1
|
Kudo A, Fukuda A, Gotoh K, Shibata H. Indoxyl Sulfate and Its Potential Role in Mineralocorticoid Receptor Transactivation in Chronic Kidney Disease. Cureus 2024; 16:e75236. [PMID: 39759603 PMCID: PMC11700523 DOI: 10.7759/cureus.75236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2024] [Indexed: 01/07/2025] Open
Abstract
BACKGROUND The uremic toxin indoxyl sulfate (IS) is an important factor in chronic kidney disease (CKD) progression. Inhibitors of the renin-angiotensin system and add-on therapy with mineralocorticoid receptor (MR) antagonists can help reduce proteinuria and suppress CKD progression. However, the association between IS and MR activation remains unknown. MATERIALS AND METHODS In vivo experiments utilized the 5/6 nephrectomy model to assess mineralocorticoid receptor (MR) activation in chronic kidney disease (CKD). The clinical parameters and immunohistochemical analysis of IS and MR proteins were investigated. In vitro experiments involved transfecting COS-7 cells with MR expression plasmids and MR response element-luciferase reporter plasmids. The cells were then treated with aldosterone (10⁻¹⁰ mol/L), indoxyl sulfate (IS, 500 μmol/L), and α-lipoic acid (10⁻³ mol/L). MR transcriptional activity was investigated by luciferase assays, and protein levels were measured by Western blotting. RESULTS In the 5/6 nephrectomy model, the serum IS concentration was significantly increased; however, the plasma aldosterone levels were decreased. Immunohistochemistry showed that the expression of IS protein increased in injured tubular cells in the 5/6 nephrectomy group compared with that in the sham group. Furthermore, evaluations of serial kidney sections revealed that the expression site of IS protein was colocalized with the distal nephron, where the expression of MR protein was observed. MR-mediated transcriptional activity in COS-7 cells was increased in an aldosterone concentration-dependent manner. IS increased MR-mediated transcriptional activity and protein levels with and without aldosterone, and α-lipoic acid attenuated this increase. CONCLUSIONS IS could enhance MR transactivation by increasing MR protein levels through oxidative stress in CKD rats, indicating that treatment with MR antagonists and antioxidants may play a permissive role in inhibiting IS-induced CKD progression.
Collapse
Affiliation(s)
- Akiko Kudo
- Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of Medicine, Oita University, Yufu, JPN
| | - Akihiro Fukuda
- Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of Medicine, Oita University, Yufu, JPN
| | - Koro Gotoh
- Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of Medicine, Oita University, Yufu, JPN
- Faculty of Welfare and Health Science, Oita University, Oita, JPN
| | - Hirotaka Shibata
- Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of Medicine, Oita University, Yufu, JPN
| |
Collapse
|
2
|
Pieniazek A, Bernasinska-Slomczewska J, Hikisz P. Indoxyl sulfate induces apoptosis in mononuclear blood cells via mitochondrial pathway. Sci Rep 2023; 13:14044. [PMID: 37640757 PMCID: PMC10462746 DOI: 10.1038/s41598-023-40824-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 08/17/2023] [Indexed: 08/31/2023] Open
Abstract
The consequence of chronic kidney disease is the accumulation of metabolic products called uremic toxins in the body. Indoxyl sulfate (IS) is a toxin with a high affinity for proteins. This study focuses on the deleterious effect of IS, especially apoptosis induction, in mononuclear blood cells (MNCs). Thus, in MNCs treated with IS at three different concentrations for 24 h, the survival, mitochondrial potential, caspases activity and expression, Bcl-2 and Bax protein expression, DNA damage, and PARP degradation were estimated. The study showed a decrease in survival and mitochondrial potential of MNCs treated with IS compared to the control. IS increased the activity of caspase 2-, 3-, 9-, and the expression of caspase 3-, and 9- in MNCs but does not affect the activity of caspase 6- and 8. The treatment of MNCs with IS also increased DNA damage and degradation of PARP. Indoxyl sulfate significantly influences the expression of Bcl-2 and Bax proteins. Indoxyl sulfate induces the programmed death of MNCs through the intrinsic mitochondrial apoptotic pathway. The observed cellular changes are mostly dose-dependent.
