You are on page 1of 40

16 FLUID, ELECTROLYTE AND

METABOLIC MANAGEMENT
Peter D. Thomas

16.1 Introduction injury and any of six potentially preventable second-


ary insults: intracranial hematoma, raised ICP, seiz-
The medical management of the severely head-injured ures, meningitis, hypoxia and hypotension (Price,
patient involves manipulations of the intracranial 1992). About 20% of patients with head injuries also
pressure (ICP), cerebral perfusion pressure (CPP), suffer injury to other systems. The injured brain is
seizure control, fluid and electrolyte therapy and highly vulnerable to hypoxia and ischemia, which
nutritional support. The goal of fluid management is may exacerbate neuronal damage by a number of
homeostasis, i.e. to provide appropriate parenteral mechanisms, including the release of excitotoxic neu-
and/or enteral fluid to maintain intravascular vol- rotransmitters such as glutamate (Zwimpfer and
ume, left ventricular filling pressure, cardiac output, Moulton, 1993). Extracranial injury that does not result
blood pressure and ultimately oxygen delivery (ḊO2) in hypotension or hypoxia appears to have little
to tissues, when normal physiological functions are influence on outcome (Jennett, Teasdale and Braak-
often altered by surgical and traumatic stress and by man, 1979). Hence preventing and aggressively treat-
anesthetic agents. A specific aim is to prevent second- ing hypotension and hypoxia are essential parts of the
ary neuronal damage due to inadequate ḊO2 to the early management of all patients with head injury.
brain; this requires adequate ventilation and oxy- Approximately 50% of patients admitted to the
genation as well as cardiac output. The most obvious hospital in a coma following head injury will require
consequences of inappropriate fluid resuscitation are craniotomy (Butterworth and De Witt, 1989). Others
shock from insufficient volume replacement, and who do not require craniotomy may require anes-
pulmonary edema from overhydration. thesia and surgery for other major injuries. The
Glucose, lipids, protein, vitamins and essential trace anesthesia and surgery can both affect fluid and
elements are provided in order to retard the ‘auto- electrolyte status.
cannibalism’ of visceral protein for gluconeogenesis. Intravenous therapy is an integral part of both the
Fluid planning must be individualized and take into initial resuscitation phase and the later maintenance or
account the patient’s cardiac, renal, pulmonary, nutri- supportive phase of management. The primary aim of
tional, functional and premorbid state. resuscitation is to achieve satisfactory ventilation and
Fluid and electrolyte abnormalities may result from restore intravascular volume, cardiac output and tissue
the brain injury itself, e.g. diabetes insipidus, from perfusion, thereby preventing the sequelae of shock,
therapy and from other injuries. The relationship acute renal and multiple organ failure and secondary
between systemic abnormalities of water, electrolytes neuronal damage. The secondary aim is correction of
and acid–base metabolism and the brain can be any underlying disorder. In adults prolonged hypoten-
approached from two vantage points: sion is rarely due to head injury alone, unless the injury
is massive and brain death is imminent, and concealed
 the effects of CNS lesions on renal function, water
hemorrhage must be sought. In infants and young
and electrolytes;
children, hypovolemic shock may result from blood
 the effects of metabolic abnormalities on the CNS.
loss in a large extradural hematoma, especially if there
is additional bleeding under the scalp. Shoemaker has
demonstrated that O2 transport variables (ḊO2 and
16.2 Rationale of metabolic support
oxygen consumption – V̇O2) can reliably predict
The outcome after a severe head injury is influenced outcome in postoperative surgical patients. Conversely
by age, pre-accident health, the severity of the primary when these variables are manipulated in a controlled

Head Injury. Edited by Peter Reilly and Ross Bullock. Published in 1997 by Chapman & Hall, London. ISBN 0 412 58540 5
294 FLUID, ELECTROLYTE AND METABOLIC MANAGEMENT

prospective manner, outcome can be improved (Shoe- electrolyte problems, the factors controlling electrolyte
maker et al. 1982, 1988). The determinants of oxygen distribution and extracellular fluid volume and the
delivery are summarized in Figure 16.1. Fluid resusci- distribution of administered fluids since the choice of
tation augments preload, increases stroke output and intravenous fluids will influence brain metabolism
total systemic flow, thereby increasing oxygen delivery. and volume (Sutin, Ruskin and Kaufman, 1992). From
Care must be taken to avoid hemodilution, which may this knowledge appropriate therapeutic strategies can
simultaneously reduce arterial oxygen content and be developed.
prevent an increase in ḊO2 despite improvement in
cardiac index. Oxygen saturation should be maintained
16.3 Basic principles
at 90% or greater in accordance with the recommenda-
tion of the American College of Chest Physicians
16.3.1 FLUID DISTRIBUTION
Consensus Conference on Mechanical Ventilation
(Slutsky, 1993). The total body water content of humans is approx-
Following resuscitation, fluid therapy aims to main- imately 60% of body weight (Figure 16.2). Two-
tain euvolemia, provide normal daily requirements thirds is located in the intracellular and one-third in
and replace any continuing losses. It is usually the extracellular compartment (Harrison, Darrow
combined with nutritional support. and Yannet, 1936). The extracellular compartment
Cerebral autoregulation determines the relationship can be further subdivided into interstitial (protein-
between cerebral blood flow (CBF) and metabolic poor) and intravascular (protein-rich) compartments.
needs (metabolic coupling) and systemic hemody- Normally, 75% of the extracellular water is contained
namics (pressure autoregulation). When metabolic within the interstitium and 25% in the vasculature.
coupling is intact, cerebral oxygen delivery is deter- There are marked differences in electrolyte content
mined by metabolic need and is unaffected by minor between intracellular and extracellular fluids: potas-
changes in blood pressure or viscosity. Both aspects of sium is predominantly intracellular, and sodium and
cerebral autoregulatory function may be impaired chloride extracellular (Gamble, Ross and Tisdall,
following traumatic brain injury, so that cerebral 1923). The energy-consuming Na+ –K+ pump is
oxygen delivery is influenced by cerebral perfusion
pressure (CPP) and blood viscosity (Shoemaker et al.,
1988). Thus maintaining a normal CPP is a key aspect
of the management of the head-injured patient.
Most severely injured patients develop only small
changes in serum electrolyte concentrations, even
though there are large fluid and electrolyte shifts
between body compartments. Thus it is important to
understand the physiological basis of the common

Figure 16.1 Systemic oxygen delivery – ḊO2 (ml/min) – is


determined by the product of the oxygen content of the
blood and total systemic blood flow. The major determinants
of each component are pictured. Hb = hemoglobin; % SAT =
percentage saturation of Hb; PaO2 = partial pressure of
oxygen in arterial blood; 0.0031 = solubility coefficient for Figure 16.2 Distribution of body water in a 70 kg man (45.5
oxygen in plasma. liters).
BASIC PRINCIPLES 295
required to maintain these concentration gradients, Cell membranes are normally relatively impermea-
which are sustained during sodium overload or ble to ions but freely permeable to water. Water,
depletion states. Distribution of water between the electrolytes and energy substrates continually move in
intracellular and extracellular compartments is and out of cells to maintain a dynamic steady-state
determined by osmosis. relationship and major fluid and electrolyte shifts
Extracellular fluid includes water ‘attached’ to bone occurring with disease are often associated with only
and dense connective tissue, as well as free trans- minimal serum concentration changes. For example, a
cellular or ‘third-space’ fluid formed by secretions decrease in extracellular sodium concentration and
from epithelia and other membranes. Free trans- osmolarity following an infusion of 5% dextrose in
cellular fluid includes cerebrospinal fluid (CSF), urine, water is followed by diffusion of water into the cells
gastrointestinal and other secretory fluid, which may until the osmotic pressure between the two compart-
have strikingly different osmolalities from the two ments is equal. Conversely, an increase in extracellular
major body fluid compartments (Shoemaker, 1982). sodium concentration and osmolarity after infusion of
Although this third space may be greatly expanded in hypertonic saline is followed by movement of water
disease states such as ascites, pleural effusion, para- from the intracellular to the extracellular compart-
lytic ileus or bowel obstruction, it is separated from ment. Infusion of an isotonic electrolyte solution does
other body fluid compartments by epithelium and not alter the osmolarity of the extracellular compart-
therefore does not communicate freely with them. The ment and therefore does not lead to shifts of water into
third space can be thought of as ‘parasitic’ – it is or out from the intracellular compartment.
generated at the expense of the other two compart- Water movement between the two components of
ments but is not available to ‘come to their rescue’ in the extravascular space is determined primarily by
states of hypovolemia. Since most forms of expansion differences in protein concentration. Colloid osmotic
of the extracellular fluid do not involve the third space pressure (COP) is the net osmotic pressure across the
fluids and the amount of water attached to bone and capillary membrane resulting from the impermeabil-
dense connective tissue is relatively fixed, the normal ity of the endothelium to plasma proteins. Because
body water distribution can be simplified as shown in physical confinement of protein molecules produces
Figure 16.3. an osmotic imbalance, water moves from interstitial
space into the blood stream. Crystalloid molecules
cannot establish a pressure gradient because they
move freely across the capillary membrane.
Protein molecules are negatively charged and
attract a small number of positive ions, preventing
them from diffusing across the endothelium. These
retained ions produce an additional osmotic pressure
(called the Donnan equilibrium effect). Some 60% of
normal COP is produced by protein molecules (75%
by albumin, the rest mainly by globulins and fibrino-
gen); 40% is produced by the electrostatically held
cations. However, if the plasma protein concentration
rises, the influence of the Donnan equilibrium effect
on COP increases disproportionately, producing a
curvilinear relationship between total protein concen-
tration and COP.
COP is measured by placing a protein solution on
one side of a semipermeable membrane and a protein-
free solution on the other side. Fluid moves through
the membrane until a pressure is generated that
prevents further osmosis. This pressure is defined as
the COP (Morissette, 1977). Plasma COP in normal
ambulatory patients is approximately 25 mmHg, in
supine patients 22 mmHg and in an Intensive Care
Unit patient 18–20 mmH (Weil et al., 1974, 1981;
Rackow, Fein and Leppo, 1977). Although protein is
Figure 16.3 Simplified distribution of body water in a 70 kg the primary determinant of osmotic pressure in
man. The forces that influence fluid distribution between the peripheral tissues, it contributes very little to the total
compartments are shown in italics. number of particles dissolved in plasma. Total plasma
296 FLUID, ELECTROLYTE AND METABOLIC MANAGEMENT

osmolality is normally 285 mosmol/kg, of which proteins. If  = 1, the membrane is totally impermea-
plasma protein accounts for less than 1 mosmol/kg. ble and proteins are able to exert their full oncotic
Infusing an iso-oncotic solution such as 5% albumin force; if  = 0, the membrane permits protein to pass
in isotonic saline expands the intravascular space without impedance (Peters and Hargens, 1981). 
without producing a shift of water from other com- varies in capillary membranes throughout the body,
partments. A hyperoncotic solution such as 25% being approximately 0.9 for systemic and 0.7 for lung
albumin expands blood volume to a extent greater capillaries (Wittmers, Bartlett and Johnson, 1976). The
than the volume infused by drawing water and net effect of the Starling forces across capillaries is to
electrolytes from the interstitium. However water produce fluid movement from the intravascular to the
does not diffuse from the intracellular space if interstitial space. Accumulation of interstitial fluid in
extracellular osmolarity remains unchanged. the lung is much more life-threatening than peripheral
Starling defined the forces influencing the bulk edema formation and there has been great interest in
movement of water between the vascular and inter- evaluating factors that are likely to reduce the risk of
stitial compartments (Starling, 1896). Pappenheimer clinically significant pulmonary edema (Allen et al.
and Soto-Rivera derived a mathematical expression 1987; Civetta, 1979; Crandall et al., 1983).
for these forces, referred to as the Starling equation of Modest increases in pulmonary capillary hydro-
transcapillary exchange: (Pappenheimer and Soto- static pressure or decreases in plasma oncotic pressure
Rivera, 1948). do not result in pulmonary edema because the lung
has mechanisms for resisting accumulation of inter-
Qf = Kf [(Pc – Pi) –  (c – i)] stitial fluid. In the first place, the pulmonary inter-
where Qf = total fluid flow across the capillary stitial hydrostatic pressure (Pi) is normally slightly
membrane; Kf = fluid filtration coefficient; Pc = negative (Civetta, 1979; Levine et al., 1967; Taylor et al.,
capillary hydrostatic pressure; Pi = interstitial hydro- 1982). When fluid first begins to accumulate in the
static pressure;  = osmotic reflection coefficient; c = interstitium, the pressure rapidly increases (i.e. com-
capillary oncotic pressure; i = interstitial oncotic pliance is low). This increase in hydrostatic pressure
pressure. opposes any further fluid entry by decreasing the
hydrostatic pressure gradient, Pc – Pi (Allen et al.,
The four pressures in this equation are called the 1987). However, as interstitial fluid pressure rises
Starling forces. The net driving pressure favoring above atmospheric, resistance to fluid transport
filtration is Pc – Pi. The hydrostatic pressure within the through the interstitium decreases markedly and
capillary is the major force driving fluid into the further increases in fluid volume cause only minimal
interstitium and is essentially unopposed by the increases in interstitial hydrostatic pressure (Levine et
interstitial hydrostatic pressure. This is usually al., 1967; Goldberg, 1980). Pi appears to level off
slightly negative and approaches zero or becomes between 1 and 5 mmHg in severe pulmonary edema
slightly positive only when substantial amounts of (Battacharya, Gropper and Staub, 1984).
edema accumulate (Meyer, Meyer and Guyton, 1968; A second mechanism which resists the accumula-
Guyton, Granger and Taylor, 1971). The plasma COP tion of pulmonary interstitial fluid is a decrease in the
is thus the only force acting to retain fluid within the interstitial oncotic pressure resulting from the accu-
intravascular space. The interstitial COP works in the mulation of protein-poor fluid (Granger,1979). Pulmo-
opposite direction; the net effect of these opposing nary interstitial oncotic pressure is normally about
forces is described by c – i 75% of the plasma level and the oncotic gradient
Kf, the filtration coefficient, has two components: Lp, across the pulmonary capillary membrane (c – i) is
the hydraulic conductivity, which describes how only 4–6 mmHg. If serum COP decreases, interstitial
rapidly fluid can pass through the microvascular oncotic pressure will decrease proportionately as fluid
exchange barrier (capillary membrane, interstitial gel enters the interstitial space, avoiding a change in the
and terminal lymphatics) and S, the capillary surface oncotic gradient (Granger, 1979; Demling, 1980; Deml-
area available for filtration (Granger, 1979; Harms et ing et al., 1979).
al., 1981; Demling et al., 1982). If either component of Thirdly, large protein molecules such as albumin are
Kf increases, e.g. through damage to the endothelial normally excluded from a substantial part of the
membrane or dilatation of precapillary vasculature in interstitial fluid volume by the density of the interstitial
response to increased cardiac output (CO), the rate matrix. However, as the degree of hydration increases
and amount of fluid filtered increase independently of the fibers in the interstitial matrix are stretched apart,
changes in the Starling forces (Peters and Hargens, allowing protein molecules to enter previously
1981). unavailable space (Granger, 1979). This lowers i,
The reflection coefficient  defines the capacity of widens the transcapillary oncotic gradient (c – i), and
the capillary membrane to prevent translocation of thus opposes further pulmonary edema formation.
BASIC PRINCIPLES 297
Lymphatic drainage is the fourth and probably POSM = 2PNa+ + glucose + urea
most important safety factor. Experimental studies
where the concentration of sodium, glucose and urea
have shown that the lymph flow rate can increase
are all expressed in mmol/l (Worthley, Guerin and
tenfold when interstitial fluid volume or pressure
Pain, 1987).
increases.
The blood–brain barrier is formed by a closely The plasma sodium (PNa+) level is multiplied by
woven mesh of capillary cells joined by a continuous two to account for anions. This rule of thumb does not
array of tight junctions (zonula occludens) with an take into account the low-concentration cations such
effective pore size of 0.7–0.9 nm. Because the blood– as potassium, calcium and magnesium or organic
brain barrier is a lipid membrane with small pores, it molecules such as creatinine, amino acids and lactate.
is easily permeated by water and very small or This error of exclusion is balanced by the presence of
lipophilic molecules. The blood–brain barrier is rela- multivalent anions such as sulfate and phosphate
tively impermeable to electrolytes, water-soluble non- which contribute fewer particles per equivalent than
electrolytes and plasma proteins. The endothelial cells do univalent ions.
in peripheral tissues are joined by interspersed tight Although urea is included in the calculation of
junctions and the effective endothelial pore size is osmolality, it diffuses readily across cell membranes
about 4–5 nm. Because of the large pore size in and does not influence relative compartment volumes
peripheral tissues, the endothelial membrane is freely (Brobeck,1973). Exogenous solutes such as mannitol or
permeable to water and electrolytes, but only partially alcohol may create an ‘osmolar gap’ between the
permeable to plasma proteins. In the peripheral measured and unmeasured osmolalities (Table 16.1).
tissues there is an active lymphatic system, which This formula must be interpreted in conjunction
helps reabsorb interstitial fluid. In the brain, however, with the clinical assessment of fluid status as, for
there are no lymphatics. When the blood–brain barrier example, an increased osmolality may result either
is injured, the barrier becomes permeable to electro- from excess sodium or a deficit of water.
lytes and large plasma proteins such as albumin Tonicity defines effective osmolality, i.e. the osmotic
(Zornow et al., 1988). Hypertonic fluids will only force due to the particles which cannot permeate
remove interstitial and intracellular fluid from regions between compartments. Because all body fluids must
of the brain where the blood–brain barrier is intact, be in osmotic equilibrium, changes in tonicity, regard-
and will have no effect on areas where the blood– less of their cause, will result in fluid shifts between
brain barrier is permeable to electrolytes. the ECF and the ICF in order to re-establish osmotic
The difference in membrane function between the equilibrium. The administration of hypertonic fluids
blood–brain barrier and the endothelium in periph- such as 50% dextrose or hypertonic saline will increase
eral tissues is of practical importance. For example, if the tonicity of the ECF and result in fluid shifts into
the intravascular concentration of sodium is increased the extracellular compartment. Thus patients with
from 140 mmol/l to 150 mmol/l, there will only be a osmotic hypertonicity will have a contracted intra-
small and short-lasting effect on the peripheral inter- cellular compartment.
stitial tissues, owing to the free movement of both This effect had been applied to the treatment of
sodium and water across the endothelium. However patients with cerebral edema in whom intracellular
such a change in sodium concentration will have a and extracellular volumes can be decreased by achiev-
pronounced effect on the brain. The osmotic pressure ing hypernatremia. Conversely in hyponatremic states
gradient will increase drawing water from the brain the tonicity of the ECF is decreased and water must
interstitium to the intravascular space. shift into and expand the intracellular fluid to main-
tain osmolality. As brain expansion is limited by the
confines of the cranial vault, severe hyponatremia
16.3.2 OSMOLALITY AND TONICITY

Osmolality is the number of dissolved particles in a


kilogram of solvent (water) and determines the Table 16.1 Osmotically active particles
osmotic force of the solution (Shoemaker, 1982). Cell
membranes are highly permeable to water, which will Extracellular fluid Intracellular fluid
diffuse across them rapidly to maintain osmotic Sodium Potassium
equilibrium between the extracellular and intracel- Glucose Magnesium
lular fluid compartments (Brobeck,1973). Hence the (Mannitol) Inorganic phosphate
relative volumes of the compartments will be deter- Urea* Urea*
Ethanol* Ethanol*
mined by their sodium and potassium content.
Plasma osmolality can be measured directly or *These particles are distributed freely in body water. Other
calculated approximately by means of the formula: particles are unable to permeate compartments.
298 FLUID, ELECTROLYTE AND METABOLIC MANAGEMENT

may result in raised intracranial pressure (ICP). Urea tissue oxygenation, but the focus in clinical medicine
increases osmolality but not tonicity because it passes has been on the level of serum hemoglobin or the
freely through biological membranes, and will not hematocrit. It has been estimated that tissue ḊO2 is
alter the intracellular volume,. optimal when the hematocrit is 33% (Sutin, Ruskin
and Kaufman, 1992), but the effect on ḊO2 may not be
the same as the effect on oxygen utilization. A
16.3.3 DISTRIBUTION OF ADMINISTERED FLUID IN
consensus development conference on perioperative
THE BODY
red cell transfusion concluded that available evidence
All infused fluids may be considered as being com- did not support the recommended practice of main-
posed of plasma equivalents (colloidal solutions), taining hemoglobin levels at 10 g/dl or hematocrits at
saline equivalents (crystalloid solutions) or water 30% (Consensus Development Conference, 1988). In
equivalents. Plasma equivalents remain essentially the United States, the National Blood Resource Educa-
confined to the circulation as a result of their oncotic tion Program guidelines for red blood cell transfusion
pressure. Saline equivalents are distributed between state that adequate oxygen-carrying capacity can be
the interstitium and plasma, but remain restricted to met by a hemoglobin of 7 g/dl (hematocrit of approx-
the extracellular fluid compartment because of their imately 21%) or even less when the intravascular
sodium content. Water equivalents are distributed volume is adequate for perfusion. The guidelines add
according to the distribution of water in the body the caveat that the decision to transfuse a given
(Figure 16.2). patient should be based on consideration of the
patient’s age, etiology and degree of anemia, hemody-
namic stability and presence of coexisting cardiac,
(a) Plasma equivalents
pulmonary or vascular disease. While the normal
Plasma equivalents include blood, its components and brain is very tolerant to anemia, the limits of iso-
colloids. Blood may be administered either whole, or volemic anemia are unknown. Because autoregulation
in its component form; the latter is preferred, as may be impaired after injury, it cannot be assumed
fractionation optimizes the availability of the various that the safe lower limit of the hematocrit is the same
blood components for special situations and improves as in the neurologically normal subject.
the survival of the stored blood elements. Its use is Colloidal solutions comprise preparations of plasma
limited by cost and availability, the risks of allergic or high-molecular-weight synthetic substances (Table
reactions and infection, the delays necessary for cross- 16.2). These fluids do not cross capillary membranes
matching and a limited shelf life. readily and are initially confined mainly to the
Patients who are bleeding acutely are best treated in intravascular space. If the colloidal solution has an
part with whole blood since this also replaces clotting oncotic pressure that is higher than that of the plasma
components and platelets. Otherwise hypovolemia interstitial fluid will move into the intravascular space
can be treated effectively by colloid or crystalloid and expand the plasma volume by more than the
solutions, and oxygen transport function can be administered volume of colloidal solution, hence the
restored by red-cell concentrates, although these take term ‘plasma expander’.
several hours to be fully effective since storage impairs
oxygen dissociation.
Plasma-protein derivatives
The absolute minimum acceptable hematocrit in the
trauma patient is difficult to determine. The goal of These include 5% normal serum albumin, which
blood transfusion is to maintain an adequate level of contains human albumin 50 g/l and sodium

