Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.003
Filter
1.
Clin J Am Soc Nephrol ; 16(10): 1531-1538, 2021 10.
Article in English | MEDLINE | ID: mdl-34233922

ABSTRACT

BACKGROUND AND OBJECTIVES: Residual native kidney function confers health benefits in patients on dialysis. It can facilitate control of extracellular volume and inorganic ion concentrations. Residual kidney function can also limit the accumulation of uremic solutes. This study assessed whether lower plasma concentrations of uremic solutes were associated with residual kidney function in pediatric patients on peritoneal dialysis. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: Samples were analyzed from 29 pediatric patients on peritoneal dialysis, including 13 without residual kidney function and ten with residual kidney function. Metabolomic analysis by untargeted mass spectrometry compared plasma solute levels in patients with and without residual kidney function. Dialytic and residual clearances of selected solutes were also measured by assays using chemical standards. RESULTS: Metabolomic analysis showed that plasma levels of 256 uremic solutes in patients with residual kidney function averaged 64% (interquartile range, 51%-81%) of the values in patients without residual kidney function who had similar total Kt/Vurea. The plasma levels were significantly lower for 59 of the 256 solutes in the patients with residual kidney function and significantly higher for none. Assays using chemical standards showed that residual kidney function provides a higher portion of the total clearance for nonurea solutes than it does for urea. CONCLUSIONS: Concentrations of many uremic solutes are lower in patients on peritoneal dialysis with residual kidney function than in those without residual kidney function receiving similar treatment as assessed by Kt/Vurea.


Subject(s)
Kidney Diseases/therapy , Kidney Function Tests , Kidney/physiopathology , Mass Spectrometry , Metabolome , Metabolomics , Peritoneal Dialysis , Uremia/therapy , Adolescent , Age Factors , Biomarkers/blood , Child , Child, Preschool , Female , Humans , Infant , Kidney Diseases/blood , Kidney Diseases/diagnosis , Kidney Diseases/physiopathology , Male , Peritoneal Dialysis/adverse effects , Predictive Value of Tests , Treatment Outcome , United States , Uremia/blood , Uremia/diagnosis , Uremia/physiopathology
2.
Toxicol Lett ; 350: 133-142, 2021 Oct 10.
Article in English | MEDLINE | ID: mdl-34303789

ABSTRACT

Non-dialysable protein-bound uremic toxins (PBUTs) contribute to the development of cardiovascular disease (CVD) in chronic kidney disease (CKD) and vice versa. PBUTs have been shown to alter sphingolipid imbalance. Dihydroceramide desaturase 1 (Des1) is an important gatekeeper enzyme which controls the non-reversible conversion of sphingolipids, dihydroceramide, into ceramide. The present study assessed the effect of Des1 inhibition on PBUT-induced cardiac and renal effects in vitro, using a selective Des1 inhibitor (CIN038). Des1 inhibition attenuated hypertrophy in neonatal rat cardiac myocytes and collagen synthesis in neonatal rat cardiac fibroblasts and renal mesangial cells induced by the PBUTs, indoxyl sulfate and p-cresol sulfate. This is at least attributable to modulation of NF-κB signalling and reductions in ß-MHC, Collagen I and TNF-α gene expression. Lipidomic analyses revealed Des1 inhibition restored C16-dihydroceramide levels reduced by indoxyl sulfate. In conclusion, PBUTs play a critical role in mediating sphingolipid imbalance and inflammatory responses in heart and kidney cells, and these effects were attenuated by Des1 inhibition. Therefore, sphingolipid modifying agents may have therapeutic potential for the treatment of CVD and CKD and warrant further investigation.


Subject(s)
Cardiovascular Diseases/chemically induced , Oxidoreductases/therapeutic use , Sphingolipids/metabolism , Toxins, Biological/adverse effects , Toxins, Biological/metabolism , Uremia/blood , Uremia/physiopathology , Animals , Enzyme Inhibitors/therapeutic use , Humans , Models, Animal , Rats , Rats, Sprague-Dawley , Renal Insufficiency, Chronic/complications , Sphingolipids/blood , Toxins, Biological/blood
3.
Am J Physiol Renal Physiol ; 321(1): F106-F119, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34121452

