You are on page 1of 17

Reviews

Evolving concepts in the pathogenesis


of uraemic cardiomyopathy
Xiaoliang Wang and Joseph I. Shapiro*
Abstract | The term uraemic cardiomyopathy refers to the cardiac abnormalities that are seen
in patients with chronic kidney disease (CKD). Historically , this term was used to describe
a severe cardiomyopathy that was associated with end-​stage renal disease and characterized
by severe functional abnormalities that could be reversed following renal transplantation. In a
modern context, uraemic cardiomyopathy describes the clinical phenotype of cardiac disease
that accompanies CKD and is perhaps best characterized as diastolic dysfunction seen in
conjunction with left ventricular hypertrophy and fibrosis. A multitude of factors may contribute
to the pathogenesis of uraemic cardiomyopathy , and current treatments only modestly improve
outcomes. In this Review , we focus on evolving concepts regarding the roles of fibroblast growth
factor 23 (FGF23), inflammation and systemic oxidant stress and their interactions with more
established mechanisms such as pressure and volume overload resulting from hypertension and
anaemia, respectively , activation of the renin–angiotensin and sympathetic nervous systems,
activation of the transforming growth factor-​β (TGFβ) pathway , abnormal mineral metabolism
and increased levels of endogenous cardiotonic steroids.

Endogenous cardiotonic Chronic kidney disease (CKD) is a global public health accumulation of type I collagen within the interstitium,
steroids problem that shortens lifespan primarily by increasing in particular around the intramyocardial arteries18.
A class of steroid hormones the risk of cardiovascular disease1. A wide range of car- A number of mechanical and humoral factors that act
with important roles in health diovascular diseases are more common in patients with during the progression of CKD are involved in the devel-
and disease. Endogenous
cardiotonic steroids such as
end-​stage renal disease (ESRD) than in healthy individ- opment of myocardial fibrosis, including haemodynamic
cardenolide and bufadienolide uals, including coronary artery disease, heart failure, overload, insulin resistance, inflammation, transforming
signal through the valvular heart disease, stroke, atrial fibrillation, sudden growth factor-​β (TGFβ), endogenous cardiotonic steroids
Na+/K+-ATPase. cardiac arrest and ventricular arrhythmias and vascular (CTS) and uraemic toxins19.
calcification2–8. The major phenotype of fatal cardiac dis- The pathogenesis of uraemic cardiomyopathy in
ease in patients with ESRD — uraemic cardiomyopathy9 patients with CKD seems to be multifactorial and to
— is characterized by diastolic dysfunction and marked involve mechanisms that are not unique to the CKD
left ventricular hypertrophy (LVH) that is histologically milieu, such as hypertension, volume overload (for
notable for profound fibrosis10. example, as a result of anaemia or hypervolaemia), acti-
LVH (Box 1) is the most commonly diagnosed cardio- vation of the renin–angiotensin–aldosterone system
vascular abnormality and the strongest independent pre- (RAAS), stimulation of the β-​adrenergic system, TGFβ
dictor of cardiovascular mortality in patients with CKD signalling and insulin resistance, as well as CKD-​specific
or ESRD11–13. However, LVH does not itself result in mechanisms and mediators such as hyperparathy-
severe symptoms. Rather, the diastolic dysfunction that roidism, hyperphosphataemia, 1,25-dihydroxy-​vitamin D
seems to accompany if not antedate the development of (vitamin D) deficiency, increased production of fibroblast
LVH produces the symptoms of heart failure. This dias- growth factor 23 (FGF23), Klotho deficiency, circulating
tolic dysfunction seems to be related to a decrease in the uraemic toxins and increases in the concentrations of
active reuptake of calcium into the sarcoplasmic retic- CTS. The Na+/K+-ATPase–reactive oxygen species (ROS)
Joan C. Edwards School of ulum as well as to ventricular fibrosis, which impairs amplification pathway might be involved in both CKD-​
Medicine, Marshall passive relaxation14–16. specific (endogenous CTS) and non-​CKD-specific mech-
University, Huntington,
WV, USA.
A potential explanation for the increased prevalence anisms (hypertension or insulin-​resistance-associated
of left ventricular diastolic dysfunction in patients with oxidative stress)10,20–26. The relationships between the
*e-​mail: shapiroj@
marshall.edu CKD17 is that CKD facilitates the development of myo- different mechanisms and outcomes are still unclear.
https://doi.org/10.1038/ cardial fibrosis, which in turn impairs left ventricular pas- Moreover, treatments that are effective in other car-
s41581-018-0101-8 sive relaxation6. The cause of this fibrosis is an excessive diomyopathic conditions, such as antihypertensive or

Nature Reviews | Nephrology


Reviews

Key points Evidence from in vivo studies that support the view
that mechanical stress is sufficient for cardiac hyper­
• Patients with chronic kidney disease or end-​stage renal disease have an increased risk trophy in response to haemodynamic overload includes
of cardiovascular disease and mortality. the finding that rodents subjected to transverse aortic
• Uraemic cardiomyopathy is characterized by diastolic dysfunction and marked left constriction (resulting in pressure overload) exhibited
ventricular hypertrophy with profound ventricular fibrosis. rapid expression of hypertrophic genes (for example,
• Factors that have been implicated in the development and progression of uraemic fos and jun) and an increase in heart weight:body weight
cardiomyopathy include haemodynamic overload, alterations in mineral metabolism, ratio38. In a pig model of volume overload induced by
insulin resistance, circulating uraemic toxins and endogenous cardiotonic steroids. aortocaval fistula, LVH was observed only 96 h after
• Oxidative stress seems to have a role in all of the putative molecular pathways that surgery39. Moreover, rodents subjected to 5/6 nephrec-
are involved in the pathogenesis of uraemic cardiomyopathy. tomy (a model of renal failure) showed substantial
• Treatments that are effective in other cardiomyopathic conditions such as and persistent hypertension accompanied by LVH40,41.
antihypertensive drugs improve clinical outcomes in uraemic cardiomyopathy only Interestingly, the development of oxidative stress also
modestly at best.
occurs in mechanical stress-induced cardiac hyper­
• The available data suggest that targeting oxidative stress might be a beneficial trophy. Mice under transverse aorta constriction pro-
therapeutic strategy for patients with uraemic cardiomyopathy.
duced significant increases in LVH and ROS production,
both of which were attenuated by antioxidant therapy42.
lipid-​lowering therapy, improve clinical outcomes in Oxidative stress was also apparent within hours of aor-
uraemic cardiomyopathy only modestly at best. tocaval shunt surgery in a porcine model39. The resulting
In this Review, we focus on the molecular mech- volume overload induced multiple apo­ptotic and hyper-
anisms of uraemic cardiomyopathy that are directly trophic responses in cardiomyocytes through the acti-
involved in the induction or progression of cardiac vation of c-​Jun-terminal kinase (JNK) and extracellular
hypertrophy, potential crosstalk between these mecha- signal-​regulated kinase (ERK).
nisms and implications for therapy. Owing to space lim- The cardiac hypertrophy response that is induced
itations, some potentially important mechanisms, such as by mechanical stress is characterized by two main sig-
inflammation and anaemia, are not extensively discussed. nalling pathways: the mitogen-​activated protein kinase
(MAPK) pathway, which is activated by tyrosine and/
Haemodynamic overload or threonine phosphorylation in the cytosol and ulti-
Haemodynamic overload is a well-​known stimulus mately results in regulation of nuclear hypertrophic
for the development of LVH in patients with CKD or factors43–45, and the Janus-​associated kinase (JAK)–signal
ESRD27,28 (Fig. 1). Pressure overload frequently occurs as trans­ducer and activator of transcription (STAT) path-
a result of hypertension and arteriosclerosis, whereas way, which is activated by stretch-​induced binding of
volume overload occurs as a result of the presence of cardiotrophin 1 and leads to phosphorylation of the
anaemia and hypervolaemia. Anaemia and hyperten- receptor–JAK complex (Fig. 1), which in turn promotes
sion are common comorbidities in patients with CKD. the expression of proliferative genes46–48.
Hypertension or hypervolaemia leads to the development
of LVH as a mechanism to maintain vascular wall stress. The renin–angiotensin–aldosterone system
However, continuing left ventricular overload results in The RAAS and its key mediator Ang II have impor-
Pressure overload maladaptive cardiomyocyte hypertrophy and eventually tant roles in signalling pathways that are involved in
Refers to the pathological state fibrosis and cardiomyocyte apoptosis. Whether the initial the pathogenesis of hypertension, CKD and cardiac
of cardiac muscle in which it stimulus for cardiac hypertrophy in CKD is mechanical remodelling49,50. However, Ang II can also induce car-
has to contract against
excessive pressure.
stress or humoral factors such as angiotensin II (Ang II), diac hypertrophy independently of its effects on blood
catecholamines or TGFβ is unclear. pressure51,52 (Fig. 1). These hypertrophic effects of Ang II
Volume overload are mediated by Ang II receptor type 1 (AT1R) on
Refers to the pathological state Mechanical stress cardiomyocytes53,54. Following stimulation by Ang II,
of the heart in which an
Several in vitro studies have demonstrated that mechan- AT1R activates protein kinase C (PKC), which in turn
abnormally large volume of
blood must be pumped. ical stress is sufficient to induce cardiac hypertrophy in activates MAPKs. Activation of the MAPK family mem-
response to substantial haemodynamic overload29–33. For bers, ERK1 and/or ERK2, JNK and p38 kinases, has
Hypervolaemia example, stretching of neonatal cardiac cells grown in been identified in cardiomyocytes involved in cardiac
Also known as fluid overload, serum-​free media stimulated the expression of skeletal remodelling55,56.
hypervolaemia is a pathological
condition in which there is too
α-​actinin, atrial natriuretic peptide (ANP) and myosin The generation of ROS might also have a role in the
much fluid in the blood. heavy chain-​β (β-​MHC) and induced proto-​oncogenes, pathogenesis of Ang-​II-mediated cardiac hypertrophy57,58
Hypervolaemia is common in such as fos, myc and jun, which are associated with car- (Fig. 1). Specifically, Ang II can induce cardiomyocyte
the setting of renal failure. diac hypertrophy, in cardiomyocytes and other cardiac hypertrophy and cardiac fibrosis via the upregulation
cells30,34. Similarly, stretching of cardiomyocytes in vitro of NADPH oxidase 4 (NOX4)59. In a rat renovascular
Aortocaval fistula
A surgically created resulted in increased protein synthesis and expression model of cardiac hypertrophy, blocking Ang II signal-
arteriovenous fistula between of proto-​oncogenes without involvement of growth ling by losartan attenuated LVH through downregulation
the abdominal aorta and factors and hormones30–32. Mechanical stretch can also of oxidative stress59. In another study, Ang II infusion
inferior vena cava, distal to the induce cardiomyocytes to release Ang II, which acts into wild-​type mice increased cardiac NOX4 expression
origin of the renal arteries.
Aortocaval fistula is used as an
as a mediator of the stretch-​induced hypertrophic and induced LVH and fibrosis, whereas in mice with
experimental model of volume response35–37. The role of Ang II in the pathogenesis of cardiac-​specific overexpression of NOX4, administra-
overload. cardiac hypertrophy is discussed further below. tion of Ang II resulted in exaggerated cardiac fibrosis

www.nature.com/nrneph
Reviews

Box 1 | evaluation of left ventricular hypertrophy exerts its effects by binding to and activating adrenergic
receptors. Consistent evidence indicates that high SNS
Evaluation of the left ventricle involves the quantification of ventricular size, wall activity, as measured by plasma noradrenaline levels,
thickness and function. Echocardiography is the most commonly used method to contributes to morbidity and mortality in a variety of
determine ventricle function because of its noninvasive nature, general applicability
cardiovascular diseases, including chronic heart failure73.
and fairly low cost310,311. Several echocardiographic methods can reproducibly measure
Patients with CKD have elevated levels of plasma
left ventricle mass (LVM), including 2D-​guided M-​mode, 2D linear measurement and 3D
echocardiography311,312. noradrenaline74, and noradrenaline infusion has been
LVM varies with gender, age and body size. However, the LVM index (LVMI), which is shown to induce LVH independently of changes in blood
calculated by dividing LVM by the body surface area, enables comparisons to be made pressure in animal models70,75.
between patients with different body sizes312. The American Society of Echocardiography Increased concentrations of catecholamines such
(ASE) guidelines state that LVMI >115 g/m2 in men and >95 g/m2 in women are diagnostic as noradrenaline in patients with CKD likely induce
of left ventricular hypertrophy (LVH) in patients with end-​stage renal disease312. Changes sustained activation of β-​adrenergic receptors (β-​ARs),
in left ventricle geometry, such as posterior and septal wall thickness <1.0 cm, also which may contribute to the development of the urae-
suggest hypertrophy. The ventricular relative wall thickness (RWT; defined as either mic cardiomyopathy phenotype76 (Fig. 1). Activation
posterior wall thickness multiplied by 2 or the sum of the septal and posterior wall
of β-​AR in the mouse heart has been shown to lead
thicknesses divided by the left ventricle diastolic dimension) can be used to further
to phosphorylation of ERK1 and ERK2 and nuclear
characterize changes in left ventricle geometry as concentric or eccentric. An RWT >0.42
together with an elevated LVMI suggest concentric LVH, whereas an RWT <0.42 in the accumulation of ERK, which ultimately results in car-
setting of increased LVMI indicates eccentric hypertrophy311,312. diomyocyte hypertrophy77. Overexpression of β-​AR in
The ASE guidelines state that Doppler echocardiographic measurements including the mouse heart also results in cardiomyocyte hyper-
the annular eʹ velocity, average E/eʹ ratio (defined as the mitral valve (MV) E velocity trophy and progressive heart failure72. These findings
divided by the mitral annular eʹ velocity), left ventricle volume index (defined as the MV indicate that increases in circulating catecholamines
E velocity divided by the A-​wave velocity) and peak tricuspid regurgitation (TR) velocity and increased expression of β-​ARs can produce similar
can be used to assess diastolic function15. However, none of these clinical pathological phenotypes.
measurements are perfect, and the clinical assessment of diastolic function using Interestingly, cardiac β-​AR signalling also has a role
echocardiography remains challenging313.
in the generation of intracellular ROS78 (Fig. 1). In mice,
overexpression of β-​AR led to increased generation of
and hypertrophy together with the expression of cardiac NOX-​derived ROS, which activated the p38 MAPK
fetal genes and activation of the NF-​κB signalling path- pathway and induced the expression of proliferative and
way60. Moreover, treatment of Ang-​II-infused mice profibrotic genes72. Furthermore, in rabbit ventricular
that overexpress NOX4 with a NOX4 inhibitor abol- myocytes β-AR stimulation increased the production
ished the increase in oxidative stress and attenuated of mitochondrial ROS, which resulted in oxidation of
cardiac remodelling60. the ryanodine receptor (which has a role in calcium
Our understanding of the RAAS has deepened in the release from the endoplasmic reticulum) and impaired
past decade with the identification of novel angiotensin calcium cycling79.
peptides61,62. Angioprotectin is an endogenous octapep-
tide that is generated enzymatically from Ang II and has Transforming growth factor-​β
been identified in the plasma of healthy people and Pressure-​overload-induced cardiac hypertrophy is
patients with ESRD63. Angioprotectin antagonizes the accompanied by increases in the expression of TGFβ and
contractile actions of Ang II in the isolated aortic ring the deposition of extracellular matrix proteins including
through its action on the Mas receptor63. Another novel fibronectin and collagen80,81. Additionally, TGFβ has been
component of the RAAS, alamandine, was identified in increasingly recognized as an important contributor to the
2013 (ref.64). Alamandine is a heptapeptide that is gen- progression of CKD82. TGFβ is a multifunctional cytokine
erated from decarboxylation of Ang (1–7) and induces and important profibrotic factor that is also upregulated in
vasodilatation and antihypertensive and antihyper- dilated cardiomyopathy and pressure-​overloaded human
trophic effects in spontaneously hypertensive rats or heart83,84. Several investigations have demonstrated that
isoprenaline-​treated rats64,65. the fibrogenic and hypertrophic actions of TGFβ may be
involved in the pathogenesis of cardiomyopathic condi-
The adrenergic system tions. In the pressure-​overloaded heart, administration
Overactivity of the sympathetic nervous system (SNS) is of anti-​TGFβ antibody prevented collagen accumulation
a common feature of CKD that might contribute to the and attenuated diastolic dysfunction85. Mechanistically,
increased prevalence of cardiovascular diseases in this upregulation of TGFβ in CKD induces the expression of
population66,67. Moreover, epidemiological studies have fibrosis genes such as collagen I and II in cardiac tissue
reported that SNS activity is associated with cardiovascular via activation of the serine/threonine kinase receptor and
mortality in patients with CKD68–70. The mechanisms that phosphorylation of SMAD85.
underlie this association can be characterized into indirect Interestingly, TGFβ-​mediated cardiac fibrosis may
and direct effects of SNS overactivation on the heart. interact with other signalling pathways. Ang-​II-induced
Hypertension induced by α-​adrenergic vasoconstric- induction of TGFβ in cultured cardiomyocytes was
tion indirectly causes LVH71, and SNS overactivation is reduced in the presence of an antioxidant or NOX
associated with the development and progression of inhibitor, indicating a role for ROS in TGFβ signalling86.
chronic heart failure67,72. Noradrenaline is an organic Our laboratory found that stimulation of rat cardiac
chemical in the catecholamine family that functions fibroblasts with CTS led to increases in collagen pro-
in the body as a hormone and neurotransmitter and duction but had no effect on the expression of TGFβ or

Nature Reviews | Nephrology


Reviews

↑ Mechanical stress ↑ Ang II ↑ Catecholamines ↑ TGFβ patients with ESRD have confirmed a positive association
Cytokines between high PTH levels and cardiovascular events93.
Integrin gp130 AT1R NOX β-AR TGFβR Secondary hyperparathyroidism in CKD is generally
thought to be induced by phosphate accumulation.
However, the role of FGF23 in the regulation of PTH
levels is unclear. Increases in FGF23 levels can inhibit
1α-​hydroxylase, which results in a reduction in the lev-
Gαq Gαs els of vitamin D94. As vitamin D regulates the response
of the parathyroid gland to extracellular calcium con-
SMAD2 centrations, this mechanism might result in indirect
MAPKs JAK PLC ROS PKA and SMAD3 stimulation of PTH production. Conversely, treatment
of parathyroid cells with FGF23 has been shown to
reduce PTH mRNA levels and protein secretion95, thus
STAT InsP3 DAG opposing the indirect effect of FGF23.
In cardiomyocytes, PTH binding activates the
G protein-coupled receptor (GPCR) PTH type 1 recep-
Ca2+ PKC MAPKs Ca2+
tor (PTH1R), leading to activation of adenylyl cyclase
and subsequent calcium influx into cardiac cells96 (Fig. 2).
Calcineurin This calcium influx is thought to trigger activation of
the phospholipase C (PLC)–PKC pathway. The down-
NFAT stream effects of PKC on gene expression and cell prolif-
eration then result in myocardial hypertrophy96–98. PTH
might also indirectly cause cardiovascular disease via its
Cardiac hypertrophy Cardiac fibrosis
effects on blood vessels. Although the exact mechanism
Fig. 1 | Molecular mechanisms of haemodynamic overload in uraemic cardiomyopathy. is unclear, one hypothesis is that increased PTH levels
In response to kidney-​injury-induced haemodynamic overload, cardiac myocytes are cause dysfunction of the vascular endothelium, which
subjected to mechanical stress, which activates integrins and leads to the induction of in turn triggers the secretion of collagen and β1 integ-
downstream hypertrophic signalling. Haemodynamic overload also causes the release rin from smooth muscle cells and thus leads to vascular
of humoral factors such as angiotensin II (Ang II), transforming growth factor-​β (TGFβ) remodelling and fibrosis99.
and catecholamines that stimulate their specific cell membrane receptors. Activation of
these receptors triggers intracellular signalling cascades that lead to the development
of a hypertrophic and fibrotic cardiac phenotype. Crosstalk also occurs between
Hyperphosphataemia
these pathways. β-​AR , β-​adrenergic receptor ; AT1R , angiotensin II receptor type 1; DAG, Regulation of phosphorus excretion by the kidney is
diacylglycerol; Gαs and Gαq, Gs and Gq α-​subunits, respectively ; gp130, glyco­protein 130; critical for maintaining phosphorus homeostasis; loss of
InsP3, inositol trisphosphate; JAK , Janus-​associated kinase; MAPK , mitogen-activated this homeostasis due to excretion failure in CKD results
protein kinase; NFAT, nuclear target of activated T cells; NOX, NADPH oxidase; in hyperphosphataemia100. Many studies suggest that
PKA , protein kinase A ; PKC, protein kinase C; PLC, phospholipase C; ROS, reactive oxygen hyperphosphataemia is an important cardiovascular risk
species; STAT, signal transducer and activator of transcription; TGFβR , TGFβ receptor. factor in patients with CKD101. For example, a study that
included >14,000 patients on dialysis reported a posi-
downstream SMAD proteins87. However, these increases tive correlation between serum phosphate concentration
in collagen production were abrogated by TGFβ receptor and cardiovascular disease102. The mechanisms by which
antagonists. Taken together, upregulation of TGFβ in the serum phosphate could contribute to cardiovascular dis-
setting of either haemodynamic overload or renal failure ease are complex. One hypothesis is that hyperphospha-
seems to have an important role in the pathogenesis of taemia induces vascular calcification and cardiovascular
cardiac fibrotic and hypertrophic remodelling (that is, abnormalities through the type III sodium-​dependent
uraemic cardiomyopathy). phosphate cotransporter 1 (PiT1) in vascular smooth
muscle cells. Hyperphosphataemia leads to increased
Alterations in mineral metabolism uptake of phosphate by vascular smooth muscle cells
Disorders of mineral metabolism in patients with CKD via PiT1 and the resulting increase in intracellular phos-
have implications not only for renal osteodystrophy phate levels induces an osteochondrogenic change103–105.
but also for cardiovascular disease in this population88 Additionally, after entry via PiT1 and/or PiT2, phos-
(Fig. 2). Alterations in the levels of parathyroid hormone phate is able to induce apoptosis in vascular smooth
(PTH), serum phosphate, vitamin D, FGF23 and Klotho muscle cells and endothelial cells106,107. Indirect conse-
might all contribute to the pathogenesis of uraemic quences of elevated phosphate concentrations include
cardiomyopathy. stimulation of the PTH and FGF23 systems108 (Fig. 2).

Secondary Secondary hyperparathyroidism Vitamin D deficiency


hyperparathyroidism In patients with CKD, persistently elevated concentra- Patients with CKD have a high rate of severe vitamin D defi-
Refers to excessive secretion of tions of PTH are associated with vascular calcification, ciency owing to an inability to convert 25-hydroxy-​vitamin D
parathyroid hormone by the myocardial hypertrophy and cardiac dysfunction89–91. into the active form of the hormone, 1,25-dihydroxy-​
parathyroid gland in response
to low serum calcium level and
Hyperparathyroidism is suspected to be an important vitamin D109. Several epidemiological and clinical studies
high phosphorus level in the contributor to cardiovascular mortality in CKD, particu- have reported associations between vitamin D deficiency
setting of renal failure. larly in the advanced stages of the disease92, and studies in and cardiovascular disease in these patients109,110.

www.nature.com/nrneph
Reviews

↑ PTH ↑ Phosphate ↑ FGF23 ↓ Vitamin D ↓ Soluble Klotho

PTH1R–cAMP– PiT-induced FGFR4–PLC– VDR RAAS TRPC6 • RAAS


PKA signalling vascular calcineurin–NFAT signalling activation signalling activation
calcification signalling • SNS activation
• TGFβ
• ROS
• Insulin–IGF1
signalling

Cardiac hypertrophy and fibrosis

Fig. 2 | The role of mineral and bone disorder in uraemic cardiomyopathy. In patients with chronic kidney disease or
end-​stage renal disease, the levels of circulating phosphate and fibroblast growth factor 23 (FGF23) are elevated,
whereas serum levels of 1,25-dihydroxy-​vitamin D (vitamin D) and soluble Klotho are reduced. Alterations in the levels
of all four of these factors might independently contribute to cardiac hypertrophy by directly and/or indirectly
interfering with cardiac remodelling. Direct effects of phosphate are not well understood; however, uptake of
phosphate via the type III sodium–phosphate cotransporter (PiT) might lead to endothelial cell damage and thereby
indirectly induce vascular calcification. Elevated phosphate levels can also lead to hyperparathyroidism. Increased
levels of parathyroid hormone (PTH) cause cardiac hypertrophy via activation of the PTH type 1 receptor (PTH1R) on
cardiomyocytes. FGF23 can directly target the myocardium via binding to FGF receptor 4 (FGFR4) and thereby induce
hypertrophic growth of cardiomyocytes and potentially subsequent damage, including cardiac fibrosis. Soluble Klotho
can directly target the heart and prevent the development of left ventricular hypertrophy via blocking transient
receptor potential cation channel, subfamily C, member 6 (TRPC6) and/or an unknown receptor. Soluble Klotho can
also block the activation of the renin–angiotensin–aldosterone system (RAAS), the sympathetic nervous system (SNS),
transforming growth factor-​β (TGFβ) and the reactive oxygen species (ROS)–insulin–insulin-​like growth factor I (IGF1)
signalling pathway and thereby protect the myocardium from pathological stress-​induced cardiac hypertrophy and
fibrosis. Vitamin D can directly target the heart via the vitamin D receptor (VDR) and downregulate cardiomyocyte
proliferation and hypertrophy. In addition, vitamin D might indirectly protect the heart against RAAS-​induced cardiac
damage, at least in part through the suppression of renal renin expression. As phosphate, vitamin D, FGF23 and soluble
Klotho regulate each other’s expression, they might also be able to amplify each other’s cardiac actions. For example,
FGF23 expression is increased by phosphate and reduced by vitamin D, and the expression of soluble Klotho is
increased by vitamin D. NFAT, nuclear target of activated T cells; PKA , protein kinase A ; PLC, phospholipase C.

The interaction of vitamin D with its receptor full-​length, biologically active form, which can be proteo-
increases the efficiency of intestinal calcium and phos- lytically cleaved into amino-​terminal and carboxy-​terminal
phorus absorption to ~40% and 80%, respectively111. fragments120,121. Binding of FGF23 to the FGF receptor 1
In addition to its role in mineral metabolism, ample (FGFR1)–Klotho co-​receptor complex leads to a decrease
evidence suggests that vitamin D may have a role in car- in the renal tubular reabsorption of phosphate and hence
diovascular disease112. For example, vitamin D may act as a net increase in phosphate excretion122. FGF23 inhibits
an antiproliferative factor in the heart113. Proposed anti- the secretion of PTH and the activation of 1,25-vitamin D3
hypertrophic mechanisms of vitamin D include direct via suppression of 1α-​hydroxylase in the kidney119,122–126.
effects on cardiomyocytes as well as indirect effects In patients with CKD, disordered phosphate metab-
via the RAAS and insulin systems (Fig. 2). Activated olism leads to overproduction of FGF23 (ref.127). Other
vitamin D has been shown to downregulate proliferation factors that are altered in CKD such as vitamin D128,
and hypertrophy in cultured cardiomyocytes114. By con- PTH129, calcium130 and leptin131 might also contribute to
trast, vitamin D receptor-​knockout mice had increased FGF23 regulation. The serum level of FGF23 progres-
levels of renin mRNA and plasma Ang II and developed sively increases as kidney function declines, resulting in
hypertension and cardiac hypertrophy, which could be levels in patients with ESRD that are 1,000-fold higher
attenuated using an angiotensin-​converting enzyme than the normal range127,132–134. Although this enormous
(ACE) inhibitor115. Vitamin D infusion also inhibited elevation in serum FGF23 levels (together with increases
renin mRNA expression in these mice115. In rats sub- in PTH) can initially maintain phosphate homeostasis,
jected to partial nephrectomy, the cardioprotective clinical studies in patients with CKD demonstrate a sig-
effect of vitamin D was accompanied by a substantial nificant association between the level of serum FGF23
reduction in FGF23 production and its downstream and the prevalence of cardiovascular disease, especially
signalling116. The role of the FGF23 system in cardiac cardiac hypertrophy135–140. Moreover, FGF23 is signifi-
hypertrophy is discussed in greater detail below. cantly upregulated in the cardiac tissue of patients with
CKD, suggesting that it functions as a paracrine factor
Overproduction of FGF23 with a role in cardiac remodelling120,141.
FGF23 is produced by osteocytes in response to elevated Detailed analyses of FGF23-induced signalling cas-
levels of calcitriol (the active form of vitamin D3) and has cades in cultured cardiac cells and in animal models sug-
important roles in the regulation of vitamin D and phos- gest an important role of FGF23 in the pathophysiology
phate metabolism117–119. FGF23 is normally present in its of uraemic cardiomyopathy (Fig. 2). For example, FGF23

Nature Reviews | Nephrology


Reviews

has been shown to induce hypertrophic growth of iso- been established. However, in human coronary artery
lated neonatal rat ventricular cardiomyocytes and to endothelial cells, both FGFR-​dependent ROS generation
activate a multitude of genes with roles in hypertrophy, via activation of NOX2 and ROS degradation via SOD2
including α-​actinin, α-​MHC, β-​MHC, ANP and brain are stimulated by FGF23 (ref.152).
natriuretic peptide (BNP)139,142. These effects of FGF23
seem to be mediated by FGFR-​dependent activation Klotho deficiency
of the PLCγ–calcineurin–nuclear target of activated Klotho is a transmembrane protein that acts as a co-​
T cells (NFAT) signalling pathway139,142. Furthermore, receptor for the binding of specific FGFs to their
intramyocardial or intravenous injection of FGF23 in receptors123,153. Three isoforms of Klotho have been
mice induced a left ventricular hypertrophic gene profile identified: αKlotho, βKlotho and γKlotho (which is also
and LVH independent of changes in cardiac contractility known as Klotho/lactase-​phlorizin hydrolase-​related
and blood pressure139. protein)119,154. αKlotho (which we refer to as Klotho
In the 5/6 partial nephrectomy rat model of renal in this Review) is expressed in the kidney and para-
failure, serum FGF23 levels are significantly increased thyroid glands; forms complexes with FGFR1, FGFR3
and administration of FGFR inhibitor has been shown and FGFR4; and is a co-​receptor for FGF23 (ref.153).
to attenuate increases in left ventricular mass, wall thick- βKlotho is expressed in liver and fat, forms complexes
ness and cardiomyocyte size139,143. The FGF23-mediated with FGFR1 and FGFR4 and is a co-​receptor for FGF19
PLCγ–calcineurin–NFAT signalling cascade in rat cardio­ and FGF21 (ref.155). γKlotho is expressed in the eye,
myocytes seems to be dependent on activation of FGFR4 fat and kidney; forms complexes with FGFR1, FGFR2
(refs116,144,145). In mice fed a high-​phosphate diet (which and FGFR4; and is a co-​receptor for FGF19 (ref.156).
induces upregulation of FGF23), FGFR4 knockout pro- A reduction in Klotho expression has been reported
tected against the development of LVH and fibrosis144. in patients with CKD157. This reduction may be due to
Similarly, administration of specific anti-​FGFR4 antibod- either defective renal production of Klotho, vitamin D
ies attenuated the development of cardiomyopathy in rats and erythropoietin158–160 or overproduction of other fac-
subjected to 5/6 partial nephrectomy144. By contrast, trans- tors such as inflammatory cytokines161, ROS162, Ang II163
genic mice that expressed constitutively activated FGFR4 and phosphate164.
developed increases in left ventricle wall thickness, cardio­ Mice with Klotho deficiency spontaneously develop
myocyte size and hypertrophic gene expression as well as premature ageing, vascular calcification, LVH and car-
increased serum levels of FGF23 (ref.144). diac fibrosis164. Klotho-​deficient animals also show a
Clinical studies also support the importance of tenfold increase in the level of FGF23, a fivefold increase
FGF23–FGFR4–calcineurin–NFAT signalling in the in vitamin D and cardiac overexpression of hyper-
development of uraemia141,146,147. A case-​c ontrolled trophic genes139,164,165. However, the mechanism by which
study in patients with childhood-​onset ESRD reported Klotho deficiency might contribute to the development
that cardiac FGF23 and FGFR4 were highly expressed in of uraemic cardiomyopathy remains unclear139,165,166.
heart tissue samples and that this increased expression One potential mechanism is via the loss of its antioxi-
was positively associated with the presence of LVH141. dative effects, as mice that overexpress Klotho have an
FGF23 levels are also highly associated with cardio- extended lifespan167 and show a reduction in systemic
vascular diseases in genetic disorders such as X-​linked oxidative stress168,169. As membrane-​bound Klotho is not
hypophosphataemia148. directly expressed in the heart139,165, its cardioprotective
Despite the evidence discussed above, some findings effects must either be indirectly related to its effects in
suggest that FGF23 is not an independent risk factor for other target organs or be mediated by soluble Klotho.
uraemic cardiomyopathy. Instead of a protective effect in Soluble Klotho arises from alternative splicing170,171
CKD, FGF23-null mice showed severe growth retarda- or proteolytic cleavage171 of full-​length Klotho. Although
tion and died by the age of 12 weeks126. Administration soluble Klotho is mainly produced in the kidney, it
of an FGF23-blocking antibody resulted in severe vas- seems to target a variety of cell types, including vascular
cular calcification in CKD rats on a high-​phosphate smooth muscle cells, cardiomyocytes and endothelial
diet149. A 2018 study reported that in the absence of cells, and to protect against inflammation, fibrosis, apop-
renal impairment, the presence of high levels of circu- tosis and oxidative stress169,172–174. In patients with CKD,
lating FGF23 was not sufficient to cause cardiovascular levels of soluble Klotho were reduced and correlated
disease150. Moreover, the non-​calcium-based phosphate positively with estimated glomerular filtration rate175.
binder sevelamer carbonate significantly reduced serum Several experimental studies suggest that soluble
FGF23 levels but failed to improve LVH or cardiac func- Klotho could directly protect against LVH and cardiac
tion in patients with CKD151. Clearly, the molecular fibrosis (Fig. 2). Soluble Klotho blunted hypertrophic
mechanisms by which FGF23 affects cardiac structure effects induced by TGFβ and Ang II in rat cardiomyo-
and function are not yet fully understood. It is likely that cytes164,176 and attenuated Ang II and TGFβ-​mediated
in addition to its direct cardiac effects, systemic changes activation of cardiac fibroblasts in vitro164. Moreover,
induced by FGF23, such as inflammation and hypophos- treatment of mice with soluble Klotho ameliorated
phataemia, contribute to cardiac injury, suggesting that indoxyl-​sulfate-induced cardiomyocyte hypertrophy
CKD-​associated high circulating FGF23 levels can have by blocking ROS generation and inhibiting the p38 and
adverse effects on the heart via several mechanisms that MAPK pathway176. In normal mice, a high-​phosphate diet
might act synergistically. Whether a direct link exists led to a reduction in the serum levels of soluble Klotho,
between FGF23 and cardiac ROS generation has not which were strongly associated with LVH and cardiac

www.nature.com/nrneph
Reviews

fibrosis164. By contrast, intraperitoneal administration of The role of the insulin–PI3K–AKT pathway in the
soluble Klotho protected against pathological changes in uraemic state is not fully understood (Fig. 3). However,
cardiac function, morphology and biochemistry in mice uraemia causes increases in AKT signalling that have
treated with isoprenaline177,178. Although the specific been implicated in the functional, metabolic and
receptor of soluble Klotho has not yet been identified, morphological changes that are seen in uraemic cardio­
soluble Klotho has been reported to inhibit transient myopathy199–201. A study that involved modulation of
receptor potential cation channel, subfamily C, member 6 AKT expression in mice demonstrated that insulin-​
(TRPC6)179. In isolated cardiac myocytes that overex- mediated cardiac hypertrophy predominantly occurs
pressed TRPC6, soluble Klotho inhibited the cell surface via activation of AKT1 rather than AKT2 (refs202,203).
expression of TRPC6 and TRPC6 currents177. Mice with In the uraemic rat model, perturbations in the level
cardiac-​specific TRPC6 overexpression spontaneously of both total and phosphorylated AKT were observed
developed cardiac hypertrophy and remodelling, which when compared with normal controls204. A clinical study
could be prevented by overexpression of Klotho177. reported that AKT was activated in the hearts of patients
Several studies suggest that the protective effects of with heart failure but not in those of patients with com-
soluble Klotho are mediated through the attenuation pensated cardiac hypertrophy or healthy individuals;
of oxidative stress. In a mouse model of CKD, treat- however, whether there is a causal relationship between
ment with exogenous soluble Klotho reduced oxidative AKT activity and heart failure is unclear205.
stress in myocardial tissue through inhibition of NOX176. The importance of persistent AKT signalling in the
Similarly, Klotho attenuated Ang-​II-mediated NOX2 pathogenesis of cardiac hypertrophy was demonstrated
expression, superoxide production, oxidative damage by a study in adult mice that showed a clear transi-
and apoptosis in cultured aortic smooth muscle cells via tion from physiological to pathological hypertrophy
activation of the cAMP–protein kinase A pathway180. with short-​term (2 weeks) versus long-​term (6 weeks)
Administration of Klotho also attenuated isoprenaline-​ induction of activated AKT1 (ref.206). Interestingly, this
induced cardiac remodelling and ROS generation in a
mouse model178. However, oxidative stress was shown Insulin ↑ Insulin
to inhibit Klotho production in this study. resistance

Insulin resistance Insulin receptor


Insulin resistance is one of the most common and earli-
est metabolic alterations that is identified in patients with
CKD181. This resistance becomes more pronounced as PtdInsP2 PtdInsP3
renal function declines182. Moreover, ESRD (glomerular Ras IRS PDK
filtration rate <15 ml/min/1.73 m2) is associated with Raf PI3K
impaired insulin clearance, which further exacerbates MAPK AKT
hyperinsulinaemia183.
The aetiology of insulin resistance in CKD is multi- Cardiac hypertrophy and fibrosis
factorial and involves risk factors that are associated with
renal failure, including metabolic acidosis, inflamma- Fig. 3 | The role of insulin resistance in uraemic
tion, oxidative stress, microbial toxins, uraemic toxins, cardiomyopathy. Insulin resistance and hyperinsulinaemia
anaemia and vitamin D deficiency184–190. Clinical stud- are early metabolic alterations that occur in patients with
ies have demonstrated that insulin resistance is highly renal insufficiency. The aetiology of insulin resistance in
associated with cardiovascular disease in patients with this population is multifactorial and risk factors include
CKD and is an independent predictor of cardiovascular inflammation, oxidative stress, uraemic toxins,
mortality in patients with ESRD22,191,192. 1,25-dihydroxy-​vitamin D (vitamin D) deficiency , metabolic
Insulin resistance is associated with altered insulin– acidosis, anaemia, microbial toxins and reduced insulin
PI3K–AKT signal transduction within the pathway that clearance. Upon binding to insulin, the insulin receptor
undergoes autophosphorylation, which increases its
regulates insulin-​mediated glucose metabolism, lipid
tyrosine kinase activity. Tyrosine phosphorylation and
metabolism, protein synthesis and cell growth193,194. activation of the docking proteins insulin receptor
Protein kinase B (AKT) is a key effector of insulin-​ substrates 1 and 2 (IRS1 and IRS2, respectively) engage
mediated signal transduction. Three isoforms of AKT regulatory subunits of phosphatidylinositol 3-kinase (PI3K)
have been identified in mammals: AKT1 and AKT2 are that generate phosphatidylinositol-3,4,5-trisphosphate
predominately expressed in the heart195,196 and AKT3 (PtdIns(3,4,5)P3) from phosphatidylinositol-3,4-bisphos-
is highly expressed in the central nervous system197. phate (PtdIns(3,4)P2). Protein kinase B (AKT) is then
Physiologically, stimulation of phosphoinositide 3-kinase recruited to the plasma membrane by PtdIns(3,4,5)P3,
(PI3K) through insulin leads to the generation of phos- enabling phosphorylation by phosphoinositide-​dependent
phatidylinositol-3,4,5-trisphosphate (PtdIns(3,4,5)P3) kinase (PDK). Activated AKT phosphorylates multiple
targets including vascular endothelial growth factor
and the recruitment of AKT to the plasma mem-
(VEGF) and angiotensin II (Ang II), which ultimately leads
brane197. AKT is then phosphorylated and activated by to the development of cardiac hypertrophy and fibrosis.
phosphoinositide-​dependent kinase (PDK). The con- Insulin might also regulate cell growth via activation of
sequence of AKT activation is dependent on the iso- the Ras (small GTPase)–mitogen-​activated protein
form as well as the route (physiological or pathological) kinase (MAPK) pathway. Raf, RAF proto-​oncogene
and duration of stimulation198. serine/threonine-​protein kinase.

Nature Reviews | Nephrology


Reviews

transition occurred in association with a decrease in capil- these toxins (for example, indoxyl sulfate and p-​cresyl
lary density. Physiological cardiac hypertrophy is accom- sulfate) are independent predictors of overall mor-
panied by increased coronary angiogenesis to maintain tality and cardiovascular outcomes in patients with
capillary density207. However, in CKD models, coronary ESRD227. In addition, uraemic toxins, such as indoxyl
angiogenesis fails to keep pace with cardiac hypertrophy, sulfate and p-​cresyl sulfate, have been shown to produce
resulting in reduced capillary density208,209. Coronary pro-​hypertrophic and pro-​inflammatory effects and to
angiogenesis is mediated by vascular endothelial growth increase arrhythmias in cardiac tissues by increasing
factor (VEGF) and Ang II, both of which are upregulated oxidative stress228–230.
by the insulin–PI3K–AKT pathway. Conversely, chronic The accumulation of asymmetrical dimethylarginine
overexpression of AKT1 leads to pathological hypertro- (ADMA) is a well-​defined risk factor for LVH in patients
phy and reduced capillary density with downregulation with CKD231. ADMA is a small, water-​soluble uraemic
of VEGF and Ang II (ref.206). Additional studies suggest toxin that results from the degradation of methylated
that signalling through the PI3K–AKT–mechanistic tar- proteins. As ADMA is an endogenous competitive inhib-
get of rapamycin (mTOR) pathway is involved in a mouse itor of nitric oxide synthase (NOS), it may cause LVH
model of CKD210 and results in the chronic overexpres- and fibrosis through accentuation of oxidative stress in
sion of AKT1, ultimately leading to the cardiac fibrosis cardiomyocytes231. A study that included 189 patients
that is associated with LVH196,211. on haemodialysis reported that the level of ADMA was
Our understanding of the role that insulin resist- significantly associated with the degree of concentric
ance plays in the development of oxidative stress is LVH232. In addition, endothelial dysfunction was shown
incomplete. Numerous studies have shown that patients to be strongly and independently associated with ADMA
with CKD and insulin resistance have higher levels of concentration in patients with atherosclerosis231–233.
oxidative stress than healthy individuals212,213. In three The intestinal tract is an important source of poten-
large, cross-​sectional studies performed in patients on tial uraemic toxins including indoxyl sulfate and p-​cresyl
dialysis, insulin resistance (measured using Homeostatic sulfate25. Indoxyl sulfate is a protein-​bound uraemic
Model Assessment (HOMA-​IR)) was positively associ- toxin that is produced by intestinal bacteria and is nor-
ated with both inflammation and oxidative stress214–216. mally absorbed and metabolized in the liver but accu-
However, the molecular pathophysiology is still unclear. mulates in patients with CKD and is poorly removed by
Elevated insulin concentrations (as seen in CKD) induce dialysis24,234. A growing number of clinical studies sug-
a shift in the activities of PI3K. Under these conditions, gest that serum levels of indoxyl sulfate in these patients
PI3K phosphorylates Rac rather than PtdInsP2, lead- are positively associated with heart failure, ischaemic
ing to an amplification of the activity of NOX4 and heart disease and diastolic dysfunction235–237.
increased oxidative stress217. Moreover, adipocytes are Indoxyl sulfate seems to function as an inducer of
now recognized as a major site for production of inflam- oxidative stress that directly targets cardiomyocytes. In
matory cytokines and a source of oxidative stress. In the cultured neonatal rat cardiomyocytes, indoxyl sulfate
general population, insulin resistance positively cor- induced hypertrophy through inhibition of AMPK–
relates with body fat mass218. In cultured adipocytes, UCP2 signalling and amplification of oxidative stress238.
insulin-​stimulated oxidative stress can be attenuated Similar effects of indoxyl sulfate have been observed in
by adenovirus-​mediated NOX4 deletion219. Conversely, cultured rat cardiomyocytes176. Moreover, in a rat model
evidence suggests that oxidative stress activates a series of CKD, oral administration of a charcoal adsorbent
of signalling pathways that have negative effects on insu- (AST-120) led to reductions in the levels of indoxyl sul-
lin sensitivity220. As mentioned above, overexpression of fate, oxidative stress and TGFβ signalling and to atten-
Klotho extends the lifespan of mice. This effect has been uation of left ventricle fibrosis and LVH230,239. On the
attributed to inhibition of insulin–insulin-​like growth other hand, research has suggested that indoxyl-​sulfate-
factor I (IGF1) signalling as soluble Klotho suppressed induced oxidative stress might be mediated by impaired
autophosphorylation of the insulin receptor and acti- activation of nuclear factor erythroid 2-related factor 2
vation of the PI3K pathway in vitro167 as well as to the (NRF2) and impaired expression of major antioxi-
antioxidative effects of Klotho164. dant signalling pathways240. In cultured kidney cells,
indoxyl sulfate downregulates the expression of haem
Uraemic toxins oxygenase 1 (HO1) and NAD(P)H quinone dehydroge-
Increasing evidence suggests that the uraemic state has nase 1 (NQO1) with attendant accumulation of ROS241.
a critical role in the development and progression of Conversely, in a rat model of CKD, a reduction in the
uraemic cardiomyopathy24,25. Numerous compounds levels of indoxyl sulfate using AST-120 resulted in upreg-
that are excreted or metabolized by the kidney in healthy ulation of NRF2, HO1 and NQO1 with decreases in
individuals accumulate in patients with renal insuf- oxidant stress241.
ficiency25,221–226. These putative uraemic toxins can be As mentioned above, Klotho has been reported to
characterized as small water-​soluble compounds with protect against indoxyl-​sulfate-induced myocardial
molecular mass <500 Da that can be easily removed hypertrophy176. In a mouse model of uraemic cardio­
by haemodialysis; molecules with molecular masses of myopathy, renal Klotho expression was reduced,
500–60,000 Da that can be removed only by haemo- whereas the serum indoxyl sulfate concentration was
dialysis using membranes with large pore sizes; and increased compared with healthy controls176. In addition,
protein-​bound molecules that are difficult to remove infusion of indoxyl sulfate resulted in a greater severity
via haemodialysis (Supplementary Table 1). Some of of LVH in heterozygous Klotho-​deficient mice than in

www.nature.com/nrneph
Reviews

normal mice176. These findings suggest that an imbal- marinobufagenin through a minipump to achieve sim-
ance between the levels of Klotho and indoxyl sulfate ilar plasma levels to those in mice with partial nephrec-
might contribute to the development of LVH and fibrosis tomy produced a similar cardiac phenotype to that seen
in CKD. in the partially nephrectomized animals14,87. We also
Upregulation of inflammasomes has also been found increases in oxidative stress and hypertrophic
reported in a mouse model of indoxyI-​sulfate-induced gene expression in partial nephrectomy and marinob-
uraemic cardiomyopathy242, and clinical studies have ufagenin infusion models260. Marinobufagenin derived
demonstrated increased levels of circulating NOD-, from cardiac tissues activated fibroblasts, and this effect
LRR- and pyrin domain-​containing 3 (NLRP3) in a could be abrogated by administration of a nonspe-
variety of kidney diseases243. In rat cardiomyocytes, cific tyrosine kinase inhibitor (herbimycin), a specific
treatment with indoxyl sulfate resulted in activation of tyrosine-​protein kinase Src inhibitor (PP2) or nonspe-
the NLRP3 inflammasome–IL-1–IL-18 axis, which leads cific antioxidant (N-​acetylcysteine)87. This observation
to apoptosis through the activation of NF-​κB242,243. suggests a pro-​fibrotic role of endogenous CTS in the
Indoxyl sulfate might also target other cell types and development of uraemic cardiomyopathy. Neutralization
indirectly cause cardiac hypertrophy and remodelling. of CTS by passive immunization and blockade of CTS
For example, in vitro studies showed that indoxyl sulfate binding to the Na+/K+-ATPase using spironolactone
reduces the viability of human vascular endothelial cells (which acts as a competitive inhibitor of CTS binding)
via the activation of NOX228. By contrast, indoxyl sulfate or rapamycin (which blocks mTOR) were effective in
has been reported to promote the proliferation of vas- blocking cardiac hypertrophy and fibrosis induced by
cular smooth muscle cells by inducing oxidative stress partial nephrectomy261–263.
in a concentration-​dependent manner244. Similarly, Endogenous CTS have been reported to activate the
indoxyl sulfate was shown to stimulate the proliferation Na+/K+-ATPase-​mediated signalling cascade through
of rat vascular smooth muscle cells via the activation of protein tyrosine phosphorylation and to ultimately
MAPK245. Our laboratory has found that indoxyl sulfate alter gene expression without affecting cellular Na+/K+-
and other putative uraemic toxins induce adipocytes ATPase pumping capacity264,265 (Fig. 4). This observation
to produce cytokines (for example, TNF and IL-6) that suggests a receptor-​like function of Na+/K+-ATPase.
could potentially cause adverse cardiac changes such Similar to cytokine receptors and GPCRs, Na +/K+-
as LVH and cardiac fibrosis246–248. In addition, clinical ATPase does not have intrinsic kinase activity; therefore,
evidence suggests that increased adiposity may amplify binding to a non-​receptor tyrosine kinase is required to
the oxidative stress and inflammation that accompanies enable tyrosine phosphorylation of the Na+/K+-ATPase
moderate to severe CKD249,250. in response to the binding of CTS266. The receptor
Similar to indoxyl sulfate, p-​cresyl sulfate is albumin-​ function of α1 Na+/K+-ATPase requires Src-​mediated
bound and its plasma concentration is determined transactivation of epidermal growth factor receptor
mainly by renal excretion251. In patients with CKD, and subsequent recruitment and assembly of signalling
impaired renal function and reductions in albumin con- proteins266,267. Binding of ouabain to the Na+/K+-ATPase
centrations lead to high concentrations of free p-​cresyl releases the Src kinase domain from a binding site in
sulfate. An increasing body of clinical evidence demon- the α1 Na+/K+-ATPase amino domain and disinhibits
strates that serum concentration of p-​cresyl sulfate is (or activates) this Src266. Activation of Src results in
a predictor of cardiovascular disease and mortality in transactivation of multiple downstream effectors such
patients with CKD25,252. Mechanistically, the cardiac tox- as ERK1, ERK2, mTOR and MAPK268,269.
icity of p-​cresyl sulfate may be related to the induction A study using transgenic mice that expressed either
of ROS generation. In a mouse model of CKD, increase ouabain-​s ensitive or ouabain-​resistant isoforms of
in the levels of p-​cresyl sulfate promoted cardiac apopto- Na+/K+-ATPase reported that the ouabain-​sensitive mice
sis and diastolic dysfunction, which were ameliorated by developed substantially more severe cardiac dysfunction
the concomitant administration of a NOX inhibitor253. in response to left ventricle pressure overload than did
the ouabain-​resistant mice270. Moreover, knockdown of
Endogenous cardiotonic steroids α1 Na+/K+-ATPase resulted in significant increases in
The concentrations of endogenous CTS are elevated in cardiac cell death and in the abundance of cardiac pro-
various diseases such as CKD, ESRD, heart failure and genitor cells in mice following partial nephrectomy271.
hypertension and are associated with clinical meas- These studies provide convincing support for a role
urements such as LVH and diastolic dysfunction254–258. of endogenous CTS-​mediated Na+/K+-ATPase signal
Several laboratories have reported significantly elevated transduction in uraemic cardiomyopathy.
concentrations of endogenous ouabain, marinobufa-
genin and telocinobufagin in patients with CKD248 The Na+/K+-ATPase–Src–ROS amplification loop
and ESRD256 and in animal models of uraemic cardio­ A link between Na+/K+-ATPase-​mediated signal trans-
Inflammasomes myopathy, suggesting an important role of these CTS in duction and ROS generation was first identified in an
A multiprotein intracellular the development of this phenotype26,258,259. early study that showed that ROS generation was required
complex that detects We found that experimental CKD induced by partial for ouabain-​induced hypertrophy of cardiomyocytes
pathogenic microorganisms nephrectomy resulted in increased levels of circulating in vitro and that this hypertrophy could be attenuated
and activates inflammatory
responses via the activation of
marinobufagenin and that active immunization against by antioxidants such as N-​acetylcysteine or vitamin E272.
pro-​inflammatory cytokines marinobufagenin attenuated the development of exper- We subsequently showed that ouabain could stimulate
such as IL-1β and IL-18. imental uraemic cardiomyopathy87. Administration of ROS generation in a Ras (small GTPase)-dependent

Nature Reviews | Nephrology


Reviews

↑ Endogenous (oxLDL)–CD36–Na+/K+-ATPase–Lyn kinase signalling


cardiotonic steroids complex in macrophages that enables oxLDL to activate
Na+/K+-ATPase EGFR downstream signalling277. The CD36–Na+/K+-ATPase
pro-​inflammatory signalling loop has also been reported
to be involved in the development of chronic inflam-
P P mation, oxidative stress and fibrosis in the kidney278.
P P Our animal studies utilizing pNaKtide (an antagonist
Src
FAK of Na+/K+-ATPase–Src signalling complex) support a
SOS direct role of the Na+/K+-ATPase–Src–ROS amplifica-
Ras tion loop in the development of uraemic cardiomyopa-
↑ ROS thy275. We found that in partially nephrectomized mice,
treatment with pNaKtide reduced systemic oxidative
ERK1, ERK2
or MAPK stress and cardiac Na+/K+-ATPase–Src–ROS amplifica-
tion (as evidenced by a decrease in protein carbonylation
and HO1 expression), cardiac hypertrophy and fibro-
Cardiac hypertrophy and fibrosis sis; improved left ventricular diastolic function; and,
somewhat surprisingly, ameliorated anaemia. pNaKtide
Fig. 4 | endogenous cardiotonic steroids and the
Na+/K+-ATpase–Src–roS amplification loop in uraemic also reversed established uraemic cardiomyopathy in a
cardiomyopathy. Patients with chronic kidney disease or dose-​dependent manner275.
end-​stage renal disease have increased levels of circulating
endogenous cardiotonic steroids (for example, ouabain, Crosstalk between signalling pathways
marinobufagenin and telocinobufagin) and reactive oxygen Although each of the aforementioned mechanisms can
species (ROS). Upon binding to endogenous cardiotonic produce some features that are consistent with uraemic
steroids, the Na+/K+-ATPase changes its conformation and cardiomyopathy, it is clear that interactions and cross-
activates proto-​oncogene tyrosine-​protein kinase Src (Src). talk occur between the different signalling pathways. For
Src then transactivates epidermal growth factor receptor example, FGF23-mediated cellular hypertrophy is asso-
(EGFR), which leads to a signal cascade involving focal
ciated with increased production and secretion of Ang II
adhesion kinase (FAK), son-​of-sevenless 1 (SOS1) and Ras
(small GTPase). This cascade results in the generation of by cardiomyocytes279. In human and rat models of kid-
ROS, which in turn activate additional Na+/K+-ATPase–Src ney injury, treatment with the Ang II receptor antagonist
signalling complexes to form a positive feedback loop. valsartan is associated with an increase in the levels of
Elevated levels of ROS lead to downstream activation of soluble Klotho280,281. Indoxyl sulfate increases Ang II sig-
extracellular-​signal-regulated kinase 1 (ERK1), ERK2 and nalling and the levels of this uraemic toxin are strongly
mitogen-​activated protein kinase (MAPK), which induce associated with the development of arteriosclerosis in
cardiac hypertrophy and fibrosis. CKD282 and positively correlate with the levels of FGF23
in advanced CKD283. Moreover, TGFβ-​induced collagen
but intracellular Ca2+-independent manner in cultured production in cardiac fibroblasts and the accumulation
pig kidney cells273. Moreover, an increase in ROS stress of α-​actinin protein in cardiomyocytes are attenuated by
correlated with elevated levels of endogenous CTS in soluble Klotho164. Determining the relative contribution
partially nephrectomized rats87. of each pathway to the development of the uraemic car-
The finding that ROS could modulate the structure diomyopathy phenotype remains challenging. However,
and activity of the α1 Na+/K+-ATPase274 led to studies of as detailed above, oxidative stress has a key role in each
potential interactions between ouabain-​induced ROS gen- of the major mechanisms that induce LVH and cardiac
eration and ROS-​induced Na+/K+-ATPase signalling. ROS fibrosis in CKD (Fig. 5).
and ouabain were found to potentiate each other in the Accumulating data indicate that the major signal-
stimulation of hypertrophic signalling in cultured cardio- ling pathways that are operant in uraemic cardiomyo-
myocytes, leading to the hypothesis that ROS and ouabain pathy involve derangements of cellular redox10,26,41,275,284.
might form a feedforward amplification loop through the For example, Ang II and α-​adrenoceptor agonists all cause
Na+/K+-ATPase–Src signalling complex10,272,273,275 (Fig. 4). increased generation of NOX-​derived ROS through its
In pig kidney cells, we observed that both ouabain and unique GPCR, which modulates specific signalling path-
increases in ROS levels (induced by glucose oxidase) can ways during the development of cardiac hypertrophy285.
induce carbonylation of α1 Na+/K+-ATPase at Pro222 and Excessive oxidative stress is a major cause of endothelial
Thr224 (refs274,276). Moreover, inhibition of this carbony­ dysfunction in CKD286 and results in oxidation of LDL
lation using antioxidants or mutation of Pro222 attenu- cholesterol, which triggers inflammation and progression
ated the activation of protein kinase cascades in response of atherosclerotic vascular disease286.
to ouabain or glucose oxidase.
In view of the well-​characterized role of oxidative Therapeutic approaches
stress in the progression of chronic disease, several As LVH is the most commonly diagnosed cardiovascu-
laboratories have investigated whether the Na+/K+- lar abnormality and an extremely strong, independent
ATPase–Src signalling complex is critical for ROS predictor of cardiovascular mortality in patients with
amplification in disease models. A study using genet- CKD11–13, targeting the mechanisms that are involved in
ically modified mice demonstrated that α1 Na+/K+- the development of uraemic cardiomyopathy is a logi­
ATPase is required for the formation of an oxidized LDL cal approach to potentially improve outcomes in this

www.nature.com/nrneph
Reviews

population. Conventional haemodialysis is the most diabetic nephropathy288. Several clinical studies have
common treatment option for patients with ESRD. attempted to delineate the potential cardiovascular ben-
However, uraemic cardiomyopathy tends to persist and efits of targeting the RAAS in patients with CKD. The
even worsen once haemodialysis is initiated. PROGRESS study reported that treatment with the ACE
inhibitor perindopril reduced the risk of major cardio-
Renin–angiotensin system vascular events by 30% in patients with CKD (n = 1,757)
Targeting the RAAS is an effective antihypertensive during 4 years of follow-​up289. Similarly, the HOPE ran-
strategy287, and treatment with either an ACE inhibi- domized trial, which enrolled 980 patients with CKD,
tor or an angiotensin receptor blocker (ARB) reduces reported that treatment with the ACE inhibitor rami-
the risk of heart failure in patients with proteinuria and pril reduced the incidence of cardiovascular death and

Haemodynamic Hyperphosphataemia Retention ↑ Endogenous Insulin


overload and vitamin D deficiency of uraemic cardiotonic resistance
toxins steroids
Mechanical
stress
↓ Soluble
↑ FGF23 Klotho
FGFR4 Ca2+ ↑ Insulin
↑ TGFβ ↑ Ang II ↑ Catecholamines ↑ PTH ↑ IS
Cytokines
Integrin gp130 TGFβR AT1R β-AR PTH1R NOX Na+/K+-ATPase Insulin
receptor

TRPC6
FAK Src Src IRS1
JAK SMAD2 Gαq Gαs Gαs PLCγ
Ras and IS SOS PI3K
STAT PLC
Raf SMAD3 Ras PtdInsP3
InsP3 and
MEK DAG PKA Raf PDK
ERK1, PKC Glutathione MEK AKT
ERK2, JUN
and MAPK ERK1 and MAPK CaM ERK1
ERK2 and
Calcineurin ERK2
NFAT

ROS

Uraemic
Expression of hypertrophy and fibrosis genes
cardiomyopathy
Nucleus

Fig. 5 | Crosstalk between signalling pathways involved in uraemic generation. Insulin induces ROS generation through the insulin receptor
cardiomyopathy. All of the major mechanisms that are involved in the substrate 1 (IRS1)–Ras (small GTPase)–Raf (RAF proto-​oncogene serine/
pathogenesis of uraemic cardiomyopathy — haemodynamic overload, threonine-​protein kinase) pathway. However, ROS can themselves inhibit
mineral metabolic disorder, insulin resistance and increased levels of the insulin–phosphoinositide 3-kinase (PI3K)–protein kinase B (AKT)
uraemic toxins and endogenous cardiotonic steroids (CTS) — result in the pathway and thereby exacerbate insulin resistance. Accumulation of
generation of reactive oxygen species (ROS). Mechanical stress stimulates endogenous CTS signalling through Na +/K +-ATPase results in the
ROS production through integrin-​mediated Rac–mitogen-​activated generation of ROS, which in turn are capable of initiating further Na+/K+-
protein kinase (MAPK) signalling, whereas activation of G protein-​coupled ATPase–tyrosine-​p rotein kinase Src signalling, forming an oxidant
receptors, such as angiotensin II (Ang II) receptor type 1 (AT1R) and the amplification loop10,272,275,314,315. In addition to their role in regulating some
β-​adrenergic receptor (β-​AR), leads to ROS production via the phospholipase of the mediators of the signalling pathways that are involved in these
C (PLC)–MAPK and protein kinase A (PKA)–Ca2+ signalling pathways, mechanisms, ROS induce the expression of genes that cause cardiac
respectively. Transforming growth factor-​β (TGFβ) stimulates mitochondrial hypertrophy and fibrosis. CaM, calmodulin; DAG, diacylglycerol; ERK ,
oxidative phosphorylation and further generation of ROS. The involvement extracellular-​signal-regulated kinase; FAK , focal adhesion kinase; FGFR4,
of ROS in hyperphosphataemia, hyperparathyroidism and elevated fibroblast growth factor receptor 4; Gαs and Gαq, Gs and Gq α-subunits,
fibroblast growth factor 23 (FGF23)-induced cardiac hypertrophy is not respectively ; gp130, glycoprotein 130; InsP3, inositol trisphosphate; JAK ,
clear. However, all of these mechanisms may produce ROS through Janus-​associated kinase; MEK , mitogen-activated protein kinase kinase;
activation of Ca2+-mediated oxidative stress. Soluble Klotho, on the other NFAT, nuclear target of activated T cells; PDK , phosphoinositide-​dependent
hand, not only directly blocks NADPH oxidase (NOX)-mediated ROS kinase 1; PtdIns(3,4,5)P3, phosphatidylinositol-3,4,5-trisphosphate; PKC,
generation but also inhibits transient receptor potential cation channel, protein kinase C; PTH, parathyroid hormone; SOS, son-​of-sevenless; STAT,
subfamily C, member 6 (TRPC6) and indoxyl sulfate (IS)-mediated ROS signal transducer and activator of transcription; TGFβR , TGFβ receptor.

Nature Reviews | Nephrology


Reviews

myocardial infarction during 3.5–5.5 years of follow-​up290. serum level of FGF23 and seemed to have beneficial
However, no significant cardiovascular benefit of effects on cardiovascular end points in older patients146.
treatment with another ACE inhibitor, fosinopril, was A meta-​analysis that included data from 24 randomized
observed in patients with ESRD (n = 397) in another controlled trials reported that treatment with cinacalcet
well-​conducted study291. lowered the serum levels of PTH, calcium, phosphate
Targeting the RAAS using the mineralocorticoid and FGF23 in patients on dialysis, but this analysis did
receptor blocker spironolactone in addition to an ACE not identify a beneficial effect on cardiovascular out-
inhibitor or an ARB in patients with CKD is associated comes (such as cardiac hypertrophy, ischaemic heart
with increased risk of hyperkalaemia292. This risk is not diseases or cardiac function) or show that cinacalcet
limited to patients with ESRD as indicated by the finding significantly improved all-​cause mortality301. Inhibitors
that hyperkalaemia-​associated mortality in Canadian of FGF23 synthesis, FGFR4 blockers and soluble Klotho
patients with congestive heart failure increased fol- mimetics might be potential novel therapeutic agents for
lowing publication of the RALES study, which led to uraemic cardiomyopathy.
widespread use of spironolactone in addition to other
RAAS therapy293. Insulin resistance
The effectiveness of treatment for insulin resistance in
The adrenergic system patients with CKD is still under debate. In a cohort of
β-Blockers are the most commonly prescribed cardio­ 5,290 patients on dialysis with diabetes, thiazolidinedi-
vascular medications among patients on dialysis294. one treatment was associated with significantly lower
However, data on their utility in improving cardiovascular cardiovascular mortality among insulin-​independent
outcomes in this population are conflicting. but not insulin-​dependent patients302. However, con-
A small randomized trial with 2 years of follow-​up cern exists that rosiglitazone might increase the risk of
reported that carvedilol attenuated pathological cardiac adverse outcomes in patients with diabetes who are on
remodelling and reduced morbidity and mortality dialysis. Among 2,393 patients on long-​term haemo-
in patients with dilated cardiomyopathy on dialysis dialysis who were monitored for a median of 1.1 years
(n = 114)295. In the SENIORS trial, nebivolol therapy in the DOPPS trial, rosiglitazone use was significantly
reduced cardiovascular hospital admission among associated with higher cardiovascular disease and
elderly patients (n = 2,112) with heart failure and mild all-​cause mortality303.
or moderate renal impairment during a follow-​up
period of ~21 months296. In addition, a meta-​analysis Uraemic toxins
that included data from eight trials with a total of 6,949 An orally administered carbon adsorbent, AST-120,
participants identified cardiac benefits of β-​blockers in that can adsorb various uraemic toxins has been
patients with CKD and chronic heart failure297. A rand- approved in Japan, Korea and the Philippines for use
omized controlled trial in patients with LVH on haemo- in delaying the initiation of dialysis and ameliorating
dialysis (n = 200) reported that treatment with atenolol the symptoms of uraemia in patients with progressive
seemed to be superior to ACE inhibitor therapy using CKD. Although a small clinical trial in Japan confirmed
lisinopril in terms of cardiovascular morbidity; how- that AST-120 was effective in slowing the progression
ever, changes in left ventricular mass index were simi- of renal disease304, two randomized placebo-​controlled
lar in the study groups298. By contrast, an observational trials involving 2,035 patients with moderate to severe
study from Canada reported no beneficial effects of CKD did not show a beneficial effect of adding AST-120
β-blockers on cardiovascular outcomes among patients to standard therapy305.
on dialysis aged >66 years299. Similar to RAAS inhibi-
tors, use of β-​blockers is associated with increased risk Oxidative stress
of hyperkalaemia in patients with CKD. Further studies of Increased levels of oxidative stress and inflammation are
β-​blockers in the setting of different stages of CKD are prevalent in patients undergoing haemodialysis and are
certainly indicated. associated with increased cardiovascular mortality. A study
conducted in patients with diabetes (n = 1,161) with ESRD
FGF23 and Klotho undergoing haemodialysis showed that carbonylated albu-
Although the molecular mechanisms by which FGF23 min was strongly associated with 1-year adjusted risk of
and Klotho affect cardiac function in patients with CKD cardiovascular mortality and 4-year risk of congestive
remain unclear, the available evidence strongly suggests heart failure306. A study that included 65 children with
that these factors are involved in the development of CKD showed a positive correlation between serum levels
uraemic cardiomyopathy and might therefore serve as of oxLDL and LVH307. However, the results of studies of
potential therapeutic targets. antioxidative approaches have been disappointing. In a
In a small open-​label single-​arm trial (n = 55), treat- prospective, placebo-​controlled, double-​blind clinical trial
ment with the calcimimetic cinacalcet lowered serum that involved 353 patients on haemodialysis, administra-
FGF23 levels in patients with secondary hyperparath- tion of antioxidant therapy (mixed tocopherols (666 inter-
yroidism on dialysis300. Unfortunately, cardiovascular national units (IU) per day) and α-​lipoic acid (600 mg per
outcomes were not assessed in this study. The EVOLVE day)) did not have a significant effect on biomarkers of
trial, which included 2,602 patients with secondary inflammation or oxidative stress at 6-month follow-​up308.
hyperparathyroidism on haemodialysis, reported that Moreover, a meta-​analysis of 14 studies that investigated
treatment with cinacalcet significantly reduced the the effectiveness of antioxidant agents in patients with

www.nature.com/nrneph
Reviews

diabetic kidney disease found that antioxidant treatment Conclusions


significantly decreased albuminuria but had no beneficial The pathophysiology of uraemic cardiomyopathy
effect on cardiovascular outcomes309. involves multiple interacting mechanisms and current
To date, most clinical antioxidant strategies employ clinical treatment is inadequate. The available evidence
the use of antioxidant or free radical scavengers that strongly suggests that oxidative stress has an important
neutralize oxidants but do not affect the factors pathophysiological role and is involved in a variety
that result in oxidative stress (for example, increased of molecular pathways that lead to the development of
production or impaired detoxification). We propose uraemic cardiomyopathy. To date, the beneficial effects
that the results of targeting the amplification of ROS of antioxidant therapy in patients with CKD in general
generation might be more promising as we have shown and in those with uraemic cardiomyopathy have not
in experimental animals275. The Na+/K+-ATPase–Src– been demonstrated. However, we believe that controlling
ROS signalling amplification loop might be a prom- pathological ROS amplification by targeting the Na+/K+-
ising therapeutic target (pNaKtide) for the prevention ATPase–Src signalling complex or other pathways that
and treatment of uraemic cardiomyopathy and other generate ROS might be promising therapeutic targets
comorbidities that are derived from ROS in CKD. to address uraemic cardiomyopathy and other com-
However, it must be stressed that this approach is still plications that result from increased oxidative stress in
at proof-​of-concept stage, and other strategies that also patients with CKD
once seemed promising have previously failed in the
clinical arena. Published online xx xx xxxx

1. Collins, A. J., Foley, R. N., Gilbertson, D. T. & Chen, S. C. 16. Otsuka, T., Suzuki, M., Yoshikawa, H. & Sugi, K. Left 32. Komuro, I. et al. Stretching cardiac myocytes stimulates
United States Renal Data System public health ventricular diastolic dysfunction in the early stage of protooncogene expression. J. Biol. Chem. 265,
surveillance of chronic kidney disease and chronic kidney disease. J. Cardiol. 54, 199–204 3595–3598 (1990).
end-​stage renal disease. Kidney Int. Suppl. 5, 2–7 (2009). 33. Cooper, G. t., Kent, R. L., Uboh, C. E., Thompson, E. W.
(2015). 17. de Almeida, E. A. et al. Diastolic function in several & Marino, T. A. Hemodynamic versus adrenergic
2. Silverberg, D., Wexler, D., Blum, M., Schwartz, D. & stages of chronic kidney disease in patients with control of cat right ventricular hypertrophy.
Iaina, A. The association between congestive heart autosomal dominant polycystic kidney disease: J. Clin. Invest. 75, 1403–1414 (1985).
failure and chronic renal disease. Curr. Opin. Nephrol. a tissue Doppler imaging study. Kidney Blood 34. Komuro, I., Kurabayashi, M., Takaku, F. & Yazaki, Y.
Hypertens. 13, 163–170 (2004). Press. Res. 30, 234–239 (2007). Expression of cellular oncogenes in the myocardium
3. London, G. M., Pannier, B., Marchais, S. J. 18. Diez, J. Mechanisms of cardiac fibrosis in hypertension. during the developmental stage and pressure-​
& Guerin, A. P. Calcification of the aortic valve in the J. Clin. Hypertens. 9, 546–550 (2007). overloaded hypertrophy of the rat heart. Circ. Res. 62,
dialyzed patient. J. Am. Soc. Nephrol. 11, 778–783 19. Lopez, B., Gonzalez, A., Hermida, N., Laviades, C. & 1075–1079 (1988).
(2000). Diez, J. Myocardial fibrosis in chronic kidney disease: 35. Sadoshima, J., Xu, Y., Slayter, H. S. & Izumo, S.
4. Dad, T. & Weiner, D. E. Stroke and chronic kidney potential benefits of torasemide. Kidney Int. Autocrine release of angiotensin II mediates stretch-​
disease: epidemiology, pathogenesis, and management 74 (Suppl. 111), S19–S23 (2008). induced hypertrophy of cardiac myocytes in vitro.
across kidney disease stages. Semin. Nephrol. 35, 20. Hung, S. C., Lai, Y. S., Kuo, K. L. & Tarng, D. C. Volume Cell 75, 977–984 (1993).
311–322 (2015). overload and adverse outcomes in chronic kidney 36. Tamura, K. et al. Activation of angiotensinogen gene in
5. Kulkarni, N., Gukathasan, N., Sartori, S. & Baber, U. disease: clinical observational and animal studies. cardiac myocytes by angiotensin II and mechanical
Chronic kidney disease and atrial fibrillation: J. Am. Heart Assoc. 4, e001918 (2015). stretch. Am. J. Physiol. 275, R1–R9 (1998).
a contemporary overview. J. Atr. Fibrillation 5, 448 21. Grabner, A. & Faul, C. The role of fibroblast growth 37. Malhotra, R., Sadoshima, J. & Brosius, F. C. 3rd &
(2012). factor 23 and Klotho in uremic cardiomyopathy. Izumo, S. Mechanical stretch and angiotensin II
6. Whitman, I. R., Feldman, H. I. & Deo, R. CKD and Curr. Opin. Nephrol. Hypertens. 25, 314–324 differentially upregulate the renin-​angiotensin system
sudden cardiac death: epidemiology, mechanisms, (2016). in cardiac myocytes in vitro. Circ. Res. 85, 137–146
and therapeutic approaches. J. Am. Soc. Nephrol. 23, 22. Shinohara, K. et al. Insulin resistance as an (1999).
1929–1939 (2012). independent predictor of cardiovascular mortality in 38. Rockman, H. A. et al. Segregation of atrial-​specific and
7. Dennis, V. W. Coronary heart disease in patients with patients with end-​stage renal disease. J. Am. Soc. inducible expression of an atrial natriuretic factor
chronic kidney disease. J. Am. Soc. Nephrol. 16 Nephrol. 13, 1894–1900 (2002). transgene in an in vivo murine model of cardiac
(Suppl. 2), 103–106 (2005). 23. Spoto, B., Pisano, A. & Zoccali, C. Insulin resistance hypertrophy. Proc. Natl Acad. Sci. USA 88,
8. Moe, S. M. & Chen, N. X. Mechanisms of vascular in chronic kidney disease: a systematic review. 8277–8281 (1991).
calcification in chronic kidney disease. J. Am. Am. J. Physiol. Renal Physiol. 311, F1087–F1108 39. Fiorillo, C. et al. Cardiac volume overload rapidly
Soc. Nephrol. 19, 213–216 (2008). (2016). induces oxidative stress-​mediated myocyte apoptosis
9. Khan, N. A. et al. Kidney function and mortality among 24. Hung, S. C., Kuo, K. L., Wu, C. C. & Tarng, D. C. and hypertrophy. Biochim. Biophys. Acta 1741,
patients with left ventricular systolic dysfunction. Indoxyl sulfate: a novel cardiovascular risk factor in 173–182 (2005).
J. Am. Soc. Nephrol. 17, 244–253 (2006). chronic kidney disease. J. Am. Heart Assoc. 6, 40. Kennedy, D. J. et al. Partial nephrectomy as a
10. Wang, X., Liu, J., Drummond, C. A. & Shapiro, J. I. e005022 (2017). model for uremic cardiomyopathy in the mouse.
Sodium potassium adenosine triphosphatase 25. Vanholder, R., Schepers, E., Pletinck, A., Nagler, E. V. Am. J. Physiol. Renal Physiol. 294, F450–F454 (2008).
(Na/K-ATPase) as a therapeutic target for uremic & Glorieux, G. The uremic toxicity of indoxyl sulfate 41. Kennedy, D. J. et al. Central role for the cardiotonic
cardiomyopathy. Expert Opin. Ther. Targets 21, and p-​cresyl sulfate: a systematic review. J. Am. Soc. steroid marinobufagenin in the pathogenesis of
531–541 (2017). Nephrol. 25, 1897–1907 (2014). experimental uremic cardiomyopathy. Hypertension
11. London, G. M. et al. Alterations of left ventricular 26. Kennedy, D. J., Malhotra, D. & Shapiro, J. I. Molecular 47, 488–495 (2006).
hypertrophy in and survival of patients receiving insights into uremic cardiomyopathy: cardiotonic 42. Tsujimoto, I. et al. The antioxidant edaravone
hemodialysis: follow-​up of an interventional study. steroids and Na/K ATPase signaling. Cell. Mol. Biol. attenuates pressure overload-​induced left ventricular
J. Am. Soc. Nephrol. 12, 2759–2767 (2001). 52, 3–14 (2006). hypertrophy. Hypertension 45, 921–926 (2005).
12. Middleton, R. J., Parfrey, P. S. & Foley, R. N. Left 27. Alhaj, E. et al. Uremic cardiomyopathy: 43. Hotamisligil, G. S. & Davis, R. J. Cell signaling and
ventricular hypertrophy in the renal patient. an underdiagnosed disease. Congest. Heart Fail. stress responses. Cold Spring Harb. Perspect. Biol. 8,
J. Am. Soc. Nephrol. 12, 1079–1084 (2001). 19, E40–E45 (2013). a006072 (2016).
13. Zoccali, C. et al. Prognostic value of echocardiographic 28. Zoccali, C. et al. Chronic fluid overload and mortality in 44. Cohen, P. The search for physiological substrates
indicators of left ventricular systolic function in ESRD. J. Am. Soc. Nephrol. 28, 2491–2497 (2017). of MAP and SAP kinases in mammalian cells.
asymptomatic dialysis patients. J. Am. Soc. Nephrol. 29. Ruwhof, C. & van der Laarse, A. Mechanical stress-​ Trends Cell Biol. 7, 353–361 (1997).
15, 1029–1037 (2004). induced cardiac hypertrophy: mechanisms and signal 45. Dingar, D. et al. Effect of pressure overload-​induced
14. Kennedy, D. et al. Effect of chronic renal failure on transduction pathways. Cardiovasc. Res. 47, 23–37 hypertrophy on the expression and localization of p38
cardiac contractile function, calcium cycling, and gene (2000). MAP kinase isoforms in the mouse heart. Cell. Signal.
expression of proteins important for calcium 30. Sadoshima, J., Jahn, L., Takahashi, T., Kulik, T. J. & 22, 1634–1644 (2010).
homeostasis in the rat. J. Am. Soc. Nephrol. 14, Izumo, S. Molecular characterization of the 46. Ihle, J. N. Cytokine receptor signalling. Nature 377,
90–97 (2003). stretch-induced adaptation of cultured cardiac cells. 591–594 (1995).
15. Nagueh, S. F. et al. Recommendations for the An in vitro model of load-​induced cardiac hypertrophy. 47. Schindler, C. & Darnell, J. E. Jr. Transcriptional
evaluation of left ventricular diastolic function by J. Biol. Chem. 267, 10551–10560 (1992). responses to polypeptide ligands: the JAK-​STAT
echocardiography: an update from the American 31. Kira, Y. et al. Effect of long-​term cyclic mechanical load pathway. Annu. Rev. Biochem. 64, 621–651 (1995).
Society of Echocardiography and the European on protein synthesis and morphological changes in 48. Pan, J. et al. Role of angiotensin II in activation of the
Association of Cardiovascular Imaging. Eur. Heart J. cultured myocardial cells from neonatal rat. JAK/STAT pathway induced by acute pressure overload
Cardiovasc. Imaging 17, 1321–1360 (2016). Cardiovasc. Drugs Ther. 8, 251–262 (1994). in the rat heart. Circ. Res. 81, 611–617 (1997).

Nature Reviews | Nephrology


Reviews

49. Lambers Heerspink, H. J., de Borst, M. H., 75. Patel, M. B. et al. Altered function and structure 99. Yao, L. et al. Parathyroid hormone and the risk of
Bakker, S. J. & Navis, G. J. Improving the efficacy of of the heart in dogs with chronic elevation in plasma incident hypertension: the Atherosclerosis Risk in
RAAS blockade in patients with chronic kidney norepinephrine. Circulation 84, 2091–2100 (1991). Communities study. J. Hypertens. 34, 196–203
disease. Nat. Rev. Nephrol. 9, 112–121 (2013). 76. Barki-​Harrington, L., Perrino, C. & Rockman, H. A. (2016).
50. Franssen, C. F. & Navis, G. Chronic kidney disease: Network integration of the adrenergic system in 100. Craver, L. et al. Mineral metabolism parameters
RAAS blockade and diastolic heart failure in chronic cardiac hypertrophy. Cardiovasc. Res. 63, 391–402 throughout chronic kidney disease stages 1-5—
kidney disease. Nat. Rev. Nephrol. 9, 190–192 (2013). (2004). achievement of K/DOQI target ranges. Nephrol. Dial.
51. Hudlicka, O., Brown, M. & Egginton, S. Angiogenesis 77. Vidal, M., Wieland, T., Lohse, M. J. & Lorenz, K. Transplant. 22, 1171–1176 (2007).
in skeletal and cardiac muscle. Physiol. Rev. 72, beta-Adrenergic receptor stimulation causes cardiac 101. Hruska, K. A., Mathew, S., Lund, R., Qiu, P.
369–417 (1992). hypertrophy via a Gbetagamma/Erk-​dependent & Pratt, R. Hyperphosphatemia of chronic kidney
52. Kurdi, M. & Booz, G. W. New take on the role of pathway. Cardiovasc. Res. 96, 255–264 (2012). disease. Kidney Int. 74, 148–157 (2008).
angiotensin II in cardiac hypertrophy and fibrosis. 78. Moniri, N. H. & Daaka, Y. Agonist-​stimulated reactive 102. Slinin, Y., Foley, R. N. & Collins, A. J. Calcium,
Hypertension 57, 1034–1038 (2011). oxygen species formation regulates beta2-adrenergic phosphorus, parathyroid hormone, and cardiovascular
53. Mehta, P. K. & Griendling, K. K. Angiotensin II cell receptor signal transduction. Biochem. Pharmacol. disease in hemodialysis patients: the USRDS waves 1,
signaling: physiological and pathological effects in the 74, 64–73 (2007). 3, and 4 study. J. Am. Soc. Nephrol. 16, 1788–1793
cardiovascular system. Am. J. Physiol. Cell Physiol. 79. Bovo, E., Lipsius, S. L. & Zima, A. V. Reactive oxygen (2005).
292, C82–C97 (2007). species contribute to the development of 103. Shaman, A. M. & Kowalski, S. R. Hyperphosphatemia
54. Fernandez-​Ruiz, I. Pharmacotherapy: angiotensin II — arrhythmogenic Ca(2)(+) waves during beta-​adrenergic management in patients with chronic kidney disease.
a new tool in vasodilatory shock. Nat. Rev. Cardiol. receptor stimulation in rabbit cardiomyocytes. Saudi Pharm. J. 24, 494–505 (2016).
14, 384 (2017). J. Physiol. 590, 3291–3304 (2012). 104. Vervloet, M. G. et al. The role of phosphate in kidney
55. Taniyama, Y. et al. Role of p38 MAPK and MAPKAPK-2 80. Villarreal, F. J. & Dillmann, W. H. Cardiac disease. Nat. Rev. Nephrol. 13, 27–38 (2017).
in angiotensin II-​induced Akt activation in vascular hypertrophy-induced changes in mRNA levels for 105. Yamazaki-​Nakazawa, A. et al. Correction of
smooth muscle cells. Am. J. Physiol. Cell Physiol. 287, TGF-beta 1, fibronectin, and collagen. Am. J. Physiol. hyperphosphatemia suppresses cardiac remodeling in
C494–C499 (2004). 262, H1861–H1866 (1992). uremic rats. Clin. Exp. Nephrol. 18, 56–64 (2014).
56. Pellieux, C. et al. Dilated cardiomyopathy and impaired 81. Lee, A. A., Dillmann, W. H., McCulloch, A. D. & 106. Rahabi-​Layachi, H., Ourouda, R., Boullier, A.,
cardiac hypertrophic response to angiotensin II in mice Villarreal, F. J. Angiotensin II stimulates the autocrine Massy, Z. A. & Amant, C. Distinct effects of inorganic
lacking FGF-2. J. Clin. Invest. 108, 1843–1851 (2001). production of transforming growth factor-​beta 1 in phosphate on cell cycle and apoptosis in human
57. Zablocki, D. & Sadoshima, J. Angiotensin II and adult rat cardiac fibroblasts. J. Mol. Cell. Cardiol. 27, vascular smooth muscle cells. J. Cell. Physiol. 230,
oxidative stress in the failing heart. Antioxid. Redox 2347–2357 (1995). 347–355 (2015).
Signal. 19, 1095–1109 (2013). 82. Murakami, K., Takemura, T., Hino, S. & Yoshioka, K. 107. Di Marco, G. S. et al. Increased inorganic phosphate
58. Kawada, N., Imai, E., Karber, A., Welch, W. J. & Urinary transforming growth factor-​beta in patients induces human endothelial cell apoptosis in vitro.
Wilcox, C. S. A mouse model of angiotensin II slow with glomerular diseases. Pediatr. Nephrol. 11, Am. J. Physiol. Renal Physiol. 294, F1381–F1387
pressor response: role of oxidative stress. J. Am. 334–336 (1997). (2008).
Soc. Nephrol. 13, 2860–2868 (2002). 83. Bottinger, E. P. & Bitzer, M. TGF-​beta signaling in 108. Gupta, D., Brietzke, S., Hayden, M. R.,
59. Polizio, A. H. et al. Angiotensin II regulates cardiac renal disease. J. Am. Soc. Nephrol. 13, 2600–2610 Kurukulasuriya, L. R. & Sowers, J. R. Phosphate
hypertrophy via oxidative stress but not antioxidant (2002). metabolism in cardiorenal metabolic disease.
enzyme activities in experimental renovascular 84. Dobaczewski, M., Chen, W. & Frangogiannis, N. G. Cardiorenal Med. 1, 261–270 (2011).
hypertension. Hypertens. Res. 31, 325–334 (2008). Transforming growth factor (TGF)-beta signaling in 109. Mehrotra, R. et al. Chronic kidney disease,
60. Zhao, Q. D. et al. NADPH oxidase 4 induces cardiac cardiac remodeling. J. Mol. Cell. Cardiol. 51, hypovitaminosis D, and mortality in the United States.
fibrosis and hypertrophy through activating Akt/mTOR 600–606 (2011). Kidney Int. 76, 977–983 (2009).
and NFkappaB signaling pathways. Circulation 131, 85. Kuwahara, F. et al. Transforming growth factor-​beta 110. Gluba-​Brzozka, A., Franczyk, B., Cialkowska-​Rysz, A.,
643–655 (2015). function blocking prevents myocardial fibrosis and Olszewski, R. & Rysz, J. Impact of vitamin D on the
61. Ferrario, C. M., Chappell, M. C., Dean, R. H. & Iyer, S. N. diastolic dysfunction in pressure-​overloaded rats. cardiovascular system in advanced chronic kidney
Novel angiotensin peptides regulate blood pressure, Circulation 106, 130–135 (2002). disease (CKD) and dialysis patients. Nutrients 10, 709
endothelial function, and natriuresis. J. Am. Soc. 86. Saito, K. et al. Iron chelation and a free radical (2018).
Nephrol. 9, 1716–1722 (1998). scavenger suppress angiotensin II-​induced 111. Holick, M. F. Vitamin D deficiency. N. Engl. J. Med.
62. Balakumar, P. & Jagadeesh, G. A century old renin-​ upregulation of TGF-​beta1 in the heart. Am. J. Physiol. 357, 266–281 (2007).
angiotensin system still grows with endless possibilities: Heart Circ. Physiol. 288, H1836–H1843 (2005). 112. Jones, G. Expanding role for vitamin D in chronic
AT1 receptor signaling cascades in cardiovascular 87. Elkareh, J. et al. Marinobufagenin stimulates kidney disease: importance of blood 25-OH-​D levels
physiopathology. Cell. Signal. 26, 2147–2160 (2014). fibroblast collagen production and causes fibrosis in and extra-​renal 1alpha-​hydroxylase in the classical and
63. Jankowski, V. et al. Angioprotectin: an angiotensin experimental uremic cardiomyopathy. Hypertension nonclassical actions of 1alpha, 25-dihydroxyvitamin
II-​like peptide causing vasodilatory effects. FASEB J. 49, 215–224 (2007). D(3). Semin. Dial. 20, 316–324 (2007).
25, 2987–2995 (2011). 88. de Albuquerque Suassuna, P. G., Sanders-​Pinheiro, H. 113. Nitsa, A. et al. Vitamin D in cardiovascular disease.
64. Lautner, R. Q. et al. Discovery and characterization & de Paula, R. B. Uremic cardiomyopathy: a new piece In Vivo 32, 977–981 (2018).
of alamandine: a novel component of the renin-​ in the chronic kidney disease-​mineral and bone 114. Wu, J., Garami, M., Cheng, T. & Gardner, D. G.
angiotensin system. Circ. Res. 112, 1104–1111 disorder puzzle. Front. Med. 5, 206 (2018). 1,25(OH)2 vitamin D3, and retinoic acid antagonize
(2013). 89. London, G. M. et al. Uremic cardiomyopathy: endothelin-​stimulated hypertrophy of neonatal rat
65. Liu, C. et al. Alamandine attenuates hypertension and an inadequate left ventricular hypertrophy. Kidney Int. cardiac myocytes. J. Clin. Invest. 97, 1577–1588
cardiac hypertrophy in hypertensive rats. Amino Acids 31, 973–980 (1987). (1996).
50, 1071–1081 (2018). 90. Goodman, W. G. The consequences of uncontrolled 115. Li, Y. C. et al. 1,25-Dihydroxyvitamin D(3) is a negative
66. Schlaich, M. P. et al. Sympathetic activation in chronic secondary hyperparathyroidism and its treatment in endocrine regulator of the renin-​angiotensin system.
renal failure. J. Am. Soc. Nephrol. 20, 933–939 chronic kidney disease. Semin. Dial. 17, 209–216 J. Clin. Invest. 110, 229–238 (2002).
(2009). (2004). 116. Leifheit-​Nestler, M. et al. Vitamin D treatment
67. Park, J. Cardiovascular risk in chronic kidney disease: 91. Andersson, P., Rydberg, E. & Willenheimer, R. Primary attenuates cardiac FGF23/FGFR4 signaling and
role of the sympathetic nervous system. Cardiol. Res. hyperparathyroidism and heart disease—a review. hypertrophy in uremic rats. Nephrol. Dial. Transplant.
Pract. 2012, 319432 (2012). Eur. Heart J. 25, 1776–1787 (2004). 32, 1493–1503 (2017).
68. Fisher, J. P., Young, C. N. & Fadel, P. J. Central 92. Rostand, S. G. & Drueke, T. B. Parathyroid hormone, 117. Benet-​Pages, A. et al. FGF23 is processed by proprotein
sympathetic overactivity: maladies and mechanisms. vitamin D, and cardiovascular disease in chronic renal convertases but not by PHEX. Bone 35, 455–462
Auton. Neurosci. 148, 5–15 (2009). failure. Kidney Int. 56, 383–392 (1999). (2004).
69. Zoccali, C. et al. Plasma norepinephrine predicts 93. Lishmanov, A., Dorairajan, S., Pak, Y., Chaudhary, K. & 118. Quarles, L. D. Endocrine functions of bone in mineral
survival and incident cardiovascular events in patients Chockalingam, A. Elevated serum parathyroid metabolism regulation. J. Clin. Invest. 118,
with end-​stage renal disease. Circulation 105, hormone is a cardiovascular risk factor in moderate 3820–3828 (2008).
1354–1359 (2002). chronic kidney disease. Int. Urol. Nephrol. 44, 119. Hu, M. C., Shiizaki, K., Kuro-​o, M. & Moe, O. W.
70. Grassi, G., Seravalle, G., Dell’Oro, R. & Mancia, G. 541–547 (2012). Fibroblast growth factor 23 and Klotho: physiology and
Sympathetic mechanisms, organ damage, and 94. Silver, J. & Naveh-​Many, T. FGF-23 and secondary pathophysiology of an endocrine network of mineral
antihypertensive treatment. Curr. Hypertens. Rep. 13, hyperparathyroidism in chronic kidney disease. metabolism. Annu. Rev. Physiol. 75, 503–533 (2013).
303–308 (2011). Nat. Rev. Nephrol. 9, 641–649 (2013). 120. Leifheit-​Nestler, M. & Haffner, D. Paracrine effects of
71. Chalothorn, D. et al. Differential cardiovascular 95. Krajisnik, T. et al. Fibroblast growth factor-23 FGF23 on the heart. Front. Endocrinol. 9, 278
regulatory activities of the alpha 1B- and alpha 1D-​ regulates parathyroid hormone and 1alpha-​ (2018).
adrenoceptor subtypes. J. Pharmacol. Exp. Ther. 305, hydroxylase expression in cultured bovine parathyroid 121. Tagliabracci, V. S. et al. Dynamic regulation of FGF23
1045–1053 (2003). cells. J. Endocrinol. 195, 125–131 (2007). by Fam20C phosphorylation, GalNAc-​T3 glycosylation,
72. Xu, Q. et al. Myocardial oxidative stress contributes to 96. Schluter, K. D. & Piper, H. M. Cardiovascular actions and furin proteolysis. Proc. Natl Acad. Sci. USA 111,
transgenic beta(2)-adrenoceptor activation-​induced of parathyroid hormone and parathyroid hormone-​ 5520–5525 (2014).
cardiomyopathy and heart failure. Br. J. Pharmacol. related peptide. Cardiovasc. Res. 37, 34–41 (1998). 122. Shimada, T. et al. FGF-23 is a potent regulator of
162, 1012–1028 (2011). 97. Urena, P. et al. Parathyroid hormone (PTH)/PTH-​ vitamin D metabolism and phosphate homeostasis.
73. Cohn, J. N. et al. Plasma norepinephrine as a guide related peptide receptor messenger ribonucleic acids J. Bone Miner. Res. 19, 429–435 (2004).
to prognosis in patients with chronic congestive heart are widely distributed in rat tissues. Endocrinology 123. Urakawa, I. et al. Klotho converts canonical FGF
failure. N. Engl. J. Med. 311, 819–823 (1984). 133, 617–623 (1993). receptor into a specific receptor for FGF23. Nature
74. Elias, A. N., Vaziri, N. D. & Maksy, M. Plasma 98. Smogorzewski, M., Zayed, M., Zhang, Y. B., Roe, J. & 444, 770–774 (2006).
norepinephrine, epinephrine, and dopamine levels Massry, S. G. Parathyroid hormone increases cytosolic 124. Saito, H. et al. Circulating FGF-23 is regulated by
in end-​stage renal disease. Effect of hemodialysis. calcium concentration in adult rat cardiac myocytes. 1alpha, 25-dihydroxyvitamin D3 and phosphorus
Arch. Intern. Med. 145, 1013–1015 (1985). Am. J. Physiol. 264, H1998–H2006 (1993). in vivo. J. Biol. Chem. 280, 2543–2549 (2005).

www.nature.com/nrneph
Reviews

125. Ben-​Dov, I. Z. et al. The parathyroid is a target organ does not increase cardiovascular disease risk. chronic kidney disease, making it a probable novel
for FGF23 in rats. J. Clin. Invest. 117, 4003–4008 Kidney Int. 94, 49–59 (2018). biomarker for early diagnosis. Clin. Exp. Nephrol. 16,
(2007). 151. Chue, C. D. et al. Cardiovascular effects of sevelamer 722–729 (2012).
126. Shimada, T. et al. Targeted ablation of Fgf23 in stage 3 CKD. J. Am. Soc. Nephrol. 24, 842–852 176. Yang, K. et al. Klotho protects against indoxyl
demonstrates an essential physiological role of (2013). sulphate-​induced myocardial hypertrophy. J. Am.
FGF23 in phosphate and vitamin D metabolism. 152. Richter, B., Haller, J., Haffner, D. & Leifheit-​Nestler, M. Soc. Nephrol. 26, 2434–2446 (2015).
J. Clin. Invest. 113, 561–568 (2004). Klotho modulates FGF23-mediated NO synthesis and 177. Xie, J. et al. Cardioprotection by Klotho through
127. Hasegawa, H. et al. Direct evidence for a causative oxidative stress in human coronary artery endothelial downregulation of TRPC6 channels in the mouse
role of FGF23 in the abnormal renal phosphate cells. Pflugers Arch. 468, 1621–1635 (2016). heart. Nat. Commun. 3, 1238 (2012).
handling and vitamin D metabolism in rats with 153. Kurosu, H. et al. Regulation of fibroblast growth 178. Song, S. & Si, L. Y. Klotho ameliorated isoproterenol-​
early-stage chronic kidney disease. Kidney Int. 78, factor-23 signaling by klotho. J. Biol. Chem. 281, induced pathological changes in cardiomyocytes via
975–980 (2010). 6120–6123 (2006). the regulation of oxidative stress. Life Sci. 135,
128. Liu, S. et al. Fibroblast growth factor 23 is a 154. Ito, S., Fujimori, T., Hayashizaki, Y. & Nabeshima, Y. 118–123 (2015).
counter-regulatory phosphaturic hormone for vitamin D. Identification of a novel mouse membrane-​bound 179. Kuwahara, K. et al. TRPC6 fulfills a calcineurin
J. Am. Soc. Nephrol. 17, 1305–1315 (2006). family 1 glycosidase-​like protein, which carries an signaling circuit during pathologic cardiac remodeling.
129. Lopez, I. et al. Direct and indirect effects of parathyroid atypical active site structure. Biochim. Biophys. Acta J. Clin. Invest. 116, 3114–3126 (2006).
hormone on circulating levels of fibroblast growth 1576, 341–345 (2002). 180. Wang, Y., Kuro-​o, M. & Sun, Z. Klotho gene delivery
factor 23 in vivo. Kidney Int. 80, 475–482 (2011). 155. Ogawa, Y. et al. BetaKlotho is required for metabolic suppresses Nox2 expression and attenuates oxidative
130. Rodriguez-​Ortiz, M. E. et al. Calcium deficiency reduces activity of fibroblast growth factor 21. Proc. Natl Acad. stress in rat aortic smooth muscle cells via the
circulating levels of FGF23. J. Am. Soc. Nephrol. 23, Sci. USA 104, 7432–7437 (2007). cAMP-PKA pathway. Aging Cell 11, 410–417 (2012).
1190–1197 (2012). 156. Fon Tacer, K. et al. Research resource: comprehensive 181. Fliser, D. et al. Insulin resistance and hyperinsulinemia
131. Tsuji, K., Maeda, T., Kawane, T., Matsunuma, A. & expression atlas of the fibroblast growth factor system are already present in patients with incipient renal
Horiuchi, N. Leptin stimulates fibroblast growth in adult mouse. Mol. Endocrinol. 24, 2050–2064 disease. Kidney Int. 53, 1343–1347 (1998).
factor 23 expression in bone and suppresses renal (2010). 182. DeFronzo, R. A. et al. Insulin resistance in uremia.
1alpha, 25-dihydroxyvitamin D3 synthesis in leptin-​ 157. Barker, S. L. et al. The demonstration of alphaKlotho J. Clin. Invest. 67, 563–568 (1981).
deficient mice. J. Bone Miner. Res. 25, 1711–1723 deficiency in human chronic kidney disease with a 183. Feneberg, R., Schaefer, F. & Veldhuis, J. D.
(2010). novel synthetic antibody. Nephrol. Dial. Transplant. Neuroendocrine adaptations in renal disease.
132. Gutierrez, O. et al. Fibroblast growth factor-23 30, 223–233 (2015). Pediatr. Nephrol. 18, 492–497 (2003).
mitigates hyperphosphatemia but accentuates 158. Leone, F. et al. Soluble Klotho levels in adult renal 184. Walker, B. G., Phear, D. N., Martin, F. I. & Baird, C. W.
calcitriol deficiency in chronic kidney disease. J. Am. transplant recipients are modulated by recombinant Inhibition of insulin by acidosis. Lancet 2, 964–965
Soc. Nephrol. 16, 2205–2215 (2005). human erythropoietin. J. Nephrol. 27, 577–585 (1963).
133. Isakova, T. et al. Fibroblast growth factor 23 is (2014). 185. Hotamisligil, G. S. et al. IRS-1-mediated inhibition of
elevated before parathyroid hormone and phosphate 159. Ritter, C. S., Zhang, S., Delmez, J., Finch, J. L. & insulin receptor tyrosine kinase activity in TNF-​alpha-
in chronic kidney disease. Kidney Int. 79, 1370–1378 Slatopolsky, E. Differential expression and regulation and obesity-​induced insulin resistance. Science 271,
(2011). of Klotho by paricalcitol in the kidney, parathyroid, 665–668 (1996).
134. Isakova, T. et al. Postprandial mineral metabolism and and aorta of uremic rats. Kidney Int. 87, 1141–1152 186. Khedr, E. et al. Effect of recombinant human
secondary hyperparathyroidism in early CKD. J. Am. (2015). erythropoietin on insulin resistance in hemodialysis
Soc. Nephrol. 19, 615–623 (2008). 160. Lau, W. L. et al. Vitamin D receptor agonists increase patients. Hemodial. Int. 13, 340–346 (2009).
135. Gutierrez, O. M. et al. Fibroblast growth factor 23 and klotho and osteopontin while decreasing aortic 187. Vaziri, N. D. et al. Chronic kidney disease alters
left ventricular hypertrophy in chronic kidney disease. calcification in mice with chronic kidney disease fed a intestinal microbial flora. Kidney Int. 83, 308–315
Circulation 119, 2545–2552 (2009). high phosphate diet. Kidney Int. 82, 1261–1270 (2013).
136. Hsu, H. J. & Wu, M. S. Fibroblast growth factor 23: (2012). 188. Levin, A. et al. Prevalence of abnormal serum vitamin D,
a possible cause of left ventricular hypertrophy in 161. Moreno, J. A. et al. The inflammatory cytokines PTH, calcium, and phosphorus in patients with chronic
hemodialysis patients. Am. J. Med. Sci. 337, 116–122 TWEAK and TNFalpha reduce renal klotho expression kidney disease: results of the study to evaluate early
(2009). through NFkappaB. J. Am. Soc. Nephrol. 22, kidney disease. Kidney Int. 71, 31–38 (2007).
137. Isakova, T. et al. Fibroblast growth factor 23 and risks 1315–1325 (2011). 189. Koppe, L. et al. Urea impairs beta cell glycolysis and
of mortality and end-​stage renal disease in patients 162. Zhu, H., Gao, Y., Zhu, S., Cui, Q. & Du, J. Klotho insulin secretion in chronic kidney disease. J. Clin.
with chronic kidney disease. JAMA 305, 2432–2439 improves cardiac function by suppressing reactive Invest. 126, 3598–3612 (2016).
(2011). oxygen species (ROS) mediated apoptosis by 190. Koppe, L. et al. p-​Cresyl sulfate promotes insulin
138. Gutierrez, O. M. et al. Fibroblast growth factor 23 and modulating MAPKs/Nrf2 signaling in doxorubicin-​ resistance associated with CKD. J. Am. Soc. Nephrol.
mortality among patients undergoing hemodialysis. induced cardiotoxicity. Med. Sci. Monit. 23, 24, 88–99 (2013).
N. Engl. J. Med. 359, 584–592 (2008). 5283–5293 (2017). 191. Xu, H. et al. Clinical correlates of insulin sensitivity and
139. Faul, C. et al. FGF23 induces left ventricular 163. Mitani, H. et al. In vivo klotho gene transfer its association with mortality among men with CKD
hypertrophy. J. Clin. Invest. 121, 4393–4408 (2011). ameliorates angiotensin II-​induced renal damage. stages 3 and 4. Clin. J. Am. Soc. Nephrol. 9, 690–697
140. Seeherunvong, W. et al. Fibroblast growth factor 23 Hypertension 39, 838–843 (2002). (2014).
and left ventricular hypertrophy in children on dialysis. 164. Hu, M. C. et al. Klotho and phosphate are modulators 192. Li, Y., Zhang, L., Gu, Y., Hao, C. & Zhu, T. Insulin
Pediatr. Nephrol. 27, 2129–2136 (2012). of pathologic uremic cardiac remodeling. J. Am. resistance as a predictor of cardiovascular disease
141. Leifheit-​Nestler, M. et al. Induction of cardiac FGF23/ Soc. Nephrol. 26, 1290–1302 (2015). in patients on peritoneal dialysis. Perit. Dial. Int. 33,
FGFR4 expression is associated with left ventricular 165. Kuro-​o, M. et al. Mutation of the mouse klotho gene 411–418 (2013).
hypertrophy in patients with chronic kidney disease. leads to a syndrome resembling ageing. Nature 390, 193. Cusi, K. et al. Insulin resistance differentially affects
Nephrol. Dial. Transplant. 31, 1088–1099 (2016). 45–51 (1997). the PI 3-kinase- and MAP kinase-​mediated signaling in
142. Touchberry, C. D. et al. FGF23 is a novel regulator 166. Corsetti, G. et al. Decreased expression of Klotho in human muscle. J. Clin. Invest. 105, 311–320 (2000).
of intracellular calcium and cardiac contractility in cardiac atria biopsy samples from patients at higher 194. Aroor, A. R., Mandavia, C. H. & Sowers, J. R. Insulin
addition to cardiac hypertrophy. Am. J. Physiol. risk of atherosclerotic cardiovascular disease. resistance and heart failure: molecular mechanisms.
Endocrinol. Metab. 304, E863–E873 (2013). J. Geriatr. Cardiol. 13, 701–711 (2016). Heart Fail. Clin. 8, 609–617 (2012).
143. Faul, C. Fibroblast growth factor 23 and the heart. 167. Kurosu, H. et al. Suppression of aging in mice by the 195. Boucher, J., Kleinridders, A. & Kahn, C. R. Insulin
Curr. Opin. Nephrol. Hypertens. 21, 369–375 (2012). hormone Klotho. Science 309, 1829–1833 (2005). receptor signaling in normal and insulin-​resistant
144. Grabner, A. et al. Activation of cardiac fibroblast 168. Couzin, J. Boosting gene extends mouse life span. states. Cold Spring Harb. Perspect. Biol. 6, a009191
growth factor receptor 4 causes left ventricular Science 309, 1310–1311 (2005). (2014).
hypertrophy. Cell Metab. 22, 1020–1032 (2015). 169. Yamamoto, M. et al. Regulation of oxidative stress by 196. Matsui, T. & Rosenzweig, A. Convergent signal
145. Grabner, A. et al. FGF23/FGFR4-mediated left the anti-​aging hormone klotho. J. Biol. Chem. 280, transduction pathways controlling cardiomyocyte
ventricular hypertrophy is reversible. Sci. Rep. 7, 38029–38034 (2005). survival and function: the role of PI 3-kinase and Akt.
1993 (2017). 170. Shiraki-​Iida, T. et al. Structure of the mouse klotho J. Mol. Cell. Cardiol. 38, 63–71 (2005).
146. Moe, S. M. et al. Cinacalcet, fibroblast growth gene and its two transcripts encoding membrane and 197. Manning, B. D. & Toker, A. AKT/PKB signaling:
factor-23, and cardiovascular disease in hemodialysis: secreted protein. FEBS Lett. 424, 6–10 (1998). navigating the network. Cell 169, 381–405 (2017).
the evaluation of cinacalcet HCl therapy to lower 171. Matsumura, Y. et al. Identification of the human klotho 198. Alessi, D. R. et al. Mechanism of activation of protein
cardiovascular events (EVOLVE) trial. Circulation 132, gene and its two transcripts encoding membrane kinase B by insulin and IGF-1. EMBO J. 15,
27–39 (2015). and secreted klotho protein. Biochem. Biophys. 6541–6551 (1996).
147. Isakova, T. et al. Associations between fibroblast Res. Commun. 242, 626–630 (1998). 199. Semple, D., Smith, K., Bhandari, S. & Seymour, A. M.
growth factor 23 and cardiac characteristics in 172. Lindberg, K. et al. The kidney is the principal organ Uremic cardiomyopathy and insulin resistance:
pediatric heart failure. Pediatr. Nephrol. 28, mediating klotho effects. J. Am. Soc. Nephrol. 25, a critical role for Akt? J. Am. Soc. Nephrol. 22,
2035–2042 (2013). 2169–2175 (2014). 207–215 (2011).
148. Nehgme, R., Fahey, J. T., Smith, C. & Carpenter, T. O. 173. Maekawa, Y. et al. Klotho protein diminishes 200. McMullen, J. R. et al. Phosphoinositide
Cardiovascular abnormalities in patients with X-​linked endothelial apoptosis and senescence via a mitogen-​ 3-kinase(p110alpha) plays a critical role for the
hypophosphatemia. J. Clin. Endocrinol. Metab. 82, activated kinase pathway. Geriatr. Gerontol. Int. 11, induction of physiological, but not pathological,
2450–2454 (1997). 510–516 (2011). cardiac hypertrophy. Proc. Natl Acad. Sci. USA 100,
149. Shalhoub, V. et al. FGF23 neutralization improves 174. Hui, H. et al. Klotho suppresses the inflammatory 12355–12360 (2003).
chronic kidney disease-​associated hyperparathyroidism responses and ameliorates cardiac dysfunction in 201. Samuelsson, A. M. et al. Hyperinsulinemia: effect
yet increases mortality. J. Clin. Invest. 122, 2543–2553 aging endotoxemic mice. Oncotarget 8, on cardiac mass/function, angiotensin II receptor
(2012). 15663–15676 (2017). expression, and insulin signaling pathways.
150. Pastor-​Arroyo, E. M. et al. The elevation of circulating 175. Shimamura, Y. et al. Serum levels of soluble secreted Am. J. Physiol. Heart Circ. Physiol. 291, H787–H796
fibroblast growth factor 23 without kidney disease alpha-​Klotho are decreased in the early stages of (2006).

Nature Reviews | Nephrology


Reviews

202. Cho, H., Thorvaldsen, J. L., Chu, Q., Feng, F. & 228. Tumur, Z. & Niwa, T. Indoxyl sulfate inhibits nitric 252. Meijers, B. K. et al. Free p-​cresol is associated with
Birnbaum, M. J. Akt1/PKBalpha is required for normal oxide production and cell viability by inducing cardiovascular disease in hemodialysis patients.
growth but dispensable for maintenance of glucose oxidative stress in vascular endothelial cells. Kidney Int. 73, 1174–1180 (2008).
homeostasis in mice. J. Biol. Chem. 276, 38349–38352 Am. J. Nephrol. 29, 551–557 (2009). 253. Han, H. et al. p-​Cresyl sulfate aggravates cardiac
(2001). 229. Huang, C. Y. et al. Effects of pamidronate and calcitriol dysfunction associated with chronic kidney disease
203. DeBosch, B. et al. Akt1 is required for physiological on the set point of the parathyroid gland in by enhancing apoptosis of cardiomyocytes. J. Am.
cardiac growth. Circulation 113, 2097–2104 (2006). postmenopausal hemodialysis patients with secondary Heart Assoc. 4, e001852 (2015).
204. Li, Y. et al. Molecular signaling mediated by hyperparathyroidism. Nephron Clin. Pract. 122, 254. Manunta, P. et al. Left ventricular mass, stroke
angiotensin II type 1A receptor blockade leading to 93–101 (2012). volume, and ouabain-​like factor in essential
attenuation of renal dysfunction-​associated heart 230. Fujii, H. et al. Oral charcoal adsorbent (AST-120) hypertension. Hypertension 34, 450–456 (1999).
failure. J. Card Fail. 13, 155–162 (2007). prevents progression of cardiac damage in chronic 255. Kennedy, D. J. et al. Elevated plasma
205. Haq, S. et al. Differential activation of signal kidney disease through suppression of oxidative marinobufagenin, an endogenous cardiotonic steroid,
transduction pathways in human hearts with stress. Nephrol. Dial. Transplant. 24, 2089–2095 is associated with right ventricular dysfunction and
hypertrophy versus advanced heart failure. Circulation (2009). nitrative stress in heart failure. Circ. Heart Fail. 8,
103, 670–677 (2001). 231. Sibal, L., Agarwal, S. C., Home, P. D. & Boger, R. H. 1068–1076 (2015).
206. Shiojima, I. et al. Disruption of coordinated cardiac The role of asymmetric dimethylarginine (ADMA) in 256. Komiyama, Y. et al. A novel endogenous digitalis,
hypertrophy and angiogenesis contributes to the endothelial dysfunction and cardiovascular disease. telocinobufagin, exhibits elevated plasma levels in
transition to heart failure. J. Clin. Invest. 115, Curr. Cardiol. Rev. 6, 82–90 (2010). patients with terminal renal failure. Clin. Biochem. 38,
2108–2118 (2005). 232. Zoccali, C. et al. Left ventricular hypertrophy, cardiac 36–45 (2005).
207. Maillet, M., van Berlo, J. H. & Molkentin, J. D. remodeling and asymmetric dimethylarginine (ADMA) 257. Bagrov, A. Y. et al. Characterization of a urinary
Molecular basis of physiological heart growth: in hemodialysis patients. Kidney Int. 62, 339–345 bufodienolide Na+,K+-ATPase inhibitor in patients
fundamental concepts and new players. Nat. Rev. (2002). after acute myocardial infarction. Hypertension 31,
Mol. Cell Biol. 14, 38–48 (2013). 233. Elesber, A. A. et al. Coronary endothelial dysfunction 1097–1103 (1998).
208. Amann, K. et al. Reduced capillary density in the is associated with erectile dysfunction and elevated 258. Kolmakova, E. V. et al. Endogenous cardiotonic
myocardium of uremic rats—a stereological study. asymmetric dimethylarginine in patients with early steroids in chronic renal failure. Nephrol. Dial.
Kidney Int. 42, 1079–1085 (1992). atherosclerosis. Eur. Heart J. 27, 824–831 (2006). Transplant. 26, 2912–2919 (2011).
209. Amann, K., Breitbach, M., Ritz, E. & Mall, G. 234. Wu, I. W. et al. p-​Cresyl sulphate and indoxyl sulphate 259. Hamlyn, J. M. & Manunta, P. Endogenous cardiotonic
Myocyte/capillary mismatch in the heart of uremic predict progression of chronic kidney disease. steroids in kidney failure: a review and an hypothesis.
patients. J. Am. Soc. Nephrol. 9, 1018–1022 (1998). Nephrol. Dial. Transplant. 26, 938–947 (2011). Adv. Chron. Kidney Dis. 22, 232–244 (2015).
210. Siedlecki, A. M., Jin, X. & Muslin, A. J. Uremic cardiac 235. Barreto, F. C. et al. Serum indoxyl sulfate is associated 260. Haller, S. T. et al. Monoclonal antibody against
hypertrophy is reversed by rapamycin but not by with vascular disease and mortality in chronic kidney marinobufagenin reverses cardiac fibrosis in rats with
lowering of blood pressure. Kidney Int. 75, 800–808 disease patients. Clin. J. Am. Soc. Nephrol. 4, chronic renal failure. Am. J. Hypertens. 25, 690–696
(2009). 1551–1558 (2009). (2012).
211. Matsui, T. et al. Phenotypic spectrum caused by 236. Cao, X. S. et al. Association of indoxyl sulfate with 261. Tian, J. et al. Spironolactone attenuates experimental
transgenic overexpression of activated Akt in the heart failure among patients on hemodialysis. Clin. J. uremic cardiomyopathy by antagonizing
heart. J. Biol. Chem. 277, 22896–22901 (2002). Am. Soc. Nephrol. 10, 111–119 (2015). marinobufagenin. Hypertension 54, 1313–1320
212. Kataria, A., Trasande, L. & Trachtman, H. The effects 237. Wu, C. C. et al. Serum indoxyl sulfate associates with (2009).
of environmental chemicals on renal function. Nat Rev. postangioplasty thrombosis of dialysis grafts. J. Am. 262. Haller, S. T. et al. Rapamycin attenuates cardiac
Nephrol. 11, 610–625 (2015). Soc. Nephrol. 27, 1254–1264 (2016). fibrosis in experimental uremic cardiomyopathy by
213. Thomas, S. S., Zhang, L. & Mitch, W. E. Molecular 238. Yang, K. et al. Indoxyl sulfate induces oxidative stress reducing marinobufagenin levels and inhibiting
mechanisms of insulin resistance in chronic kidney and hypertrophy in cardiomyocytes by inhibiting the downstream pro-​fibrotic signaling. J. Am. Heart Assoc.
disease. Kidney Int. 88, 1233–1239 (2015). AMPK/UCP2 signaling pathway. Toxicol. Lett. 234, 5, e004106 (2016).
214. Borazan, A. & Binici, D. N. Relationship between 110–119 (2015). 263. Haller, S. T. et al. Passive immunization against
insulin resistance and inflamation markers in 239. Lekawanvijit, S. et al. Chronic kidney disease-​induced marinobufagenin attenuates renal fibrosis and
hemodialysis patients. Ren. Fail. 32, 198–202 (2010). cardiac fibrosis is ameliorated by reducing circulating improves renal function in experimental renal disease.
215. Kursat, S. et al. Relationship of insulin resistance in levels of a non-​dialysable uremic toxin, indoxyl sulfate. Am. J. Hypertens. 27, 603–609 (2014).
chronic haemodialysis patients with inflammatory PLOS ONE 7, e41281 (2012). 264. Arnon, A., Hamlyn, J. M. & Blaustein, M. P. Ouabain
indicators, malnutrition, echocardiographic 240. Stockler-​Pinto, M. B., Fouque, D., Soulage, C. O., augments Ca(2+) transients in arterial smooth muscle
parameters and 24 hour ambulatory blood pressure Croze, M. & Mafra, D. Indoxyl sulfate and p-​cresyl without raising cytosolic Na(+). Am. J. Physiol. Heart
monitoring. Scand. J. Urol. Nephrol. 44, 257–264 sulfate in chronic kidney disease. Could these toxins Circ. Physiol. 279, H679–H691 (2000).
(2010). modulate the antioxidant Nrf2-Keap1 pathway? 265. Skou, J. C. The identification of the sodium pump.
216. Martins, C. et al. Insulin resistance is associated with J. Ren. Nutr. 24, 286–291 (2014). Biosci. Rep. 24, 436–451 (2004).
circulating fibrinogen levels in nondiabetic patients 241. Bolati, D., Shimizu, H., Yisireyili, M., Nishijima, F. & 266. Tian, J. et al. Binding of Src to Na+/K+-ATPase forms
receiving peritoneal dialysis. J. Ren. Nutr. 17, Niwa, T. Indoxyl sulfate, a uremic toxin, downregulates a functional signaling complex. Mol. Biol. Cell 17,
132–137 (2007). renal expression of Nrf2 through activation of 317–326 (2006).
217. Campa, C. C., Ciraolo, E., Ghigo, A., Germena, G. NF-kappaB. BMC Nephrol. 14, 56 (2013). 267. Haas, M., Wang, H., Tian, J. & Xie, Z. Src-​mediated
& Hirsch, E. Crossroads of PI3K and Rac pathways. 242. Chin, L. H. et al. The regulation of NLRP3 inter-​receptor cross-​talk between the Na+/K+-ATPase
Small GTPases 6, 71–80 (2015). inflammasome expression during the development and the epidermal growth factor receptor relays
218. Trirogoff, M. L., Shintani, A., Himmelfarb, J. of cardiac contractile dysfunction in chronic kidney the signal from ouabain to mitogen-​activated
& Ikizler, T. A. Body mass index and fat mass are the disease. Oncotarget 8, 113303–113317 (2017). protein kinases. J. Biol. Chem. 277, 18694–18702
primary correlates of insulin resistance in nondiabetic 243. Vilaysane, A. et al. The NLRP3 inflammasome (2002).
stage 3–4 chronic kidney disease patients. Am. J. promotes renal inflammation and contributes to CKD. 268. Liu, J. et al. Ouabain induces endocytosis of
Clin. Nutr. 86, 1642–1648 (2007). J. Am. Soc. Nephrol. 21, 1732–1744 (2010). plasmalemmal Na/K-​ATPase in LLC-​PK1 cells by a
219. Mahadev, K. et al. The NAD(P)H oxidase homolog 244. Muteliefu, G., Enomoto, A. & Niwa, T. Indoxyl sulfate clathrin-​dependent mechanism. Kidney Int. 66,
Nox4 modulates insulin-​stimulated generation of promotes proliferation of human aortic smooth muscle 227–241 (2004).
H2O2 and plays an integral role in insulin signal cells by inducing oxidative stress. J. Ren. Nutr. 19, 269. Tian, J., Gong, X. & Xie, Z. Signal-​transducing function
transduction. Mol. Cell. Biol. 24, 1844–1854 (2004). 29–32 (2009). of Na+-K+-ATPase is essential for ouabain’s effect on
220. Morino, K., Petersen, K. F. & Shulman, G. I. Molecular 245. Yamamoto, H. et al. Indoxyl sulfate stimulates [Ca2+]i in rat cardiac myocytes. Am. J. Physiol. Heart
mechanisms of insulin resistance in humans and their proliferation of rat vascular smooth muscle cells. Circ. Physiol. 281, H1899–H1907 (2001).
potential links with mitochondrial dysfunction. Kidney Int. 69, 1780–1785 (2006). 270. Wansapura, A. N., Lasko, V. M., Lingrel, J. B. &
Diabetes 55 (Suppl. 2), 9–15 (2006). 246. Bartlett, D. E. et al. Uremic toxins activates Lorenz, J. N. Mice expressing ouabain-​sensitive alpha1-
221. Fujii, H., Goto, S. & Fukagawa, M. Role of uremic Na/K-ATPase oxidant amplification loop causing Na, K-​ATPase have increased susceptibility to pressure
toxins for kidney, cardiovascular, and bone dysfunction. phenotypic changes in adipocytes in in vitro models. overload-​induced cardiac hypertrophy. Am. J. Physiol.
Toxins 10, 202 (2018). Int. J. Mol. Sci. 19, E2685 (2018). Heart Circ. Physiol. 300, H347–H355 (2011).
222. Dobre, M., Meyer, T. W. & Hostetter, T. H. Searching 247. Zhao, L. et al. Deletion of interleukin-6 attenuates 271. Drummond, C. A. et al. Reduction of Na/K-​ATPase
for uremic toxins. Clin. J. Am. Soc. Nephrol. 8, pressure overload-​induced left ventricular affects cardiac remodeling and increases c-​kit cell
322–327 (2013). hypertrophy and dysfunction. Circ. Res. 118, abundance in partial nephrectomized mice. Am. J.
223. Neirynck, N. et al. An update on uremic toxins. 1918–1929 (2016). Physiol. Heart Circ. Physiol. 306, H1631–H1643
Int. Urol. Nephrol. 45, 139–150 (2013). 248. Sriramula, S. & Francis, J. Tumor necrosis factor-​alpha (2014).
224. Koppe, L. & Fouque, D. Microbiota and prebiotics is essential for angiotensin II-​induced ventricular 272. Xie, Z. et al. Intracellular reactive oxygen species
modulation of uremic toxin generation. remodeling: role for oxidative stress. PLOS ONE 10, mediate the linkage of Na+/K+-ATPase to hypertrophy
Panminerva Med. 59, 173–187 (2017). e0138372 (2015). and its marker genes in cardiac myocytes.
225. Vanholder, R. et al. Review on uremic toxins: 249. Furukawa, S. et al. Increased oxidative stress in J. Biol. Chem. 274, 19323–19328 (1999).
classification, concentration, and interindividual obesity and its impact on metabolic syndrome. 273. Liu, J. et al. Ouabain interaction with cardiac Na+/K+-
variability. Kidney Int. 63, 1934–1943 (2003). J. Clin. Invest. 114, 1752–1761 (2004). ATPase initiates signal cascades independent of
226. Duranton, F. et al. Normal and pathologic 250. Ramos, L. F., Shintani, A., Ikizler, T. A. & Himmelfarb, J. changes in intracellular Na+ and Ca2+ concentrations.
concentrations of uremic toxins. J. Am. Soc. Nephrol. Oxidative stress and inflammation are associated J. Biol. Chem. 275, 27838–27844 (2000).
23, 1258–1270 (2012). with adiposity in moderate to severe CKD. J. Am. 274. Yan, Y. et al. Protein carbonylation of an amino acid
227. Zoccali, C. et al. Plasma concentration of asymmetrical Soc. Nephrol. 19, 593–599 (2008). residue of the Na/K-​ATPase alpha1 subunit determines
dimethylarginine and mortality in patients with end-​ 251. Viaene, L. et al. Albumin is the main plasma binding Na/K-​ATPase signaling and sodium transport in renal
stage renal disease: a prospective study. Lancet 358, protein for indoxyl sulfate and p-​cresyl sulfate. proximal tubular cells. J. Am. Heart Assoc. 5,
2113–2117 (2001). Biopharm. Drug Dispos. 34, 165–175 (2013). e003675 (2016).

www.nature.com/nrneph
Reviews

275. Liu, J. et al. Attenuation of Na/K-​ATPase mediated of fosinopril and implications for future studies. disease. Oxid. Med. Cell. Longev. 2016, 7520231
oxidant amplification with pNaKtide ameliorates Kidney Int. 70, 1318–1324 (2006). (2016).
experimental uremic cardiomyopathy. Sci. Rep. 6, 292. Marquez, D. F., Ruiz-​Hurtado, G., Ruilope, L. M. & 308. Himmelfarb, J. et al. Provision of antioxidant
34592 (2016). Segura, J. An update of the blockade of the renin therapy in hemodialysis (PATH): a randomized
276. Yan, Y. et al. Involvement of reactive oxygen species in angiotensin aldosterone system in clinical clinical trial. J. Am. Soc. Nephrol. 25, 623–633
a feed-​forward mechanism of Na/K-​ATPase-mediated practice. Expert Opin. Pharmacother. 16, (2014).
signaling transduction. J. Biol. Chem. 288, 2283–2292 (2015). 309. Bolignano, D. et al. Antioxidant agents for delaying
34249–34258 (2013). 293. Juurlink, D. N. et al. Rates of hyperkalemia after diabetic kidney disease progression: a systematic
277. Chen, Y. et al. Oxidized LDL-​bound CD36 recruits an publication of the Randomized Aldactone Evaluation review and meta-​analysis. PLOS ONE 12, e0178699
Na+/K+-ATPase-​Lyn complex in macrophages that Study. N. Engl. J. Med. 351, 543–551 (2004). (2017).
promotes atherosclerosis. Sci. Signal. 8, ra91 (2015). 294. Frankenfield, D. L. et al. Utilization and costs of 310. Cheitlin, M. D. et al. ACC/AHA/ASE 2003
278. Kennedy, D. J. et al. CD36 and Na/K-​ATPase-alpha1 cardiovascular disease medications in dialysis patients guideline update for the clinical application of
form a proinflammatory signaling loop in kidney. in Medicare Part D. Am. J. Kidney Dis. 59, 670–681 echocardiography: summary article. J. Am. Soc.
Hypertension 61, 216–224 (2013). (2012). Echocardiogr. 16, 1091–1110 (2003).
279. Mhatre, K. N. et al. Crosstalk between FGF23- and 295. Cice, G. et al. Carvedilol increases two-​year survivalin 311. Marwick, T. H. et al. Recommendations on the use of
angiotensin II-​mediated Ca(2+) signaling in dialysis patients with dilated cardiomyopathy: echocardiography in adult hypertension: a report from
pathological cardiac hypertrophy. Cell. Mol. Life Sci. a prospective, placebo-​controlled trial. J. Am. Coll. the European Association of Cardiovascular Imaging
75, 4403–4416 (2018). Cardiol. 41, 1438–1444 (2003). (EACVI) and the American Society of Echocardiography
280. Raeisi, S. et al. Effects of angiotensin II receptor 296. Cohen-​Solal, A. et al. Efficacy and safety of nebivolol in (ASE). J. Am. Soc. Echocardiogr. 28, 727–754
blockade on soluble Klotho and oxidative stress in elderly heart failure patients with impaired renal (2015).
calcineurin inhibitor nephrotoxicity in rats. Iran. J. function: insights from the SENIORS trial. Eur. J. 312. Lang, R. M. et al. Recommendations for cardiac
Kidney Dis. 10, 358–363 (2016). Heart Fail. 11, 872–880 (2009). chamber quantification by echocardiography in
281. Karalliedde, J., Maltese, G., Hill, B., Viberti, G. & 297. Badve, S. V. et al. Effects of beta-​adrenergic adults: an update from the American Society of
Gnudi, L. Effect of renin-​angiotensin system blockade antagonists in patients with chronic kidney disease: Echocardiography and the European Association
on soluble Klotho in patients with type 2 diabetes, a systematic review and meta-​analysis. J. Am. Coll. of Cardiovascular Imaging. J. Am. Soc. Echocardiogr.
systolic hypertension, and albuminuria. Clin. J. Am. Cardiol. 58, 1152–1161 (2011). 28, 1–39 (2015).
Soc. Nephrol. 8, 1899–1905 (2013). 298. Agarwal, R., Sinha, A. D., Pappas, M. K., Abraham, T. N. 313. Khouri, S. J., Maly, G. T., Suh, D. D. & Walsh, T. E.
282. Shimizu, H. et al. Indoxyl sulfate enhances angiotensin & Tegegne, G. G. Hypertension in hemodialysis patients A practical approach to the echocardiographic
II signaling through upregulation of epidermal growth treated with atenolol or lisinopril: a randomized evaluation of diastolic function. J. Am. Soc.
factor receptor expression in vascular smooth muscle controlled trial. Nephrol. Dial. Transplant. 29, Echocardiogr. 17, 290–297 (2004).
cells. Life Sci. 91, 172–177 (2012). 672–681 (2014). 314. Sodhi, K. et al. pNaKtide attenuates
283. Lin, C. J. et al. Association of indoxyl sulfate with 299. Kitchlu, A. et al. Beta-​blockers and cardiovascular steatohepatitis and atherosclerosis by blocking
fibroblast growth factor 23 in patients with advanced outcomes in dialysis patients: a cohort study in Na/K-ATPase/ROS amplification in C57Bl6 and
chronic kidney disease. Am. J. Med. Sci. 347, Ontario, Canada. Nephrol. Dial. Transplant. 27, ApoE knockout mice fed a Western diet. Sci. Rep. 7,
370–376 (2014). 1591–1598 (2012). 193 (2017).
284. Taylor, D., Bhandari, S. & Seymour, A. M. Mitochondrial 300. Koizumi, M., Komaba, H., Nakanishi, S., Fujimori, A. & 315. Sodhi, K. et al. pNaKtide inhibits Na/K-​ATPase
dysfunction in uremic cardiomyopathy. Am. J. Physiol. Fukagawa, M. Cinacalcet treatment and serum FGF23 reactive oxygen species amplification and
Renal Physiol. 308, F579–F587 (2015). levels in haemodialysis patients with secondary attenuates adipogenesis. Sci. Adv. 1, e1500781 (2015).
285. Burgoyne, J. R., Mongue-​Din, H., Eaton, P. & hyperparathyroidism. Nephrol. Dial. Transplant. 27,
Shah, A. M. Redox signaling in cardiac physiology 784–790 (2012). Acknowledgements
and pathology. Circ. Res. 111, 1091–1106 301. Greeviroj, P. et al. Cinacalcet for treatment of chronic The authors’ work was supported by US National Institutes
(2012). kidney disease-​mineral and bone disorder: a meta-​ of Health grants HL109015, HL071556 and HL105649;
286. Annuk, M., Zilmer, M., Lind, L., Linde, T. & Fellstrom, B. analysis of randomized controlled trials. Nephron 139, by the Brickstreet Foundation; and by the Huntington
Oxidative stress and endothelial function in chronic 197–210 (2018). Foundation, Inc.
renal failure. J. Am. Soc. Nephrol. 12, 2747–2752 302. Brunelli, S. M., Thadhani, R., Ikizler, T. A. &
(2001). Feldman, H. I. Thiazolidinedione use is associated Author contributions
287. Ruggenenti, P., Cravedi, P. & Remuzzi, G. Mechanisms with better survival in hemodialysis patients with Both authors researched the data discussed in this article,
and treatment of CKD. J. Am. Soc. Nephrol. 23, non-insulin dependent diabetes. Kidney Int. 75, discussed the content, wrote the article and reviewed or
1917–1928 (2012). 961–968 (2009). edited the text before submission.
288. Balamuthusamy, S. et al. Renin angiotensin system 303. Ramirez, S. P. et al. Rosiglitazone is associated with
blockade and cardiovascular outcomes in patients with mortality in chronic hemodialysis patients. J. Am. Competing interests
chronic kidney disease and proteinuria: a meta-​ Soc. Nephrol. 20, 1094–1101 (2009). The authors declare no competing interests.
analysis. Am. Heart J. 155, 791–805 (2008). 304. Hatakeyama, S. et al. Effect of an oral adsorbent,
289. Perkovic, V. et al. Chronic kidney disease, AST-120, on dialysis initiation and survival in patients Publisher’s note
cardiovascular events, and the effects of perindopril-​ with chronic kidney disease. Int. J. Nephrol. 2012, Springer Nature remains neutral with regard to jurisdictional
based blood pressure lowering: data from the 376128 (2012). claims in published maps and institutional affiliations.
PROGRESS study. J. Am. Soc. Nephrol. 18, 305. Schulman, G. et al. Randomized placebo-​controlled
2766–2772 (2007). EPPIC trials of AST-120 in CKD. J. Am. Soc. Nephrol. Reviewer information
290. Mann, J. F., Gerstein, H. C., Pogue, J., Bosch, J. & 26, 1732–1746 (2015). Nature Reviews Nephrology thanks C. Faul, J. Jankowski and
Yusuf, S. Renal insufficiency as a predictor of 306. Drechsler, C. et al. Protein carbamylation is associated the other anonymous reviewer(s), for their contribution to the
cardiovascular outcomes and the impact of ramipril: with heart failure and mortality in diabetic patients peer review of this work.
the HOPE randomized trial. Ann. Intern. Med. 134, with end-​stage renal disease. Kidney Int. 87,
629–636 (2001). 1201–1208 (2015). Supplementary information
291. Zannad, F. et al. Prevention of cardiovascular events in 307. Drozdz, D. et al. Oxidative stress biomarkers and left Supplementary information is available for this paper at
end-​stage renal disease: results of a randomized trial ventricular hypertrophy in children with chronic kidney https://doi.org/10.1038/s41581-018-0101-8.

Nature Reviews | Nephrology

You might also like