The Sensory Psychobiology of Thirst and Salt Appetite

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Psychobiology and Behavioral Strategies

The Sensory Psychobiology of Thirst


and Salt Appetite
ALAN KIM JOHNSON
Departments of Psychology, Integrative Physiology, and Pharmacology, and the Cardiovascular Center, University
of Iowa, Iowa City, IA

ABSTRACT
JOHNSON, A. K. The Sensory Psychobiology of Thirst and Salt Appetite. Med. Sci. Sports Exerc., Vol. 39, No. 8, pp. 1388–1400,
2007. Thirst and the hunger for sodium containing fluids and food (i.e., sodium appetite) are the consequences of the generation of
unique central nervous system states. Altered body fluid homeostasis produces sensory and perceptional changes that arise from signals
generated in the body that serve as indices of body fluid balance and distribution. These signaling mechanisms activate networks of
brain neurons that use specific neurochemicals to communicate between cells and process information. The brain integrates information
derived from various bodily sources so that thirst and sodium appetite are in a true sense the synthetic products of the nervous system.
In recent years much has been learned about the stimuli and receptor systems involved in signaling the brain to reflect the status of
bodily fluids and about the central neural substrates that process such inputs to generate thirst and sodium appetite. Knowledge about
the sensory nature of thirst and sodium appetite provides a basis for understanding the biological constraints under which thirst and
sodium appetite operate. This information is important for appreciating the extent to which thirst and sodium appetite motivational
states and behaviors can be relied on to maintain and repair disruptions of body fluid homeostasis. Key Words: ANGIOTENSIN II,
BARORECEPTORS, SENSORY CIRCUMVENTRICULAR ORGANS, EXERCISE-INDUCED HYPONATREMIA, HEAT INJURY

T
he perceptions of the thirst for water or a hunger for or the hunger for a particular substance, the brain receives
specific minerals or macronutrients that result from inputs from multiple sources that are related to the phy-
homeostatic deficits are the result of unique patterns siological status of the body. This information is conveyed
APPLIED SCIENCES

of neural activity in the central nervous system (CNS). A to the brain in the form of various chemical and neural
useful heuristic for understanding the biological under- signals over multiple modalities or input pathways and then
pinnings of states of thirst and various hungers is to con- is processed by the brain in an extensive neural network. In
sider them to be similar to the perceptions generated by effect, thirst and specific hungers are the synthetic products
sensory modalities such as vision, audition, olfaction, taste, of the CNS—they are created in the brain.
or the somatic senses (i.e., the classic senses). The neuro- This review will discuss the biological nature of the
biology underlying thirst and related appetites is similar to psychological states of thirst and salt appetite from the
that of many sensory systems. However, unlike primary perspective of the signals that generate them and the brain
sensory systems, thirst and specific hungers do not employ neuronal and chemical systems responsible for creating
unique sensory organs (eyes, ears, tongue, nose, and skin) them. By considering thirst and similar motivated states
or a single type of dedicated receptor (rods, cones, taste from the perspective of sensory and perceptual systems, it is
buds, hair cells) sensing the information necessary to gen- easier to appreciate that these behavioral controls of f luid
erate the central states associated with the respective per- homeostasis operate under unique sets of biological con-
ception. To produce the neural states associated with thirst straints. Understanding the sensory and perceptual neuro-
biology of thirst and specific hungers allows an appreciation
of the probable limits of these processes and the restrictions
they have in restoring homeostasis under conditions of en-
Address for correspondence: Alan Kim Johnson, Department of Psychol-
ogy, University of Iowa, 11 Seashore Hall E., Iowa City, IA 52242-1407;
vironmental and physical challenges.
E-mail: alan-johnson@uiowa.edu.
Submitted for publication December 2006.
Accepted for publication April 2007.
TOTAL BODY WATER AND BODY FLUID
COMPARTMENTS
0195-9131/07/3908-1388/0
MEDICINE & SCIENCE IN SPORTS & EXERCISEÒ Fluid homeostasis requires mechanisms that maintain
Copyright Ó 2007 by the American College of Sports Medicine overall sodium and water balance as well as the appropriate
DOI: 10.1249/mss.0b013e3180686de8 distributions of these substances within the intracellular

1388

Copyright @ 2007 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
(ICF) and extracellular fluid (ECF) compartments of the Body fluid balance is determined by interacting physio-
body. Consistency of the f luid matrix of organisms is logical and behavioral effector systems. Behavioral mech-
achieved by multiple control systems with physiological anisms participate in the intake of sodium and water, and
(i.e., autonomic nervous system; hormonal) and behavioral hormonal and autonomic mediators establish the rate of loss
effectors that are coordinated by the CNS. The level of ac- of these substances from the body. Both autonomic and en-
tivation of each of these mediators participates in the process docrine mechanisms target the kidney and the sweat and
of maintaining body f luid homeostasis and the patterned salivary glands to adjust the rates of sodium and water loss
output of each response system is adjusted by the brain. from the body. In the kidney, the antidiuretic hormone,
When separate sources of sodium and water are available, it vasopressin, is a well-def ined endocrine factor acting on
is possible to establish rates of sodium and water intake and renal collecting ducts to increase water permeability and
loss that are largely independent of one another. water reabsorption. Aldosterone, a mineralocorticoid, acts
Although the importance of water for maintaining f luid on renal tubules to promote sodium reabsorption. These
homeostasis seems obvious, the contribution of sodium (or actions of hormones are complemented by renal tubule
related extracellular ions and molecules) to overall fluid sympathetic innervation that, when activated, increases the
balance and distribution in the body may not be so apparent. reabsorption of sodium and water.
Sodium ions do not readily cross cell membranes and are Hormonal and autonomic mechanisms act on the kidneys
the key to determining the partitioning of water between the and on sweat and salivary glands to reduce the rate of
ICF and the ECF compartments. That is, the sodium con- sodium and water loss in the face of accruing fluid deficits.
centration of the ECF, in large part, determines the osmotic However, despite renal- and endocrine-targeted defenses, it
gradients and forces that move water in and out of cells. is only by engaging motivational mechanisms for the
Animals drink water and ingest impermeable ions and acquisition and consumption of water (thirst) and of sodium
molecules intermittently. Between the postabsorptive phase, (sodium or salt appetite) that restoration of sodium and
a time when the hydromineral milieu is theoretically closest water lost to the environment can be attained.
to balance and the conclusion of the next drinking episode,
physiological and behavioral control systems work together
SIGNALS REFLECTING DEHYDRATION OF BODY
to optimize the distribution of available fluid resources and
FLUID COMPARTMENTS GENERATE THIRST
to restore lost body water and sodium. When animals are
AND SODIUM APPETITE
challenged by physiological (e.g., exercise), environmental
(e.g., increased ambient temperature), or pathophysiological In classic studies in dogs conducted in the 1930s, Gilman
(e.g., emesis; diarrhea) conditions, afferent, neural, and (34) demonstrated that intravenous infusions of hypertonic
humoral signals directed to the CNS are generated and saline, but not hypertonic urea, induce thirst. These findings

APPLIED SCIENCES
serve as independent indices ref lecting the degree of f luid indicated that substances excluded from cells generate an
loss from the ICF and ECF compartments (Fig. 1). effective osmotic pressure and, thereby, produce drinking
for the simple reason that they dehydrate the ICF compart-
ment. The additional discovery that water intake was
evoked after experimental procedures in animals that
allowed investigators to selectively deplete either ICF or
ECF led to the formulation of the so-called double depletion
hypothesis of thirst in the early 1970s (23,31).
Hypovolemia, the depletion of ECF produced by exper-
imental procedures that generate a localized edema or
remove isotonic fluid from the body (e.g., hemorrhage or
intraperitoneal dialysis), is also an effective condition for
inducing thirst. For example, Fitzsimons (30) produced
drinking in rats by intraperitoneal injections of large
molecular weight substances (hyperoncotic colloids, such
as polyethylene glycol and gum acacia) that reverse the
normal Starling forces present at the capillaries. Increased
oncotic pressure in the extracellular space at the injection
site of such colloids progressively sequesters isotonic ECF
to dehydrate interstitial f luid and reduce blood volume.
With hemorrhage, intraperitoneal dialysis or hyperoncotic
colloid–induced dehydration, the osmotic equilibrium
across cell membranes is not affected. Therefore there is
no movement of water out of the cellular compartment and
FIGURE 1—Gain and loss of water and sodium. hence no reduction of ICF.

BEHAVIORAL MECHANISMS IN BODY FLUID BALANCE Medicine & Science in Sports & Exercised 1389

Copyright @ 2007 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
Although thirst can be induced by selective dehydration ning 6–9 h after polyethylene glycol–induced hypovolemia.
of either ECF or ICF (i.e., the double depletion hypothesis), However, if the hypovolemic animals are given access to
under the majority of physiological conditions thirst arises isotonic saline rather than water, they continue to ingest the
as a result of a simultaneous depletion of the two fluid salty fluid at high rates (69). This indicates that sodium, in
compartments. Conjoint dehydration of the ICF and ECF by addition to water, is required to correct hypovolemia.
coadministered hypertonic saline and polyethylene glycol In fact, Stricker_s experiments (68,69) model what had
produces a total water intake that equals the sum of what been described in man by Adolph (1) as voluntary de-
either dipsogenic treatment would produce when adminis- hydration. Later, the term involuntary dehydration was
tered independently (13). suggested to be more appropriate to describe this phenom-
Under physiological or pathophysiological conditions, enon (35). Dehydrated humans with access only to water
various hydration challenges generate different degrees of stop drinking before the fluid deficit is repaired. Hypovo-
dehydration of the ICF and ECF compartments. For exam- lemic rats will not only restore fluid balance by ingesting
ple, transpiration of water through the skin or loss by evapo- isotonic saline but will consume nonpreferred, unpalatable
ration from the respiratory system or oral mucosa results concentrations of hypertonic saline if they are offered in
in a depletion of only water and, therefore, a pure dehydra- conjunction with water (68,70,71) (Fig. 2). A significant
tion of the ICF compartment; loss of hypotonic fluid as increase in the intake of a concentrated, normally rejected or
sweat or saliva produces loss of both ECF and ICF; fluid unpreferred solution of NaCl is a commonly employed
depletion as a result of bleeding (e.g., hemorrhage due to operational definition of sodium appetite (i.e., salt appetite).
tissue damage; menstruation) produces a selective ECF loss The experimental study of sodium appetite dates from Curt
(i.e., hypovolemia). Richter_s (58) pioneering demonstration that adrenalectomy
Disproportionate dehydration in one body compartment in the rat results in the ingestion of significant volumes of
can become translated into a loss from the other compart- sodium solution. Adrenalectomy removes the body_s pri-
ment. For example, an initial loss of hypotonic f luid results mary source of aldosterone and therefore causes a loss of
in a relative hypernatremia that, in turn, dehydrates the in- sodium in the urine, saliva, and feces. This absence of
tracellular compartment. Ultimately, further changes to ap- sodium-retaining hormone causes compensatory sodium
portion the distribution of dehydration to each of the body ingestion to replace the lost sodium. Numerous other
fluid compartments can occur as a result of renal mecha-
nisms, because the kidney has the capacity to excrete either
hypotonic or hypertonic urine.
Total water loss can be differentially shared by the ICF or
ECF compartments over time and under different physiolog-
APPLIED SCIENCES

ical tasks or environmental conditions. Classic demonstra-


tions of thirst being maintained after the administration of
hypertonic saline while water was withheld were made by
Adolph and colleagues (2) and Holmes and Gregersen
(36,37). After delays of 3–8 h, rats and dogs drank amounts
of water that were similar to those under experimental
conditions where they were given immediate access to water.
During the time when water was not accessible, sufficient
amounts of solute were excreted so that plasma osmolarity
was nearly normalized. In other words, by excreting Na+,
the animals exchanged the prevailing stimulus for thirst, an
ICF depletion, for an ECF dehydration.
Not only are the processes underlying water intake
induced by depletion of ICF versus ECF compartments dif-
ferent, so are the mechanisms of satiety. Fitzsimons (29)
demonstrated that nephrectomized rats infused with effec-
tive osmotic agents drink sufficient water to restore body
fluids to isotonicity. Anephric rats that cannot use renal
mechanisms to excrete Na+ behave as near-perfect osmom-
eters. In contrast, animals with pure hypovolemia do not
FIGURE 2—Mean hourly cumulative intakes, in total licks, showing
restore fluid balance by ingesting only water. Studying rats the pattern of water and 0.5 M NaCl solution intake by rats during the
made hypovolemic by subcutaneous administration of 24-h period after subcutaneous injection of 30% polyethylene glycol
polyethylene glycol, Stricker (68,69) demonstrated that rats in isotonic saline to induce hypovolemia (N = 5). (Reprinted from
Stricker, E. M., K. S. Gannon, and J. C. Smith. Thirst and salt appetite
do not maintain high rates of water intake in the face of induced by hypovolemia in rats: analysis of drinking behavior. Physiol.
persisting hypovolemia. That is, water intake slows begin- Behav. 51:27–37, 1992. Used with permission.)

1390 Official Journal of the American College of Sports Medicine http://www.acsm-msse.org

Copyright @ 2007 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
TABLE 1. Experimental manipulations commonly employed to induce salt appetite. state of) sodium appetite as indexed by a significant
Primary salt appetite increase in hypertonic saline solution intake. Although in
Absolute sodium deficit induced by depletion
Dietary sodium restriction principle both thirst and sodium appetite can exist simulta-
Perspiration (exercise in the heat with restoration of water loss neously, under common experimental conditions only one
without sodium)
Adrenalectomy
behavior can be expressed and measured at a time. The
Peritoneal dialysis investigation of the patterns of water and NaCl intake after
Natriuretic treatment
dehydration proves instructive and has increased the under-
Salivary sodium loss
Relative sodium deficit standing of the neural and endocrine mechanisms that
Sequestration of extracellular fluid (e.g., polyethylene glycol) control the generation of the hydromineral-related ingestive
Pharmacological simulation
Combined natriuretic–hypotensive drug (e.g., furosemide + captopril; behaviors involved in maintaining body fluid homeostasis.
furosemide + minoxidil) Stricker and colleagues (70,71) characterized the time
Aldosterone or deoxycorticosteroid acetate (DOCA)
Intracerebroventricular angiotensin or renin treatment
course of the onset of thirst (water intake) and sodium
Intracerebroventricular angiotensin + priming with low-dose aldosterone appetite (NaCl intake) after the induction of hypovolemia
>2-adrenoceptor antagonist treatment (yohimbine) induced by subcutaneous polyethylene glycol treatment.
The general result from such studies indicates that although
experimental manipulations have been shown to enhance significant increases in water intake are apparent within an
the ingestion of concentrated solutions of NaCl in animals. hour or two after polyethylene glycol treatment, it usually
These treatments range from pharmacological doses of takes on the order of 5–7 h before significant increases in
aldosterone to the induction of sodium depletion by any of the consumption of concentrated (i.e., 2 or 5%) NaCl
several methods including dietary restriction (Table 1). It solutions are observed (Fig. 2). The reasons for this relative
is interesting to note that the majority of the experimental delay in onset are probably because of the need for an
manipulations used to induce sodium appetite require sub- increase in one or more facilitory factors or a decrease in
stantial periods of time to elapse, often on the order of inhibitory mechanisms. Such processes may be related to
several hours to days, before a significant increase in con- the degree or duration of hypovolemia, the consequences of
centrated NaCl becomes apparent. This is in contrast to first drinking water (i.e., to produce a decrease in
the shorter latencies for the onset of thirst (i.e., signifi- osmolarity of ECF), increased hormonal levels (e.g., ANG
cant water intake) after experimental depletion of the ICF II; aldosterone), the development of hypotension, or some
or ECF compartments. combination of these factors (44,45,72).
The first time animals are made sodium def icient, they will After NaCl intake commences, animals alternate between
consume large quantities of NaCl on being given access to drinking water and ingesting concentrated sodium solution
salty fluids even though they have had no previous ex- (Fig. 2). By adjusting the volumes of water and hypertonic

APPLIED SCIENCES
perience with such solutions (25). Display of this appetite saline consumed over time, rats end up consuming the
for sodium by naBve animals provides evidence that the equivalent of a near-isotonic (0.9–1.2%) mixture (70).
appetitive and consummatory behaviors associated with Thus, two distant behaviors driven by two different moti-
sodium appetite are innate. However, this native behavior is vational states provide the optimal strategy for the hypo-
also modifiable as a result of prior experience. A single volemic animal to correct an ECF def icit. Sodium appetite
sodium depletion, regardless of whether the animal does or as demonstrated by a significant increase in hypertonic
does not orally consume sodium, produces an enhanced NaCl solution intake has been demonstrated in many avian
NaCl intake response on subsequent tests in response to and mammalian species. This paradigm has not been
sodium depletion (26,62). The phenomenon of enhanced applied to study the recovery from dehydration in humans.
NaCl intake after an initial treatment can be mimicked by The classical studies of human sodium deficiency were
administration of exogenous angiotensin II (ANG II) and conducted by McCance (50), who used himself and three
aldosterone (62). volunteers as subjects. Severe sodium deficits were induced
Many of the treatments that induce sodium appetite also by low-sodium diet and episodes of sweating. During the
induce water intake; however, the converse does not hold. sodium-depleted state, which was maintained for 11 d, the
For example, depletion of ICF with hypertonic saline induces subjects reported excessive fatigue, lethargy, and a general
water intake but little, if any, intake of hypertonic NaCl (57). feeling of exhaustion. The sense of taste was generally
Such observations led Peck (57) to suggest that ICF affected. Food and cigarettes were described as tasteless.
depletion–induced water drinking and ECF depletion– The subjects differed in their desire for sodium. One subject
induced water drinking and sodium ingestion may be the described a distinct longing for salt and often went to sleep
reflection of different types of thirst. In experimental thinking about it, but McCance reported having no specific
animals, it is impossible to test whether a thirst induced craving for sodium.
by ICF depletion is qualitatively different than a thirst More recently, better-controlled experiments employing
induced by ECF depletion. It is probably best to use more objective methods have been used to assess sodium
objective operational definitions of (the state of) thirst as appetite in humans after sodium depletion. Beauchamp and
measured by a significant increase in water intake and (the colleagues (6) studied the effects of a very-low-sodium diet

BEHAVIORAL MECHANISMS IN BODY FLUID BALANCE Medicine & Science in Sports & Exercised 1391

Copyright @ 2007 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
and diuretic treatment on taste in normal subjects for a 10 d bouts during 7 h), which was followed by a rehydration
period. As assessed after 5 and 10 d of depletion, thresholds period providing only ad libitum water. After a delay of
for the taste of salt decreased in the majority of subjects, and several hours, the subjects began to express a significant
the preference judgments for salt in foods tended to be increase in palatability ratings for hypertonic saline sol-
greater. It is interesting to note that during the depletion utions. The increased acceptance over the hypertensive
period, despite increased plasma aldosterone and renin range of solutions became most apparent 17–23 h after the
activity, supine and upright blood pressure were slightly start of rehydration (Fig. 3). It is unclear whether factors
decreased. Together, the data are interpreted as indicating such as reduced plasma osmolarity or the presence of
that sodium depletion in humans produces moderate sensory hypotension were responsible for increasing perceived
(salt taste) changes and increased preference for salty foods. sodium palatability during the course of rehydration.
The time course and development of sodium appetite in
humans rehydrating without sodium replacement has been
VISCERAL AND BRAIN SENSORY RECEPTORS
studied by Takamata and colleagues (74). These investiga-
DETECT THE STATUS OF BODY FLUID BALANCE
tors induced H2O and Na+ depletion by light exercise (eight
Both chemo- and mechanoreceptors sense the status of
body f luids. These receptors are located in the systemic viscera
and in the brain. Increasing the osmolarity of brain ECF by
injection of hypertonic cerebrospinal fluid into the cerebral
ventricles activates vasopressin release, sympathetic outf low,
and thirst. These physiological and behavioral responses are
biologically consistent with one another and cooperatively
produce an increase in total body water and a tendency to
elevate blood pressure. Such responses have been proposed
to be mediated by neuron-like cells, which sense their own
volume and are referred to as osmoreceptors (80,81).
Although there are many osmosensitive regions in the
CNS and body, including the hepatoportal region, one of
the key areas housing osmoreceptors for thirst and vaso-
pressin release lies within the periventricular tissue sur-
rounding the anteroventral portion of the third cerebral
ventricle (AV3V) (40,44,45). AV3V-associated structures
APPLIED SCIENCES

that play particularly important roles in osmoreception and/


or processing are the ventral median preoptic nucleus and
the organum vasculosum of the lamina terminals (OVLT).
The OVLT along with two other anatomically and func-
tionally unique structures, the subfornical organ (SFO) and
the area postrema (AP), are referred to as sensory circum-
ventricular organs (43). Sensory circumventricular organs
like the other similar structures are devoid of a blood–brain
barrier, but whereas most of the other circumventricular
organs function as sites of secretion, sensory circumven-
tricular organs have been implicated as structures sensing
humoral signals in blood or interstitial fluid. Cells with
stretch-sensitive ion channels have been identified electro-
physiologically within the OVLT (7) and are, therefore,
primary candidates as osmoreceptors.
A second key humoral signal that acts on the brain to
effect responses consistent with restoration or expansion of
body fluids and elevation of blood pressure is circulating
FIGURE 3—Subjective palatability rating as a function of tasted NaCl ANG II. Systemic administration of ANG II elevates
solution molarity in subjects initially dehydrated and then rehydrated arterial blood pressure, releases vasopressin, and increases
(beginning at 0-h rehydration) with only water. Concentrations of
tasted solutions are expressed on a logarithmic scale. Data are means T the ingestion of water and sodium (44,45). These ingestive,
SE (n = 7). * Significant difference from control, P G 0.05. Reprinted pressor, and secretory responses are also produced by ANG
from Takamata, A., G. W. Mack, C. M. Gillen, and E. R. Nadel. II administered directly into the brain ventricles.
Sodium appetite, thirst, and body fluid regulation in humans during
rehydration without sodium replacement. Am. J. Physiol. Regul. Integr. There are several regions in the CNS sensitive to ANG II.
Comp. Physiol. 266:R1493–R1502, 1994. Used with permission. The SFO in particular has been implicated in the activation

1392 Official Journal of the American College of Sports Medicine http://www.acsm-msse.org

Copyright @ 2007 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
of thirst, salt appetite and vasopressin release by circulating requires the integration of information derived from several
ANG II (44,45). It has been hypothesized that the brain types of sensory systems. Visceral afferent signals must
renin–angiotensin system (i.e., a system with its compo- communicate the body’s need for water and sodium to the
nents synthesized de novo in the brain) is coupled with the brain. In the process of seeking water and sodium, sensory
systemic (i.e., renal) renin–angiotensin system through the systems are engaged for identif ication of potential sources
SFO (38). Circulating angiotensin in the mode of a in the external environment. When in contact with food and
hormone acts on sensory circumventricular organs to fluids, gustatory mechanisms assess the taste qualities of the
activate brain neural pathways. Brain interneurons, in turn, ingesta to identify the commodity as an acceptable source
release angiotensin from their axon terminals, which then of water or sodium for consumption.
functions like a traditional neurotransmitter (38,39,43). The VIIth, IXth, and Xth cranial nerves carrying taste and
Considerable attention has been paid to the contribution visceral information enter the brain stem and synapse
of brain steroid receptors in the generation of sodium primarily in the nucleus of the tractus solitarius (NTS).
appetite (24). Mineralocorticoids (e.g., aldosterone), when Axons from cells in the NTS, in turn, project to other brain
given in pharmacological doses (82) or in more physio- nuclei. Similarly, input detected by sensory circumventric-
logical concentrations in conjunction with ANG II (32), ular organs (SFO, OVLT, AP) is also carried into the brain
produce significant NaCl ingestion. The facilitory interac- by nerves emanating from these structures. Many of the
tion that can be achieved with systemic injections of very brain pathways and regions carrying and processing input
low doses of aldosterone and small amounts of ANG II from the systemic chemo- and mechanoreceptors and from
injected into the brain has been referred to as the synergy sensory circumventricular organs have been defined and
hypothesis for sodium appetite (24). characterized by anatomical, neurochemical, and functional
Mechanoreceptors identif ied as key for sensing blood techniques. The result has been the identif ication of a neu-
volume and arterial blood pressure are located in the ral network that both carries and integrates information
periphery. The great veins and atria (i.e., the low-pressure critical for the regulation of fluid balance. Pathways and
side of the circulation) contain what are often referred to information initially processed by the AP and NTS ascend
as cardiopulmonary receptors (a misleading terminology, the neuraxis to meet information deriving from the SFO and
because there are many different types of visceral sensory OVLT. Between the forebrain and hindbrain portals of
receptors associated with the heart and lungs) that are input, there are several nodes (brain nuclei) where informa-
responsive to stretch. These sensors generate afferent nerve tion from such various sources converges.
traffic to the CNS in proportion to distention of the walls of In principle, activity in any pathway and nucleus in the
the great veins (e.g., superior vena cava) and atria. Similar brain can influence information handling (processing and
stretch receptors located in the aortic arch and carotid sinus storage) within the neural network that controls the

APPLIED SCIENCES
sense changes in arterial blood pressure. Output from these effectors maintaining body fluid and cardiovascular homeo-
receptors change in proportion to arterial blood pressure. stasis. However, there are brain areas that have been found
Information from low- and high-pressure baroreceptors as to be especially critical in these processes. Most notable
well as from cardiac ventricular mechano-/chemoreceptors among these brain regions are 1) the sensory circum-
reaches the brain by the IXth and Xth cranial nerves. When ventricular organs (i.e., SFO, OVLT, and AP), 2) the NTS,
blood pressure and/or blood volume falls below a ‘‘normal’’ 3) caudal and rostral ventrolateral medulla, 4) parabrachial
set point, nerve activity declines and results in triggering the nucleus (PBN), 5) the median preoptic nucleus, 6)
reflex release of vasopressin, increased sympathetic outflow, parvocellular hypothalamic paraventricular nucleus, 7)
and, ultimately, thirst and sodium appetite. A moderate magnocellular paraventricular nucleus, 8) supraoptic
reduction in blood pressure (induced by antihypertensive nucleus, 9) amygdala (particularly the central and medial
agents) in the presence of hypovolemia synergizes to rapidly nuclei of the amygdala), 10) bed nucleus of the stria
generate sodium appetite and thirst, which are dependent on terminalis, 11) lateral hypothalamus, 12) ventrolateral
a renin–angiotensin system that is intrinsic to the brain (76). medulla, and 13) the spinal cord intermediolateral cell
Maintaining blood pressure or cutting neural afferents from column (42,44,45). Reciprocal pathways connect most of
high-pressure baroreceptors (sinoaortic denervation) blocks these structures with one another and provide feedforward
this type of experimentally induced sodium appetite (77). and feedback neural circuits that are necessary for achieving
Blood pressure at hypertensive or normotensive levels is the processing of information within the CNS. Information
likely to inhibit thirst and sodium appetite (44,45,59). handling and processing within this hydromineral–neural
network is necessary for both acute and chronic adjustments
involved in regulating body f luid balance and distribution
A BRAIN NEURAL NETWORK PROCESSES FLUID
(e.g., hemodynamics). There are many neurotransmitter
BALANCE-RELATED INPUT TO MAINTAIN
systems associated with this body f luid–related brain
BODY FLUID HOMEOSTASIS
network. Two of the most important of the central neuro-
The generation of the appetitive and consummatory chemical mediators are norepinephrine and angiotensin.
behaviors necessary to acquire and ingest water and sodium Pathways containing these putative neurotransmitters are

BEHAVIORAL MECHANISMS IN BODY FLUID BALANCE Medicine & Science in Sports & Exercised 1393

Copyright @ 2007 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
distributed throughout the CNS and are associated with most, Both the SFO and OVLT project to the median preoptic
if not all, of the key structures involved in f luid balance and nucleus that lies between them and is located inside the blood–
cardiovascular control. Evidence indicates that these two brain barrier. The median preoptic nucleus is an important
brain neurochemical systems are important for mobilizing integrative node, processing both angiotensin-derived and
effector systems that expand ECF volume (39,44,45). osmotic information. Neural activity in the median preoptic
In addition to facilitory mechanisms, there are inhibitory nucleus partly determines the degree and pattern of activation
counterparts represented in the central f luid regulatory of the physiological and behavioral control systems for body
network that retard overexpansion of the ECF compartment. fluid homeostasis (41). Numerous studies have implicated the
Evidence indicates that such inhibition may involve brain SFO (66,75), the ventral median preoptic nucleus (27,28),
oxytocin (72), serotonin (44,45,51–53), tachykinins (49), and the combined ventral median preoptic nucleus and
cholecystokinin (54), and bombesin (15,16). OVLT region in the control of thirst and sodium appetite
In recent years, signif icant progress has been made in (5,8,19,40). Several pathways descend from lamina termi-
several laboratories in identifying forebrain and hindbrain nalis structures to innervate key forebrain nuclei that have
components of the hydromineral neural network subserving been implicated in thirst (e.g., lateral hypothalamic/periforn-
thirst and sodium appetite as well as those for physio- ical area), sodium appetite (central nucleus of the amygdala),
logical control systems (i.e., sympathetic and endocrine). anterior pituitary hormone control (parvocellular paraven-
Systemically derived information has immediate access to tricular nucleus; PVN), posterior pituitary function (magno-
the forebrain via actions on rostral sensory circumventric- cellular PVN and supraoptic nuclei), and sympathetic
ular organs associated with the lamina terminalis (Fig. 4). outflow (parvocellular PVN) (Fig. 4).
The lamina terminalis is comprised of a layer of ependymal In contrast to the forebrain facilitory actions of ANG II,
cells that during early development covers the rostral end of hyperosmolarity, and mineralocorticoid binding, other sig-
the neural tube. In mature animals, this sheet of cells forms nals act through a hindbrain system to keep water and
the rostral wall of the third ventricle, and four midline sodium intake in check. This inhibitory system involves the
structures, the SFO, the median preoptic nucleus, the neural circuitry of the AP, the medial portion of the NTS
anterior commissure and the OVLT, lie adjacent to the (AP/mNTS), and the lateral PBN (44,45).
lamina terminalis. The SFO is situated in the dorsal aspect Ablation of the AP/mNTS dramatically increases the
of the third ventricle and the OVLT is located along the intake of concentrated sodium chloride solution both in
ventral portion of the lamina terminalis, both of these sense chronic (11) and acute (21) experimental tests. An
plasma ANG II levels and extracellular osmolarity. important component of a neural pathway projecting from
APPLIED SCIENCES

FIGURE 4—Organization of structures of the lamina terminalis region (midsagittal representation) and projections from the lamina terminalis to
key forebrain area involved in the control of the endocrine, autonomic, and behavioral systems that participate in body f luid regulation. The
subfornical organ, organum vasculosum of the lamina terminalis (OVLT), the median preoptic nucleus, and anterior commissure are adjacent to the
lamina terminalis and can be referred to as the structures of the lamina terminalis. CRF, corticotropin-releasing factor; ACTH, adrenocorticotropic
hormone; PVN, paraventricular nucleus.

1394 Official Journal of the American College of Sports Medicine http://www.acsm-msse.org

Copyright @ 2007 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
the AP/mNTS to the lateral PBN (14,64,79) contains recently been demonstrated. Inflating a small balloon at the
serotonin (48). Bilateral injections of serotonin antagonists junction of the vena cava and right atrium in rats inhibits
directly into the lateral PBN markedly enhance both thirst thirst (46) and sodium appetite (78). The expansion of such
and sodium appetite induced by many dipsogenic and a balloon in rats with lesions of the lateral PBN is no longer
natriorexigenic challenges (44,45,53). Several recent stud- effective in inhibiting isoproterenol-induced drinking (56).
ies have also implicated the actions of cholecystokinin, This observation is consistent with other findings (22,54)
GABA, corticotropin-releasing hormone, and norepineph- indicating that the lateral PBN is a key brain region
rine in the lateral PBN on sodium and water intake controlling behaviors that inf luence ECF volume. Informa-
(3,9,17,18,54). In many respects the LPBN seems to be a tion processed within the lateral PBN is carried deeper into
switch to determine whether water intake versus sodium the neural network for hydromineral balance where it
intake becomes the predominant behavior used to correct interacts with other types of input, particularly those from
body f luid deficits. sites receiving humorally derived signals from the lamina
The lateral PBN (Fig. 5) is an integrative region involved terminalis (Fig. 5).
in processing viscerally derived inhibitory input. An Although much of the neural circuitry of thirst and sodium
example of the importance of the lateral PBN in the appetite is contained within the brain stem and limbic
inhibition of behaviors related to volume expansion has system, we probably become aware of our thirst and hunger
for salty substances because of activation of parts of the
cerebral cortex. This is a reasonable expectation (hypothesis)
because the highest levels of information processing for
perception in other sensory systems occur at various cortical
loci. In recent studies using positron emission tomography,
Derek Denton and his colleagues (20) have identified in
human volunteers several brain regions that are activated in
conjunction with a strong sensation of thirst produced by
intravenous infusions of hypertonic saline. Among the brain
regions showing changes in activity associated with the
desire to drink were the anterior and posterior portions of
the cingulate cortex. This is a brain region associated with
motivational and affective processes.

APPLIED SCIENCES
EXERCISE, CONDITIONING, AND BIOLOGICAL
CONSTRAINTS ON THIRST AND
SODIUM APPETITE
There is a substantial body of experimental evidence
indicating that maintaining adequate hydration is critical for
health and well-being (4,63). The excessive loss of water
and sodium in extreme environments or while engaging in
athletics not only impairs performance but also increases
the likelihood of thermal injury (73). Significant water and
sodium losses can occur very quickly in both fit and
untrained individuals, especially if exercising in a hot
environment. The speed of onset of severe dehydration
under extreme conditions can exceed the onset of thirst and
sodium appetite. Thirst drive resulting from dehydration
FIGURE 5—A schematic depicting a hindbrain system involved in the
inhibitory control of thirst and sodium appetite. The key hindbrain may occur on the order of minutes to tens of minutes, but
structures implicated are the area postrema (AP), nucleus of the the onset of sodium appetite is likely to require many hours
solitary tract (NTS), and the lateral parabrachial nucleus (LPBN). To before a significant hunger develops (70,74). Impaired
simplify the diagram, pathways are only presented unilaterally.
Lesions of the AP and LPBN produce overdrinking to thirst-inducing thirst and sodium appetite mechanisms that accompany the
stimuli that simulate hypovolemia. In the presence of thirst- or salt normal aging process (47) are likely to only complicate this
appetite-inducing stimuli, bilateral injections of the nonselective problem. Although behavioral mechanisms of drinking and
serotonergic receptor antagonist, methysergide, into the LPBN causes
profound overdrinking and sodium intake. The LPBN, in turn, salt ingestion can act in concert with the kidney to restore
projects to forebrain structures such as the central nucleus of the body f luid homeostasis over the long-term, the mobilization
amygdala (CeA), bed nucleus of the stria terminalis (BNST), and of thirst- and especially sodium appetite-related behaviors
median preoptic nucleus (not shown). Therefore, the AP–LPBN has
been proposed to be a hindbrain circuit that guards against hyper- do not have sufficiently rapid response times to prevent
volemia or expanded blood volume. VII, IX, X, input of cranial nerves. immediate consequences of severe water and sodium losses.

BEHAVIORAL MECHANISMS IN BODY FLUID BALANCE Medicine & Science in Sports & Exercised 1395

Copyright @ 2007 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
Beyond concerns of the extended lag times for the onset exercise-induced hyponatremia, a dilutional hyponatremia
of thirst and sodium appetite that constrain the efficiency in associated with positive f luid balance. This disorder is
converting exercise-induced fluid deficits into corrective produced in part by the excess secretion of antidiuretic
behaviors, there are exercise-induced fluid shifts and hormone (i.e., arginine vasopressin (65)) and can be life
hemodynamic changes that are likely to also compromise threatening. Excessive ingestion of fluid prompted only by
the reliability of rehydration-related behaviors for def icit a cognitively based formula clearly carries a risk of
correction. The nature of circulatory changes and of fluid untoward consequences produced by hyponatremia.
movement among lymphatic, intravascular and interstitial By viewing thirst and sodium appetite from the perspec-
spaces, as well as the movement between ICF and ECF tive of sensory psychobiology, it becomes easier to appre-
compartments (60) that accompany exercise, needs to be ciate that there are biological limits on the functional
considered. Unfortunately, little is known about how capacities of motivational systems. That is, just as there are
changes in fluid distribution produced either as a result of limitations in the classic senses in terms of thresholds for
chronic exercise and physical conditioning (e.g., expansion detection and discrimination, there are restrictions as to how
of blood volume (12)) or by acute exercise will alter af- fast and how reliably the receptors and neural systems
ferent signaling mechanisms. At present, one can only subserving thirst and salt appetite can sense and respond to
speculate about the effects of exercise and conditioning on changes in hydrational status. Just as we employ micro-
thirst and sodium appetite based on hydromineral-related scopes, telescopes, pressure transducers, audio amplif iers,
hormonal or autonomic responses under such conditions. and other instrumentation to enhance and resolve physical
For example, Nose and colleagues (55) have demonstrated stimuli, which normally serve to excite the classic senses, it
that during graded exercise the threshold for the release of is not unreasonable to expect that it may be necessary to
vasopressin is increased. Therefore, one might also suspect employ instrumentation (scales, osmometers, sodium ana-
that increased inhibition of water or sodium intake would lyzers) to assess hydrational status under extreme and
accompany exercise. Perhaps increases in arterial pressure rapidly changing conditions.
and venous return associated with exercise would contribute
to such inhibition, but this has not been studied in relation
SUMMARY AND CONCLUSIONS
to thirst or sodium appetite.
The degree of loading of cardiopulmonary baroreceptors Maintaining body fluid and cardiovascular homeostasis
have been demonstrated to affect both thirst and vasopressin requires the integrative capacity of the CNS. A thirst for
secretion. Head-out water immersion produces a shift of water or a hunger for sodium are perceptions that result
blood volume toward the thorax stretching the large from synthetic processes carried out in the brain. The CNS
capacitance vessels and the atria produces a reduction of receives visceral and somatic sensory inputs and integrates
APPLIED SCIENCES

thirst, water intake, and vasopressin secretion in dehydrated this information. Through this process, calculated decisions
subjects (61,67). Interestingly, this inhibitory effect on thirst are made by the brain and the best pattern of reflexes and
and vasopressin release does not occur in older subjects behaviors are generated to optimize available fluid distri-
(67). It has been suggested by Stachenfeld and colleagues bution between body compartments and to restore hydro-
(67) that the attenuated inhibitory mechanisms may be due mineral balance. The key behaviors that are necessary for
to impaired cardiopulmonary reflexes (10). From this per- the maintenance and recovery of normal fluid balance are
spective, one might consider the possibility that thirst in those that lead to the acquisition and consumption of both
athletes with cardiac hypertrophy and diminished cardio- water (thirst) and sodium (sodium appetite).
vascular reflex function (33) may have impaired fluid- Figure 6 summarizes several of the major points
related behavior and endocrine responses to altered stretch identifying the afferent stimuli and central circuitry relevant
of vessels containing cardiopulmonary receptors. to the behavioral controls of ECF volume. To the left of the
Despite the limitations imposed by the sensory nature of box, one of the primary sites for osmoreception related to
thirst and sodium appetite, it is important to recognize that thirst and vasopressin release is identified as a component
the cognitive capacities of humans provide a means of of the lamina terminalis. To the right and above the large
compensating for limitations inherent in intrinsic biological box representing the brain are the neural and humoral inputs
motivational processes. That is, cognitive mechanisms can that communicate the status of body fluid and cardiovas-
activate behaviors to drink or consume salt and override an cular homeostasis to the CNS. Under a range of physio-
absence of thirst and sodium appetite. However, f luid intake logical conditions, circulating ANG II is a participant in the
resulting from only cognitive processes without the activa- generation of thirst and sodium appetite and its actions are
tion of thirst- or sodium appetite–related mechanisms may complemented by additional visceral afferent inputs. ANG
lead to inappropriate water and salt intake. In this context it is II accesses the CNS through brain circumventricular organs,
important to note that not only is hypohydration caused by particularly the SFO.
inadequate thirst and sodium appetite a concern, but under Systemic input carried over nerves from the viscera
conditions of extreme athletic challenges there is also the risk originates in vascular baroreceptors. Hypovolemia and/or
of cognitively overdriven f luid intake. The result can be hypotension result in input to the CNS that enhances

1396 Official Journal of the American College of Sports Medicine http://www.acsm-msse.org

Copyright @ 2007 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
FIGURE 6—Diagram depicting neural and hormonal inputs into the brain and the central neural pathways that mediate the sensory integration of
signals for generating drinking (thirst) and sodium ingestion (salt appetite). Both inhibitory and excitatory inputs from the periphery are derived from
arterial and cardiopulmonary baroreceptors as well as other visceral receptors (e.g., gastric, hepatic/portal, renal). Information carried in afferent
nerves projects mainly to the nucleus of the tractus solitarius (NTS). Angiotensin (ANG) acts in the form of ANG II on angiotensin type 1 receptors in the
subfornical organ (SFO). Osmoreception takes place in structures along the ventral lamina terminalis (OVLT; median preoptic nucleus (MePO)).
Hormonal information to the SFO is subsequently carried in descending pathways, some of which are likely to use ANG in the mode of a
neurotransmitter, to forebrain structures such as those in the tissue surrounding the anteroventral third ventricle. Ascending information to the
forebrain is carried in projections from noradrenergic cell groups in the hindbrain which are activated by arterial and cardiopulmonary receptor input
under conditions of hypotension and/or hypovolemia (not shown). ANG and noradrenergic inputs act synergistically in forebrain nuclei. A hindbrain
inhibitory pathway originating in the area postrema (AP) and medial NTS ascends to the lateral parabrachial nucleus. This projection uses serotonin
(5-HT) as a neurotransmitter and prevents excessive sodium and water intake thereby limiting excess expansion of blood volume. Inhibitory input is
likely to ascend the neuraxis either directly or indirectly to interact with forebrain structures. FAC, facilitation; INH, inhibition; AMYG, amygdala;
BNST, bed nucleus of the stria terminalis; LPBN, lateral parabrachial nucleus; SNS, sympathetic nervous system; JG, juxtaglomerular.

APPLIED SCIENCES
activity in central facilitory systems. Conversely, hyper- lateral PBN have been associated with a hindbrain
volemia and/or hypertension activate inhibitory mecha- projection from the AP/mNTS to the lateral PBN that has
nisms. Other types of facilitory and inhibitory input from been proposed to protect from overexpansion of the ECF
the viscerally derived gastrointestinal tract, splanchnic and compartment.
hepatoportal circulation and the kidneys also are likely to The integration of information derived from facilitory
feed into the neural network for fluid balance. and inhibitory humoral and visceral neural inputs is
In the brain, angiotensin acting within the SFO has been conducted within many different nodes (nuclei) in a sensory
proposed to activate several descending projections to key network that regulates hydromineral balance. The outputs of
forebrain structures such as the median preoptic nucleus, this network both controls motor-pattern generators respon-
hypothalamic perifornical area, central nucleus of the sible for the appetitive and consummatory behaviors and
amygdala, supraoptic nucleus, and paraventricular nucleus. give rise to a sensory experience perceived as thirst and
Some of the fibers from the SFO and other lamina sodium appetite.
terminalis structures are angiotensinergic. These pathways Taking into consideration the basic neurobiology of the
and their termination sites are associated with the control of brain systems maintaining fluid balance, it becomes
the behaviors and reflexes that maintain body fluid balance apparent that generation of thirst or sodium appetite results
and cardiovascular homeostasis (Fig. 4). Ascending facili- from a set of neural computations based on a complex of
tory and inhibitory inputs project into many of these same facilitory and inhibitory afferent signals. Phenomena such
forebrain regions. For example, norepinephrine is released as voluntary (or involuntary) dehydration arise from failure
at key forebrain sites and acts to reinforce the actions of to consume adequate sodium in conjunction with water.
angiotensin to generate thirst and sodium appetite. It is important to recognize that there are limits to the
Several recent studies have described the role of operating capabilities of the behavioral controls of body
inhibitory influences arising in the periphery and activating fluid homeostasis. Like any sensory system, there are
neural circuitry in the hindbrain. Central structures impli- thresholds for detection and discrimination of the pattern
cated in this inhibitory system are the AP/mNTS and the of stimuli that are perceived as thirst or sodium appetite. It
lateral PBN. The inhibitory actions of serotonin within the should be recognized that cognitive instructions derived

BEHAVIORAL MECHANISMS IN BODY FLUID BALANCE Medicine & Science in Sports & Exercised 1397

Copyright @ 2007 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
from the cortex can supplement the mechanisms of thirst controls affecting thirst and sodium appetite and how these
and sodium appetite which arise from phylogenetically old are influenced by the environment and exercise, a body of
parts of the nervous system. Appropriately informed, cog- knowledge will be developed to provide the best strategies
nitive mechanisms can be useful in maintaining adequate for maintenance and restoration of body fluid homeostasis
hydration. However, the strength of cognitive mechanisms for health and well-being.
can also lead to either inappropriate hypo- or hyperhydra-
tion, conditions with their own attendant risks.
Under extreme conditions all sensory systems fail. The work was supported in part by grants from the National
Heart, Lung, and Blood Institute HL 14388 and HL-57472, and from
Technology to enhance sensory capacity is appropriate in the National Institute of Diabetes and Digestive and Kidney
many circumstances. As more is learned about the nature of Diseases DK-066086.

REFERENCES
1. ADOLPH, E. F. Physiology of Man in the Desert, New York, NY: 18. DE GOBBI, J. I. F., L. A. DE LUCA JR., A. K. JOHNSON, and J. V.
Interscience Publishers, pp. 254–270, 1947. MENANI. Interaction of serotonin and cholecystokinin in the lateral
2. ADOLPH, E. F., J. P. BARKER, and P. A. HOY. Multiple factors in parabrachial nucleus to control sodium intake. Am. J. Physiol.
thirst. Am. J. Physiol. 178:538–562, 1954. Regul. Integr. Comp. Physiol. 280:R1301–R1307, 2001.
3. ANDRADE, C. A., S. P. BARBOSA, L. A. DE LUCA JR., and J. V. 19. DE LUCA, L. A. JR., O. GALAVERNA, J. SCHULKIN, S. Z. YAO, and
MENANI. Activation of alpha2-adrenergic receptors into the lateral A. N. EPSTEIN. The anteroventral wall of the third ventricle and the
parabrachial nucleus enhances NaCl intake in rats. Neuroscience angiotensinergic component of need-induced sodium intake in the
129:25–34, 2004. rat. Brain Res. Bull. 28:73–87, 1992.
4. BARR, S. I. Effects of dehydration on exercise performance. Can. 20. DENTON, D., R. SHADE, F. ZAMARIPPA, et al. Neuroimaging of
J. Appl. Physiol. 24:164–172, 1999. genesis and satiation of thirst and an interoceptor-driven theory of
5. BEALER, S. L., and A. K. JOHNSON. Sodium consumption following origins of primary consciousness. Proc. Natl. Acad. Sci. U. S. A.
lesions surrounding the anteroventral third ventricle. Brain Res. 96:5304–5309, 1999.
Bull. 4:287–290, 1979. 21. EDWARDS, G. L., T. G. BELTZ, J. D. POWER, and A. K. JOHNSON.
6. BEAUCHAMP, G. K., M. BERTINO, D. BURKE, and K. ENGELMAN. Rapid-onset ‘‘need-free’’ sodium appetite after lesions of the
Experimental sodium depletion and salt taste in normal human dorsomedial medulla. Am. J. Physiol. Regul. Integr. Comp.
volunteers. Am. J. Clin. Nutr. 51:881–889, 1990. Physiol. 264:R1242–R1247, 1993.
7. BOURQUE, C. W., S. H. OLIET, and D. RICHARD. Osmoreceptors, 22. EDWARDS, G. L., and A. K. JOHNSON. Enhanced drinking after
osmoreception, and osmoregulation. Front. Neuroendocrinol. excitotoxic lesions of the parabrachial nucleus in the rat. Am. J.
15:231–274, 1994. Physiol. Regul. Integr. Comp. Physiol. 261:R1039–R1044, 1991.
8. BUGGY, J., and A. K. JOHNSON. Preoptic-hypothalamic periven- 23. EPSTEIN, A. N. Epilogue: retrospect and prognosis. In: The
tricular lesions: thirst deficits and hypernatremia. Am. J. Physiol. Neuropsychology of Thirst: New Findings and Advances in
APPLIED SCIENCES

Regul. Integr. Comp. Physiol. 233:R44–R52, 1977. Concepts, A. N. Epstein, H. R. Kissileff, and E. Stellar (Eds.).
9. CALLERA, J. C., L. B. OLIVEIRA, S. P. BARBOSA, D. S COLOMBARI, Washington, DC: V.H. Winston, pp. 315–332, 1973.
L. A. DE LUCA JR., and J. V. MENANI. GABA(A) receptor 24. EPSTEIN, A. N. Mineralocorticoids and cerebral angiotensin may
activation in the lateral parabrachial nucleus induces water and act together to produce sodium appetite. Peptides 3:493–494,
hypertonic NaCl intake. Neuroscience 134:725–735, 2005. 1982.
10. CLEROUX, J., C. GIANNATTASIO, G. BOLLA, et al. Decreased 25. EPSTEIN, A. N., and E. STELLAR. The control of salt preference in
cardiopulmonary ref lexes with aging in normotensive humans. the adrenalectomized rat. J. Comp. Physiol. Psychol. 48:167–172,
Am. J. Physiol. Heart Circ. Physiol. 257:H961–H968, 1989. 1955.
11. CONTRERAS, R. J., and P. W. STETSON. Changes in salt intake 26. FALK, J. L. Serial sodium depletion and NaCl solution. Physiol.
lesions of the area postrema and the nucleus of the solitary tract in Behav. 1:75–77, 1966.
rats. Brain Res. 211:355–366, 1981. 27. FITTS, D. A. Effects of lesions of the ventral ventral median
12. CONVERTINO, V. A. Blood volume: its adaptation to endurance preoptic nucleus or subfornical organ on drinking and salt appetite
training. Med. Sci. Sports Exerc. 23:1338–1348, 1991. after deoxycorticosterone acetate or yohimbine. Behav. Neurosci.
13. CORBIT, J. D. Cellular dehydration and hypovolaemia are additive 105:721–726, 1991.
in producing thirst. Nature 218:886–887, 1968. 28. FITTS, D. A., D. S. TJEPKES, and R. O. BRIGHT. Salt appetite and
14. CUNNINGHAM, E. T. JR., R. R. MISELIS, and P. E. SAWCHENKO. The lesions of the ventral part of the ventral median preoptic nucleus.
relationship of efferent projections from the area postrema to vagal Behav. Neurosci. 104:818–827, 1990.
motor and brain stem catecholamine-containing cell groups: an 29. FITZSIMONS, J. T. Drinking by nephrectomized rats injected with
axonal transport and immunohistochemical study in the rat. various substances. J. Physiol. (Lond.) 155:563–579, 1961.
Neuroscience 58:635–648, 1994. 30. FITZSIMONS, J. T. Drinking by rats depleted of body fluid without
15. DE CARO, G., C. POLIDORI, T. G. BELTZ, and A. K. JOHNSON. Area increase in osmotic pressure. J. Physiol. (Lond.) 159:297–309,
postrema, lateral parabrachial nucleus, and the antinatriorexic 1961.
effect of bombesin. Peptides 19:1399–1406, 1998. 31. FITZSIMONS, J. T. Some historical perspectives in the physiology of
16. DE CARO, G., C. POLIDORI, J. V. MENANI, and A. K. JOHNSON. thirst. In: The Neuropsychology of Thirst: New Findings and
Bombesin affects the central nervous system to produce sodium Advances in Concepts, A. N. Epstein, H. R. Kissileff, and E.
intake inhibition in rats. Physiol. Behav. 63:15–23, 1998. Stellar (Eds.). Washington, DC: V.H. Winston, pp. 3–33, 1973.
17. DE CASTRO E SILVA, E., J. B. FREGONEZE, and A. K. JOHNSON. 32. FLUHARTY, S. J., and A. N. EPSTEIN. Sodium appetite elicited by
Corticotropin-releasing hormone in the lateral parabrachial intracerebroventricular infusion of angiotensin II in the rat: II.
nucleus inhibits sodium appetite in rats. Am. J. Physiol. Regul. Synergistic interaction with systemic mineralocorticoids. Behav.
Integr. Comp. Physiol. 290:R1136–R1141, 2006. Neurosci. 97:746–758, 1983.

1398 Official Journal of the American College of Sports Medicine http://www.acsm-msse.org

Copyright @ 2007 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
33. GIANNATTASIO, C., G. SERAVALLE, G. B. BOLLA, et al. Cardiopulmo- gic mechanisms: angiotensin-induced pressor and drinking
nary receptor reflexes in normotensive athletes with cardiac responses. Am. J. Physiol. Regul. Integr. Comp. Physiol. 269:
hypertrophy. Circulation 82:1222–1229, 1990. R1044–R1049, 1995.
34. GILMAN, A. The relation between blood osmotic pressure, fluid 54. MENANI, J. V., and A. K. JOHNSON. Cholecystokinin actions in
distribution and voluntary water intake. Am. J. Physiol. 120: the parabrachial nucleus: effects on thirst and salt appetite.
323–328, 1937. Am. J. Physiol. Regul. Integr. Comp. Physiol. 275:R1431–R1437,
35. GREENLEAF, J. E. Problem: thirst, drinking behavior, and involun- 1998.
tary dehydration. Med. Sci. Sports Exerc. 24:645–656, 1992. 55. NOSE, H., A. TAKAMATA, G. W. MACK, et al. Water and electrolyte
36. HOLMES, J. H., and M. I. GREGERSEN. Observations on drinking balance in the vascular space during graded exercise in humans.
induced by hypertonic solutions. Am. J. Physiol. 162:326–337, J. Appl. Physiol. 70:2757–2762, 1991.
1950. 56. OHMAN, L. E., and A. K. JOHNSON. Role of lateral parabrachial
37. HOLMES, J. H., and M. I. GREGERSEN. Role of sodium and chloride nucleus in the inhibition of water intake produced by right atrial
in thirst. Am. J. Physiol. 162:338–347, 1950. stretch. Brain Res. 695:275–278, 1995.
38. JOHNSON, A. K. The periventricular anteroventral third ventricle 57. PECK, J. W. Discussion: Thirst(s) resulting from bodily water
(AV3V): its relationship with the subfornical organ and neural imbalances. In: The Neuropsychology of Thirst: New Findings
systems involved in maintaining body fluid homeostasis. Brain and Advances in Concepts, A. N. Epstein, H. R. Kissileff,
Res. Bull. 15:595–601, 1985. and E. Stellar (Eds.). Washington, DC: V.H. Winston & Sons,
39. JOHNSON, A. K. Brain mechanisms in the control of body fluid pp. 99–112, 1973.
homeostasis. In: Perspectives in Exercise Science and Sports 58. RICHTER, C. P. Increased salt appetite in adrenalectomized rats.
Medicine, Volume 3: Fluid Homeostasis During Exercise, C. V. Am. J. Physiol. 115:155–161, 1936.
Gisolfi and D. R. Lamb (Eds.). Carmel, IN: Benchmark Press, pp. 59. ROBINSON, M. M., and M. D. EVERED. Pressor action of intra-
347–419, 1990. venous angiotensin II reduces drinking response in rats. Am. J.
40. JOHNSON, A. K., and J. BUGGY. Periventricular preoptic-hypothal- Physiol. Regul. Integr. Comp. Physiol. 252:R754–R759, 1987.
amus is vital for thirst and normal water economy. Am. J. Physiol. 60. ROWELL, L. B. Human Cardiovascular Control. New York, NY:
Regul. Integr. Comp. Physiol. 234:R122–R129, 1978. Oxford University Press, 1993.
41. JOHNSON, A. K., J. T. CUNNINGHAM, and R. L. THUNHORST. 61. SAGAWA, S., K. MIKI, F. TAJIMA, et al. Effect of dehydration on
Integrative role of the lamina terminalis in the regulation of thirst and drinking during immersion in men. J. Appl. Physiol.
cardiovascular and body fluid homeostasis. Clin. Exp. Pharmacol. 72:128–134, 1992.
Physiol. 23:183–191, 1996. 62. SAKAI, R. R., W. B. FINE, A. N. EPSTEIN, and S. P. FRANKMANN. Salt
42. JOHNSON, A. K., J. DE OLMOS, C. V. PASTUSKOVAS, A. M. ZARDETTO- appetite is enhanced by one prior episode of sodium depletion in
SMITH, and L. VIVAS. The extended amygdala and salt appetite. the rat. Behav. Neurosci. 101:724–731, 1987.
Ann. N. Y. Acad. Sci. 877:258–280, 1999. 63. SAWKA, M. N., and E. F. COYLE. Influence of body water and
43. JOHNSON, A. K., and P. M. GROSS. Sensory circumventricular blood volume on thermoregulation and exercise performance in
organs and brain homeostatic pathways. FASEB J. 7:678–686, the heat. Exerc. Sport Sci. Rev. 27:167–218, 1999.
1993. 64. SHAPIRO, R. E., and R. R. MISELIS. The central neural connections
44. JOHNSON, A. K., and R. L. THUNHORST. The neuroendocrinol- of the area postrema of the rat. J. Comp. Neurol. 234:344–364,
ogy of thirst and salt appetite: visceral sensory signals and mecha- 1985.

APPLIED SCIENCES
nisms of central integration. Front. Neuroendocrinol. 18:292–353, 65. SIEGEL, A. J. Exercise-associated hyponatremia: role of cytokines.
1997. Am. J. Med. 119:S74–S78, 2006.
45. JOHNSON, A. K., and R. L. THUNHORST. The neuroendocrinology, 66. SIMPSON, J. B., and A. ROUTTENBERG. Subfornical organ: site of
neurochemistry and molecular biology of thirst and salt appetite. drinking elicitation by angiotensin II. Science 181:1172–1175,
In: Handbook of Neurochemistry and Molecular Neurobiology: 1973.
Behavioral Neurochemistry, Neuroendocrinology and Molecular 67. STACHENFELD, N. S., L. DIPIETRO, E. R. NADEL, and G. W. MACK.
Neurobiology, 3rd ed., A. Lajtha and J. D. Blaustein (Eds.). New Mechanisms of attenuated thirst in aging: role of central volume
York, NY: Springer, pp. 641–688, 2007. receptors. Am. J. Physiol. Regul. Integr. Comp. Physiol. 272:
46. KAUFMAN, S. Role of right atrial receptors in the control of R148–R157, 1997.
drinking in the rat. J. Physiol. (Lond.) 349:389–396, 1984. 68. STRICKER, E. M. Osmoregulation and volume regulation in rats:
47. KENNEY, W. L., and P. CHIU. Influence of age on thirst and fluid inhibition of hypovolemic thirst by water. Am. J. Physiol. 217:
intake. Med. Sci. Sports Exerc. 33:1524–1532, 2001. 98–105, 1969.
48. LAN0A, A. J., and D. VAN DER KOOY. A serotonin-containing 69. STRICKER, E. M. Inhibition of thirst in rats following hypovolemia
pathway from the area postrema to the parabrachial nucleus in the and/or caval ligation. Physiol. Behav. 6:293–298, 1971.
rat. Neuroscience 14:1117–1126, 1985. 70. STRICKER, E. M., K. S. GANNON, and J. C. SMITH. Thirst and salt
49. MASSI, M., L. GENTILI, M. PERFUMI, G. DE CARO, and J. SCHULKIN. appetite induced by hypovolemia in rats: analysis of drinking
Inhibition of salt appetite in the rat following injection of behavior. Physiol. Behav. 51:27–37, 1992.
tachykinins into the medial amygdala. Brain Res. 513:1–7, 71. STRICKER, E. M., and J. E. JALOWIEC. Restoration of intravascular
1990. fluid volume following acute hypovolemia in rats. Am. J. Physiol.
50. MCCANCE, R. A. Medical problems in mineral metabolism: III. 218:191–196, 1970.
Experimental human salt deficiency. Lancet 1:823–830, 1936. 72. STRICKER, E. M., and J. G. VERBALIS. Sodium appetite. In:
51. MENANI, J. V., L. A. DE LUCA JR., and A. K. JOHNSON. Lateral Handbook of Behavioral Neurobiology, Vol. 10, Neurobiology of
parabrachial nucleus serotonergic mechanisms and salt appetite Food and Fluid Intake, E. M. Stricker (Ed.). New York, NY:
induced by sodium depletion. Am. J. Physiol. Regul. Integr. Plenum Press, pp. 387–419, 1990.
Comp. Physiol. 274:R555–R560, 1998. 73. SUTTON, J. R. Clinical implications of fluid imbalance. In:
52. MENANI, J. V., L. A. DE LUCA JR., R. L. THUNHORST, and A. K. Perspectives in Exercise Science and Sports Medicine, Volume
JOHNSON. Hindbrain serotonin and the rapid induction of sodium 3: Fluid Homeostasis During Exercise, C. V. Gisolfi and D. R.
appetite. Am. J. Physiol. Regul. Integr. Comp. Physiol. 279: Lamb (Eds.). Carmel, IN: Benchmark, pp. 425–444, 1990.
R126–R131, 2000. 74. TAKAMATA, A., G. W. MACK, C. M. GILLEN, and E. R. NADEL.
53. MENANI, J. V., and A. K. JOHNSON. Lateral parabrachial serotoner- Sodium appetite, thirst, and body fluid regulation in humans

BEHAVIORAL MECHANISMS IN BODY FLUID BALANCE Medicine & Science in Sports & Exercised 1399

Copyright @ 2007 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.
during rehydration without sodium replacement. Am. J. Physiol. appetite. Am. J. Physiol. Regul. Integr. Comp. Physiol. 252:
Regul. Integr. Comp. Physiol. 266:R1493–R1502, 1994. R925–R929, 1987.
75. THUNHORST, R. L., T. G. BELTZ, and A. K. JOHNSON. Effects of 79. VAN DER KOOY, D., and L. Y. KODA. Organization of the projec-
subfornical organ lesions on acutely induced thirst and salt appetite. tions of a circumventricular organ: the area postrema in the rat.
Am. J. Physiol. Regul. Integr. Comp. Physiol. 277:R56–R65, 1999. J. Comp. Neurol. 219:328–338, 1983.
76. THUNHORST, R. L., and A. K. JOHNSON. Renin-angiotensin, arterial 80. VERNEY, E. B. The antidiuretic hormone and the factor which
blood pressure, and salt appetite in rats. Am. J. Physiol. 266: determines its release. Proc. R. Soc. Lond. B Biol. Sci. 135:27–106,
R458–R465, 1994. 1947.
77. THUNHORST, R. L., S. J. LEWIS, and A. K. JOHNSON. Effects of 81. WOLF, A. V. Osmometric analysis of thirst in man and dog. Am. J.
sinoaortic baroreceptor denervation on depletion-induced salt Physiol. 161:75–86, 1950.
appetite. Am. J. Physiol. 267:R1043–R1049, 1994. 82. WOLF, G. Sodium appetite elicited by aldosterone. Psychon. Sci.
78. TOTH, E., J. STELFOX, and S. KAUFMAN. Cardiac control of salt I:211–212, 1964.
APPLIED SCIENCES

1400 Official Journal of the American College of Sports Medicine http://www.acsm-msse.org

Copyright @ 2007 by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.

You might also like