Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
3.
Biomed Pharmacother ; 153: 113515, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36068956

ABSTRACT

BACKGROUND: Renal ischemia and reperfusion injury (IRI) is the main cause of acute kidney injury (AKI). AKI induces the development of cardiac hypertrophy (CH) during cardiorenal syndrome (CRS), and cardiomyocyte calcium mishandling though systemic inflammation after 8 days of renal IRI. Klotho has recently been described as an anti-inflammatory component. Given this, Klotho treatment could prevent or attenuate the inflammation, thereby also preventing electrical cardiac outcomes incurred by CRS. The aim of this study was to investigate the therapeutic role of Klotho in CRS after unilateral renal IRI through its anti-inflammatory action. METHODS: We examined renal tissue structure and function, intracellular Ca2+ dynamics in adult ventricular cardiomyocytes and serum cytokine levels from C57BL/6 mice that suffered unilateral renal IRI by occluding the left pedicle for 60 min and reperfusion for 8 days. The animals were treated with recombinant Klotho protein starting from the day of the surgery, then daily for 8 days. RESULTS: After Klotho treatment for 8 days, the left renal tissue remained damaged, however the renal function was restored due to the right kidney tissue preservation. In parallel, Klotho also prevented an increase in serum interleukin (IL-) 6, IL-1ß, and tumor necrosis factor alpha (TNF-α) levels. CH and low cell contraction were also prevented, as well as a decrease in systolic Ca2+ transients and sarco/endoplasmic reticulum Ca2+-ATPase (SERCA2a) activity measured as Ca2+ transient decay, an increase in spontaneous Ca2+ release and the incidence of pro-arrhythmic events. CONCLUSIONS: The Klotho treatment showed promise, playing an important role in the pathophysiology of CRS. We were unable to observe a total renoprotective role of the compound in the model; in turn, a cardioprotective role of Klotho was demonstrated through the prevention of hypertrophy and normalization of the Ca2+ cycle dysfunction of cardiomyocytes. We propose that Klotho acts in the cardiorenal syndrome by systematically preventing inflammation and increased FGF23, alleviating cardiac outcomes.


Subject(s)
Acute Kidney Injury , Cardio-Renal Syndrome , Reperfusion Injury , Acute Kidney Injury/etiology , Acute Kidney Injury/metabolism , Acute Kidney Injury/prevention & control , Animals , Cardio-Renal Syndrome/drug therapy , Cardio-Renal Syndrome/prevention & control , Inflammation/metabolism , Ischemia/metabolism , Kidney , Mice , Mice, Inbred C57BL , Reperfusion , Reperfusion Injury/complications , Reperfusion Injury/drug therapy , Reperfusion Injury/prevention & control
4.
G Ital Cardiol (Rome) ; 23(9): 716-727, 2022 Sep.
Article in Italian | MEDLINE | ID: mdl-36039723

ABSTRACT

Chronic kidney disease (CKD) and cardiovascular (CV) disease are highly prevalent conditions in the general population and are strictly connected to each other with a bidirectional interaction. In patients affected by CKD, the leading cause of morbidity and mortality is represented by CV disease, since CKD promotes the atherosclerotic process increasing inflammation, and modifying lipid and bone mineral metabolism. On the other side, a strict relationship exists between CKD and CV risk factors, which are prevalent in nephropathic patients and impose a stringent assessment of the risk of CV events in this population together with an optimized pharmacological approach, complicated by the coexistence of the two pathological conditions. The first part of this consensus document focuses on the mechanisms of cardio-renal damage and on the impact, as well as the management, of the main CV risk factors in the context of CKD.


Subject(s)
Cardio-Renal Syndrome , Cardiology , Cardiovascular Diseases , Nephrology , Renal Insufficiency, Chronic , Cardio-Renal Syndrome/etiology , Cardio-Renal Syndrome/prevention & control , Cardiovascular Diseases/etiology , Cardiovascular Diseases/prevention & control , Consensus , Heart Disease Risk Factors , Humans , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/epidemiology , Risk Factors
6.
Cardiovasc Diabetol ; 20(1): 189, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34526024

ABSTRACT

BACKGROUND: A meta-analysis is presented of cardiovascular outcome trials (CVOTs) comparing glucagon-like peptide-1 receptor agonists (GLP-1RA) versus placebo on cardiorenal outcomes in patients with type 2 diabetes mellitus (T2DM). METHODS: We did an electronic search up to June 30, 2021, for eligible trials. We did a meta-analysis of available trial data using a random-effects model to calculate overall hazard ratios (HRs) and 95% CI (confidence intervals). We included data from 8 CVOTs and 60,080 patients (72.4% with established cardiovascular disease). RESULTS: GLP-1RA reduced major cardiovascular events (MACE) by 14% (HR = 0.86, 95% CI 0.79-0.94, P = 0.006) with a non-significant heterogeneity between subgroups of patients with and without cardiovascular disease (P = 0.127). GLP-1RA also reduced the risk of cardiovascular death by 13% (P = 0.016), nonfatal stroke by 16% (P = 0.007), hospitalization for heart failure by 10% (P = 0.023), all-cause mortality by 12% (P = 0.012), and the broad composite kidney outcome by 17% (P = 0.012), which was driven by a reduction in macroalbuminuria only (HR = 0.74, 0.67-0.82, P < 0.001). CONCLUSIONS: GLP-1RA have moderate benefits on MACE, and also reduce hospitalization for heart failure and all-cause mortality; they also have robust benefits on reducing the incidence of macroalbuminuria.


Subject(s)
Cardio-Renal Syndrome/prevention & control , Cardiovascular Diseases/prevention & control , Diabetes Mellitus, Type 2/drug therapy , Glucagon-Like Peptide-1 Receptor/agonists , Hypoglycemic Agents/therapeutic use , Incretins/therapeutic use , Kidney Diseases/prevention & control , Aged , Cardio-Renal Syndrome/diagnosis , Cardio-Renal Syndrome/mortality , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/mortality , Cause of Death , Clinical Trials as Topic , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/mortality , Female , Hospitalization , Humans , Hypoglycemic Agents/adverse effects , Incidence , Incretins/adverse effects , Kidney Diseases/diagnosis , Kidney Diseases/mortality , Male , Middle Aged , Risk Assessment , Risk Factors , Treatment Outcome
7.
J Am Geriatr Soc ; 69(2): 539-546, 2021 02.
Article in English | MEDLINE | ID: mdl-33104236

ABSTRACT

The cardiovascular and renal systems share an intimate physiological relationship, wherein a perturbance in one system may have an adverse effect on the other. Since the burden of renal disease increases with age, there is a considerable interest in the pathophysiology of kidney disease in the geriatric patient population. This review will explore the physiological dynamics behind the increased susceptibility to kidney disease in this population. A better understanding of these pathophysiological changes may lead to improved prevention and management strategies.


Subject(s)
Aging/physiology , Cardio-Renal Syndrome , Kidney Diseases , Aged , Cardio-Renal Syndrome/physiopathology , Cardio-Renal Syndrome/prevention & control , Heart Disease Risk Factors , Humans , Kidney Diseases/physiopathology , Kidney Diseases/therapy , Patient Care Management
8.
JAMA Netw Open ; 2(8): e199307, 2019 08 02.
Article in English | MEDLINE | ID: mdl-31441935

ABSTRACT

Importance: There are few studies comparing the optimal level of treated blood pressure (BP) between high- and low-risk patients. Objective: To examine whether optimally treated BP is different according to risk status. Design, Setting, and Participants: Population-based cohort study using data from the National Health Information Database in Korea from 2002 to 2015 and 2006 to 2017. A total of 1 402 975 adults aged 40 to 79 years who had no known cardiorenal disease were included. Exposures: Systolic BP treated with antihypertensive medication. Main Outcomes and Measures: The yearly rates of critical cardiorenal events and all-cause death were estimated according to the levels of treated systolic BP and the presence of 5 risk factors (hypertension, diabetes, hyperlipidemia, proteinuria, and smoking). Results: During the study periods, 225 103 of 487 412 participants (54.0% male; median [interquartile range] age, 50 [44-59] years) in the primary cohort and 360 503 of 915 563 participants (50.1% male; median [interquartile range] age, 52 [46-60] years) in the secondary cohort received antihypertensive treatment. In total, 28 411 of 51 292 cardiorenal incidents and 33 102 of 72 500 deaths were noted in ever-treated participants. The absolute increase in cardiorenal and mortality risk associated with inadequately treated BP was greater in participants with multiple risk factors than in those with 1 or 0 risk factors. The hazard ratios for critical cardiorenal events increased as the treated systolic BP increased to more than 130 to 140 mm Hg. The hazard ratio for all-cause mortality for patients with 3 or more risk factors and treated systolic BP within the range of 110 to 119 mm Hg was 1.21 (95% CI, 1.07-1.37); 130 to 139 mm Hg, 1.04 (95% CI, 0.98-1.11); 140 to 149 mm Hg, 1.12 (95% CI, 1.05-1.20); 150 to 159 mm Hg, 1.21 (95% CI, 1.11-1.32); and 160 mm Hg or greater, 1.46 (95% CI, 1.32-1.62) compared with high-risk patients with BP of 120 to 129 mm Hg. For participants with 1 or 0 risk factors and treated systolic BP within the range of 110 to 119 mm Hg, the hazard ratio was 1.14 (95% CI, 1.07-1.22); 130 to 139 mm Hg, 0.97 (95% CI, 0.93-1.02); 140 to 149 mm Hg, 1.00 (95% CI, 0.91-1.09); 150 to 159 mm Hg, 1.06 (95% CI, 0.99-1.14); and 160 mm Hg or greater, 1.26 (95% CI, 1.15-1.37). However, when categorized using cardiovascular risk calculators, there was no consistent trend in mortality thresholds of BP across the risk score categories. Conclusions and Relevance: These results suggest that intensive BP control is appropriate for reducing all-cause mortality in addition to cardiorenal risk in higher- rather than lower-risk patients. However, caution may be required when determining BP targets using current risk calculators.


