Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Nephron overload as a therapeutic target to maximize kidney lifespan

Abstract

Kidney lifespan is a patient-oriented outcome that provides much needed context for understanding chronic kidney disease (CKD). Nephron endowment, age-associated decline in nephron number, kidney injury history and the intrinsic capacity of nephrons to adapt to haemodynamic and metabolic overload vary widely within the population. Defining percentiles of kidney function might therefore help to predict individual kidney lifespan and distinguish healthy ageing from progressive forms of CKD. In response to nephron loss, the remaining nephrons undergo functional and structural adaptations to meet the ongoing haemodynamic and metabolic demands of the organism. When these changes are no longer sufficient to maintain kidney cell homeostasis, remnant nephron demise occurs and CKD progression ensues. An individual’s trajectory of glomerular filtration rate and albuminuria reflects the extent of nephron loss and adaptation of the remaining nephrons. Nephron overload represents the final common pathway of CKD progression and is largely independent of upstream disease mechanisms. Thus, interventions that efficiently attenuate nephron overload in early disease stages can protect remnant kidney cells and nephrons, and delay CKD progression. This Review provides a conceptual framework for individualized diagnosis, monitoring and treatment of CKD with the goal of maximizing kidney lifespan.

Key points

  • The current chronic kidney disease (CKD) classification is useful from the perspective of epidemiology, public health care and advocacy. Kidney lifespan is a more individualized, patient-oriented outcome that takes into account linear and non-linear declines in estimated glomerular filtration rate (eGFR) for the prediction of individual prognoses.

  • Population percentiles of eGFR acknowledge its age-specific spectrum and, along with individualized eGFR slopes, could help distinguish between healthy kidney ageing and progressive CKD. Percentiles are population-specific and should help identify patients at risk of CKD, as well as improve patient prognosis and management.

  • Adaptation to haemodynamic and metabolic overload is observed in the remaining nephrons in CKD but not in physiological kidney ageing. Adaptation is first evidenced by nephron hypertrophy and later by albuminuria.

  • Haemodynamic stress promotes podocyte loss directly and metabolic stress is a key driver of loss of tubular epithelial cells. Both types of stress can lead to secondary focal segmental glomerulosclerosis, tubular atrophy and interstitial fibrosis.

  • Progressive nephron loss reduces kidney lifespan. Dual blockade of the renin–angiotensin–aldosterone system and of sodium–glucose co-transporter 2 is very potent in both alleviating mechanisms of stress and prolonging kidney lifespan, and hence CKD has become a treatable disease.

  • Prolonging kidney lifespan with novel combination therapies is effective in patients with glomerular forms of CKD. Broad implementation of this approach requires effort at all levels, including improving our ability to assess and predict individual kidney lifespan, implementing remnant nephron overload as a pathophysiological concept and a treatment target, and raising awareness that CKD is a treatable disease.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Gaussian distribution of nephron number and GFR percentiles.
Fig. 2: Adaptations to nephron overload and CKD progression.
Fig. 3: Structural adaptations of nephrons.
Fig. 4: Modulation of haemodynamic and metabolic overload, and its effect on kidney lifespan.
Fig. 5: Pathophysiological mechanisms and treatment targets in glomerular versus tubular forms of CKD.

Similar content being viewed by others

References

  1. Kidney Disease: Improving Global Outcomes CKD Work Group. KDIGO 2012 clinical practice guideline for the evaluation and management of chronic kidney disease. Kidney Int. Suppl. 3, 1–150 (2013).

    Google Scholar 

  2. Benghanem Gharbi, M. et al. Chronic kidney disease, hypertension, diabetes, and obesity in the adult population of Morocco: how to avoid “over”- and “under”-diagnosis of CKD. Kidney Int. 89, 1363–1371 (2016).

    PubMed  Google Scholar 

  3. Jonsson, A. J., Lund, S. H., Eriksen, B. O., Palsson, R. & Indridason, O. S. The prevalence of chronic kidney disease in Iceland according to KDIGO criteria and age-adapted estimated glomerular filtration rate thresholds. Kidney Int. 98, 1286–1295 (2020).

    CAS  PubMed  Google Scholar 

  4. Hill, N. R. et al. Global prevalence of chronic kidney disease–a systematic review and meta-analysis. PLoS ONE 11, e0158765 (2016).

    PubMed  PubMed Central  Google Scholar 

  5. Matsushita, K. et al. Estimated glomerular filtration rate and albuminuria for prediction of cardiovascular outcomes: a collaborative meta-analysis of individual participant data. Lancet Diabetes Endocrinol. 3, 514–525 (2015).

    PubMed  PubMed Central  Google Scholar 

  6. Figurek, A., Luyckx, V. A. & Mueller, T. F. A systematic review of renal functional reserve in adult living kidney donors. Kidney Int. Rep. 5, 448–458 (2020).

    PubMed  PubMed Central  Google Scholar 

  7. Zelmer, J. L. The economic burden of end-stage renal disease in Canada. Kidney Int. 72, 1122–1129 (2007).

    CAS  PubMed  Google Scholar 

  8. Levey, A. S., Stevens, L. A. & Hostetter, T. Automatic reporting of estimated glomerular filtration rate–just what the doctor ordered. Clin. Chem. 52, 2188–2193 (2006).

    CAS  PubMed  Google Scholar 

  9. Delanaye, P., Cavalier, E. & Pottel, H. Serum creatinine: not so simple! Nephron 136, 302–308 (2017).

    CAS  PubMed  Google Scholar 

  10. Porrini, E. et al. Estimated GFR: time for a critical appraisal. Nat. Rev. Nephrol. 15, 177–190 (2019).

    CAS  PubMed  Google Scholar 

  11. Bello, A. K. et al. Quality of chronic kidney disease management in Canadian primary care. JAMA Netw. Open 2, e1910704 (2019).

    PubMed  PubMed Central  Google Scholar 

  12. Hallan, S. I. et al. Age and association of kidney measures with mortality and end-stage renal disease. JAMA 308, 2349–2360 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Delanaye, P. et al. CKD: a call for an age-adapted definition. J. Am. Soc. Nephrol. 30, 1785–1805 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Denic, A., Glassock, R. J. & Rule, A. D. Structural and functional changes with the aging kidney. Adv. Chronic Kidney Dis. 23, 19–28 (2016).

    PubMed  PubMed Central  Google Scholar 

  15. Welch, H. G., Schwartz, L. M. & Woloshin, S. Overdiagnosed: Making People Sick in the Pursuit of Health (Beacon Press, 2011).

  16. Liu, P. et al. Accounting for age in the definition of chronic kidney disease. JAMA Intern. Med. 181, 1359–1366 (2021).

    PubMed  Google Scholar 

  17. Daugirdas, J. T., Meyer, K., Greene, T., Butler, R. S. & Poggio, E. D. Scaling of measured glomerular filtration rate in kidney donor candidates by anthropometric estimates of body surface area, body water, metabolic rate, or liver size. Clin. J. Am. Soc. Nephrol. 4, 1575–1583 (2009).

    PubMed  PubMed Central  Google Scholar 

  18. Denic, A. et al. Single-nephron glomerular filtration rate in healthy adults. N. Engl. J. Med. 376, 2349–2357 (2017).

    PubMed  PubMed Central  Google Scholar 

  19. Khan, S., Loi, V. & Rosner, M. H. Drug-induced kidney injury in the elderly. Drugs Aging 34, 729–741 (2017).

    CAS  PubMed  Google Scholar 

  20. Eriksen, B. O. et al. Blood pressure and age-related GFR decline in the general population. BMC Nephrol. 18, 77 (2017).

    PubMed  PubMed Central  Google Scholar 

  21. Palsson, R. & Waikar, S. S. Renal functional reserve revisited. Adv. Chronic Kidney Dis. 25, e1–e8 (2018).

    PubMed  Google Scholar 

  22. Hughson, M., Farris, A. B. III, Douglas-Denton, R., Hoy, W. E. & Bertram, J. F. Glomerular number and size in autopsy kidneys: the relationship to birth weight. Kidney Int. 63, 2113–2122 (2003).

    PubMed  Google Scholar 

  23. Luyckx, V. A. et al. Effect of fetal and child health on kidney development and long-term risk of hypertension and kidney disease. Lancet 382, 273–283 (2013).

    PubMed  Google Scholar 

  24. Harer, M. W., Charlton, J. R., Tipple, T. E. & Reidy, K. J. Preterm birth and neonatal acute kidney injury: implications on adolescent and adult outcomes. J. Perinatol. 40, 1286–1295 (2020).

    PubMed  Google Scholar 

  25. Crump, C., Sundquist, J., Winkleby, M. A. & Sundquist, K. Preterm birth and risk of chronic kidney disease from childhood into mid-adulthood: national cohort study. BMJ 365, l1346 (2019).

    PubMed  PubMed Central  Google Scholar 

  26. Ruggajo, P. et al. Familial factors, low birth weight, and development of ESRD: a nationwide registry study. Am. J. Kidney Dis. 67, 601–608 (2016).

    PubMed  Google Scholar 

  27. Low Birth Weight and Nephron Number Working Group The impact of kidney development on the life course: a consensus document for action. Nephron 136, 3–49 (2017).

    Google Scholar 

  28. Abitbol, C. L. & Ingelfinger, J. R. Nephron mass and cardiovascular and renal disease risks. Semin. Nephrol. 29, 445–454 (2009).

    PubMed  Google Scholar 

  29. Wiles, K. et al. The impact of chronic kidney disease stages 3-5 on pregnancy outcomes. Nephrol. Dialysis Transpl. https://doi.org/10.1093/ndt/gfaa247 (2020).

    Article  Google Scholar 

  30. Coca, S. G., Singanamala, S. & Parikh, C. R. Chronic kidney disease after acute kidney injury: a systematic review and meta-analysis. Kidney Int. 81, 442–448 (2012).

    PubMed  Google Scholar 

  31. Kellum, J. A. et al. Acute kidney injury. Nat. Rev. Dis. Prim. 7, 52 (2021).

    Google Scholar 

  32. Newsome, B. B. et al. Long-term risk of mortality and end-stage renal disease among the elderly after small increases in serum creatinine level during hospitalization for acute myocardial infarction. Arch. Intern. Med. 168, 609–616 (2008).

    CAS  PubMed  Google Scholar 

  33. Murphy, D. et al. Trends in prevalence of chronic kidney disease in the United States. Ann. Intern. Med. 165, 473–481 (2016).

    PubMed  PubMed Central  Google Scholar 

  34. Gregg, E. W. et al. Changes in diabetes-related complications in the United States, 1990-2010. N. Engl. J. Med. 370, 1514–1523 (2014).

    CAS  PubMed  Google Scholar 

  35. Wakasugi, M., Kazama, J. J. & Narita, I. Secular trends in end-stage kidney disease requiring renal replacement therapy in Japan: Japanese Society of Dialysis Therapy Registry data from 1983 to 2016. Nephrology 25, 172–178 (2020).

    PubMed  Google Scholar 

  36. Romagnani, P. et al. Chronic kidney disease. Nat. Rev. Dis. Prim. 3, 17088 (2017).

    PubMed  Google Scholar 

  37. Brenner, B. M., Garcia, D. L. & Anderson, S. Glomeruli and blood pressure. Less of one, more the other? Am. J. Hypertens. 1, 335–347 (1988).

    CAS  PubMed  Google Scholar 

  38. Freedman, B. I., Limou, S., Ma, L. & Kopp, J. B. APOL1-associated nephropathy: a key contributor to racial disparities in CKD. Am. J. Kidney Dis. 72, S8–S16 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Luyckx, V. A. et al. Sustainable development goals relevant to kidney health: an update on progress. Nat. Rev. Nephrol. 17, 15–32 (2021).

    PubMed  Google Scholar 

  40. Hostetter, T. H., Olson, J. L., Rennke, H. G., Venkatachalam, M. A. & Brenner, B. M. Hyperfiltration in remnant nephrons: a potentially adverse response to renal ablation. J. Am. Soc. Nephrol. 12, 1315–1325 (2001).

    Google Scholar 

  41. Giral, M. et al. Kidney and recipient weight incompatibility reduces long-term graft survival. J. Am. Soc. Nephrol. 21, 1022–1029 (2010).

    PubMed  PubMed Central  Google Scholar 

  42. Al-Sehli, R. et al. What should the serum creatinine be after transplantation? An approach to integrate donor and recipient information to assess posttransplant kidney function. Transplant 99, 1960–1967 (2015).

    CAS  Google Scholar 

  43. Diao, J. A. et al. In search of a better equation–performance and equity in estimates of kidney function. N. Engl. J. Med. 384, 396–399 (2021).

