You are on page 1of 7

REVIEW

CURRENT
OPINION Sodium homeostasis and bone
Mark J. Hannon and Joseph G. Verbalis

Purpose of review
Sodium balance is primarily regulated through the renin–angiotensin–aldosterone system. Extracellular
fluid (ECF) sodium concentrations ([Naþ]) reflect the overall body sodium content, but are also influenced
by the osmoregulatory system, which is regulated by the posterior pituitary hormone arginine vasopressin
(AVP). Consequently, changes in total body sodium content are not always accurately reflected by the ECF
[Naþ]. This review summarizes the growing evidence base suggesting that skeletal bone, which is rich in
sodium, may play a key role in overall body sodium homeostasis.
Recent findings
Hyponatremia, even when relatively mild, leads to increased morbidity and mortality in diverse clinical
scenarios. In particular, hyponatremia has been shown to increase gait instability, falls, and fracture risk. It
now appears likely that at least part of the fracture risk is because of the adverse effects of hyponatremia
on bone density and quality. The mechanisms through which this occurs are not yet completely understood,
but prominently involve increased bone osteoclast formation and activity. An additional direct effect of AVP
on bone remodeling has also been recently suggested.
Summary
Recent evidence expands upon the previously accepted concepts of body sodium homeostasis and suggests
that sodium balance can be augmented by inputs from skeletal bone, which acts as a sodium-rich reservoir
that can be deployed during times of sodium deficiency. However, this evolutionarily adaptive mechanism
to maintain sodium homeostasis during times of environmental sodium deprivation also has adverse
consequences by negatively impacting bone quality and increasing fracture risk.
Keywords
bone, falls, fracture risk, osteoporosis, sodium balance, vasopressin

INTRODUCTION balance studies are not always reflected by changes


Sodium is the major extracellular cation and, there- in the sodium concentration ([Naþ]) of serum or
fore, is the most important contributor to plasma the extracellular fluid (ECF) [3–6]. It has also been
osmolality, which in healthy humans is tightly known for many years that bone mineral is
regulated and varies by only 1–2% during physio- extremely rich in sodium [3,7], up to 40% of which
logical conditions in which there is free access to is exchangeable with circulating sodium within a
water. This precise regulation of plasma osmolality relatively short time period [4,8]. Bone mineral
is maintained by the homeostatic process of osmor- undergoes constant turnover throughout life [9]
egulation [1]; increases in plasma osmolality are and is the only tissue in which the sodium concen-
detected by specialized osmoreceptor cells in the tration is actually higher than in the ECF [3]. This
anterior hypothalamus, causing increased synthesis leads to the possibility of bone acting as an internal
and secretion of the pituitary hormone arginine sodium ‘reservoir’. The sodium content of bone
vasopressin (AVP), which leads to renal water re- increases with age, whereas the proportion available
absorption. There is a linear relationship between
plasma osmolality and plasma AVP concentrations
Division of Endocrinology and Metabolism, Georgetown University
throughout the physiological range of plasma ton- Medical Center, Washington, District of Columbia, USA
icity [2], and this homeostatic process occurs con- Correspondence to Mark J. Hannon, MD, Georgetown University Medical
tinuously to maintain plasma osmolality within a Center, 233 Building D, 4000 Reservoir Road NW, Washington, DC
relatively narrow reference range. 20007, USA. Tel: +1 202 687 4923; e-mail: markjhannon2002@yahoo.
The classic sodium balance studies of Harrison, co.uk
Kaltreider, Forbes, and others showed that losses Curr Opin Nephrol Hypertens 2014, 23:370–376
of total body sodium demonstrated by external DOI:10.1097/01.mnh.0000447022.51722.f4

www.co-nephrolhypertens.com Volume 23  Number 4  July 2014

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Sodium homeostasis and bone Hannon and Verbalis

HYPONATREMIA AND FRACTURE RISK


KEY POINTS Hyponatremia is the most common electrolyte
 Hyponatremia is the most common electrolyte disorder imbalance encountered in clinical practice [12]
encountered in clinical practice, and even mild and is especially common in the elderly [13]. Hypo-
hyponatremia is associated with cognitive deficits, natremia may progress to a chronic disorder, which
increased mortality, and increased risk of falls is found in up to 11% of ambulatory outpatients
and fractures. [14]. Hyponatremia from multiple causes and in
 Bone mineral is rich in sodium content, which may act heterogeneous clinical settings is associated with
&

as an internal sodium ‘reservoir’ that can be mobilized increased morbidity and mortality [15,16,17 ,18,
&
during states of sodium deficiency. 19,20 ]. Although some of the adverse outcomes
associated with hyponatremia can be accounted
 What is an evolutionarily protective mechanism to
for by the serious medical illness causing the hypo-
maintain sodium homeostasis during sodium deficiency
becomes maladaptive during dilutional hyponatremias natremia [19], data show that hyponatremia itself is
such as SIADH, because the sodium released from associated with adverse outcomes independently of
bone is excreted by the kidneys in the absence of the underlying condition [21]. It is also becoming
aldosterone-mediated renal sodium conservation in increasingly apparent that patients with even mild
this condition. hyponatremia, which was historically thought to be
 This theory is supported by recent basic and clinical relatively asymptomatic, also have excess mortality
evidence showing a clear association between compared with patients with normal plasma [Naþ]
hyponatremia and increased bone resorption, leading [15,16,22,23].
to osteoporosis and increased fracture risk. Furthermore, even mild hyponatremia has been
linked to a significantly increased risk of gait insta-
 The mechanism through which hyponatremia increases
bone resorption is not fully understood but appears to bility, falls, and fractures in the elderly [24–29]. The
be primarily related to osteoclast sensing of serum and adaptive process of the brain to chronic hyponatre-
ECF [Naþ] rather than plasma AVP levels, with mia initially involves cellular loss of electrolytes
subsequent stimulation of osteoclast proliferation and (potassium, sodium, and chloride) to the ECF, fol-
resorptive activity. lowed by subsequent loss of organic osmolytes,
largely amino acids [30], including glutamate which
is the most prevalent excitatory neurotransmitter in
the brain. Loss of intracellular glutamate could con-
for rapid mobilization declines. Subsequent radio- tribute to some of the gait instability and falls found
isotope studies of acute acidosis and sodium in patients with chronic hyponatremia [31], by vir-
depletion in rats showed that bone sodium content tue of delaying the synaptic neurotransmission of
was acutely depleted by these procedures [10,11], action potentials in motor neurons important for
supporting the theory that bone acts as a sodium the maintenance of postural muscle tone. Further-
storage reservoir that could be mobilized to main- more, a rat model of the syndrome of inappropriate
tain the sodium content of the ECF at levels antidiuretic hormone secretion (SIADH) in aged rats
adequate to maintain blood volume, blood pressure, showed that chronic hyponatremia exacerbated
and tissue perfusion during times of sodium multiple manifestations of senescence including
deficiency, similar to the release of bone calcium hypogonadism, decreased body fat, skeletal muscle
to maintain calcium homeostasis during calcium
&
sarcopenia, and cardiomyopathy [32 ]. Although
deprivation. falls alone are an obvious risk factor for fractures
However, little work was done in subsequent in the elderly, the earlier data already outlined
years to further evaluate this possibility. This relating to the importance of sodium in the crystal-
changed over the last decade, as more advanced line structure of bone for sodium homeostasis
data from both rat studies and large population suggested that hyponatremia might also contribute
databases have re-advanced the concept of bone to bone loss, thereby further increasing the fracture
as a storage reservoir for sodium, with chronic alter- risk.
ations in serum sodium balance conversely having
adverse effects on bone quality and fracture risk.
This review will summarize the classic 20th century HYPONATREMIA-INDUCED
studies which first demonstrated the high sodium OSTEOPOROSIS
content of bone, and then will describe the more Increasing interest in hyponatremia as a potential
recent human and experimental data that has new independent risk factor for fractures led to the
revealed strong associations between hyponatremia, publication of a study in 2010 that examined the
bone density and quality, and fracture risk. effects of chronic hyponatremia on bone density in

1062-4821 ß 2014 Wolters Kluwer Health | Lippincott Williams & Wilkins www.co-nephrolhypertens.com 371

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Mineral metabolism

both an animal model and a large human popu- trabecular and cortical bone contents and an
lation database [33]. The investigators utilized an increase in the number of osteoclasts per bone area.
established rat model of SIADH to produce chronic In contrast, mineralizing surface per bone surface,
severe hyponatremia with a mean serum [Naþ] of as analyzed by dynamic histomorphometry, and
110 mmol/l, which was maintained for 3 months. osteoblast surface per bone surface, as analyzed by
Analysis of excised rat femora using dual-energy static histomorphometry, were not significantly
X-ray absorptiometry (DXA) revealed a 30% different between the control and hyponatremic
reduction in bone mineral density (BMD) in the groups. Similarly, analyses of serum and urinary
hyponatremic rats. Subsequent analysis using markers of renal and liver function did not reveal
microcomputed tomography measurements showed any significant differences. Although hyponatremic
marked reductions in both trabecular and cortical animals had a more pronounced reduction in
bone. The second part of this study examined the 1,25(OH)2D3 concentrations, there was no signifi-
effect of chronic hyponatremia on humans using cant increase in parathyroid hormone levels, nor did
the National Health and Nutrition Examination the bone histomorphometry suggest causative
Survey (NHANES) III database, which includes vitamin D deficiency. Furthermore, treatment with
serum [Naþ] and hip BMD data from an ethnically vitamin D only slightly reduced the degree of bone
diverse ambulatory U.S. population. NHANES indi- loss in hyponatremic rats, although it did decrease
viduals who were hyponatremic had an increased further bone loss in a subsequent study of aged
&
risk of osteoporosis of the total hip (odds ratio hyponatremic rats [32 ]. Male hyponatremic animals
2.85) and the femoral neck (2.87), after adjustment also had a mild degree of primary hypogonadism.
for multiple known osteoporosis risk factors, despite Bone formation markers were also reduced, even after
the fact that the overall level of hyponatremia was vitamin D treatment, suggesting an uncoupling of
quite mild (mean ¼ 133.0  0.2 mmol/l). A subse- formation from resorption in hyponatremic animals.
quent study examining the effects of sustained hypo- These data, in aggregate, strongly support osteoclast-
natremia over 18 weeks on aged rats found a mediated bone resorption as the primary causal fac-
progressive reduction of BMD and sodium content tor involved in hyponatremia-induced bone loss.
&
of the tibia and lumbar vertebrae [32 ]. More detailed in-vitro studies of osteoclast num-
Although the above clinical and biochemical ber and function in the presence of low extracellular
findings make teleological sense given the high [Naþ] showed that hyponatremia increased both
concentration of sodium known to be stored in number and resorptive activity of murine RAW
skeletal bone, the above data are mostly associative 264.7 preosteoclastic cells cultured in vitro, and
in nature, with limited insight into the precise primary rat bone marrow monocyte (BMM) cultures
mechanism of sodium mobilization from bone. taken from rats maintained hyponatremic using a
Nor are the data consistent, as Hoorn et al. [29] well described in-vivo model [34]. Importantly, the
found that mild hyponatremia in the elderly was effect of hyponatremia was specific to sodium
associated with an increased risk of vertebral and rather than related to overall osmolality, as normal-
nonvertebral fractures but not with BMD ization of the osmolality of the low sodium
reductions. However, the increased fracture risk culture media using mannitol did not prevent the
was also independent of the number of recent falls, increased proliferation and resorptive activity of
suggesting that although BMD was unchanged, the osteoclasts in cell culture [34] (Fig. 1). This is
there may have been a subtle reduction in bone in contrast to the activity of brain osmoreceptors,
quality as a result of chronic hyponatremia [29]. which clearly react to the changes in extracellular
Recent research has focused on this issue by explor- osmolality rather than individual solute concen-
ing potential causative links between chronic hypo- trations [35].
natremia and low BMD because of reabsorption of Gradual chronic reduction in [Naþ] in the cell
stored bone sodium. culture medium led to an increase in oxidative stress
and free radical accumulation, dose-dependent
decreases in intracellular calcium, and decreases
POTENTIAL MECHANISMS OF in cellular uptake of ascorbic acid. Similarly, a
HYPONATREMIA-INDUCED reduction in ascorbic acid in the culture medium
OSTEOPOROSIS led to increased osteoclast activity. Ascorbic acid
More recent studies have examined the potential exerts its effects on the osteoclast through increased
mechanisms through which hyponatremia could intracellular free oxygen radical accumulation,
affect BMD and bone quality. In the study of and proportional changes in protein expression
Verbalis et al. [33], bone histomorphology was and phosphorylation. This suggested that ascorbic
highly abnormal, with a reduction in both acid may play a role in the sodium signaling

372 www.co-nephrolhypertens.com Volume 23  Number 4  July 2014

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Sodium homeostasis and bone Hannon and Verbalis

RELATION OF HYPONATREMIA-INDUCED
6000
OSTEOPOROSIS TO ARGININE
VASOPRESSIN LEVELS
5000 **
The majority of cases of chronic hyponatremia in
humans are due to SIADH [14], and most patients
TRAP+ MNC/mm2

4000
with chronic hyponatremia have osmotically
inappropriately elevated AVP levels, even if the cause
3000 **
of the hyponatremia is not SIADH. For example,
patients with diuretic-induced hypovolemic hypo-
2000
natremia [41], congestive cardiac failure [42], and
cirrhosis [43] all have nonsuppressed AVP levels. This
1000
has led some to hypothesize that the relative excess of
AVP, rather than the hyponatremia itself, may be the
0
[Na+]=136 [Na+]=112 [Na+]=112
cause of reduced BMD and bone quality in hypo-
&
Osm corrected natremic patients [44 ]. Previous studies have ident-
ified receptors on skeletal bone for other pituitary
FIGURE 1. RAW 264.7 cells were grown in medium with hormones, such as thyroid stimulating hormone
normal [Naþ] (136 mmol/l), in medium with both low (TSH) [45], follicle stimulating hormone [46],
[Naþ] (112 mmol/l) and uncorrected low osmolality adrenocorticotrophic hormone [47], and oxytocin
(237 mOsm/kg), and in medium with low [Naþ] [48]. Deletion of the oxytocin receptor Oxtr causes
(112 mmol/l) and corrected osmolality (290 mOsm/kg). The severe osteoporosis in mice, primarily because
number of tartrate-resistant acid phosphatase (TRAP)-positive of reduced osteoblast function [48]. Reduced TSH
multinucleated cells (i.e., the number of mature osteoclasts) receptor signaling, as seen in primary hyperthyroid-
was significantly higher in both hyponatremic groups, ism, may contribute to the bone loss seen in this
regardless of the medium osmolality (P < 0.001 when condition. Both AVP receptors, AVPR1a and AVPR2,
compared with cells cultured in eunatremic media). MNC, can be found on osteoclasts and osteoblasts, and exert
multinucleated cells. Reproduced with permission [34]. their effects through activation of the intracellular
&
Erk signaling cascade [44 ]. Analysis of AVP receptor
&
mechanism within osteoclasts, leading to increased function by Tamma et al. [44 ] using both wildtype
osteoclast activity in the presence of hyponatremia. and AVPR1a-/- mice and an AVPR1a inhibitor
revealed increased osteoblast number and function,
and reduced osteoclast function and bone resorption
POTENTIAL MEDIATORS OF OSTEOCLAST in mice with knocked-out or pharmacologically
SODIUM SENSING blocked AVPR1a. Specifically, there was a significant
Studies to date do not fully explain the actual mech- increase in bone volume, trabecular thickness, and
anism by which osteoclasts first sense and then number, and a decrease in trabecular spacing follow-
respond to ECF hyponatremia. A sodium-activated ing knockout or inhibition of AVPR1a. Inhibition of
sodium channel, which acts as a sensor of ECF AVPR2 in AVPR1a-/- knockout mice led to similar
sodium, has recently been described on the thick increases in osteoblastogenesis. Conversely, injec-
ascending limb of the loop of Henle in rats [36]. This tion of AVP into wildtype mice led to significant
sensor was previously identified in specialized central upregulation of osteoclast differentiation genes,
nervous system neurons, sensory neurons in the including Cfms, Rank, and Trap.
peripheral nervous system, specialized ependymal However, the data from this study were not
and glial cells in the central nervous system, non- entirely consistent. The expression of Runx2, which
myelinating Schwann cells, and epithelial cells, indicates increased osteoblast differentiation, has
including the alveolar type II cells in the lung [37]. been shown to be increased in both AVPR1a-/- mice
[44 ] and Oxtr-/- mice [48], even though AVP recep-
&
Alternatively, a recent study identified a previously
unknown mechanism controlling vertebrate regen- tor knockout leads to increased bone formation
eration by modulating in-vivo sodium transport, and oxytocin receptor knockout leads to profound
endogenously mediated by the voltage-gated sodium osteoporosis. Also, hyponatremic patients may not
channel, NaV1.2. Direct inhibition of this channel have an absolute AVP elevation above that of euna-
using tricaine, a well described voltage-gated channel tremic patients; rather, the problem is that the AVP
blocker [38,39], reduced vertebrate regeneration, is inappropriately elevated for the patient’s osmo-
suggesting that reduced ECF [Naþ] might lead to
&
lality and sodium level [49,50,51 ]. Furthermore, a
reduced sodium influx, impaired bone healing, and significant relation has been shown between
eventual reduction in bone density and quality [40]. patients’ serum [Naþ] and BMD at the femoral neck

1062-4821 ß 2014 Wolters Kluwer Health | Lippincott Williams & Wilkins www.co-nephrolhypertens.com 373

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Mineral metabolism

[33], which has not been shown for AVP. In fact,


SOLID + DDAVP
AVP levels are highly variable in hyponatremia, and,
as already mentioned, they often do not even exceed 0.24 LIQUID + DDAVP
that of eunatremic patients. The methodological
difficulties involved in measuring AVP in clinical 0.22
*
practice means that it is difficult to relate plasma
AVP concentrations to variables such as serum [Naþ] 0.20

TRAP+ MNC/mm2
or BMD. Copeptin, which is the C-terminal glyco-
protein of the AVP prohormone, has recently been 0.18
established as an easy to measure and stable surro- *
gate for endogenous AVP secretion [52–54], which 0.16

may enable future studies to examine the relation-


ship between AVP and BMD. 0.14

It should also be noted that multiple previous


0.12
results indicate that the phenomenon of hypo-
natremia-induced osteoporosis is independent of
0.10
AVP levels both in vivo and in vitro. In the model
of SIADH used by Verbalis and Drutarosky [55], FIGURE 3. The number of osteoclasts per bone area,
chronic hyponatremia is induced by a combination defined as the number of TRAP-positive multinucleated cells,
of continuous infusion of the AVPR2 agonist des- was five times higher (P < 0.001) in hyponatremic rats
mopressin (DDAVP) in combination with a liquid (mean serum [Naþ] ¼ 110  2 mmol/l) receiving a liquid diet
diet to produce water loading and retention. One of and DDAVP, when compared with eunatremic rats (mean
the controls included in the study was a group of rats serum [Naþ] ¼ 141  1 mmol/l) receiving a solid diet and
infused with the same dose of DDAVP but fed an DDAVP, despite the presence of equivalently excess AVP in
isocaloric solid diet. In the absence of water loading, both groups. AVP, arginine vasopressin; DDAVP, 1-
this group did not become hyponatremic, their desamino-8-D-arginine; MNC, multinucleated cells; TRAP,
BMD was not significantly reduced compared with tartrate-resistant acid phosphatase. Reproduced with
the hyponatremic group (Fig. 2), and the number of permission [33].
osteoclasts present in excised bones was also not
increased (Fig. 3) despite identical DDAVP infusions
[33]. Furthermore, in the in-vitro studies, the culture medium was formulated identically between the
groups of cultured cells except for the sodium con-
centration, so all cells experienced the same extra-
SOLID + DDAVP cellular AVP levels [34]. Consequently, although
0.24
elevated AVP levels may contribute yet another
LIQUID + DDAVP
component to bone loss in some patients with
0.22
SIADH, they are clearly not the primary cause of
0.20 hyponatremia-induced bone loss and osteoporosis.
BMD g/cm2

0.18
RELATION OF SODIUM DEFICIENCY TO
*
0.16 THE SYNDROME OF INAPPROPRIATE
ANTIDIURETIC HORMONE SECRETION
0.14 AND HYPONATREMIA-INDUCED
OSTEOPOROSIS
0.12
The data accumulated to date support the hypo-
0.10 thesis that osteoclast-mediated bone resorption
during hyponatremic conditions occurs to preserve
FIGURE 2. Bone mineral density was significantly reduced sodium homeostasis. Key to this hypothesis is that
in hyponatremic rats (mean serum [Naþ] ¼ 110  2 mmol/l) low body sodium content is sensed by osteoclasts via
receiving a liquid diet and DDAVP, when compared with a low ECF [Naþ]. The sodium resorbed from bone is
eunatremic rats (mean serum [Naþ] ¼ 141  1 mmol/l) retained by the kidney because of activation of the
receiving a solid diet and DDAVP, despite the presence of renin–angiotensin–aldosterone system (RAAS) and
equivalently excess AVP in both groups. AVP, arginine the subsequent actions of aldosterone in the kidney.
vasopressin; BMD, bone mineral density; DDAVP, 1- Thus, mobilization of internal sodium stores would
desamino-8-D-arginine. Reproduced with permission [33]. help to stabilize ECF volume and blood pressure,

374 www.co-nephrolhypertens.com Volume 23  Number 4  July 2014

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Sodium homeostasis and bone Hannon and Verbalis

Table 1. Bone ash analyses in bones from aged rats

L1–L2 vertebrae Tibia


Normo [Naþ] Hypo [Naþ] Normo [Naþ] Hypo [Naþ]

Wet weight (g) 1.998  0.161 1.664  0.051 1.173  0.027 1.168  0.037
Dry weight (g) 0.818  0.099 0.746  0.027 0.767  0.019 0.728  0.019
Ash weight (mg/bone) 307.8  27.8 270.0  16.2 401.1  10.9 375.4  9.0
Calcium mass (mg) 121.8  7.4 113.4  10.1 169.6  6.9 156.0  3.3

Sodium mass (mg) 22.2  0.6 19.1  1.5 19.1  0.5 15.5  0.2

Hypo [Naþ], hyponatremic aged rats; Normo [Naþ], normonatremic aged rats. Adapted with permission [32 ].
&


P < 0.05 compared to normonatremic group values.

P < 0.01 compared to normonatremic group values.

and, in that sense, would be evolutionarily protec- CONCLUSION


tive under conditions of environmental sodium It has long been recognized that bone mineral
deficiency. In contrast, SIADH is not a state of sodium content is rich in sodium, some of which is readily
deficiency, but rather one of dilutional hyponatremia exchangeable with serum [Naþ]. However, it has
due to water retention. Consequently, the low serum only become clear relatively recently that hypo-
[Naþ] in SIADH is primarily because of excess water, natremia is an independent predictive risk factor
not sodium deficiency. However, if serum [Naþ] is for falls and fractures. This is partly because of the
the signal by which osteoclasts sense ECF and total fact that even mild hyponatremia negatively
body sodium, then this would represent a patho- impacts on cognition and gait stability, and also
logical ‘misinterpretation’ of the low serum [Naþ] because hyponatremia itself leads to reduced
as signifying deficient total body sodium. Further- BMD. The mechanism(s) through which hypo-
more, any sodium resorbed from bone during natremia impacts on bone are as yet unknown,
SIADH is not retained, but is instead excreted by but clearly involve stimulation of osteoclast for-
the kidneys as the RAAS is downregulated in SIADH. mation and activity. AVP itself may impact directly
Consequently, there is no brake to the stimulated on bone to exacerbate the hyponatremia-induced
bone resorption, as the serum [Naþ] remains low no bone loss. It therefore appears increasingly likely
matter how much bone is resorbed. This would lead that sodium homeostasis is intrinsically related to
to continued osteoclast-mediated bone resorption as bone physiology, but further research will be needed
long as the hyponatremia persists. Direct evidence of to more accurately delineate the complex interplay
this was demonstrated by measuring bone sodium between these two systems.
content after 3 months of sustained hyponatremia
in rats; bone sodium mass decreased by 14%, even Acknowledgements
greater than the 7% decrease in bone calcium mass The studies in this review were supported by the National
&
[32 ] (Table 1). Institutes of Health (grants R01-AG029477 and UL1-
TR000101 to J.G.V.) and an unrestricted educational
FUTURE DIRECTIONS grant from the Irish Endocrine Society (to M.J.H.).
It is increasingly clear that hyponatremia is an inde-
pendent risk factor for falls and fractures. Given that Conflicts of interest
bone is rich in sodium which is readily exchangeable Disclosures: M.J.H.: nothing to declare; J.G.V. serves as a
with plasma, it makes sense that the risk of fractures consultant to Cornerstone Pharmaceuticals, Ferring
associated with hyponatremia may not just be Pharmaceuticals and Otsuka Pharmaceuticals, and has
related to falls, but also to innate changes in bone received research grants from Otsuka Pharmaceuticals.
density and quality. The molecular and cellular
mechanisms by which this happens are currently
unknown. Further research must identify the poten- REFERENCES AND RECOMMENDED
tial receptors and pathways through which hypo- READING
Papers of particular interest, published within the annual period of review, have
natremia leads to abnormal osteoclast function. been highlighted as:
Similarly, more robust studies of cellular AVP action & of special interest
&& of outstanding interest
are needed to verify whether AVP itself impacts on
bone remodeling, independently of the serum 1. Robertson GL, Aycinena P, Zerbe RL. Neurogenic disorders of osmoregula-
[Naþ], and how these different pathological stimuli tion. Am J Med 1982; 72:339–353.
2. Baylis PH, Thompson CJ. Osmoregulation of vasopressin secretion and thirst
might interact to increase bone fragility. in health and disease. Clin Endocrinol (Oxf) 1988; 29:549–576.

1062-4821 ß 2014 Wolters Kluwer Health | Lippincott Williams & Wilkins www.co-nephrolhypertens.com 375

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Mineral metabolism

3. Harrison HE, Darrow DC, Yannet H. The total electrolyte content of animals 32. Barsony J, Manigrasso MB, Xu Q, et al. Chronic hyponatremia exacerbates
and its probable relation to the distribution of body water. J Biol Chem 1936; & multiple manifestations of senescence in male rats. Age (Dordr) 2013;
113:515–529. 35:271–288.
4. Kaltreider NL, Meneely GR, Allen JR, Bale WF. Determination of the volume of This study showed that, in aged rats with chronic hyponatremia because of
the extracellular fluid of the body with radioactive sodium. J Exp Med 1941; SIADH, the presence of hyponatremia increased bone resorption and decreased
74:569–590. bone ash sodium content, and also led to hypogonadism, reduced body fat,
5. Forbes GB, Perley A. Estimation of total body sodium by isotopic dilution. I. sarcopenia, and cardiomyopathy. This is the first study to show causality in an
Studies on young adults. J Clin Invest 1951; 30:558–565. animal model of the effects of hyponatremia previously seen in various human
6. Bergstrom WH, Wallace WM. Bone as a sodium and potassium reservoir. association studies.
J Clin Invest 1954; 33:867–873. 33. Verbalis JG, Barsony J, Sugimura Y, et al. Hyponatremia-induced osteoporo-
7. Gabriel S. Chemische Untersuchungen uber die Mineralstoffe der Knochen sis. J Bone Miner Res 2010; 25:554–563.
und Zahne. Ztschr f Physiol Chem 1894; 18:257. 34. Barsony J, Sugimura Y, Verbalis JG. Osteoclast response to low extracellular
8. Stern TN, Cole VV, Bass AC, Overman RR. Dynamic aspects of sodium sodium and the mechanism of hyponatremia-induced bone loss. J Biol Chem
metabolism in experimental adrenal insufficiency using radioactive sodium. 2011; 286:10864–10875.
Am J Physiol 1951; 164:437–449. 35. Zerbe RL, Robertson GL. Osmoregulation of thirst and vasopressin secretion
9. Shohl AT. Mineral metabolism. New York: Reinhold Publishing; 1939. in human subjects: effect of various solutes. Am J Physiol 1983; 244:E607–
10. Bergstrom WH. The participation of bone in total body sodium metabolism in E614.
the rat. J Clin Invest 1955; 34 (Part 1):997–1004. 36. Lara LS, Satou R, Bourgeois CR, et al. The sodium-activated sodium channel
11. Nichols G Jr, Nichols N. Electrolyte equilibria in erythrocytes during diabetic is expressed in the rat kidney thick ascending limb and collecting duct cells
acidosis. J Clin Invest 1953; 32:113–120. and is upregulated during high salt intake. Am J Physiol Renal Physiol 2012;
12. Baran D, Hutchinson TA. The outcome of hyponatremia in a general hospital 303:F105–F109.
population. Clin Nephrol 1984; 22:72–76. 37. Watanabe E, Hiyama TY, Kodama R, Noda M. NaX sodium channel is
13. Flear CT, Gill GV, Burn J. Hyponatraemia: mechanisms and management. expressed in nonmyelinating Schwann cells and alveolar type II cells in mice.
Lancet 1981; 2:26–31. Neurosci Lett 2002; 330:109–113.
14. Hannon MJ, Thompson CJ. The syndrome of inappropriate antidiuretic hor- 38. Frazier DT, Narahashi T. Tricaine (MS-222): effects on ionic conductances of
mone: prevalence, causes and consequences. Eur J Endocrinol 2010; 162 squid axon membranes. Eur J Pharmacol 1975; 33:313–317.
(Suppl. 1):S5–S12. 39. Hedrick MS, Winmill RE. Excitatory and inhibitory effects of tricaine (MS-222)
15. Stelfox HT, Ahmed SB, Khandwala F, et al. The epidemiology of intensive care on fictive breathing in isolated bullfrog brain stem. Am J Physiol Regul Integr
unit-acquired hyponatraemia and hypernatraemia in medical-surgical intensive Comp Physiol 2003; 284:R405–R412.
care units. Crit Care 2008; 12:R162. 40. Tseng AS, Beane WS, Lemire JM, et al. Induction of vertebrate regeneration
16. Zilberberg MD, Exuzides A, Spalding J, et al. Hyponatremia and hospital by a transient sodium current. J Neurosci 2010; 30:13192–13200.
outcomes among patients with pneumonia: a retrospective cohort study. 41. Johnson JA, Zehr JE, Moore WW. Effects of separate and concurrent osmotic
BMC Pulm Med 2008; 8:16. and volume stimuli on plasma ADH in sheep. Am J Physiol 1970; 218:1273–
17. Gankam-Kengne F, Ayers C, Khera A, et al. Mild hyponatremia is associated 1280.
& with an increased risk of death in an ambulatory setting. Kidney Int 2013; 42. Schrier RW, Fassett RG. Pathogenesis of sodium and water retention in
83:700–706. cardiac failure. Ren Fail 1998; 20:773–781.
This study showed that hyponatremia is an independent risk factor for mortality in a 43. Cardenas A, Arroyo V. Mechanisms of water and sodium retention in cirrhosis
young, ambulatory, ethnically diverse U.S. population even after adjustment for and the pathogenesis of ascites. Best Pract Res Clin Endocrinol Metab 2003;
multiple risk factors. 17:607–622.
18. Gill G, Huda B, Boyd A, et al. Characteristics and mortality of severe hypona- 44. Tamma R, Sun L, Cuscito C, et al. Regulation of bone remodeling by
traemia – a hospital-based study. Clin Endocrinol (Oxf) 2006; 65:246–249. & vasopressin explains the bone loss in hyponatremia. Proc Natl Acad Sci
19. Clayton JA, Le Jeune IR, Hall IP. Severe hyponatraemia in medical in-patients: USA 2013; 110:18644–18649.
aetiology, assessment and outcome. QJM 2006; 99:505–511. This study showed that AVP receptors are expressed in both osteoclasts and
20. Corona G, Giuliani C, Parenti G, et al. Moderate hyponatremia is associated osteoblasts. Exposure of osteoblast precursors to AVP receptor antagonism or
& with increased risk of mortality: evidence from a meta-analysis. PLoS One deletion led to increased osteoblastogenesis, whereas AVP directly stimulated
2013; 8:e80451. osteoclastogenesis, establishing a direct role for AVP in the causation of hypona-
This large meta-analysis, encompassing 81 studies and 850 222 patients, showed tremia-induced osteoporosis.
that hyponatremia of multiple causes was associated with increased mortality in 45. Abe E, Marians RC, Yu W, et al. TSH is a negative regulator of skeletal
various heterogeneous patient groups, and hyponatremia-related risk of overall remodeling. Cell 2003; 115:151–162.
mortality was inversely correlated with plasma [Naþ]. 46. Sun L, Peng Y, Sharrow AC, et al. FSH directly regulates bone mass. Cell
21. Hoorn EJ, Lindemans J, Zietse R. Development of severe hyponatraemia in 2006; 125:247–260.
hospitalized patients: treatment-related risk factors and inadequate manage- 47. Zaidi M, Sun L, Robinson LJ, et al. ACTH protects against glucocorticoid-
ment. Nephrol Dial Transplant 2006; 21:70–76. induced osteonecrosis of bone. Proc Natl Acad Sci USA 2010; 107:8782–
22. Sajadieh A, Binici Z, Mouridsen MR, et al. Mild hyponatremia carries a poor 8787.
prognosis in community subjects. Am J Med 2009; 122:679–686. 48. Tamma R, Colaianni G, Zhu LL, et al. Oxytocin is an anabolic bone hormone.
23. Waikar SS, Mount DB, Curhan GC. Mortality after hospitalization with mild, Proc Natl Acad Sci USA 2009; 106:7149–7154.
moderate, and severe hyponatremia. Am J Med 2009; 122:857–865. 49. Verbalis JG. Hyponatraemia. Balliere’s Clin Endocrinol Metab 1989; 3:499–
24. Renneboog B, Musch W, Vandemergel X, et al. Mild chronic hyponatremia is 530.
associated with falls, unsteadiness, and attention deficits. Am J Med 2006; 50. Smith DM, McKenna K, Thompson CJ. Hyponatraemia. Clin Endocrinol (Oxf)
119:71.e1–71.e8. 2000; 52:667–678.
25. Gankam Kengne F, Andres C, Sattar L, et al. Mild hyponatremia and risk of 51. Verbalis JG, Goldsmith SR, Greenberg A, et al. Diagnosis, evaluation, and
fracture in the ambulatory elderly. QJM 2008; 101:583–588. & treatment of hyponatremia: expert panel recommendations. Am J Med 2013;
26. Sandhu HS, Gilles E, DeVita MV, et al. Hyponatremia associated with large- 126 (10 Suppl. 1):S1–S42.
bone fracture in elderly patients. Int Urol Nephrol 2009; 41:733–737. This guideline uses the most recent published data and the consensus opinions of
27. Kinsella S, Moran S, Sullivan MO, et al. Hyponatremia independent of various international experts in the field to give updated recommendations for the
osteoporosis is associated with fracture occurrence. Clin J Am Soc Nephrol optimal management of hyponatremia of multiple causes.
2010; 5:275–280. 52. Morgenthaler NG, Struck J, Alonso C, Bergmann A. Assay for the measure-
28. Tolouian R, Alhamad T, Farazmand M, Mulla ZD. The correlation of hip fracture ment of copeptin, a stable peptide derived from the precursor of vasopressin.
and hyponatremia in the elderly. J Nephrol 2012; 25:789–793. Clin Chem 2006; 52:112–119.
29. Hoorn EJ, Rivadeneira F, van Meurs JB, et al. Mild hyponatremia as a risk factor 53. Morgenthaler NG, Struck J, Jochberger S, Dunser MW. Copeptin: clinical use
for fractures: the Rotterdam Study. J Bone Miner Res 2011; 26:1822–1828. of a new biomarker. Trends Endocrinol Metab 2008; 19:43–49.
30. Lien YH, Shapiro JI, Chan L. Study of brain electrolytes and organic osmolytes 54. Balanescu S, Kopp P, Gaskill MB, et al. Correlation of plasma copeptin and
during correction of chronic hyponatremia. Implications for the pathogenesis vasopressin concentrations in hypo-, iso-, and hyperosmolar states. J Clin
of central pontine myelinolysis. J Clin Invest 1991; 88:303–309. Endocrinol Metab 2011; 96:1046–1052.
31. Lipton SA, Rosenberg PA. Excitatory amino acids as a final common pathway 55. Verbalis JG, Drutarosky MD. Adaptation to chronic hypoosmolality in rats.
for neurologic disorders. N Engl J Med 1994; 330:613–622. Kidney Int 1988; 34:351–360.

376 www.co-nephrolhypertens.com Volume 23  Number 4  July 2014

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

You might also like