Subject(s)
Antihypertensive Agents/therapeutic use , Blood Pressure/drug effects , Cardio-Renal Syndrome/prevention & control , Hypertension/drug therapy , Adult , Aged , Antihypertensive Agents/pharmacology , Blood Pressure Determination , Cardio-Renal Syndrome/etiology , Cardio-Renal Syndrome/mortality , Databases, Factual , Female , Follow-Up Studies , Humans , Hypertension/complications , Hypertension/diagnosis , Hypertension/mortality , Male , Middle Aged , Risk Assessment , Risk Factors , Treatment Outcome
10.
J Diabetes Investig ; 10(4): 933-946, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30663266

ABSTRACT

AIMS/INTRODUCTION: Type 2 diabetes mellitus is a risk factor of acute kidney injury after myocardial infarction (MI), a form of cardiorenal syndrome. Recent clinical trials have shown that a sodium-glucose cotransporter 2 (SGLT2) inhibitor improved both cardiac and renal outcomes in patients with type 2 diabetes mellitus, but effects of an SGLT2 inhibitor on cardiorenal syndrome remain unclear. MATERIALS AND METHODS: Type 2 diabetes mellitus (Otsuka Long-Evans Tokushima Fatty rats [OLETF]) and control (Long-Evans Tokushima Otsuka rats [LETO]) were treated with canagliflozin, an SGLT2 inhibitor, for 2 weeks. Renal tissues were analyzed at 12 h after MI with or without preoperative fasting. RESULTS: Canagliflozin reduced blood glucose levels in OLETF, and blood ß-hydroxybutyrate levels were increased by canagliflozin only with fasting. MI increased neutrophil gelatinase-associated lipocalin and kidney injury molecule-1 protein levels in the kidney by 3.2- and 1.6-fold, respectively, in OLETF, but not in LETO. The renal messenger ribonucleic acid level of Toll-like receptor 4 was higher in OLETF than in LETO after MI, whereas messenger ribonucleic acid levels of cytokines/chemokines were not significantly different. Levels of lipid peroxides, nicotinamide adenine dinucleotide phosphate oxidase (NOX)2 and NOX4 proteins after MI were significantly higher in OLETF than in LETO. Canagliflozin with pre-MI fasting suppressed MI-induced renal expression of neutrophil gelatinase-associated lipocalin and kidney injury molecule-1 in OLETF, together with reductions in lipid peroxides and NOX proteins in the kidney. Blood ß-hydroxybutyrate levels before MI were inversely correlated with neutrophil gelatinase-associated lipocalin protein levels in OLETF. Pre-incubation with ß-hydroxybutyrate attenuated angiotensin II-induced upregulation of NOX4 in NRK-52E cells. CONCLUSIONS: The findings suggest that SGLT2 inhibitor treatment with a fasting period protects kidneys from MI-induced cardiorenal syndrome, possibly by ß-hydroxybutyrate-mediated reduction of NOXs and oxidative stress, in type 2 diabetic rats.


Subject(s)
Acute Kidney Injury/prevention & control , Canagliflozin/pharmacology , Cardio-Renal Syndrome/prevention & control , Diabetes Mellitus, Type 2/complications , Myocardial Infarction/prevention & control , Oxidative Stress/drug effects , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Acute Kidney Injury/etiology , Animals , Cardio-Renal Syndrome/etiology , Lipid Peroxidation/drug effects , Male , Myocardial Infarction/etiology , Rats , Rats, Inbred OLETF
11.
Am J Hypertens ; 32(1): 34-44, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30329020

ABSTRACT

BACKGROUND: In uremic animals, vitamin D receptor (VDR) agonists like paricalcitol (Pc) attenuate cardiac hypertrophy, but this effect has not been replicated consistently in humans with chronic kidney disease. Elevated fibroblast growth factor 23 (FGF23) levels cause cardiac hypertrophy with activation of the myocardial calcineurin/nuclear factor of activated T cell (NFAT) axis and may antagonize the cardioprotective effects of VDR agonist therapy. We hypothesized that the effectiveness of Pc may depend on the prevailing circulating levels of FGF23 and could be potentiated by the combined administration of a pan-FGF23 receptor (FGFR) blocker agent (PD173074). METHODS: In rats with 5/6 nephrectomy treated with Pc or PD173074 or both agents concurrently, myocardial mRNA expression of renin-angiotensin system, VDR, FGFR4, and calcineurin/NFAT target genes was determined. In adolescents on hemodialysis, we analyzed sequential echocardiograms, blood pressures and serial FGF23 measurements, and their relations to the cumulative administered dose of parenteral Pc. RESULTS: The ratio of Pc dose/plasma levels of FGF23 correlated inversely (P < 0.005) with the cardiac mass in uremic rats and in hemodialysis patients, independently of hypertension. Despite persistently elevated FGF23 levels and myocardial FGFR4 activation, Pc suppressed upregulated myocardial calcineurin/NFAT target genes, and the effects were amplified by coadministration of PD173074. CONCLUSIONS: The beneficial effects of Pc on uremic cardiac hypertrophy are counterbalanced by the increased FGF23 levels. Blockade of FGF23-mediated signaling increased the Pc-induced suppression of the myocardial calcineurin/NFAT system. Higher doses of Pc should be considered in the treatment of patients with uremic cardiomyopathy.


Subject(s)
Cardio-Renal Syndrome/prevention & control , Cardiomyopathies/prevention & control , Ergocalciferols/pharmacology , Fibroblast Growth Factors/blood , Heart Ventricles/drug effects , Kidney Failure, Chronic/drug therapy , Pyrimidines/pharmacology , Receptor, Fibroblast Growth Factor, Type 4/antagonists & inhibitors , Receptors, Calcitriol/agonists , Adolescent , Animals , Cardio-Renal Syndrome/metabolism , Cardio-Renal Syndrome/physiopathology , Cardiomyopathies/metabolism , Cardiomyopathies/physiopathology , Child , Disease Models, Animal , Drug Therapy, Combination , Female , Fibroblast Growth Factor-23 , Heart Ventricles/metabolism , Heart Ventricles/physiopathology , Humans , Kidney Failure, Chronic/metabolism , Kidney Failure, Chronic/physiopathology , Male , Rats, Sprague-Dawley , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Receptors, Calcitriol/metabolism , Retrospective Studies , Signal Transduction , Uremia/drug therapy , Uremia/metabolism , Uremia/pathology , Uremia/physiopathology , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects
12.
Clin Cardiol ; 41(9): 1259-1267, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30125365

ABSTRACT

Heart failure (HF) and type 2 diabetes mellitus (T2DM) are two global pandemics, affecting over 25 and 420 million people, respectively. The prevalence of comorbid HF and T2DM is rising, and the prognosis remains poor. One central area of overlap of these two disease processes is renal dysfunction, which contributes to poor cardiovascular outcomes and mortality. As such, there is a growing need for antihyperglycemic agents with cardio- and renoprotective effects. Three classes of novel antihyperglycemic agents, sodium-glucose cotransporter-2 (SGLT2) inhibitors, glucagon-like peptide-1 receptor agonists (GLP-1RA), and dipeptidyl peptidase-4 (DPP4) inhibitors have demonstrated varied cardiorenal outcomes in recent cardiovascular outcomes trials. Understanding the differential effects of these agents, together with their proposed mechanisms, is crucial for the development of safe and effective treatment regimens and future pharmacologic targets for HF and T2DM. In this review, we discuss the overlapping pathophysiology of HF and T2DM, summarize outcomes data for the novel antihyperglycemic agents and proposed mechanisms of action, and review how the current evidence informs future management of comorbid HF and T2DM.