    PubMed  PubMed Central  Google Scholar 

  44. Inker, L. A. et al. New creatinine- and cystatin C-based equations to estimate GFR without race. N. Engl. J. Med. 385, 1737–1749 (2021).

    CAS  PubMed  Google Scholar 

  45. Peralta, C. A. et al. Racial and ethnic differences in kidney function decline among persons without chronic kidney disease. J. Am. Soc. Nephrol. 22, 1327–1334 (2011).

    PubMed  PubMed Central  Google Scholar 

  46. Glassock, R., Denic, A. & Rule, A. D. In Brenner and Rector’s The Kidney 11th edn, Ch. 22 (ed. Yu, A. et al.) 710–730 (Elsevier, 2019).

  47. Baba, M. et al. Longitudinal study of the decline in renal function in healthy subjects. PLoS ONE 10, e0129036 (2015).

    PubMed  PubMed Central  Google Scholar 

  48. Carlström, M., Wilcox, C. S. & Arendshorst, W. J. Renal autoregulation in health and disease. Physiol. Rev. 95, 405–511 (2015).

    PubMed  PubMed Central  Google Scholar 

  49. Anders, H. J., Davis, J. M. & Thurau, K. Nephron protection in diabetic kidney disease. N. Engl. J. Med. 375, 2096–2098 (2016).

    PubMed  Google Scholar 

  50. Vallon, V. The mechanisms and therapeutic potential of SGLT2 inhibitors in diabetes mellitus. Annu. Rev. Med. 66, 255–270 (2015).

    CAS  PubMed  Google Scholar 

  51. Firsov, D. & Bonny, O. Circadian rhythms and the kidney. Nat. Rev. Nephrol. 14, 626–635 (2018).

    CAS  PubMed  Google Scholar 

  52. Bosch, J. P. et al. Renal functional reserve in humans. Effect of protein intake on glomerular filtration rate. Am. J. Med. 75, 943–950 (1983).

    CAS  PubMed  Google Scholar 

  53. Kriz, W. & Lemley, K. V. A potential role for mechanical forces in the detachment of podocytes and the progression of CKD. J. Am. Soc. Nephrol. 26, 258–269 (2015).

    PubMed  Google Scholar 

  54. Ichikawa, I., Hoyer, J. R., Seiler, M. W. & Brenner, B. M. Mechanism of glomerulotubular balance in the setting of heterogeneous glomerular injury. Preservation of a close functional linkage between individual nephrons and surrounding microvasculature. J. Clin. Invest. 69, 185–198 (1982).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Rule, A. D. et al. The association between age and nephrosclerosis on renal biopsy among healthy adults. Ann. Intern. Med. 152, 561–567 (2010).

    PubMed  PubMed Central  Google Scholar 

  56. Pontzer, H. et al. Daily energy expenditure through the human life course. Science 373, 808–812 (2021).

    CAS  PubMed  Google Scholar 

  57. Locke, J. E. et al. Obesity increases the risk of end-stage renal disease among living kidney donors. Kidney Int. 91, 699–703 (2017).

    PubMed  Google Scholar 

  58. Mueller, T. F. & Luyckx, V. A. The natural history of residual renal function in transplant donors. J. Am. Soc. Nephrol. 23, 1462–1466 (2012).

    PubMed  Google Scholar 

  59. Lenihan, C. R. et al. Longitudinal study of living kidney donor glomerular dynamics after nephrectomy. J. Clin. Invest. 125, 1311–1318 (2015).

    PubMed  PubMed Central  Google Scholar 

  60. Strieder, T. et al. Effects of perfusion pressures on podocyte loss in the isolated perfused mouse kidney. Cell. Physiol. Biochem. 55, 1–12 (2021).

    PubMed  Google Scholar 

  61. Hughson, M. D., Hoy, W. E., Douglas-Denton, R. N., Zimanyi, M. A. & Bertram, J. F. Towards a definition of glomerulomegaly: clinical-pathological and methodological considerations. Nephrol. Dialysis Transpl. 26, 2202–2208 (2011).

    Google Scholar 

  62. Liapis, H., Romagnani, P. & Anders, H. J. New insights into the pathology of podocyte loss: mitotic catastrophe. Am. J. Pathol. 183, 1364–1374 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Hodgin, J. B. et al. Glomerular aging and focal global glomerulosclerosis: a podometric perspective. J. Am. Soc. Nephrol. 26, 3162–3178 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Kopp, J. B. et al. Podocytopathies. Nat. Rev. Dis. Prim. 6, 68 (2020).

    PubMed  Google Scholar 

  65. Benz, K. et al. Early glomerular alterations in genetically determined low nephron number. Am. J. Physiol. Ren. Physiol. 300, F521–F530 (2011).

    CAS  Google Scholar 

  66. Butt, L. et al. A molecular mechanism explaining albuminuria in kidney disease. Nat. Metab. 2, 461–474 (2020).

    CAS  PubMed  Google Scholar 

  67. Fine, L. G. & Norman, J. Cellular events in renal hypertrophy. Annu. Rev. Physiol. 51, 19–32 (1989).

    CAS  PubMed  Google Scholar 

  68. Fine, L. G., Schlondorff, D., Trizna, W., Gilbert, R. M. & Bricker, N. S. Functional profile of the isolated uremic nephron. Impaired water permeability and adenylate cyclase responsiveness of the cortical collecting tubule to vasopressin. J. Clin. Invest. 61, 1519–1527 (1978).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Denic, A. et al. Clinical and pathology findings associate consistently with larger glomerular volume. J. Am. Soc. Nephrol. 29, 1960–1969 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Menn-Josephy, H. et al. Renal interstitial fibrosis: an imperfect predictor of kidney disease progression in some patient cohorts. Am. J. Nephrol. 44, 289–299 (2016).

    PubMed  Google Scholar 

  71. Abedini, A. et al. Urinary single-cell profiling captures the cellular diversity of the kidney. J. Am. Soc. Nephrol. 32, 614–627 (2021).

    CAS  PubMed  Google Scholar 

  72. Kriz, W. & Lemley, K. V. Potential relevance of shear stress for slit diaphragm and podocyte function. Kidney Int. 91, 1283–1286 (2017).

    PubMed  Google Scholar 

  73. Ryu, M., Mulay, S. R., Miosge, N., Gross, O. & Anders, H. J. Tumour necrosis factor-α drives Alport glomerulosclerosis in mice by promoting podocyte apoptosis. J. Pathol. 226, 120–131 (2012).

    CAS  PubMed  Google Scholar 

  74. Tao, J., Polumbo, C., Reidy, K., Sweetwyne, M. & Susztak, K. A multicolor podocyte reporter highlights heterogeneous podocyte changes in focal segmental glomerulosclerosis. Kidney Int. 85, 972–980 (2014).

    CAS  PubMed  Google Scholar 

  75. Wickman, L. et al. Urine podocyte mRNAs, proteinuria, and progression in human glomerular diseases. J. Am. Soc. Nephrol. 24, 2081–2095 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Ruggenenti, P., Cravedi, P. & Remuzzi, G. Mechanisms and treatment of CKD. J. Am. Soc. Nephrol. 23, 1917–1928 (2012).

    CAS  PubMed  Google Scholar 

  77. Ma, Q., Steiger, S. & Anders, H. J. Sodium glucose transporter-2 inhibition has no renoprotective effects on non-diabetic chronic kidney disease. Physiol. Rep. 5, e13228 (2017).

    PubMed  PubMed Central  Google Scholar 

  78. De Chiara, L., Lazzeri, E. & Romagnani, P. Tubular epithelial cell polyploidy is essential to survive AKI but it contributes to CKD progression. Nephrol. Dialysis Transpl. Assoc. 36, i29–i31 (2021).

    Google Scholar 

  79. Câmara, N. O., Iseki, K., Kramer, H., Liu, Z. H. & Sharma, K. Kidney disease and obesity: epidemiology, mechanisms and treatment. Nat. Rev. Nephrol. 13, 181–190 (2017).

    PubMed  Google Scholar 

  80. Helal, I., Fick-Brosnahan, G. M., Reed-Gitomer, B. & Schrier, R. W. Glomerular hyperfiltration: definitions, mechanisms and clinical implications. Nat. Rev. Nephrol. 8, 293–300 (2012).

    CAS  PubMed  Google Scholar 

  81. Tuttle, K. R. et al. Effect of strict glycemic control on renal hemodynamic response to amino acids and renal enlargement in insulin-dependent diabetes mellitus. N. Engl. J. Med. 324, 1626–1632 (1991).

    CAS  PubMed  Google Scholar 

  82. Anders, H. J., Huber, T. B., Isermann, B. & Schiffer, M. CKD in diabetes: diabetic kidney disease versus nondiabetic kidney disease. Nat. Rev. Nephrol. 14, 361–377 (2018).

    CAS  PubMed  Google Scholar 

  83. Brenner, B. M., Meyer, T. W. & Hostetter, T. H. Dietary protein intake and the progressive nature of kidney disease: the role of hemodynamically mediated glomerular injury in the pathogenesis of progressive glomerular sclerosis in aging, renal ablation, and intrinsic renal disease. N. Engl. J. Med. 307, 652–659 (1982).

    CAS  PubMed  Google Scholar 

  84. Hummel, D. et al. Dihydropyridine calcium antagonists are associated with increased albuminuria in treatment-resistant hypertensives. J. Nephrol. 23, 563–568 (2010).

    PubMed  Google Scholar 

  85. Richardson, K. L. et al. L-type calcium channel blocker use and proteinuria among children with chronic kidney diseases. Pediatr. Nephrol. 36, 2411–2419 (2021).

    PubMed  Google Scholar 

  86. Gashti, C. N. & Bakris, G. L. The role of calcium antagonists in chronic kidney disease. Curr. Opin. Nephrol. Hypertens. 13, 155–161 (2004).

    CAS  PubMed  Google Scholar 

  87. Pergola, P. E. et al. Bardoxolone methyl and kidney function in CKD with type 2 diabetes. N. Engl. J. Med. 365, 327–336 (2011).

    CAS  PubMed  Google Scholar 

  88. Baigent, C. & Lennon, R. Should we increase GFR with bardoxolone in Alport syndrome? J. Am. Soc. Nephrol. 29, 357–359 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Himmelfarb, J. & Tuttle, K. R. New therapies for diabetic kidney disease. N. Engl. J. Med. 369, 2549–2550 (2013).

    CAS  PubMed  Google Scholar 

  90. Silva-Islas, C. A. & Maldonado, P. D. Canonical and non-canonical mechanisms of Nrf2 activation. Pharmacol. Res. 134, 92–99 (2018).

    CAS  PubMed  Google Scholar 

  91. Heerspink, H. J. L. et al. Dapagliflozin in patients with chronic kidney disease. N. Eng. J. Med. 383, 1436–1446 (2020).

    CAS  Google Scholar 

  92. Perkovic, V. et al. Canagliflozin and renal outcomes in type 2 diabetes and nephropathy. N. Engl. J. Med. 380, 2295–2306 (2019).

    CAS  PubMed  Google Scholar 

  93. Wanner, C. et al. Empagliflozin and progression of kidney disease in type 2 diabetes. N. Engl. J. Med. 375, 323–334 (2016).

    CAS  PubMed  Google Scholar 

  94. Gross, O. et al. Early angiotensin-converting enzyme inhibition in Alport syndrome delays renal failure and improves life expectancy. Kidney Int. 81, 494–501 (2012).

    CAS  PubMed  Google Scholar 

  95. Kidney Disease: Improving Global Outcomes Anemia Work Group. KDIGO clinical practice guideline for anemia in chronic kidney disease. Kidney Int. Suppl. 2, 279–335 (2012).

    Google Scholar 

  96. Kidney Disease: Improving Global Outcomes Blood Pressure Work Group. KDIGO clinical practice guideline for the management of blood pressure in chronic kidney disease. Kidney Int. Suppl. 2, 337–414 (2012).

    Google Scholar 

  97. Kidney Disease: Improving Global Outcomes Lipid Work Group. KDIGO clinical practice guideline for lipid management in chronic kidney disease. Kidney Int. Suppl. 3, 259–305 (2013).

    Google Scholar 

  98. Kidney Disease: Improving Global Outcomes Lipid Work Group. KDIGO 2020 clinical practice guideline for diabetes management in chronic kidney Disease. Kidney Int. 98, S1–S115 (2020).

    Google Scholar 

  99. Neal, B. et al. Canagliflozin and cardiovascular and renal events in type 2 diabetes. N. Engl. J. Med. 377, 644–657 (2017).

    CAS  PubMed  Google Scholar 

  100. Neuen, B. L. et al. SGLT2 inhibitors for the prevention of kidney failure in patients with type 2 diabetes: a systematic review and meta-analysis. Lancet Diabetes Endocrinol. 7, 845–854 (2019).

    CAS  PubMed  Google Scholar 

  101. Wiviott, S. D. et al. Dapagliflozin and cardiovascular outcomes in type 2 diabetes. N. Engl. J. Med. 380, 347–357 (2019).

    CAS  PubMed  Google Scholar 

  102. Wheeler, D. C. et al. Effects of dapagliflozin on major adverse kidney and cardiovascular events in patients with diabetic and non-diabetic chronic kidney disease: a prespecified analysis from the DAPA-CKD trial. Lancet Diabetes Endocrinol. 9, 22–31 (2021).

    CAS  PubMed  Google Scholar 

  103. Heerspink, H. J. L. et al. Effects of dapagliflozin on mortality in patients with chronic kidney disease: a pre-specified analysis from the DAPA-CKD randomized controlled trial. Eur. Heart J. 42, 1216–1227 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Alicic, R. Z., Cox, E. J., Neumiller, J. J. & Tuttle, K. R. Incretin drugs in diabetic kidney disease: biological mechanisms and clinical evidence. Nat. Rev. Nephrol. 17, 227–244 (2021).

    CAS  PubMed  Google Scholar 

  105. Bakris, G. L. et al. Effect of finerenone on chronic kidney disease outcomes in type 2 diabetes. N. Engl. J. Med. 383, 2219–2229 (2020).

    CAS  PubMed  Google Scholar 

  106. González-Blázquez, R. et al. Finerenone attenuates endothelial dysfunction and albuminuria in a chronic kidney disease model by a reduction in oxidative stress. Front. Pharmacol. 9, 1131 (2018).

    PubMed  PubMed Central  Google Scholar 

  107. Heerspink, H. J. L. et al. Atrasentan and renal events in patients with type 2 diabetes and chronic kidney disease (SONAR): a double-blind, randomised, placebo-controlled trial. Lancet 393, 1937–1947 (2019).

    CAS  PubMed  Google Scholar 

  108. Kohan, D. E. & Barton, M. Endothelin and endothelin antagonists in chronic kidney disease. Kidney Int. 86, 896–904 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Heerspink, H. J. L., Kohan, D. E. & de Zeeuw, D. New insights from SONAR indicate adding sodium glucose co-transporter 2 inhibitors to an endothelin receptor antagonist mitigates fluid retention and enhances albuminuria reduction. Kidney Int. 99, 346–349 (2021).

    CAS  PubMed  Google Scholar 

  110. Rangaswami, J., Tuttle, K. & Vaduganathan, M. Cardio-renal-metabolic care models: toward achieving effective interdisciplinary care. Circ. Cardiovasc. Qual. Outcomes 13, e007264 (2020).

    PubMed  Google Scholar 

  111. Liyanage, T. et al. Worldwide access to treatment for end-stage kidney disease: a systematic review. Lancet 385, 1975–1982 (2015).

    PubMed  Google Scholar 

  112. Elshahat, S. et al. The impact of chronic kidney disease on developed countries from a health economics perspective: a systematic scoping review. PLoS ONE 15, e0230512 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Gandjour, A., Armsen, W., Wehmeyer, W., Multmeier, J. & Tschulena, U. Costs of patients with chronic kidney disease in Germany. PLoS ONE 15, e0231375 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Eriksson, J. K., Neovius, M., Jacobson, S. H., Elinder, C. G. & Hylander, B. Healthcare costs in chronic kidney disease and renal replacement therapy: a population-based cohort study in Sweden. BMJ Open 6, e012062 (2016).

    PubMed  PubMed Central  Google Scholar 

  115. Subramanian, S. et al. Cost and affordability of non-communicable disease screening, diagnosis and treatment in Kenya: patient payments in the private and public sectors. PLoS ONE 13, e0190113 (2018).

    PubMed  PubMed Central  Google Scholar 

  116. Vanholder, R. et al. Reducing the costs of chronic kidney disease while delivering quality health care: a call to action. Nat. Rev. Nephrol. 13, 393–409 (2017).

    PubMed  Google Scholar 

  117. Tonelli, M. & Dickinson, J. A. Early detection of CKD: implications for low-income, middle-income, and high-income countries. J. Am. Soc. Nephrol. 31, 1931–1940 (2020).

    PubMed  PubMed Central  Google Scholar 

  118. Komenda, P. et al. Cost-effectiveness of primary screening for CKD: a systematic review. Am. J. Kidney Dis. 63, 789–797 (2014).

    PubMed  Google Scholar 

  119. Willis, M. et al. Cost-effectiveness of canagliflozin added to standard of care for treating diabetic kidney disease (DKD) in patients with type 2 diabetes mellitus (T2DM) in England: estimates using the CREDEM-DKD model. Diabetes Ther. 12, 313–328 (2021).

    PubMed  Google Scholar 

  120. Ashuntantang, G. et al. Outcomes in adults and children with end-stage kidney disease requiring dialysis in sub-Saharan Africa: a systematic review. Lancet Glob. Health 5, e408–e417 (2017).

    PubMed  Google Scholar 

  121. Chow, C. K. et al. Availability and affordability of medicines and cardiovascular outcomes in 21 high-income, middle-income and low-income countries. BMJ Glob. Health 5, e002640 (2020).

    PubMed  PubMed Central  Google Scholar 

  122. Rook, M. et al. Nephrectomy elicits impact of age and BMI on renal hemodynamics: lower postdonation reserve capacity in older or overweight kidney donors. Am. J. Transplant. 8, 2077–2085 (2008).

    CAS  PubMed  Google Scholar 

  123. Pivin, E. et al. Uromodulin and nephron mass. Clin. J. Am. Soc. Nephrol. 13, 1556–1557 (2018).

    PubMed  PubMed Central  Google Scholar 

  124. Denic, A., Elsherbiny, H. & Rule, A. D. In-vivo techniques for determining nephron number. Curr. Opin. Nephrol. Hypertens. 28, 545–551 (2019).

    PubMed  PubMed Central  Google Scholar 

  125. Tofte, N. 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. 8, 301–312 (2020).

    CAS  PubMed  Google Scholar 

  126. Pruijm, M. et al. Reduced cortical oxygenation predicts a progressive decline of renal function in patients with chronic kidney disease. Kidney Int. 93, 932–940 (2018).

    PubMed  Google Scholar 

  127. Ruiz-Ortega, M., Rayego-Mateos, S., Lamas, S., Ortiz, A. & Rodrigues-Diez, R. R. Targeting the progression of chronic kidney disease. Nat. Rev. Nephrol. 16, 269–288 (2020).

    PubMed  Google Scholar 

  128. Wesson, D. E. The continuum of acid stress. Clin. J. Am. Soc. Nephrol. 16, 1292–1299 (2021).

    CAS  PubMed  Google Scholar 

  129. Anders, H.-J., Peired, A. J. & Romagnani, P. SGLT2 inhibition requires reconsideration of fundamental paradigms in chronic kidney disease, ‘diabetic nephropathy’, IgA nephropathy and podocytopathies with FSGS lesions. Nephrol. Dial. Transplant. https://doi.org/10.1093/ndt/gfaa329 (2020).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

K.R.T. is supported by four NIDDK/NIH grants, one NCATS/NIH grant, one NIMHD/NIH grant, and a CDC contract all from the US Government, as well as research grants from Goldfinch Bio, Bayer and Travere. A.D.R. is supported by funding from the National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases (R01 DK090358). H.-J.A. is supported by the Deutsche Forschungsgemeinschaft (AN372/30-1).

Author information

Authors and Affiliations

Authors

Contributions

All authors researched data for the article, made substantial contributions to discussions of the content and wrote, reviewed and edited the manuscript before submission.

Corresponding author

Correspondence to Hans-Joachim Anders.

Ethics declarations

Competing interests

P.D. has received consultancy fees from Bayer and AstraZeneca. K.R.T. has received consulting fees for diabetes and CKD from Eli Lilly and Company, Boehringer Ingelheim, AstraZeneca, Gilead, Goldfinch Bio, Novo Nordisk and Bayer. A.G. has received consulting and lecture fees from Fresenius Medical Care. H.-J.A. has received consultancy fees from Bayer, Janssen, GSK, Novartis, Boehringer, AstraZeneca and PreviPharma. The other authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Nephrology thanks M. Canney, A. Levin, R. Liu, P. Rossing, J. Wei and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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

Luyckx, V.A., Rule, A.D., Tuttle, K.R. et al. Nephron overload as a therapeutic target to maximize kidney lifespan. Nat Rev Nephrol 18, 171–183 (2022). https://doi.org/10.1038/s41581-021-00510-7

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41581-021-00510-7

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing