You are on page 1of 9

CONCISE REVIEW IN MECHANISMS OF DISEASE

of approximately 0.6 mmol/L. HSA is a small (66 kd)


Albumin: Biochemical globular protein composed of 585 amino acids, with few
Properties and Therapeutic tryptophan or methionine residues but an abundance of
charged residues such as lysine, and aspartic acids and no
Potential prosthetic groups or carbohydrate. X-ray crystallography
has shown albumin to possess a heart-shaped tertiary
structure, but in solution HSA is ellipsoid. Some 67% of
Gregory J. Quinlan,1 Greg S. Martin,2 and the tertiary structure of HSA is composed of ␣-helices.
Timothy W. Evans1 Indeed, the protein is composed of 3 homologous do-
mains (I-III), each containing two sub-domains (A and B)

H
uman serum albumin (HSA) is an abundant mul- composed of 4 and 6 ␣-helices respectively. The sub-
tifunctional non-glycosylated, negatively charged domains move relative to one another by means of flexible
plasma protein, with ascribed ligand-binding and loops provided by proline residues, which helps accom-
transport properties, antioxidant functions, and enzymatic modate the binding of an array of substances, as does the
activities.1 It is synthesized primarily in the liver and is flexibility provided by domain-linking disulfide bridges.
thought to be a negative acute-phase protein. Physiologi- Figure 1 depicts the tertiary structure with bound fatty
cally, albumin is responsible for maintaining colloid osmotic acids. HSA contains 35 cysteine residues, most of which
pressure and may influence microvascular integrity and as- form disulfide bridges (17 in all), contributing to overall
pects of the inflammatory pathway, including neutrophil tertiary structure. However, it also contains 1 free cys-
adhesion and the activity of cell signaling moieties. Clini- teine-derived, redox active, thiol (-SH) group (Cys-34),
cally, albumin has been employed as a plasma expander in which accounts for 80% (500 ␮mol/L) thiols in plasma .
many patient populations, although the evidence from meta The thiol moiety of Cys-34 is reactive and capable of
analyses2,3 and the recently published SAFE investigation4 thiolation (HSA-S-R) and nitrosylation (HSA-S-NO),
suggests it does not afford a survival benefit over crystalloid processes that are thought to contribute to several in vivo
solutions when administered to the critically ill. However, functions.
studies of albumin usage as a volume expander and albumin Physiologically, HSA exists predominantly in a re-
dialysis therapy in patients with liver disease have led to some duced form (that is, with a free thiol, HSA-SH) and is
encouraging results. This review aims to highlight current known as mercaptoalbumin. However, a small but signif-
thinking regarding albumin therapy in the critical care and icant proportion of the albumin pool exists as mixed dis-
hepatological setting and also discusses other potential ther- ulfides (HSA-S-S-R); where (R) represents low-
apeutic applications for its use based around the complex molecular-weight, thiol-containing substances in plasma,
biochemistry of this multifunctional plasma protein. Poten- chiefly cysteine and glutathione. Mixed disulfide forma-
tial contraindications are also discussed. tion also increases as part of the aging process (reviewed by
Droge5) and during disease processes characterized by ox-
Tertiary Structure and Biochemical idative stress. Dimer formation is also theoretically possi-
Characteristics of Albumin ble (HSA-S-S-ASH), but in practice is unlikely to occur in
Albumin normally accounts for over 50% of total vivo because of stearic interference. However, this process
plasma protein content, being present at concentrations is known to occur ex vivo on purification and storage and
therefore may have implications related to certain aspects
of the therapeutic use of albumin.
Abbreviations: HSA, human serum albumin; NO, nitric oxide; ROS, reactive
oxygen species; RNS, reactive nitrogen species; COP, colloid oncotic pressure.
From the 1Department of Critical Care Medicine, Imperial College School of Properties of Albumin
Medicine, Royal Brompton Hospital, London, UK; and the 2Department of Pul-
monary and Critical Care Medicine, Emory University School of Medicine, At-
lanta, GA. Ligand-Binding. HSA binds many endogenous and
Received September 21, 2004; accepted March 21, 2005. exogenous compounds, including fatty acids, metal ions,
Address reprint requests to: Professor TW Evans, Royal Brompton Hospital, pharmaceuticals, and metabolites, with implications for
Sydney Street, London SW3 6NP, United Kingdom. E-mail: t.evans@
rbh.nthames.nhs.uk; fax: (44) 207 351 8524.
drug delivery and efficacy, detoxification, and antioxidant
Copyright © 2005 by the American Association for the Study of Liver Diseases. protection. Several low- and high-affinity ligand-binding
Published online in Wiley InterScience (www.interscience.wiley.com). sites have been identified on HSA, the first of which to be
DOI 10.1002/hep.20720
identified (termed site I and II) are responsible for the
Potential conflict of interest: G.J.Q., G.S.M., and T.W.E. have received non-
directed grants from agencies representing albumin manufacturers (Plasma Protein binding of most pharmaceuticals that interact with the
Therapuetics Association). protein. Sites I and II are located in different domains and
1211
1212 QUINLAN, MARTIN, AND EVANS HEPATOLOGY, June 2005

(NO⫹). Indeed, until recently NO was thought to circu-


late in plasma primarily as an S-nitroso HSA adduct and
to possess vasodilatory properties , augmented by NO
transfer to low-molecular-weight thiols.8 However, recent
in vitro and in vivo studies indicate that levels of s-nitroso-
albumin that form under biologically relevant conditions
in normal plasma are in the low nanomolar range (⬍10
nmol/L) and that several other reaction products of NO
contribute to the NO plasma sink.9 It is less clear to what
extent HSA contributes to NO binding in vivo under
pathological conditions, or to what extent the availability
of catalysts and or other NO-derived species impacts on
s-nitrosolation. Furthermore, recent studies using a tar-
geted s-nitrosoglutathione reductase murine model have
demonstrated the importance of nitroso-thiol turnover in
endotoxic shock.10 Further studies are required to deter-
mine HSA’s role under such circumstances. Key reactions
are summarized in Fig. 3.
Metal Cations. The N-terminal portion of HSA (N-
Fig. 1. Tertiary structure of albumin showing the binding of seven Asp-Ala-His-Lys-) binds Cu, Ni, and Co ions with high
archidonic acid ligands is depicted. Illustration obtained from the RCSB
protein data bank PDB ID:1gnj by David S Goodsell Scripps Research affinity, whereas Au, Ag, and Hg ions bind to cysteine-34
Institute. Primary reference source: Petitpas I, Gruene T, Bhattacharya AA, (reviewed in Kragh-Hansen et al.6) HSA is also the major
Curry S. Crystal structures of human serum albumin complexed with Zn binding protein in plasma, although there is some
monounsaturated and polyunsaturated fatty acids. J Mol Biol
2001;314:955. debate as to the nature of and location of its binding site.11
HSA has also been reported to possess a relatively weak,
nonspecific, latent iron-binding capacity.12 This is, how-
exhibit differential, but not always exclusive, ligand-bind- ever, unlikely to be of significance under normal circum-
ing affinities. Site I tends to bind relatively large hetero- stances in plasma, because the specific, high-affinity, iron-
cyclic compounds or dicarboxylic acids. A diverse array of binding protein transferrin binds all low-molecular-mass
unrelated compounds bind with high affinity to various ferric iron.
locations within this site, indicating adaptability. More- Antioxidant Functions. Aerobic metabolism is en-
over, this site is large and able to bind bulky endogenous ergy efficient. However, whereas oxygen-containing end
substances, including bilirubin and porphyrins. By con- products of these processes are relatively innocuous, many
trast, site II (also known as the indole-benzodiazepine
site) is smaller and less flexible in nature, because binding
is more stereo-specific. Importantly, additional high-af-
finity binding areas are present within HSA for some
drugs and compounds that do not conform to either site I
or II. Furthermore, the binding domains of some sub-
stances such as digitoxin and the bile acids remains to be
elucidated; for a concise review on ligand-binding, see
Kragh-Hansen et al.6
Cysteine-34 binds drugs including cisplatin, D-peni-
cillamine, and N-acetyl-cysteine.6 Covalent interactions
(thiolations) also occur with endogenous, low-molecular-
weight, thiol-containing substances via disulphide bridge
formation. Higher oxidation states of cys-34 can also oc- Fig. 2. Scheme gives an overview of the steps involved (highlighted in
cur, resulting in the formation of sulfenic, sulfinic, or blue) for the nitrosylation of Cys-34 of human serum albumin (HSA).
sulfonic acid residues (Fig. 2), although levels seen in Nitric oxide (NO) requires an electron accepting catalyst (reactive tran-
normal plasma (bovine) are low.7 Both endogenous and sition metal ion, or metal-containing proteins) to favor such reactions.
Steps leading to Cys-34 oxidation and thiolation are highlighted in red.
exogenous nitric oxide (NO) are known to interact with Formation of higher oxidation states of HSA are also shown. RSH,
cys-34 via electrophilic addition of the nitrosonium ion glutathione or free cysteine; Alb, albumin.
HEPATOLOGY, Vol. 41, No. 6, 2005 QUINLAN, MARTIN, AND EVANS 1213

more recent studies, hydrogen peroxide (H2O2) and the


RNS peroxynitrite (ONOO⫺) have been shown to oxi-
dize cys-34 to a sulfenic acid derivative (HSA-SOH).15
This is subsequently converted to a disulfide with the
potential to be redox cycled to mercapto-albumin (HSA-
SH), thereby restoring antioxidant function (Fig. 2). In-
creased ROS and RNS formation have been implicated as
contributory factors in the onset and progression of crit-
ical illness.16 Albumin may provide effective extracellular
scavenging antioxidant protection under such circum-
stances. Thus, albumin supplementation has been shown
to replenish extracellular thiol status in patients with sep-
Fig. 3. Scheme depicts antioxidant (highlighted in blue) and the sis by means other than that which would be expected on
pro-oxidant potential (highlighted in red) of human serum albumin (HSA).
The potential of iron and copper ions to catalyze the formation of the purely stoichiometric grounds.17 Moreover, such supple-
extremely aggressive and damaging hydroxyl radical 䡠 OH (the Fenton mentation was shown to improve thiol-dependent anti-
reaction) is shown. The potential ability of HSA to redox cycle these metal oxidant protection in plasma obtained from patients with
catalysts exacerbates this pro-oxidant response when these metals have
access to Cys-34, in other words, when they are not bound at protected acute lung injury and to be associated with decreased lev-
sites on this protein or elsewhere. As shown, such metal salts also can els of oxidative markers (protein carbonyls),18 although
propagate membrane lipid peroxidation directly if stable lipid peroxides there was no difference in survival rates between groups.
are already present. Nitric oxide and bilirubin binding may provide an
indirect (supportive) antioxidant response attributable to albumin, as Persistent hypoalbuminemia is also associated with per-
both compounds have reported lipid-phase antioxidant function. oxidation of erythrocyte membranes in patients undergo-
ing chronic hemodialysis, indicating that HSA protects
against lipid oxidation.19
intermediates thereby formed are potentially, or directly, In vitro studies have shown that bovine serum albumin
extremely reactive in nature. Such reactive oxygen species scavenges neutrophil-derived ROS, including hydrogen
(ROS) can inflict damage on molecules, leading to the peroxide, superoxide, and hypochlorous acid.20 Inflam-
accumulation of toxic end products and cellular dysfunc- matory cell-derived oxidants contribute to oxidative stress
tion or damage. Normally, the body uses protective (i.e., during acute inflammation and the consequences thereof.
antioxidant) and reparative systems that limit the effects HSA could potentially reduce such effects through scav-
of oxidative stress. An antioxidant is any substance that enging antioxidant actions in humans, which may, also
when present at low levels significantly diminishes or pre- through modifying redox balance, regulate cell signaling
vents the oxidation of an oxidizable substrate, and may be moieties active in mediating pro-inflammatory forces
dietary, constitutive, or inducible in origin. Primary anti- (Fig. 3). In vitro, albumin has been shown to offer anti-
oxidants prevent ROS formation and include the iron- oxidant protection against the oxidative effects of carbon
binding antioxidant transferrin. Secondary antioxidants tetrachloride and uremic toxins,21,22 findings with impli-
scavenge pre-formed ROS. Examples include ascorbate cations for both hepatic and renal failure. HSA may pro-
and superoxide dismutase. For the definitive text on ROS vide a supportive antioxidant role in vivo, through its
in biology, see Gutteridge and Halliwell.13 Reactive nitro- ability to bind and transport substances with known an-
gen species (RNS) are nitrogen-centered species analo- tioxidant function, specifically, bilirubin and NO, which
gous to ROS. Evidence indicates that such species are are effective lipid phase antioxidants23,24 (Fig. 3). Biliru-
formed in vivo; some, such as nitric oxide, contribute to bin may also protect albumin from oxidant-mediated
various biological signaling responses. Others, however, damage.25
are powerful oxidants and nitrating species capable of Metal-Binding. Heme is thought to possess pro-oxi-
damaging biomolecules; antioxidant protection also lim- dant properties through the redox properties of iron. HSA
its the damage inflicted by RNS. Several such antioxidant is an effective heme-binding protein.26 Once bound to
functions have been ascribed to HSA. albumin, such pro-oxidant properties are decreased, indi-
ROS/RNS Scavenging. HSA in plasma, or bovine se- cating an antioxidant function,27 although under physio-
rum albumin in artificial systems, provides protection logical circumstances the heme-binding plasma protein
from lipid peroxidation propagated by inorganic ROS hemopexin provides most of this form of antioxidant pro-
generated from xanthine oxidase/hypoxanthine.14 How- tection.28 Free, or loosely bound, redox-active transition
ever, thiol oxidation occurs, indicating the cys-34 moiety metal ions (low molecular mass) are potentially extremely
to be the source of the antioxidant protection afforded. In pro-oxidant, having the ability to catalyze the formation
1214 QUINLAN, MARTIN, AND EVANS HEPATOLOGY, June 2005

of damaging and aggressive ROS from much more innoc- cycling of iron. Intravenous albumin therefore may be
uous organic and inorganic species (Fig. 3). In strictly inadvisable in circumstances when pronounced extracel-
biological terms the 2 most important such metals are lular iron mobilization or overload are complicating fac-
iron and copper. In specific circumstances (certain disease tors.
states and poisoning), these metal ions can become free of Synthesis and Turnover. In healthy adults, albumin
constraints, which normally limit and control their reac- synthesis occurs predominantly in polysomes of hepato-
tivity. By virtue of its high-affinity copper-binding site, cytes (10-15 g/day) and accounts for 10% of total liver
HSA limits copper-catalyzed oxidative damage to other protein synthesis. Relatively small amounts of albumin
biomolecules by directing damage toward the albumin are hepatologically stored (⬍2 g), the majority being re-
molecule itself in a sacrificial fashion.29 In similar fashion, leased into the vascular space. Approximately 30%-40%
HSA can limit damage caused by accidental biological of albumin synthesized is maintained within the plasma
contamination by redox active metal ions such as vana- compartment. The remaining pool is located within tis-
dium, cobalt, and nickel. Although HSA iron-binding is sues such as muscle and skin. Studies of radiolabeled al-
weak and nonspecific, it may offer antioxidant protection bumin catabolism in normal healthy young adult males
when other specific protective stratagems become over- indicate a variation of half-life of between 12.7 and 18.2
whelmed, such as under conditions of iron overload or days (mean, 14.8 days),36 although a dynamic exchange
pronounced hemolysis (Fig. 3). between plasma and the interstitium occurs. Albumin
Anti-Inflammatory Properties. Evidence indicates leaks from plasma at a rate of 5%/hour and is returned to
that accessible thiol groups can signal inflammatory cell the vascular space at an equivalent rate through the lym-
regulatory changes dependent on their redox state.30 phatic system. Synthesis is a constant process, regulated at
Thus, 25% albumin has been shown to modulate neutro- both transcriptional and posttranscriptional levels by spe-
phil/endothelial cell interactions after shock and resusci- cific stimuli, but change in interstitial colloid oncotic
tation and to attenuate lung injury.31 Furthermore, HSA pressure is thought to be the predominant regulatory in-
augments intracellular glutathione levels and influences fluence.37 Albumin homeostasis is maintained by bal-
activation of the ubiquitous transcription factor nuclear anced catabolism that is not well characterized, occurring
factor-kappa B using both in vitro and in vivo protocols.32 in all tissues. However, most albumin (40%-60%) is de-
Moreover, several recent studies using a rat model of graded in muscle, liver, and kidney. Plasma hyperalbu-
hemorrhagic shock have indicated that the type of resus- minemia is rare, whereas hypoalbuminemia is a feature of
citation fluid administered greatly influences proinflam- a variety of pathological processes, including liver disor-
matory responses, including lung apoptosis and rates of ders, cancer, and severe sepsis. Ascites formation is a com-
neutrophil activation. Plasma albumin was found to be mon complication of cirrhosis and contributes to vascular
the least proinflammatory of the fluids utilized.33 The hypoalbuminemia. Perturbations in hepatic vascular con-
formation of sulfenic acid residues by cys-34 oxidation15 trol are thought to be responsible for ascites formation,
also may be a key factor determining signaling responses, although the precise mechanisms remain a matter of de-
because recent evidence indicates that such groups impact bate (reviewed in Arroyo38). However, cirrhosis in the
on cellular signaling functions, reviewed in Poole et al.34 advanced stages is also characterized by protein wasting
Pro-oxidant Implications. Paradoxically, and in and hence albumin depletion. The reasons for such dys-
common with other redox active antioxidant substances, function remain unresolved, but nutritional, metabolic,
albumin can display pro-oxidant properties, through its and hormonal abnormalities and uncharacterized re-
ability to redox cycle/recycle transition metal ions such as sponses to the release of bioactive substances including
iron and copper from the less reactive (ferric/cupric) to chemokines may contribute to hyopalbuminemia (re-
more pro-oxidant (ferrous/cuprious) states (Fig. 3). Thus, viewed in Tessari39).
a recent study has shown that copper/HSA could become The capillary bed is known to be hyperpermeable in
pro-oxidant after fatty acid binding and subsequent patients with sepsis, thereby leaking albumin. However,
cys-34 oxidation.35 Iron has much less binding affinity for the extent to which altered liver biosynthesis and rates of
HSA and is more likely to be recycled as a free agent able catabolism contribute to the plasma albumin deficiency
to catalyze damaging ROS formation at sites distant from seen in sepsis and critical illness remains uncertain. In-
HSA. Such an action is, therefore, potentially more dele- deed, the half-life of HSA in patients with hypoalbumin-
terious. Indeed, HSA administration was reported re- emia supported with total parenteral nutrition is only 9
cently to be adversely associated with a decline in iron- days, although rates of catabolism are normal.40 More-
binding antioxidant protection in patients with acute over, catabolism actually may be decreased while the ex-
lung injury,18 an effect thought to be related to the redox tracellular pool is increased, suggesting that HSA may be
HEPATOLOGY, Vol. 41, No. 6, 2005 QUINLAN, MARTIN, AND EVANS 1215

spared or protected.41 Studies in healthy individuals given larger meta-analysis published subsequently found no dif-
endotoxin, as well as in the critically ill, indicate that ference in outcome.2 Systematic reviews have attempted
albumin synthesis increases under these circumstances, to elucidate potential reasons as to why albumin would
even though hypoalbuminemia is common.42,43 By con- adversely affect outcome, but have failed to identify the
trast, in animal models of sepsis and endotoxemia, de- mechanism.50 However, in one of the largest clinical trials
creased rates of liver albumin synthesis at the expense of ever conducted in critically ill patients, the SAFE study
acute phase protein synthesis is detectable regardless of randomized 7,000 critically ill patients needing fluid re-
nutritional support.44 Further studies in humans are re- suscitation to receive either 4% albumin or normal saline.
quired to resolve this controversy. There were no differences between groups in survival,
Colloid Oncotic Pressure. HSA is a relatively small measures of morbidity, and organ dysfunction, nor in
protein that accounts for some 75% of protein molecules length of stay in the intensive care unit or hospital.4
in plasma in healthy individuals. Because of its dispropor- Is albumin neither beneficial nor harmful in a thera-
tionate contribution to the plasma protein pool, albumin peutic sense in the critically ill, despite its wide range of
is also responsible for approximately 75% of plasma col- potentially significant properties? We suggest that uncer-
loid oncotic pressure (COP). Oncotic pressure becomes tainty remains for a number of reasons. First, the wide
osmotic pressure as the negative charges surrounding the range of patients with critical illness that have been stud-
protein molecules attract sodium, thus holding water. Its ied to date may conceal specific populations in which
remaining contribution to COP is due to the Donnan HSA may be definitely beneficial or harmful. Indeed,
effect attributable to its overall negative charge. More- HSA is increasingly recognized as a “niche” drug, evi-
over, HSA may influence directly vascular integrity by denced by its ability to significantly improve outcomes in
binding in the interstitial matrix and subendothelium and patients with cirrhosis complicated by spontaneous bac-
reducing the permeability of these layers to large mole- terial peritonitis.51 This is further supported by subgroup
cules, and indirectly through its scavenging properties.
analysis of the SAFE study,4 which has indicated that
Albumin as Therapy in Critical Illness. HSA has
albumin may have varying effects in different patient
long been used to combat vascular collapse in severely ill
groups. From these data, patients with traumatic brain
patients, customarily as an iso-oncotic (4%-5%) solution
injuries may actually be harmed by albumin administra-
for intravascular volume expansion; and as an hyperon-
tion (relative risk for death ⫽ 1.62, P ⫽ .009), whereas
cotic (20%-25%) solution for the maintenance of fluid
sepsis patients may derive benefit (relative risk ⫽ 0.87,
balance between compartments and the restoration of
P ⫽ .06). Second, the use of albumin in all trials pub-
COP. More recently, the benefits of HSA in supporting
the critically ill have been debated, not least because it is lished to date has concentrated on its volume-expanding
expensive compared with synthetic colloid solutions. effects. The total dose administered may therefore be in-
Concerns have been raised regarding the distribution of appropriate if other, non-circulatory properties such as
albumin in states of altered capillary permeability, such as those relating to antioxidant or anti-inflammatory capac-
sepsis. However, clinical trials have confirmed that albu- ities are the desired end point. Such properties might
min remains a potent volume expander compared with account for any disease-specific (i.e., niche) effects of HSA
crystalloid solutions even under these circumstances.45 therapy. Third, albumin may have valuable properties
However, a recent study finding that bolus administra- when used as an adjunct with other therapies, as in hy-
tion of 20% HSA to patients with sepsis led to a signifi- poproteinemic patients with acute lung injury and acute
cantly faster decline in plasma albumin compared with respiratory distress syndrome, where the combination of
healthy controls raises questions about longer-term effi- HSA and furosemide therapy has been shown to improve
cacy of albumin administration.46 Furthermore, in these fluid balance, oxygenation, and hemodynamics.48 The
states of altered capillary permeability, the formation of mechanisms responsible for the benefits observed in acute
edema (such as pulmonary edema) is governed more by lung injury and acute respiratory distress syndrome are
hydrostatic pressure than COP.47 Colloid administration incompletely understood and may be specific to albumin
to patients with acute lung injury does not worsen pul- compared with synthetic colloids. There is no clear evi-
monary edema and may in fact reduce edema-producing dence that pulmonary edema may be influenced by one
forces.48 A variety of meta-analyses have produced con- colloid more than another.47 The ability of HSA to bind
flicting results regarding the risk and benefits of albumin many drugs, sometimes irreversibly, is a complicated issue
administration to critically ill patients. Specifically, the that may impact on combinational therapies.52,53 Fur-
1998 Cochrane Injuries Group concluded that albumin thermore, binding of biologically active moieties such as
administration may increase the risk of death,49 whereas a NO (NO⫹), levels of which become elevated during crit-
1216 QUINLAN, MARTIN, AND EVANS HEPATOLOGY, June 2005

Table 1. Study Findings: Treatment of Cirrhosis by Albumin


Cirrhosis Treatment Albumin Comments

Ascites Paracentesis Plasma volume expansion Paracentesis alone results in pronounced renal impairment
Independent risk for renal in a significant proportion of patients. In combination
impairment with albumin volume expansion, this is almost
completely prevented
Albumin more effective than other volume expanders
Hepatorenal syndrome Vasoconstrictor drugs Plasma volume expansion Long-term albumin treatment together with the
Common complication of Renal replacement therapy administration of arterial vasoconstrictors reverses
advanced cirrhosis hepatorenal syndrome in most patients, use of
resulting in renal vasoconstrictors alone is much less effective
failure
Spontaneous bacterial Non-nephrotoxic antibiotics Combinational use Renal impairment develops in a third of patients treated
peritonitis with antibiotics, but this was substantially decreased if
Risk of renal impairment albumin is given in combination with antibiotics at time
potentiated of diagnosis, mortality was also lower

NOTE. Summarizes findings of studies into the use of intravenous albumin for the treatment of cirrhosis. It is not an exhaustive list. More detailed reviews of current
literature can be found in Arroyo38 and Grange and Amiot69 (used to compile this table).

ical illness, may influence the ability of HSA to bind other posttransplantation complications. The ability of MARS
ligands.54 to remove other toxins and pro-inflammatory stimuli
such as lipopolysaccharides, chemokines, and lipid per-
Albumin as Therapy in Liver Disease oxidation end-products, xenobiotics and free heme/
Historically albumin was used in patients with cirrho- hemoglobin, may have implications for limiting the
sis for vascular volume maintenance, because of its on- inflammatory response. Ample evidence exists of en-
cotic properties. As theories regarding the nature of hanced NO production during both chronic and acute
vascular control and ascites formation developed, and liver failure61 that may relate to adverse clinical events
with a better understanding of the use of diuretics and such as renal impairment and circulatory dysfunction,
other management strategies, the use of albumin for the including hepatopulmonary syndrome. NO is specifi-
treatment of this condition declined. However, it is now cally bound by HSA at position cys-34, MARS treat-
apparent that the volume-expanding properties of albu- ment may therefore also help modulate circulatory NO
min, in combination with other therapeutic approaches, levels during liver failure, thereby protecting against
is of clinical benefit to patients with cirrhosis, thereby the cascade of other organ failures that accompany
reducing the renal impairment that complicates liver dis- acute liver disease. However, as previously stated, the
ease38 (Table 1). levels of s-nitroso-albumin found in healthy plasma
Recently albumin has been used not as a medium for subjected to relevant physiological exposure with NO
administration, but as a part of a hemodialysis regimen in are low. Caution should therefore be used when inter-
patients with hepatic failure, the so-called molecular ad- preting results of MARS trials in this context, with the
sorbent recirculating system (MARS, for a review see Sen need for the true extent of s-nitrosylation to be deter-
et al.55). MARS uses an albumin-containing dialysate that mined under such pathological circumstances. There
is regenerated by dialysis against buffered bicarbonate so- may well be implications for hepatopulmonary syn-
lution subsequent to carbon and anion exchange column drome, although studies in this area are somewhat lim-
treatment to combine the removal of toxins normally ited. However, the MARS system has been used to treat
cleared by the kidneys with those removed by the liver. a small number of patients with ARDS.62
MARS has been used to treat liver dysfunction and failure Contraindications. Recognized contraindications to
in more than 4,000 patients over the last 4 years56 and has albumin therapy include a known allergy to albumin and
been shown to improve renal function and hemodynam- states in which fluid overload could be harmful (decom-
ics, and to decrease brain edema and hepatic encephalop- pensated congestive heart failure or hypertension, severe
athy (reviewed in Mitzner et al.57). HSA avidly binds anemia, etc.). Administration of certain colloids may be
toxins, including bilirubin, copper ions, and protein contraindicated in specific patient populations, such as
breakdown products, substances that accumulate in pri- dextrans or starches in patients with clinically significant
mary liver diseases including cirrhosis,58 hepatitis C infec- coagulopathies, or hetastarch to patients with severe sep-
tion,59 and Wilson’s disease.60 There are also indications sis.63 Whereas albumin has similar colligative properties
for the treatment of secondary liver disease and during to these synthetic colloids, the adverse effect profile is less
HEPATOLOGY, Vol. 41, No. 6, 2005 QUINLAN, MARTIN, AND EVANS 1217

Table 2. Theoretical Pros and Cons Associated with Intravenous HSA Use
Theoretical Benefits Potential Adverse Effects Comments

Intravascular maintenance and volume Fluid overload as plasma volume responds No enhanced outcome benefit compared with other
expansion linearly to levels of HSA administered colloids, but less likely to see adverse drug reactions
Allergic reactions A rare occurrence may be associated with polymer
formation on storage of HSA
Manufacturer and batch variation in albumin Variable levels of reduced thiols present, higher oxidation
for clinical use. states seen. Bound reactive metal ions including
copper, iron, vanadium, and aluminum seen
Drug interactions Drug interactions The ability to bind an array of drugs may in some
instances aid drug transport and delivery but equally
some covalent interactions may be irreversible, thereby
compromising drug efficacy. Binding of other ligands
may also influence drug interactions with HSA
Increased interstitial clearance Enhanced albumin clearance when administered to
patients with sepsis
Removal of endogenous toxins Liver dysfunction: HSA binds bilirubin, copper and nitric
oxide as well as other liver-derived toxins. May also
help remove pro-inflammatory substances such as
chemokines
Removal of exogenous toxins Metal poisoning, and certain drugs
Maintenance of extracellular redox status Plasma thiol repletion
Scavenging antioxidant protection Cys-34–mediated removal of hydrogen peroxide and other
ROS
Heme-binding antioxidant protection High-affinity heme-binding may come into play when other
(hemopexin) antioxidant protection is overwhelmed or
deficient.
Brain trauma Adverse indications
N-terminal metal-binding and antioxidant High-affinity binding of reactive transition metal ions;
protection Copper, cobalt and nickel directs oxidative damage to
albumin rather than other biomolecules
Nonspecific iron-binding and antioxidant Also pro-oxidant implications May offer antioxidant protection when other (transferrin)
protection iron-binding antioxidant protection is overwhelmed or
deficient. However, as HSA is redox active it can
potentially recycle iron to the more catalytically active
ferrous state
Nitric oxide-binding Nitric oxide-binding NO bound to HSA may represent a proportion of an in
vivo sink for this vasoactive gas, the actions of which
may be either beneficial or deleterious dependant on
circumstances and organ type.

NOTE. Treatment of the critically ill are described. This is not an exhaustive list, but it summarizes areas highlighted and cited in this review.

cumbersome, although it could be pro-oxidant under de- effects from colloid administration can be found in a re-
fined circumstances with as yet unknown consequences. cent review.68
Finally, HSA from different manufacturers may differ
markedly in terms of the types of metals bound to it and in Summary
levels of oxidation.64 Albumin employed for clinical use
therefore may differ markedly from endogenous HSA. Human serum albumin (HSA) has many physiological
Such batch differences may influence biochemical prop- and biochemical properties that render it relevant to many
erties, and HSA can thereby vary in its ability to influence aspects of the disordered vascular and cellular functions
adhesion molecule expression from endothelial cells in that characterize the critically ill; particularly those af-
culture.65 Furthermore, dimer and polymer formation on flicted by the systemic inflammatory response syn-
storage may contribute to rare instances of allergic reac- drome—sepsis, severe sepsis, and septic shock. The use of
tions being seen,66 and vanadium contamination of com- albumin as a routine volume expanding agent in the in-
mercial batches of HSA has been shown to adversely tensive care setting cannot be justified in terms of a mor-
influence renal function in patients with coronary heart tality or morbidity advantage over crystalloid solutions.
disease.67 Table 2 describes theoretical/potential implica- However, albumin may be beneficial in specific clinical
tions associated with intravenous albumin administra- circumstances, such as in patients with cirrhosis compli-
tion. A more complete description of possible adverse cated by SBP, and its potential to modulate the inflam-
1218 QUINLAN, MARTIN, AND EVANS HEPATOLOGY, June 2005

matory response is, we suggest, worthy of further 20. Kouoh F, Gressier B, Luyckx M, Brunet C, Dine T, Cazin M, et al.
Antioxidant properties of albumin: effect on oxidative metabolism of hu-
exploration. man neutrophil granulocytes. Farmaco 1999;54:695-699.
21. Wratten ML, Sereni L, Tetta C. Oxidation of albumin is enhanced in the
Acknowledgment: The authors thank the British presence of uremic toxins. Ren Fail 2001;23:563-571.
Heart Foundation, The Dunhill Medical Trust, The 22. Ogasawara Y, Isoda S, Tanabe S. Antioxidant effects of albumin-bound
Garfield Weston Trust and the David Boston Trust for sulfur in lipid peroxidation of rat liver microsomes. Biol Pharm Bull 1999;
their support, and the U.S. National Institutes of Health 22:441-445.
23. Rubbo H, Parthasarathy S, Barnes S, Kirk M, Kalyanaraman B, Freeman
for support via grant HL-67739.
BA. Nitric oxide inhibition of lipoxygenase-dependent liposome and low-
density lipoprotein oxidation: termination of radical chain propagation
References reactions and formation of nitrogen-containing oxidized lipid derivatives.
Arch Biochem Biophys 1995;324:15-25.
1. Peters T. All About Albumin: Biochemistry, Genetics, and Medical Appli- 24. Stocker R, Glazer AN, Ames BN. Antioxidant activity of albumin-bound
cations. San Diego, CA: Academic Press, 1996. bilirubin. Proc Natl Acad Sci U S A 1987;84:5918-5922.
2. Wilkes MM, Navickis RJ. Patient survival after human albumin adminis- 25. Neuzil J, Stocker R. Bilirubin attenuates radical-mediated damage to se-
tration: a meta-analysis of randomized, controlled trials. Ann Intern Med rum albumin. FEBS Lett 1993;331:281-284.
2001;135:149-164. 26. Wardell M, Wang Z, Ho JX, Robert J, Ruker F, Ruble J, et al. The atomic
3. Finfer S, Bellomo R, Boyce N, French J, Myburgh J, Norton R. A com- structure of human methemalbumin at 1.9 A. Biochem Biophys Res Com-
parison of albumin and saline for fluid resuscitation in the intensive care mun 2002;291:813-819.
unit. N Engl J Med 2004;350:2247-2256. 27. Vincent SH, Grady RW, Shaklai N, Snider JM, Muller-Eberhard U. The
4. Finfer S, Norton R, Bellomo R, Boyce N, French J, Myburgh J. The SAFE influence of heme-binding proteins in heme-catalyzed oxidations. Arch
study: saline vs. albumin for fluid resuscitation in the critically ill. Vox Sang Biochem Biophys 1988;265:539-550.
2004;87(Suppl 2):123-131. 28. Gutteridge JM, Smith A. Antioxidant protection by haemopexin of haem-
5. Droge W. Aging-related changes in the thiol/disulfide redox state: impli- stimulated lipid peroxidation. Biochem J 1988;256:861-865.
cations for the use of thiol antioxidants. Exp Gerontol 2002;37:1333- 29. Gutteridge JM, Wilkins S. Copper salt-dependent hydroxyl radical forma-
1345. tion: damage to proteins acting as antioxidants. Biochim Biophys Acta
6. Kragh-Hansen U, Chuang VT, Otagiri M. Practical aspects of the ligand- 1983;759:38-41.
binding and enzymatic properties of human serum albumin. Biol Pharm 30. Haddad JJ. Oxygen homeostasis, thiol equilibrium and redox regulation of
Bull 2002;25:695-704. signalling transcription factors in the alveolar epithelium. Cell Signal 2002;
7. Janatova J, Fuller JK, Hunter MJ. The heterogeneity of bovine albumin 14:799-810.
with respect to sulfhydryl and dimer content. J Biol Chem 1968;243: 31. Powers KA, Kapus A, Khadaroo RG, He R, Marshall JC, Lindsay TF, et al.
3612-3622. Twenty-five percent albumin prevents lung injury following shock/resus-
8. Scharfstein JS, Keaney JF Jr, Slivka A, Welch GN, Vita JA, Stamler JS, et citation. Crit Care Med 2003;31:2355-2363.
al. In vivo transfer of nitric oxide between a plasma protein-bound reservoir 32. Cantin AM, Paquette B, Richter M, Larivee P. Albumin-mediated regula-
and low molecular weight thiols. J Clin Invest 1994;94:1432-1439. tion of cellular glutathione and nuclear factor kappa B activation. Am J
9. Wang X, Tanus-Santos JE, Reiter CD, Dejam A, Shiva S, Smith RD, et al. Respir Crit Care Med 2000;162:1539-1546.
Biological activity of nitric oxide in the plasmatic compartment. Proc Natl 33. Alam HB, Stanton K, Koustova E, Burris D, Rich N, Rhee P. Effect of
Acad Sci U S A 2004;101:11477-11482. different resuscitation strategies on neutrophil activation in a swine model
10. Liu L, Yan Y, Zeng M, Zhang J, Hanes MA, Ahearn G, et al. Essential roles of hemorrhagic shock. Resuscitation 2004;60:91-99.
of S-nitrosothiols in vascular homeostasis and endotoxic shock. Cell 2004; 34. Poole LB, Karplus PA, Claiborne A. Protein sulfenic acids in redox signal-
116:617-628. ing. Annu Rev Pharmacol Toxicol 2004;44:325-347.
35. Gryzunov YA, Arroyo A, Vigne JL, Zhao Q, Tyurin VA, Hubel CA, et al.
11. Masuoka J, Saltman P. Zinc(II) and copper(II) binding to serum albumin:
Binding of fatty acids facilitates oxidation of cysteine-34 and converts
a comparative study of dog, bovine, and human albumin. J Biol Chem
copper-albumin complexes from antioxidants to prooxidants. Arch Bio-
1994;269:25557-25561.
chem Biophys 2003;413:53-66.
12. Loban A, Kime R, Powers H. Iron-binding antioxidant potential of plasma
36. Beeken WL, Volwiler W, Goldsworthy PD, Garby LE, Reynolds WE,
albumin. Clin Sci (Lond) 1997;93:445-451.
Stogsdill R, et al. Studies of I-131-albumin catabolism and distribution in
13. Gutteridge John MC, Halliwell B. Free Radicals in Biology and Medicine.
normal young male adults. J Clin Invest 1962;41:1312-1333.
3rd ed. Oxford: Oxford University Press, 1998.
37. Nicholson JP, Wolmarans MR, Park GR. The role of albumin in critical
14. Radi R, Bush KM, Cosgrove TP, Freeman BA. Reaction of xanthine oxi-
illness. Br J Anaesth 2000;85:599-610.
dase-derived oxidants with lipid and protein of human plasma. Arch Bio- 38. Arroyo V. Review article: albumin in the treatment of liver diseases—new
chem Biophys 1991;286:117-125. features of a classical treatment. Aliment Pharmacol Ther 2002;16(Suppl
15. Carballal S, Radi R, Kirk MC, Barnes S, Freeman BA, Alvarez B. Sulfenic 5):1-5.
acid formation in human serum albumin by hydrogen peroxide and per- 39. Tessari P. Protein metabolism in liver cirrhosis: from albumin to muscle
oxynitrite. Biochemistry 2003;42:9906-9914. myofibrils. Curr Opin Clin Nutr Metab Care 2003;6:79-85.
16. Lang JD, McArdle PJ, O’Reilly PJ, Matalon S. Oxidant-antioxidant bal- 40. Spiess A, Mikalunas V, Carlson S, Zimmer M, Craig RM. Albumin kinet-
ance in acute lung injury. Chest 2002;122:314S-320S. ics in hypoalbuminemic patients receiving total parenteral nutrition. JPEN
17. Quinlan GJ, Margarson MP, Mumby S, Evans TW, Gutteridge JM. Ad- J Parenter Enteral Nutr 1996;20:424-428.
ministration of albumin to patients with sepsis syndrome: a possible ben- 41. Rothschild MA, Oratz M, Schreiber SS. Extravascular albumin. N Engl
eficial role in plasma thiol repletion. Clin Sci (Lond) 1998;95:459-465. J Med 1979;301:497-498.
18. Quinlan GJ, Mumby S, Martin GS, Bernard GR, Gutteridge JM, Evans 42. Barle H, Januszkiewicz A, Hallstrom L, Essen P, McNurlan MA, Garlick
TW. Albumin influences total plasma antioxidant capacity favorably in PJ, et al. Albumin synthesis in humans increases immediately following the
patients with acute lung injury. Crit Care Med 2004;32:755-759. administration of endotoxin. Clin Sci (Lond) 2002;103:525-531.
19. Soejima A, Matsuzawa N, Miyake N, Karube M, Fukuoka K, Nakabayashi 43. Essen P, McNurlan MA, Gamrin L, Hunter K, Calder G, Garlick PJ, et al.
K, et al. Hypoalbuminemia accelerates erythrocyte membrane lipid peroxi- Tissue protein synthesis rates in critically ill patients. Crit Care Med 1998;
dation in chronic hemodialysis patients. Clin Nephrol 1999;51:92-97. 26:92-100.
HEPATOLOGY, Vol. 41, No. 6, 2005 QUINLAN, MARTIN, AND EVANS 1219

44. O’Leary MJ, Koll M, Ferguson CN, Coakley JH, Hinds CJ, Preedy VR, et during treatment of liver failure with albumin dialysis MARS: a review of
al. Liver albumin synthesis in sepsis in the rat: influence of parenteral clinical, biochemical, and electrophysiological data. Metab Brain Dis
nutrition, glutamine and growth hormone. Clin Sci (Lond) 2003;105: 2002;17:463-475.
691-698. 58. Pares A, Cisneros L, Salmeron JM, Caballeria L, Mas A, Torras A, et al.
45. Ernest D, Belzberg AS, Dodek PM. Distribution of normal saline and 5% Extracorporeal albumin dialysis: a procedure for prolonged relief of intrac-
albumin infusions in septic patients. Crit Care Med 1999;27:46-50. table pruritus in patients with primary biliary cirrhosis. Am J Gastroenterol
46. Margarson MP, Soni NC. Changes in serum albumin concentration and 2004;99:1105-1110.
volume expanding effects following a bolus of albumin 20% in septic 59. Doria C, Mandala L, Smith J, Vitale CH, Lauro A, Gruttadauria S, et al.
patients. Br J Anaesth 2004;92:821-826. Effect of molecular adsorbent recirculating system in hepatitis C virus-
47. Lewis CA, Martin GS. Understanding and managing fluid balance in related intractable pruritus. Liver Transplant 2003;9:437-443.
patients with acute lung injury. Curr Opin Crit Care 2004;10:13-17. 60. Sen S, Felldin M, Steiner C, Larsson B, Gillett GT, Olausson M, et al.
48. Martin GS, Mangialardi RJ, Wheeler AP, Dupont WD, Morris JA, Ber- Albumin dialysis and Molecular Adsorbents Recirculating System (MARS)
nard GR. Albumin and furosemide therapy in hypoproteinemic patients for acute Wilson’s disease. Liver Transpl 2002;8:962-967.
with acute lung injury. Crit Care Med 2002;30:2175-2182. 61. Leifeld L, Fielenbach M, Dumoulin FL, Speidel N, Sauerbruch T, Spen-
49. Human albumin administration in critically ill patients: systematic review gler U. Inducible nitric oxide synthase (iNOS) and endothelial nitric oxide
of randomised controlled trials. Cochrane Injuries Group Albumin Re- synthase (eNOS) expression in fulminant hepatic failure. J Hepatol 2002;
viewers. BMJ 1998;317:235-240.
37:613-619.
50. Choi PT, Yip G, Quinonez LG, Cook DJ. Crystalloids vs. colloids in fluid
62. Peek GJ, Killer HM, Sosnowski MA, Firmin RK. Modular extracorpo-
resuscitation: a systematic review. Crit Care Med 1999;27:200-210.
real life support for multiorgan failure patients. Liver 2002;22(Suppl
51. Sort P, Navasa M, Arroyo V, Aldeguer X, Planas R, Ruiz-del-Arbol L, et al.
2):69-71.
Effect of intravenous albumin on renal impairment and mortality in pa-
63. Schortgen F, Lacherade JC, Bruneel F, Cattaneo I, Hemery F, Lemaire F,
tients with cirrhosis and spontaneous bacterial peritonitis. N Engl J Med
et al. Effects of hydroxyethylstarch and gelatin on renal function in severe
1999;341:403-409.
sepsis: a multicentre randomised study. Lancet 2001;357:911-916.
52. Bertucci C, Domenici E. Reversible and covalent binding of drugs to
human serum albumin: methodological approaches and physiological rel- 64. Quinlan GJ, Coudray C, Hubbard A, Gutteridge JM. Vanadium and
evance. Curr Med Chem 2002;9:1463-1481. copper in clinical solutions of albumin and their potential to damage
53. De Paepe P, Belpaire FM, Buylaert WA. Pharmacokinetic and pharmaco- protein structure. J Pharm Sci 1992;81:611-614.
dynamic considerations when treating patients with sepsis and septic 65. Nohe B, Dieterich HJ, Eichner M, Unertl K. Certain batches of albumin
shock. Clin Pharmacokinet 2002;41:1135-1151. solutions influence the expression of endothelial cell adhesion molecules.
54. Kashiba-Iwatsuki M, Miyamoto M, Inoue M. Effect of nitric oxide on the Intensive Care Med 1999;25:1381-1385.
ligand-binding activity of albumin. Arch Biochem Biophys 1997;345:237- 66. Doweiko JP, Nompleggi DJ. Use of albumin as a volume expander. JPEN
242. J Parenter Enteral Nutr 1991;15:484-487.
55. Sen S, Jalan R, Williams R. Liver failure: basis of benefit of therapy with the 67. Heinemann G, Braun S, Overbeck M, Page M, Michel J, Vogt W. The
molecular adsorbents recirculating system. Int J Biochem Cell Biol 2003; effect of vanadium-contaminated commercially available albumin solu-
35:1306-1311. tions on renal tubular function. Clin Nephrol 2000;53:473-478.
56. Steiner C, Mitzner S. Experiences with MARS liver support therapy in liver 68. Evidence-based colloid use in the critically ill: American Thoracic Society
failure: analysis of 176 patients of the International MARS Registry. Liver Consensus Statement. Am J Respir Crit Care Med 2004;170:1247-1259.
2002;22(Suppl 2):20-25. 69. Grange JD, Amiot X. Nitric oxide and renal function in cirrhotic patients
57. Mitzner S, Loock J, Peszynski P, Klammt S, Majcher-Peszynska J, with ascites: from physiopathology to practice. Eur J Gastroenterol Hepa-
Gramowski A, et al. Improvement in central nervous system functions tol 2004;16:567-570.

You might also like