Skip to main content
Log in

RAS inhibition modulates kynurenine levels in a CKD population with and without type 2 diabetes mellitus

  • Nephrology - Original Paper
  • Published:
International Urology and Nephrology Aims and scope Submit manuscript

Abstract

Kynurenine pathway of tryptophan metabolism is involved in the pathophysiology of chronic kidney disease (CKD) and diabetes mellitus, mainly through the inflammation-induced activity of indoleamine 2,3-dioxygenase (IDO), and few studies have investigated its potential link with proteinuria. Renin–angiotensin system inhibitors (RASis) are recommended in these patients to decrease proteinuria, slow CKD progression and reduce cardiovascular risk, but whether these drugs influence kynurenine levels in humans is unknown. We evaluated serum tryptophan and kynurenine in patients suffering from CKD with or without type 2 diabetes mellitus, their correlations with markers of reduced kidney function, and their relationship with RAS-inhibiting therapy. Of 72 adult patients enrolled, 55 were receiving RASis, whereas 17 were not. Tryptophan was assessed by HPLC (high-performance liquid chromatography); kynurenine was measured using an enzyme-linked immunosorbent assay kit; IDO activity (%) was calculated with the formula (kynurenine/tryptophan) × 100. Kynurenine levels were significantly lower in the group under RASis compared to the untreated group (1.56 ± 0.79 vs 2.16 ± 1.51 µmol/l; P = 0.0378). In patients not receiving RASis, kynurenine was inversely related to estimated glomerular filtration rate (eGFR) (r = − 0.4862; P = 0.0478) and directly related to both proteinuria (ρ = 0.493; P = 0.0444) and albuminuria (ρ = 0.542; P = 0.0247). IDO activity was higher in patients with history of cardiovascular disease compared to patients with no such history, and it negatively correlated with eGFR (ρ = − 0.554; P = 0.0210) in the same group. These findings may contribute to explain the well-known beneficial effects of RAS inhibition in CKD population, especially considering that kynurenine is emerging as a potential new biomarker of CKD.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8

Similar content being viewed by others

References

  1. Cernaro V, Medici MA, Leonello G et al (2015) Auxin induces cell proliferation in an experimental model of mammalian renal tubular epithelial cells. Ren Fail 37(5):911–913

    Article  CAS  Google Scholar 

  2. Law KP, Zhang H (2017) The pathogenesis and pathophysiology of gestational diabetes mellitus: deductions from a three-part longitudinal metabolomics study in China. Clin Chim Acta 468:60–70

    Article  CAS  Google Scholar 

  3. Schwarcz R, Bruno JP, Muchowski PJ et al (2012) Kynurenines in the mammalian brain: when physiology meets pathology. Nat Rev Neurosci 13(7):465–477

    Article  CAS  Google Scholar 

  4. Oxenkrug GF (2007) Genetic and hormonal regulation of tryptophan kynurenine metabolism: implications for vascular cognitive impairment, major depressive disorder, and aging. Ann N Y Acad Sci 1122:35–49

    Article  CAS  Google Scholar 

  5. Jayawickrama GS, Nematollahi A, Sun G et al (2018) Improvement of kynurenine aminotransferase-II inhibitors guided by mimicking sulfate esters. PLoS ONE 13(4):e0196404

    Article  Google Scholar 

  6. Larkin PB, Sathyasaikumar KV, Notarangelo FM et al (2016) Tryptophan 2,3-dioxygenase and indoleamine 2,3-dioxygenase 1 make separate, tissue-specific contributions to basal and inflammation-induced kynurenine pathway metabolism in mice. Biochim Biophys Acta 1860(11 Pt A):2345–2354

    Article  CAS  Google Scholar 

  7. Oxenkrug G (2013) Insulin resistance and dysregulation of tryptophan-kynurenine and kynurenine-nicotinamide adenine dinucleotide metabolic pathways. Mol Neurobiol 48(2):294–301

    Article  CAS  Google Scholar 

  8. Debnath S, Velagapudi C, Redus L et al (2017) Tryptophan metabolism in patients with chronic kidney disease secondary to type 2 diabetes: relationship to inflammatory markers. Int J Tryptophan Res 10:1178646917694600

    Article  Google Scholar 

  9. Schefold JC, Zeden JP, Fotopoulou C et al (2009) Increased indoleamine 2,3-dioxygenase (IDO) activity and elevated serum levels of tryptophan catabolites in patients with chronic kidney disease: a possible link between chronic inflammation and uraemic symptoms. Nephrol Dial Transplant 24(6):1901–1908

    Article  CAS  Google Scholar 

  10. Vanholder R, De Smet R, Glorieux G et al (2003) Review on uremic toxins: classification, concentration, and interindividual variability. Kidney Int 63(5):1934–1943

    Article  CAS  Google Scholar 

  11. Korstanje R, Deutsch K, Bolanos-Palmieri P et al (2016) Loss of kynurenine 3-mono-oxygenase causes proteinuria. J Am Soc Nephrol 27(11):3271–3277

    Article  CAS  Google Scholar 

  12. Hirayama A, Nakashima E, Sugimoto M et al (2012) Metabolic profiling reveals new serum biomarkers for differentiating diabetic nephropathy. Anal Bioanal Chem 404(10):3101–3109

    Article  CAS  Google Scholar 

  13. Group KDIGOKCW (2013) KDIGO clinical practice guideline for the evaluation and management of chronic kidney disease. Kidney Int Suppl 3(1):1–150

    Article  Google Scholar 

  14. Levey AS, Stevens LA, Schmid CH et al (2009) A new equation to estimate glomerular filtration rate. Ann Intern Med 150(9):604–612

    Article  Google Scholar 

  15. Zhang Y, Ruan Y, Zhang P et al (2017) Increased indoleamine 2,3-dioxygenase activity in type 2 diabetic nephropathy. J Diabetes Complicat 31(1):223–227

    Article  Google Scholar 

  16. Atlas SA (2007) The renin–angiotensin aldosterone system: pathophysiological role and pharmacologic inhibition. J Manag Care Pharm. 13(8 Suppl B):9–20

    PubMed  Google Scholar 

  17. Pacurari M, Kafoury R, Tchounwou PB et al (2014) The Renin–Angiotensin–aldosterone system in vascular inflammation and remodeling. Int J Inflam 2014:689360

    Article  Google Scholar 

  18. Nishiyama A, Kim-Mitsuyama S (2010) New approaches to blockade of the renin-angiotensin-aldosterone system: overview of regulation of the renin–angiotensin-aldosterone system. J Pharmacol Sci 113(4):289–291

    Article  CAS  Google Scholar 

  19. Roscioni SS, Heerspink HJ, de Zeeuw D (2014) The effect of RAAS blockade on the progression of diabetic nephropathy. Nat Rev Nephrol 10(2):77–87

    Article  CAS  Google Scholar 

  20. Cernaro V, Trifirò G, Lorenzano G et al (2014) New therapeutic strategies under development to halt the progression of renal failure. Expert Opin Investig Drugs 23(5):693–709

    Article  CAS  Google Scholar 

  21. Lytvyn Y, Bjornstad P, Pun N et al (2016) New and old agents in the management of diabetic nephropathy. Curr Opin Nephrol Hypertens 25(3):232–239

    Article  CAS  Google Scholar 

  22. Mair RD, Sirich TL, Meyer TW (2018) Uremic toxin clearance and cardiovascular toxicities. Toxins (Basel) 10(6):226

    Article  Google Scholar 

  23. Bartosiewicz J, Kaminski T, Pawlak K et al (2017) The activation of the kynurenine pathway in a rat model with renovascular hypertension. Exp Biol Med (Maywood) 242(7):750–761

    Article  CAS  Google Scholar 

  24. Zakrocka I, Kocki T, Turski WA (2017) The effect of three angiotensin-converting enzyme inhibitors on kynurenic acid production in rat kidney in vitro. Pharmacol Rep 69(3):536–541

    Article  CAS  Google Scholar 

  25. Zakrocka I, Targowska-Duda KM, Wnorowski A et al (2019) Angiotensin II type 1 receptor blockers decrease kynurenic acid production in rat kidney in vitro. Naunyn Schmiedebergs Arch Pharmacol 392(2):209–217

    Article  CAS  Google Scholar 

  26. Widner B, Werner ER, Schennach H et al (1997) Simultaneous measurement of serum tryptophan and kynurenine by HPLC. Clin Chem 43(12):2424–2426

    Article  CAS  Google Scholar 

  27. Badawy AA, Dougherty DM (2016) Assessment of the human kynurenine pathway: comparisons and clinical implications of ethnic and gender differences in plasma tryptophan, kynurenine metabolites, and enzyme expressions at baseline and after acute tryptophan loading and depletion. Int J Tryptophan Res 9:31–49

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Baratta AM, Buck SA, Buchla AD et al (2018) Sex differences in hippocampal memory and kynurenic acid formation following acute sleep deprivation in rats. Sci Rep 8(1):6963

    Article  Google Scholar 

  29. Chauvel V, Schoenen J, Multon S (2013) Influence of ovarian hormones on cortical spreading depression and its suppression by l-kynurenine in rat. PLoS ONE 8(12):e82279

    Article  Google Scholar 

  30. Fertan E, Stover KRJ, Brant MG et al (2019) Effects of the novel IDO inhibitor DWG-1036 on the behavior of male and female 3xTg-AD mice. Front Pharmacol 10:1044

    Article  CAS  Google Scholar 

  31. Meier TB, Drevets WC, Teague TK et al (2018) Kynurenic acid is reduced in females and oral contraceptive users: Implications for depression. Brain Behav Immun 67:59–64

    Article  CAS  Google Scholar 

  32. Reininghaus EZ, Dalkner N, Riedrich K et al (2019) Sex specific changes in tryptophan breakdown over a 6 week treatment period. Front Psychiatry 10:74

    Article  Google Scholar 

  33. Songtachalert T, Roomruangwong C, Carvalho AF et al (2018) Anxiety disorders: sex differences in serotonin and tryptophan metabolism. Curr Top Med Chem 18(19):1704–1715

    Article  CAS  Google Scholar 

  34. Naz S, Bhat M, Stahl S et al (2019) Dysregulation of the tryptophan pathway evidences gender differences in COPD. Metabolites 9(10):212

    Article  CAS  Google Scholar 

  35. Sas K, Szabo E, Vecsei L (2018) Mitochondria, oxidative stress and the kynurenine system, with a focus on ageing and neuroprotection. Molecules 23(1):191

    Article  Google Scholar 

  36. Oxenkrug GF (2010) Metabolic syndrome, age-associated neuroendocrine disorders, and dysregulation of tryptophan-kynurenine metabolism. Ann N Y Acad Sci 1199:1–14

    Article  CAS  Google Scholar 

  37. Pawlak K, Domaniewski T, Mysliwiec M et al (2009) The kynurenines are associated with oxidative stress, inflammation and the prevalence of cardiovascular disease in patients with end-stage renal disease. Atherosclerosis 204(1):309–314

    Article  CAS  Google Scholar 

  38. Silva RE, Baldim JL, Chagas-Paula DA et al (2018) Predictive metabolomic signatures of end-stage renal disease: a multivariate analysis of population-based data. Biochimie 152:14–30

    Article  CAS  Google Scholar 

  39. Canadas-Garre M, Anderson K, McGoldrick J et al (2019) Proteomic and metabolomic approaches in the search for biomarkers in chronic kidney disease. J Proteomics 193:93–122

    Article  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Valeria Cernaro.

Ethics declarations

Conflict of interest

On behalf of all authors, the corresponding author states that there is no conflict of interest.

Research involving human participants

There is no research involving human participants.

Informed consent

Written informed consent was obtained from all recruited patients.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Cernaro, V., Loddo, S., Macaione, V. et al. RAS inhibition modulates kynurenine levels in a CKD population with and without type 2 diabetes mellitus. Int Urol Nephrol 52, 1125–1133 (2020). https://doi.org/10.1007/s11255-020-02469-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11255-020-02469-z

Keywords

Navigation