Collapse
Affiliation(s)
- Anna Pieniazek
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, Ul. Pomorska 141/143, 90-236, Lodz, Poland.
| | - Joanna Bernasinska-Slomczewska
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, Ul. Pomorska 141/143, 90-236, Lodz, Poland
| | - Pawel Hikisz
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, Ul. Pomorska 141/143, 90-236, Lodz, Poland
| |
Collapse
|
3
|
Caggiano G, Stasi A, Franzin R, Fiorentino M, Cimmarusti MT, Deleonardis A, Palieri R, Pontrelli P, Gesualdo L. Fecal Microbiota Transplantation in Reducing Uremic Toxins Accumulation in Kidney Disease: Current Understanding and Future Perspectives. Toxins (Basel) 2023; 15:toxins15020115. [PMID: 36828429 PMCID: PMC9965504 DOI: 10.3390/toxins15020115] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/21/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
During the past decades, the gut microbiome emerged as a key player in kidney disease. Dysbiosis-related uremic toxins together with pro-inflammatory mediators are the main factors in a deteriorating kidney function. The toxicity of uremic compounds has been well-documented in a plethora of pathophysiological mechanisms in kidney disease, such as cardiovascular injury (CVI), metabolic dysfunction, and inflammation. Accumulating data on the detrimental effect of uremic solutes in kidney disease supported the development of many strategies to restore eubiosis. Fecal microbiota transplantation (FMT) spread as an encouraging treatment for different dysbiosis-associated disorders. In this scenario, flourishing studies indicate that fecal transplantation could represent a novel treatment to reduce the uremic toxins accumulation. Here, we present the state-of-the-art concerning the application of FMT on kidney disease to restore eubiosis and reverse the retention of uremic toxins.
Collapse
|
4
|
Martin WP, Nair M, Chuah YH, Malmodin D, Pedersen A, Abrahamsson S, Hutter M, Abdelaal M, Elliott JA, Fearon N, Eckhardt H, Godson C, Brennan EP, Fändriks L, le Roux CW, Docherty NG. Dietary restriction and medical therapy drives PPARα-regulated improvements in early diabetic kidney disease in male rats. Clin Sci (Lond) 2022; 136:1485-1511. [PMID: 36259366 PMCID: PMC7613831 DOI: 10.1042/cs20220205] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 10/14/2022] [Accepted: 10/18/2022] [Indexed: 11/17/2022]
Abstract
The attenuation of diabetic kidney disease (DKD) by metabolic surgery is enhanced by pharmacotherapy promoting renal fatty acid oxidation (FAO). Using the Zucker Diabetic Fatty and Zucker Diabetic Sprague Dawley rat models of DKD, we conducted studies to determine if these effects could be replicated with a non-invasive bariatric mimetic intervention. Metabolic control and renal injury were compared in rats undergoing a dietary restriction plus medical therapy protocol (DMT; fenofibrate, liraglutide, metformin, ramipril, and rosuvastatin) and ad libitum-fed controls. The global renal cortical transcriptome and urinary 1H-NMR metabolomic profiles were also compared. Kidney cell type-specific and medication-specific transcriptomic responses were explored through in silico deconvolution. Transcriptomic and metabolomic correlates of improvements in kidney structure were defined using a molecular morphometric approach. The DMT protocol led to ∼20% weight loss, normalized metabolic parameters and was associated with reductions in indices of glomerular and proximal tubular injury. The transcriptomic response to DMT was dominated by changes in fenofibrate- and peroxisome proliferator-activated receptor-α (PPARα)-governed peroxisomal and mitochondrial FAO transcripts localizing to the proximal tubule. DMT induced urinary excretion of PPARα-regulated metabolites involved in nicotinamide metabolism and reversed DKD-associated changes in the urinary excretion of tricarboxylic acid (TCA) cycle intermediates. FAO transcripts and urinary nicotinamide and TCA cycle metabolites were moderately to strongly correlated with improvements in glomerular and proximal tubular injury. Weight loss plus pharmacological PPARα agonism is a promising means of attenuating DKD.
