Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

S P E C I A L F E A T U R E

R e v i e w

Endocrine Responses to Critical Illness: Novel Insights


and Therapeutic Implications

Downloaded from https://academic.oup.com/jcem/article/99/5/1569/2537306 by National Science & Technology Library user on 10 March 2023
Eva Boonen and Greet Van den Berghe
Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular
Medicine, KU Leuven, B-3000 Leuven, Belgium

Context: Critical illness, an extreme form of severe physical stress, is characterized by important
endocrine and metabolic changes. Due to critical care medicine, survival from previously lethal
conditions has become possible, but many patients now enter a chronic phase of critical illness. The
role of the endocrine and metabolic responses to acute and prolonged critical illness in mediating
or hampering recovery remains highly debated.

Evidence Acquisition: The recent literature on changes within the hypothalamic-pituitary-thyroid


axis and the hypothalamic-pituitary-adrenal axis and on hyperglycemia in relation to recovery from
critical illness was critically appraised and interpreted against previous insights. Possible thera-
peutic implications of the novel insights were analyzed. Specific remaining questions were
formulated.

Evidence Synthesis: In recent years, important novel insights in the pathophysiology and the
consequences of some of these endocrine responses to acute and chronic critical illness were
generated. Acute endocrine adaptations are directed toward providing energy and substrates for
the vital fight-or-flight response in a context of exogenous substrate deprivation. Distinct endo-
crine and metabolic alterations characterize the chronic phase of critical illness, which seems to be
no longer solely beneficial and could hamper recovery and rehabilitation.

Conclusions: Important novel insights reshape the current view on endocrine and metabolic re-
sponses to critical illness and further clarify underlying pathways. Although many issues remain
unresolved, some therapeutic implications were already identified. More work is required to find
better treatments, and the optimal timing for such treatments, to further prevent protracted
critical illness, to enhance recovery thereof, and to optimize rehabilitation. (J Clin Endocrinol
Metab 99: 1569 –1582, 2014)

ritical illness is defined as any life-threatening condi- are presumably directed toward providing the required
C tion requiring support of vital organ functions to
prevent imminent death. This condition can be evoked by
energy for the fight-or-flight response in a context of ex-
ogenous substrate deprivation. Indeed, alterations within
a variety of insults such as multiple trauma, complicated the different hypothalamic-pituitary axes bring about li-
surgery, and severe medical illnesses. Without modern polysis, proteolysis, and gluconeogenesis and redirect en-
critical care medicine, critically ill patients would not sur- ergy consumption toward those processes that mediate
vive. Critical illness is thus the ultimate form of severe acute survival, whereas anabolism is postponed to more
physical stress, and all the immediate biological responses prosperous times.
that are evoked are expected to be of greater magnitude in Although survival from previously lethal conditions is
critically ill patients. These immediate stress responses nowadays possible, often recovery does not swiftly follow,
comprise many orchestrated endocrine adaptations that and patients enter a chronic phase of critical illness during

ISSN Print 0021-972X ISSN Online 1945-7197 Abbreviations: CBG, cortisol binding globulin; D1, type 1 deiodinase; D2, type 2 deiodi-
Printed in U.S.A. nase; D3, type 3 deiodinase; GR, glucocorticoid receptor; ICU, intensive care unit; SIRS,
Copyright © 2014 by the Endocrine Society systemic inflammatory response syndrome; TR, thyroid hormone receptor.
Received November 15, 2013. Accepted February 6, 2014.
First Published Online February 11, 2014

doi: 10.1210/jc.2013-4115 J Clin Endocrinol Metab, May 2014, 99(5):1569 –1582 jcem.endojournals.org 1569
1570 Boonen and Van den Berghe Novel Insights on Endocrine Changes in the ICU J Clin Endocrinol Metab, May 2014, 99(5):1569 –1582

which they continue to depend upon vital organ support


for weeks, whereas the original trigger of the critical illness
has long been resolved. This stage is characterized by dis-
tinct endocrine and metabolic alterations that may no lon-
ger be solely beneficial because they may hamper recovery.
An example is the relative maintenance of fat stores while

Downloaded from https://academic.oup.com/jcem/article/99/5/1569/2537306 by National Science & Technology Library user on 10 March 2023
large amounts of proteins continue to be wasted from skel-
etal muscle and organs (1). This response may impair re-
covery of vital organ functions, extend weakness, hamper
rehabilitation (2), and expose patients to severe, often in-
fectious, complications (3). The understanding of the
mechanisms determining why certain patients recover and
others don’t remains very limited, but recent studies point
to variable abilities to remove cell damage as playing a key
role (4, 5). When patients remain dependent upon critical
care support, it is ultimately decided to withdraw care
because of futility. Hence, further understanding the un-
derlying pathways of recovery and investigating whether
these pathways can be beneficially affected by treatment is
of high clinical relevance.
In recent years, important novel insights in the patho-
physiology and the consequences of these endocrine re-
sponses to critical illness were generated. This review sum-
marizes these insights with a specific focus on the
hypothalamic-pituitary-thyroid axis, the hypothalamic-
pituitary-adrenal axis, and the impact of the hyperglyce-
mic response on recovery from critical illness. Any thera-
peutic implications of these novel insights are critically
analyzed. Figure 1. Changes in the central and peripheral thyroid axis in acute
vs prolonged critical illness. The top panel shows reduced TRH gene
expression in the hypothalamus of prolonged ill patients. The second
panel illustrates adaptations in nocturnal TSH secretion with a loss of
Hypothalamic-Pituitary-Thyroid Axis pulsatility during prolonged critical illness. The lower panels summarize
schematically the changes in circulating thyroid hormone
Responses within the hypothalamic-pituitary- concentrations and changes in peripheral deiodinase enzyme activity
thyroid axis during acute critical illness levels. [Figure was drafted from original data in E. Fliers et al:
Decreased hypothalamic thyrotropin-releasing hormone gene
It has long been known that both fasting and acute expression in patients with nonthyroidal illness. J Clin Endocrinol
illnesses immediately affect circulating levels of thyroid Metab. 1997;82:4032– 4036 (26). © The Endocrine Society. Y.
hormones. Most typically, plasma concentrations of T3 Debaveye et al: Regulation of tissue iodothyronine deiodinase activity
in a model of prolonged critical illness. Thyroid. 2008;18:551–560 (46),
decrease and plasma concentrations of rT3 rise, suggesting with permission. © American Thyroid Association. I. Vanhorebeek et al:
an immediate inactivation of thyroid hormone in periph- Endocrine aspects of acute and prolonged critical illness. Nat Clin Pract
eral tissues such as the liver, likely mediated by a sup- Endocrinol Metab. 2006;2:20 –31 (125), with permission. © Macmillan
Publishers Limited. L. Mebis et al: Thyroid axis function and dysfunction
pressed activity of the type 1 deiodinase (D1) and/or an in critical illness. Best Pract Res Clin Endocrinol Metab. 2011;25:745–
activated type 3 deiodinase (D3) (6, 7). Concentrations of 757 (126), with permission. © Elsevier Ltd.
T4 and TSH have been shown to be briefly increased im-
mediately after surgery (7). Thereafter, plasma TSH and Several possible mediators of the acute fall in plasma T3
T4 concentrations often return to “normal,” although a concentrations in critically ill patients include the lack of
normal nocturnal TSH surge is absent (8, 9). This con- nutrients, the release of cytokines, or hypoxia (10 –12).
stellation of low plasma T3 concentrations and elevated TNF-␣, IL-1, and IL-6 are capable of mimicking the acute
rT3 is generally referred to as the acute low-T3 syndrome, stress-induced alterations within the thyroid axis. How-
the euthyroid-sick syndrome, or the nonthyroidal illness ever, neutralizing antibodies to these cytokines in a human
syndrome (Figure 1). experiment of lipopolysaccharide-induced inflammation
doi: 10.1210/jc.2013-4115 jcem.endojournals.org 1571

failed to restore normal thyroid hormone concentrations sampling revealed that the pulsatility of TSH secretion is
(13). Acute decreases in plasma concentrations of thyroid virtually lost, which relates to low plasma thyroid hor-
hormone binding proteins and the inhibition of hormone mone levels, a presentation resembling central hypothy-
binding, transport, and metabolism by elevated levels of roidism (Figure 1) (25). In line with this interpretation,
free fatty acids and bilirubin may also play a role (14). Fliers et al (26) demonstrated in postmortem brain sam-
The low T3 concentrations that occur with fasting have ples of chronic critically ill patients that the gene expres-

