Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
JMA J ; 6(4): 523-526, 2023 Oct 16.
Article in English | MEDLINE | ID: mdl-37941684

ABSTRACT

Clinical practice guidelines (CPGs) consist of clinical questions (CQs) and corresponding recommendations. Considering the estimation of body of evidence, patients' opinions, and medical economics, recommendations can vary depending on the votes of the committee members of CPGs. Taking this into consideration, concerns have already been raised on how financial conflict of interest (COI) potentially influences recommendations. In this study, we developed the third edition of guideline for the management of hyperuricemia and gout. This CPG was composed of seven CQs and recommendations. The direction and strength of the recommendations were determined by votes. There are three CQs. Individual questions asked whether uric acid-lowering-agents (ULAs) could be applied to hyperuricemic patients with chronic kidney disease (CKD) (CQ A), hypertension (CQ B), or heart failure (CQ C) to prevent organ damage. We examined whether the absence (18 members) or presence (8 members) of COIs of committee members could influence the votes. In total, 26 committee members with and without COI have equally determined the direction and strength of recommendations. In CQ A, members without financial COIs and those with financial COI selected conditional recommendation for the use of ULAs in patients with CKD (without COI, 17/18; with COI, 7/8). In CQ B, members without financial COIs and those with financial COI selected conditional recommendation against the use of ULAs in hypertensive patients (without COI, 14/18; with COI, 5/8). In CQ C, members without financial COIs and those with financial COIs have selected conditional recommendation against the use of ULAs in patients suffering from heart failure (without COI, 15/18; with COI, 4/8). We found that members with financial COIs have determined their recommendations in the same direction and strength as those without financial COIs.

2.
Biomolecules ; 13(10)2023 10 13.
Article in English | MEDLINE | ID: mdl-37892201

ABSTRACT

The prevalence of patients with hyperuricemia or gout is increasing worldwide. Hyperuricemia and gout are primarily attributed to genetic factors, along with lifestyle factors like consuming a purine-rich diet, alcohol and/or fructose intake, and physical activity. While numerous studies have reported various comorbidities linked to hyperuricemia or gout, the range of these associations is extensive. This review article focuses on the relationship between uric acid and thirteen specific domains: transporters, genetic factors, diet, lifestyle, gout, diabetes mellitus, metabolic syndrome, atherosclerosis, hypertension, kidney diseases, cardiovascular diseases, neurological diseases, and malignancies. The present article provides a comprehensive review of recent developments in these areas, compiled by experts from the Young Committee of the Japanese Society of Gout and Uric and Nucleic Acids. The consolidated summary serves to enhance the global comprehension of uric acid-related matters.


Subject(s)
Gout , Hyperuricemia , Metabolic Syndrome , Humans , Uric Acid , Diet
3.
Biomedicines ; 11(5)2023 Apr 24.
Article in English | MEDLINE | ID: mdl-37238929

ABSTRACT

Uric acid (UA) forms monosodium urate (MSU) crystals to exert proinflammatory actions, thus causing gout arthritis, urolithiasis, kidney disease, and cardiovascular disease. UA is also one of the most potent antioxidants that suppresses oxidative stress. Hyper andhypouricemia are caused by genetic mutations or polymorphism. Hyperuricemia increases urinary UA concentration and is frequently associated with urolithiasis, which is augmented by low urinary pH. Renal hypouricemia (RHU) is associated with renal stones by increased level of urinary UA, which correlates with the impaired tubular reabsorption of UA. Hyperuricemia causes gout nephropathy, characterized by renal interstitium and tubular damage because MSU precipitates in the tubules. RHU is also frequently associated with tubular damage with elevated urinary beta2-microglobulin due to increased urinary UA concentration, which is related to impaired tubular UA reabsorption through URAT1. Hyperuricemia could induce renal arteriopathy and reduce renal blood flow, while increasing urinary albumin excretion, which is correlated with plasma xanthine oxidoreductase (XOR) activity. RHU is associated with exercise-induced kidney injury, since low levels of SUA could induce the vasoconstriction of the kidney and the enhanced urinary UA excretion could form intratubular precipitation. A U-shaped association of SUA with organ damage is observed in patients with kidney diseases related to impaired endothelial function. Under hyperuricemia, intracellular UA, MSU crystals, and XOR could reduce NO and activate several proinflammatory signals, impairing endothelial functions. Under hypouricemia, the genetic and pharmacological depletion of UA could impair the NO-dependent and independent endothelial functions, suggesting that RHU and secondary hypouricemia might be a risk factor for the loss of kidney functions. In order to protect kidney functions in hyperuricemic patients, the use of urate lowering agents could be recommended to target SUA below 6 mg/dL. In order to protect the kidney functions in RHU patients, hydration and urinary alkalization may be recommended, and in some cases an XOR inhibitor might be recommended in order to reduce oxidative stress.

4.
Intern Med ; 62(13): 1915-1920, 2023 Jul 01.
Article in English | MEDLINE | ID: mdl-36418091

ABSTRACT

Objective Both renal hypouricemia (RHU) and gout are associated with renal dysfunction and urolithiasis. The difference in renal complications associated with RHU and gout, however, has not been studied. We characterized the urate metabolism and complications of patients with RHU and compared them with patients with gout. Methods Eighteen patients with RHU who had a serum uric acid (SUA) level <2 mg/dL (10 men and 8 women), 44 patients with gout (44 men) and 16 normouricemic patients (4 men and 12 women) were included. The blood and urinary biochemical data were evaluated. A genetic analysis of uric acid transporter 1 (URAT1) was also conducted in 15 cases with RHU. Results The SUA level of RHU was 0.9±0.5/mg/dL, and the Uur/Ucr and Cur/Ccr were 0.56±0.14% and 45.7±18.0%, respectively. A genetic analysis of URAT1 in 15 RHU patients showed that 13 harbored a URAT1 gene mutation, whereas 2 harbored the wild-type gene. The SUA level was significantly lower in RHU patients (n=11) than in either gout patients (n=44) or normouricemic patients (n=16). This reduction was accompanied by the elevation of Cua/Ccr. Urinary beta 2-microglobulin levels were higher in RHU patients than in gout or normouricemia patients. Cua/Ccr correlated with normalized urinary beta 2-microglobulin levels. The prevalence of urolithiasis was 18.2% in RHU cases and 6.8% in gout cases. A homozygous URAT1 mutation was associated with urolithiasis. Conclusion Besides urolithiasis, RHU can be associated with tubular dysfunction, such as elevated urinary beta 2-microglobulin levels.


Subject(s)
Gout , Urinary Calculi , Male , Humans , Female , Uric Acid , beta 2-Microglobulin , Gout/complications , Gout/genetics , Urinary Calculi/complications , Urinary Calculi/genetics
5.
Intern Med ; 61(9): 1383-1386, 2022.
Article in English | MEDLINE | ID: mdl-35491177

ABSTRACT

Whether or not extremely low levels of serum uric acid (SUA) in xanthinuria are associated with impairment of the endothelial function and exercise-induced acute kidney injury (EIAKI) is unclear. A 59-year-old woman without EIAKI or urolithiasis had undetectable levels of UA in serum and urine and elevated levels of hypoxanthine and xanthine in urine. A genetic analysis revealed homozygous mutations in the XDH gene [c.1585 C>T (p. Gln529*)]. Flow-mediated dilation was within the normal range. This is the first report of a case with extremely low levels of SUA, xanthinuria with novel mutations of xanthine dehydrogenase (XDH) and a normal endothelial function.


Subject(s)
Metabolism, Inborn Errors , Xanthine Dehydrogenase , Female , Humans , Metabolism, Inborn Errors/genetics , Middle Aged , Mutation/genetics , Uric Acid , Xanthine Dehydrogenase/deficiency , Xanthine Dehydrogenase/genetics
6.
Hum Cell ; 32(2): 83-87, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30783949

ABSTRACT

Renal hypouricemia (RHUC) is a disease caused by dysfunction of renal urate reabsorption transporters; however, diagnostic guidance and guidelines for RHUC have been lacking, partly due to the low evidence level of studies on RHUC. This review describes a world-first clinical practice guideline (CPG) and its first version in English for this condition. It was developed following the "MINDS Manual for Guideline Development" methodology, which prioritizes evidence-based medicine. It was published in Japanese in 2017 and later translated into English. The primary goal of this CPG is to clarify the criteria for diagnosing RHUC; another aim is to work towards a consensus on clinical decision-making. One of the CPG's unique points is that it contains textbook descriptions at the expert consensus level, in addition to two clinical questions and recommendations derived from a systematic review of the literature. The guidance shown in this CPG makes it easy to diagnose RHUC from simple blood and urine tests. This CPG contains almost all of the clinical foci of RHUC: epidemiology, pathophysiology, diagnostic guidance, clinical examinations, differential diagnosis, and complications, including exercise-induced acute kidney injury and urolithiasis. A CPG summary as well as a clinical algorithm to assist healthcare providers with a quick reference and notes from an athlete for both physicians and patients are included. We hope that this CPG will help healthcare providers and patients to make clinical decisions, and that it will promote further research on RHUC.


Subject(s)
Practice Guidelines as Topic , Renal Tubular Transport, Inborn Errors , Urinary Calculi , Acute Kidney Injury/etiology , Algorithms , Clinical Decision-Making , Diagnosis, Differential , Evidence-Based Medicine , Exercise , Health Personnel , Humans , Renal Tubular Transport, Inborn Errors/diagnosis , Renal Tubular Transport, Inborn Errors/therapy , Urinary Calculi/diagnosis , Urinary Calculi/therapy , Urolithiasis/etiology
7.
Regen Ther ; 9: 79-88, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30525078

ABSTRACT

INTRODUCTION: Cell sheets using myoblasts have been developed for the treatment of heart failure after myocardial infarction (MI) bridging to heart transplantation. Stem cells are supposed to be better than myoblasts as a source of cells, since they possess a potential to proliferate and differentiate into cardiomyocytes, and also have capacity to secrete angiogenic factors. Adipose-derived stem cells (ASCs) obtained from fat tissues are expected to be a new cell source for ASC sheet therapies. Administration of angiotensin II receptor blockers (ARBs) is a standard therapy for heart failure after MI. However, it is not known whether ARBs affect the cell sheet therapy. This study aimed to examine ameliorating effects of ASC sheets on heart failure and remodeling after MI, and how pretreatment with ARBs prior to the creation of MI and ASC sheet transplantation modifies the effects of ASC sheets. METHODS: ASCs were isolated from fat tissues of wild-type rats, and ASC sheets were engineered on temperature-responsive dishes. In in vitro studies using cultured cells, mRNA levels of vascular endothelial growth factor (VEGF) in ASCs were determined by RT-PCR in the presence of angiotensin II and/or an ARB, irbesartan, under normoxia and hypoxia; mRNA and protein levels of angiotensin II receptor type 1a (AT1aR), type 1b (AT1bR) and type 2 (AT2R) were also determined by RT-PCR and western blotting. In in vivo studies using a rat MI model, effects of transplanted ASC sheets and/or irbesartan on cardiac functions and remodeling after MI were evaluated by echocardiography, histological analysis and molecular biological techniques. RESULTS: In the in vitro studies, ASCs expressed higher levels of VEGF mRNA under hypoxia. They also expressed mRNA and protein of AT1aR but not AT1bR or AT2R. Under normoxia, angiotensin II increased the level of VEGF mRNA in ASCs, which was abolished by irbesartan. Under hypoxia, irbesartan reduced the level of VEGF mRNA in ASCs regardless of whether angiotensin II was present or not. In the in vivo studies, ASC sheets improved cardiac functions after MI, leading to decreased interstitial fibrosis and increased capillary density in border zones. These effects of ASC sheets were abolished by oral administration of irbesartan before MI and their transplantation. CONCLUSIONS: ASC sheets ameliorated cardiac dysfunctions and remodeling after MI via increasing VEGF expression, which was abolished by pretreatment with irbesartan before the creation of MI and transplantation.

8.
Circ J ; 82(4): 1101-1111, 2018 03 23.
Article in English | MEDLINE | ID: mdl-29491325

ABSTRACT

BACKGROUND: Ischemia/reperfusion (I/R) injury triggers cardiac dysfunctions via creating reactive oxygen species (ROS). Because xanthine oxidase (XO) is one of the major enzymes that generate ROS, inhibition of XO is expected to suppress ROS-induced I/R injury. However, it remains unclear whether XO inhibition really yields cardioprotection during I/R. The protective effects of the XO inhibitors, topiroxostat and allopurinol, on cardiac I/R injury were evaluated.Methods and Results:Using isolated rat hearts, ventricular functions, occurrence of arrhythmias, XO activities and thiobarbituric acid reactive substances (TBARS) productions and myocardial levels of adenine nucleotides before and after I/R, and cardiomyocyte death markers during reperfusion, were evaluated. Topiroxostat prevented left ventricular dysfunctions and facilitated recovery from arrhythmias during I/R. Allopurinol and the antioxidant, N-acetylcysteine (NAC), exhibited similar effects at higher concentrations. Topiroxostat inhibited myocardial XO activities and TBARS productions after I/R. I/R decreased myocardial levels of ATP, ADP and AMP, but increased that of xanthine. While topiroxostat, allopurinol or NAC did not change myocardial levels of ATP, ADP or AMP after I/R, all of the agents decreased the level of xanthine. They also decreased releases of CPK and LDH during reperfusion. CONCLUSIONS: Topiroxostat showed protective effects against I/R injury with higher potency than allopurinol or NAC. It dramatically inhibited XO activity and TBARS production, suggesting suppression of ROS generation.


