Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 300
Filter
1.
Clin Transl Oncol ; 23(8): 1585-1592, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33502740

ABSTRACT

PURPOSE: This study aimed to determine the outcomes and prognostic factors associated with octogenarians who underwent pulmonary resection for lung cancer. METHODS/PATIENTS: From 2009 to 2018, 76 octogenarians underwent pulmonary surgery for lung cancer at the Kanazawa Medical University, Japan. They were divided into two groups (early and late octogenarians), and their clinicopathological characteristics and outcomes were investigated. Overall survival rates and recurrence-free survival rates were determined using Kaplan-Meier curves. Univariate and multivariate analyses were performed to identify prognostic factors. RESULTS: Limited surgery was performed more often in the late octogenarian group; however, most perioperative factors were not significantly different between the two groups. The 3-year overall survival and recurrence-free survival rates were 61.2% and 52.8%, respectively. The median observation period was 37.5 (8.9-112.3) months postoperatively. Kaplan-Meier curves showed that age ≥ 85 years (late octogenarian), smoking history, and squamous cell carcinoma on histology were associated with worse survival rates. Multivariate analysis identified age ≥ 85 years (late octogenarian) (p = 0.011) and cigarette smoking (p = 0.025) as unfavorable prognostic factors for overall survival and recurrence-free survival, respectively. CONCLUSIONS: Most octogenarians with an indication for surgery can tolerate pulmonary surgery. However, owing to the limitations of this retrospective, single-center study, future studies involving multiple-institutions are required to confirm our findings.


Subject(s)
Carcinoma, Squamous Cell/surgery , Lung Neoplasms/surgery , Lung/surgery , Age Factors , Aged, 80 and over , Analysis of Variance , Carcinoma, Squamous Cell/mortality , Female , Humans , Japan , Kaplan-Meier Estimate , Lung Neoplasms/mortality , Male , Prognosis , Progression-Free Survival , Retrospective Studies , Smoking/adverse effects , Survival Rate , Thoracic Surgery, Video-Assisted , Treatment Outcome
2.
Horm Metab Res ; 48(9): 613-9, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27214310

ABSTRACT

Pigment epithelium-derived factor (PEDF) is a glycoprotein with complex neuroprotective, anti-angiogenic, and anti-inflammatory properties, all of which could potentially be exploited as a therapeutic option for vascular complications in diabetes. We have previously shown that PEDF-derived synthetic peptide, P5-3 (FIFVLRD) has a comparable ability with full PEDF protein to inhibit rat corneal neovascularization induced by chemical cauterization. However, the effects of PEDF peptide on experimental diabetic nephropathy remain unknown. To address the issue, we modified P5-3 to stabilize and administered the modified peptide (d-Lys-d-Lys-d-Lys-Gln-d-Pro-P5-3-Cys-amide, 0.2 nmol/day) or vehicle to streptozotocin-induced diabetic rats (STZ-rats) intraperitoneally by an osmotic mini pump for 2 weeks. We further examined the effects of modified peptide on human proximal tubular cells. Renal PEDF expression was decreased in STZ-rats. Although the peptide administration did not affect blood glucose or blood pressure, it decreased urinary excretion levels of 8-hydroxy-2'-deoxyguanosine, an oxidative stress marker, and reduced plasminogen activator inhibitor-1 (PAI-1) gene expression, and suppressed glomerular expansion in the diabetic kidneys. High glucose or advanced glycation end products stimulated oxidative stress generation and PAI-1 gene expression in tubular cells, all of which were significantly suppressed by 10 nM modified P5-3 peptide. Our present study suggests that PEDF-derived synthetic modified peptide could protect against experimental diabetic nephropathy and inhibit tubular cell damage under diabetes-like conditions through its anti-oxidative properties. Supplementation of modified P5-3 peptide may be a novel therapeutic strategy for diabetic nephropathy.


Subject(s)
Diabetes Mellitus, Experimental/complications , Diabetic Nephropathies/prevention & control , Eye Proteins/metabolism , Nerve Growth Factors/metabolism , Peptide Fragments/pharmacology , Serpins/metabolism , Animals , Diabetes Mellitus, Experimental/physiopathology , Diabetic Nephropathies/etiology , Humans , Kidney/metabolism , Male , Oxidative Stress , Rats , Rats, Wistar
3.
Nutr Metab Cardiovasc Dis ; 26(9): 797-807, 2016 09.
Article in English | MEDLINE | ID: mdl-27212619

ABSTRACT

BACKGROUND AND AIMS: Advanced glycation end products (AGEs)-receptor RAGE interaction evokes oxidative stress and inflammatory reactions, thereby being involved in endothelial cell (EC) damage in diabetes. Sulforaphane is generated from glucoraphanin, a naturally occurring isothiocyanate found in widely consumed cruciferous vegetables, by myrosinase. Sulforaphane has been reported to protect against oxidative stress-mediated cell and tissue injury. However, effects of sulforaphane on AGEs-induced vascular damage remain unclear. METHODS AND RESULTS: In this study, we investigated whether and how sulforaphane could inhibit inflammation in AGEs-exposed human umbilical vein ECs (HUVECs) and AGEs-injected rat aorta. Sulforaphane treatment for 4 or 24 h dose-dependently inhibited the AGEs-induced increase in RAGE, monocyte chemoattractant protein-1 (MCP-1), intercellular adhesion molecule-1 (ICAM-1), and vascular cell adhesion molecular-1 (VCAM-1) gene expression in HUVECs. AGEs significantly stimulated MCP-1 production by, and THP-1 cell adhesion to, HUVECs, both of which were prevented by 1.6 µM sulforaphane. Sulforaphane significantly suppressed oxidative stress generation and NADPH oxidase activation evoked by AGEs in HUVECs. Furthermore, aortic RAGE, ICAM-1 and VCAM-1 expression in AGEs-injected rats were increased, which were suppressed by simultaneous infusion of sulforaphane. CONCLUSION: The present study demonstrated for the first time that sulforaphane could inhibit inflammation in AGEs-exposed HUVECs and AGEs-infused rat aorta partly by suppressing RAGE expression through its anti-oxidative properties. Inhibition of the AGEs-RAGE axis by sulforaphane might be a novel therapeutic target for vascular injury in diabetes.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Aorta/drug effects , Aortitis/prevention & control , Endothelial Cells/drug effects , Glycation End Products, Advanced , Isothiocyanates/pharmacology , Animals , Aorta/metabolism , Aortitis/chemically induced , Aortitis/metabolism , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Inflammation Mediators/metabolism , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Wistar , Receptor for Advanced Glycation End Products/drug effects , Receptor for Advanced Glycation End Products/genetics , Receptor for Advanced Glycation End Products/metabolism , Sulfoxides , Time Factors
4.
Horm Metab Res ; 48(3): 191-5, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26158396

ABSTRACT

Ninety percent of glucose filtered by the glomerulus is reabsorbed by a sodium-glucose cotransporter 2 (SGLT2), which is mainly expressed on S1 and S2 segment of renal proximal tubules. Since SGLT-2-mediated glucose reabsorption is increased under diabetic conditions, selective inhibition of SGLT2 is a potential therapeutic target for the treatment of diabetes. We have recently shown that an inhibitor of SGLT2 has anti-inflammatory and antifibrotic effects on experimental diabetic nephropathy partly by suppressing advanced glycation end products formation and oxidative stress generation in the kidney. However, the direct effects of SGLT2 inhibitor on tubular cell damage remain unclear. In this study, we investigated the effects of tofogliflozin, a highly selective inhibitor of SGLT2 on oxidative stress generation, inflammatory and proapoptotic reactions in cultured human proximal tubular cells exposed to high glucose. Tofogliflozin dose-dependently suppressed glucose entry into tubular cells. High glucose exposure (30 mM) for 4 and 24 h significantly increased oxidative stress generation in tubular cells, which were suppressed by the treatment of tofogliflozin or an antioxidant N-acetylcysteine (NAC). Monocyte chemoattractant protein-1 (MCP-1) gene expression and apoptotic cell death were induced by 4 h- and 8 day-exposure to high glucose, respectively, both of which were also blocked by tofogliflozin or NAC. The present study suggests that SGLT2-mediated glucose entry into tubular cells could stimulate oxidative stress and evoke inflammatory and proapoptotic reactions in this cell type. Blockade of glucose reabsorption in tubular cells by SGLT2 inhibitor might exert beneficial effects on tubulointerstitial damage in diabetic nephropathy.


Subject(s)
Apoptosis/drug effects , Benzhydryl Compounds/therapeutic use , Glucose/pharmacology , Glucosides/therapeutic use , Inflammation/drug therapy , Kidney Tubules, Proximal/pathology , Oxidative Stress/drug effects , Sodium-Glucose Transporter 2 Inhibitors , Apoptosis/genetics , Benzhydryl Compounds/pharmacology , Cell Line , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Gene Expression Regulation/drug effects , Glucosides/pharmacology , Humans , Inflammation/pathology , Kidney Tubules, Proximal/drug effects , Oxidative Stress/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Sodium-Glucose Transporter 2/metabolism
5.
Hepatogastroenterology ; 62(137): 30-3, 2015.
Article in English | MEDLINE | ID: mdl-25911862

ABSTRACT

BACKGROUND/AIMS: Anastomotic leakage is major complication of colorectal surgery. Total parenteral nutrition (TPN) and fasting are conservative treatments for leakage in the absence of peritonitis in Japan. Elemental diet (ED) jelly is a completely digested formula and is easily absorbed without secretion of digestive juices. The purpose of this study was to assess the safety of ED jelly in management of anastomotic leakage. METHODOLOGY: Six hundred and two patients who underwent elective surgery for left side colorectal cancer from January 2008 to December 2011 were included in the study. Pelvic drainage was performed for all patients. Sixty-three (10.5%) patients were diagnosed with an anastomotic leakage, and of these, 31 (5.2%) without diverting stoma were enrolled in this study. RESULTS: Sixteen patients received TPN (TPN group) and 15 patients received ED jelly (ED group). The duration of intravenous infusion was significantly shorter in the ED group than in the TPN group (15 days versus 25 days, P= 0.008). In the TPN group, catheter infection was occurred in 2 patients who required re-insertion of the catheter. CONCLUSION: Conservative management of anastomotic leakage after colorectal surgery with ED jelly appears to be a safe and useful approach.


Subject(s)
Anastomotic Leak/therapy , Colectomy/adverse effects , Colorectal Neoplasms/surgery , Food, Formulated , Parenteral Nutrition, Total , Administration, Oral , Adult , Aged , Aged, 80 and over , Anastomotic Leak/diagnosis , Anastomotic Leak/diet therapy , Anastomotic Leak/etiology , Elective Surgical Procedures , Female , Food, Formulated/adverse effects , Gels , Humans , Male , Middle Aged , Parenteral Nutrition, Total/adverse effects , Retrospective Studies , Time Factors , Treatment Outcome
6.
Med Hypotheses ; 84(5): 490-3, 2015 May.
Article in English | MEDLINE | ID: mdl-25697114

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) ranges from simple steatosis to nonalcoholic steatohepatitis (NASH), leads to fibrosis and potentially cirrhosis, liver failure, and hepatocellular carcinoma, and is one of the most common causes of liver disease worldwide. NAFLD has also been implicated in other medical conditions such as insulin resistance, obesity, metabolic syndrome, hyperlipemia, hypertension, cardiovascular disease, and diabetes. Continuous hyperglycemia has been implicated in the pathogenesis of diabetic micro- and macro-vascular complications via various metabolic pathways, and numerous hyperglycemia-induced metabolic and hemodynamic conditions exist, including the increased generation of various types of advanced glycation end-products (AGEs). We recently demonstrated that glyceraldehyde-derived AGEs (Glycer-AGEs), the predominant components of toxic AGEs (TAGE), played an important role in the pathogenesis of angiopathy in diabetic patients. Moreover, a growing body of evidence suggests that the interaction between TAGE and the receptor for AGEs may alter intracellular signaling, gene expression, and the release of pro-inflammatory molecules and also elicits the generation of oxidative stress in numerous types of cells including hepatocytes and hepatic stellate cells. Serum levels of TAGE were significantly higher in NASH patients than in those with simple steatosis and healthy controls. Moreover, serum levels of TAGE inversely correlated with adiponectin (adiponectin is produced by adipose tissue and is an anti-inflammatory adipokine that can increase insulin sensitivity). Furthermore, immunohistochemical staining of TAGE showed intense staining in the livers of patients with NASH. Serum levels of TAGE may be a useful biomarker for discriminating NASH from simple steatosis. The administration of atorvastatin (10 mg daily) for 12 months significantly improved NASH-related metabolic parameters and significantly decreased serum levels of TAGE. The steatosis grade and NAFLD activity score were also significantly improved. These results demonstrated that atorvastatin decreased the serum levels of TAGE in NASH patients with dyslipidemia and suggest the usefulness of TAGE as a biomarker for the attenuation of NASH. Serum levels of TAGE were significantly higher in non-B or non-C hepatocellular carcinoma (NBNC-HCC) patients than in NASH subjects without HCC or control subjects. TAGE may be involved in the pathogenesis of NBNC-HCC, and could, therefore, be a biomarker that could discriminate NBNC-HCC from NASH. We propose that serum levels of TAGE are promising novel targets for the diagnosis of and therapeutic interventions against NASH.


Subject(s)
Biomarkers/blood , Glycation End Products, Advanced/blood , Models, Biological , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/etiology , Disease Progression , Glycation End Products, Advanced/toxicity , Humans
7.
Horm Metab Res ; 47(9): 686-92, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25611208

ABSTRACT

Advanced glycation end products (AGEs) and receptor RAGE play a role in diabetic nephropathy. We have previously shown that increased glucose uptake into proximal tubular cells via sodium-glucose cotransporter 2 (SGLT2) stimulates oxidative stress generation and RAGE expression, thereby exacerbating the AGE-induced apoptosis in this cell type. However, the protective role of SGLT2 inhibition against the AGE-RAGE-induced renal damage in diabetic animals remains unclear. In this study, we investigated the effects of empagliflozin, SGLT2 inhibitor on AGE-RAGE axis, inflammatory and fibrotic reactions, and tubular injury in the kidney of streptozotocin-induced diabetic rats.Administration of empagliflozin for 4 weeks significantly improved hyperglycemia and HbA1c, and decreased expression levels of AGEs, RAGE, 8-hydroxydeoxyguanosine (8-OHdG), and F4/80, markers of oxidative stress and macrophages, respectively, in the diabetic kidney. Although empagliflozin did not reduce albuminuria, it significantly decreased urinary excretion levels of 8-OHdG and L-fatty acid binding protein, a marker of tubular injury. Moreover, inflammatory and fibrotic gene expression such as monocyte chemoattractant protein-1, intercellular adhesion molecule-1, plasminogen activator inhibitor-1, transforming growth factor-ß, and connective tissue growth factor was enhanced in the diabetic kidney, all of which were prevented by empagliflozin. The present study suggests that empagliflozin could inhibit oxidative, inflammatory and fibrotic reactions in the kidney of diabetic rats partly via suppression of the AGE-RAGE axis. Blockade of the increased glucose uptake into renal proximal tubular cells by empagliflozin might be a novel therapeutic target for tubulointerstitial damage in diabetic nephropathy.


