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:

Mechanisms of vascular calcification in CKD—evidence for premature ageing?

Abstract

Ageing is a potent, independent risk factor for cardiovascular disease. Calcification of the vascular smooth muscle cell (VSMC) layer of the vessel media is a hallmark of vascular ageing. Young patients with chronic kidney disease (CKD) exhibit an extremely high cardiovascular mortality, equivalent to that seen in octogenarians in the general population. Even children on dialysis develop accelerated medial vascular calcification and arterial stiffening, leading to the suggestion that patients with CKD exhibit a 'premature ageing' phenotype. It is now well documented that uraemic toxins, particularly those associated with dysregulated mineral metabolism, can drive VSMC damage and phenotypic changes that promote vascular calcification; epidemiological data suggest that some of these same risk factors associate with cardiovascular mortality in the aged general population. Importantly, emerging evidence suggests that uraemic toxins may promote DNA damage, a key factor driving cellular ageing, and moreover, that these ageing mechanisms may reiterate some of those seen in patients with genetically induced progeric syndromes caused by nuclear lamina disruption. This new knowledge should pave the way for the development of novel therapies that target tissue-specific ageing mechanisms to treat vascular decline in CKD.

Key Points

  • Vascular calcification is an age-associated pathology that is accelerated in patients with chronic kidney disease (CKD) and associated with increased mortality

  • Vascular calcification is a cell-mediated process driven by vascular smooth muscle cell (VSMC) death and maladaptation

  • Emerging evidence suggests that defects in the nuclear lamina occur during VSMC ageing and in CKD, causing accelerated DNA damage and premature senescence

  • Senescent VSMCs develop a secretory phenotype and release factors that drive VSMC osteogenic differentiation both locally and potentially at remote sites

  • DNA damage may also link the parallel decline of the vasculature–kidney–bone axis in CKD

  • Prelamin A accumulation and/or DNA damage signalling pathways may represent novel therapeutic targets for vascular calcification in CKD

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

Figure 1: Mechanisms leading to VSMC phenotypic change and calcification in response to dysregulated mineral metabolism in chronic kidney disease.
Figure 2: DNA damage drives the SASP in VSMCs.
Figure 3: Prelamin A accumulation promotes vascular ageing and dysfunction.
Figure 4: Tissue ageing is driven by DNA damage and inflammatory mediators released from senescent tissues.

Similar content being viewed by others

References

  1. Go, A. S. et al. Chronic kidney disease and the risks of death, cardiovascular events, and hospitalization. N. Engl. J. Med. 351, 1296–1305 (2004).

    Article  CAS  PubMed  Google Scholar 

  2. Goodman, W. G. et al. Coronary-artery calcification in young adults with end-stage renal disease who are undergoing dialysis. N. Engl. J. Med. 342, 1478–1483 (2000).

    Article  CAS  PubMed  Google Scholar 

  3. Foley, R. N. & Parfrey, P. S. Cardiovascular disease and mortality in ESRD. J. Nephrol. 11, 239–245 (1998).

    CAS  PubMed  Google Scholar 

  4. Mitsnefes, M. M. Cardiovascular disease in children with chronic kidney disease. J. Am. Soc. Nephrol. 23, 578–585 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Virchow, R. Cellular pathology. As based upon physiological and pathological histology. Lecture XVI--Atheromatous affection of arteries. 1858. Nutr. Rev. 47, 23–25 (1989).

    Article  CAS  PubMed  Google Scholar 

  6. McEniery, C. M. et al. The impact of cardiovascular risk factors on aortic stiffness and wave reflections depends on age: the Anglo-Cardiff Collaborative Trial (ACCT III). Hypertension 56, 591–597 (2010).

    Article  CAS  PubMed  Google Scholar 

  7. Kovacic, J. C. et al. Cellular senescence, vascular disease, and aging: part 1 of a 2-part review. Circulation 123, 1650–1660 (2011).

    Article  PubMed  Google Scholar 

  8. Kovacic, J. C. et al. Cellular senescence, vascular disease, and aging: part 2 of a 2-part review: clinical vascular disease in the elderly. Circulation 123, 1900–1910 (2011).

    Article  PubMed  Google Scholar 

  9. London, G. M. Arterial calcification: cardiovascular function and clinical outcome. Nefrologia 31, 644–647 (2011).

    CAS  PubMed  Google Scholar 

  10. London, G. M. et al. Arteriosclerosis, vascular calcifications and cardiovascular disease in uremia. Curr. Opin. Nephrol. Hypertens. 14, 525–531 (2005).

    Article  CAS  PubMed  Google Scholar 

  11. Shaw, L. J. et al. Coronary artery calcium as a measure of biologic age. Atherosclerosis 188, 112–119 (2006).

    Article  CAS  PubMed  Google Scholar 

  12. Girndt, M. & Seibert, E. Premature cardiovascular disease in chronic renal failure (CRF): a model for an advanced ageing process. Exp. Gerontol. 45, 797–800 (2010).

    Article  CAS  PubMed  Google Scholar 

  13. Block, G. A. et al. Association of serum phosphorus and calcium x phosphate product with mortality risk in chronic hemodialysis patients: a national study. Am. J. Kidney Dis. 31, 607–617 (1998).

    Article  CAS  PubMed  Google Scholar 

  14. Tonelli, M. et al. Relation between alkaline phosphatase, serum phosphate, and all-cause or cardiovascular mortality. Circulation 120, 1784–1792 (2009).

    Article  CAS  PubMed  Google Scholar 

  15. London, G. M. Bone-vascular cross-talk. J. Nephrol. 25, 619–625 (2012).

    Article  PubMed  Google Scholar 

  16. Mackenzie, I. S. et al. Comparison of the effects of antihypertensive agents on central blood pressure and arterial stiffness in isolated systolic hypertension. Hypertension 54, 409–413 (2009).

    Article  CAS  PubMed  Google Scholar 

  17. Wanner, C. et al. Atorvastatin in patients with type 2 diabetes mellitus undergoing hemodialysis. N. Engl. J. Med. 353, 238–248 (2005).

    Article  CAS  PubMed  Google Scholar 

  18. Shanahan, C. M. et al. Medial localization of mineralization-regulating proteins in association with Monckeberg's sclerosis: evidence for smooth muscle cell-mediated vascular calcification. Circulation 100, 2168–2176 (1999).

    Article  CAS  PubMed  Google Scholar 

  19. Shroff, R., Long, D. A. & Shanahan, C. Mechanistic insights into vascular calcification in CKD. J. Am. Soc. Nephrol. 24, 179–189 (2013).

    Article  CAS  PubMed  Google Scholar 

  20. Liu, Y. et al. Prelamin A accelerates vascular calcification via activation of the DNA damage response and senescence associated secretory phenotype in vascular smooth muscle cells. Circ. Res. 112, e99–e109 (2013).

    Article  CAS  PubMed  Google Scholar 

  21. Nakano-Kurimoto, R. et al. Replicative senescence of vascular smooth muscle cells enhances the calcification through initiating the osteoblastic transition. Am. J. Physiol. Heart Circ. Physiol. 297, H1673–H1684 (2009).

    Article  CAS  PubMed  Google Scholar 

  22. Marchand, A. et al. The Wnt/beta-catenin pathway is activated during advanced arterial aging in humans. Aging Cell 10, 220–232 (2011).

    Article  CAS  PubMed  Google Scholar 

  23. Burton, D. G. et al. Microarray analysis of senescent vascular smooth muscle cells: a link to atherosclerosis and vascular calcification. Exp. Gerontol. 44, 659–665 (2009).

    Article  CAS  PubMed  Google Scholar 

  24. Hutchison, C. J. The role of DNA damage in laminopathy progeroid syndromes. Biochem. Soc. Trans. 39, 1715–1718 (2011).

    Article  CAS  PubMed  Google Scholar 

  25. Warren, D. T. & Shanahan, C. M. Defective DNA-damage repair induced by nuclear lamina dysfunction is a key mediator of smooth muscle cell aging. Biochem. Soc. Trans. 39, 1780–1785 (2011).

    Article  CAS  PubMed  Google Scholar 

  26. Shanahan, C. M. Inflammation ushers in calcification: a cycle of damage and protection? Circulation 116, 2782–2785 (2007).

    Article  PubMed  Google Scholar 

  27. Iyemere, V. P. et al. Vascular smooth muscle cell phenotypic plasticity and the regulation of vascular calcification. J. Intern. Med. 260, 192–210 (2006).

    Article  CAS  PubMed  Google Scholar 

  28. Shanahan, C. M. et al. Arterial calcification in chronic kidney disease: key roles for calcium and phosphate. Circ. Res. 109, 697–711 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Jahnen-Dechent, W. et al. Fetuin-A regulation of calcified matrix metabolism. Circ. Res. 108, 1494–1509 (2011).

    Article  CAS  PubMed  Google Scholar 

  30. McCabe, K. M. et al. Dietary vitamin K and therapeutic warfarin alter the susceptibility to vascular calcification in experimental chronic kidney disease. Kidney Int. 83, 835–844 (2013).

    Article  CAS  PubMed  Google Scholar 

  31. Yamada, S. et al. The antioxidant tempol ameliorates arterial medial calcification in uremic rats: important role of oxidative stress in the pathogenesis of vascular calcification in chronic kidney disease. J. Bone Miner. Res. 27, 474–485 (2012).

    Article  CAS  PubMed  Google Scholar 

  32. Vaziri, N. D. Oxidative stress in uremia: nature, mechanisms, and potential consequences. Semin. Nephrol. 24, 469–473 (2004).

    Article  CAS  PubMed  Google Scholar 

  33. Zhao, M.-M. et al. Mitochondrial reactive oxygen species promote p65 nuclear translocation mediating high-phosphate-induced vascular calcification in vitro and in vivo. Kidney Int. 79, 1071–1079 (2011).

    Article  CAS  PubMed  Google Scholar 

  34. Shroff, R. C. et al. Dialysis accelerates medial vascular calcification in part by triggering smooth muscle cell apoptosis. Circulation 118, 1748–1757 (2008).

    Article  CAS  PubMed  Google Scholar 

  35. Reynolds, J. L. et al. Human vascular smooth muscle cells undergo vesicle-mediated calcification in response to changes in extracellular calcium and phosphate concentrations: a potential mechanism for accelerated vascular calcification in ESRD. J. Am. Soc. Nephrol. 15, 2857–2867 (2004).

    Article  CAS  PubMed  Google Scholar 

  36. Kapustin, A. N. et al. Calcium regulates key components of vascular smooth muscle cell-derived matrix vesicles to enhance mineralization. Circ. Res. 109, e1–e12 (2011).

    Article  CAS  PubMed  Google Scholar 

  37. Schlieper, G. et al. Ultrastructural analysis of vascular calcifications in uremia. J. Am. Soc. Nephrol. 21, 689–696 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Kapustin, A. N. & Shanahan, C. M. Calcium regulation of vascular smooth muscle cell-derived matrix vesicles. Trends Cardiovasc. Med. 22, 133–137 (2012).

    Article  CAS  PubMed  Google Scholar 

  39. Reynolds, J. L. et al. Multifunctional roles for serum protein fetuin-a in inhibition of human vascular smooth muscle cell calcification. J. Am. Soc. Nephrol. 16, 2920–2930 (2005).

    Article  CAS  PubMed  Google Scholar 

  40. Ewence, A. E. et al. Calcium phosphate crystals induce cell death in human vascular smooth muscle cells: a potential mechanism in atherosclerotic plaque destabilization. Circ. Res. 103, e28–e34 (2008).

    Article  CAS  PubMed  Google Scholar 

  41. Sage, A. P. et al. Hyperphosphatemia-induced nanocrystals upregulate the expression of bone morphogenetic protein-2 and osteopontin genes in mouse smooth muscle cells in vitro. Kidney Int. 79, 414–422 (2011).

    Article  CAS  PubMed  Google Scholar 

  42. Bellasi, A. & Raggi, P. Vascular imaging in chronic kidney disease. Curr. Opin. Nephrol. Hypertens. 21, 382–388 (2012).

    Article  PubMed  Google Scholar 

  43. Speer, M. Y. et al. Smooth muscle cells give rise to osteochondrogenic precursors and chondrocytes in calcifying arteries. Circ. Res. 104, 733–741 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Luo, G. et al. Spontaneous calcification of arteries and cartilage in mice lacking matrix GLA protein. Nature 386, 78–81 (1997).

    Article  CAS  PubMed  Google Scholar 

  45. Yao, Y. et al. Inhibition of bone morphogenetic proteins protects against atherosclerosis and vascular calcification. Circ. Res. 107, 485–494 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Westenfeld, R. et al. Effect of vitamin K2 supplementation on functional vitamin K deficiency in hemodialysis patients: a randomized trial. Am. J. Kidney Dis. 59, 186–195 (2012).

    Article  CAS  PubMed  Google Scholar 

  47. Hermans, M. M. et al. Association of serum fetuin-A levels with mortality in dialysis patients. Kidney Int. 72, 202–207 (2007).

    Article  CAS  PubMed  Google Scholar 

  48. Fusaro, M. et al. Vitamin K, vertebral fractures, vascular calcifications, and mortality: VItamin K Italian (VIKI) dialysis study. J. Bone Miner. Res. 27, 2271–2278 (2012).

    Article  CAS  PubMed  Google Scholar 

  49. Fish, R. S. et al. ATP and arterial calcification. Eur. J. Clin. Invest. 43, 405–412 (2013).

    Article  CAS  PubMed  Google Scholar 

  50. Rutsch, F. et al. Mutations in ENPP1 are associated with 'idiopathic' infantile arterial calcification. Nat. Genet. 34, 379–381 (2003).

    Article  CAS  PubMed  Google Scholar 

  51. Lomashvili, K. A. et al. Upregulation of alkaline phosphatase and pyrophosphate hydrolysis: potential mechanism for uremic vascular calcification. Kidney Int. 73, 1024–1030 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Narisawa, S. et al. Novel inhibitors of alkaline phosphatase suppress vascular smooth muscle cell calcification. J. Bone Miner. Res. 22, 1700–1710 (2007).

    Article  CAS  PubMed  Google Scholar 

  53. O'Neill, W. C. Pyrophosphate, alkaline phosphatase, and vascular calcification. Circ. Res. 99, e2 (2006).

    CAS  PubMed  Google Scholar 

  54. Lencel, P. et al. Cell-specific effects of TNF-alpha and IL-1beta on alkaline phosphatase: implication for syndesmophyte formation and vascular calcification. Lab. Invest. 91, 1434–1442 (2011).

    Article  CAS  PubMed  Google Scholar 

  55. Ding, J. et al. TNF-alpha and IL-1beta inhibit RUNX2 and collagen expression but increase alkaline phosphatase activity and mineralization in human mesenchymal stem cells. Life Sci. 84, 499–504 (2009).

    Article  CAS  PubMed  Google Scholar 

  56. Zebboudj, A. F., Shin, V. & Bostrom, K. Matrix GLA protein and BMP-2 regulate osteoinduction in calcifying vascular cells. J. Cell. Biochem. 90, 756–765 (2003).

    Article  CAS  PubMed  Google Scholar 

  57. Schinke, T. et al. The serum protein alpha2-HS glycoprotein/fetuin inhibits apatite formation in vitro and in mineralizing calvaria cells. A possible role in mineralization and calcium homeostasis. J. Biol. Chem. 271, 20789–20796 (1996).

    Article  CAS  PubMed  Google Scholar 

  58. Schafer, C. et al. The serum protein alpha 2-Heremans-Schmid glycoprotein/fetuin-A is a systemically acting inhibitor of ectopic calcification. J. Clin. Invest. 112, 357–366 (2003).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Westenfeld, R. et al. Fetuin-A protects against atherosclerotic calcification in CKD. J. Am. Soc. Nephrol. 20, 1264–1274 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Ketteler, M. et al. Association of low fetuin-A (AHSG) concentrations in serum with cardiovascular mortality in patients on dialysis: a cross-sectional study. Lancet 361, 827–833 (2003).

    Article  CAS  PubMed  Google Scholar 

  61. Smith, E. R. et al. Serum fetuin-A concentration and fetuin-A-containing calciprotein particles in patients with chronic inflammatory disease and renal failure. Nephrology (Carlton) 18, 215–221 (2013).

    Article  CAS  Google Scholar 

  62. Smith, E. R. et al. Phosphorylated fetuin-A-containing calciprotein particles are associated with aortic stiffness and a procalcific milieu in patients with pre-dialysis CKD. Nephrol. Dial. Transplant. 27, 1957–1966 (2012).

    Article  CAS  PubMed  Google Scholar 

  63. Hamano, T. et al. Fetuin-mineral complex reflects extraosseous calcification stress in CKD. J. Am. Soc. Nephrol. 21, 1998–2007 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Pasch, A. et al. Nanoparticle-based test measures overall propensity for calcification in serum. J. Am. Soc. Nephrol. 23, 1744–1752 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Kuro-o, M. Klotho, phosphate and FGF-23 in ageing and disturbed mineral metabolism. Nat. Rev. Nephrol. http://dx.doi.org/10.1038/nrneph.2013.111.

  66. Louvet, L. et al. Magnesium prevents phosphate-induced calcification in human aortic vascular smooth muscle cells. Nephrol. Dial. Transplant. 28, 869–878 (2013).

    Article  CAS  PubMed  Google Scholar 

  67. Schoppet, M. et al. Exploring the biology of vascular calcification in chronic kidney disease: what's circulating? Kidney Int. 73, 384–390 (2008).

    Article  CAS  PubMed  Google Scholar 

  68. Salem, S. et al. Relationship between magnesium and clinical biomarkers on inhibition of vascular calcification. Am. J. Nephrol. 35, 31–39 (2012).

    Article  CAS  PubMed  Google Scholar 

  69. Tyson, K. L. et al. Osteo/chondrocytic transcription factors and their target genes exhibit distinct patterns of expression in human arterial calcification. Arterioscler. Thromb. Vasc. Biol. 23, 489–494 (2003).

    Article  CAS  PubMed  Google Scholar 

  70. Tintut, Y. et al. Multilineage potential of cells from the artery wall. Circulation 108, 2505–2510 (2003).

    Article  PubMed  Google Scholar 

  71. Bostrom, K. I., Rajamannan, N. M. & Towler, D. A. The regulation of valvular and vascular sclerosis by osteogenic morphogens. Circ. Res. 109, 564–577 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Liu, Y. & Shanahan, C. M. Signalling pathways and vascular calcification. Front. Biosci. 16, 1302–1314 (2011).

    Article  CAS  Google Scholar 

  73. Byon, C. H. et al. Oxidative stress induces vascular calcification through modulation of the osteogenic transcription factor Runx2 by AKT signaling. J. Biol. Chem. 283, 15319–15327 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Li, X. & Giachelli, C. M. Sodium-dependent phosphate cotransporters and vascular calcification. Curr. Opin. Nephrol. Hypertens. 16, 325–328 (2007).

    Article  PubMed  Google Scholar 

  75. Voelkl, J. et al. Spironolactone ameliorates PIT1-dependent vascular osteoinduction in klotho-hypomorphic mice. J. Clin. Invest. 123, 812–822 (2013).

    PubMed  PubMed Central  Google Scholar 

  76. Cheng, S. L. et al. Activation of vascular smooth muscle parathyroid hormone receptor inhibits Wnt/beta-catenin signaling and aortic fibrosis in diabetic arteriosclerosis. Circ. Res. 107, 271–282 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Sun, Y. et al. Smooth muscle cell-specific runx2 deficiency inhibits vascular calcification. Circ. Res. 111, 543–552 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Naik, V. et al. Sources of cells that contribute to atherosclerotic intimal calcification: an in vivo genetic fate mapping study. Cardiovasc. Res. 94, 545–554 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Lai, C. F. et al. TNFR1-activated reactive oxidative species signals up-regulate osteogenic Msx2 programs in aortic myofibroblasts. Endocrinology 153, 3897–3910 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Shroff, R. C. et al. Chronic mineral dysregulation promotes vascular smooth muscle cell adaptation and extracellular matrix calcification. J. Am. Soc. Nephrol. 21, 103–112 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Ohyama, Y. et al. Molecular cloning of rat klotho cDNA: markedly decreased expression of klotho by acute inflammatory stress. Biochem. Biophys. Res. Commun. 251, 920–925 (1998).

    Article  CAS  PubMed  Google Scholar 

  82. Shimada, T. et al. Targeted ablation of Fgf23 demonstrates an essential physiological role of FGF23 in phosphate and vitamin D metabolism. J. Clin. Invest. 113, 561–568 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. John, G. B., Cheng, C. Y. & Kuro-o, M. Role of Klotho in aging, phosphate metabolism, and CKD. Am. J. Kidney Dis. 58, 127–134 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Moe, S. M. Klotho: a master regulator of cardiovascular disease? Circulation 125, 2181–2183 (2012).

    Article  PubMed  Google Scholar 

  85. Hu, M. C. et al. Klotho deficiency causes vascular calcification in chronic kidney disease. J. Am. Soc. Nephrol. 22, 124–136 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Kuro-o, M. Klotho as a regulator of oxidative stress and senescence. Biol. Chem. 389, 233–241 (2008).

    Article  CAS  PubMed  Google Scholar 

  87. Liu, F. et al. Klotho suppresses RIG-I-mediated senescence-associated inflammation. Nat. Cell Biol. 13, 254–262 (2011).

    Article  CAS  PubMed  Google Scholar 

  88. Shiloh, Y. & Ziv, Y. The ATM protein kinase: regulating the cellular response to genotoxic stress, and more. Nat. Rev. Mol. Cell Biol. 14, 197–210 (2013).

    Article  CAS  PubMed  Google Scholar 

  89. d'Adda di Fagagna, F. et al. A DNA damage checkpoint response in telomere-initiated senescence. Nature 426, 194–198 (2003).

    Article  CAS  PubMed  Google Scholar 

  90. Toussaint, O. et al. Stress-induced premature senescence and tissue ageing. Biochem. Pharmacol. 64, 1007–1009 (2002).

    Article  CAS  PubMed  Google Scholar 

  91. Rodier, F. et al. Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion. Nat. Cell Biol. 11, 973–979 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Song, Y. et al. Aging enhances the basal production of IL-6 and CCL2 in vascular smooth muscle cells. Arterioscler. Thromb. Vasc. Biol. 32, 103–109 (2012).

    Article  CAS  PubMed  Google Scholar 

  93. Fyhrquist, F., Saijonmaa, O. & Strandberg, T. The roles of senescence and telomere shortening in cardiovascular disease. Nat. Rev. Cardiol. 10, 274–283 (2013).

    Article  CAS  PubMed  Google Scholar 

  94. Wang, J. C. & Bennett, M. Aging and atherosclerosis: mechanisms, functional consequences, and potential therapeutics for cellular senescence. Circ. Res. 111, 245–259 (2012).

    Article  CAS  PubMed  Google Scholar 

  95. Jacobi, C., Hömme, M. & Melk, A. Is cellular senescence important in pediatric kidney disease? Pediatr. Nephrol. 26, 2121–2131 (2011).

    Article  PubMed  Google Scholar 

  96. Boxall, M. C. et al. Telomere shortening and haemodialysis. Blood Purif. 24, 185–189 (2006).

    Article  PubMed  Google Scholar 

  97. Stenvinkel, P. & Larsson, T. E. Chronic kidney disease: a clinical model of premature aging. Am. J. Kidney Dis. 62, 339–351 (2013).

    Article  PubMed  Google Scholar 

  98. Ragnauth, C. D. et al. Prelamin A acts to accelerate smooth muscle cell senescence and is a novel biomarker of human vascular aging. Circulation 121, 2200–2210 (2010).

    Article  CAS  PubMed  Google Scholar 

  99. Gerhard-Herman, M. et al. Mechanisms of premature vascular aging in children with Hutchinson-Gilford progeria syndrome. Hypertension 59, 92–97 (2012).

    Article  CAS  PubMed  Google Scholar 

  100. Mounkes, L. C. & Stewart, C. L. Aging and nuclear organization: lamins and progeria. Curr. Opin. Cell Biol. 16, 322–327 (2004).

    Article  CAS  PubMed  Google Scholar 

  101. McClintock, D., Gordon, L. B. & Djabali, K. Hutchinson-Gilford progeria mutant lamin A primarily targets human vascular cells as detected by an anti-Lamin A G608G antibody. Proc. Natl Acad. Sci. USA 103, 2154–2159 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Richards, S. A. et al. The accumulation of un-repairable DNA damage in laminopathy progeria fibroblasts is caused by ROS generation and is prevented by treatment with N-acetyl cysteine. Hum. Mol. Genet. 20, 3997–4004 (2011).

    Article  CAS  PubMed  Google Scholar 

  103. Muteliefu, G. et al. Indoxyl sulfate promotes vascular smooth muscle cell senescence with upregulation of p53, p21, and prelamin A through oxidative stress. Am. J. Physiol. Cell Physiol. 303, C126–C134 (2012).

    Article  CAS  PubMed  Google Scholar 

  104. Liu, B. et al. Genomic instability in laminopathy-based premature aging. Nat. Med. 11, 780–785 (2005).

    Article  CAS  PubMed  Google Scholar 

  105. Ugalde, A. P. et al. Aging and chronic DNA damage response activate a regulatory pathway involving miR-29 and p53. EMBO J. 30, 2219–2232 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Gordon, L. B. et al. Clinical trial of a farnesyltransferase inhibitor in children with Hutchinson-Gilford progeria syndrome. Proc. Natl Acad. Sci. USA 109, 16666–16671 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  107. Varela, I. et al. Combined treatment with statins and aminobisphosphonates extends longevity in a mouse model of human premature aging. Nat. Med. 14, 767–772 (2008).

    Article  CAS  PubMed  Google Scholar 

  108. Liu, Y. et al. DNA damage responses in progeroid syndromes arise from defective maturation of prelamin A. J. Cell Sci. 119, 4644–4649 (2006).

    CAS  PubMed  Google Scholar 

  109. Chaki, M. et al. Exome capture reveals ZNF423 and CEP164 mutations, linking renal ciliopathies to DNA damage response signaling. Cell 150, 533–548 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Zhou, W. et al. FAN1 mutations cause karyomegalic interstitial nephritis, linking chronic kidney failure to defective DNA damage repair. Nat. Genet. 44, 910–915 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Rasheed, N. et al. Atm-deficient mice: an osteoporosis model with defective osteoblast differentiation and increased osteoclastogenesis. Hum. Mol. Genet. 15, 1938–1948 (2006).

    Article  CAS  PubMed  Google Scholar 

  112. Durik, M. et al. Nucleotide excision DNA repair is associated with age-related vascular dysfunction. Circulation 126, 468–478 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Zhang, J. et al. A human iPSC model of Hutchinson Gilford Progeria reveals vascular smooth muscle and mesenchymal stem cell defects. Cell Stem Cell 8, 31–45 (2011).

    Article  CAS  PubMed  Google Scholar 

  114. Scaffidi, P. & Misteli, T. Lamin A-dependent misregulation of adult stem cells associated with accelerated ageing. Nat. Cell Biol. 10, 452–459 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Agarwal, A. K. et al. Focal segmental glomerulosclerosis in patients with mandibuloacral dysplasia owing to ZMPSTE24 deficiency. J. Investig. Med. 54, 208–213 (2006).

    Article  PubMed  Google Scholar 

  116. Imachi, H. et al. A case of Dunnigan-type familial partial lipodystrophy (FPLD) due to lamin A/C (LMNA) mutations complicated by end-stage renal disease. Endocrine 35, 18–21 (2009).

    Article  CAS  PubMed  Google Scholar 

  117. Ahmad, Z. et al. Early onset mandibuloacral dysplasia due to compound heterozygous mutations in ZMPSTE24. Am. J. Med. Genet. A 152A, 2703–2710 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  118. Osorio, F. G. et al. Nuclear lamina defects cause ATM-dependent NF-kappaB activation and link accelerated aging to a systemic inflammatory response. Genes Dev. 26, 2311–2324 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Dalfino, G. et al. Bone morphogenetic protein-2 may represent the molecular link between oxidative stress and vascular stiffness in chronic kidney disease. Atherosclerosis 211, 418–423 (2010).

    Article  CAS  PubMed  Google Scholar 

  120. Wei, J. et al. Increase of plasma IL-6 concentration with age in healthy subjects. Life Sci. 51, 1953–1956 (1992).

    Article  CAS  PubMed  Google Scholar 

  121. Nitta, K. et al. Serum osteoprotegerin levels and the extent of vascular calcification in haemodialysis patients. Nephrol. Dial. Transplant. 19, 1886–1889 (2004).

    Article  CAS  PubMed  Google Scholar 

  122. Takemura, A. et al. Sirtuin 1 retards hyperphosphatemia-induced calcification of vascular smooth muscle cells. Arterioscler. Thromb. Vasc. Biol. 31, 2054–2062 (2011).

    Article  CAS  PubMed  Google Scholar 

  123. Tang, Y. et al. Resveratrol reduces vascular cell senescence through attenuation of oxidative stress by SIRT1/NADPH oxidase-dependent mechanisms. J. Nutr. Biochem. 23, 1410–1416 (2012).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The author's work is supported by the British Heart Foundation.

Author information

Authors and Affiliations

Authors

Ethics declarations

Competing interests

The author declares no competing financial interests.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Shanahan, C. Mechanisms of vascular calcification in CKD—evidence for premature ageing?. Nat Rev Nephrol 9, 661–670 (2013). https://doi.org/10.1038/nrneph.2013.176

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrneph.2013.176

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