ABSTRACT

Preclinical animal models of chronic kidney disease (CKD) are critical to investigate the underlying mechanisms of disease and to evaluate the efficacy of novel therapeutics aimed to treat CKD-associated pathologies. The objective of the present study was to compare the adenine diet and 5/6 nephrectomy (Nx) CKD models in mice. Male and female 10-wk-old C57BL/6J mice (n = 5-9 mice/sex/group) were randomly allocated to CKD groups (0.2-0.15% adenine-supplemented diet or 5/6 Nx surgery) or the corresponding control groups (casein diet or sham surgery). Following the induction of CKD, the glomerular filtration rate was reduced to a similar level in both adenine and 5/6 Nx mice (adenine diet-fed male mice: 81.1 ± 41.9 µL/min vs. 5/6 Nx male mice: 160 ± 80.9 µL/min, P = 0.5875; adenine diet-fed female mice: 112.9 ± 32.4 µL/min vs. 5/6 Nx female mice: 107.0 ± 45.7 µL/min, P = 0.9995). Serum metabolomics analysis indicated that established uremic toxins were robustly elevated in both CKD models, although some differences were observed between CKD models (i.e., p-cresol sulfate). Dysregulated phosphate homeostasis was observed in the adenine model only, whereas Ca2+ homeostasis was disturbed in male mice with both CKD models. Compared with control mice, muscle mass and myofiber cross-sectional areas of the extensor digitorum longus and soleus muscles were ∼18-24% smaller in male CKD mice regardless of the model but were not different in female CKD mice (P > 0.05). Skeletal muscle mitochondrial respiratory function was significantly decreased (19-24%) in CKD mice in both models and sexes. These findings demonstrate that adenine diet and 5/6 Nx models of CKD have similar levels of renal dysfunction and skeletal myopathy. However, the adenine diet model demonstrated superior performance with regard to mortality (∼20-50% mortality for 5/6 Nx vs. 0% mortality for the adenine diet, P < 0.05 for both sexes) compared with the 5/6 Nx surgical model.NEW & NOTEWORTHY Numerous preclinical models of chronic kidney disease have been used to evaluate skeletal muscle pathology; however, direct comparisons of popular models are not available. In this study, we compared adenine-induced nephropathy and 5/6 nephrectomy models. Both models produced equivalent levels of muscle atrophy and mitochondrial impairment, but the adenine model exhibited lower mortality rates, higher consistency in uremic toxin levels, and dysregulated phosphate homeostasis compared with the 5/6 nephrectomy model.


Subject(s)
Adenine/pharmacology , Glomerular Filtration Rate/genetics , Muscle, Skeletal/metabolism , Renal Insufficiency, Chronic/metabolism , Animals , Disease Models, Animal , Kidney/metabolism , Kidney/pathology , Male , Mice, Inbred C57BL , Muscle, Skeletal/pathology , Muscular Diseases/pathology , Muscular Diseases/physiopathology , Nephrectomy/methods , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/pathology , Uremia/physiopathology
4.
Sci Rep ; 11(1): 12159, 2021 06 09.
Article in English | MEDLINE | ID: mdl-34108522

ABSTRACT

Patients with chronic kidney disease (CKD) commonly exhibit hypercoagulability. Increased levels of uremic toxins cause thrombogenicity by increasing tissue factor (TF) expression and activating the extrinsic coagulation cascade. TF is induced in monocytes and macrophages under pathological conditions, such as inflammatory diseases. However, the role of monocyte myeloid cell TF in CKD progression remains unclear. We aimed to clarify this issue, and the present study found that patients with CKD had elevated levels of D-dimer, a marker of fibrin degradation, which was associated with decreased estimated glomerular filtration rate and increased serum levels of uremic toxins, such as indoxyl sulfate. In vitro studies showed that several uremic toxins increased cellular TF levels in monocytic THP-1 cells. Mice with TF specifically deleted in myeloid cells were fed an adenine diet to cause uremic kidney injury. Myeloid TF deletion reduced tubular injury and pro-inflammatory gene expression in the kidneys of adenine-induced CKD but did not improve renal function as measured by plasma creatinine or blood urea nitrogen. Collectively, our findings suggest a novel concept of pathogenesis of coagulation-mediated kidney injury, in which elevated TF levels in monocytes under uremic conditions is partly involved in the development of CKD.


Subject(s)
Adenine/toxicity , Kidney Tubules/pathology , Myeloid Cells/metabolism , Renal Insufficiency, Chronic/prevention & control , Thromboplastin/physiology , Toxins, Biological/metabolism , Uremia/physiopathology , Animals , Fibrin Fibrinogen Degradation Products/metabolism , Glomerular Filtration Rate , Humans , Kidney Tubules/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology
5.
J Cell Mol Med ; 25(12): 5602-5614, 2021 06.
Article in English | MEDLINE | ID: mdl-33960650

ABSTRACT

Extracellular vesicles (EV) function as messengers between endothelial cells (EC) and vascular smooth muscle cells (VSMC). Since chronic kidney disease (CKD) increases the risk for vascular calcifications, we investigated whether EV derived from uraemic milieu-stimulated EC and derived from uraemic rats impact the osteogenic transdifferentiation/calcification of VSMC. For that purpose, human EC were treated with urea and indoxyl sulphate or left untreated. Experimental uraemia in rats was induced by adenine feeding. 'Uraemic' and control EV (EVUR ; EVCTRL ) were isolated from supernatants and plasma by using an exosome isolation reagent. Rat VSMC were treated with a pro-calcifying medium (CM) with or without EV supplementation. Gene expressions, miRNA contents and protein expressions were determined by qPCR and Western blots, respectively. Calcifications were determined by colorimetric assays. Delivery of miRNA inhibitors/mimics to EV and siRNA to VSMC was achieved via transfection. EVCTRL and EVUR differed in size and miRNA contents. Contrary to EVCTRL , EC- and plasma-derived EVUR significantly increased the pro-calcifying effects of CM, including altered gene expressions of osterix, runx2, osteocalcin and SM22α. Further, EVUR enhanced the protein expression of the phosphate transporter PiT-1 in VSMC and induced a phosphorylation of AKT and ERK. Knock down of PiT-1 and individual inhibition of AKT and ERK signalling in VSMC blocked the pro-calcifying effects of EVUR . Similar effects were achieved by inhibition of miR-221/-222 and mimicking of miR-143/-145 in EVUR . In conclusion, EVUR might represent an additional puzzle piece of the complex pathophysiology of vascular calcifications in CKD.


Subject(s)
Cell Transdifferentiation , Extracellular Vesicles/pathology , Muscle, Smooth, Vascular/pathology , Proto-Oncogene Proteins c-akt/metabolism , Transcription Factor Pit-1/metabolism , Uremia/physiopathology , Vascular Calcification/pathology , Animals , Endothelial Cells/metabolism , Endothelial Cells/pathology , Extracellular Vesicles/metabolism , Gene Expression Regulation , Humans , Muscle, Smooth, Vascular/metabolism , Osteogenesis , Proto-Oncogene Proteins c-akt/genetics , Rats , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Transcription Factor Pit-1/genetics , Vascular Calcification/metabolism
6.
Toxins (Basel) ; 13(4)2021 04 19.
Article in English | MEDLINE | ID: mdl-33921862

ABSTRACT

The retention of uremic toxins and their pathological effects occurs in the advanced phases of chronic kidney disease (CKD), mainly in stage 5, when the implementation of conventional thrice-weekly hemodialysis is the prevalent and life-saving treatment. However, the start of hemodialysis is associated with both an acceleration of the loss of residual kidney function (RKF) and the shift to an increased intake of proteins, which are precursors of uremic toxins. In this phase, hemodialysis treatment is the only way to remove toxins from the body, but it can be largely inefficient in the case of high molecular weight and/or protein-bound molecules. Instead, even very low levels of RKF are crucial for uremic toxins excretion, which in most cases are protein-derived waste products generated by the intestinal microbiota. Protection of RKF can be obtained even in patients with end-stage kidney disease (ESKD) by a gradual and soft shift to kidney replacement therapy (KRT), for example by combining a once-a-week hemodialysis program with a low or very low-protein diet on the extra-dialysis days. This approach could represent a tailored strategy aimed at limiting the retention of both inorganic and organic toxins. In this paper, we discuss the combination of upstream (i.e., reduced production) and downstream (i.e., increased removal) strategies to reduce the concentration of uremic toxins in patients with ESKD during the transition phase from pure conservative management to full hemodialysis treatment.


Subject(s)
Diet, Protein-Restricted , Kidney Failure, Chronic/therapy , Renal Dialysis , Toxins, Biological/blood , Uremia/therapy , Biomarkers/blood , Combined Modality Therapy , Diet, Protein-Restricted/adverse effects , Disease Progression , Humans , Kidney Failure, Chronic/blood , Kidney Failure, Chronic/diagnosis , Kidney Failure, Chronic/physiopathology , Renal Dialysis/adverse effects , Treatment Outcome , Uremia/blood , Uremia/diagnosis , Uremia/physiopathology
7.
J Stroke Cerebrovasc Dis ; 30(9): 105819, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33926796

ABSTRACT

Patients with renal disease have increased rates of admission to the neurological intensive care unit related to overlapping risk factors for renal and cerebrovascular disease as well as unique risks associated with renal dysfunction alone. Management of acute neurological injury in these patients requires individualized attention to diagnostic and management factors as they relate to coagulopathy, disorders of immune function, encephalopathy and renal replacement modalities. Careful consideration of these brain-kidney interactions is necessary to optimize care for this special patient population and improve neurological and renal outcomes.


Subject(s)
Infections/therapy , Intensive Care Units , Intracranial Hemorrhages/therapy , Ischemic Stroke/therapy , Renal Dialysis , Renal Insufficiency, Chronic/therapy , Uremia/therapy , Brain/physiopathology , Humans , Infections/diagnosis , Infections/mortality , Infections/physiopathology , Intracranial Hemorrhages/diagnosis , Intracranial Hemorrhages/mortality , Intracranial Hemorrhages/physiopathology , Ischemic Stroke/diagnosis , Ischemic Stroke/mortality , Ischemic Stroke/physiopathology , Kidney/physiopathology , Recovery of Function , Renal Dialysis/adverse effects , Renal Dialysis/mortality , Renal Insufficiency, Chronic/diagnosis , Renal Insufficiency, Chronic/mortality , Renal Insufficiency, Chronic/physiopathology , Risk Assessment , Risk Factors , Treatment Outcome , Uremia/diagnosis , Uremia/mortality , Uremia/physiopathology
8.
Int J Mol Sci ; 22(6)2021 Mar 11.
Article in English | MEDLINE | ID: mdl-33799511

ABSTRACT

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.