Subject(s)
Benzhydryl Compounds/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diabetic Nephropathies/drug therapy , Fibromyalgia/prevention & control , Glucosides/pharmacology , Glycation End Products, Advanced/drug effects , Hypoglycemic Agents/pharmacology , Inflammation/prevention & control , Oxidative Stress/drug effects , Receptor for Advanced Glycation End Products/drug effects , Sodium-Glucose Transporter 2 Inhibitors , Animals , Diabetes Mellitus, Experimental/chemically induced , Diabetic Nephropathies/chemically induced , Gene Expression/drug effects , Male , Rats , Rats, Sprague-Dawley
8.
Horm Metab Res ; 47(4): 253-8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25105541

ABSTRACT

Advanced glycation end products (AGEs) decrease adiponectin expression and suppress insulin signaling in cultured adipocytes through the interaction with a receptor for AGEs (RAGE) via oxidative stress generation. We have recently found that high-affinity DNA aptamer directed against AGE (AGE-aptamer) prevents the progression of experimental diabetic nephropathy by blocking the harmful actions of AGEs in the kidney. This study examined the effects of AGE-aptamer on adipocyte remodeling, AGE-RAGE-oxidative stress axis, and adiponectin expression in fructose-fed rats. Although AGE-aptamer treatment by an osmotic mini pump for 8 weeks did not affect serum insulin levels, it significantly decreased average fasting blood glucose and had a tendency to inhibit body weight gain in fructose-fed rats. Furthermore, AGE-aptamer significantly suppressed the increase in adipocyte size and prevented the elevation in AGEs, RAGE, and an oxidative stress marker, 8-hydroxydeoxyguanosine (8-OHdG), levels in adipose tissues of fructose-fed rats at 14-week-old, while it restored the decrease in adiponectin mRNA levels. Our present study suggests that AGE-aptamer could improve glycemic control and prevent adipocyte remodeling in fructose-fed rats partly by suppressing the AGE-RAGE-mediated oxidative stress generation. AGE-aptamer might be a novel therapeutic strategy for fructose-induced metabolic derangements.


Subject(s)
Adipocytes/pathology , Aptamers, Nucleotide/administration & dosage , Blood Glucose/analysis , Fructose/administration & dosage , Glycation End Products, Advanced/antagonists & inhibitors , Receptor for Advanced Glycation End Products/antagonists & inhibitors , Adipocytes/drug effects , Adiponectin/genetics , Animals , Cell Size/drug effects , Diet , Gene Expression/drug effects , Glycation End Products, Advanced/genetics , Insulin Resistance , Male , Oxidative Stress/drug effects , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Weight Gain/drug effects
9.
Climacteric ; 18(3): 426-30, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25340263

ABSTRACT

Advanced glycation endproducts (AGEs) and their receptor (RAGE) play a role in vascular complications in diabetes. We have previously shown that 17ß-estradiol at 10 nmol/l, a nearly identical plasma concentration to that during mid-pregnancy, up-regulates RAGE expression in endothelial cells. The finding might suggest the involvement of 17ß-estradiol in the deterioration of vascular complications in diabetes during pregnancy. However, the effects of the selective estrogen receptor modulator, bazedoxifene, on oxidative and inflammatory reactions in AGEs-exposed endothelial cells remain unknown. In this study, we addressed the issue. Ten nmol/l 17ß-estradiol increased RAGE and monocyte chemoattractant protein-1 (MCP-1) gene and protein expression in human umbilical vein endothelial cells (HUVECs), both of which were blocked by 10 nmol/l bazedoxifene. Bazedoxifene at 10 nmol/l also significantly inhibited the AGEs-induced superoxide generation, RAGE and MCP-1 gene and protein expression in HUVECs. The present study suggests that blockade of the AGEs-RAGE axis by bazedoxifene might be a novel therapeutic target for preventing vascular damage in diabetes, especially in postmenopausal women.


Subject(s)
Chemokine CCL2/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Indoles/administration & dosage , Receptor for Advanced Glycation End Products/metabolism , Selective Estrogen Receptor Modulators/administration & dosage , Superoxides/chemistry , Cells, Cultured , Chemokine CCL2/genetics , Humans , Oxidative Stress/drug effects , Receptor for Advanced Glycation End Products/genetics
10.
Horm Metab Res ; 46(10): 717-21, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24710699

ABSTRACT

Advanced glycation end products (AGEs) and their receptor (RAGE) play a role in diabetic nephropathy. We have recently found that linagliptin, an inhibitor of dipeptidyl peptidase-4 (DPP-4) suppresses the AGE-induced oxidative stress generation and intercellular adhesion molecule-1 (ICAM-1) gene expression in endothelial cells. However, whether linagliptin could have beneficial effects on experimental diabetic nephropathy in a glucose-lowering independent manner remains unknown. To address the issue, this study examined the effects of linagliptin on renal damage in streptozotocin-induced diabetic rats. Serum levels of DPP-4 were significantly elevated in diabetic rats compared with control rats. Although linagliptin treatment for 2 weeks did not improve hyperglycemia in diabetic rats, linagliptin significantly reduced AGEs levels, RAGE gene expression, and 8-hydroxy-2'-deoxyguanosine, a marker of oxidative stress in the kidney of diabetic rats. Furthermore, linagliptin significantly reduced albuminuria, renal ICAM-1 mRNA levels, and lymphocyte infiltration into the glomeruli of diabetic rats. Our present study suggests that linagliptin could exert beneficial effects on diabetic nephropathy partly by blocking the AGE-RAGE-evoked oxidative stress generation in the kidney of streptozotocin-induced diabetic rats. Inhibition of DPP-4 by linagliptin might be a promising strategy for the treatment of diabetic nephropathy.


Subject(s)
Diabetes Mellitus, Type 1/complications , Diabetic Nephropathies/drug therapy , Purines/administration & dosage , Quinazolines/administration & dosage , Receptors, Immunologic/metabolism , Animals , Diabetic Nephropathies/etiology , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Dipeptidyl Peptidase 4/genetics , Glycation End Products, Advanced/metabolism , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Kidney/drug effects , Kidney/metabolism , Linagliptin , Male , Rats , Rats, Sprague-Dawley , Receptor for Advanced Glycation End Products , Receptors, Immunologic/genetics
11.
Horm Metab Res ; 46(6): 379-83, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24297485

ABSTRACT

Advanced glycation end products (AGEs) and their receptor (RAGE) system are involved in diabetic nephropathy. Angiotensin-converting enzyme 2 (ACE 2) plays a protective role against cardiovascular and renal injury by stimulating the production of angiotensin-(1-7) [Ang-(1-7)], an antagonist of angiotensin II (Ang II). However, effects of the AGEs-RAGE axis on ACE 2 expression in mesangial cells remain unknown. We examined here the role of ACE 2 in the AGEs-RAGE-induced mesangial cell damage and investigated whether olmesartan, one of the Ang II type 1 receptor blockers (ARB), prevented the deleterious effects of AGEs via restoration of ACE 2 and Ang-(1-7) level. AGEs significantly increased superoxide generation, upregulated RAGE mRNA level, and decreased ACE 2 gene expression and Ang-(1-7) production in mesangial cells, all of which were blocked by olmesartan, but not by a different type of ARB, azilsartan. An antioxidant, N-acetylcysteine or RAGE-antibodies also restored the decrease in ACE 2 mRNA level in AGEs-exposed mesangial cells. Moreover, olmesartan, but not azilsartan completely inhibited the AGEs-induced increase in vascular cell adhesion molecule-1 (VCAM-1) mRNA level in mesangial cells, which was abolished by the treatment with A-779, an antagonist of Ang-(1-7) receptor, Mas receptor. Our present study suggests that olmesartan could block the AGEs-induced VCAM-1 gene induction in mesangial cells by restoring the downregulated ACE 2 levels and subsequently stimulating the Ang-(1-7)-Mas receptor axis. Restoration of ACE 2 levels and blockade of renin-angiotensin system by olmesartan might be a promising strategy for the treatment of diabetic nephropathy.


Subject(s)
Glycation End Products, Advanced/pharmacology , Imidazoles/pharmacology , Mesangial Cells/metabolism , Peptidyl-Dipeptidase A/metabolism , Tetrazoles/pharmacology , Vascular Cell Adhesion Molecule-1/genetics , Angiotensin I , Angiotensin-Converting Enzyme 2 , Cell Line , Gene Expression Regulation/drug effects , Humans , Mesangial Cells/drug effects , Mesangial Cells/enzymology , Peptide Fragments , Receptor for Advanced Glycation End Products , Receptors, Immunologic/metabolism , Superoxides/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
12.
Reprod Domest Anim ; 48(6): 1001-5, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23808530

ABSTRACT

This study aimed to develop a polymerase chain reaction (PCR)-based sexing and effective semen collection methods for black-headed and straw-necked ibis species. However, most birds are not sexually dimorphic, that is, the sexes appear similar. Therefore, the gender should be determined before semen collection. DNA was extracted from the blood samples of 11 black-headed and 4 straw-necked ibis. The sex was determined after PCR amplification of the EE0.6 region of W-chromosome. The PCR products were separated using gel electrophoresis. A single band indicated the presence of the EE0.6 region and that the individual was a female, while no band indicated that the individual was a male. Further, the single bands from seven specimens were amplified. Semen collection was performed by massage or a combination of massage with electro-ejaculation and was attempted during all four seasons. The semen was successfully collected in March from male straw-necked ibis using the massage method. Limited motility, viability and concentration of straw-necked ibis sperm were observed. The sperm length was 180 µm and that of the nucleus was 30 µm with acrosome located at the tip of the nucleus. Thus, the PCR-based sexing proved to be an accurate molecular sexing method for black-headed and straw-necked ibis. Furthermore, we successfully collected semen and observed the stained sperm nucleus and acrosome of the straw-necked ibis sperm. We propose that the use of this PCR methodology can be applied as a routine method for sex determination and semen collection in ibis species for future ecological research. However, considering our limited success, further studies on semen collection method are required.


Subject(s)
Birds/physiology , Polymerase Chain Reaction/veterinary , Semen/physiology , Sex Determination Analysis/veterinary , Specimen Handling/veterinary , Animals , Female , Male , Polymerase Chain Reaction/methods , Specimen Handling/methods , Spermatozoa/cytology
13.
Horm Metab Res ; 45(5): 387-90, 2013 May.
Article in English | MEDLINE | ID: mdl-23225247

ABSTRACT

Metformin use has been reported to decrease breast cancer incidence and mortality in diabetic patients. We have previously shown that advanced glycation end products (AGEs) and their receptor (RAGE) interaction stimulate growth and/or migration of pancreatic cancer and melanoma cells. However, effects of metformin on AGEs-RAGE axis in breast cancers remain unknown. We examined here whether and how metformin could block the AGEs-induced growth and vascular endothelial growth factor (VEGF) expression in MCF-7 breast cancer cells. Cell proliferation was measured with an electron coupling reagent WST-1 based colorimetric assay. Gene expression level was evaluated by real-time reverse-transcription polymerase chain reactions. AGEs significantly increased cell proliferation of MCF-7 cells, which was completely prevented by the treatment with 0.01 or 0.1 mM metformin or anti-RAGE antibodies. Furthermore, metformin at 0.01 mM completely suppressed the AGEs-induced upregulation of RAGE and VEGF mRNA levels in MCF-7 cells. An inhibitor of AMP-activated protein kinase, compound C significantly blocked the growth-inhibitory and RAGE and VEGF suppressing effects of metformin in AGEs-exposed MCF-7 cells. Our present study suggests that metformin could inhibit the AGEs-induced growth and VEGF expression in MCF-7 breast cancer cells by suppressing RAGE gene expression via AMP-activated protein kinase pathway. Metformin may protect against breast cancer expansion in diabetic patients by blocking the AGEs-RAGE axis.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Glycation End Products, Advanced/pharmacology , Metformin/pharmacology , Receptors, Immunologic/metabolism , Vascular Endothelial Growth Factor A/genetics , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Receptor for Advanced Glycation End Products , Receptors, Immunologic/genetics , Vascular Endothelial Growth Factor A/metabolism
14.
Horm Metab Res ; 44(12): 891-5, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22864903

ABSTRACT

Advanced glycation end products (AGEs) and their receptor (RAGE) play a role in tubulointerstitial damage in diabetic nephropathy. Recently, metformin has been shown to ameliorate tubular injury both in cell culture and diabetic animal model. However, effects of metformin on AGEs-induced tubular cell apoptosis and damage remain unknown. We examined here whether and how metformin could block the AGEs-RAGE-elicited tubular cell injury in vitro. Gene expression level was evaluated by real-time reverse-transcription polymerase chain reactions. Reactive oxygen species (ROS) generation was measured with dihydroethidium staining. Apoptosis was evaluated by DNA fragmentation and annexin V expression level. AGEs upregulated RAGE mRNA levels and subsequently increased ROS generation and intercellular adhesion molecule-1, monocyte chemoattractant protein-1 and transforming growth factor-ß gene expression in human renal proximal tubular cells, all of which were significantly blocked by the treatment of 0.01 and 0.1 mM metformin. Compound C, an inhibitor of AMP-activated protein kinase significantly blocked the effects of metformin on RAGE gene expression and ROS generation in AGEs-exposed tubular cells. Furthermore, metformin dose-dependently inhibited the AGEs-induced apoptotic cell death of tubular cells; 1 mM metformin completely suppressed the pro-apoptotic effects of AGEs in 2 different assay systems. Our present study suggests that metformin could inhibit the AGEs-induced apoptosis and inflammatory and fibrotic reactions in tubular cells probably by reducing ROS generation via suppression of RAGE expression through AMP-activated protein kinase activation. Metformin may protect against tubular cell injury in diabetic nephropathy by blocking the AGEs-RAGE-ROS axis.


Subject(s)
Glycation End Products, Advanced/antagonists & inhibitors , Hypoglycemic Agents/pharmacology , Kidney Tubules, Proximal/drug effects , Metformin/pharmacology , Reactive Oxygen Species/antagonists & inhibitors , Receptors, Immunologic/metabolism , Up-Regulation/drug effects , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Apoptosis/drug effects , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , DNA Fragmentation/drug effects , Diabetic Nephropathies/prevention & control , Glycation End Products, Advanced/metabolism , Humans , Hypoglycemic Agents/antagonists & inhibitors , Hypoglycemic Agents/therapeutic use , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Metformin/antagonists & inhibitors , Metformin/therapeutic use , Molecular Targeted Therapy , Osmolar Concentration , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Receptor for Advanced Glycation End Products , Receptors, Immunologic/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
15.
Horm Metab Res ; 44(7): 501-5, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22581648

ABSTRACT

Glucose-dependent insulinotropic polypeptide (GIP) is one of the incretins, a gut hormone secreted from K cells in the intestine in response to food intake. It could be a potential therapeutic target for the treatment of patients with type 2 diabetes. However, effects of GIP on vascular injury remain unknown. Since interaction of advanced glycation end products (AGEs) with their receptor RAGE has been shown to play a crucial role in vascular damage in diabetes, this study investigated whether and how GIP blocked the deleterious effects of AGEs on human umbilical vein endothelial cells (HUVECs). GIP receptor was expressed in HUVECs. GIP, an analogue of cyclic AMP or inhibitors of NADPH oxidase inhibited the AGE-induced reactive oxygen species (ROS) generation in HUVECs. Furthermore, GIP reduced both RAGE mRNA and protein levels in HUVECs. GLP-1 also blocked the AGE-induced increase in mRNA levels of vascular cell adhesion molecule-1 (VCAM-1) and plasminogen activator inhibitor-1 in HUVECs. In addition, an antioxidant N-acetylcysteine mimicked the effects of GIP on RAGE and VCAM-1 gene expression in HUVECs. Our present study suggests that GIP could block the signal pathways of AGEs in HUVECs by reducing ROS generation and subsequent RAGE expression probably via GIP receptor-cyclic AMP axis.


Subject(s)
Antioxidants/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gastric Inhibitory Polypeptide/pharmacology , Glycation End Products, Advanced/pharmacology , Signal Transduction/drug effects , Gene Expression Regulation/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Reactive Oxygen Species/metabolism , Receptor for Advanced Glycation End Products , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
16.
J Int Med Res ; 40(2): 798-803, 2012.
Article in English | MEDLINE | ID: mdl-22613445

ABSTRACT

OBJECTIVE: This study investigated the effects of ezetimibe, an inhibitor of intestinal cholesterol absorption, on early phase diabetic nephropathy. METHODS: A total of 32 hypercholesterolaemic type 2 diabetes patients with microalbuminuria, defined as a urinary albumin excretion (UAE) 30 but < 300 mg/g creatinine, were enrolled. Various clinical and laboratory parameters were determined at baseline and after 6 months of treatment with 10 mg/day ezetimibe. RESULTS: Ezetimibe treatment significantly decreased glycated haemoglobin (HbA(1c)), low-density lipoprotein-cholesterol (LDL-C), triglycerides and UAE, and significantly increased high-density lipoprotein-cholesterol and albumin. It also decreased the serum level of monocyte chemoattractant protein-1 (MCP-1), but this difference was not statistically significant. Univariate analyses showed a correlation between UAE and body mass index, systolic and diastolic blood pressures, HbA(1c), LDL-C, estimated glomerular filtration rate (inverse), creatinine and MCP-1. Since these parameters may be closely correlated with each other, multiple stepwise regression analysis was performed and demonstrated that HbA(1c) and MCP-1 were independent determinants of UAE. CONCLUSIONS: Ezetimibe may be a promising therapeutic strategy for improving albumin excretion, partly through its anti-inflammatory properties, and for reducing LDL-C in hypercholesterolaemic type 2 diabetes patients with microalbuminuria.


Subject(s)
Albuminuria/drug therapy , Azetidines/therapeutic use , Cholesterol, LDL/blood , Hypercholesterolemia/drug therapy , Albumins/analysis , Azetidines/pharmacology , Blood Pressure , Body Mass Index , Chemokine CCL2/blood , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Diabetic Nephropathies/complications , Diabetic Nephropathies/drug therapy , Ezetimibe , Female , Glycated Hemoglobin/analysis , Humans , Hypercholesterolemia/complications , Male , Middle Aged , Triglycerides/blood
17.
Horm Metab Res ; 43(10): 731-4, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21932180

ABSTRACT

Sitagliptin is a stable inhibitor of dipeptidyl peptidase-IV, a responsible enzyme that mainly inactivates glucagon-like peptide-1 (GLP-1), and now one of the widely used agents for the treatment of diabetes. However, effects of sitagliptin on vascular injury are largely unknown. Since advanced glycation end products (AGEs) and their receptor (RAGE) axis contribute to vascular damage in diabetes, we investigated here whether sitagliptin inhibits the AGE-RAGE-induced endothelial cell damage in vitro. Although effects of 10 pM GLP-1 or 0.5 µM sitagliptin monotherapy on RAGE gene and protein expression were modest, combination therapy completely blocked the AGE-induced increase in RAGE mRNA and protein levels in human umbilical vein endothelial cells (HUVEC). AGEs induced reactive oxygen species (ROS) generation and reduced endothelial nitric oxide synthase (eNOS) mRNA level in HUVEC, both of which were also completely blocked by the treatment with 10 pM GLP-1 and 0.5 µM sitagliptin, but not with GLP-1 or sitagliptin monotherapy. Further, anti-RAGE antibody restored the decrease in eNOS mRNA level in AGE-exposed HUVEC. The present study suggests that sitagliptin augments the effects of GLP-1 on eNOS mRNA level in AGE-exposed HUVEC by suppressing RAGE expression and subsequent ROS generation. Sitagliptin may work as a vasoprotecitve agent in diabetes by blocking the AGE-RAGE axis.


Subject(s)
Glucagon-Like Peptide 1/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Protective Agents/pharmacology , Pyrazines/pharmacology , Receptors, Immunologic/metabolism , Triazoles/pharmacology , Gene Expression Regulation/drug effects , Glycation End Products, Advanced/pharmacology , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Reactive Oxygen Species/metabolism , Receptor for Advanced Glycation End Products , Receptors, Immunologic/genetics , Serum Albumin, Bovine/pharmacology , Sitagliptin Phosphate
18.
J Int Med Res ; 39(3): 880-90, 2011.
Article in English | MEDLINE | ID: mdl-21819721

ABSTRACT

Little is known about the association between plasma levels of pigment epithelium-derived factor (PEDF), coronary artery disease (CAD) and/or chronic kidney disease (CKD). This study evaluated 289 consecutive patients with chest pain or at least one coronary risk factor who underwent coronary angiography using multidetector row computed tomography (MDCT). Presence of CAD and CKD, CAD severity (i.e. number of significantly stenosed coronary vessels, described as vessel disease [VD]), coronary calcification scores, visceral fat area (VFA), subcutaneous fat area on MDCT, and metabolic biomarkers were recorded. PEDF levels correlated significantly with sex, VFA, CKD presence/hyperuricaemia and high-density lipoprotein cholesterol levels. PEDF levels were closely associated with CKD and were significantly higher in CKD patients than in non-CKD patients, regardless of the presence of CAD. CKD patients with two-VD or three-VD had higher plasma PEDF levels than non-CKD patients with two-VD or three-VD. It is concluded that PEDF may be associated with CKD regardless of the presence of CAD.


Subject(s)
Coronary Artery Disease/blood , Eye Proteins/blood , Kidney Diseases/blood , Multidetector Computed Tomography , Nerve Growth Factors/blood , Serpins/blood , Adult , Aged , Aged, 80 and over , Chronic Disease , Cross-Sectional Studies , Female , Humans , Male , Middle Aged , Risk Factors
19.
Horm Metab Res ; 43(9): 619-24, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21823057

ABSTRACT

Adenosine triphosphate-binding membrane cassette transporter A1 (ABCA1) and ABCG1 play a crucial role in macrophage cholesterol efflux, which is a novel therapeutic target for atherosclerosis. Advanced glycation end products (AGE) and their receptor RAGE axis is involved in accelerated atherosclerosis in diabetes as well. However, the role of AGE-RAGE axis in macrophage cholesterol efflux is not fully understood. We examined here whether AGE-RAGE axis could impair cholesterol efflux from human macrophage cells, THP-1 cells by suppressing ABCA1 and ABCG1 expression. We further investigated the effects of rosuvastatin on cholesterol efflux from AGE-exposed THP-1 cells. AGE increased reactive oxygen species generation in THP-1 cells, which was completely inhibited by rosuvastatin, anti-RAGE-antibody or diphenylene iodonium chloride (DPI), an inhibitor of NADPH oxidase. The antioxidative effect of rosuvastatin on AGE-exposed THP-1 cells was significantly prevented by geranylgeranyl pyrophosphate (GGPP). AGE decreased ABCA1 and ABCG1 mRNA levels, and subsequently reduced cholesterol efflux from THP-1 cells, which was prevented by GGPP. DPI mimicked the effects of rosuvastain. The results demonstrated that rosuvastatin could inhibit the AGE-induced reduction of THP-1 macrophage cholesterol efflux by suppressing NADPH oxidase activity via inhibition of geranylgeranylation of Rac-1. Our present study provides a novel beneficial aspect of rosuvastatin in diabetes; rosuvastain may prevent the development and progression of atherosclerosis in diabetes by not only reducing serum cholesterol level, but also by improving cholesterol efflux from foam cells of the arterial wall via blocking the harmful effects of AGE on macrophages.


Subject(s)
Cholesterol/metabolism , Down-Regulation/drug effects , Fluorobenzenes/pharmacology , Glycation End Products, Advanced/metabolism , Macrophages/metabolism , NADPH Oxidases/metabolism , Pyrimidines/pharmacology , Sulfonamides/pharmacology , rac1 GTP-Binding Protein/metabolism , ATP Binding Cassette Transporter 1 , ATP Binding Cassette Transporter, Subfamily G, Member 1 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Biological Transport/drug effects , Cell Line, Tumor , Humans , Macrophages/drug effects , NADPH Oxidases/genetics , Prenylation , Rosuvastatin Calcium , rac1 GTP-Binding Protein/genetics
20.
Horm Metab Res ; 43(3): 226-8, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20972939

ABSTRACT

Advanced glycation end products (AGEs) and their receptor (RAGE) have been shown to play a role in insulin resistance. We have previously shown that combination therapy with nateglinide (NAT) and telmisartan (TEL) improves postprandial metabolic derangements in Zucker fatty (ZF) rats, an animal model of insulin resistance with obesity. However, effects of combination therapy on insulin resistance remain unknown. We investigated here whether combination therapy with TEL and NAT could ameliorate insulin resistance in ZF rats by suppressing AGE-RAGE axis. NAT and/or TEL inhibited insulin receptor substrate-1 (IRS-1) serine phosphorylations at 307 and 636/639 residues in the liver of ZF rats. Further, combination therapy with NAT and TEL, but not each monotherapy alone, significantly restored the decrease in hepatic IRS-1 tyrosine phosphorylation in these animals. In addition, serum levels of AGEs, RAGE expression levels in the liver and hepatic AGE-RAGE index were decreased in NAT plus TEL-treated ZF rats. The present study suggests that combination therapy with NAT and TEL could ameliorate insulin resistance in ZF rats by suppressing the AGE-RAGE axis in the liver.


Subject(s)
Benzimidazoles/administration & dosage , Benzoates/administration & dosage , Cyclohexanes/administration & dosage , Down-Regulation , Insulin Resistance , Phenylalanine/analogs & derivatives , Receptors, Immunologic/metabolism , Animals , Disease Models, Animal , Drug Therapy, Combination , Glycation End Products, Advanced/metabolism , Humans , Liver/drug effects , Liver/metabolism , Male , Nateglinide , Phenylalanine/administration & dosage , Rats , Rats, Zucker , Receptor for Advanced Glycation End Products , Telmisartan
SELECTION OF CITATIONS
SEARCH DETAIL
...