Subject(s)
Amino Acid Metabolism, Inborn Errors/drug therapy , Cardio-Renal Syndrome , Diabetes Mellitus, Type 2/epidemiology , Heart Failure , Hypoglycemic Agents/therapeutic use , Triglycerides/blood , Amino Acid Metabolism, Inborn Errors/blood , Amino Acid Metabolism, Inborn Errors/epidemiology , Cardio-Renal Syndrome/blood , Cardio-Renal Syndrome/epidemiology , Cardio-Renal Syndrome/prevention & control , Comorbidity/trends , Diabetes Mellitus, Type 2/blood , Global Health , Heart Failure/blood , Heart Failure/drug therapy , Heart Failure/epidemiology , Humans
13.
Kidney Int ; 93(2): 375-389, 2018 02.
Article in English | MEDLINE | ID: mdl-28760335

ABSTRACT

Cardiovascular events are the leading cause of death in patients with chronic kidney disease (CKD), although the pathological mechanisms are poorly understood. Here we longitudinally characterized left ventricle pathology in a 5/6 nephrectomy rat model of CKD and identify novel molecular mediators. Next-generation sequencing of left ventricle mRNA and microRNA (miRNA) was performed at physiologically distinct points in disease progression, identifying alterations in genes in numerous immune, lipid metabolism, and inflammatory pathways, as well as several miRNAs. MiRNA miR-21-5p was increased in our dataset and has been reported to regulate many identified pathways. Suppression of miR-21-5p protected rats with 5/6 nephrectomy from developing left ventricle hypertrophy and improved left ventricle function. Next-generation mRNA sequencing revealed that miR-21-5p suppression altered gene expression in peroxisome proliferator-activated receptor alpha (PPARα) regulated pathways in the left ventricle. PPARα, a miR-21-5p target, is the primary PPAR isoform in the heart, importantly involved in regulating fatty acid metabolism. Therapeutic delivery of low-dose PPARα agonist (clofibrate) to rats with 5/6 nephrectomy improved cardiac function and prevented left ventricle dilation. Thus, comprehensive characterization of left ventricle molecular changes highlights the involvement of numerous signaling pathways not previously explored in CKD models and identified PPARα as a potential therapeutic target for CKD-related cardiac dysfunction.


Subject(s)
Cardio-Renal Syndrome/metabolism , Heart Ventricles/metabolism , Hypertrophy, Left Ventricular/metabolism , MicroRNAs/metabolism , PPAR alpha/metabolism , Ventricular Dysfunction, Left/metabolism , Ventricular Function, Left , Ventricular Remodeling , Animals , Cardio-Renal Syndrome/genetics , Cardio-Renal Syndrome/pathology , Cardio-Renal Syndrome/prevention & control , Clofibrate/pharmacology , Disease Models, Animal , Fatty Acids/metabolism , Fibrosis , Gene Expression Regulation , Heart Ventricles/drug effects , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/prevention & control , Male , MicroRNAs/genetics , PPAR alpha/agonists , PPAR alpha/genetics , Rats, Sprague-Dawley , Signal Transduction , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/pathology , Ventricular Dysfunction, Left/prevention & control , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects
14.
Rev Port Cardiol ; 36(9): 619-625, 2017 Sep.
Article in English, Portuguese | MEDLINE | ID: mdl-28826934

ABSTRACT

INTRODUCTION: Cardiorenal syndrome (CRS) is common in acute heart failure (AHF), and is associated with dire prognosis. Levosimendan, a positive inotrope that also has diuretic effects, may improve patients' renal profile. Published results are conflicting. OBJECTIVES: We aimed to assess the incidence of CRS in AHF patients according to the inotrope used and to determine its predictors in order to identify patients who could benefit from the most renoprotective inotrope. METHODS: In a retrospective study, 108 consecutive patients with AHF who required inotropes were divided into two groups according to the inotrope used (levosimendan vs. dobutamine). The primary endpoint was CRS incidence. Follow-up for mortality and readmission for AHF was conducted. RESULTS: Seventy-one percent of the study population were treated with levosimendan and the remainder with dobutamine. No differences were found in heart failure etiology or chronic kidney disease. At admission, the dobutamine group had lower blood pressure; there were no differences in estimated glomerular filtration rate or cystatin C levels. The levosimendan group had lower left ventricular ejection fraction. CRS incidence was higher in the dobutamine group, and they more often had incomplete recovery of renal function at discharge. In multivariate analysis, cystatin C levels predicted CRS. The dobutamine group had higher in-hospital mortality, of which CRS and the inotrope used were predictors. CONCLUSIONS: Levosimendan appears to have some renoprotective effect, as it was associated with a lower incidence of CRS and better recovery of renal function at discharge. Identification of patients at increased risk of renal dysfunction by assessing cystatin C may enable more tailored therapy, minimizing the incidence of CRS and its negative impact on outcome in AHF.


Subject(s)
Cardio-Renal Syndrome/etiology , Cardio-Renal Syndrome/prevention & control , Cardiotonic Agents/therapeutic use , Dobutamine/therapeutic use , Heart Failure/complications , Heart Failure/drug therapy , Hydrazones/therapeutic use , Pyridazines/therapeutic use , Acute Disease , Aged , Cardio-Renal Syndrome/epidemiology , Female , Humans , Incidence , Male , Middle Aged , Retrospective Studies , Simendan
15.
J Complement Integr Med ; 14(3)2017 Mar 22.
Article in English | MEDLINE | ID: mdl-28333655

ABSTRACT

Background Cardiac toxicity is one of the life-threatening complications of cancer therapy. Cyclophosphamide (CYP) is an alkylating agent with potent antineoplastic and immunosuppressive properties and possibly the most widely used antineoplastic agent. Chronic cardiotoxicity associated with CYP is characterized by progressive heart failure developing from weeks to years after therapy. Methods In this study, rats were administered with (60 mg/kg and 120 mg/kg) alone or in combination with single intraperitoneal (200 mg/kg) administration of CYP for 7 days. CYP was only administered on day 1. Results The administration of CYP led to a significant (p<0.05) increase in cardiac and renal malondialdehyde (MDA) contents and hydrogen peroxide (H2O2) generation. Also, the activities of catalase (CAT), glutathione peroxidase (GPx), glutathione-S-transferase (GST) and reduced glutathione (GSH) levels were significantly (p<0.05) reduced following CYP treatment. A significant (p<0.05) increase in serum myeloperoxidase (MPO) activity was recorded in rats administered CYP only. Electrocardiogram (ECG) showed a significant (p<0.05) increase in heart rate (HR) accompanied by transient decrease in QRS duration. Histologic examination revealed architectural anarchy of both heart and kidney of rats that received only CYP. Conclusions In this study, treatment with gallic acid (60 mg/kg and 120 mg/kg) restored the enzymic and non-enzymic antioxidants and also attenuated cardiotoxic and nephrotoxic effect of CYP through free radical scavenging activity, anti-inflammatory and improvement of antioxidant defence system.


Subject(s)
Antineoplastic Agents/adverse effects , Antioxidants/therapeutic use , Cardio-Renal Syndrome/prevention & control , Cyclophosphamide/adverse effects , Gallic Acid/therapeutic use , Protective Agents/therapeutic use , Animals , Biomarkers/metabolism , Cardio-Renal Syndrome/chemically induced , Cardio-Renal Syndrome/diagnosis , Cardio-Renal Syndrome/metabolism , Electrocardiography , Oxidative Stress , Rats , Treatment Outcome
16.
Int J Cardiol ; 238: 22-30, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28341374

ABSTRACT

Female gender, post-menopause, chronic kidney disease (CKD) and (CKD linked) microvascular disease are important risk factors for developing heart failure with preserved ejection fraction (HFpEF). Enhancing our understanding of the interrelation between these risk factors could greatly benefit the identification of new drug targets for future therapy. This review discusses the evidence for the protective role of estradiol (E2) in CKD-associated microvascular disease and related HFpEF. Elevated circulating levels of uremic toxins (UTs) during CKD may act in synergy with hormonal changes during post-menopause and could lead to coronary microvascular endothelial dysfunction in HFpEF. To elucidate the molecular mechanism involved, published transcriptome datasets of indoxyl sulfate (IS), high inorganic phosphate (HP) or E2 treated human derived endothelial cells from the NCBI Gene Expression Omnibus database were analyzed. In total, 36 genes overlapped in both IS- and HP-activated gene sets, 188 genes were increased by UTs (HP and/or IS) and decreased by E2, and 572 genes were decreased by UTs and increased by E2. Based on a comprehensive in silico analysis and literature studies of collected gene sets, we conclude that CKD-accumulated UTs could negatively impact renal and cardiac endothelial homeostasis by triggering extensive inflammatory responses and initiating dysregulation of angiogenesis. E2 may protect (myo)endothelium by inhibiting UTs-induced inflammation and ameliorating UTs-related uremic bleeding and thrombotic diathesis via restored coagulation capacity and hemostasis in injured vessels.


Subject(s)
Cardio-Renal Syndrome/blood , Estrogens/blood , Microvessels/metabolism , Neovascularization, Physiologic/physiology , Postmenopause/blood , Uremia/blood , Cardio-Renal Syndrome/epidemiology , Cardio-Renal Syndrome/prevention & control , Estrogens/therapeutic use , Female , Gene Regulatory Networks/physiology , Humans , Postmenopause/drug effects , Uremia/epidemiology , Uremia/prevention & control , Vascular Diseases/blood , Vascular Diseases/epidemiology , Vascular Diseases/prevention & control
17.
BMC Nephrol ; 18(1): 79, 2017 Feb 28.
Article in English | MEDLINE | ID: mdl-28245790

ABSTRACT

BACKGROUND: Chronic kidney disease (CKD) is an independent risk factor for cardiovascular disease (CVD) events, and a number of reports have shown a relationship between CKD and CVD in pre-dialysis or maintenance dialysis patients. However, few studies have reported serial observations during dialysis initiation and maintenance. Therefore, we examined whether the incidence of heart disease events differed between CKD patients with and without a history of coronary heart disease (CHD) at dialysis initiation. METHODS: The subjects were patients in the 17 centers participating in the Aichi Cohort Study of Prognosis in Patients Newly Initiated into Dialysis (AICOPP) from October 2011 to September 2013. We excluded nine patients whose outcomes were unknown, as determined by a survey conducted at the end of March 2015. Thus, we enrolled 1,515 subjects into the study. We classified patients into 2 groups according to the history of CHD (i.e., a CHD group and a non-CHD group). Propensity scores (PS) represented the probability of being assigned to a group with or without a history of CHD. Onset of heart disease events and associated mortality and all-cause mortality were compared in PS-matched patients by using the log-rank test for Kaplan-Meier curves. Factors contributing to heart disease events were examined using stepwise multivariate Cox proportional hazards analysis. RESULTS: There were 254 patients in each group after PS-matching. During observation, heart disease events occurred in 85 patients (33.5%) in the CHD group and 48 (18.9%) patients in the non-CHD group. The incidence was significantly higher in the CHD group (p < 0.0001). The CHD group was associated with higher incidence of heart disease events (vs. the non-CHD group, hazard ratio = 1.750, 95% confidence interval = 1.160-2.639). In addition, comorbidities such as diabetes mellitus, low body mass index, and low serum high-density lipoprotein cholesterol were associated with higher incidence of events. CONCLUSION: History of CHD at dialysis initiation was associated with a higher incidence of heart disease events and mortality and all-cause mortality. TRIAL REGISTRATION: UMIN 000007096 . Registered 18 January 2012.


Subject(s)
Cardio-Renal Syndrome/mortality , Coronary Disease/mortality , Renal Dialysis/mortality , Renal Dialysis/statistics & numerical data , Renal Insufficiency, Chronic/mortality , Renal Insufficiency, Chronic/therapy , Aged , Cardio-Renal Syndrome/prevention & control , Cohort Studies , Comorbidity , Coronary Disease/diagnosis , Female , Hospitalization/statistics & numerical data , Humans , Incidence , Japan/epidemiology , Male , Prognosis , Propensity Score , Prospective Studies , Risk Factors , Survival Rate , Time-to-Treatment
18.
J Urol ; 197(3 Pt 1): 566-573, 2017 03.
Article in English | MEDLINE | ID: mdl-27746281

ABSTRACT

PURPOSE: Currently no data exist to guide renal surgeons on the perioperative use of renin-angiotensin blockers despite potential cardiorenal benefits. We aimed to assess the impact of resuming renin-angiotensin blockers on postoperative renal function and adverse cardiac events following partial nephrectomy. MATERIALS AND METHODS: This is an observational analysis of patients who underwent robot-assisted laparoscopic partial nephrectomy from 2006 to 2014 at a single institution. The Wilcoxon rank sum and chi-square tests, and logistic regression were used to assess the risk of adverse renal and cardiac events stratified by history and pattern of renin-angiotensin blockade perioperatively. RESULTS: We identified 900 patients with a median followup of 16.3 months (IQR 1.4-39.1). There were no significant differences in severe renal dysfunction at last followup on univariate analysis or adverse cardiac events at 30 days on multivariate analysis in patients stratified by a history of renin-angiotensin blockade. Of the 338 patients 137 (41.9%) resumed renin-angiotensin blockade immediately after surgery, which did not result in any significant difference in the postoperative glomerular filtration rate (p >0.05). Resuming renin-angiotensin blockade at discharge home was associated with a decreased risk of heart failure within 30 days of surgery (0.3% vs 11.8% of cases) and stage IV/V chronic kidney disease at last followup (2.6% vs 25.5%, each p <0.001). CONCLUSIONS: Renin-angiotensin blockers appear safe to continue immediately after renal surgery. Discharge home with angiotensin converting enzyme inhibitors/angiotensin receptor blockers was associated with a decreased risk of heart failure and severe renal dysfunction. However, this risk may be overstated as a result of the small number of patients discharged without resuming the home medication.


Subject(s)
Angiotensin Receptor Antagonists/therapeutic use , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Cardio-Renal Syndrome/prevention & control , Laparoscopy/methods , Nephrectomy/methods , Robotic Surgical Procedures/methods , Aged , Female , Humans , Kidney Function Tests , Male , Middle Aged , Treatment Outcome
19.
Hypertension ; 69(2): 259-266, 2017 02.
Article in English | MEDLINE | ID: mdl-27872232

ABSTRACT

Hyperhomocysteinemia (hHcys) is an important independent risk factor for the development of cardiovascular disease and end-stage renal disease. Although multiple approaches lowering the levels of homocysteine have been used in experimental studies and clinical trials, there is no effective therapy available to fully prevent homocysteine-induced injury. Therefore, identifying key molecules in the pathogenic pathways may provide clues to develop new therapeutic strategies for the treatment of hHcys-associated injury beyond lowering the plasma homocysteine levels. In this study, we found that the levels of progranulin (PGRN), an autocrine growth factor, were significantly reduced in the kidney and heart from a mouse model of hHcys. We further observed that in hHcys, PGRN-deficient mice significantly exacerbated cardiorenal injury as evidenced by higher levels of urinary albumin excretion, more severe renal morphological injuries, including pronounced glomerular basement membrane thickening and podocyte foot process effacement, and adverse myocardial remodeling versus wild-type mice. Mechanistically, we found that PGRN-medicated Wnt/ß-catenin signaling was one of the critical signal transduction pathways that links homocysteine to cardiorenal injury. Importantly, we finally provided direct evidence for the therapeutic potential of PGRN in mice with hHcys by pretreatment with recombinant human PGRN. Collectively, our results suggest that PGRN may be an innovative therapeutic strategy for treating patients with hHcys.


Subject(s)
Cardio-Renal Syndrome/prevention & control , Hyperhomocysteinemia/complications , Intercellular Signaling Peptides and Proteins/therapeutic use , Animals , Cardio-Renal Syndrome/diagnosis , Cardio-Renal Syndrome/etiology , Disease Models, Animal , Echocardiography , Granulins , Homocysteine/blood , Hyperhomocysteinemia/blood , Hyperhomocysteinemia/drug therapy , Kidney Glomerulus/drug effects , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Progranulins , Recombinant Proteins/therapeutic use
20.
Dtsch Med Wochenschr ; 141(24): 1771-1776, 2016 Nov.
Article in German | MEDLINE | ID: mdl-27903028

ABSTRACT

Patients with kidney disease have a significantly increased cardiovascular morbidity and mortality. Especially diabetics have an increased risk to develop renal insufficiency and cardiovascular events. Two recent studies show that the SGLT2 inhibitor Empagliflozin and the GLP1 agonist Liraglutid are able to lower the cardiovascular risk of type2 diabetics with renal insufficiency. Recent observations suggest that bradycardia and asystole are main triggers for sudden cardiac death in patients with chronic kidney disease. In line with these results registry data failed to confirm benefits of cardioverters/defibrillators in hemodialysis patients. Whether cardiac resynchronization therapy may slower pathomechanisms of cardiomyopathy in patients with chronic kidney disease still needs to be confirmed in prospective trials. Taken together implementation of device therapy in hemodialysis patients should integrate indications of current guidelines with individual patient risks and life expectancy. Everolimus-eluting stents have shown superior results compared to bare metal stents in terms of ischemia-driven target-lesion-revascularization in patients with chronic kidney disease in a prospective trial. These results were corroborated by registry data reporting survival benefit associated with the use of drug-eluting stents in patients with chronic renal disease.


Subject(s)
Cardiovascular Diseases/mortality , Cardiovascular Diseases/prevention & control , Kidney Diseases/mortality , Kidney Diseases/therapy , Cardio-Renal Syndrome/mortality , Cardio-Renal Syndrome/prevention & control , Comorbidity , Evidence-Based Medicine , Germany , Humans , Prevalence , Risk Factors , Survival Rate , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...