mesenchymal stem cells (MSCs) potentially differentiate into mature musculoskeletal tissues during tissue repair, but the pathogenetic role of AGEs on MSCs is unclear. MATERIALS AND METHODS: AGEs were prepared by incubating BSA with glucose, glyceraldehydes, or glycolaldehyde (designated as AGE-1, AGE-2, or AGE-3, respectively). Proliferation, apoptosis, and reactive oxygen species (ROS) generation were assayed in AGE-treated cells. The expression of the receptor for AGE (RAGE) was examined by immunohistochemistry and Western blotting. Involvement of RAGE-mediated signaling was examined using a neutralizing antiserum against RAGE. Differentiation into adipose tissue, cartilage, and bone were morphologically and biochemically monitored with specific markers for each. RESULTS: AGE-2 and AGE-3, but not control nonglycated BSA and AGE-1, reduced the viable cell number and 5-bromo-2'deoxyuridine (BrdU) incorporation with increased intracellular ROS generation and the percentage of apoptotic cells. MSCs expressed RAGE and its induction was stimulated by AGE-2 and AGE-3. These AGEs inhibited adipogenic differentiation (assayed by oil red O staining, lipoprotein lipase production, and intracellular triglyceride content) and chondrogenic differentiation (assayed by safranin O staining and type II collagen production). On osteogenic differentiation, AGE-2 and AGE-3 increased alkaline phosphatase activity and intracellular calcium content; however, von Kossa staining revealed the loss of mineralization and mature bone nodule formation. The antiserum against RAGE partially prevented AGE-induced cellular events. CONCLUSION: AGE-2 and AGE-3 may lead to the in vivo loss of MSC mass and the delay of tissue repair by inhibiting the maturation of MSC-derived cells. The AGE-RAGE interaction may be involved in the deleterious effect of AGEs on MSCs.


Subject(s)
Adipose Tissue/pathology , Bone and Bones/pathology , Cartilage/pathology , Cell Differentiation/drug effects , Glycation End Products, Advanced/physiology , Mesenchymal Stem Cells/cytology , Acetaldehyde/analogs & derivatives , Acetaldehyde/pharmacology , Adipocytes/cytology , Adipose Tissue/cytology , Apoptosis , Blotting, Western , Bone and Bones/cytology , Bromodeoxyuridine/pharmacology , Calcium/metabolism , Cartilage/cytology , Cell Line , Cell Proliferation , Cell Survival , Cells, Cultured , Chondrocytes/cytology , Collagen Type II/biosynthesis , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Glucose/pharmacology , Glycation End Products, Advanced/metabolism , Glyceraldehyde/pharmacology , Humans , Immunohistochemistry , Lipoprotein Lipase/biosynthesis , Microscopy, Fluorescence , Microscopy, Immunoelectron , Microscopy, Phase-Contrast , Osteocytes/metabolism , Phenazines/pharmacology , Reactive Oxygen Species , Stem Cells/cytology , Time Factors , Wound Healing
8.
Microvasc Res ; 69(1-2): 45-55, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15797260

ABSTRACT

Pigment epithelium-derived factor (PEDF) is a potent inhibitor of angiogenesis in the mammalian eye, suggesting that loss of PEDF is implicated in the pathogenesis of proliferative diabetic retinopathy. However, a role for PEDF in early diabetic retinopathy remains to be elucidated. Since oxidative stress is thought to be involved in pericyte loss and dysfunction, one of the changes characteristic of early diabetic retinopathy, we investigated whether and how PEDF could protect cultured retinal pericyte against oxidative stress injury. High glucose (30 mM) increased intracellular reactive oxygen species (ROS) generation in pericytes, which was completely blocked by PEDF. High glucose or H2O2 was found to induce growth retardation and apoptotic cell death of pericytes. PEDF completely restored these cytopathic effects on pericytes. An increased ratio of bax to bcl-2 mRNA level with subsequent activation of caspase-3 was observed in high-glucose- or H2O2-exposed pericytes, which was also completely prevented by PEDF. PEDF significantly increased glutathione peroxidase (GPx) mRNA levels and activity in pericytes. Further, PEDF was found to completely inhibit high-glucose- or H2O2-induced increase in a mRNA ratio of angiopoietin-2 to angiopoietin-1 and up-regulation of VEGF mRNA levels in pericytes. PEDF mRNA levels themselves were down-regulated in high-glucose- or H2O2-exposed pericytes. These results demonstrate that PEDF protects against high-glucose- or H2O2-induced pericyte apoptosis and dysfunction through its anti-oxidative properties via GPx induction. Our present study suggests that substitution of PEDF proteins might be a promising therapeutic strategy for treatment of patients with early diabetic retinopathy.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Eye Proteins/metabolism , Nerve Growth Factors/metabolism , Oxidative Stress/drug effects , Pericytes/metabolism , Retina/cytology , Serpins/metabolism , Animals , Cattle , Cells, Cultured , Down-Regulation , Eye Proteins/genetics , Eye Proteins/pharmacology , Gene Expression Regulation/drug effects , Nerve Growth Factors/genetics , Nerve Growth Factors/pharmacology , Pericytes/cytology , Serpins/genetics , Serpins/pharmacology
9.
Am J Pathol ; 165(6): 1865-74, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15579431

ABSTRACT

Angiogenesis, a process by which new vascular networks are formed from pre-existing capillaries, is required for tumors to grow, invade, and metastasize. Vascular endothelial growth factor (VEGF), a specific mitogen to endothelial cells, is a crucial factor for tumor angiogenesis. In this study, we investigated whether minodronate, a newly developed nitrogen-containing bisphosphonate, could inhibit melanoma growth and improve survival in nude mice by suppressing the VEGF signaling. We found here that minodronate inhibited melanoma growth and improved survival in nude mice by suppressing the tumor-associated angiogenesis and macrophage infiltration. Minodronate completely inhibited the VEGF-induced increase in DNA synthesis and tube formation in endothelial cells by suppressing NADPH oxidase-mediated reactive oxygen species generation and Ras activation. Furthermore, minodronate inhibited the VEGF-induced expression of intercellular adhesion molecule-1 and monocyte chemoattractant protein-1 in endothelial cells. Minodronate decreased DNA synthesis and increased apoptotic cell death of cultured melanoma cells as well. Our present study suggests that minodronate might suppress melanoma growth and improve survival in nude mice by two independent mechanisms; one is by blocking the VEGF signaling in endothelial cells, and the other is by inducing apoptotic cell death of melanoma. The present study provides a novel potential therapeutic strategy for the treatment of melanoma.


Subject(s)
Apoptosis/drug effects , Diphosphonates/therapeutic use , Imidazoles/therapeutic use , Melanoma, Experimental/prevention & control , Skin Neoplasms/prevention & control , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Cell Adhesion/drug effects , DNA/drug effects , DNA/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Female , Humans , Melanoma, Experimental/blood supply , Melanoma, Experimental/mortality , Mice , Mice, Nude , NADPH Oxidases/metabolism , Neovascularization, Pathologic/prevention & control , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Skin Neoplasms/blood supply , Skin Neoplasms/mortality , Survival Rate , rac1 GTP-Binding Protein/metabolism
10.
Kidney Int ; 66(6): 2137-47, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15569303

ABSTRACT

BACKGROUND: The renin-angiotensin system (RAS) and the accumulation of advanced glycation end products (AGEs) have been implicated in the pathogenesis of diabetic nephropathy. Whether there is a functional interaction between the RAS and AGEs in diabetic nephropathy is not known. In this study, we investigated whether AGEs could activate autocrine angiotensin II (Ang II) signaling and subsequently induce transforming growth factor-beta (TGF-beta)-Smad signaling in cultured rat mesangial cells. METHODS: The intracellular formation of reactive oxygen species (ROS) was detected using the fluorescent probe CM-H2DCFDA. Ang II was measured by radioimmunoassay. TGF-beta released into media was quantitatively analyzed in an enzyme-linked immunosorbent assay (ELISA). Smad2, p27(Kip1) (p27), fibronectin, and receptor for AGEs (RAGE) protein expression were determined by Western blot analysis. TGF-beta-inducible promoter activity was analyzed by a luciferase assay. DNA synthesis was evaluated by 5-bomo-2'-deoxyuridine (BrdU) incorporation and de novo protein synthesis was determined by [3H]leucine incorporation. RESULTS: AGEs increased intracellular ROS generation in mesangial cells, and this effect was significantly inhibited by an antiserum against RAGE. AGEs also were found to stimulate Ang II production in a time- and dose-dependent manner, which was completely prevented by an antioxidant, N-acetylcysteine (NAC). AGE-induced TGF-beta overproduction was completely blocked by candesartan, an Ang II type 1 receptor (AT1R) antagonist. Both candesartan and neutralizing antibody against TGF-beta completely prevented AGEs-induced Smad2 phosphorylation and TGF-beta-inducible promoter activity. Furthermore, AGEs were found to inhibit DNA synthesis and to stimulate de novo protein synthesis and fibronectin production in association with up-regulation of p27. All of these phenomena were completely prevented by candesartan or a polyclonal antibody against TGF-beta. CONCLUSION: The present study suggests that AGE-RAGE-mediated ROS generation activates TGF-beta-Smad signaling and subsequently induces mesangial cell hypertrophy and fibronectin synthesis by autocrine production of Ang II. This pathway may provide an important link between metabolic and haemodynamic factors in promoting the development and progression of diabetic nephropathy.


Subject(s)
DNA-Binding Proteins/metabolism , Glycation End Products, Advanced/pharmacology , Receptor, Angiotensin, Type 1/metabolism , Signal Transduction/drug effects , Trans-Activators/metabolism , Transforming Growth Factor beta/metabolism , Angiotensin II/metabolism , Animals , Cells, Cultured , DNA/biosynthesis , Fibronectins/metabolism , Glomerular Mesangium/cytology , Humans , Male , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Receptor for Advanced Glycation End Products , Receptors, Immunologic/metabolism , Smad Proteins
11.
J Mol Cell Cardiol ; 37(2): 497-506, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15276019

ABSTRACT

Pigment epithelium-derived factor (PEDF) has recently been shown to be involved in the pathogenesis of proliferative diabetic retinopathy. Atherosclerosis is an inflammatory-fibroproliferative disease as well. Oxidative stress plays a major role in retinopathy and atherosclerosis. Accordingly, we investigated effects of PEDF on reactive oxygen species (ROS) generation, NF-kappaB activation and interleukin (IL)-6 expression in TNF-alpha-exposed HUVEC. TNF-alpha significantly increased intracellular ROS generation, which was completely blocked by PEDF or diphenylene iodonium, an inhibitor of NADPH oxidase. Further, PEDF completely prevented the TNF-alpha-induced increase in NADPH oxidase activity. PEDF or an antioxidant, N-acetylcysteine, significantly inhibited the TNF-alpha-induced NF-kappaB activation. PEDF inhibited TNF-alpha-induced expression of IL-6 at both mRNA and protein levels. Moreover, TNF-alpha downregulated PEDF mRNA levels. Ligand blot analysis revealed that HUVEC possessed a membrane protein with binding affinity for PEDF. The results demonstrated that PEDF inhibited TNF-alpha-induced NF-kappaB activation and subsequent IL-6 overexpression in HUVEC by suppressing NADPH oxidase-mediated ROS generation. Our present study suggests that PEDF may play an important role in the development and progression of atherosclerosis.


Subject(s)
Endothelium, Vascular/metabolism , Eye Proteins/pharmacology , Interleukin-6/biosynthesis , NADPH Oxidases/metabolism , Nerve Growth Factors/pharmacology , Reactive Oxygen Species/metabolism , Serpins/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Cells, Cultured , Endothelial Cells/chemistry , Endothelial Cells/drug effects , Endothelium, Vascular/cytology , Gene Expression/drug effects , Humans , Interleukin-6/genetics , NADPH Oxidases/analysis , NF-kappa B/analysis , NF-kappa B/metabolism , RNA, Messenger/analysis , RNA, Messenger/metabolism , Reactive Oxygen Species/analysis , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
12.
J Cardiovasc Pharmacol ; 43(5): 724-30, 2004 May.
Article in English | MEDLINE | ID: mdl-15071361

