403
Views
2
CrossRef citations to date
0
Altmetric
Review

An Update of Urine and Blood Metabolomics in Chronic Kidney Disease

&
Pages 577-596 | Received 06 Jun 2018, Accepted 12 Mar 2019, Published online: 29 May 2019

References

  • Bouatra S , AziatF, MandalRet al. The human urine metabolome. PloS ONE8(9), e73076 (2013).
  • Patti GJ , YanesO, SiuzdakG. Innovation: metabolomics: the apogee of the omics trilogy. Nat. Rev. Mol. Cell Biol.13(4), 263 (2012).
  • Turi KN , Romick-RosendaleL, RyckmanKK, HartertTV. A review of metabolomics approaches and their application in identifying causal pathways of childhood asthma. J. Allergy. Clin. Immunol.141(4), 1191–1201 (2018).
  • Khamis MM , AdamkoDJ, El-AneedA. Mass spectrometric based approaches in urine metabolomics and biomarker discovery. Mass. Spectrom. Rev.36(2), 115–134 (2017).
  • Kettunen J , TukiainenT, SarinA-Pet al. Genome-wide association study identifies multiple loci influencing human serum metabolite levels. Nat. Genet.44(3), 269 (2012).
  • López-López Á , López-GonzálvezÁ, Barker-TejedaTC, BarbasC. A review of validated biomarkers obtained through metabolomics. Expert. Rev. Mol. Diagn.18(6), 557–575 (2018).
  • Dettmer K , AronovPA, HammockBD. Mass spectrometry-based metabolomics. Mass. Spectrom. Rev.26(1), 51–78 (2007).
  • Roberts LD , SouzaAL, GersztenRE, ClishCB. Targeted metabolomics. Curr. Protoc. Mol.98(1), 30–32 (2012).
  • Ioannidis JP , KhouryMJ. Improving validation practices in “omics” research. Science334(6060), 1230–1232 (2011).
  • Johnson CH , IvanisevicJ, SiuzdakG. Metabolomics: beyond biomarkers and towards mechanisms. Nat. Rev. Mol. Cell Biol.17(7), 451 (2016).
  • Emwas A-HM . The strengths and weaknesses of NMR spectroscopy and mass spectrometry with particular focus on metabolomics research. Methods. Mol. Biol.1277, 161–193 (2015).
  • Hocher B , AdamskiJ. Metabolomics for clinical use and research in chronic kidney disease. Nat. Rev. Nephrol.13(5), 269 (2017).
  • Wilmes A , LimoncielA, AschauerLet al. Application of integrated transcriptomic, proteomic and metabolomic profiling for the delineation of mechanisms of drug induced cell stress. J. Proteomics.79, 180–194 (2013).
  • Ussher JR , ElmariahS, GersztenRE, DyckJR. The emerging role of metabolomics in the diagnosis and prognosis of cardiovascular disease. J. Am. Coll. Cardiol.68(25), 2850–2870 (2016).
  • Boelaert J , LynenF, GlorieuxG, SchepersE, NeirynckN, VanholderR. Metabolic profiling of human plasma and urine in chronic kidney disease by hydrophilic interaction liquid chromatography coupled with time-of-flight mass spectrometry: a pilot study. Anal. Bioanal. Chem.409(8), 2201–2211 (2017).
  • Jha V , Garcia-GarciaG, IsekiKet al. Chronic kidney disease: global dimension and perspectives. Lancet382(9888), 260–272 (2013).
  • Eddy AA . Progression in chronic kidney disease. Adv Chronic Kidney Dis.12(4), 353–365 (2005).
  • Hill NR , FatobaST, OkeJLet al. Global prevalence of chronic kidney disease – a systematic review and meta-analysis. PloS ONE11(7), e0158765 (2016).
  • Chen D-Q , CaoG, ChenHet al. Gene and protein expressions and metabolomics exhibit activated redox signaling and wnt/β-catenin pathway are associated with metabolite dysfunction in patients with chronic kidney disease. Redox. Biol.12, 505–521 (2017).
  • Zhang ZH , ChenH, VaziriNDet al. Metabolomic signatures of chronic kidney disease of diverse etiologies in the rats and humans. J. Proteome Res.15(10), 3802–3812 (2016).
  • Niewczas MA , SirichTL, MathewAVet al. Uremic solutes and risk of end-stage renal disease in Type 2 diabetes: metabolomic study. Kidney. Int.85(5), 1214–1224 (2014).
  • Kienana M , LydieND, Jean-MichelHet al. Elucidating time-dependent changes in the urinary metabolome of renal transplant patients by a combined 1H NMR and GC–MS approach. Mol. Biosyst.11(9), 2493–2510 (2015).
  • Sharma K , KarlB, MathewAVet al. Metabolomics reveals signature of mitochondrial dysfunction in diabetic kidney disease. J. Am. Soc. Nephrol.24(11), 1901–1912 (2013).
