scholarly journals Molecular mechanisms underlying uremic toxin-related systemic disorders in chronic kidney disease: focused on β2-microglobulin-related amyloidosis and indoxyl sulfate-induced atherosclerosis—Oshima Award Address 2016

2018 ◽  
Vol 23 (2) ◽  
pp. 151-157 ◽  
Author(s):  
Suguru Yamamoto
Author(s):  
Ying Li ◽  
Jing Yan ◽  
Minjia Wang ◽  
Jing Lv ◽  
Fei Yan ◽  
...  

AbstractEvidence has been shown that indoxyl sulfate (IS) could impair kidney and cardiac functions. Moreover, macrophage polarization played important roles in chronic kidney disease and cardiovascular disease. IS acts as a nephron-vascular toxin, whereas its effect on macrophage polarization during inflammation is still not fully elucidated. In this study, we aimed to investigate the effect of IS on macrophage polarization during lipopolysaccharide (LPS) challenge. THP-1 monocytes were incubated with phorbol 12-myristate-13-acetate (PMA) to differentiate into macrophages, and then incubated with LPS and IS for 24 h. ELISA was used to detect the levels of TNFα, IL-6, IL-1β in THP-1-derived macrophages. Western blot assay was used to detect the levels of arginase1 and iNOS in THP-1-derived macrophages. Percentages of HLA-DR-positive cells (M1 macrophages) and CD206-positive cells (M2 macrophages) were detected by flow cytometry. IS markedly increased the production of the pro-inflammatory factors TNFα, IL-6, IL-1β in LPS-stimulated THP-1-derived macrophages. In addition, IS induced M1 macrophage polarization in response to LPS, as evidenced by the increased expression of iNOS and the increased proportion of HLA-DR+ macrophages. Moreover, IS downregulated the level of β-catenin, and upregulated the level of YAP in LPS-stimulated macrophages. Activating β-catenin signaling or inhibiting YAP signaling suppressed the IS-induced inflammatory response in LPS-stimulated macrophages by inhibiting M1 polarization. IS induced M1 macrophage polarization in LPS-stimulated macrophages via inhibiting β-catenin and activating YAP signaling. In addition, this study provided evidences that activation of β-catenin or inhibition of YAP could alleviate IS-induced inflammatory response in LPS-stimulated macrophages. This finding may contribute to the understanding of immune dysfunction observed in chronic kidney disease and cardiovascular disease.


Antioxidants ◽  
2021 ◽  
Vol 10 (6) ◽  
pp. 936
Author(s):  
Chien-Lin Lu ◽  
Cai-Mei Zheng ◽  
Kuo-Cheng Lu ◽  
Min-Tser Liao ◽  
Kun-Lin Wu ◽  
...  

The accumulation of the uremic toxin indoxyl sulfate (IS) induces target organ damage in chronic kidney disease (CKD) patients, and causes complications including cardiovascular diseases, renal osteodystrophy, muscle wasting, and anemia. IS stimulates reactive oxygen species (ROS) production in CKD, which impairs glomerular filtration by a direct cytotoxic effect on the mesangial cells. IS further reduces antioxidant capacity in renal proximal tubular cells and contributes to tubulointerstitial injury. IS-induced ROS formation triggers the switching of vascular smooth muscular cells to the osteoblastic phenotype, which induces cardiovascular risk. Low-turnover bone disease seen in early CKD relies on the inhibitory effects of IS on osteoblast viability and differentiation, and osteoblastic signaling via the parathyroid hormone. Excessive ROS and inflammatory cytokine releases caused by IS directly inhibit myocyte growth in muscle wasting via myokines’ effects. Moreover, IS triggers eryptosis via ROS-mediated oxidative stress, and elevates hepcidin levels in order to prevent iron flux in circulation in renal anemia. Thus, IS-induced oxidative stress underlies the mechanisms in CKD-related complications. This review summarizes the underlying mechanisms of how IS mediates oxidative stress in the pathogenesis of CKD’s complications. Furthermore, we also discuss the potential role of oral AST-120 in attenuating IS-mediated oxidative stress after gastrointestinal adsorption of the IS precursor indole.


Cells ◽  
2021 ◽  
Vol 10 (1) ◽  
pp. 114
Author(s):  
Chih-Yu Yang ◽  
Ting-Wen Chen ◽  
Wan-Lun Lu ◽  
Shih-Shin Liang ◽  
Hsien-Da Huang ◽  
...  

Chronic kidney disease (CKD) has long been known to cause significant digestive tract pathology. Of note, indoxyl sulfate is a gut microbe-derived uremic toxin that accumulates in CKD patients. Nevertheless, the relationship between gut microbiota, fecal indole content, and blood indoxyl sulfate level remains unknown. In our study, we established an adenine-induced CKD rat model, which recapitulates human CKD-related gut dysbiosis. Synbiotic treatment in CKD rats showed a significant reduction in both the indole-producing bacterium Clostridium and fecal indole amount. Furthermore, gut microbiota diversity was reduced in CKD rats but was restored after synbiotic treatment. Intriguingly, in our end-stage kidney disease (ESKD) patients, the abundance of indole-producing bacteria, Bacteroides, Prevotella, and Clostridium, is similar to that of healthy controls. Consistently, the fecal indole tends to be higher in the ESKD patients, but the difference did not achieve statistical significance. However, the blood level of indoxyl sulfate was significantly higher than that of healthy controls, implicating that under an equivalent indole production rate, the impaired renal excretion contributes to the accumulation of this notorious uremic toxin. On the other hand, we did identify two short-chain fatty acid-producing bacteria, Faecalibacterium and Roseburia, were reduced in ESKD patients as compared to the healthy controls. This may contribute to gut dysbiosis. We also identified that three genera Fusobacterium, Shewanella, and Erwinia, in the ESKD patients but not in the healthy controls. Building up gut symbiosis to treat CKD is a novel concept, but once proved effective, it will provide an additional treatment strategy for CKD patients.


2021 ◽  
Vol 36 (Supplement_1) ◽  
Author(s):  
Laurent Metzinger

Abstract Background and Aims The gene program is controlled at the post-transcriptional level by the action of small non-coding RNAs known as microRNAs (miRNAs), short, single-stranded molecules that control mRNA stability or translational repression via base pairing with regions in the 3' untranslated region of their target mRNAs. Recently, considerable progress has been made to elucidate the roles of miRNAs in vascular pathogenesis and develop the use of miRNAs as biomarkers, and innovative drugs. We demonstrated during the last decade that miRNAs miR-126 and miR-223 are implicated in the course of chronic kidney disease (CKD) and cardiovascular damage. miR-223 expression is enhanced in vascular smooth muscle cells (VSMCs) subjected to an uremic toxin and also in aortas of a murine model of CKD. As restenosis is a common complication of angioplasty, in which neointimal hyperplasia results from migration of VSMCs into the vessel lumen we measured the effect of miR-223 modulation on restenosis in a rat model of carotid artery after balloon injury. We over-expressed and inhibited miR-223 expression using adenoviral vectors, coding a pre-miR-223 sequence or a sponge sequence, used to trap endogenous microRNA, respectively. We demonstrated that inhibiting miR-223 function significantly reduced neointimal hyperplasia by almost half in carotids. Thus down-regulating miR-223 could be a potential therapeutic approach to prevent restenosis after angioplasty. We also correlated miR-126 and miR-223 expression with clinical outcomes in a large cohort of CKD patients, in collaboration with the University Hospital of Ghent (Belgium) and Ambroise Paré Hospital, France. We evaluated both miRNA’s link with all-cause mortality and cardiovascular and renal events over a 6-year follow-up period. The serum levels of miR-126 and miR-223 were decreased as CKD stage advanced, and patients with higher levels of miR-126 and miR-223 had a higher survival rate. Similar results were observed for cardiovascular and renal events. In conclusion, CKD is associated with a decrease in circulating miR-126 and miR-223 levels in CKD patients. We will also present links between several uremic toxin concentrations and miRNA concentration in the patients of this cohort. Finally, anemia is a common feature of CKD that is associated with cardiovascular disease and poor clinical outcomes. A mixture of uremic toxins accumulates in the blood of CKD patients during the course of the disease, and there is good evidence that they modulate erythropoiesis, explaining at least partly anemia. The exact molecular mechanisms implicated are however poorly understood, although recent progresses have been made to identify key components in the CKD process. We will present results on the effect of uremic toxins on erythropoiesis, having an impact on cell metabolism during this process. Taken together, our findings could be of interest to both researchers and clinicians working in the field since they might shed new light on the molecular mechanisms involved in the CKD process. MicroRNAs implicated in Chronic Kidney Disease Pr. Laurent Metzinger, UR-UPJV 4666 HEMATIM, CURS, Université de Picardie Jules Verne, CHU Amiens Sud, Avenue René Laënnec, Salouel, F-80054, Amiens, France. Tel: (+33) 22 82 53 56, Email: [email protected]


2020 ◽  
Vol 35 (Supplement_3) ◽  
Author(s):  
Jun Chul Kim ◽  
Seok Hui Kang ◽  
Miyeun Han ◽  
Su-Hyun Kim ◽  
Ran-Hui Cha ◽  
...  

Abstract Background and Aims Sarcopenia in patients with chronic kidney disease (CKD) is highly prevalent and leads to high rate of morbidity and mortality. The role of indoxyl sulfate (IS) to develop muscle wasting has been researched and proved in several animal model studies. However, there is no human data showing this relationship in CKD population. The aim of the present study was to evaluate the association between serum IS levels and each component of sarcopenia in nondialysis dependent-CKD (NDD-CKD) patients. Method We enrolled 150 NDD-CKD adult patients from 6 medical centers and collected data of demographics, blood chemistry such as indoxyl sulfate, interleukin (IL)-6, and estimated glomerular filtration rate using MDRD equation (eGFR), and body mass index (BMI, kg/m2). We also measured hand-grip strength (HGS, kg), walking speed (WS, m/s), skeletal muscle mass (SMM, kg) by bioelectrical impedance analysis (BIA). Results The numbers of male sex was 97 (64.7%). Mean age was 63.7±10.8 years old. The numbers of patients with diabetes mellitus was 77 (52.0%). Charlson comorbidity index (CCI) score was 3.9 ± 1.9. The stage of CKD ranged from 3 to 5 (eGFR=33.7±12.0 ml/min/1.73m2, mean±SD). Correlation coefficients with indoxyl sulfate levels were 0.211 for serum IL-6 level (P = 0.010), -0.212 for HGS (P = 0.009), -0.188 for WS (P = 0.021), -0.237 for SMM (P = 0.004), and -0.168 for BMI (P = 0.041), respectively. Correlation analysis showed that indoxyl sulfate levels had inverse association significantly with HGS, WS, SMM, and BMI and were positively associated with serum IL-6 levels. Conclusion Our study shows that higher serum indoxyl sulfate level was significantly associated with lower levels of muscle mass, strength, and physical performance function and higher inflammation status in non-dialysis dependent CKD patients. We suggest that the role of AST120 in prevention or treatment of sarcopenia be studied in this CKD population.


2020 ◽  
Author(s):  
Cheng-Hsu Chen ◽  
Shih-Chien Huang ◽  
Pei-Chih Lin ◽  
Shang-Feng Tsai ◽  
Yi-Chia Huang

Abstract Background: Increased levels of uremic toxins and decreased antioxidant capacities have a significant impact on the progression of chronic kidney disease (CKD). However, it is unclear whether they interact with each other in order to mediate the damage of renal function. The purpose of this study was to determine whether uremic toxins [i.e., homocysteine and indoxyl sulfate (IS)] and glutathione-dependent antioxidant enzyme activities are dependently or independently associated with each other in affecting renal function during different stages of CKD patients.Methods: One hundred thirty-two patients diagnosed with CKD stage 1 to 5 participated in this cross-sectional study.Results: Patients who had reached an advanced CKD stage experienced a gradual increase in plasma uremic toxin levels, along with decreased glutathione peroxidase (GSH-Px) activities. Plasma homocysteine, cysteine and IS concentrations were positively associated with each other, but negatively correlated to GSH-Px activity levels after adjusting potential confounders in all CKD patients. Although plasma homocysteine, cysteine, IS and GSH-Px levels were significantly associated with renal function, only plasma IS levels still had a significant association with renal function after these parameters were simultaneously adjusted.Conclusions: IS plays a more dominant role than other factors in affecting renal function, where a higher IS concentration needs to be controlled in order to defer the progressive loss of renal function.


2017 ◽  
Vol 33 (4) ◽  
pp. 586-597 ◽  
Author(s):  
Hirofumi Hamano ◽  
Yasumasa Ikeda ◽  
Hiroaki Watanabe ◽  
Yuya Horinouchi ◽  
Yuki Izawa-Ishizawa ◽  
...  

2020 ◽  
Vol 35 (Supplement_3) ◽  
Author(s):  
Natasha Rogers ◽  
Sohel Julovi ◽  
Kedar Ghimire ◽  
Nikita Minhas

Abstract Background and Aims Patients with chronic kidney disease (CKD) are at significantly greater risk of cardiovascular disease (CVD), which remains the leading cause of hospitalisation and mortality. Cardiovascular complications are 20-30 times more likely to occur in these patients than progression to end-stage kidney disease. Traditional cardiovascular risk factors are prevalent in patients with CKD, but clinical trials targeting standard cardiovascular risks have failed to improve outcomes in patients with CKD. This has led to the investigation of non-traditional pathways that drive the development of the CVD in CKD. The matrix protein thrombospondin-1 (TSP1), and its receptor CD47, regulates cell responses and we recently identified their role in acute kidney injury. We now investigate this pathway in vascular changes in CKD. Method Human plasma TSP1 was collected from n=120 patients attending outpatient clinics and measured by ELISA. Age matched male C57BL/6 (WT) and TSP1KO mice (on a C57BL/6 background) were used to develop a model of CKD employing a 5/6 nephrectomy technique (5/6Nx). Blood pressure and weight were checked weekly, and echocardiography analysed cardiac function at baseline. After 12 weeks, we performed echocardiography and biomolecular phenotyping on tissues. Human cardiomyocytes were used to assess the effect of TSP1 and the uremic toxin indoxyl sulfate on cell proliferation and senescence. Results Linear regression analysis demonstrate that plasma TSP1 correlated inversely with eGFR (r2=-0.556, p<0.001). WT mice undergoing 5/6Nx developed renal interstitial fibrosis, hypertension, and left ventricular (LV) hypertrophy after 12 weeks. Echocardiography confirmed LV thickness with a concurrent reduction in LV ejection fraction compared to baseline. Histologic examination of WT myocardium revealed interstitial and perivascular fibrosis, and increased myocardial TSP1 expression. These findings were mitigated in TSP1KO mice who had preserved LV function, reduced LV hypertrophy and reduced cardiac fibrosis despite equivalent changes in renal mass and blood pressure. mRNA levels of cardiac-based TNF-α, IL-6, collagen-1, fibronectin and α-smooth muscle actin were increased in WT 5/6Nx mice compared to sham-operated controls, and transcript expression was robustly reduced in TSP1KO mice. In vitro, the addition of uremic toxin (indoxyl sulfate) to cardiomyocytes robustly upregulated expression of TSP1, and this was dependent upon activation of the aryl hydrocarbon receptor. TSP1 significantly down-regulated cardiomyocte proliferation and increased β-galactosidase activity. Pre-incubation with a blocking antibody reversed the proliferative and senescent effects of indoxyl sulfate and TSP1. Conclusion Our findings show that TSP1 is elevated in CKD and may drive the cardiovascular manifestations of uremia. TSP1KO mice were protected from disease, suggestion this protein has a crucial role in the development of CVD. Manipulation of TSP1 signaling is an attractive target to potentially reduce the excessive burden of CVD seen in the CKD population.


Sign in / Sign up

Export Citation Format

Share Document