Subject(s)
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
9.
J Stroke Cerebrovasc Dis ; 30(9): 105651, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33581988

ABSTRACT

Chronic kidney disease and seizures often co-exist. When seizures are provoked in patients with kidney disease, their treatment poses a particular challenge. Seizures may be provoked in the context of uremia, and toxic substances associated with uremic encephalopathy. In that case, the mainstay of therapy is to treat the uremia before consideration for anticonvulsant therapy. Treatment of seizures in the setting of chronic kidney disease requires special attention to selection of anticonvulsant medications and knowledge of the altered pharmacokinetics of these medications, which may require special titration schedule in that setting. The purpose of this review is to summarize the current knowledge about inter-relation of seizures and kidney disease. The review will also help practitioners who treat patients with renal failure and coexisting seizures in choosing the best treatment options.


Subject(s)
Anticonvulsants/therapeutic use , Brain Waves/drug effects , Brain/drug effects , Epilepsy/drug therapy , Kidney Diseases/therapy , Renal Dialysis , Uremia/therapy , Anticonvulsants/adverse effects , Anticonvulsants/pharmacokinetics , Brain/physiopathology , Electroencephalography , Epilepsy/diagnosis , Epilepsy/etiology , Epilepsy/physiopathology , Humans , Kidney Diseases/complications , Kidney Diseases/diagnosis , Kidney Diseases/physiopathology , Male , Middle Aged , Predictive Value of Tests , Renal Dialysis/adverse effects , Risk Factors , Treatment Outcome , Uremia/complications , Uremia/diagnosis , Uremia/physiopathology
10.
Sci Rep ; 11(1): 512, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33436654

ABSTRACT

Uraemic toxins increase in serum parallel to a decline in the glomerular filtration rate and the development of sarcopenia in patients with chronic kidney disease (CKD). This study analyses the role of uraemic toxins in sarcopenia at different stages of CKD, evaluating changes in the muscular regeneration process. Cultured C2C12 cells were incubated with a combination of indoxyl sulphate and p-cresol at high doses (100 µg/mL) or low doses (25 µg/mL and 10 µg/mL) resembling late or early CKD stages, respectively. Cell proliferation (analysed by scratch assays and flow cytometry) was inhibited only by high doses of uraemic toxins, which inactivated the cdc2-cyclin B complex, inhibiting mitosis and inducing apoptosis (analysed by annexin V staining). By contrast, low doses of uraemic toxins did not affect proliferation, but reduced myogenic differentiation, primed with 2% horse serum, by inhibiting myogenin expression and promoting fibro-adipogenic differentiation. Finally, to assess the in vivo relevance of these results, studies were performed in gastrocnemii from uraemic rats, which showed higher collagen expression and lower myosin heavy chain expression than those from healthy rats. In conclusion, uraemic toxins impair the skeletal muscular regeneration process, even at low concentrations, suggesting that sarcopenia can progress from the early stages of CKD.


Subject(s)
Cell Differentiation/drug effects , Cell Proliferation/drug effects , Muscle Development/drug effects , Myoblasts/physiology , Regeneration/drug effects , Toxins, Biological/adverse effects , Uremia/physiopathology , Animals , Cells, Cultured , Disease Models, Animal , Fibrosis , Mice , Muscle, Skeletal/physiology , Rats
11.
Cardiovasc Drugs Ther ; 35(3): 521-532, 2021 06.
Article in English | MEDLINE | ID: mdl-32651897

ABSTRACT

PURPOSE: Chronic kidney disease (CKD) associates with inflammatory and prothrombotic phenotypes, resulting in higher cardiovascular risk. Factor Xa displays functions beyond coagulation, exhibiting proinflammatory effects. The aim of the present study was to investigate whether a direct FXa inhibitor protects from the endothelial dysfunction (ED) caused by uremia. METHODS: Macro (HUVEC) and microvascular (HMEC) endothelial cells (ECs) were exposed to serum from uremic patients or healthy donors, in absence and presence of apixaban (60 ng/ml). We evaluated changes in surface VCAM-1 and ICAM-1, intracellular eNOS, reactive oxygen species (ROS), and von Willebrand Factor (VWF) production by immunofluorescence, reactivity of the extracellular matrix (ECM) towards platelets, and intracellular signaling. RESULTS: ECs exposed to uremic serum triggered dysregulation of all the parameters. Presence of apixaban resulted in decreased expression of VCAM-1 (178 ± 14 to 89 ± 2% on HMEC and 324 ± 71 to 142 ± 25% on HUVEC) and ICAM-1 (388 ± 60 to 111 ± 10% on HMEC and 148 ± 9% to 90 ± 7% on HUVEC); increased eNOS (72 ± 8% to 95 ± 10% on HMEC); normalization of ROS levels (173 ± 21 to 114 ± 13% on HMEC and 165 ± 14 to 127 ± 7% on HUVEC); lower production of VWF (168 ± 14 to 92 ± 4% on HMEC and 151 ± 22 to 99 ± 11% on HUVEC); and decreased platelet adhesion onto ECM (134 ± 22 to 93 ± 23% on HMEC and 161 ± 14 to 117 ± 7% on HUVEC). Apixaban inhibited p38MAPK and p42/44 activation in HUVEC (139 ± 15 to 48 ± 15% and 411 ± 66 to 177 ± 57%, respectively) (p < 0.05 vs control for all parameters). CONCLUSION: Anti-FXa strategies, such as apixaban, prevented ED caused by the uremic milieu, exhibiting anti-inflammatory and antioxidant properties and modulating the reactivity of the ECM.


Subject(s)
Factor Xa Inhibitors/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Pyrazoles/pharmacology , Pyridones/pharmacology , Uremia/physiopathology , Endothelial Cells/drug effects , Extracellular Matrix/drug effects , Humans , Inflammation/physiopathology , Intercellular Adhesion Molecule-1/drug effects , Nitric Oxide Synthase Type III/drug effects , Phenotype , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Vascular Cell Adhesion Molecule-1/drug effects , von Willebrand Factor/drug effects
12.
Curr Vasc Pharmacol ; 19(1): 63-76, 2021.
Article in English | MEDLINE | ID: mdl-32600233

ABSTRACT

Adverse innate immune responses have been implicated in several disease processes, including cardiovascular disease (CVD) and chronic kidney disease (CKD). The monocyte subsets natural killer (NK) cells and natural killer T (NKT) cells are involved in innate immunity. Monocytes subsets are key in atherogenesis and the inflammatory cascade occurring in heart failure. Upregulated activity and counts of proinflammatory CD16+ monocyte subsets are associated with clinical indices of atherosclerosis, heart failure syndromes and CKD. Advanced CKD is a complex state of persistent systemic inflammation characterized by elevated expression of proinflammatory and pro-atherogenic CD14++CD16+ monocytes, which are associated with cardiovascular events and death both in the general population and among patients with CKD. Diminished NK cells and NKT cells counts and aberrant activity are observed in both coronary artery disease and end-stage kidney disease. However, evidence of the roles of NK cells and NKT cells in atherogenesis in advanced CKD is circumstantial and remains to be clarified. This review describes the available evidence regarding the roles of specific immune cell subsets in the pathogenesis of CVD in patients with CKD. Future research is expected to further uncover the links between CKD associated innate immune system dysregulation and accelerated CVD and will ideally be translated into therapeutic targets.


Subject(s)
Cardiovascular Diseases/immunology , Cardiovascular System/immunology , Immune System/immunology , Immunity, Innate , Kidney Failure, Chronic/immunology , Kidney/immunology , Animals , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/physiopathology , Cardiovascular System/metabolism , Cardiovascular System/physiopathology , Humans , Immune System/metabolism , Immune System/physiopathology , Inflammation Mediators/metabolism , Kidney/metabolism , Kidney/physiopathology , Kidney Failure, Chronic/metabolism , Kidney Failure, Chronic/physiopathology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Monocytes/immunology , Monocytes/metabolism , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Prognosis , Uremia/immunology , Uremia/metabolism , Uremia/physiopathology
13.
Transl Res ; 229: 115-134, 2021 03.
Article in English | MEDLINE | ID: mdl-32891787

ABSTRACT

The prevalence of chronic kidney disease (CKD) in the worldwide population is currently estimated between 11% and 13%. Adequate renal clearance is compromised in these patients and the accumulation of a large number of uremic retention solutes results in an irreversible worsening of renal function which can lead to end stage renal disease (ESRD). Approximately three million ESRD patients currently receive renal replacement therapies (RRTs), such as hemodialysis, which only partially restore kidney function, as they are only efficient in removing mainly small, unbound solutes from the circulation while leaving larger and protein-bound uremic toxins (PBUTs) untouched. The accumulation of PBUTs in patients highly increases the risk of cardiovascular events and is associated with higher mortality and morbidity in CKD and ESRD. In this review, we address several strategies currently being explored toward reducing PBUT concentrations, including clinical and medical approaches, therapeutic techniques, and recent developments in RRT technology. These include preservation of renal function, limitation of colon derived PBUTs, oral sorbents, adsorbent RRT technology, and use of albumin displacers. Despite the promising results of the different approaches to promote enhanced removal of a small percentage of the more than 30 identified PBUTs, on their own, none of them provide a treatment with the required efficiency, safety and cost-effectiveness to prevent CKD-related complications and decrease mortality and morbidity in ESRD.


Subject(s)
Kidney Failure, Chronic/therapy , Renal Insufficiency, Chronic/therapy , Renal Replacement Therapy/methods , Toxins, Biological/metabolism , Uremia/therapy , Administration, Oral , Carbon/therapeutic use , Colon/metabolism , Humans , Kidney/physiology , Kidney Failure, Chronic/metabolism , Oxides/therapeutic use , Renal Dialysis/methods , Renal Insufficiency, Chronic/metabolism , Uremia/metabolism , Uremia/physiopathology
14.
Curr Vasc Pharmacol ; 19(1): 55-62, 2021.
Article in English | MEDLINE | ID: mdl-32310050

ABSTRACT

Alterations of fibroblast growth factor 23 (FGF-23) and Klotho levels are considered to be the earliest biochemical abnormality of chronic kidney disease - mineral and bone disease (CKDMBD) syndrome. Moreover, emerging data suggests that the dysregulated FGF-23 and Klotho axis has many effects on the cardiovascular (CV) system and contributes significantly to the increased CV morbidity and mortality rates of CKD patients. This review examines recent evidence on the role of FGF-23 and Klotho in the development and progression of CV complications of uremia namely cardiac hypertrophy, uremic cardiomyopathy, and atherosclerotic and arteriosclerotic vascular lesions. Moreover, the available evidence on their associations with adverse clinical outcomes are summarized. Undoubtedly, more studies are needed to further elucidate the effects of FGF-23 and Klotho on the heart and vessels and to gain insights into their prognostic value as CV risk factors. Finally, large prospective studies are required to test the hypothesis that modification of their levels would have a favourable impact on the unacceptably high mortality rates of these patient populations.


Subject(s)
Cardiovascular Diseases/blood , Cardiovascular System/metabolism , Fibroblast Growth Factors/blood , Glucuronidase/blood , Kidney/metabolism , Renal Insufficiency, Chronic/blood , Uremia/blood , Animals , Biomarkers/blood , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/mortality , Cardiovascular Diseases/physiopathology , Cardiovascular System/physiopathology , Fibroblast Growth Factor-23 , Humans , Kidney/physiopathology , Klotho Proteins , Prognosis , Renal Insufficiency, Chronic/diagnosis , Renal Insufficiency, Chronic/mortality , Renal Insufficiency, Chronic/physiopathology , Risk Factors , Uremia/diagnosis , Uremia/mortality , Uremia/physiopathology
15.
J Stroke Cerebrovasc Dis ; 30(9): 105376, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33214054

ABSTRACT

Patients with renal failure have extremely high cardiovascular risk; in dialysis patients the risk of stroke is increased approximately 10-fold over that in the general population. Reasons include not only a high prevalence of traditional risk factors such as diabetes, hypertension and dyslipidemia, but also the accumulation of toxic substances that are eliminated by the kidneys, so have very high levels in patients with renal failure. These include plasma total homocysteine, asymmetric dimethylarginine, thiocyanate, and toxic products of the intestinal microbiome (Gut-Derived Uremic Toxins; GDUT), which include trimethylamine N- oxide (TMAO), produced from phosphatidylcholine (largely from egg yolk) and carnitine (largely from red meat). Other GDUT are produced from amino acids, largely from meat consumption. Deficiency of vitamin B12 is very common, raises plasma tHcy, and is easily treated. However, cyanocobalamin is toxic in patients with renal failure. To reduce the risk of stroke in renal failure it is important to limit the intake of meat, avoid egg yolk, and use methylcobalamin instead of cyanocobalamin, in addition to folic acid.


Subject(s)
Diet , Dietary Supplements , Kidney/physiopathology , Nutritional Status , Renal Insufficiency/diet therapy , Stroke/prevention & control , Vitamin B 12 Deficiency/diet therapy , Vitamin B 12/therapeutic use , Bacteria/metabolism , Biomarkers/blood , Comorbidity , Diet/adverse effects , Dietary Supplements/adverse effects , Gastrointestinal Microbiome , Homocysteine/blood , Humans , Protective Factors , Renal Insufficiency/diagnosis , Renal Insufficiency/epidemiology , Renal Insufficiency/physiopathology , Risk Assessment , Risk Factors , Stroke/diagnosis , Stroke/epidemiology , Stroke/physiopathology , Treatment Outcome , Uremia/diet therapy , Uremia/epidemiology , Uremia/physiopathology , Vitamin B 12/adverse effects , Vitamin B 12 Deficiency/diagnosis , Vitamin B 12 Deficiency/epidemiology , Vitamin B 12 Deficiency/physiopathology
16.
Toxins (Basel) ; 12(12)2020 Dec 21.
Article in English | MEDLINE | ID: mdl-33371477

ABSTRACT

Uremic vascular calcification (VC) commonly occurs during advanced chronic kidney disease (CKD) and significantly increases cardiovascular morbidity and mortality. Uremic toxins are integral within VC pathogenesis, as they exhibit adverse vascular influences ranging from atherosclerosis, vascular inflammation, to VC. Experimental removal of these toxins, including small molecular (phosphate, trimethylamine-N-oxide), large molecular (fibroblast growth factor-23, cytokines), and protein-bound ones (indoxyl sulfate, p-cresyl sulfate), ameliorates VC. As most uremic toxins share a gut origin, interventions through gastrointestinal tract are expected to demonstrate particular efficacy. The "gastrointestinal decontamination" through the removal of toxin in situ or impediment of toxin absorption within the gastrointestinal tract is a practical and potential strategy to reduce uremic toxins. First and foremost, the modulation of gut microbiota through optimizing dietary composition, the use of prebiotics or probiotics, can be implemented. Other promising strategies such as reducing calcium load, minimizing intestinal phosphate absorption through the optimization of phosphate binders and the inhibition of gut luminal phosphate transporters, the administration of magnesium, and the use of oral toxin adsorbent for protein-bound uremic toxins may potentially counteract uremic VC. Novel agents such as tenapanor have been actively tested in clinical trials for their potential vascular benefits. Further advanced studies are still warranted to validate the beneficial effects of gastrointestinal decontamination in the retardation and treatment of uremic VC.


Subject(s)
Decontamination/methods , Gastrointestinal Tract/metabolism , Renal Insufficiency, Chronic/metabolism , Toxins, Biological/toxicity , Uremia/metabolism , Vascular Calcification/metabolism , Clinical Trials as Topic/methods , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/physiopathology , Humans , Indican/pharmacology , Indican/therapeutic use , Isoquinolines/pharmacology , Isoquinolines/therapeutic use , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/physiopathology , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Uremia/drug therapy , Uremia/physiopathology , Vascular Calcification/drug therapy , Vascular Calcification/physiopathology
17.
Toxins (Basel) ; 12(12)2020 12 11.
Article in English | MEDLINE | ID: mdl-33322362

ABSTRACT

The last years have brought an abundance of data on the existence of a gut-kidney axis and the importance of microbiome in kidney injury. Data on kidney-gut crosstalk suggest the possibility that microbiota alter renal inflammation; we therefore aimed to answer questions about the role of microbiome and gut-derived toxins in acute kidney injury. PubMed and Cochrane Library were searched from inception to October 10, 2020 for relevant studies with an additional search performed on ClinicalTrials.gov. We identified 33 eligible articles and one ongoing trial (21 original studies and 12 reviews/commentaries), which were included in this systematic review. Experimental studies prove the existence of a kidney-gut axis, focusing on the role of gut-derived uremic toxins and providing concepts that modification of the microbiota composition may result in better AKI outcomes. Small interventional studies in animal models and in humans show promising results, therefore, microbiome-targeted therapy for AKI treatment might be a promising possibility.


Subject(s)
Acute Kidney Injury/chemically induced , Gastrointestinal Microbiome/drug effects , Toxins, Biological/toxicity , Uremia/chemically induced , Acute Kidney Injury/microbiology , Acute Kidney Injury/physiopathology , Animals , Gastrointestinal Microbiome/physiology , Humans , Microbiota/drug effects , Microbiota/physiology , Uremia/microbiology , Uremia/physiopathology
18.
Drug Metab Pharmacokinet ; 35(6): 555-562, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33191090

ABSTRACT

The hepatic uptake transporter organic anion transporting polypeptide (OATP) 1B1 is inhibited by some uremic toxins; however, direct inhibition can only partially explain the delayed systemic elimination of substrate drugs in renal failure patients. This study aimed to examine the long-lasting inhibition of OATP1B1 by uremic toxins and their metabolites. Preincubation of HEK293/OATP1B1 cells with 21 uremic toxins resulted in almost no change in the uptake of a typical substrate [3H]estrone-3-sulfate (E1S), although some directly inhibited [3H]E1S uptake. In contrast, preincubation with an indole metabolite, 6-hydroxyindole, reduced [3H]E1S uptake, even after the inhibitor was washed out before [3H]E1S incubation. Such long-lasting inhibition by 6-hydroxyindole was time-dependent and recovered after a 3-h incubation without 6-hydroxyindole. Preincubation with 6-hydroxyindole increased the Km for [3H]E1S uptake with minimal change in Vmax. This was compatible with no change in the cell-surface expression of OATP1B1, as assessed by a biotinylation assay. Preincubation with 6-hydroxyindole reduced [3H]E1S uptake in human hepatocytes without changes in OATP1B1 mRNA. Plasma concentration of 6-hydroxyindole in renal failure patients increased as renal function decreased, but might be insufficient to exhibit potent OATP1B1 inhibition. In conclusion, 6-hydroxyindole is an endogenous long-lasting OATP1B1 inhibitor with elevated plasma concentrations in renal failure patients.


Subject(s)
Hepatocytes/drug effects , Indoles/pharmacology , Liver-Specific Organic Anion Transporter 1/antagonists & inhibitors , Renal Insufficiency/blood , Uremia/blood , Biological Transport , Dose-Response Relationship, Drug , Estrone/analogs & derivatives , Estrone/metabolism , HEK293 Cells , Hepatocytes/metabolism , Humans , Indoles/blood , Kinetics , Liver-Specific Organic Anion Transporter 1/genetics , Liver-Specific Organic Anion Transporter 1/metabolism , Renal Insufficiency/diagnosis , Renal Insufficiency/physiopathology , Up-Regulation , Uremia/diagnosis , Uremia/physiopathology
19.
Toxins (Basel) ; 12(10)2020 09 29.
Article in English | MEDLINE | ID: mdl-33003628

ABSTRACT

The cardiorenal syndrome relates to the detrimental interplay between the vascular system and the kidney. The uremic milieu induced by reduced kidney function alters the phenotype of vascular smooth muscle cells (VSMC) and promotes vascular calcification, a condition which is strongly linked to cardiovascular morbidity and mortality. Biological mechanisms involved include generation of reactive oxygen species, inflammation and accelerated senescence. A better understanding of the vasotoxic effects of uremic retention molecules may reveal novel avenues to reduce vascular calcification in CKD. The present review aims to present a state of the art on the role of uremic toxins in pathogenesis of vascular calcification. Evidence, so far, is fragmentary and limited with only a few uremic toxins being investigated, often by a single group of investigators. Experimental heterogeneity furthermore hampers comparison. There is a clear need for a concerted action harmonizing and standardizing experimental protocols and combining efforts of basic and clinical researchers to solve the complex puzzle of uremic vascular calcification.


Subject(s)
Cardio-Renal Syndrome/metabolism , Kidney/metabolism , Muscle, Smooth, Vascular/metabolism , Renal Insufficiency, Chronic/metabolism , Toxins, Biological/metabolism , Uremia/metabolism , Vascular Calcification/metabolism , Animals , Cardio-Renal Syndrome/pathology , Cardio-Renal Syndrome/physiopathology , Cardio-Renal Syndrome/therapy , Humans , Kidney/pathology , Kidney/physiopathology , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology , Prognosis , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/physiopathology , Renal Insufficiency, Chronic/therapy , Uremia/pathology , Uremia/physiopathology , Uremia/therapy , Vascular Calcification/pathology , Vascular Calcification/physiopathology , Vascular Calcification/therapy
20.
Biochem Biophys Res Commun ; 532(1): 11-18, 2020 10 29.
Article in English | MEDLINE | ID: mdl-32826057

ABSTRACT

Severe secondary hyperparathyroidism (SHPT) represents a high turnover bone disease, osteitis fibrosa, but the pathogenesis of osteitis fibrosa remains to be fully elucidated. We examined the characteristics of the differentiation of bone marrow mesenchymal stem cells (BMSCs) into osteoblasts in uremic rats. We bred 5/6 nephrectomized (Nx) rats with a high phosphorus (P) diet to induce SHPT (Nx + HP), or Nx (Nx + ND) and normal rats (Nc + ND) fed a standard diet (ND). After 8 weeks, BMSCs were isolated from the femur and serum were analyzed. BMSCs underwent flow cytometric examination for the expression patterns of cell surface markers (CD90+, CD29+, CD45-, and CD31-). Serum creatinine (Cre) levels were significantly elevated in the Nx + NP rats compared with the Nc + NP rats. Cre levels in the Nx + HP rats were levels to those in the Nx + ND rats. Serum P and PTH levels were significantly elevated in the Nx + HP rats compared with the Nx + ND rats. Bone morphometrical analysis showed increases in both osteoid volume and eroded surfaces in the Nx + HP but not in the Nx + ND rats. The populations of harvested BMSCs were similar between all three groups. Alp, Runx2, Pth1r and Cyclin D1 mRNA expression in the BMSCs from the Nx + ND rats were significantly suppressed compared with those isolated from the Nc + ND groups. Alizarin red staining tended to be similar to the expression of these mRNA. These results suggest that the BMSCs differentiation into osteoblasts was disturbed in the uremic rats.


Subject(s)
Mesenchymal Stem Cells/pathology , Osteoblasts/pathology , Uremia/pathology , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Animals , Calcification, Physiologic , Cell Differentiation/genetics , Cell Differentiation/physiology , Creatinine/blood , Disease Models, Animal , Hyperparathyroidism, Secondary/etiology , Hyperparathyroidism, Secondary/pathology , Hyperparathyroidism, Secondary/physiopathology , Male , Mesenchymal Stem Cells/metabolism , Osteoblasts/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/physiopathology , Uremia/complications , Uremia/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...