Diabetes Metab J.  2022 Mar;46(2):181-197. 10.4093/dmj.2021.0329.

Pathophysiologic Mechanisms and Potential Biomarkers in Diabetic Kidney Disease

Affiliations
  • 1Department of Internal Medicine and Institute of Kidney Disease Research, Yonsei University College of Medicine, Seoul, Korea

Abstract

Although diabetic kidney disease (DKD) remains the leading cause of end-stage kidney disease eventually requiring chronic kidney replacement therapy, the prevalence of DKD has failed to decline over the past 30 years. In order to reduce disease prevalence, extensive research has been ongoing to improve prediction of DKD onset and progression. Although the most commonly used markers of DKD are albuminuria and estimated glomerular filtration rate, their limitations have encouraged researchers to search for novel biomarkers that could improve risk stratification. Considering that DKD is a complex disease process that involves several pathophysiologic mechanisms such as hyperglycemia induced inflammation, oxidative stress, tubular damage, eventually leading to kidney damage and fibrosis, many novel biomarkers that capture one specific mechanism of the disease have been developed. Moreover, the increasing use of high-throughput omic approaches to analyze biological samples that include proteomics, metabolomics, and transcriptomics has emerged as a strong tool in biomarker discovery. This review will first describe recent advances in the understanding of the pathophysiology of DKD, and second, describe the current clinical biomarkers for DKD, as well as the current status of multiple potential novel biomarkers with respect to protein biomarkers, proteomics, metabolomics, and transcriptomics.

Keyword

Biomarkers; Diabetic nephropathies; Inflammation; Metabolomics; Oxidative stress; Pathophysiology; Proteomics; Transcriptome

Figure

  • Fig. 1. The pathophysiology of potential and novel biomarkers of diabetic kidney disease classified by their target pathophysiological pathways. AGE, advanced glycation end products; PKC, protein kinase C; IGF-1, insulin like growth factor-1; EGF, epidermal growth factor; PDGF, platelet-derived growth factor; VEGF, vascular endothelial growth factor; TGF-β, transforming growth factor-β; Ang II, angiotensin II; TNF-α, tissue necrosis factor-α; INF-γ, interferon-γ; IL-1, interleukin-1; IL-6, interleukin-6; Nlrp3, nod-like receptor protein-3; ROS, reactive oxygen species; CCL2, C-C motif chemokine 2; ICAM-1, intracellular adhesion molecule 1; TNFR1, tissue necrosis factor receptor 1; TNFR2, tissue necrosis factor receptor 2; PAI-1, plasminogen activator inhibitor-1; VCAM-1, vascular cell adhesion protein-1; CRP, C-reactive protein; Nrf2, nuclear factor erythroid 2-related factor; NOS, nitric oxide synthase; 8-OHdG, 8-Hydroxy-2’-deoxyguanosine.


Cited by  1 articles

Renoprotective Mechanism of Sodium-Glucose Cotransporter 2 Inhibitors: Focusing on Renal Hemodynamics
Nam Hoon Kim, Nan Hee Kim
Diabetes Metab J. 2022;46(4):543-551.    doi: 10.4093/dmj.2022.0209.


Reference

1. Alicic RZ, Rooney MT, Tuttle KR. Diabetic kidney disease: challenges, progress, and possibilities. Clin J Am Soc Nephrol. 2017; 12:2032–45.
2. Gregg EW, Li Y, Wang J, Burrows NR, Ali MK, Rolka D, et al. Changes in diabetes-related complications in the United States, 1990-2010. N Engl J Med. 2014; 370:1514–23.
Article
3. Thomas MC, Brownlee M, Susztak K, Sharma K, JandeleitDahm KA, Zoungas S, et al. Diabetic kidney disease. Nat Rev Dis Primers. 2015; 1:15018.
Article
4. Anders HJ, Huber TB, Isermann B, Schiffer M. CKD in diabetes: diabetic kidney disease versus nondiabetic kidney disease. Nat Rev Nephrol. 2018; 14:361–77.
Article
5. Kidney Disease: Improving Global Outcomes (KDIGO). KDIGO 2012 clinical practice guideline for the evaluation and management of chronic kidney disease. Kidney Int Suppl. 2013; 3:1–150.
6. Jones RH, Hayakawa H, Mackay JD, Parsons V, Watkins PJ. Progression of diabetic nephropathy. Lancet. 1979; 1:1105–6.
Article
7. Diabetes Control and Complications Trial Research Group, Nathan DM, Genuth S, Lachin J, Cleary P, Crofford O, et al. The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulindependent diabetes mellitus. N Engl J Med. 1993; 329:977–86.
Article
8. American Diabetes Association. 6. Glycemic targets: standards of medical care in diabetes-2018. Diabetes Care. 2018; 41(Suppl 1):S55–64.
9. ADVANCE Collaborative Group, Patel A, MacMahon S, Chalmers J, Neal B, Billot L, et al. Intensive blood glucose control and vascular outcomes in patients with type 2 diabetes. N Engl J Med. 2008; 358:2560–72.
Article
10. Ismail-Beigi F, Craven T, Banerji MA, Basile J, Calles J, Cohen RM, et al. Effect of intensive treatment of hyperglycaemia on microvascular outcomes in type 2 diabetes: an analysis of the ACCORD randomised trial. Lancet. 2010; 376:419–30.
Article
11. Tamborlane WV, Puklin JE, Bergman M, Verdonk C, Rudolf MC, Felig P, et al. Long-term improvement of metabolic control with the insulin pump does not reverse diabetic microangiopathy. Diabetes Care. 1982; 5 Suppl 1:58–64.
12. Ciavarella A, Vannini P, Flammini M, Bacci L, Forlani G, Borgnino LC. Effect of long-term near-normoglycemia on the progression of diabetic nephropathy. Diabete Metab. 1985; 11:3–8.
13. Caramori ML, Fioretto P, Mauer M. Low glomerular filtration rate in normoalbuminuric type 1 diabetic patients: an indicator of more advanced glomerular lesions. Diabetes. 2003; 52:1036–40.
14. Brownlee M, Vlassara H, Cerami A. Nonenzymatic glycosylation and the pathogenesis of diabetic complications. Ann Intern Med. 1984; 101:527–37.
Article
15. Lin YC, Chang YH, Yang SY, Wu KD, Chu TS. Update of pathophysiology and management of diabetic kidney disease. J Formos Med Assoc. 2018; 117:662–75.
Article
16. Sharma D, Bhattacharya P, Kalia K, Tiwari V. Diabetic nephropathy: new insights into established therapeutic paradigms and novel molecular targets. Diabetes Res Clin Pract. 2017; 128:91–108.
Article
17. Warren AM, Knudsen ST, Cooper ME. Diabetic nephropathy: an insight into molecular mechanisms and emerging therapies. Expert Opin Ther Targets. 2019; 23:579–91.
Article
18. Sharma K, Jin Y, Guo J, Ziyadeh FN. Neutralization of TGFbeta by anti-TGF-beta antibody attenuates kidney hypertrophy and the enhanced extracellular matrix gene expression in STZ-induced diabetic mice. Diabetes. 1996; 45:522–30.
Article
19. Ziyadeh FN, Hoffman BB, Han DC, Iglesias-De La Cruz MC, Hong SW, Isono M, et al. Long-term prevention of renal insufficiency, excess matrix gene expression, and glomerular mesangial matrix expansion by treatment with monoclonal antitransforming growth factor-beta antibody in db/db diabetic mice. Proc Natl Acad Sci U S A. 2000; 97:8015–20.
20. Sharma K, Ix JH, Mathew AV, Cho M, Pflueger A, Dunn SR, et al. Pirfenidone for diabetic nephropathy. J Am Soc Nephrol. 2011; 22:1144–51.
Article
21. Parving HH, Andersen AR, Smidt UM, Svendsen PA. Early aggressive antihypertensive treatment reduces rate of decline in kidney function in diabetic nephropathy. Lancet. 1983; 1:1175–9.
Article
22. Xie X, Atkins E, Lv J, Bennett A, Neal B, Ninomiya T, et al. Effects of intensive blood pressure lowering on cardiovascular and renal outcomes: updated systematic review and metaanalysis. Lancet. 2016; 387:435–43.
