Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 160
Filter
1.
Biochem J ; 477(10): 1907-1921, 2020 05 29.
Article in English | MEDLINE | ID: mdl-32255479

ABSTRACT

Continuous exposure to peritoneal dialysis (PD) fluid results in peritoneal fibrosis and ultimately causes ultrafiltration failure. Noncoding RNAs, including long noncoding RNAs (lncRNAs) and microRNAs (miRNAs), have been reported to participate in ultrafiltration failure in PD. Therefore, our study aimed to investigate the mechanism of lncRNA 6030408B16RIK in association with miR-326-3p in ultrafiltration failure in PD. Peritoneal tissues were collected from uremic patients with or without PD. A uremic rat model with PD was first established by 5/6 nephrectomy. The relationship between lncRNA 6030408B16RIK, miR-326-3p and WISP2 was identified using luciferase reporter, RNA pull-down and RIP assays. After ectopic expression and depletion treatments in cells, expression of α-SMA, phosphorylated ß-catenin, FSP1, E-cadherin and Vimentin was evaluated by RT-qPCR and Western blot analyses, and Collagen III and CD31 expression by immunohistochemistry. Ultrafiltration volume and glucose transport capacity were assessed by the peritoneal equilibration test. Expression of lncRNA 6030408B16RIK and WISP2 was up-regulated and miR-326-3p expression was poor in peritoneal tissues of uremic PD patients and model rats. LncRNA 6030408B16RIK competitively bound to miR-326-3p and then elevated WISP2 expression. Silencing of lncRNA 6030408B16RIK and WISP2 or overexpression of miR-326-3p was shown to decrease the expression of α-SMA, phosphorylated ß-catenin, FSP1, Vimentin, Collagen III and CD31, while reducing glucose transport capacity and increasing E-cadherin expression and ultrafiltration volume in uremic PD rats. In summary, lncRNA 6030408B16RIK silencing exerts an anti-fibrotic effect on uremic PD rats with ultrafiltration failure by inactivating the WISP2-dependent Wnt/ß-catenin pathway via miR-326-3p.


Subject(s)
MicroRNAs/metabolism , RNA, Long Noncoding/genetics , Uremia , Actins/metabolism , Animals , CCN Intercellular Signaling Proteins/metabolism , Cadherins/metabolism , Extracellular Matrix/metabolism , Gene Silencing , Humans , Models, Animal , Peritoneal Dialysis/adverse effects , RNA, Long Noncoding/metabolism , Rats , Repressor Proteins/metabolism , Ultrafiltration , Uremia/prevention & control , Vimentin/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism
2.
Semin Dial ; 32(4): 368-378, 2019 07.
Article in English | MEDLINE | ID: mdl-30968465

ABSTRACT

Dialysis adequacy is an independent predictor of high mortality rates in hemodialysis patients. Intradialytic exercise is a potential strategy to increase uremic solute removal by increasing blood flow to low perfusion tissue beds. The purpose of this review is to establish the efficacy of intradialytic exercise for hemodialysis adequacy. Additionally, this review aims to provide practical information to aid health care professionals implement intradialytic exercise for dialysis adequacy. Database and hand searches identified 15 published interventional studies that implemented intradialytic exercise for dialysis adequacy as a primary outcome measure in adult maintenance hemodialysis patients. Data pertaining to dialytic solute clearance of urea, creatinine, beta2 microglobulin, phosphate, and potassium were extracted. Mean differences, normalized to percentages, and effect sizes were calculated and reported. The current data pertaining to the use of intradialytic exercise for improving dialysis adequacy in terms of Kt/Vurea or small molecule uremic toxin clearance are equivocal. Limited data showed that intradialytic exercise has no effect middle molecule toxin (beta2 - microglobulin) clearance. Intradialytic exercise favored increased phosphate removal showing medium to large effects for reduced serum concentrations, reduced rebound and increased clearance. In summary, supervised light to moderate intradialytic aerobic cycling appears to be beneficial for increasing phosphate removal and may be an adjunct therapy for patients failing to meet clinical phosphate targets. Further work is required to establish the effect of intradialytic exercise on Kt/Vurea and other middle molecule and protein bound solutes. Research aimed at establishing the most effective exercise prescription for improved solute clearance is warranted.


Subject(s)
Dialysis Solutions/pharmacokinetics , Exercise Therapy/methods , Kidney Failure, Chronic/therapy , Quality of Life , Renal Dialysis/methods , Uremia/prevention & control , Aged , Combined Modality Therapy , Creatinine/blood , Female , Glomerular Filtration Rate , Humans , Kidney Failure, Chronic/diagnosis , Kidney Function Tests , Male , Middle Aged , Prognosis , Risk Assessment , Treatment Outcome , Urea/blood
3.
Cardiovasc Drugs Ther ; 33(3): 277-286, 2019 06.
Article in English | MEDLINE | ID: mdl-30903544

ABSTRACT

PURPOSE: Several lines of evidence suggest that renal dysfunction is associated with cardiovascular toxicity through the action of uremic toxins. The levels of those uremic toxins can be reportedly reduced by the spherical carbon adsorbent AST-120. Because heart failure (HF) causes renal dysfunction by low cardiac output and renal edema, the removal of uremic toxins could be cardioprotective. METHOD: To determine whether blood levels of the uremic toxin indoxyl sulfate (IS) increase in HF and whether AST-120 can reduce those levels and improve HF. We induced HF in 12 beagle dogs by 6 weeks of rapid right ventricular pacing at 230 beats per min. We treated six dogs with a 1-g/kg/day oral dosage of AST-120 for 14 days from week 4 after the start of rapid ventricular pacing. The other six dogs did not receive any treatment (control group). RESULTS: In the untreated dogs, IS levels increased as cardiac function deteriorated. In contrast, plasma IS levels in the treated dogs decreased to baseline levels, with both left ventricular fractional shortening and pulmonary capillary wedge pressure also improving when compared with untreated dogs. Finally, AST-120 treatment was shown to reduce both myocardial apoptosis and fibrosis along with decreases in extracellular signal-regulated kinase phosphorylation, the Bax/Bcl-2 ratio, and TGF-ß1 expression and increases in AKT phosphorylation. CONCLUSIONS: IS levels are increased in HF. AST-120 treatment reduces the levels of IS and improves the pathophysiology of HF in a canine model. AST-120 could be a novel candidate for the treatment of HF.


Subject(s)
Carbon/administration & dosage , Cardio-Renal Syndrome/therapy , Heart Failure/therapy , Indican/blood , Kidney Diseases/prevention & control , Oxides/administration & dosage , Sorption Detoxification/methods , Uremia/prevention & control , Adsorption , Animals , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Cardio-Renal Syndrome/blood , Cardio-Renal Syndrome/physiopathology , Consciousness , Disease Models, Animal , Dogs , Fibrosis , Heart Failure/blood , Heart Failure/complications , Heart Failure/physiopathology , Hemodynamics , Kidney Diseases/blood , Kidney Diseases/etiology , Kidney Diseases/physiopathology , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Myocardium/metabolism , Myocardium/pathology , Signal Transduction , Uremia/blood , Uremia/etiology , Uremia/physiopathology , Ventricular Function, Left
4.
J Intensive Care Med ; 34(2): 94-103, 2019 Feb.
Article in English | MEDLINE | ID: mdl-29739260

ABSTRACT

Acute kidney injury (AKI) is common in critically ill patients and associated with increased morbidity and mortality. With the increased use of renal replacement therapy (RRT) for severe AKI, the optimal time for initiation of RRT has become one of the most probed and debated topic in the field of nephrology and critical care. There appears to be an increased trend toward earlier initiation of RRT to avoid life-threatening complications associated with AKI. Despite the presence of a plethora of studies in this field, the lack of uniformity in study design, patient population types, definition of early and late initiation, modality of RRT, and results, the optimal time for starting RRT in AKI still remains unknown. The beneficial effects reported in observational studies have not been supported by clinical trials. Recently, 2 of the largest randomized control trials evaluating the timing of RRT in critically ill patients with AKI showed differing results. We provide an in-depth review of the available data on the timing of dialysis in patients with AKI.


Subject(s)
Acute Kidney Injury/therapy , Critical Care/methods , Renal Replacement Therapy , Time-to-Treatment , Acute Kidney Injury/complications , Humans , Observational Studies as Topic , Randomized Controlled Trials as Topic , Research Design , Uremia/prevention & control , Water-Electrolyte Imbalance/prevention & control
5.
Clin Exp Nephrol ; 23(7): 908-919, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30895529

ABSTRACT

BACKGROUND: Constipation is frequently observed in patients with chronic kidney disease (CKD). Lactulose is expected to improve the intestinal environment by stimulating bowel movements as a disaccharide laxative and prebiotic. We studied the effect of lactulose on renal function in adenine-induced CKD rats and monitored uremic toxins and gut microbiota. METHODS: Wistar/ST male rats (10-week-old) were fed 0.75% adenine-containing diet for 3 weeks to induce CKD. Then, they were divided into three groups and fed as follows: control, normal diet; and 3.0- and 7.5-Lac, 3.0% and 7.5% lactulose-containing diets, respectively, for 4 weeks. Normal diet group was fed normal diet for 7 weeks. The rats were observed for parameters including renal function, uremic toxins, and gut microbiota. RESULTS: The control group showed significantly higher serum creatinine (sCr) and blood urea nitrogen (BUN) 3 weeks after adenine feeding than at baseline, with a 8.5-fold increase in serum indoxyl sulfate (IS). After switching to 4 weeks of normal diet following adenine feeding, the sCr and BUN in control group remained high with a further increase in serum IS. In addition, tubulointerstitial fibrosis area was increased in control group. On the other hand, 3.0- and 7.5-Lac groups improved sCr and BUN levels, and suppressed tubulointerstitial fibrosis, suggesting preventing of CKD progression by lactulose. Lac groups also lowered level of serum IS and proportions of gut microbiota producing IS precursor. CONCLUSION: Lactulose modifies gut microbiota and ameliorates CKD progression by suppressing uremic toxin production.


