Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Nephrol Dial Transplant ; 39(1): 103-113, 2023 Dec 20.
Article in English | MEDLINE | ID: mdl-37349959

ABSTRACT

BACKGROUND: Chronic kidney disease (CKD) is associated with a significant decrease in muscle strength and mass, possibly related to muscle cell damage by uremic toxins. Here, we studied in vitro and in vivo the effect of indoxyl sulfate (IS), an indolic uremic toxin, on myoblast proliferation, differentiation and expression of myogenic regulatory factors (MRF)-myoblast determination protein 1 (MyoD1), myogenin (Myog), Myogenic Factor 5 (Myf5) and myogenic regulatory factor 4 (Myf6/MRF4)-and expression of myosin heavy chain, Myh2. METHODS: C2C12 myoblasts were cultured in vitro and differentiated in myotubes for 7 days in the presence of IS at a uremic concentration of 200 µM. Myocytes morphology and differentiation was analyzed after hematoxylin-eosin staining. MRF genes' expression was studied using reverse transcription polymerase chain reaction in myocytes and 5/6th nephrectomized mice muscle. Myf6/MRF4 protein expression was studied using enzyme-linked immunosorbent assay; MYH2 protein expression was studied using western blotting. The role of Aryl Hydrocarbon Receptor (AHR)-the cell receptor of IS-was studied by adding an AHR inhibitor into the cell culture milieu. RESULTS: In the presence of IS, the myotubes obtained were narrower and had fewer nuclei than control myotubes. The presence of IS during differentiation did not modify the gene expression of the MRFs Myf5, MyoD1 and Myog, but induced a decrease in expression of Myf6/MRF4 and MYH2 at the mRNA and the protein level. AHR inhibition by CH223191 did not reverse the decrease in Myf6/MRF4 mRNA expression induced by IS, which rules out the implication of the ARH genomic pathway. In 5/6th nephrectomized mice, the Myf6/MRF4 gene was down-regulated in striated muscles. CONCLUSION: In conclusion, IS inhibits Myf6/MRF4 and MYH2 expression during differentiation of muscle cells, which could lead to a defect in myotube structure. Through these new mechanisms, IS could participate in muscle atrophy observed in CKD.


Subject(s)
Indican , Renal Insufficiency, Chronic , Animals , Mice , Indican/pharmacology , Down-Regulation , Cell Differentiation/genetics , Muscle, Skeletal , RNA, Messenger
2.
Biomedicines ; 11(2)2023 Jan 27.
Article in English | MEDLINE | ID: mdl-36830910

ABSTRACT

Many hypotheses could explain the mortality decrease observed using hemodiafiltration, such as reduction of intradialytic hypotension and more efficient toxin removal. We led a systematic analysis of representative uremic toxin removal with hemodialysis (HD), online postdilution hemodiafiltration (postHDF) and online predilution hemodiafiltration (preHDF), in a single-center crossover and prospective observational study. The primary outcome was the reduction ratio of uremic toxins of the three categories defined by the Eutox group. Twenty-six patients were treated by those three techniques of extra renal epuration. Mean Kt/Vurea was not different between the treatment methods. Mean reduction ratio of beta2microglobulin was significantly higher for both HDF treatments than for HD (p < 0.001). Myoglobin, kappa, and lambda free light chain reduction ratio was significantly different between the modes: 37.75 ± 11.95%, 45.31 ± 11% and 61.22 ± 10.56%/57.21 ± 12.5%, 63.53 ± 7.93%, and 68.40 ± 11.79%/29.12 ± 8.44%, 34.73 ± 9.01%, and 45.55 ± 12.31% HD, preHDF, and postHDF, respectively (p < 0.001). Mean protein-bound solutes reduction ratio was not different between the different treatments except for PCS with a higher reduction ratio during HDF treatments. Mean albumin loss was always less than 2 g. HDF improved removal of middle molecules but had no effect on indoles concentration without any difference between synthetic dialysis membranes.

3.
Toxins (Basel) ; 14(9)2022 08 27.
Article in English | MEDLINE | ID: mdl-36136527

ABSTRACT

BACKGROUND: Indoxyl sulfate (IS) and p-cresyl sulfate (PCS), two uremic toxins (UTs), are associated with increased mortality in patients with chronic kidney disease (CKD). These toxins are produced by the microbiota from the diet and excreted by the kidney. The purpose of this study was to analyze the effect of diet on IS and PCS concentration in hemodialysis (HD) patients. METHODS: We performed a prospective monocentric study using a seven-day diet record and determination of serum IS and PCS levels in HD patients. We tested the association between toxin concentrations and nutritional data. RESULTS: A total of 58/75 patients (77%) completed the diet record. Mean caloric intake was 22 ± 9.2 kcal/kg/day. The protein/fiber index was 4.9 ± 1.8. No correlation between IS or PCS concentration and protein/fiber index was highlighted. In the 18 anuric patients (31%) in whom residual renal function could not affect toxin concentrations, IS and PCS concentrations were negatively correlated with fiber intake and positively correlated with the protein/fiber index. In a multivariate analysis, IS serum concentration was positively associated with the protein/fiber index (p = 0.03). CONCLUSIONS: A low protein/fiber index is associated with low concentrations of uremic toxins in anuric HD patients. Diets with an increased fiber intake must be tested to determine whether they reduce PCS and IS serum concentrations.


Subject(s)
Renal Insufficiency, Chronic , Toxins, Biological , Uremia , Cresols , Dietary Fiber , Humans , Indican , Prospective Studies , Proteins , Renal Dialysis , Renal Insufficiency, Chronic/therapy , Sulfates , Sulfuric Acid Esters , Uremia/therapy , Uremic Toxins
4.
J Invest Dermatol ; 142(12): 3200-3210.e5, 2022 12.
Article in English | MEDLINE | ID: mdl-35690141

ABSTRACT

CD146 involvement was recently described in skin fibrosis of systemic sclerosis through its regulation of the Wnt pathway. Because the interaction between Wnt and ROS signaling plays a major role in fibrosis, we hypothesized that in systemic sclerosis, CD146 may regulate Wnt/ROS crosstalk. Using a transcriptomic and western blot analysis performed on CD146 wild-type or knockout mouse embryonic fibroblasts, we showed a procanonical Wnt hallmark in the absence of CD146 that is reversed when CD146 expression is restored. We found an elevated ROS content in knockout cells and an increase in DNA oxidative damage in the skin sections of knockout mice compared with those of wild-type mice. We also showed that ROS increased CD146 and its noncanonical Wnt ligand, WNT5A, only in wild-type cells. In humans, fibroblasts from patients with systemic sclerosis presented higher ROS content and expressed CD146, whereas control fibroblasts did not. Moreover, CD146 and its ligand were upregulated by ROS in both human fibroblasts. The increase in bleomycin-induced WNT5A expression was abrogated when CD146 was silenced. We showed an interplay between Wnt and ROS signaling in systemic sclerosis, regulated by CD146, which promotes the noncanonical Wnt pathway and prevents ROS signaling, opening the way for innovative therapeutic strategies.


Subject(s)
Scleroderma, Systemic , Wnt Signaling Pathway , Humans , Animals , Mice , Wnt Signaling Pathway/physiology , CD146 Antigen/genetics , CD146 Antigen/metabolism , Reactive Oxygen Species/metabolism , Ligands , Fibroblasts/metabolism , Scleroderma, Systemic/genetics , Scleroderma, Systemic/metabolism , Fibrosis , Oxidative Stress
5.
Nephrol Dial Transplant ; 37(7): 1249-1260, 2022 06 23.
Article in English | MEDLINE | ID: mdl-35333341

ABSTRACT

BACKGROUND: Myostatin and activin A induce muscle wasting by activating the ubiquitin proteasome system and inhibiting the Akt/mammalian target of rapamycin pathway. In chronic kidney disease (CKD), myostatin and activin A plasma concentrations are increased, but it is unclear if there is increased production or decreased renal clearance. METHODS: We measured myostatin and activin A concentrations in 232 CKD patients and studied their correlation with estimated glomerular filtration rate (eGFR). We analyzed the myostatin gene (MSTN) expression in muscle biopsies of hemodialysis (HD) patients. We then measured circulating myostatin and activin A in plasma and the Mstn and Inhba expression in muscles, kidney, liver and heart of two CKD mice models (adenine and 5/6 nephrectomy models). Finally, we analyzed whether the uremic toxin indoxyl sulfate (IS) increased Mstn expression in mice and cultured muscle cells. RESULTS: In patients, myostatin and activin A were inversely correlated with eGFR. MSTN expression was lower in HD patients' muscles (vastus lateralis) than in controls. In mice with CKD, myostatin and activin A blood concentrations were increased. Mstn was not upregulated in CKD mice tissues. Inha was upregulated in kidney and heart. Exposure to IS did not induce Mstn upregulation in mouse muscles and in cultured myoblasts and myocytes. CONCLUSION: During CKD, myostatin and activin A blood concentrations are increased. Myostatin is not overproduced, suggesting only an impaired renal clearance, but activin A is overproduced in the kidney and heart. We propose to add myostatin and activin A to the list of uremic toxins.


Subject(s)
Myostatin , Renal Insufficiency, Chronic , Activins/metabolism , Animals , Humans , Indican , Mammals/metabolism , Mice , Muscle, Skeletal/metabolism , Myostatin/genetics , Renal Insufficiency, Chronic/pathology
6.
Nephrol Dial Transplant ; 37(12): 2528-2537, 2022 11 23.
Article in English | MEDLINE | ID: mdl-35146525

ABSTRACT

BACKGROUND: Chronic kidney disease (CKD) is a major public health issue associated with increased cardiovascular, infectious and all-cause mortality. The neutrophil:lymphocyte ratio (NLR) is a predictive marker of the risk of death and cardiovascular events. Uremic toxins, notably indoxyl sulfate (IS), are involved in immune deficiency and cardiovascular complications associated with CKD. The aim of this study was to assess whether the NLR was related to uremic toxins and could predict clinical outcome in hemodialysis (HD) patients. METHODS: We conducted a prospective cohort study of 183 patients on chronic HD. The main objective was to study the correlation between the NLR and uremic toxin serum levels. The secondary objective was to test if the NLR can predict the incidence of mortality, cardiovascular events and infectious events. RESULTS: Patients were separated into two groups according to the NLR median value (3.49). The NLR at inclusion was correlated with the NLR at the 6-month (r = 0.55, P < 0.0001) and 12-month (r = 0.62, P < 0.0001) follow-up. Among uremic toxins, IS levels were higher in the group with high NLR (104 µmol/L versus 81 µmol/L; P = 0.004). In multivariate analysis, the NLR remained correlated with IS (P = 0.03). The incidence of death, cardiovascular events and severe infectious events was higher in the group with high NLR [respectively, 38% versus 18% (P = 0.004), 45% versus 26% (P = 0.01) and 33% versus 21% (P = 0.02)] than in the low NLR group. Multivariate analysis showed an independent association of the NLR with mortality (P = 0.02) and cardiovascular events (P = 0.03) but not with severe infectious events. CONCLUSIONS: In HD patients, the NLR predicted mortality and cardiovascular events but not severe infections and correlated positively with the level of the uremic toxin IS. The NLR could be an interesting marker for monitoring the risk of clinical events in CKD patients.


Subject(s)
Cardiovascular Diseases , Renal Insufficiency, Chronic , Toxins, Biological , Humans , Indican , Neutrophils , Uremic Toxins , Prospective Studies , Renal Dialysis/adverse effects , Lymphocytes , Renal Insufficiency, Chronic/complications , Biomarkers
7.
Kidney Int ; 99(5): 1062-1064, 2021 05.
Article in English | MEDLINE | ID: mdl-33892855

ABSTRACT

Sodium-glucose cotransporter 2 inhibitors offer cardiovascular and renal benefits in patients with chronic kidney disease through not yet clearly defined mechanisms. Juni et al. showed that sodium-glucose cotransporter 2 inhibitor empagliflozin exposure in vitro can restore cardiomyocyte function by counteracting harmful effects of uremic serum on the endothelium-cardiomyocyte crosstalk between endothelial cells and cardiomyocytes. The author's findings improved our understanding of cardiovascular impairment in chronic kidney disease and provided new perspectives for the beneficial effects of sodium-glucose cotransporter 2 inhibitor therapy.


Subject(s)
Cardio-Renal Syndrome , Renal Insufficiency, Chronic , Sodium-Glucose Transporter 2 Inhibitors , Humans , Benzhydryl Compounds/therapeutic use , Cardio-Renal Syndrome/drug therapy , Endothelial Cells , Endothelium , Glucosides , Myocytes, Cardiac , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/drug therapy , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use
8.
Int J Mol Sci ; 21(7)2020 Mar 31.
Article in English | MEDLINE | ID: mdl-32244284

ABSTRACT

Endogenous agonists of the transcription factor aryl hydrocarbon receptor (AHR) such as the indolic uremic toxin, indoxyl sulfate (IS), accumulate in patients with chronic kidney disease. AHR activation by indolic toxins has prothrombotic effects on the endothelium, especially via tissue factor (TF) induction. In contrast, physiological AHR activation by laminar shear stress (SS) is atheroprotective. We studied the activation of AHR and the regulation of TF by IS in cultured human umbilical vein endothelial cells subjected to laminar fluid SS (5 dynes/cm2). SS and IS markedly increased the expression of AHR target genes PTGS2 (encoding for COX2), AHRR, CYP1A1, and CYP1B1, as well as F3 (encoding for TF), in an AHR-dependent way. IS amplified SS-induced TF mRNA and protein expression and upregulation of AHR target genes. Interestingly, tyrosine kinase inhibition by genistein decreased SS- but not IS-induced TF expression. Finally, the increase in TF expression induced by laminar SS was not associated with increased TF activity. In contrast, IS increased TF activity, even under antithrombotic SS conditions. In conclusion, IS and SS induce AHR activation and AHR-dependent TF upregulation by different mechanisms. Impairment of the antithrombotic properties of shear stressed endothelium by toxic AHR agonists could favor cardiovascular diseases in CKD.


Subject(s)
Human Umbilical Vein Endothelial Cells/metabolism , Indican/agonists , Indican/toxicity , Receptors, Aryl Hydrocarbon/drug effects , Receptors, Aryl Hydrocarbon/metabolism , Thromboplastin/drug effects , Thromboplastin/metabolism , Basic Helix-Loop-Helix Transcription Factors , Cyclooxygenase 2/genetics , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1B1/genetics , Gene Expression Regulation , Gene Knockdown Techniques , Humans , RNA, Messenger/metabolism , Renal Insufficiency, Chronic/metabolism , Stress, Mechanical
9.
Toxins (Basel) ; 11(10)2019 10 05.
Article in English | MEDLINE | ID: mdl-31590361

ABSTRACT

Alterations of renal endothelial cells play a crucial role in the initiation and progression of diabetic kidney disease. High glucose per se, as well as glucose by-products, induce endothelial dysfunction in both large vessels and the microvasculature. Toxic glucose by-products include advanced glycation end products (AGEs), a group of modified proteins and/or lipids that become glycated after exposure to sugars, and glucose metabolites produced via the polyol pathway. These glucose-related endothelio-toxins notably induce an alteration of the glomerular filtration barrier by increasing the permeability of glomerular endothelial cells, altering endothelial glycocalyx, and finally, inducing endothelial cell apoptosis. The glomerular endothelial dysfunction results in albuminuria. In addition, high glucose and by-products impair the endothelial repair capacities by reducing the number and function of endothelial progenitor cells. In this review, we summarize the mechanisms of renal endothelial toxicity of high glucose/glucose by-products, which encompass changes in synthesis of growth factors like TGF-ß and VEGF, induction of oxidative stress and inflammation, and reduction of NO bioavailability. We finally present potential therapies to reduce endothelial dysfunction in diabetic kidney disease.


Subject(s)
Diabetic Nephropathies/metabolism , Endothelial Cells/metabolism , Glucose/metabolism , Animals , Humans , Kidney/cytology , Kidney/metabolism
10.
Nat Rev Nephrol ; 15(2): 87-108, 2019 02.
Article in English | MEDLINE | ID: mdl-30607032

ABSTRACT

The kidney harbours different types of endothelia, each with specific structural and functional characteristics. The glomerular endothelium, which is highly fenestrated and covered by a rich glycocalyx, participates in the sieving properties of the glomerular filtration barrier and in the maintenance of podocyte structure. The microvascular endothelium in peritubular capillaries, which is also fenestrated, transports reabsorbed components and participates in epithelial cell function. The endothelium of large and small vessels supports the renal vasculature. These renal endothelia are protected by regulators of thrombosis, inflammation and complement, but endothelial injury (for example, induced by toxins, antibodies, immune cells or inflammatory cytokines) or defects in factors that provide endothelial protection (for example, regulators of complement or angiogenesis) can lead to acute or chronic renal injury. Moreover, renal endothelial cells can transition towards a mesenchymal phenotype, favouring renal fibrosis and the development of chronic kidney disease. Thus, the renal endothelium is both a target and a driver of kidney and systemic cardiovascular complications. Emerging therapeutic strategies that target the renal endothelium may lead to improved outcomes for both rare and common renal diseases.


Subject(s)
Endothelium , Kidney Diseases/pathology , Kidney Diseases/physiopathology , Kidney , Endothelial Cells/pathology , Endothelial Cells/physiology , Endothelium/anatomy & histology , Endothelium/pathology , Endothelium/physiology , Endothelium/physiopathology , Humans , Kidney/anatomy & histology , Kidney/pathology , Kidney/physiology , Kidney/physiopathology , Kidney Diseases/therapy
11.
Arch Toxicol ; 93(1): 121-136, 2019 01.
Article in English | MEDLINE | ID: mdl-30324315

ABSTRACT

Chronic kidney disease (CKD) is associated with high risk of thrombosis. Indole-3 acetic acid (IAA), an indolic uremic toxin, induces the expression of tissue factor (TF) in human umbilical vein endothelial cells (HUVEC) via the transcription factor aryl hydrocarbon receptor (AhR). This study aimed to understand the signaling pathways involved in AhR-mediated TF induction by IAA. We incubated human endothelial cells with IAA at 50 µM, the maximal concentration found in patients with CKD. IAA induced TF expression in different types of human endothelial cells: umbilical vein (HUVEC), aortic (HAoEC), and cardiac-derived microvascular (HMVEC-C). Using AhR inhibition and chromatin immunoprecipitation experiments, we showed that TF induction by IAA in HUVEC was controlled by AhR and that AhR did not bind to the TF promoter. The analysis of TF promoter activity using luciferase reporter plasmids showed that the NF-κB site was essential in TF induction by IAA. In addition, TF induction by IAA was drastically decreased by an inhibitor of the NF-κB pathway. IAA induced the nuclear translocation of NF-κB p50 subunit, which was decreased by AhR and p38MAPK inhibition. Finally, in a cohort of 92 CKD patients on hemodialysis, circulating TF was independently related to serum IAA in multivariate analysis. In conclusion, TF up-regulation by IAA in human endothelial cells involves a non-genomic AhR/p38 MAPK/NF-κB pathway. The understanding of signal transduction pathways related to AhR thrombotic/inflammatory pathway is of interest to find therapeutic targets to reduce TF expression and thrombotic risk in patients with CKD.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Indoleacetic Acids/toxicity , NF-kappa B/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction , Thromboplastin/metabolism , Adult , Aged , Aged, 80 and over , Basic Helix-Loop-Helix Transcription Factors/genetics , Cells, Cultured , Female , Gene Knockdown Techniques , Humans , Male , Middle Aged , Promoter Regions, Genetic , Prospective Studies , RNA, Small Interfering , Receptors, Aryl Hydrocarbon/genetics , Renal Insufficiency, Chronic , Young Adult
12.
Toxins (Basel) ; 10(10)2018 10 12.
Article in English | MEDLINE | ID: mdl-30322010

ABSTRACT

Patients with chronic kidney disease (CKD) display an elevated risk of thrombosis. Thrombosis occurs in cardiovascular events, such as venous thromboembolism, stroke, and acute coronary syndrome, and is a cause of hemodialysis vascular access dysfunction. CKD leads to the accumulation of uremic toxins, which exerts toxic effects on blood and the vessel wall. Some uremic toxins result from tryptophan metabolization in the gut through the indolic and the kynurenine pathways. An increasing number of studies are highlighting the link between such uremic toxins and thrombosis in CKD. In this review, we describe the thrombotic mechanisms induced by tryptophan-derived uremic toxins (TDUT). These mechanisms include an increase in plasma levels of procoagulant factors, induction of platelet hyperactivity, induction of endothelial dysfunction/ impairment of endothelial healing, decrease in nitric oxide (NO) bioavailability, and production of procoagulant microparticles. We focus on one important prothrombotic mechanism: The induction of tissue factor (TF), the initiator of the extrinsic pathway of the blood coagulation. This induction occurs via a new pathway, dependent on the transcription factor Aryl hydrocarbon receptor (AhR), the receptor of TDUT in cells. A better understanding of the prothrombotic mechanisms of uremic toxins could help to find novel therapeutic targets to prevent thrombosis in CKD.


Subject(s)
Renal Insufficiency, Chronic/metabolism , Thrombosis/metabolism , Toxins, Biological/metabolism , Tryptophan/metabolism , Uremia/metabolism , Animals , Diet , Humans , Microbiota
13.
Kidney Int ; 93(4): 986-999, 2018 04.
Article in English | MEDLINE | ID: mdl-29395338

ABSTRACT

Patients with chronic kidney disease (CKD) are exposed to uremic toxins and have an increased risk of cardiovascular disease. Some uremic toxins, like indoxyl sulfate, are agonists of the transcription factor aryl hydrocarbon receptor (AHR). These toxins induce a vascular procoagulant phenotype. Here we investigated AHR activation in patients with CKD and in a murine model of CKD. We performed a prospective study in 116 patients with CKD stage 3 to 5D and measured the AHR-Activating Potential of serum by bioassay. Compared to sera from healthy controls, sera from CKD patients displayed a strong AHR-Activating Potential; strongly correlated with eGFR and with the indoxyl sulfate concentration. The expression of the AHR target genes Cyp1A1 and AHRR was up-regulated in whole blood from patients with CKD. Survival analyses revealed that cardiovascular events were more frequent in CKD patients with an AHR-Activating Potential above the median. In mice with 5/6 nephrectomy, there was an increased serum AHR-Activating Potential, and an induction of Cyp1a1 mRNA in the aorta and heart, absent in AhR-/- CKD mice. After serial indoxyl sulfate injections, we observed an increase in serum AHR-AP and in expression of Cyp1a1 mRNA in aorta and heart in WT mice, but not in AhR-/- mice. Thus, the AHR pathway is activated both in patients and mice with CKD. Hence, AHR activation could be a key mechanism involved in the deleterious cardiovascular effects observed in CKD.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/blood , Receptors, Aryl Hydrocarbon/blood , Renal Insufficiency, Chronic/blood , Adult , Aged , Aged, 80 and over , Animals , Basic Helix-Loop-Helix Transcription Factors/agonists , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cardiovascular Diseases/blood , Cardiovascular Diseases/mortality , Case-Control Studies , Cause of Death , Cell Line, Tumor , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Disease Models, Animal , Female , Humans , Indican/administration & dosage , Indican/blood , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Prospective Studies , Receptors, Aryl Hydrocarbon/agonists , Receptors, Aryl Hydrocarbon/deficiency , Receptors, Aryl Hydrocarbon/genetics , Renal Dialysis , Renal Insufficiency, Chronic/diagnosis , Renal Insufficiency, Chronic/mortality , Renal Insufficiency, Chronic/therapy , Repressor Proteins/genetics , Repressor Proteins/metabolism , Risk Factors , Treatment Outcome
14.
Kidney Int ; 89(3): 532-4, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26880447

ABSTRACT

Patients with chronic kidney disease display an impairment of neovascularization in ischemic tissues. Studies have suggested the involvement of the uremic toxin indoxyl sulfate by demonstrating that indoxyl sulfate affects endothelial progenitor cells. However, few data are available on the effects of indoxyl sulfate on neovascularization and on the mechanisms involved. The article by Hung et al. shows that indoxyl sulfate suppresses neovascularization in uremic mice by impairing endothelial progenitor cell function via the inhibition of hypoxia-induced hypoxia-inducible factor/interleukin-10/vascular endothelial growth factor signaling.


Subject(s)
Indican/metabolism , Vascular Endothelial Growth Factor A , Animals , Humans , Interleukin-10 , Renal Insufficiency, Chronic , Signal Transduction/drug effects
15.
Nephron ; 131(3): 167-74, 2015.
Article in English | MEDLINE | ID: mdl-26426087

ABSTRACT

BACKGROUND: Chronic kidney disease (CKD) is associated with increased cardiovascular morbidity and mortality. Oxidative stress seems to play a pivotal role in this process, and purine metabolism may be involved in CKD-related oxidative stress. Xanthine oxidase (XO) is an enzyme involved in purine metabolism and is also responsible for the production of reactive oxygen species. METHODS: This prospective study aimed to analyze the relation between plasma dosages of molecules involved in redox balance, purine metabolism and cardiovascular events in patients with non-diabetic CKD stages 3-5 or on chronic hemodialysis (HD). CKD (n = 51) and HD (n = 50) patients were compared to matched healthy controls (n = 38) and followed-up for 3 years. RESULTS: Both CKD and HD patients had decreased plasma levels of antioxidants (selenium, zinc, vitamin C). HD patients had decreased levels of the antioxidant enzyme superoxide dismutase and increased levels of oxidation products (ischemia-modified albumin, malondialdehyde [MDA]). The following substrates and enzymes involved in purine metabolism were increased in the HD cohort: adenosine, adenosine deaminase and the pro-oxidant XO. XO activity was negatively correlated with super oxide dismutase and positively with MDA. Interestingly, XO activity was an independent predictor of cardiovascular events in CKD and HD patients, regardless of uric acid levels. Uric acid was not predictive of events. CONCLUSION: This highlights a possible role of XO itself in CKD-related cardiovascular disease (CVD) and raises the hypothesis that beneficial effects observed with XO inhibitors on CVD in CKD may also be due to the reduction of oxidative stress.


Subject(s)
Cardiovascular Diseases/blood , Cardiovascular Diseases/etiology , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/complications , Uric Acid/blood , Xanthine Oxidase/blood , Adult , Aged , Aged, 80 and over , Cardiovascular Diseases/mortality , Female , Follow-Up Studies , Humans , Kaplan-Meier Estimate , Longitudinal Studies , Male , Middle Aged , Oxidation-Reduction , Oxidative Stress , Predictive Value of Tests , Prospective Studies , Purines/metabolism , Renal Dialysis , Renal Insufficiency, Chronic/mortality , Risk Factors
16.
J Am Soc Nephrol ; 26(4): 876-87, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25145928

ABSTRACT

In CKD, uremic solutes may induce endothelial dysfunction, inflammation, and oxidative stress, leading to increased cardiovascular risk. We investigated whether the uremic solute indole-3 acetic acid (IAA) predicts clinical outcomes in patients with CKD and has prooxidant and proinflammatory effects. We studied 120 patients with CKD. During the median study period of 966 days, 29 patients died and 35 experienced a major cardiovascular event. Kaplan-Meier analysis revealed that mortality and cardiovascular events were significantly higher in the higher IAA group (IAA>3.73 µM) than in the lower IAA group (IAA<3.73 µM). Multivariate Cox regression analysis demonstrated that serum IAA was a significant predictor of mortality and cardiovascular events after adjustments for age and sex; cholesterol, systolic BP, and smoking; C-reactive protein, phosphate, body mass index, and albumin; diastolic BP and history of cardiovascular disease; and uremic toxins p-cresyl sulfate and indoxyl sulfate. Notably, IAA level remained predictive of mortality when adjusted for CKD stage. IAA levels were positively correlated with markers of inflammation and oxidative stress: C-reactive protein and malondialdehyde, respectively. In cultured human endothelial cells, IAA activated an inflammatory nongenomic aryl hydrocarbon receptor (AhR)/p38MAPK/NF-κB pathway that induced the proinflammatory enzyme cyclooxygenase-2. Additionally, IAA increased production of endothelial reactive oxygen species. In conclusion, serum IAA may be an independent predictor of mortality and cardiovascular events in patients with CKD. In vitro, IAA induces endothelial inflammation and oxidative stress and activates an inflammatory AhR/p38MAPK/NF-κB pathway.