ABSTRACT

Interleukin-8 (IL-8), a member of CXC chemokine family, has been found to play an important role in the pathogenesis of atherosclerosis. Tumor necrosis factor-alpha (TNF-alpha) is involved in the development and progression of atherosclerosis as well. In this study, we investigated whether and how azelnidipine, a newly developed long-acting calcium antagonist, could inhibit TNF-alpha-induced IL-8 expression in human umbilical vein endothelial cells (HUVEC). TNF-alpha significantly increased intracellular reactive oxygen species (ROS) generation in HUVEC, which was completely blocked by azelnidipine or apocynin, an inhibitor of NADPH oxidase. Azelnidipine also completely prevented TNF-alpha-induced increase in NADPH oxidase activity in HUVEC. Further, azelnidipine was found to significantly inhibit activator protein-1 (AP-1) promoter activity and IL-8 expression in TNF-alpha-exposed HUVEC. An inhibitor of AP-1, curcumin, or an anti-oxidant, N-acetylcysteine, also inhibited the TNF-alpha-induced IL-8 expression in HUVEC. These results demonstrated that azelnidipine inhibited TNF-alpha-induced IL-8 expression in HUVEC by blocking NADPH oxidase-mediated ROS generation and subsequent AP-1 activation. Our present study suggests that azelnidipine may play a protective role in the development and progression of atherosclerosis through its anti-oxidative properties.


Subject(s)
Antioxidants/pharmacology , Azetidinecarboxylic Acid/analogs & derivatives , Azetidinecarboxylic Acid/pharmacology , Calcium Channel Blockers/pharmacology , Dihydropyridines/pharmacology , Endothelium, Vascular/drug effects , Interleukin-8/biosynthesis , Tumor Necrosis Factor-alpha/pharmacology , Amlodipine/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans , In Vitro Techniques , Radioligand Assay , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor AP-1/metabolism , Tumor Necrosis Factor-alpha/physiology , Umbilical Veins/enzymology
13.
Am J Pathol ; 164(4): 1225-32, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15039211

ABSTRACT

Pigment epithelium-derived factor (PEDF) has recently been shown to be the most potent inhibitor of angiogenesis in the mammalian eye, and is involved in the pathogenesis of angiogenic eye disease such as proliferative diabetic retinopathy. However, a functional role for PEDF in tumor growth and angiogenesis remains to be determined. In this study, we have investigated both the in vitro and in vivo growth characteristics of human malignant melanoma G361 cell lines, stably transfected to overexpress human PEDF. Expression levels of PEDF proteins in melanoma cell lines G361 and A375 were comparable with that of human cultured melanocytes, whereas vascular endothelial growth factor levels in two tumor cell lines were much stronger than that in normal melanocytes. Overexpression of PEDF was found to significantly inhibit tumor growth and vessel formation in G361 nude mice xenografts. Furthermore, in vitro proliferation rates of G361 cells were decreased in PEDF-transfected cells. PEDF proteins showed dose-dependent induced growth retardation and apoptotic cell death in nontransfected G361 cells, which were completely prevented by treatment with antibodies against the Fas ligand. Our present study highlights two beneficial effects of PEDF treatment on melanoma growth and expansion; one is the suppression of tumor angiogenesis, and the other is induction of Fas ligand-dependent apoptosis in tumor cells. PEDF therefore might be a promising novel therapeutic agent for treatment of patients with melanoma.


Subject(s)
Eye Proteins , Melanoma, Experimental/pathology , Neovascularization, Pathologic/physiopathology , Nerve Growth Factors , Protein Biosynthesis , Serpins/biosynthesis , Animals , Apoptosis/physiology , Blotting, Western , Cell Line, Tumor , Humans , Mice , Microscopy, Fluorescence , Proteins/genetics , Serpins/genetics , Transfection , Vascular Endothelial Growth Factor A/metabolism
14.
J Neuropathol Exp Neurol ; 62(5): 486-96, 2003 May.
Article in English | MEDLINE | ID: mdl-12769188

ABSTRACT

The Maillard reaction that leads to the formation of advanced glycation end products (AGEs) plays an important role in the pathogenesis of angiopathy in diabetic patients, in aging, and in neurodegenerative processes. We hypothesize that acetaldehyde (AA), one of the main metabolites of alcohol, may be involved in alcohol-induced neurotoxicity in vivo by formation of AA-derived AGEs (AA-AGEs) with brain proteins. Incubation of cortical neurons with AA-AGE produced a dose-dependent increase in neuronal cell-death, and the neurotoxicity of AA-AGE was neutralized by the addition of an anti-AA-AGE-specific antibody, but not by anti-N-ethyllysine (NEL) antibody. The AA-AGE epitope was detected in human brain of alcoholism. We propose that the structural epitope AA-AGE is an important toxic moiety for neuronal cells in alcoholism.


Subject(s)
Acetaldehyde/toxicity , Cerebral Cortex/metabolism , Glycation End Products, Advanced/toxicity , Neurons/drug effects , Acetaldehyde/chemistry , Acetaldehyde/metabolism , Albumins/metabolism , Alcoholism/metabolism , Alcoholism/pathology , Animals , Antibodies/metabolism , Apoptosis/physiology , Cattle , Cells, Cultured , Cerebral Cortex/cytology , Glycation End Products, Advanced/chemistry , Glycation End Products, Advanced/metabolism , Humans , Neurons/cytology , Neurons/metabolism , Rabbits , Rats
15.
Microvasc Res ; 65(3): 186-90, 2003 May.
Article in English | MEDLINE | ID: mdl-12711260

ABSTRACT

Leptin, a circulating hormone secreted mainly from adipose tissues, is involved in the control of body weight. Recently, leptin was found to be an angiogenic factor, and its vitreous levels are associated with angiogenic eye diseases such as proliferative diabetic retinopathy. However, the molecular mechanism for leptin-elicited angiogenesis remains to be elucidated. Pigment epithelium-derived factor (PEDF) has been shown to be the most potent natural inhibitor of angiogenesis in the mammalian eye, and its levels in the vitreous were decreased in angiogenic eye diseases. In this study, we investigated whether and how PEDF could inhibit the leptin-induced DNA synthesis in microvascular endothelial cells (EC), a key step of angiogenesis. Leptin significantly increased intracellular reactive oxygen species (ROS) generation in microvascular EC. PEDF was found to inhibit the leptin-induced ROS generation in EC. An anti-oxidant, N-acetylcysteine, or PEDF completely prevented the leptin-induced upregulation of vascular endothelial growth factor (VEGF) mRNA levels as well as any increase in DNA synthesis in microvascular EC. Polyclonal antibodies against human VEGF were also found to completely inhibit DNA synthesis in leptin-exposed EC. The present study suggests that leptin could elicit angiogenesis through autocrine VEGF production via intracellular ROS generation. PEDF may block the angiogenic effects of leptin through its anti-oxidative properties.


Subject(s)
Antioxidants/chemistry , Eye Proteins , Gene Expression Regulation , Leptin/chemistry , Neovascularization, Pathologic , Nerve Growth Factors , Retinal Neovascularization , Vascular Endothelial Growth Factor A/biosynthesis , Acetylcysteine/chemistry , Animals , Antioxidants/metabolism , Brain/metabolism , Cattle , DNA/biosynthesis , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Eye Diseases/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Leptin/metabolism , Microcirculation/metabolism , Proteins/chemistry , Proteins/metabolism , RNA, Messenger/metabolism , Reactive Oxygen Species , Receptors, Leptin , Receptors, Vascular Endothelial Growth Factor/metabolism , Retinal Vessels/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Serpins/chemistry , Serpins/metabolism , Thymidine/chemistry , Time Factors , Vascular Endothelial Growth Factor A/metabolism
16.
Kidney Int ; 63(2): 464-73, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12631112

ABSTRACT

BACKGROUND: We have shown previously that OPB-9195, a novel inhibitor of advanced glycation end products (AGE), significantly prevented renal tubular injury and tubulointerstitial fibrosis in spontaneous diabetic rats. However, the molecular mechanisms underlying this have not been fully elucidated. METHODS: Three immunochemically distinct AGE were prepared by incubating bovine serum albumin (BSA) with glucose, glyceraldehyde, or methylglyoxal. Then, the effects of AGE on human proximal tubular epithelial cells were examined. The intracellular formation of reactive oxygen species (ROS) was detected using the fluorescent probe CM-H2DCFDA. DNA synthesis was evaluated by thymidine uptake, and de novo protein synthesis was determined by [3H]leucine incorporation. Prostaglandin E2 (PGE2) and transforming growth factor-beta (TGF-beta) released into media were quantitatively analyzed in an enzyme-linked immunosorbent assay. TGF-beta gene expression was analyzed by quantitative reverse transcription-polymerase chain reaction (RT-PCR). RESULTS: When these AGE-BSA were administered to tubular cells, each of them increased generation of intracellular ROS. All of the AGE-BSA, but not non-glycated BSA, were found to induce statistically significant decreases in de novo protein synthesis and PGE2 secretion by tubular cells. Furthermore, AGE-BSA up-regulated the levels of mRNAs for TGF-beta in tubular cells. The structural epitope designated glucose-derived AGE was found to have the greatest cytopathic effects on tubular cells. These AGE-induced inhibition of protein synthesis and PGE2 secretion as well as the up-regulation of TGF-beta mRNA were found to be completely prevented by N-acetylcysteine. Furthermore, H2O2 was shown to inhibit protein synthesis and PGE2 secretion by proximal tubular cells in a dose-dependent manner. CONCLUSION: The results suggest that AGE inhibits de novo protein synthesis and stimulates TGF-beta mRNA expression in proximal tubular epithelial cells through overgeneration of intracellular ROS. Thus, AGE are involved in the pathogenesis of tubular injury in diabetic nephropathy.


Subject(s)
Glycation End Products, Advanced/pharmacology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Protein Synthesis Inhibitors/pharmacology , Transforming Growth Factor beta/metabolism , Cells, Cultured , Diabetes Mellitus/blood , Dinoprostone/metabolism , Humans , Hydrogen Peroxide/pharmacology , Intracellular Membranes/metabolism , Kidney Tubules, Proximal/cytology , Leucine/metabolism , RNA, Messenger/metabolism , Reactive Oxygen Species , Serum Albumin, Bovine/pharmacology , Thymidine/metabolism , Transforming Growth Factor beta/genetics
17.
Mol Med ; 8(10): 591-9, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12477969

ABSTRACT

BACKGROUND: Diabetic nephropathy is a leading cause of end-stage renal disease in industrialized countries. Previous studies have documented that angiotensin converting enzyme (ACE) inhibitors consistently reduce albuminuria and retard the progression of diabetic nephropathy. However, the involvement of angiotensin II in diabetic nephropathy is not fully understood. MATERIALS AND METHODS: In this study we compared the effects of CS-866, a new angiotensin II type 1 receptor antagonist, to that of an ACE inhibitor, temocapril hydrochloride, on the development and progression of diabetic nephropathy using Otsuka Long-Evans Tokushima fatty rats, a type II diabetes mellitus model animal. RESULTS: High doses of CS-866 or temocapril treatment were found to significantly improve urinary protein and beta(2)-microglobulin excretions in diabetic rats. In electron microscopic analysis, loss of glomerular anionic sites, one of the causes of glomerular hyperpermeability in diabetic nephropathy, was found to be significantly prevented by CS-866 treatment. Light microscopic examinations revealed that both treatments ameliorated glomerular sclerosis and tubulointerstitial injury in diabetic rats. Furthermore, high doses of CS-866 or temocapril treatment significantly reduced the positive stainings for transforming growth factor-beta (TGF-beta), vascular endothelial growth factor, and type IV collagen in glomeruli of diabetic rats. CONCLUSIONS: These results indicate that intrarenal angiotensin II type 1 receptor activation plays a dominant role in the development and progression of diabetic nephropathy. Our study suggests that CS-866 represents a valuable new drug for the treatment of diabetic patients with nephropathy.


Subject(s)
Angiotensin II Type 1 Receptor Blockers , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/pathology , Imidazoles/therapeutic use , Kidney Glomerulus/drug effects , Kidney Glomerulus/pathology , Tetrazoles/therapeutic use , Animals , Anions , Diabetic Nephropathies/prevention & control , Disease Models, Animal , Imidazoles/pharmacology , Immunohistochemistry , Kidney Glomerulus/ultrastructure , Male , Olmesartan Medoxomil , Proteinuria/drug therapy , Rats , Rats, Inbred OLETF , Severity of Illness Index , Tetrazoles/pharmacology , Thiazepines/pharmacology , Transforming Growth Factor beta/analysis , Vascular Endothelial Growth Factor A/analysis
18.
Biochem Biophys Res Commun ; 299(2): 183-8, 2002 Nov 29.
Article in English | MEDLINE | ID: mdl-12437967

ABSTRACT

The polyol pathway consists of two enzymes, aldose reductase (AR) and sorbitol dehydrogenase (SDH). There is a growing body of evidence to suggest that acceleration of the polyol pathway is implicated in the pathogenesis of diabetic vascular complications. However, a functional role remains to be elucidated for SDH in the development and progression of diabetic retinopathy. In this study, cultured bovine retinal capillary pericytes were used to investigate the effects of SDH overexpression on glucose toxicity. High glucose modestly increased reactive oxygen species (ROS) generation, decreased DNA synthesis, and up-regulated vascular endothelial growth factor (VEGF) mRNA levels in cultured pericytes. SDH overexpression was found to significantly stimulate ROS generation in high glucose-exposed pericytes and subsequently potentiate the cytopathic effects of glucose. Fidarestat, a newly developed AR inhibitor, and N-acetylcysteine, an antioxidant, completely prevented these deleterious effects of SDH overexpression on pericytes. Furthermore, fidarestat administration was found to significantly prevent vascular hyperpermeability, the characteristic changes of the early phase of diabetic retinopathy, in streptozotocin-induced diabetic rats. Our present results suggest that SDH-mediated conversion of sorbitol to fructose and the resultant ROS generation may play an active role in the pathogenesis of diabetic retinopathy. Blockage of sorbitol formation by fidarestat could be a promising therapeutic strategy for the treatment of early phase of diabetic retinopathy.


Subject(s)
Blood-Retinal Barrier , Glucose/toxicity , Imidazolidines , L-Iditol 2-Dehydrogenase/physiology , Pericytes/metabolism , Retina/cytology , Aldehyde Reductase/antagonists & inhibitors , Animals , Blood-Retinal Barrier/drug effects , Capillary Permeability/drug effects , Cattle , Cells, Cultured , DNA/biosynthesis , Diabetes Mellitus, Experimental/blood , Diabetic Nephropathies/etiology , Endothelial Growth Factors/biosynthesis , Endothelial Growth Factors/genetics , Enzyme Inhibitors/pharmacology , Fructose/metabolism , Imidazoles/pharmacology , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/genetics , L-Iditol 2-Dehydrogenase/genetics , L-Iditol 2-Dehydrogenase/metabolism , Lymphokines/biosynthesis , Lymphokines/genetics , Male , Pericytes/drug effects , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Sorbitol/metabolism , Transfection , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
19.
Mol Med ; 8(9): 546-50, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12456993

ABSTRACT

BACKGROUND: Beraprost sodium, a prostaglandin I2 analogue, has been recently reported to exhibit beneficial effects on atherosclerosis in patients with diabetes. However, effects of beraprost sodium on microvascular injury in diabetes remain to be elucidated. We have previously shown that advanced glycation end products (AGE), senescent macroproteins formed at an accelerated rate in diabetes, caused pericyte apoptosis, thus being involved in the pathogenesis of the early phase of diabetic retinopathy. In this study, we examined whether beraprost sodium can protect against AGE-induced cytotoxicity in cultured retinal pericytes. MATERIALS AND METHODS: Intracellular formation of reactive oxygen species (ROS) was detected using a fluorescent probe. DNA synthesis was determined by measuring [3H]thymidine incorporation into cells. Apoptosis was determined by DNA fragmentations, which were quantitatively measured in an enzyme-linked immunosorbent assay. RESULTS: Beraprost sodium or forskolin, a stimulator of adenylate cyclase, was found to significantly inhibit AGE-induced ROS generation and the subsequent decrease in DNA synthesis in pericytes. Both treatments significantly prevented AGE-induced apoptotic cell death in pericytes. Furthermore, beraprost sodium was found to down-regulate AGE receptor mRNA levels in pericytes. CONCLUSION: The results demonstrated that cyclic AMP-elevating agents such as beraprost sodium and forskolin protected retinal pericytes from AGE-induced cytotoxicity through its anti-oxidative properties. Our present study suggests that beraprost sodium may have therapeutic potentials in treatment of patients with early diabetic retinopathy.


Subject(s)
Epoprostenol/analogs & derivatives , Epoprostenol/pharmacology , Glycation End Products, Advanced/metabolism , Pericytes/drug effects , Retina/drug effects , Vasodilator Agents/pharmacology , Animals , Apoptosis/physiology , Cattle , Colforsin/pharmacology , DNA/biosynthesis , Reactive Oxygen Species/metabolism , Receptor for Advanced Glycation End Products , Receptors, Immunologic/biosynthesis , Receptors, Immunologic/genetics
20.
FASEB J ; 16(14): 1928-30, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12368225

ABSTRACT

We previously have found that advanced glycation end products (AGE), senescent macroproteins formed at an accelerated rate in diabetes, arise in vivo not only from glucose but also from reducing sugars. Furthermore, we recently have shown that glyceraldehyde- and glycolaldehyde-derived AGE (glycer- and glycol-AGE) are mainly involved in loss of pericytes, the earliest histopathological hallmark of diabetic retinopathy. However, the effects of these AGE proteins on angiogenesis, another vascular derangement in diabetic retinopathy, remain to be elucidated. In this study, we investigated whether these AGE proteins elicit changes in cultured endothelial cells that are associated with angiogenesis. When human skin microvascular endothelial cells (EC) were cultured with glycer-AGE or glycol-AGE, growth and tube formation of EC, the key steps of angiogenesis, were significantly stimulated. The AGE-induced growth stimulation was significantly enhanced in AGE receptor (RAGE)-overexpressed EC. Furthermore, AGE increased transcriptional activity of nuclear factor-kB (NF-kB) and activator protein-1 (AP-1) and then up-regulated mRNA levels of vascular endothelial growth factor (VEGF) and angiopoietin-2 (Ang-2) in EC. Cerivastatin, a hydroxymethylglutaryl CoA reductase inhibitor; pyrrolidinedithiocarbamate; or curcumin was found to completely prevent the AGE-induced increase in NF-kB and AP-1 activity, VEGF mRNA up-regulation, and the resultant increase in DNA synthesis in microvascular EC. These results suggest that the AGE-RAGE interaction elicited angiogenesis through the transcriptional activation of the VEGF gene via NF-kB and AP-1 factors. By blocking AGE-RAGE signaling pathways, cerivastatin might be a promising remedy for treating patients with proliferative diabetic retinopathy.


Subject(s)
Endothelium, Vascular/growth & development , Glycation End Products, Advanced/antagonists & inhibitors , Glycation End Products, Advanced/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Pyridines/pharmacology , Angiogenesis Inducing Agents/biosynthesis , Angiogenesis Inducing Agents/genetics , Angiopoietin-2 , Cells, Cultured , DNA/biosynthesis , Endothelial Growth Factors/biosynthesis , Endothelial Growth Factors/genetics , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Glycation End Products, Advanced/metabolism , Humans , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/genetics , Lymphokines/biosynthesis , Lymphokines/genetics , Microcirculation/cytology , Models, Biological , NF-kappa B/metabolism , Neovascularization, Physiologic , RNA, Messenger/biosynthesis , Receptor for Advanced Glycation End Products , Receptors, Immunologic/metabolism , Signal Transduction/drug effects , Transcription Factor AP-1/metabolism , Transcriptional Activation , Up-Regulation , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...