  • Qi S , OuyangX, WangL, PengW, WenJ, DaiY. A Pilot metabolic profiling study in serum of patients with chronic kidney disease based on 1H-NMR-spectroscopy. Clin. Transl. Sci.5(5), 379–385 (2012).
  • Posada-Ayala M , ZubiriI, Martin-LorenzoMet al. Identification of a urine metabolomic signature in patients with advanced-stage chronic kidney disease. Kidney Int.85(1), 103–111 (2014).
  • Gil R , HeinzmannS, WörmannKet al. FP292 non-targeted metabolomics approaches for elucidation of novel diagnostic markers in chronic kidney disease. Nephrol. Dial. Transplant.30(Suppl. 3), iii164–iii165 (2015).
  • Toyohara T , AkiyamaY, SuzukiTet al. Metabolomic profiling of uremic solutes in CKD patients. Hypertens. Res.33(9), 944 (2010).
  • Goek ON , PrehnC, SekulaPet al. Metabolites associate with kidney function decline and incident chronic kidney disease in the general population. Nephrol. Dial. Transplant.28(8), 2131–2138 (2013).
  • Hirayama A , NakashimaE, SugimotoMet al. Metabolic profiling reveals new serum biomarkers for differentiating diabetic nephropathy. Anal. Bioanal. Chem.404(10), 3101–3109 (2012).
  • Pena M , LambersHeerspink H, HellemonsMet al. Urine and plasma metabolites predict the development of diabetic nephropathy in individuals with Type 2 diabetes mellitus. Diabet. Med.31(9), 1138–1147 (2014).
  • Shah VO , TownsendRR, FeldmanHI, PappanKL, KensickiE, Vander JagtDL. Plasma metabolomic profiles in different stages of CKD. Clin. J. Am. Soc. Nephrol.8(3), 363–370 (2013).
  • Benito S , SánchezA, UncetaNet al. LC–QTOF–MS-based targeted metabolomics of arginine–creatine metabolic pathway-related compounds in plasma: application to identify potential biomarkers in pediatric chronic kidney disease. Anal. Bioanal. Chem.408(3), 747–760 (2016).
  • Afshinnia F , RajendiranTM, SoniTet al. Impaired β-oxidation and altered complex lipid fatty acid partitioning with advancing CKD. J. Am. Soc. Nephrol.29(1), 295–306 (2018).
  • Han LD , XiaJF, LiangQLet al. Plasma esterified and non-esterified fatty acids metabolic profiling using gas chromatography–mass spectrometry and its application in the study of diabetic mellitus and diabetic nephropathy. Anal. Chim. Acta689(1), 85–91 (2011).
  • Wang L , HuC, LiuSet al. Plasma lipidomics investigation of hemodialysis effects by using liquid chromatography–mass spectrometry. J. Proteome. Res.15(6), 1986–1994 (2016).
  • Emwas A-HM , SalekRM, GriffinJL, MerzabanJ. NMR-based metabolomics in human disease diagnosis: applications, limitations, and recommendations. Metabolomics9(5), 1048–1072 (2013).
  • Markley JL , BrüschweilerR, EdisonASet al. The future of NMR-based metabolomics. Curr. Opin. Biotechnol.43, 34–40 (2017).
  • Daly RA , BortonMA, WilkinsMJet al. Microbial metabolisms in a 2.5-km-deep ecosystem created by hydraulic fracturing in shales. Nat. Microbiol.1(10), 16146 (2016).
  • Bingol K . Recent advances in targeted and untargeted metabolomics by NMR and MS/NMR methods. High-Throughput7(2), 9 (2018).
  • McKay RT . How the 1D-NOESY suppresses solvent signal in metabonomics NMR spectroscopy: an examination of the pulse sequence components and evolution. Concepts. Magn. Reson. Part A38(5), 197–220 (2011).
  • Kalantari S , NafarM, SamavatSet al.. 1H NMR-based metabolomics exploring urinary biomarkers correlated with proteinuria in focal segmental glomerulosclerosis: a pilot study. Magn. Reson. Chem.54(10), 821–826 (2016).
  • Sandusky P , RafteryD. Use of selective TOCSY NMR experiments for quantifying minor components in complex mixtures: application to the metabonomics of amino acids in honey. Anal. Chem.77(8), 2455–2463 (2005).
  • Xi Y , DeRopp JS, ViantMR, WoodruffDL, YuP. Automated screening for metabolites in complex mixtures using 2D COSY NMR spectroscopy. Metabolomics2(4), 221–233 (2006).
  • Mannina L , SobolevA, CapitaniDet al. NMR metabolic profiling of organic and aqueous sea bass extracts: implications in the discrimination of wild and cultured sea bass. Talanta77(1), 433–444 (2008).
  • Gowda GN , DjukovicD. Overview of mass spectrometry-based metabolomics: opportunities and challenges. Methods Mol Biol.1198, 3–12 (2014).