Downloaded from https://academic.oup.com/jcem/article/99/5/1569/2537306 by National Science & Technology Library user on 10 March 2023
been shown to be adaptive because they appear to protect sion of TRH in the hypothalamic paraventricular nuclei
the organism against the deleterious catabolic conse- was much lower than in patients who died after acute
quences of a lack of macronutrients (15, 16). In critical insults (Figure 1). Furthermore, a positive correlation was
illness, it was suggested that the low T3 concentrations observed between the TRH mRNA expression and the
could be maladaptive because the magnitude of the acute plasma concentrations of TSH and T3. Together, these
T3 decrease was associated with the severity of illness and data indicate that production and/or release of thyroid
with the risk of death (17, 18). However, the acute fall in hormones is reduced in prolonged critical illness due to
circulating levels of thyroid hormone in response to illness reduced hypothalamic stimulation of the thyrotropes, in
could also be an adaptive attempt to reduce energy ex- turn leading to reduced stimulation of the thyroid gland.
penditure, as happens with fasting in healthy subjects, in The observation that a rise in TSH levels precedes the onset
which case it should be left untreated (15). Improved post- of recovery from severe illness further supports this inter-
operative cardiac function was observed after short-term pretation (27).
iv administration of T3 to patients during elective cardiac The factors triggering hypothalamic suppression dur-
surgery (19, 20). However, supranormal plasma T3 con- ing prolonged critical illness are unknown. Because
centrations were evoked, and thus it is uncertain whether plasma cytokine concentrations are usually much lower in
these findings were merely due to a pharmacological ef- the prolonged phase of critical illness (28), other mecha-
fect. Recently, the results of a large randomized controlled nisms likely play a role, like endogenous dopamine or el-
trial investigating the impact of early parenteral nutrition, evated cortisol levels in the hypothalamus, because exog-
as compared with tolerating pronounced caloric deficit in enous dopamine and hydrocortisone are known to
critically ill patients, provided indirect evidence for an provoke or aggravate hypothyroidism in critical illness
adaptive nature of the low T3 levels (21, 22). This study (29 –31). A local increase in type 2 deiodinase (D2) activity
revealed that providing nutrition in the acute phase of in the hypothalamus could elevate local thyroid hormone
critical illness impaired rather than improved outcome. levels, whereby the setpoint for feedback inhibition could
The provision of macronutrients partially prevented the be altered (32). Indeed, in a rabbit model of prolonged
acute thyroid hormone changes, which were also recently critical illness and low thyroid hormone plasma concen-
observed in a rabbit model of critical illness (23). In the trations, hypothalamic TRH mRNA was low and D2
clinical study, specifically, the rise in T3 and in the ratio of mRNA was high. However, the hypothalamic T4 and T3
T3 over rT3 with early forceful feeding statistically ex- concentrations were not increased (33). Increased pitu-
plained the worsening of the outcome (22). These data itary D2 could also play a role in suppressing local TSH
therefore suggest that at least part of the acute fall in T3 mRNA (34), although this was not confirmed in an animal
concentrations during critical illness is related to the con- model of prolonged critical illness (35).
comitant fasting, and that this part of the response is likely During prolonged critical illness, peripheral tissues
adaptive. Benefits include the expected reduction in energy seem to respond to low T3 levels to increase local hormone
expenditure with low T3 levels, or a direct effect of in- availability and effects. For example, in skeletal muscle
creased D3 activity locally in granulocytes, which could and liver biopsies from prolonged critically ill patients, the
optimize bacterial killing capacity (12, 24). monocarboxylate transporter MCT-8 was overexpressed
(Figure 2) (36). This was confirmed in an animal model,
Responses within the hypothalamic-pituitary- where the up-regulation of the monocarboxylate trans-
thyroid axis during prolonged critical illness porters in liver and kidney was reversible by treatment
However, when patients are treated in intensive care with thyroid hormones (36, 37). Also, in skeletal muscle
units (ICUs) for several weeks, receiving full enteral and/or biopsies from prolonged critically ill patients, D2 expres-
parenteral nutrition, the alterations within the thyroid sion and activity were up-regulated as compared with
axis appear different. In this phase of critical illness, low healthy controls and with acutely ill patients (Figure 1)
plasma T3 concentrations now coincide with low T4 con- (37). Up-regulation of D2 in lungs was recently found to
centrations and low-normal TSH concentrations in a sin- be adaptive in sepsis and acute lung injury, further accen-
gle morning sample (25). Moreover, overnight repeated tuated by the observation that a D2 polymorphism was
1572 Boonen and Van den Berghe Novel Insights on Endocrine Changes in the ICU J Clin Endocrinol Metab, May 2014, 99(5):1569 –1582

longed critically ill patients (39). Together, the data sug-


Plasma Hormone Concentrations gest that when the production of thyroid hormones falls in
160
2.0 prolonged critical illness, peripheral tissues adapt by in-
T3 (nmol/l)

120 creasing thyroid hormone transporters, local activation of

T4 (nmol/l)
1.5
thyroid hormone, and gene expression of the active recep-
1.0 * 80 * tor isoform.
HUMAN PATIENTS

Downloaded from https://academic.oup.com/jcem/article/99/5/1569/2537306 by National Science & Technology Library user on 10 March 2023
0.5 40 In protracted critical illness, low T3 levels were found to
0.0 0 correlate inversely with markers of muscle breakdown and
of bone loss, which could indicate either an adaptive and
Tissue Expression protective response against catabolism or a causal mal-
adaptive relationship (40). Because the cause of the low
Liver Muscle
2.0 * 12 thyroid hormone levels during prolonged critical illness
*
MCT 8 mRNA

10 appears to be a suppressed TRH expression, and therefore


1.5
8 reduced thyroid hormone production, the question could
1.0 6 be addressed by assessing the effect of TRH treatment.
4 When patients were given a TRH infusion, plasma T3 and
0.5 T4 could be increased, but rT3 concentrations also rose
2
0.0 0 (41). However, when TRH was combined with a GH-
secretagogue, this rise in rT3 was prevented, explained by
* * a GH-mediated effect on the inactivating D3 (42). This
3.0 3.0
MCT8 mRNA

treatment also induced an anabolic response, which sug-


gested a causal relationship between low thyroid hormone
2.0 2.0
levels and the impaired anabolism during prolonged crit-
ical illness (40). Furthermore, the negative feedback ex-
RABBIT MODEL

1.0 1.0
erted by thyroid hormones on the thyrotropes was found
to be maintained during TRH infusion, a self-limitation
0.0 0.0
that precludes overstimulation of the thyroid axis (41, 43).
* *
3.0 3.0 Diagnostic implications
MCT10 mRNA

Given the nature of the changes within the thyroid axis


2.0 * 2.0 evoked by critical illness, the diagnosis of pre-existing thy-
* roid disease during critical illness is very difficult. Patients
1.0 1.0 with pre-existing primary hypothyroidism are expected to
reveal low serum levels of T4 and T3 in combination with
0.0 0.0 high TSH concentrations. However, when primary hypo-
Figure 2. The top panel represents the circulating thyroid hormone thyroidism and severe nonthyroidal critical illness coin-
parameters in acutely stressed (light gray bars, n ⫽ 22) and chronically cide, TSH levels may be lower than anticipated. Moreover,
ill (dark gray bars, n ⫽ 64) patients. The white bars designate the
serum TSH may be paradoxically low because of iatro-
normal ranges. The second panel shows the relative MCT8 mRNA
expression levels measured in liver and skeletal muscle of acutely genic factors such as iodine wound dressings, iodine-con-
stressed (light gray) and chronically ill (dark gray) patients. The lower taining contrast agents, and drugs such as high-dose cor-
panels represents the relative expression levels of MCT8 and MCT10 in
ticosteroids, dopamine, somatostatin, and amiodarone
liver and muscle of healthy control rabbits (white bars), saline-treated
prolonged ill rabbits (dark gray bars), and T3⫹T4 treated (black bars) ill (30, 44). So, a normal or low TSH during critical illness
rabbits. Data are expressed as mean ⫾ SEM. *, P ⬍ .05 vs acute does not exclude primary hypothyroidism. Also, the low
values. [Figure was drafted from original data in L. Mebis et al: T4 and T3 levels in patients with severe hypothyroidism
Expression of thyroid hormone transporters during critical illness. Eur J
Endocrinol. 2009;161:243–250 (36), with permission. © European can be indistinguishable from those values observed in
Society of Endocrinology.] prolonged nonthyroidal critical illness. A high ratio of
T3/T4 in serum, a low thyroid hormone-binding ratio, and
associated with less sepsis susceptibility (38). At the level a low serum rT3 may favor the presence of primary hy-
of the thyroid hormone receptor (TR), an inverse corre- pothyroidism. However, the diagnostic accuracy is lim-
lation was observed between the active TR-1/inactive ited. In these patients, history, physical examination, and
TR-2 ratio, a surrogate marker of thyroid hormone sen- the possible presence of thyroid autoantibodies may give
sitivity, and the ratio of T3/rT3 in liver biopsies of pro- further clues to the presence of thyroid disease. Repeated
doi: 10.1210/jc.2013-4115 jcem.endojournals.org 1573

thyroid function tests after improvement of the nonthy- loading dose of 300 –500 ␮g of T4 to quickly reach 50%
roidal illness are required to confirm the diagnosis. of the euthyroid value of T4 (53–55), followed by 50 –100
Elevated plasma T4 and T3 concentrations are so un- ␮g of iv T4 daily until oral medication can be given. Some
usual during critical illness that they should always raise authors have suggested the use of a co-infusion of the
concern of pre-existing hyperthyroidism. However, unde- biologically active form of T3 and T4. Escobar-Morreale et
tectable TSH has no diagnostic value for hyperthyroidism al (56) showed in an animal study that T4 alone did not

Downloaded from https://academic.oup.com/jcem/article/99/5/1569/2537306 by National Science & Technology Library user on 10 March 2023
during critical illness. ensure euthyroidism in all tissues, which was achieved by
combined treatment with T4 and T3. An experimental pro-
Therapeutic implications tocol for thyroid hormone therapy during prolonged in-
Because the available evidence now indicates that the tensive care of presumed hypothyroidism advises admin-
acute “low T3 syndrome” appears to be an adaptive re- istering a 100- to 200-␮g bolus of T4 iv per 24 hours alone
sponse partially explained by fasting, treatment is likely or, when required to also increase plasma T3, combined with
not indicated (22, 23). In contrast, the low T4 and T3 levels T3 at 0.6 ␮g/kg ideal body weight per 24 hours in a contin-
during the prolonged phase of critical illness could be mal- uous iv infusion, targeting serum thyroid hormone levels in
adaptive. Experimental studies showed that in animal the low-normal range (57). When the patients start to re-
models of prolonged critical illness and in prolonged crit- cover, a prompt tapering of this dose may be required.
ically ill patients who are receiving nutrition, the syn- The treatment for primary hyperthyroidism is less af-
drome can be reversed via hypothalamic-releasing factors, fected by concomitant critical illness, except that treat-
with an anabolic response at the tissue level (40). How- ment requirements could be lower in the presence of in-
ever, the effect on clinical outcome of such a treatment creased thyroid hormone metabolism. Furthermore, when
remains to be investigated, so therapeutic implications are patients are receiving active treatment for hyperthyroid-
currently lacking. ism, they should be monitored because of potential tox-
An alternative option for treatment could be the ad- icity of the medication and the impact of other frequently
ministration of thyroid hormones T4 or T3 or the combi- used medication on thyroid hormone levels.
nation to normalize the plasma concentrations. In animal
studies, substitution doses of T4, T3, or their combination
were unable to alter circulating levels of thyroid hor- Hypothalamic-Pituitary-Adrenal Axis
mones, likely explained by the increased metabolism of
thyroid hormones during critical illness, perhaps in part Responses within the hypothalamic-pituitary-
mediated by sulfo-conjugation as was also shown in pa- adrenal axis during acute and prolonged critical
tients (45– 48). Three times the substitution dose of T4 illness
normalized plasma T3 concentrations in this model but The stress hormone cortisol is an essential component
resulted in supranormal T4 levels and a rise in rT3. A dose of the fight-or-flight reaction to the stress of illness and
of T3 that was able to normalize the plasma T3 concen- trauma, and both very high and low cortisol levels have
trations, five times the substitution dose, suppressed TSH been associated with the risk of death in such patients (58).
and T4 to subnormal levels via negative feedback inhibi- Whenever the brain senses a stressful event, activation of
tion. A combination of these doses of T4 and T3 resulted the hypothalamic-pituitary-adrenal axis initiates the re-
in dramatic overtreatment. Similar dosing issues were lease of the CRH and arginine vasopressin from the hy-
present in the few available small randomized studies in pothalamus, which stimulates the anterior pituitary cor-
critically ill patients, which also did not show outcome ticotrophs to secrete ACTH. High cortisol levels during
benefits (49 –52). critical illness likely contribute to the provision of extra
When and how to treat primary hypothyroidism during energy to vital organs by acutely shifting carbohydrate,
critical illness also remains controversial . When patients fat, and protein metabolism and by delaying anabolism.
were receiving active treatment for hypothyroidism before Moreover, cortisol likely affects the hemodynamic system
critical illness, it seems wise to continue their usual dose of by intravascular fluid retention and by enhancing inotro-
thyroid hormone. For myxedema coma, it is generally ac- pic and vasopressor responses, respectively, to cat-
cepted that patients should be treated with parenteral in- echolamines and angiotensin II. In addition, the anti-in-
fusion of thyroid hormones. However, the proper initia- flammatory effects of cortisol can be interpreted as an
tion of replacement therapy during other types of critical attempt to prevent overactivation of the inflammatory
illnesses remains controversial. There is no consensus on cascade (59, 60).
the type of thyroid hormone or on the optimal initial dose During critical illness, plasma cortisol concentrations
for replacement therapy. Many clinicians prefer a high iv are substantially elevated, which is traditionally explained
1574 Boonen and Van den Berghe Novel Insights on Endocrine Changes in the ICU J Clin Endocrinol Metab, May 2014, 99(5):1569 –1582

by severalfold elevated cortisol production in the adrenal work that used a state of the art cortisol-tracer technique
cortex driven by ACTH. However, Vermes et al (61) re- showed that daytime cortisol production during critical
ported only transiently elevated ACTH concentrations in illness was only slightly higher than in healthy subjects.
patients with multiple trauma or sepsis, whereas cortisol Furthermore, cortisol production was only increased in
concentrations remained high. This was recently con- patients with excessive inflammation, whereas it was un-
firmed in a more heterogeneous critically ill patient pop- altered in other critically ill patients (Figure 3) (62). Cor-

Downloaded from https://academic.oup.com/jcem/article/99/5/1569/2537306 by National Science & Technology Library user on 10 March 2023
ulation. In this study (62), plasma ACTH concentrations tisol breakdown on the other hand was substantially
were found to be suppressed already from ICU admission reduced, irrespective of the inflammatory status, attribut-
onward and stayed below the lower limit of normality able to suppressed expression and activity of A-ring re-
throughout the first week of critical illness. It remains un- ductases in the liver and by suppressed activity of 11␤-
known whether the expected initial ACTH rise in response hydroxysteroid dehydrogenase type 2 in kidney (62). It
to stress was missed in this study and had already occurred remains unclear, however, what is driving the suppression
before ICU admission, for example, in the operating room of these enzymes, but an inverse correlation between ele-
or emergency department. vated plasma concentrations of bile acids and the expres-
Low plasma ACTH in the presence of high plasma cor- sion level of the A-ring reductases could point to bile acids
tisol concentrations has been interpreted as non-ACTH- playing a role (Figure 3) (62, 64). Indeed, bile acids are
driven cortisol production, among which cytokines could potent inhibitors of the cortisol-metabolizing enzymes,
play a role (61, 63). Alternatively, this constellation could both via competitive inhibition and by suppression of gene
be caused by reduced cortisol breakdown suppressing the and protein expression (65– 67).
production of adrenocortical hormones via feedback in- The concept of increasing the bioavailability of cortisol
hibition. In fact, direct evidence of increased cortisol pro- levels primarily in tissues that produce these enzymes, and
duction during critical illness has been lacking. Recent to a lesser extent in the circulation, could be interpreted as

A B
Cortisol Production (mg/h)

60 mg / day P=0.01 P=0.03


of D4-cortisol (liter/min)

4
.6
Plasma Clearance

3.5
3 P=0.34
2.5 .4
30 mg / day 2
1.5
.2
1
.5
0 0
Controls No SIRS SIRS Controls Patients

C P<0.001 D P<0.001 E
1.6 5 P<0.001
5β-reductase protein (10log)

R²=0.36
5β–reductase protein

4 1
5β-reductase mRNA

1.2

3 0
.8
2
-1
.4
1
-2
0 0
Controls Patients Controls Patients -0.25 0.25 0.75 1.25 1.75 2.25
Total Bile Acids (μmol/liter) (10log)
Figure 3. A, Cortisol production in critically ill patients with the SIRS (n ⫽ 7; dark gray bar) and no SIRS (n ⫽ 4; light gray bar) compared to
controls (n ⫽ 9; white bar). Based on these results, 24-hour cortisol production was estimated and depicted with arrows. B, Cortisol plasma
clearance as assessed with a small dose of deuterated-cortisol tracer. Bar charts represent means and SE values. C–E, mRNA and protein expression
of 5␤-reductase in liver of 20 controls (white bars) and 44 patients (gray bars) and the relation to plasma total bile acid concentrations. Bar charts
represent means and SE values. The mRNA data are expressed, normalized to glyceraldehyde-3-phosphate dehydrogenase, as a fold difference
from the mean of the controls. Protein data are expressed, normalized for CK-18 protein expression, as a fold difference from the mean of the
controls. [Figure was drafted from original data from E. Boonen et al: Reduced cortisol metabolism during critical illness. N Engl J Med. 2013;368:
1477–1488 (62), with permission. © Massachusetts Medical Society.]
doi: 10.1210/jc.2013-4115 jcem.endojournals.org 1575

a highly economic way to keep cortisol levels high without much elevated, if at all, and that instead reduced cortisol
spending too much energy producing it. This concept is breakdown plays a major role during critical illness, fur-
further supported by low plasma cortisol binding globulin ther complicate the issue of diagnostic criteria for adrenal
levels in critical illness, causing increased levels of free failure in that setting. Moreover, it was recently shown
cortisol, the biologically active form. Furthermore, as that cortisol responses to ACTH stimulation in critically ill
such, cortisol is elevated locally in liver and kidney, where patients correlated positively with both cortisol produc-

Downloaded from https://academic.oup.com/jcem/article/99/5/1569/2537306 by National Science & Technology Library user on 10 March 2023
it is needed for an optimal fight-or-flight response, with- tion rate and cortisol plasma clearance, but patients who
out an undue exposure of immune cells and vulnerable revealed the lowest response to ACTH, to the extent of
target tissues such as skeletal muscle or brain to the del- absolute adrenal failure, were the ones with the most sup-
eterious side effects of hypercortisolism. The local effects pressed cortisol breakdown, whereas their cortisol pro-
of cortisol appear to be further regulated at the level of duction was similar to healthy subjects (65). These find-
glucocorticoid receptor (GR) expression. Previous work ings hint that a low cortisol response to an ACTH injection
indeed showed suppressed expression of GR in white reflects the degree of negative feedback inhibition exerted
blood cells of critically ill children, which could be a way by the high levels of circulating cortisol, a situation similar
to allow the innate immune response to effectively protect to patients treated with exogenous glucocorticoids for an
the host against infections in the presence of hypercorti- extended time, who also reveal a suppressed response to
solism (68). Clearly, this novel concept of tissue-specific ACTH injection. Whether this low response during critical
regulation of glucocorticoid activity during critical illness illness indicates that cortisol availability would be “insuf-
requires further investigation. ficient” to cope with the stress of illness remains unclear.
The new insight that during critical illness cortisol me- Alternatively, a random total cortisol of ⬍ 10 ␮g/dL
tabolism is suppressed, contributing to hypercortisolism, during critical illness has been suggested for the diagnosis
could theoretically explain the concomitantly low plasma of “relative adrenal insufficiency” (75). However, total
ACTH concentrations via negative feedback inhibition at plasma cortisol concentration is the net effect of adrenal
the level of the pituitary gland and/or the hypothalamus, production and secretion, distribution, binding, and elim-
but studies assessing this at the tissue level are currently ination of cortisol. Judging the adequacy of the adrenal
lacking. It remains unclear whether such a sustained sup- cortisol production in response to critical illness based on
pressed ACTH secretion could cause adrenal atrophy in a single measurement of total plasma cortisol is merely
the prolonged phase of critical illness. However, this could indicative. Furthermore, circulating total cortisol concen-
explain the reported 20-fold higher incidence of symp- trations do not reveal the glucocorticoid effect. During
tomatic adrenal insufficiency in critically ill patients being crucial illness suppressed circulating levels of the binding
treated in the ICU for more than 14 days (69). Other fac- proteins, cortisol binding globulin (CBG) and albumin, as
tors contributing to adrenal failure are also possible, such well as decreased CBG binding affinity via increased cleav-
as endothelial dysfunction (70, 71), although conforma- age from CBG at inflammatory loci or by increased tem-
tional human studies are lacking. perature were established (76 –79). Since only free cortisol
can pass the cell membrane to bind to GR and plasma, free
Diagnostic implications cortisol may be more appropriate to assess HPA-axis func-
Since the last decade, reference is made to “relative tion. However, more research is needed because plasma
adrenal insufficiency” in the context of critical illness (72– free cortisol assays are not readily available, and normal
74). It refers to the condition in which, despite a maximally ranges for plasma free cortisol during critical illness have
ACTH-activated adrenal cortex in response to critical ill- not been defined. Additionally, increasing evidence from
ness, the cortisol production is still insufficient to generate both animal and human experiments suggests altered GR
enough GR and mineralocorticoid receptor activation to regulation during critical illness (68, 80 – 84), precluding
maintain hemodynamic stability. From large association conclusions about “adequacy” of cortisol availability and
studies, such a condition is thought to be identifiable by an function during illness. Finally, assays to quantify plasma
insufficient rise (⬍9 ␮g/dL) in plasma cortisol in response cortisol concentrations are often inaccurate and vary sub-
to a 250-␮g ACTH bolus, irrespective of the baseline stantially (85), making it impossible to identify one cutoff
plasma cortisol concentration, which is usually much value for clinical practice.
higher than in healthy humans (72). In such a condition of Recently, measuring interstitial cortisol levels was in-
insufficiently increased cortisol production, a very high troduced to assess the amount of active tissue cortisol lev-
plasma ACTH concentration would be expected. How- els in critically ill patients (86, 87). Therefore, a microdi-
ever, the recent robust findings that ACTH plasma con- alysis catheter is inserted into the sc adipose tissue.
centrations are suppressed, that cortisol production is not However, critical illness presents frequently with edema,
1576 Boonen and Van den Berghe Novel Insights on Endocrine Changes in the ICU J Clin Endocrinol Metab, May 2014, 99(5):1569 –1582

and regional blood flow is variable. Furthermore, the sc Based on the results of stable isotope studies (62), a dose
adipose tissue is not the main target tissue for cortisol, nor of ⫾ 60 mg of hydrocortisone, equivalent to about a dou-
is it the main cortisol-metabolizing organ during critical bling of the normal daily cortisol production, may be in-
illness (62). teresting to investigate further when patients at risk can be
identified. A fast tapering down to the lowest effective
Therapeutic implications dose should limit the adverse effects of excessive amounts

Downloaded from https://academic.oup.com/jcem/article/99/5/1569/2537306 by National Science & Technology Library user on 10 March 2023
It is generally accepted that patients with an established of glucocorticoids during critical illness.
diagnosis of primary or central adrenal failure or patients
on chronic treatment with systemic glucocorticoids before
critical illness should receive additional coverage to cope The Hyperglycemic Response to Critical
with the acute stress (53, 88). Also, patients who are di- Illness: To Treat or Not to Treat?
agnosed with an acute Addisonian crisis in the ICU are
typically treated with high doses of glucocorticoids. This Blood glucose and critical illness: robust
therapeutic strategy is based on the assumption that cor- associative data
tisol production is increased severalfold in critical illness. In humans, the natural endocrine and immunological
The conventional treatment proposes the administration responses to stress ensure adequate availability of glucose
of a bolus of 100-mg hydrocortisone followed by 50 to by activating gluconeogenesis and by reducing the sensi-
100 mg every 6 hours on the first day, 50 mg every 6 hours tivity to insulin for those organs and tissues that predom-
on the second day, and 25 mg every 6 hours on the third inantly rely upon glucose as metabolic substrate, such as
day, tapering to a maintenance dose by the fourth to fifth the brain and blood cells. In young and lean patients not
day (53, 88). receiving macronutrients, this stress response will main-
The dose of hydrocortisone advised for treatment of tain normoglycemia. However, when patients are older,
“relative adrenal failure” is another controversial issue. are overweight, suffer from chronic comorbidity, or re-
The proposed dose of 300 mg of hydrocortisone per day, ceive drugs that affect insulin sensitivity or enteral/paren-
referred to as “low dose” in the literature, is in fact ap- teral nutrition, the circulating glucose concentrations usu-
proximately 10 times higher than the normal amount of ally rise quickly above the upper limit of normality (98 –
daily cortisol production in healthy humans (89 –92) and 102), which could be adaptive or maladaptive. In the
between 3- and 6-fold higher than the production that condition of prolonged critical illness, stress-induced hy-
now has been quantified in critically ill patients (Figure 3). perglycemia may be quite severe and may persist for a long
In view of the substantially reduced cortisol breakdown period of time. Hyperglycemia in critically ill patients has
during critical illness, the currently proposed doses for repeatedly been shown to be associated with a risk of mor-
adrenal failure during critical illness may be too high. This tality, an association that appears to have a J-shape with
may further explain why the multicenter randomized con- the lowest risk in the normoglycemic zone (Figure 4) (103).
trolled study that assessed the effect of hydrocortisone In critically ill patients with established diabetes mellitus,
treatment could not confirm the benefit that was originally the J-shaped curve is significantly blunted in the hyper-
observed in the pioneer trial (89, 92). glycemic zone, and the nadir is shifted to higher blood
Also, the duration of treatment is under debate. Treat- glucose levels (103, 104).
ing critically ill patients with glucocorticoids in too high of
a dose for too long of a period could inferentially aggra- Hyperglycemia and adverse outcome: cause or
vate the loss of lean tissue, increase the risk of myopathy, consequence?
and prolong the ICU dependence, which could increase the The first randomized controlled trial on blood glucose
susceptibility to potentially lethal complications (93, 94). management was the 2001 Leuven Surgical ICU study
Finally, because glucocorticoid sensitivity likely varies (105). In this study, a “strictly normal level for fasting
among individuals (95) and among cell types in critically blood glucose,” ie, 80 –110 mg/dL, was targeted in the
ill patients (68, 81, 83) and glucocorticoid treatment may intervention group, as compared to the “usual care” of
down-regulate GR-␣ via induction of miR-124, the dosing adult surgical ICU patients in the year 2000, which was to
issue is further complicated (96). Moreover, single nucle- tolerate hyperglycemia up to 215 mg/dL. The study was
otide polymorphisms in the GR gene, with an altered re- highly standardized, resulting in a strong internal validity.
sponse to glucocorticoids, have been identified (97). How- For example, frequent blood glucose measurements (in-
ever, it remains a challenge to identify specific clinical terval, 0.5– 4 h) on whole arterial blood by an accurate
biomarkers of GR activation to guide optimal glucocor- blood gas analyzer were done by well-trained nurse, and
ticoid therapy for individual patients and illnesses. insulin was continuously infused exclusively via a dedi-
doi: 10.1210/jc.2013-4115 jcem.endojournals.org 1577

DIFFERENCES IN DESIGN EXPECTED OUTCOMES BASED ON LEUVEN TRIALS

The Leuven comparison Cumulative risk in-hospital mortality

“Don’t touch”
The NICE-SUGAR
MORTALITY

comparison .3

Downloaded from https://academic.oup.com/jcem/article/99/5/1569/2537306 by National Science & Technology Library user on 10 March 2023
.2

.1

0 100 200 300 400 500 600


hypo normal for age “renal threshold”
BLOOD GLUCOSE days
Figure 4. Different designs of key intervention trials and expected outcome benefits. The left panel shows J-shaped association curve between
blood glucose and risk of death. The NICE-SUGAR trial was executed in the flatter part of the J-shaped curve. A very small benefit from aiming at
lowering blood glucose further down from an intermediate level to strict normoglycemia was hereby traded off against a similar risk of harm by
hypoglycemia, particularly when using inaccurate tools. The right panel shows the dose response in the two adult Leuven trials compared to the
NICE-SUGAR trial. Black circles represent blood glucose ⬎ 150mg/dl, dark grey circles represent blood glucose 110 –150mg/dl and light grey circles
represent blood glucose ⬍ 110mg/dl. The maximal benefit that could be expected from lowering blood glucose from an intermediate level to
normoglycemia is ⬍ 1%, provided blood glucose could be perfectly separated between the two study arms. To confidently conclude that such a
small benefit is not present, 70 000 patients should have been included. Hence, NICE-SUGAR, with 6100 patients, was in fact underpowered to
address this hypothesis. [Reproduced from G. Van den Berghe: Intensive insulin therapy in the ICU–reconciling the evidence. Nat Rev Endocrinol.
2012;8:374 –378 (127), with permission. © Macmillan Publishers Limited.]

cated lumen of a central venous line with an accurate sy- worldwide (114, 115). After several smaller studies, the
ringe pump. Maintaining strict normoglycemia lowered NICE-SUGAR (Normoglycemia in Intensive Care Evalu-
ICU and in-hospital mortality and reduced morbidity by ation and Survival Using Glucose Algorithm Regulation)
preventing organ failure, reflected in a shorter duration of multicenter trial was designed to be the definitive study to
mechanical ventilation, a decreased incidence of acute kid- answer this question. The study compared tight blood glu-
ney failure, severe infections, and critical illness polyneu- cose control to a normoglycemic target (80 –100 mg/dL) in
ropathy. In a second study performed in patients admitted the intervention group with an intermediate target of 140 –
to a medical ICU in Leuven, these morbidity benefits were 180 mg/dL in the control group (116). The study revealed
confirmed (106). A subsequent randomized controlled that blood glucose control to a normoglycemic target in-
study was performed in critically ill children, in which the creased mortality as compared with the intermediate level
intervention group was targeted to normal fasting glucose in the control group (116), subsequently explained by a
levels for the age groups (50 – 80 mg/dL for infants, and 13-fold increase in hypoglycemia (117). Because this study
70 –100 mg/dL for children) as compared with tolerating was designed for a high external validity, the first conclu-
hyperglycemia up to 215 mg/dL (107). Also in this young sion is that very tight blood glucose control is not readily
patient population, the intervention reduced ICU morbid- applicable in general daily clinical practice. However, the
ity and mortality and also had long-term beneficial effects usual care had already evolved significantly between the
on neurocognitive development up to 4 years after inclu- first Leuven study and the start of NICE-SUGAR; toler-
sion in the study (107, 108). In a subsequent study, tar- ating excessive hyperglycemia was now the new no-go
geting the much higher adult range for normal fasting zone, compared to the 215 mg/dL tolerance threshold 5
blood glucose levels in such young infants in the ICU did years earlier. Second, due to its pragmatic nature, there
not alter the level of blood glucose concentration or the was no emphasis on standardization in NICE-SUGAR. All
outcome (109), suggesting that the normal fasting level is sorts of glucose measurement methodologies were al-
key to preventing toxicity of hyperglycemia in each age lowed, and practitioners were not specifically trained to
group. The underlying mechanisms of hyperglycemia-in- perform the complex treatment. Now, it has become clear
duced toxicity were identified to involve cellular damage that tight blood glucose control requires accurate blood
occurring in those cells that do not require insulin for gas analyzers, like those used in the Leuven studies, to
glucose uptake, such as hepatocytes, renal tubular cells, target a narrow range of blood glucose (118). It is also
the endothelium, immune cells, and neurons (93, 110 – clear that extensive experience is crucial to avoid unde-
113). Soon after the first Leuven study was published, the tected episodes of hypoglycemia and to treat hypoglyce-
intervention was swiftly implemented in clinical practice mia when it occurs. Certainly profound, prolonged/unde-
1578 Boonen and Van den Berghe Novel Insights on Endocrine Changes in the ICU J Clin Endocrinol Metab, May 2014, 99(5):1569 –1582

tected hypoglycemia can have grave consequences and the acute endocrine responses are likely adaptive and thus
may even result in death. Hence, hypoglycemia should be should probably not be treated. Nevertheless, many pa-
avoided as much as possible. Nevertheless, recent data tients who survived the initial phase of critical illness still
show that in cardiac patients and in critically ill children, remain in the ICU for long periods and face a risk of death
iatrogenic hypoglycemia may not by itself affect outcome that increases steadily with every day that recovery does
(108, 119, 120). Spontaneous hypoglycemia is in contrast not set in. Hence, more work is required to find better

Downloaded from https://academic.oup.com/jcem/article/99/5/1569/2537306 by National Science & Technology Library user on 10 March 2023
a strong predictor of poor outcome. For example, patients treatments to further prevent protracted critical illness, to
with liver failure, acute kidney injury requiring renal re- enhance recovery from organ failure, and to optimize
placement therapy, diabetes mellitus, and septic shock rehabilitation.
have higher risk to develop spontaneous hypoglycemia.
Furthermore, adequate treatment of hypoglycemia is es-
sential to avoid rebound hyperglycemia, which causes Novel Insights in Endocrine Changes in
brain damage (121). Detailed protocols for prompt and Critical Illness
gentle correction of hypoglycemia are often not in place, • Part of the acute fall in T3 plasma concentrations during
which again contrasts with the Leuven studies (116). critical illness is related to the concomitant fasting, and
this part of the response seems adaptive.
Therapeutic implications: how to translate this • Cortisol production is only moderately increased dur-
into general clinical practice? ing critical illness and is only increased in patients suf-
What then could be a sensible approach for daily prac- fering from the systemic inflammatory response syn-
tice? Tight blood glucose control with current technolo- drome (SIRS). Cortisol production is unaltered in
gies and experience is not yet ready to be broadly imple- patients without SIRS, in the face of severalfold higher
mented in every ICU, as clearly demonstrated by NICE- plasma cortisol in all patients.
SUGAR. Post hoc analyses of the Leuven clinical trials • Cortisol plasma clearance is substantially reduced in all
revealed that the bulk of the beneficial effects of blood critically ill patients and contributes substantially to
glucose control lay in bringing overt hyperglycemia to hypercortisolism during critical illness, irrespective of
moderate levels (Figure 4) (122, 123). More can be gained the type and severity of illness and irrespective of the
by further tightening the glycemic control, but it requires inflammation status.
a substantial investment in training and technology to do • The largest benefit of blood glucose control may be
this safely. Hence, targeting blood glucose below 145 brought about by preventing overt hyperglycemia;
mg/dL seems a reasonable compromise (115). Critically ill hence, targeting blood glucose to intermediate ranges
diabetic patients may benefit from treatment to somewhat during critical illness seems a reasonable compromise.
higher glycemic targets, depending on their premorbid lev-
els (102–103). However, irrespective of the chosen target
level, several methodological aspects ought to be taken Acknowledgments
into account to assure patient safety whenever insulin
treatment is used. These include frequent blood glucose Address all correspondence and requests for reprints to: Greet
Van den Berghe, Clinical Division and Laboratory of Intensive
measurements, the use of on-site blood gas analyzers as the
Care Medicine, KU Leuven, Herestraat 49, B-3000 Leuven, Bel-
preferred measurement tool, the avoidance of capillary
gium. E-mail: greet.vandenberghe@med.kuleuven.be.
blood samples, and the continuous infusion of insulin with This work was supported by research grants from the Fund
accurate syringe pumps through a dedicated lumen of a for Scientific Research Flanders Belgium, by the Methusalem
central venous catheter. Finally, insulin dosing decisions Program funded by the Flemish Government, and by the Euro-
should not be based on a sliding scale system, but instead pean Research Council under the European Union’s Seventh
on a (computerized) algorithm that was clinically vali- Framework Program (FP7/2007–2013 ERC Advanced Grant
dated for critically ill patients (124). Agreement no. 307523).
Disclosure Summary: The authors have no conflict of interest
to declare.

Conclusions
Recent studies generated important novel insights in the References
endocrine and metabolic responses to critical illness. Al-
1. Casaer MP, Langouche L, Coudyzer W, et al. Impact of early par-
though many aspects remain unresolved, an important enteral nutrition on muscle and adipose tissue compartments during
recent insight with therapeutic implications is that most of critical illness. Crit Care Med. 2013;41:2298 –2309.
doi: 10.1210/jc.2013-4115 jcem.endojournals.org 1579

2. Puthucheary ZA, Rawal J, McPhail M, et al. Acute skeletal muscle enteral nutrition in critically ill adults. N Engl J Med. 2011;365:
wasting in critical illness. JAMA. 2013;310:1591–1600. 506 –517.
3. Villet S, Chiolero RL, Bollmann MD, et al. Negative impact of hy- 22. Langouche L, Vander Perre S, Marques M, et al. Impact of early
pocaloric feeding and energy balance on clinical outcome in ICU nutrient restriction during critical illness on the nonthyroidal illness
patients. Clin Nutr. 2005;24:502–509. syndrome and its relation with outcome: a randomized, controlled
4. Hermans G, Casaer MP, Clerckx B, et al. Effect of tolerating ma- clinical study. J Clin Endocrinol Metab. 2013;98:1006 –1013.
cronutrient deficit on the development of intensive-care unit ac- 23. Mebis L, Eerdekens A, Güiza F, et al. Contribution of nutritional
quired weakness: a subanalysis of the EPaNIC trial. Lancet Respir deficit to the pathogenesis of the nonthyroidal illness syndrome in

Downloaded from https://academic.oup.com/jcem/article/99/5/1569/2537306 by National Science & Technology Library user on 10 March 2023
Med. 2013;1:621– 629. critical illness: a rabbit model study. Endocrinology. 2012;153:973–
5. Vanhorebeek I, Gunst J, Derde S, et al. Insufficient activation of 984.
autophagy allows cellular damage to accumulate in critically ill pa- 24. Boelen A, Boorsma J, Kwakkel J, et al. Type 3 deiodinase is highly
tients. J Clin Endocrinol Metab. 2011;96:E633–E645. expressed in infiltrating neutrophilic granulocytes in response to
6. Chopra IJ, Huang TS, Beredo A, Solomon DH, Chua Teco GN, acute bacterial infection. Thyroid. 2008;18:1095–1103.
Mead JF. Evidence for an inhibitor of extrathyroidal conversion of 25. Van den Berghe G, de Zegher F, Veldhuis JD, et al. Thyrotrophin
thyroxine to 3,5,3⬘-triiodothyronine in sera of patients with non- and prolactin release in prolonged critical illness: dynamics of spon-
thyroidal illnesses. J Clin Endocrinol Metab. 1985;60:666 – 672. taneous secretion and effects of growth hormone-secretagogues.
7. Michalaki M, Vagenakis AG, Makri M, Kalfarentzos F, Kyriazo- Clin Endocrinol (Oxf). 1997;47:599 – 612.
poulou V. Dissociation of the early decline in serum T(3) concen- 26. Fliers E, Guldenaar SE, Wiersinga WM, Swaab DF. Decreased hy-
tration and serum IL-6 rise and TNF␣ in nonthyroidal illness syn- pothalamic thyrotropin-releasing hormone gene expression in pa-
drome induced by abdominal surgery. J Clin Endocrinol Metab. tients with nonthyroidal illness. J Clin Endocrinol Metab. 1997;82:
2001;86:4198 – 4205. 4032– 4036.
8. Bartalena L, Martino E, Brandi LS, et al. Lack of nocturnal serum 27. Bacci V, Schussler GC, Kaplan TB. The relationship between serum
thyrotropin surge after surgery. J Clin Endocrinol Metab. 1990;70: triiodothyronine and thyrotropin during systemic illness. J Clin En-
293–296. docrinol Metab. 1982;54:1229 –1235.
9. Romijn JA, Wiersinga WM. Decreased nocturnal surge of thyrotro- 28. Damas P, Reuter A, Gysen P, Demonty J, Lamy M, Franchimont P.
pin in nonthyroidal illness. J Clin Endocrinol Metab. 1990;70:35– Tumor necrosis factor and interleukin-1 serum levels during severe
42. sepsis in humans. Crit Care Med. 1989;17:975–978.
10. Redout EM, van der Toorn A, Zuidwijk MJ, et al. Antioxidant 29. Faglia G, Ferrari C, Beck-Peccoz P, Spada A, Travaglini P, Ambrosi
treatment attenuates pulmonary arterial hypertension-induced B. Reduced plasma thyrotropin response to thyrotropin releasing
heart failure. Am J Physiol Heart Circ Physiol. 2010;298:H1038 – hormone after dexamethasone administration in normal subjects.
H1047. Horm Metab Res. 1973;5:289 –292.
11. Wajner SM, Goemann IM, Bueno AL, Larsen PR, Maia AL. IL-6 30. Van den Berghe G, de Zegher F, Lauwers P. Dopamine and the sick
promotes nonthyroidal illness syndrome by blocking thyroxine ac- euthyroid syndrome in critical illness. Clin Endocrinol (Oxf). 1994;
tivation while promoting thyroid hormone inactivation in human 41:731–737.
cells. J Clin Invest. 2011;121:1834 –1845. 31. Van den Berghe G, de Zegher F, Lauwers P. Dopamine suppresses
12. Boelen A, Kwakkel J, Fliers E. Beyond low plasma T3: local thyroid pituitary function in infants and children. Crit Care Med. 1994;22:
hormone metabolism during inflammation and infection. Endocr 1747–1753.
Rev. 2011;32:670 – 693. 32. Boelen A, Kwakkel J, Thijssen-Timmer DC, Alkemade A, Fliers E,
13. van der Poll T, Van Zee KJ, Endert E, et al. Interleukin-1 receptor Wiersinga WM. Simultaneous changes in central and peripheral
blockade does not affect endotoxin-induced changes in plasma thy- components of the hypothalamus-pituitary-thyroid axis in lipopo-
roid hormone and thyrotropin concentrations in man. J Clin Endo- lysaccharide-induced acute illness in mice. J Endocrinol. 2004;182:
crinol Metab. 1995;80:1341–1346. 315–323.
14. Lim CF, Docter R, Visser TJ, et al. Inhibition of thyroxine transport 33. Mebis L, Debaveye Y, Ellger B, et al. Changes in the central com-
into cultured rat hepatocytes by serum of nonuremic critically ill ponent of the hypothalamus-pituitary-thyroid axis in a rabbit model
patients: effects of bilirubin and nonesterified fatty acids. J Clin of prolonged critical illness. Crit Care. 2009;13:R147.
Endocrinol Metab. 1993;76:1165–1172. 34. Alkemade A, Friesema EC, Kuiper GG, et al. Novel neuroanatomi-
15. Gardner DF, Kaplan MM, Stanley CA, Utiger RD. Effect of tri- cal pathways for thyroid hormone action in the human anterior
iodothyronine replacement on the metabolic and pituitary responses pituitary. Eur J Endocrinol. 2006;154:491–500.
to starvation. N Engl J Med. 1979;300:579 –584. 35. Langouche L, Princen L, Gunst J, Güiza F, Derde S, Van den Berghe
16. Moshang T Jr, Parks JS, Baker L, et al. Low serum triiodothyronine G. Anterior pituitary morphology and hormone production during
in patients with anorexia nervosa. J Clin Endocrinol Metab. 1975; sustained critical illness in a rabbit model. Horm Metab Res. 2013;
40:470 – 473. 45:277–282.
17. Peeters RP, Wouters PJ, van Toor H, Kaptein E, Visser TJ, Van den 36. Mebis L, Paletta D, Debaveye Y, et al. Expression of thyroid hor-
Berghe G. Serum 3,3⬘,5⬘-triiodothyronine (rT3) and 3,5,3⬘-triiodo- mone transporters during critical illness. Eur J Endocrinol. 2009;
thyronine/rT3 are prognostic markers in critically ill patients and are 161:243–250.
associated with postmortem tissue deiodinase activities. J Clin En- 37. Mebis L, Langouche L, Visser TJ, Van den Berghe G. The type II
docrinol Metab. 2005;90:4559 – 4565. iodothyronine deiodinase is up-regulated in skeletal muscle during
18. Rothwell PM, Lawler PG. Prediction of outcome in intensive care prolonged critical illness. J Clin Endocrinol Metab. 2007;92:3330 –
patients using endocrine parameters. Crit Care Med. 1995;23:78 – 3333.
83. 38. Ma SF, Xie L, Pino-Yanes M, et al. Type 2 deiodinase and host
19. Klemperer JD, Klein I, Gomez M, et al. Thyroid hormone treatment responses of sepsis and acute lung injury. Am J Respir Cell Mol Biol.
after coronary-artery bypass surgery. N Engl J Med. 1995;333: 2011;45:1203–1211.
1522–1527. 39. Thijssen-Timmer DC, Peeters RP, Wouters P, et al. Thyroid hor-
20. Mullis-Jansson SL, Argenziano M, Corwin S, et al. A randomized mone receptor isoform expression in livers of critically ill patients.
double-blind study of the effect of triiodothyronine on cardiac func- Thyroid. 2007;17:105–112.
tion and morbidity after coronary bypass surgery. J Thorac Cardio- 40. Van den Berghe G, Wouters P, Weekers F, et al. Reactivation of
vasc Surg. 1999;117:1128 –1134. pituitary hormone release and metabolic improvement by infusion
21. Casaer MP, Mesotten D, Hermans G, et al. Early versus late par- of growth hormone-releasing peptide and thyrotropin-releasing
1580 Boonen and Van den Berghe Novel Insights on Endocrine Changes in the ICU J Clin Endocrinol Metab, May 2014, 99(5):1569 –1582

hormone in patients with protracted critical illness. J Clin Endocri- livered before The Royal College of Physicians of London on St.
nol Metab. 1999;84:1311–1323. Luke’s Day, 1923. Br Med J. 1923;2:685– 690.
41. Van den Berghe G, de Zegher F, Baxter RC, et al. Neuroendocri- 61. Vermes I, Beishuizen A, Hampsink RM, Haanen C. Dissociation of
nology of prolonged critical illness: effects of exogenous thyrotro- plasma adrenocorticotropin and cortisol levels in critically ill pa-
pin-releasing hormone and its combination with growth hormone tients: possible role of endothelin and atrial natriuretic hormone.
secretagogues. J Clin Endocrinol Metab. 1998;83:309 –319. J Clin Endocrinol Metab. 1995;80:1238 –1242.
42. Weekers F, Michalaki M, Coopmans W, et al. Endocrine and met- 62. Boonen E, Vervenne H, Meersseman P, et al. Reduced cortisol me-
abolic effects of growth hormone (GH) compared with GH-releas- tabolism during critical illness. N Engl J Med. 2013;368:1477–

Downloaded from https://academic.oup.com/jcem/article/99/5/1569/2537306 by National Science & Technology Library user on 10 March 2023
ing peptide, thyrotropin-releasing hormone, and insulin infusion in 1488.
a rabbit model of prolonged critical illness. Endocrinology. 2004; 63. Bornstein SR, Engeland WC, Ehrhart-Bornstein M, Herman JP. Dis-
145:205–213. sociation of ACTH and glucocorticoids. Trends Endocrinol Metab.
43. Van den Berghe G, Baxter RC, Weekers F, Wouters P, Bowers CY, 2008;19:175–180.
Iranmanesh A, Veldhuis JD, Bouillon R. The combined administra- 64. Vanwijngaerden YM, Wauters J, Langouche L, et al. Critical illness
tion of GH-releasing peptide-2 (GHRP-2), TRH and GnRH to men evokes elevated circulating bile acids related to altered hepatic trans-
with prolonged critical illness evokes superior endocrine and met- porter and nuclear receptor expression. Hepatology. 2011;54:
abolic effects compared to treatment with GHRP-2 alone. Clin En- 1741–1752.
docrinol (Oxf). 2002;56:655– 669. 65. Ackermann D, Vogt B, Escher G, et al. Inhibition of 11␤-hydrox-
44. Mebis L, van den Berghe G. The hypothalamus-pituitary-thyroid ysteroid dehydrogenase by bile acids in rats with cirrhosis. Hepa-
axis in critical illness. Neth J Med. 2009;67:332–340. tology. 1999;30:623– 629.
45. Debaveye Y, Ellger B, Mebis L, et al. Tissue deiodinase activity 66. McNeilly AD, Macfarlane DP, O’Flaherty E, et al. Bile acids mod-
during prolonged critical illness: effects of exogenous thyrotropin- ulate glucocorticoid metabolism and the hypothalamic-pituitary-
releasing hormone and its combination with growth hormone-re- adrenal axis in obstructive jaundice. J Hepatol. 2010;52:705–711.
leasing peptide-2. Endocrinology. 2005;146:5604 –5611. 67. Stauffer AT, Rochat MK, Dick B, Frey FJ, Odermatt A. Chenode-
46. Debaveye Y, Ellger B, Mebis L, Darras VM, Van den Berghe G. oxycholic acid and deoxycholic acid inhibit 11 ␤-hydroxysteroid
Regulation of tissue iodothyronine deiodinase activity in a model of dehydrogenase type 2 and cause cortisol-induced transcriptional
prolonged critical illness. Thyroid. 2008;18:551–560. activation of the mineralocorticoid receptor. J Biol Chem. 2002;
47. Debaveye Y, Ellger B, Mebis L, Visser TJ, Darras VM, Van den 277:26286 –26292.
Berghe G. Effects of substitution and high-dose thyroid hormone 68. van den Akker EL, Koper JW, Joosten K, et al. Glucocorticoid re-
therapy on deiodination, sulfoconjugation, and tissue thyroid hor- ceptor mRNA levels are selectively decreased in neutrophils of chil-
mone levels in prolonged critically ill rabbits. Endocrinology. 2008; dren with sepsis. Intensive Care Med. 2009;35:1247–1254.
149:4218 – 4228. 69. Barquist E, Kirton O. Adrenal insufficiency in the surgical intensive
48. Peeters RP, van der Geyten S, Wouters PJ, et al. Tissue thyroid care unit patient. J Trauma. 1997;42:27–31.
hormone levels in critical illness. J Clin Endocrinol Metab. 2005; 70. Kanczkowski W, Chatzigeorgiou A, Grossklaus S, Sprott D, Born-
90:6498 – 6507. stein SR, Chavakis T. Role of the endothelial-derived endogenous
49. Acker CG, Singh AR, Flick RP, Bernardini J, Greenberg A, Johnson anti-inflammatory factor Del-1 in inflammation-mediated adrenal
JP. A trial of thyroxine in acute renal failure. Kidney Int. 2000;57: gland dysfunction. Endocrinology. 2013;154:1181–1189.
293–298. 71. Prigent H, Maxime V, Annane D. Science review: mechanisms of
50. Becker RA, Vaughan GM, Ziegler MG, et al. Hypermetabolic low impaired adrenal function in sepsis and molecular actions of gluco-
triiodothyronine syndrome of burn injury. Crit Care Med. 1982; corticoids. Crit Care. 2004;8:243–252.
10:870 – 875. 72. Annane D, Sébille V, Troché G, Raphaël JC, Gajdos P, Bellissant E.
51. Brent GA, Hershman JM. Thyroxine therapy in patients with severe A 3-level prognostic classification in septic shock based on cortisol
nonthyroidal illnesses and low serum thyroxine concentration. levels and cortisol response to corticotropin. JAMA. 2000;283:
J Clin Endocrinol Metab. 1986;63:1– 8. 1038 –1045.
52. Sirlak M, Yazicioglu L, Inan MB, et al. Oral thyroid hormone pre- 73. Beishuizen A, Vermes I, Hylkema BS, Haanen C. Relative eosino-
treatment in left ventricular dysfunction. Eur J Cardiothorac Surg. philia and functional adrenal insufficiency in critically ill patients.
2004;26:720 –725. Lancet. 1999;353:1675–1676.
53. Debaveye Y, Vandenbrande J, Van den Berghe G. Endocrine emer- 74. Richards ML, Caplan RH, Wickus GG, Lambert PJ, Kisken WA.
gencies. In: Tubaro M, Danchin N, Filippatos G, Goldstein P, The rapid low-dose (1 microgram) cosyntropin test in the immediate
Vranckx P, Zahger D, eds. The ESC Textbook of Intensive and postoperative period: results in elderly subjects after major abdom-
Acute Cardiac Care. New York: Oxford University Press; 2011: inal surgery. Surgery. 1999;125:431– 440.
709 –717. 75. Marik PE, Pastores SM, Annane D, et al. Recommendations for the
54. Nicoloff JT. Thyroid storm and myxedema coma. Med Clin North diagnosis and management of corticosteroid insufficiency in criti-
Am. 1985;69:1005–1017. cally ill adult patients: consensus statements from an international
55. Ringel MD. Management of hypothyroidism and hyperthyroidism task force by the American College of Critical Care Medicine. Crit
in the intensive care unit. Crit Care Clin. 2001;17:59 –74. Care Med. 2008;36:1937–1949.
56. Escobar-Morreale HF, Obregón MJ, Escobar del Rey F, Morreale de 76. Chan WL, Carrell RW, Zhou A, Read RJ. How changes in affinity
Escobar G. Replacement therapy for hypothyroidism with thyrox- of corticosteroid-binding globulin modulate free cortisol concen-
ine alone does not ensure euthyroidism in all tissues, as studied in tration. J Clin Endocrinol Metab. 2013;98:3315–3322.
thyroidectomized rats. J Clin Invest. 1995;96:2828 –2838. 77. Hamrahian AH, Oseni TS, Arafah BM. Measurements of serum free
57. Van den Berghe G. Endocrine aspects of critical care medicine. In: cortisol in critically ill patients. N Engl J Med. 2004;350:1629 –
Jameson JL, De Groot LJ, eds. Endocrinology: Adult and Pediatric. 1638.
St Louis, MO: WB Saunders; 2010:2084 –2085. 78. Holland PC, Hancock SW, Hodge D, Thompson D, Shires S, Evans
58. Finlay WE, McKee JI. Serum cortisol levels in severely stressed pa- S. Degradation of albumin in meningococcal sepsis. Lancet. 2001;
tients. Lancet. 1982;1:1414 –1415. 357:2102–2104.
59. Munck A, Guyre PM, Holbrook NJ. Physiological functions of glu- 79. Pugeat M, Bonneton A, Perrot D, et al. Decreased immunoreactivity
cocorticoids in stress and their relation to pharmacological actions. and binding activity of corticosteroid-binding globulin in serum in
Endocr Rev. 1984;5:25– 44. septic shock. Clin Chem. 1989;35:1675–1679.
60. Starling EH. The wisdom of the body: the Harveian Oration, de- 80. Bergquist M, Nurkkala M, Rylander C, Kristiansson E, Hedensti-
doi: 10.1210/jc.2013-4115 jcem.endojournals.org 1581

erna G, Lindholm C. Expression of the glucocorticoid receptor is 100. Pasquel FJ, Smiley D, Spiegelman R, Lin E, Peng L, Umpierrez GE.
decreased in experimental Staphylococcus aureus sepsis. J Infect. Hyperglycemia is associated with increased hospital complications
2013;67:574 –583. and mortality during parenteral nutrition. Hosp Pract. 2011;39:
81. Guerrero J, Gatica HA, Rodriguez M, Estay R, Goecke IA. Septic 81– 88.
serum induces glucocorticoid resistance and modifies the expression 101. Pieracci F, Hydo L, Eachempati S, Pomp A, Shou J, Barie PS.
of glucocorticoid isoforms receptors: a prospective cohort study and Higher body mass index predicts need for insulin but not hyper-
in vitro experimental assay. Crit Care. 2013;17:R107. glycemia, nosocomial infection, or death in critically ill surgical
82. Indyk JA, Candido-Vitto C, Wolf IM, et al. Reduced glucocorticoid patients. Surg Infect (Larchmt). 2008;9:121–130.

Downloaded from https://academic.oup.com/jcem/article/99/5/1569/2537306 by National Science & Technology Library user on 10 March 2023
receptor protein expression in children with critical illness. Horm 102. Weiss M, Kron M, Hay B, Taenzer M, Radermacher P, Georgieff
Res Paediatr. 2013;79:169 –178. M. Which variables are associated with blood glucose levels out-
83. Peeters RP, Hagendorf A, Vanhorebeek I, et al. Tissue mRNA ex- side the target range in surgical critically ill patients? A retrospec-
pression of the glucocorticoid receptor and its splice variants in fatal tive observational study. Patient Saf Surg. 2011;5:5.
critical illness. Clin Endocrinol (Oxf). 2009;71:145–153. 103. Krinsley JS. Glycemic control in the critically ill - 3 domains and
84. Siebig S, Meinel A, Rogler G, et al. Decreased cytosolic glucocorti- diabetic status means one size does not fit all! Crit Care. 2013;17:
131.
coid receptor levels in critically ill patients. Anaesth Intensive Care.
104. Falciglia M, Freyberg RW, Almenoff PL, D’Alessio DA, Render
2010;38:133–140.
ML. Hyperglycemia-related mortality in critically ill patients varies
85. Cohen J, Ward G, Prins J, Jones M, Venkatesh B. Variability of
with admission diagnosis. Crit Care Med. 2009;37:3001–3009.
cortisol assays can confound the diagnosis of adrenal insufficiency
105. van den Berghe G, Wouters P, Weekers F, et al. Intensive insulin
in the critically ill population. Intensive Care Med. 2006;32:1901–
therapy in critically ill patients. N Engl J Med. 2001;345:1359 –
1905.
1367.
86. Vassiliadi DA, Ilias I, Tzanela M, et al. Interstitial cortisol obtained 106. Van den Berghe G, Wilmer A, Hermans G, et al. Intensive insulin
by microdialysis in mechanically ventilated septic patients: correla- therapy in the medical ICU. N Engl J Med. 2006;354:449 – 461.
tions with total and free serum cortisol. J Crit Care. 2013;28:158 – 107. Vlasselaers D, Milants I, Desmet L, et al. Intensive insulin therapy
165. for patients in paediatric intensive care: a prospective, randomised
87. Venkatesh B, Morgan TJ, Cohen J. Interstitium: the next diagnostic controlled study. Lancet. 2009;373:547–556.
and therapeutic platform in critical illness. Crit Care Med. 2010; 108. Mesotten D, Gielen M, Sterken C, et al. Neurocognitive develop-
38:S630 –S636. ment of children 4 years after critical illness and treatment with
88. Chung TT, Grossman A, Clark AJ. Adrenal insufficiency. In: Jame- tight glucose control: a randomized controlled trial. JAMA. 2012;
son JL, De Groot LJ, eds. Endocrinology: Adult and Pediatric. St 308:1641–1650.
Louis, MO: WB Saunders; 2010;1853–1863. 109. Agus MS, Steil GM, Wypij D, et al. Tight glycemic control versus
89. Annane D, Sébille V, Charpentier C, et al. Effect of treatment with standard care after pediatric cardiac surgery. N Engl J Med. 2012;
low doses of hydrocortisone and fludrocortisone on mortality in 367:1208 –1219.
patients with septic shock. JAMA. 2002;288:862– 871. 110. Gunst J, Derese I, Aertgeerts A, et al. Insufficient autophagy con-
90. Bollaert PE, Charpentier C, Levy B, Debouverie M, Audibert G, tributes to mitochondrial dysfunction, organ failure, and adverse
Larcan A. Reversal of late septic shock with supraphysiologic doses outcome in an animal model of critical illness. Crit Care Med.
of hydrocortisone. Crit Care Med. 1998;26:645– 650. 2013;41:182–194.
91. Briegel J, Forst H, Haller M, et al. Stress doses of hydrocortisone 111. Langouche L, Vanhorebeek I, Vlasselaers D, et al. Intensive insulin
reverse hyperdynamic septic shock: a prospective, randomized, dou- therapy protects the endothelium of critically ill patients. J Clin
ble-blind, single-center study. Crit Care Med. 1999;27:723–732. Invest. 2005;115:2277–2286.
92. Sprung CL, Annane D, Keh D, et al. Hydrocortisone therapy for 112. Vanhorebeek I, De Vos R, Mesotten D, Wouters PJ, De Wolf-
patients with septic shock. N Engl J Med. 2008;358:111–124. Peeters C, Van den Berghe G. Protection of hepatocyte mitochon-
93. Hermans G, Wilmer A, Meersseman W, et al. Impact of intensive drial ultrastructure and function by strict blood glucose control
insulin therapy on neuromuscular complications and ventilator de- with insulin in critically ill patients. Lancet. 2005;365:53–59.
pendency in the medical intensive care unit. Am J Respir Crit Care 113. Vanhorebeek I, Gunst J, Ellger B, et al. Hyperglycemic kidney
Med. 2007;175:480 – 489. damage in an animal model of prolonged critical illness. Kidney
94. Hermans G, De Jonghe B, Bruyninckx F, Van den Berghe G. Clinical Int. 2009;76:512–520.
114. Garber AJ, Moghissi ES, Bransome ED Jr, et al. American College
review: critical illness polyneuropathy and myopathy. Crit Care.
of Endocrinology position statement on inpatient diabetes and
2008;12:238.
metabolic control. Endocr Pract. 2004;10:77– 82.
95. Hauer D, Weis F, Papassotiropoulos A, et al. Relationship of a com-
115. Umpierrez GE, Hellman R, Korytkowski MT, et al. Management
mon polymorphism of the glucocorticoid receptor gene to traumatic
of hyperglycemia in hospitalized patients in non-critical care set-
memories and posttraumatic stress disorder in patients after inten-
ting: an Endocrine Society Clinical Practice Guideline. J Clin En-
sive care therapy. Crit Care Med. 2011;39:643– 650.
docrinol Metab. 2012;97:16 –38.
96. Ledderose C, Möhnle P, Limbeck E, et al. Corticosteroid resistance 116. Finfer S, Chittock DR, Su SY, et al. Intensive versus conventional
in sepsis is influenced by microRNA-124 –induced downregulation glucose control in critically ill patients. N Engl J Med. 2009;360:
of glucocorticoid receptor-␣. Crit Care Med. 2012;40:2745–2753. 1283–1297.
97. Baker AC, Chew VW, Green TL, et al. Single nucleotide polymor- 117. Finfer S, Liu B, Chittock DR, et al. Hypoglycemia and risk of death
phisms and type of steroid impact the functional response of the in critically ill patients. N Engl J Med. 2012;367:1108 –1118.
human glucocorticoid receptor. J Surg Res. 2013;180:27–34. 118. Finfer S, Billot L. Managing blood glucose in critically ill patients
98. Ahrens CL, Barletta JF, Kanji S, et al. Effect of low-calorie parenteral with or without diabetes. Crit Care. 2013;17:134.
nutrition on the incidence and severity of hyperglycemia in surgical 119. Kosiborod M, Inzucchi SE, Goyal A, et al. Relationship between
patients: a randomized, controlled trial. Crit Care Med. 2005;33: spontaneous and iatrogenic hypoglycemia and mortality in pa-
2507–2512. tients hospitalized with acute myocardial infarction. JAMA. 2009;
99. Olveira G, Tapia MJ, Ocón J, et al. Parenteral nutrition-associated 301:1556 –1564.
hyperglycemia in non-critically ill inpatients increases the risk of 120. Tin W, Brunskill G, Kelly T, Fritz S. 15-year follow-up of recurrent
in-hospital mortality (multicenter study). Diabetes Care. 2013;36: “hypoglycemia” in preterm infants. Pediatrics. 2012;130:e1497–
1061–1066. e1503.
1582 Boonen and Van den Berghe Novel Insights on Endocrine Changes in the ICU J Clin Endocrinol Metab, May 2014, 99(5):1569 –1582

121. Suh SW, Gum ET, Hamby AM, Chan PH, Swanson RA. Hypo- 124. Kavanagh BP, McCowen KC. Clinical practice. Glycemic control
glycemic neuronal death is triggered by glucose reperfusion and in the ICU. N Engl J Med. 2010;363:2540 –2546.
activation of neuronal NADPH oxidase. J Clin Invest. 2007;117: 125. Vanhorebeek I, Langouche L, Van den Berghe G. Endocrine as-
910 –918. pects of acute and prolonged critical illness. Nat Clin Pract Endo-
122. Van den Berghe G, Wouters PJ, Bouillon R, et al. Outcome benefit crinol Metab. 2006;2:20 –31.
of intensive insulin therapy in the critically ill: insulin dose versus 126. Mebis L, Van den Berghe G. Thyroid axis function and dysfunction
glycemic control. Crit Care Med. 2003;31:359 –366. in critical illness. Best Pract Res Clin Endocrinol Metab. 2011;25:
123. Van den Berghe G, Wilmer A, Milants I, et al. Intensive insulin 745–757.

Downloaded from https://academic.oup.com/jcem/article/99/5/1569/2537306 by National Science & Technology Library user on 10 March 2023
therapy in mixed medical/surgical intensive care units: benefit ver- 127. Van den Berghe G. Intensive insulin therapy in the ICU–reconciling
sus harm. Diabetes. 2006;55:3151–3159. the evidence. Nat Rev Endocrinol. 2012;8:374 –378.

Members have FREE online access to current endocrine


Clinical Practice Guidelines.
www.endocrine.org/guidelines

You might also like