Marine n-3 Polyunsaturated Fatty Acids and Cellular Senescence Markers in Incident Kidney Transplant Recipients: The Omega-3 Fatty Acids in Renal Transplantation (ORENTRA) Randomized Clinical Trial

Joe Chan, Ivar A Eide, Tone M Tannæs, Bård Waldum-Grevbo, Trond Jenssen, My Svensson, Joe Chan, Ivar A Eide, Tone M Tannæs, Bård Waldum-Grevbo, Trond Jenssen, My Svensson

Abstract

Rationale & objective: Deterioration of kidney graft function is associated with accelerated cellular senescence. Marine n-3 polyunsaturated fatty acids (PUFAs) have favorable properties that may counteract cellular senescence development and damage caused by the senescence-associated secretory phenotype (SASP) secretome. Our objective was to investigate the potential effects of marine n-3 PUFA supplementation on the SASP secretome in kidney transplant recipients.

Study design: Exploratory substudy of the Omega-3 Fatty Acids in Renal Transplantation trial.

Setting & participants: Adult kidney transplant recipients with a functional kidney graft (defined as having an estimated glomerular filtration rate of >30 mL/min/1.73 m2) 8 weeks after engraftment were included in this study conducted in Norway.

Analytical approach: The intervention consisted of 2.6 g of a marine n-3 PUFA or olive oil (placebo) daily for 44 weeks. The outcome was a predefined panel of SASP components in the plasma and urine.

Results: A total of 132 patients were enrolled in the Omega-3 Fatty Acids in Renal Transplantation trial, and 66 patients were allocated to receive either the study drug or placebo. The intervention with the marine n-3 PUFA was associated with reduced plasma levels of granulocyte colony-stimulating factor, interleukin 1α, macrophage inflammatory protein 1α, matrix metalloproteinase (MMP)-1, and MMP-13 compared with the intervention in the control group.

Limitations: Post hoc analysis.

Conclusions: The results suggest that marine n-3 PUFA supplementation has mitigating effects on the plasma SASP components granulocyte colony-stimulating factor, interleukin 1α, macrophage inflammatory protein 1α, MMP-1, and MMP-13 in kidney transplant recipients. Future studies with kidney transplant recipients in maintenance phase, combined with an evaluation of cellular senescence markers in kidney transplant biopsies, are needed to further elucidate the potential antisenescent effect of marine n-3 PUFAs. This trial is registered as NCT01744067.

Keywords: Assay validation; cellular senescence; fatty acid; interleukin; intervention; kidney transplantation; macrophage inflammatory protein; matrix metalloproteinase; monocyte chemoattractant protein; multiplex; omega-3; senescence-associated secretory phenotype; transforming growth factor; tumor necrosis factor.

© 2021 The Authors.

Figures

Figure 1
Figure 1
Study flowchart presenting patient screening, randomization, reasons for withdrawal from the trial, and the number of plasma and urine samples available from each group at baseline and at the end of the study. Abbreviations: ITT, intention to treat; PP, per protocol; PUFA, polyunsaturated fatty acid.

References

    1. van Deursen J.M. The role of senescent cells in ageing. Nature. 2014;509(7501):439–446.
    1. Childs B.G., Durik M., Baker D.J., van Deursen J.M. Cellular senescence in aging and age-related disease: from mechanisms to therapy. Nat Med. 2015;21(12):1424–1435.
    1. Burton D.G., Krizhanovsky V. Physiological and pathological consequences of cellular senescence. Cell Mol Life Sci. 2014;71(22):4373–4386.
    1. Coppé J.P., Desprez P.Y., Krtolica A., Campisi J. The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu Rev Pathol. 2010;5:99–118.
    1. Tchkonia T., Zhu Y., van Deursen J., Campisi J., Kirkland J.L. Cellular senescence and the senescent secretory phenotype: therapeutic opportunities. J Clin Invest. 2013;123(3):966–972.
    1. Melk A., Schmidt B.M., Vongwiwatana A., Rayner D.C., Halloran P.F. Increased expression of senescence-associated cell cycle inhibitor p16INK4a in deteriorating renal transplants and diseased native kidney. Am J Transplant. 2005;5(6):1375–1382.
    1. Chkhotua A.B., Gabusi E., Altimari A., et al. Increased expression of p16(INK4a) and p27(Kip1) cyclin-dependent kinase inhibitor genes in aging human kidney and chronic allograft nephropathy. Am J Kidney Dis. 2003;41(6):1303–1313.
    1. Koppelstaetter C., Schratzberger G., Perco P., et al. Markers of cellular senescence in zero hour biopsies predict outcome in renal transplantation. Aging Cell. 2008;7(4):491–497.
    1. McGlynn L.M., Stevenson K., Lamb K., et al. Cellular senescence in pretransplant renal biopsies predicts postoperative organ function. Aging Cell. 2009;8(1):45–51.
    1. Sofue T., Kushida Y., Ozaki T., et al. Tubular cell senescence in the donated kidney predicts allograft function, but not donor remnant kidney function, in living donor kidney transplantation. Am J Nephrol. 2018;47(1):8–17.
    1. Koppelstaetter C., Kern G., Leierer G., Mair S.M., Mayer G., Leierer J. Effect of cyclosporine, tacrolimus and sirolimus on cellular senescence in renal epithelial cells. Toxicol In Vitro. 2018;48:86–92.
    1. Sagiv A., Krizhanovsky V. Immunosurveillance of senescent cells: the bright side of the senescence program. Biogerontology. 2013;14(6):617–628.
    1. Hoffmann U., Neudörfl C., Daemen K., et al. NK cells of kidney transplant recipients display an activated phenotype that is influenced by immunosuppression and pathological staging. PLoS One. 2015;10(7)
    1. Jin J., Tao J., Gu X., et al. P16INK4a deletion ameliorated renal tubulointerstitial injury in a stress-induced premature senescence model of Bmi-1 deficiency. Sci Rep. 2017;7(1):7502.
    1. Braun H., Schmidt B.M., Raiss M., et al. Cellular senescence limits regenerative capacity and allograft survival. J Am Soc Nephrol. 2012;23(9):1467–1473.
    1. Docherty M.H., O'Sullivan E.D., Bonventre J.V., Ferenbach D.A. Cellular senescence in the kidney. J Am Soc Nephrol. 2019;30(5):726–736.
    1. Calder P.C. Marine omega-3 fatty acids and inflammatory processes: effects, mechanisms and clinical relevance. Biochim Biophys Acta. 2015;1851(4):469–484.
    1. Yamagata K., Suzuki S., Tagami M. Docosahexaenoic acid prevented tumor necrosis factor alpha-induced endothelial dysfunction and senescence. Prostaglandins Leukot Essent Fatty Acids. 2016;104:11–18.
    1. Sakai C., Ishida M., Ohba H., et al. Fish oil omega-3 polyunsaturated fatty acids attenuate oxidative stress-induced DNA damage in vascular endothelial cells. PLoS One. 2017;12(11)
    1. Farzaneh-Far R., Lin J., Epel E.S., Harris W.S., Blackburn E.H., Whooley M.A. Association of marine omega-3 fatty acid levels with telomeric aging in patients with coronary heart disease. JAMA. 2010;303(3):250–257.
    1. O'Callaghan N., Parletta N., Milte C.M., Benassi-Evans B., Fenech M., Howe P.R. Telomere shortening in elderly individuals with mild cognitive impairment may be attenuated with omega-3 fatty acid supplementation: a randomized controlled pilot study. Nutrition. 2014;30(4):489–491.
    1. Kiecolt-Glaser J.K., Epel E.S., Belury M.A., et al. Omega-3 fatty acids, oxidative stress, and leukocyte telomere length: a randomized controlled trial. Brain Behav Immun. 2013;28:16–24.
    1. Eide I.A., Åsberg A., Svensson M., et al. Plasma levels of marine n-3 fatty acids are inversely correlated with proinflammatory markers sTNFR1 and IL-6 in renal transplant recipients. J Ren Nutr. 2017;27(3):161–168.
    1. Eide I.A., Reinholt F.P., Jenssen T., et al. Effects of marine n-3 fatty acid supplementation in renal transplantation: a randomized controlled trial. Am J Transplant. 2019;19(3):790–800.
    1. Wang W.J., Cai G.Y., Chen X.M. Cellular senescence, senescence-associated secretory phenotype, and chronic kidney disease. Oncotarget. 2017;8(38):64520–64533.
    1. Valentijn F.A., Falke L.L., Nguyen T.Q., Goldschmeding R. Cellular senescence in the aging and diseased kidney. J Cell Commun Signal. 2018;12(1):69–82.
    1. Childs B.G., Gluscevic M., Baker D.J., et al. Senescent cells: an emerging target for diseases of ageing. Nat Rev Drug Discov. 2017;16(10):718–735.
    1. de Boer I.H., Gao X., Bebu I., et al. Biomarkers of tubulointerstitial damage and function in type 1 diabetes. BMJ Open Diabetes Res Care. 2017;5(1)
    1. Grainger D.J., Mosedale D.E., Metcalfe J.C. TGF-beta in blood: a complex problem. Cytokine Growth Factor Rev. 2000;11(1-2):133–145.
    1. Eide I.A., Jenssen T., Hartmann A., et al. The association between marine n-3 polyunsaturated fatty acid levels and survival after renal transplantation. Clin J Am Soc Nephrol. 2015;10(7):1246–1256.
    1. Taylor T.P., Janech M.G., Slate E.H., Lewis E.C., Arthur J.M., Oates J.C. Overcoming the effects of matrix interference in the measurement of urine protein analytes. Biomark Insights. 2012;7:1–8.
    1. Verzola D., Gandolfo M.T., Gaetani G., et al. Accelerated senescence in the kidneys of patients with type 2 diabetic nephropathy. Am J Physiol Ren Physiol. 2008;295(5):F1563–F1573.
    1. Westhoff J.H., Hilgers K.F., Steinbach M.P., et al. Hypertension induces somatic cellular senescence in rats and humans by induction of cell cycle inhibitor p16INK4a. Hypertension. 2008;52(1):123–129.
    1. Sis B., Tasanarong A., Khoshjou F., Dadras F., Solez K., Halloran P.F. Accelerated expression of senescence associated cell cycle inhibitor p16INK4A in kidneys with glomerular disease. Kidney Int. 2007;71(3):218–226.
    1. Salminen A., Kauppinen A., Kaarniranta K. Emerging role of NF-κB signaling in the induction of senescence-associated secretory phenotype (SASP) Cell Signal. 2012;24(4):835–845.
    1. Szanto A., Nagy L. The many faces of PPARgamma: anti-inflammatory by any means? Immunobiology. 2008;213(9-10):789–803.
    1. Serhan C.N., Chiang N., Dalli J. New pro-resolving n-3 mediators bridge resolution of infectious inflammation to tissue regeneration. Mol Aspects Med. 2018;64:1–17.
    1. Di Paolo N.C., Shayakhmetov D.M. Interleukin 1alpha and the inflammatory process. Nat Immunol. 2016;17(8):906–913.
    1. Nankivell B.J., Borrows R.J., Fung C.L., O'Connell P.J., Allen R.D., Chapman J.R. The natural history of chronic allograft nephropathy. N Engl J Med. 2003;349(24):2326–2333.
    1. Recio-Mayoral A., Banerjee D., Streather C., Kaski J.C. Endothelial dysfunction, inflammation and atherosclerosis in chronic kidney disease—a cross-sectional study of predialysis, dialysis and kidney-transplantation patients. Atherosclerosis. 2011;216(2):446–451.
    1. Segerer S., Schlondörff D. Role of chemokines for the localization of leukocyte subsets in the kidney. Semin Nephrol. 2007;27(3):260–274.
    1. Zakiyanov O., Kalousová M., Zima T., Tesař V. Matrix metalloproteinases in renal diseases: a critical appraisal. Kidney Blood Press Res. 2019;44(3):298–330.
    1. Spinale F.G. Matrix metalloproteinases: regulation and dysregulation in the failing heart. Circ Res. 2002;90(5):520–530.
    1. Li M.O., Wan Y.Y., Sanjabi S., Robertson A.K., Flavell R.A. Transforming growth factor-beta regulation of immune responses. Annu Rev Immunol. 2006;24:99–146.
    1. Goumenos D.S., Tsakas S., El Nahas A.M., et al. Transforming growth factor-beta(1) in the kidney and urine of patients with glomerular disease and proteinuria. Nephrol Dial Transplant. 2002;17(12):2145–2152.
    1. Rogier E., Durrbach A., Abecassis L., et al. A novel biological assay to detect the active form of TGF-beta in urine to monitor renal allograft rejection. Kidney Int. 2005;68(4):1875–1883.
    1. Stark K.D., Van Elswyk M.E., Higgins M.R., Weatherford C.A., Salem N., Jr. Global survey of the omega-3 fatty acids, docosahexaenoic acid and eicosapentaenoic acid in the blood stream of healthy adults. Prog Lipid Res. 2016;63:132–152.
    1. Idda M.L., McClusky W.G., Lodde V., et al. Survey of senescent cell markers with age in human tissues. Aging (Albany NY) 2020;12(5):4052–4066.
    1. Gorgoulis V., Adams P.D., Alimonti A., et al. Cellular senescence: defining a path forward. Cell. 2019;179(4):813–827.

Source: PubMed

3
Iratkozz fel