  • Sparkman OD , PentonZ, KitsonFG. Gas Chromatography and Mass Spectrometry: A Practical Guide. Academic Press, USA (2011).
  • Zhao YY , ChengXL, VaziriND, LiuS, LinRC. UPLC–based metabonomic applications for discovering biomarkers of diseases in clinical chemistry. Clin. Biochem.47(15), 16–26 (2014).
  • Zhou B , XiaoJF, TuliL, RessomHW. LC–MS-based metabolomics. Mol. Biosyst.8(2), 470–481 (2012).
  • Bajad SU , LuW, KimballEH, YuanJ, PetersonC, RabinowitzJD. Separation and quantitation of water soluble cellular metabolites by hydrophilic interaction chromatography–tandem mass spectrometry. J. Chromatogr. A1125(1), 76–88 (2006).
  • Zhao YY , ShenX, ChengXL, WeiF, BaiX, LinRC. Urinary metabonomics study on the protective effects of ergosta-4, 6, 8 (14), 22-tetraen-3-one on chronic renal failure in rats using UPLC Q-TOF/MS and a novel MSE data collection technique. Process Biochem.47(12), 1980–1987 (2012).
  • Wrona M , MaurialaT, BatemanKP, Mortishire-SmithRJ, O'ConnorD. ‘All-in-one’ analysis for metabolite identification using liquid chromatography/hybrid quadrupole time-of-flight mass spectrometry with collision energy switching. Rapid. Commun. Mass. Spectrom.19(18), 2597–2602 (2005).
  • Zhao YY , LiuJ, ChengXL, BaiX, LinRC. Urinary metabonomics study on biochemical changes in an experimental model of chronic renal failure by adenine based on UPLC Q-TOF/MS. Clin. Chim. Acta.413(5–6), 642–649 (2012).
  • Zhao YY , FengYL, BaiX, TanXJ, LinRC, MeiQ. Ultra performance liquid chromatography-based metabonomic study of therapeutic effect of the surface layer of Poria cocos on adenine-induced chronic kidney disease provides new insight into anti-fibrosis mechanism. PloS ONE8(3), e59617 (2013).
  • Zhao YY , ChenH, TianT, ChenDQ, BaiX, WeiF. A pharmaco-metabonomic study on chronic kidney disease and therapeutic effect of ergone by UPLC–QTOF/HDMS. PLoS ONE9(12), e115467 (2014).
  • Zhao YY , ChengXL, WeiFet al. Intrarenal metabolomic investigation of chronic kidney disease and its TGF-β1 mechanism in induced-adenine rats using UPLC Q-TOF/HSMS/MSE. J. Proteome Res.12(2), 692–703 (2013).
  • Zhao YY , LinRC. UPLC–MSE application in disease biomarker discovery: the discoveries in proteomics to metabolomics. Chem. Biol. Interact.215, 7–16 (2014).
  • Psychogios N , HauDD, PengJet al. The human serum metabolome. PloS ONE6(2), e16957 (2011).
  • West JB . Study Guide and Self-examination Review for Best and Taylor's Physiological Basis of Medical Practice. Williams & Wilkins, USA (1985).
  • Fischer S . Analysis of cardiovascular eicosanoids in man with special reference to HPLC. Chromatographia22(7–12), 416–420 (1986).
  • Yu Z , KastenmüllerG, HeYet al. Differences between human plasma and serum metabolite profiles. PloS ONE6(7), e21230 (2011).
  • Liu L , AaJ, WangGet al. Differences in metabolite profile between blood plasma and serum. Anal. Biochem.406(2), 105–112 (2010).
  • Suarez-Diez M , AdamJ, AdamskiJet al. Plasma and serum metabolite association networks: comparability within and between studies using NMR and MS profiling. J. Proteome. Res.16(7), 2547–2559 (2017).
  • Boudah S , OlivierM-F, Aros-CaltSet al. Annotation of the human serum metabolome by coupling three liquid chromatography methods to high-resolution mass spectrometry. J. Chromatogr. B966, 34–47 (2014).
  • Tizianello A , DeFerrari G, GaribottoG, GurreriG, RobaudoC. Renal metabolism of amino acids and ammonia in subjects with normal renal function and in patients with chronic renal insufficiency. J. Clin. Invest.65(5), 1162–1173 (1980).
  • Rhee EP , ClishCB, GhorbaniAet al. A combined epidemiologic and metabolomic approach improves CKD prediction. J. Am. Soc. Nephrol.24(8), 1330–1338 (2013).
  • Kalim S , RheeEP. An overview of renal metabolomics. Kidney. Int.91(1), 61–69 (2017).
  • Debnath S , VelagapudiC, RedusLet al. Tryptophan metabolism in patients with chronic kidney disease secondary to Type 2 diabetes: relationship to inflammatory markers. Int. J. Tryptophan. Res.10, 1178646917694600 (2017).
  • Wang Y , LiuH, McKenzieGet al. Kynurenine is an endothelium-derived relaxing factor produced during inflammation. Nat. Med.16(3), 279 (2010).