Subject(s)
Myocardial Reperfusion Injury/drug therapy , Nitriles/therapeutic use , Pyridines/therapeutic use , Allopurinol/pharmacology , Allopurinol/therapeutic use , Animals , Arrhythmias, Cardiac/drug therapy , Nitriles/pharmacology , Protective Agents/pharmacology , Protective Agents/therapeutic use , Pyridines/pharmacology , Rats , Reactive Oxygen Species/metabolism , Thiobarbituric Acid Reactive Substances/metabolism , Ventricular Dysfunction, Left/prevention & control , Xanthine Dehydrogenase/antagonists & inhibitors
9.
PLoS One ; 12(4): e0176055, 2017.
Article in English | MEDLINE | ID: mdl-28448596

ABSTRACT

BACKGROUNDS: Hypouricemia was reported as a risk factor for exercise-induced acute renal injury (EIAKI) and urinary stones. However, the prevalence of kidney diseases among hypouricemic subjects has not been evaluated. This study was conducted to clarify the prevalence of hypouricemia and the association of hypouricemia with kidney diseases by using a large-scale Japanese population data. METHODS: This study is a retrospective cross-sectional study at the Center for Preventive Medicine, St. Luke's International Hospital, Tokyo, Japan, and Sanin Rousai Hospital, Yonago, Japan. We analyzed the medical records of 90,143 Japanese subjects at the center in St. Luke's International Hospital, Tokyo, and 4,837 subjects in Sanin Rousai Hospital, Yonago, who underwent annual regular health check-up between January 2004 and June 2010. We defined hypouricemia as serum uric acid level of ≤2.0 mg/dL. We checked the medical history of all the study subjects and compared the rates of complications including urinary stones and kidney diseases among those with or without hypouricemia. RESULTS: The prevalence of hypouricemia was 0.19% in St. Luke's International Hospital, Tokyo, and 0.58% in Sanin Rousai Hospital, Yonago. The prevalence of hypouricemia in women was larger than that in men both in Tokyo (0.31% vs 0.068%, p<0.001) and in Yonago (1.237% vs 0.318%, p<0.001). Among 172 hypouricemic subjects (30 men), the rates of previous urinary stones and kidney diseases (including nephritis/nephrosis) were 1.2% (3.3% men, 0.7% women) and 2.3% (10% men, 0.7% women), respectively. Hypouricemic men had a 9-fold higher rate of previously having kidney diseases compared to non-hypouricemic men (p<0.001). However, the rates of other diseases including urinary stones were not significantly different between the two groups. CONCLUSIONS: Hypouricemia was associated with a history of kidney disease especially in men.


Subject(s)
Kidney Diseases/complications , Renal Tubular Transport, Inborn Errors/epidemiology , Urinary Calculi/epidemiology , Adult , Cross-Sectional Studies , Female , Hospitals , Humans , Japan/epidemiology , Male , Middle Aged , Prevalence , Renal Tubular Transport, Inborn Errors/etiology , Retrospective Studies , Sex Factors , Uric Acid/blood , Urinary Calculi/etiology
10.
Circ J ; 79(5): 1125-32, 2015.
Article in English | MEDLINE | ID: mdl-25739858

ABSTRACT

BACKGROUND: Uric acid (UA) serves as an antioxidant in vascular endothelial cells. UA transporter 1 (URAT1) encoded by SLC22A12 is expressed in the kidney and vessels and its loss of function causes hypouricemia. The purpose of this study was to examine whether there is any endothelial dysfunction in patients with hypouricemia. METHODS AND RESULTS: Twenty-six patients with hypouricemia (<2.5 mg/dl) and 13 healthy control subjects were enrolled. Endothelial function was evaluated using flow-mediated dilation (FMD). mRNA of UA transporters expressed in cultured human umbilical endothelial cells (HUVEC) was detected on RT-PCR. There was a positive correlation between FMD and serum UA in the hypouricemia group. URAT1 loss-of-function mutations were found in the genome of 21 of 26 patients with hypouricemia, and not in the other 5. In the hypouricemia groups, serum UA in homozygous and compound heterozygous patients was significantly lower than in other groups, suggesting that severity of URAT1 dysfunction may influence the severity of hypouricemia. Thirteen of 16 hypouricemia subjects with homozygous and compound heterozygote mutations had SUA <0.8 mg/dl and their FMD was lower than in other groups. HUVEC do not express mRNA of URAT1, suggesting the null role of URAT1 in endothelial function. CONCLUSIONS: Depletion of UA due to SLC22A12/URAT1 loss-of-function mutations causes endothelial dysfunction in hypouricemia patients.


Subject(s)
Endothelium, Vascular , Heterozygote , Human Umbilical Vein Endothelial Cells/metabolism , Organic Anion Transporters , Organic Cation Transport Proteins , Renal Tubular Transport, Inborn Errors , Uric Acid/blood , Urinary Calculi , Adult , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Female , Human Umbilical Vein Endothelial Cells/pathology , Humans , Male , Middle Aged , Mutation , Organic Anion Transporters/genetics , Organic Anion Transporters/metabolism , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism , Renal Tubular Transport, Inborn Errors/blood , Renal Tubular Transport, Inborn Errors/genetics , Renal Tubular Transport, Inborn Errors/physiopathology , Urinary Calculi/blood , Urinary Calculi/genetics , Urinary Calculi/physiopathology , Vasodilation
11.
Biomed Res ; 36(1): 11-9, 2015.
Article in English | MEDLINE | ID: mdl-25749147

ABSTRACT

Adipose stem cells (ASCs) are a source of regenerative cells available for autologous transplantation to hearts. We compared protective actions of ASC sheets on rat myocardial infarction (MI) in comparison with those of skeletal myoblast cell sheets. Their effects on infarcted hearts were evaluated by biological, histochemical as well as physiological analyses. ASC sheets secreted higher concentrations of angiogenic factors (HGF, VEGF, and bFGF; P < 0.05) under normoxic and hypoxic conditions than those of myoblast cell sheets, associated with reduction of cell apoptosis (P < 0.05). Like myoblast cell sheets, ASC sheets improved cardiac function (P < 0.05) and decreased the plasma level of ANP (P < 0.05) in MI hearts. ASC sheets restored cardiac remodeling characterized by fibrosis, cardiac hypertrophy and impaired angiogenesis (P < 0.05), which was associated with increases in angiogenic factors (P < 0.05). In isolated perfused rat hearts, ASC sheets improved both systolic and diastolic functions, which was comparable to cardiac functions of myoblast cell sheets, while both cell sheets failed to restore cardiac contractile response to either isoproterenol, pimobendan or dibutyryl cAMP. These results indicated that ASC sheets improved cardiac function and remodeling of MI hearts mediated by their paracrine action and this improvement was comparable to those by myoblast cell sheets.


Subject(s)
Adipocytes/cytology , Cardiomegaly/therapy , Myocardial Contraction/drug effects , Myocardial Infarction/therapy , Stem Cells/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Atrial Natriuretic Factor/metabolism , Atrial Remodeling/drug effects , Bucladesine/pharmacology , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cell Differentiation , Diastole/drug effects , Fibroblast Growth Factors/metabolism , Hepatocyte Growth Factor/metabolism , Isoproterenol/pharmacology , Male , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/drug effects , Myoblasts, Skeletal/metabolism , Myoblasts, Skeletal/transplantation , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/metabolism , Myocardium/pathology , Organ Culture Techniques , Paracrine Communication/drug effects , Pyridazines/pharmacology , Rats , Rats, Inbred Lew , Stem Cell Transplantation , Stem Cells/drug effects , Stem Cells/metabolism , Systole/drug effects , Vascular Endothelial Growth Factor A/metabolism
12.
Int J Cardiol ; 171(3): 398-403, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24405838

ABSTRACT

BACKGROUND/OBJECTIVES: Insulin resistance plays an important role in the pathophysiology in chronic heart failure (CHF). Diuretics generally have harmful effects on glucose metabolism, however, the effect of mineral corticoid receptor blockers on insulin resistance in CHF is unclear. This study aimed to evaluate the effects of the aldosterone blocker spironolactone, in comparison with furosemide, on insulin resistance in CHF patients. METHODS: The effect of spironolactone (25mg/day) and furosemide (20mg/day) on IR for 16 weeks each was analyzed in 16 CHF patients using a double-blind, placebo-controlled, randomized cross-over study design. RESULTS: Plasma BNP and left ventricular ejection fraction were improved with both treatments (furosemide: p=0.02 and p=0.009, respectively, spironolactone: p=0.03 and p=0.007, respectively). Fasting plasma glucose was not changed; however, plasma insulin levels decreased and insulin sensitivity (by homeostasis model assessment: HOMA-IR) improved with spironolactone as compared to furosemide (p<0.0005). TNF-α, IL-6 and MCP-1 decreased with spironolactone (p=0.002, p=0.02 and p=0.02 vs. baseline, respectively), but not with furosemide. Matrix metalloproteinase (MMP)-2 and MMP-9 were decreased with spironolactone (p=0.003 and p=0.04 vs. baseline, respectively), but not furosemide. Changes in TNF-α, IL-6 and MCP-1 levels after spironolactone treatment were significantly correlated with changes in HOMA-IR (r=0.61, r=0.55 and r=0.65, respectively; p=0.01, p=0.03 and p=0.01, respectively). Furthermore, changes in MMP-2 and MMP-9 levels were significantly correlated with changes in HOMA-IR (r=0.58 and r=0.58, respectively; p=0.02 and p=0.02, respectively). CONCLUSIONS: Spironolactone, not furosemide, improved insulin resistance in CHF patients probably by the inhibition of inflammatory cytokines and MMPs.


Subject(s)
Furosemide/therapeutic use , Heart Failure/blood , Heart Failure/drug therapy , Insulin Resistance/physiology , Spironolactone/therapeutic use , Aged , Blood Glucose/drug effects , Blood Glucose/metabolism , Chronic Disease , Cross-Over Studies , Double-Blind Method , Female , Furosemide/pharmacology , Humans , Male , Middle Aged , Spironolactone/pharmacology
13.
Clin Exp Hypertens ; 36(7): 447-53, 2014.
Article in English | MEDLINE | ID: mdl-24433018

ABSTRACT

PURPOSE: To examine effects of a long-acting calcium channel blocker (CCB) azelnidipine on uric acid metabolism in hypertensive patients. METHODS: Azelnidipine was administered to 72 patients at a daily dose of 8 mg or 16 mg. In 22 cases out of the 72 patients, a different CCB was switched to azelnidipine. Blood pressure was measured and biochemical parameters of blood and urine were evaluated before and 2-3 months after the administration. RESULTS: Azelnidipine significantly decreased both systolic and diastolic blood pressure and the heart rate. It decreased both serum urate levels and the urinary uric acid to creatinine ratio (Uur/Ucr), but did not affect the uric acid clearance to creatinine clearance ratio (Cur/Ccr). Azelnidipine decreased both Uur/Ucr and Cur/Ccr in patients with Uur/Ucr ≥ 0.5 or ≥ 0.34, although it did not change these clearance parameters in patients with Uur/Ucr <0.5 or <0.34. Azelnidipine decreased the serum urate levels and Uur/Ucr in hyperuricemic patients with uric acid levels ≥ 7.0 mg/dL in males and ≥ 6.0 mg/dL in females. It did not change these parameters in normouricemic patients with serum urate levels <7.0 mg/dL in males and <6.0 mg/dL in females. Azelnidipine decreased Uur/Ucr and Cur/Ccr in hyperuricemic patients with normal or over excretion of uric acid, although it did not change these clearance parameters in hyperuricemic patients with uric acid hypoexcretion. CONCLUSIONS: Azelnidipine decreased the serum urate acid levels and Uur/Ucr, and this response was most prominent in hyperuricemic patients or patients with normal and over excretion of uric acid.


Subject(s)
Azetidinecarboxylic Acid/analogs & derivatives , Calcium Channel Blockers/therapeutic use , Dihydropyridines/therapeutic use , Hypertension/drug therapy , Hypertension/metabolism , Hyperuricemia/drug therapy , Uric Acid/metabolism , Aged , Aged, 80 and over , Azetidinecarboxylic Acid/therapeutic use , Blood Pressure/drug effects , Creatinine/metabolism , Essential Hypertension , Female , Humans , Hypertension/complications , Hyperuricemia/complications , Hyperuricemia/metabolism , Male , Uric Acid/blood , Uric Acid/urine
14.
Clin Exp Hypertens ; 36(4): 251-7, 2014.
Article in English | MEDLINE | ID: mdl-23865441

ABSTRACT

PURPOSE: Long-term effects of a low-dose hydrochlorothiazide (HCTZ) with losartan (LOS) on uric acid (UA) metabolism as well as glucose metabolism have been studied in hypertensive patients in comparison with those of a low-dose HCTZ with telmisartan (TEL). METHOD: Fifty-nine hypertensive patients were allocated to a combination therapy with either losartan (50 mg/day)/HCTZ (12.5 mg/day) (LOS + HCTZ group: n = 37) or telmisartan (40 mg/day)/HCTZ (12.5 mg/day) (TEL + HCTZ group: n = 22), respectively. Before and 1 year after the treatment, blood pressure and biochemical parameters of blood and urine were evaluated. RESULTS: Both systolic and diastolic blood pressures significantly decreased in two groups, without any statistical differences among them. LOS + HCTZ caused no changes in the serum UA level or the ratio of UA clearance to creatinine clearance (CUA/Ccr), whereas TEL + HCTZ significantly increased the serum UA level and reduced CUA/Ccr. LOS + HCTZ did not influence CUA/Ccr in patients with their serum UA below 5.4 mg/dl, while LOS + HCTZ significantly increased CUA/Ccr in patients with their serum UA above 5.5 mg/dl. TEL + HCTZ significantly reduced CUA/Ccr in patients with their serum UA below and above 5.4 mg/dl to increase serum UA level significantly. Neither combination therapies caused any changes in fasting plasma glucose, HbA1c and HOMA-R. In patients with their serum UA level above 5.4 mg/dl, TEL + HCTZ increased HOMA-R, whereas LOS + HCTZ did not. CONCLUSIONS: LOS + HCTZ did not influence UA metabolism as well as glucose metabolism, likely because of inhibitory action of losartan on URAT1, although TEL + HCTZ were accompanied with impairment of the UA metabolism and glucose metabolism.


Subject(s)
Benzimidazoles/administration & dosage , Benzoates/administration & dosage , Blood Pressure/drug effects , Hydrochlorothiazide/administration & dosage , Losartan/administration & dosage , Adult , Aged , Dose-Response Relationship, Drug , Drug Combinations , Female , Follow-Up Studies , Humans , Hypertension/blood , Hypertension/drug therapy , Hypertension/physiopathology , Male , Middle Aged , Prospective Studies , Telmisartan , Treatment Outcome , Uric Acid/blood
15.
Biochem Biophys Res Commun ; 434(1): 131-6, 2013 Apr 26.
Article in English | MEDLINE | ID: mdl-23542464

ABSTRACT

Remote reperfusion lung injury following skeletal muscle ischemia and reperfusion accounts for high morbidity and mortality. AMP deaminase (AMPD), a key enzyme for nucleotide cycle, has been implicated in the regulation of this phenomenon. However, the function of Ampd2 and Ampd3 subtype has not been elucidated in remote reperfusion rodent lung injury. We utilized AMPD3 and AMPD2-deficient mice. The two types of AMPD-deficient mice and wild-type (WT) littermates were subjected to ischemia-reperfusion injury. After 3h bilateral hind-limb ischemia and reperfusion, AMPD3 mRNA, AMPD activity and inosine monophosphate (IMP) increased significantly in WT and AMPD2-deficient mice lungs, while they did not show significant alterations in AMPD3-deficient mice lungs. Genetic inactivation of Ampd3 resulted in markedly accelerated myeloperoxidase (MPO) activity along with exaggerated neutrophils infiltration and hemorrhage in the lungs compared to WT and AMPD2-deficient mice, furthermore, IMP treatment significantly attenuated MPO activity and neutrophils infiltration in WT and the two types of AMPD-deficient mice lungs after 3h reperfusion. These findings demonstrate for the first time in AMP-deficient mice models that AMPD3 plays a critical role in remote reperfusion lung injury via generation of IMP and validate the potential to use IMP into the clinical arena to attenuate remote ischemia-reperfusion lung injury.


Subject(s)
AMP Deaminase/physiology , Lung Injury/enzymology , Reperfusion Injury/enzymology , AMP Deaminase/deficiency , AMP Deaminase/genetics , Animals , Disease Models, Animal , Inosine Monophosphate/administration & dosage , Inosine Monophosphate/biosynthesis , Lung Injury/genetics , Lung Injury/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Reperfusion Injury/genetics , Reperfusion Injury/pathology
16.
Clin Exp Hypertens ; 34(7): 470-3, 2012.
Article in English | MEDLINE | ID: mdl-22681507

ABSTRACT

The effects of cilnidipine on the serum uric acid level and urinary NO excretion in hypertensive patients were investigated. Blood and urine samples of 16 hypertensive outpatients were collected before and 2 months after cilnidipine therapy (10 mg). The serum uric acid level decreased significantly after cilnidipine treatment, while the uric acid-creatinine clearance ratio was unaffected. The cilnidipine medication produced a significant increase in urinary NO excretion, although amlodipine did not change it significantly. Therefore, cilnidipine has a profound antihypertensive effect and may reduce the serum uric acid level and increase NO production in the kidney.


Subject(s)
Calcium Channel Blockers/therapeutic use , Dihydropyridines/therapeutic use , Hypertension/drug therapy , Nitric Oxide/urine , Uric Acid/blood , Aged , Aged, 80 and over , Amlodipine/therapeutic use , Female , Humans , Hypertension/blood , Hypertension/urine , Kidney/physiopathology , Male , Middle Aged , Treatment Outcome
17.
Biomed Res ; 32(5): 303-12, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22033299

ABSTRACT

It is unknown whether salicylate enhances the action of antiarrhythmic agents on human Na+ channels with state dependency and tissue specificity. We therefore investigated effects of salicylate on quinidine-induced block of human cardiac and skeletal muscle Na+ channels. Human cardiac wild-type (hH1), LQT3-related mutant (ΔKPQ), and skeletal muscle (hSkM1) Na+ channel α subunits were expressed in COS7 cells. Effects of salicylate on quinidine-induced tonic and use-dependent block of Na+ channel currents were examined by the whole-cell patch-clamp technique. Salicylate enhanced the quinidine-induced tonic and use-dependent block of both hH1 and hSkM1 currents at a holding potential (HP) of -100 mV but not at -140 mV. Salicylate decreased the IC50 value for the quinidine-induced tonic block of hH1 at an HP of -100 mV, and produced a negative shift in the steady-state inactivation curve of hH1 in the presence of quinidine. According to the modulated receptor theory, it is probable that salicylate decreases the dissociation constant for quinidine binding to inactivated-state channels. Furthermore, salicylate significantly enhanced the quinidine-induced tonic and use-dependent block of the peak and steady-state ΔKPQ channel currents. The results suggest that salicylate enhances quinidine-induced block of Na+ channels via increasing the affinity of quinidine to inactivated state channels.


Subject(s)
Quinidine/pharmacology , Salicylates/pharmacology , Sodium Channel Blockers/pharmacology , Sodium Channels/genetics , Sodium Channels/metabolism , Animals , COS Cells , Chlorocebus aethiops , Heart/drug effects , Humans , Membrane Potentials/drug effects , Membrane Potentials/genetics , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Mutation , Myocardium/metabolism , NAV1.5 Voltage-Gated Sodium Channel , Protein Binding , Quinidine/metabolism
19.
Arterioscler Thromb Vasc Biol ; 31(5): 1133-8, 2011 May.
Article in English | MEDLINE | ID: mdl-21350197

ABSTRACT

OBJECTIVE: Nitric oxide (NO) is an important modulator of cardiovascular function. In this study, we examined whether cytosolic phospholipase A2α (cPLA2α), an initial enzyme in the arachidonic acid pathway, is involved in blood pressure (BP) elevation in a murine model of chronic NO inhibition. METHODS AND RESULTS: cPLA2α gene-deficient mice (cPLA2α-/-) and wild-type mice (WT) were administered the NO synthesis inhibitor Nω-nitro-L-arginine methyl ester (L-NAME) for 4 weeks. Before treatment, BP was comparable in both groups; it increased significantly in the WT but not in the cPLA2α-/- after treatment. Bone marrow transplantation experiments showed that cPLA2α in blood cells and plasma eicosanoid concentrations were not involved in BP elevation by L-NAME treatment. Activation of cPLA2α and subsequent production of eicosanoids in the aortic endothelium but not in aortic smooth muscle cell, heart, or kidney was observed after L-NAME treatment. Aortic ring assays revealed that endothelial function was comparable in both groups of mice before treatment. L-NAME treatment disturbed endothelial function in WT but not in cPLA2α-/-. CONCLUSIONS: These results suggest that endothelial cPLA2α may play a principal role in L-NAME-induced hypertension and may be a target molecule for maintaining endothelial function under NO inhibition.


Subject(s)
Aorta, Thoracic/enzymology , Blood Pressure , Endothelium, Vascular/enzymology , Group IV Phospholipases A2/metabolism , Hypertension/enzymology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide/metabolism , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/physiopathology , Bone Marrow Transplantation , Disease Models, Animal , Dose-Response Relationship, Drug , Eicosanoids/blood , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Enzyme Inhibitors , Group IV Phospholipases A2/deficiency , Group IV Phospholipases A2/genetics , Hypertension/chemically induced , Hypertension/genetics , Hypertension/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NG-Nitroarginine Methyl Ester , Nitric Oxide Synthase/metabolism , Phosphorylation , Renin/blood , Time Factors , Tissue Culture Techniques , Vasoconstriction , Vasoconstrictor Agents/pharmacology , Vasodilation , Vasodilator Agents/pharmacology
20.
Circ J ; 74(11): 2346-52, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20827028

ABSTRACT

BACKGROUND: Angiotensin II and insulin resistance (IR) have clinical implications in the pathophysiology of chronic heart failure (CHF). However, it is still unclear whether the combination of an angiotensin-receptor blocker and angiotensin-converting enzyme inhibitor (ACEI) improves IR in CHF patients who do not receive ß-blockers. Thus, the aim of the present study was to evaluate the effects of losartan on glucose metabolism and inflammatory cytokines in CHF patients treated with ACEI but not ß-blockers. METHODS AND RESULTS: The effect of losartan treatment for 16 weeks on IR was analyzed in 16 CHF patients in a randomized crossover trial. Insulin level and homeostasis model IR index (HOMA-IR) decreased significantly (P<0.05), but fasting plasma glucose did not change significantly. Serum tumor necrosis factor (TNF)-α, interleukin (IL)-6, and monocyte chemoattractant protein (MCP)-1 levels were significantly decreased with losartan (P<0.05). Furthermore, the changes in IL-6 and MCP-1 levels were significantly correlated with the reduction in HOMA-IR (P<0.05), but the change in TNF-α levels was not significantly correlated. CONCLUSIONS: The addition of losartan to ACEI therapy improved IR and decreased inflammatory cytokines in CHF patients who did not receive ß-blockers.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/therapeutic use , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Heart Failure/drug therapy , Insulin Resistance , Losartan/therapeutic use , Adrenergic beta-Antagonists/therapeutic use , Aged , Biomarkers/blood , Blood Glucose/metabolism , Chemokine CCL2/blood , Chronic Disease , Cross-Over Studies , Drug Therapy, Combination , Enalapril/therapeutic use , Female , Heart Failure/blood , Heart Failure/physiopathology , Humans , Imidazolidines/therapeutic use , Inflammation Mediators/blood , Insulin/blood , Interleukin-6/blood , Japan , Lisinopril/therapeutic use , Male , Middle Aged , Time Factors , Treatment Outcome , Tumor Necrosis Factor-alpha/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...