Subject(s)
Adenine , Bacteria/drug effects , Gastrointestinal Microbiome/drug effects , Kidney/drug effects , Lactulose/pharmacology , Prebiotics , Renal Insufficiency, Chronic/prevention & control , Uremia/prevention & control , Animals , Bacteria/metabolism , Biomarkers/blood , Blood Urea Nitrogen , Creatinine/blood , Disease Models, Animal , Disease Progression , Fibrosis , Kidney/metabolism , Kidney/pathology , Kidney/physiopathology , Male , Oxidative Stress/drug effects , Rats, Wistar , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/microbiology , Renal Insufficiency, Chronic/physiopathology , Uremia/chemically induced , Uremia/microbiology , Uremia/physiopathology
6.
BMC Nephrol ; 20(1): 358, 2019 09 14.
Article in English | MEDLINE | ID: mdl-31521120

ABSTRACT

BACKGROUND: Cardiovascular disease is the most common complication and leading cause of death in maintenance hemodialysis patients. The protection mechanism of hydrogen sulfide (H2S) and the specific role of conventional protein kinase C ßII (cPKCßII)/Akt signaling pathway in the formation of atherosclerosis is still controversial. METHODS: 8-week-old male ApoE-/- mice were treated with 5/6 nephrectomy and high-fat diet to make uremia accelerated atherosclerosis (UAAS) model. Mice were divided into normal control group (control group), sham operation group (sham group), UAAS group, L-cysteine group (UAAS+L-cys group), sodium hydrosulfide group (UAAS+NaHS group), and propargylglycine group (UAAS+PPG group). Western blot was used to detect cPKCßII activation, Akt phosphorylation and endothelial nitric oxide synthase (eNOS) expression in mice aorta. RESULTS: The membrane translocation of cPKCßII in UAAS group was higher than sham group, and L-cys or NaHS injection could suppress the membrane translocation, but PPG treatment resulted in more membrane translocation of cPKCßII (P < 0.05, n = 6 per group). Akt phosphorylation and the eNOS expression in UAAS group was lower than sham group, and L-cys or NaHS injection could suppress the degradation of Akt phosphorylation and the eNOS expression, but PPG treatment resulted in more decrease in the Akt phosphorylation and the eNOS expression (P < 0.05, n = 6 per group). CONCLUSION: Endogenous cystathionine-γ-lyase (CSE)/H2S system protected against the formation of UAAS via cPKCßII/Akt signal pathway. The imbalance of CSE/H2S system may participate in the formation of UAAS by affecting the expression of downstream molecule eNOS, which may be mediated by cPKCßII/Akt signaling pathway.


Subject(s)
Atherosclerosis/metabolism , Hydrogen Sulfide/metabolism , Protein Kinase C beta/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Uremia/metabolism , Animals , Atherosclerosis/etiology , Atherosclerosis/prevention & control , Diet, High-Fat/adverse effects , Male , Mice , Mice, Knockout , Protein Kinase C beta/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Uremia/etiology , Uremia/prevention & control
7.
Eur J Clin Invest ; 48(11): e13020, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30144313

ABSTRACT

Chronic kidney disease (CKD), which affects 10%-15% of the population, associates with a range of complications-such as cardiovascular disease, frailty, infections, muscle and bone disorders and premature ageing-that could be related to alterations of mitochondrial number, distribution, structure and function. As mitochondrial biogenesis, bioenergetics and the dynamic mitochondrial networks directly or indirectly regulate numerous intra- and extracellular functions, the mitochondria have emerged as an important target for interventions aiming at preventing or improving the treatment of complications in CKD. In this review, we discuss the possible role of bioactive food compounds and exercise in the modulation of the disturbed mitochondrial function in a uraemic milieu.


Subject(s)
Biological Factors/therapeutic use , Exercise Therapy , Mitochondrial Diseases/prevention & control , Renal Insufficiency, Chronic/etiology , Diet , Energy Metabolism/physiology , Humans , Mitochondria/physiology , Oxidative Stress/physiology , Phytochemicals/therapeutic use , Reactive Oxygen Species/metabolism , Renal Insufficiency, Chronic/prevention & control , Uremia/prevention & control
8.
Clin Exp Nephrol ; 21(1): 7-15, 2017 Feb.
Article in English | MEDLINE | ID: mdl-26965149

ABSTRACT

It is well-established that uremic toxins are positively correlated with the risk of developing chronic kidney disease and cardiovascular disease. In addition, emerging data suggest that gut bacteria exert an influence over both the production of uremic toxins and the development of chronic kidney disease. As such, modifying the gut microbiota may have the potential as a treatment for chronic kidney disease. This is supported by data that suggest that rescuing microbiota dysbiosis may: reduce uremic toxin production; prevent toxins and pathogens from crossing the intestinal barrier; and, reduce gastrointestinal tract transit time allowing nutrients to reach the microbiota in the distal portion of the gastrointestinal tract. Despite emerging literature, the gut-kidney axis has yet to be fully explored. A special focus should be placed on examining clinically translatable strategies that might encourage improvements to the microbiome, thereby potentially reducing the risk of the development of chronic kidney disease. This review aims to present an overview of literature linking changes to the gastrointestinal tract with microbiota dysbiosis and the development and progression of chronic kidney disease.


Subject(s)
Bacteria/metabolism , Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Renal Insufficiency, Chronic/microbiology , Uremia/microbiology , Animals , Bacteria/immunology , Bacterial Translocation , Disease Progression , Dysbiosis , Host-Pathogen Interactions , Humans , Probiotics/therapeutic use , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/prevention & control , Uremia/metabolism , Uremia/prevention & control
9.
Semin Dial ; 29(5): 333-7, 2016 09.
Article in English | MEDLINE | ID: mdl-27174444

ABSTRACT

Urea is an old uremic toxin which has been used for many years as a global biomarker of CKD severity and dialysis adequacy. Old studies were not in favor of its role as a causal factor in the pathogenesis of complications associated with the uremic state. However, recent experimental and clinical evidence is compatible with both direct and indirect toxicity of urea, particularly via the deleterious actions of urea-derived carbamylated molecules. Further studies are clearly needed to explore the potential relevance of urea-related CKD complications for patient management, in particular the place of new therapeutic strategies to prevent urea toxicity.


Subject(s)
Renal Dialysis , Urea/blood , Uremia/diagnosis , Uremia/prevention & control , Biomarkers/blood , Humans
10.
J Am Soc Nephrol ; 26(8): 1787-94, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25525179

ABSTRACT

The accumulation of uremic toxins is involved in the progression of CKD. Various uremic toxins are derived from gut microbiota, and an imbalance of gut microbiota or dysbiosis is related to renal failure. However, the pathophysiologic mechanisms underlying the relationship between the gut microbiota and renal failure are still obscure. Using an adenine-induced renal failure mouse model, we evaluated the effects of the ClC-2 chloride channel activator lubiprostone (commonly used for the treatment of constipation) on CKD. Oral administration of lubiprostone (500 µg/kg per day) changed the fecal and intestinal properties in mice with renal failure. Additionally, lubiprostone treatment reduced the elevated BUN and protected against tubulointerstitial damage, renal fibrosis, and inflammation. Gut microbiome analysis of 16S rRNA genes in the renal failure mice showed that lubiprostone treatment altered their microbial composition, especially the recovery of the levels of the Lactobacillaceae family and Prevotella genus, which were significantly reduced in the renal failure mice. Furthermore, capillary electrophoresis-mass spectrometry-based metabolome analysis showed that lubiprostone treatment decreased the plasma level of uremic toxins, such as indoxyl sulfate and hippurate, which are derived from gut microbiota, and a more recently discovered uremic toxin, trans-aconitate. These results suggest that lubiprostone ameliorates the progression of CKD and the accumulation of uremic toxins by improving the gut microbiota and intestinal environment.


Subject(s)
Alprostadil/analogs & derivatives , Chloride Channel Agonists/therapeutic use , Gastrointestinal Tract/drug effects , Kidney Failure, Chronic/prevention & control , Microbiota/drug effects , Adenine , Alprostadil/pharmacology , Alprostadil/therapeutic use , Animals , Chloride Channel Agonists/pharmacology , Disease Progression , Drug Evaluation, Preclinical , Gastrointestinal Tract/microbiology , Kidney Failure, Chronic/chemically induced , Lubiprostone , Male , Mice, Inbred C57BL , Random Allocation , Uremia/prevention & control
11.
Semin Dial ; 28(1): 75-80, 2015.
Article in English | MEDLINE | ID: mdl-25319504

ABSTRACT

Prior to the availability of hemodialysis, dietary protein restriction played a large part in the treatment of uremia. This therapy was based on observations that uremic symptoms increased with high protein intake. Early investigators thus presumed that "uremic toxins" were derived from the breakdown of dietary protein; its restriction improved uremic symptoms but caused malnutrition. After the availability of hemodialysis, protein restriction was no longer recommended. Studies in healthy subjects have shown that an intake of 0.6-0.8 g/kg/day is adequate to prevent protein malnutrition. Guidelines for hemodialysis patients, however, currently recommend higher protein intakes of 1.2 g/kg/day. A downside to higher intake may be increased production of protein-derived uremic solutes that caused the symptoms observed by early investigators. Some of these solutes are produced by colon microbes acting on protein which escapes digestion in the small intestine. Increasing dietary fiber may reduce the production of colon-derived solutes in hemodialysis patients without adverse effects of protein restriction. Fiber comprises carbohydrates and related substances that are resistant to digestion in the small intestine. Upon delivery to the colon, fiber is broken down to short chain fatty acids, providing energy to both the microbes and the host. With an increased energy supply, the microbes can incorporate dietary protein for growth rather than breaking them down to uremic solutes. Increasing fiber intake in hemodialysis patients has been shown to reduce the plasma levels of selected colon-derived solutes. Further studies are needed to test whether this provides clinical benefit.


Subject(s)
Dietary Fiber/administration & dosage , Dietary Proteins/administration & dosage , Kidney Failure, Chronic/therapy , Renal Dialysis/adverse effects , Uremia/etiology , Uremia/prevention & control , Diet, Protein-Restricted , Humans
12.
J Ren Nutr ; 25(3): 316-20, 2015 May.
Article in English | MEDLINE | ID: mdl-25446837

ABSTRACT

OBJECTIVE: To determine the effects of supplemental fiber on plasma p-cresol, stool frequency, and quality of life (QoL) in chronic kidney disease (CKD) patients. DESIGN AND SETTING: In a 12-week single-blind study, participants were provided with control muffins and supplements (5.5 g sucrose/day) for 2 weeks, muffins containing 10 g/day pea hull fiber and control supplements for 4 weeks, and muffins with 10 g/day pea hull fiber and 15 g/day inulin as a supplement for 6 weeks. SUBJECTS: Individuals with CKD (n = 13; 6 males, 7 females; aged 65 ± 3 years; estimated glomerular filtration rate <50 mL/minute/1.73(2)) completed the study. MAIN OUTCOME MEASURES: Plasma p-cresol was determined by gas chromatography-mass spectrometry, stool frequency by 5-day journals, and QoL by the KDQOL-36™. RESULTS: Plasma p-cresol decreased from 7.25 ± 1.74 mg/L during week 1 to 5.82 ± 1.72 mg/L during week 12 (P < .05), and in participants with high compliance (>70% inulin intake), from 6.71 ± 1.98 mg/L to 4.22 ± 1.16 mg/L (P < .05). Total fiber intake increased from 16.6 ± 1.7 g/day during control to 26.5 ± 2.4 g/day (P < .0001) with the added pea hull and to 34.5 ± 2.2 g/day with pea hull and inulin (P < .0001). Stool frequency increased from 1.4 ± 0.2 stools/day during control to 1.9 ± 0.3 stools/day during both fiber periods (P < .05). No change in overall QoL was observed. CONCLUSIONS: Supplementing the diet of CKD patients with fiber may be a dietary therapy to reduce p-cresol and improve stool frequency.


Subject(s)
Cresols/blood , Dietary Fiber/administration & dosage , Renal Insufficiency, Chronic/blood , Aged , Defecation , Female , Humans , Male , Middle Aged , Quality of Life , Uremia/blood , Uremia/prevention & control
13.
J Mater Sci Mater Med ; 25(6): 1589-97, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24573455

ABSTRACT

Adsorbents designed with porosity which allows the removal of protein bound and high molecular weight uraemic toxins may improve the effectiveness of haemodialysis treatment of chronic kidney disease (CKD). A nanoporous activated carbon monolith prototype designed for direct blood contact was first assessed for its capacity to remove albumin bound marker toxins indoxyl sulphate (IS), p-cresyl sulphate (p-CS) and high molecular weight cytokine interleukin-6 in spiked healthy donor studies. Haemodialysis patient blood samples were then used to measure the presence of these markers in pre- and post-dialysis blood and their removal by adsorbent recirculation of post-dialysis blood samples. Nanopores (20-100 nm) were necessary for marker uraemic toxin removal during in vitro studies. Limited removal of IS and p-CS occurred during haemodialysis, whereas almost complete removal occurred following perfusion through the carbon monoliths suggesting a key role for such adsorbent therapies in CKD patient care.


Subject(s)
Charcoal/chemistry , Cresols/isolation & purification , Hemofiltration/instrumentation , Indican/isolation & purification , Interleukin-6/isolation & purification , Renal Dialysis/instrumentation , Sulfuric Acid Esters/isolation & purification , Uremia/blood , Absorption , Cresols/blood , Equipment Design , Equipment Failure Analysis , Humans , Indican/blood , Interleukin-6/blood , Materials Testing , Membranes, Artificial , Pilot Projects , Sulfuric Acid Esters/blood , Uremia/prevention & control
14.
Nephron Clin Pract ; 123(1-2): 83-92, 2013.
Article in English | MEDLINE | ID: mdl-23796960

ABSTRACT

UNLABELLED: In adults, recurrent hemodialysis (HD)-induced cardiac injury results in ischemic myocardial dysfunction. Uremic children, like adults, share the full complement of uremia-related cardiovascular abnormalities but without significant atheromatous coronary artery disease. The aim of this study was, to assess the impact of HD on left ventricular (LV) myocardial function in children. METHOD: We assessed all single-center chronic HD patients (n = 15, range 1-17 years) excluding those with overt cardiac disease. Regional LV function and mechanical synchronicity was measured echographically by two-dimensional segmental longitudinal, circumferential and radial myocardial strain. All patients were assessed pre-dialysis and at the end of dialysis. In addition, we scanned age-matched controls at rest. RESULTS: The peak longitudinal strain was lower in uremic patients compared with controls with a significant fall during HD (mean peak strain -19.9 controls, -17.9 pre-HD, -15.3 end of HD, p < 0.05). Radial strain was lower in uremic patients and increased during HD. Circumferential strain was preserved in uremic patients and fell during HD. Intrasegmental deformation synchronicity was progressively worse pre-dialysis and end of dialysis compared with controls. Intradialytic peak longitudinal strain reduction was significantly associated with systolic blood pressure and ultrafiltrate volume (p < 0.05). CONCLUSION: Uremic children have impaired regional LV function, with a predisposition to longitudinal axis dysfunction and LV mechanical dyssynchrony, both of which are established markers of ischemic injury. This is further evidence for a characteristic cardiovascular phenotype in uremic patients that predisposes them to subclinical demand ischemia during dialysis.


Subject(s)
Renal Dialysis/adverse effects , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/rehabilitation , Uremia/etiology , Uremia/prevention & control , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Dysfunction, Left/etiology , Adolescent , Child , Child, Preschool , Echocardiography/methods , Elasticity Imaging Techniques/methods , Female , Humans , Infant , Male , Renal Insufficiency, Chronic/diagnostic imaging , Treatment Outcome , Uremia/diagnosis
15.
Nephrol Dial Transplant ; 27(6): 2198-205, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22036941

ABSTRACT

BACKGROUND: Secondary hyperparathyroidism (sHPT) represents an adaptive response to progressively impaired control of calcium, phosphorus and vitamin D in chronic kidney disease (CKD). It is characterized by parathyroid hyperplasia and excessive synthesis and secretion of parathyroid hormone (PTH). Parathyroid hyperplasia in uremic rats can be prevented by calcium-sensing receptor (CaSR) activation with the calcimimetic cinacalcet (Sensipar®/Mimpara®); however, it is unknown, how long the effects of cinacalcet persist after withdrawal of treatment or if cinacalcet is efficacious in uremic rats with established sHPT. METHODS: We sought to determine the effect of cinacalcet discontinuation in uremic rats and whether cinacalcet was capable of influencing parathyroid hyperplasia in animals with established sHPT. RESULTS: Discontinuation of cinacalcet resulted in reversal of the beneficial effects on serum PTH and parathyroid hyperplasia. In rats with established sHPT, cinacalcet decreased serum PTH and mediated regression of parathyroid hyperplasia. The cinacalcet-mediated decrease in parathyroid gland size was accompanied by increased expression of the cyclin-dependent kinase inhibitor p21. Prevention of cellular proliferation with cinacalcet occurred despite increased serum phosphorus and decreased serum calcium. CONCLUSIONS: The animal data provided suggest established parathyroid hyperplasia can be reversed by modulating CaSR activity with cinacalcet and that continued treatment may be necessary to maintain reductions in PTH.


Subject(s)
Disease Models, Animal , Hyperparathyroidism, Secondary/prevention & control , Hyperplasia/prevention & control , Kidney Failure, Chronic/complications , Naphthalenes/therapeutic use , Parathyroid Glands/drug effects , Uremia/prevention & control , Animals , Calcium/blood , Cell Proliferation/drug effects , Cinacalcet , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Hyperparathyroidism, Secondary/etiology , Hyperparathyroidism, Secondary/pathology , Hyperplasia/etiology , Hyperplasia/pathology , Kidney Failure, Chronic/pathology , Male , Parathyroid Glands/metabolism , Parathyroid Glands/pathology , Parathyroid Hormone/blood , Phosphorus/blood , Rats , Rats, Sprague-Dawley , Uremia/etiology , Uremia/pathology
16.
BMC Nephrol ; 13: 131, 2012 Oct 03.
Article in English | MEDLINE | ID: mdl-23033904

ABSTRACT

BACKGROUND: A number of studies during the nineties have shown that antihypertensive therapy, particularly using RAS blockade, can reduce uremia progression, and ESRD incidence. METHODS: National incidence rates were studied of end stage renal disease (ESRD) for Denmark between 1990 and 2011, and of national prescription of antihypertensive drugs between 1995 and 2010, in order to investigate whether prescription rates had changed, and whether the expected change in ESRD had materialized. The Danish Nephrology Registry (DNR) is incident and comprehensive. Incidence rates were classified according to renal diagnosis. RESULTS: ESRD incidence was constant for age groups <60 years. Incidence rates rose during the nineties for all cohorts >60 years. Since 2001 rates for subjects 60-70 years have fallen from 400 ppm/yr to 234, and since 2002 for subjects 70-80 years from 592 to 398. The incidence of patients >80 years has increased to 341. The falling incidence for patients 60-80 years was distributed among a number of diagnoses. Since 1995 national antihypertensive drug therapy has increased from 24.5 defined daily doses (DDD)/citizen/yr to 101.3, and the proportion using renin-angiotensin system (RAS) blockade from 34 to 58%. CONCLUSIONS: This national study has shown a reduction in actively treated ESRD incidence among patients aged 60-80 years. It is possible that this is the result of increased antihypertensive prescription rates, particularly with RAS blockade. If it is assumed that therapeutic intervention is the cause of the observed reduced incidence, ESRD incidence has been reduced by 33.8 ppm/yr, prevalence by 121 ppm, and ESRD expenditure by 6 €/citizen/yr.


Subject(s)
Antihypertensive Agents/administration & dosage , Kidney Failure, Chronic/epidemiology , Kidney Failure, Chronic/prevention & control , Prescriptions/statistics & numerical data , Uremia/epidemiology , Uremia/prevention & control , Age Distribution , Aged , Aged, 80 and over , Denmark/epidemiology , Female , Humans , Incidence , Male , Middle Aged , Risk Factors , Treatment Outcome
17.
Nephrol Dial Transplant ; 26(3): 832-8, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20810455

ABSTRACT

BACKGROUND: The mechanisms of uraemia-induced atherosclerosis have not been fully delineated. The aims of this study were (i) to investigate the extent and the phenotype of atherosclerosis, including the activation of local renin-angiotensin system (RAS), in a mouse model of mild uraemia and (ii) to determine the effects of angiotensin II type1 (AT1) receptor blockade on the uraemic atherosclerosis, clarifying the mechanisms of its action. METHODS: Mild uraemia was induced by 5/6 nephrectomy in 8-week-old apo E-deficient mice (apoE-KO). After nephrectomy, the animals received either treatment with candesartan or no treatment for 12-weeks. Sham-operated apoE-KO mice were used as controls. RESULTS: Uraemia led to a two-fold increase in aortic plaque area. This was associated with a significant upregulation of aortic angiotensin-converting enzyme (ACE), AT1 receptor, connective tissue growth factor (CTGF), monocyte chemoattractant protein (MCP)-1 and vascular cell adhesion molecule (VCAM)-1. Candesartan significantly reduced aortic atherosclerosis, prevented the upregulation of the uraemia-induced genes and led to changes predicting greater stability of the plaques, without influencing blood pressure or serum lipids. CONCLUSIONS: This study indicates that uraemia leads to an acceleration of aortic atherosclerosis. The upregulation of aortic RAS and the reduced atherosclerosis following AT1 receptor blocker treatment highlights the pivotal role of the local RAS in the development and acceleration of atherosclerosis in uraemia.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/therapeutic use , Apolipoproteins E/physiology , Atherosclerosis/prevention & control , Receptor, Angiotensin, Type 1/chemistry , Renal Insufficiency/drug therapy , Renal Insufficiency/metabolism , Animals , Aorta/drug effects , Aorta/pathology , Atherosclerosis/metabolism , Benzimidazoles/therapeutic use , Biphenyl Compounds , Mice , Mice, Inbred C57BL , Mice, Knockout , Nephrectomy , Phenotype , RNA, Messenger/genetics , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/chemistry , Receptor, Angiotensin, Type 2/genetics , Receptor, Angiotensin, Type 2/metabolism , Renal Insufficiency/etiology , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tetrazoles/therapeutic use , Uremia/metabolism , Uremia/prevention & control
18.
Toxicol Mech Methods ; 21(6): 473-8, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21417628

ABSTRACT

Chronic renal failure (CRF) patients have an increased plasma level of urea, which can be a source of cyanate. This compound can cause protein carbamoylation thereby changing biological activity of proteins. Therefore, in renal failure patients, cyanate can disturb metabolism and functioning of the liver. This work presents studies demonstrating that the treatment of rats with cyanate alone causes the following changes in the liver: (1) inhibition of rhodanese (TST), cystathionase (CST) and 3-mercaptopyruvate sulfotransferase (MPST) activities, (2) decrease in sulfane sulfur level (S*), (3) lowering of nonprotein sulfhydryl groups (NPSH) group level, and (4) enhancement of prooxidant processes (rise in reactive oxygen species (ROS) and malondialdehyde (MDA) level). This indicates that cyanate inhibits anaerobic cysteine metabolism and shows prooxidant action in the liver. Out of the above-mentioned changes, lipoate administered with cyanate jointly was able to correct MDA, ROS and NPSH levels, and TST activity. It had no significant effect on MPST and CST activities. It indicates that lipoate can prevent prooxidant cyanate action and cyanate-induced TST inhibition. These observations can be promising for CRF patients since lipoate can play a dual role in these patients as an efficient antioxidant defense and a protection against cyanate and cyanide toxicity.


Subject(s)
Antioxidants/therapeutic use , Cyanates/toxicity , Cysteine/metabolism , Liver/drug effects , Oxidative Stress/drug effects , Thioctic Acid/therapeutic use , Uremia/prevention & control , Animals , Hydrogen Sulfide/metabolism , Kidney Failure, Chronic/drug therapy , Lipid Peroxidation/drug effects , Liver/enzymology , Liver/metabolism , Male , Molecular Targeted Therapy , Oxidants/toxicity , Poisons/toxicity , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Sulfhydryl Compounds/metabolism , Thiosulfate Sulfurtransferase/metabolism , Uremia/chemically induced , Uremia/metabolism
19.
Nat Rev Nephrol ; 17(3): 153-171, 2021 03.
Article in English | MEDLINE | ID: mdl-32963366

ABSTRACT

The observation that unhealthy diets (those that are low in whole grains, fruits and vegetables, and high in sugar, salt, saturated fat and ultra-processed foods) are a major risk factor for poor health outcomes has boosted interest in the concept of 'food as medicine'. This concept is especially relevant to metabolic diseases, such as chronic kidney disease (CKD), in which dietary approaches are already used to ameliorate metabolic and nutritional complications. Increased awareness that toxic uraemic metabolites originate not only from intermediary metabolism but also from gut microbial metabolism, which is directly influenced by diet, has fuelled interest in the potential of 'food as medicine' approaches in CKD beyond the current strategies of protein, sodium and phosphate restriction. Bioactive nutrients can alter the composition and metabolism of the microbiota, act as modulators of transcription factors involved in inflammation and oxidative stress, mitigate mitochondrial dysfunction, act as senolytics and impact the epigenome by altering one-carbon metabolism. As gut dysbiosis, inflammation, oxidative stress, mitochondrial dysfunction, premature ageing and epigenetic changes are common features of CKD, these findings suggest that tailored, healthy diets that include bioactive nutrients as part of the foodome could potentially be used to prevent and treat CKD and its complications.


Subject(s)
Diet, Healthy/methods , Renal Insufficiency, Chronic/therapy , Uremia/prevention & control , Disease Progression , Humans , Phenotype , Renal Insufficiency, Chronic/complications , Uremia/etiology
20.
Nat Rev Nephrol ; 17(6): 402-416, 2021 06.
Article in English | MEDLINE | ID: mdl-33758363

ABSTRACT

Chronic kidney disease (CKD) is characterized by the retention of a myriad of solutes termed uraemic (or uremic) toxins, which inflict damage to several organs, including the cardiovascular system. Uraemic toxins can induce hallmarks of cardiovascular disease (CVD), such as atherothrombosis, heart failure, dysrhythmias, vessel calcification and dysregulated angiogenesis. CVD is an important driver of mortality in patients with CKD; however, reliance on conventional approaches to managing CVD risk is insufficient in these patients, underscoring a need to target risk factors that are specific to CKD. Mounting evidence suggests that targeting uraemic toxins and/or pathways induced by uraemic toxins, including tryptophan metabolites and trimethylamine N-oxide (TMAO), can lower the risk of CVD in patients with CKD. Although tangible therapies resulting from our growing knowledge of uraemic toxicity are yet to materialize, a number of pharmacological and non-pharmacological approaches have the potential to abrogate the effects of uraemic toxins, for example, by decreasing the production of uraemic toxins, by modifying metabolic pathways induced by uraemic toxins such as those controlled by aryl hydrocarbon receptor signalling and by augmenting the clearance of uraemic toxins.


Subject(s)
Cardiovascular Diseases/etiology , Renal Insufficiency, Chronic/complications , Uremia/complications , Animals , Cardiovascular Diseases/prevention & control , Cardiovascular Diseases/therapy , Humans , Uremia/drug therapy , Uremia/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL