Elsevier

Bone

Volume 100, July 2017, Pages 80-86
Bone

Review Article
The chronic kidney disease — Mineral bone disorder (CKD-MBD): Advances in pathophysiology

https://doi.org/10.1016/j.bone.2017.01.023Get rights and content

Highlights

  • This review discusses novel aspects of CKD-MBD pathogenesis.

Abstract

The causes of excess cardiovascular mortality associated with chronic kidney disease (CKD) have been attributed in part to the CKD-mineral bone disorder syndrome (CKD-MBD), wherein, novel cardiovascular risk factors have been identified. New advances in the causes of the CKD-MBD are discussed in this review. They demonstrate that repair and disease processes in the kidneys release factors to the circulation that cause the systemic complications of CKD. The discovery of WNT inhibitors, especially Dickkopf 1 (Dkk1), produced during renal repair as participating in the pathogenesis of the vascular and skeletal components of the CKD-MBD implied that additional pathogenic factors are critical. This lead to the discovery that activin A is a second renal repair factor circulating in increased levels during CKD. Activin A derives from peritubular myofibroblasts of diseased kidneys, wherein it stimulates fibrosis, and decreases tubular klotho expression. Activin A binds to the type 2 activin A receptor, ActRIIA, which is variably affected by CKD in the vasculature. In diabetic/atherosclerotic aortas, specifically in vascular smooth muscle cells (VSMC), ActRIIA signaling is inhibited and contributes to CKD induced VSMC dedifferentiation, osteogenic transition and neointimal atherosclerotic calcification. In nondiabetic/nonatherosclerotic aortas, CKD increases VSMC ActRIIA signaling, and vascular fibroblast signaling causing the latter to undergo osteogenic transition and stimulate vascular calcification. In both vascular situations, a ligand trap for ActRIIA prevented vascular calcification. In the skeleton, activin A is responsible for CKD stimulation of osteoclastogenesis and bone remodeling increasing bone turnover. These studies demonstrate that circulating renal repair and injury factors are causal of the CKD-MBD and CKD associated cardiovascular disease.

Section snippets

Introduction and epidemiology of the CKD-MBD

The kidney disease pandemic [1] is associated with high mortality rates, in part due to cardiovascular complications [2], [3], [4], [5]. The kidney disease produced increase in cardiovascular risk extends to type 2 diabetes [6], where the presence of mild to moderate kidney disease increases atherosclerotic cardiovascular disease risk by 87% [7]. The causes of the increased cardiovascular risk associated with kidney diseases partly reside in the chronic kidney disease – mineral bone disorder

Conclusion and future directions

The CKD-MBD defines a disruption in the systems biology between the injured kidney, skeleton, and cardiovascular system that has a profoundly negative impact on survival in CKD. Recent translational discoveries have introduced a new paradigm where kidney injury directly leads to skeletal and cardiovascular injury through the production of pathogenic circulating factors during attempted renal repair, including molecules that inhibit the canonical Wnt pathway and activin, both processes that have

Financial support and sponsorship

This work was funded by NIH grants, RO1DK070790 (KAH) and RO1DK089137 (KAH), an investigator stimulated grant from Celgene, and by NIH grants UL1 TR000448, KL2 TR000450, and L40 DK099748-01 (MS).

References (114)

  • M.E. Brunkow et al.

    Bone dysplasia sclerosteosis results from loss of the SOST gene product, a novel cystine knot-containing protein

    Am. J. Hum. Genet.

    (2001)
  • T.D. Camenisch et al.

    Temporal and distinct TGFβ ligand requirements during mouse and avian endocardial cushion morphogenesis

    Dev. Biol.

    (2002)
  • O.A. Agapova et al.

    Ligand trap for the activin type IIA receptor protects against vascular disease and renal fibrosis in mice with chronic kidney disease

    Kidney Int.

    (2016)
  • H. Kurosu et al.

    Regulation of fibroblast growth factor-23 signaling by Klotho

    J. Biol. Chem.

    (2006)
  • M.-C. Hu et al.

    Klotho deficiency is an early biomarker of renal ischemia-reperfusion injury and its replacement is protective

    Kidney Int.

    (2010)
  • A. Grabner et al.

    Activation of cardiac fibroblast growth factor receptor 4 causes left ventricular hypertrophy

    Cell Metab.

    (2015)
  • K.A. Hruska et al.

    Hyperphosphatemia of chronic kidney disease

    Kidney Int.

    (2008)
  • E. Moallem et al.

    RNA-protein binding and post-transcriptional regulation of parathyroid hormone gene expression by calcium and phosphate

    J. Biol. Chem.

    (1998)
  • T. Larsson et al.

    Circulating concentration of FGF-23 increases as renal function declines in patients with chronic kidney disease, but does not change in response to variation in phosphate intake in healthy volunteers

    Kidney Int.

    (2003)
  • J. Cunningham et al.

    Osteoporosis in chronic kidney disease

    Am. J. Kidney Dis.

    (2004)
  • M. Rix et al.

    Bone mineral density and biochemical markers of bone turnover in patients with predialysis chronic renal failure

    Kidney Int.

    (1999)
  • M. Coco et al.

    Increased incidence of hip fractures in dialysis patients with low serum parathyroid hormone

    Am. J. Kidney Dis.

    (2000)
  • T. Isakova et al.

    Fibroblast growth factor 23 is elevated before parathyroid hormone and phosphate in chronic kidney disease

    Kidney Int.

    (2011)
  • M.J. Sarnak et al.

    Kidney disease as a risk factor for development of cardiovascular disease

    Circulation

    (2003)
  • A.S. Go et al.

    Chronic kidney disease and the risks of death, cardiovascular events, and hospitalization

    New Engl. J. Med.

    (2004)
  • M. Tonelli et al.

    Chronic kidney disease and mortality risk: a systematic review

    J. Am. Soc. Nephrol.

    (2006)
  • R. Vanholder et al.

    Chronic kidney disease as cause of cardiovascular morbidity and mortality

    Nephrol. Dial. Transplant.

    (2005)
  • J. Blacher et al.

    Arterial calcifications, arterial stiffness, and cardiovascular risk in end-stage renal disease

    Hypertension

    (2001)
  • O.M. Gutierrez et al.

    Fibroblast growth factor 23 and mortality among patients undergoing hemodialysis

    New Engl. J. Med.

    (2008)
  • R. Dhingra et al.

    Relations of serum phosphorus and calcium levels to the incidence of cardiovascular disease in the community

    Arch. Intern. Med.

    (2007)
  • K. Matsushita et al.

    Subclinical atherosclerosis measures for cardiovascular prediction in CKD

    J. Am. Soc. Nephrol.

    (2014)
  • M. Dalal et al.

    Relationship of serum fibroblast growth factor 23 with cardiovascular disease in older community-dwelling women

    Eur. J. Endocrinol.

    (2011)
  • Y. Fang et al.

    Early chronic kidney disease (CKD) stimulates vascular calcification (VC) and decreased bone formation rates prior to positive phosphate balance

    J. Am. Soc. Nephrol.

    (2009)
  • M.C. Hu et al.

    Klotho deficiency causes vascular calcification in chronic kidney disease

    J. Am. Soc. Nephrol.

    (2011)
  • Y. Fang et al.

    CKD-induced wingless/integration1 inhibitors and phosphorus cause the CKD-mineral and bone disorder

    J. Am. Soc. Nephrol.

    (2014)
  • R.B. de Oliveira et al.

    Disturbances of Wnt/β-catenin pathway and energy metabolism in early CKD: effect of phosphate binders

    Nephrol. Dial. Transplant.

    (2013)
  • Y. Sabbagh et al.

    Repression of osteocyte Wnt/β-catenin signaling is an early event in the progression of renal osteodystrophy

    J. Bone Miner. Res.

    (2012)
  • K. Surendran et al.

    A role for Wnt-4 in renal fibrosis

    Am. J. Physiol. Renal Physiol.

    (2002)
  • K. Surendran et al.

    Wnt-dependent-β-catenin signaling is activated after unilateral ureteral obstruction, and recombinant secreted frizzled-related protein 4 alters the progression of renal fibrosis

    J. Am. Soc. Nephrol.

    (2005)
  • Y. Terada et al.

    Expression and function of the developmental gene Wnt-4 during experimental acute renal failure in rats

    J. Am. Soc. Nephrol.

    (2003)
  • T. Kawakami et al.

    Wnt signalling in kidney diseases: dual roles in renal injury and repair

    J. Pathol.

    (2013)
  • S.-L. Lin et al.

    Macrophage Wnt7b is critical for kidney repair and regeneration

    Proc. Natl. Acad. Sci.

    (2010)
  • A. Niida et al.

    DKK1, a negative regulator of Wnt signaling, is a target of the á-catenin/TCF pathway

    Oncogene

    (2004)
  • C. Niehrs

    Function and biological roles of the Dickkopf family of Wnt modulators

    Oncogene

    (2006)
  • T. Reya et al.

    A role for Wnt signalling in self-renewal of haematopoietic stem cells

    Nature

    (2003)
  • M.N. Chamorro et al.

    FGF-20 and DKK1 are transcriptional targets of β-catenin and FGF-20 is implicated in cancer and development

    EMBO J.

    (2004)
  • J.M. Gonzalez-Sancho et al.

    The Wnt antagonist DICKKOPF-1 gene is a downstream target of [beta]-catenin//TCF and is downregulated in human colon cancer

    Oncogene

    (2004)
  • K. Surendran et al.

    A putative Wnt antagonist secreted frizzled-related protein 4 (sFRP4) suppresses the progression of renal fibrosis following unilateral ureteral obstruction

    J. Am. Soc. Nephrol.

    (2004)
  • C. Niehrs

    Function and biological roles of the Dickkopf family of Wnt modulators

    Oncogene

    (2006)
  • D.P. DiRocco et al.

    Wnt4/β  Catenin signaling in medullary kidney myofibroblasts

    J. Am. Soc. Nephrol.

    (2013)
  • Cited by (0)

    View full text