Collapse
Affiliation(s)
- William P. Martin
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | - Meera Nair
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | - Yeong H.D. Chuah
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | - Daniel Malmodin
- Swedish NMR Centre, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Anders Pedersen
- Swedish NMR Centre, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Sanna Abrahamsson
- Bioinformatics Core Facility, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Michaela Hutter
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | - Mahmoud Abdelaal
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | - Jessie A. Elliott
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | - Naomi Fearon
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | - Hans Eckhardt
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | - Catherine Godson
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | - Eoin P. Brennan
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | - Lars Fändriks
- Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Carel W. le Roux
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
- Diabetes Research Group, Ulster University, Coleraine BT52 1SA, UK
| | - Neil G. Docherty
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| |
Collapse
|
5
|
Cheng TH, Ma MC, Liao MT, Zheng CM, Lu KC, Liao CH, Hou YC, Liu WC, Lu CL. Indoxyl Sulfate, a Tubular Toxin, Contributes to the Development of Chronic Kidney Disease. Toxins (Basel) 2020; 12:E684. [PMID: 33138205 PMCID: PMC7693919 DOI: 10.3390/toxins12110684] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/27/2020] [Accepted: 10/28/2020] [Indexed: 12/22/2022] Open
Abstract
Indoxyl sulfate (IS), a uremic toxin, causes chronic kidney disease (CKD) progression via its tubulotoxicity. After cellular uptake, IS directly induces apoptotic and necrotic cell death of tubular cells. Additionally, IS increases oxidative stress and decreases antioxidant capacity, which are associated with tubulointerstitial injury. Injured tubular cells are a major source of transforming growth factor-β1 (TGF-β1), which induces myofibroblast transition from residual renal cells in damaged kidney, recruits inflammatory cells and thereby promotes extracellular matrix deposition in renal fibrosis. Moreover, IS upregulates signal transducers and activators of transcription 3 phosphorylation, followed by increases in TGF-β1, monocyte chemotactic protein-1 and α-smooth muscle actin production, which participate in interstitial inflammation, renal fibrosis and, consequently, CKD progression. Clinically, higher serum IS levels are independently associated with renal function decline and predict all-cause mortality in CKD. The poor removal of serum IS in conventional hemodialysis is also significantly associated with all-cause mortality and heart failure incidence in end-stage renal disease patients. Scavenging the IS precursor by AST-120 can markedly reduce tubular IS staining that attenuates renal tubular injury, ameliorates IS-induced oxidative stress and rescues antioxidant glutathione activity in tubular epithelial cells, thereby providing a protective role against tubular injury and ultimately retarding renal function decline.
Collapse
Affiliation(s)
- Tong-Hong Cheng
- School of Medicine, Fu Jen Catholic University, New Taipei 242, Taiwan; (T.-H.C.); (M.-C.M.); (C.-H.L.); (Y.-C.H.)
- Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan 325, Taiwan
| | - Ming-Chieh Ma
- School of Medicine, Fu Jen Catholic University, New Taipei 242, Taiwan; (T.-H.C.); (M.-C.M.); (C.-H.L.); (Y.-C.H.)
| | - Min-Tser Liao
- Department of Pediatrics, Taoyuan Armed Forces General Hospital, Taoyuan 325, Taiwan;
- Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Cai-Mei Zheng
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Division of Nephrology, Department of Internal Medicine, Taipei Medical University Shuang Ho Hospital, New Taipei 235, Taiwan
- Taipei Medical University-Research Center of Urology and Kidney, Taipei Medical University, Taipei 110, Taiwan
| | - Kuo-Cheng Lu
- Division of Nephrology, Department of Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan;
| | - Chun-Hou Liao
- School of Medicine, Fu Jen Catholic University, New Taipei 242, Taiwan; (T.-H.C.); (M.-C.M.); (C.-H.L.); (Y.-C.H.)
- Divisions of Urology, Department of Surgery, Cardinal Tien Hospital, New Taipei 23148, Taiwan
| | - Yi-Chou Hou
- School of Medicine, Fu Jen Catholic University, New Taipei 242, Taiwan; (T.-H.C.); (M.-C.M.); (C.-H.L.); (Y.-C.H.)
- Division of Nephrology, Department of Medicine, Cardinal-Tien Hospital, School of Medicine, Fu-Jen Catholic University, New Taipei 234, Taiwan
| | - Wen-Chih Liu
- Division of Nephrology, Department of Medicine, Taipei Hospital, Ministry of Health and Welfare, New Taipei 242, Taiwan;
| | - Chien-Lin Lu
- School of Medicine, Fu Jen Catholic University, New Taipei 242, Taiwan; (T.-H.C.); (M.-C.M.); (C.-H.L.); (Y.-C.H.)
- Division of Nephrology, Department of Medicine, Fu Jen Catholic University Hospital, New Taipei 242, Taiwan
| |
Collapse
|
6
|
Metabolomics study of fasudil on cisplatin-induced kidney injury. Biosci Rep 2020; 39:220982. [PMID: 31670380 PMCID: PMC6863766 DOI: 10.1042/bsr20192940] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 10/16/2019] [Accepted: 10/25/2019] [Indexed: 12/01/2022] Open
Abstract
Fasudil is a derivative of 5-isoquinoline sulfonamide, which is a Rho kinase inhibitor, a wide range of pharmacological effects. Fasudil has been shown to attenuate kidney injury caused by certain substances. In the present study, metabolomic analysis of mouse kidney tissues ultra-performance liquid chromatography/quadrupole time-of-flight mass spectrometry was used to determine the metabolomic changes in cisplatin-induced kidney injury and the fasudil-induced attenuation of cisplatin-induced kidney injury. Metabolomic profiling of kidney tissues revealed significant differences in metabolites between the control group and the cisplatin group and between the cisplatin group and the fasudil-intervention group. With metabolomic approach, 68 endogenous differential metabolites were found, and multivariate statistical analysis, accurate molecular weights, isotope tracers, mass-spectrometry secondary-fragment information, and standard-reference comparisons were used to identify these substances. Based on these differential metabolites, a metabolic-pathway network was constructed and revealed that fasudil primarily attenuated cisplatin-induced renal injury by modulating lipid and amino-acid metabolism. These results further demonstrate that kidney injury can be induced by cisplatin and, moreover, suggest that fasudil can be used to reduce kidney injury at early stages in patients treated with cisplatin.
Collapse
|
7
|
Rodrigues GGC, Dellê H, Brito RBO, Cardoso VO, Fernandes KPS, Mesquita-Ferrari RA, Cunha RS, Stinghen AEM, Dalboni MA, Barreto FC. Indoxyl Sulfate Contributes to Uremic Sarcopenia by Inducing Apoptosis in Myoblasts. Arch Med Res 2020; 51:21-29. [PMID: 32086105 DOI: 10.1016/j.arcmed.2019.12.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 11/28/2019] [Accepted: 12/13/2019] [Indexed: 01/02/2023]
Abstract
OBJETIVE Uremic sarcopenia is a complication of chronic kidney disease, particularly in its later stages, which leads to musculoskeletal disability. Uremic toxins have been linked to the pathogenesis of several manifestations of uremic syndrome. We sought to investigate whether indoxyl sulphate (IS), a protein-bound uremic toxin, is implicated in the development of uremic sarcopenia. MATERIAL AND METHODS Myoblasts were exposed to IS at normal (0.6 mg/L, IS0.6), uremic (53 mg/L, IS53) or maximum uremic (236 mg/L, IS236) concentrations for 24, 48 and 72 h. Cell viability was evaluated by MTT assay and by 7-aminoactinomycin D staining. ROS generation and apoptosis were evaluated by flow cytometry. MyoD and myogenin mRNA expression was evaluated by qRT-PCR and myosin heavy chain expression by immunocytochemistry. RESULTS Myoblast viability was reduced by IS236 in a time-dependent pattern (p <0.05; 84.4, 68.0, and 63.6%). ROS production was significantly higher (p <0.05) in cells exposed to IS53 and IS236 compared to control (untreated cells). The apoptosis rate was significantly higher in cells treated with IS53 and IS236 than in control after 48h (p <0.05; 4.7 ± 0.1% and 4.6 ± 0.3% vs. 3.1 ± 0.1%, respectively) and 72h (p <0.05; 9.6 ± 1.1% and 10.4 ± 0.3% vs. 3.1 ± 0.7%, respectively). No effect was observed on MyoD, myogenin, myosin heavy chain expression, and markers of myoblast differentiation at any IS concentration tested or time-point experiment. CONCLUSIONS These data indicate that IS has direct toxic effects on myoblast by decreasing its viability and increasing cell apoptosis. IS may be a potential target for treating uremic sarcopenia.
Collapse
Affiliation(s)
| | - Humberto Dellê
- Postgraduate Program in Medicine, Universidade Nove de Julho, São Paulo, Brazil.
| | | | | | | | | | - Regiane Stafim Cunha
- Experimental Nephrology Laboratory, Basic Pathology Department, Universidade Federal do Paraná, Curitiba, Brazil
| | | | | | - Fellype Carvalho Barreto
- Nephrology Service, Department of Internal Medicine, Universidade Federal do Paraná, Paraná, Brazil
| |
Collapse
|
8
|
Savira F, Magaye R, Hua Y, Liew D, Kaye D, Marwick T, Wang BH. Molecular mechanisms of protein-bound uremic toxin-mediated cardiac, renal and vascular effects: underpinning intracellular targets for cardiorenal syndrome therapy. Toxicol Lett 2019; 308:34-49. [PMID: 30872129 DOI: 10.1016/j.toxlet.2019.03.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 02/21/2019] [Accepted: 03/05/2019] [Indexed: 02/07/2023]
Abstract
Cardiorenal syndrome (CRS) remains a global health burden with a lack of definitive and effective treatment. Protein-bound uremic toxin (PBUT) overload has been identified as a non-traditional risk factor for cardiac, renal and vascular dysfunction due to significant albumin-binding properties, rendering these solutes non-dialyzable upon the state of irreversible kidney dysfunction. Although limited, experimental studies have investigated possible mechanisms in PBUT-mediated cardiac, renal and vascular effects. The ultimate aim is to identify relevant and efficacious targets that may translate beneficial outcomes in disease models and eventually in the clinic. This review will expand on detailed knowledge on mechanisms involved in detrimental effects of PBUT, specifically affecting the heart, kidney and vasculature, and explore potential effective intracellular targets to abolish their effects in CRS initiation and/or progression.
Collapse
Affiliation(s)
- Feby Savira
- Monash Centre of Cardiovascular Research and Education in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Ruth Magaye
- Monash Centre of Cardiovascular Research and Education in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Yue Hua
- Monash Centre of Cardiovascular Research and Education in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Danny Liew
- Monash Centre of Cardiovascular Research and Education in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - David Kaye
- Baker Heart and Diabetes Research Institute, Melbourne, Victoria, Australia
| | - Tom Marwick
- Baker Heart and Diabetes Research Institute, Melbourne, Victoria, Australia
| | - Bing Hui Wang
- Monash Centre of Cardiovascular Research and Education in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia; Baker Heart and Diabetes Research Institute, Melbourne, Victoria, Australia.
| |
Collapse
|
9
|
Tan X, Cao XS, Zhang P, Xiang FF, Teng J, Zou JZ, Ding XQ. Endoplasmic reticulum stress associated apoptosis as a novel mechanism in indoxyl sulfate‑induced cardiomyocyte toxicity. Mol Med Rep 2018; 18:5117-5122. [PMID: 30272270 DOI: 10.3892/mmr.2018.9496] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 07/05/2018] [Indexed: 11/05/2022] Open
Abstract
Indoxyl sulfate (IS), a typical uremic toxin, is of great importance in the development of chronic kidney disease. In addition to its nephrotoxicity, previous studies have provided increasing evidence for its cardiovascular toxicity. The mechanism underlying IS‑induced cardiovascular toxicity has been elusive to date. The present study aimed to evaluate whether IS treatment could induce apoptosis of H9C2 cells, and used the endoplasmic reticulum (ER) stress‑modulator 4‑phenylbutyric acid (4‑PBA) to evaluate whether IS‑induced apoptosis is indeed associated with ERS. To evaluate whether IS induces apoptosis in H9C2 cardiomyocytes, cells were exposed to increasing concentrations of IS (500, 1,000, and 2,000 µM) for 24 h, and apoptosis was detected by flow cytometry. To determine whether IS‑induced apoptosis is associated with ERS, cells were divided into 4 groups: control group, PBA group, IS group and PBA+IS group. IS dose‑dependently induced apoptosis, and increased the expression of ER chaperones in H9C2 cells. Additionally, 4‑PBA treatment decreased IS‑induced apoptosis, and reduced ERS‑associated protein expression induced by IS. Therefore, the mechanism may be associated with the CCAAT‑enhancer‑binding protein homologous protein and c‑Jun N‑terminal kinase signaling pathways.
Collapse
Affiliation(s)
- Xiao Tan
- Shanghai Key Laboratory of Kidney and Blood Purification, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Xue-Sen Cao
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Pan Zhang
- Shanghai Key Laboratory of Kidney and Blood Purification, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Fang-Fang Xiang
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Jie Teng
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Jian-Zhou Zou
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Xiao-Qiang Ding
- Shanghai Key Laboratory of Kidney and Blood Purification, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|
10
|
Wanchai K, Yasom S, Tunapong W, Chunchai T, Thiennimitr P, Chaiyasut C, Pongchaidecha A, Chatsudthipong V, Chattipakorn S, Chattipakorn N, Lungkaphin A. Prebiotic prevents impaired kidney and renal Oat3 functions in obese rats. J Endocrinol 2018; 237:29-42. [PMID: 29483238 DOI: 10.1530/joe-17-0471] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 02/08/2018] [Indexed: 01/19/2023]
Abstract
Obesity is health issue worldwide, which can lead to kidney dysfunction. Prebiotics are non-digestible foods that have beneficial effects on health. This study aimed to investigate the effects of xylooligosaccharide (XOS) on renal function, renal organic anion transporter 3 (Oat3) and the mechanisms involved. High-fat diet was provided for 12 weeks in male Wistar rats. After that, the rats were divided into normal diet (ND); normal diet treated with XOS (NDX); high-fat diet (HF) and high-fat diet treated with XOS (HFX). XOS was given daily at a dose of 1000 mg for 12 weeks. At week 24, HF rats showed a significant increase in obesity and insulin resistance associated with podocyte injury, increased microalbuminuria, decreased creatinine clearance and impaired Oat3 function. These alterations were improved by XOS supplementation. Renal MDA level and the expression of AT1R, NOX4, p67phox, 4-HNE, phosphorylated PKCα and ERK1/2 were significantly decreased after XOS treatment. In addition, Nrf2-Keap1 pathway, SOD2 and GCLC expression as well as renal apoptosis were also significantly reduced by XOS. These data suggest that XOS could indirectly restore renal function and Oat3 function via the reduction of oxidative stress and apoptosis through the modulating of AT1R-PKCα-NOXs activation in obese insulin-resistant rats. These attenuations were instigated by the improvement of obesity, hyperlipidemia and insulin resistance.
Collapse
Affiliation(s)
- Keerati Wanchai
- Department of PhysiologyFaculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- School of MedicineMae Fah Luang University, Chiang Rai, Thailand
| | - Sakawdaurn Yasom
- Department of MicrobiologyFaculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Wannipa Tunapong
- Department of PhysiologyFaculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Cardiac Electrophysiology Research and Training CenterFaculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Titikorn Chunchai
- Department of PhysiologyFaculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Cardiac Electrophysiology Research and Training CenterFaculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Parameth Thiennimitr
- Department of MicrobiologyFaculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | | | - Anchalee Pongchaidecha
- Department of PhysiologyFaculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | | | - Siriporn Chattipakorn
- Department of PhysiologyFaculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Department of Oral Biology and Diagnostic SciencesFaculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Department of PhysiologyFaculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Cardiac Electrophysiology Research and Training CenterFaculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Anusorn Lungkaphin
- Department of PhysiologyFaculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center for Research and Development of Natural Products for HealthChiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
11
|
Chin LH, Hsu YJ, Hsu SC, Chen YH, Chang YL, Huang SM, Tsai CS, Lin CY. The regulation of NLRP3 inflammasome expression during the development of cardiac contractile dysfunction in chronic kidney disease. Oncotarget 2017; 8:113303-113317. [PMID: 29371912 PMCID: PMC5768329 DOI: 10.18632/oncotarget.22964] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 11/09/2017] [Indexed: 02/07/2023] Open