Article
23. Patney V, Whaley-Connell A, Bakris G. Hypertension management in diabetic kidney disease. Diabetes Spectr. 2015; 28:175–80.
Article
24. Gross JL, de Azevedo MJ, Silveiro SP, Canani LH, Caramori ML, Zelmanovitz T. Diabetic nephropathy: diagnosis, prevention, and treatment. Diabetes Care. 2005; 28:164–76.
Article
25. ACCORD Study Group, Cushman WC, Evans GW, Byington RP, Goff DC Jr, Grimm RH Jr, et al. Effects of intensive bloodpressure control in type 2 diabetes mellitus. N Engl J Med. 2010; 362:1575–85.
Article
26. Cooper-DeHoff RM, Gong Y, Handberg EM, Bavry AA, Denardo SJ, Bakris GL, et al. Tight blood pressure control and cardiovascular outcomes among hypertensive patients with diabetes and coronary artery disease. JAMA. 2010; 304:61–8.
Article
27. Whelton PK, Carey RM, Aronow WS, Casey DE Jr, Collins KJ, Dennison Himmelfarb C, et al. 2017 ACC/AHA/AAPA/ABC/ACPM/AGS/APhA/ASH/ASPC/NMA/PCNA guideline for the prevention, detection, evaluation, and management of high blood pressure in adults: executive summary: a report of the American College of Cardiology/American Heart Association task force on clinical practice guidelines. Hypertension. 2018; 71:1269–324.
28. Passarella P, Kiseleva TA, Valeeva FV, Gosmanov AR. Hypertension management in diabetes: 2018 update. Diabetes Spectr. 2018; 31:218–24.
Article
29. Armstrong C; Joint National Committee. JNC8 guidelines for the management of hypertension in adults. Am Fam Physician. 2014; 90:503–4.
30. SPRINT Research Group, Wright JT Jr, Williamson JD, Whelton PK, Snyder JK, Sink KM, et al. A randomized trial of intensive versus standard blood-pressure control. N Engl J Med. 2015; 373:2103–16.
Article
31. Buckley LF, Dixon DL, Wohlford GF 4th, Wijesinghe DS, Baker WL, Van Tassell BW. Intensive versus standard blood pressure control in SPRINT-Eligible participants of ACCORD-BP. Diabetes Care. 2017; 40:1733–8.
Article
32. Chow F, Ozols E, Nikolic-Paterson DJ, Atkins RC, Tesch GH. Macrophages in mouse type 2 diabetic nephropathy: correlation with diabetic state and progressive renal injury. Kidney Int. 2004; 65:116–28.
Article
33. Shahzad K, Bock F, Dong W, Wang H, Kopf S, Kohli S, et al. Nlrp3-inflammasome activation in non-myeloid-derived cells aggravates diabetic nephropathy. Kidney Int. 2015; 87:74–84.
Article
34. Barrera-Chimal J, Jaisser F. Pathophysiologic mechanisms in diabetic kidney disease: a focus on current and future therapeutic targets. Diabetes Obes Metab. 2020; 22 Suppl 1:16–31.
Article
35. Okada S, Shikata K, Matsuda M, Ogawa D, Usui H, Kido Y, et al. Intercellular adhesion molecule-1-deficient mice are resistant against renal injury after induction of diabetes. Diabetes. 2003; 52:2586–93.
Article
36. Giunti S, Barutta F, Perin PC, Gruden G. Targeting the MCP1/CCR2 system in diabetic kidney disease. Curr Vasc Pharmacol. 2010; 8:849–60.
Article
37. Boels MG, Koudijs A, Avramut MC, Sol WM, Wang G, van Oeveren-Rietdijk AM, et al. Systemic monocyte chemotactic protein-1 inhibition modifies renal macrophages and restores glomerular endothelial glycocalyx and barrier function in diabetic nephropathy. Am J Pathol. 2017; 187:2430–40.
Article
38. Menne J, Eulberg D, Beyer D, Baumann M, Saudek F, Valkusz Z, et al. C-C motif-ligand 2 inhibition with emapticap pegol (NOX-E36) in type 2 diabetic patients with albuminuria. Nephrol Dial Transplant. 2017; 32:307–15.
Article
39. Elmarakby AA, Sullivan JC. Relationship between oxidative stress and inflammatory cytokines in diabetic nephropathy. Cardiovasc Ther. 2012; 30:49–59.
Article
40. Moriwaki Y, Yamamoto T, Shibutani Y, Aoki E, Tsutsumi Z, Takahashi S, et al. Elevated levels of interleukin-18 and tumor necrosis factor-alpha in serum of patients with type 2 diabetes mellitus: relationship with diabetic nephropathy. Metabolism. 2003; 52:605–8.
Article
41. Donate-Correa J, Tagua VG, Ferri C, Martin-Nunez E, Hernandez-Carballo C, Urena-Torres P, et al. Pentoxifylline for renal protection in diabetic kidney disease: a model of old drugs for new horizons. J Clin Med. 2019; 8:287.
Article
42. Navarro JF, Mora C, Muros M, Garcia J. Additive antiproteinuric effect of pentoxifylline in patients with type 2 diabetes under angiotensin II receptor blockade: a short-term, randomized, controlled trial. J Am Soc Nephrol. 2005; 16:2119–26.
Article
43. Navarro-Gonzalez JF, Sanchez-Nino MD, Donate-Correa J, Martin-Nunez E, Ferri C, Perez-Delgado N, et al. Effects of pentoxifylline on soluble klotho concentrations and renal tubular cell expression in diabetic kidney disease. Diabetes Care. 2018; 41:1817–20.
Article
44. Shan D, Wu HM, Yuan QY, Li J, Zhou RL, Liu GJ. Pentoxifylline for diabetic kidney disease. Cochrane Database Syst Rev. 2012; 2:CD006800.
Article
45. Honda T, Hirakawa Y, Nangaku M. The role of oxidative stress and hypoxia in renal disease. Kidney Res Clin Pract. 2019; 38:414–26.
Article
46. Verma S, Singh P, Khurana S, Ganguly NK, Kukreti R, Saso L, et al. Implications of oxidative stress in chronic kidney disease: a review on current concepts and therapies. Kidney Res Clin Pract. 2021; 40:183–93.
Article
47. Sasaki S, Inoguchi T. The role of oxidative stress in the pathogenesis of diabetic vascular complications. Diabetes Metab J. 2012; 36:255–61.
Article
48. Goligorsky MS, Chen J, Brodsky S. Workshop: endothelial cell dysfunction leading to diabetic nephropathy: focus on nitric oxide. Hypertension. 2001; 37(2 Pt 2):744–8.
Article
49. Xia Y, Dawson VL, Dawson TM, Snyder SH, Zweier JL. Nitric oxide synthase generates superoxide and nitric oxide in arginine-depleted cells leading to peroxynitrite-mediated cellular injury. Proc Natl Acad Sci U S A. 1996; 93:6770–4.
Article
50. Brodsky SV, Gao S, Li H, Goligorsky MS. Hyperglycemic switch from mitochondrial nitric oxide to superoxide production in endothelial cells. Am J Physiol Heart Circ Physiol. 2002; 283:H2130–9.
51. Jha JC, Gray SP, Barit D, Okabe J, El-Osta A, Namikoshi T, et al. Genetic targeting or pharmacologic inhibition of NADPH oxidase nox4 provides renoprotection in long-term diabetic nephropathy. J Am Soc Nephrol. 2014; 25:1237–54.
Article
52. Gorin Y, Cavaglieri RC, Khazim K, Lee DY, Bruno F, Thakur S, et al. Targeting NADPH oxidase with a novel dual Nox1/Nox4 inhibitor attenuates renal pathology in type 1 diabetes. Am J Physiol Renal Physiol. 2015; 308:F1276–87.
Article
53. Kwon G, Uddin MJ, Lee G, Jiang S, Cho A, Lee JH, et al. A novel pan-Nox inhibitor, APX-115, protects kidney injury in streptozotocin-induced diabetic mice: possible role of peroxisomal and mitochondrial biogenesis. Oncotarget. 2017; 8:74217–32.
Article
54. Sakashita M, Tanaka T, Inagi R. Metabolic changes and oxidative stress in diabetic kidney disease. Antioxidants (Basel). 2021; 10:1143.
Article
55. Satirapoj B, Adler SG. Comprehensive approach to diabetic nephropathy. Kidney Res Clin Pract. 2014; 33:121–31.
Article
56. Marumo T, Yagi S, Kawarazaki W, Nishimoto M, Ayuzawa N, Watanabe A, et al. Diabetes induces aberrant DNA methylation in the proximal tubules of the kidney. J Am Soc Nephrol. 2015; 26:2388–97.
Article
57. DeFronzo RA, Reeves WB, Awad AS. Pathophysiology of diabetic kidney disease: impact of SGLT2 inhibitors. Nat Rev Nephrol. 2021; 17:319–34.
Article
58. Salim HM, Fukuda D, Yagi S, Soeki T, Shimabukuro M, Sata M. Glycemic control with ipragliflozin, a novel selective SGLT2 inhibitor, ameliorated endothelial dysfunction in streptozotocin-induced diabetic mouse. Front Cardiovasc Med. 2016; 3:43.
Article
59. Shigiyama F, Kumashiro N, Miyagi M, Ikehara K, Kanda E, Uchino H, et al. Effectiveness of dapagliflozin on vascular endothelial function and glycemic control in patients with earlystage type 2 diabetes mellitus: DEFENCE study. Cardiovasc Diabetol. 2017; 16:84.
Article
60. Tanaka A, Shimabukuro M, Machii N, Teragawa H, Okada Y, Shima KR, et al. Effect of empagliflozin on endothelial function in patients with type 2 diabetes and cardiovascular disease: results from the multicenter, randomized, placebo-controlled, double-blind EMBLEM trial. Diabetes Care. 2019; 42:e159–61.
61. Zelniker TA, Wiviott SD, Raz I, Im K, Goodrich EL, Bonaca MP, et al. SGLT2 inhibitors for primary and secondary prevention of cardiovascular and renal outcomes in type 2 diabetes: a systematic review and meta-analysis of cardiovascular outcome trials. Lancet. 2019; 393:31–9.
Article
62. Perkovic V, Jardine MJ, Neal B, Bompoint S, Heerspink HJ, Charytan DM, et al. Canagliflozin and renal outcomes in type 2 diabetes and nephropathy. N Engl J Med. 2019; 380:2295–306.
Article
63. Oh TJ, Moon JY, Hur KY, Ko SH, Kim HJ, Kim T, et al. Sodium-glucose cotransporter-2 inhibitor for renal function preservation in patients with type 2 diabetes mellitus: a Korean Diabetes Association and Korean Society of Nephrology consensus statement. Kidney Res Clin Pract. 2020; 39:269–83.
Article
64. Nangaku M, Kanda H, Takama H, Ichikawa T, Hase H, Akizawa T. Randomized clinical trial on the effect of bardoxolone methyl on GFR in diabetic kidney disease patients (TSUBAKI Study). Kidney Int Rep. 2020; 5:879–90.
Article
65. de Zeeuw D, Akizawa T, Audhya P, Bakris GL, Chin M, ChristSchmidt H, et al. Bardoxolone methyl in type 2 diabetes and stage 4 chronic kidney disease. N Engl J Med. 2013; 369:2492–503.
Article
66. Yamazaki T, Mimura I, Tanaka T, Nangaku M. Treatment of diabetic kidney disease: current and future. Diabetes Metab J. 2021; 45:11–26.
Article
67. Stevens PE, Levin A; Kidney Disease: Improving Global Outcomes Chronic Kidney Disease Guideline Development Work Group Members. Evaluation and management of chronic kidney disease: synopsis of the kidney disease: improving global outcomes 2012 clinical practice guideline. Ann Intern Med. 2013; 158:825–30.
Article
68. An JH, Cho YM, Yu HG, Jang HC, Park KS, Kim SY, et al. The clinical characteristics of normoalbuminuric renal insufficiency in Korean type 2 diabetic patients: a possible early stage renal complication. J Korean Med Sci. 2009; 24(Suppl 1):S75–81.
Article
69. Thomas G, Sehgal AR, Kashyap SR, Srinivas TR, Kirwan JP, Navaneethan SD. Metabolic syndrome and kidney disease: a systematic review and meta-analysis. Clin J Am Soc Nephrol. 2011; 6:2364–73.
Article
70. Vistisen D, Andersen GS, Hulman A, Persson F, Rossing P, Jorgensen ME. Progressive decline in estimated glomerular filtration rate in patients with diabetes after moderate loss in kidney function-even without albuminuria. Diabetes Care. 2019; 42:1886–94.
Article
71. MacIsaac RJ, Ekinci EI, Jerums G. ‘Progressive diabetic nephropathy: how useful is microalbuminuria?: contra’. Kidney Int. 2014; 86:50–7.
Article
72. Moazzeni SS, Arani RH, Hasheminia M, Tohidi M, Azizi F, Hadaegh F. High incidence of chronic kidney disease among iranian diabetic adults: using CKD-EPI and MDRD equations for estimated glomerular filtration rate. Diabetes Metab J. 2021; 45:684–97.
Article
73. Dedual MA, Wueest S, Challa TD, Lucchini FC, Aeppli TR, Borsigova M, et al. Obesity-induced increase in cystatin c alleviates tissue inflammation. Diabetes. 2020; 69:1927–35.
Article
74. Colhoun HM, Marcovecchio ML. Biomarkers of diabetic kidney disease. Diabetologia. 2018; 61:996–1011.
Article
75. Pena MJ, de Zeeuw D, Mischak H, Jankowski J, Oberbauer R, Woloszczuk W, et al. Prognostic clinical and molecular biomarkers of renal disease in type 2 diabetes. Nephrol Dial Transplant. 2015; 30 Suppl 4:iv86–95.
Article
76. Han WK, Bailly V, Abichandani R, Thadhani R, Bonventre JV. Kidney Injury Molecule-1 (KIM-1): a novel biomarker for human renal proximal tubule injury. Kidney Int. 2002; 62:237–44.
Article
77. Coca SG, Nadkarni GN, Huang Y, Moledina DG, Rao V, Zhang J, et al. Plasma biomarkers and kidney function decline in early and established diabetic kidney disease. J Am Soc Nephrol. 2017; 28:2786–93.
Article
78. Schrauben SJ, Shou H, Zhang X, Anderson AH, Bonventre JV, Chen J, et al. Association of multiple plasma biomarker concentrations with progression of prevalent diabetic kidney disease: findings from the Chronic Renal Insufficiency Cohort (CRIC) Study. J Am Soc Nephrol. 2021; 32:115–26.
Article
79. de Carvalho JA, Tatsch E, Hausen BS, Bollick YS, Moretto MB, Duarte T, et al. Urinary kidney injury molecule-1 and neutrophil gelatinase-associated lipocalin as indicators of tubular damage in normoalbuminuric patients with type 2 diabetes. Clin Biochem. 2016; 49:232–6.
Article
80. Yuruk Yildirim Z, Nayir A, Yilmaz A, Gedikbasi A, Bundak R. Neutrophil gelatinase-associated lipocalin as an early sign of diabetic kidney injury in children. J Clin Res Pediatr Endocrinol. 2015; 7:274–9.
Article
81. Lacquaniti A, Donato V, Pintaudi B, Di Vieste G, Chirico V, Buemi A, et al. “Normoalbuminuric” diabetic nephropathy: tubular damage and NGAL. Acta Diabetol. 2013; 50:935–42.
Article
82. Nielsen SE, Sugaya T, Hovind P, Baba T, Parving HH, Rossing P. Urinary liver-type fatty acid-binding protein predicts progression to nephropathy in type 1 diabetic patients. Diabetes Care. 2010; 33:1320–4.
Article
83. Kamijo-Ikemori A, Sugaya T, Yasuda T, Kawata T, Ota A, Tatsunami S, et al. Clinical significance of urinary liver-type fatty acid-binding protein in diabetic nephropathy of type 2 diabetic patients. Diabetes Care. 2011; 34:691–6.
Article
84. Panduru NM, Forsblom C, Saraheimo M, Thorn L, Bierhaus A, Humpert PM, et al. Urinary liver-type fatty acid-binding protein and progression of diabetic nephropathy in type 1 diabetes. Diabetes Care. 2013; 36:2077–83.
Article
85. Kim SS, Song SH, Kim IJ, Jeon YK, Kim BH, Kwak IS, et al. Urinary cystatin C and tubular proteinuria predict progression of diabetic nephropathy. Diabetes Care. 2013; 36:656–61.
Article
86. Hasegawa G, Nakano K, Sawada M, Uno K, Shibayama Y, Ienaga K, et al. Possible role of tumor necrosis factor and interleukin-1 in the development of diabetic nephropathy. Kidney Int. 1991; 40:1007–12.
Article
87. Bertani T, Abbate M, Zoja C, Corna D, Perico N, Ghezzi P, et al. Tumor necrosis factor induces glomerular damage in the rabbit. Am J Pathol. 1989; 134:419–30.
88. Kalantarinia K, Awad AS, Siragy HM. Urinary and renal interstitial concentrations of TNF-alpha increase prior to the rise in albuminuria in diabetic rats. Kidney Int. 2003; 64:1208–13.
89. Niewczas MA, Gohda T, Skupien J, Smiles AM, Walker WH, Rosetti F, et al. Circulating TNF receptors 1 and 2 predict ESRD in type 2 diabetes. J Am Soc Nephrol. 2012; 23:507–15.
Article
90. Skupien J, Warram JH, Niewczas MA, Gohda T, Malecki M, Mychaleckyj JC, et al. Synergism between circulating tumor necrosis factor receptor 2 and HbA(1c) in determining renal decline during 5-18 years of follow-up in patients with type 1 diabetes and proteinuria. Diabetes Care. 2014; 37:2601–8.
Article
91. Lopes-Virella MF, Baker NL, Hunt KJ, Cleary PA, Klein R, Virella G, et al. Baseline markers of inflammation are associated with progression to macroalbuminuria in type 1 diabetic subjects. Diabetes Care. 2013; 36:2317–23.
Article
92. Wu LL, Chiou CC, Chang PY, Wu JT. Urinary 8-OHdG: a marker of oxidative stress to DNA and a risk factor for cancer, atherosclerosis and diabetics. Clin Chim Acta. 2004; 339:1–9.
Article
93. Xu GW, Yao QH, Weng QF, Su BL, Zhang X, Xiong JH. Study of urinary 8-hydroxydeoxyguanosine as a biomarker of oxidative DNA damage in diabetic nephropathy patients. J Pharm Biomed Anal. 2004; 36:101–4.
Article
94. Sanchez M, Roussel R, Hadjadj S, Moutairou A, Marre M, Velho G, et al. Plasma concentrations of 8-hydroxy-2’-deoxyguanosine and risk of kidney disease and death in individuals with type 1 diabetes. Diabetologia. 2018; 61:977–84.
Article
95. Serdar M, Sertoglu E, Uyanik M, Tapan S, Akin K, Bilgi C, et al. Comparison of 8-hydroxy-2’-deoxyguanosine (8-OHdG) levels using mass spectrometer and urine albumin creatinine ratio as a predictor of development of diabetic nephropathy. Free Radic Res. 2012; 46:1291–5.
Article
96. Dubin RF, Rhee EP. Proteomics and metabolomics in kidney disease, including insights into etiology, treatment, and prevention. Clin J Am Soc Nephrol. 2020; 15:404–11.
Article
97. Bhensdadia NM, Hunt KJ, Lopes-Virella MF, Michael Tucker J, Mataria MR, Alge JL, et al. Urine haptoglobin levels predict early renal functional decline in patients with type 2 diabetes. Kidney Int. 2013; 83:1136–43.
Article
98. Good DM, Zurbig P, Argiles A, Bauer HW, Behrens G, Coon JJ, et al. Naturally occurring human urinary peptides for use in diagnosis of chronic kidney disease. Mol Cell Proteomics. 2010; 9:2424–37.
Article
99. Zurbig P, Jerums G, Hovind P, Macisaac RJ, Mischak H, Nielsen SE, et al. Urinary proteomics for early diagnosis in diabetic nephropathy. Diabetes. 2012; 61:3304–13.
Article
100. Roscioni SS, de Zeeuw D, Hellemons ME, Mischak H, Zurbig P, Bakker SJ, et al. A urinary peptide biomarker set predicts worsening of albuminuria in type 2 diabetes mellitus. Diabetologia. 2013; 56:259–67.
Article
101. Zurbig P, Mischak H, Menne J, Haller H. CKD273 enables efficient prediction of diabetic nephropathy in nonalbuminuric patients. Diabetes Care. 2019; 42:e4–5.
102. Tofte N, Lindhardt M, Adamova K, Bakker SJ, Beige J, Beulens JW, et al. Early detection of diabetic kidney disease by urinary proteomics and subsequent intervention with spironolactone to delay progression (PRIORITY): a prospective observational study and embedded randomised placebo-controlled trial. Lancet Diabetes Endocrinol. 2020; 8:301–12.
103. Lindhardt M, Persson F, Zurbig P, Stalmach A, Mischak H, de Zeeuw D, et al. Urinary proteomics predict onset of microalbuminuria in normoalbuminuric type 2 diabetic patients, a sub-study of the DIRECT-Protect 2 study. Nephrol Dial Transplant. 2017; 32:1866–73.
Article
104. Kolch W, Neusüss C, Pelzing M, Mischak H. Capillary electrophoresis-mass spectrometry as a powerful tool in clinical diagnosis and biomarker discovery. Mass Spectrom Rev. 2005; 24:959–77.
Article
105. Hocher B, Adamski J. Metabolomics for clinical use and research in chronic kidney disease. Nat Rev Nephrol. 2017; 13:269–84.
Article
106. Han LD, Xia JF, Liang QL, Wang Y, Wang YM, Hu P, et 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 Acta. 2011; 689:85–91.
Article
107. Hirayama A, Nakashima E, Sugimoto M, Akiyama S, Sato W, Maruyama S, et al. Metabolic profiling reveals new serum biomarkers for differentiating diabetic nephropathy. Anal Bioanal Chem. 2012; 404:3101–9.
Article
108. Zhang J, Yan L, Chen W, Lin L, Song X, Yan X, et al. Metabonomics research of diabetic nephropathy and type 2 diabetes mellitus based on UPLC-oaTOF-MS system. Anal Chim Acta. 2009; 650:16–22.
Article
109. Pena MJ, Lambers Heerspink HJ, Hellemons ME, Friedrich T, Dallmann G, Lajer M, et al. Urine and plasma metabolites predict the development of diabetic nephropathy in individuals with type 2 diabetes mellitus. Diabet Med. 2014; 31:1138–47.
110. Looker HC, Colombo M, Hess S, Brosnan MJ, Farran B, Dalton RN, et al. Biomarkers of rapid chronic kidney disease progression in type 2 diabetes. Kidney Int. 2015; 88:888–96.
Article
111. Kwan B, Fuhrer T, Zhang J, Darshi M, Van Espen B, Montemayor D, et al. Metabolomic markers of kidney function decline in patients with diabetes: evidence from the Chronic Renal Insufficiency Cohort (CRIC) Study. Am J Kidney Dis. 2020; 76:511–20.
Article
112. Mutter S, Valo E, Aittomaki V, Nybo K, Raivonen L, Thorn LM, et al. Urinary metabolite profiling and risk of progression of diabetic nephropathy in 2670 individuals with type 1 diabetes. Diabetologia. 2022; 65:140–9.
Article
113. Pezzolesi MG, Satake E, McDonnell KP, Major M, Smiles AM, Krolewski AS. Circulating TGF-β1-regulated miRNAs and the risk of rapid progression to ESRD in type 1 diabetes. Diabetes. 2015; 64:3285–93.
Article
114. Argyropoulos C, Wang K, Bernardo J, Ellis D, Orchard T, Galas D, et al. Urinary microRNA profiling predicts the development of microalbuminuria in patients with type 1 diabetes. J Clin Med. 2015; 4:1498–517.
Article
115. Barutta F, Tricarico M, Corbelli A, Annaratone L, Pinach S, Grimaldi S, et al. Urinary exosomal microRNAs in incipient diabetic nephropathy. PLoS One. 2013; 8:e73798.
Article
116. Kato M. Noncoding RNAs as therapeutic targets in early stage diabetic kidney disease. Kidney Res Clin Pract. 2018; 37:197–209.
Article
Full Text Links
  • DMJ
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr