Download as pdf or txt
Download as pdf or txt
You are on page 1of 37

10

Posterior Pituitary
CHRISTOPHER J. THOMPSON AND JOSEPH G. VERBALIS

CHAPTER OUTLINE
Anatomy, 303 Diabetes Insipidus, 308
Synthesis and Release of Neurohypophyseal Hormones, 304 The Syndrome of Inappropriate Antidiuresis, 315
Physiology of Secretion of Vasopressin and Thirst, 305 Oxytocin, 328

KEY POINTS
• The posterior pituitary is composed of neural tissue and consists induce antidiuresis via a signal transduction cascade that results
of distal axon terminals of the hypothalamic magnocellular in insertion of aquaporin 2 water channels into the apical mem-
vasopressin and oxytocin neurons that constitute the neurohy- brane of principal cells in the renal collecting duct.
pophysis. • Diabetes insipidus is a disorder of a large volume of urine (dia-
• Regulation of neurohypophyseal hormone synthesis occurs at betes) that is hypotonic (dilute) and tasteless (insipid), leading
the level of transcription. Stimuli for the secretion of vasopressin to polyuria and polydipsia. It can be caused by deficient vaso-
or oxytocin also induce transcription and increase the hormone pressin section, increased vasopressin catabolism, or decreased
messenger ribonucleic acid (mRNA) content in the magnocel- vasopressin effect in the kidneys.
lular neurons. • The syndrome of inappropriate antidiuresis (SIAD) occurs when
• Physiologic regulation of vasopressin synthesis and secretion plasma levels of vasopressin are elevated at times when physi-
involves two systems: osmotic and pressure/volume. Each are ologic vasopressin secretion from the posterior pituitary would
controlled by separate neural inputs to the neurohypophysis normally be osmotically suppressed, leading to water retention
and have different thresholds for vasopressin secretion. and hypoosmolality. It can be caused by several diseases and
• Once released, vasopressin elicits end-organ effects via activa- drugs.
tion of vasopressin receptors. Renal vasopressin V2 receptors

Anatomy granules, membrane-bound packets of hormones stored for subse-


quent release. The blood supply for the anterior pituitary is via the
Normal hypothalamic/pituitary portal system, but the posterior pituitary
is supplied directly from the inferior hypophyseal arteries, which
The posterior pituitary is neural tissue and consists only of the are branches of the posterior communicating and internal carotid
distal axons of the hypothalamic magnocellular neurons that make arteries. The drainage is into the cavernous sinus and internal
up the neurohypophysis. The perikarya (cell bodies) of these axons jugular vein.
are located in paired paraventricular and supraoptic nuclei of the The hormones of the posterior pituitary, oxytocin and vaso-
hypothalamus. During embryogenesis neuroepithelial cells of the pressin, are for the most part synthesized in individual hormone-
lining of the third ventricle mature into magnocellular neurons specific magnocellular neurons although a small number of
while migrating laterally to and above the optic chiasm to form neurons (∼3%) express both peptides.2 The supraoptic nucleus
the supraoptic nuclei and to the walls of the third ventricle to form (SON) is relatively simple with 80% to 90% of the neurons pro-
the paraventricular nuclei.1 In the posterior pituitary the axon ducing vasopressin3 and virtually all axons projecting to the pos-
terminals of the magnocellular neurons contain neurosecretory terior pituitary.1 The organization of the paraventricular nucleus

303
Downloaded for FK UMI Makassar (mahasiswafkumi01@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier
on November 01, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
304 SE C T I O N I I    Hypothalamus and Pituitary

(PVN), however, is much more complex and varies among spe- A Arginine vasopressin
cies. In the human there are five subnuclei,3 and parvocellular
(smaller cell) divisions synthesize other peptides such as cortico- Phe3 Gln4
tropin-releasing hormone (CRH), thyrotropin-releasing hormone
Tyr2 Asn5
(TRH), and somatostatin4 as well as opioids.5 The parvocellular
neurons project to the median eminence, brainstem, and spinal H2N Cys1 S S Cys6 Pro7 L Arg8 Gly9 NH2
cord6 where they play a role in a variety of neuroendocrine auto-
nomic functions. The suprachiasmatic nucleus, which is located in
the midline at the base of and anterior to the third ventricle, also B Oxytocin
synthesizes vasopressin and controls both circadian and seasonal
Ile3 Gln4
rhythms.3
The major stimulatory neurotransmitter in the neurohypophy- Tyr2 Asn5
sis is glutamate with noradrenergic stimulatory inputs acting by
stimulation of glutamate.7,8 Glutamate receptors account for 25% H2N Cys1 S S Cys6 Pro7 Leu8 Gly9 NH2
of synapses on magnocellular neurons.7 The major inhibitory
input is γ-aminobutyric acid (GABA), which accounts for 20% to
C Desmopressin
40% of the synaptic input to the magnocellular neurons.9 Phasic
firing of vasopressin neurons is the most efficient activity pattern Phe3 Gln4
for release of vasopressin from axon terminals. Phasic activity is
controlled by glutamate stimulation and opioid inhibition. Dyn- Tyr2 Asn5
orphin is synthesized in vasopressin neurons and co-released with
vasopressin from dendrites at the somatic level where it acts in an H Cys1 S S Cys6 Pro7 D Arg8 Gly9 NH2
autocrine fashion to inhibit the activity of the vasopressin neu- • Fig. 10.1  Comparison of the chemical structures of arginine vasopres-
rons, contributing to the phasic firing pattern.10,11 One of the sin (A), oxytocin (B), and desmopressin (C). The differences are illustrated
most remarkable aspects of the magnocellular system is the plastic- by the shaded areas. Oxytocin differs from vasopressin in position 3 (Ile
ity of the system in response to prolonged stimulation. Plasticity for Phe) and position 8 (Leu for Arg). Desmopressin differs from arginine
is of most import in humans during parturition and lactation.9  vasopressin in that the terminal cystine is deaminated and the arginine
in position 8 is a d-isomer rather than an l-isomer. (From A.G. Robinson,
University of California at Los Angeles, used with permission.)
Ectopic Posterior Pituitary
With the development of magnetic resonance imaging (MRI) in which lysine is substituted for arginine in position 8 produc-
scans of the brain it was discovered that T1-weighted images with ing lysine vasopressin. Both genes are found on chromosome 2019
MRI produced a bright signal in the posterior pituitary.12 This although they are situated in a tail-to-tail position and transcribed
allowed the identification of children in whom there was abnor- in opposite directions.20 The hormones are synthesized as part of
mal anatomy of the posterior pituitary when the “bright spot” a precursor molecule consisting of the nonapeptide and a hor-
was recognized in the base of the hypothalamus. These cases are mone-specific neurophysin and, for vasopressin, a 39–amino acid
referred to as ectopic posterior pituitary or pituitary stalk interrup- peptide named copeptin.21 The precursor is packaged in neurose-
tion. Etiologies include traumatic delivery (these patients have a cretory granules and cleaved to the products during transport to
higher incidence of breech delivery and perinatal injuries) and the posterior pituitary.
genetic abnormalities of the transcription factors that regulate When a stimulus for secretion of vasopressin or oxytocin acts
pituitary embryogenesis.13 The latter is supported in cases where on the appropriate magnocellular cell body, an action potential
abnormalities of the posterior pituitary and/or stalk are associated is generated and propagates down the long axon to the posterior
with extrapituitary malformations such as septal optic dysplasia. pituitary. The action potential causes an influx of calcium, which
Cases with malformations are more likely to have diabetes insipi- induces neurosecretory granules to fuse with the cell membrane
dus or other osmotic dysfunction than simple ectopic posterior and extrude the entire contents of the neurosecretory granule into
pituitary.14,15 Cases are recognized in children with growth retar- the perivascular space and subsequently into the capillary system
dation and anterior pituitary deficiency rather than posterior pitu- of the posterior pituitary. At physiologic pH of plasma there is
itary deficiency. The degree of anterior pituitary deficit depends on no binding of hormone (vasopressin or oxytocin) to their respec-
the persistence of a pituitary stalk and a retained portal vasculature tive neurophysins, so each peptide circulates independently in the
from the hypothalamus to the anterior pituitary.16–18 Deficiency bloodstream.
of adrenocorticotropic hormone (ACTH) is common and should The control of hormone synthesis is at the level of tran-
be investigated as the patients may not respond appropriately to scription. Stimuli for secretion of vasopressin or oxytocin also
stress.13  stimulate transcription and increase the mRNA content in the
magnocellular neurons. This has been studied in most detail in
rats where dehydration22 accelerates transcription and increases
Synthesis and Release of Neurohypophyseal the levels of vasopressin (and oxytocin) mRNA23–25 and where
Hormones hypoosmolality produces a decrease in the content of vasopressin
mRNA.26
Vasopressin and oxytocin are nonapeptides consisting of a six– The transport of neurosecretory vesicles from the site of syn-
amino acid ring with a cysteine-to-cysteine bridge and a three– thesis to the posterior pituitary along microtubule tracks27 is
amino acid tail. All mammals have arginine vasopressin and also regulated. When synthesis is turned off, transport stops;
oxytocin, as illustrated in Fig. 10.1, with the exception of the pig when synthesis is increased, transport is upregulated.27 Thus

Downloaded for FK UMI Makassar (mahasiswafkumi01@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier
on November 01, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
Chapter 10  Posterior Pituitary 305

25 systems at the level of the receptors on the end organs of response.


Pressure
The V1a receptors on blood vessels are distinct from V2 receptors
on renal collecting duct epithelia. These two vasopressin recep-
Plasma vasopressin (pg/mL)
20 tor subtypes are responsible for the main physiologic actions of
Volume vasopressin. A third receptor, V1b, is responsible for the nontra-
Osmolality
15 ditional biologic action of vasopressin to stimulate ACTH secre-
tion from the anterior pituitary and has been found in numerous
peripheral tissues and areas of the brain.32 V2 receptors also regu-
10
late the nontraditional action of vasopressin to stimulate factor
VIII and von Willebrand factor production. Vasopressin is the
5 main hormone involved in the regulation of water homeostasis
and osmolality, while the renin-angiotensin-aldosterone system
0 (RAAS) is mainly responsible for regulation of blood pressure
and volume. Pathologic disorders of the neurohypophysis are
0 +10 +20 +30 (Osmolality) primarily expressed as abnormalities of osmolality produced by
0 −10 −20 −30 (Pressure or volume) abnormal excretion or retention of water. In the case of osmo-
Percent change
regulation vasopressin secretion is relatively uncomplicated with
small increases in osmolality producing a parallel increase in
• Fig. 10.2 Comparison in humans of the release of vasopressin in vasopressin secretion and small decreases in osmolality causing a
response to increased osmolality (open triangles) or to decreased blood
parallel decrease in vasopressin secretion. The regulation of vol-
pressure (filled circles) or blood volume (open circles). Plasma vasopressin
is much more sensitive to change in osmolality, responding to as little as
ume and blood pressure is significantly more complicated (see
a 1% increase, whereas a change of 10% to 15% or greater in volume or review by Thrasher33) and experimental models of vasopressin
pressure is required to stimulate release of vasopressin. (Redrawn from and baroreceptor regulation in animals often involve inhibiting
Robertson GL, Berl T. Water metabolism. In: Brenner B, Rector F Jr, eds. and/or measuring other concurrent sympathetic inputs to the
The Kidney. 3rd ed, vol 1. Philadelphia, PA: Elsevier; 1986:385. Figure by system to ascertain direct effects of any stimulus on secretion
A.G. Robinson, University of California at Los Angeles, used with permis- of vasopressin (see Fig. 10.2). Other influences on secretion of
sion.) vasopressin such as the inhibiting influence of glucocorticoids
and the potent stimulus of nausea and vomiting are less impor-
there is coordination of stimulated release of hormone, trans- tant as physiologic regulators of vasopressin but might contrib-
port of hormone, and synthesis of new hormone. There is, how- ute during pathologic situations.
ever, asynchrony in the timing of these events. The asynchrony is
demonstrated by changes in the content of vasopressin stored in
the posterior pituitary. The absolute content varies considerably
Volume and Pressure Regulation
among species but is a remarkable store, generally equivalent to High-pressure arterial baroreceptors are located in the carotid
the amount of hormone required to sustain basal release for 30 to sinus and aortic arch; low-pressure volume receptors are located
50 days or maximum release for 5 to 10 days.28 In animals, pro- in the atria and pulmonary venous system.33 The afferent signals
longed and intense stimulation of vasopressin release such as dehy- from these receptors are carried from the chest to the brainstem
dration or salt loading produces a depletion of stored hormone in through cranial nerves 9 and 10. Interruption of the vagal input
the posterior pituitary.25,29,30 Then when animals are returned to by vagal cold block in dogs34,35 and destruction in rabbits of the
normal water intake, there is in 7 to 14 days a gradual recovery of A1 area of the medulla, which receives input from cranial nerves
pituitary content back to baseline. Modeling of this phenomenon 9 and 10,36–38 leads to an increase in vasopressin secretion. These
has provided experimental evidence that a long half-life of the and other data led to the concept that baroreceptors and volume
vasopressin message, approximately 2 days, is (from a minimal- receptors normally inhibit the magnocellular neurons and that
ist point of view) a plausible explanation.31 When a strong and/ decreases in this tonic inhibition result in release of vasopressin.
or sustained stimulus releases vasopressin there is an immediate Arterial and venous constriction induced by vasopressin action on
stimulus to transcription of new mRNA, but it requires several V1a receptors will contract the vessels around the existing plasma
days for the peak level of mRNA to be reached; thus while release volume to effectively “increase” plasma volume and reestablish the
of hormone is rapid, translation increases slowly. When the stimu- inhibition of secretion of vasopressin. Vasopressin’s action at the
lus is removed, the elevated mRNA slowly declines while continu- kidney to retain water will help replace volume but, in fact, the
ing to synthesize hormone that repletes the store in the posterior major hormonal regulation to control volume is the RAAS, which
pituitary.  stimulates sodium reabsorption in the kidney (see Chapter 15).
The concept of tonic inhibition of vasopressin secretion by baro-
receptors has been questioned,33,39 but there is agreement that the
Physiology of Secretion of Vasopressin and volume/baroreceptor responses are much less sensitive than are
Thirst the osmoreceptors (see Fig. 10.2). The lesser response has been
attributed to the fact that changes in blood volume and central
The physiologic regulation of vasopressin synthesis and secretion venous pressure have little effect to increase vasopressin in humans
involves two systems: osmotic and pressure/volume (Fig. 10.2). as long as arterial pressure can be maintained by alternative regula-
Functions of these two systems are so distinct that historically it tory mechanisms such as RAAS and sympathetic reflexes.33 When
was thought there were two hormones, an antidiuretic hormone the hypovolemia is sufficient to cause a decrease in blood pres-
and a vasopressor hormone. Hence the two names that are used sure there is an exponential increase in the level of vasopressin in
interchangeably for (8-arginine) vasopressin. There are separate plasma33,40 (see Fig. 10.2). There is also agreement that changes in

Downloaded for FK UMI Makassar (mahasiswafkumi01@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier
on November 01, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
306 SE C T I O N I I    Hypothalamus and Pituitary

volume or pressure that are insufficient to cause direct increases in 320


vasopressin can nonetheless modify the response of the vasopres-
sin system to osmoregulatory inputs.40,41 Increases in pressure and 310
central volume will decrease the secretion of vasopressin,42 but the
response of the RAAS to cause sodium excretion is much more
300
sensitive to increases of pressure and volume than is the response

pOsm
to decrease secretion of vasopressin.33 Consequently, changes in
blood pressure and volume involve both excitatory and inhibitory 290
influences from the brainstem to magnocellular neurons, with the
dominant effect depending on the physiologic circumstances.  280

Osmotic Regulation 270

The primary receptors for sensing changes in osmolality are located 0 3 6 9 12


in the brain. Most of the brain is within the blood-brain barrier, A PAVP
which is generally impermeable to polar solutes. The osmostat is
1400
insensitive to urea and glucose, which readily cross cellular mem-
branes but not the blood-brain barrier; this provides strong evi-
dence that the osmoreceptors must be outside the blood-brain 1050
barrier. Experimental brain lesions in animals have implicated
cells in the organum vasculosum of the lamina terminalis (OVLT)

UOsm
and in areas of the anterior hypothalamus near the anterior wall of 700
the third cerebral ventricle as the primary osmoreceptors. Because
these and other circumventricular organs are perfused by fenes-
trated capillaries, they are outside the blood-brain barrier. Surgical 350
destruction of the OVLT abolishes vasopressin secretion and thirst
responses to hyperosmolality but not their responses to hypovole-
mia.43 Patients with brain damage that destroys the region around 0
the OVLT cannot maintain normal plasma osmolalities even 0 3 6 9 12
under basal conditions.44 In contrast, destruction of the magno- B PAVP
cellular neurons of the SON and PVN eliminates dehydration-
induced secretion of vasopressin but does not alter thirst, clearly 30
indicating that osmotically stimulated thirst must be generated at
a site proximal to the magnocellular cells. Because the osmorecep- 25
tors are relatively solute specific, they are most sensitive to altera-
U volume/24 h

tions in plasma sodium concentrations but respond less well to 20


elevations in blood urea and seem insensitive to changes in plasma
15
glucose concentration.45
Extracellular fluid osmolality (predominantly determined by 10
sodium concentration) varies from 280 to 295 mOsm/kg H2O
in normal subjects, but in any individual it is maintained within 5
a more narrow range. The ability to maintain this narrow range
is dependent upon the sensitive response of plasma vasopressin 0
to changes in plasma osmolality, the sensitive response of urine
osmolality to changes in plasma vasopressin, and the gain in the UOsm 0 350 700 1050 1400
system by the response of urine volume to change in plasma vaso-
pressin (Fig. 10.3). Basal plasma vasopressin is in the range of C PAVP 0 3 6 9 12
0.5 to 2 pg/mL. As little as a 1% increase or decrease in plasma • Fig. 10.3  Effect of change in plasma osmolality (pOsm, in mOsm/kg of
osmolality will cause a rapid increase or decrease of vasopressin H2O) on plasma arginine vasopressin (PAVP, in pg/mL) and consequent
released from the store of hormone in the posterior pituitary.40 effects on urine osmolality (UOsm, in mOsm/kg of H2O) and urine volume
(L/day). The shaded area represents the normal range. (A) Small changes
Rapid metabolism of vasopressin is also characteristic of the hor-
in pOsm induce changes in PAVP, typically between less than 0.5 and 5
mone that circulates in plasma with a half-life of approximately to 6 pg/mL. (B) Changes in PVAP induce changes in UOsm through the
15 minutes, which allows rapid changes in levels of vasopressin in full range, from maximally dilute to maximally concentrated urine. Although
plasma. Thus small increases in plasma osmolality produce a PAVP can rise to higher levels than 6 pg/mL, this does not translate into
concentrated urine and small decreases produce a water diuresis. increased UOsm, which has a maximum determined by the osmolality
Fig. 10.3A illustrates the linear relationship between plasma osmolal­ of an inner medulla of the kidney. (C) The relationship of urine volume to
ity and plasma vasopressin that has been described in humans.40 This UOsm is logarithmic, assuming a constant osmolar load and the urine
linear relationship for osmolalities persists well above the normal volume that would excrete that osmolar load at the UOsm indicated. As a
excursion of osmolalities as demonstrated when the increase is result, urine volume changes relatively little with small changes in the other
induced by infusion of hypertonic saline or is observed during parameters until there is almost complete absence of PAVP, after which the
urine volume increases dramatically. (Calculated from a formula presented
dehydration of patients with nephrogenic diabetes insipidus.46
in Robertson G, Shelton R, Athar S. The osmoregulation of vasopressin.
Similarly, Fig. 10.3B illustrates that there is a sensitive and lin- Kidney Int. 1976;10:25–37. Figure by A.G. Robinson, University of Cali-
ear relationship between the level of vasopressin in plasma and fornia at Los Angeles, used with permission of Macmillan Publishers, Ltd.)

Downloaded for FK UMI Makassar (mahasiswafkumi01@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier
on November 01, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
Chapter 10  Posterior Pituitary 307

the induced osmolality of the urine. Although plasma vasopres- Thirst


sin might increase above the normal physiologic range, the urine
osmolality plateaus at approximately 800 to 1200 mOsm/kg H2O Renal free water clearance can be reduced to a minimum by the
because the maximum concentration of fluid in the renal collect- antidiuretic actions of vasopressin, but water loss is not completely
ing duct is the osmolality of the inner medulla. Fig. 10.3C shows eliminated, and insensible water loss from respiration and sweat-
the relationship of plasma vasopressin to urine volume. This is ing is a continuous process. To maintain water homeostasis, water
a calculated relationship based on the urine volume necessary to must also be consumed to replace the obligate urinary and insen-
excrete a fixed quantity of osmolytes (800 mOsm) at the urine sible fluid losses. This is regulated by thirst. The sensation of thirst
osmolality produced by the change in plasma vasopressin. These can be stimulated by increases in plasma osmolality or by decreases
graphs demonstrate the gain in the system when considering the in intravascular volume, in a manner identical to vasopressin.
changes in urine volume relative to plasma vasopressin. When Furthermore, there is evidence that the receptors are similar (i.e.,
vasopressin is absent, 18 to 20 L of urine are excreted daily; with osmoreceptors in the anterior hypothalamus and low-pressure
an increase of vasopressin by as little as 0.5 to 1 pg/mL, how- and/or high-pressure baroreceptors in the chest [with a likely con-
ever, urine volume is reduced to less than 4 L/day. This illustrates tribution from circulating angiotensin II to stimulate thirst during
the important point that at low plasma levels of vasopressin small more severe degrees of intravascular hypovolemia and hypoten-
changes of vasopressin are much larger determinants of polyuria sion]).51 Careful physiologic studies in healthy humans, using
than are greater changes at higher plasma levels. quantitative estimates of subjective symptoms of thirst, have con-
In the kidney, water is conserved by the combined functions firmed that the characteristics of osmotically stimulated thirst are
of the loop of Henle and the collecting duct. The loop of Henle similar to those of vasopressin secretion; the osmotic thresholds
generates a high osmolality in the renal medulla via the coun- are similar, and there is a close linear relationship between thirst
tercurrent multiplier system. Vasopressin acts in the collecting scores and plasma osmolality throughout a wide range of plasma
duct to increase water (and urea) permeability, thereby allow- tonicity.52 The threshold for producing thirst by hypovolemia is
ing osmotic equilibration between the urine and the hypertonic significantly higher than that for osmotic stimulation.
medullary interstitium. The net effect of this process is to extract Although osmotic changes clearly are effective stimulants of
water from the urine (which is removed from the medulla by thirst, most humans consume the bulk of their ingested water as a
interstitial blood vessels, vasa recta), resulting in increased urine result of the relatively unregulated components of fluid intake—
concentration and decreased urine volume (antidiuresis). Vaso- that is, beverages are consumed with food for reasons of palatabil-
pressin produces antidiuresis by binding to V2 receptors on the ity, taken for desired secondary effects (e.g., caffeine), or taken for
epithelial principal cells of the renal collecting tubule. Binding social or habitual reasons (e.g., sodas or alcoholic beverages). As a
activates adenylate cyclase, increasing cyclic adenosine mono- result, humans generally ingest volumes in excess of what can be
phosphate (cAMP), which then stimulates protein kinase A. considered an actual need for fluid. In humans, plasma osmolality
This leads to phosphorylation and activation of aquaporin 2 and does not vary by more than 1% to 2% of basal levels. However, in
movement of the water channels into the luminal membrane.47 adipsic patients the absence of thirst is associated with profound
Aquaporin 2 is one of the widely expressed family of water chan- alterations of plasma water homeostasis, which emphasizes that
nels that mediate rapid water transport across cell membranes.48 even in humans who mostly drink for pleasure, thirst still plays a
In the kidney, water moves from the collecting duct into the fundamental role in physiologic osmoregulation. 
hypertonic inner medulla producing concentrated urine.49 In
addition to moving constitutively synthesized aquaporin 2 from Clinical Consequences of Osmotic and Volume
the cytoplasm to the luminal membrane, activation of the V2
receptor also increases the synthesis of aquaporin 2 and the Regulation
permeability of aquaporin 2 to water.50 Aquaporin 3 and 4 are Under physiologic situations and during most pathophysiologic
constitutively synthesized and are expressed at high levels in the conditions, the integration of osmotic and baroregulated vasopres-
basolateral plasma membranes of principal cells, where they are sin release and thirst are closely integrated and maintain plasma
responsible for the high water permeability of the basolateral osmolality and sodium concentrations within normal ranges.
plasma membrane.48,49 Dissociation of vasopressin from the V2 During dehydration, for example, plasma osmolality increases
receptor allows intracellular cAMP levels to decrease; the water and blood volume decreases, and vasopressin secretion and thirst
channels are then reinternalized, terminating the increased water sensation occur as a composite result of the two separate stimuli.
permeability. The aquaporin-containing vesicles remain just There is good evidence that a decrease in blood volume shifts the
below the apical membrane and can be quickly “shuttled” into plasma vasopressin/plasma osmolality response curve to the left,
and out of the membrane in response to changes in intracellular resulting in a greater release of vasopressin per unit rise osmolality;
cAMP levels. This mechanism allows minute-to-minute regula- in other words, hypovolemia enhances the vasopressin response
tion of renal water excretion in response to changes in ambient to an osmotic stimulus.33,53 Similarly, an excess of fluid produces
levels of vasopressin in plasma. There is also long-term regulation a decrease in osmolality and an increase in volume, and both will
of collecting duct water permeability in response to prolonged cause a decrease in vasopressin secretion and thirst.
high levels of circulating vasopressin. Chronically high levels of The clinical manifestations of osmoregulatory diseases are
vasopressin induce increased synthesis of aquaporin 2 and 3 water largely explainable on the basis of the physiology of vasopressin
channels in the collecting duct principal cells and hence high lev- secretion and its antidiuretic actions. Fig. 10.3 shows that loss of
els of these proteins. This response requires at least 24 hours and vasopressin neurons sufficient to decrease the vasopressin secretory
is not rapidly reversible. Increased numbers of aquaporin 2 and 3 capacity from that able to produce plasma concentrations of 10 to
water channels combined with the effect of vasopressin to insert 20 pg/mL down to a secretory capacity able to generate plasma
aquaporin 2 into the apical plasma membrane allows the col- concentrations of only 5 pg/mL would not compromise the abil-
lecting ducts to achieve extremely high water permeabilities and ity to maximally concentrate urine. Further losses of vasopressin
water conservation during prolonged dehydration.48,49  neurons such that maximal plasma vasopressin concentrations are

Downloaded for FK UMI Makassar (mahasiswafkumi01@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier
on November 01, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
308 SE C T I O N I I    Hypothalamus and Pituitary

in the 1 to 5 pg/mL range are associated with a linear decrease in nocturnal plasma levels of arginine vasopressin (AVP) and a pro-
the ability to maximally concentrate the urine; however, from the longed effect of administered desmopressin.67 Many other abnor-
urine volume curve it can be seen that this results in only modest malities of fluid and electrolyte balance in the elderly are due to
increases in urine volume. But when loss of vasopressin neurons to comorbid conditions and/or the numerous pharmacologic agents
produce maximal plasma vasopressin concentrations below 1 pg/ these patients are often taking.68 Studies of responses to dehydra-
mL occurs, there is a large increase in urine volume, as urine con- tion, osmolar stimulation, or volume stimulation in the elderly
centration is severely compromised. Water conservation is there- are complicated by the fact that by age 75 to 80 years total body
fore protected despite minimal ability to secrete vasopressin. In water declines to 50% of the level of normal young adults.69 The
addition, the phenomenon of recovery of diabetes insipidus after elderly have a decreased thirst with dehydration and a lesser fluid
transsphenoidal surgery or traumatic brain injury, even after pro- intake during recovery from dehydration.70,71 At the other end of
longed symptoms, might reflect a comparatively minor recovery the spectrum, elderly patients have been found to excrete a water
of vasopressin neurons sufficient to abolish polyuric symptoms. load less well than younger subjects, and at least part of this is
In contrast, in the syndrome of inappropriate antidiuresis, a due to decreased suppression of vasopressin.72 In summary, there
patient who is unable to suppress plasma vasopressin levels to less are age-related changes in body volumes and renal function that
than 1 pg/mL might be unable to excrete more than 2 to 4 L of predispose the elderly to abnormalities in water and electrolyte
urine a day at a standard osmolar load. However, because osmo- balance.73 Diseases that are more common in the elderly aggravate
regulated thirst is not suppressed despite hyponatremia in SIAD,54 this; in addition, the therapy for these diseases affects water bal-
if fluid intake increases above these levels the antidiuretic effects ance. Healthy elderly humans probably have at least a normal (or
of nonsuppressible vasopressin secretion lead to dilutional hypo- increased) ability to secrete vasopressin but a decreased apprecia-
natremia. Thus, although the regulation of vasopressin secretion tion for thirst and a decreased ability to achieve either a maximum
and thirst demonstrates a simple but elegant human system to concentration of urine to retain water or a maximum dilution of
maintain water balance, abnormal functioning of these complex urine to excrete water. This demonstrates the necessity of paying
systems leads to the development of the pathophysiologic states attention to fluid balance problems in the elderly as undetected
that will be described in this chapter.  hypernatremia or hyponatremia can lead to increased morbidity
and mortality.74 
Reset Osmostat During Pregnancy
Diabetes Insipidus
Major shifts of fluid during normal pregnancy produce a fall in
plasma osmolality of about 10 mmol/kg and an increase in plasma Diabetes insipidus is a clinical disorder that is characterized by
volume55; this is the best physiologic example of a true resetting the excretion of large volumes of urine (diabetes) that is hypo-
of the osmostat. The shift in osmotic threshold for vasopressin tonic, dilute, and tasteless (i.e., insipid). This is in contrast to the
appears at about 5 to 8 weeks of gestation and persists throughout hypertonic and sweet urine of diabetes mellitus (i.e., honey). Dia-
pregnancy, returning to normal by 2 weeks after delivery.55 The betes insipidus can result from four abnormal pathophysiologic
physiology of the reset osmostat has been considered in relation processes:
to the expanded plasma volume. Total body water in pregnant 1. Primary polydipsia, caused by excess fluid intake
women is increased by 7 to 8 L due to profound vasodilatation.56 2. Hypothalamic or central diabetes insipidus, caused by dimin-
This volume is sensed as normal and vasopressin responds appro- ished synthesis or secretion of vasopressin
priately to decreases and increases of the expanded volume.55,57 3. Diabetes insipidus of pregnancy, caused by increased enzy-
Both the changes in volume and the changes in osmolality have matic metabolism of vasopressin
been reproduced by infusion of relaxin (a normal hormone of 4. Nephrogenic diabetes insipidus, caused by renal resistance to
pregnancy that is a member of the insulin-like growth factor vasopressin
[IGF] family) into virgin female and normal rats,58,59 and reversed
in pregnant rats by immunoneutralization of relaxin.60 Increased Causes of Diabetes Insipidus
nitric oxide (NO) by relaxin is reported to increase vasodilatation,
and estrogens also increase NO synthesis.61 Diabetes Insipidus Due to Excess Fluid Intake (Primary
In women the placenta produces an enzyme, cysteine ami- Polydipsia)
nopeptidase, which is released into the plasma and is known as Primary polydipsia is associated with a wide variety of organic
oxytocinase.55,62 This enzyme is equally potent in degrading oxy- structural brain lesions, including sarcoidosis of the hypothala-
tocin and vasopressin. The activity of oxytocinase (vasopressinase) mus75 and craniopharyngioma.76 It can also be produced by drugs
increases markedly around 20 weeks of gestation and increases that cause a dry mouth or by any peripheral disorder causing an
further to 40 weeks, returning slowly to normal over a few weeks elevation of renin and/or angiotensin.77 However, there is often
after delivery.63 The potential pathology produced by oxytocinase no identifiable pathologic etiology; in this circumstance the dis-
is described in “Diabetes Insipidus.”  order can be associated with psychiatric syndromes or be habitual
throughout a lifetime. Series of patients in psychiatric hospitals
have shown that as many as 42% have some form of polydipsia,
Osmotic Regulation in Aging and for over half of those there was no obvious explanation for the
Numerous studies have reported that elderly humans are at risk polydipsia.78,79 
for both hypernatremia and hyponatremia.64,65 In older subjects
there is a decrease in glomerular filtration rate,64 and the collecting Hypothalamic/Central Diabetes Insipidus
duct in the aged kidney is less responsive to vasopressin-stimulated Hypothalamic or central diabetes insipidus (CDI) can occur sec-
increases in aquaporin 2 water channels, thus limiting the ability ondary to a large variety of genetic, immunologic, and structural
to excrete free water.66 Elderly subjects are reported to have lower conditions. The relative frequency of the causes of CDI depends on

Downloaded for FK UMI Makassar (mahasiswafkumi01@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier
on November 01, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
Chapter 10  Posterior Pituitary 309

the nature of the unit where the endocrinologist works. However, is often symptomatic and patients present with headache, nau-
as most pituitary endocrinologists work in units where their work sea and emesis, or seizure.84 When all the vasopressin from the
is integrated with that of a neurosurgical unit, neurosurgical condi- damaged neurons has been secreted, the stimulus for water reten-
tions are usually the commonest encountered causes of CDI.80 tion resolves and the retained water is excreted producing recovery
Pituitary adenomas almost never cause CDI to such an extent from the hyponatremia. Thus the clinical picture is one of hypo-
that if a patient presents with a pituitary mass and polyuria and natremia occurring around 7 to 10 days after pituitary surgery,
polydipsia, it is safe to assume that he or she does not have a pitu- persisting for a few days, and then returning to normal. This syn-
itary adenoma. However, neurosurgical intervention with trans- drome of transient hyponatremia has been referred to as “isolated
sphenoidal or transcranial surgery causes CDI in as many as 50% second phase”83 to emphasize the pathophysiologic etiology. Iso-
to 60% of patients, most of whom will recover, with only a small lated hyponatremia has been reported in 10% to 25% of patients
number having permanent diabetes insipidus.81 Permanent CDI after pituitary surgery.85–87
is more common after craniotomy for large tumors. If there is A patient presenting with a sellar mass is more likely to have
complete stalk section, patients can exhibit a pattern known as another tumor in the pituitary area or, alternatively, a granulo-
triphasic diabetes insipidus (Fig. 10.4). The first phase is diabetes matous disorder. Craniopharyngioma is particularly associated
insipidus with onset within the first 24 hours of surgery and is with CDI, particularly after extensive suprasellar surgery; it is
thought to be due to axon shock and inability of action potentials hoped that the new recommendations for a more limited surgi-
to be propagated from the cell body to the neurons terminating in cal approach in conjunction with postoperative radiotherapy for
the posterior pituitary. The second phase is an antidiuretic phase, craniopharyngioma will result in fewer hypothalamic complica-
which is due to unregulated release of vasopressin from damaged tions such as CDI, but there is a paucity of randomized controlled
neurohypophyseal neurons; this phase, which occurs typically 5 data to address this possibility. Other tumors, such as germinoma,
to 7 days following surgery, is often marked by hyponatremia, pinealoma, or a metastasis from a distant solid organ tumor, can
particularly if there is injudicious administration of intravenous also present with CDI. Metastatic disease involving the pituitary
hypotonic fluids. This phase terminates in permanent CDI, as is usually found in association with widespread metastatic disease
the damaged neurons undergo gliosis, and loss of secretory func- and may be asymptomatic and only reported at autopsy. Metas-
tion. An important observation is that the second phase of the tases are twice as likely to involve the posterior pituitary as the
triphasic response (i.e., uncontrolled release of vasopressin due to anterior pituitary,88,89 which is thought to be due to a more direct
axon trauma) can occur without preceding or subsequent diabetes arterial blood supply to the posterior pituitary.90 Most primary
insipidus.82,83 This has been reported clinically and has been pro- tumors in the hypothalamic/pituitary area that cause diabetes
duced experimentally in the rat by unilateral lesion of the supra- insipidus are relatively slow growing, and any tumor in this area
opticohypophyseal track.83 The interpretation is that if the trauma that shows rapid growth in a short period of time should be con-
is to only some of the axons coursing to the posterior pituitary sidered as a possible metastatic tumor.91,92 Pituitary abscess is a
then the remaining intact axons will have sufficient vasopressin rare cause of a pituitary mass and diabetes insipidus.93,94 Diabetes
function to avoid clinically apparent diabetes insipidus character- insipidus has been reported with lymphomas and leukemia in the
istic of the first and third phases of the triphasic response. How- hypothalamic/pituitary area.95–97 Diabetes insipidus is distinctly
ever, the store of hormone in the posterior pituitary is sufficiently more common in nonlymphocytic leukemia.98–100 MRI studies
large that necrosis of even a fraction of these vasopressin neurons in leukemia can show infiltration or an infundibular mass98 but
will cause enough uncontrolled release of vasopressin to produce often are normal even when leukemic cells are found in the cere-
hyponatremia if excess fluid is administered. The hyponatremia brospinal fluid (CSF).100
A variety of granulomatous diseases, such as sarcoidosis and
histiocytosis, have been associated with CDI; usually there is evi-
Diabetes Antidiuretic Diabetes dence of characteristic disease elsewhere in the body.101–103 The
insipidus interphase insipidus MRI often shows involvement of the hypothalamus and absence
10
of the posterior pituitary bright spot on T1-weighted images with
widening of the pituitary stalk (Table 10.1). Although there are
8
occasional reports of resolution of the diabetes insipidus with
Urine output/24 h

TABLE 10.1  Diseases Associated With Enlarged


4
Infundibular Stalk
2 1. Germinoma
2. Craniopharyngioma
3. Metastases to the hypothalamus and long portal vessels (e.g., carci-
0 noma of the breast or lung)
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 4. Granulomatosis diseases
Days post surgery a. Langerhans cell histiocytosis
b. Sarcoidosis
• Fig. 10.4  Typical triphasic response of urine volume after sectioning of
c. Wegener granulomatosis
the pituitary stalk induced by surgery or head trauma. The first phase of
d. Non–Langerhans cell histiocytosis (e.g., Erdheim-Chester dis-
diabetes insipidus occurs immediately postoperatively and continues to
ease)
day 6. The second phase of antidiuresis occurs from day 7 and continues
5. Tuberculosis
to day 12. The third stage is the recurrence of diabetes insipidus on day
6. Lymphocytic infundibulo-hypophysitis
13. Durations vary; see text for detailed discussion. (From A.G. Robinson,
University of California at Los Angeles, used with permission.)

Downloaded for FK UMI Makassar (mahasiswafkumi01@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier
on November 01, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
310 SE C T I O N I I    Hypothalamus and Pituitary

appropriate therapy primary disease, in most cases once estab- Most patients with CDI have normal osmoregulated thirst; as
lished, diabetes insipidus is permanent.104–106 Lymphocytic infun- a result, they do not develop hypernatremia unless they develop
dibuloneurohypophysitis, associated with a thickened stalk and diminished consciousness or cognitive impairment or are in an
loss of the pituitary bright spot on T1-weighted MRI, also pres- environment where water supply is insufficient.116 However,
ents commonly with CDI. Lymphocytic adenohypophysitis pres- damage to the osmoreceptors in the anterior hypothalamus117
ents classically in females around the time of a pregnancy, whereas may lead to the dangerous combination of CDI and adipsia.
infundibuloneurohypophysitis occurs in either sex. Infundibulo- Adipsic DI can be seen where surgical damage is so severe that
neurohypophysitis occurring in middle-aged to elderly males in the damage is not only to the neurohypophysis but also to the
association with IgG4-related systemic disease has recently been central anteriorly placed osmostat,117 such as following extensive
reported. IgG hypophysitis is often associated with other organ surgery for large craniopharyngioma.76 Adipsic DI can also be
involvement, including the pancreas, and other endocrine glands. caused by isolated damage to the hypothalamic osmoreceptors,
The diagnosis can be established by elevated serum IgG4 and with intact baroreceptors, as seen after surgery to an anterior
characteristic histology of biopsies. Response to steroids or other communicating artery aneurysm.118 In the former case, there is
immunosuppressive drugs is characteristic.107,108 no release of vasopressin in response to either osmotic or barore-
CDI with no obvious underlying cause has traditionally been ceptor stimulation; in the latter case, however, there is adequate
considered to be idiopathic, but evidence suggests that a signifi- synthesis of vasopressin with normal baroregulated but attenu-
cant proportion of idiopathic CDI cases are in fact autoimmune ated osmotic release of AVP.119 Adipsic DI is sometimes associ-
in origin.109,110 Several groups have reported the presence of anti- ated with widespread damage to other parts of the hypothalamus
bodies to vasopressin cells in the plasma of patients with “idio- and can be associated with other manifestations of hypothalamic
pathic” CDI, and one series reported an incidence of associated syndrome, such as hyperphagia, sleep apnea, thermoregulation,
autoimmune endocrine disease in 30% of patients with idiopathic seizures, and high mortality.117,120
CDI,111 which is higher than the rate reported in, for example, Although rare, genetic abnormalities of the vasopressin gene
type 1 diabetes mellitus. are a recognized cause of CDI in childhood. Familial CDI is
CDI has been reported in a number of vascular and traumatic characterized by the onset of classic diabetes insipidus with poly-
brain disorders. Approximately 15% of patients who have sus- dipsia and polyuria in childhood or as a young adult, but dur-
tained subarachnoid hemorrhage develop acute CDI,112 which ing infancy they may be asymptomatic.121,122 This is in contrast
nearly always resolves if the patient survives the initial insult. to cases of familial nephrogenic diabetes insipidus in which the
Very few survivors of subarachnoid hemorrhage have permanent defect is expressed as a polyuric disease at birth (see later descrip-
CDI; however, some patients who have surgical clipping of ante- tion). A rare type of familial CDI that can be present at birth
rior communicating artery aneurysms can develop permanent is in infants with homozygotes mutation of the AVP hormone
adipsic CDI due to vascular damage to the perforating arteries region of the pre-prohormone. This can produce excretion of an
of the anterior communicating artery, which provide vascular inactive vasopressin but no difficulty in folding of the pre-pro-
input to the anterior hypothalamus where the osmoreceptors are hormone.123 In the usual familial CDI, MRI findings are vari-
situated (see upcoming discussion). Patients who have moderate able even within affected family members, but the most constant
or severe traumatic brain injury (TBI) also develop CDI quite finding is the presence of a posterior pituitary bright spot in chil-
commonly in the immediate aftermath of injury: 15% to 20% dren, which progressively disappears with time.124 The genetic
develop CDI, and if close attention is not paid to fluid balance defect is usually in the biologically inactive neurophysin or in
when the patient is obtunded or has cognitive decline, hyper- the signal peptide of the pre-prohormone. Although genetically
natremia can develop. Hypernatremia and persistent CDI have heterozygous with the defect expressed in only one allele, the
been shown to be a poor prognostic indicator in acute TBI, often clinical phenotype is autosomal dominant. Lack of normal cleav-
heralding a rise in intracerebral pressure as a preterminal event. age of the signal peptide from the prohormone and abnormal
Patients who develop CDI secondary to TBI also occasionally folding of the vasopressin/neurophysin precursor are thought
go through the triphasic response of early polyuria, followed by to produce fibrillar aggregations in the endoplasmic reticulum,
transient hyponatremia due to unregulated AVP release, before which is cytotoxic to the neuron, explaining the dominant phe-
gliosis produces permanent CDI. There may be a risk in TBI notype.125,126 Autopsy studies have confirmed neuronal cell
patients during the second phase of SIAD, as hyponatremia will death.127 Genetic testing of asymptomatic children in affected
produce cerebral edema, which leads to raised intracranial pres- families will negate the need for repeated dehydration testing
sure. It is important therefore to treat early CDI with intermit- and allow early treatment.128 Wolfram syndrome is a rare auto-
tent dosing of parenteral desmopressin; the effect of one dose somal recessive disease with diabetes insipidus, diabetes mellitus,
should be allowed to wane before administering another dose to optic atrophy, and deafness (DIDMOAD). The genetic defect
ensure that the patient has not developed second phase SIAD. is for the protein wolframin that is found in the endoplasmic
Patients with moderate/severe TBI can also develop anterior reticulum and is important for folding proteins.129 Wolframin is
pituitary deficiency, including ACTH and cortisol deficiency, localized to chromosome 4. It is involved in beta-cell prolifera-
which can affect the ability to excrete water. Cortisol deficiency tion and intracellular protein processing and calcium homeo-
should therefore be considered if diabetes insipidus appears to stasis, producing a wide spectrum of endocrine and central
resolve spontaneously with reversal of polyuria. Cortisol defi- nervous system (CNS) disorders. Diabetes insipidus is usually
ciency alone decreases the ability to excrete water even in the a late manifestation and is associated with decreased magnocel-
absence of vasopressin.113 Lastly, in a long-term follow-up of lular neurons in the paraventricular and supraoptic nuclei.124,130
these patients partial diabetes insipidus can occur,114,115 but Diabetes insipidus is reported as a premorbid development
there may be return of sufficient vasopressin function that under in 50% to 90% of patients with brain death.131,132 This is most
basal conditions the patient is no longer symptomatic from likely to represent a reaction to rising intracranial pressure.
polyuria.113,114 Although some aspects of hormonal treatment of organ donors

Downloaded for FK UMI Makassar (mahasiswafkumi01@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier
on November 01, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
Chapter 10  Posterior Pituitary 311

are controversial, a consensus has emerged that treatment of dia- in males and females. The patients can be heterozygous for two
betes insipidus should be standard in donors who develop CDI.133  different recessive mutations154 or be homozygous for the same
abnormality from both parents.155 Mutations of the aquaporin 2
Diabetes Insipidus Due to Accelerated Metabolism of protein can produce an autosomal dominant nephrogenic diabe-
Vasopressin (Diabetes Insipidus of Pregnancy) tes insipidus when the mutant aquaporin 2 protein associates with
The physiologic adaptations associated with normal pregnancy the wild-type normal protein to inhibit normal intracellular rout-
include expansion of blood volume and decreased plasma osmo- ing and function of the wild type.156
lality and serum sodium. Although thirst and increased fluid The development of NDI in an adult is less likely to reflect
intake are commonly reported in pregnancy, in some patients a genetic cause. The commonest cause of acquired NDI in
the increased thirst is driven by marked polyuria, which could clinical practice is lithium therapy, with other causes (includ-
be indicative of diabetes insipidus. There are two types of tran- ing hypokalemia, hypercalcemia, and release of bilateral urinary
sient diabetes insipidus in pregnancy, both caused by the placental tract obstruction) associated with downregulation of aquaporin
enzyme cysteine aminopeptidase, named oxytocinase, which enzy- 2 and decreased function of vasopressin.153,156 Lithium produces
matically degrades oxytocin, which protects the fetal unit from a decrease in urea transporters, reducing vasopressin-stimulated
early labor.134 Because of the close structural homology between urea uptake and decreasing urea recycling, which reduces inter-
AVP and oxytocin, this enzyme also metabolizes AVP. In the first medullary osmolality.153,157 Even more dramatic is the reduction
type of pregnancy-associated DI, the activity of oxytocinase is in aquaporin 2 levels to decrease water transport in the collecting
abnormally elevated. This syndrome has been referred to as “vaso- duct.156 There is as much as a 95% decrease in aquaporin 2 con-
pressin resistant diabetes insipidus of pregnancy,”135 and has been tent and even the 5% of aquaporin 2 that persists is not normally
reported with preeclampsia, acute fatty liver, and coagulopathies. transported to the renal principal cell membrane.158 The defect of
Affected patients have decreased metabolism of vasopressinase by aquaporins with lithium is slow to correct both in experimental
the liver.56,136–138 Characteristically, subsequent pregnancies are animals and in humans and can be permanent.156,159 Demeclocy-
not complicated by diabetes insipidus or acute fatty liver. In the cline is another commonly recognized drug to cause nephrogenic
second type of pregnancy-associated DI, the accelerated metabolic diabetes insipidus and is sometimes used clinically to treat SIAD.
clearance of vasopressin produces diabetes insipidus in a patient A comprehensive review160 has documented the large number of
with borderline vasopressin function from a specific disease (e.g., drugs that can cause NDI. 
mild nephrogenic diabetes insipidus or partial hypothalamic/neu-
rohypophyseal diabetes insipidus).62,139,140 Vasopressin is rapidly Approach to the Differential Diagnosis of
destroyed and the neurohypophysis is unable to keep up with
the increased demand. Labor and parturition usually proceed Polyuric States
normally and patients have no trouble with lactation.141 There The first diagnostic step is to confirm polyuria, because up to 15%
is the threat of chronic and severe dehydration when diabetes of patients referred for investigation of polyuria have urinary fre-
insipidus is unrecognized, and this can pose a threat to a pregnant quency due to bladder wall defects, infection, or prostate disease,
woman.142 Patients with Sheehan syndrome have been reported to with normal urine volume. A 24-hour urine volume greater than
have asymptomatic partial diabetes insipidus143 but rarely develop 50 mL/kg body weight is worthy of further investigation. Diabe-
overt diabetes insipidus.144  tes mellitus, hypercalcemia, hypokalemia, and chronic renal fail-
ure are excluded by biochemical tests. Urine osmolality should
Nephrogenic Diabetes Insipidus be low in all polyuric states; however, a random urine osmolality
Genetic variants of nephrogenic diabetes insipidus (NDI) usu- above 700 mOsm/kg excludes diabetes insipidus and makes the
ally present in infancy, with vomiting, failure to thrive, and diagnosis of primary polydipsia certain. Presenting serum sodium
polyuria.145,146 NDI can occur as a result of mutations in the V2 concentration is almost always normal in diabetes insipidus, but
receptor and mutations of the aquaporin 2 water channels.145–147 results at the higher part of the reference range are more suggestive
Over 90% of cases are X-linked recessive in males, and over 200 of DI than primary polydipsia. However, it is difficult to confi-
different mutations of the V2 receptor have been reported.148 dently distinguish between CDI, NDI, and thirst disorders solely
Three general categories of V2 receptor mutations have been on baseline laboratory measurements, so osmotic stimulation of
described148: the posterior pituitary is needed to secure a firm diagnosis.
1. Type 1, characterized by impaired AVP binding The water deprivation test, which incorporates a dehydration
2. Type 2, typified by defective transport step followed by a desmopressin challenge, is usually the first line
3. Type 3, where unstable receptors are rapidly degraded of investigation. The interpretation of the initial period of water
Most of the reported cases are of type 2.149 Ten percent of deprivation is based on the understanding that a functioning
the V2 receptor defects causing congenital nephrogenic diabetes osmoregulatory system will respond to elevation of plasma osmo-
insipidus occur de novo. The majority of female carriers of the lality with the secretion of vasopressin and subsequent concentra-
X-linked V2 receptor mutation are asymptomatic, though careful tion of the urine. As the test progresses, urine volume decreases
physiologic testing reveals that some might have attenuated maxi- and urine osmolality rises usually to over 750 mOsm/kg H2O.
mum urine osmolality in response to vasopressin.150 In rare cases Importantly, an adequate osmotic stimulus to AVP secretion is
where heterozygous females have a defect as severe as males, it is necessary; a plasma osmolality of over 295 mOsm/kg H2O is
thought that there is coexisting inactivation of the function of the needed to stimulate sufficient AVP to maximally concentrate the
X chromosome.151,152 urine. In primary polydipsia urine should concentrate relatively
When the proband is a female it is likely the defect is a muta- normally, as AVP secretion is normal, whereas in patients with
tion of the aquaporin 2 water channel gene due to an autosomal either CDI or NDI, urine remains dilute at the end of dehydra-
recessive disease.153 This should be especially considered where tion. The second part of the test, the administration of desmopres-
consanguinity is known in the family and the disease is expressed sin, is designed to differentiate CDI from NDI. As patients with

Downloaded for FK UMI Makassar (mahasiswafkumi01@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier
on November 01, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
312 SE C T I O N I I    Hypothalamus and Pituitary

CDI are deficient in AVP, exogenous desmopressin concentrates quick, reliable turnover of results. Careful studies have revealed
urine osmolality, whereas patients with NDI do not respond to that the copeptin responses to changes in plasma osmolality are
the antidiuretic effects of desmopressin and do not concentrate similar to those seen with plasma AVP levels when measured dur-
urine appropriately. The water deprivation test is best performed ing water deprivation.165,166 In a prospective multicenter study,
in a specialized center. Careful patient supervision to monitor for a postoperative copeptin concentration of less than 2.5 pmol/L
surreptitious drinking is important, and body weight is checked as had a post-transsphenoidal surgery predictive value of 81% for
patients with severe CDI can develop hypernatremia when fluid is CDI, with a specificity of 97%. Conversely, high plasma copeptin
withheld due to excess urination. In addition, there are subtleties concentrations effectively excluded the diagnosis of central DI; a
of interpretation of results that must be considered. Although the plasma copeptin concentration over 30 pmol/L on day 1 follow-
water deprivation test differentiates between primary polydipsia, ing transsphenoidal surgery was associated with a negative pre-
CDI, and NDI in classic and complete cases, published data have dictive value of 95%, with a sensitivity of 94%.167 In addition,
shown that accurate diagnosis was made in only 70% of polyuric a single copeptin concentration of over 21.4 pmol/L has been
patients, and the correct diagnosis in primary polydipsia was con- shown to differentiate NDI from other causes of polyuria, with a
cluded in only 41% of cases.161 A number of confounding factors sensitivity and specificity of 100%, thus mitigating the need for
can produce misleading or uninterpretable results: water deprivation testing.168 Conversely, a baseline copeptin less
1. Patients with prolonged severe polyuria may not concentrate than 2.6 pmol/L diagnoses complete CDI with 95% sensitivity
urine in response to endogenous or exogenous vasopressin. and 100% specificity. Copeptin is therefore a promising marker
Chronic hypoosmolality suppresses AVP secretion, and in the of plasma AVP secretion, which offers a convenient, rapid, and
absence of V2 receptor stimulation intracellular aquaporin 2 is sensitive diagnostic tool for polyuric states. The assay should be
not generated. Patients with thirst disorders may therefore be manageable in most clinical laboratories, and it is likely to replace
classified as partial DI on the basis of a subnormal rise in urine measurement of plasma AVP in routine diagnostic practice.
osmolality during the water deprivation step, despite normal Measurement of thirst, using a simple visual analogue scale, in
AVP responses. For the same reason, receptor stimulation with osmotic studies has demonstrated that thirst onset occurs at the
desmopressin in step 2 might not stimulate increased urine same osmotic threshold as AVP secretion52 and is rapidly switched
concentration in patients with CDI, leading to an erroneous off by drinking.169 The characteristics of osmoregulated thirst are
diagnosis of NDI. surprisingly reproducible.170 When patients with CDI are stud-
2. In patients with partial CDI, upregulation of V2 receptors can ied during water deprivation testing or hypertonic saline infusion,
result in urine concentration with relatively low plasma AVP they show a similar pattern of linear elevation during osmotic
concentrations. stimulation, and they show suppression after drinking.116
3. High plasma AVP concentrations achieved by fluid deprivation Measurement of thirst is valuable diagnostically in two clini-
in NDI can partially overcome renal resistance; urine osmolal- cal situations. In adipsic DI, absent osmoregulated thirst during
ity can rise to over 300 mOsm/kg H2O, leading to diagnostic water deprivation or hypertonic saline infusion is the gold stan-
confusion with partial CDI.162 dard for diagnosis of the condition.116,117 Patients with primary
Several studies have demonstrated that the diagnostic accu- polydipsia have three clearly defined abnormalities of thirst: (1)
racy of the water deprivation test was enhanced by the inclusion a low osmotic threshold for thirst, which is disconnected from
of measurement of plasma AVP concentration using a sensitive that of AVP release; (2) exaggerated thirst responses to elevation of
radioimmunoassay.163,164 However, AVP is unstable after veni- plasma osmolality; and (3) failure to suppress thirst during drink-
puncture and requires careful sample handling, immediate centrif- ing after osmotic stimulation.171 Failure to suppress thirst after
ugation, and storage at -70°C. In addition, commercially available drinking by more than 50% of stimulated levels is the most use-
AVP antibodies for use in clinical radioimmunoassays are poorly ful of these abnormalities, as it is a strong diagnostic indicator of
sensitive and have lower limits of detection for the hormone that primary polydipsia. 
exceeds low physiologic plasma concentrations. Good-quality,
sensitive and specific assays are available in a few specialized cen- Further Investigations of Diabetes Insipidus
ters, but results are often only available weeks after testing has
been performed. A recent analysis claimed a diagnostic accuracy of Once a diagnosis of CDI is established, MRI of the hypothalamo-
only 46% when direct AVP measurement was prospectively tested neurohypophyseal region is indicated. T1-weighted MRI images
in 50 patients with polyuria,161 but the results may have reflected show a classic bright spot in the posterior pituitary12 caused by
suboptimal performance of the AVP assay that was used. Alterna- AVP stored in neurosecretory granules.29,172–175 The bright spot
tively, measurement of the AVP responses during the intravenous is present in over 80% of normal subjects176,177 and it is absent
infusion of hypertonic (3–5%) sodium chloride solution at the in most patients with central diabetes insipidus.178 Some patients
end of a water deprivation test enables more confident distinction with familial hypothalamic/neurohypophyseal diabetes insipidus
between CDI and NDI or primary polydipsia. can have a posterior pituitary bright spot early in the disease, when
Because the logistic difficulty of accurate and sensitive plasma diabetes insipidus is partial; however, the bright spot disappears as
AVP measurements make routine use of the assay impractical for AVP-secreting neurons diminish in numbers and AVP deficiency
most clinical laboratories, there has been much interest in the mea- gets worse.179
surement of plasma copeptin, which offers a biologic surrogate for The posterior pituitary bright spot decreases with a prolonged
the measurement of plasma AVP. Copeptin is the C-terminal locus stimulus to vasopressin secretion180 and has been variably reported
of the AVP precursor provasopressin, and it is co-secreted from in other polyuric disorders. In primary polydipsia the bright spot
the neurohypophysis in equimolar amounts with AVP in response usually is seen.174,181 In nephrogenic diabetes insipidus the bright
to the same stimuli. It is much more stable ex  vivo, so sample spot has been reported to be absent in some patients174 but pres-
handling is straightforward. A sandwich immunoluminomet- ent in others.174,182 Patients with nephrogenic diabetes insipi-
ric assay is used rather than a radioimmunoassay, which enables dus have high levels of vasopressin in plasma and are chronically

Downloaded for FK UMI Makassar (mahasiswafkumi01@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier
on November 01, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
Chapter 10  Posterior Pituitary 313

dehydrated, so the posterior pituitary might be depleted of vaso- Most patients prefer desmopressin tablets (0.1 and 0.2 mg); the
pressin stores. Similarly, with the osmotic stress of untreated dia- oral preparations are effective, and unlike nasal sprays absorption
betes mellitus or the transient diabetes insipidus of pregnancy the is not impaired with nasal infections. However, intestinal pepti-
posterior pituitary can be depleted of vasopressin, and the bright dase activity can result in increased degradation before absorption,
spot is transiently lost only to reappear with resolution of the so the tablets should be taken 1 hour before or 2 hours after meals.
underlying condition.180,183 Desmopressin melt (60, 120, and 240 μg) is reported to be more
Imaging of the hypothalamus is also important to establish acceptable in some children.195 As the degree of AVP deficiency is
whether structural abnormalities of the neurohypophysis are variable among patients, the dosage and frequency of desmopres-
responsible for CDI. Because 90% of the vasopressin neurons sin are also highly variable.196–198 Some patients with partial CDI
must be destroyed to produce symptomatic CDI,113,184 a mass need a single dose before bed to prevent nocturia, whereas some
lesion must either destroy a large area of the hypothalamus or patients with complete AVP deficiency need desmopressin two to
be located where the tracks of the supraoptic and paraventricu- four times daily. The total duration of action of desmopressin will
lar nuclei converge at the base of the hypothalamus, superior to usually be 6 to 18 hours depending on the route of administration.
the pituitary stalk. Pituitary adenomas confined within the sella When a dose of desmopressin is sufficient to elicit a stable ther-
do not cause diabetes insipidus.113 Diseases of the pituitary stalk apeutic response, further increasing (e.g., doubling) the dose pro-
commonly cause CDI and are listed in Table 10.1. When there is duces a moderate increase in duration of only a few hours,196,197
a diagnosis of CDI, thickening of the stalk is usually associated consistent with the half-life of desmopressin in plasma.196 The
with absence of the posterior pituitary bright spot, and a search for maximum dose rarely exceeds 0.2 mg orally or 20 μg intrana-
systemic diseases is indicated.101 A thickened stalk with coexistent sally (two sprays) given two or three times a day (usually three
anterior pituitary deficiency is especially suggestive of etiologic for tablets and two for intranasal).197 For greater flexibility with
systemic disease.185,186 A recent retrospective study has suggested intranasal administration, the patient can be taught to use the
that T2 DRIVE sequence images of the pituitary stalk are so reli- rhinal catheter.192 Parenterally administered desmopressin (2-mL
able that the need for gadolinium enhancement is unnecessary.187 vials of 4 μg/mL) is only needed for ambulatory patients during
In the absence of structural causes of CDI on MRI, it is impor- an intercurrent illness.197 Parenterally administered desmopres-
tant to recognize that in some cases of germinoma the tumor may sin gives virtually identical therapeutic response when given as an
not be detectable on the initial scans. Therefore, particularly in intravenous bolus, intramuscularly or subcutaneously,197 and the
children and young adults who are more vulnerable to germ cell parenteral administration is 5 to 20 times as potent as an intrana-
tumors, markers such as beta human chorionic gonadotropin sally administrated dose.192,197 Published studies have reported an
(βhCG) and alpha-fetoprotein (AFP) should be measured in the increased antidiuretic response to desmopressin in women and in
blood and in the cerebrospinal fluid if the index of clinical suspi- the elderly.199
cion is high enough. In addition, repeat MRI should be obtained Hyponatremia is a common complication of desmopressin
every 3 to 6 months for the first 2 years of follow-up.185,186,188,189 therapy. Mild hyponatremia (plasma sodium 131–134 mmol/L)
If follow-up imaging shows a decrease in size of the stalk, a likely occurs in 27% of ambulatory blood samples in patients with
diagnosis is infundibulo-neurohypophysitis.190 Increases in size intact thirst, and in 15% of blood samples serum sodium is less
often occur initially with infundibuloneurohypophysitis, but than 130 mmol/L.80 This reflects the tendency in humans to drink
sustained increases over a 2-year period necessitate biopsy of the for pleasure or to drink socially; as desmopressin produces a con-
pituitary stalk. tinuous antidiuretic effect, the usual physiologic suppression of
Other investigations are dictated by clinical circumstances; if endogenous AVP that occurs during drinking does not occur, with
histiocytosis X is suspected, a radiologic skeletal survey should consequent dilutional hyponatremia. The hyponatremia seen due
be performed. Sarcoidosis can be revealed by classic radiographic to desmopressin is severe enough to precipitate hospital admis-
or computed tomography (CT) thorax changes or elevation of sion in 6% of patients.80 Hyponatremia is particularly an issue
serum angiotensin-converting enzyme activity. When MRI of in infants, who consume a large part of their calories as liquid
the pituitary is normal, autoimmune variants of CDI should be formula or breast milk, and in whom treatment with desmopres-
considered; studies of patients classified as “idiopathic” CDI have sin can cause erratic serum sodium consequences and the risk of
revealed the presence of antibodies to AVP-neurons and a high symptomatic hyponatremia.200 The risk of dilutional hypona-
incidence of related autoimmune conditions, most commonly tremia can be reduced by any one of three desmopressin dosing
thyroid disease.111  schedules:
1. Omitting a full dose once a week to allow an aquaresis to occur;
Treatment of Polyuric Conditions this is effective, but unpleasant for the patient
2. Delaying a desmopressin dose once or twice weekly until the
Central Diabetes Insipidus in Ambulatory Patients patient urinates two or three times
Treatment of CDI depends on regulation of water intake and 3. Delaying each dose of desmopressin until the patient begins to
excretion. Because thirst is normal in CDI, water intake self-reg- urinate
ulates. Replacement of absent AVP is generally done with des- In practice, individual patients vary as to which of these meth-
mopressin,191,192 a synthetic analog of vasopressin in which the ods they prefer.
substitution of d-arginine markedly reduces pressor activity and Hypernatremia is much less common in ambulant patients,
removing the terminal amine increases the half-life (see Fig. 10.1). as the intact thirst mechanism ensures that fluid intake is
These two changes produce an agent nearly 2000 times more spe- sufficient to supply physiologic needs; only 1% of ambulant
cific for antidiuresis than naturally occurring l-arginine vasopres- plasma sodium concentrations are above the normal reference
sin.193 Desmopressin is available as tablets for oral administration, range.80 In patients with adipsic DI, ambulatory hypernatre-
a lyophilate for sublingual administration (oral melt), a solution mia is much more common, occurring in up to 20% of blood
for intranasal administration, and a solution for parenteral use.194 samples. 

Downloaded for FK UMI Makassar (mahasiswafkumi01@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier
on November 01, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
314 SE C T I O N I I    Hypothalamus and Pituitary

Central Diabetes Insipidus in Hospitalized Patients desmopressin they are at risk for developing hyponatremia. A care-
Although hypernatremia is rare in ambulatory patients with nor- fully monitored regimen of a fixed dose of desmopressin to main-
mal thirst, the incidence is much higher during intercurrent ill- tain chronic antidiuresis and a prescribed volume of fluid intake
ness, particularly if the patient is vomiting, and unable to absorb is preferable.205,206 Daily weight can be used to guide intake, and
oral desmopressin. As a result, close attention must be paid to fluid regular follow-up with measurement of serum sodium is essential
balance when patients are admitted as emergencies. If the patient to ensure that these patients do not develop water intoxication
develops severe hypernatremic dehydration, consideration should with hyponatremia or recurrent dehydration with hypernatremia.
be given to anticoagulation to prevent thrombotic complications. Patients with adipsic DI have been reported to develop throm-
Equally, if a patient is on intravenous fluids, regular electrolyte botic complications when severely dehydrated, and they should
estimations are necessary to ensure that dilutional hyponatremia be prophylactically anticoagulated when admitted with severe
does not develop, particularly if hypotonic fluids such as dextrose intercurrent illness. Although management of water balance is the
solutions are used. In patients with abnormal thirst, hypernatre- clinical priority for endocrinologists, it is important to holistically
mia and hyponatremia are both much more common, and the approach the hypothalamic complication associated with adipsic
need for careful monitoring of electrolytes and fluid balance is DI. Sleep apnea can respond to treatment with continuous positive
more crucial.80  airway pressure (CPAP) or modafinil, and treatment for associated
hypothalamic obesity is also important.117 This group of patients
Central Diabetes Insipidus in Neurosurgical Patients has high mortality rates and requires very careful management. 
Transient polyuria due to CDI is not uncommon after pituitary
surgery; it is more common after extensive surgery for larger pitu-
itary adenomas or for craniopharyngioma. The diagnosis is made
Treatment of Diabetes Insipidus in Pregnancy
by the presence of the classic triad of polyuria, hypernatremia, and Desmopressin is the only therapy recommended for treatment
dilute urine after exclusion of other possibilities such as diabetes of diabetes insipidus during pregnancy. Desmopressin has 2% to
mellitus and mannitol therapy. Sometimes diuresis after surgery 25% the oxytocic activity of lysine vasopressin or arginine vaso-
is the result of water retention during the procedure. Vasopressin pressin193 and can be used with minimal stimulation of the oxy-
is released during surgical procedures and administered fluid is tocin receptors in the uterus.141,207 Desmopressin is not destroyed
retained. When the stress of surgery abates, the vasopressin level by the cysteine aminopeptidase (oxytocinase) of pregnancy141,208
falls and retained fluid is excreted. If an attempt is made to match and is reported to be safe for both the mother and the child.209,210
the urine output with further fluid infusion, persistent polyuria During delivery, patients can maintain adequate oral intake and
might be mistaken for diabetes insipidus. If in doubt, fluid should are therefore safe to continue administration of desmopressin.
be withheld until there is a modest increase in serum sodium. If Physicians should be cautious about overadministration of fluid
the urine output decreases and the serum sodium remains nor- parenterally during delivery because these patients will not be
mal, the polyuria was due to excretion of physiologically retained able to excrete the fluid and can develop water intoxication and
fluid. If the serum sodium begins to rise while urine osmolality is hyponatremia. After delivery, plasma oxytocinase decreases and
low and there is a positive response to administered ­desmopressin, patients can recover completely or be asymptomatic with regard
the diagnosis of diabetes insipidus can be established.201 If the to fluid intake and urine excretion. 
duration of diabetes insipidus is quite transient, it is acceptable
to treat this simply with fluid replacement, parenterally or orally.
However, most patients who develop diabetes insipidus require
Treatment of Nephrogenic Diabetes Insipidus
desmopressin 0.5 to 2 μg subcutaneously, intramuscularly, or Adequate water intake is the mainstay of treatment of NDI as phar-
intravenously. Urine output will be reduced in 1 to 2 hours and macologic reversal of the renal resistance to AVP is rarely possible.
the duration of effect is 6 to 24 hours. Care should be taken that Nephrogenic diabetes insipidus does not respond to desmopressin
hypotonic intravenous fluids are not given excessively after admin- therapy, although occasionally partial defects have limited response
istering desmopressin, as the combination can lead to profound to high doses of desmopressin.211,212 In congenital NDI, therapy
hyponatremia.202 Because there is always the possibility of devel- aimed at reducing symptomatic polyuria is addressed primarily by
oping the triphasic response due to pituitary stalk damage, it is inducing plasma volume contraction via a low sodium diet and
recommended that polyuria should be recurrent before a decision a thiazide diuretic. It has been hypothesized that contraction of
to administer subsequent doses of desmopressin is made.203 extracellular fluid volume, decreased glomerular filtration rate,
Acute diabetes insipidus after traumatic brain injury can be proximal tubular sodium and water reabsorption, and decreased
treated similarly to that recommended for patients following pitu- delivery of fluid to the collecting duct result in a decreased volume
itary surgery; as the patient with head injury is more likely to be of urine.213 Studies have also demonstrated that thiazide diuret-
comatose or cognitively impaired, and thus unable to depend on ics can increase aquaporin 2 independent of vasopressin.214 All the
thirst, hypernatremia is more likely to develop. Because a coma- thiazide diuretics appear to have similar effects. Potassium replace-
tose patient must be given fluids parenterally, it is important to ment and/or coadministration of a potassium-sparing antidiuretic
monitor serum sodium concentration regularly to check for dilu- may be necessary, as hypokalemia can worsen renal resistance to
tional hyponatremia. Persistent diabetes insipidus has been dem- AVP. The effect of thiazides can be augmented by coadministration
onstrated in prospective studies to be a predictor of fatal outcome of nonsteroidal anti-inflammatory drugs (NSAIDs), but this com-
after traumatic brain injury, perhaps because it is a manifestation bination is nephrotoxic, and careful monitoring of renal function
of rising intracranial pressure and imminent herniation.204  is mandatory. Selective cyclooxygenase 2 (COX2) inhibitors with
less gastrointestinal (GI) effect have been reported to decrease water
Adipsic Diabetes Insipidus loss, but long-term safety has not been documented.156
The combination of attenuated thirst and diabetes insipidus con- Drug-induced nephrogenic diabetes insipidus should be
fers high risk of severe hypernatremia. As well, when treated with treated by stopping the offending agent if possible. Persistence

Downloaded for FK UMI Makassar (mahasiswafkumi01@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier
on November 01, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
Chapter 10  Posterior Pituitary 315

of nephrogenic diabetes insipidus can be treated by hydrochloro- a greater emphasis on fluid replacement to avoid dehydration and
thiazide and amiloride. Plasma volume contraction produced by hypernatremia.222 
thiazide diuretics can decrease lithium excretion and predispose
to lithium toxicity.160,215 The diuretic amiloride blocks sodium Panhypopituitarism
channels in the luminal membrane of the collecting duct cells Patients with coexistent diabetes and anterior pituitary deficiency
and inhibits lithium reabsorption, a unique advantage in treat- are at risk of developing hyponatremia, as cortisol deficiency is
ing lithium-induced nephrogenic diabetes insipidus.216 In animal associated with impaired water excretion. Patients who miss their
studies of lithium-induced nephrogenic diabetes insipidus, treat- hydrocortisone doses are vulnerable to desmopressin-induced
ment with amiloride increased levels of both aquaporin 2 and urea hyponatremia. 
transporters.157
Studies have reported the possibility of rescuing mutant recep- Hypertonic Encephalopathy
tors in NDI. In autosomal dominant type 2 NDI, the misfolded Hypertonic encephalopathy is uncommon in diabetes insipidus
receptor protein is trapped in the quality control system of the and is only seen when there is inadequate fluid intake in an adip-
endoplasmic reticulum, but in some cases the defect involves sic patient or in a patient who is unconscious and not receiving
transport of the receptor, and were the receptor to reach the cell adequate fluid supplementation. Conditions other than diabetes
membrane it would respond to vasopressin. V2 receptor antago- insipidus are more common causes of hypernatremic encephalop-
nists (vaptans) have been reported to act as pharmacologic chap- athy. The condition is associated with loss of hypotonic fluids by
erones that combine with the misfolded receptor, changing the the kidney or the intestines, or from insensible losses; or, it might
confirmation to allow maturation and transport to the plasma occur following administration of hypertonic sodium containing
membrane where vasopressin (in excess of the vaptans) would fluids or hyperalimentation.223 Hypernatremia leads to osmotic
cause the receptor to be activated.217–219 Similar studies of rescue movement of water out of cells and cellular dehydration. Stud-
have recently been reported with nonpeptide V2 receptor agonists. ies indicate that in the brain idiogenic osmoles, mainly polyols,
These agonists combine with the mutant receptor trapped in the trimethylamines, and amino acids, are generated intracellularly so
endoplasmic reticulum and allow maturation of the mutant recep- the degree of cell shrinkage is less than would occur based on the
tor. The rescued receptor is then inserted into the cell membrane degree of hypernatremia.224 Loss of water from the brain occurs
and when stimulated by vasopressin or desmopressin generates in minutes and electrolytes enter the brain in a few hours, but
sufficient cAMP to move aquaporin 2 from the cytoplasm to the the increase in organic osmoles occurs over several days.225 When
cell membrane to enhance water transport.218,219 Nonpeptide fluid is replaced, intracellular organic osmoles dissipate more
antagonists working as chaperones is a potential new treatment of slowly than the decrease in osmolality of extracellular fluid. This
nephrogenic diabetes insipidus, especially in patients with a partial asynchrony increases the potential for cerebral edema and wors-
disorder.220 Sequencing of genes in all families with NDI is rec- ening of the neurologic condition with overzealous treatment of
ommended because of the small size of the genes to be sequenced hypernatremia.225 In most cases of diabetes insipidus seen follow-
and because of the value of the information.149 In X-linked disor- ing neurosurgery or head injury the diagnosis will be made within
ders, carrier females can be distinguished from noncarrier females, a few hours and therapy should be instituted promptly. In cases
so it will be known which sibling’s children are at risk and require where the duration of the hypernatremia is not known, the degree
special observation at birth. Molecular testing of newborns will of correction of hypernatremia should not exceed 0.5 mEq/L/
confirm the need for long-term treatment to avoid complications hour to prevent cerebral edema and convulsions.223,225 
in the affected children and obviate the need for water deprivation
or other testing in unaffected children.152,221 The Syndrome of Inappropriate Antidiuresis
Treatment of Primary Polydipsia The SIAD is produced when plasma levels of vasopressin are ele-
Treatment of primary polydipsia entails reduction of excessive vated at times when the physiologic secretion of vasopressin from
fluid intakes, best done in a graded fashion to allow patients to the posterior pituitary would normally be osmotically suppressed.
slowly achieve a level of intake that reduced urine volume below The clinical abnormality is a decrease in the osmotic pressure of
polyuric levels (50 mL/kg BW). Measures to reduce mouth dry- body fluids, so the hallmark of SIAD is hypoosmolality. This led
ness (e.g., ice chips, hard candy to stimulate salivary flow) are use- to the identification of the first well-described cases of this dis-
ful adjuncts to reduce thirst. Pharmacologic therapies have been order in 1957226 and the subsequent clinical investigations that
tried but without consistent evidence of success.  resulted in delineation of the essential characteristics of the syn-
drome.227 Although initially called the syndrome of inappropriate
Diabetes Insipidus in Association With Other antidiuretic hormone (SIADH) secretion, the term SIAD is more
Therapeutic Decisions appropriate in view of the inability to measure elevated vasopres-
sin levels in some patients with this disorder. It is necessary to first
Routine Surgical Procedures review hypoosmolality and hyponatremia before discussing details
In all cases there should be preoperative consultation of the sur- that are specific to SIAD.
geon, the anesthesiologist, and the endocrinologist/nephrologist.
For most routine surgical procedures the patient is not uncon- Hypoosmolality and Hyponatremia
scious for a sufficiently long period of time to require anything
more that administration of the usual dose of desmopressin and Incidence
careful monitoring of fluids during the surgery to prevent overhy- Hypoosmolality is the most common disorder of fluid and
dration. If the patient is taking desmopressin orally and is NPO, electrolyte balance encountered in hospitalized patients. The
a parenteral dose can be administered before the procedure. Close incidence and prevalence of hypoosmolar disorders depend on
monitoring of serum sodium is essential. In NDI there should be the nature of the patient population studied as well as on the

Downloaded for FK UMI Makassar (mahasiswafkumi01@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier
on November 01, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
316 SE C T I O N I I    Hypothalamus and Pituitary

laboratory methods and criteria used to diagnose hyponatremia. relative proportion of plasma volume that is occupied by the
Most investigators have used the serum sodium concentration excess lipids or proteins.236 However, the increased protein or
([Na+]) to determine the clinical incidence of hypoosmolality. lipid will not appreciably change the total number of solute par-
When hyponatremia is defined as a serum [Na+] of less than ticles in solution, so the directly measured plasma osmolality will
135 mEq/L, prevalences as high as 15% to 38% have been not be significantly affected. Measurement of serum [Na+] by
observed in studies of both acutely and chronically hospital- ion-specific electrodes, which is now commonly used by most
ized patients.228,229 However, incidences decrease to the range clinical laboratories, is less influenced by high concentrations
of 1% to 4% when only patients with serum [Na+] under 130 to of lipids or proteins than is measurement of serum [Na+] by
131 mEq/L are included, which represents a more appropriate flame photometry. However, this can still occur if the electrode
level at which to define the occurrence of clinically significant measurement is done using a diluted sample of the serum. The
cases of this disorder.230 Even using these more stringent criteria, second situation in which hyponatremia does not reflect true
incidences from 7% to 53% have been reported in institution- plasma hypoosmolality occurs when high concentrations of
alized geriatric patients.231 Although hyponatremia and hypoos- effective solutes other than Na+ are present in the plasma. The
molality are quite common, the majority of cases are relatively initial hyperosmolality produced by the additional solute causes
mild and most are acquired during the course of hospitaliza- an osmotic shift of water from the ICF to the ECF, which in turn
tion. Nonetheless, hyponatremia is important clinically because produces a dilutional decrease in serum [Na+]. Once equilibrium
(1) severe hypoosmolality (serum [Na+] levels <120 mEq/L) is between both fluid compartments is achieved, the total effective
associated with substantial morbidity and mortality,232 (2) even osmolality remains relatively unchanged. This situation most
relatively mild hypoosmolality can quickly progress to more commonly occurs with hyperglycemia and represents a frequent
dangerous levels during the course of therapeutic management cause of hyponatremia in hospitalized patients, accounting for
of other disorders, (3) overly rapid correction of hyponatremia up to 10% to 20% of all cases.234 Misdiagnosis of true hypoos-
can itself cause severe neurologic morbidity and mortality,233 molality in such cases can be avoided by measuring plasma
and (4) it has been observed that mortality rates are from 3-fold osmolality directly or, alternatively, by correcting the measured
to 60-fold higher in patients with even asymptomatic degrees serum [Na+] for the glucose elevation. Traditionally this correc-
of hypoosmolality compared to normonatremic patients.234,235  tion factor has been 1.6 mEq/L for each 100-mg/dL increase in
serum glucose concentration above normal levels,237 but some
Osmolality, Tonicity, and Serum [Na+] studies have shown a more complex relation between hyper-
As discussed, the osmolality of body fluid normally is maintained glycemia and serum [Na+] and reported that a more accurate
within narrow limits for each individual by osmotically regulated correction factor is closer to 2.4 mEq/L.238 When the plasma
vasopressin secretion and thirst. Plasma osmolality can be deter- contains significant amounts of unmeasured solutes, such as
mined directly by measuring the freezing-point depression or osmotic diuretics, radiographic contrast agents, and some toxins
the vapor pressure of plasma. Alternatively, it can be calculated (ethanol, methanol, and ethylene glycol), plasma osmolality can-
indirectly from the concentrations of the three major solutes in not be calculated accurately, and in these situations osmolality
plasma: must be ascertained by direct measurement. 
pOsm mOsm/kg H2 O = 2 × [Na + ] (mEq/L)
+ glucose (mg/dL) / 18 + blood urea
Pathogenesis of Hypoosmolality
nitrogen (mg/dL) / 2.8 Water moves freely between the ICF and ECF, consequently
osmolality will always be equivalent in both of these fluid com-
Direct measure and calculation produce comparable results partments. Since the bulk of body solute is comprised of elec-
under most conditions. However, while either of these meth- trolytes, namely the exchangeable Na+ (Na+E) in the ECF and
ods produce valid measures of total osmolality, this is not always the exchangeable K+ (K+E) in the ICF along with their associated
equivalent to the effective osmolality, which is commonly referred anions, total body osmolality (OSMT) will largely be a function of
to as the “tonicity” of the plasma. Only solutes such as Na+ and Cl- these parameters239:
that are impermeable to the cell membrane and remain relatively
compartmentalized within the extracellular fluid (ECF) space are OSMT = OSMECF = OSMICF
effective solutes, because these solutes create osmotic gradients
across cell membranes and regulate the osmotic movement of OSMT = (ECF solute + ICF solute) / body water
water between the intracellular fluid (ICF) compartment and the
ECF compartment. Solutes that readily permeate cell membranes OSMT (2 × NaE+ + 2 × KE+ + nonelectrolyte solute) / body water
(e.g., urea, ethanol, and methanol) are not effective solutes. There-
fore only the concentrations of effective solutes in plasma should
be used to ascertain whether clinically significant hyperosmolality According to this definition, the spresence of plasma hypoos-
or hypoosmolality is present. molality indicates a relative excess of water to solute in the ECF.
Sodium and its accompanying anions represent the major This can be produced either by an excess of body water, resulting
effective plasma solutes, so hyponatremia and hypoosmolality in a dilution of remaining body solute, or by a depletion of body
are usually synonymous. However, there are two situations in solute, either Na+ or K+, relative to body water. This classification
which hyponatremia will not reflect true hypoosmolality. The is an oversimplification, because most hypoosmolar states involve
first is pseudohyponatremia, which is produced by marked eleva- significant components of both solute depletion and water reten-
tions of either lipids or proteins in plasma. If serum [Na+] is tion. Nonetheless, it is conceptually useful for understanding the
measured by flame photometry, the concentration of sodium mechanisms underlying the pathogenesis of hypoosmolality and
per liter of plasma is artifactually decreased because of the larger as a framework for therapy of hypoosmolar disorders.

Downloaded for FK UMI Makassar (mahasiswafkumi01@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier
on November 01, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
Chapter 10  Posterior Pituitary 317

Solute Depletion Some dilutional disorders do not fit well into either category,
Depletion of body solute can result from any significant losses of specifically the hyponatremia that sometimes occurs in patients
ECF. Body fluid losses by themselves rarely cause hypoosmolality who ingest large volumes of beer with little food intake for pro-
because excreted or secreted body fluids are usually isotonic or longed periods, called “beer potomania.”243 Even though the
hypotonic relative to plasma and therefore tend to increase plasma volume of fluid ingested may not seem sufficiently excessive to
osmolality. When hypoosmolality accompanies ECF losses, it is overwhelm renal diluting mechanisms, free water excretion is lim-
the result of replacement of body fluid losses by more hypotonic ited by very low urinary solute excretion, thereby causing water
solutions either by drinking or by infusion, thereby diluting the retention and dilutional hyponatremia. 
remaining body solutes. If the solute losses are marked, these
patients show signs of volume depletion (e.g., addisonian crisis). Adaptation to Hyponatremia: ICF and ECF Volume
However, such patients often have a more deceptive clinical pre- Regulation
sentation because the volume deficits have been partially replaced. Many past studies have suggested that the combined effects of water
Moreover, they may not manifest signs or symptoms of cellular retention plus urinary solute excretion cannot adequately explain
dehydration because osmotic gradients will draw water into the the degree of plasma hypoosmolality observed in patients.227,244
ICF, which is relatively hypertonic to the solute-depleted ECF. This observation led to the theory of cellular inactivation of solute,
Therefore clinical evidence of hypovolemia strongly supports sol- which suggested that as ECF osmolality falls, water moves into
ute depletion as the cause of plasma hypoosmolality, but absence cells along osmotic gradients, thereby causing the cells to swell;
of clinically evident hypovolemia never completely eliminates this at some point during this volume expansion, the cells theoreti-
as a possibility. Although ECF solute losses are responsible for cally osmotically “inactivate” some of their intracellular solutes as
most cases of depletion-induced hypoosmolality, ICF solute loss a defense mechanism to prevent continued cellular swelling with
can also cause hypoosmolality as a result of osmotic water shifts subsequent detrimental effects on cell function and survival. This
from the ICF into the ECF. This mechanism contributes to some effect would decrease the intracellular osmolality allowing water to
cases of diuretic-induced hypoosmolality in which depletion of shift back out of the ICF into the ECF, thereby further worsening
total body K+ often occurs.240  the dilution-induced hypoosmolality. Despite the appeal of this
theory, its validity has never been demonstrated conclusively in
Water Retention either human or animal studies. An alternative theory is that cell
Despite the importance of solute depletion in some patients, volume is maintained under hypoosmolar conditions by extrusion
most cases of clinically significant hypoosmolality are caused by of intracellular solutes such as potassium.245 Whole brain volume
increases in total body water rather than by primary losses of regulation via electrolyte losses was first described by Yannet246
extracellular solute. This can occur because of either impaired and has long been recognized as the mechanism by which the
renal free water excretion or excessive free water intake. The for- brain is able to adapt to hyponatremia and limit brain edema to
mer accounts for most hypoosmolar disorders because normal sublethal levels.247 Following the recognition that low molecular
kidneys have sufficient diluting capacity to allow excretion of 18 weight organic compounds, called organic osmolytes, also consti-
to 24 L/day of free water. Intakes of this magnitude are occasion- tuted a significant osmotic component of a wide variety of cells,
ally seen in some psychiatric patients but not in most patients studies demonstrated the accumulation of these compounds in
with SIAD in whom fluid intakes average 2 to 3 L/day.241 Con- response to hyperosmolality in both kidney248 and brain249 tis-
sequently, dilutional hypoosmolality usually is the result of an sue and conversely that the brain also loses organic osmolytes in
abnormality of renal free water excretion. The renal mechanisms addition to electrolytes during volume regulation to hypoosmolar
responsible for impairments in free water excretion can be sub- conditions in experimental animals250,251 and human patients.252
grouped according to whether the major impairment in free These losses occur relatively quickly (within 24–48 hours in rats)
water excretion occurs in proximal or distal parts of the nephron, and can account for as much as one-third of the brain solute losses
or both. Any disorder that leads to a decrease in glomerular filtra- during hyponatremia.253 Such coordinate losses of both electro-
tion rate causes increased reabsorption of both Na+ and water in lytes and organic osmolytes from brain cells allow effective regula-
the proximal tubule. As a result, the ability to excrete free water tion of brain volume during chronic hyponatremia.
is limited because of decreased delivery of tubular fluid to the Although recent studies of volume regulation during hypo-
distal nephron. Disorders that cause a decreased glomerular fil- natremia have focused on the brain, all cells regulate volume by
tration rate in the absence of significant ECF fluid losses are, for cellular losses of both electrolyte and organic solutes to varying
the most part, edema-forming states associated with decreased degrees. However, volume regulatory processes are not limited to
effective arterial blood volume (EABV) and secondary hyperal- cells. In most cases of hyponatremia induced by stimulated antidi-
dosteronism.242 Even though these conditions are characterized uresis and water retention, natriuresis also regulates the volumes of
by increased proximal reabsorption of both Na+ and fluid, water the ECF and intravascular spaces. Both experimental and clinical
retention also results from increased distal reabsorption caused observations are consistent with ECF volume regulation via sec-
by nonosmotic baroreceptor-mediated stimulated increases in ondary solute losses. First, the concentrations of most blood con-
plasma vasopressin levels. Distal nephron impairments in free stituents other than Na+ and Cl- are not decreased in patients with
water excretion are characterized by an inability to dilute tubu- SIAD,254 suggesting that plasma volume is not nearly as expanded
lar fluid maximally. These disorders are usually associated with as would be predicted simply by the measured decreases in serum
abnormalities in the secretion of vasopressin. Just as depletion- [Na+]. Second, an increased incidence of hypertension has never
induced hypoosmolar disorders usually include an important been observed in patients with SIAD, again evidence against sig-
component of secondary impairments of free water excretion, nificant expansion of the arterial blood volume. Third, results of
most dilution-induced hypoosmolar disorders also involve sig- animal studies in both dogs255 and rats256 have indicated that a
nificant degrees of secondary solute depletion. This is described significant component of chronic hyponatremia is attributable
later with SIAD. to secondary Na+ losses rather than water retention; the relative

Downloaded for FK UMI Makassar (mahasiswafkumi01@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier
on November 01, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
318 SE C T I O N I I    Hypothalamus and Pituitary

contributions from water retention versus sodium loss vary with


the duration and severity of the hyponatremia: Water retention
was found to be the major cause of decreased serum [Na+] in the [S]ICF [S]ECF
first 24 hours of induced hyponatremia in rats, but Na+ depletion
then became the predominant etiologic factor after longer periods A
(7–14 days) of sustained hyponatremia, particularly at very low
H2O
(<115 mEq/L) serum [Na+] levels.256 Finally, multiple studies of
body fluid compartment volumes in hyponatremic patients have
not demonstrated either plasma or ECF expansion. For example,
a report of body fluid space measurements using isotope dilution [S]ICF [S]ECF
techniques in hyponatremic and normonatremic patients with
small cell lung carcinoma showed no differences between the two
groups with regard to exchangeable sodium space, ECF volume by B
35SO distribution, or total body water.257 Fig. 10.5 schematically
4
illustrates the volume regulatory processes that occur in response
to water retention induced by inappropriate antidiuresis. 
K+
Differential Diagnosis of Hyponatremia and ICF osmolytes
Hypoosmolality VRD H 2O
H 2O
ECF
Because of the multiplicity of disorders causing hypoosmolality VRD
and the fact that many involve more than one pathologic mecha- C Na+, Cl–
nism, a definitive diagnosis is not always possible at the time of
initial presentation. Nonetheless, an approach based on clinical
parameters of ECF volume status and urine sodium concentration
generally allows a sufficient categorization for appropriate deci- [S]ECF
[S]ICF [S]ECF [S]ICF
sions regarding initial therapy and further evaluation.

Decreased Extracellular Fluid Volume D E


Clinically detectable hypovolemia always signifies total body sol-
ute depletion. A low urine [Na+] indicates a nonrenal cause and • Fig. 10.5  Schematic illustration of potential changes in whole-body fluid
compartment volumes at various times during adaptation to hyponatre-
an appropriate renal response. A high urine [Na+] indicates renal mia. (A) Under basal conditions, the concentrations of effective solutes
causes of solute depletion are more likely. Therapy with thiazide in the extracellular fluid ([S]ECF) and in the intracellular fluid ([S]ICF) are in
diuretics is the most common cause of renal solute losses,240 par- osmotic balance. (B) During the first phase of water retention resulting from
ticularly in the elderly,258 but mineralocorticoid deficiency as a inappropriate antidiuresis, the excess water distributes across total body
result of adrenal insufficiency or mineralocorticoid resistance must water, causing expansion of both ECF and ICF volumes (dotted lines), with
be considered as well as (less commonly) renal solute losses due to equivalent dilutional decreases in both [S]ICF and [S]ECF. (C) In response to
salt-wasting nephropathy (e.g., polycystic kidney disease, intersti- the volume expansion, compensatory volume regulatory decreases (VRD)
tial nephritis, or chemotherapy).  occur to reduce the effective solute content of the ECF (via pressure diure-
sis and natriuretic factors) and the ICF (via increased electrolyte and osmo-
lyte extrusion mediated by stretch-activated channels and downregulation
Increased Extracellular Fluid Volume
of synthesis of osmolytes and osmolyte uptake transporters). (D and E) If
Clinically detectable hypervolemia always signifies total body both processes go to completion, such as under conditions of fluid restric-
Na+ excess. In these patients hypoosmolality results from an even tion, a final steady state can be reached in which ICF and ECF volumes
greater expansion of total body water caused by a marked reduc- have returned to normal levels but [S]ICF and [S]ECF remain low. In most
tion in the rate of water excretion (and sometimes an increased cases, this final steady state is not reached, and moderate degrees of ECF
rate of water ingestion). The impairment in water excretion is sec- and ICF expansion persist, although they are significantly less than would
ondary to a decreased EABV,242 which increases the reabsorption be predicted from the decrease in body osmolality (D). Consequently, the
of glomerular filtrate not only in the proximal nephron but also in degree to which hyponatremia is the result of dilution due to water reten-
the distal and collecting tubules by stimulated secretion of vaso- tion versus solute depletion from volume regulatory processes can vary
pressin. These patients generally have a low urine [Na+] because markedly, depending on which phase of adaptation the patient is in and
the relative rates at which the different compensatory processes occur. For
of secondary hyperaldosteronism. However, under certain condi-
example, delayed ICF VRD can worsen hyponatremia because of shifts
tions urine [Na+] may be elevated if there is concurrent diuretic of intracellular water into the ECF as intracellular organic osmolytes are
therapy, a solute diuresis (e.g., glucosuria in diabetics), or after extruded and subsequently metabolized; this likely accounts for some
successful treatment of the underlying disease (e.g., improved car- component of the hyponatremia that was unexplained by the combina-
diac output in patients with congestive heart failure).  tion of water retention and sodium excretion in early clinical studies. (From
Verbalis JG. Hyponatremia: epidemiology, pathophysiology, and therapy.
Normal Extracellular Fluid Volume Curr Opin Nephrol Hypertens. 1993;2:626–652.)
Many different hypoosmolar disorders present with euvolemia,
and measure of urinary [Na+] is an especially important first Hyponatremia from diuretic use also can present without clini-
step.259 A high urine [Na+] usually implies a distally mediated, cally evident hypovolemia, and the urine [Na+] will usually be
dilution-induced hypoosmolality such as SIAD. However, gluco- elevated.240 A low urine [Na+] suggests a depletion-induced
corticoid deficiency can mimic SIAD so closely that these two hypoosmolality from ECF losses with subsequent volume replace-
disorders are often indistinguishable in terms of water balance. ment by water or other hypotonic fluids. The solute loss often

Downloaded for FK UMI Makassar (mahasiswafkumi01@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier
on November 01, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
Chapter 10  Posterior Pituitary 319

is nonrenal, but an important exception is recent cessation of


diuretic therapy, because urine [Na+] can decrease to low values
within 12 to 24 hours after discontinuation of the drug. A low 11

Plasma vasopressin (pg/mL)


urine [Na+] also can also be seen during the recovery phase from 10
SIAD.  9
8
7 Normal
Syndrome of Inappropriate Antidiuresis 6 range
SIAD is the most common cause of euvolemic hypoosmolality 5
and it is also the single most common cause of hypoosmolality 4
of all etiologies encountered in clinical practice with prevalence 3
rates from 20% to 40% among all hypoosmolar patients.234,241 2
The clinical criteria necessary to diagnose SIAD remain basically 1
as set forth by Schwartz and Bartter in 1967:227 0
1. Decreased effective osmolality of the extracellular fluid (Posm
<275 mOsm/kg H2O). Pseudohyponatremia or hyperglycemia 230 240 250 260 270 280 290 300 310
alone must be excluded. Plasma osmolality (mOsm/kg)
2. Inappropriate urinary concentration at some level of hypoos- • Fig. 10.6  Plasma arginine vasopressin (AVP) levels in patients with the
molality. This does not mean that urine osmolality is greater syndrome of inappropriate antidiuretic hormone (SIADH) secretion as a
than plasma osmolality, only less than maximally dilute (i.e., function of plasma osmolality. Each point depicts one patient at a single
urine osmolality >100 mOsm/kg H2O). Also, urine osmolal- point in time. The shaded area represents AVP levels in normal subjects
ity need not be elevated inappropriately at all levels of plasma over physiologic ranges of plasma osmolality. The lowest measurable
osmolality, because in the reset osmostat variant form of SIAD, plasma AVP level that could be detected with this radioimmunoassay was
vasopressin secretion can be suppressed with resultant maximal 0.5 pg/mL. (From Robertson GL, Aycinena P, Zerbe RL. Neurogenic disor-
ders of osmoregulation. Am J Med. 1982;2:339–353.)
urinary dilution if plasma osmolality is decreased to sufficiently
low levels.260
3. Clinical euvolemia, as defined by the absence of signs of hypo- mOsm/kg H2O) confirms the presence of an underlying defect
volemia (orthostasis, tachycardia, decreased skin turgor, dry in free water excretion. However, water excretion is abnormal
mucous membranes) or hypervolemia (subcutaneous edema, in almost all disorders that cause hypoosmolality, whether dilu-
ascites). Hypovolemia or hypervolemia strongly suggests differ- tional or depletion induced with secondary impairments in free
ent causes of hypoosmolality. Patients with SIAD can become water excretion. Two exceptions are primary polydipsia, in which
hypovolemic or hypervolemic for other reasons, but in such hypoosmolality can rarely be secondary to excessive water intake
cases it is impossible to diagnose the underlying inappropri- alone, and the reset osmostat variant of SIAD, in which normal
ate antidiuresis until the patient is rendered euvolemic and is excretion of a water load can occur once plasma osmolality falls
found to have persistent hypoosmolality. below the new set-point for vasopressin secretion.
4. Elevated urinary sodium excretion while on a normal salt and Another supportive criterion is an inappropriately elevated
water intake. This criterion is included because of its utility in plasma vasopressin level in relation to plasma osmolality. How-
differentiating between hypoosmolality caused by a decreased ever, several factors limit the utility of vasopressin measurements
EABV, in which case renal Na+ conservation occurs, and dis- to diagnose SIAD. First, although plasma vasopressin levels are
tal dilution-induced disorders, in which urine Na+ excretion elevated in most patients with this syndrome, the elevations
is normal or increased secondary to ECF volume expansion. generally remain within the normal physiologic range and are
Patients with SIAD can have low urine Na+ excretion if they abnormal only in relation to plasma osmolality (Fig. 10.6). Sec-
subsequently become hypovolemic or solute depleted, condi- ond, 10% to 20% of patients with SIAD do not have measurably
tions that sometimes follow severe salt and water restriction. elevated plasma vasopressin levels and are at the limits of detec-
Consequently, a high urine Na+ excretion is the rule in most tion by radioimmunoassay (see Fig. 10.6).262 Third, and perhaps
patients with SIAD; its presence does not guarantee this diag- most importantly, most disorders causing solute and volume
nosis, and its absence does not rule out the diagnosis. depletion or decreased EABV are associated with elevations of
5. Absence of other potential causes of euvolemic hypoosmolality. plasma vasopressin levels secondary to nonosmotic hemody-
Most notably are hypothyroidism, hypocortisolism (Addison namic stimuli. 
disease or pituitary ACTH insufficiency), and diuretic use.
Several other criteria support but are not essential for a diag-
nosis of SIAD. Volume expansion and vasopressin acting on V1
Etiology
receptors in the kidney increase the clearance of uric acid, so hyp- Although the list of disorders associated with SIAD is long (Table
ouricemia is found with SIAD. When patients are hyponatremic, 10.2), they can be divided into several major etiologic groups.
values of uric acid are generally <4 mg/dL (<0.24 mmol/L).261 A
water-loading test is of value when there is uncertainty regarding Tumors
the etiology of modest degrees of hypoosmolality in euvolemic The most common association of SIAD is with tumors. Many
patients, but it does not add useful information if the plasma osmo- different types of tumors have been associated with SIAD, but
lality is already under 275 mOsm/kg H2O. Inability to excrete a bronchogenic carcinoma of the lung has been uniquely asso-
standard water load normally (with normal excretion defined as a ciated with SIAD since the first description of this disorder in
cumulative urine output of at least 90% of the administered water 1957.226 In virtually all cases, the bronchogenic carcinomas caus-
load within 4 hours and suppression of urine osmolality to <100 ing this syndrome have been of the small cell variety. Incidence

Downloaded for FK UMI Makassar (mahasiswafkumi01@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier
on November 01, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
320 SE C T I O N I I    Hypothalamus and Pituitary

TABLE 10.2  Common Causes of the Syndrome of but also a major innervation from brainstem cardiovascular regu-
Inappropriate Antidiuretic Hormone latory and emetic centers. Although various components of these
(SIADH) Secretion pathways have yet to be elucidated fully, many of them appear to
have inhibitory as well as excitatory components. Consequently,
Tumors any CNS disorder can potentially cause vasopressin hypersecre-
Pulmonary/mediastinal (bronchogenic carcinoma, mesothelioma, tion either by nonspecifically exciting these pathways via irritative
thymoma) foci or alternatively by disrupting them and thereby decreasing the
Nonchest (duodenal carcinoma, pancreatic carcinoma, ureteral/prostate level of inhibition. 
carcinoma, uterine carcinoma, nasopharyngeal carcinoma, leukemia)
Central Nervous System Disorders Drugs
Drug-induced hyponatremia is a common cause of hypoosmolal-
Mass lesions (tumors, brain abscesses, subdural hematoma)
Inflammatory diseases (encephalitis, meningitis, systemic lupus erythe-
ity.268 Table 10.2 lists some of the agents that have been associated
matosus, acute intermittent porphyria, multiple sclerosis) with SIAD, but new drugs are added continually. Pharmacologic
Degenerative/demyelinative diseases (Guillain-Barré syndrome, spinal agents may stimulate secretion of vasopressin, activate renal V2
cord lesions) receptors, or potentiate the antidiuretic effect of vasopressin. Not
Miscellaneous (subarachnoid hemorrhage, head trauma, acute psy- all of the drug effects are fully understood, and many appear to
chosis, delirium tremens, pituitary stalk section, transsphenoidal work through a combination of mechanisms. A particularly inter-
adenomectomy, hydrocephalus) esting and clinically important class of agents is the selective sero-
Drug Related
tonin reuptake inhibitors (SSRIs). Hyponatremia following SSRI
administration has been reported almost exclusively in the elderly,
Stimulated release of AVP (nicotine, phenothiazines, tricyclics) with rates as high as 22% to 28%, although in larger series the
Direct renal effects or potentiation of AVP antidiuretic effects (dDAVP,
oxytocin, prostaglandin synthesis inhibitors)
incidence was closer to 1 in 200.269 A similar effect is likely also
Mixed or uncertain actions (ACE inhibitors, carbamazepine and oxcar- responsible for the recent reports of severe fatal hyponatremia
bazepine, chlorpropamide, clofibrate, clozapine, cyclophosphamide, caused by use of the recreational drug 3,4-methylenedioxymeth-
3,4-methylenedioxymethamphetamine [ecstasy], omeprazole; amphetamine, “ecstasy,” which possesses substantial serotonergic
serotonin reuptake inhibitors, vincristine) activity.270 
Pulmonary Pulmonary Disorders
Infections (tuberculosis, acute bacterial and viral pneumonia, aspergil- A variety of pulmonary disorders have been associated with
losis, empyema) this syndrome, but other than tuberculosis, acute pneumonia,
Mechanical/ventilatory causes (acute respiratory failure, COPD, positive- and advanced chronic obstructive lung disease, the occur-
pressure ventilation)
rence of hypoosmolality has been noted only sporadically.
Other Causes Hypoxia stimulates secretion of vasopressin in animals,271
Acquired immunodeficiency syndrome (AIDS) and AIDS-related complex but in humans hypercarbia is more associated with abnormal
Prolonged strenuous exercise (marathon, triathlon, ultramarathon, hot- water retention. Elevated vasopressin may be limited to the
weather hiking) initial days of hospitalization, when respiratory failure is most
Senile atrophy marked. Therefore, with SIAD in nontumor pulmonary dis-
Idiopathic ease, the pulmonary disease is obvious with severe dyspnea
or extensive radiographically evident infiltrates, and the inap-
ACE, Angiotensin-converting enzyme; AVP, arginine vasopressin; COPD, chronic obstructive
pulmonary disease; dDAVP, desmopressin.
propriate antidiuresis will usually be limited to the period of
respiratory failure. Mechanical ventilation can cause SIAD via
   inappropriate secretion of vasopressin via decreased venous
return. 
of hyponatremia is as high as 11% of all patients with small cell
carcinoma,263 or 33% of cases with more extensive disease,264 Other Causes
have been reported. The high incidence of small cell carcinoma In acquired immunodeficiency syndrome (AIDS) or AIDS-related
of the lung makes it imperative that all adult patients presenting complex (ARC) and in patients with human immunodeficiency
with an otherwise unexplained SIAD be investigated thoroughly virus (HIV) infection, hyponatremia has been reported as high
and aggressively for a possible lung tumor. Head and neck can- as 30% to 38% in adults and children.272 Although there are
cers account for another group of malignancies associated with many potential etiologies, including dehydration, adrenal insuf-
relatively higher incidences of SIAD,265 and some of these tumors ficiency, and pneumonitis, from 12% to 68% of AIDS patients
have clearly been shown to synthesize vasopressin.266 A report who develop hyponatremia appear to meet criteria for a diagnosis
from a large cancer hospital showed an incidence of hyponatre- of SIAD.272 Not unexpectedly, some of the medications used to
mia for all malignancies of 3.7%, with approximately one-third of treat these patients may cause the hyponatremia, either via direct
these due to SIAD.267  renal tubular toxicity or induced SIAD.273
Elderly patients often develop SIAD without any apparent
Central Nervous System Disorders underlying etiology, and the high incidence of hyponatremia in
A large number of different CNS disorders are associated with geriatric patients231,274 suggests that the normal aging process may
SIAD, but without a common denominator linking them. This be accompanied by abnormalities of regulation of water balance
is not surprising when one considers the neuroanatomy described and of secretion of vasopressin as noted earlier. Such an effect
earlier. Magnocellular vasopressin neurons receive excitatory inputs could potentially account for the fact that drug-induced hypo-
from osmoreceptive cells located in the anterior hypothalamus, natremia occurs much more frequently in elderly patients. In a

Downloaded for FK UMI Makassar (mahasiswafkumi01@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier
on November 01, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
Chapter 10  Posterior Pituitary 321

series of 50 consecutive elderly patients meeting criteria for SIAD, of inappropriate antidiuresis, reserving SIAD only for those cases
60% remained idiopathic despite rigorous evaluation, leading the where measured plasma vasopressin levels are really inappropriate.
authors to conclude that extensive diagnostic procedures were Although the incidence of nephrogenic syndrome of inappropri-
not warranted in such elderly patients if routine history, physi- ate antidiuresis in the general population is unknown, the descrip-
cal examination, and laboratory evaluation failed to suggest an tion of Belgian kindred with this mutation suggests that it can
underlying etiology.275  present later in life as well as in childhood. 

Contribution of Natriuresis to the Hyponatremia of SIAD


Pathophysiology Since the original cases studied by Schwartz and Bartter, increased
Sources of Vasopressin Secretion renal Na+ excretion has been one of the cardinal manifestations of
Elevated plasma levels of vasopressin can be broadly divided SIAD, indeed one which later became embedded in the require-
into those associated with paraneoplastic (“ectopic”) secretion ments for its diagnosis.227 Demonstration that the natriuresis
of vasopressin or pituitary hypersecretion of vasopressin. There accompanying administration of antidiuretic hormone is not due
is substantial cumulative evidence that tumor tissue can in fact to vasopressin itself but to the volume expansion produced as a
synthesize vasopressin,276 but it is not certain that all tumors result of water retention was unequivocally shown by Leaf and
associated with SIAD do so because only about half of small cell associates even before the description of the disorder.279 Although
carcinomas have been found to contain vasopressin immunore- a negative Na+ balance occurs during the development of hypo-
activity, and many of the tumors listed in Table 10.2 have not natremia in patients with SIAD, eventually urinary sodium excre-
been carefully studied.  tion simply reflects daily sodium intake.226 Thus the term renal
sodium wasting is used to describe continued excretion of sodium
Pituitary Vasopressin Secretion—Inappropriate Versus despite being hyponatremic, but in reality there is a new steady
Appropriate state in which patients are in neutral sodium balance. Studies of
In the majority of cases of SIAD, the vasopressin secretion origi- long-term antidiuretic-induced hyponatremia in both dogs and
nates from the posterior pituitary. This is also true of over 90% of rats have indicated that a large proportion of the hyponatremia
all cases of hyponatremia, including patients with hypovolemic was attributable to secondary Na+ losses rather than to water
and hypervolemic hyponatremia.234 This raises the question of retention,255,256 but the natriuresis did not actually worsen the
what constitutes “inappropriate” secretion of vasopressin. Secre- hyponatremia; rather, it allowed volume regulation of ECF.280
tion of vasopressin in response to a hypovolemic stimulus is clearly Secondary natriuresis in patients with SIAD likely explains the
physiologically “appropriate,” but when it leads to symptomatic failure to find expanded plasma or ECF volumes using tracer dilu-
hyponatremia it could be considered inappropriate for the ECF tion techniques (see Fig. 10.5).257 
osmolality. Despite these semantic conundrums, the diagnosis of
SIAD should be based upon the original Schwartz-Bartter criteria Cerebral Salt Wasting
and specifically exclude other clinical conditions that cause known The degree to which hyponatremia might occur primarily as a
impairments in free water excretion even when these are mediated result of primary natriuresis is controversial. Cerebral salt wast-
by a secondary nonosmotic physiologic stimulation of vasopres- ing syndrome (CSWS) was proposed by Peters and associates in
sin secretion. Without maintaining these distinctions, arguable as 1950281 as an explanation for the natriuresis and hyponatremia
some may be, the definition of SIAD becomes too broad to retain that sometimes accompanies intracranial disease, particularly sub-
any practical clinical utility.  arachnoid hemorrhage in which up to one-third of patients often
develop hyponatremia. After the description of SIAD in 1957,
Patterns of Vasopressin Secretion such patients were generally assumed to have hyponatremia sec-
Studies of plasma vasopressin levels in patients with SIAD dur- ondary to vasopressin hypersecretion with a secondary natriuresis.
ing graded increases in plasma osmolality produced by hypertonic However, over the last decade clinical and experimental data have
saline administration have defined four patterns of secretion: (1) been interpreted to indicate that some patients with subarachnoid
random hypersecretion of vasopressin; (2) inappropriate non- hemorrhage and other intracranial diseases indeed have a primary
suppressible basal vasopressin release, but normal secretion in natriuresis leading to volume contraction rather than SIAD,282,283
response to osmolar changes above basal plasma osmolality; (3) and the elevated plasma vasopressin levels may be physiologically
a reset osmostat system, whereby vasopressin is secreted at an appropriate for the degree of volume contraction. With regard to
abnormally low threshold of plasma osmolality but otherwise dis- the potential mechanisms of natriuresis, both plasma and CSF lev-
plays a normal response to relative changes in osmolality; and (4) els of atrial natriuretic peptide are elevated in many patients with
low or even undetectable plasma vasopressin levels despite clas- subarachnoid hemorrhage and have been found to correlate vari-
sic clinical characteristics of SIAD.262 The pattern of SIAD that ably with hyponatremia in patients with intracranial diseases.284
occurs without measurable vasopressin secretion is not yet well However, clearly documented SIAD also is frequently associated
understood, but the positive response of one such patient to a with elevated plasma levels of atrial natriuretic peptide, so this
vasopressin V2-receptor antagonist would suggest that this may finding does not prove causality. In other disorders of hypona-
represent increased renal sensitivity to low circulating levels of tremia due to Na+ wasting (e.g., Addison disease) and diuretic-
vasopressin.277 It is surprising that no correlation has been found induced hyponatremia, infusion of saline restores normal ECF
between any of these patterns of secretion of vasopressin and the volume and plasma tonicity by shutting off the secondary vaso-
various etiologies of SIAD.262 Recent studies of pediatric patients pressin secretion. In subarachnoid hemorrhage, however, large
with hyponatremia and unmeasurable plasma vasopressin levels volumes of isotonic saline sufficient to maintain plasma volume
led to the discovery of an activating mutation of the vasopressin did not change the incidence of hyponatremia.285
V2 receptor as the cause of their inappropriate antidiuresis.278 It Those authors who have distinguished CSWS from SIAD
is more appropriate to call these cases the nephrogenic syndrome have emphasized that in CSWS the primary disorder, salt

Downloaded for FK UMI Makassar (mahasiswafkumi01@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier
on November 01, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
322 SE C T I O N I I    Hypothalamus and Pituitary

wasting, produces convincing evidence of decreased extracellular process takes a finite period of time to complete, and the more
fluid volume.286,287 There are only a few case reports of patients rapid the fall in serum [Na+] the more brain edema will be accu-
after traumatic brain injury or neurosurgery who while being mulated before the brain is able to volume regulate. Thus there
observed in the hospital have acute onset of massive diuresis and is a much higher incidence of neurologic symptoms as well as
natriuresis with clear evidence of volume contraction by weight a higher mortality in patients with acute hyponatremia than in
loss, decreased central venous pressure, increased blood urea those with chronic hyponatremia.293 For example, the most dra-
nitrogen (BUN), or increased hematocrit. Most of these cases matic cases of death due to hyponatremic encephalopathy have
have been in children288,289 and have responded to replacement generally been reported in postoperative patients in whom hypo-
with normal or hypertonic saline, but concurrent treatment natremia develops rapidly as a result of intravenous infusion of
with fludrocortisone has also been advocated.288,290 However, a hypotonic fluids.293,296 In such cases nausea and vomiting are
recent study of 100 consecutive adult patients with acute non- frequently overlooked as potential early signs of increased intra-
traumatic aneurysmal subarachnoid hemorrhage found that the cranial pressure. Critically ill patients with unexplained seizures
cause of the hyponatremia was attributable to SIAD in 71.4% also should be immediately evaluated for possible hyponatremia,
and acute glucocorticoid deficiency in 8.2%, with the remain- since as many as one-third of such patients have a serum [Na+]
ing cases caused by incorrect intravenous fluid administration or below 125 mEq/L as the cause of the seizure activity.297 Under-
hypovolemia. Most significantly, no cases were found that met lying neurologic disease and nonneurologic metabolic disorders
historically accepted criteria for a diagnosis of CSWS.112 This (e.g., hypoxia,298 acidosis, hypercalcemia) can raise the level of
suggests that CSWS is an exceedingly rare cause of the hypona- plasma osmolality at which CNS symptoms occur.
tremia with intracranial disorders.  In the most severe cases of hyponatremic encephalopathy,
death results from respiratory failure after tentorial cerebral her-
Renal Escape From Antidiuresis niation and brainstem compression. One-quarter of patients with
In addition to excreting osmoles to bring volumes back toward severe postoperative hyponatremic encephalopathy manifested
normal, there are intrarenal adaptations that allow excretion of hypercapneic respiratory failure, the expected result of brainstem
more water. Chronic stimulation by vasopressin in SIAD produces compression, but three-quarters had pulmonary edema as the
dramatic increases of aquaporin 2 content and insertion into the apparent cause of the hypoxia.299 Studies of acute hyponatremia
collecting duct principal cell membranes, which increases the effi- after marathon races have shown hypoxia and pulmonary edema
ciency of water retention and aggravates the pathology. However, in association with brain edema.300 These results suggest the pos-
the induced volume expansion and hypotonicity act on the tubu- sibility that hypoxia from noncardiogenic pulmonary edema
lar cells of the collecting duct to decrease the content and action may represent an early sign of developing cerebral edema even
of aquaporin 2 substantially, thus decreasing the amount of water before brainstem compression and tentorial herniation. Clinical
resorbed in spite of high vasopressin levels. Experimental studies studies have suggested that menstruating women296 and young
have suggested this may be due to downregulation of vasopres- children301 may be particularly susceptible to the development of
sin V2-receptor expression in the kidney.291 This renal “escape” neurologic morbidity and mortality during hyponatremia, espe-
therefore represents another (in addition to natriuresis) adapta- cially in the acute postoperative setting.294 However, other studies
tion that allows patients with persistent SIAD to come into a new have failed to corroborate these findings.302,303
steady state of Na+ and water balance despite low serum sodium Although some studies have suggested that hyponatremia
concentrations.292  is an indicator of severe underlying disease and poor prognosis
rather than a cause of the increased mortality in such patients, a
Hypoosmolar Symptoms, Morbidity, and meta-analysis of studies in which some patients had correction of
their hyponatremia indicated that improvement in serum [Na+]
Mortality was associated with a 50% reduction in mortality of the cor-
Regardless of the etiology of hypoosmolality, most clinical rected groups compared to patients in whom the hyponatremia
manifestations are similar. Nonneurologic symptoms are rela- remained uncorrected, suggesting that hyponatremia may in fact
tively uncommon, although a number of cases of rhabdomy- be causally related to increased mortality.304 Recent studies have
olysis have been reported presumably secondary to osmotically further indicated that even mild hyponatremia is an independent
induced swelling of muscle fibers. Hypoosmolality is primarily predictor of higher mortality across a wide variety of disorders,
associated with a broad spectrum of neurologic manifestations including patients with acute ST-elevation myocardial infarction,
ranging from mild nonspecific symptoms (e.g., headache, nau- heart failure, and liver disease.228,235A large study of over 55,000
sea) to more significant disorders (e.g., disorientation, confu- electronic heath records from a single Boston hospital showed
sion, obtundation, focal neurologic deficits, and seizures).293 that the association of hyponatremia with inpatient mortality was
This neurologic symptom complex has been termed “hypona- significant across all levels of hyponatremia and even began at
tremic encephalopathy”294 and primarily reflects brain edema serum [Na+] levels in the lower part of the normal range.229 These
resulting from osmotic water shifts into the brain because of findings were corroborated in studies of 249,000 Danish patients
decreased effective plasma osmolality. Significant neurologic hospitalized over a 5-year period that showed increased 30-day
symptoms generally do not occur until serum [Na+] falls below and 1-year mortality associated with all levels of hyponatremia,
125 mmol/L, and the severity of symptoms are roughly corre- including the range of 130 to 134.9 mmol/L,305 and analyses of
lated with the degree of hypoosmolality.293,295 However, indi- 2.3 million hospitalized patients enrolled in the Cerner Health
vidual variability is marked, and for any single patient the level Facts database.306 The mortality associated with chronic hypo-
of serum [Na+] at which symptoms appear cannot be predicted. natremia is much less well studied, but results of the Rotterdam
The rate of fall of serum [Na+] is often more strongly correlated Longitudinal Aging Study noted significantly decreased survival
with morbidity and mortality than is the actual magnitude of the of elderly patients with hyponatremia over a 12-year period of
decrease.293,295 This is due to the fact that the volume-adaptation observation.307

Downloaded for FK UMI Makassar (mahasiswafkumi01@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier
on November 01, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
Chapter 10  Posterior Pituitary 323

Once the brain has volume regulated via solute losses, thereby and a retrospective study of 4900 hyponatremic patients in
reducing brain edema, neurologic symptoms are not as promi- Taiwan found that the hyponatremic patients had a 2.36-
nent and may even be virtually absent. This accounts for the fairly fold higher hazard ratio of developing dementias compared to
common finding of relatively asymptomatic patients even with matched controls, which increased to a hazard ratio of 4.29
severe levels of hyponatremia.295,308 Despite this powerful adapta- for patients with more severe hyponatremia. Thus the major
tion process, chronic hyponatremia is frequently associated with clinical significance of chronic hyponatremia may lie in the
neurocognitive symptomatology, albeit milder and more subtle increased morbidity and mortality associated with falls, frac-
in nature, such as headaches, nausea, mood disturbances, depres- tures, neurocognitive impairments, and dementias in our older
sion, difficulty concentrating, slowed reaction times, unstable population as well as potential adverse effects not yet studied
gait, increased falls, confusion, and disorientation.309,310 Even in humans.323 
in patients adjudged to be “asymptomatic” by virtue of a normal
neurologic examination, accumulating evidence suggests that Therapy of SIAD and Other Hypoosmolar
there may be previously unrecognized adverse effects as a result Disorders
of chronic hyponatremia. In one study, 16 patients with hypona-
tremia secondary to SIAD, in the range of 124 to 130 mmol/L, General Principles
demonstrated a significant gait instability that normalized after Correction of hyponatremia is associated with markedly improved
correction of the hyponatremia to a normal range.309 The func- neurologic outcomes in patients with severely symptomatic hypo-
tional significance of the gait instability was illustrated in a study natremia. In a retrospective review of patients who presented with
of 122 Belgian patients with a variety of levels of hyponatremia all severe neurologic symptoms and serum [Na+] below 125 mmol/L,
judged to be asymptomatic at the time of visit to an emergency prompt therapy with isotonic or hypertonic saline resulted in a
department (ED). These patients were compared with 244 age- correction in the range of 20 mEq/L over several days and neu-
matched, gender-matched, and disease-matched controls also pre- rologic recovery in almost all cases; in contrast, in patients who
senting to the ED during the same time period. Researchers found were treated with fluid restriction alone, there was very little cor-
that 21% of the hyponatremic patients came to the ED because of rection over the study period (<5 mmol/L over 72 hours), and
a recent fall, compared to only 5% of the controls; this difference the neurologic outcomes were much worse, with most of these
was highly significant and remained so after multivariable adjust- patients either dying or entering a persistently vegetative state.324
ment.309 Analogous results were found in a study of admissions Based on this and similar retrospective analyses, prompt therapy
to a US hospital geriatric trauma unit over a 3-year period; when to rapidly increase the serum [Na+] represents the standard of
patients admitted because of a fall (n = 1841) were analyzed for care for treatment of patients presenting with severe symptoms of
risk factors associated with fall, the odds ratio for serum [Na+] hyponatremia.
below 135 mmol/L was 1.81 (p <0.001), which was greater than Brain herniation, the most dreaded complication of hyponatre-
all other risk factors except age over 85 years.311 Consequently, mia, is seen almost exclusively in patients with acute hyponatremia
these studies clearly document an increased incidence of falls in (usually <24 hours) or in patients with intracranial pathology.325
so-called asymptomatic hyponatremic patients. Recent studies in In postoperative patients and in patients with self-induced water
both experimental animals312 and humans313 have demonstrated intoxication associated with marathon running, psychosis, or
decreases in nerve conduction associated with hyponatremia as a use of “ecstasy” (3,4-methylenedioxy-N-methamphetamine
potential cause of the gait disturbances. [MDMA]), nonspecific symptoms such as headache, nausea and
The clinical significance of the gait instability and fall data have vomiting, or confusion can rapidly progress to seizures, respira-
been indicated by multiple independent international studies that tory arrest, and ultimately death or a permanent vegetative state
have demonstrated increased rates of bone fractures in patients as a complication of cerebral edema.232 Hypoxia from noncardio-
with hyponatremia.307,314–316 Other studies have shown that genic pulmonary edema and/or hypoventilation can exacerbate
hyponatremia is associated with increased bone loss in experimen- brain swelling caused by the low serum [Na+].299,300 Although
tal animals and a significantly increased odds ratio for osteoporo- usually self-limited, hyponatremic seizures may be refractory to
sis of the femoral neck in those older than 50 years in the Third anticonvulsants.
National Health and Nutrition Examination Survey (NHANES As discussed earlier, chronic hyponatremia is much less
III) database.317 These findings have been corroborated by mul- symptomatic as a result of the process of brain volume regula-
tiple epidemiologic studies that have demonstrated decreased tion. Because of this adaptation process, chronic hyponatremia
bone mineral density in human subjects.318,319 In the largest epi- is arguably a condition that clinicians feel less concerned about,
demiologic analysis to date (2.9 million independent electronic which has been reinforced by the common usage of the descrip-
health records), the odds ratios for osteoporosis and fractures were tor asymptomatic hyponatremia for many such patients. However,
significantly greater for hyponatremia (3.99 and 3.05, respec- as discussed, it is clear that many such patients very often have
tively) than for any other diseases or medications associated with neurologic symptoms, even if milder and more subtle in nature.
increased bone loss and fracture risk.320 Of particular note, the Consequently, all patients with hyponatremia who manifest any
odds ratios for both osteoporosis and fractures were the highest neurologic symptoms that could possibly be related to the hypo-
in patients with chronic persistent hyponatremia, indicating that natremia should be considered candidates for treatment of the
duration of hyponatremia represents an important risk factor for hyponatremia, regardless of the chronicity of the hyponatremia
bone disease and fractures.320 These findings were supported by a or the level of serum [Na+]. An additional reason to treat even
subsequent clinical study of hip fractures in Argentina.321 asymptomatic hyponatremia effectively is to prevent a lowering
Recent studies of a cohort of 5435 community dwelling of the serum [Na+] to more symptomatic and dangerous levels
men over the age of 65 in the United States have found that during treatment of underlying conditions (e.g., increased fluid
hyponatremia is independently associated with greater cogni- administration via parenteral nutrition, treatment of heart failure
tive impairment and cognitive decline in this population,322 with diuretics). 

Downloaded for FK UMI Makassar (mahasiswafkumi01@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier
on November 01, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
324 SE C T I O N I I    Hypothalamus and Pituitary

drips. Depending on individual hospital policies, the administra-


Change in serum sodium concentration

20 tion of hypertonic solutions may require special considerations


18 Relowering Relowering Relowering (e.g., placement in the intensive care unit [ICU], sign-off by a
in first 24 hours (mmol/L)

16 unnecessary optional recommended


consultant), which each clinician needs to take into account to
14 optimize patient care. One barrier to the use of hypertonic saline
12 that appears to be overstated and unfounded is the frequent
10 requirement for a central intravenous catheter for chronic infu-
8 sion. A recent study demonstrated a low rate of complications
6 Goal using peripheral infusions of 3% NaCl (6% infiltration and 3%
Goal
4 thrombophlebitis) and concluded that peripheral administration
2 of 3% NaCl carries a low risk of minor nonlimb or life-threaten-
0 ing complications.327
Acute water Low to moderate High An alternative option for more emergent situations is admin-
intoxication risk of ODS risk of ODS
istration of a 100-mL bolus of 3% NaCl, repeated twice if there
• Fig. 10.7  Recommended goals (green) and limits (red) for correction of is no clinical improvement in 30 minutes, which has been recom-
hyponatremia based on risk of producing ODS and recommendations mended by a consensus conference organized to develop guidelines
for relowering of serum sodium concentration ([Na+]) to goals for patients
for the prevention and treatment of exercise-induced hyponatre-
presenting with serum [Na+] lower than 120 mmol/L who exceed the rec-
ommended limits of correction in the first 24 hours. ODS, osmotic demy-
mia328 and adopted as a general recommendation by several expert
elination syndrome. (From Verbalis JG, Goldsmith SR, Greenberg A, et al. panels.310,329 Injecting this amount of hypertonic saline intrave-
Diagnosis, evaluation, and treatment of hyponatremia: expert panel rec- nously raises the serum [Na+] by an average of 2 to 4 mmol/L,
ommendations. Am J Med. 2013;126:S1–S42.) which is well below the recommended maximal daily rate of
change of 10 to 12 mmol/L/24 hours or 8 mmol/L/24 hours for
patients with increased risk factors for ODS (serum [Na+] ≤105
Therapies for Treatment of Hyponatremia mmol/L, hypokalemia, advanced liver disease, malnutrition, or a
Conventional management strategies for hyponatremia range history of alcoholism)310,330 (see Fig. 10.7). Because the adult brain
from saline infusion and fluid restriction to pharmacologic mea- can only accommodate an average increase of approximately 8%
sures to adjust fluid balance. Although the number of available in brain volume before herniation occurs, quickly increasing the
treatments for hyponatremia is large, some are not appropriate serum [Na+] by as little as 2 to 4 mmol/L in acute hyponatremia
for correction of symptomatic hyponatremia because they work can effectively reduce brain swelling and intracranial pressure.331 
too slowly or inconsistently to be effective in hospitalized patients
(e.g., demeclocycline, mineralocorticoids). Consideration of treat- Isotonic Saline
ment options should always include an evaluation of the benefits The treatment of choice for depletional hyponatremia (i.e., hypo-
as well as the potential toxicities of any therapy and must be indi- volemic hyponatremia) is isotonic saline ([Na+] = 154 mmol/L)
vidualized for each patient.310,326 For all therapies, careful atten- to restore ECF volume and ensure adequate organ perfusion. This
tion should be paid to recommendations for goals and limits of initial therapy is appropriate for patients who either have clinical
correction of the serum [Na+] to reduce the risk of the osmotic signs of hypovolemia or in whom a spot urine [Na+] concentra-
demyelination syndrome (ODS)310 (Fig. 10.7). It should also be tion is below 20 to 30 mEq/L.310 Such patients often develop a
remembered that sometimes simply stopping treatment with an free water diuresis (aquaresis) as their ECF volume is corrected,
agent that is associated with causing hyponatremia is sufficient to potentially leading to an overly rapid correction with increased
correct a low serum [Na+]. risk of ODS, so the serum [Na+] and urine output should be fol-
lowed carefully during the first 24 to 48 hours of therapy. How-
Hypertonic Saline ever, isotonic saline is ineffective for dilutional hyponatremias
Acute hyponatremia presenting with severe neurologic symp- such as SIAD,226 and continued administration of isotonic saline
toms is life threatening and should be treated promptly with to a euvolemic patient may worsen their hyponatremia332 and/
hypertonic solutions, typically 3% NaCl ([Na+] = 513 mmol/L), or cause fluid overload. Although saline may improve the serum
as this represents the most reliable method to quickly raise the [Na+] in some patients with hypervolemic hyponatremia, their
serum [Na+]. A continuous infusion of hypertonic NaCl is usu- volume status will generally worsen with this therapy, so unless
ally utilized in inpatient settings. Various formulae have been the hyponatremia is profound both hypertonic and isotonic saline
suggested for calculating the initial rate of infusion of hypertonic should be avoided. 
solutions,325 but until now there has been no consensus regard-
ing optimal infusion rates of 3% NaCl. One of the simplest Fluid Restriction
methods to estimate an initial 3% NaCl infusion rate utilizes the For patients with chronic hyponatremia, fluid restriction has been
following relationship310: the most popular and most widely accepted treatment. When
SIAD is present, fluids should generally be limited to 500 to 1000
Patient’s weight (kg) × desired correction rate (mEq/L/hr) mL/24 hours. Because fluid restriction increases the serum [Na+]
= infusion rate of 3% NaCl (mL/hr) by underreplacing the excretion of fluid by the kidneys, some have
advocated an initial restriction to 500 mL less than the 24-hour
urine output.333 When instituting a fluid restriction, it is impor-
This may not achieve the desired correction rate, but frequent tant for the nursing staff and the patient to understand that this
monitoring of the serum [Na+] will inform the clinician whether includes all fluids that are consumed, not just water (Table 10.3).
the rate should be increased or decreased, similar to using mea- Generally the water content of ingested food is not included in
surement of serum glucose to guide the infusion rate of insulin the restriction because this is balanced by insensible water losses

Downloaded for FK UMI Makassar (mahasiswafkumi01@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier
on November 01, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
Chapter 10  Posterior Pituitary 325

TABLE 10.3  General Recommendations for Use of hours to gauge the initial response. If the correction of serum [Na+]
Fluid Restriction and Predictors of the is thought to be inadequate (e.g., <5 mmol/L), the infusion rate
Increased Likelihood of Failure of Fluid can be increased to 40 mg/day. Clinical studies have supported the
efficacy of bolus infusions of conivaptan rather than continuous
Restriction
infusions.337 Therapy is limited to a maximum duration of 4 days
General Recommendations because of drug interaction effects with other agents metabolized
• Restrict all intake that is consumed by drinking, not just water. by the CYP3A4 hepatic isoenzyme. Importantly, for conivaptan
• Aim for a fluid restriction that is 500 mL/day below the 24-hour and all other vaptans, it is critical that the serum [Na+] be mea-
urine volume. sured frequently during the active phase of correction of the hypo-
• Do not restrict sodium or protein intake unless indicated. natremia—a minimum of every 6 to 8 hours for conivaptan but
Predictors of the Likely Failure of Fluid Restriction
more frequently in patients with risk factors for the ODS.310 If the
correction exceeds 10 to 12 mmol/L in the first 24 hours, the infu-
• High urine osmolality (≥500 mOsm/kg H2O). sion should be stopped and the patient monitored closely. Consid-
• Sum of the urine [Na+] and [K+] concentrations exceeds the serum
[Na+] concentration.
eration should be given to administering sufficient water, orally or
• 24-hour urine volume <1500 mL/day. as intravenous 5% dextrose in water, to avoid a correction of more
• Increase in serum [Na+] concentration <2 mmol/L/day in 24–48 than 12 mmol/L/day. The maximum correction limit should be
hours on a fluid restriction of ≤1 L/day. reduced to 8 mmol/L over the first 24 hours in patients with risk
factors for ODS310 (see Fig. 10.7). The most common side effects
of conivaptan include headache, thirst, and hypokalemia.338
Tolvaptan, an oral vasopressin receptor antagonist, is also FDA
(perspiration, exhaled air, feces, etc.), but caution should be exer- approved for the treatment of euvolemic and hypervolemic hypo-
cised with foods that have high fluid concentrations (such as fruits natremia. In contrast to conivaptan, the availability of tolvaptan
and soups). Restricting fluid intake can be effective when prop- in tablet form allows short-term and long-term use.339 Similar to
erly applied and managed in selected patients, but serum [Na+] conivaptan, tolvaptan treatment must be initiated in the hospi-
is generally increased only slowly (1–2 mmol/L/day) even with tal so that the rate of correction can be monitored carefully. In
severe fluid restriction.226 In addition, this therapy is often poorly the United States, patients with a serum [Na+] lower than 125
tolerated because of an associated increase in thirst leading to poor mmol/L are eligible for therapy with tolvaptan as primary therapy;
compliance with long-term therapy. if the serum [Na+] is 125 mmol/L or higher, tolvaptan therapy is
Fluid restriction should not be used with hypovolemic patients only indicated if the patient has symptoms that could be attrib-
and is particularly difficult to maintain in hospitalized patients utable to the hyponatremia and is resistant to attempts at fluid
with very elevated urine osmolalities secondary to high vasopres- restriction.340 In the European Union, tolvaptan is approved only
sin levels; similarly, if the sum of urine [Na+] and [K+] exceeds for the treatment of euvolemic hyponatremia due to SIAD, but
the serum [Na+], most patients will not respond to a fluid restric- any symptomatic euvolemic patient is eligible for tolvaptan ther-
tion since an electrolyte-free water clearance will be difficult to apy regardless of the level of hyponatremia or response to previ-
achieve.334,335 These and other known predictors of failure of ous fluid restriction. The starting dose of tolvaptan is 15 mg on
fluid restriction are summarized in Table 10.3; the presence of the first day, although in clinical practice some clinicians recom-
any of these factors in hospitalized patients with symptomatic mend starting with a lower dose of 7.5 mg,341 and the dose can be
hyponatremia make this therapy less than ideal. In addition, fluid titrated to 30 and 60 mg at 24-hour intervals if the serum [Na+]
restriction is not practical for some patients, particularly including remains lower than 135 mmol/L or the increase in serum [Na+]
patients in intensive care settings who often require administra- has been less than 5 mmol/L in the previous 24 hours. As with
tion of significant volumes of fluids as part of their therapies. Such conivaptan, it is essential that the serum [Na+] be measured fre-
patients are candidates for more effective pharmacologic or saline quently during the active phase of correction of the hyponatremia
treatment strategies.  at a minimum of every 6 to 8 hours, particularly in patients with
risk factors for ODS. Goals and limits for the safe correction of
Arginine Vasopressin Receptor Antagonists hyponatremia and methods to compensate for overly rapid correc-
Conventional therapies for hyponatremia, although effective in tions are the same as described previously for conivaptan (see Fig.
specific circumstances, are suboptimal for many different rea- 10.7). One additional factor that helps avoid overly rapid correc-
sons, including variable efficacy, slow responses, intolerable side tion with tolvaptan is the recommendation that fluid restriction
effects, and serious toxicities. But perhaps the greatest deficiency should not be used during the active phase of correction, thereby
of most conventional therapies is that they do not directly target allowing the patient’s thirst to compensate for an overly vigorous
the underlying cause of most dilutional hyponatremias, namely aquaresis. Common side effects of tolvaptan include dry mouth,
inappropriately elevated plasma vasopressin levels. A new class of thirst, increased urinary frequency, dizziness, nausea, and ortho-
pharmacologic agents (vasopressin receptor antagonists [vaptans]) static hypotension.340
that directly block vasopressin-mediated receptor activation have Vaptans are not needed in the treatment of hypovolemic hypo-
been approved for the treatment of euvolemic hyponatremia and natremia because simple volume expansion would be expected to
hypervolemic hyponatremia in many countries.326,336 abolish the nonosmotic stimulus to AVP secretion and lead to a
Conivaptan has been approved by the US Food and Drug prompt aquaresis. Furthermore, inducing increased renal fluid
Administration (FDA) for euvolemic and hypervolemic hypona- excretion via diuresis or aquaresis can cause or worsen hypoten-
tremia in hospitalized patients. It is available only as an intrave- sion in such patients. This possibility has resulted in the labeling
nous preparation and is given as a 20-mg loading dose over 30 of these drugs as contraindicated for hypovolemic hyponatremia.
minutes, followed by a continuous infusion of 20 or 40 mg/day. Importantly, clinically significant hypotension was not observed
Generally, the 20-mg continuous infusion is used for the first 24 in the conivaptan or tolvaptan clinical trials in euvolemic and

Downloaded for FK UMI Makassar (mahasiswafkumi01@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier
on November 01, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
326 SE C T I O N I I    Hypothalamus and Pituitary

hypervolemic hyponatremic patients. Although vaptans are not during treatment,346 but it is important to remember that this
contraindicated with decreased renal function, these agents gener- does not represent renal impairment.
ally will not be effective if the serum creatinine level is more than Reports of retrospective, uncontrolled studies suggest that
3 mg/dL. the use of urea has been effective in treating SIAD in patients
The FDA has issued a caution about hepatic injury that was with hyponatremia due to subarachnoid hemorrhage and in criti-
noted in patients who received tolvaptan in a 3-year clinical cal care patients,347 and case reports have documented success in
trial examining the effect of tolvaptan on autosomal dominant infants with chronic SIAD348 and the nephrogenic syndrome of
polycystic kidney disease, the Tolvaptan Efficacy and Safety in inappropriate antidiuresis.349 More recent evidence from a short
Management of Autosomal Dominant Polycystic Kidney Dis- study in a small cohort of SIAD patients suggests that urea may
ease and Its Outcomes (TEMPO) study.342 Although the doses have a comparable efficacy to vaptans in reversing hyponatremia
used in the TEMPO study were up to twice the maximum dose due to chronic SIAD.350 Although these reports suggest that urea
approved for hyponatremia (e.g., tolvaptan, 120 mg/day) and might be an acceptable alternative for treatment of chronic hypo-
in clinical trials of tolvaptan at doses approved by the FDA for natremia, data regarding efficacy and safety of long-term urea
treatment of clinically significant euvolemic or hypervolemic treatment of hyponatremia are lacking.351 
hyponatremia liver damage was not reported, the FDA recom-
mended that “Samsca treatment should be stopped if the patient Furosemide and NaCl
develops signs of liver disease. Treatment duration should be The use of furosemide (20–40 mg/day) coupled with a high sodium
limited to 30 days or less, and use should be avoided in patients intake (200 mEq/day) represents an extension of the treatment of
with underlying liver disease, including cirrhosis.” The European acute symptomatic hyponatremia352 in selected cases.329,353 How-
Medicines Agency (EMA) has approved the use of tolvaptan for ever, similar to urea, the efficacy of this approach to correct symp-
SIAD but not for hyponatremia due to heart failure or cirrhosis. tomatic hyponatremia promptly and within accepted goals limits
Based on the TEMPO trial results, the EMA also issued a warn- is unknown. 
ing about the possible occurrence of hepatic injury in patients
treated with tolvaptan but did not recommend any restriction Efficacy of Hyponatremia Treatment
on the duration of treatment of SIAD patients with tolvaptan. There have been no adequately powered randomized controlled
Accordingly, appropriate caution should be exercised in patients trials to compare the efficacy and safety of different treatments
treated with tolvaptan for hyponatremia for extended periods utilized to correct hyponatremia. However, results of a prospective
(e.g., >30 days), but this decision should be based on the clinical observational study in a large number of hospitalized patients in
judgment of the treating physician. Patients who are refractory the United States and the European Union provide useful data
to or unable to tolerate or obtain other therapies for hypona- about the success rates of different therapies in euvolemic hypona-
tremia, and for whom the benefit of tolvaptan treatment out- tremic patients.354,355 In this study, success was defined by three
weighs the risks, remain candidates for long-term therapy with different criteria, from least to most stringent: (1) an increase
tolvaptan. in serum [Na+] of at least 5 mmol/L, (2) correction to a serum
An additional barrier to the use of vasopressin antagonists for [Na+] of 130 mmol/L or above, and (3) correction to a normal
treatment of hyponatremia is the high cost of the drug. This is true serum [Na+] of 135 mmol/L or above. Only 3% NaCl and tolvap-
in the United States and the European Union but interestingly not tan had success rates significantly greater than 50% for the least
in Asian countries. Despite this pronounced geographic disparity, stringent criterion, and only tolvaptan achieved this level for the
many economic analyses have confirmed the increased economic next most stringent criteria and a significantly higher rate for the
burden of hyponatremia, which is largely driven by longer hospi- most stringent criteria of normalization of the serum [Na+]. Of
tal and intensive care stays.343,344 An analysis of use of tolvaptan particular note, fluid restriction, the most frequently prescribed
compared with fluid restriction showed a favorable cost savings therapy in the Hyponatremia Registry patients, achieved a cor-
that offset the high cost of tolvaptan, suggesting that selective use rection of serum [Na+] in only 44% of patients treated with this
of these agents in appropriate inpatients may in fact be cost effec- therapy and isotonic saline in only 36% of patients. This is con-
tive in hospitalized patients in the United States and the European sistent with a prospective study of 183 hyponatremic patients that
Union.345  demonstrated that up to 60% of patients with SIAD had one or
more criteria predicting nonresponse to fluid restriction (see Table
Urea 10.3).356 These data underscore the importance of carefully select-
Urea has been described as an alternative oral treatment for SIAD ing therapy for individual patients to meet predefined goals for
and other hyponatremic disorders. The mode of action is to cor- correction of serum [Na+]. 
rect hypoosmolality not only by increasing solute-free water excre-
tion but also by decreasing urinary sodium excretion. Doses of Hyponatremia Treatment Guidelines Based on Symptom
15 to 60 g/day are generally effective; the dose can be titrated in Severity
increments of 15 g/day at weekly intervals as necessary to achieve Although many authors have published recommendations on the
normalization of the serum [Na+]. It is advisable to dissolve the treatment of hyponatremia,310,329,357–361 no standardized treat-
urea in orange juice or some other strongly flavored liquid to cam- ment algorithms have yet been universally accepted, and some
ouflage the bitter taste. Even if completely normal water balance major differences between the various guidelines and expert rec-
is not achieved, it is often possible to allow the patient to main- ommendations exist.326,362 For almost all treatment recommenda-
tain a less strict regimen of fluid restriction while receiving urea. tions, the initial evaluation includes an assessment of the ECF
The disadvantages associated with the use of urea include poor volume status of the patient because treatment recommendations
palatability, the development of azotemia at higher doses, and differ for hypovolemic, euvolemic, and hypervolemic hypona-
the unavailability of a convenient or FDA-approved form of the tremic patients.310 Euvolemic patients, mainly including those
agent. Data suggest that blood urea concentrations may double with SIAD, represent a unique challenge because of the multiplicity

Downloaded for FK UMI Makassar (mahasiswafkumi01@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier
on November 01, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
Chapter 10  Posterior Pituitary 327

of causes and presentations of patients with SIAD. Recent expert can indicate that a patient is in an early stage of acute hyponatre-
opinion recommendations are based primarily on the neurologic mia, they more often indicate a chronically hyponatremic state
symptomatology of hyponatremic patients rather than the serum with sufficient brain volume adaptation to prevent marked symp-
[Na+] or the chronicity of the hyponatremia, because the latter tomatology from cerebral edema. Most patients with moderate
is often difficult to ascertain accurately.310 A careful neurologic hyponatremic symptoms have a more chronic form of hypona-
history and assessment should always be conducted to identify tremia, so guidelines for goals and limits of correction should be
potential causes for the patient’s symptoms other than hyponatre- followed closely (see Fig. 10.7), and close monitoring of these
mia, although it will not always be possible to exclude an additive patients in a hospital setting is warranted until the symptoms
contribution from the hyponatremia to an underlying neurologic improve or stabilize.
condition. In this algorithm, patients are divided into three major Patients with no or minimal symptoms should be managed
groups based on their presenting symptoms. initially with fluid restriction, although treatment with pharma-
Severe Symptoms. Coma, obtundation, seizures, respiratory cologic therapy, such as vaptans or urea, may be appropriate for
distress or arrest, and unexplained vomiting usually imply a more a wide range of specific clinical conditions. Foremost of these is a
acute onset or worsening of hyponatremia that requires immedi- failure to improve the serum [Na+], despite reasonable attempts at
ate active treatment. Therapies that will quickly raise serum [Na+] fluid restriction, or the presence of clinical characteristics associ-
are necessary to reduce cerebral edema and decrease the risk of ated with poor responses to fluid restriction (see Table 10.3).
potentially fatal brain herniation.  A special case is when spontaneous correction of hyponatre-
Moderate Symptoms. Altered mental status, disorientation, mia occurs at an undesirably rapid rate because of the onset of
confusion, unexplained nausea, gait instability, and falls gener- water diuresis. This can occur following cessation of desmopressin
ally indicate some degree of brain volume regulation and absence therapy in a patient who has become hyponatremic, replacement
of clinically significant cerebral edema. These symptoms can be of glucocorticoids in a patient with adrenal insufficiency, replace-
chronic or acute, but allow more time to elaborate a deliberate ment of solutes in a patient with diuretic-induced hyponatremia,
approach to the choice of therapies.  or spontaneous resolution of transient SIAD. Brain damage from
Mild or Absent Symptoms. Minimal symptoms, such as dif- ODS can clearly ensue in this setting if the preceding period of
ficulty concentrating, irritability, altered mood, depression, or hyponatremia has been long enough (usually ≥48 hours) to allow
unexplained headache, or a virtual absence of discernible symp- brain volume regulation to occur. If the correction parameters dis-
toms, indicate that the patient may have chronic or slowly cussed earlier have been exceeded and the correction is proceeding
evolving hyponatremia. These symptoms necessitate a cautious more rapidly than planned (usually because of continued excretion
approach, especially when patients have underlying comorbidi- of hypotonic urine), the pathologic events leading to demyelin-
ties, to prevent both worsening of the hyponatremia and overly ation can be reversed by administration of hypotonic fluids, with
rapid correction with production of ODS. or without desmopressin. The efficacy of this approach has been
Patients with severe neurologic symptoms should be treated suggested from animal studies369 and case reports in humans,370
with hypertonic (3%) NaCl as first-line therapy, followed after even when patients are overtly symptomatic.371 However, relow-
24 to 48 hours by fluid restriction and/or vaptan therapy. Because ering the serum [Na+] after an initial, overly rapid correction is
overly rapid correction of serum [Na+] occurs in more than 10% only strongly recommended for patients at high risk of ODS; it
of patients treated with hypertonic NaCl,363 such patients are at is considered optional for patients with a low-to-moderate risk of
risk for ODS unless carefully monitored. For this reason, some ODS, and unnecessary for patients with acute water intoxication
authors have proposed simultaneous treatment with desmopres- (see Fig. 10.7). 
sin to reduce the rate of correction to only that produced by the
hypertonic NaCl infusion itself.364 Whether sufficient clinical Monitoring the Serum [Na+] in Hyponatremic Patients
data eventually prove that this approach is effective and safe in The frequency of serum [Na+] monitoring is dependent on both
larger numbers of patients remains to be determined.365,366 Only the severity of the hyponatremia and the therapy chosen. All
one case of ODS has been reported in a patient receiving vap- patients undergoing active treatment with hypertonic saline for
tan monotherapy,367 and two abstracts have reported ODS when symptomatic hyponatremia should have frequent monitoring of
vaptans were used directly following hypertonic saline administra- serum [Na+], urine output, and ECF volume status (every 2–4
tion within the same 24-hour period. Consequently, no additional hours) to ensure that the serum [Na+] does not exceed the limits of
active hyponatremia therapy should be administered until at least safe correction during the active phase of correction310 since overly
24 hours following successful increases in serum [Na+] using rapid correction of serum [Na+] will increase the risk of ODS.233
hypertonic NaCl. Patients treated with vaptans for moderate or mild symptoms
The choice of treatment for patients with moderate symptoms should have serum [Na+] monitored every 6 to 8 hours during the
will depend on their ECF volume status. Hypovolemic patients active phase of correction, which will generally be the first 24 to
should be treated with solute repletion via isotonic NaCl infusion 48 hours of therapy. Active treatment with any therapy should be
or oral sodium replacement. Euvolemic patients, typically with stopped when the patient’s symptoms are no longer present, a safe
SIAD, will benefit from vaptan therapy, limited hypertonic saline serum [Na+] (usually >125 mmol/L) has been achieved, or the rate
administration, or in some cases urea (where available). This can of correction has reached maximum limits of 10 to 12 mmol/L
be followed by fluid restriction or long-term vaptan therapy when within 24 hours, 18 mmol/L within 48 hours,310,330 or 8 mmol/L
the cause of the SIAD is expected to be chronic. In hypervolemic over any 24-hour period in patients at high risk of ODS (see Fig.
patients with heart failure, vaptans are usually the best choice 10.7). In patients with a stable level of serum [Na+] treated with
because fluid restriction is rarely successful in this group,368 saline fluid restriction or therapies other than hypertonic saline, mea-
administration can cause fluid retention with increased edema, surement of serum [Na+] daily is generally sufficient since levels
and urea can lead to ammonia buildup in the GI tract if hepatic will not change that quickly in the absence of active therapy or
function is impaired. Although moderate neurologic symptoms large changes in fluid intake or administration. 

Downloaded for FK UMI Makassar (mahasiswafkumi01@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier
on November 01, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
328 SE C T I O N I I    Hypothalamus and Pituitary

Long-Term Treatment of Chronic Hyponatremia When an infant begins sucking at the breast, an afferent signal
Some patients will benefit from continued treatment of hypona- is transmitted from the mechanoreceptors or tactile receptors in
tremia following discharge from the hospital. In many cases, this the breast to the spinal cord and eventually ascend to the oxytocin
will consist of a continued fluid restriction, but long-term com- magnocellular neurons in the supraoptic nucleus and the paraven-
pliance with this therapy is poor because of the increased thirst tricular nucleus.375,376 Pulsatile release of oxytocin produces a pul-
that occurs with more severe degrees of fluid restriction. Thus for satile pumping action on the alveoli, which promotes maximum
select patients who have responded to tolvaptan in the hospital, emptying of milk from the alveoli.374 The importance of oxytocin
consideration should be given to continuing the treatment as an in maintaining milk secretion is demonstrated by transgenic mice
outpatient after discharge. In patients with established chronic with a knockout that inhibits oxytocin synthesis. These animals
hyponatremia, tolvaptan has shown to be effective for maintaining deliver their young normally and have normal milk production,
a normal [Na+] for as long as 3 years of continued daily therapy.372 but there is no milk release in spite of normal suckling. The pups
However, many patients with inpatient hyponatremia have a tran- die of dehydration with no milk in the stomach.377 Adminis-
sient form of SIAD without the need for long-term therapy. Selec- tration of oxytocin to these oxytocin-deficient mice rescues the
tion of which patients with inpatient hyponatremia are candidates ability to secrete milk and allows the pups to survive. Similarly
for long-term therapy should be based on the cause of the SIAD. oxytocin may promote successful lactation in women who have
In all cases, consideration should be given to a trial of stopping the lactational insufficiency.378
drug 2 to 4 weeks after discharge to determine if hyponatremia is As breastfeeding continues in humans, the basal levels of oxy-
still present. A reasonable period of tolvaptan cessation to evaluate tocin decrease but pulses of oxytocin in response to suckling con-
the presence of continued SIAD is 7 days because this period was tinue and may increase.379 Humans with diabetes insipidus have
found to be sufficient for demonstration of a recurrence of hypo- been able to successfully breastfeed infants, and this has caused
natremia in the tolvaptan SALT339 and SALT-WATER trials.372 some to question the importance of oxytocin in humans.380 How-
Findings of hepatotoxicity in a small number of patients on ever, oxytocin secretion may be preserved in the absence of vaso-
high doses of tolvaptan in a clinical trial of polycystic kidney dis- pressin in patients with diabetes insipidus. 
ease led to a recent FDA recommendation that tolvaptan not be
used for longer than 30 days. If tolvaptan is used for longer than Parturition
30 days, liver function should be assessed at regular intervals (e.g.,
every 3 months) at least for the first year of therapy. As is always The isolation of oxytocin was followed quickly by the description
the case, decisions about appropriate treatment of hyponatremia of oxytocin to stimulate uterine contractions, and this was fol-
should be based on clinical judgment and risk/benefit analysis that lowed shortly by clinical use of oxytocin as a uterotonic agent.381
is individualized for specific patients, because there is no single Parturition in humans is much more complex than just the role of
treatment that represents the “best” therapy for all patients with oxytocin.382 In all species the uterus must grow during pregnancy,
SIAD.326  and estrogen is a promoter of this growth. Levels of oxytocin in
humans are not well defined in pregnancy but are not reported to
Oxytocin increase until the expulsive stage at term.382–384 The uterine myo-
metrial cells have intrinsic contractile activity, but during preg-
Study of the normal physiologic regulation of oxytocin secretion nancy the uterus is maintained in a quiescent state by the actions
and action is complicated by the fact that secretion and func- of progesterone and relaxin (produced by the corpus luteum and
tion of oxytocin varies markedly among different experimental decidual tissue).385–387 The initiation of labor is accomplished by
mammals. There are varying sites of synthesis in the ovary and a relative increase in estrogen activation and a decrease in proges­
in tissues of the uterus that are different among species. It is dif- terone activation. Changes in oxytocin receptors and oxytocin
ficult to study pregnant women and human tissue, so physiologic produced by the placenta may be more important than levels of
regulation of oxytocin secretion and function is less well known oxytocin in the circulation. During early labor there is an upregu-
in humans than other species. The classic roles of oxytocin are lation in the uterus of oxytocin receptor mRNA, and oxytocin
smooth muscle activation promoting milk letdown with nursing receptor numbers increase.384,388 Oxytocin receptors are promi-
and uterine myometrial contraction at parturition. nent in the fundus of the uterus where they stimulate myometrial
contraction and in decidual cells where they stimulate the produc-
tion of prostaglandins. At parturition increased oxytocin activity
Lactation in the fundus will push the fetus toward the cervix that is thinned
A characteristic of all mammals is lactation, and all mammals and relaxed by the effects of prostaglandins.383,389 Prostaglandins
secrete oxytocin to stimulate milk letdown associated with nurs- play a key role in an inflammatory process that is important in the
ing.373 The other hormone critical to lactation is prolactin. Each of uterus at parturition. Cytokines induce enzymes that digest extra-
these pituitary/hypothalamic hormones is importantly influenced cellular matrix to soften and ripen the cervix.383 The role of pro-
and regulated by gonadal steroid hormones. The milk-producing gesterone in maintaining uterine quiescence is not only the action
unit of the breast is the alveolar system with multiple clusters of on oxytocin receptors but also the antagonizing of the inflamma-
milk-producing cells surrounded by specialized myoepithelial tory response that softens the lower uterus and cervix.384
cells. The alveoli are directly connected to ductules, and then There are three situations in pregnancy where a pharmacologic
ducts converge and lead to the nipple. Milk is synthesized in the role of oxytocin is of interest. The first situation and most widely
glandular cells of the alveoli.374 Oxytocin receptors are localized used role of oxytocin is to induce and augment labor.390 This has
on glandular cells, and oxytocin in the systemic circulation acts on received increased interest in an effort to decrease the number of
these receptors to cause myoepithelial contraction. Oxytocin also and morbidity of cesarean sections.391 Oxytocin may be delivered
acts on myoepithelial cells along the duct to shorten and widen alone or in combination with another pharmacologic agent such
the ducts to enhance milk flow through the ducts to the nipple.375 as propranolol or prostaglandins.391,392 The second area of interest

Downloaded for FK UMI Makassar (mahasiswafkumi01@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier
on November 01, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
Chapter 10  Posterior Pituitary 329

is preterm labor with an effort toward prevention by decreasing for appropriate administration,397 prompting a search for other
contractile activity of the uterus and/or inhibiting the inflamma- agents. Promising results have been reported with prostaglandin
tory response.393 Peptide and nonpeptide oxytocin antagonists analogues (e.g., misoprostol).394,395,398 
have been of special interest to inhibit myometrial contractions,
but widespread clinical use awaits the development of antagonist Behavior
with better risk/benefit activity.394 The third pharmacologic inter-
est in oxytocin is as a uterotonic to decrease postpartum hemor- This chapter is about functions of vasopressin and oxytocin as tra-
rhage associated with uterine atony.395 Postpartum hemorrhage is ditional endocrine hormones secreted by the posterior pituitary.
the major cause of maternal deaths worldwide and ranks second For further discussion related to these hormones in purported
to embolism as a cause of maternal death in the United States.396 functions as neurotransmitters, especially with regard to influenc-
Mechanical options in the active management of the third stage of ing social and ingestive behaviors, the reader is referred to Chap-
labor include cord traction to reduce the risk of retained placenta ters 9 and 20.
and uterine massage, which has been augmented by pharmacologic
agents, most commonly oxytocin and/or ergotamine.395 Maternal References
death by postpartum hemorrhage is most significant in develop-
ing countries. Oxytocin is heat labile and requires a trained staff The complete list of references is available online at ExpertConsult.com.

Downloaded for FK UMI Makassar (mahasiswafkumi01@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier
on November 01, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
This page intentionally left blank
     

Downloaded for FK UMI Makassar (mahasiswafkumi01@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier
on November 01, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
References 21. Acher R. Evolution of neurohypophysial control of water homeo-
stasis: integrative biology of molecular, cellular and organismal
1. Makarenko IG, Ugrumov MV, Derer P, Calas A. Projections from aspects. In: Saito T, Kurokawa K, Yoshida S, eds. Neurohypophysis:
the hypothalamus to the posterior lobe in rats during ontogenesis: Recent Progress of Vasopressin and Oxytocin Research. Amsterdam:
1,1’-dioctadecyl-3,3,3’, 3’-tetramethylindocarbocyanine perchlo- Elsevier; 1995:39–54.
rate tracing study. J Comp Neurol. 2000;422(3):327–337. 22. Herman JP, Schafer MK, Watson SJ, Sherman TG. In situ hybrid-
2. Gainer H. Cell-type specific expression of oxytocin and vaso- ization analysis of arginine vasopressin gene transcription using
pressin genes: an experimental odyssey. J Neuroendocrinol. intron-specific probes. Mol Endocrinol. 1991;5(10):1447–1456.
2012;24(4):528–538. 23. Majzoub J. Vasopressin biosynthesis. In: Schrier RW, ed. Vasopres-
3. Sofroniew MV. Morphology of vasopressin and oxytocin neu- sin. New York: Raven Press; 1985:465–474.
rones and their central and vascular projections. Prog Brain Res. 24. Sherman T, Akil H, Watson S. Vasopressin mRNA expression: a
1983;60:101–114. Northern and in situ hybridization analysis. In: Schrier RW, ed.
4. Treier M, Rosenfeld MG. The hypothalamic-pituitary axis: Vasopressin. New York: Raven Press; 1985:475–483.
­co-development of two organs. Curr Opin Cell Biol. 1996;8(6): 25. Zingg HH, Lefebvre D, Almazan G. Regulation of vasopressin
833–843. gene expression in rat hypothalamic neurons: response to osmotic
5. Sawchenko PE, Swanson LW. The organization and biochemical stimulation. J Biol Chem. 1986;261(28):12956–12959.
specificity of afferent projections to the paraventricular and supra- 26. Robinson AG, Roberts MM, Evron WA, et al. Hyponatremia in
optic nuclei. Prog Brain Res. 1983;60:19–29. rats induces downregulation of vasopressin synthesis. J Clin Invest.
6. Swanson LW, Sawchenko PE. Paraventricular nucleus: a site for the 1990;86(4):1023–1029.
integration of neuroendocrine and autonomic mechanisms. Neuro- 27. Roberts MM, Robinson AG, Hoffman GE, Fitzsimmons MD.
endocrinology. 1980;31(6):410–417. Vasopressin transport regulation is coupled to the synthesis rate.
7. Oliet SH. Functional consequences of morphological neuroglial Neuroendocrinology. 1991;53(4):416–422.
changes in the magnocellular nuclei of the hypothalamus. J Neuro- 28. Lederis K, Jayasena K. Storage of neurohypophysial hormones and
endocrinol. 2002;14(3):241–246. the mechanism for their release. In: Heller H, Pickering BT, eds.
8. Theodosis DT, Poulain DA. Maternity leads to morphologi- Pharmacology of the Endocrine System and Related Drugs. London:
cal synaptic plasticity in the oxytocin system. Prog Brain Res. Pergamon; 1970:111–154.
2001;133:49–58. 29. Kurokawa H, Fujisawa I, Nakano Y, et al. Posterior lobe of the pitu-
9. Oliet SH, Piet R, Poulain DA, Theodosis DT. Glial modulation of itary gland: correlation between signal intensity on T1-weighted MR
synaptic transmission: insights from the supraoptic nucleus of the images and vasopressin concentration. Radiology. 1998;207(1):79–83.
hypothalamus. Glia. 2004;47(3):258–267. 30. Sato N, Tanaka S, Tateno M, et al. Origin of posterior pituitary high
10. Israel JM, Poulain DA, Oliet SH. Glutamatergic inputs con- intensity on T1-weighted magnetic resonance imaging. Immuno-
tribute to phasic activity in vasopressin neurons. J Neurosci. histochemical, electron microscopic, and magnetic resonance stud-
2010;30(4):1221–1232. ies of posterior pituitary lobe of dehydrated rabbits. Invest Radiol.
11. Scott V, Bishop VR, Leng G, Brown CH. Dehydration-induced 1995;30(10):567–571.
modulation of kappa-opioid inhibition of vasopressin neurone 31. Fitzsimmons MD, Roberts MM, Sherman TG, Robinson AG.
activity. J Physiol. 2009;587(Pt 23):5679–5689. Models of neurohypophyseal homeostasis. Am J Physiol. 1992;262(6
12. Mark L, Pech P, Daniels D, et al. The pituitary fossa: a correlative Pt 2):R1121–R1130.
anatomic and MR study. Radiology. 1984;153(2):453–457. 32. Barberis C, Mouillac B, Durroux T. Structural bases of vasopressin/
13. Melo ME, Marui S, Carvalho LR, et al. Hormonal, pituitary mag- oxytocin receptor function. J Endocrinol. 1998;156(2):223–229.
netic resonance, LHX4 and HESX1 evaluation in patients with 33. Thrasher TN. Baroreceptor regulation of vasopressin and renin
hypopituitarism and ectopic posterior pituitary lobe. Clin Endocri- secretion: low-pressure versus high-pressure receptors. Front Neu-
nol (Oxf ). 2007;66(1):95–102. roendocrinol. 1994;15(2):157–196.
14. Reynaud R, Albarel F, Saveanu A, et  al. Pituitary stalk interrup- 34. Bishop VS, Thames MD, Schmid PG. Effects of bilateral vagal cold
tion syndrome in 83 patients: novel HESX1 mutation and severe block on vasopressin in conscious dogs. Am J Physiol. 1984;246(4
hormonal prognosis in malformative forms. Eur J Endocrinol. Pt 2):R566–R569.
2011;164(4):457–465. 35. Thames MD, Schmid PG. Cardiopulmonary receptors with vagal
15. Secco A, Allegri AE, di Iorgi N, et  al. Posterior pituitary (PP) afferents tonically inhibit ADH release in the dog. Am J Physiol.
evaluation in patients with anterior pituitary defect associated 1979;237(3):H299–H304.
with ectopic PP and septo-optic dysplasia. Eur J Endocrinol. 36. Blessing WW, Sved AF, Reis DJ. Destruction of noradrenergic
2011;165(3):411–420. neurons in rabbit brainstem elevates plasma vasopressin, causing
16. Garel C, Leger J. Contribution of magnetic resonance imaging in non- hypertension. Science. 1982;217(4560):661–663.
tumoral hypopituitarism in children. Horm Res. 2007;67(4):194–202. 37. Blessing WW, Sved AF, Reis DJ. Arterial pressure and plasma vaso-
17. Maghnie M, Genovese E, Villa A, et al. Dynamic MRI in the con- pressin: regulation by neurons in the caudal ventrolateral medulla
genital agenesis of the neural pituitary stalk syndrome: the role of of the rabbit. Clin Exp Hypertens A. 1984;6(1–2):149–156.
the vascular pituitary stalk in predicting residual anterior pituitary 38. Sved AF, Blessing WW, Reis DJ. Caudal ventrolateral medulla
function. Clin Endocrinol (Oxf ). 1996;45(3):281–290. can alter vasopressin and arterial pressure. Brain Res Bull.
18. Ultmann MC, Siegel SF, Hirsch WL, et  al. Pituitary stalk and 1985;14(3):227–232.
ectopic hyperintense T1 signal on magnetic resonance imaging. 39. Schreihofer AM, Stricker EM, Sved AF. Chronic nucleus tractus
Implications for anterior pituitary dysfunction. Am J Dis Child. solitarius lesions do not prevent hypovolemia-induced vasopressin
1993;147(6):647–652. secretion in rats. Am J Physiol. 1994;267(4 Pt 2):R965–R973.
19. Gainer H, Wray S. Cellular and molecular biology of oxytocin and 40. Robertson GL. The regulation of vasopressin function in health
vasopressin. In: Knobil E, Neill JD, eds. The Physiology of Reproduc- and disease. Recent Prog Horm Res. 1976;33:333–385.
tion. 2nd ed. New York: Raven Press; 1994:1099–1129. 41. Callahan MF, Ludwig M, Tsai KP, et al. Baroreceptor input regu-
20. Chen LQ, Rose JP, Breslow E, et  al. Crystal structure of a lates osmotic control of central vasopressin secretion. Neuroendocri-
bovine neurophysin II dipeptide complex at 2.8 A deter- nology. 1997;65(4):238–245.
mined from the single-wavelength anomalous scattering sig- 42. Pump B, Gabrielsen A, Christensen NJ, et al. Mechanisms of inhibi-
nal of an incorporated iodine atom. Proc Natl Acad Sci U S A. tion of vasopressin release during moderate antiorthostatic posture
1991;88(10):4240–4244. change in humans. Am J Physiol. 1999;277(1 Pt 2):R229–R235.

330.e1
Downloaded for FK UMI Makassar (mahasiswafkumi01@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier
on November 01, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
330.e2 References

43. Johnson AK, Thunhorst RL. The neuroendocrinology of thirst and 67. Hvistendahl GM, Frokiaer J, Nielsen S, Djurhuus JC. Gender
salt appetite: visceral sensory signals and mechanisms of central differences in nighttime plasma arginine vasopressin and delayed
integration. Front Neuroendocrinol. 1997;18(3):292–353. compensatory urine output in the elderly population after desmo-
44. Baylis PH, Thompson CJ. Osmoregulation of vasopressin secre- pressin. J Urol. 2007;178(6):2671–2676.
tion and thirst in health and disease. Clin Endocrinol (Oxf ). 68. Schlanger LE, Bailey JL, Sands JM. Electrolytes in the aging. Adv
1988;29(5):549–576. Chronic Kidney Dis. 2010;17(4):308–319.
45. Zerbe RL, Robertson GL. Osmoregulation of thirst and AVP secre- 69. Fulop T Jr., Worum I, Csongor J, et al. Body composition in elderly
tion in human subjects: effect of various solutes. Am J Physiol. people. II. Comparison of measured and predicted body composi-
1983;244(6):E607–E614. tion in healthy elderly subjects. Gerontology. 1985;31(3):150–157.
46. Baylis PH. Investigation of suspected hypothalamic diabetes insipi- 70. Phillips PA, Johnston CI, Gray L. Disturbed fluid and electrolyte
dus. Clin Endocrinol (Oxf ). 1995;43(4):507–510. homoeostasis following dehydration in elderly people. Age Ageing.
47. Majzoub JA, Srivatsa A. Diabetes insipidus: clinical and basic 1993;22(1):S26–S33.
aspects. Pediatr Endocrinol Rev. 2006;4(suppl 1):60–65. 71. Takamata A, Ito T, Yaegashi K, et al. Effect of an exercise-heat accli-
48. Nielsen S, Frokiaer J, Marples D, et  al. Aquaporins in the mation program on body fluid regulatory responses to dehydration
kidney: from molecules to medicine. Physiol Rev. 2002;82(1): in older men. Am J Physiol. 1999;277(4 Pt 2):R1041–R1050.
205–244. 72. Crowe MJ, Forsling ML, Rolls BJ, et al. Altered water excretion in
49. Brown D. The ins and outs of aquaporin-2 trafficking. Am J Physiol healthy elderly men. Age Ageing. 1987;16(5):285–293.
Renal Physiol. 2003;284(5):F893–F901. 73. Sands JM. Urine concentrating and diluting ability during aging. J
50. Eto K, Noda Y, Horikawa S, et al. Phosphorylation of aquapo- Gerontol A Biol Sci Med Sci. 2012;67(12):1352–1357.
rin-2 regulates its water permeability. J Biol Chem. 2010;285(52): 74. Roberts MM, Robinson AG. Hyponatremia in the elderly: diagno-
40777–40784. sis and management. Geriatr Nephrol Urol. 1993;3(1):43–50.
51. Stocker SD, Sved AF, Stricker EM. Role of renin-angiotensin sys- 75. Bell NH. Endocrine complications of sarcoidosis. Endocrinol Metab
tem in hypotension-evoked thirst: studies with hydralazine. Am J Clin North Am. 1991;20(3):645–654.
Physiol Regul Integr Comp Physiol. 2000;279(2):R576–R585. 76. Smith D, Finucane F, Phillips J, et al. Abnormal regulation of thirst
52. Thompson CJ, Bland J, Burd J, Baylis PH. The osmotic thresholds and AVP secretion following surgery for craniopharyngioma. Clin
for thirst and vasopressin release are similar in a healthy man. Clin Endocrinol. 2004;61(2):273–279.
Sci. 1986;71(6):651–656. 77. Moses AM. Clinical and laboratory observations in the adult with dia-
53. Robertson G, Berl T. Water metabolism. In: Brenner BM, Rec- betes insipidus and related syndromes. In: Czernichow P, Robinson
tor FC Jr., eds. The Kidney. 3rd ed. Philadelphia: W.B. Saunders; AG, eds. Diabetes Insipidus in Man. Basel: Karger; 1985:156–175.
1986:385–432. 78. de Leon J, Dadvand M, Canuso C, et  al. Polydipsia and water
54. Smith D, Moore K, TormeyW, et  al. Downward resetting of the intoxication in a long-term psychiatric hospital. Biol Psychiatry. 1.
osmotic thresholds for thirst and vasopressin in patients with 1996;40(1):28–34.
SIAD. Am J Physiol Endocrinol Metab. 2004;287(5):E1019–E1023. 79. Siegel AJ, Baldessarini RJ, Klepser MB, McDonald JC. Primary
55. Lindheimer MD, Davison JM. Osmoregulation, the secretion of and drug-induced disorders of water homeostasis in psychiatric
arginine vasopressin and its metabolism during pregnancy. Eur J patients: principles of diagnosis and management. Harv Rev Psy-
Endocrinol. 1995;132(2):133–143. chiatry. 1998;6(4):190–200.
56. Lindheimer MD, Barron WM. Water metabolism and vasopres- 80. Behan LA, Sherlock M, Moyles P, et al. Abnormal plasma sodium
sin secretion during pregnancy. Baillieres Clin Obstet Gynaecol. concentrations in patients treated with desmopressin for cranial
1994;8(2):311–331. diabetes insipidus: results of a long-term retrospective study. Eur J
57. Barron WM, Stamoutsos BA, Lindheimer MD. Role of volume in Endocrinol. 2015;172(3):243–250.
the regulation of vasopressin secretion during pregnancy in the rat. 81. Fatemi N, Dusick JR, Mattozo C, et  al. Pituitary hormonal loss
J Clin Invest. 1984;73(4):923–932. and recovery after transsphenoidal adenoma removal. Neurosurgery.
58. Danielson LA, Kercher LJ, Conrad KP. Impact of gender and 2008;63(4):709–718, discussion 718–709.
endothelin on renal vasodilation and hyperfiltration induced by 82. Cusick JF, Hagen TC, Findling JW. Inappropriate secretion of
relaxin in conscious rats. Am J Physiol Regul Integr Comp Physiol. antidiuretic hormone after transsphenoidal surgery for pituitary
2000;279(4):R1298–R1304. tumors. N Engl J Med. 1984;311(1):36–38.
59. Danielson LA, Sherwood OD, Conrad KP. Relaxin is a potent 83. Ultmann MC, Hoffman GE, Nelson PB, Robinson AG. Transient
renal vasodilator in conscious rats. J Clin Invest. 1999;103(4): hyponatremia after damage to the neurohypophyseal tracts. Neuro-
525–533. endocrinology. 1992;56(6):803–811.
60. Novak J, Danielson LA, Kerchner LJ, et al. Relaxin is essential for 84. Olson BR, Rubino D, Gumowski J, Oldfield EH. Isolated hypo-
renal vasodilation during pregnancy in conscious rats. J Clin Invest. natremia after transsphenoidal pituitary surgery. J Clin Endocrinol
2001;107(11):1469–1475. Metab. 1995;80(1):85–91.
61. Schrier RW. Systemic arterial vasodilation, vasopressin, and vaso- 85. Kristof RA, Rother M, Neuloh G, Klingmuller D. Incidence, clini-
pressinase in pregnancy. J Am Soc Nephrol. 2010;21(4):570–572. cal manifestations, and course of water and electrolyte metabolism
62. Robinson AG, Fitzsimmons M. Diabetes insipidus. In: Mazzaf- disturbances following transsphenoidal pituitary adenoma sur-
erri EL, Bar RS, Kreisberg RA, eds. Advances in Endocrinology and gery: a prospective observational study. J Neurosurg. 2009;111(3):
Metabolism. St. Louis: Mosby; 1994:261–296. 555–562.
63. Davison JM, Sheills EA, Barron WM, et al. Changes in the meta- 86. Olson BR, Gumowski J, Rubino D, Oldfield EH. Pathophysiology
bolic clearance of vasopressin and in plasma vasopressinase through- of hyponatremia after transsphenoidal pituitary surgery. J Neuro-
out human pregnancy. J Clin Invest. 1989;83(4):1313–1318. surg. 1997;87(4):499–507.
64. Davis PJ, Davis FB. Water excretion in the elderly. Endocrinol 87. Sata A, Hizuka N, Kawamata T, et al. Hyponatremia after trans-
Metab Clin North Am. 1987;16(4):867–875. sphenoidal surgery for hypothalamo-pituitary tumors. Neuroendo-
65. Stout NR, Kenny RA, Baylis PH. A review of water balance in age- crinology. 2006;83(2):117–122.
ing in health and disease. Gerontology. 1999;45(2):61–66. 88. Kovacs K. Metastatic cancer of the pituitary gland. Oncology.
66. Tian Y, Serino R, Verbalis JG. Downregulation of renal vasopres- 1973;27(6):533–542.
sin V2 receptor and aquaporin-2 expression parallels age-asso- 89. Max MB, Deck MD, Rottenberg DA. Pituitary metastasis: inci-
ciated defects in urine concentration. Am J Physiol Renal Physiol. dence in cancer patients and clinical differentiation from pituitary
2004;287(4):F797–F805. adenoma. Neurology. 1981;31(8):998–1002.

Downloaded for FK UMI Makassar (mahasiswafkumi01@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier
on November 01, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
References 330.e3

90. Basaria S, Westra WH, Brem H, Salvatori R. Metastatic renal cell 111. Hannon MJ, Orr C, Moran C, et al. Anterior hypopituitarism is
carcinoma to the pituitary presenting with hyperprolactinemia. J rare and autoimmune disease is common in adults with idiopathic
Endocrinol Invest. 2004;27(5):471–474. central diabetes insipidus. Clin Endocrinol. 2012;76(5):725–728.
91. Fassett DR, Couldwell WT. Metastases to the pituitary gland. Neu- 112. Hannon MJ, Behan LA, O’Brien MM, et  al. Hyponatremia fol-
rosurg Focus. 2004;16(4):E8. lowing mild/moderate subarachnoid hemorrhage is due to SIAD
92. Komninos J, Vlassopoulou V, Protopapa D, et  al. Tumors meta- and glucocorticoid deficiency and not cerebral salt wasting. J Clin
static to the pituitary gland: case report and literature review. J Clin Endocrinol Metab. 2014;99(1):291–298.
Endocrinol Metab. 2004;89(2):574–580. 113. Verbalis J, Robinson A, Moses M. Postoperative and post-traumatic
93. Vuillermet P, Cauliez B, Freger P, et al. Simultaneous suprasellar diabetes insipidus. In: Czernichow P, Robinson AG, eds. Diabetes
and pineal germ cell tumors in five late stage adolescents: endo- Insipdus in Man. Basel: Karger; 1985:247–265.
crinological studies and prolonged follow-up. J Pediatr Endocrinol 114. Agha A, Sherlock M, Phillips J, et  al. The natural history of
Metab. 2008;21(12):1169–1178. post-traumatic neurohypophysial dysfunction. Eur J Endocrinol.
94. Dutta P, Bhansali A, Singh P, Bhat MH. Suprasellar tubercu- 2005;152(3):371–377.
lar abscess presenting as panhypopituitarism: a common lesion 115. Agha A, Thornton E, O’Kelly P, et  al. Posterior pituitary dys-
in an uncommon site with a brief review of literature. Pituitary. function after traumatic brain injury. J Clin Endocrinol Metab.
2006;9(1):73–77. 2004;89(12):5987–5992.
95. Pascual JM, Gonzalez-Llanos F, Roda JM. Primary hypothalamic- 116. Thompson CJ, Baylis PH. Thirst in diabetes insipidus: clinical rele-
third ventricle lymphoma. Case report and review of the literature. vance of quantitative assessment. Q J Med. 1987;65(246):853–862.
Neurocirugia (Astur). 2002;13(4):305–310. 117. Crowley RK, Sherlock M, Agha A, et  al. Clinical insights into
96. Castagnola C, Morra E, Bernasconi P, et al. Acute myeloid leuke- adipsic diabetes insipidus: a large case series. Clin Endocrinol (Oxf ).
mia and diabetes insipidus: results in 5 patients. Acta Haematol. 2007;66(4):475–482.
1995;93(1):1–4. 118. McIver B, Connacher A, Whittle I, et al. Adipsic hypothalamic dia-
97. Foresti V, Casati O, Villa A, et al. Central diabetes insipidus due to betes insipidus after clipping of an anterior communicating artery
acute monocytic leukemia: case report and review of the literature. aneurysm. BMJ. 1991;303(6815):1465–1467.
J Endocrinol Invest. 1992;15(2):127–130. 119. Smith D, McKenna K, Moore K, et al. Baroregulation of vasopres-
98. Dilek I, Uysal A, Demirer T, et  al. Acute myeloblastic leukemia sin release in adipsic diabetes insipidus. J Clin Endocrinol Metab.
associated with hyperleukocytosis and diabetes insipidus. Leuk 2002;87(10):4564–4568.
Lymphoma. 1998;30(5–6):657–660. 120. Cuesta M, Hannon MJ, Thompson CJ. Adipsic diabetes insipidus
99. Nieboer P, Vellenga E, Adriaanse R, van de Loosdrecht AA. Central in adult patients. Pituitary. 2017;20(3):372–380.
diabetes insipidus preceding acute myeloid leukemia with t(3;12) 121. Baylis PH, Robertson GL. Vasopressin function in familial cranial
(q26;p12). Neth J Med. 2000;56(2):45–47. diabetes insipidus. Postgrad Med J. 1981;57(663):36–40.
100. Ra’anani P, Shpilberg O, Berezin M, Ben-Bassat I. Acute leu- 122. Christensen JH, Rittig S. Familial neurohypophyseal diabetes
kemia relapse presenting as central diabetes insipidus. Cancer. insipidus—an update. Semin Nephrol. 2006;26(3):209–223.
1994;73(9):2312–2316. 123. Abu Libdeh A, Levy-Khademi F, Abdulhadi-Atwan M, et al. Auto-
101. Prosch H, Grois N, Prayer D, et al. Central diabetes insipidus as somal recessive familial neurohypophyseal diabetes insipidus: onset
presenting symptom of Langerhans cell histiocytosis. Pediatr Blood in early infancy. Eur J Endocrinol. 2010;162(2):221–226.
Cancer. 2004;43(5):594–599. 124. Ghirardello S, Garre ML, Rossi A, Maghnie M. The diagnosis of
102. Demaerel P, Van Gool S. Paediatric neuroradiological aspects children with central diabetes insipidus. J Pediatr Endocrinol Metab.
of Langerhans cell histiocytosis. Neuroradiology. 2008;50(1): 2007;20(3):359–375.
85–92. 125. Birk J, Friberg MA, Prescianotto-Baschong C, et al. Dominant pro-
103. Donadieu J, Rolon MA, Thomas C, et al. Endocrine involvement vasopressin mutants that cause diabetes insipidus form disulfide-
in pediatric-onset Langerhans’ cell histiocytosis: a population-based linked fibrillar aggregates in the endoplasmic reticulum. J Cell Sci.
study. J Pediatr. 2004;144(3):344–350. 2009;122(Pt 21):3994–4002.
104. Abla O, Weitzman S, Minkov M, et al. Diabetes insipidus in Lang- 126. Brachet C, Birk J, Christophe C, et  al. Growth retardation in
erhans cell histiocytosis: When is treatment indicated? Pediatr untreated autosomal dominant familial neurohypophyseal dia-
Blood Cancer. 2009;52(5):555–556. betes insipidus caused by one recurring and two novel muta-
105. Amato MC, Elias LL, Elias J, et al. Endocrine disorders in pediatric- tions in the vasopressin-neurophysin II gene. Eur J Endocrinol.
onset Langerhans Cell Histiocytosis. Horm Metab Res. 2006;38(11): 2011;164(2):179–187.
746–751. 127. Christensen JH, Siggaard C, Rittig S. Autosomal dominant
106. Tabuena RP, Nagai S, Handa T, et al. Diabetes insipidus from neu- familial neurohypophyseal diabetes insipidus. APMIS Suppl.
rosarcoidosis: long-term follow-up for more than eight years. Intern 2003;(109):92–95.
Med. 2004;43(10):960–966. 128. Chitturi S, Harris M, Thomsett MJ, et al. Utility of AVP gene test-
107. Caputo C, Bazargan A, McKelvie PA, et al. Hypophysitis due to ing in familial neurohypophyseal diabetes insipidus. Clin Endocri-
IgG4-related disease responding to treatment with azathioprine: nol (Oxf ). 2008;69(6):926–930.
an alternative to corticosteroid therapy. Pituitary. 2014;17(3): 129. Rohayem J, Ehlers C, Wiedemann B, et al. Diabetes and neurode-
251–256. generation in Wolfram syndrome: a multicenter study of pheno-
108. Shimatsu A, Oki Y, Fujisawa I, Sano T. Pituitary and stalk lesions type and genotype. Diabetes Care. 2011;34(7):1503–1510.
(infundibulo-hypophysitis) associated with immunoglobulin 130. Hilson JB, Merchant SN, Adams JC, Joseph JT. Wolfram
G4-related systemic disease: an emerging clinical entity. Endocr J. syndrome: a clinicopathologic correlation. Acta Neuropathol.
2009;56(9):1033–1041. 2009;118(3):415–428.
109. Abe T. Lymphocytic infundibulo-neurohypophysitis and infundib- 131. Dominguez-Roldan JM, Garcia-Alfaro C, Diaz-Parejo P, et al. Risk
ulo-panhypophysitis regarded as lymphocytic hypophysitis variant. factors associated with diabetes insipidus in brain dead patients.
Brain Tumor Pathol. 2008;25(2):59–66. Transplant Proc. 2002;34(1):13–14.
110. Maghnie M, Ghirardello S, De Bellis A, et  al. Idiopathic central 132. Saner FH, Kavuk I, Lang H, et al. Organ protective management
diabetes insipidus in children and young adults is commonly asso- of the brain-dead donor. Eur J Med Res. 2004;9(10):485–490.
ciated with vasopressin-cell antibodies and markers of autoimmu- 133. Chamorro C, Falcon JA, Michelena JC. Controversial points in
nity. Clin Endocrinol (Oxf ). 2006;65(4):470–478. organ donor management. Transplant Proc. 2009;41(8):3473–3475.

Downloaded for FK UMI Makassar (mahasiswafkumi01@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier
on November 01, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
330.e4 References

134. Oiso Y. Transient diabetes insipidus during pregnancy. Intern Med. 157. Bedford JJ, Leader JP, Jing R, et al. Amiloride restores renal medul-
2003;42(6):459–460. lary osmolytes in lithium-induced nephrogenic diabetes insipidus.
135. Barron WM, Cohen LH, Ulland LA, et  al. Transient vasopres- Am J Physiol Renal Physiol. 2008;294(4):F812–F820.
sin-resistant diabetes insipidus of pregnancy. N Engl J Med. 158. Marples D, Frokiaer J, Knepper MA, Nielsen S. Disordered water
1984;310(7):442–444. channel expression and distribution in acquired nephrogenic diabe-
136. Kennedy S, Hall PM, Seymour AE, Hague WM. Transient diabetes tes insipidus. Proc Assoc Am Physicians. 1998;110(5):401–406.
insipidus and acute fatty liver of pregnancy. Br J Obstet Gynaecol. 159. Bendz H, Sjodin I, Aurell M. Renal function on and off lithium
1994;101(5):387–391. in patients treated with lithium for 15 years or more. A controlled,
137. Krege J, Katz VL, Bowes WA Jr. Transient diabetes insipidus of prospective lithium-withdrawal study. Nephrol Dial Transplant.
pregnancy. Obstet Gynecol Surv. 1989;44(11):789–795. 1996;11(3):457–460.
138. Krysiak R, Kobielusz-Gembala I, Okopien B. Recurrent preg- 160. Bendz H, Aurell M. Drug-induced diabetes insipidus: incidence,
nancy-induced diabetes insipidus in a woman with hemochroma- prevention and management. Drug Saf. 1999;21(6):449–456.
tosis. Endocr J. 2010;57(12):1023–1028. 161. Fenske W, Quinkler M, Lorenz D, et al. Copeptin in the differ-
139. Hashimoto M, Ogura T, Otsuka F, et  al. Manifestation of sub- ential diagnosis of the polydipsia-polyuria syndrome–revisiting the
clinical diabetes insipidus due to pituitary tumor during pregnancy. direct and indirect water deprivation tests. J Clin Endocrinol Metab.
Endocr J. 1996;43(5):577–583. 2011;96(5):1506–1515.
140. Kalelioglu I, Kubat Uzum A, Yildirim A, et al. Transient gestational 162. Moses AM, Scheinman SJ, Oppenheim A. Marked hypo-
diabetes insipidus diagnosed in successive pregnancies: review of tonic polyuria resulting from nephrogenic diabetes insipidus
pathophysiology, diagnosis, treatment, and management of deliv- with partial sensitivity to vasopressin. J Clin Endocrinol Metab.
ery. Pituitary. 2007;10(1):87–93. 1984;59(6):1044–1049.
141. Amico J. Diabetes insipidus and pregnancy. In: Czernichow P, 163. Zerbe RL, Robertson GL. A comparison of plasma vasopressin
Robinson AG, eds. Diabetes Insipidus in Man. Basel: Karger; measurements with a standard indirect test in the differential diag-
1985:266–277. nosis of polyuria. N Engl J Med. 1981;305(26):1539–1546.
142. Sherer DM, Cutler J, Santoso P, et al. Severe hypernatremia after 164. Milles JJ, Spruce B, Baylis PH. A comparison of diagnostic methods
cesarean delivery secondary to transient diabetes insipidus of preg- to differentiate diabetes insipidus from primary polyuria; a review
nancy. Obstet Gynecol. 2003;102(5 Pt 2):1166–1168. of 21 patients. Acta Endocrinol (Copenh). 1983;104(4):410–416.
143. Atmaca H, Tanriverdi F, Gokce C, et  al. Posterior pituitary 165. Szinnai G, Morgenthaler NG, Berneis K, et al. Changes in plasma
function in Sheehan’s syndrome. Eur J Endocrinol. 2007;156(5): copeptin, the c-terminal portion of arginine vasopressin during
563–567. water deprivation and excess in healthy subjects. J Clin Endocrinol
144. Laway BA, Mir SA, Dar MI, Zargar AH. Sheehan’s syndrome Metab. 2007;92(10):3973–3978.
with central diabetes insipidus. Arq Bras Endocrinol Metabol. 166. Balanescu S, Kopp P, Gaskill MB, et  al. Correlation of plasma
2011;55(2):171–174. copeptin and vasopressin concentrations in hypo-, iso-, and hyper-
145. Bichet DG. Nephrogenic diabetes insipidus. Am J Med. osmolar states. J Clin Endocrinol Metab. 2011;96(4):1046–1052.
1998;105(5):431–442. 167. Winzeler B, Zweifel C, Nigro N, et al. Postoperative copeptin con-
146. van Lieburg AF, Knoers NV, Monnens LA. Clinical presentation centration predicts diabetes insipidus after pituitary surgery. J Clin
and follow-up of 30 patients with congenital nephrogenic diabetes Endocrinol Metab. 2015;100(6):2275–2282.
insipidus. J Am Soc Nephrol. 1999;10(9):1958–1964. 168. Timper K, Fenske W, Kühn F, et  al. Diagnostic accuracy of
147. Bichet DG. Hereditary polyuric disorders: new concepts and dif- copeptin in the differential diagnosis of the polyuria-polydipsia
ferential diagnosis. Semin Nephrol. 2006;26(3):224–233. syndrome: a prospective multicenter study. J Clin Endocrinol
148. Spanakis E, Milord E, Gragnoli C. AVPR2 variants and mutations Metab. 2015;100(6):2268–2274.
in nephrogenic diabetes insipidus: review and missense mutation 169. Thompson CJ, Burd JM, Baylis PH. Acute suppression of plasma
significance. J Cell Physiol. 2008;217(3):605–617. vasopressin and thirst after drinking in hypernatraemic man. Am J
149. Bichet DG. Physiopathology of hereditary polyuric states: a molec- Physiol. 1987;252(6 Pt 2):R1138–R1142.
ular view of renal function. Swiss Med Wkly. 2012;142:w13613. 170. Thompson CJ, Selby P, Baylis PH. Reproducibility of osmotic and
150. Schoneberg T, Schulz A, Biebermann H, et  al. V2 vasopressin nonosmotic tests of vasopressin secretion in men. Am J Physiol.
receptor dysfunction in nephrogenic diabetes insipidus caused 1991;260(3 Pt 2):R533–R539.
by different molecular mechanisms. Hum Mutat. 1998;12(3): 171. Thompson CJ, Edwards CR, Baylis PH. Osmotic and non-osmotic
196–205. regulation of thirst and vasopressin secretion in patients with
151. Satoh M, Ogikubo S, Yoshizawa-Ogasawara A. Correlation between compulsive water drinking. Clin Endocrinol (Oxf ). 1991;35(3):
clinical phenotypes and X-inactivation patterns in six female carri- 221–228.
ers with heterozygote vasopressin type 2 receptor gene mutations. 172. Arslan A, Karaarslan E, Dincer A. High intensity signal of the
Endocr J. 2008;55(2):277–284. posterior pituitary. A study with horizontal direction of fre-
152. Wildin RS, Cogdell DE. Clinical utility of direct mutation testing quency-encoding and fat suppression MR techniques. Acta Radiol.
for congenital nephrogenic diabetes insipidus in families. Pediatrics. 1999;40(2):142–145.
1999;103(3):632–639. 173. Gudinchet F, Brunelle F, Barth MO, et  al. MR imaging of
153. Sands JM, Bichet DG. Nephrogenic diabetes insipidus. Ann Intern the posterior hypophysis in children. AJR Am J Roentgenol.
Med. 2006;144(3):186–194. 1989;153(2):351–354.
154. Iolascon A, Aglio V, Tamma G, et al. Characterization of two novel 174. Moses AM, Clayton B, Hochhauser L. Use of T1-weighted MR
missense mutations in the AQP2 gene causing nephrogenic diabe- imaging to differentiate between primary polydipsia and cen-
tes insipidus. Nephron Physiol. 2007;105(3):33–41. tral diabetes insipidus. AJNR Am J Neuroradiol. 1992;13(5):
155. van Os CH, Deen PM. Aquaporin-2 water channel mutations 1273–1277.
causing nephrogenic diabetes insipidus. Proc Assoc Am Physicians. 175. Tien R, Kucharczyk J, Kucharczyk W. MR imaging of the brain
1998;110(5):395–400. in patients with diabetes insipidus. AJNR Am J Neuroradiol.
156. Robben JH, Knoers NV, Deen PM. Cell biological aspects of 1991;12(3):533–542.
the vasopressin type-2 receptor and aquaporin 2 water channel 176. Brooks BS, el Gammal T, Allison JD, Hoffman WH. Frequency
in nephrogenic diabetes insipidus. Am J Physiol Renal Physiol. and variation of the posterior pituitary bright signal on MR images.
2006;291(2):F257–F270. AJNR Am J Neuroradiol. 1989;10(5):943–948.

Downloaded for FK UMI Makassar (mahasiswafkumi01@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier
on November 01, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
References 330.e5

177. Saeki N, Tokunaga H, Wagai N, et  al. MRI of ectopic posterior 199. Juul KV, Klein BM, Sandstrom R, et  al. Gender difference in
pituitary bright spot with large adenomas: appearances and rela- antidiuretic response to desmopressin. Am J Physiol Renal Physiol.
tionship to transient postoperative diabetes insipidus. Neuroradiol- 2011;300(5):F1116–F1122.
ogy. 2003;45(10):713–716. 200. Rivkees SA, Dunbar N, Wilson TA. The management of cen-
178. Maghnie M, Genovese E, Bernasconi S, et al. Persistent high MR tral diabetes insipidus in infancy: desmopressin, low renal sol-
signal of the posterior pituitary gland in central diabetes insipidus. ute load formula, thiazide diuretics. J Pediatr Endocrinol Metab.
AJNR Am J Neuroradiol. 1997;18(9):1749–1752. 2007;20(4):459–469.
179. Miyamoto S, Sasaki N, Tanabe Y. Magnetic resonance imaging 201. Bononi PL, Robinson AG. Central diabetes insipidus: management
in familial central diabetes insipidus. Neuroradiology. 1991;33(3): in the postoperative period. Endocrinologist. 1991;1(3):180–186.
272–273. 202. Campigotto MJ, Koczmara C, Greenall J, Hyland S. Desmopres-
180. Fujisawa I. Magnetic resonance imaging of the hypothalamic- sin (dDAVP) incident signals the need for enhanced monitoring
neurohypophyseal system. J Neuroendocrinol. 2004;16(4): protocols. Dynamics. 2008;19(3):34–36.
297–302. 203. Loh JA, Verbalis JG. Disorders of water and salt metabolism asso-
181. Maghnie M, Villa A, Arico M, et  al. Correlation between mag- ciated with pituitary disease. Endocrinol Metab Clin North Am.
netic resonance imaging of posterior pituitary and neurohypophy- 2008;37(1):213–234, x.
seal function in children with diabetes insipidus. J Clin Endocrinol 204. Hannon MJ, Crowley RK, Behan LA, et al. Acute glucocorticoid
Metab. 1992;74(4):795–800. deficiency and diabetes insipidus are common after acute trau-
182. Kubota T, Yamamoto T, Ozono K, Shimotsuji T. Hyperintensity matic brain injury and predict mortality. J Clin Endocrinol Metab.
of posterior pituitary on MR T1WI in a boy with central diabe- 2013;98(8):3229–3237.
tes insipidus caused by missense mutation of neurophysin II gene. 205. Mavrakis AN, Tritos NA. Diabetes insipidus with deficient thirst:
Endocr J. 2001;48(4):459–463. report of a patient and review of the literature. Am J Kidney Dis.
183. Yamamoto T, Ishii T, Yoshioka K, et  al. Transient central diabe- 2008;51(5):851–859.
tes insipidus in pregnancy with a peculiar change in signal inten- 206. Ball SG, Vaidja B, Baylis PH. Hypothalamic adipsic syndrome: diag-
sity on T1-weighted magnetic resonance images. Intern Med. nosis and management. Clin Endocrinol (Oxf). 1997;47(4):405–409.
2003;42(6):513–516. 207. Edwards CR, Kitau MJ, Chard T, Besser GM. Vasopressin ana-
184. Heinbecker P, White HL. Hypothalamico-hypophysial sys- logue DDAVP in diabetes insipidus: clinical and laboratory studies.
tem and its relation to water balance in the dog. Am J Physiol. Br Med J. 1973;3(5876):375–378.
1941;133(3):582–593. 208. Davison JM, Sheills EA, Philips PR, et al. Metabolic clearance of
185. Di Iorgi N, Allegri AE, Napoli F, et  al. Central diabetes insipi- vasopressin and an analogue resistant to vasopressinase in human
dus in children and young adults: etiological diagnosis and pregnancy. Am J Physiol. 1993;264(2 Pt 2):F348–F353.
long-term outcome of idiopathic cases. J Clin Endocrinol Metab. 209. Kallen BA, Carlsson SS, Bengtsson BK. Diabetes insipidus and
2014;99(4):1264–1272. use of desmopressin (Minirin) during pregnancy. Eur J Endocrinol.
186. Di Iorgi N, Napoli F, Allegri AE, et al. Diabetes insipidus—diag- 1995;132(2):144–146.
nosis and management. Horm Res Paediatr. 2012;77(2):69–84. 210. Ray JG. DDAVP use during pregnancy: an analysis of its safety for
187. Godano E, Morana G, Di Iorgi N, et al. Role of MRI T2-DRIVE mother and child. Obstet Gynecol Surv. 1998;53(7):450–455.
in the assessment of pituitary stalk abnormalities without gado- 211. Faerch M, Christensen JH, Corydon TJ, et al. Partial nephrogenic
linium in pituitary diseases. Eur J Endocrinol. 2018;178(6): diabetes insipidus caused by a novel mutation in the AVPR2 gene.
613–622. Clin Endocrinol (Oxf ). 2008;68(3):395–403.
188. Czernichow P, Garel C, Leger J. Thickened pituitary stalk on mag- 212. Postina R, Ufer E, Pfeiffer R, et  al. Misfolded vasopressin V2
netic resonance imaging in children with central diabetes insipidus. receptors caused by extracellular point mutations entail congential
Horm Res. 2000;53(suppl 3):61–64. nephrogenic diabetes insipidus. Mol Cell Endocrinol. 2000;164(1–
189. Leger J, Velasquez A, Garel C, et al. Thickened pituitary stalk on 2):31–39.
magnetic resonance imaging in children with central diabetes insip- 213. Magaldi AJ. New insights into the paradoxical effect of thia-
idus. J Clin Endocrinol Metab. 1999;84(6):1954–1960. zides in diabetes insipidus therapy. Nephrol Dial Transplant.
190. Maghnie M. Diabetes insipidus. Horm Res. 2003;59(suppl 1):42–54. 2000;15(12):1903–1905.
191. Robertson GL. Diabetes insipidus. Endocrinol Metab Clin North 214. Loffing J. Paradoxical antidiuretic effect of thiazides in diabe-
Am. 1995;24(3):549–572. tes insipidus: another piece in the puzzle. J Am Soc Nephrol.
192. Robinson AG, Verbalis JG. Diabetes insipidus. Curr Ther Endocri- 2004;15(11):2948–2950.
nol Metab. 1997;6:1–7. 215. Singer I, Oster JR, Fishman LM. The management of diabetes
193. Robinson AG. DDAVP in the treatment of central diabetes insipi- insipidus in adults. Arch Intern Med. 1997;157(12):1293–1301.
dus. N Engl J Med. 1976;294(10):507–511. 216. Nguyen MK, Nielsen S, Kurtz I. Molecular pathogenesis of neph-
194. Oiso Y, Robertson GL, Norgaard JP, Juul KV. Clinical review: rogenic diabetes insipidus. Clin Exp Nephrol. 2003;7(1):9–17.
Treatment of neurohypophyseal diabetes insipidus. J Clin Endocri- 217. Robben JH, Deen PM. Pharmacological chaperones in nephro-
nol Metab. 2013;98(10):3958–3967. genic diabetes insipidus: possibilities for clinical application. Bio-
195. Juul KV, Van Herzeele C, De Bruyne P, et al. Desmopressin melt Drugs. 2007;21(3):157–166.
improves response and compliance compared with tablet in treat- 218. Jean-Alphonse F, Perkovska S, Frantz MC, et  al. Biased ago-
ment of primary monosymptomatic nocturnal enuresis. Eur J Pedi- nist pharmacochaperones of the AVP V2 receptor may treat
atr. 2013;172(9):1235–1242. congenital nephrogenic diabetes insipidus. J Am Soc Nephrol.
196. Lam KS, Wat MS, Choi KL, et al. Pharmacokinetics, pharmacody- 2009;20(10):2190–2203.
namics, long-term efficacy and safety of oral 1-deamino-8-D-argi- 219. Robben JH, Kortenoeven ML, Sze M, et  al. Intracellular activa-
nine vasopressin in adult patients with central diabetes insipidus. Br tion of vasopressin V2 receptor mutants in nephrogenic diabe-
J Clin Pharmacol. 1996;42(3):379–385. tes insipidus by nonpeptide agonists. Proc Natl Acad Sci U S A.
197. Richardson DW, Robinson AG. Desmopressin. Ann Intern Med. 2009;106(29):12195–12200.
1985;103(2):228–239. 220. Takahashi K, Makita N, Manaka K, et  al. V2 vasopressin recep-
198. Juul KV, Erichsen L, Robertson GL. Temporal delays and individ- tor (V2R) mutations in partial nephrogenic diabetes insipidus
ual variation in antidiuretic response to desmopressin. Am J Physiol highlight protean agonism of V2R antagonists. J Biol Chem.
Renal Physiol. 2013;304(3):F268–F278. 2012;287(3):2099–2106.

Downloaded for FK UMI Makassar (mahasiswafkumi01@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier
on November 01, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
330.e6 References

221. Bichet DG. Vasopressin receptor mutations in nephrogenic diabe- 247. Holliday MA, Kalayci MN, Harrah J. Factors that limit brain vol-
tes insipidus. Semin Nephrol. 2008;28(3):245–251. ume changes in response to acute and sustained hyper- and hypo-
222. Moug SJ, McKee RF, O’Reilly DS, et al. The perioperative chal- natremia. J Clin Invest. 1968;47(8):1916–1928.
lenge of nephrogenic diabetes insipidus: a multidisciplinary 248. Garcia-Perez A, Burg MB. Renal medullary organic osmolytes.
approach. Surgeon. 2005;3(2):89–94. Physiol Rev. 1991;71(4):1081–1115.
223. Bagshaw SM, Townsend DR, McDermid RC. Disorders of 249. Heilig CW, Stromski ME, Blumenfeld JD, et al. Characterization
sodium and water balance in hospitalized patients. Can J Anaesth. of the major brain osmolytes that accumulate in salt-loaded rats.
2009;56(2):151–167. Am J Physiol. 1989;257(6 Pt 2):F1108–F1116.
224. Lien YH, Shapiro JI, Chan L. Effects of hypernatremia on organic 250. Lien YH, Shapiro JI, Chan L. Study of brain electrolytes and
brain osmoles. J Clin Invest. 1990;85(5):1427–1435. organic osmolytes during correction of chronic hyponatremia.
225. Adrogue HJ, Madias NE. Hypernatremia. N Engl J Med. Implications for the pathogenesis of central pontine myelinolysis. J
2000;342(20):1493–1499. Clin Invest. 1991;88(1):303–309.
226. Schwartz WB, Bennett W, Curelop S, Bartter FC. A syndrome 251. Verbalis JG, Gullans SR. Hyponatremia causes large sustained
of renal sodium loss and hyponatremia probably resulting from reductions in brain content of multiple organic osmolytes in rats.
inappropriate secretion of antidiuretic hormone. Am J Med. Brain Res. 1991;567(2):274–282.
1957;23(4):529–542. 252. Videen JS, Michaelis T, Pinto P, Ross BD. Human cerebral osmo-
227. Bartter FC, Schwartz WB. The syndrome of inappropriate secre- lytes during chronic hyponatremia. A proton magnetic resonance
tion of antidiuretic hormone. Am J Med. 1967;42(5):790–806. spectroscopy study. J Clin Invest. 1995;95(2):788–793.
228. Upadhyay A, Jaber BL, Madias NE. Incidence and prevalence of 253. Gullans SR, Verbalis JG. Control of brain volume during hyperosmo-
hyponatremia. The Am J Med. 2006;119(7 suppl 1):S30–S35. lar and hypoosmolar conditions. Annu Rev Med. 1993;44:289–301.
229. Wald R, Jaber BL, Price LL, et  al. Impact of hospital-associ- 254. Graber M, Corish D. The electrolytes in hyponatremia. Am J Kid-
ated hyponatremia on selected outcomes. Arch Intern Med. ney Dis. 1991;18(5):527–545.
2010;170(3):294–302. 255. Smith MJ Jr., Cowley MJ Jr., Guyton AC, Manning RD Jr. Acute
230. Hawkins RC. Age and gender as risk factors for hyponatremia and and chronic effects of vasopressin on blood pressure, electro-
hypernatremia. Clin Chim Acta. 2003;337(1–2):169–172. lytes, and fluid volumes. Am J Physiol. 1979;237(3):F232–F240.
231. Miller M, Morley JE, Rubenstein LZ. Hyponatremia in a nursing 256. Verbalis JG. Pathogenesis of hyponatremia in an experimental
home population. J Am Geriatr Soc. 1995;43(12):1410–1413. model of the syndrome of inappropriate antidiuresis. Am J Physiol.
232. Arieff AI. Hyponatremia, convulsions, respiratory arrest, and per- 1994;267(6 Pt 2):R1617–R1625.
manent brain damage after elective surgery in healthy women. N 257. Southgate HJ, Burke BJ, Walters G. Body space measurements in the
Engl J Med. 1986;314(24):1529–1535. hyponatraemia of carcinoma of the bronchus: evidence for the chronic
233. Sterns RH, Riggs JE, Schochet SS Jr. Osmotic demyelination ‘sick cell’ syndrome? Ann Clin Biochem. 1992;29(Pt 1):90–95.
syndrome following correction of hyponatremia. N Engl J Med. 258. Clark BA, Shannon RP, Rosa RM, Epstein FH. Increased suscep-
1986;314(24):1535–1542. tibility to thiazide-induced hyponatremia in the elderly. J Am Soc
234. Anderson RJ, Chung HM, Kluge R, Schrier RW. Hyponatremia: Nephrol. 1994;5(4):1106–1111.
a prospective analysis of its epidemiology and the pathogenetic role 259. Chung HM, Kluge R, Schrier RW, Anderson RJ. Clinical assess-
of vasopressin. Ann Intern Med. 1985;102(2):164–168. ment of extracellular fluid volume in hyponatremia. Am J Med.
235. Corona G, Giuliani C, Parenti G, et al. Moderate hyponatremia is 1987;83(5):905–908.
associated with increased risk of mortality: evidence from a meta- 260. Michelis MF, Fusco RD, Bragdon RW, Davis BB. Reset of osmore-
analysis. PLoS One. 2013;8(12):e80451. ceptors in association with normovolemic hyponatremia. Am J Med
236. Weisberg LS. Pseudohyponatremia: a reappraisal. Am J Med. Sci. 1974;267(5):267–273.
1989;86(3):315–318. 261. Beck LH. Hypouricemia in the syndrome of inappropriate secretion
237. Katz MA. Hyperglycemia-induced hyponatremia—calcula- of antidiuretic hormone. N Engl J Med. 1979;301(10):528–530.
tion of expected serum sodium depression. N Engl J Med. 262. Zerbe R, Stropes L, Robertson G. Vasopressin function in the syn-
1973;289(16):843–844. drome of inappropriate antidiuresis. Annu Rev Med. 1980;31:315–
238. Hillier TA, Abbott RD, Barrett EJ. Hyponatremia: evaluating 327.
the correction factor for hyperglycemia. Am J Med. 1999;106(4): 263. List AF, Hainsworth JD, Davis BW, et al. The syndrome of inap-
399–403. propriate secretion of antidiuretic hormone (SIAD) in small-cell
239. Rose BD. New approach to disturbances in the plasma sodium lung cancer. J Clin Oncol. 1986;4(8):1191–1198.
concentration. Am J Med. 1986;81(6):1033–1040. 264. Maurer LH, O’Donnell JF, Kennedy S, et al. Human neurophy-
240. Spital A. Diuretic-induced hyponatremia. Am J Med. sins in carcinoma of the lung: relation to histology, disease stage,
1999;19(4):447–452. response rate, survival, and syndrome of inappropriate antidiuretic
241. Gross PA, Pehrisch H, Rascher W, et al. Pathogenesis of clinical hypo- hormone secretion. Cancer Treat Rep. 1983;67(11):971–976.
natremia: observations of vasopressin and fluid intake in 100 hypona- 265. Ferlito A, Rinaldo A, Devaney KO. Syndrome of inappropriate
tremic medical patients. Eur J Clin Invest. 1987;17(2):123–129. antidiuretic hormone secretion associated with head neck cancers:
242. Schrier RW. Body fluid volume regulation in health and disease: a review of the literature. Ann Otol Rhinol Laryngol. 1997;106(10 Pt
unifying hypothesis. Ann Intern Med. 1990;113(2):155–159. 1):878–883.
243. Demanet JC, Bonnyns M, Bleiberg H, Stevens-Rocmans 266. Kavanagh BD, Halperin EC, Rosenbaum LC, et al. Syndrome of
C. Coma due to water intoxication in beer drinkers. Lancet. inappropriate secretion of antidiuretic hormone in a patient with
1971;2(7734):1115–1117. carcinoma of the nasopharynx. Cancer. 1992;69(6):1315–1319.
244. Cooke CR, Turin MD, Walker WG. The syndrome of inappro- 267. Berghmans T, Paesmans M, Body JJ. A prospective study on hypo-
priate antidiuretic hormone secretion (SIADH): pathophysiologic natraemia in medical cancer patients: epidemiology, aetiology and
mechanisms in solute and volume regulation. Medicine (Baltimore). differential diagnosis. Support Care Cancer. 2000;8(3):192–197.
1979;58(3):240–251. 268. Moses AM, Miller M. Drug-induced dilutional hyponatremia. N
245. Grantham J, Linshaw M. The effect of hyponatremia on the regu- Engl J Med. 1974;291(23):1234–1239.
lation of intracellular volume and solute composition. Circ Res. 269. Wilkinson TJ, Begg EJ, Winter AC, Sainsbury R. Incidence
1984;54(5):483–491. and risk factors for hyponatraemia following treatment with
246. Yannet H. Changes in the brain resulting from depletion of extra- fluoxetine or paroxetine in elderly people. Br J Clin Pharmacol.
cellular electrolytes. Am J Physiol. 1940;128(4):683–689. 1999;47(2):211–217.

Downloaded for FK UMI Makassar (mahasiswafkumi01@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier
on November 01, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
References 330.e7

270. Burgess C, O’Donohoe A, Gill M. Agony and ecstasy: a review of 292. Verbalis JG, Murase T, Ecelbarger CA, et al. Studies of renal aqua-
MDMA effects and toxicity. Eur Psychiatry. 2000;15(5):287–294. porin-2 expression during renal escape from vasopressin-induced
271. Kelestimur H, Leach RM, Ward JP, Forsling ML. Vasopressin and antidiuresis. Adv Exp Med Biol. 1998;449:395–406.
oxytocin release during prolonged environmental hypoxia in the 293. Arieff AI, Llach F, Massry SG. Neurological manifestations and
rat. Thorax. 1997;52(1):84–88. morbidity of hyponatremia: correlation with brain water and elec-
272. Tang WW, Kaptein EM, Feinstein EI, Massry SG. Hyponatre- trolytes. Medicine (Baltimore). 1976;55(2):121–129.
mia in hospitalized patients with the acquired immunodeficiency 294. Fraser CL, Arieff AI. Epidemiology, pathophysiology, and manage-
syndrome (AIDS) and the AIDS-related complex. Am J Med. ment of hyponatremic encephalopathy. Am J Med. 1997;102(1):
1993;94(2):169–174. 67–77.
273. Yeung KT, Chan M, Chan CK. The safety of i.v. pentamidine admin- 295. Daggett P, Deanfield J, Moss F. Neurological aspects of hypona-
istered in an ambulatory setting. Chest. 1996;110(1):136–140. traemia. Postgrad Med J. 1982;58(686):737–740.
274. Miller M, Hecker MS, Friedlander DA, Carter JM. Apparent idio- 296. Ayus JC, Wheeler JM, Arieff AI. Postoperative hyponatremic
pathic hyponatremia in an ambulatory geriatric population. J Am encephalopathy in menstruant women. Ann Intern Med.
Geriatr Soc. 1996;44(4):404–408. 1992;117(11):891–897.
275. Hirshberg B, Ben-Yehuda A. The syndrome of inappropri- 297. Wijdicks EF, Sharbrough FW. New-onset seizures in critically ill
ate antidiuretic hormone secretion in the elderly. Am J Med. patients. Neurology. 1993;43(5):1042–1044.
1997;103(4):270–273. 298. Vexler ZS, Ayus JC, Roberts TP, et  al. Hypoxic and ischemic
276. Ishikawa S, Kuratomi Y, Saito T. A case of oat cell carcinoma of the hypoxia exacerbate brain injury associated with metabolic encepha-
lung associated with ectopic production of ADH, neurophysin and lopathy in laboratory animals. J Clin Invest. 1994;93(1):256–264.
ACTH. Endocrinol Jpn. 1980;27(2):257–263. 299. Ayus JC, Arieff AI. Pulmonary complications of hyponatremic
277. Kamoi K. Syndrome of inappropriate antidiuresis without encephalopathy. Noncardiogenic pulmonary edema and hypercap-
involving inappropriate secretion of vasopressin in an elderly nic respiratory failure. Chest. 1995;107(2):517–521.
woman: effect of intravenous administration of the nonpep- 300. Ayus JC, Varon J, Arieff AI. Hyponatremia, cerebral edema, and
tide vasopressin V2 receptor antagonist OPC-31260. Nephron. noncardiogenic pulmonary edema in marathon runners. Ann
1997;76(1):111–115. Intern Med. 2000;132(9):711–714.
278. Feldman BJ, Rosenthal SM, Vargas GA, et al. Nephrogenic syndrome 301. Arieff AI, Ayus JC, Fraser CL. Hyponatraemia and death or per-
of inappropriate antidiuresis. N Engl J Med. 2005;352(18):1884– manent brain damage in healthy children. BMJ. 1992;304(6836):
1890. 1218–1222.
279. Leaf A, Bartter FC, Santos RF, Wrong O. Evidence in man that 302. Wattad A, Chiang ML, Hill LL. Hyponatremia in hospitalized chil-
urinary electrolyte loss induced by pitressin is a function of water dren. Clin Pediatr (Phila). 1992;31(3):153–157.
retention. J Clin Invest. 1953;32(9):868–878. 303. Wijdicks EF, Larson TS. Absence of postoperative hyponatremia
280. Verbalis JG. Whole-body volume regulation and escape from antid- syndrome in young, healthy females. Ann Neurol. 1994;35(5):626–
iuresis. Am J Med. 2006;119(7 suppl 1):S21–S29. 628.
281. Peters JP, Welt LG, Sims EA, et  al. A salt-wasting syndrome 304. Corona G, Giuliani C, Verbalis JG, et al. Hyponatremia improve-
associated with cerebral disease. Trans Assoc Am Physicians. ment is associated with a reduced risk of mortality: evidence from a
1950;63:57–64. meta-analysis. PLoS One. 2015;10(4):e0124105.
282. Nelson PB, Seif S, Gutai J, Robinson AG. Hyponatremia and natri- 305. Holland-Bill L, Christiansen CF, Heide-Jørgensen U, et al. Hypo-
uresis following subarachnoid hemorrhage in a monkey model. J natremia and mortality risk: a Danish cohort study of 279508
Neurosurg. 1984;60(2):233–237. acutely hospitalized patients. Eur J Endocrinol. 2015;173(1):71–81.
283. Wijdicks EF, Ropper AH, Hunnicutt EJ, et  al. Atrial natriuretic 306. Al Mawed S, Pankratz VS, Chong K, et al. Low serum sodium lev-
factor and salt wasting after aneurysmal subarachnoid hemorrhage. els at hospital admission: outcomes among 2.3 million hospitalized
Stroke. 1991;22(12):1519–1524. patients. PLoS One. 2018;13(3):e0194379.
284. Diringer MN, Lim JS, Kirsch JR, Hanley DF. Suprasellar and 307. Hoorn EJ, Rivadeneira F, van Meurs JB, et al. Mild hyponatremia
intraventricular blood predict elevated plasma atrial natriuretic fac- as a risk factor for fractures: the Rotterdam Study. J Bone Miner Res.
tor in subarachnoid hemorrhage. Stroke. 1991;22(5):577–581. 2011;26(8):1822–1828.
285. Diringer MN, Wu KC, Verbalis JG, Hanley DF. Hypervolemic 308. Sterns RH. Severe symptomatic hyponatremia: treatment and out-
therapy prevents volume contraction but not hyponatremia come. A study of 64 cases. Ann Intern Med. 1987;107(5):656–664.
following subarachnoid hemorrhage. Ann Neurol. 1992;31(5): 309. Renneboog B, Musch W, Vandemergel X, et  al. Mild chronic
543–550. hyponatremia is associated with falls, unsteadiness, and attention
286. Cerda-Esteve M, Cuadrado-Godia E, Chillaron JJ, et al. Cere- deficits. Am J Med. 2006;119(1):71 e71–78.
bral salt wasting syndrome: review. Eur J Intern Med. 2008;19(4): 310. Verbalis JG, Goldsmith SR, Greenberg A, et al. Diagnosis, evalu-
249–254. ation, and treatment of hyponatremia: expert panel recommenda-
287. Palmer BF. Hyponatremia in patients with central nervous tions. Am J Med. 2013;126(10 suppl 1):S1–S42.
system disease: SIAD versus CSW. Trends Endocrinol Metab. 311. Rittenhouse KJ, To T, Rogers A, et  al. Hyponatremia as a fall
2003;14(4):182–187. predictor in a geriatric trauma population. Injury. 2015;46(1):
288. Kinik ST, Kandemir N, Baykan A, et  al. Fludrocortisone treat- 119–123.
ment in a child with severe cerebral salt wasting. Pediatr Neurosurg. 312. Fujisawa H, Sugimura Y, Takagi H, et al. Chronic hyponatremia
2001;35(4):216–219. causes neurologic and psychologic impairments. J Am Soc Nephrol.
289. von Bismarck P, Ankermann T, Eggert P, et  al. Diagnosis and 2016;27(3):766–780.
management of cerebral salt wasting (CSW) in children: the role 313. Vandergheynst F, Gombeir Y, Bellante F, et al. Impact of hypona-
of atrial natriuretic peptide (ANP) and brain natriuretic peptide tremia on nerve conduction and muscle strength. Eur J Clin Invest.
(BNP). Childs Nerv Syst. 2006;22(10):1275–1281. 2016;46(4):328–333.
290. Taplin CE, Cowell CT, Silink M, Ambler GR. Fludrocorti- 314. Gankam Kengne F, Andres C, Sattar L, et  al. Mild hypona-
sone therapy in cerebral salt wasting. Pediatrics. 2006;118(6): tremia and risk of fracture in the ambulatory elderly. QJM.
e1904–1908. 2008;101(7):583–588.
291. Tian Y, Sandberg K, Murase T. Bet al. Vasopressin V2 receptor 315. Kinsella S, Moran S, Sullivan MO, et al. Hyponatremia indepen-
binding is down-regulated during renal escape from vasopressin- dent of osteoporosis is associated with fracture occurrence. Clin J
induced antidiuresis. Endocrinology. 2000;141(1):307–314. Am Soc Nephrol. 2010;5(2):275–280.

Downloaded for FK UMI Makassar (mahasiswafkumi01@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier
on November 01, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
330.e8 References

316. Sandhu HS, Gilles E, DeVita MV, et  al. Hyponatremia associ- 339. Schrier RW, Gross P, Gheorghiade M, et al. Tolvaptan, a selective
ated with large-bone fracture in elderly patients. Int Urol Nephrol. oral vasopressin V2-receptor antagonist, for hyponatremia. N Engl
2009;41(3):733–737. J Med. 2006;355(20):2099–2112.
317. Verbalis JG, Barsony J, Sugimura Y, et al. Hyponatremia-induced 340. Otsuka Pharmaceutical Company. Samsca (Tolvaptan) Prescribing
osteoporosis. J Bone Miner Res. 2010;25(3):554–563. Information; 2009.
318. Kruse C, Eiken P, Verbalis J, Vestergaard P. The effect of chronic 341. Castello LM, Baldrighi M, Panizza A, et al. Efficacy and safety of two
mild hyponatremia on bone mineral loss evaluated by retrospective different tolvaptan doses in the treatment of hyponatremia in the
national Danish patient data. Bone. 2016;84:9–14. Emergency Department. Intern Emerg Med. 2017;12(7):993–1001.
319. Afshinnia F, Sundaram B, Ackermann RJ, Wong KK. Hyponatre- 342. Torres VE, Chapman AB, Devuyst O, et al. Tolvaptan in patients
mia and osteoporosis: reappraisal of a novel association. Osteoporos with autosomal dominant polycystic kidney disease. N Engl J Med.
Int. 2015;26(9):2291–2298. 2012;367(25):2407–2418.
320. Usala RL, Fernandez SJ, Mete M, et al. Hyponatremia is associated 343. Corona G, Giuliani C, Parenti G, et al. The economic burden of
with increased osteoporosis and bone fractures in a large US health hyponatremia: systematic review and meta-analysis. Am J Med.
system population. J Clin Endocrinol Metab. 2015;100(8):3021–3031. 2016;129(8):823–835.e4.
321. Ayus JC, Fuentes NA, Negri AL, et  al. Mild prolonged chronic 344. Abola MV, Tanenbaum JE, Bomberger TT, et  al. Preoperative
hyponatremia and risk of hip fracture in the elderly. Nephrol Dial hyponatremia is associated with reoperation and prolonged length
Transplant. 2016;31(10):1662–1669. of hospital stay following total knee arthroplasty. J Knee Surg. 2018.
322. Nowak KL, Yaffe K, Orwoll ES, et al. Serum sodium and cogni- [Epub ahead of print].
tion in older community-dwelling men. Clin J Am Soc Nephrol. 345. Ramamohan V, Mladsi D, Ronquest N, et al. An economic analysis of
2018;13(3):366–374. tolvaptan compared with fluid restriction among hospitalized patients
323. Barsony J, Manigrasso MB, Xu Q, et  al. Chronic hyponatremia with hyponatremia. Hosp Pract (1995). 2017;45(3):111–117.
exacerbates multiple manifestations of senescence in male rats. Age 346. Coussement J, Danguy C, Zouaoui-Boudjeltia K, et al. Treatment of
(Dordr). 2013;35(2):271–288. the syndrome of inappropriate secretion of antidiuretic hormone with
324. Ayus JC. Diuretic-induced hyponatremia. Arch Intern Med. urea in critically ill patients. Am J Nephrol. 2012;35(3):265–270.
1986;146(7):1295–1296. 347. Decaux G, Andres C, Gankam Kengne F, Soupart A. Treatment
325. Adrogue HJ, Madias NE. Hyponatremia. N Engl J Med. of euvolemic hyponatremia in the intensive care unit by urea. Crit
2000;342(21):1581–1589. Care. 2010;14(5):R184.
326. Verbalis JG, Grossman A, Hoybye C, Runkle I. Review and analy- 348. Chehade H, Rosato L, Girardin E, Cachat F. Inappropriate antidi-
sis of differing regulatory indications and expert panel guide- uretic hormone secretion: long-term successful urea treatment. Acta
lines for the treatment of hyponatremia. Curr Med Res Opin. Paediatr. 2012;101(1):e39–e42.
2014;30(7):1201–1207. 349. Levtchenko EN, Monnens LA. Nephrogenic syndrome of inap-
327. Perez CA, Figueroa SA. Complication rates of 3% hypertonic saline propriate antidiuresis. Nephrol Dial Transplant. 2010;25(9):
infusion through peripheral intravenous access. J Neurosci Nurs. 2839–2843.
2017;49(3):191–195. 350. Soupart A, Coffernils M, Couturier B, et al. Efficacy and tolerance
328. Hew-Butler T, Ayus JC, Kipps C, et  al. Statement of the Sec- of urea compared with vaptans for long-term treatment of patients
ond International Exercise-Associated Hyponatremia Consensus with SIAD. Clin J Am Soc Nephrol. 2012;7(5):742–747.
Development Conference, New Zealand, 2007. Clin J Sport Med. 351. Sterns RH, Silver SM, Hix JK. Urea for hyponatremia? Kidney Int.
2008;18(2):111–121. 2015;87(2):268–270.
329. Spasovski G, Vanholder R, Allolio B, et al. Clinical practice guide- 352. Hantman D, Rossier B, Zohlman R, Schrier R. Rapid correction of
line on diagnosis and treatment of hyponatraemia. Nephrol Dial hyponatremia in the syndrome of inappropriate secretion of antidi-
Transplant. 2014;29(suppl 2):i1–i39. uretic hormone. An alternative treatment to hypertonic saline. Ann
330. Sterns RH, Cappuccio JD, Silver SM, Cohen EP. Neurologic Intern Med. 1973;78(6):870–875.
sequelae after treatment of severe hyponatremia: a multicenter per- 353. Decaux G, Waterlot Y, Genette F, Mockel J. Treatment of the
spective. J Am Soc Nephrol. 1994;4(8):1522–1530. syndrome of inappropriate secretion of antidiuretic hormone with
331. Battison C, Andrews PJ, Graham C, Petty T. Randomized, controlled furosemide. N Engl J Med. 1981;304(6):329–330.
trial on the effect of a 20% mannitol solution and a 7.5% saline/6% 354. Greenberg A, Verbalis JG, Amin AN, et al. Current treatment prac-
dextran solution on increased intracranial pressure after brain injury. tice and outcomes. Report of the hyponatremia registry. Kidney Int.
Crit Care Med. 2005;33(1):196–202, discussion 257–198. 2015;88(1):167–177.
332. Steele A, Gowrishankar M, Abrahamson S, et  al. Postoperative 355. Verbalis JG, Greenberg A, Burst V, et al. Diagnosing and treating
hyponatremia despite near-isotonic saline infusion: a phenomenon the syndrome of inappropriate antidiuretic hormone secretion. Am
of desalination. Ann Intern Med. 1997;126(1):20–25. J Med. 2016;129(5):537.e9–537.e23.
333. Robertson GL. Regulation of arginine vasopressin in the syn- 356. Cuesta M, Ortolá A, Garrahy A, et  al. Predictors of failure to
drome of inappropriate antidiuresis. Am J Med. 2006;119(7 suppl respond to fluid restriction in SIAD in clinical practice; time to
1):S36–42. re-evaluate clinical guidelines? QJM. 2017;110(8):489–492.
334. Berl T. Impact of solute intake on urine flow and water excretion. J 357. Ellison DH, Berl T. Clinical practice. The syndrome of inappropri-
Am Soc Nephrol. 2008;19(6):1076–1078. ate antidiuresis. The N Engl J Med. 2007;356(20):2064–2072.
335. Furst H, Hallows KR, Post J, et  al. The urine/plasma electro- 358. Adrogué HJ, Madias NE. Diagnosis and treatment of hyponatre-
lyte ratio: a predictive guide to water restriction. Am J Med Sci. mia. Am J Kidney Dis. 2014;64(5):681–684.
2000;319(4):240–244. 359. Sterns RH, Silver SM. Complications and management of hypona-
336. Greenberg A, Verbalis JG. Vasopressin receptor antagonists. Kidney tremia. Curr Opin Nephrol Hypertens. 2016;25(2):114–119.
Int. 2006;69(12):2124–2130. 360. Cuesta M, Garrahy A, Thompson CJ. SIAD: practical recom-
337. Murphy T, Dhar R, Diringer M. Conivaptan bolus dosing for the mendations for diagnosis and management. J Endocrinol Invest.
correction of hyponatremia in the neurointensive care unit. Neuro- 2016;39(9):991–1001.
crit Care. 2009;11(1):14–19. 361. Sbardella E, Isidori AM, Arnaldi G, et al. Approach to hyponatre-
338. Palmer BF, Rock AD, Woodward EJ. Dose comparison of conivap- mia according to the clinical setting: Consensus statement from the
tan (Vaprisol®) in patients with euvolemic or hypervolemic hypo- Italian Society of Endocrinology (SIE), Italian Society of Nephrol-
natremia—efficacy, safety, and pharmacokinetics. Drug Des Devel ogy (SIN), and Italian Association of Medical Oncology (AIOM).
Ther. 2016;10:339–351. J Endocrinol Invest. 2018;41(1):3–19.

Downloaded for FK UMI Makassar (mahasiswafkumi01@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier
on November 01, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
References 330.e9

362. Hoorn EJ, Zietse R. Diagnosis and treatment of hyponatremia: 380. De Coopman J. Breastfeeding after pituitary resection: support
compilation of the guidelines. J Am Soc Nephrol. 2017;28(5): for a theory of autocrine control of milk supply? J Hum Lact.
1340–1349. 1993;9(1):35–40.
363. Mohmand HK, Issa D, Ahmad Z, et  al. Hypertonic saline for 381. Robinson A, Amico J. Remarks on the history of oxytocin. In:
hyponatremia: risk of inadvertent overcorrection. Clin J Am Soc Robinson A, Amico J, eds. Oxytocin: Clinical and Laboratory Stud-
Nephrol. 2007;2(6):1110–1117. ies/Proceedings of the Second International Conference on Oxytocin,
364. Sterns RH, Hix JK, Silver S. Treating profound hyponatre- Lac Beauport, Quebec, Canada. Amsterdam: Excerpta Medica;
mia: a strategy for controlled correction. Am J Kidney Dis. 1985:xvii–xxiv.
2010;56(4):774–779. 382. Smith R. Parturition. N Engl J Med. 2007;356(3):271–283.
365. MacMillan TE, Tang T, Cavalcanti RB. Desmopressin to prevent 383. Terzidou V. Preterm labour: biochemical and endocrinological
rapid sodium correction in severe hyponatremia: a systematic preparation for parturition. Best Pract Res Clin Obstet Gynaecol.
review. Am J Med. 2015;128(12). 1362.e15–24. 2007;21(5):729–756.
366. Ward FL, Tobe SW, Naimark DMJ. The role of desmopressin in 384. Vrachnis N, Malamas FM, Sifakis S, et al. The oxytocin-oxytocin
the management of severe, hypovolemic hyponatremia: a single- receptor system and its antagonists as tocolytic agents. Int J Endo-
center, comparative analysis. Can J Kidney Health Dis. 2018;5. crinol. 2011;2011:350546.
2054358118761051. 385. Evans JJ. Oxytocin in the human: regulation of derivations and
367. Malhotra I, Gopinath S, Janga KC, et al. Unpredictable nature of destinations. Eur J Endocrinol. 1997;137(6):559–571.
tolvaptan in treatment of hypervolemic hyponatremia: case review 386. Hertelendy F, Zakár T. Prostaglandins and the myometrium
on role of vaptans. Case Rep Endocrinol. 2014;2014:807054. and cervix. Prostaglandins Leukot Essent Fatty Acids. 2004;70(2):
368. Dunlap ME, Hauptman PJ, Amin AN, et al. Current management 207–222.
of hyponatremia in acute heart failure: a report from the Hypo- 387. Olson DM, Mijovic JE, Sadowsky DW. Control of human parturi-
natremia Registry for Patients With Euvolemic and Hypervolemic tion. Semin Perinatol. 1995;19(1):52–63.
Hyponatremia (HN Registry). J Am Heart Assoc. 2017;6(8). 388. Terzidou V, Blanks AM, Kim SH, et al. Labor and inflammation
369. Soupart A, Penninckx R, Crenier L, et al. Prevention of brain demy- increase the expression of oxytocin receptor in human amnion. Biol
elination in rats after excessive correction of chronic hyponatremia Reprod. 2011;84(3):546–552.
by serum sodium lowering. Kidney Int. 1994;45(1):193–200. 389. Lockwood CJ. The initiation of parturition at term. Obstet Gynecol
370. Goldszmidt MA, Iliescu EA. DDAVP to prevent rapid correction Clin North Am. 2004;31(4):935–947, xii.
in hyponatremia. Clin Nephrol. 2000;53(3):226–229. 390. Smith JG, Merrill DC. Oxytocin for induction of labor. Clin Obstet
371. Oya S, Tsutsumi K, Ueki K, Kirino T. Reinduction of hypo- Gynecol. 2006;49(3):594–608.
natremia to treat central pontine myelinolysis. Neurology. 391. Moghadam AD, Jaafarpour M, Khani A. Comparison effect of oral
2001;57(10):1931–1932. propranolol and oxytocin versus oxytocin only on induction of
372. Berl T, Quittnat-Pelletier F, Verbalis JG, et  al. Oral tolvaptan labour in nulliparous women (a double blind randomized trial). J
is safe and effective in chronic hyponatremia. J Am Soc Nephrol. Clin Diagn Res. 2013;7(11):2567–2569.
2010;21(4):705–712. 392. Mozurkewich EL, Chilimigras JL, Berman DR, et al. Methods of
373. Insel TR, Gingrich BS, Young LJ. Oxytocin: who needs it? Prog induction of labour: a systematic review. BMC Pregnancy Child-
Brain Res. 2001;133:59–66. birth. 2011;11:84.
374. Glasier A, McNeilly AS. Physiology of lactation. Baillieres Clin 393. Mendelson CR. Minireview: fetal-maternal hormonal signaling in
Endocrinol Metab. 1990;4(2):379–395. pregnancy and labor. Mol Endocrinol. 2009;23(7):947–954.
375. Uvnas-Moberg K, Eriksson M. Breastfeeding: physiological, endo- 394. Prata N, Bell S, Weidert K. Prevention of postpartum hemorrhage
crine and behavioural adaptations caused by oxytocin and local in low-resource settings: current perspectives. Int J Womens Health.
neurogenic activity in the nipple and mammary gland. Acta Paedi- 2013;5:737–752.
atr. 1996;85(5):525–530. 395. Chelmow D. Postpartum haemorrhage: prevention. BMJ Clin
376. Giraldi A, Enevoldsen AS, Wagner G. Oxytocin and the initiation Evid. 2011:1410. pii.
of parturition: a review. Dan Med Bull. 1990;37(4):377–383. 396. Grotegut CA, Paglia MJ, Johnson LN, et  al. Oxytocin exposure
377. Young 3rd WS, Shepard E, DeVries AC, et al. Targeted reduction during labor among women with postpartum hemorrhage second-
of oxytocin expression provides insights into its physiological roles. ary to uterine atony. Am J Obstet Gynecol. 2011;204(1):56 e51–56.
Adv Exp Med Biol. 1998;449:231–240. 397. Afolabi EO, Kuti O, Orji EO, Ogunniyi SO. Oral misoprostol ver-
378. Renfrew MJ, Lang S, Woolridge M. Oxytocin for promoting suc- sus intramuscular oxytocin in the active management of the third
cessful lactation. Cochrane Database Syst Rev. 2000;2:CD000156. stage of labour. Singapore Med J. 2010;51(3):207–211.
379. Johnston JM, Amico JA. A prospective longitudinal study of the 398. Firouzbakht M, Kiapour A, Omidvar S. Prevention of post-partum
release of oxytocin and prolactin in response to infant suckling in hemorrhage by rectal Misoprostol: a randomized clinical trial. J Nat
long term lactation. J Clin Endocrinol Metab. 1986;62(4):653–657. Sci Biol Med. 2013;4(1):134–137.

Downloaded for FK UMI Makassar (mahasiswafkumi01@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier
on November 01, 2021. For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.

You might also like