Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 191
1.
Ren Fail ; 44(1): 1819-1832, 2022 Dec.
Article En | MEDLINE | ID: mdl-36299239

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.


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
2.
Kidney Int ; 101(6): 1232-1250, 2022 06.
Article En | MEDLINE | ID: mdl-35276205

The molecular circadian clock is an evolutionary adaptation to anticipate recurring changes in the environment and to coordinate variations in activity, metabolism and hormone secretion. Parathyroid hyperplasia in uremia is a significant clinical challenge. Here, we examined changes in the transcriptome of the murine parathyroid gland over 24 hours and found a rhythmic expression of parathyroid signature genes, such as Casr, Vdr, Fgfr1 and Gcm2. Overall, 1455 genes corresponding to 6.9% of all expressed genes had significant circadian rhythmicity. Biological pathway analysis indicated that the circadian clock system is essential for the regulation of parathyroid cell function. To study this, a novel mouse strain with parathyroid gland-specific knockdown of the core clock gene Bmal1 (PTHcre;Bmal1flox/flox) was created. Dampening of the parathyroid circadian clock rhythmicity was found in these knockdown mice, resulting in abrogated rhythmicity of regulators of parathyroid cell proliferation such as Sp1, Mafb, Gcm2 and Gata3, indicating circadian clock regulation of these genes. Furthermore, the knockdown resulted in downregulation of genes involved in mitochondrial function and synthesis of ATP. When superimposed by uremia, these PTHcre;Bmal1flox/flox mice had an increased parathyroid cell proliferative response, compared to wild type mice. Thus, our findings indicate a role of the internal parathyroid circadian clock in the development of parathyroid gland hyperplasia in uremia.


Circadian Clocks , Uremia , Animals , Cell Proliferation , Circadian Clocks/genetics , Circadian Rhythm/genetics , Gene Expression Regulation , Hyperplasia , Mice , Parathyroid Glands , Uremia/genetics
3.
Anal Cell Pathol (Amst) ; 2022: 5437560, 2022.
Article En | MEDLINE | ID: mdl-36618529

Uremia is a common syndrome that happens to nearly all end-stage kidney diseases, which profound have changes in human gene expressions, but the related pathways are poorly understood. Gene Ontology categories and Kyoto Encyclopedia of Genes and Genomes pathways enriched in the differentially expressed genes (DEGs) were analyzed by using clusterProfiler, org.Hs.eg.db, and Pathview, and protein-protein interaction (PPI) network was built by Cytoscape. We identified 3432 DEGs (including 3368 down- and 64 up-regulated genes), of which there were 52 different molecular functions, and 178 genes were identified as immune genes controlled by the four transcription factors (POU domain class 6 transcription factor 1 (POU6F1), interferon regulator factor 7 [IRF7], forkhead box D3 (FOXD3), and interferon-stimulated response element [ISRE]). In the gender research, no significant difference was observed. The top 15 proteins of 178 immune-related genes were identified with the highest degree in PPI network. The DEG analysis of uremia patients was expected to provide fundamental information to relieve pain and add years to their life.


Gene Expression Profiling , Uremia , Humans , Protein Interaction Maps/genetics , Transcription Factors/metabolism , Uremia/genetics , Computational Biology , Gene Regulatory Networks , POU Domain Factors/genetics , POU Domain Factors/metabolism
4.
PLoS One ; 16(9): e0256734, 2021.
Article En | MEDLINE | ID: mdl-34469488

OBJECTIVES: This study is to investigate whether the cardiac microvascular endothelial cells (CMECs) can regulate the autophagy of cardiomyocytes (CMs) by secreting lncRNA-ANRIL/miR-181b exosomes, thus participating in the occurrence of uremic cardiovascular disease (CVD). METHODS: A 5/6 nephrectomy uremia model was established, with the mice injected with ANRIL-shRNA lentivirus vector, miR-181b agomir, and related control reagents, containing the serum creatinine and urea nitrogen measured. The renal tissue sections of mice were stained with Periodic Acid-Schiff (PAS), TUNEL, and Hematoxylin-Eosin (HE) performed on myocardial tissue sections of mice. ANRIL-shRNA, miR-181b mimics, and related control reagents were transfected into CMECs, in which the exosomes were extracted and co-cultured with CMs. The expressions of ANRIL, miR-181b and ATG5 were detected by qRT-PCR, and the expressions of autophagy related proteins by Western blot, as well as the binding of ANRIL and miR-181b by the double luciferase reporter gene experiment. RESULTS: ANRIL down-regulation or miR-181b up-regulation can increase the weight of mice with uremia, as well as the expressions of p62 and miR-181b, and reduce the content of serum creatinine and urea nitrogen, the damage of kidney and myocardial tissues, the number of apoptotic cells in myocardial tissues, as well as the expressions of ANRIL, ATG5, Beclin1, and LC3. CMs can absorb the exosomes of CMECs. Compared with IS+ CMEC-Exo group, the expressions of ANRIL and ATG5 in CMs of IS+ CMEC-Exo + sh lncRNA ANRIL and IS+CMEC-Exo+miR-181b mimics groups was down-regulated, as well as the expressions of ATG5, Beclin1, and LC3, while miR-181b expression was up-regulated as well as P62 expression. CONCLUSIONS: CMECs can regulate autophagy of CMs by releasing exosomes containing ANRIL and miR-181b.


Autophagy-Related Protein 5/genetics , Autophagy/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Uremia/immunology , Animals , Autophagy-Related Protein 5/metabolism , Coronary Vessels/cytology , Coronary Vessels/metabolism , Disease Models, Animal , Down-Regulation/immunology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Exosomes/metabolism , Humans , Male , Mice , MicroRNAs/genetics , Microvessels/cytology , Myocardium/cytology , Myocardium/immunology , Myocardium/pathology , Myocytes, Cardiac/immunology , Myocytes, Cardiac/pathology , RNA, Long Noncoding/genetics , Up-Regulation/immunology , Uremia/genetics , Uremia/pathology
5.
Int J Mol Sci ; 22(6)2021 Mar 11.
Article En | MEDLINE | ID: mdl-33799511

In uremic patients, high-density lipoprotein (HDL) loses its anti-inflammatory features and can even become pro-inflammatory due to an altered protein composition. In chronic kidney disease (CKD), impaired functions of polymorphonuclear leukocytes (PMNLs) contribute to inflammation and an increased risk of cardiovascular disease. This study investigated the effect of HDL from CKD and hemodialysis (HD) patients on the CD14 expression on PMNLs. HDL was isolated using a one-step density gradient centrifugation. Isolation of PMNLs was carried out by discontinuous Ficoll-Hypaque density gradient centrifugation. CD14 surface expression was quantified by flow cytometry. The activity of the small GTPase Rac1 was determined by means of an activation pull-down assay. HDL increased the CD14 surface expression on PMNLs. This effect was more pronounced for HDL isolated from uremic patients. The acute phase protein serum amyloid A (SAA) caused higher CD14 expression, while SAA as part of an HDL particle did not. Lipid raft disruption with methyl-ß-cyclodextrin led to a reduced CD14 expression in the absence and presence of HDL. HDL from healthy subjects but not from HD patients decreased the activity of Rac1. Considering the known anti-inflammatory effects of HDL, the finding that even HDL from healthy subjects increased the CD14 expression was unexpected. The pathophysiological relevance of this result needs further investigation.


Lipopolysaccharide Receptors/genetics , Lipoproteins, HDL/pharmacology , Neutrophils/drug effects , Renal Insufficiency, Chronic/genetics , Uremia/genetics , Aged , Case-Control Studies , Female , Humans , Lipopolysaccharide Receptors/metabolism , Lipoproteins, HDL/isolation & purification , Male , Membrane Microdomains/chemistry , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Middle Aged , Neutrophils/metabolism , Neutrophils/pathology , Primary Cell Culture , Renal Dialysis , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/physiopathology , Renal Insufficiency, Chronic/therapy , Serum Amyloid A Protein/genetics , Serum Amyloid A Protein/metabolism , Uremia/metabolism , Uremia/physiopathology , Uremia/therapy , beta-Cyclodextrins/pharmacology , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
6.
Cardiovasc Res ; 117(8): 1958-1973, 2021 07 07.
Article En | MEDLINE | ID: mdl-32866261

AIMS: Vascular calcification (VC) increases the future risk of cardiovascular events in uraemic patients, but effective therapies are still unavailable. Accurate identification of those at risk of developing VC using pathogenesis-based biomarkers is of particular interest and may facilitate individualized risk stratification. We aimed to uncover microRNA (miRNA)-target protein-based biomarker panels for evaluating uraemic VC probability and severity. METHODS AND RESULTS: We created a three-tiered in vitro VC model and an in vivo uraemic rat model receiving high phosphate diet to mimic uraemic VC. RNAs from the three-tiered in vitro and in vivo uraemic VC models underwent miRNA and mRNA microarray, with results screened for differentially expressed miRNAs and their target genes as biomarkers. Findings were validated in original models and additionally in an ex vivo VC model and human cells, followed by functional assays of identified miRNAs and target proteins, and tests of sera from end-stage renal disease (ESRD) and non-dialysis-dependent chronic kidney disease (CKD) patients without and with VC. Totally 122 down-regulated and 119 up-regulated miRNAs during calcification progression were identified initially; further list narrowing based on miRNA-mRNA pairing, anti-correlation, and functional enrichment left 16 and 14 differentially expressed miRNAs and mRNAs. Levels of four miRNAs (miR-10b-5p, miR-195, miR-125b-2-3p, and miR-378a-3p) were shown to decrease throughout all models tested, while one mRNA (SULF1, a potential target of miR-378a-3p) exhibited the opposite trend concurrently. Among 96 ESRD (70.8% with VC) and 59 CKD patients (61% with VC), serum miR-125b2-3p and miR-378a-3p decreased with greater VC severity, while serum SULF1 levels increased. Adding serum miR-125b-2-3p, miR-378a-3p, and SULF1 into regression models for VC substantially improved performance compared to using clinical variables alone. CONCLUSION: Using a translational approach, we discovered a novel panel of biomarkers for gauging the probability/severity of uraemic VC based on miRNAs/target proteins, which improved the diagnostic accuracy.


Gene Expression Profiling , MicroRNAs/genetics , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Proteome , Proteomics , Transcriptome , Translational Research, Biomedical , Uremia/complications , Vascular Calcification/etiology , Adult , Aged , Aged, 80 and over , Animals , Biomarkers/blood , Cells, Cultured , Disease Models, Animal , Female , Gene Regulatory Networks , Humans , Male , MicroRNAs/metabolism , Middle Aged , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Organ Culture Techniques , Predictive Value of Tests , Protein Interaction Maps , Rats, Sprague-Dawley , Risk Assessment , Risk Factors , Severity of Illness Index , Signal Transduction , Sulfotransferases/blood , Uremia/genetics , Uremia/metabolism , Vascular Calcification/genetics , Vascular Calcification/metabolism , Vascular Calcification/pathology
7.
Oxid Med Cell Longev ; 2020: 3431597, 2020.
Article En | MEDLINE | ID: mdl-33343805

Vascular calcification is a major complication of maintenance hemodialysis patients. Studies have confirmed that calcification mainly occurs in the vascular smooth muscle cells (VSMC) of the vascular media. However, the exact pathogenesis of VSMC calcification is still unknown. This study shows that the crosstalk between calcium and aldosterone via the allograft inflammatory factor 1 (AIF-1) pathway contributes to calcium homeostasis and VSMC calcification, which is a novel mechanism of vascular calcification in uremia. In vivo results showed that the level of aldosterone and inflammatory factors increased in calcified arteries, whereas no significant changes were observed in peripheral blood. However, the expression of inflammatory factors markedly increased in the peripheral blood of uremic rats without aortic calcification and gradually returned to normal levels with aggravation of aortic calcification. In vitro results showed that there was an interaction between calcium ions and aldosterone in macrophages or VSMC. Calcium induced aldosterone synthesis, and in turn, aldosterone also triggered intracellular calcium content upregulation in macrophages or VSMC. Furthermore, activated macrophages induced inflammation, apoptosis, and calcification of VSMC. Activated VSMC also imparted a similar effect on untreated VSMC. Finally, AIF-1 enhanced aldosterone- or calcium-induced VSMC calcification, and NF-κB inhibitors inhibited the effect of AIF-1 on VSMC. These in vivo and in vitro results suggest that the crosstalk between calcium ions and aldosterone plays an important role in VSMC calcification in uremia via the AIF-1/NF-κB pathway. Local calcified VSMC induced the same pathological process in surrounding VSMC, thereby contributing to calcium homeostasis and accelerating vascular calcification.


Aldosterone/metabolism , Apoptosis , Calcium-Binding Proteins/metabolism , Calcium/metabolism , Microfilament Proteins/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , NF-kappa B/metabolism , Signal Transduction , Uremia/metabolism , Vascular Calcification/metabolism , Animals , Calcium-Binding Proteins/genetics , Cell Line , Female , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Male , Microfilament Proteins/genetics , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , NF-kappa B/genetics , Rats , Rats, Sprague-Dawley , Uremia/genetics , Uremia/pathology , Vascular Calcification/genetics , Vascular Calcification/pathology
8.
Toxins (Basel) ; 12(10)2020 09 28.
Article En | MEDLINE | ID: mdl-32998431

End-stage renal disease (ESRD) is the final stage of chronic kidney disease, which is increasingly prevalent worldwide and is associated with the progression of cardiovascular disease (CVD). Indoxyl sulfate (IS), a major uremic toxin, plays a key role in the pathology of CVD via adverse effects in endothelial and immune cells. Thus, there is a need for a transcriptomic overview of IS responsive genes in immune cells of ESRD patients. Here, we investigated IS-mediated alterations in gene expression in monocytes from ESRD patients. Transcriptomic analysis of ESRD patient-derived monocytes and IS-stimulated monocytes from healthy controls was performed, followed by analysis of differentially expressed genes (DEGs) and gene ontology (GO). We found that 148 upregulated and 139 downregulated genes were shared between ESRD patient-derived and IS-stimulated monocytes. Interaction network analysis using STRING and ClueGo suggests that mainly metabolic pathways, such as the pentose phosphate pathway, are modified by IS in ESRD patient-derived monocytes. These findings were confirmed in IS-stimulated monocytes by the increased mRNA expression of genes including G6PD, PGD, and TALDO1. Our data suggest that IS causes alteration of metabolic pathways in monocytes of ESRD patients and, thus, these altered genes may be therapeutic targets.


Energy Metabolism/genetics , Indican/blood , Kidney Failure, Chronic/genetics , Monocytes/metabolism , Toxins, Biological/blood , Transcriptome , Uremia/genetics , Aged , Case-Control Studies , Female , Gene Expression Profiling , Gene Regulatory Networks , Humans , Kidney Failure, Chronic/blood , Kidney Failure, Chronic/diagnosis , Kidney Failure, Chronic/immunology , Male , Middle Aged , Monocytes/immunology , Protein Interaction Maps , Up-Regulation , Uremia/blood , Uremia/diagnosis , Uremia/immunology
9.
Nutrients ; 12(5)2020 Apr 27.
Article En | MEDLINE | ID: mdl-32349312

Cardiovascular morbidity and mortality are several-fold higher in patients with advanced chronic kidney disease (CKD) and end-stage renal disease (ESRD) than in the general population. Hyperhomocysteinemia has undoubtedly a central role in such a prominent cardiovascular burden. The levels of homocysteine are regulated by methyl donors (folate, methionine, choline, betaine), and cofactors (vitamin B6, vitamin B12,). Uremia-induced hyperhomocysteinemia has as its main targets DNA methyltransferases, and this leads to an altered epigenetic control of genes regulated through methylation. In renal patients, the epigenetic landscape is strictly correlated with the uremic phenotype and dependent on dietary intake of micronutrients, inflammation, gut microbiome, inflammatory status, oxidative stress, and lifestyle habits. All these factors are key contributors in methylome maintenance and in the modulation of gene transcription through DNA hypo- or hypermethylation in CKD. This is an overview of the epigenetic changes related to DNA methylation in patients with advanced CKD and ESRD. We explored the currently available data on the molecular dysregulations resulting from altered gene expression in uremia. Special attention was paid to the efficacy of B-vitamins supplementation and dietary intake of methyl donors on homocysteine lowering and cardiovascular protection.


Betaine/administration & dosage , Choline/administration & dosage , DNA Methylation/genetics , Dietary Supplements , Eating/physiology , Epigenesis, Genetic , Folic Acid/administration & dosage , Methionine/administration & dosage , Nutritional Physiological Phenomena/genetics , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/metabolism , Vitamin B 12/administration & dosage , Vitamin B 6/administration & dosage , Cardiovascular Diseases/etiology , Cardiovascular Diseases/prevention & control , Humans , Hyperhomocysteinemia/etiology , Hyperhomocysteinemia/prevention & control , Kidney Failure, Chronic , Nutritional Physiological Phenomena/physiology , Renal Insufficiency, Chronic/complications , Uremia/complications , Uremia/genetics
10.
Immun Inflamm Dis ; 8(2): 228-235, 2020 06.
Article En | MEDLINE | ID: mdl-32180373

INTRODUCTION: We previously reported that fibroblast growth factor 23 (FGF23)-klotho signaling plays a role in B cell immunity. Despite high serum levels of FGF23, a decline in immunity is frequently observed in patients on hemodialysis (HD); thus, abnormalities in the FGF23-klotho signaling pathway in immune cells may occur in these patients. METHODS: We analyzed the number of klotho-positive cells in peripheral blood mononuclear cells from 10 male and 6 female patients on HD and 5 healthy male subjects using flow cytometry. We analyzed the abundance of cleaved klotho protein in the murine B cell line, A20, and in the serum of HD patients and healthy subjects (HS) using flow cytometry and Western blotting. The serum level of A disintegrin and metalloprotease 17 (ADAM17) was measured in HD patients and HS using enzyme-linked immunosorbent assay. RESULTS: The number of klotho-positive B cells was reduced in HD patients. Serum ADAM17 was responsible for the reduction in klotho, as a specific ADAM17 inhibitor reversed this change. The total serum levels of ADAM17 were similar in HD patients and HS; however, activated ADAM17 was increased in the serum of HD patients. CONCLUSIONS: We concluded that abnormal ADAM17 activation could contribute to the immunocompromised status in patients on HD, in line with the reported role of ADAM17 as an anti-inflammatory and immunosuppressive factor.


ADAM17 Protein/blood , Fibroblast Growth Factors/blood , Glucuronidase/blood , Leukocytes, Mononuclear/metabolism , Renal Insufficiency, Chronic/genetics , ADAM17 Protein/genetics , Aged , Aged, 80 and over , Animals , Case-Control Studies , Cell Line , Female , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/genetics , Glucuronidase/genetics , Humans , Immunocompromised Host , Klotho Proteins , Male , Mice , Middle Aged , Renal Dialysis , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/therapy , Uremia/blood , Uremia/genetics
11.
Int J Mol Sci ; 21(4)2020 Feb 13.
Article En | MEDLINE | ID: mdl-32069992

In recent years, Na/K-ATPase signaling has been implicated in different physiological and pathophysiological conditions, including cardiac hypertrophy and uremic cardiomyopathy. Cardiotonic steroids (CTS), specific ligands of Na/K-ATPase, regulate its enzymatic activity (at higher concentrations) and signaling function (at lower concentrations without significantly affecting its enzymatic activity) and increase reactive oxygen species (ROS) generation. On the other hand, an increase in ROS alone also regulates the Na/K-ATPase enzymatic activity and signaling function. We termed this phenomenon the Na/K-ATPase-mediated oxidant-amplification loop, in which oxidative stress regulates both the Na/K-ATPase activity and signaling. Most recently, we also demonstrated that this amplification loop is involved in the development of uremic cardiomyopathy. This review aims to evaluate the redox-sensitive Na/K-ATPase-mediated oxidant amplification loop and uremic cardiomyopathy.


Cardiomyopathies/genetics , Oxidative Stress/genetics , Sodium-Potassium-Exchanging ATPase/genetics , Uremia/genetics , Cardiac Glycosides/therapeutic use , Cardiomyopathies/drug therapy , Cardiomyopathies/etiology , Cardiomyopathies/pathology , Humans , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Signal Transduction , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Uremia/complications , Uremia/drug therapy , Uremia/pathology
12.
Med Sci Monit ; 26: e919086, 2020 Feb 10.
Article En | MEDLINE | ID: mdl-32040471

BACKGROUND This study aimed to investigate the effects of resveratrol on kidney function in a rat model of uremia and the expression of heat shock proteins. MATERIAL AND METHODS The rat model of uremia was developed by 5/6 nephrectomy of Sprague-Dawley rats. The Hsp70 inhibitor MKT-077, a rhodacyanine dye, was used. The study groups included rats with sham surgery (the sham group), the rat model of uremia (the model group), the solvent-treated control group (the control group), the rat model treated with resveratrol group (the resveratrol group), the rat model treated with MKT-077 (the MKT-077 group), and the resveratrol+MKT-077 group. Kidney tissues were studied histologically. Renal cell apoptosis was detected by the TUNEL method. Expression of p53, Bax, and Bcl-2 mRNA and protein were detected by quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and immunohistochemistry, respectively. RESULTS Compared with the sham group, the expression levels of heat shock proteins Hsp70, Hsp90, Hsp27, Hsp25, Hsp40, and Hsp60 in the kidney of the rat model group increased to different degrees. Compared with the model group, the Hsp70 levels in the resveratrol group were significantly increased (p<0.05). Compared with the model group, treatment with MKT-077 reduced the survival rate of rats, which was increased following resveratrol treatment. Compared with the resveratrol group, renal function in the resveratrol+MKT-077 group was significantly reduced (p<0.05). CONCLUSIONS In a rat model of uremia, resveratrol reduced renal injury and improved both renal function and survival, which were associated with increased expression of Hsp70.


HSP70 Heat-Shock Proteins/metabolism , Kidney/injuries , Kidney/metabolism , Resveratrol/therapeutic use , Uremia/complications , Animals , Apoptosis/drug effects , Apoptosis/genetics , Blood Urea Nitrogen , Creatinine/blood , HSP70 Heat-Shock Proteins/genetics , Kidney/pathology , Kidney/physiopathology , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Resveratrol/pharmacology , Survival Analysis , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Uremia/blood , Uremia/genetics , Uremia/physiopathology , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
13.
Biosci Rep ; 39(12)2019 12 20.
Article En | MEDLINE | ID: mdl-31789348

Endothelial dysfunction and vitamin D deficiency are prevalent in patients with cardiovascular disease (CVD) and chronic kidney disease (CKD). Both are risk factors for cardiovascular events in patients with CKD. No studies have investigated the effect of nutritional forms of vitamin D on endothelial function in earlier stages of CKD, when vascular endothelium may be more amenable to this therapy. We studied the effect of ergocalciferol in a pre-clinical model of mild uraemia. Male Wistar rats underwent either a 5/6th nephrectomy or sham surgery. Four weeks after the final stage of the surgery, these two groups were randomly allocated to placebo or an oral dose of 1000 iu of ergocalcfierol at day 7 and 2 pre sacrifice. Vascular responses to acetylcholine, Spermine NONOate and phenylephrine were determined in aortic rings. Blood pressure, calcium, phosphate and parathyroid hormone were measured in all groups. Ergocalciferol significantly improved the endothelium-dependent responses to acetylcholine and overcame the blunting of the contractile response to phenylephrine seen in uraemic animals. Ergocalciferol improved the contractile response to potassium chloride in uraemic, but not sham animals. All effects occurred independently of changes to calcium, phosphate, parathyroid hormone and systolic blood pressure. There were no differences in endothelium-independent relaxation to Spermine NONOate. In summary, in a model of mild uraemia, ergocalciferol improved vasodilator and vasoconstrictor tone independently of blood pressure and bone mineral parameters suggesting a direct effect of ergocalciferol on the endothelium.


Ergocalciferols/pharmacology , Uremia/drug therapy , Vasoconstrictor Agents/pharmacology , Vasodilation/drug effects , Animals , Aorta/drug effects , Aorta/pathology , Disease Models, Animal , Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Humans , Parathyroid Hormone/genetics , Rats , Renal Insufficiency/drug therapy , Renal Insufficiency/pathology , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/pathology , Uremia/genetics , Uremia/pathology , Vitamin D/genetics , Vitamin D Deficiency/drug therapy , Vitamin D Deficiency/genetics , Vitamin D Deficiency/pathology
14.
J Cell Physiol ; 234(12): 21825-21837, 2019 12.
Article En | MEDLINE | ID: mdl-31297803

Uremia largely results from the accumulation of organic waste products normally cleared by the kidneys, which commonly accompanies kidney failure and chronic kidney disease. However, genetic investigations in a uremia remain largely unclear. This study aimed to determine the expression patterns of distal-less homeobox 5 (DLX5) in uremia rat model and further to study its effects on glomerulosclerosis and interstitial fibrosis. Uremic expression chip was applied to screen differentially expressed genes in uremia. Next, we used small interfering RNA-mediated RNA interference to specifically silence DLX5 in experimental uremic rats to understand the regulatory mechanism of DLX5. To understand effect of Notch1 signaling pathway in uremia, we also treated experimental uremic rats with γ-secretase inhibitor (GSI), an inhibitor of Notch1 signaling pathway. The expression of fibronectin (FN), laminin (LN), transforming growth factor-ß1 (TGF-ß1), Hes1, Hes5, and Jagged2 was determined. The semiquantitative assessment was applied to verify the effects of DLX5 on glomerulosclerosis. In the uremic expression chip, we found that DLX5 was upregulated in uremia samples, and considered to regulate the Notch signaling pathway. We found that small interfering RNA-mediated DLX5 inhibition or Notch1 signaling pathway inhibitory treatment relieved and delayed the kidney injury and glomerulosclerosis in uremia. Meanwhile, inhibition of DLX5 or Nothch1 signaling pathway reduced expression of FN, LN, Nothch1, TGF-ß1, Hes1, Hes5, and Jagged2. Intriguingly, we discovered that Notch1 signaling pathway was inhibited after silencing DLX5. In conclusion, these findings highlight that DLX5 regulates Notch signaling, which may, in turn, promote complications of uremia such as kidney fibrosis, providing a novel therapeutic target for treating uremia.


Homeodomain Proteins/genetics , Kidney Diseases/genetics , Receptors, Notch/metabolism , Transcription Factors/genetics , Transcriptome/genetics , Animals , Genes, Homeobox/genetics , Kidney/pathology , Kidney Diseases/pathology , Male , Rats, Wistar , Uremia/genetics , Uremia/pathology
15.
Int J Mol Sci ; 20(9)2019 May 08.
Article En | MEDLINE | ID: mdl-31071929

(1) The beneficial effects of hydrogen sulfide (H2S) on the cardiovascular and nervous system have recently been re-evaluated. It has been shown that lanthionine, a side product of H2S biosynthesis, previously used as a marker for H2S production, is dramatically increased in circulation in uremia, while H2S release is impaired. Thus, lanthionine could be classified as a novel uremic toxin. Our research was aimed at defining the mechanism(s) for lanthionine toxicity. (2) The effect of lanthionine on H2S release was tested by a novel lead acetate strip test (LAST) in EA.hy926 cell cultures. Effects of glutathione, as a redox agent, were assayed. Levels of sulfane sulfur were evaluated using the SSP4 probe and flow cytometry. Protein content and glutathionylation were analyzed by Western Blotting and immunoprecipitation, respectively. Gene expression and miRNA levels were assessed by qPCR. (3) We demonstrated that, in endothelial cells, lanthionine hampers H2S release; reduces protein content and glutathionylation of transsulfuration enzyme cystathionine-ß-synthase; modifies the expression of miR-200c and miR-423; lowers expression of vascular endothelial growth factor VEGF; increases Ca2+ levels. (4) Lanthionine-induced alterations in cell cultures, which involve both sulfur amino acid metabolism and calcium homeostasis, are consistent with uremic dysfunctional characteristics and further support the uremic toxin role of this amino acid.


Alanine/analogs & derivatives , Calcium/metabolism , Renal Insufficiency, Chronic/drug therapy , Sulfides/pharmacology , Uremia/drug therapy , Alanine/chemistry , Alanine/pharmacology , Amino Acids, Sulfur/drug effects , Amino Acids, Sulfur/metabolism , Cell Line , Cystathionine beta-Synthase/genetics , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Flow Cytometry , Gene Expression Regulation/drug effects , Glutathione/metabolism , Humans , Hydrogen Sulfide/metabolism , MicroRNAs/genetics , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/genetics , Oxidation-Reduction , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/metabolism , Sulfides/chemistry , Uremia/genetics , Uremia/metabolism , Vascular Endothelial Growth Factor A/genetics
16.
Nephron ; 141(1): 61-74, 2019.
Article En | MEDLINE | ID: mdl-30326474

BACKGROUND: Decreased levels of leucocytic angiotensin converting enzyme 2 (ACE2) relate to atherosclerosis in patients with chronic kidney disease (CKD). Recently, micro RNA 421 (miR-421) was found to target and down-regulate ACE2 in human cardiac myofibroblasts. In this study, we investigated the correlation between serum levels of miR-421 and ACE2 transcripts in circulating leukocytes of healthy individuals (NP), CKD (3-5) and haemodialysis (HD) patients. Furthermore, we tested the possible interaction between miR-421 and 3'-UTR of ACE2 under normal and uremic milieu. METHODS: The levels of circulating miR-421, serum Ang1-7 and expression of leucocytic ACE2, ACE, MASR, AT1R and AT2R were investigated in 16 CKD3-5 (76 ± 10 years), 32 HD patients (65 ± 13 years) and 23 NP (51 ± 5 years) by employment of specific primers, TaqMan and competitive enzyme-linked immunosorbent assay assays. Interaction between miR-421 and ACE2-3'-UTR was investigated on THP-1 cells by employment of normal and uremic sera, reporter vectors and miR-421 inhibitor. Effects of uremic toxins indoxyl sulphate, p-cresol and p-cresyl sulphate on ACE2 and miR-421 levels were investigated in THP-1 monocytes. RESULTS: The levels of serum miR-421 were significantly elevated, while Ang1-7 was significantly decreased in CKD3-5 and HD patients as compared with NP. Serum Ang1-7 correlated positively with leucocytic ACE2 (r2 = 0.213, p < 0.001). We found a significant and inverse correlation between the levels of circulating miR-421 and the expression of leucocytic ACE2 (r2 = 0.223, p < 0.0001). Further significant and positive correlations could be demonstrated between miR-421 and the transcripts of leucocytic AT1R (r2 = 0.094, p < 0.05) or eGFR (r2 = 0.231, p < 0.0001) or CRP (r2 = 0.092, p < 0.01). We found no correlations between miR-421 and ACE or AT2R or MASR expression. Treatment with miR-421 or uremic serum led to noticeable decrease of reporter 3'UTR-ACE2. Anti-miR-421 treatment resulted in the up-regulation of ACE2 protein. All uremic toxins tested were able to significantly elevate miR-421 levels and decrease the monocytic ACE2 transcripts. CONCLUSIONS: Uremic patients show an enhanced expression of serum miR-421 as compared to healthy individuals. A strong association of circulating miR-421 with decreased transcripts of ACE2 may contribute to the low expression of the enzyme in leukocytes of CKD patients, further supporting the development of atherosclerotic events.


Leukocytes/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Peptidyl-Dipeptidase A/blood , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/genetics , 3' Untranslated Regions/genetics , Aged , Aged, 80 and over , Angiotensin I/blood , Angiotensin-Converting Enzyme 2 , Cell Line , Female , Humans , Male , Middle Aged , Peptide Fragments/blood , Proto-Oncogene Mas , Renal Dialysis , Uremia/genetics , Uremia/metabolism
17.
Atherosclerosis ; 280: 28-36, 2019 01.
Article En | MEDLINE | ID: mdl-30453118

BACKGROUND AND AIMS: Chronic kidney disease (CKD) is strongly associated with a high burden of cardiovascular morbidity and mortality. Therefore, we aimed to characterize the putative role of microRNAs (miR)s in uremic vascular remodelling and endothelial dysfunction. METHODS: We investigated the expression pattern of miRs in two independent end-stage renal disease (ESRD) cohorts and in the animal model of uremic DBA/2 mice via quantitative RT-PCR. Moreover, DBA/2 mice were treated with intravenous injections of synthetic miR-142-3p mimic and were analysed for functional and morphological vascular changes by mass spectrometry and wire myography. RESULTS: The expression pattern of miRs was regulated in ESRD patients and was reversible after kidney transplantation. Out of tested miRs, only blood miR-142-3p was negatively associated with carotid-femoral pulse-wave velocity in CKD 5D patients. We validated these findings in a murine uremic model and found similar suppression of miR-142-3p as well as decreased acetylcholine-mediated vascular relaxation of the aorta. Therefore, we designed experiments to restore bioavailability of aortic miR-142-3p in vivo via intravenous injection of synthetic miR-142-3p mimic. This intervention restored acetylcholine-mediated vascular relaxation. CONCLUSIONS: Taken together, we provide compelling evidence, both in humans and in mice, that miR-142-3p constitutes a potential pharmacological agent to prevent endothelial dysfunction and increased arterial stiffness in ESRD.


Acetylcholine/metabolism , Endothelium, Vascular/pathology , MicroRNAs/metabolism , Uremia/blood , Uremia/genetics , Vascular Stiffness , Adult , Animals , Aorta/metabolism , Case-Control Studies , Female , Humans , Kidney Failure, Chronic/metabolism , Kidney Transplantation , Male , Mice , Mice, Inbred DBA , Middle Aged , Muscle, Smooth, Vascular/metabolism , Peritoneal Dialysis , Phenotype , Prospective Studies , Pulse Wave Analysis , Renal Dialysis
18.
Cytokine ; 106: 19-28, 2018 06.
Article En | MEDLINE | ID: mdl-29529595

Osteoprotegerin (OPG), receptor activator of NF-κB ligand (RANKL), and parathyroid hormone (PTH) play a central role in the regulation of bone turnover in chronic kidney disease (CKD), but their influence on bone mineral density (BMD) and strength remains unclear, particularly in children. We studied the clinical significance of OPG and RANKL in relation to PTH, femur weight, BMD, and bone biomechanical properties in growing rats after one month (CKD-1) and three months (CKD-3) of surgically-induced mild CKD. Gene expression of parathyroid hormone 1 receptor (PTH1R) and activating transcription factor 4 (ATF4), major regulators of anabolic PTH response in bone, was also determined. Serum PTH and bone PTH1R/ATF4 expression was elevated in CKD-3 compared with other groups, and it positively correlated with femur weight, BMD, and the biomechanical properties of the femoral diaphysis reflecting cortical bone strength. In contrast, bone RANKL/OPG ratios were decreased in CKD-3 rats compared with other groups, and they were inversely correlated with PTH and the other abovementioned bone parameters. However, the PTH-PTH1R-ATF4 axis exerted an unfavorable effect on the biomechanical properties of the femoral neck. In conclusion, this study showed for the first time an inverse association between serum PTH and the bone RANKL/OPG system in growing rats with mild CKD. A decrease in the RANKL/OPG ratio, associated with PTH-dependent activation of the anabolic PTH1R/ATF4 pathway, seems to be responsible for the unexpected, beneficial effect of PTH on cortical bone accrual and strength. Simultaneously, impaired biomechanical properties of the femoral neck were observed, making this bone site more susceptible to fractures.


Bone and Bones/metabolism , Osteoprotegerin/metabolism , Parathyroid Hormone/metabolism , RANK Ligand/metabolism , Receptor, Parathyroid Hormone, Type 1/metabolism , Uremia/metabolism , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Animals , Biomechanical Phenomena , Bone Density , Femur/metabolism , Gene Expression Regulation , Male , Organ Size , Osteoprotegerin/blood , Parathyroid Hormone/blood , RANK Ligand/blood , Rats, Wistar , Receptor, Parathyroid Hormone, Type 1/genetics , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/diagnostic imaging , Renal Insufficiency, Chronic/pathology , Uremia/blood , Uremia/genetics , X-Ray Microtomography
19.
PLoS One ; 13(1): e0190820, 2018.
Article En | MEDLINE | ID: mdl-29304096

Hyperphosphatemia and vascular calcification are frequent complications of chronic renal failure and bone morphogenetic protein 7 (BMP7) has been shown to protect against development of vascular calcification in uremia. The present investigation examined the potential reversibility of established uremic vascular calcification by treatment of uremic rats with BMP7. A control model of isogenic transplantation of a calcified aorta from uremic rats into healthy littermates examined whether normalization of the uremic environment reversed vascular calcification. Uremia and vascular calcification were induced in rats by 5/6 nephrectomy, high phosphate diet and alfacalcidol treatment. After 14 weeks severe vascular calcification was present and rats were allocated to BMP7, vehicle or aorta transplantation. BMP7 treatment caused a significant decrease of plasma phosphate to 1.56 ± 0.17 mmol/L vs 2.06 ± 0.34 mmol/L in the vehicle group even in the setting of uremia and high phosphate diet. Uremia and alfacalcidol resulted in an increase in aortic expression of genes related to fibrosis, osteogenic transformation and extracellular matrix calcification, and the BMP7 treatment resulted in a decrease in the expression of profibrotic genes. The total Ca-content of the aorta was however unchanged both in the abdominal aorta: 1.9 ± 0.6 µg/mg tissue in the vehicle group vs 2.2 ± 0.6 µg/mg tissue in the BMP7 group and in the thoracic aorta: 71 ± 27 µg/mg tissue in the vehicle group vs 54 ± 18 µg/mg tissue in the BMP7 group. Likewise, normalization of the uremic environment by aorta transplantation had no effect on the Ca-content of the calcified aorta: 16.3 ± 0.6 µg/mg tissue pre-transplantation vs 15.9 ± 2.3 µg/mg tissue post-transplantation. Aortic expression of genes directly linked to extracellular matrix calcification was not affected by BMP7 treatment, which hypothetically might explain persistent high Ca-content in established vascular calcification. The present results highlight the importance of preventing the development of vascular calcification in chronic kidney disease. Once established, vascular calcification persists even in the setting when hyperphosphatemia or the uremic milieu is abolished.


Bone Morphogenetic Protein 7/pharmacology , Gene Expression Regulation/drug effects , Uremia/drug therapy , Vascular Calcification/drug therapy , Animals , Aorta/drug effects , Aorta/metabolism , Bone Morphogenetic Protein 7/therapeutic use , Chronic Disease , Fibrosis , Male , Phosphates/blood , Rats , Real-Time Polymerase Chain Reaction , Uremia/genetics , X-Ray Microtomography
20.
Front Biosci (Landmark Ed) ; 23(2): 348-387, 2018 01 01.
Article En | MEDLINE | ID: mdl-28930551

We mined novel uremic toxin (UT) metabolomics/gene databases, and analyzed the expression changes of UT receptors and UT synthases in chronic kidney disease (CKD) and cardiovascular disease (CVD). We made the following observations: 1) UTs represent only 1/80th of human serum small-molecule metabolome; 2) Some UTs are increased in CKD and CVD; 3) UTs either induce or suppress the expression of inflammatory molecules; 4) The expression of UT genes is significantly modulated in CKD patients, and coronary artery disease (CAD) patients; 5) The expression of UT genes is upregulated by caspase-1 and TNF-alpha pathways but is inhibited in regulatory T cells. These results demonstrate that UTs are selectively increased, and serve as danger signal-associated molecular patterns (DAMPs) and homeostasis-associated molecular patterns (HAMPs) that modulate inflammation. These results also show that some UT genes are upregulated in CKD and CAD via caspase-1/inflammatory cytokine pathways, rather than by purely passive accumulation.


Cardiovascular Diseases/metabolism , Homeostasis , Toxins, Biological/metabolism , Uremia/metabolism , Alarmins/genetics , Alarmins/metabolism , Cardiovascular Diseases/genetics , Caspase 1/genetics , Caspase 1/metabolism , Cytokines/genetics , Cytokines/metabolism , Gene Expression Profiling/methods , Humans , Inflammation Mediators/metabolism , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/metabolism , Signal Transduction/genetics , Toxins, Biological/genetics , Uremia/genetics
...