Subject(s)
Cardiovascular Diseases/blood , Endothelium, Vascular/metabolism , Indoleacetic Acids/blood , Oxidative Stress , Renal Insufficiency, Chronic/blood , Adult , Aged , Aged, 80 and over , Cardiovascular Diseases/etiology , Cyclooxygenase 2/metabolism , Female , Human Umbilical Vein Endothelial Cells , Humans , Male , Middle Aged , Reactive Oxygen Species/metabolism , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/mortality , Signal Transduction , Uremia/complications , Young Adult
17.
Toxins (Basel) ; 6(3): 934-49, 2014 Mar 04.
Article in English | MEDLINE | ID: mdl-24599232

ABSTRACT

Patients with chronic kidney disease (CKD) have a higher risk of cardiovascular diseases and suffer from accelerated atherosclerosis. CKD patients are permanently exposed to uremic toxins, making them good candidates as pathogenic agents. We focus here on uremic toxins from tryptophan metabolism because of their potential involvement in cardiovascular toxicity: indolic uremic toxins (indoxyl sulfate, indole-3 acetic acid, and indoxyl-ß-d-glucuronide) and uremic toxins from the kynurenine pathway (kynurenine, kynurenic acid, anthranilic acid, 3-hydroxykynurenine, 3-hydroxyanthranilic acid, and quinolinic acid). Uremic toxins derived from tryptophan are endogenous ligands of the transcription factor aryl hydrocarbon receptor (AhR). AhR, also known as the dioxin receptor, interacts with various regulatory and signaling proteins, including protein kinases and phosphatases, and Nuclear Factor-Kappa-B. AhR activation by 2,3,7,8-tetrachlorodibenzo-p-dioxin and some polychlorinated biphenyls is associated with an increase in cardiovascular disease in humans and in mice. In addition, this AhR activation mediates cardiotoxicity, vascular inflammation, and a procoagulant and prooxidant phenotype of vascular cells. Uremic toxins derived from tryptophan have prooxidant, proinflammatory, procoagulant, and pro-apoptotic effects on cells involved in the cardiovascular system, and some of them are related with cardiovascular complications in CKD. We discuss here how the cardiovascular effects of these uremic toxins could be mediated by AhR activation, in a "dioxin-like" effect.


Subject(s)
Cardiovascular Diseases/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Renal Insufficiency, Chronic/metabolism , Toxins, Biological/metabolism , Tryptophan/metabolism , Uremia/metabolism , Animals , Cardiovascular Diseases/etiology , Endothelium, Vascular/physiopathology , Humans , Leukocytes/immunology , Oxidative Stress , Renal Insufficiency, Chronic/complications
18.
Kidney Int ; 84(4): 733-44, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23636172

ABSTRACT

In chronic kidney disease (CKD), uremic solutes accumulate in blood and tissues. These compounds probably contribute to the marked increase in cardiovascular risk during the progression of CKD. The uremic solutes indoxyl sulfate and indole-3-acetic acid (IAA) are particularly deleterious for endothelial cells. Here we performed microarray and comparative PCR analyses to identify genes in endothelial cells targeted by these two uremic solutes. We found an increase in endothelial expression of tissue factor in response to indoxyl sulfate and IAA and upregulation of eight genes regulated by the transcription factor aryl hydrocarbon receptor (AHR). The suggestion by microarray analysis of an involvement of AHR in tissue factor production was confirmed by siRNA inhibition and the indirect AHR inhibitor geldanamycin. These observations were extended to peripheral blood mononuclear cells. Tissue factor expression and activity were also increased by AHR agonist dioxin. Finally, we measured circulating tissue factor concentration and activity in healthy control subjects and in patients with CKD (stages 3-5d), and found that each was elevated in patients with CKD. Circulating tissue factor levels were positively correlated with plasma indoxyl sulfate and IAA. Thus, indolic uremic solutes increase tissue factor production in endothelial and peripheral blood mononuclear cells by AHR activation, evoking a 'dioxin-like' effect. This newly described mechanism of uremic solute toxicity may help understand the high cardiovascular risk of CKD patients.


Subject(s)
Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Indican/pharmacology , Indoleacetic Acids/pharmacology , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction/drug effects , Thromboplastin/metabolism , Adult , Aged , Aged, 80 and over , Benzoquinones/pharmacology , Case-Control Studies , Cells, Cultured , Dioxins/pharmacology , Endothelium, Vascular/cytology , Female , Humans , In Vitro Techniques , Indican/metabolism , Indoleacetic Acids/metabolism , Lactams, Macrocyclic/pharmacology , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Polymerase Chain Reaction , RNA, Small Interfering/pharmacology , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/drug effects , Renal Insufficiency, Chronic/metabolism , Signal Transduction/physiology , Tissue Array Analysis , Umbilical Veins/cytology , Umbilical Veins/drug effects , Umbilical Veins/metabolism , Up-Regulation/drug effects , Up-Regulation/physiology
19.
J Chromatogr B Analyt Technol Biomed Life Sci ; 879(23): 2281-6, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21727042

ABSTRACT

During chronic kidney disease (CKD), solutes called uremic solutes, accumulate in blood and tissues of patients. We developed an HPLC method for the simultaneous determination of several uremic solutes of clinical interest in biological fluids: phenol (Pol), indole-3-acetic acid (3-IAA), p-cresol (p-C), indoxyl sulfate (3-INDS) and p-cresol sulfate (p-CS). These solutes were separated by ion-pairing HPLC using an isocratic flow and quantified with a fluorescence detection. The mean serum concentrations of 3-IAA, 3-INDS and p-CS were 2.12, 1.03 and 13.03 µM respectively in healthy subjects, 3.21, 17.45 and 73.47 µM in non hemodialyzed stage 3-5 CKD patients and 5.9, 81.04 and 120.54 µM in hemodialyzed patients (stage 5D). We found no Pol and no p-C in any population. The limits of quantification for 3-IAA, 3-INDS, and p-CS were 0.83, 0.72, and 3.2 µM respectively. The within-day CVs were between 1.23 and 3.12% for 3-IAA, 0.98 and 2% for 3-INDS, and 1.25 and 3.01% for p-CS. The between-day CVs were between 1.78 and 5.48% for 3-IAA, 1.45 and 4.54% for 3-INDS, and 1.19 and 6.36% for p-CS. This HPLC method permits the simultaneous and quick quantification of several uremic solutes for daily analysis of large numbers of samples.


Subject(s)
Chromatography, High Pressure Liquid/methods , Cresols/blood , Indican/blood , Indoleacetic Acids/blood , Kidney Failure, Chronic/blood , Phenol/blood , Phenols/blood , Sulfuric Acid Esters/blood , Uremia/blood , Aged , Female , Humans , Male , Middle Aged
20.
Semin Dial ; 24(3): 327-37, 2011.
Article in English | MEDLINE | ID: mdl-21682773

ABSTRACT

Patients with chronic kidney disease (CKD) have a much higher risk of cardiovascular diseases than the general population. Endothelial dysfunction, which participates in accelerated atherosclerosis, is a hallmark of CKD. Patients with CKD display impaired endothelium-dependent vasodilatation, elevated soluble biomarkers of endothelial dysfunction, and increased oxidative stress. They also present an imbalance between circulating endothelial populations reflecting endothelial injury (endothelial microparticles and circulating endothelial cells) and repair (endothelial progenitor cells). Endothelial damage induced by a uremic environment suggests an involvement of uremia-specific factors. Several uremic toxins, mostly protein-bound, have been shown to have specific endothelial toxicity: ADMA, homocysteine, AGEs, and more recently, p-cresyl sulfate and indoxyl sulfate. These toxins, all poorly removed by hemodialysis therapies, share mechanisms of endothelial toxicity: they promote pro-oxidant and pro-inflammatory response and inhibit endothelial repair. This article (i) reviews the evidence for endothelial dysfunction in CKD, (ii) specifies the involvement of protein-bound uremic toxins in this dysfunction, and (iii) discusses therapeutic strategies for lowering uremic toxin concentrations or for countering the effects of uremic toxins on the endothelium.


Subject(s)
Endothelium, Vascular/physiopathology , Renal Dialysis , Toxins, Biological/blood , Uremia/blood , Angiotensin Receptor Antagonists/therapeutic use , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Antioxidants/therapeutic use , Arginine/analogs & derivatives , Arginine/blood , Carbon/therapeutic use , Cell-Derived Microparticles/metabolism , Cresols/blood , Endothelial Cells/metabolism , Erythropoietin/therapeutic use , Glycation End Products, Advanced/blood , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hyperhomocysteinemia/physiopathology , Hyperhomocysteinemia/prevention & control , Indican/blood , Indoleacetic Acids/blood , Kidney Diseases/physiopathology , Oxides/therapeutic use , Protein Binding , Recombinant Proteins , Sulfuric Acid Esters/blood , Surface-Active Agents/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...