Table 16.2 Colloidal solutions

Volume Mean mol. Protein Sodium content Oncotic Half-life Side effects Cost/bottle
Type (ml) wt (kDa) content (mmol/bottle) pressure Other constituents (%)* (Aust$)

Normal serum 500 69 5% (25 g) 70 Iso-oncotic – 5–10 d 0.02 (0.004) 60.00


albumin
Concentrated 100 69 20% (20 g) 10 5 – 5–10 d 0.01 (0.003) 85.00
albumin
Dextran 70 500 70 6% 77 or 0  2.5 – 12 h 0.07 (0.017) 8.00
(Macrodex)
Polygeline 500 35 3.5% 72 Iso-oncotic Potassium, 5 mmol/l; 4h 0.15 (0.05) 12.50
(Haemaccel) calcium, 6 mmol/l

*Percentage incidence of all reactions. Percentage incidence of major (grade-3 and grade-4) reactions in parentheses.
BASIC PRINCIPLES 299
140 mmol/l, concentrated (20%) albumin, which con- kinin activation, are rare. Because of their long half-life
tains 200 g/l of albumin and less sodium (100 mmol/l). care must be taken to avoid circulatory overload in
The latter is usually restricted to hypoproteinemic patients who have been resuscitated with plasma-
patients with either hypernatremia or fluid overload. A protein solutions and appropriate hemodynamic mon-
500 ml infusion of 5% albumin expands the intra- itoring is required.
vascular volume by 450–500 ml (Lamke and Liljedahl,
1976). After 2 hours, under conditions of normal
Synthetic colloidal solutions
capillary permeability, 90% remains within the intra-
vascular space (Rainey and English, 1988). Eventually Dextrans are polysaccharides composed of glucose
the administered albumin is distributed throughout the molecules polymerized into chains of various lengths.
extra cellular space (Rainey and English, 1988). After They are classified according to molecular weight and
infusion of 100 ml of concentrated albumin (20%), the are available in isotonic saline or 5% dextrose. Because
plasma volume continues to increase over the next of their shape dextran molecules are hyperoncotic –
30–60 minutes to achieve a final blood volume i.e. they have the water-attracting equivalent of
expansion of 400–450 ml. Redistribution of 350 ml of several individual molecules; thus they produce a
interstitial fluid to the intravascular space is necessary plasma-volume expansion of about 120% of the
for this to occur. In patients with extracellular or total infused volume (Scheinkestel et al., 1989).
body water depletion, equilibration is slow and Potentially life-threatening toxic effects may compli-
incomplete (Beecher, 1945). Therefore, in acute hypovo- cate dextran administration, including acute renal
lemia, 5% albumin should be given rather than the failure, anaphylaxis and bleeding diathesis (Atik,
hyperoncotic form. The 20% albumin solution is 1969). Dextran 40 has been associated with acute renal
usually used in patients with concomitant hypovole- failure due to irreversible plugging of the renal
mia and elevated extracellular fluid volume. tubules; Dextran 70 is rarely associated with the
The duration of vascular retention and the hemody- development of acute renal failure.
namic effects of infused albumin solutions depend The incidence of anaphylactoid reactions after dex-
greatly on the patient’s disease state. This variability tran administration was reported to be 0.032%. Dextran
may result from ‘leakage’ into the interstitium, prefer- 40 produced fewer reactions than Dextran 70 and severe
ential binding of albumin in the skin and wound, or reactions were uncommon (Ring and Messmer, 1977).
increased catabolism. Dextran 70 and, to a lesser extent, Dextran 40
Albumin has several unique effects that differentiate produce a dose-related hemostatic defect. This has a
it from other colloids as well as from crystalloids number of mechanisms but is due primarily to a
(Emerson, 1989). Albumin can bind reversibly with reduction in platelet adhesion and aggregation nor-
both anions and cations, which allows it to regulate mally mediated by factor VIIIR:antigen activity (Alex-
the extracellular concentration of various substances ander et al., 1975). Dextran also lowers the levels of all
such as iron, lipids and bilirubin (Emerson, 1989). clotting factors by hemodilution, coats blood vessel
Albumin also has the ability to act as a free radical walls and cellular elements and impairs the elasticity
scavenger and may limit lipid peroxidation (Holt, and tensile strength of fibrin clots (Atik, 1967; Weiss,
Ryall and Campbell, 1984; Wasil et al., 1987; Stocker, 1967; Adelson, Crosby and Roeder, 1955; Karlson et al.,
Glazer and Ames, 1987; Pirisino et al., 1988). These 1967; Muzaffar et al., 1973). Primary hemostasis is
properties may in the future become the major reason affected and clinically the picture mimics von Will-
for choosing albumin as a resuscitative fluid. ebrand’s disease. Bleeding is more likely in patients
Albumin may also play a role in maintaining with pre-existing coagulation abnormalities. To mini-
normal microvascular permeability to protein mole- mize this risk, the volume of dextran infused should
cules. Endothelial cells contain pores through which be limited to 20 ml/kg/d or 1.5 g/kg/d (Atik, 1967).
protein molecules may leave the vascular space. Some 50–70% of infused dextran is excreted by the
Albumin may help regulate the permeability of these kidneys and the rest is slowly metabolized.
pores. Hypoalbuminemia increases capillary permea- Because of their interference with coagulation,
bility and restoring the albumin level reduces permea- dextrans are rarely used in the management of
bility to normal (Harms et al., 1981; Demling et al., severely injured patients.
1982). However, the intravascular albumin level neces- Polygeline (Haemaccel, Hoechst) consists of urea-
sary to maintain normal microvascular permeability is bridged gelatin, molecular weight range 5000–50 000;
not known. mean 35 000, derived from cattle-bone gelatin and
Plasma-protein solutions are heated to 60°C for 10 suspended in saline. It is an effective plasma volume
hours during processing and are free of transmissible expander in critically ill patients (Edwards et al., 1989;
diseases (Rainey and English, 1988). Severe side- Mishler, 1984). The low-molecular-weight gelatin por-
effects, such as hypotension, which is probably due to tion distributes readily through the ECF and produces
300 FLUID, ELECTROLYTE AND METABOLIC MANAGEMENT

Table 16.3 Composition of commonly prescribed electrolyte solutions

Crystalloid solutions Water-equivalent solutions


Isotonic Hartmann‘s (Ringer’s 5% dextrose 4% dextrose
(0.9%) saline lactate) solution in water in 0.18% saline

Glucose (g) – – 50 40
Calories (kcal) – – 205 164
Na+ (mmol/l) 150 131 – 30
K+ (mmol/l) – 5 – –
Cl– (mmol/l) 150 111 – 30
Ca2+ (mmol/l) – 2 – –
Lactate (mmol/l) – 29 – –
Osmolarity (mosmol/l) 300 279 277 282
pH 6.0 5.1 3.5–6.5 3.5–6.5

a plasma volume expansion of only about 70% of the Sodium is the major component of crystalloid fluids
infused volume. Renal excretion accounts for 85% of and the distribution of infused sodium will determine
elimination, fecal excretion for 10% and the remainder the distribution of infused crystalloid fluids. Since
is metabolized to non-essential amino acids. Polyge- sodium is the major solute in the extracellular space
line contains potassium and calcium and this must be and 80% is extravascular (Edelman and Leibman,
remembered when large volumes are infused. Rapid 1959), infused sodium will reside primarily in the
infusions cause histamine release, which usually takes extravascular compartment.
the form of urticaria, but anaphylaxis has been Physiological saline and Hartmann’s solution are
reported (Scheinkestel et al., 1989). Gelatin products the two most frequently used crystalloids and their
are not associated with renal failure or coagulopathy. volume-expanding effects are identical (Cervera and
Moss, 1975). Figure 16.4 shows the effect of colloid
and crystalloid infusions on blood volume in criti-
(b) Crystalloid solutions
cally ill adults. Physiological saline and Hartmann’s
Crystalloids are isotonic mixtures of sodium chloride solution are both freely permeable across the vas-
and other physiologically active solutes (Table 16.3). cular membrane and distribute evenly throughout

Figure 16.4 The influence of colloid and crystalloid infusion on blood volume in critically ill adults. (Source: redrawn from
Shippy, Appel and Shoemaker, 1984.)
BASIC PRINCIPLES 301
the extracellular space. In normal individuals accumulating locally, can reduce flow even further.
approximately 25% of the infused volume remains Thus dextrose infusion during experimental cardio-
within the blood vessels when equilibrium is reached pulmonary resuscitation was associated with a much
(usually within 20–30 minutes). In other words, for higher mortality (Lundy et al., 1987). At this stage, the
every liter of infused crystalloid approximately routine infusion of glucose is not recommended in
750 ml will pass into the interstitium and 250 ml will patients at risk of cerebral insufficiency.
remain in the plasma. Thus interstitial edema is a Other fluids such as 1 liter of half-isotonic 0.45%
necessary consequence of volume resuscitation with saline can be regarded as comprising 500 ml of isotonic
crystalloids and should not, unless excessive, be saline and 500 ml of free water.
interpreted as evidence of fluid overload. However,
the volume of crystalloid used in resuscitating
patients with head injury should be tempered by the (d) Hypertonic saline
possibility of worsening brain edema.
Hypertonic saline will increase the osmolality of the
Crystalloids are well suited for replacing extrac-
ECF, causing a water shift from cells to re-establish
ellular fluid losses (dehydration); they are also used to
osmotic equilibrium. One liter of a 3% saline solution
replace blood loss, based on the notion that acute
contains 510 mmol each of sodium and chloride – a
hemorrhage (or hypovolemia) causes an interstitial
total of 1020 osmotically-active particles. These will be
fluid deficit that must also be replaced. Indeed,
remain in the ECF, thus increasing its number of
crystalloids have proven effective in resuscitating
osmotically active particles by 25% and its volume by
patients following acute hemorrhage and continue to
7%. ECF osmolality will rise to 339 mmol/kg. To re-
be widely used for this purpose (Moss and Gould,
establish osmotic equilibrium, about 1.5 liters of water
1988; Dodge and Glass, 1982; Tranbaugh and Lewis,
will shift from the ICF into the ECF. Thus the 1 liter of
1985; Shackford, 1987; Horton, Landreau and Tuggle,
a 3% saline solution will increase the ECF volume by
1985; Lowe et al., 1979; Virgilio et al., 1979).
about 2.5 liters in total. As in other states of ECF
Crystalloids are non-toxic and do not produce
expansion, three-quarters of this volume (1.9 l) is
anaphylactoid reactions.
distributed to the interstitium and one-quarter (0.6 l)
to the plasma, explaining the propensity of hypertonic
(c) Water-equivalent solutions saline to cause pulmonary and peripheral edema
(Table 16.4).
Solutions of 5% dextrose or 4% dextrose in 0.18% Hypertonic saline is effective in restoring volume
isotonic saline are essentially water rendered isotonic after hemorrhage (Gala et al., 1991), endotoxic shock
to prevent local red-cell lysis at the infusion site. At (Horton and Walker, 1991), trauma (Mattox et al., 1991;
most infusion rates insufficient dextrose is present to Vassar et al., 1991) and burn injury (Griswold et al.
alter blood glucose levels, so the fluid is distributed 1991; Monafo, Halverson and Schechtman, 1984).
evenly throughout total body water. Thus for every Several clinical studies have suggested a better sur-
liter of solution administered, two-thirds will enter the vival after resuscitation with hypertonic solutions
intracellular space and one-third the extracellular compared with isotonic fluids (Mattox et al., 1991;
space. Three-quarters of the extracellular fluid will be Monafo, Halverson and Schechtman, 1984; Vassar et
in the interstitium and one-quarter in the plasma. al., 1991).
Thus about 8% only of the infused volume remains in Hypertonic solutions have a greater volume-
the circulation. For this reason 5% dextrose solution is expanding capacity than equal volumes of isotonic
the fluid of choice for patients with ischemic heart crystalloid solutions. Approximately three times the
disease or congestive cardiac failure, since it does not volume of 0.9% NaCl compared with 3.0% NaCl was
expand the circulation and increase the cardiac work-
load.
Table 16.4 Body compartment expansion according to
type of resuscitation fluid: compartment expansion (ml)
Dextrose and cerebral ischemia per 1000 ml of administered fluid

The observation that carbohydrates promote ischemic Fluid Plasma Interstitium Intracellular
damage in the central nervous system is not new but Colloid 1000 0 0
seems forgotten (Voll and Auer, 1988). The central Isotonic (0.9%) saline 250 750 0
nervous system relies on glucose for much of its 5% dextrose 83 250 667
energy needs. When cerebral ischemia develops, Half isotonic (0.45%) 167 500 133
glucose infusion will promote anaerobic glycolysis saline
3% saline 600 1900 –1500
and produce large quantities of lactic acid, which,
302 FLUID, ELECTROLYTE AND METABOLIC MANAGEMENT

required to restore MAP in a canine hemorrhage 16.4 Fluid resuscitation


model, although the overall amount of sodium admin-
istered did not differ between the two experimental 16.4.1 SHOCK
groups. Because small infusion volumes cause rela-
tively large increases in intravascular fluid volume, When hypovolemia is severe, tissue oxygenation
blood volume restitution is rapid; however this effect becomes impaired and the clinical and metabolic
is short-lived (Gala et al., 1991). features of shock appear. The need for prompt
Hypertonic solutions may reduce edema formation restoration of adequate plasma volume under such
in non-injured tissues (Battistella and Wisner, 1991; circumstances is well recognized (Shoemaker, 1976;
Cross et al., 1988; Wisner, Schuster and Quinn, 1990). Shires and Canizaro, 1973). Hypotension has a mark-
This may be particularly useful in head-injured edly deleterious effect on the outcome from traumatic
patients. Several studies of resuscitation in experi- head injury and indeed from other injuries (Price and
mental brain injury have found that hypertonic Murray, 1972; Siegel et al., 1991). Hence patients with
solutions are more effective at lowering intracranial head injury should receive sufficient intravenous fluid
pressure and decreasing edema in the uninjured resuscitation to avoid hypovolemia and hypotension
cerebral hemisphere than isotonic fluids (Battistella (Buchman, Menker and Lipsett, 1991; Levison and
and Wisner, 1991; Wisner, Schuster and Quinn, 1990). Trunkey, 1982). Few issues in critical care medicine
In a study of trauma victims with concomitant head have prompted as much diversity of opinion as the
injuries, hypertonic saline resuscitation was associated most appropriate asanguineous fluid to achieve this
with increased survival compared with Hartmann’s goal (Virgilio, Smith and Zarins, 1979; Shoemaker and
solution (Vassar et al., 1991). Hauser, 1979; Velanovich, 1989).
At present hypertonic saline, with or without the The primary abnormality in hypovolemic shock is a
addition of colloid, is most commonly used for reduction of plasma volume. Absolute or relative
volume resuscitation after trauma or burns. The plasma volume deficits contribute to the disturbances
efficacy of small volumes and the rapidity of admin- of tissue oxygenation in the other three types of
istration make its use convenient. Other reported circulatory shock, cardiogenic, obstructive and septic,
benefits of hypertonic compared to isotonic solutions and these also may occur in the head-injured patient.
are improved pulmonary gas exchange (Boldt et al., Decreased plasma volume produces a decrease in left
1991), increased renal cortical and gut mucosal blood ventricular end-diastolic volume and stroke volume.
flow (Behrman et al., 1991), decreased bacterial trans- Although sympathetic nervous system activation can
location (Reed et al., 1991a) and the induction of initially maintain cardiac output (CO) by inducing
natriuresis (Gala et al., 1991) and kaliuresis (Battistella tachycardia and arterial pressure by producing vaso-
and Wisner, 1991). constriction, at some point compensatory limits are
Although these properties would be advantageous reached and CO and systemic blood pressure may
in many clinical situations, the use of hypertonic suddenly fall.
solutions is not without problems. In an experimental There is clear agreement that the major goal of
model, increased bleeding, probably due to vasodila- treatment of circulatory shock associated with hypo-
tation, was reported if hypertonic fluid was admin- volemia is rapid restoration of blood volume and
istered within 15 minutes of injury (Krausz et al., tissue oxygenation (Rackow et al., 1983; Weil and
1992). If more than 10% of normal plasma volume is Henning, 1979). The use of asanguineous fluids for
replaced with hypertonic saline, increases in pro- initial resuscitation provides better restoration of
thrombin time and activated partial thromboplastin capillary blood flow than does immediate transfusion.
time and a decrease in platelet aggregation are Moderate hemodilution, for example, to a hemoglobin
observed (Reed et al., 1991b). If large volumes of level of 10–12 g/dl, is well tolerated by most patients,
hypertonic saline are used, potential complications and does not lower ḊO2 if intravascular volume is
include hypernatremia, hyperchloremia, hyperosmo- maintained (Messmer, 1975).
larity, hypokalemia, metabolic acidosis, intracellular
dehydration, cerebral hemorrhage, inhibition of lipol- 16.4.2 CHOICE OF FLUID
ysis, hyperosmolar coma and central pontine myeli-
nolysis (Carvajal and Parks, 1988; Cross et al., 1988; The hemodynamic response to fluid infusion is influ-
Griffel and Kaufman,1992). These complications are enced by the choice of fluid, vascular tone and cardiac
related directly to the solute load infused and compliance. Several studies have directly compared
inversely to the patient’s volume of distribution. the plasma volume-expanding and hemodynamic
When using hypertonic saline serum sodium levels effects of colloids and crystalloids. In these studies, a
(less than 160 mmol/l) and serum osmolarity (less preselected volume of fluid was administered rather
than 320 mosmol/l) require close monitoring. than a volume adjusted on the basis of physiological
FLUID RESUSCITATION 303
response. In stable postoperative patients plasma
volume responses were compared to 1 liter infusions
of 6% Dextran 70, 6% hetastarch, 5% albumin, 3.5%
urea-bridged gelatin (Haemaccel) and physiological
saline. Significant increases in plasma volume occur-
red with all colloids but not with physiological saline.
The greatest increases occurred with Dextran 70
(790 ml) and 6% hetastarch (710 ml; Figure 16.5; Lamke
and Liljedahl, 1976).
In acutely ill postoperative patients Lazrove and
associates evaluated responses to infusion of 500 ml of
5% albumin and 6% hetastarch over 1 hour, using a
prospective, randomized crossover design (Lazrove et
al., 1980). Both solutions significantly increased
plasma volume, CO, ḊO2 and V̇O2. Plasma volume
expansion lasted for 1–5 hours with albumin and for Figure 16.6 Maximal changes in blood volume (ml) versus
at least 3 hours with hetastarch. Similar hemodynamic change in cardiac output (l/min) after infusion of 500 ml of
whole blood or colloids and 1000 ml of RL in critically ill
effects of 5% albumin and 6% hetastarch were repor- patients. (Source: reproduced from Shoemaker, 1976, with
ted in a series of critically ill patients by Puri and permission.)
associates (1983).
The relative effectiveness in critically ill patients of 1
hour infusions of 500 ml of whole blood, 5% albumin,
6% Dextran 70, 10% Dextran 40 and 1 liter of Ringer’s Thus the more potent volume expanding properties
lactate were compared (Shoemaker, 1976). Blood and of colloids compared to crystalloids may permit more
colloid infusion produced significant increases in rapid restoration of hemodynamic stability in hypo-
blood volume, CO, stroke volume, left ventricular tensive critically ill patients (Shoemaker et al., 1981;
stroke work index, central venous pressure (CVP) and Modig, 1986). Our current clinical practice is to use
pulmonary arterial wedge pressure (PAWP), but both. In the resuscitative phase the emphasis is on
improvements after crystalloid infusion were insignif- volume restoration and we use mainly colloid; in the
icant, even though twice the volume was infused maintenance phase we use mainly crystalloid.
(Figure 16.6). In a later study by the same group
involving critically ill patients with ARDS, 100 ml of
16.4.3 MONITORING
25% albumin was compared with 1000 ml of Ringer’s
lactate (Hauser et al., 1980). Plasma volume increased Changing vascular and cardiac compliances in the
by an average of 465 ml with 25% albumin but by only shock patient make it difficult to rely on any single
194 ml with Ringer’s lactate. Significant increases in hemodynamic measurement to define adequate resus-
ḊO2 and V̇O2 occurred only in the albumin group. citation. The adequacy of volume resuscitation is
measured by blood pressure, heart rate and urine
output, which reflect a summation of many different
processes and are only gross estimates of end-organ
perfusion (Lowell et al., 1990). Mean arterial pressure
(MAP), heart rate, central venous pressure (CVP) and
pulmonary arterial wedge pressure (PAWP) do not
reflect intravascular volumes in a linear fashion
(Dawidson et al., 1991; Shippy, Appel and Shoemaker,
1984). CVP may fall during fluid infusion if blood
volume expansion results in decreased autonomic-
nervous-system-induced arteriolar and venous vaso-
constriction (Baek et al., 1975). Changes in left ven-
tricular compliance during fluid resuscitation may
also limit the usefulness of PAWP as a measure of left
ventricular end-diastolic volume (preload; Calvin,
Figure 16.5 Plasma volume expansion after infusion of 1 Driedger and Sibbald, 1981).
liter of (1) Dextran 70, (2) 6% hexastarch, (3) 5% albumin, (4)
modified fluid gelatin and (5) NS in postoperative patients. Even with sophisticated monitoring techniques,
(Source: reproduced from Lamke and Liljedahl, 1976, with such as Swan–Ganz catheterization and oxygen sat-
permission.) uration monitoring, the adequacy of resuscitation and
304 FLUID, ELECTROLYTE AND METABOLIC MANAGEMENT

Table 16.5 Guidelines for fluid challenge utilizing pulmonary arterial diastolic or pulmonary arterial wedge pressure
monitoring: 7/3 rule for fluid challenge. PAWP = pulmonary arterial wedge pressure (mmHg); PADP = pulmonary
arterial diastolic pressure (mmHg). (Source: adapted from Weil and Henning, 1979)

PAWP/PADP Fluid infusion rate

For 10 min before challenge < 12 200 ml  10 min


< 16 100 ml  10 min
≥ 16 50 ml  10min
During 10 min infusion Change > 7 Stop
Change ≥ 3 Continue infusion without interruption
Immediately after 10 min infusion Change > 3, ≤ 7 Wait 10 min
After 10 min wait Change > 3 Stop
Change ≤ 3 Repeat fluid challenge

oxygen delivery to individual tissue beds in vital end animals resuscitated with whole blood and 50 ml/kg
organs cannot be determined. Poole et al. demon- Ringer’s lactate (Figure 16.7; Shires et al., 1964). In a
strated this in an experimental canine shock model. baboon model of hemorrhagic shock, microelectrodes
Despite restoration of MAP and cardiac output (CO), were used to monitor transmembrane potential gra-
cerebral blood flow did not return to preshock levels dients across individual skeletal muscle cells. Sus-
(Poole et al., 1986). tained cell depolarization, a 49% decrease in extrac-
Nevertheless for clinical purposes the hemody- ellular water, and 6% increase in intracellular water
namic response to fluid resuscitation should be increased intracellular sodium and decreased intra-
assessed by using the fluid challenge technique cellular potassium were all recorded, suggesting a
originally described by Weil and Henning (Table 16.5; failure of the ATPase-dependent Na+ –K+ pump
Weil and Henning, 1979). When this is combined with (Shires et al., 1972). Resuscitation with balanced salt
sequential measurements of CO and measurements of solution produced a return of the potential difference
tissue oxygenation (lactate, ḊO2, V̇O2 and mixed to normal together with a decrease in intracellular and
venous oxygen saturation) the risks of under- and an increase in extracellular volume. Similar alterations
over-resuscitation are minimized (Shoemaker and in water distribution were described in patients with
Czer, 1979). circulatory shock and those undergoing major oper-
ative procedures (Shires, 1965; Shires, Williams and
Brown, 1960). These changes also resolved with
16.4.4 CRYSTALLOIDS VERSUS COLLOIDS
infusion of balanced salt solutions.
The basic disagreement that initiated the colloid versus
crystalloid controversy was a conceptual one. Those
favoring crystalloids believe that an extracellular fluid
deficit plays a primary role in the pathophysiology of
hypovolemic shock and therefore repletion of this
deficit is essential. Colloid proponents believe that
decreased blood volume and reduced oxygen trans-
port are the critical pathophysiological factors in
hypovolemic shock; therefore, rapid and complete
repletion of the intravascular compartment is critical
for resuscitation.
In a series of experimental and clinical studies
initiated in the early 1960s, Shires and associates
showed that severe hemorrhagic shock was associated
with a large decrease in extracellular volume, caused
predominantly by intracellular shift of interstitial fluid Figure 16.7 Survival after resuscitation of dogs from
in addition to external losses or shift in the intra- hemorrhagic shock. Each group consisted of 10 dogs, bled
vascular space (transcapillary refill; Shires et al., 1964, into shock by the Wiggers method. The RL group received
1972; Carrico, Canizaro and Shires, 1976). In a canine fluid equivalent to 5% of body weight followed by return of
shed blood. The plasma group received 10 ml/kg of donor
hemorrhagic shock model, animals resuscitated with plasma plus shed blood. The blood group received shed
shed whole blood alone or whole blood plus 10 ml/kg blood alone. (Source: reproduced from Shires et al., 1964, ©
plasma had a significantly higher mortality than 1964, American Medical Association, with permission.)
EFFECTS OF INTRAVENOUS FLUIDS ON THE BRAIN 305
There is however, considerable controversy regard- rapid infusion of up to 2 liters of Ringer’s lactate until
ing the methods used by Shires and colleagues to hemodynamic stability is restored. If the patient
measure extracellular fluid volume (Shoemaker, 1976; remains unstable packed red blood cells are then
Roth, Lax and Malone, 1969). Using different tech- infused. The use of two to four large-bore intravenous
niques and models of hemorrhagic shock, other groups lines can compensate for the much more limited
have found either a minimal decrease in extracellular volume expansion produced by crystalloids. Compli-
volume accountable by the plasma volume loss or else cations of this approach to fluid infusion are minimal
an increase in extracellular volume (Roth, Lax and (Moss et al., 1981).
Malone, 1969; Serkes and Lang, 1966; Moore et al., The choice of fluids in patients with massive
1966). There is similar debate about the changes in bleeding that is difficult to control, or those with pre-
extracellular volume after major surgery, where no existing medical problems, is more controversial. In
significant change was found in patients after chol- these situations fluid administration may result in
ecystectomy and increases were found in patients after fluid overload and subsequent morbidity. The risk of
cardiac surgery (Roth, Lax and Malone, 1969). pulmonary edema has been central to the debate over
Despite this there is considerable evidence, from the relative merits of crystalloids versus colloids. As
experimental models of severe hemorrhagic shock and mentioned earlier, capillary membrane permeability,
from clinical studies of traumatic shock, that subjects capillary hydrostatic pressure, colloid osmotic pres-
can be successfully resuscitated with balanced salt sure (COP) and pulmonary lymph flow are all
solutions and blood (either whole blood or packed red important factors in the development of pulmonary
blood cells) without the need for colloidal solutions edema. Despite claims to the contrary (Virgilio et al.,
(Lowe et al., 1979; Moss et al., 1981; Weaver et al., 1978). 1979), weight gain and systemic edema due to fluid
In the study of Lowe et al. (1979), patients with infusion are not benign problems (Falk, 1991; Lowell et
traumatic abdominal injuries requiring laparotomy al., 1990). Decreased chest wall compliance due to
were resuscitated randomly with either Ringer’s tissue edema may occur following hydration with
lactate or 4% albumin together with washed red blood crystalloid (Brinkmeyer et al., 1981). Peripheral edema,
cells for the entire emergency room and intraoperative particularly in a debilitated patient, can cause
period. Crystalloids alone were used postoperatively. decreased mobility and subsequent skin breakdown.
Clinical criteria (systemic blood pressure, pulse and Tissue oxygenation is decreased when edema is
urine output) were used as the end points for present (Heughan, Niinikoski and Hunt, 1972) and
resuscitation. Both groups of patients were resuscitated wound healing may be impaired (Falk, 1991; Man-
successfully and overall mortality was low (4.3%). galore and Hunt, 1972; Niinikoski, Hengan and Hunt,
There was no difference between the two groups in the 1972).
need for postoperative ventilatory support. Since most Edema of the gastrointestinal tract may result in
patients in this study were not in shock on admission, ileus and intolerance for enteric alimentation (Falk,
the results might not be applicable to patients who are 1991). The potential for bacterial translocation and the
in severe hemorrhagic shock. Moss et al. (1981) repeated development of systemic sepsis and multiple systems
the study in a group of patients with traumatic shock, organ failure may also be increased (Baker et al., 1988;
defined as systolic arterial pressure of 80 mmHg or the Wilmore et al., 1988).
need for five or more red blood cell transfusions before In addition, systemic edema can increase time on
surgery, with similar results. Pulmonary edema did not mechanical ventilation, require diuresis or dialysis
complicate the use of colloid or crystalloid in either of and lengthen the stay in the ICU
these studies. In both studies similar volumes of colloid
and crystalloid were needed for resuscitation. This 16.5 Effects of intravenous fluids on the
surprising finding may be due to the use of clinical
rather than physiological end points for fluid resuscita-
brain
tion. Whether the successful outcome with crystalloid Parenteral fluid therapy, particularly in patients with
resuscitation in these studies is related to replacement head injury, may increase brain swelling, intracranial
of an interstitial fluid deficit or to the limited amount of pressure and neurological dysfunction and this may
the infused solution that remains in the circulation has be reflected in increased mortality and morbidity
not been ascertained. Despite the numerous studies (Vassar et al., 1991; Battistella and Wisner, 1991; Falk,
comparing crystalloids to colloids, none has unequivo- 1991; Fein et al., 1982). However, there is little
cally demonstrated a distinct survival advantage with information available on the relative effects of colloid
either therapy. or crystalloid administration on cerebral edema for-
The current recommendations of the American mation in normal subjects or in patients with brain
College of Surgeons for initial resuscitation of patients injury and decreased intracranial compliance (Zornow
with traumatic injury (hemorrhagic shock) include et al., 1988).
306 FLUID, ELECTROLYTE AND METABOLIC MANAGEMENT

16.5.1 THE EFFECTS OF CRYSTALLOIDS AND resuscitation with hypertonic crystalloid solutions was
COLLOIDS ON THE NORMAL BRAIN consistently associated with a lower ICP than fluid
resuscitation with Ringer’s lactate, physiological saline
Early studies in animals demonstrated brain shrinkage
or 10% Dextran 40 (Prough et al., 1985; Gunnar et al.,
in response to infusion of hypertonic solutions and
1986, 1988). Resuscitation with 6% hetastarch was
expansion in response to hypotonic solutions (Weed
associated with significantly lower ICP than resuscita-
and McKibben, 1919a, b). In general, hypertonic fluids
tion with Ringer’s lactate (Poole et al., 1986).
decrease both brain interstitial and intracellular vol-
The unique structural characteristics of the cerebral
ume and thus decrease ICP, and hypotonic fluids have
capillaries (the blood–brain barrier) limit the effects
the opposite effects. It was not possible to determine
of changes in COP on cerebral water distribution.
from these studies whether the increase in ICP and
The intercellular pores of non-cerebral capillaries are
cerebral water content with hypotonic solutions was
approximately 65 Å in diameter. Ionic solutes such as
due to a decrease in plasma osmolality or a decrease in
sodium and chloride are able to migrate freely
colloid oncotic pressure (COP). To clarify this issue,
between the interstitium and the intravascular space
Zornow, Todd and Moore (1987) examined the acute
and therefore have no influence on water distribu-
effects of changes in plasma osmolality and plasma
tion (Zornow, Todd and Moore, 1987). High-molec-
COP in normal rabbits with an intact blood–brain
ular-weight compounds like albumin and urea-
barrier. In one group COP was decreased without a
bridged gelatin are retained in the intravascular
change in plasma osmolarity, whereas in a second
space and produce an oncotic gradient that tends to
group a hypo-osmolar condition was produced with-
retain water in the capillaries. Because of the small
out change in the COP. Cerebral edema, estimated by
pore size (8 Å) of cerebral capillaries, changes in
changes in brain specific gravity, developed only in the
serum electrolyte content (osmolarity) have signifi-
hypo-osmolar group, suggesting that changes in COP
cant effects on water movement. An osmotic gradient
do not influence intracranial water distribution in the
of 1 mosmol/l produces a hydrostatic gradient of
absence of brain injury (Figure 16.8). These and other
19.3 mmHg. Because there are so few protein mole-
studies demonstrate that when the blood–brain barrier
cules compared with the number of inorganic ions,
is intact, brain volume is strongly influenced by plasma
their effect is minimal (Albright, Latchaw and Robin-
osmotic pressure and is unaffected by changes in
son, 1984). A 10 mm decrease in COP during crys-
plasma oncotic pressure (Hindman et al., 1990). The
talloid resuscitation has the same effect on cerebral
brain, like a red blood cell, swells in a hypotonic
water distribution as a decrease in osmolality of only
solution and contracts in a hypertonic solution.
0.5 mosmol/l. Clearly the influence of changes in
The effects of fluid resuscitation in experimental
osmolarity on cerebral water distribution dwarfs the
hemorrhagic shock on ICP and cerebral edema in
effects of alteration of COP.
animals with intact blood–brain barriers have been
evaluated in several studies (Poole et al., 1986; Prough et
al., 1985; Gunnar et al., 1986). In different models, fluid 16.5.2 THE EFFECTS OF CRYSTALLOIDS AND
COLLOIDS ON THE BRAIN AFTER INJURY

Patients with severe traumatic injuries often require


resuscitation with large volumes of intravenous fluid
to restore hemodynamic stability. When a head injury
is also present, the choice of fluid for resuscitation may
influence the development of brain edema. Animal
studies confirm that damage to the blood–brain
barrier (e.g. by cryogenic injury) increases the permea-
bility of capillaries to both proteins and electrolytes so
that osmotic and oncotic gradients cannot be estab-
lished (Zornow et al., 1988; Weed and McKibben,
1919a). In these circumstances capillary hydrostatic
pressure determines the rate at which edema forms.
When hemorrhagic shock was combined with cryo-
genic brain injury, brain water fell in uninjured brain
after resuscitation with hypertonic saline but not in
Figure 16.8 Acute cerebral effects of changes in plasma areas of brain injury where the blood–brain barrier
osmolality and oncotic pressure in normal rabbits. The
lower the specific gravity, the greater the brain water was damaged (Wisner, Schuster and Quinn, 1990).
content **p < 0.01 versus other groups. (Source: from Zor- Other animal studies evaluating the effects of fluid
now, Todd and Moore, 1987, with permission.) resuscitation in hemorrhagic shock on neurological
METABOLIC RESPONSE TO INJURY 307
function have shown that, although ICP in animals reflecting accelerated net protein breakdown (catabol-
resuscitated with hypertonic saline was significantly ism). The magnitude of these changes is proportional
lower, cerebral perfusion pressure was significantly to the extent of the injury (Weissman, 1990).
higher in those receiving 6% hetastarch compared
with those receiving hypertonic saline or physio-
16.6.2 STARVATION
logical saline. Neurological function was also best in
the hetastarch group, suggesting that restoration of Starvation, i.e. lack of nutrient input, often accom-
cerebral perfusion pressure is a more important goal panies injury, while nutrient utilization is normal at
than change in ICP alone. The vasodilatory effect of the cellular level. During starvation, metabolic adapta-
hypertonic saline may have resulted in a lower mean tion occurs, with the aim of conserving essential
arterial pressure compared with hetastarch. tissues. There is little or no activation of metabolic
These experimental results reaffirm that hypertonic mediators and the system is still able to respond to
saline will shift water from the intracellular to the normal physiological stimuli. Glycogen reserves are
extracellular space. Ringer’s lactate is a hypo-osmolar only small, approximately 200–400 g; thus liver glyco-
solution and 6% hetastarch is iso-osmolar; hence the genolysis is only useful for 24 hours. Decrease in
differing effects of these solutions on ICP can be insulin and increase in glucagon secretion result in
explained by their osmolarities. protein and fat breakdown to provide energy. Amino
The lower ICP after resuscitation with hypertonic acids are converted to pyruvic acid, acetyl CoA and
crystalloid solutions may assist in restoring of cerebral tricarboxylic acid (Krebs) cycle intermediates. Glyc-
blood flow and ḊO2, but there is not universal erol from fat is fed into the glycolytic pathway and
agreement on this issue (Prough et al., 1986). fatty acids form acetyl CoA, some of which enters the
The days of keeping the patient with a head injury Krebs cycle inside the mitochondria; some is con-
‘dry’ are over. There is no good evidence supporting verted to ketones, which are used by skeletal muscle
fluid restriction as a means of limiting cerebral edema and brain. Protein catabolism results in increased
after brain injury (Morse et al., 1985). Dehydration urinary loss of urea, sodium, potassium, magnesium
increases sympathetic stimulation, metabolism and O2 and calcium. As the new energy pathways become
demand (Beckstead et al., 1978). Indeed, the ther- established, protein breakdown decreases and fat
apeutic aim is now to maintain euvolemia and normal becomes the chief energy source, supplying 75–90% of
physiological indices, especially cerebral perfusion the calories. The basal metabolic rate decreases as a
pressure. Animal studies support this approach result of decreasing lean body mass, decreased phys-
(Smith et al., 1982; Ito et al., 1979). ical activity, and decreased thyroxine production.
These changes are summarized in Figure 16.9.
In the absence of sepsis or injury, the starved patient
16.6 Metabolic response to injury can usually be rapidly converted to the anabolic state
by administering moderate amounts of nutritional
16.6.1 OVERVIEW substrate.

Major injury, whether surgical or accidental, evokes


16.6.3 METABOLIC STRESS
predictable metabolic, hormonal and hemodynamic
responses (Buckingham, 1985; Table 16.6). Changes in When surgery, trauma or sepsis occur, a new process is
carbohydrate metabolism include increased endoge- activated (Cerra, 1986). Injured or dead tissue, divid-
nous hepatic glucose production (gluconeogenesis) ing organisms, severe perfusion deficits and resolving
and reduced glucose clearance (insulin resistance), hematomas activate mediator systems that affect end-
which results in hyperglycemia. Fat becomes the organ function, producing the clinical, physiological
major body fuel; therefore lipolysis is increased and and metabolic manifestations of the response to stress.
lipogenesis is retarded. Changes in protein metabo- This process is dynamic, and begins with a lag or ebb
lism are manifested by negative nitrogen balance phase during which there is little metabolic activity.
This is followed by a flow phase during which
metabolic activity increases and peaks, usually on day
3 or 4 after injury. The processes then abate over
Table 16.6 Metabolic response to injury
another 3–4 days, unless a complication ensues. In the
 Altered protein homeostasis event of a new stress, the process reactivates and
 Hypermetabolism reaches a new peak.
 Altered carbohydrate metabolism The resting energy expenditure rises to an amount
 Sodium and water retention that depends on the type and severity of the stress.
 Increased lipolysis
At high levels of stress, 30% or more of the increased
308 FLUID, ELECTROLYTE AND METABOLIC MANAGEMENT

Figure 16.9 Acute non-stressed fasting metabolism. The initial flow of substrate is designed to provide glucose for obligate
and non-obligate glucose users. With time, ketones and fats become the main energy substrates, resulting in daily urinary
nitrogen excretion.

energy expenditure appears to come from the oxida- excretion appears to reflect the stress response,
tion of amino acids, 30–40% from glucose and whereas the persistent nitrogen excretion may be due
30–40% from fat. With activation of metabolic media- to bedrest and disuse in a largely young and mus-
tors by trauma, gluconeogenesis becomes less cular group of patients.
responsive to exogenous nutritional substances (sec- In some instances under new stimuli the process
tion 16.6.4(b)). Thus, administered glucose has pro- reactivates and the state of persistent hypermetabol-
gressively less inhibiting effect on lipolysis, proteol- ism with or without the development of multiple
ysis and gluconeogenesis. Glucose calories, as well as system organ failure ensues (Cerra, 1986). Risk fac-
other calories, which are in excess of the existing tors include severe perfusion shock, severe septic
demands promote lipogenesis, with excess CO2 pro- shock, persistent septic sources, and recurrent septic
duction and hepatic dysfunction. episodes.
Proteolysis increases and amino-acid flux attempts
to meet the demands of energy production and
protein synthesis. Ureagenesis is increased and more 16.6.4 MEDIATORS OF THE RESPONSE TO INJURY
nitrogen is present in the urine. Gluconeogenesis is
increased; peripheral glucose uptake is normal, but (a) The neuroendocrine axis
much of the glucose is recycled as lactate and
alanine. Thus, increased plasma glucose and lactate The mechanisms initiating, regulating and sustaining
levels are a normal part of the response. Ketosis is this response are not all identified. It is known that
relatively depressed. These changes are summarized injured patients have elevated levels of the anti-
in Figure 16.10. insulin hormones: cortisol, glucagon, and the cate-
Malnutrition usually develops rapidly in post- cholamines. Insulin levels are usually elevated, but
traumatic patients, taking days instead of the weeks not sufficiently to prevent the commonly noted
needed in simple starvation. It is difficult to exclude hyperglycemia. Growth hormone, aldosterone and
the effects of bedrest (disuse) on some of the para- ADH are also elevated. These elevations may in part
meters. Indeed, in patients with isolated closed-head be neurally mediated via the hypothalamus (Hume
injuries many reports have described persistent and Egdahl, 1959). Patients with severe head injury
excretion of large amounts of urinary nitrogen up to may develop abnormalities of fluid and electrolyte
weeks after severe head injury. However some stud- balance because of damage to the neuroendocrine
ies indicate that the response abates in 5–7 days axis (e.g. diabetes insipidus following pituitary dam-
unless a complication ensues. The early nitrogen age; Figure 16.11).
METABOLIC RESPONSE TO INJURY 309

Figure 16.10 Summary of moderate to severe stress metabolism. The neuroendocrine and intrinsic mediator systems
modulate the metabolic machinery to mobilize fat and amino acid stores, which provide a continuous supply of fuel to meet
increased energy demands and provide an increased supply of substrate for the production of stress proteins. Increased
urinary nitrogen excretion is one result of the process. BCAA = branched-chain amino acids.

(b) Cytokines lemia), nausea and emesis, and hypoglycemia. Of


these, osmotic stimulation is the most important for
Several non-endocrine factors play important roles in
fine control of ADH secretion and maintenance of
the response to stress. These include interleukin-1 (IL-
water balance. A number of drugs used in critically ill
1), tumor necrosis factor (TNF), IL-2 and gamma-
patients affect ADH secretion. Halothane and mor-
interferon (IFN). The reader is referred to the extensive
phine in high doses increase ADH release, while
review of this subject by Weissmann (1990).
phenytoin and chlorpromazine inhibit ADH release.
Positive pressure ventilation also increases secretion.
16.6.5 EFFECT OF STRESS ON FLUID AND The thirst mechanism assists by controlling fluid
ELECTROLYTE MOVEMENTS intake to some extent in the conscious patient.
The second objective is to keep the total sodium
The changes in electrolyte concentrations and fluid
content of the ECF within normal limits and thus
distribution after trauma and critical illness have been
maintain a normal ECF volume. Since sodium is the
extensively studied. In the normal subject neural,
major cation of the ECF and the body adjusts the water
hormonal, hemodynamic and renal mechanisms func-
around it to maintain a normal sodium concentration,
tion in a highly integrated manner to preserve sodium
the total number of sodium ions in the ECF will
and water homeostasis. There are two main objectives.
determine the ECF volume. Large deviations from
The first is to keep the concentration of sodium in the
normal in the ECF sodium content cause fluctuations
ECF within a very narrow range. Together with its
in the circulating blood volume. Both volume contrac-
associated anions, sodium constitutes more than 90%
tion and expansion can have serious effects on brain
of the total solute in the ECF and controls the
function, particularly in the presence of pre-existing
distribution of water between the cells and the
brain damage (Aubry and Nankin, 1965). Normally,
extracellular space. Large deviations from normal in
ECF sodium is regulated by several closely coordi-
ECF sodium concentration cause cells to shrink or
nated mechanisms that adjust the amount of sodium
swell, which may have serious consequences for brain
lost through the kidneys.
function. Sodium concentration is kept constant by
finely adjusting the water content of the ECF through  The relative fullness of the ECF is sensed by
the secretion of antidiuretic hormone (ADH, vasopres- receptors located in low- (intrathoracic) and high-
sin; Figure 16.12). At least four independent physio- pressure (intra-arterial) areas of the cardiovascular
logical stimuli release ADH into the blood stream: system. When changes in ECF volume occur, renal,
osmotic, hemodynamic (hypotension and/or hypovo- neural and hormonal mechanisms modulate renal
310 FLUID, ELECTROLYTE AND METABOLIC MANAGEMENT

(a)

(b)

Figure 16.11 Hypothalamic control of pituitary secretion. (a) Vasopressin and oxytocin are secreted in the paraventricular
and supraoptic nuclei of the hypothalamus and transported in the axons of the supraoptico-hypophyseal tract to the
posterior lobe of the pituitary gland; these hormones are then released into the circulation. (b) Hypothalamic releasing
hormones are formed in the hypothalamus and are transported to a capillary plexus formed by the superior hypophyseal
arteries around the median eminence and infundibular stem of the hypothalamus; these hormones there enter into the
vascular system and are taken by the portal veins to the adenohypophysis. (Source: reproduced from Brown, David and
Reilly, 1995, with permission.)
METABOLIC RESPONSE TO INJURY 311

Figure 16.12 Control of body fluids. This shows the pituitary gland at the top of the diagram, the heart in the center and
the two kidneys. The factors affecting ACTH, ADH and catecholamine release are shown and the effects on the kidney of
these two hormones are simplified at the bottom of the diagram. The possible results of the kallikrein system, natriuretic
hormone and angiotensin are also included.

sodium excretion such that renal sodium and water occurs. Changes in the resistance of the glomerular
excretion fall when absolute or relative ECF volume afferent or efferent arteriole affect renal sodium
is low and increase when ECF volume is high. The reabsorption independent of changes in renal perfu-
renal mechanisms influencing sodium excretion in sion pressure or oncotic pressure. Vasodilatation
response to changes in ECF volume are listed in (e.g. with low-dose dopamine) increases capillary
Table 16.7.
 Physical factors in the peritubular capillary environ-
ment are one of the principal mechanisms influenc- Table 16.7 Renal mechanisms influencing sodium
ing Na+ excretion in response to changes in ECF excretion in response to changes in ECF volume
volume. When renal perfusion pressure falls or  Physical factors in peritubular capillary environment
oncotic pressure in the peritubular capillary increa- (i.e. hydrostatic and oncotic pressure)
ses, the balance of Starling forces favors sodium and  Changes in resistance of afferent or efferent arteriole
water reabsorption in the proximal tubule. Con-  Renal adrenergic nerve fibers
versely, if the hydrostatic pressure in the peri-  Hormonal mechanisms–renin–angiotensin–aldosterone;
atrial natriuretic peptide (ANP)
tubular capillary is increased, sodium excretion
312 FLUID, ELECTROLYTE AND METABOLIC MANAGEMENT

hydrostatic pressure and renal sodium excretion,  retention of Na+ and water; water is retained
while vasoconstriction (e.g. heart failure) reduces relatively more than Na+, leading to dilutional
hydrostatic pressure and renal sodium excretion. hyponatremia.
Adrenergic nerve fibers innervate the renal tubules
These changes are maximum on the first and second
and may influence renal sodium excretion inde-
day after injury and persist for 4–7 days. They are
pendent of changes in renal hemodynamics. Stim-
more pronounced after severe trauma (Verney, 1954).
ulation enhances sodium reabsorption, while dener-
vation reduces renal Na+ reabsorption.
 The major hormonal mechanism influencing renal 16.7 Fluid therapy in uncomplicated
sodium excretion is the renin–angiotensin–aldos- postoperative and post-traumatic states
terone system. The juxtaglomerular apparatus
senses a reduction in effective blood volume, renin 16.7.1 FLUID AND ELECTROLYTE REQUIREMENTS
is released from the kidney, ultimately increasing
angiotensin II and aldosterone levels. Aldosterone Abnormal fluid losses occur through:
increases distal tubular sodium reabsorption and
 external routes, such as hemorrhage, GI losses,
potassium excretion. Angiotensin II also increases
wounds and burns; CSF otorrhea in young children
systemic vascular resistance and proximal tubular
can sometime be clinically significant;
sodium reabsorption by increasing resistance in the
 internal shifts into the interstitial water or extra-
efferent arteriole.
cellular water;
ANP is produced both in atrial myocytes and within  local edema due to surgical wounds, crushing
the CNS is also important in body sodium regulation injuries, burns, venous thrombosis;
(Needleman and Greenwald, 1986; Samson, 1987). Its  fluid accumulation in the transcellular space, e.g.
actions tend to oppose those of ADH and angiotensin, pleural effusion, ascites and paralytic ileus.
and so it appears to play a role in regulating fluid and
Fluid replacement must take into account basal losses,
electrolyte balance. ANP may play a role in local
additional extrarenal losses and distributional chan-
control of brain volume as well as electrolyte and
ges, based on an understanding of the underlying
water content. ANP is released into the systemic
physiological and metabolic changes.
circulation from the atria, stimulated by increased
Once a stable circulation has been established, water
intravascular volume or by increased atrial pressure
replacement in adult patients with no oral intake
independent of intravascular volume, and under salt-
should equal urinary losses, usually 1000–1500 ml/d,
loading conditions as with infusion of hypertonic
plus insensible water losses, usually 600–800 ml/d,
saline. ANP decreases renal vascular resistance and
minus the volume of endogenous water released by
increases glomerular filtration rate. It induces natriur-
tissue breakdown during semistarvation. In uncom-
esis and diuresis and inhibits the renin–angiotensin–
plicated, afebrile postoperative patients, the latter is
aldosterone axis. ANP also reduces systemic vascular
usually only 100–200 ml but may be as much as
resistance and blood pressure.
1000 ml in a critically ill trauma patient with sepsis.
Fluid balance disturbances are common among
Thus, replacement volume is approximately
patients with head injury and are often multifactorial.
1500–3000 ml/d, depending on weight, age, sex and
Hyponatremia has often been attributed to a cerebral
overall status. Elderly patients with underlying car-
salt-wasting state or to inappropriate secretion of
diac or renal disease usually require less.
antidiuretic hormone (SIADH; as noted earlier, a
Water requirements increase by about 300 ml/d for
moderate degree of ADH secretion is part of the
each 1°C elevation in body temperature. Fluid require-
response to stress). However, hyponatremia may in
ments are greatly increased by shock, multiple
fact often be iatrogenic (Bouzarth et al., 1982; Nelson et
trauma, sepsis, postoperative complications and other
al., 1981). Hypernatremia and hyperosmolarity are
critical illnesses. Under these conditions, criteria for
present in many head-injured patients as a con-
fluid therapy are based primarily on hemodynamic
sequence of the fluid restriction and hyperosmolar
factors and adequate oxygen delivery.
agents used to manage the increases in intracranial
Basal sodium requirements are met by the daily
pressure. Hypothalamic dysfunction can cause diabe-
administration of 500 ml of physiological saline
tes insipidus with hypernatremia.
(75 mmol/d) plus the estimated volume of extrarenal
The major fluid/electrolyte changes immediately
fluid losses (e.g. nasogastric suction, diarrhea, CSF
after acute injury are:
and other fistulae, and pleural or peritoneal drains).
 release of K+ by the cells, leading to transient In an uncomplicated postoperative patient, 40 mmol
relative hyperkalemia, increased K+ excretion and potassium per day is usually sufficient, unless the
subsequent reduction of total body K+; patient is suffering from renal failure or upper GIT
FLUID THERAPY IN UNCOMPLICATED STATES 313
bleeding. Such patients may develop hyperkalemia greater caloric intake is unnecessary in most patients
and thus usually need less K+ replacement. Fluid and may be detrimental in those with severe respira-
therapy of brief duration does not usually require tory failure, particularly if infusion rates of greater than
magnesium, but debilitated patients who do not have 7 mg/kg/min (equivalent to 700 g of dextrose or
renal failure may benefit from 10–15 mmol Mg2+ 2800 kcal/d in a 70 kg subject) are used. Excessive
daily. glucose administration may lead to hepatic steatosis,
Nutritional support plays an integral role in the hyperbilirubinemia and an elevated alkaline
management of metabolic stress. The time to initiate phosphatase.
nutritional support is not completely settled. It is In patients requiring parenteral nutrition, glucose
generally agreed that oxygen transport must be should be used as the major caloric source and infused
restored, stabilized and maintained prior to beginning at a rate no greater than 30 kcal/kg/d (equivalent to
nutritional therapy. In a previously well nourished 500 g of dextrose or 2000 kcal/d in a 70 kg subject).
patient, our practice is to wait a few days, whereas in Each gram of glucose provides approximately 4 kcal.
a severely malnourished patient, therapy should begin
as soon as O2 transport is stabilized.
Lipid
Intravenous lipids are available as emulsions similar
16 7.2 NUTRITIONAL REQUIREMENTS
in particle size to chylomicrons, and are cleared as
Nutrition may be administered via the enteral or such by the body. Chylomicrons are acted upon by
parenteral route. The essential requirements are as plasma lipoprotein lipase to form free fatty acids and
follows. glycerol, which are either oxidized or stored as lipid.
Although there is an increasing trend toward the
routine use of lipid solutions as an energy source,
(a) Calories
these expensive substances have no advantages over
Energy as carbohydrate or fat is given to meet the dextrose. Lipid solutions are only necessary to provide
metabolic needs of the patient (i.e. basal metabolic rate the essential fatty acids linolenic and linoleic. In the
plus additional demands). Sufficient energy of non- absence of essential fatty acids, linoleic and hence
protein origin must be given to prevent infused arachidonic acid levels fall and skin lesions (desqua-
protein being used as an energy source. Energy is matous dermatitis) may result. Approximately 9 kcal
measured in kilocalories (kcal) or kilojoules (kJ) – one is provided for each gram of fat used by the body.
kilocalorie = 4.184 kilojoules. A satisfactory ratio of
non-nitrogen kilocalories per gram of nitrogen is 150:1
(b) Protein
(each 6.25 g protein yields 1 g nitrogen).
Although numerous equations and nomograms The normal adult only needs an oral intake of 20 g of
have been used to estimate metabolic rate and protein to meet daily protein requirements. Protein
nitrogen requirements, none predict accurately which ingested in excess of this amount is broken down to
caloric or nitrogen substrate will be utilized most provide energy and nitrogenous wastes. In the acutely
effectively. Furthermore, substrate utilization varies ill patient, the minimal quantity of protein required is
during the course of an illness. unknown, but it is unlikely to exceed 50 g/d in a
The increase in metabolism during the acute phase patient who is not losing protein externally. The
of injury is short-lived, and nutritional requirements usually quoted requirement is 1–2 g/kg body weight
fall rapidly within 2–5 days as shock, pain and sepsis per day.
resolve and recovery begins. During convalescence Normal adults require 20 L-amino acids for protein
very few hospital patients expend more than synthesis, although only leucine, isoleucine, valine,
1500–2000 kcal (6300–8400 kJ) per day. lysine, threonine, phenylalanine, methionine and tryp-
tophan cannot be synthesized and are therefore
essential. In patients requiring parenteral nutrition,
Glucose
histidine, arginine and cysteine may also be
Glucose and lipid solutions are the standard intra- required.
venous energy substrates. In a normal person a daily For effective nitrogen utilization, a ratio of essential
infusion of 4 mg/kg/min of glucose suppresses gluco- to total amino acids of 2:5 is advisable, and should
neogenesis maximally Approximately one-third of the reflect the amino acid profile of a normal diet. To
infused glucose is oxidized immediately and the reduce the oxidation of amino acids as an energy
remainder is stored as glycogen or lipid. Increasing the substrate, a source of alternate energy should be
infusion rate increases the amount of glucose stored, administered simultaneously in a ratio of 150 kcal for
thus increasing O2 utilization and CO2 production. A each 1 g of nitrogen.
314 FLUID, ELECTROLYTE AND METABOLIC MANAGEMENT

(c) Vitamins and trace elements Table 16.8 Recommended daily allowance (RDA) of
vitamins for adults (MVI-12 contents)
Vitamins are dietary compounds that act as cofactors Vitamin RDA (mg) MVI-12(mg)
for intermediary metabolism. Patients may develop a
deficiency of the water-soluble C and B group vitamins A 1 1
and vitamin K within 4 weeks of parenteral nutrition; D 0.005 0.005
E 10 10
hence they should be administered to all patients K 0.1 –
receiving intravenous nutrition at doses equivalent to B1 (thiamin) 1.5 3
normal daily requirement (Table 16.8). Vitamin defi- B2 (riboflavin) 1.6 3.6
ciency may rarely affect neurological assessment. B6 (pyridoxine) 2.2 4
The essential trace elements are those present in the Niacin 18 40
Folic acid 0.4 0.4
body in amounts of less than 50 g/g tissue, and are B12 0.003 0.005
required in the diet in milligram quantities or less per Biotin 0.05 0.06
day. They are usually defined by their deficiency states, Pantothenic acid 5 15
i.e. a reproducible functional and/or structural abnor- Ascorbic acid 60 100
mality associated with a specific biochemical change MVI-12 does not contain vitamin K
which is prevented or reversed by the element.
With the exception of zinc, the body stores of the
essential trace elements, copper, iodine, iron, man-
ganese, cobalt, selenium, chromium and molybdenum possible. In many patients the gut provides an
are usually adequate for patients requiring parenteral adequate portal of entry for nutritional support at a
nutrition for less than 3 months. A total of 2.5 mg of zinc much reduced risk. By keeping bacteria and toxins
per day will normally maintain the zinc balance, within its lumen, the gut has a role in host defense. In
although, in patients with diarrhea, up to five times this states of metabolic stress, especially coupled with gut
amount may be required. Table 16.9 summarizes starvation, mucosal permeability increases, allowing
parenteral nutrient intake for adults. Trace element movement of enteric bacteria to the regional lymph
deficiency can result in neurological manifestations, nodes and thence to the systemic circulation.
which may interfere with the neurological assessment Proven benefits of enteral feeding include main-
of the head-injured patient and wound healing, as is the tenance of an intestinal ‘barrier’ and better immune
case with some vitamin deficiency syndromes. There is function, improved liver blood flow following shock
little data regarding the optimal amounts of provision and better wound healing. Gut feeding can be started
of trace elements to critically ill patients. Guidelines early even in the presence of a mild ileus, although
will only be established when more accurate methods early feeding does not attenuate the metabolic
are developed for assessing and monitoring trace response to blunt trauma (Eyer et al., 1993). Exercise,
element status in patients in intensive care. even when done passively, is a potent anabolic
Gastric atony is usually present during states of stimulus and retards nitrogen mobilization in criti-
metabolic stress and responds poorly to motility cally ill and injured patients. While vigorous nutri-
enhancers. Gastric feeding is associated with a very tional support appears to affect mortality favorably
high incidence of aspiration and great care must be and speed recovery from head injury, nutrient admin-
taken to avoid this. These problems are alleviated by istration alone cannot override the metabolic response
using sites for feeding distal to the pylorus and in our to head injury (Clifton et al., 1984; Young et al., 1985).
view enteral nutrition should be used whenever Despite adequate caloric administration, severe head

Table 16.9 Usual daily dose range of parenteral nutrient intake for the adult

Nutrient Amount Comment

Water 2500–3500 ml 30–40 ml/kg/d


Calories 2500–3500 30–40 kcal/kg/d
Glucose 600–900 g
Protein equivalent (as amino acids) 60–130 g 1–2 g/kg/d
Na+ 100–120 mmol ↑ with GI losses; ↓ in elderly, CCF
K+ 80–120 mmol ↓ with renal failure
Ca2+ 4–10 mmol
Mg2+ 12–15 mmol ↑ with GI losses
PO4 10–15 mmol ↑ when glucose alone is given; ↓ with renal failure
DISORDERS OF WATER AND ELECTROLYTE BALANCE 315
injury is followed by negative nitrogen balance, Table 16.10 Pathogenesis of hyponatremia (ADH =
weight loss and depression of immune status and of antidiuretic hormone; GI = gastrointestinal)
plasma protein levels.  Shift of water from the cell (e.g. hyperglycemia,
mannitol infusion)
(d) Nutrition and outcome  Shift of sodium into the cell (e.g. potassium loss)
 Retention of water (e.g. syndrome of inappropriate
Since protein-calorie malnutrition can adversely affect ADH secretion, edematous states)
the structure and function of many organs, including  Loss of sodium (e.g. through kidneys, GI tract, skin)
the lungs (Arora and Rochester, 1982), heart (Heyms-
field et al., 1978), gastrointestinal tract (James, 1971)
and musculoskeletal system (Jeejeebhoy, 1986) as well
as wound healing (Haydock and Hill, 1987) and this type of hyponatremia is due to renal salt wasting
immune function, nutritional support might be expec- caused by an unknown humoral substance released
ted to have a beneficial effect on mortality and in response to cerebral injury (‘cerebral salt wasting’);
morbidity following head injury. however, the evidence is not strong and the reported
However, although there are studies that suggest cases most probably represent SIADH (Oh and Car-
that nutritional support is beneficial in supporting roll, 1992). Deterioration in level of consciousness,
general metabolism following head injury (McMahon new focal deficits, myoclonus, seizures or increasing
et al., 1993), a significant relationship between nutrient ICP should alert the physician to the possibility of
intake and outcome has not been proved. hyponatremia and hypo-osmolality in the patient
with a severe brain injury.
16.8 Disorders of water and electrolyte
(a) Isotonic hyponatremia
balance
An apparent decrease in serum sodium concentration
Hypovolemia (from aggressive use of diuretics),
(measured by flame photometry) occurs when the
hyperosmolarity (from osmotic diuretics, hyperglyce-
mass of the non-aqueous components of serum is
mia resulting in glycosuria, and diabetes insipidus)
increased by severe hyperlipidemia or hyperproteine-
and disorders of sodium metabolism (especially the
mia. This is usually referred to as pseudo-hypona-
syndrome of inappropriate ADH secretion – SIADH)
tremia and can be readily identified by finding a
are common problems in head-injured patients. These
normal serum osmolality (Weisberg, 1989).
and other disorders may cause secondary brain injury
through their effects on cerebral perfusion, intra-
(b) Hypertonic hyponatremia
cranial pressure and neuronal function.
In patients with an increased amount of an imper-
16.8.1 HYPONATREMIA meant solute (i.e. one that is unable to cross the blood–
Hyponatremia (plasma Na+ < 135 mmol/l) is seen in
5–12% of patients with severe head injury (Steinbok
and Thompson, 1978). It is especially important, Table 16.11 Common causes of hyponatremia
since the osmotic buffering capacity of the brain may
be impaired following primary brain injuries. ECF Isotonic
 Pseudohyponatremia
osmolality is reduced in all cases of hyponatremia in – Hyperlipidemia
the absence of abnormal solute accumulation, and – Hyperproteinemia
even small decreases in osmolality may cause brain
Hypertonic
edema, impairing brain function and increasing ICP.  Hyperglycemia
The pathogenesis of hyponatremia is summarized in  Mannitol, glycerol or sorbitol excess
Table 16.10. Hypotonic
Hyponatremia may occur in association with dif-  Water retention
ferent levels of body fluid tonicity (Table 16.11) and – Syndrome of inappropriate antidiuretic hormone
so be classified as isotonic, hypertonic or hypotonic, secretion
based on the measured plasma osmolality. Hypona- – Syndrome of inappropriate antidiuresis
– Psychogenic polydipsia
tremia following head injury is hypotonic and is – Transurethral resection of the prostate (TURP)
usually thought to be due to SIADH. The risk of syndrome
hyponatremia seems greater in those with severe  Salt depletion
head injuries, chronic subdural hematoma and basal – Adrenocortical failure
skull fracture (Steinbok and Thompson, 1978; Doczi – Diuretic excess
 Potassium depletion
et al., 1982). There are numerous reports alleging that
316 FLUID, ELECTROLYTE AND METABOLIC MANAGEMENT

brain barrier), such as glucose, mannitol, glycerol or Table 16.12 Criteria for the diagnosis of the syndrome of
sorbitol, osmotic equilibration occurs by water shift- inappropriate secretion of antidiuretic hormome (SIADH)
ing from the ICF to the ECF, thus diluting the ECF  Hypotonic hyponatremia
sodium. In such circumstances, hyponatremia is often  Urine osmolality greater than plasma osmolality
associated with an elevated measured osmolality. For  Urine sodium excretion greater than 20 mmol/l
every 1 mmol/l rise in glucose above a plasma glucose  Normal renal, hepatic, cardiac, pituitary, adrenal and
thyroid function
level of 5.6 mmol/l, the plasma sodium decreases by
 Absence of hypotension, hypovolemia, edema and
0.288 mmol/l (Katz, 1973; Crandall, 1974; Jenkins and drugs
Larmore, 1974; Robin et al., 1979). The corrected  Correction by water restriction
sodium value is derived from the formula:
Corrected Na value = measured plasma Na+
(plasma glucose value – SIADH is a form of dilutional hyponatremia; ECF
5.6)  0.288. volume is usually increased by 3–4 liters and periph-
eral edema does not occur. Because of the expanded
(c) Hypotonic hyponatremia ECF volume, glomerular filtration rate is increased
and the renin–angiotensin–aldosterone mechanism is
Hyponatremia is almost always caused by an excess of
suppressed, resulting in a decrease in the renal
total body water. This may be due to excessive
absorption of sodium.
infusion of hypotonic or water-generating intravenous
fluids such as 5% dextrose, 1.5% glycine or 0.45%
saline, particularly when administered rapidly or in Syndrome of inappropriate antidiuresis (SIAD)
the presence of high circulating ADH levels. Hypona-
This syndrome is defined as hyponatremia due either
tremia may rarely be due to loss of exchangeable
to increased ADH secretion initiated by stimulation of
sodium or potassium (Fuisz, 1963).
high- or low-pressure baroreceptors, or to a reduction
Since hyponatremia may be associated with an
in non-ADH renal water excretion mechanisms
alteration in both total body water and total body
(Robertson, 1989). The causes of SIAD are listed in
solute the ECF volume may be increased (hyper-
Table 16.13.
volemia), decreased (hypovolemia) or unchanged
(isovolemia; Humes, 1986).
In health, a fluid intake of up to 15–20 liters may be (d) Clinical features of hyponatremia
tolerated before water is retained. However, less water
Hypotonic hyponatremia is almost always caused by
is needed to produce hyponatremia in individuals in
excess total body water, and the symptoms are due
whom there is also an increase in non-osmotic
stimulation of ADH by hypovolemia, hypotension,
pain, nausea or postoperative stress (Hayes, 1968;
Chung et al.,1986; Sinnatamby et al., 1974), or a Table 16.13 Causes of syndrome of inappropriate
antidiuresis
reduction in urinary osmotic excretion, as in patients
with a reduced urea production, or a reduction in Baroreceptor-mediated
renal capacity to respond to ADH.  Hypovolemia
The proposal that disease may cause a ‘sick cell’ – Addison’s disease
syndrome (Flear and Singh, 1973; Flear, Gill and Burn, – Hepatic failure
– Nephrotic syndrome
1981) in which ECF sodium leaks into cells and – Drugs: frusemide, thiazides
produces hyponatremia is a misconception (Leaf,  Cardiac failure
1974; Bichet and Schrier, 1982).
Cortical influences
Loss of potassium may cause hyponatremia as  Pain, anxiety and nausea
sodium shifts into the cell in exchange for K+. – Postoperative
– Trauma
Syndrome of inappropriate ADH secretion (SIADH) Drugs
 Narcotics, barbiturates, chlorpropamide, phenothiazines,
This syndrome is defined as hyponatremia due to an carbamazepine, tricyclics, nicotine derivatives, clofibrate,
elevated level of ADH inappropriate for the prevailing vincristine, vinblastine, cyclophosphamide
osmotic or volume stimuli (Robinson, 1985; Bartter Reduction in renal response to antidiuretic hormone
and Schwartz, 1967). ADH secretion from the neu-  Renal failure
rohypophysis is no longer under normal regulatory  Hypothyroidism
 Addison’s disease
influences. The criteria for the diagnosis of SIADH are
 Drugs: indomethacin, frusemide, thiazides
listed in Table 16.12.
DISORDERS OF WATER AND ELECTROLYTE BALANCE 317
to the cerebral water excess (cerebral edema; Waste- difference between the measured osmolality and the
rlain and Posner, 1968). Normally the brain partially calculated osmolality (2  Na+ + glucose + urea, where
adapts to the hypo-osmolality within 24 hours, plasma sodium, glucose and urea are measured in
reducing the cerebral water excess by losing or millimoles per liter; Worthley, Guerin and Pain, 1987).
inactivating intracellular osmotically active solutes When there is an excess of total body water, urinary
(Arieff, 1984, 1985; Arieff and Guisado, 1976; Arieff, sodium is usually greater than 40 mmol/l and plasma
Liach and Massry, 1976). urea is low. In the rare case of hyponatremia due to
If the patient develops hyponatremia of 125 mmol/l salt depletion, urinary sodium is usually less than
(osmolality 260 mosmol/kg) acutely, that is within 3 20 mmol/l and the plasma urea and uric acid are often
days, then symptoms of cerebral edema usually occur. high (Humes, 1986). In patients with head injuries
These include headache, anorexia, nausea, weakness, hyponatremia will almost always be associated with a
lethargy, confusion, disorientation, blurred vision, urinary osmolality of greater than 100 mosmol/kg.
muscle cramps, coma and seizures (Arieff, 1984; The presumptive diagnosis of SIADH can be con-
Arieff, Liach and Massry, 1976; Plum and Posner, firmed if simply restricting fluids results in a reduc-
1980). tion of urinary Na+ losses and correction of hypona-
Chronic hyponatremia, on the other hand, with tremia. The clinical picture most often confused with
plasma sodium values well below 125 mmol/l, may be SIADH is a patient with isotonic fluid loss, usually
well tolerated because of cerebral osmotic compensa- due to diuretics, who is treated with hypotonic fluid
tion (Arieff, 1984). Thus the mortality associated with replacement. In this situation, the body preserves
hyponatremia is usually related to its rate of develop- volume at the expense of tonicity – a form of
ment rather than its severity. Chronic hyponatremia hypotonic dehydration. It should be stressed that the
has a mortality of less than 10% (Sterns, 1987), absence of an increase in ECF volume excludes a
whereas a mortality up to 50% has been reported with diagnosis of SIADH.
acute hyponatremia (Robertson, 1989). Chronic hyponatremia accompanies other diseases
and it is important to differentiate SIADH from
sodium depletion due to prolonged vomiting, adrenal
(e) Diagnosis of hyponatremia
insufficiency or hypopituitarism.
The evaluation of hyponatremia with hypo-osmolality
begins with a clinical assessment of volume status and
measurement of urinary indices (Table 16.14). How- (f) Treatment of hyponatremia
ever, volume status can be difficult to assess clinically
Treatment aims to remove excess water and treat of
in a critically ill patient. ECF volume may be affected
the underlying cause.
by blood loss, the amount and type of fluid admin-
istered and the use of diuretics. Urinary Na+ measure-
ments are affected by the use of osmotic and non- Chronic hyponatremia (> 3 days’ duration)
osmotic diuretics. Figure 16.13 summarizes our
diagnostic approach to hyponatremia. If the patient is asymptomatic and hyponatremia has
Acute hyponatremia is usually due to excess water, been present for more than 3 days, fluid restriction
as in dextrose solutions administered inappropriately and removal of any precipitating factor is often all that
to patients in the postoperative period. Diagnosis is is required (Cluitmans and Meinders, 1990). Fluid
based on measurement of plasma osmolality, osmolar should be restricted to less than 500 ml/d. The rate of
gap and urinary sodium. The osmolar gap is the correction should be no greater than 12 mmol/l/d
(0.5 mmol/l/h) and is continued only until the plasma
sodium is 130 mmol/l (Laureno and Karp, 1988;
Table 16.14 Extracellular volume (ECV) and urinary Sterns, Riggs and Schochet, 1986; Plum and Posner,
sodium (uNa+) in hyponatremia and hypo-osmolality 1980; Cluitmans and Meinders, 1990; Norenberg and
Papendick, 1984). If fluid deprivation is difficult to
uNa+ sustain in patients with SIADH or SIAD then patients
Diagnosis ECV (mmol/l)
with hyponatremia and chronic CCF may benefit from
Dehydration Decreased < 10 an ACE inhibitor added to frusemide (furosamide).
Congestive heart failure, cirrhosis Increased < 10 This will inhibit the stimulation of thirst and ADH
release by angiotensin II (Dzau and Hollenberg, 1984;
Renal disease, adrenal insufficiency, Decreased > 25
Packer, Medina and Yushak, 1984). However, in these
diuretics
patients a direct ADH inhibitor such as phenytoin may
SIADH Normal or > 25 be of greater value (Mulinari et al., 1990). This has been
increased
used to reduce ADH release from the hypophysis in
318 FLUID, ELECTROLYTE AND METABOLIC MANAGEMENT

Figure 16.13 Diagnostic approach to hyponatremia. Arrows indicate the direction of change; single and double arrows
indicate the magnitude of change. Iso = isotonic; N = normal; V = variable. In hypertonic hyponatremia due to
hyperglycemia, the corrected plasma Na+ = plasma Na+ + (plasma glucose – 10)/3 mmol/l.

patients with SIADH due to CNS disorders including onset may be from one to several days after the
head injury (Martinez-Maldonado, 1980). hyponatremia has been corrected and may require
MRI to confirm the diagnosis (Brunner et al., 1990).
While an association with CPM and correction of
Complications of treatment of chronic hyponatremia
hyponatremia has been reported, the relationship
In chronic hyponatremia the brain is less able to between CPM and the rapidity of correction of
tolerate rapid correction and may develop central hyponatremia has not been firmly established in
pontine and extrapontine myelinolysis if hypertonic clinical practice. In 170 cases of CPM, hyponatremia
saline is used. The lesions of central pontine myelinol- occurred in only 28% of patients, most of whom were
ysis (CPM) are caused by the destruction of myelin corrected slowly (Ayus, Krothapalli and Arieff, 1985),
sheaths in the center of the basilar portion of the pons an observation that has been confirmed by others
and may extend from the midbrain to the lower pons. (Tormey, 1990). Nevertheless, there is persuasive
The clinical features range from coma, flaccid quad- experimental evidence for the association between
riplegia, facial weakness and pseudobulbar palsy to rapidity of correction of chronic hyponatremia and
minor behavioral changes without focal findings. The CPM (Norenberg and Papendick, 1984).
DISORDERS OF WATER AND ELECTROLYTE BALANCE 319
Acute hyponatremia (< 3 days duration) Complications of treatment of acute hyponatremia
In this condition, the rate of reduction of plasma The complications reported with the use of hypertonic
sodium is 0.5 mmol/l/h or greater and/or the hypo- saline include congestive cardiac failure, cerebral
natremia is of less than 3 days duration (Cluitmans hemorrhage – intracerebral and subdural, presumed
and Meinders, 1990). It is often associated with due to tearing of bridging veins – and CPM. To reduce
inappropriate intravenous fluid administration and the incidence of congestive cardiac failure, CVP or
with the postoperative period (Arieff, 1986). The total PAWP should be monitored throughout saline admin-
water excess in adults ranges from 5–10 liters. istration. Cerebral hemorrhage will only occur if there
The rate of correction of acute hyponatremia should is an inappropriate administration of hypertonic
be no greater than 2 mmol/l/h of sodium until the saline in normonatremic states (Finberg, Luttrell and
plasma level has increased to 120 mmol/l or by a Redd, 1959).
maximum of 20 mmol/l during the first 24 hours
(Arieff and Guisado, 1976; Ayus, Krothapalli and
Arieff, 1987, 1988). 16.8.2 HYPERNATREMIA
This is achieved initially by intravenous administra-
Hypernatremia is defined as a plasma sodium greater
tion of hypertonic saline given at 50–70 mmol/h. It is
than 145 mmol/l. It is always associated with hyper-
important to note that the purpose of using hypertonic
osmolality and may be caused by water depletion,
saline is not to correct a saline deficit, since there is in
excessive administration of sodium salts or both
fact not a total Na+ deficiency, but rather the hyper-
(Table 16.16).
tonic saline draws water into the intravascular com-
In water-depleted states, the thirst mechanism
partment and reduces brain edema. A spontaneous or
normally stimulates a conscious individual to drink
frusemide-induced diuresis (e.g. by 20–40 mg intra-
water. Thus water loss only produces hypernatremia if
venously) is then required to excrete the water excess.
the patient is unconscious or otherwise physically
When the plasma sodium has increased to 120 mmol/
unable to drink. Excessive administration of sodium
l, precautions should be taken to prevent the plasma
salts is a rare cause of hypernatremia and usually only
sodium rising to greater than 130 mmol/l over the
occurs through therapeutic misadventure.
next 24 hours. If the hyponatremia presents with
Hypernatremia is the usual cause of hyperosmolar
convulsions, then urgent correction of the cerebral
states in neurosurgical patients, although marked
edema using 250 mmol of hypertonic sodium chloride
over 10 minutes (equivalent to 500 ml of 20% man-
nitol), has been used. This will immediately elevate Table 16.16 Causes of hypernatremia
the plasma sodium in adults by about 7 mmol/l (Table
16 15; Worthley and Thomas, 1986). Water depletion
Extrarenal loss
 Exposure
Table 16.15 Treatment of severe hyponatremia (ACE =  GIT losses
angiotensin-converting enzyme; ADH = antidiuretic Renal loss
hormone)  Osmotic diuresis
– Urea, mannitol, glycosuria
Acute hyponatremia  Diabetes insipidus
 Fluid restrict to 500 ml/d or less – Central (neurogenic)
 Hypertonic saline (50–70 mmol/h) Post-traumatic, postoperative
 Diuresis of 160 ml/h or greater Metastatic tumors, craniopharyngioma, pinealoma,
 Rate of correction cysts
– First day: no greater than 20 mmol/l/d (2 mmol/l/h) Meningitis, encephalitis
until plasma sodium 120 mmol/l Fat embolism
– Second day: attempt to keep plasma sodium between Granulomas (TB sarcoid)
130 and 135 mmol/l Idiopathic
 Seizures: 100–250 mmol hypertonic saline over 10 min – Nephrogenic
Congenital
Chronic hyponatremia
Hypercalcemia
 Fluid restrict to 500 ml/d or less
Hypokalemia
 Rate of correction:
Drugs: lithium, amphotericin B
– No greater than 12 mmol/l/d (0.5 mmol/l/h) until
Renal diseases: chronic pyelonephritis; medullary
the plasma sodium is 130 mmol/l
sponge kidney; polycystic kidney; analgesic
 Drugs
nephropathy; postobstructive uropathy; multiple
– Lithium, demeclocycline
myeloma; amyloid; sarcoid
– Phenytoin
– ACE inhibitor Salt gain
– ADH inhibitor  Hypertonic saline or sodium bicarbonate
320 FLUID, ELECTROLYTE AND METABOLIC MANAGEMENT

hyperglycemia may contribute to hyperosmolality in solute-free water in the face of appropriate stimuli.
states of insulin resistance, such as in sepsis, or as a Head injury is a common cause of central DI. A very
result of inadequate insulin administration with par- early onset is characteristic of major hypothalamic
enteral nutrition. damage and is associated with a high mortality (Seckl,
Dunger and Lightman, 1987).
Head-injured patients with fractures involving the
(a) Clinical features
base of the skull and sella turcica appear to be at
Hypernatremia is usually symptomatic if plasma increased risk of DI (Crompton, 1971; Edwards and
sodium is 155–160 mmol/l or greater (equivalent to an Clark, 1986). Diabetes insipidus is common in
osmolality of 330 mosmol/kg or greater). This equates patients prior to brain death. Traumatic DI is thought
to a water depletion of 6 liters or greater in a 70 kg to be caused by a shearing injury to the pituitary
man (Arieff, 1984). Clinical features include pyrexia, stalk. The time of onset is variable, but it usually
restlessness, weakness, paralysis, irritability, drowsi- appears after 5–10 days (Kern and Meislin, 1984). In
ness, lethargy, confusion, tremor, hyper-reflexia, seiz- most cases of post-traumatic DI the onset is charac-
ures and coma (Ross and Christie, 1969). The signs of terized by polyuria, hypernatremia and plasma
hypernatremia are due mainly to increased effective hyperosmolality, sometimes as early as 12–24 hours
osmolality, which reduces brain cell volume (Oh and after injury. If the damage is limited to the pituitary
Carroll, 1992). As already noted, hypernatremia is or lower pituitary stalk, the DI may only be tran-
much better tolerated if it develops slowly. Increase of sient, and this is usually the case. High stalk lesions
the brain volume may be accomplished by intra- or injury to the hypothalamus usually cause perma-
cellular shift of sodium, potassium and chloride, and nent DI. Since most patients in ICU are unable to
the accumulation of organic solutes, mainly taurine control their own water intake, hemodynamic insta-
and other amino acids (Lohr, McReynolds and Gri- bility may occur with untreated DI.
maldi, 1988). Normalizing brain volume in chronic Nephrogenic DI is often mild because the patient
hypernatremia therefore requires an increase in the can usually produce an isotonic urine; thus the
total solute content of the brain. A sudden lowering of diuresis is limited to 3–5 l/d.
the osmolality of the ECF may cause a shift of water
into the brain cells with resultant cerebral edema.
(c) Diagnosis
Every patient with hypernatremia may be considered
(b) Diabetes insipidus
to have an inadequate water intake, either absolute or
Diabetes insipidus (DI) is characterized by polyuria relative. Excessive sodium administration as the cause
(3–15 l/d) and polydipsia due to the complete or of hypernatremia is usually obvious from the history.
partial failure of ADH secretion (central or neurogenic Whether hypernatremia is due to insufficient water
DI), or decrease in the renal response to ADH intake or to excessive water loss can be determined by
(nephrogenic DI; Table 16.16). Both situations result in measurements of urine osmolality (Figure 16.14).
an inability to concentrate urine and to conserve Normal concentration of urine (urine osmolality

Figure 16.14 Differential diagnosis of hypernatremia. Posm = plasma osmolality; DI = diabetes insipidus.
DISORDERS OF WATER AND ELECTROLYTE BALANCE 321
> 700 mosmol/l) suggests that the main problem is Table 16.17 Treatment of diabetes insipidus (DI) in the
insufficient water intake, with or without excessive critically ill ICU patient
extrarenal water loss. Urine osmolality between Remarks
700 mosmol/l and the osmolality of plasma suggests
partial central DI, osmotic diuresis, diuretic therapy, 1 Confirm diagnosis Altered consious state or inability
acquired (partial) nephrogenic DI or renal failure. A to detect thirst or take oral fluids.
Hypertonicity (> 295 mosmol/kg),
high (> 30 ml/kg/h) urine volume may be the first sign hypernatremia, inappropriately
of diabetes insipidus, but is also seen in osmotic dilute urine. DI may be transient
diuresis. A urine osmolality below that of plasma or permanent; permanent form
suggests either complete central DI or nephrogenic DI. seen in one-third of patients with
Laboratory investigations show hypernatremia, per- trauma or neurosurgery. Presence
of associated anterior pituitary
sistently hypotonic urine and low urine osmolality, hypofunction not uncommon in
usually less than 200 mosmol/l. In milder cases urine patients with severe head trauma.
osmolality may not be below that of plasma, and may 2 Replace free water Free H2O deficit (liters) =
be 300–600 mosmol/l. Patients with central DI are deficit 0.6  (body weight [kg])  ([Na+/
unable to increase urinary osmolality during fluid 140] – 1)
restriction, but remain sensitive to exogenous ADH. Parenteral replacement of half of
Patients with partial DI have urinary volumes much deficit immediately; rest over
24–36 hours. Use 5% dextrose, not
less than those with complete DI. Other causes of saline solutions
polyuria which should be considered in the head-
injured patient include osmotic diuretics (mannitol, 3 Give maintenance Labor-intensive; inaccurate fluid
fluids and replace balance may occur with high
iodinated contrast-medium), severe hyperglycemia urine output volumes of urine
and fluid overload. In contrast to DI, a solute diuresis hourly with
is usually accompanied by a higher urinary osmolality dextrose solutions
(between 250 and 320 mosmol/l; Shucart and Jackson, 4 Daily weight Not always available in adult ICU
1976).
5 Close monitoring
of electrolytes and
(d) Treatment of hypernatremia fluid balance
6 Aqueous If urine output remains excessive
Hypotension due to a reduction in intravascular fluid vasopressin (AVP) (> 200–250 ml/h) in the absence of
volume should be treated by isotonic saline infusions diuretics or, if maintenance of fluid
to replace the intravascular volume and improve balance is difficult or
tissue perfusion before hypotonic solutions are used. hyperosmolality is present give
5–10 U subcutaneously or i.m.
Pure water depletion is treated by water administra-
tion. (In a conscious patient with normal gastro- 7 Desmopressin More recent regimen. Synthetic
intestinal function, oral ingestion of water is encour- (DDAVP) for analog of AVP with longer half-life
patients with and fewer vasoconstrictive effects.
aged.) If intravenous fluid is required, 5% dextrose or severe DI Give 1–2 µg subcutaneously or i.v.
hypotonic saline solutions (e.g. 0.45% saline) are often every 12–24 h or by nasal
used, as sterile water through a peripheral vein causes insufflation of 5–20 µg every 12 h.
hemolysis (Krumbhaar, 1914). In rare cases, where Watch for water intoxication
saline and dextrose solutions are deemed inadvisable; (hyponatremia and elevated
urinary osmolality) during
for example, in a patient who is volume-depleted and therapy; withhold DDAVP
severely hypernatremic, central venous administra- periodically to document
tion of sterile water can be used without causing persistence of DI.
hemolysis (Worthley, 1986).
The rate of increase in osmolality should be no
greater than 2 mosmol/kg/h (Arieff, 1984; Arieff and
Guisado, 1976). Hypernatremia due to excess sodium lower ICP. The aim is not to induce hypovolemia,
is treated by water and diuretic administration which may be detrimental, but rather to maintain a
(Addleman, Pollard and Grossman, 1985). The man- normovolemic state while increasing serum sodium,
agement of DI depends on the cause (Table 16.17). osmolality and tonicity, thus decreasing the extrac-
ellular fluid volume within the brain. A serum
sodium level of 145–155 mmol/l and serum osmolal-
(e) Hyperosmolar therapy
ity of 300–320 mosmol/l are often generated, and
Intravenous 20% mannitol (1098 mosmol/l) a six- excessive dehydration may precipitate prerenal renal
carbon sugar similar to glucose, is often used to failure. Fluids are administered to maintain CVP
322 FLUID, ELECTROLYTE AND METABOLIC MANAGEMENT

between 5 and 10 mmHg. In young, otherwise fit lesion. Hypertonic saline has the additional benefit of
head-injured patients, pulmonary artery catheteriza- rapid small volume resuscitation of associated hemor-
tion is rarely indicated. rhagic shock (section 16.5.2).
Mannitol is administered as an intermittent intra-
venous bolus (0.25–0.5 g/kg body weight) up to
16.8.3 HYPOKALEMIA
4-hourly. Where the blood–brain barrier is intact,
mannitol remains within the intravascular and extrac- Hypokalemia is defined as a serum potassium of less
ellular spaces (Sutin, Ruskin and Kaufman, 1992). It than 3.5 mmol/l or plasma potassium less than
has several effects, including an immediate increase in 3.0 mmol/l. It may be due to a reduction in total
circulating blood volume and arterial blood pressure body potassium, caused by a decreased oral intake,
(Wise and Chater, 1962; Roberts et al., 1987). It also increased renal or gastrointestinal loss, or to a com-
reduces blood viscosity by hemodilution (Burke et al. , partmental shift of potassium from the ECF to the
1981; Muizelaar et al., 1983) and by increasing red ICF (Tannen, 1986). In contrast to Na+ and C1–,
blood cell deformability. It causes osmotic dehydra- which are predominantly in the ECF and regulated
tion of the brain, which reduces the volume of by the kidneys, K+ is predominantly in the ICF; its
extracellular free water (Roberts et al., 1987), thus movements are primarily determined by nutritional
decreasing brain volume and ICP as well as causing a and metabolic factors (e.g. pH) and are mediated by
urinary osmotic diuresis. Repeated administration of neural and hormonal influences (Schultze and Nis-
mannitol may result in a systemic hyperosmolar state senson, 1980).
and dehydration, which should be avoided since Hypokalemia may be caused by osmotic diuretics,
cerebral perfusion may decrease and renal failure may diabetes insipidus, steroids, catabolic losses in
occur. Repeated administration of mannitol may result trauma and secondary hyperaldosteronism (com-
in its eventual movement through the blood–brain monly caused by hypovolemia or cardiac failure).
barrier into the extracellular space, where the Excessive renal K+ loss also occurs in the presence of
increased oncotic pressure may retain free water and alkalemia and hypomagnesemia. Gastrointestinal K+
worsen edema. Thus mannitol use should be restricted losses may be due to vomiting, nasogastric suction or
to the minimum amount needed to control ICP and diarrhea. Intracellular shift of K+ occurs in metabolic
CPP. Our practice is to measure serum osmolality or respiratory alkalosis, exogenous insulin admin-
daily and cease mannitol if serum osmolality exceeds istration and endogenous or exogenous catechola-
320 mosmol/l. mine administration.
Mannitol induces diuresis primarily by elevating
the osmotic pressure of the glomerular filtrate to such
(a) Clinical features
an extent that the tubular reabsorption of water and
solutes is hindered. It promotes the excretion of The clinical features of hypokalemia are listed in
sodium; however, proportionately more water than Table 16.18. These include ventricular and supraven-
sodium is excreted. Excretion of potassium, chloride, tricular arrhythmias, ileus, enhancement of digitalis
calcium, phosphorus, magnesium, urea and uric acid toxicity and hypokalemic nephropathy; nephropathy
are also increased. Mannitol is only very slightly occurs if hypokalemia is severe and prolonged and
metabolized, if at all, to glycogen in the liver. It is results in impaired renal water conservation and
freely filtered by the glomeruli, with less than 10% polyuria. If K+ depletion is severe neuromuscular
tubular reabsorption, and is not secreted by tubular manifestations include weakness, hyporeflexia and
cells. The elimination half life in adults is about 100 even paralysis. The muscular weakness can interfere
minutes. Approximately 80% of a 100 g dose is with attempts to wean patients from ventilatory
excreted unchanged in the urine within 3 hours. In the support in the ICU.
presence of renal disease in which glomerular function
is impaired or in conditions that impair small vessel
(b) Treatment
circulation, such as congestive cardiac failure, cir-
rhosis with ascites, shock or dehydration, mannitol Treatment of hypokalemia is usually commenced
clearance is lower than normal. Mannitol does not when plasma K+ is less than 3.0 mmol/l. While there is
cross the intact blood–brain barrier unless very high normally a linear relationship between serum K+ and
concentrations are present in the plasma or the patient total body K+ (Sterns et al., 1981), acid–base dis-
has acidosis. turbances and other causes of compartmental K+ shift
Hypertonic saline (7.5%) has also been used to make estimates of total body K+ difficult in severely ill
lower ICP in animal models of head injury (Freshman patients. Thus K+ therapy must be monitored by
et al., 1993). It was as effective as 20% mannitol in frequent estimations of serum K+. Serum magnesium
treating elevated ICP caused by a space-occupying levels should be measured in patients with refractory
DISORDERS OF WATER AND ELECTROLYTE BALANCE 323
Table 16.18 Clinical features of hypokalemia compartment shift, or may be artifactual, due to
hemolysis after collection.
Cardiac
 Predisposition to digoxin toxicity Hyperkalemia is uncommon in patients with an
 Ventricular and atrial tachycardias isolated head injury unless overzealous use of osmotic
 Torsades de pointes diuretics has caused renal failure. Apparent hyper-
 ECG changes: PR prolongation, T-wave inversion, kalemia may be seen with hemolysis or acidosis. In
prominent U waves
patients with additional trauma, rhabdomyolysis and
Skeletal muscle massive transfusion of old blood may result in
 Weakness, hypotonicity
hyperkalemia.
 Ascending paralysis, ventilatory failure
 Cramps, rhabdomyolysis

Gastointesinal (a) Hypoaldosteronism (Barratt, 1981)


 Constipation, ileus

Central nervous system Hyporeninemic hypoaldosteronism


 Depression, coma
This usually occurs in patients who have non-insulin-
Renal dependent diabetes mellitus and mild renal failure,
 Nephrogenic diabetes insipidus (polyuria, polydipsia)
 Metabolic alkalosis
and may be provoked by cyclo-oxygenase inhibitors.
 Interstitial nephritis The patient is hyperkalemic with metabolic acidosis
 Predisposition to urinary tract infection (Type 4 renal tubular acidosis, RTA) and has an
Endocrine elevated plasma creatinine and urea nitrogen. The
 Glucose intolerance etiology is not known, although hyporeninemia, pros-
tacyclin deficiency and excess atrial natriuretic hor-
mone have all been implicated. Non-steroidal anti-
inflammatory drugs (NSAIDs), calcium-channel
hypokalemia, since both electrolytes may be disturbed blockers and beta-adrenergic blockers should not be
together and hypokalemia may be difficult to correct administered as they will contribute to the hypor-
without concomitant correction of hypomagnesemia eninemic state (Williams, 1986).
(Tannen, 1986). Hypokalemia protects against hypo-
calcemic tetany; thus hypocalcemia should be cor-
Primary hypoaldosteronism
rected before or together with the correction of
hypokalemia. This is characterized by normal or high renin levels
When hypokalemia is associated with metabolic and is due to an enzymatic defect in the aldosterone
alkalosis the standard treatment is intravenous or oral pathway.
potassium chloride. If the patient has renal tubular
acidosis and hypokalemia, then potassium acetate is
Pseudo-hypoaldosteronism
given. If phosphate depletion is also present, potas-
sium phosphate may be administered. Phosphate can This is characterized by normal or high levels of
only be infused slowly (at a maximum rate of aldosterone with a renal tubular resistance to the
2–4 mmol/h/70 kg ) and is usually given to correct a action of the hormone. This disorder is commonly
phosphate deficiency rather than potassium defi- caused by spironolactone and amiloride. The suppres-
ciency; thus potassium chloride is often given sion of aldosterone production with hyperkalemia
concomitantly. (Edes and Sunderrajan, 1985) described with heparin
Intravenous replacement of potassium should not use is due to the antiseptic in commercial heparin
normally exceed 40 mmol/h. Plasma potassium levels solutions and not to heparin (Jaques, 1985).
should be monitored at 2–6-hourly intervals (Tannen,
1986; Stockigt, 1977). Spironolactone, amiloride or
(b) Clinical features of hyperkalemia
ACE inhibitors are more useful for preventing renal
potassium loss than for correcting an existing deficit The clinical features of hyperkalemia include tingling,
(Tannen, 1986). paresthesias, weakness, flaccid paralysis and hypoten-
sion. ECG changes include T-wave peaking, P-wave
flattening, PR prolongation (until sinus arrest with
16.8.4 HYPERKALEMIA
nodal rhythm occurs), QRS widening, shortened QT
Hyperkalemia is defined as a serum potassium greater interval and deep S wave. Finally, a sine-wave ECG
than 5.0 mmol/l or plasma potassium greater than pattern develops, which deteriorates to asystole,
4.5 mmol/l. It may be due to excessive intake, severe occurring at plasma potassium levels of 7 mmol/l or
tissue damage, decreased excretion or body fluid greater.
324 FLUID, ELECTROLYTE AND METABOLIC MANAGEMENT

(c) Treatment 15–20% of patients in the ICU (Zaloga, Chernow and


+ Cook, 1985) but cannot be predicted by estimation of
For mild hyperkalemia (i.e. plasma K > 4.5 and
either total serum calcium or of ionized calcium
< 5.5 mmol/l) treatment usually involves reducing the
corrected for pH and serum albumin concentration
potassium intake, correcting renal failure, and treating
(Desai, Carlson and Geheb, 1987).
the cause (e.g. ceasing NSAIDs or spironolactone).
Common causes of hypocalcemia in head-injured
Treatment of severe hyperkalemia (i.e. plasma
patients include respiratory alkalosis, sepsis, hypo-
K+ > 5.5 mmol/l) often involves the administration of
magnesemia and rhabdomyolysis in the polytrauma
intravenous glucose and insulin, sodium bicarbonate
patient. Hypomagnesemia impairs both parathyroid
and calcium chloride (Table 16.19; Tannen, 1986),
hormone (PTH) release and action. Critically ill
although nebulized salbutamol (10–20 mg) may also
patients who are septic or who have unexplained
be useful as it can reduce the plasma potassium by
ionized hypocalcemia may be vitamin-D-deficient,
0.6–1.0 mmol/l after 30 minutes and will last for 2
have an impairment of vitamin D metabolism or
hours (Williams, 1986). Resonium A has the capacity
action, or have impaired release of PTH (Desai,
to bind 3.1 mmol of potassium per gram by releasing
Carlson and Geheb, 1987; Zaloga et al., 1984; Knochel,
3.1 mmol of sodium. Calcium chloride (or gluconate)
1977). Hyperphosphatemia in severe rhabdomyolysis
does not reduce the plasma potassium and is used
causes hypocalcemia by calcium precipitation or
only to counteract the toxic cardiac effects of hyper-
impaired vitamin D metabolism.
kalemia. Since all the methods of reducing the
potassium level are only temporary, treatment should
also be directed at the underlying cause, and may also (a) Clinical features
include dialysis.
Clinical manifestations of ionized hypocalcemia are
generally not evident until ionized calcium is less than
16.8.5 HYPOCALCEMIA 0.7 mmol/l. Signs include hypotension, impaired ven-
tricular function, bradycardia, bronchospasm and
Hypocalcemia occurs in up to 70% of patients in ICU laryngospasm. Weakness, tetany, hyper-reflexia, agi-
(Desai, Carlson and Geheb, 1988; Zaloga and Cher- tation, confusion and seizures may occur, but are less
now, 1986). It is defined as a total plasma calcium less common than in patients with hypoparathyroidism.
than 2.2 mmol/l and may be caused by either a The response to digoxin and catecholamines may also
reduced ionized or protein-bound calcium. In criti- be impaired by hypocalcemia.
cally ill patients calcium loss due to increased tissue
sequestration of calcium may be associated with
(b) Treatment
pancreatitis, septicemia, burns or toxic shock (Cher-
now et al., 1982a; Zaloga, 1991). However, hypocalce- Treatment is only undertaken if the patient is symp-
mia is only important clinically when the physio- tomatic. Acute hypocalcemia may be corrected by
logically active, ionized fraction (40% of total) is intravenous 10% calcium chloride (0.7 mmol calcium
reduced. Ionized calcium is lowered by alkalosis, by per milliliter). Before initiating treatment, hypomag-
high circulating free fatty acids, as in pancreatitis or nesemia should be excluded, as hypomagnesemic
sepsis, and by agents that chelate calcium, such as patients respond poorly to calcium. Calcium chloride
albumin infusions, bicarbonate administration and 10%, 5 ml intravenously (i.e. 3.4 mmol) or 10 ml of
high citrate loads administered with massive blood calcium gluconate 10% (i.e. 2.3 mmol) may be admin-
transfusion (Zaloga and Chernow, 1986). Ionized istered over 3–5 minutes. Rapid infusion of calcium
hypocalcemia (less than 1.15 mmol/l) occurs in may result in unpleasant flushing, hypertension,

Table 16.19 Treatment of life-threatening hyperkalemia

Agent Dose Onset (min) Duration (h)

Intravenous
Dextrose and insulin 50 g dextrose 10–15 3
20 IU insulin
Sodium bicarbonate 50–100 mmol 60–240 1–2
Calcium chloride (10%) 5–10 ml (3.4–6.8 mmol) 1–5 1–2
Oral or rectal resonium A 50 g 120–240 3–12
Nebulized salbutamol 10–20 mg 30 2
DISORDERS OF WATER AND ELECTROLYTE BALANCE 325
bradycardia and atrioventricular nodal blockade, and necessary. Generally, however, in the critically ill
may precipitate digitalis toxicity. Persistent unex- patient, the phosphate level should be kept above
plained hypocalcemia requires further evaluation to 0.8 mmol/l, to ensure adequate respiratory, cardiac and
exclude hypoparathyroidism and vitamin D intracellular function. Phosphate at 40–80 mmol/24 h
deficiency. may be administered orally or intravenously as sodium
or potassium phosphate (Chernow et al., 1989). If an
intravenous solution is administered as the dihydrogen
16.8.6 HYPOPHOSPHATEMIA
salt, an equimolar amount of Na+ or K+ and 80% of the
Hypophosphatemia is defined as a fasting plasma molar amount as H+ is administered with the phos-
phosphate level of less than 0.8 mmol/l. Moderate phate. If it is administered as the monohydrogen salt,
hypophosphatemia exists if the phosphate level is then twice the molar amounts of sodium or potassium
between 0.32 and 0.8 mmol/l (Bohannon, 1989). are administered with the phosphate.
Severe hypophosphatemia exists when the plasma
level is less than 0.32 mmol/l (Hayek and Eisenberg,
16.8.7 HYPOMAGNESEMIA
1989). It may be caused by decreased intake, increased
excretion or intracellular redistribution. Hypomagnesemia is defined as a plasma level of less
Parenteral or enteral nutrition induces a transcellular than 0.7 mmol/l and is associated with a 24-hour urine
shift of phosphate from the ECF to the ICF via insulin- magnesium of less than 1 mmol/l in the absence of
mediated intracellular transport of phosphate with abnormal renal magnesium losses. This is a common
glucose (‘refeeding syndrome’; Solomon and Kirby, electrolyte disturbance in ICU patients (Chernow et al.,
1990; Aubier et al., 1985). Respiratory alkalosis causes 1982b; Kingston, Al-Siba and Skooge, 1986; Paymaster,
intracellular shift of phosphate by stimulating glycol- 1976) and is caused by decreased intake or increased
ysis and intracellular phosphate trapping. Hypophos- loss. The causes are similar to those for hypophospha-
phatemia induced by transcellular shift is associated temia. Common causes in patients with head injury
with hypophosphaturia and the normal body stores of include diuretics (osmotic and non-osmotic), diabetes
phosphate are maintained. Hypokalemia and hypo- insipidus, respiratory alkalosis, sepsis and certain
magnesemia may also promote hypophosphatemia by drugs including aminoglycosides.
impairing renal phosphate reabsorption.
(a) Clinical features
(a) Clinical features
Clinical features tend only to occur when the plasma
Clinical manifestations of hypophosphatemia are not level is less than 0.5 mmol/l (Table 16.20; Paymaster,
usually seen unless levels are markedly reduced, i.e. to 1976; Fishman, 1965; Whang et al., 1984). The clinical
less than 0.3 mmol/l. Patients who are hyperventi- diagnosis of magnesium depletion is difficult because
lated or malnourished, alcoholic, septic or have only 1% of total body magnesium is contained in
diabetic ketoacidosis are at increased risk of severe plasma and because total plasma magnesium does not
hypophosphatemia, and diaphragmatic weakness and accurately reflect the ionized fraction (55% of the
respiratory failure may occur (Lau, 1986). total), which is the physiologically active form. The
remaining plasma magnesium is bound to protein
(33%) or is chelated. Measurement of ionized magne-
(b) Investigations
sium is not available. Hypomagnesemia may be
Patients with hypophosphatemia in the absence of associated with hypokalemia (due to renal loss),
alkalosis and a urinary phosphate greater than
25 mmol/d,are suffering excessive renal loss. If the
urinary loss is less than 5 mmol/d, then renal loss is
Table 16.20 Clinical features of hypomagnesemia
excluded (Knochel, 1981). If hypophosphatemia is
associated with hypercalcemia, hyperparathyroidism Neurological
should be considered.  Confusion, irritability, delirium, convulsions
 Ataxia, athetoid movements
 Weakness, tremors, cramps, tetany
(c) Treatment
Cardiovascular
Hypophosphatemia is usually asymptomatic. If hypo-  Tachyarrhythmias (torsades de pointes)
 Enhanced digoxin toxicity
phosphatemia is due to compartmental shift resulting
from respiratory alkalosis or to catecholamine infu- Biochemical
 Resistant hypokalemia, hypocalcemia, renal tubular
sions, even though phosphate levels may decrease to
acidosis
0.4 mmol/l, treatment with phosphate may not be
326 FLUID, ELECTROLYTE AND METABOLIC MANAGEMENT

hypocalemia (due to impaired release of PTH and


impaired peripheral action of PTH) and renal tubular
acidosis (RTA). In one study, more than 40% of
hypokalemic patients revealed an associated hypo-
magnesemia (Gums, 1987).

(b) Investigations
With magnesium deficiency, the renal loss is usually
minimal (0.05–0.10 mmol/d). An intravenous dose of
magnesium is normally excreted in the urine in 24
hours; thus 30 mmol administered over 8–12 hours
and a 24-hour collection of urine can be used as an
estimate of magnesium balance. Normally more than
60% of the dose is excreted; if less than 50% is excreted
the patient has a magnesium deficiency (Salem,
Munoz and Chernow, 1991).

(c) Treatment (a)

Symptomatic hypomagnesemia (e.g. causing refrac-


tory arrhythmias or seizures) is treated by intravenous
bolus injection of 5–10 ml of 49.3% MgSO4 solution
(2 mmol/ml magnesium; Cohen, Gambill and Eggers,
1977). Less severe degrees of hypomagnesemia are
treated by adding magnesium salts to the enteral or
parenteral solutions.

16.9 Special fluid and electrolyte


problems in the neurosurgical patient
16.9.1 NEUROGENIC PULMONARY EDEMA

Patients with a variety of central nervous system (b)


disorders, including head injury, may develop acute
Figure 16.15 A 24-year-old man suffered a gunshot wound
pulmonary edema in the absence of cardiopulmonary to the head. Shortly after admission to the ICU he developed
disease or other apparent causes. This neurogenic marked frothy sputum and oxygen desaturation. (a) The CT
pulmonary edema may follow head trauma (Ducker, scan shows the missile track and a ‘tight’ brain. (b) The chest
Simmons and Martin, 1969), intracranial tumors, X-ray shows fluffy exudates bilaterally. There is a (Swan–
subarachnoid hemorrhage, stroke, seizures (Bonbrest, Ganz) catheter. Hemodynamic measurement confirmed a
non-cardiogenic basis for the pulmonary edema. Mechanical
1965), Guillain-Barré syndrome and meningitis. ventilation and diuresis resulted in a successful outcome.
Although the cause is unclear, it is thought that
neurogenic pulmonary edema may be due to massive
alpha-adrenergic discharge (Wray and Nicotra, 1978).
The sympathetic discharge increases systemic and
pulmonary vascular resistances. This transiently issued from the endotracheal tube and marked oxygen
increases left atrial pressure and pulmonary artery desaturation was noted on pulse oximetry. He
wedge pressure (PAWP), and pulmonary capillary required ventilation with 100% O2 and positive end-
disruption occurs. An animal model has also impli- expiratory pressure (PEEP); chest X-ray confirmed the
cated endothelial injury in its development (Minnear presence of pulmonary edema (Figure 16.15(b)). The
et al., 1987). The altered pulmonary capillary permea- CVP was 10 mmHg. A Swan–Ganz catheter was
bility results in the accumulation of pulmonary edema inserted to facilitate fluid management in this patient,
fluid, which has a high protein content. who subsequently made a satisfactory recovery.
A previously healthy 24-year-old male was admit- The treatment of neurogenic pulmonary edema is
ted to ICU following a gunshot wound to the head similar to the treatment of adult respiratory distress
(Figure 16.15(a)). Frothy pulmonary edema fluid syndrome (ARDS) from any cause. In addition to
REFERENCES 327
ventilatory support with positive-pressure ventilation injury, because cerebral autoregulation may be over-
and positive end-expiratory pressure (PEEP), fluid come and cerebral edema exacerbated.
management is of the utmost importance. Fluid intake Because the heart cannot compensate for over-
should be crystalloid solution, rather than protein- transfusion or increased venous return, careful mon-
containing solutions, and should be minimal. If PAWP itoring is mandatory. Circulating volume is critical,
is monitored, it should be kept low (5–7 mmHg) to and observation of the CVP or PAWP to fluid
minimize further extravasation of proteinacious fluid. challenge may be used to assess the volume status of
If mean arterial pressure or perfusion is inadequate, the patient.
inotropes should be used to maintain cerebral and Electrolyte abnormalities may develop as a result of
peripheral tissue perfusion. For a detailed account of immobility and absent muscle activity. Mobilization of
the respiratory management of neurogenic pulmonary calcium may increase, causing hypercalcemia and
edema see Chapter 17. hypercalciuria, from about 10 days after injury. The
increase in serum calcium levels may predispose to
ventricular arrhythmias during anesthesia. Thus cal-
16.9.2 NON-KETOTIC HYPERGLYCEMIC
cium levels should be measured preoperatively in
HYPEROSMOLAR COMA
these patients.
Non-ketotic hyperglycemic hyperosmolar coma
(NHHC) may occur following head injury, intra-
cerebral hemorrhage, brain tumor or cerebral infarc-
16.10 Conclusions
tion (Park, Meacham and Netsky, 1976). About two-  The goal of fluid therapy in the critically ill patient
thirds of patients who develop NHHC have no history is to restore and maintain normal O2 delivery to the
of diabetes mellitus. Many have intercurrent infec- tissues before the sequelae of shock (including
tions, or are taking drugs that alter glucose tolerance, secondary neuronal damage) occur.
such as steroids, phenytoin and thiazides, or are  Abnormalities of fluid, electrolyte and acid–base
dehydrated. metabolism may result in alterations in neurological
The diagnosis of NHHC is based on the findings of status and thus interfere with neurological assess-
hyperglycemia, glycosuria and a plasma osmolality ment, as well as contributing to secondary neuronal
greater than 330 mosmol/l in the absence of ketosis. damage and thus worsening outcome.
Patients who have NHHC and are dehydrated  Maintenance of normal cerebral perfusion pressure
because of the osmotic diuresis are often potassium- (CPP) is a key aspect of the management of patients
depleted. They may also have evidence of prerenal with head injuries, since impairment of cerebral
renal failure which, if untreated, can progress to acute autoregulation following traumatic brain injury
renal failure. causes cerebral oxygen delivery to be influenced by
Treatment is primarily correction of dehydration CPP.
and hypertonicity. Physiological saline is used for  Correction of hypovolemia is best achieved with
volume replacement, together with careful monitoring colloid, physiological saline or blood transfusion (if
of serum and urine electrolytes and osmolality. After necessary). At least two animal studies have indi-
sodium deficits have been corrected with physio- cated that the amount of sodium infused does not
logical saline, half physiological saline or dextrose/ adversely affect intracranial pressure (Ramming et
saline (4% dextrose in N/5 saline) should be used to al., 1994). In one of the studies, infusion of a
replace water deficits. The hyperglycemia is usually dextrose solution in amounts similar to the infused
responsive to insulin. sodium load significantly decreased serum sodium,
increased brain edema and decreased neurological
16.9.3 SPINAL-CORD-INJURED PATIENTS
outcome (Shapira et al., 1995).

Cervical cord injury may cause spinal shock due to


anatomical or functional transection of the cord and 16.11 References
this may last for 1–3 weeks. The interruption to Addleman, M., Pollard, A. and Grossman, R. F. (1985) Survival after severe
hypernatremia due to salt ingestion by an adult. American Journal of
sympathetic outflow results in vasodilatation, pooling Medicine, 78, 176–178.
of blood in peripheral vascular beds and hypotension. Adelson, E., Crosby, W. H. and Roeder, W. H. (1955) Further studies of a
hemostatic defect caused by intravenous dextran. Journal of Laboratory and
Reflex bradycardia also occurs, because the cardiac Clinical Medicine, 45, 441.
accelerator nerves arise from segments T1–T4. The Albright, A. L., Latchaw, R. E. and Robinson, A. G. (1984) Intracranial and
systemic effects of osmotic and oncotic therapy in experimental cerebral
hypotensive phase is usually preceded by a rise in edema. Journal of Neurosurgery, 60, 481.
systemic pressure over 3–4 minutes as a result of Alexander, B., Odake, K., Lawlor, D. et al. (1975) Coagulation, haemostasis and
plasma expanders: a quarter century enigma. Federal Proceedings, 34, 1429.
widespread initial sympathetic activation. This may Allen, S. J., Drake, R. E., Williams, J. P. et al. (1987) Recent advances in
be particularly important in the presence of brain pulmonary oedema. Critical Care Medicine, 15, 963.
328 FLUID, ELECTROLYTE AND METABOLIC MANAGEMENT

Arieff, A. I. (1984) Central nervous system manifestations of disordered Burke, A. M., Quest, D. O., Chein, S. and Cerri, C. (1981) The effects of
sodium metabolism. Clinics in Endocrinology and Metabolism, 13, 269–294. mannitol on blood viscosity. Journal of Neurosurgery, 55, 550–553.
Arieff, A. I. (1985) Effects of water, acid–base and electrolyte disorders on the Butterworth, J. F. and De Witt, D. S. (1989) Severe head trauma: pathophysiol-
central nervous system: hyponatremia, in Fluid, Electrolyte and Acid–base ogy and management, in Critical Care Clinics, vol. 5(4), (ed. D. S. Prough), W.
Disorders, (eds A. I. Arieff and R. A. De Fronzo), Churchill Livingstone, New B. Saunders, Philadelphia, PA, pp. 807–820.
York, pp. 992–1007. Calvin, J. E., Driedger, A. A. and Sibbald, W. J. (1981) The hemodynamic effect
Arieff, A. I. (1986) Hyponatraemia, convulsions, respiratory arrest and of rapid fluid infusion in critically ill patients. Surgery, 90, 61.
permanent brain damage after elective surgery in healthy women. New Carrico, C. J., Canizaro, P. C. and Shires, G. T. (1976) Fluid resuscitation
England Journal of Medicine, 314, 1529–1535. following injury: Rationale for the use of balanced salt solutions. Critical
Arieff, A. I. and Guisado, R. (1976) Effects on the central nervous system of Care Medicine, 4, 46.
hypernatremic and hyponatremic states. Kidney International, 10, 104–116. Carvajal, H. F. and Parks, D. K. (1988) Optimal composition of burn
Arieff, A. I., Liach, F. and Massry, S. G. (1976) Neurological manifestations and resuscitation fluids. Critical Care Medicine, 16, 695.
morbidity of hyponatraemia: correlation with brain water and electrolytes. Cerra, F. B. (1986) The role of nutrition in the management of metabolic stress,
Medicine, 55, 121–129. in Critical Care Clinics, vol. 2(4), (ed. A. A. Meyer), W. B. Saunders,
Arora, N. S. and Rochester, D. F. (1982) Respiratory muscle strength and Philadelphia, PA, pp. 807–819.
maximal voluntary ventilation in undernourished patients. American Cervera, A. L. and Moss, G. (1975) Dilutional re-expansion with crystalloid
Review of Respiratory Disease, 126, 5–8. after massive haemorrhage: saline versus balanced electrolyte solution for
Atik, M. (1967) Dextran 40 and dextran 70. Archives of Surgery, 94, 664. maintenance of normal blood volume and arterial pH. Journal of Trauma, 15,
Atik, M. (1969) The uses of dextran in surgery: a current evaluation. Surgery, 498.
65, 548. Chernow, B., Zaloga, G., McFadden, E. et al. (1982a). Hypocalemia in critically
Aubier, M., Murciano, D., Lecocguic, Y. et al. (1985) Effect of hypophosphate- ill patients. Critical Care Medicine, 10, 848–851.
mia on diaphragmatic contractility in patients with acute respiratory Chernow, B., Smith, J., Rainey, T. G. et al. (1982b). Hypomagnesemia:
failure. New England Journal of Medicine, 313, 420–424. Implications for the critical care specialist. Critical Care Medicine, 10,
Aubry, R. H. and Nankin, H. R. (1965) Measurement of the osmotic threshold 193–196.
for vasopressin release in human subjects, and its modification by cortisol. Chernow, B., Bamberger, S., Stoiko, M. et al. (1989) Hypomagnesemia in
Journal of Clinical Endocrinology and Metabolism, 25, 1481–1490. patients in postoperative intensive care. Chest, 95, 391–397.
Ayus, J. C., Krothapalli, R. K. and Arieff, A. I. (1985) Changing concepts in Chung, H. M., Kluge, R., Schrier, R. W. and Anderson, R. J. (1986)
treatment of severe symptomatic hyponatremia. Rapid correction and Postoperative hyponatraemia. A prospective study. Archives of Internal
possible relation to central pontine myelinolysis. American Journal of Medicine, 146, 333–336.
Medicine, 78, 897–902. Civetta, J. M. (1979) A new look at the Starling equation. Critical Care Medicine,
Ayus, J. C., Krothapalli, R. K. and Arieff, A. I. (1987) Treatment of symptomatic 7, 84.
hyponatremia and its relation to brain damage. New England Journal of Clifton, G. L., Robertson, C. S., Grossman, R. G. et al. (1984) The metabolic
Medicine, 317, 1190–1195. response to severe head injury. Journal of Neurosurgery, 60, 687–696.
Ayus, J. C., Krothapalli, R. K. and Arieff, A. I. (1988) Correction of Cluitmans, F. H. M. and Meinders, A. E. (1990) Management of severe
hyponatraemia and its relation to brain damage. New England Journal of hyponatremia: Rapid or slow correction. American Journal of Medicine, 88,
Medicine, 318, 1336–1337. 161–166.
Baek, S. M., Makabali, G. G., Bryan-Brown, C. W. et al. (1975) Plasma Cohen, H. B., Gambill, A. F. and Eggers, G. W. N. (1977) Acute pulmonary
expansion in surgical patients with high central venous pressure (CVP): the oedema following head injury: two case reports. Anesthesia and Analgesia,
relationship of blood volume to hematocrit, CVP, pulmonary wedge 56, 136–139.
pressure and cardiorespiratory changes. Surgery, 78, 304. Consensus Development Conference (1988) Perioperative red blood cell
Baker, J. W., Deitch, E. D., Ma, L. M. et al. (1988) Hemorrhagic shock induces transfusion. Report of consensus development conference on perioperative
bacterial translocation from the gut. Journal of Trauma, 28, 896. red cell transfusion. Journal of the American Medical Association, 260,
Barratt, L. J. (1981) Potassium handling by the kidney. Hyperkalaemia. 2700–2703.
Australian and New Zealand Journal of Medicine, 11, 16–22. Crandall, E. D. (1974) Serum sodium response to hyperglycemia. New England
Bartter, F. C. and Schwartz, W. B. (1967) The syndrome of inappropriate Journal of Medicine, 290, 465.
secretion of antidiuretic hormone. American Journal of Medicine, 42, Crandall, E. D., Staub, N. C., Goldberg, H. S. et al. (1983) Recent developments
790–806. in pulmonary oedema. Annals of Internal Medicine, 99, 808.
Battacharya, J., Gropper, M. A. and Staub, N. S. (1984) Interstitial fluid Crompton, M. R. (1971) Hypothalamic lesions following closed head injury.
pressure gradient measured by micropuncture in excised dog lung. Journal Brain, 94, 165–172.
of Applied Physiology, 56, 271. Cross, J. S., Griber, D. P., Burchard, K. W. et al. (1988) Hypertonic saline fluid
Battistella, F. D. and Wisner, D. H. (1991) Combined haemorrhagic shock and therapy following surgery: a prospective study. Journal of Trauma, 29, 817.
head injury: effects of hypertonic saline (7.5%) resuscitation. Journal of Dawidson, I. J. A., Willms, C. D., Sandor, Z. F. et al. (1991) Ringer’s lactate with
Trauma, 31, 182. or without 3% dextran-60 as volume expanders during abdominal aortic
Beckstead, J. E., Tweed, W. A., Lee, J. et al. (1978) Cerebral blood flow and surgery. Critical Care Medicine, 19, 36.
metabolism in man following cardiac arrest. Stroke, 9(6), 569. Demling, R. H. (1980) Lung fluid and protein dynamics during haemorrhagic
Beecher, H. K. (1945) Preparation of battle casualties for surgery. Annals of shock, resuscitation and recovery. Circulatory Shock, 7, 149.
Surgery, 121, 769. Demling, R. H., Manohar, M., Will, J. A. et al. (1979) The effect of plasma
Behrman, S. W., Fabian, T. C., Kudsk, K. A. et al. (1991) Microcirculatory flow oncotic pressure on the pulmonary microcirculation after haemorrhagic
changes after initial resuscitation of haemorrhagic shock with 7.5% shock. Surgery, 86, 323.
hypertonic saline/6% dextran 70. Journal of Trauma, 31, 589. Demling, R. H., Harms, B., Kramer, G. et al. (1982) Acute versus sustained
Bichet, D. and Schrier, R. W. (1982) Evidence against concept of hypona- hypoproteinaemia and post traumatic pulmonary oedema. Surgery, 92, 79.
traemia and ‘sick cells’. Lancet, i, 742. Desai, T. K., Carlson, R. W. and Geheb, M. A. (1987) Parathyroid-vitamin D
Bohannon, N. J. V. (1989) Large phosphate shifts with treatment for axis in critically ill patients with unexplained hypocalcaemia. Kidney
hyperglycaemia. Archives of Internal Medicine, 149, 1423–1425. International, 32(22), S225.
Boldt, J., Zickmann, B., Ballesteros, M. et al. (1991) Cardiorespiratory Desai, T. K., Carlson, R. W. and Geheb, M. A. (1988) Prevalence and clinical
responses to hypertonic saline solution in cardiac operations. Annals of implications of hypocalcaemia in acutely ill patients in a medical intensive
Thoracic Surgery, 51, 610. care setting. American Journal of Medicine, 84, 209–214.
Bonbrest, H. C. (1965) Pulmonary oedema following an epileptic seizure. Doczi, T., Tarjanyi, J., Huszka, E. and Kiss, J. (1982) Syndrome of inappropriate
American Review of Respiratory Disease, 91, 97–100. secretion of antidiuretic hormone (SIADH) after head injury. Neurosurgery,
Bouzarth, W. F., Shenkin, H. A. et al. (1982) Is cerebral hyponatraemia 10, 685–688.
iatrogenic? Lancet, i, 1061–1062. Dodge, C. and Glass, D. D. (1982) Crystalloid and colloid therapy. Seminars in
Brinkmeyer, S., Safar, P., Motoyama, E. et al. (1981) Superiority of colloid over Anaesthesia, 1, 293–301.
electrolyte solution for fluid resuscitation (severe normovolemic hemodilu- Ducker, T. B., Simmons, R. L. and Martin, A. M. (1969) Pulmonary oedema as
tion). Critical Care Medicine, 9, 369. a complication of intracranial disease. American Journal of Diseases of
Brobeck, J. R. (ed.) (1973) Best and Taylor’s Physiological Basis of Medical Practice, Children, 118, 638–641.
9th edn, Williams & Wilkins, Baltimore, MD, pp. 4–113–4-123; 1–15–1-29. Dzau, V. J. and Hollenberg, N. K. (1984) Renal response to captopril in severe
Brunner, J. E., Redmond, J. M., Haggar, A. M. et al. (1990) Central pontine heart failure: role of frusemide in natriuresis and reversal of hyponatremia.
myelinolysis and pontine lesions after rapid correction of hyponatraemia: a Annals of Internal Medicine, 100, 777–782.
prospective magnetic resonance imaging study. Annals of Neurology, 27, Edelman, I. S. and Leibman, J. (1959) Anatomy of body water and electrolytes.
61–66. American Journal of Medicine, 27, 256–263.
Buchman, T. G., Menker, J. and Lipsett, P. A. (1991) Strategies for trauma Edes, T. E. and Sunderrajan, E. V. (1985) Heparin-induced hyperkalemia.
resuscitation. Surgery, Gynecology, and Obstetrics, 172, 8–12. Archives of Internal Medicine, 145, 1070–1072.
Buckingham, J. C. (1985) Hypothalamo-pituitary responses to trauma. British Edwards, O. M. and Clark, J. D. A. (1986) Post-traumatic hypopituitarism.
Medical Bulletin, 41, 203–211. Medicine, 65, 281–290.
REFERENCES 329
Edwards, J. D., Nightingale, P., Wilkins, R. G. et al. (1989) Hemodynamic and Hume, D. M. and Egdahl, R. H. (1959) The importance of the brain in the
oxygen transport response to modified fluid gelatin in critically ill patients. endocrine response to injury. Annals of Surgery, 150, 697–712.
Critical Care Medicine, 17, 996. Humes, H. D. (1986) Disorders of water metabolism, in Fluids and Electrolytes,
Emerson, T. E. (1989) Unique features of albumin: a brief review. Critical Care (eds J. P. Kokko and R. L. Tannen), W. B. Saunders, Philadelphia, PA, pp.
Medicine, 17, 690. 118–149.
Eyer, S. D., Micon, L. T., Konstantinides, F. N. et al. (1993) Early enteral feeding Ito, U., Ohno, K., Nakamura, R. et al. (1979) Brain edema during ischaemia
does not attenuate metabolic response after blunt trauma. Journal of Trauma, and after restoration of blood flow. Stroke, 10(5), 542.
34(5), 639–644. James, W. P. T. (1971) Effects of protein-calorie malnutrition on intestinal
Falk, J. L. (1991) Fluid resuscitation and colloid-crystalloid controversy: new absorption. Annals of the New York Academy of Science, 176, 224–261.
thoughts on an old debate. Critical Care Medicine, 19, 451. Jaques, L. B. (1985) The new understanding of the drug heparin. Chest, 88,
Fein, I. A., Rackow, E. C., Sprung, C. L. et al. (1982) Relation of COP to arterial 751–757.
hypoxaemia and cerebral oedema during crystalloid volume loading of Jeejeebhoy, K. N. (1986) Muscle function and nutrition. Gut, 27(Suppl.),
patients. Annals of Internal Medicine, 96, 570. 25–39.
Finberg, L., Luttrell, C. N. and Redd, H. (1959) Pathogenesis of lesions in the Jenkins, P. G. and Larmore, C. (1974) Hyperglycemia-induced hyponatremia.
nervous system in hypernatraemic states. Pediatrics, 23, 46–53. New England Journal of Medicine, 290, 573.
Fishman, R. A. (1965) Neurological aspects of magnesium metabolism. Jennett, B., Teasdale, G. and Braakman, R. (1979) Prognosis of patients with
Archives of Neurology, 12, 562–570. severe head injury. Neurosurgery, 4, 283–289.
Flear, C. T. G., Gill, G. V. and Burn, J. (1981) Hyponatraemia: mechanisms and Karlson, K. E., Garzon, A. A., Shaftan, G. W. et al. (1967) Increased blood loss
management. Lancet, ii, 26–31. associated with administration of certain plasma expanders: Dextran 75,
Flear, C. T. G. and Singh, C. M. (1973) Hyponatraemia and sick cells. British dextran 40 and hydroxyethyl starch. Surgery, 62, 670.
Journal of Anaesthesia, 45, 976–994. Katz, M. A. (1973) Hyperglycemia-induced hyponatraemia – calculation of
Freshman, S. P., Battistella, F. D., Matteucci, M. et al. (1993) Hypertonic saline expected serum sodium depression. New England Journal of Medicine, 289,
(7.5%) versus mannitol: a comparison for treatment of acute head injuries. 843–844.
Journal of Trauma, 35(3). Kee, D. B. and Wood, J. H. (1984) Rheology of the circulation. Neurosurgery,
Fuisz, R. E. (1963) Hyponatremia. Medicine, 42, 149–168. 15(1), 125–131.
Gala, G. J., Lilly, M. P., Thomas, S. E. et al. (1991) Interaction of sodium and Kern, K. and Meislin, H. (1984) Diabetes insipidus: occurrence after minor
volume in fluid resuscitation after haemorrhage. Journal of Trauma, 31, 545. head trauma. Journal of Trauma, 24, 69–72.
Gamble, J., Ross, G. and Tisdall, F. (1923) The metabolism of fixed base during Kingston, M. E., Al-Siba, M. B. and Skooge, W. C. (1986) Clinical manifesta-
fasting. Journal of Biological Chemistry, 57, 633. tions of hypomagnesemia. Critical Care Medicine, 14, 950–954.
Goldberg, H. S. (1980) Pulmonary interstitial compliance and microvascular Knochel, J. P. (1977) The pathophysiology and clinical characteristics of severe
filtration coefficient. American Journal of Physiology, 239, H189. hypophosphataemia. Archives of Internal Medicine, 137, 203–220.
Granger, J. H. (1979) Role of the interstitial matrix and lymphatic pump in Knochel, J. P. (1981) Hypophosphatemia. Western Journal of Medicine, 134,
regulation of transvascular fluid balance. Microvascular Research, 18, 207. 15–26.
Griffel, M. I. and Kaufman, B. S. (1992) Pharmacology of colloids and Krausz, M. M., Landau, E. H., Klin, B. et al. (1992) Hypertonic saline treatment
crystalloids. Critical Care Clinics, 8, 235. of uncontrolled hemorrhagic shock at different periods from bleeding.
Griswold, J. A., Anglin, B. L., Love, R. T. et al. (1991) Hypertonic saline Archives of Surgery, 127, 93.
resuscitation: efficacy in a community-based burn unit. Southern Medical Krumbhaar, E. B. (1914) Hemolysis due to intravenous injection of distilled
Journal, 84, 692. water. Journal of the American Medical Association, 62, 992–996.
Gums, J. G. (1987) Clinical significance of magnesium: a review. Drug Lamke, L. O. and Liljedahl, S. O. (1976) Plasma volume changes after infusion
Intelligence and Clinical Pharmacology, 21, 240–246. of various plasma expanders. Resuscitation, 5, 93–100.
Gunnar, W., Merlotti, G. J., Jonasson, O. et al. (1986) Resuscitation from Lau, K. (1986) Phosphate disorders, in Fluids and Electrolytes, (eds J. P. Kokko
hemorrhagic shock: Alterations of the intracranial pressure after normal and R. L. Tannen), W. B. Saunders, Philadelphia, PA, pp. 398–471.
saline, 3% saline and dextran 40. Annals of Surgery, 204, 686. Laureno, R. and Karp, B. I. (1988) Pontine and extrapontine myelinolysis
Gunnar, W., Jonasson, O., Merlotti, G. et al. (1988) Head injury and following rapid correction of hyponatraemia. Lancet, i, 1439–1441.
hemorrhagic shock: Studies of the blood brain barrier and intracranial Lazrove, S., Waxman, K., Shippy, C. et al. (1980) Hemodynamic, blood volume
pressure after resuscitation with normal saline, 3% saline solution and and oxygen transport responses to albumin and hydroxyethyl starch
dextran-40. Surgery, 103, 398–407. infusions in critically ill postoperative patients. Critical Care Medicine, 8, 302.
Guyton, A. C., Granger, H. J. and Taylor, A. E. (1971) Interstitial fluid pressure. Leaf, A. (1974) Hyponatraemia. Lancet, i, 1119–1120.
Physiology Review, 51, 527. Levine, O. R., Mellins, R. B., Senior, R. M. et al. (1967) The application of
Harms, B. A., Kramer, G. C., Bodal, B. I. et al. (1981) Effect of hypoproteinae- Starling’s law of capillary exchange to the lungs. Journal of Clinical
mia on pulmonary and soft tissue oedema formation. Critical Care Medicine, Investigation, 46, 934.
9, 503. Levison, M. and Trunkey, D. D. (1982) Initial assessment and resuscitation
Harrison, N., Darrow, D. and Yannet, H. (1936) The total electrolyte content of Surgical Clinics of North America, 62, 9–16.
animals and its probable relation to distribution of body water. Journal of Lohr, J. W., McReynolds, J. and Grimaldi, T. (1988) Effect of acute and chronic
Biological Chemistry, 113, 515. hypernatraemia on myoinositol and sorbitol concentration in rat brain and
Hauser, C. J., Shoemaker, W. C., Turpin, I. et al. (1980) Oxygen transport kidney. Life Science, 271–276.
responses to colloids and crystalloids in critically ill surgical patients. Lowe, R. J., Moss, G. S., Jilek, J. et al. (1979) Crystalloid vs colloid in the
Surgery Gynecology and Obstetrics, 150, 811. etiology of pulmonary failure after trauma. A randomized trial in man.
Haydock, D. A. and Hill, G. L. (1987) Improved wound healing response in Critical Care Medicine, 7, 107.
surgical patients receiving intravenous nutrition. British Journal of Surgery, Lowell, J. A., Schifferdecker, C., Driscoll, D. F. et al. (1990) Postoperative fluid
74, 320–323. overload: Not a benign problem. Critical Care Medicine, 18(Suppl.), S193.
Hayek, M. E. and Eisenberg, P. G. (1989) Severe hypophosphataemia Lundy, E. F., Kuhn, J. E., Kwon, J. M. et al. (1987) Infusion of 5% dextrose
following the institution of enteral feedings. Archives of Surgery, 124, increases mortality and morbidity following six minutes of cardiac arrest in
1325–1328. resuscitated dogs. Journal of Critical Care, 2, 4–14.
Hayes, M. A. (1968) Water and electrolyte therapy after operation. New McMahon, M. M., Farnell, M. B. and Murray, M. J. (1993) Nutritional support
England Journal of Medicine, 278, 1054–1056. of critically ill patients. Mayo Clinic Proceedings, 68, 911–920.
Heughan, C., Niinikoski, J. and Hunt, T. K. (1972) Effect of excessive infusion Mangalore, P. P. and Hunt, T. K. (1972) Effect of varying oxygen tensions on
of saline on tissue oxygen transport. Surgery, Gynecology and Obstetrics, 135, healing open wounds. Surgery, Gynecology, and Obstetrics, 135, 756.
257. Martinez-Maldonado, M. (1980) Inappropriate antidiuretic hormone secretion
Heymsfield, S. B., Bethel, R. A., Ansley, J. D. et al. (1978) Cardiac abnormalities of unknown origin. Kidney International, 17, 554–567.
in cachectic patients before and during nutritional repletion. American Heart Mattox, K. L., Maningas, P. A., Moore, E. E. et al. (1991) Pre-hospital
Journal, 95, 584–594. hypertonic saline/dextran infusion for post-traumatic hypotension. Annals
Hindman, B. J., Funatsu, N., Cheng, D. C. H. et al. (1990) Differential effect of of Surgery, 213, 482.
oncotic pressure on cerebral and extracerebral water content during Messmer, K. (1975) Hemodilution. Surgical Clinics of North America, 55, 659.
cardiopulmonary bypass in rabbits. Anesthesiology, 73, 951. Meye, B. J., Meyer, A. and Guyton, A. (1968) Interstitial fluid pressure. Clinical
Holt, M. E., Ryall, M. E. and Campbell, A. K. (1984) Albumin inhibits human Research, 22, 263.
polymorphonuclear leucocyte luminol-dependent chemiluminescence: evi- Minnear, F. L., Kite, C., Hill, L. A. and Hoyte, Z. (1987) Endothelial injury and
dence for oxygen radical scavenging. British Journal of Experimental pulmonary congestion characterize neurogenic pulmonary oedema in
Pathology, 65, 231. rabbits. Journal of Applied Physiology, 63(1), 335–341.
Horton, J., Landreau, R. and Tuggle, T. (1985) Cardiac response to fluid Mishle, J. (1984) Synthetic plasma volume expanders – their pharmacology,
resuscitation from haemorrhagic shock. Surgery, Gynecology, and Obstetrics, safety and clinical efficacy. Clinical Haematology, 13, 75.
160, 444–452. Modig, J. (1986) Effectiveness of dextran 70 versus Ringer’s acetate in
Horton, J. W. and Walker, P. B. (1991) Small volume hypertonic saline dextran traumatic shock and adult respiratory distress syndrome. Critical Care
resuscitation from canine endotoxin shock. Annals of Surgery, 214, 64. Medicine, 14, 454.
330 FLUID, ELECTROLYTE AND METABOLIC MANAGEMENT

Monafo, W. W., Halverson, J. D. and Schechtman, K. (1984) The role of Rainey, T. G. and English, J. F. (1988) Pharmacology of colloids and
concentrated sodium solutions in the resuscitation of patients with severe crystalloids, in The Pharmacologic Approach to the Critically Ill Patient, (ed. B.
burns. Surgery, 95, 129. Chernow), Williams & Wilkins, Baltimore, MD, p. 219.
Moore, F. D., Dagher, F. J., Boyden, C. M. et al. (1966) Hemorrhage in normal Ramming, S., Shackford, S. R., Zhuang, J. and Schmoker, J. D. (1994) The
man. 1. Distribution and dispersal of saline infusions following acute relationship of fluid balance and sodium administration to cerebral oedema
blood loss: clinical kinetics of blood volume support. Annals of Surgery, formation and intracranial pressure in a porcine model of brain injury.
163, 485. Journal of Trauma, 37, 705–713.
Morissette, M. P. (1977) Colloid osmotic pressure: its measurement and Reed, L. L., Manglano, R., Martin, M. et al. (1991a) The effect of hypertonic
clinical value. Canadian Medical Association Journal, 116, 897. saline resuscitation on bacterial translocation after haemorrhagic shock in
Morse, M. L., Milstein, J. M., Haas, J. E. et al. (1985) Effect of hydration on rats. Surgery, 110, 685.
experimentally induced cerebral edema. Critical Care Medicine, 13(7), 563. Reed, R. L., Johnston, T. D., Chen, Y. et al. (1991b) Hypertonic saline alters
Moss, G. S. and Gould, S. A. (1988) Plasma expanders. American Journal of plasma clotting times and platelet aggregation. Journal of Trauma, 31, 8.
Surgery, 155, 425–434. Ring, J. and Messmer, K. (1977) Incidence and severity of anaphylactoid
Moss, G. S., Lowe, R. J., Jilek, J. et al. (1981) Colloid or crystalloid in the reactions to colloid volume substitutes. Lancet, i, 466.
resuscitation of haemorrhagic shock: a controlled clinical trial. Surgery, 89, Roberts, P., Pollay, M., Engles, L. et al. (1987) Effect on intracranial pressure of
434. furosemide combined with varying doses and administration of mannitol.
Muizelaar, J. P., Wei, E. P., Kontos, H. A. and Becker, D. P. (1983) Mannitol Journal of Neurosurgery, 66, 440–446.
causes compensatory cerebral vasoconstriction and vasodilation in Robertson, G. L. (1989) Syndrome of inappropriate antidiuresis. New England
response to blood viscosity changes. Journal of Neurosurgery, 59, 822–828. Journal of Medicine, 321, 538–539.
Mulinari, R. A., Gavras, I., Wang, Y. X. et al. (1990) Effects of a vasopressin Robin, A. P., Ing, T. S., Lancaster, G. A. et al. (1979) Hyperglycemia-induced
antagonist with combined antipressor and antidiuretic activities in rats hyponatremia: a fresh look. Clinical Chemistry, 25, 496–497.
with left ventricular dysfunction. Circulation, 81, 308–311. Robinson, A. G. (1985) Disorders of antidiuretic hormone secretion. Clinics in
Muzaffar, T. Z., Stalker, A. L., Bryce, W. A. J. et al. (1973) Quantitative studies Endocrinology and Metabolism, 14, 55–88.
on fibrin formation and effects of dextran. Bibliotheca Anatomica, 12, 340. Ross, E. J. and Christie, S. B. M. (1969) Hypernatremia. Medicine, 48,
Needleman, P. and Greenwald, J. E. (1986) Atriopeptin: a cardiac hormone 441–472.
intimately involved in fluid, electrolyte, and blood-pressure homeostasis. Roth, E., Lax, L. C. and Malone, J. V. (1969) Ringer’s lactate solution and
New England Journal of Medicine, 314, 828–834. extracellular fluid volume in the surgical patient: a critical analysis. Annals
Nelson, P. B., Seif, S. M., Maroon, J. C. and Robinson, A. G. (1981) of Surgery, 169, 149.
Hyponatraemia in intracranial disease. Perhaps not the syndrome of Salem, M., Munoz, R. and Chernow, B. (1991) Hypomagnesemia in critical
inappropriate secretion of antidiuretic hormone (SIADH). Journal of illness: a common and clinically important problem. Critical Care Clinics,
Neurosurgery, 55, 938–941. 7(1), 225–252.
Niinikoski, J., Hengan, C. and Hunt, T. K. (1972) Oxygen tensions in human Samson, W. K. (1987) Atrial natriuretic factor and the central nervous system.
wounds. Journal of Surgical Research, 12, 1977. Endocrinology and Metabolism Clinics of North America, 16, 145–161.
Norenberg, M. D. and Papendick, R. E. (1984) Chronicity of hyponatraemia as Scheinkestel, C. D., Tuxen, D. V., Cade, J. F. and Shann, F. A. (1989) Fluid
a factor in experimental myelinolysis. Annals of Neurology, 15, 544–547. management of shock in critically ill patients. Medical Journal of Australia,
Oh, M. S. and Carroll, H. J. (1992) Disorders of sodium metabolism: 150, 508–517.
hypernatraemia and hyponatraemia. Critical Care Medicine, 20(1), 94–103. Schultze, R. G. and Nissenson, A. R. (1980) Potassium: physiology and patho-
Packer, M., Medina, M. and Yushak, M. (1984) Correction of dilutional physiology, in Clinical Disorders of Fluid And Electrolyte Metabolism, 3rd edn,
hyponatremia in severe chronic heart failure by converting-enzyme (eds M. H. Maxwell and C. R. Kleeman), McGraw-Hill, New York, pp.
inhibition. Annals of Internal Medicine, 100, 782–789. 113–143.
Pappenheimer, J. R. and Soto-Rivera, A. (1948) Effective osmotic pressure of Seckl, J. and Dunger, D. (1989) Postoperative diabetes insipidus. British
the plasma proteins and other quantities associated with the capillary Medical Journal, 298, 2–3.
circulation in the hind limbs of cats and dogs. American Journal of Physiology, Seckl, J. R., Dunger, D. B. and Lightman, S. L. (1987) Neurohypophyseal
152, 471. function during early post- operative diabetes insipidus. Brain, 110,
Park, B. E., Meacham, W. F. and Netsky, N. G. (1976) Nonketotic hyper- 737–746.
glycaemic hyperosmolar coma: report of neurosurgical cases with a review Serkes, K. D. and Lang, S. (1966) Changes in extracellular fluid volume after
of mechanisms and treatment. Journal of Neurosurgery, 44, 409–417. hemorrhage and tourniquet trauma. Surgery Forum, 17, 58.
Paymaster, N. J. (1976) Magnesium metabolism: a brief review. Annals of the Shackford, S. R. (1987) Fluid resuscitation of the trauma victim, in Trauma
Royal College of Surgeons of England, 58, 309–314. Problems in Critical Care, (eds S. R. Shackford and A. Perel), J. B. Lippincott,
Peters, R. M. and Hargens, A. R. (1981) Protein vs electrolytes and all of the Philadelphia, PA, vol. 1 pp. 576–587.
Starling forces. Archives of Surgery, 116, 1293. Shapira, Y., Artae, A. A., Qassam, N. et al. (1995) Brain oedema and neurologic
Pirisino, R., Di Simplicio, P., Ignesti, G. et al. (1988) Sulfhydryl groups and status with rapid infusion of 0.9% saline or 5% dextrose after head trauma.
peroxidase-like activity of albumin as scavenger of organic peroxides. Journal of Neurosurgical Anesthesia, 7,17–25.
Pharmacology Research Community, 20, 545. Shippy, C. R., Appel, P. L. and Shoemaker, W. C. (1984) Reliability of clinical
Plum, F. and Posner, J. B. (1980) The Diagnosis of Stupor and Coma, 3rd edn, F. monitoring to assess blood volume in critically ill patients. Critical Care
A. Davis, Philadelphia, PA. Medicine, 12, 107.
Poole, G. V., Johnson, J. C., Prough, D. S. et al. (1986) Cerebral haemodynamics Shires, G. T. (1965) The role of sodium containing solution in the treatment of
after haemorrhagic shock: effects of the type of resuscitation fluid. Critical oligemic shock. Surgical Clinics of North America, 45, 365.
Care Medicine, 14, 629. Shires, G. T. and Canizaro, P. C. (1973) Fluid resuscitation in the severely
Poole, G. V., Miller, J. D., Agnew, S. G. and Griswold, J. A. (1992) Lower injured. Surgical Clinics of North America, 53, 1341.
extremity fracture fixation in head injured patients. Journal of Trauma, 32, Shires, G. T., Williams, J. and Brown, F. (1960) Simultaneous measurement of
654–659. plasma volume, extracellular fluid volume and red blood cell mass in man
Price, D. J. (1992) The intensive care of head-injured patients, in Care of the utilizing I131, S35, O4, Cr51. Journal of Laboratory and Clinical Medicine, 55,
Critically Ill Patient, 2nd edn, (eds J. Tinker and W. M. Zapol), Springer- 776.
Verlag, London, pp. 831–872. Shires, T., Coln, D., Carrico, J. et al. (1964) Fluid therapy in haemorrhagic
Price, D. J. E. and Murray, A. (1972) The influence of hypoxia and hypotension shock. Archives of Surgery, 88, 688.
on recovery from head injury. Injury, 3, 218–224. Shires, G. T., Cunningham, J. N., Baker, C. R. F. et al. (1972) Alterations in
Prough, D. S., Johnson, J. C., Poole, G. V. et al. (1985) Effects on intracranial cellular membrane function during hemorrhagic shock in primates. Annals
pressure of resuscitation from haemorrhagic shock with hypertonic saline of Surgery, 176, 288.
versus lactated Ringer’s solution. Critical Care Medicine, 13, 407. Shoemaker, W. C. (1976) Comparison of the relative effectiveness of whole
Prough, D. S., Johnson, J. C., Stump, D. A. et al. (1986) Effects of hypertonic blood transfusions and various types of fluid therapy in resuscitation.
saline versus lactated Ringer’s solution on cerebral oxygen transport Critical Care Medicine, 4, 71.
during resuscitation from hemorrhagic shock. Journal of Neurosurgery, 64, Shoemaker, W. C. (1982) Fluids and electrolytes in the adult, in Critical Care:
627. State of the Art, (eds W. C. Shoemaker and W. L. Thompson), vol. 3, Society
Puri, V. K., Howard, M., Pidipaty, B. B. et al. (1983) Resuscitation in of Critical Care Medicine, Fullerton, CA.
hypovolemia and shock: a prospective study of hydroxyethyl starch and Shoemaker, W. C. and Czer, L. S. (1979) Evaluation of the biologic importance
albumin. Critical Care Medicine, 11, 518. of various hemodynamic and oxygen transport variables: which variables
Rackow, E. C., Falk, J. L., Fein, I. A. et al. (1983) Fluid resuscitation in should be monitored in postoperative shock? Critical Care Medicine, 7,
circulatory shock: a comparison of the cardiorespiratory effects of albumin, 424.
hetastarch and saline solutions in patients with hypovolaemic and septic Shoemaker, W. C. and Hauser, C. J. (1979) Critique of crystalloid versus
shock. Critical Care Medicine, 11, 839. colloid therapy in shock and shock lung. Critical Care Medicine, 7, 117.
Rackow, E. C., Fein, I. A. and Leppo, J. (1977) Colloid osmotic pressure as a Shoemaker, W. C., Schluchter, M., Hopkins, J. A. et al. (1981) Comparison of
prognostic indicator of pulmonary oedema and mortality in the critically ill. the relative effectiveness of colloids and crystalloids in emergency
Chest, 72, 709. resuscitation. American Journal of Surgery, 142, 73.
REFERENCES 331
Shoemaker, W. C., Appel, P. L., Waxman, K. et al. (1982) Clinical trials of Wasterlain, C. G. and Posner, J. B. (1968) Cerebral oedema in water
survivors. 7 cardiorespiratory patterns as therapeutic goals in critically ill intoxication: I. Clinical and chemical observations. Archives of Neurology, 19,
post-operative patients. Critical Care Medicine, 10, 398–403. 71–78.
Shoemaker, W. C., Appel, P., Kram, H. et al. (1988) Prospective trial of supra Weaver, D. W., Ledgerwood, A. M., Lucas, C. E. et al. (1978) Pulmonary effects
normal values of survivors as therapeutic goals in high risk surgical of albumin resuscitation for severe hypovolaemic shock. Surgery, 113, 387.
patients. Chest, 94, 1176–1186. Weed, L. H. and McKibben, P. S. (1919a) Experimental alteration of brain bulk.
Shucart, W. A. and Jackson, I. (1976) Management of diabetes insipidus in American Journal of Physiology, 48, 531.
neurosurgical patients. Journal of Neurosurgery, 44, 65–71. Weed, L. H. and McKibben, P. S. (1919b) Pressure changes in the cerebrospinal
Siegel, J. H., Gens, D. R., Mamantov, T. et al. (1991) Effect of associated injuries fluid following intravenous injection of solutions of various concentrations.
and blood volume replacement on death, rehabilitation needs, and American Journal of Physiology, 48(4):512.
disability in blunt traumatic brain injury. Critical Care Medicine, 19, Weil, M. H. and Henning, R. J. (1979) New concepts in the diagnosis and fluid
1252–1265. treatment of circulatory shock. Anesthesia and Analgesia, 58, 124.
Sinnatamby, C., Edwards, C. R. W., Kitau, M. and Irving, M. H. (1974) Weil, M. H., Morissette, M., Michaels, S. et al. (1974) Routine plasma colloid
Antidiuretic hormone response to high and conservative fluid regimens in osmotic pressure measurements. Critical Care Medicine, 2, 229.
patients undergoing operation. Surgery, Gynecology, and Obstetrics, 139, Weil, M. H., Michaels, S., Puri, V. K. et al. (1981) The stat laboratory: facilitating
715–719. blood gas and biochemical measurements for the critically ill and injured.
Slutsky, A. S. (1993) American College of Chest Physicians Consensus American Journal of Clinical Pathology, 76, 34.
Conference. Mechanical ventilation. Chest, 104, 1833–59. Weisberg, L. S. (1989) Pseudohyponatraemia: a reappraisal. American Journal of
Smith, S. D., Cone, J. B., Bowser, B. H. et al. (1982) Cerebral edema following Medicine, 86, 315–318.
acute haemorrhage in a murine model: the role of crystalloid resuscitation. Weiss, H. J. (1967) The effect of clinical dextran on platelet aggregation,
Journal of Trauma, 22(7), 588. adhesion and ADP release in man: in vivo and in vitro studies. Journal of
Solomon, S. M. and Kirby, D. F. (1990) The refeeding syndrome: a review. Laboratory and Clinical Medicine, 69, 37.
Journal of Parenteral and Enteral Nutrition, 14, 90–97. Weissman, C. (1990) The metabolic response to stress: an overview and
Starling, E. H. (1896) On the absorption of fluids from the connective tissue update. Anaesthesiology, 73, 308–327.
spaces. Journal of Physiology (London), 9, 312. Whang, R., Oei, T. O., Aikawa, J. K. et al. (1984) Predictors of clinical
Steinbok, P. and Thompson, G. B. (1978) Metabolic disturbances after head hypomagnesemia. Hypokalemia, hypophosphatemia, hyponatremia and
injury: Abnormalities of sodium and water balance with special reference to hypocalcemia. Archives of Internal Medicine, 144, 1794–1796.
the effects of alcohol intoxication. Neurosurgery, 3(1), 9–15. Williams, G. H. (1986) Hyporeninemic hypoaldosteronism. New England
Sterns, R. H. (1987) Severe symptomatic hyponatraemia: treatment and Journal of Medicine, 1041–1042.
outcome. Annals of Internal Medicine, 107, 656–664. Wilmore, D. W., Smith, R. J., O’Dwyer, S. T. et al. (1988) The gut – a central
Sterns, R. H., Cox, M., Felig, P. U. et al. (1981) Internal potassium balance and organ following surgical stress. Surgery, 104, 917.
the control of the plasma potassium concentration. Medicine, 60, 339–354. Wise, B. L. and Chater, N. (1962) The value of hypertonic mannitol solution in
Sterns, R. H., Riggs, J. E. and Schochet, S. S. Jr (1986) Osmotic demyelination decreasing brain mass and lowering cerebrospinal fluid pressure. Journal of
syndrome following correction of hyponatraemia. New England Journal of Neurosurgery, 19, 1038–1040.
Medicine, 314, 1535. Wisner, D. K., Schuster, L. and Quinn, C. (1990) Hypertonic saline resuscita-
Stocker, R., Glazer, A. N. and Ames, B. N. (1987) Antioxidant activity of tion of head injury: Effects on cerebral water content. Journal of Trauma, 30,
albumin-bound bilirubin. Proceedings of the National Academy of Sciences of 75.
the USA, 84, 5918. Wittmers, L. E., Bartlett, M. and Johnson, J. A. (1976) Estimation of capillary
Stockigt, J. R. (1977) Potassium metabolism. Anaesthesia and Intensive Care, 5, permeability coefficient of inulin in various tissues of the rabbit. Micro-
317–325. vascular Research, 11, 67.
Sutin, K. M., Ruskin, K. J. and Kaufman, B. S. (1992) Intravenous fluid therapy Worthley, L. I. G. (1986) Hyperosmolar coma treated with intravenous sterile
in neurologic injury, in Critical Care Clinics vol. 8(2), (ed. B. S. Kaufman), W. water. A study of three cases. Archives of Internal Medicine, 146, 945–947.
B. Saunders, Philadelphia, PA, pp. 367–408. Worthley, L. I. G., Guerin, M. and Pain, R. W. (1987) For calculating osmolality,
Tannen, R. L. (1986) Potassium disorders, in Fluids and Electrolytes, (eds J. P. the simplest formula is the best. Anaesthesia and Intensive Care, 15,
Kokko and R. L. Tannen), W. B. Saunders, Philadelphia, PA, pp. 199–202.
150–2228. Worthley, L. I. G. and Thomas, P. D. (1986) Treatment of hyponatraemic
Taylor, A. E., Parker, J. C., Kvietys, P. R. et al. (1982) The pulmonary seizures with intravenous, 29.2% saline. British Medical Journal, 292,
interstitium in capillary exchange. Annals of the New York Academy of Science, 168–170.
384, 146. Wray, N. P. and Nicotra, M. B. (1978) Pathogenesis of neurogenic pulmonary
Tormey, W. P. (1990) Central pontine myelinolysis and changes in serum oedema. American Review of Respiratory Disease, 118, 783–786.
sodium. Lancet, 335, 1169. Young, B., Ott, L., Norton, J. et al. (1985) Metabolic and nutritional sequelae in
Tranbaugh, R. F. and Lewis, F. R. (1985) Crystalloid fluid, in Controversies in the nonsteroid-treated head injury patient. Neurosurgery, 17, 784–791.
Trauma Management (Clinics in Emergency Medicine), (eds R. N. Dailey and Zaloga, G. P. (1991) Hypocalcaemic crisis. Critical Care Clinics, 7, 191–200.
M. Callaham), Churchill Livingstone, pp. 121–133. Zaloga, G. P. and Chernow, B. (1986) Hypocalcemia in critical illness. Journal
Vassar, M. J., Perry, C. A., Gannaway, W. L. et al. (1991) 7.5% sodium chloride/ of the American Medical Association, 256(14), 1924–1929.
dextran for resuscitation of trauma patients undergoing helicopter trans- Zaloga, G. P., Chernow, B. and Cook, D. (1985) Assessment of calcium
port. Archives of Surgery, 126, 1065. homeostasis in the critically ill patient: the diagnostic pitfalls of the McLean
Velanovich, V. (1989) Crystalloid versus colloid fluid resuscitation: a meta- Hastings nomogram. Annals of Surgery, 202, 587–594.
analysis of mortality. Surgery, 105, 65. Zaloga, G. P., Chernow, B., Cook, D. et al. (1984) The importance of measured
Verney, E. B. (1954) Water diuresis (John Malet Purser Lecture). Irish Journal of serum ionized calcium levels in critically ill patients. Critical Care Medicine,
Medical Science, 345–377. 12(3), 236.
Virgilio, R. W., Smith, D. E. and Zarins, C. K. (1979) Balanced electrolyte Zornow, M. H., Scheller, M. S., Todd, M. M. et al. (1988) Acute cerebral effects
solutions: Experimental and clinical studies. Critical Care Medicine, 7, 98. of isotonic crystalloid and colloid solutions following cryogenic brain
Virgilio, R. W., Rice, C. L., Smithe, D. E. et al. (1979) Crystalloid vs colloid injury in the rabbit. Anesthesiology, 69, 180.
resuscitation. Is one better? Surgery, 85, 129–139. Zornow, M. H., Todd, M. M. and Moore, S. S. (1987) The acute cerebral effects
Voll, C. L. and Auer, R. N. (1988) The effect of post ischaemic blood glucose of changes in plasma osmolality and oncotic pressure. Anaesthesiology, 67,
levels on ischaemic brain damage in the rat. Annals of Neurology, 24, 936.
638–646. Zwimpfer, T. J. and Moulton, R. J. (1993) Neurologic trauma concerns, in
Wasil, M., Halliwell, B., Hutchison, D. C. et al. (1987) The antioxidant action of Critical Care Clinics, vol. 9(4), (eds D. P. Milzman, B. R. Boulanger and A.
human extracellular fluids. Biochemistry Journal, 243, 219. Rodriguez), W. B. Saunders, Philadelphia, PA, pp. 727–739.

You might also like