Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
PLoS One ; 11(6): e0157012, 2016.
Article in English | MEDLINE | ID: mdl-27258009

ABSTRACT

Diabetic nephropathy is the leading cause of end-stage renal disease worldwide, but no effective therapeutic strategy is available. Because plasminogen activator inhibitor-1 (PAI-1) is increasingly recognized as a key factor in extracellular matrix (ECM) accumulation in diabetic nephropathy, this study examined the renoprotective effects of TM5275 and TM5441, two novel orally active PAI-1 inhibitors that do not trigger bleeding episodes, in streptozotocin (STZ)-induced diabetic mice. TM5275 (50 mg/kg) and TM5441 (10 mg/kg) were administered orally for 16 weeks to STZ-induced diabetic and age-matched control mice. Relative to the control mice, the diabetic mice showed significantly increased (p < 0.05) plasma glucose and creatinine levels, urinary albumin excretion, kidney-to-bodyweight ratios, glomerular volume, and fractional mesangial area. Markers of fibrosis and inflammation along with PAI-1 were also upregulated in the kidney of diabetic mice, and treatment with TM5275 and TM5441 effectively inhibited albuminuria, mesangial expansion, ECM accumulation, and macrophage infiltration in diabetic kidneys. Furthermore, in mouse proximal tubular epithelial (mProx24) cells, both TM5275 and TM5441 effectively inhibited PAI-1-induced mRNA expression of fibrosis and inflammation markers and also reversed PAI-1-induced inhibition of plasmin activity, which confirmed the efficacy of the TM compounds as PAI-1 inhibitors. These data suggest that TM compounds could be used to prevent diabetic kidney injury.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diabetic Nephropathies/drug therapy , Plasminogen Activator Inhibitor 1/metabolism , Animals , Cells, Cultured , Diabetes Mellitus, Experimental/complications , Diabetic Nephropathies/etiology , Disease Models, Animal , Extracellular Matrix/metabolism , Fibrinolytic Agents/therapeutic use , Glomerular Mesangium/metabolism , Immunohistochemistry , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Kidney Glomerulus/metabolism , Male , Mice , Mice, Inbred C57BL , Piperazines/therapeutic use , Reverse Transcriptase Polymerase Chain Reaction , para-Aminobenzoates/therapeutic use
2.
PLoS One ; 11(6): e0155564, 2016.
Article in English | MEDLINE | ID: mdl-27249020

ABSTRACT

BACKGROUND AND OBJECTIVES: Glucose control is a significant predictor of mortality in diabetic peritoneal dialysis (PD) patients. During PD, the local toxic effects of intra-peritoneal glucose are well recognized, but despite large amounts of glucose being absorbed, the systemic effects of this in non-diabetic patients are not clear. We sought to clarify whether dialysate glucose has an effect upon systemic glucose metabolism. METHODS AND MATERIALS: We analysed the Global Fluid Study cohort, a prospective, observational cohort study initiated in 2002. A subset of 10 centres from 3 countries with high data quality were selected (368 incident and 272 prevalent non-diabetic patients), with multilevel, multivariable analysis of the reciprocal of random glucose levels, and a stratified-by-centre Cox survival analysis. RESULTS: The median follow up was 5.6 and 6.4 years respectively in incident and prevalent patients. On multivariate analysis, serum glucose increased with age (ß = -0.007, 95%CI -0.010, -0.004) and decreased with higher serum sodium (ß = 0.002, 95%CI 0.0005, 0.003) in incident patients and increased with dialysate glucose (ß = -0.0002, 95%CI -0.0004, -0.00006) in prevalent patients. Levels suggested undiagnosed diabetes in 5.4% of prevalent patients. Glucose levels predicted death in unadjusted analyses of both incident and prevalent groups but in an adjusted survival analysis they did not (for random glucose 6-10 compared with <6, Incident group HR 0.92, 95%CI 0.58, 1.46, Prevalent group HR 1.42, 95%CI 0.86, 2.34). CONCLUSIONS: In prevalent non-diabetic patients, random glucose levels at a diabetic level are under-recognised and increase with dialysate glucose load. Random glucose levels predict mortality in unadjusted analyses, but this association has not been proven in adjusted analyses.


Subject(s)
Glucose/metabolism , Peritoneal Dialysis , Adult , Aged , Diabetes Mellitus/metabolism , Female , Humans , Male , Middle Aged , Prospective Studies
3.
Int Urol Nephrol ; 47(6): 1017-24, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25917484

ABSTRACT

PURPOSE: Poor glycemic control associates with increased mortality in diabetic (DM) dialysis patients, but it is less well established whether high blood glucose (BG) independent of pre-existing diabetic status associates with mortality in dialysis patients. We assessed factors affecting BG at the start of peritoneal dialysis (PD) and its mortality-predictive impact in Korean PD patients. METHODS: In 174 PD patients (55 % males, 56 % DM), BG, nutritional status, comorbidity (CMD), and residual renal function (RRF) were assessed in conjunction with dialysis initiation. Determinants of BG and its association with mortality after a mean follow-up period of 30 ± 24 months were analyzed. RESULTS: On Cox proportional hazards analysis comprising all patients, old age, high CMD score, presence of protein energy wasting, and low serum albumin (Salb) concentration were independent predictors of mortality but not a high-BG level, while in patients without pre-existing diabetic status, high BG, together with old age and high CMD score, was an independent predictor of mortality. After adjustment for age, CMD score, and Salb, the risk ratio for mortality increased by 12 % per 1 mg/dL increase in BG in the non-DM patients. Patient survival in patients without pre-existing diabetic status with high BG did not differ from DM patients, but the survival of patients with high BG was significantly lower than in patients with low BG. In patients without pre-existing diabetic status, in multiple regression analysis, high BG at initiation of PD associated with high age, high body mass index, and low RRF. CONCLUSIONS: High blood glucose at initiation of PD associated with an increased mortality risk in PD patients without pre-existing diabetic status suggesting that blood glucose monitoring and surveillance of factors contributing to poor glycemic control are warranted in patients initiating PD therapy.


Subject(s)
Hyperglycemia/complications , Peritoneal Dialysis , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/mortality , Blood Glucose/analysis , Diabetes Mellitus , Female , Humans , Male , Middle Aged , Predictive Value of Tests , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/therapy , Retrospective Studies
4.
J Ren Nutr ; 24(5): 336-42, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25167998

ABSTRACT

OBJECTIVES: It is controversial to what extent serum uric acid (SUA) is associated with mortality in patients with chronic kidney disease undergoing hemodialysis (HD). We analyzed the predictive role of SUA in the mortality of diabetic and nondiabetic chronic kidney disease patients starting on maintenance HD therapy. DESIGN AND SUBJECTS: SUA was measured at the initiation of HD therapy in 319 patients (137 females and 193 diabetic patients) with mean age of 60 ± 14 years and mean estimated glomerular filtration rate of 7.5 ± 3.8 mL/min/1.73 m(2). The patients were divided into 2 groups, hyperuricemia (HUA; n = 165) and non-HUA (n = 154) groups based on laboratory limit for normal SUA. Mortality was recorded during 31.5 ± 24.8 months. RESULTS: Among the 193 diabetic patients, but not among the whole group of 319 patients, survival was significantly lower in HUA than in non-HUA patients. Among diabetic patients 2-year patient survival was worse in patients with HUA and cardiovascular disease (CVD; 52.3%; n = 30) than in non-HUA patients with CVD (81.1%; n = 36), HUA without CVD (88.6%; n = 62), and non-HUA without CVD (93.9%; n = 65). Cox analysis in all 319 patients showed that, old age, CVD, other comorbidity, and low serum albumin but not high SUA predicted mortality. Among diabetic patients, predictors of increased mortality risk were old age, CVD, other comorbidity but also high SUA with adjusted hazard ratio of 1.12 (95% confidence interval 1.02-1.22) per 1 mg/dL increase in SUA. In diabetic patients with HUA and CVD, adjusted hazard ratio for mortality was 5.98 times that of diabetic non-HUA patients without CVD. CONCLUSIONS: High SUA is associated with poor survival in diabetic patients undergoing HD but not in nondiabetic patients undergoing HD. High SUA was found to be a risk marker especially in diabetic HD patients with concurrent CVD.


Subject(s)
Cardiovascular Diseases/epidemiology , Diabetes Mellitus/blood , Hyperuricemia/epidemiology , Renal Dialysis/mortality , Renal Insufficiency, Chronic/epidemiology , Uric Acid/blood , Aged , Biomarkers/blood , Cardiovascular Diseases/blood , Comorbidity , Female , Follow-Up Studies , Glomerular Filtration Rate , Humans , Hyperuricemia/blood , Male , Middle Aged , Proportional Hazards Models , Renal Insufficiency, Chronic/blood , Retrospective Studies , Risk Factors
5.
J Am Soc Nephrol ; 24(12): 2071-80, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24009237

ABSTRACT

Systemic inflammation, as evidenced by elevated inflammatory cytokines, is a feature of advanced renal failure and predicts worse survival. Dialysate IL-6 concentrations associate with variability in peritoneal small solute transport rate (PSTR), which has also been linked to patient survival. Here, we determined the link between systemic and intraperitoneal inflammation with regards to peritoneal membrane function and patient survival as part of the Global Fluid Study, a multinational, multicenter, prospective, combined incident and prevalent cohort study (n=959 patients) with up to 8 years of follow-up. Data collected included patient demographic characteristics, comorbidity, modality, dialysis prescription, and peritoneal membrane function. Dialysate and plasma cytokines were measured by electrochemiluminescence. A total of 426 survival endpoints occurred in 559 incident and 358 prevalent patients from 10 centers in Korea, Canada, and the United Kingdom. On patient entry to the study, systemic and intraperitoneal cytokine networks were dissociated, with evidence of local cytokine production within the peritoneum. After adjustment for multiple covariates, systemic inflammation was associated with age and comorbidity and independently predicted patient survival in both incident and prevalent cohorts. In contrast, intraperitoneal inflammation was the most important determinant of PSTR but did not affect survival. In prevalent patients, the relationship between local inflammation and membrane function persisted but did not account for an increased mortality associated with faster PSTR. These data suggest that systemic and local intraperitoneal inflammation reflect distinct processes and consequences in patients treated with peritoneal dialysis, so their prevention may require different therapeutic approaches; the significance of intraperitoneal inflammation requires further elucidation.


Subject(s)
Inflammation/mortality , Kidney Failure, Chronic/mortality , Peritoneal Dialysis/mortality , Peritonitis/mortality , Adult , Aged , Cohort Studies , Comorbidity , Cytokines/blood , Cytokines/immunology , Female , Humans , Incidence , Inflammation/immunology , Kidney Failure, Chronic/therapy , Male , Middle Aged , Multivariate Analysis , Peritoneum/immunology , Peritonitis/immunology , Predictive Value of Tests , Prevalence
6.
Diabetes ; 61(3): 728-38, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22315314

ABSTRACT

Mitochondrial reactive oxygen species (ROS) play an important role in diabetes complications, including diabetic nephropathy (DN). Plasma free fatty acids (FFAs) as well as glucose are increased in diabetes, and peroxisomes and mitochondria participate in FFA oxidation in an interconnected fashion. Therefore, we investigated whether deficiency of catalase, a major peroxisomal antioxidant, accelerates DN through peroxisomal dysfunction and abnormal renal FFA metabolism. Diabetes was induced by multiple injections of low-dose streptozotocin into catalase knock-out (CKO) and wild-type (WT) C57BL/6 mice. Murine mesangial cells (MMCs) transfected with catalase small interfering RNA followed by catalase overexpression were used to further elucidate the role of endogenous catalase. Despite equivalent hyperglycemia, parameters of DN, along with markers of oxidative stress, were more accelerated in diabetic CKO mice than in diabetic WT mice up to 10 weeks of diabetes. CKO mice and MMCs showed impaired peroxisomal/mitochondrial biogenesis and FFA oxidation. Catalase deficiency increased mitochondrial ROS and fibronectin expression in response to FFAs, which were effectively restored by catalase overexpression or N-acetylcysteine. These data provide unprecedented evidence that FFA-induced peroxisomal dysfunction exacerbates DN and that endogenous catalase plays an important role in protecting the kidney from diabetic stress through maintaining peroxisomal and mitochondrial fitness.


Subject(s)
Catalase/physiology , Diabetic Nephropathies/etiology , Peroxisomes/physiology , Animals , Cells, Cultured , Fatty Acids, Nonesterified/blood , Fibrosis , Kidney/pathology , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/physiology , Oxidative Stress , Reactive Oxygen Species/metabolism
7.
Nephrol Dial Transplant ; 27(1): 218-25, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21622994

ABSTRACT

BACKGROUND: Chronic kidney disease (CKD) is associated with increased risk for cardiovascular diseases (CVD). We hypothesized that inadequate angiogenic response in uremic patients could result from dysfunction of bone marrow-derived stromal cells [mesenchymal stem cells (MSCs)]. METHODS: We investigated whether MSCs are functionally competent in uremia induced by partial kidney ablation in C57Bl/6J mice. RESULTS: Uremic MSCs showed decreased expression of vascular endothelial growth factor (VEGF), VEGF receptor (VEGFR)1 and stromal cell-derived factor (SDF)-1α, increased cellular senescence, decreased proliferation, defects in migration in response to VEGF and SDF-1α and in vitro tube formation. Interestingly, the expression of fibroblast-specific protein-1 was higher in uremic MSCs. Uremia decreased hypoxia-inducible factor-1α, VEGF and VEGFR1 expression under hypoxia and Akt phosphorylation in both basal and VEGF-stimulated states. A diminished mitogenic effect on endothelial proliferation was observed in conditioned media from uremic MSCs. In addition, intravital microscopic analysis showed decreased angiogenesis in uremic MSCs. CONCLUSION: These results clearly demonstrate the functional incompetence in MSCs under uremic conditions and may significantly contribute to the disproportionately high risk for CVD in patients with CKD.


Subject(s)
Bone Marrow/pathology , Kidney Failure, Chronic/complications , Mesenchymal Stem Cells/pathology , Neovascularization, Pathologic , Uremia/etiology , Uremia/pathology , Animals , Biomarkers/metabolism , Blotting, Western , Bone Marrow/metabolism , Cell Adhesion , Cell Differentiation , Cell Movement , Cell Proliferation , Cells, Cultured , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Enzyme-Linked Immunosorbent Assay , Human Umbilical Vein Endothelial Cells/metabolism , Hypoxia/genetics , Hypoxia/metabolism , Kidney Failure, Chronic/etiology , Male , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Uremia/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism
8.
Perit Dial Int ; 32(1): 20-8, 2012.
Article in English | MEDLINE | ID: mdl-20798331

ABSTRACT

OBJECTIVE: Accumulation of visceral fat is one of the major risk factors for the development of cardiovascular disease in peritoneal dialysis (PD) patients. Adiponectin, an adipokine commonly regarded as a negative indicator of metabolic disease, is reported to be downregulated in its gene level in end-stage renal disease patients. Since excessive fat deposit is involved in increased reactive oxygen species (ROS), PD solution (PDS) may contribute to ROS production, resulting in dysregulation of adiponectin. In this study, we tested our hypothesis that oxidative stress induced by PDS may play a role in the regulation of adiponectin. METHODS: Commercial PDS containing 3.86% glucose (20 - 30 mL) was administered to SD rats for 12 weeks with and without N-acetylcysteine (NAC; 10 mmol/L). ELISA was used to quantify adiponectin in plasma and spent dialysate. For in vitro studies, fully differentiated 3T3-L1 adipocytes and adipocytes isolated from abdominal fat were treated with a high glucose solution, PDS, and H(2)O(2). Adiponectin levels in the conditioned media were measured by ELISA and immunoblot assays. The mRNA levels of adiponectin in mature adipocytes were examined using real-time RT-PCR. RESULTS: The levels of adiponectin in plasma and spent dialysate were significantly downregulated by PDS and this effect was suppressed by NAC. In 3T3-L1 adipocytes, adiponectin secretion was inhibited by 50 mmol/L glucose, PDS diluted 2-fold, and H(2)O(2) (200 µmol/L). In addition, H(2)O(2) downregulated expression of adiponectin mRNA and secretion of adiponectin oligomer complexes. CONCLUSIONS: Our data suggest that ROS induced by conventional glucose-based PDS may contribute to pathophysiological changes in abdominal fat and downregulate adiponectin secreted from adipocytes during long-term PD.


Subject(s)
Adiponectin/biosynthesis , Dialysis Solutions/administration & dosage , Glucose/administration & dosage , Oxidative Stress/drug effects , Peritoneal Dialysis , Peritoneum/metabolism , Adipocytes/metabolism , Adipocytes/pathology , Adiponectin/genetics , Animals , Blotting, Western , Cells, Cultured , Disease Management , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation/drug effects , Male , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Sweetening Agents/pharmacology
9.
J Nephrol ; 24 Suppl 17: S93-6, 2011.
Article in English | MEDLINE | ID: mdl-21614787

ABSTRACT

Korea has a long tradition of Eastern (Korean) medicine. Modern nephrology in Korea began in the 20th century after the Second World War. The first nephrology report was presented in 1949 at the third annual meeting of the Korean Society of Internal Medicine. The first hemodialysis was performed in 1952 during the Korean War, the first acute peritoneal dialysis in 1957, the first kidney biopsy in 1959, the first kidney transplantation in 1969 and the first continuous ambulatory peritoneal dialysis in 1981. National Health Insurance was launched in 1976 with a limited coverage, which was gradually expanded to cover all Korean nationals in 1989. The Korean Society of Nephrology (KSN) was inaugurated in 1980. The KSN End-Stage Renal Disease (ESRD) Registry was started in 1985. Brain death legislation was enacted, and the Korean network for organ sharing launched in 2000. During 2009, a total of 8,906 patients or 175.9 patients per million population (PMP) began renal replacement therapy (RRT), and at the end of the year a total of 56,396 patients or 1,113.6 patients PMP were on RRT in Korea. The prevalence of ESRD continues to grow in Korea while its incidence is stable or declining as compared with the previous year.


Subject(s)
Nephrology/history , Renal Replacement Therapy/history , History, 20th Century , History, 21st Century , Humans , Kidney Failure, Chronic/epidemiology , Kidney Failure, Chronic/therapy , Kidney Transplantation/history , Korea/epidemiology , Peritoneal Dialysis/history , Prevalence , Renal Dialysis/history
10.
Nephrol Dial Transplant ; 26(11): 3474-84, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21478303

ABSTRACT

BACKGROUND: Despite the beneficial effects of alagebrium (ALA), a putative advanced glycation end-product (AGE) breaker, on diabetic nephropathy, its renoprotective mechanisms are incompletely understood. Since oxidative stress exacerbates diabetic renal injury through interaction with AGE, the present study examined the antioxidative property of ALA in db/db mice, mesangial cells cultured under high glucose or H(2)O(2) and a test tube. METHODS: ALA (2 mg/kg/day) was administered intraperitoneally for 12 weeks to 8-week-old db/m and db/db (D(ALA)E) mice or for 4 weeks to 16-week-old db/db mice (D(ALA)L). Oxidative stress markers (nitrotyrosine accumulation, expression and translocation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase subunits, cellular DCF-DA fluorescence) together with urinary albumin excretion and histological changes including mesangial expansion were measured. The concentration of H(2)O(2) in the presence and absence of ALA was measured by iodometric analysis in a test tube. RESULTS: ALA significantly reduced not only urinary albumin excretion and renal pathological changes but also accumulation of pentosidine and nitrotyrosine and expression of NADPH oxidase subunits in db/db mice regardless of treatment protocol. In mesangial cells, ALA effectively prevented not only high glucose- but also H(2)O(2)-induced membrane translocation of NADPH oxidase subunit (p47 phox, p67 phox and rac1) and protein kinase C isoform (α, ßI and ßII) and Nox4 messenger RNA expression concomitant with cellular reactive oxygen species. Furthermore, ALA directly decreased H(2)O(2) in a test tube. CONCLUSION: ALA has both direct and indirect antioxidant effects that may play important roles in ALA's renoprotective effect in diabetic kidneys.


Subject(s)
Acute Kidney Injury/prevention & control , Antioxidants/therapeutic use , Diabetes Mellitus, Experimental/physiopathology , Diabetic Nephropathies/prevention & control , Mesangial Cells/drug effects , Oxidative Stress/drug effects , Thiazoles/therapeutic use , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Albuminuria/etiology , Albuminuria/prevention & control , Animals , Blotting, Western , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Hydrogen Peroxide/pharmacology , Immunoenzyme Techniques , Male , Mesangial Cells/metabolism , Mice , Mice, Inbred C57BL , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Protein Kinase C/genetics , Protein Kinase C/metabolism , RNA, Messenger/genetics , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction
11.
Nephrol Dial Transplant ; 25(11): 3742-8, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20466690

ABSTRACT

BACKGROUND: It is well established that the survival rate of diabetic end-stage renal disease patients remains the lowest among all primary diagnoses probably because of higher prevalence of cardiovascular diseases (CVD) associated with diabetes. This study was designed to evaluate the impact of CVD and other risk factors individually or in combination on mortality in diabetic peritoneal dialysis (PD) patients. METHODS: In a retrospective study, 213 incident PD patients [118 had diabetes mellitus (DM), 94 were female, mean age 55 ± 13 years] underwent initial assessment of nutritional status, comorbid disease (CMD) survey, residual renal function (RRF), dialysis adequacy and peritoneal transport characteristics at a mean of 9 days (range, 3-24 days) after start of PD and were then followed for 30 ± 24 months (range, 3-115 months). Of 213 patients, 154 patients were reassessed after a mean of 11 months (range, 6-19 months). Nutritional status was assessed by subjective global assessment and other methods. CMD was graded by Davies index and included DM, CVD, liver disease and respiratory disease. RESULTS: On Kaplan-Meier analysis, patient survival was significantly lower in female DM patients compared to other groups. The 3-year patient survival rate was 46, 70, 82 and 83% for female DM, male DM, male non-DM and female non-DM, respectively (P = 0.003). On Cox proportional hazards multivariate analysis including all patients, old age, presence of CVD or protein-energy wasting (PEW), low serum albumin concentration and low RRF were independent predictors of mortality but not DM per se or female gender. In DM patients, old age, female gender, presence of CVD or PEW and low RRF were independent predictors of mortality while old age was the only risk factor in non-DM patients. After adjustment for age, gender and RRF, DM patients with both CVD and PEW had a risk of mortality that was 3.3 times that of DM patients without CVD and PEW. In DM patients without CVD and PEW, patient survival was not different from that of non-DM patients without CVD and PEW. CONCLUSIONS: DM per se was not a risk factor for mortality in this group of PD patients. Instead, the higher mortality rate in diabetic PD patients, in particular among female patients, was mainly attributable to concurrent morbidity such as CVD and PEW, together with low RRF.


Subject(s)
Diabetic Nephropathies/mortality , Peritoneal Dialysis/mortality , Adult , Aged , Aged, 80 and over , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/mortality , Cause of Death , Diabetic Nephropathies/therapy , Female , Humans , Kidney/physiopathology , Kidney Failure, Chronic/mortality , Kidney Failure, Chronic/therapy , Male , Middle Aged , Prevalence , Proportional Hazards Models , Protein-Energy Malnutrition/epidemiology , Retrospective Studies , Risk Factors , Survival Rate
13.
Am J Nephrol ; 30(6): 481-90, 2009.
Article in English | MEDLINE | ID: mdl-19786738

ABSTRACT

BACKGROUND/AIMS: Plasminogen activator inhibitor (PAI)-1 is increasingly recognized as a profibrotic factor but the mechanisms are not entirely clear. The present study examined the profibrotic mechanism of PAI-1 focusing on its effect on transforming growth factor (TGF)-beta1 in experimental diabetes. METHODS: PAI-1 knockout (KO) mesangial cells cultured under high glucose (HG) in addition to streptozotocin-induced diabetic PAI-1 KO mice were used. RESULTS: PAI-1 deficiency did not affect plasma glucose significantly but reduced the fractional mesangial area, fibronectin and collagen I expression in the renal cortex after 20 weeks of diabetes as well as in HG-stimulated mesangial cells along with suppression of TGF-beta1 mRNA expression. PAI-1 deficiency also reduced HG-induced betaig-h3, a TGF-beta1-induced gene product, mRNA expression. All these losses-of-function in PAI-1 KO mesangial cells were effectively gained by recombinant PAI-1. Recombinant PAI-1-induced fibronectin and collagen I expression was abrogated by TGF-beta1 receptor inhibitor or anti-TGF-beta antibody suggesting that the effect of PAI-1 was mediated by TGF-beta1. In a similar context, recombinant PAI-1 stimulated TGF-beta1 promoter activity to the same extent as TGF-beta1 itself. CONCLUSION: Since TGF-beta1 is well known to stimulate the PAI-1 promoter, we suggest that TGF-beta1 and PAI-1 together constitute a positive feedback loop in the development of renal fibrosis in diabetes.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/metabolism , Feedback, Physiological/physiology , Serpins/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Blood Glucose/metabolism , Cell Line, Transformed , Cells, Cultured , Collagen Type I/metabolism , Collagen Type IV/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/physiopathology , Diabetic Nephropathies/pathology , Diabetic Nephropathies/physiopathology , Fibrinolysin/metabolism , Fibronectins/metabolism , Fibrosis , Humans , Male , Matrix Metalloproteinase 9/metabolism , Mesangial Cells/metabolism , Mesangial Cells/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Osmotic Pressure/physiology , Promoter Regions, Genetic/physiology , Proteinuria/metabolism , Proteinuria/pathology , Proteinuria/physiopathology , RNA, Messenger/metabolism , Serpin E2 , Serpins/genetics , Transforming Growth Factor beta1/genetics
14.
Am J Physiol Renal Physiol ; 297(3): F729-39, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19553350

ABSTRACT

Excessive accumulation of extracellular matrix (ECM) in the kidneys and epithelial-to-mesenchymal transition (EMT) of renal tubular epithelial cells contributes to the renal fibrosis that is associated with diabetic nephropathy. Histone deacetylase (HDAC) determines the acetylation status of histones and thereby controls the regulation of gene expression. This study examined the effect of HDAC inhibition on renal fibrosis induced by diabetes or transforming growth factor (TGF)-beta1 and determined the role of reactive oxygen species (ROS) as mediators of HDAC activation. In streptozotocin (STZ)-induced diabetic kidneys and TGF-beta1-treated normal rat kidney tubular epithelial cells (NRK52-E), we found that trichostatin A, a nonselective HDAC inhibitor, decreased mRNA and protein expressions of ECM components and prevented EMT. Valproic acid and class I-selective HDAC inhibitor SK-7041 also showed similar effects in NRK52-E cells. Among the six HDACs tested (HDAC-1 through -5 and HDAC-8), HDAC-2 activity significantly increased in the kidneys of STZ-induced diabetic rats and db/db mice and TGF-beta1-treated NRK52-E cells. Levels of mRNA expression of fibronectin and alpha-smooth muscle actin were decreased, whereas E-cadherin mRNA was increased when HDAC-2 was knocked down using RNA interference in NRK52-E cells. Interestingly, hydrogen peroxide increased HDAC-2 activity, and the treatment with an antioxidant, N-acetylcysteine, almost completely reduced TGF-beta1-induced activation of HDAC-2. These findings suggest that HDAC-2 plays an important role in the development of ECM accumulation and EMT in diabetic kidney and that ROS mediate TGF-beta1-induced activation of HDAC-2.


Subject(s)
Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 2/complications , Diabetic Nephropathies/enzymology , Histone Deacetylases/metabolism , Kidney/enzymology , Repressor Proteins/metabolism , Transforming Growth Factor beta1/metabolism , Acetylcysteine/pharmacology , Amides/pharmacology , Animals , Antioxidants/pharmacology , Biphenyl Compounds/pharmacology , Cell Line , Cell Transdifferentiation , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/enzymology , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/pathology , Diabetic Nephropathies/etiology , Diabetic Nephropathies/pathology , Diabetic Nephropathies/prevention & control , Enzyme Inhibitors/pharmacology , Extracellular Matrix Proteins/genetics , Fibrosis , Gene Expression Regulation , Histone Deacetylase 2 , Histone Deacetylase Inhibitors , Histone Deacetylases/genetics , Humans , Hydroxamic Acids/pharmacology , Kidney/drug effects , Kidney/pathology , Male , Mice , RNA Interference , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Recombinant Proteins/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Valproic Acid/pharmacology
15.
Perit Dial Int ; 29 Suppl 2: S132-4, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19270202

ABSTRACT

The survival of patients with end-stage renal disease (ESRD) resulting from diabetes continues to improve, but the survival rate among diabetic ESRD patients remains the lowest among all primary diagnoses probably because of the higher prevalence of cardiovascular comorbidity associated with diabetes. Diabetes, age, and comorbidity all significantly modify the effect of treatment modality on patient survival. As compared with hemodialysis (HD), peritoneal dialysis (PD) offers an equal or lower risk of death across all subgroups during the first 1-2 years of dialysis. The association of PD with better outcomes than are seen with HD is probably a result of a lower prevalence of infections and congestive heart failure and better preservation of residual renal function (RRF) in PD patients. Use of angiotensin converting-enzyme inhibitor (ACEI) or angiotensin II receptor blocker (ARB) helps to preserve RRF in ESRD patients and to maintain peritoneal membrane integrity longer in PD patients. Antioxidants can also support preservation of peritoneal membrane function. Peritoneal dialysis should be the initial modality of dialysis in all ESRD patients. Older patients (age > or = 45 years) with diabetes and patients without diabetes may switch to HD or receive a kidney graft in 1-2 years' time; younger patients (age < 45 years) with diabetes may stay on PD longer. Use of ACEI and ARB or antioxidants can help to maintain peritoneal membrane function longer.


Subject(s)
Diabetes Mellitus/therapy , Kidney Failure, Chronic/therapy , Peritoneal Dialysis/standards , Diabetes Mellitus/mortality , Global Health , Glomerular Filtration Rate/physiology , Humans , Kidney Failure, Chronic/complications , Kidney Failure, Chronic/mortality , Survival Rate/trends , Treatment Outcome
16.
Blood Purif ; 27(2): 165-71, 2009.
Article in English | MEDLINE | ID: mdl-19141994

ABSTRACT

BACKGROUND/AIM: Peritoneal dialysis (PD) patient survival is influenced by many factors and there is no consensus on the relative importance of these predictors, independently or combined. This study was designed to evaluate how these independent factors, alone or in various combinations, may influence PD patient survival. METHODS: A peritoneal equilibration test, subjective global assessment (SGA), and comorbid diseases (CMD) were assessed. RESULTS: On multivariate analysis, age (>60 years), CMD, malnutrition, and low RRF (

Subject(s)
Peritoneal Dialysis/mortality , Adult , Age Factors , Aged , Aged, 80 and over , Comorbidity , Female , Humans , Kidney Failure, Chronic/mortality , Kidney Function Tests , Korea/epidemiology , Male , Malnutrition/complications , Middle Aged , Nutritional Status , Peritoneal Dialysis/adverse effects , Risk Factors
17.
Diabetes Res Clin Pract ; 82 Suppl 1: S42-5, 2008 Nov 13.
Article in English | MEDLINE | ID: mdl-18845352

ABSTRACT

There is an increasing evidence that reactive oxygen species (ROS) play a major role in the development of diabetic complications. Oxidative stress is increased in diabetes and the overproduction of ROS in diabetes is a direct consequence of hyperglycemia. Various types of vascular cells including renal cells are able to produce ROS under hyperglycemic condition. Both NADPH oxidase and mitochondrial electron gradient play roles in hyperglycemia-induced ROS generation. In addition to their ability to directly inflict macromolecular damage, ROS can function as signaling molecules. ROS mediate hyperglycemia-induced activation of signal transduction cascades and transcription factors leading to transcriptional activation of profibrotic genes in the kidney. Furthermore, ROS-activated signaling molecules generate and signal through ROS and thus ROS act as a signal amplifier. Intensive glycemic control and inhibition of angiotensin II delay the onset and progression of diabetic nephropathy, in part, through prevention of overproduction of ROS. Conventional and catalytic antioxidants have been shown to prevent or delay the onset of diabetic nephropathy. Combination of strategies to prevent overproduction of ROS and to increase the removal of preformed ROS may prove to be effective in preventing the development and progression of diabetic nephropathy.


Subject(s)
Diabetic Nephropathies/metabolism , Reactive Oxygen Species/metabolism , Animals , Antioxidants/therapeutic use , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/etiology , Humans , Mitochondria/metabolism , Signal Transduction
18.
J Korean Med Sci ; 22(6): 943-5, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18162703

ABSTRACT

A growing body of evidence indicates that epithelial-mesenchymal transition (EMT) of human peritoneal mesothelial cells (HPMC) may play an important role in the development and progression of peritoneal fibrosis during long-term peritoneal dialysis (PD) leading to failure of peritoneal membrane function. Here, we review our own observations and those of others on the mechanisms of EMT of HPMC and suggest potential therapeutic strategies to prevent EMT and peritoneal fibrosis during long-term PD. We found that high glucose and H2O2 as well as transforming growth factor-beta1 (TGF-beta1) induced EMT in HPMC and that high glucoseinduced EMT was blocked not only by inhibition of TGF-beta1 but also by antioxidants or inhibitors of mitogen-activated protein kinases (MAPK). Since MAPKs are downstream target molecules of reactive oxygen species (ROS), these data suggest that high glucose-induced generation of ROS and subsequent MAPK activation mediate high glucose-induced EMT in HPMC. We and others also observed that bone morphogenetic protein-7 (BMP-7) prevented EMT in HPMC. Glucose degradation products (GDP) were shown to play a role in inducing EMT. Involvement of a mammalian target of rapamycin (mTOR) in TGF-beta1-induced EMT has also been proposed in cultured HPMC. A better understanding of the precise mechanisms involved in EMT of HPMC may provide new therapeutic strategies for inhibiting peritoneal fibrosis in long-term PD patients.


Subject(s)
Epithelial Cells/pathology , Mesoderm/pathology , Peritoneal Dialysis/adverse effects , Peritoneum/pathology , Fibrosis , Humans
19.
Nephrology (Carlton) ; 10 Suppl: S11-3, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16174279

ABSTRACT

Diabetic nephropathy is characterized by excessive accumulation of extracellular matrix (ECM) in the kidney. Decreased ECM degradation as well as increased ECM synthesis plays an important role in ECM remodeling that favours tissue fibrosis. Plasminogen activator (PA)/plasmin/PA inhibitor (PAI) system is involved in ECM degradation and PAI-1 plays a critical role in ECM remodeling in the kidney. Normal human kidneys do not express PAI-1 but PAI-1 is overexpressed in pathologic conditions associated with renal fibrosis including diabetic nephropathy. Reactive oxygen species mediate PAI-1 up-regulation in renal cells cultured under high glucose, hypoxia, and TGF-beta1. Recent studies utilizing PAI-1 deficient mice suggest that PAI-1 induce ECM deposition in diabetic kidney through increased ECM synthesis by TGF-beta1 up-regulation as well as through decreased ECM degradation by suppression of plasmin and MMP-2 activity.


Subject(s)
Diabetic Nephropathies/metabolism , Kidney/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Animals , Extracellular Matrix/metabolism , Humans
20.
Nephrology (Carlton) ; 10 Suppl: S7-10, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16174288

ABSTRACT

Diabetic nephropathy is characterized by excessive accumulation of extracellular matrix (ECM) in the kidney. Reactive oxygen species (ROS) play a central role in the ECM synthesis and degradation in the glomeruli and tubulointerstitium leading to renal fibrosis. High glucose (HG) induces cellular ROS through protein kinase C (PKC)-dependent activation of NADPH oxidase and through mitochondrial metabolism. ROS thus generated activate signal transduction cascade (PKC, mitogen-activated protein kinases, and janus kinase/signal transducers and activators of transcription) and transcription factors (nuclear factor-kappaB, activated protein-1, and specificity protein-1), up-regulate transforming growth factor-beta1 (TGF-beta1), angiotensin II (Ang II), monocyte chemoattractant protein-1 (MCP-1), and plasminogen activator inhibitor-1 (PAI-1) gene and protein expression, and promote formation of advanced glycation end-products (AGE). PKC, TGF-beta1, Ang II, and AGE also induce cellular ROS and signal through ROS leading to enhanced ECM synthesis. NF-kappaB-MCP-1 pathway is activated by ROS and promotes monocyte recruitment and profibrotic process in the kidney. HG- and TGF-beta1-induced PAI-1 up-regulation is mediated by ROS and contribute to ECM accumulation via suppression of plasmin ativity. TGF-beta1-induced myofibroblast transformation of renal tubular epithelial cells (epithelial-mesenchymal transition) is also mediated by ROS and contribute to tubulointerstitial fibrosis. In summary, ROS transduce and amplify glucose signalling in renal cells under high glucose environment and play a critical role in excessive ECM deposition in the diabetic kidney. A better understanding of ROS production and removal will allow more effective therapeutic strategies in diabetic renal and other vascular complications.


Subject(s)
Diabetic Nephropathies/metabolism , Glucose/administration & dosage , Glucose/metabolism , Kidney/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Animals , Cells, Cultured , Diabetic Nephropathies/pathology , Dose-Response Relationship, Drug , Glucose/pharmacology , Humans , Kidney/cytology , Kidney/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...