Abstract
Chronic inflammation plays a crucial role in the long-term complications in patients with chronic kidney disease (CKD). This study aimed to assess the role of NLR pyrin domain-containing protein (NLRP3) inflammasome in cardiac contractile dysfunctions in CKD. The cardiac contractile function was evaluated and the expression of NLRP3 inflammasome and related cytokines in the heart was assessed in a murine sham-operated and 5/6 nephrectomy CKD model in vivo. In vitro, H9c2 cells were treated with uremic toxin indoxyl sulfate (IS), with or without NLRP3 inflammasome inhibition, which was achieved by using small interfering RNA (siRNA)-mediated knockdown of the NLRP3 gene. Moreover, the activation of nuclear factor κB (NF-κB) signaling and apoptosis marker levels were assessed in the IS-treated H9c2 cells. The results demonstrated that CKD can lead to the development of cardiac contractile dysfunction in vivo associated with the upregulation of NLRP3 inflammasome, IL-1β, IL-18, and contribute to the myocardial apoptosis. In vitro experiments showed the upregulation of inflammasome, IL-1β, and IL-18 levels, and cell apoptosis in the IS-treated H9c2 cells through the activation of NF-κB signaling pathway. The transfection of cells with si-NLRP3 was shown to alleviate IL-1β, IL-18, and cell apoptosis. Moreover, decreased cell viability induced by IS was shown to be attenuated by IL-1β or IL-18-neutralizing antibody. In summary, CKD can result in the development of cardiac contractile dysfunction associated with the upregulation of NLRP3 inflammasome/IL-1β/IL-18 axis induced by the uremic toxins.
Collapse
Affiliation(s)
- Li-Han Chin
- Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Juei Hsu
- Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Che Hsu
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| | - Yen-Hui Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yung-Lung Chang
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Ming Huang
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| | - Chien-Sung Tsai
- Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Superintendent's Office, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan
| | - Chih-Yuan Lin
- Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
12
|
Carmona A, Guerrero F, Buendia P, Obrero T, Aljama P, Carracedo J. Microvesicles Derived from Indoxyl Sulfate Treated Endothelial Cells Induce Endothelial Progenitor Cells Dysfunction. Front Physiol 2017; 8:666. [PMID: 28951723 PMCID: PMC5599774 DOI: 10.3389/fphys.2017.00666] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 08/22/2017] [Indexed: 12/31/2022] Open
Abstract
Cardiovascular disease is a major cause of mortality in chronic kidney disease patients. Indoxyl sulfate (IS) is a typical protein-bound uremic toxin that cannot be effectively cleared by conventional dialysis. Increased IS is associated with the progression of chronic kidney disease and development of cardiovascular disease. After endothelial activation by IS, cells release endothelial microvesicles (EMV) that can induce endothelial dysfunction. We developed an in vitro model of endothelial damage mediated by IS to evaluate the functional effect of EMV on the endothelial repair process developed by endothelial progenitor cells (EPCs). EMV derived from IS-treated endothelial cells were isolated by ultracentrifugation and characterized for miRNAs content. The effects of EMV on healthy EPCs in culture were studied. We observed that IS activates endothelial cells and the generated microvesicles (IsEMV) can modulate the classic endothelial roles of progenitor cells as colony forming units and form new vessels in vitro. Moreover, 23 miRNAs were contained in IsEMV including four (miR-181a-5p, miR-4454, miR-150-5p, and hsa-let-7i-5p) that were upregulated in IsEMV compared with control endothelial microvesicles. Other authors have found that miR-181a-5p, miR-4454, and miR-150-5p are involved in promoting inflammation, apoptosis, and cellular senescence. Interestingly, we observed an increase in NFκB and p53, and a decrease in IκBα in EPCs treated with IsEMV. Our data suggest that IS is capable of inducing endothelial vesiculation with different membrane characteristics, miRNAs and other molecules, which makes maintaining of vascular homeostasis of EPCs not fully functional. These specific characteristics of EMV could be used as novel biomarkers for diagnosis and prognosis of vascular disease.
Collapse
Affiliation(s)
- Andres Carmona
- Maimonides Institute of Biomedical Research of CordobaCordoba, Spain
| | - Fatima Guerrero
- Maimonides Institute of Biomedical Research of CordobaCordoba, Spain
| | - Paula Buendia
- Maimonides Institute of Biomedical Research of CordobaCordoba, Spain
| | - Teresa Obrero
- Maimonides Institute of Biomedical Research of CordobaCordoba, Spain
| | - Pedro Aljama
- Maimonides Institute of Biomedical Research of CordobaCordoba, Spain.,Department of Nephrology, Nephrology Service, Reina Sofia University HospitalCordoba, Spain.,RETICs Red Renal, Instituto de Salud Carlos IIIMadrid, Spain
| | - Julia Carracedo
- Department of Animal Physiology II, Faculty of Biology, Complutense University of MadridMadrid, Spain
| |
Collapse
|
13
|
Ellis RJ, Small DM, Vesey DA, Johnson DW, Francis R, Vitetta L, Gobe GC, Morais C. Indoxyl sulphate and kidney disease: Causes, consequences and interventions. Nephrology (Carlton) 2016; 21:170-7. [PMID: 26239363 DOI: 10.1111/nep.12580] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/30/2015] [Indexed: 12/28/2022]
Abstract
In the last decade, chronic kidney disease (CKD), defined as reduced renal function (glomerular filtration rate (GFR) < 60 mL/min per 1.73 m(2) ) and/or evidence of kidney damage (typically manifested as albuminuria) for at least 3 months, has become one of the fastest-growing public health concerns worldwide. CKD is characterized by reduced clearance and increased serum accumulation of metabolic waste products (uremic retention solutes). At least 152 uremic retention solutes have been reported. This review focuses on indoxyl sulphate (IS), a protein-bound, tryptophan-derived metabolite that is generated by intestinal micro-organisms (microbiota). Animal studies have demonstrated an association between IS accumulation and increased fibrosis, and oxidative stress. This has been mirrored by in vitro studies, many of which report cytotoxic effects in kidney proximal tubular cells following IS exposure. Clinical studies have associated IS accumulation with deleterious effects, such as kidney functional decline and adverse cardiovascular events, although causality has not been conclusively established. The aims of this review are to: (i) establish factors associated with increased serum accumulation of IS; (ii) report effects of IS accumulation in clinical studies; (iii) critique the reported effects of IS in the kidney, when administered both in vivo and in vitro; and (iv) summarize both established and hypothetical therapeutic options for reducing serum IS or antagonizing its reported downstream effects in the kidney.
Collapse
Affiliation(s)
- Robert J Ellis
- Centre for Kidney Disease Research, Translational Research Institute, School of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - David M Small
- Centre for Kidney Disease Research, Translational Research Institute, School of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - David A Vesey
- Centre for Kidney Disease Research, Translational Research Institute, School of Medicine, University of Queensland, Brisbane, Queensland, Australia.,Department of Renal Medicine, University of Queensland at Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - David W Johnson
- Centre for Kidney Disease Research, Translational Research Institute, School of Medicine, University of Queensland, Brisbane, Queensland, Australia.,Department of Renal Medicine, University of Queensland at Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Ross Francis
- Department of Renal Medicine, University of Queensland at Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Luis Vitetta
- Sydney Medical School - Medical Sciences, Medlab, Sydney, New South Wales, Australia.,Medlab Clinical Ltd., Medlab, Sydney, New South Wales, Australia
| | - Glenda C Gobe
- Centre for Kidney Disease Research, Translational Research Institute, School of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Christudas Morais
- Centre for Kidney Disease Research, Translational Research Institute, School of Medicine, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
14
|
Yang K, Xu X, Nie L, Xiao T, Guan X, He T, Yu Y, Liu L, Huang Y, Zhang J, Zhao J. Indoxyl sulfate induces oxidative stress and hypertrophy in cardiomyocytes by inhibiting the AMPK/UCP2 signaling pathway. Toxicol Lett 2015; 234:110-9. [DOI: 10.1016/j.toxlet.2015.01.021] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Revised: 01/24/2015] [Accepted: 01/28/2015] [Indexed: 12/24/2022]
|