  • Roshanravan B , ZelnickLR, DjucovicDet al. Chronic kidney disease attenuates the plasma metabolome response to insulin. JCI Insight.3(16), 122219 (2018).
  • Kimura T , YasudaK, YamamotoRet al. Identification of biomarkers for development of end-stage kidney disease in chronic kidney disease by metabolomic profiling. Sci. Rep.6, 26138 (2016).
  • Kobayashi T , YoshidaT, FujisawaTet al. A metabolomics-based approach for predicting stages of chronic kidney disease. Biochem. Biophys. Res. Commun.445(2), 412–416 (2014).
  • Karu N , McKercherC, NicholsDSet al. Tryptophan metabolism, its relation to inflammation and stress markers and association with psychological and cognitive functioning: Tasmanian chronic kidney disease pilot study. BMC Nephrol.17(1), 171 (2016).
  • Niewczas MA , MathewAV, CroallSet al. Circulating modified metabolites and a risk of ESRD in patients with Type 1 diabetes and chronic kidney disease. Diabetes. Care.40 (3), 383–390 (2017).
  • Mika A , WojtowiczW, ZąbekAet al. Application of nuclear magnetic resonance spectroscopy for the detection of metabolic disorders in patients with moderate kidney insufficiency. J. Pharm. Biomed. Anal.149, 1–8 (2018).
  • Benito S , Sanchez-OrtegaA, UncetaNet al. Untargeted metabolomics for plasma biomarker discovery for early chronic kidney disease diagnosis in pediatric patients using LC–QTOF–MS. The Analyst143(18), 4448–4458 (2018).
  • Rysz J , Gluba-BrzózkaA, FranczykB, JabłonowskiZ, Ciałkowska-RyszA. Novel biomarkers in the diagnosis of chronic kidney disease and the prediction of its outcome. Int. J. Mol. Sci.18(8), 1702 (2017).
  • Barrios C , SpectorTD, MenniC. Blood, urine and faecal metabolite profiles in the study of adult renal disease. Arch. Biochem. Biophys.589, 81–92 (2016).
  • Zuppi C , MessanaI, ForniFet al. 1H NMR spectra of normal urines: reference ranges of the major metabolites. Clin. Chim. Acta265(1), 85–97 (1997).
  • Slupsky CM , RankinKN, WagnerJet al. Investigations of the effects of gender, diurnal variation, and age in human urinary metabolomic profiles. Anal. Chem.79(18), 6995–7004 (2007).
  • Mandal R , GuoAC, ChaudharyKKet al. Multi-platform characterization of the human cerebrospinal fluid metabolome: a comprehensive and quantitative update. Genome Med.4(4), 38 (2012).
  • Takeda I , StretchC, BarnabyPet al. Understanding the human salivary metabolome. NMR Biomedicine22(6), 577–584 (2009).
  • Kielstein A , TsikasD, GallowayGP, MendelsonJE. Asymmetric dimethylarginine (ADMA) – a modulator of nociception in opiate tolerance and addiction?Nitric Oxide17(2), 55–59 (2007).
  • Mcbride AE , SilverPA. State of the arg: protein methylation at arginine comes of age. Cell106(1), 5–8 (2001).
  • Achan V , BroadheadM, MalakiMet al. Asymmetric dimethylarginine causes hypertension and cardiac dysfunction in humans and is actively metabolized by dimethylarginine dimethylaminohydrolase. Arterioscler. Thromb. Vasc. Biol.23(8), 1455–1459 (2003).
  • Kielstein JT , FliserD. The past, presence and future of ADMA in nephrology. Nephrol. Ther.3(2), 47–54 (2007).
  • Leone A , MoncadaS, VallanceP, CalverA, CollierJ. Accumulation of an endogenous inhibitor of nitric oxide synthesis in chronic renal failure. Lancet339(8793), 572–575 (1992).
  • Osamu S , MasatoT, TsuyoshiM. Asymmetric dimethylarginine produces vascular lesions in endothelial nitric oxide synthase-deficient mice. Involvement of renin-angiotensin system and oxidative stress. Arterioscler. Thromb. Vasc. Biol24, 1682–1688 (2004).
  • Wang D , GillPS, ChabrashviliTet al. Isoform-specific regulation by NG, NG-dimethylarginine dimethylaminohydrolase of rat serum asymmetric dimethylarginine and vascular endothelium-derived relaxing factor/NO. Circ. Res.101(6), 627–635 (2007).
  • Fleck C , JanzA, SchweitzerF, KargeE, SchwertfegerM, SteinG. Serum concentrations of asymmetric (ADMA) and symmetric (SDMA) dimethylarginine in renal failure patients. Kidney. Int.59, S14–S18 (2001).
  • Baylis C . Arginine, arginine analogs and nitric oxide production in chronic kidney disease. Nat. Rev. Nephrol.2(4), 209 (2006).
  • Raptis V , KapoulasS, GrekasD. Role of asymmetrical dimethylarginine in the progression of renal disease. Nephrology18(1), 11–21 (2013).
  • Zoccali C , KielsteinJT. Asymmetric dimethylarginine: a new player in the pathogenesis of renal disease?Curr. Opin. Nephrol. Hypertens.15(3), 314–320 (2006).
  • Zoccali C , BenedettoFA, MaasRet al. Asymmetric dimethylarginine, C-reactive protein, and carotid intima-media thickness in end-stage renal disease. J. Am. Soc. Nephrol.13(2), 490–496 (2002).
  • Lundin U . Biomarker discovery in diabetic nephropathy by targeted metabolomics [Masters thesis]. Linköping University, Linköping, Sweden (2008).
  • Kuo HC , HsuCN, HuangCF, LoMH, ChienSJ, TainYL. Urinary arginine methylation index associated with ambulatory blood pressure abnormalities in children with chronic kidney disease. J. Am. Soc. Hypertens.6(6), 385–392 (2012).
  • Duranton F , LundinU, GayrardNet al. Plasma and urinary amino acid metabolomic profiling in patients with different levels of kidney function. Clin. J. Am. Soc. Nephrol.9(1), 37–45 (2014).
  • Kanzelmeyer NK , PapeL, Chobanyan-JürgensKet al. L-arginine/NO pathway is altered in children with haemolytic-uraemic syndrome (HUS). Oxid. Med. Cell. Longev.2014, Article ID 203512 (2014).
  • Mihout F , ShwekeN, BigéNet al. Asymmetric dimethylarginine (ADMA) induces chronic kidney disease through a mechanism involving collagen and TGF-β1 synthesis. J. Pathol.223(1), 37–45 (2011).
  • Benito S , Sanchez-OrtegaA, UncetaNet al. Plasma biomarker discovery for early chronic kidney disease diagnosis based on chemometric approaches using LC–QTOF targeted metabolomics data. J. Pharm. Biomed. Anal.149, 46–56 (2018).
  • Breit M , WeinbergerKM. Metabolic biomarkers for chronic kidney disease. Arch. Biochem. Biophys.589, 62–80 (2016).
  • Stubbs JR , HouseJA, OcqueAJet al. Serum trimethylamine-N-oxide is elevated in CKD and correlates with coronary atherosclerosis burden. J. Am. Soc. Nephrol.27(1), 305–313 (2016).
  • Cho CE , TaesuwanS, MalyshevaOVet al. Trimethylamine-N-oxide (TMAO) response to animal source foods varies among healthy young men and is influenced by their gut microbiota composition: a randomized controlled trial. Mol. Nutr. Food. Res.61(1), 1600324 (2017).
  • De Filippis F , PellegriniN, VanniniLet al. High-level adherence to a Mediterranean diet beneficially impacts the gut microbiota and associated metabolome. Gut65(11), 1812–1821 (2016).
  • Lang D , YeungC, PeterRet al. Isoform specificity of trimethylamine N-oxygenation by human flavin-containing monooxygenase (FMO) and P450 enzymes: selective catalysis by FMO3. Biochem. Pharmacol.56(8), 1005–1012 (1998).
  • Mitchell S , ZhangA, NobletJ, GillespieS, JonesN, SmithR. Metabolic disposition of [14C]-trimethylamine N-oxide in rat: variation with dose and route of administration. Xenobiotica27(11), 1187–1197 (1997).
  • Cho CE , CaudillMA. Trimethylamine-N-oxide: friend, foe, or simply caught in the cross-fire?Trends. Endocrinol. Metab.28(2), 121–130 (2017).
  • Yancey PH . Organic osmolytes as compatible, metabolic and counteracting cytoprotectants in high osmolarity and other stresses. J. Exp. Biol.208(15), 2819–2830 (2005).
  • Kumemoto R , YusaK, ShibayamaT, HatoriK. Trimethylamine N-oxide suppresses the activity of the actomyosin motor. Biochim. Biophys. Acta1820(10), 1597–1604 (2012).
  • Lin T-Y , TimasheffSN. Why do some organisms use a urea-methylamine mixture as osmolyte? Thermodynamic compensation of urea and trimethylamine N-oxide interactions with protein. Biochemistry33(42), 12695–12701 (1994).
  • Ma J , PazosIM, GaiF. Microscopic insights into the protein-stabilizing effect of trimethylamine N-oxide (TMAO). Proc. Natil Acad. Sci. USA111(23), 8476–8481 (2014).
  • Velasquez MT , RamezaniA, ManalA, RajDS. Trimethylamine N-oxide: the good, the bad and the unknown. Toxins8(11), 326 (2016).
  • Yamamoto S , KazamaJJ, WakamatsuTet al. Removal of uremic toxins by renal replacement therapies: a review of current progress and future perspectives. Ren. Replace. Ther.2(1), 43 (2016).
  • Bain MA , FaullR, FornasiniG, MilneRW, EvansAM. Accumulation of trimethylamine and trimethylamine-N-oxide in end-stage renal disease patients undergoing haemodialysis. Nephrol. Dial. Transplant.21(5), 1300–1304 (2006).
  • Missailidis C , HallqvistJ, QureshiARet al. Serum trimethylamine-N-oxide is strongly related to renal function and predicts outcome in chronic kidney disease. PLoS ONE11(1), e0141738 (2016).
  • Manor O , ZubairN, ConomosMPet al. A multi-omic association study of trimethylamine N-oxide. Cell. Rep.24(4), 935–946 (2018).
  • Xu K-Y , XiaG-H, LuJ-Qet al. Impaired renal function and dysbiosis of gut microbiota contribute to increased trimethylamine-N-oxide in chronic kidney disease patients. Sci. Rep.7(1), 1445–1445 (2017).
  • Gruppen EG , GarciaE, ConnellyMAet al. TMAO is associated with mortality: impact of modestly impaired renal function. Sci. Rep.7(1), 13781 (2017).
  • Nakajima T , MatsuokaY, KakimotoY. Isolation and identification of NG-monomethyl NG, NG-dimethyl-and NG, NG-dimethylarginine from the hydrolysate of proteins of bovine brain. Biochim. Biophys. Acta230(2), 212–222 (1971).
  • Morales Y , CáceresT, MayK, HevelJM. Biochemistry and regulation of the protein arginine methyltransferases (PRMTs). Arch. Biochem. Biophys.590, 138–152 (2016).
  • Tain YL , HsuCN. Toxic dimethylarginines: asymmetric dimethylarginine (ADMA) and symmetric dimethylarginine (SDMA). Toxins9(3), 92 (2017).
  • Kakimoto Y , AkazawaS. Isolation and identification of NG, NG-and NG, N'G-dimethylarginine, Nε-mono-, di-, and trimethyllysine, and glucosylgalactosyl-and galactosyl-δ-hydroxylysine from human urine. J. Biol. Chem.245(21), 5751–5758 (1970).
  • Mcdermott J . Studies on the catabolism of Ng-methylarginine, Ng, Ng-dimethylarginine and Ng, Ng-dimethylarginine in the rabbit. Biochem. J.154(1), 179–184 (1976).
  • Bode-Böger SM , ScaleraF, KielsteinJTet al. Symmetrical dimethylarginine: a new combined parameter for renal function and extent of coronary artery disease. J. Am. Soc. Nephrol.17(4), 1128–1134 (2006).
  • Closs EI , BashaFZ, HabermeierA, FörstermannU. Interference of L-arginine analogues with L-arginine transport mediated by the y+ carrier hCAT-2B. Nitric Oxide1(1), 65–73 (1997).
  • Kielstein JT , SalpeterSR, Bode-BoegerSM, CookeJP, FliserD. Symmetric dimethylarginine (SDMA) as endogenous marker of renal function – a meta-analysis. Nephrol. Dial. Transplant.21(9), 2446–2451 (2006).
  • MaCallister R , RambausekM, VallanceP, WilliamsD, HoffmannKH, RitzE. Concentration of dimethyl-L-arginine in the plasma of patients with end-stage renal failure. Nephrol. Dial. Transplant.11(12), 2449–2452 (1996).
  • El-Sadek AE , BeheryEG, AzabAAet al. Arginine dimethylation products in pediatric patients with chronic kidney disease. Ann. Med. Surg.9, 22–27 (2016).
  • El-Khoury JM , BunchDR, HuB, PaytoD, ReineksEZ, WangS. Comparison of symmetric dimethylarginine with creatinine, cystatin C and their eGFR equations as markers of kidney function. Clin. Biochem.49(15), 1140–1143 (2016).
  • Torrent E , PlanellasM, OrdeixL, PastorJ, RodonJ, Solano-GallegoL. Serum symmetric dimethylarginine as an early marker of excretory dysfunction in canine leishmaniosis (L. infantum) induced nephropathy. Vet. Med. Int. 2018, 7517359 (2018).
  • Peterson ME , VarelaFV, RishniwM, PolzinDJ. Evaluation of serum symmetric dimethylarginine concentration as a marker for masked chronic kidney disease in cats with hyperthyroidism. J. Vet. Intern. Med.32(1), 295–304 (2018).
  • Van Den Brand JA , MutsaersHA, Van ZuilenADet al. Uremic solutes in chronic kidney disease and their role in progression. PLoS ONE.11(12), e0168117 (2016).
  • Vanholder R , DeSmet R, GlorieuxGet al. Review on uremic toxins: classification, concentration, and interindividual variability. Kidney. Int.63(5), 1934–1943 (2003).
  • Duranton F , CohenG, DeSmet Ret al. Normal and pathologic concentrations of uremic toxins. J. Am. Soc. Nephrol.23(7), 1258–1270 (2012).
  • Kim SH , YuMA, RyuES, JangYH, KangDH. Indoxyl sulfate-induced epithelial-to-mesenchymal transition and apoptosis of renal tubular cells as novel mechanisms of progression of renal disease. Lab Invest.92(4), 488–498 (2012).
  • Mutsaers HA , Caetano-PintoP, SeegersAEet al. Proximal tubular efflux transporters involved in renal excretion of p-cresyl sulfate and p-cresyl glucuronide: implications for chronic kidney disease pathophysiology. Toxicol. In Vitro29(7), 1868–1877 (2015).
  • Owada S , MaebaT, SuganoYet al. Spherical carbon adsorbent (AST-120) protects deterioration of renal function in chronic kidney disease rats through inhibition of reactive oxygen species production from mitochondria and reduction of serum lipid peroxidation. Nephron. Exp. Nephrol.115(4), e101–111 (2010).
  • Mutsaers HA , WilmerMJ, ReijndersDet al. Uremic toxins inhibit renal metabolic capacity through interference with glucuronidation and mitochondrial respiration. Biochim. Biophys. Acta.1832(1), 142–150 (2013).
  • Taes YE , MarescauB, DeVriese Aet al. Guanidino compounds after creatine supplementation in renal failure patients and their relation to inflammatory status. Nephrol. Dial. Transplant.23(4), 1330–1335 (2008).
  • Muller C , EisenbrandG, GradingerMet al. Effects of hemodialysis, dialyser type and iron infusion on oxidative stress in uremic patients. Free. Radic. Res.38(10), 1093–1100 (2004).
  • Ficheux A , GayrardN, SzwarcIet al. The use of SDS-PAGE scanning of spent dialysate to assess uraemic toxin removal by dialysis. Nephrol. Dial. Transplant.26(7), 2281–2289 (2011).
  • Moon SJ , KimDK, ChangJHet al. The impact of dialysis modality on skin hyperpigmentation in haemodialysis patients. Nephrol. Dial. Transplant.24(9), 2803–2809 (2009).
  • Rhee EP , SouzaA, FarrellLet al. Metabolite profiling identifies markers of uremia. J. Am. Soc. Nephrol.21(6), 1041–1051 (2010).
  • Meyer TW , HostetterTH. Uremic solutes from colon microbes. Kidney. Int.81(10), 949–954 (2012).
  • Meijers BK , DeLoor H, BammensB, VerbekeK, VanrenterghemY, EvenepoelP. p-cresyl sulfate and indoxyl sulfate in hemodialysis patients. Clin. J. Am. Soc. Nephrol.4(12), 1932–1938 (2009).
  • Poveda J , Sanchez-NinoMD, GlorieuxGet al. p-cresyl sulphate has pro-inflammatory and cytotoxic actions on human proximal tubular epithelial cells. Nephrol. Dial. Transplant29(1), 56–64 (2014).
  • Sun CY , ChangSC, WuMS. Suppression of Klotho expression by protein-bound uremic toxins is associated with increased DNA methyltransferase expression and DNA hypermethylation. Kidney. Int.81(7), 640–650 (2012).
  • Koppe L , PillonNJ, VellaREet al. p-cresyl sulfate promotes insulin resistance associated with CKD. J. Am. Soc. Nephrol.24(1), 88–99 (2013).
  • Wikoff WR , AnforaAT, LiuJet al. Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proc. Natl Acad. Sci. USA106(10), 3698–3703 (2009).
  • Aronov PA , LuoFJ, PlummerNSet al. Colonic contribution to uremic solutes. J. Am. Soc. Nephrol.22(9), 1769–1776 (2011).
  • Hung SC , KuoKL, WuCC, TarngDC. Indoxyl sulfate: a novel cardiovascular risk factor in chronic kidney disease. J. Am. Heart. Assoc.6(2), e005022 (2017).
  • Lin CJ , LiuHL, PanCFet al. Indoxyl sulfate predicts cardiovascular disease and renal function deterioration in advanced chronic kidney disease. Arch. Med. Res.43(6), 451–456 (2012).
  • Yu M , KimYJ, KangDH. Indoxyl sulfate-induced endothelial dysfunction in patients with chronic kidney disease via an induction of oxidative stress. Clin. J. Am. Soc. Nephrol.6(1), 30–39 (2011).
  • Chitalia VC , ShivannaS, MartorellJet al. Uremic serum and solutes increase post-vascular interventional thrombotic risk through altered stability of smooth muscle cell tissue factor. Circulation127(3), 365–376 (2013).
  • Ito S , OsakaM, HiguchiY, NishijimaF, IshiiH, YoshidaM. Indoxyl sulfate induces leukocyte-endothelial interactions through up-regulation of E-selectin. J. Biol. Chem.285(50), 38869–38875 (2010).
  • Lekawanvijit S , AdrahtasA, KellyDJ, KompaAR, WangBH, KrumH. Does indoxyl sulfate, a uraemic toxin, have direct effects on cardiac fibroblasts and myocytes?Eur. Heart. J.31(14), 1771–1779 (2010).
  • Sun CY , ChangSC, WuMS. Uremic toxins induce kidney fibrosis by activating intrarenal renin–angiotensin–aldosterone system associated epithelial-to-mesenchymal transition. PLoS ONE.7(3), e34026 (2012).
  • Shimizu H , BolatiD, HigashiyamaY, NishijimaF, ShimizuK, NiwaT. Indoxyl sulfate upregulates renal expression of MCP-1 via production of ROS and activation of NF-kappaB, p53, ERK, and JNK in proximal tubular cells. Life. Sci.90(13–14), 525–530 (2012).
  • Takada T , YamamotoT, MatsuoHet al. Identification of ABCG2 as an exporter of uremic toxin indoxyl sulfate in mice and as a crucial factor influencing CKD progression. Sci. Rep.8(1), 11147 (2018).
  • Jourde-Chiche N , DouL, CeriniC, Dignat-GeorgeF, VanholderR, BrunetP. Protein-bound toxins – update 2009. Semin. Dial.22(4), 334–339 (2009).
  • Brito JS , BorgesNA, EsgalhadoM, MaglianoDC, SoulageCO, MafraD. Aryl hydrocarbon receptor activation in chronic kidney disease: role of uremic toxins. Nephron.137(1), 1–7 (2017).
  • Dou L , SalleeM, CeriniCet al. The cardiovascular effect of the uremic solute indole-3 acetic acid. J. Am. Soc. Nephrol.26(4), 876–887 (2015).
  • Borges NA , BarrosAF, NakaoLS, DolengaCJ, FouqueD, MafraD. Protein-bound uremic toxins from gut microbiota and inflammatory markers in chronic kidney disease. J. Ren. Nutr.26(6), 396–400 (2016).
  • Tanaka H , SirichTL, PlummerNS, WeaverDS, MeyerTW. An enlarged profile of uremic solutes. PLoS ONE10(8), e0135657 (2015).
  • Silva RE , BaldimJL, Chagas-PaulaDAet al. Predictive metabolomic signatures of end-stage renal disease: a multivariate analysis of population-based data. Biochimie152, 14–30 (2018).
  • Zhao YY , ChengXL, WeiFet al. Serum metabonomics study of adenine-induced chronic renal failure in rats by ultra performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry. Biomarkers17(1), 48–55 (2012).
  • Zhao YY , ChengXL, CuiJHet al. Effect of ergosta-4, 6, 8 (14), 22-tetraen-3-one (ergone) on adenine-induced chronic renal failure rat: a serum metabonomic study based on ultra performance liquid chromatography/high-sensitivity mass spectrometry coupled with MassLynx i-FIT algorithm. Clin. Chim. Acta413(19–20), 1438–1445 (2012).
  • Zhang ZH , WeiF, VaziriNDet al. Metabolomics insights into chronic kidney disease and modulatory effect of rhubarb against tubulointerstitial fibrosis. Sci. Rep.5, 14472 (2015).
  • Zhang ZH , ChenH, VaziriNDet al. Metabolomic signatures of chronic kidney disease of diverse etiologies in the rats and humans. J. Proteome. Res.15(10), 3802–3812 (2016).
  • Velenosi TJ , HennopA, FeereDAet al. Untargeted plasma and tissue metabolomics in rats with chronic kidney disease given AST-120. Sci. Rep.6, 22526 (2016).
  • Mathew AV , ZengL, ByunJ, PennathurS. Metabolomic profiling of arginine metabolome links altered methylation to chronic kidney disease accelerated atherosclerosis. J. Proteomics Bioinform.Suppl 14, pii: 001 (2015).
  • Kim JA , ChoiHJ, KwonYK, RyuDH, KwonTH, HwangGS. 1H NMR-based metabolite profiling of plasma in a rat model of chronic kidney disease. PLoS ONE.9(1), e85445 (2014).
  • Titan SM , VenturiniG, PadilhaKet al. Metabolites related to eGFR: Evaluation of candidate molecules for GFR estimation using untargeted metabolomics. Clin. Chim. Acta.489, 242–248 (2019).
  • Sekula P , GoekON, QuayeLet al. A metabolome-wide association study of kidney function and disease in the general population. J. Am. Soc. Nephrol.27(4), 1175–1188 (2016).
  • Yu B , ZhengY, NettletonJA, AlexanderD, CoreshJ, BoerwinkleE. Serum metabolomic profiling and incident CKD among African Americans. Clin. J. Am. Soc. Nephrol.9(8), 1410–1417 (2014).
  • Tavares G , VenturiniG, PadilhaKet al. 1,5-Anhydroglucitol predicts CKD progression in macroalbuminuric diabetic kidney disease: results from non-targeted metabolomics. Metabolomics14(4), 39 (2018).
  • Levin A , TonelliM, BonventreJet al. Global kidney health 2017 and beyond: a roadmap for closing gaps in care, research, and policy. Lancet390(10105), 1888–1917 (2017).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.