Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.228
Filter
Add more filters

Publication year range
1.
Cell Mol Biol (Noisy-le-grand) ; 70(5): 209-213, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38814214

ABSTRACT

Uremia (UR) is a terminal renal failure manifestation with a very high risk of death, high-flux hemodialysis (HFHD) is currently the most common treatment for UR in clinical practice. In this study, we analysed the therapeutic efficacy of HFHD plus Compound-α Ketoacid Tablets for UR under humanistic care. Firstly, we randomised 100 patients with UR into a research group (RG) for HFHD plus Compound-α Ketoacid Tablets therapy and a control group (CG) for HFHD treatment, with both therapies implemented under humanistic care. By way of comparison, we found that the study group had significantly better renal function after treatment and they had a lower inflammatory response. Also, the study group showed lower calcium and phosphorus metabolism and better immune function. Based on these results, we believe that HFHD + Compound-α Ketoacid Tablets under humanistic care have high clinical value.


Subject(s)
Calcium , Phosphorus , Renal Dialysis , Tablets , Uremia , Humans , Renal Dialysis/methods , Uremia/therapy , Uremia/metabolism , Female , Male , Calcium/metabolism , Middle Aged , Aged , Adult
2.
J Biochem Mol Toxicol ; 38(8): e23785, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39051181

ABSTRACT

An arteriovenous fistula (AVF) is the preferred vascular access for hemodialysis in uremic patients, yet its dysfunction poses a significant clinical challenge. Venous stenosis, primarily caused by venous neointimal hyperplasia, is a key factor in the failure of vascular access. During vascular access dysfunction, endothelial cells (ECs) transform mechanical stimuli into intracellular signals and interact with vascular smooth muscle cells. Tanshinone IIA, an important compound derived from Salvia miltiorrhiza, has been widely used to treat cardiovascular diseases. However, its role in modulating ECs under uremic conditions remains incompletely understood. In this research, ECs were exposed to sodium tanshinone IIA sulfonate (STS) and subjected to shear stress and uremic conditions. The results indicate that STS can reduce the suppressive effects on the expression of NF-κB p65, JNK and Collagen I in uremia-induced ECs. Moreover, the downregulation of NF-κB p65, JNK and Collagen I can be enhanced through the inhibition of ERK1/2 and the upregulation of Caveolin-1. These findings suggest that tanshinone IIA may improve EC function under uremic conditions by targeting the Caveolin-1/ERK1/2 pathway, presenting tanshinone IIA as a potential therapeutic agent against AVF immaturity caused by EC dysfunction.


Subject(s)
Abietanes , Caveolin 1 , Uremia , Uremia/metabolism , Uremia/drug therapy , Uremia/pathology , Humans , Abietanes/pharmacology , Abietanes/therapeutic use , Caveolin 1/metabolism , MAP Kinase Signaling System/drug effects , Collagen Type I/metabolism , Transcription Factor RelA/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Phenanthrenes
3.
Acta Pharmacol Sin ; 45(7): 1406-1424, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38589687

ABSTRACT

Acute kidney injury (AKI) is often accompanied by uremic encephalopathy resulting from accumulation of uremic toxins in brain possibly due to impaired blood-brain barrier (BBB) function. Anionic uremic toxins are substrates or inhibitors of organic anionic transporters (OATs). In this study we investigated the CNS behaviors and expression/function of BBB OAT3 in AKI rats and mice, which received intraperitoneal injection of cisplatin 8 and 20 mg/kg, respectively. We showed that cisplatin treatment significantly inhibited the expressions of OAT3, synaptophysin and microtubule-associated protein 2 (MAP2), impaired locomotor and exploration activities, and increased accumulation of uremic toxins in the brain of AKI rats and mice. In vitro studies showed that uremic toxins neither alter OAT3 expression in human cerebral microvascular endothelial cells, nor synaptophysin and MAP2 expressions in human neuroblastoma (SH-SY5Y) cells. In contrast, tumour necrosis factor alpha (TNFα) and the conditioned medium (CM) from RAW264.7 cells treated with indoxyl sulfate (IS) significantly impaired OAT3 expression. TNFα and CM from IS-treated BV-2 cells also inhibited synaptophysin and MAP2 expressions in SH-SY5Y cells. The alterations caused by TNFα and CMs in vitro, and by AKI and TNFα in vivo were abolished by infliximab, a monoclonal antibody designed to intercept and neutralize TNFα, suggesting that AKI impaired the expressions of OAT3, synaptophysin and MAP2 in the brain via IS-induced TNFα release from macrophages or microglia (termed as IS-TNFα axis). Treatment of mice with TNFα (0.5 mg·kg-1·d-1, i.p. for 3 days) significantly increased p-p65 expression and reduced the expressions of Nrf2 and HO-1. Inhibiting NF-κB pathway, silencing p65, or activating Nrf2 and HO-1 obviously attenuated TNFα-induced downregulation of OAT3, synaptophysin and MAP2 expressions. Significantly increased p-p65 and decreased Nrf2 and HO-1 protein levels were also detected in brain of AKI mice and rats. We conclude that AKI inhibits the expressions of OAT3, synaptophysin and MAP2 due to IS-induced TNFα release from macrophages or microglia. TNFα impairs the expressions of OAT3, synaptophysin and MAP2 partly via activating NF-κB pathway and inhibiting Nrf2-HO-1 pathway.


Subject(s)
Acute Kidney Injury , Cisplatin , Indican , Tumor Necrosis Factor-alpha , Animals , Acute Kidney Injury/metabolism , Tumor Necrosis Factor-alpha/metabolism , Humans , Mice , Male , RAW 264.7 Cells , Rats , Mice, Inbred C57BL , Organic Anion Transporters, Sodium-Independent/metabolism , Rats, Sprague-Dawley , Synaptophysin/metabolism , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Uremia/metabolism , Uremia/complications , Cell Line, Tumor
4.
J Am Soc Nephrol ; 33(10): 1841-1856, 2022 10.
Article in English | MEDLINE | ID: mdl-36038265

ABSTRACT

BACKGROUND: Bleeding diatheses, common among patients with ESKD, can lead to serious complications, particularly during invasive procedures. Chronic urea overload significantly increases cyanate concentrations in patients with ESKD, leading to carbamylation, an irreversible modification of proteins and peptides. METHODS: To investigate carbamylation as a potential mechanistic link between uremia and platelet dysfunction in ESKD, we used liquid chromatography-tandem mass spectrometry (HPLC-MS/MS) to quantify total homocitrulline, and biotin-conjugated phenylglyoxal labeling and Western blot to detect carbamylated integrin α IIb ß 3 (a receptor required for platelet aggregation). Flow cytometry was used to study activation of isolated platelets and platelet-rich plasma. In a transient transfection system, we tested activity and fibrinogen binding of different mutated forms of the receptor. We assessed platelet adhesion and aggregation in microplate assays. RESULTS: Carbamylation inhibited platelet activation, adhesion, and aggregation. Patients on hemodialysis exhibited significantly reduced activation of α IIb ß 3 compared with healthy controls. We found significant carbamylation of both subunits of α IIb ß 3 on platelets from patients receiving hemodialysis versus only minor modification in controls. In the transient transfection system, modification of lysine 185 in the ß 3 subunit was associated with loss of receptor activity and fibrinogen binding. Supplementation of free amino acids, which was shown to protect plasma proteins from carbamylation-induced damage in patients on hemodialysis, prevented loss of α IIb ß 3 activity in vitro. CONCLUSIONS: Carbamylation of α IIb ß 3-specifically modification of the K185 residue-might represent a mechanistic link between uremia and dysfunctional primary hemostasis in patients on hemodialysis. The observation that free amino acids prevented the carbamylation-induced loss of α IIb ß 3 activity suggests amino acid administration during dialysis may help to normalize platelet function.


Subject(s)
Platelet Glycoprotein GPIIb-IIIa Complex , Uremia , Humans , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Protein Carbamylation , Tandem Mass Spectrometry , Blood Platelets , Uremia/complications , Uremia/metabolism , Fibrinogen/chemistry , Fibrinogen/metabolism , Amino Acids
5.
Int J Mol Sci ; 24(8)2023 Apr 18.
Article in English | MEDLINE | ID: mdl-37108613

ABSTRACT

Chronic kidney disease is the gradual progression of kidney dysfunction and involves numerous co-morbidities, one of the leading causes of mortality. One of the primary complications of kidney dysfunction is the accumulation of toxins in the bloodstream, particularly protein-bound uremic toxins (PBUTs), which have a high affinity for plasma proteins. The buildup of PBUTs in the blood reduces the effectiveness of conventional treatments, such as hemodialysis. Moreover, PBUTs can bind to blood plasma proteins, such as human serum albumin, alter their conformational structure, block binding sites for other valuable endogenous or exogenous substances, and exacerbate the co-existing medical conditions associated with kidney disease. The inadequacy of hemodialysis in clearing PBUTs underscores the significance of researching the binding mechanisms of these toxins with blood proteins, with a critical analysis of the methods used to obtain this information. Here, we gathered the available data on the binding of indoxyl sulfate, p-cresyl sulfate, indole 3-acetic acid, hippuric acid, 3-carboxyl-4-methyl-5-propyl-2-furan propanoic acid, and phenylacetic acid to human serum albumin and reviewed the common techniques used to investigate the thermodynamics and structure of the PBUT-albumin interaction. These findings can be critical in investigating molecules that can displace toxins on HSA and improve their clearance by standard dialysis or designing adsorbents with greater affinity for PBUTs than HSA.


Subject(s)
Toxins, Biological , Uremia , Humans , Serum Albumin, Human/metabolism , Uremic Toxins , Renal Dialysis/adverse effects , Uremia/metabolism , Protein Binding , Blood Proteins/metabolism , Toxins, Biological/metabolism
6.
Kidney Int ; 101(2): 227-241, 2022 02.
Article in English | MEDLINE | ID: mdl-34736971

ABSTRACT

Uremic encephalopathy encompasses a wide range of central nervous system abnormalities associated with poor kidney function occurring with either progressive chronic kidney disease or acute kidney injury. The syndrome is likely caused by retention of uremic solutes, alterations in hormonal metabolism, changes in electrolyte and acid-base homeostasis, as well as changes in vascular reactivity, blood-brain barrier transport, and inflammation. There are no defining clinical, laboratory, or imaging findings, and the diagnosis is often made retrospectively when symptoms improve after dialysis or transplantation. The diagnosis is also made difficult because of the many confounding and overlapping conditions seen in patients with chronic kidney disease and acute kidney injury. Thus, institution of kidney replacement therapy should be considered as a trial to improve symptoms in the right clinical context. Neurological symptoms that do not improve after improvement in clearance should prompt a search for other explanations. Further knowledge linking possible uremic retention solutes with neurological symptoms is needed to better understand this syndrome as well as to develop more tailored treatments that aim to improve cognitive function.


Subject(s)
Brain Diseases , Renal Insufficiency, Chronic , Uremia , Brain Diseases/complications , Humans , Renal Dialysis , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/diagnosis , Renal Insufficiency, Chronic/therapy , Retrospective Studies , Uremia/complications , Uremia/metabolism , Uremia/therapy
7.
FASEB J ; 35(8): e21761, 2021 08.
Article in English | MEDLINE | ID: mdl-34245616

ABSTRACT

Uremic cardiomyopathy is a common complication in chronic kidney disease (CKD) patients, accounting for a high mortality rate. Several mechanisms have been proposed to link CKD and cardiac alterations; however, the early cardiac modifications that occur in CKD that may trigger cardiac remodeling and dysfunction remain largely unexplored. Here, in a mouse model of CKD induced by 5/6 nephrectomy, we first analyzed the early transcriptional and inflammatory changes that occur in the heart. Five days after 5/6 nephrectomy, RNA-sequencing showed the upregulation of 54 genes in the cardiac tissue of CKD mice and the enrichment of biological processes related to immune system processes. Increased cardiac infiltration of T-CD4+ lymphocytes, myeloid cells, and macrophages during early CKD was observed. Next, since CC chemokine ligand-8 (CCL8) was one of the most upregulated genes in the heart of mice with early CKD, we investigated the effect of acute and transient CCL8 inhibition on uremic cardiomyopathy severity. An increase in CCL8 protein levels was confirmed in the heart of early CKD mice. CCL8 inhibition attenuated the early infiltration of T-CD4+ lymphocytes and macrophages to the cardiac tissue, leading to a protection against chronic cardiac fibrotic remodeling, inflammation and cardiac dysfunction induced by CKD. Altogether, our data show the occurrence of transcriptional and inflammatory changes in the heart during the early phases of CKD and identify CCL8 as a key contributor to the early cardiac inflammatory state that triggers further cardiac remodeling and dysfunction in uremic cardiomyopathy.


Subject(s)
Cardiomyopathies/metabolism , Chemokine CCL8/biosynthesis , Myocardium/metabolism , Renal Insufficiency, Chronic/metabolism , Up-Regulation , Uremia/metabolism , Animals , Cardiomyopathies/pathology , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Myocardium/pathology , Renal Insufficiency, Chronic/pathology , Uremia/pathology
8.
Mol Biol Rep ; 49(8): 8139-8143, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35661049

ABSTRACT

Acetaminophen (APAP) is commonly prescribed as an antipyretic and analgesic agent in the practical field. Like every other drug(s), APAP also undergo metabolism by oxidation or conjugation by glucuronate and sulphate to form the toxic metabolite N-acetyl-p-benzoquinone imine (NAPQI). Moreover, the NAPQI is detoxified by conjugation with reduced glutathione (GSH). Interestingly, APAP is also metabolized in the kidney by deacetylation reaction in the presence of N-deacetylase enzyme into another severely toxic but minor metabolite, p-aminophenol. Both NAPQI and p-aminophenol shows nephrotoxicity as well as hepatotoxicity. Hence, the long-term therapeutic dose use and unnecessary overdose of APAP are of great concern as prolonged negligence may cost the nephrotoxicity that may lead to uremia and finally to kidney failure. It has recently been investigated that probiotic supplementation inhibits the sequential events associated with APAP-induced nephrotoxicity. This review emphasizes the role of different probiotics that have already been investigated in nephrotoxicity or uremia caused by APAP overdose.


Subject(s)
Probiotics , Renal Insufficiency , Uremia , Acetaminophen/adverse effects , Glutathione/metabolism , Humans , Probiotics/therapeutic use , Uremia/metabolism
9.
Cell Biochem Funct ; 40(8): 856-864, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36121199

ABSTRACT

Oxidative stress (OS) is essential in uremia-associated comorbidities, including renal anemia. Complications experienced by hemodialysis (HD) patients, such as hypoxemia and uremic toxins accumulation, induce OS and premature death of red blood cells (RBC). We aimed to characterize reactive oxygen species (ROS) production and antioxidant pathways in HD-RBC and RBC from healthy controls (CON-RBC) and evaluate the role of uremia and hypoxia in these pathways. ROS production, xanthine oxidase (XO) and superoxide dismutase (SOD) activities, glutathione (GSH), and heme oxygenase-1 (HO-1) levels were measured using flow cytometry or spectrophotometry in CON-RBC and HD-RBC (pre- and post-HD), at baseline and after 24 h incubation with uremic serum (S-HD) and/or under hypoxic conditions (5% O2 ). Ketoprofen was used to inhibit RBC uremic toxins uptake. HD-RBC showed higher ROS levels and lower XO activity than CON-RBC, particularly post-HD. GSH levels were lower, while SOD activity and HO-1 levels of HD-RBC were higher than control. Hypoxia per se triggered ROS production in CON-RBC and HD-RBC. S-HD, on top of hypoxia, increased ROS levels. Inhibition of uremic toxins uptake attenuated ROS of CON and HD-RBC under hypoxia and uremia. CON-RBC in uremia and hypoxia showed lower GSH levels than cells in normoxia and non-uremic conditions. Redox mechanisms of HD-RBC are altered and prone to oxidation. Uremic toxins and hypoxia play a role in unbalancing these systems. Hypoxia and uremia participate in the pathogenesis of OS in HD-RBC and might induce RBC death and thus compound anemia.


Subject(s)
Anemia , Uremia , Humans , Erythrocytes/metabolism , Uremia/metabolism , Renal Dialysis , Oxidative Stress , Glutathione/metabolism , Hypoxia/metabolism , Anemia/metabolism
10.
J Ren Nutr ; 32(2): 178-188, 2022 03.
Article in English | MEDLINE | ID: mdl-34688540

ABSTRACT

OBJECTIVE: Vascular calcification (VC) is an important risk factor for cardiovascular disease in maintenance hemodialysis (MHD) patients. Hyperphosphatemia and microinflammation statement are known major contributors to the development of VC; however, the mechanisms are unknown. The aims of this study were to explore the risk factors of VC in MHD patients and to explore whether high phosphate could increase the secretion of inflammatory cytokines via PiT-1 in monocytes. METHODS: A cross-sectional study was conducted on 65 MHD patients to assess the relevance of coronary artery calcification (CAC), inflammatory factors, serum phosphate, and sodium-dependent phosphate cotransporter (NPT) mRNA expression of peripheral blood mononuclear cells (PBMCs). Multivariate logistic regression analysis was used to analyze the predictors of CAC. The calcification effects of high phosphate (HP), TNF-α, and supernatants of healthy human monocytes treated with HP were further evaluated in cultured HASMCs. RESULTS: Diabetes, longer dialysis vintage, higher serum TNF-α levels, and PiT-1 mRNA expression of PBMCs) were independent risk factors of CAC in MHD patients. The mRNA levels of PiT-1 in PBMCs were positively correlated with serum phosphate, CAC scores, and Pit-2 mRNA levels of PBMCs. The expressions of TNF-α, IL-6, and PiT-1 in human monocytes were significantly increased in a dose-dependent manner after treatment with HP, which was subsequently inhibited by NPT antagonist phosphonoformic acid. Neither TNF-α alone nor supernatants of monocytes stimulated with HP promoted the expression of osteopontin and Runt-related transcription factor 2 (Runx2) or caused mineralization in human aortic smooth muscle cells, but combined with HP intervention, the calcification effects were markedly increased in human aortic smooth muscle cells and ameliorated by phosphonoformic acid treatment. CONCLUSION: Hyperphosphatemia directly increased the synthesis and secretion of TNF-α by monocytes may via PiT-1 pathway, resulting in elevated systemic inflammatory response, which may further aggravate VC induced by phosphate overload in MHD patients.


Subject(s)
Hyperphosphatemia , Uremia , Vascular Calcification , Cells, Cultured , Cross-Sectional Studies , Female , Foscarnet/adverse effects , Foscarnet/metabolism , Humans , Hyperphosphatemia/complications , Leukocytes, Mononuclear/metabolism , Male , Muscle, Smooth, Vascular/metabolism , Phosphates/metabolism , RNA, Messenger/metabolism , RNA, Messenger/pharmacology , Sodium-Phosphate Cotransporter Proteins, Type III/genetics , Sodium-Phosphate Cotransporter Proteins, Type III/metabolism , Tumor Necrosis Factor-alpha/genetics , Uremia/complications , Uremia/metabolism , Vascular Calcification/etiology
11.
Ren Fail ; 44(1): 1819-1832, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36299239

ABSTRACT

BACKGROUND/AIM: Uremic cardiomyopathy (UCM) is a characteristic cardiac pathology that is commonly found in patients with chronic kidney disease. This study dissected the mechanism of SPI1 in myocardial fibrosis and inflammation induced by UCM through S100A8/A9. METHODS: An UCM rat model was established, followed by qRT-PCR and western blot analyses of SPI1 and S100A8/A9 expression in myocardial tissues. After alterations of SPI1 and S100A8/A9 expression in UCM rats, the blood specimens were harvested from the cardiac apex of rats. The levels of creatine phosphokinase-MB (CK-MB), blood creatinine, blood urea nitrogen (BUN), and inflammatory cytokines (interleukin [IL]-6, IL-1ß, and tumor necrosis factor-α [TNF-α]) were examined in the collected blood. Collagen fibrosis was assessed by Masson staining. The expression of fibrosis markers [transforming growth factor (TGF)-ß1, α-smooth muscle actin (SMA), Collagen 4a1, and Fibronectin], IL-6, IL-1ß, and TNF-α was measured in myocardial tissues. Chromatin immunoprecipitation and dual-luciferase reporter gene assays were conducted to test the binding relationship between SPI1 and S100A8/A9. RESULTS: S100A8/A9 and SPI1 were highly expressed in the myocardial tissues of UCM rats. Mechanistically, SPI1 bound to the promoter of S100A8/A9 to facilitate S100A8/A9 transcription. S100A8/A9 or SPI1 knockdown reduced myocardial fibrosis and inflammation and the levels of CK-MB, blood creatinine, and BUN, as well as the expression of TGF-ß1, α-SMA, Collagen 4a1, Fibronectin, IL-6, TNF-α, and IL-1ß in UCM rats. CONCLUSION: SPI1 knockdown diminished S100A8/A9 transcription, thus suppressing myocardial fibrosis and inflammation caused by UCM.


Subject(s)
Calgranulin A , Calgranulin B , Cardiomyopathies , Animals , Rats , Actins/metabolism , Calgranulin A/genetics , Calgranulin A/metabolism , Calgranulin B/genetics , Calgranulin B/metabolism , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cardiomyopathies/prevention & control , Creatine Kinase , Creatinine , Cytokines/metabolism , Down-Regulation , Fibronectins/metabolism , Fibrosis/genetics , Fibrosis/metabolism , Genomic Islands , Inflammation/genetics , Inflammation/metabolism , Interleukin-6/metabolism , Luciferases/genetics , Luciferases/metabolism , SUMO-1 Protein/genetics , SUMO-1 Protein/metabolism , Transforming Growth Factor beta1/metabolism , Transforming Growth Factors/genetics , Transforming Growth Factors/metabolism , Tumor Necrosis Factor-alpha/metabolism , Uremia/complications , Uremia/genetics , Uremia/metabolism
12.
Int J Mol Sci ; 23(4)2022 Feb 21.
Article in English | MEDLINE | ID: mdl-35216489

ABSTRACT

In patients with chronic kidney disease (CKD) and in animal models of CKD, the transcription factor Aryl Hydrocabon Receptor (AhR) is overactivated. In addition to the canonical AhR targets constituting the AhR signature, numerous other genes are regulated by this factor. We identified neuronal pentraxin 1 (NPTX1) as a new AhR target. Belonging to the inflammatory protein family, NPTX1 seems of prime interest regarding the inflammatory state observed in CKD. Endothelial cells were exposed to tryptophan-derived toxins, indoxyl sulfate (IS) and indole-3-acetic acid (IAA). The adenine mouse model of CKD was used to analyze NPTX1 expression in the burden of uremia. NPTX1 expression was quantified by RT-PCR and western blot. AhR involvement was analyzed using silencing RNA. We found that IS and IAA upregulated NPTX1 expression in an AhR-dependent way. Furthermore, this effect was not restricted to uremic indolic toxins since the dioxin 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) and the tryptophan photoproduct 6-formylindolo[3,2-b]carbazole (FICZ) do the same. In CKD mice, NPTX1 expression was increased in the aorta. Therefore, NPTX1 is a new target of AhR and further work is necessary to elucidate its exact role during CKD.


Subject(s)
C-Reactive Protein/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Nerve Tissue Proteins/metabolism , Tryptophan/metabolism , Animals , Carbazoles/metabolism , Cells, Cultured , Disease Models, Animal , Humans , Indican/metabolism , Indoleacetic Acids/metabolism , Male , Mice , Mice, Inbred C57BL , Polychlorinated Dibenzodioxins/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Renal Insufficiency, Chronic/metabolism , Toxins, Biological/metabolism , Uremia/metabolism
13.
Eur J Immunol ; 50(2): 220-233, 2020 02.
Article in English | MEDLINE | ID: mdl-31777959

ABSTRACT

The role of complement system in heart diseases is controversial. Besides, the mechanisms by which complement components participate in cardiac remodeling (CR) and heart failure during uremia are unclear. In this study, 5/6 nephrectomy was performed to adult mice to establish the uremic model and CR deteriorated over the course of uremia. Although complement pathways were not further activated over the course of the disease, soluble complement factor B (CFB) was upregulated at post-nephrectomy day 90 (PNx90) compared with PNx30. Further, CFB notably deteriorated CR in uremic mice but this effect was reversed by depletion of macrophages with liposomal clodronate. In vivo and in vitro CFB upregulated arginase 1 (ARG1) expression, increased ARG1 enzymatic activity, and stimulated the syntheses of ornithine, leading to polyamine overproduction in macrophages. Putrescine, an important polyamine, promoted cardiac fibroblast proliferation and collagen production, resulting in progressive CR. In vivo the inhibition of ARG1 activity with Nω -hydroxyl-l-arginine remarkably improved the general survival rates, inhibited the infiltration of cardiac fibroblasts, and alleviated progression of CR in uremic mice. Taken together, the CFB-ARG1-putrescine axis is related to progression of CR and ARG1 hyperactivity in macrophages may provide a novel therapeutic target against the heart injury in uremia.


Subject(s)
Arginase/metabolism , Complement Factor B/metabolism , Uremia/metabolism , Ventricular Remodeling/physiology , Animals , Arginine/metabolism , Macrophages/metabolism , Mice , Nitric Oxide/metabolism , Ornithine/metabolism , Polyamines/metabolism , Signal Transduction/physiology
14.
Opt Lett ; 46(5): 1101-1104, 2021 Mar 01.
Article in English | MEDLINE | ID: mdl-33649667

ABSTRACT

In this Letter, we propose a microstructured in-fiber optofluidic surface-enhanced Raman spectroscopy (SERS) sensor for the initial inspection of uremia through the detection of unlabeled urea and creatinine. As a natural microfluidic device, microstructured hollow fiber has a special structure inside. Through chemical bonds, the SERS substrate can be modified and grown on the surface of the suspended core. Here, the silver nanoparticles (Ag NPs) are embedded on the poly diallyl dimethyl ammonium chloride-modified graphene oxide sheet to achieve the self-assembled SERS substrate. The reduced distance between Ag NPs can increase the strong hot spots that generate enhanced Raman signals. Therefore, it can effectively detect the Raman signal of unlabeled trace uremic toxin analytes (urea, creatinine) inside the optical fiber. The results show that under simulated biophysical conditions, the limit detection (LOD) for urea is 10-4M and the linearity is good, especially at the clinical conventional concentration range (2.5-6.5×10-3M). In addition, the online Raman detection of creatinine aqueous solution LOD is 10-6M, which also has good linearity. Significantly, this Letter provides a microstructured optofluidic in-fiber Raman sensor for the preliminary detection of uremia, which will have good development prospects in the field of clinical biomedicine.


Subject(s)
Limit of Detection , Optical Fibers , Spectrum Analysis, Raman/instrumentation , Toxins, Biological/metabolism , Uremia/metabolism
15.
Kidney Blood Press Res ; 46(3): 377-386, 2021.
Article in English | MEDLINE | ID: mdl-34044409

ABSTRACT

INTRODUCTION: Cardiovascular disease is the most common cause of morbidity and mortality in patients with ESRD. In addition to phosphate overload, oxalate, a common uremic toxin, is also involved in vascular calcification in patients with ESRD. The present study investigated the role and mechanism of hyperoxalemia in vascular calcification in mice with uremia. METHODS: A uremic atherosclerosis (UA) model was established by left renal excision and right renal electrocoagulation in apoE-/- mice to investigate the relationship between oxalate loading and vascular calcification. After 12 weeks, serum and vascular levels of oxalate, vascular calcification, inflammatory factors (TNF-α and IL-6), oxidative stress markers (malondialdehyde [MDA], and advanced oxidation protein products [AOPP]) were assessed in UA mice. The oral oxalate-degrading microbe Oxalobacter formigenes (O. formigenes) was used to evaluate the effect of a reduction in oxalate levels on vascular calcification. The mechanism underlying the effect of oxalate loading on vascular calcification was assessed in cultured human aortic endothelial cells (HAECs) and human aortic smooth muscle cells (HASMCs). RESULTS: Serum oxalate levels were significantly increased in UA mice. Compared to the control mice, UA mice developed more areas of aortic calcification and showed significant increases in aortic oxalate levels and serum levels of oxidative stress markers and inflammatory factors. The correlation analysis showed that serum oxalate levels were positively correlated with the vascular oxalate levels and serum MDA, AOPP, and TNF-α levels, and negatively correlated with superoxide dismutase activity. The O. formigenes intervention decreased serum and vascular oxalate levels, while did not improve vascular calcification significantly. In addition, systemic inflammation and oxidative stress were also improved in the O. formigenes group. In vitro, high concentrations of oxalate dose-dependently increased oxidative stress and inflammatory factor expression in HAECs, but not in HASMCs. CONCLUSIONS: Our results indicated that hyperoxalemia led to the systemic inflammation and the activation of oxidative stress. The reduction in oxalate levels by O. formigenes might be a promising treatment for the prevention of oxalate deposition in calcified areas of patients with ESRD.


Subject(s)
Endothelial Cells/pathology , Oxalates/metabolism , Oxidative Stress , Renal Insufficiency, Chronic/pathology , Animals , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cell Line , Disease Models, Animal , Endothelial Cells/metabolism , Humans , Male , Mice , Renal Insufficiency, Chronic/metabolism , Uremia/metabolism , Uremia/pathology , Vascular Calcification/metabolism , Vascular Calcification/pathology
16.
Int J Med Sci ; 18(3): 744-755, 2021.
Article in English | MEDLINE | ID: mdl-33437209

ABSTRACT

Chronic kidney disease (CKD) patients obtained high levels of uremic toxins progressively develop several complications including bone fractures. Protein-bound uremic toxins especially p-cresol and indoxyl sulfate are hardly eliminated due to their high molecular weight. Thus, the abnormality of bone in CKD patient could be potentially resulted from the accumulation of uremic toxins. To determine whether protein-bound uremic toxins have an impact on osteogenesis, mesenchymal stem cells were treated with either p-cresol or indoxyl sulfate under in vitro osteogenic differentiation. The effects of uremic toxins on MSC-osteoblastic differentiation were investigated by evaluation of bone phenotype. The results demonstrated that p-cresol and indoxyl sulfate down-regulated the transcriptional level of collagen type I, deceased alkaline phosphatase activity, and impaired mineralization of MSC-osteoblastic cells. Furthermore, p-cresol and indoxyl sulfate gradually increased senescence-associated beta-galactosidase positive cells while upregulated the expression of p21 which participate in senescent process. Our findings clearly revealed that the presence of uremic toxins dose-dependently influenced a gradual deterioration of osteogenesis. The effects partially mediate through the activation of senescence-associated gene lead to the impairment of osteogenesis. Therefore, the management of cellular senescence triggered by uremic toxins could be considered as an alternative therapeutic approach to prevent bone abnormality in CKD patients.


Subject(s)
Chronic Kidney Disease-Mineral and Bone Disorder/metabolism , Mesenchymal Stem Cells/pathology , Renal Insufficiency, Chronic/complications , Toxins, Biological/metabolism , Uremia/metabolism , Cells, Cultured , Cellular Senescence , Chronic Kidney Disease-Mineral and Bone Disorder/etiology , Chronic Kidney Disease-Mineral and Bone Disorder/urine , Cresols/metabolism , Cresols/urine , Healthy Volunteers , Humans , Indican/metabolism , Indican/urine , Osteogenesis/physiology , Primary Cell Culture , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/urine , Toxins, Biological/urine , Uremia/etiology , Uremia/urine
17.
J Am Soc Nephrol ; 31(7): 1423-1434, 2020 07.
Article in English | MEDLINE | ID: mdl-32527977

ABSTRACT

Cardiac hypertrophy is a common feature in patients with CKD. Recent studies revealed that two phosphate regulators, fibroblast growth factor-23 and α-Klotho, are highly involved in the pathophysiologic process of CKD-induced cardiac hypertrophy. With decreasing renal function, elevated fibroblast growth factor-23 and decreased α-Klotho may contribute to cardiac hypertrophy by targeting the heart directly or by inducing systemic changes, such as vascular injury, hemodynamic disorders, and inflammation. However, several studies have demonstrated that disturbances in the fibroblast growth factor-23/α-Klotho axis do not lead to cardiac hypertrophy. In this review, we describe the cardiac effects of the fibroblast growth factor-23/α-Klotho axis and summarize recent progress in this field. In addition, we present not only the main controversies in this field but also provide possible directions to resolve these disputes.


Subject(s)
Fibroblast Growth Factors/metabolism , Glucuronidase/metabolism , Hypertrophy, Left Ventricular/metabolism , Renal Insufficiency, Chronic/metabolism , Uremia/metabolism , Animals , Endothelium, Vascular/physiopathology , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/blood , Glucuronidase/blood , Humans , Hypertrophy, Left Ventricular/etiology , Klotho Proteins , Paracrine Communication , Receptors, Fibroblast Growth Factor/metabolism , Uremia/complications
18.
Ren Fail ; 43(1): 401-405, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33641611

ABSTRACT

BACKGROUND: Uremia induces various pathologic changes in the endothelium. However, there is limited information about the differences of these effects in endothelial cells originating from different parts of the vascular tree. METHODS: The effect of uremic serum obtained from patients with end stage renal failure on the gene expression and secretory activity of venous endothelial cells (VEC) and aortic endothelial cells (AEC) was studied in in vitro culture. RESULTS: In VEC, the expression of genes regulating the synthesis of von Willebrand factor (vWF) was increased by 254% (p<.005), vascular endothelial growth factor (VEGF) synthesis by 150% (p<.001), tissue plasminogen activator (t-PA) synthesis by 62% (p<.005), platelet endothelial cell adhesion molecule by 89% (p<.005), and the expression of gene regulating interleukin-6 (IL-6) synthesis was reduced. In AEC, the expression of the gene regulating synthesis of IL-6 was increased by 174% (p<.001), and the expression of the other genes was reduced. The secretion of IL-6 was reduced in VEC by 38% (p<.01) and increased in AEC by 55% (p<.005). In VEC, increased synthesis of VEGF 64% (p<.001) vWF (+34%, p<.01), and t-PA (+53%, p<.002) was observed, and in AEC it was reduced. CONCLUSIONS: VEC and AEC respond in different ways after exposure to uremic serum. VEC acquires the prothrombotic phenotype, whereas in AEC the inflammatory phenotype appears.


Subject(s)
Arteries/pathology , Endothelial Cells/pathology , Endothelium, Vascular/cytology , Inflammation/pathology , Uremia/metabolism , Cells, Cultured , Endothelial Cells/metabolism , Gene Expression Regulation , Humans , Inflammation/genetics , Interleukin-6/genetics , Interleukin-6/metabolism , Kidney Failure, Chronic/blood , Kidney Failure, Chronic/complications , Tissue Plasminogen Activator/genetics , Tissue Plasminogen Activator/metabolism , Uremia/blood , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , von Willebrand Factor/genetics , von Willebrand Factor/metabolism
19.
Int J Mol Sci ; 22(12)2021 Jun 18.
Article in English | MEDLINE | ID: mdl-34207309

ABSTRACT

Secondary hyperparathyroidism and abnormalities in tryptophan (TRP) metabolism are commonly observed in chronic kidney disease (CKD). The present study aimed to establish potential interactions between endogenous parathyroid hormone (PTH) and activation of the bone kynurenine (KYN) pathway in relation to bone turnover and strength in young rats after one month (CKD-1) and three months (CKD-3) of experimental CKD. TRP, KYN, KYN/TRP ratio and bone turnover markers (BTMs) were measured in trabecular and cortical bone tissue. Expression of aryl hydrocarbon receptor (AhR) and the genes involved in osteogenesis was determined in femoral bone. Biomechanical testing of femoral diaphysis and femoral neck was also performed. Activation of the KYN pathway in trabecular bone during CKD development intensified the expression of genes related to osteogenesis, which led to a decrease in cyclic adenosine monophosphate (cAMP) and BTMs levels, resulting in a stiffer and mechanically weaker femoral neck. In contrast, reduction of the KYN pathway in cortical bone allowed to unblock the PTH-dependent anabolic activating transcription factor 4/parathyroid hormone 1 receptor (PTH1R/ATF4) axis, led to cAMP accumulation, better bone turnover and strength in the course of CKD development. In summary, the paracrine KYN pathway in bone can interfere with the anabolic effects of PTH on bone through disrupting PTH-dependent molecular signaling.


Subject(s)
Femur/metabolism , Kynurenine/metabolism , Paracrine Communication , Parathyroid Hormone/metabolism , Signal Transduction , Uremia/metabolism , Activating Transcription Factor 4/metabolism , Animals , Cancellous Bone/metabolism , Cortical Bone/metabolism , Cyclic AMP/metabolism , Male , Rats , Rats, Wistar , Receptor, Parathyroid Hormone, Type 1/metabolism
20.
Int J Mol Sci ; 22(19)2021 Sep 29.
Article in English | MEDLINE | ID: mdl-34638892

ABSTRACT

Chronic kidney disease (CKD) is a major cause of death worldwide and is associated with a high risk for cardiovascular and all-cause mortality. In CKD, endothelial dysfunction occurs and uremic toxins accumulate in the blood. miR-126 is a regulator of endothelial dysfunction and its blood level is decreased in CKD patients. In order to obtain a better understanding of the physiopathology of the disease, we correlated the levels of miR-126 with several markers of endothelial dysfunction, as well as the representative uremic toxins, in a large cohort of CKD patients at all stages of the disease. Using a univariate analysis, we found a correlation between eGFR and most markers of endothelial dysfunction markers evaluated in this study. An association of miR-126 with all the evaluated uremic toxins was also found, while uremic toxins were not associated with the internal control, specifically cel-miR-39. The correlation between the expression of endothelial dysfunction biomarker Syndecan-1, free indoxyl sulfate, and total p-cresyl glucuronide on one side, and miR-126 on the other side was confirmed using multivariate analysis. As CKD is associated with reduced endothelial glycocalyx (eGC), our results justify further evaluation of the role of correlated parameters in the pathophysiology of CKD.


Subject(s)
Biomarkers/metabolism , Indican/metabolism , MicroRNAs/genetics , Renal Insufficiency, Chronic/genetics , Syndecan-1/metabolism , Adult , Aged , Biomarkers/blood , Cohort Studies , Female , Glomerular Filtration Rate , Glycocalyx/metabolism , Humans , Indican/blood , Linear Models , Male , Middle Aged , Multivariate Analysis , Renal Insufficiency, Chronic/diagnosis , Renal Insufficiency, Chronic/metabolism , Toxins, Biological/metabolism , Uremia/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL