Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

CONCISE CLINICAL REVIEW

On the Neuroendocrinopathy of Critical Illness


Perspectives for Feeding and Novel Treatments
Greet Van den Berghe
Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven University and
Hospitals, Leuven, Belgium

Abstract accentuated by the concomitant fasting. Accepting the lack of


macronutrients as well as the neuroendocrine responses to such
Typical for critical illnesses are substantial alterations within the fasting in the acute phase of critical illness has shown to beneficially
hypothalamic–anterior pituitary–peripheral hormonal axes that are affect outcome. In contrast, the neuroendocrine alterations that
proportionate to the risk of poor outcome. These neuroendocrine occur in the chronic phase of illness while patients are fully
responses to critical illness follow a biphasic pattern. The acute phase fed contribute to bone and skeletal muscle wasting and impose risk
(first hours to days) is characterized by an increased release of of adrenocortical atrophy. The combined administration of those
anterior pituitary hormones, whereas altered target-organ sensitivity hypothalamic releasing factors, which have been identified as
and hormone metabolism result in low levels of the anabolic suppressed or deficient during prolonged critical illness, may be a
peripheral effector hormones and contribute to the substantially promising strategy to enhance recovery. The potential impact of
elevated levels of the catabolic hormone cortisol. The prolonged treatment with such hypothalamic releasing factors on recovery from
phase of critical illness is hallmarked by a uniform suppression of the critical illness as well as on long-term rehabilitation should be
neuroendocrine axes, predominantly of central/hypothalamic origin, investigated in future randomized controlled clinical trials.
which contributes to the low (or insufficiently high in the case of
cortisol) circulating levels of the target-organ hormones. Several Keywords: growth hormone; cortisol; adrenocorticotropic
of the acute-phase adaptations to critical illness are due to or hormone; thyrotrophin-releasing hormone; ghrelin

Thanks to the development of modern for which patients required vital mass and induces weakness, whereas
intensive care medicine over the past organ support, ICU-acquired weakness acromegaly results in an inappropriately
7 decades, patients nowadays survive develops in the chronic phase. Ongoing large bone and muscle mass with loss
previously lethal trauma or illnesses. The hypercatabolism despite the provision of of body fat. Normal availability of thyroid
majority of patients treated in ICUs artificial nutrition is believed to result hormone is also crucial for the balance
worldwide recover within a week. About in ongoing loss of lean body mass, which is between anabolism and catabolism.
25% of ICU patients, however, enter the assumed to contribute to the weakness Hypothyroidism causes weakness and
chronic phase of critical illness, during and frailty and to prolong the dependency cognitive and organ dysfunction,
which they require vital organ support on intensive medical care. and hyperthyroidism results in increased
often for weeks, sometimes months. On The hypothalamic–anterior pituitary energy expenditure, hypercatabolism, and
average, between 15% and 20% of those axes play a central role in the endocrine wasting. Also, the pituitary–adrenocortical
prolonged critically ill patients do not regulation of metabolic homeostasis. axis is a major switch in controlling the
survive the ICU phase, and the risk of not Outside the context of critical illness, it is metabolic balance, as evidenced by the
leaving the hospital alive is even higher. well known that growth hormone (GH) phenotype of patients with Addison disease
Irrespective of the type of the initial disease deficiency causes loss of muscle and bone and those with Cushing syndrome. Lack

( Received in original form July 26, 2016; accepted in final form September 8, 2016 )
Supported by the Research Foundation Flanders grant G041712N, the Methusalem Program of the Flemish Government grant METH/14/06, and the
European Research Council under the European Union’s Seventh Framework Program grant FP7/2013-2018 ERC Advanced Grant Agreement 321670.
Correspondence and requests for reprints should be addressed to Greet Van den Berghe, M.D., Ph.D. Intensive Care Medicine, UZ Leuven, Herestraat 49,
B-3000 Leuven, Belgium. E-mail: greet.vandenberghe@kuleuven.be
CME will be available for this article at www.atsjournals.org
Am J Respir Crit Care Med Vol 194, Iss 11, pp 1337–1348, Dec 1, 2016
Copyright © 2016 by the American Thoracic Society
Originally Published in Press as DOI: 10.1164/rccm.201607-1516CI on September 9, 2016
Internet address: www.atsjournals.org

Concise Clinical Review 1337


CONCISE CLINICAL REVIEW

of cortisol causes weakness and impaired changes substantially (Figure 1A). More GH GHRP-2 reveals that the somatotropes
cognition, and hypercortisolism causes is released into the circulation, with high are more than able to synthesize GH.
muscle and bone wasting, with GH peaks and interpulse concentrations Hence, it is more likely that the cause of
inappropriate fat storage. and an increased GH pulse frequency the relative hyposomatotropism in the
Critical illness is characterized by (1, 10). Concomitantly, in part triggered by chronic phase of illness lies within the
profound alterations within these normally proinflammatory cytokines, peripheral hypothalamus. With GH release in
tightly feedback-controlled neuroendocrine GH resistance develops, with loss of response to GHRH that is much lower than
systems. Research performed over the last functional GH receptors as revealed by low that to GHRP-2 in prolonged critical
2 decades has revealed that these changes circulating levels of GH-binding protein, illness (9), a hypothalamic inactivity of
follow a biphasic pattern, with the and as a consequence enhanced GH ghrelin is likely to play a key role. This
neuroendocrine responses to acute and secretion in the face of low levels of IGF-I, “hyposomatotropism,” with loss of
prolonged critical illness being distinct GH-dependent IGFBP-3, and acid-labile pulsatile GH secretion, contributes to
(1, 2). Recent research has shed some light subunit (8, 10–12). Enhanced clearance muscle and bone wasting typically present
on which of these neuroendocrine of IGF-I, due to changes in its binding in prolonged critical illness (16).
alterations are likely adaptive and beneficial proteins, further lowers its circulating
at a certain time point in the course of levels. Reduced expression of the GH
critical illness and which may represent a receptor (12, 13), whereby low-circulating The Pituitary–Thyroid Axis
therapeutic opportunity that should IGF-I and thus loss of negative feedback
be further explored in clinical trials. inhibition is believed to drive the abundant Thyrotrophin-releasing hormone (TRH)
In this concise review, the new insights release of GH in the acute phase of illness. secreted by the hypothalamus stimulates
are integrated with the previous This constellation is also evoked by the pituitary thyrotropes to produce
knowledge on the alterations within fasting, where it results in amplified direct thyrotrophin (TSH) in a pulsatile fashion,
the neuroendocrine system observed lipolytic and insulin-antagonizing effects which in turn regulates the thyroidal
during the acute and the chronic phases of of GH, leading to a release of endogenous synthesis and secretion of thyroid
critical illness, with a special focus on the fatty acids and glucose into the circulation, hormones. Although the thyroid gland
GH axis, the pituitary–thyroid axis and whereas indirect, IGF-I–mediated effects predominantly produces thyroxin (T4), the
the pituitary–adrenocortical axis. of GH are switched off. As a result, energy- biological activity of thyroid hormones is
Opportunities for research are highlighted. consuming anabolism, mediated by IGF-I, largely exerted by triiodothyronine (T3)
is postponed. From an evolutionary (17). Different types of deiodinating
viewpoint, such a GH-axis response to enzymes control the peripheral activation
The GH Axis acute illnesses appears appropriate for of T4 to T3 or the alternative conversion
survival. of T4 into the biologically inactive reverse
GH is released into the circulation by the T3 (rT3) (18). Thyroid hormones enter
somatotrope cells of the anterior The GH Axis in the Prolonged Phase cells through specific monocarboxylate
pituitary gland in a pulsatile fashion, with of Critical Illness transporters, and T3 acts on thyroid
peak GH levels alternating with virtually Unlike in the acute phase of illness, the hormone nuclear receptors that
undetectable troughs. The regulation of pulsatile release of GH is suppressed in heterodimerize with the Retinoid-X
pulsatile GH secretion is complex, with prolonged (fed) critically ill patients Receptor and control gene expression in
hypothalamic GH-releasing hormone (Figure 1A) (14–16). Although it was various peripheral tissues (19). Circulating
(GHRH) stimulating, and somatostatin previously assumed that GH resistance and local thyroid hormones exert
inhibiting, the synthesis and secretion of GH persists throughout critical illness, GH feedback control on both hypothalamic
(3). In addition, ghrelin, a highly conserved responsiveness was later shown to recover TRH and pituitary TSH secretion.
hormone expressed in the hypothalamus in the chronic phase when patients are
and in the gut, is a third key driver receiving nutrition. In this chronic phase, The Pituitary–Thyroid Axis in the
of pulsatile GH release (4, 5). The the pulsatile fraction of GH release Acute Phase of Critical Illness
hypothalamic and pituitary G-protein– correlates positively with circulating IGF-I, The initial response of the thyroid axis to
coupled ghrelin receptor can also IGFBP-3, and acid-labile subunit levels illness in humans consists of a rapid decline
be activated by several synthetic (14–16), which suggests that the loss of in the circulating levels of T3 and an
GH-releasing peptides (GHRPs) (6, 7). pulsatile GH release contributes to these increase in rT3 levels (Figure 2). This is
GH exerts direct and indirect effects, the low levels of IGF-I and binding proteins in explained by an immediately decreased
latter exerted by insulin-like growth factor-I prolonged critical illness. Furthermore, type 1 deiodinase (D1) activity and an
(IGF-I), of which the bioactivity is the infusion of a ghrelin-like GH-releasing increased type 3 deiodinase (D3) activity
regulated via a series of IGF-binding peptide (GHRP-2) was found to reactivate (18,19), whereby T4 is converted to rT3
proteins (IGFBPs) (8, 9). pulsatile GH secretion back to the level rather than to T3 (20, 21). TSH and
of activation seen in the acute phase of T4 levels are transiently elevated but
The GH Axis in the Acute Phase of illness and to proportionally increase subsequently return to normal (Figure 2)
Critical Illness IGF-I and GH-dependent IGFBP levels (22). Low T3 levels without an increase
Within hours up to days after onset of (Figure 1B) (14, 15). The higher-than- in TSH is referred to as “the low T3
acute illnesses, the GH secretion pattern normal release of GH in response to syndrome.” Such a low T3 syndrome is also

1338 American Journal of Respiratory and Critical Care Medicine Volume 194 Number 11 | December 1 2016
CONCISE CLINICAL REVIEW

A Health Acute Critical Illness Prolonged Critical Illness


14 14 14
GH (μg/l)

7 7 7

0 0 0
21:00 06:00 21:00 06:00 21:00 06:00
Time Time Time

B
50 50 50 50
Placebo GHRH GHRP-2 GHRH + GHRP-2
40 40 40 40
GH (μg/l)

30 30 30 30
20 20 20 20
10 10 10 10
0 0 0 0
21 06h 21 06h 21 06h 21 06h
Time Time Time Time

150
IGF-1 R2= 0.67

100 ALS R2= 0.76


concentration (%)
Change in

IGFBP-3 R2= 0.57


50

–50
0 100 200 300 400 500 600
Pulsatile GH
(μg/lv over 9h)
Figure 1. Illustration of the growth hormone (GH) axis responses to acute and prolonged critical illness as compared with the healthy control condition.
(A) Nocturnal GH plasma concentration time series during health and during acute and prolonged critical illness. Pulsatile GH secretion is activated
during acute critical illness and suppressed during prolonged critical illness. (B) The suppressed pulsatile GH secretion during prolonged critical illness can
be reactivated. As compared with placebo, continuous infusion of GH-releasing hormone (GHRH; 1 mg/kg/h) was only partially able to reactive GH
secretion, whereas infusion of GH-releasing peptide 2 (GHRP-2; 1 mg/kg/h) and even more so the combination of GHRH and GHRP-2 (1 1 1 mg/kg/h)
could restore the increased pulsatile GH secretion as observed in the acute phase of critical illness. Unlike during acute illness, primarily a condition of
GH resistance, reactivated pulsatile GH secretion in the prolonged phase of critical illness is able to generate peripheral GH responses. This is illustrated by
the regression lines between pulsatile GH secretion and the changes in circulating insulin-like growth factor I (IGF-I), acid-labile substrate (ALS), and
IGF-binding protein-3 (IGFBP-3). These lines indicate that GH responses during prolonged critical illness are proportionate to GH secretion up to a
certain point, beyond which a further increase in GH secretion has apparently little or no additional effect. This point corresponds to a pulsatile GH
secretion that can be evoked by the infusion of GHRP-2 alone. lv = liter of distribution volume. Adapted by permission from Reference 2.

Concise Clinical Review 1339


CONCISE CLINICAL REVIEW

A effects (27). Recent indirect evidence from


Health Acute Critical Illness Prolonged Critical Illness two large randomized controlled trials
7 (RCTs), the EPaNIC (Early versus Late
Parenteral Nutrition in Critically Ill Adults)
trial and the pediatric PEPaNIC trial,
TSH (mlU/l)

supports this possibility. It was shown that


tolerating the substantial macronutrient
deficit that occurs during the first week in
the ICU improves outcomes as compared
with preventing it via early initiation of
0 parenteral nutrition (29, 30). Accepting
21h 06h 21h 06h 21h 06h such “relative fasting” early during critical
illness reduced nosocomial infections,
B prevented liver function abnormalities,
Acute Critical Illness Prolonged Critical Illness prevented muscle weakness, and enhanced
changes in peripheral + neuroendocrine component recovery (29–33). The clinical “harm” by
metabolism and binding
giving parenteral feeding early coincided
with a reversal of the decrease in plasma
TRH TRH ↓ TSH and T3 as well as of the increase in
rT3 (34). Such an effect of early parenteral
nutrition was also shown in an animal
model of critical illness, where it prevented
TSH (↑) = the suppressed expression and activity of
TSH ↓
liver D1 and the activated expression and
activity of D3 and reversed the plasma
phenotype of the low T3 syndrome
(Figure 3) (35). Statistically, part of the
T4 (↑) = T4 ↓ clinical benefit of not feeding patients early
D1
+ D3, D1 D1
+
D3, D1 was explained specifically by the steeper
acute decrease in T3 and in the T3/rT3
ratio. Hence, part of the immediate
↑ T3 rT3 ↑ ↓↓ T3 rT3 (↑)= decrease in T3 and increase in rT3
concentrations during acute critical illness
D3, D1 + D1 D3, D1 + D1 is in fact elicited by the concomitant fasting
rather than the illness and is likely
T2 T2
beneficial (34). Besides less energy
Figure 2. Illustration of the pituitary–thyroid axis responses to acute and prolonged critical illness as expenditure with low T3 availability evoked
compared with the healthy control condition. (A) The nocturnal pulsatile thyroid-stimulating hormone by the shortage of macronutrients,
(TSH) secretion is suppressed during prolonged critical illness. (B) In the acute phase of critical illness, optimized bacterial killing capacity via a
the peripheral changes in thyroid hormone metabolism predominate, whereas in the chronic phase, direct impact of high D3 activity locally
a hypothalamic suppression is superimposed, with (partial) recovery of the peripheral changes.
within granulocytes (36, 37) could also
Adapted by permission from Reference 2. D1–3 = iodothyronine deiodinase type 1–3; rT3 = reverse
T3; T2 = diiodothyronine; T3 = triiodothyronine; T4 = thyroxin; TRH = thyrotropin-releasing hormone.
explain fewer nosocomial infections (29, 30).

The Pituitary–Thyroid Axis in the


present when healthy subjects are fasted. metabolism by free fatty acids and Prolonged Phase of Critical Illness
In critical illness, the severity of the bilirubin (26). In prolonged critically ill patients who for
disease is reflected by the degree of the The immediate decrease in circulating weeks or longer receive, among other
acute decrease in serum T3 (23), and T3 that occurs when otherwise healthy vital support, also full enteral and/or
nonsurvivors have lower T3 levels than subjects fast protects the organism against parenteral nutrition, different changes
survivors (24). hypercatabolism (27, 28). Patients with within the thyroid axis are present (1).
Cytokines have been proposed to illnesses requiring intensive medical care In this later phase, low plasma T4
trigger the low T3 syndrome, although cannot normally feed by mouth and often concentrations accompany the low
cytokine antagonists administered after an are not yet given artificial nutrition. T3 levels, tightly linked with a virtually
endotoxemic challenge cannot restore Inferentially, at least part of the decrease complete loss of pulsatile TSH secretion
normal thyroid function (25). Other factors in circulating thyroid hormone levels in (Figure 2) (38). Also, TRH gene expression
contributing to the acute low T3 syndrome response to acute illness could thus be in the hypothalamic paraventricular nuclei
are decreased concentrations of thyroid fasting induced and hence could represent of patients who died after a prolonged
hormone–binding proteins and inhibition an adaptation to the low availability of critical illness was found to be much lower
of hormone binding, transport, and nutrients and could mediate beneficial than in patients who died suddenly (39).

1340 American Journal of Respiratory and Critical Care Medicine Volume 194 Number 11 | December 1 2016
CONCISE CLINICAL REVIEW

TSH d7
2.5 12 P = 0.001
2.0 11
1.5 10
Δ TSH (mlU/l)

1.0 9
Fasted

mlU/l
0.5 Fed 8
0.0 7 **
–0.5 6
–1.0 5
P = 0.03
–1.5 4
d0 d3 d5 d7 Fasted Fed

T4
T4 d7
5.0 80
P = 0.1
0.0 70
–5.0 60
ΔT4 (nmol/l)

–10.0 50 *

Nmol/l
Fasted
–15.0 40
Fed
–20.0 30
–25.0 20
–30.0 10
P = 0.7
–35.0 0
d0 d3 d5 d7 Fasted Fed

T3 T3 d7

1.0 3.5 P = 0.0001

0.8 *
3.0
0.6
Fasted 2.5
ΔT3 (nmol/l)

0.4 Fed
2.0
Nmol/l

0.2
1.5 *
0.0
1.0
–0.2
–0.4 0.5
P = 0.007
–0.6 0.0
d0 d3 d5 d7 Fasted Fed
Figure 3. Effect of feeding during critical illness, as compared with fasting, on the “low T3 syndrome” in a rabbit model. Feeding critically ill rabbits
intravenously reversed the phenotype of the low T3 syndrome as compared with full fasting for 7 days after onset of illness. The gray areas indicate the
normal reference ranges for healthy rabbits. Asterisks indicate significant differences from values in healthy rabbits. Adapted by permission from Reference
35. d = day; T3 = triiodothyronine; T4 = thyroxin; TSH = thyrotrophin.

In this post mortem study, the suppressed lack of hypothalamic TRH-mediated of a reversible central hypothyroidism in
TRH mRNA expression correlated stimulation of the thyrotropes with prolonged critical illness (40).
positively with the low plasma T3 (39). suppressed TSH-mediated activation of It remains obscure which molecules
Thyroidal production and/or release of the thyroid gland as a result. The finding of drive the hypothalamic suppression in
thyroid hormones thus appears reduced in a supranormal TSH that precedes onset prolonged critical illness. Suspects other
prolonged critical illness because there is of recovery further supports this concept than cytokines must be involved, as plasma

Concise Clinical Review 1341


CONCISE CLINICAL REVIEW

cytokine concentrations are usually much concentrations were elevated (15, 53). corticosteroid-binding globulin and
lower in this late phase (41). High However, in combination with GHRP-2, albumin, but only free cortisol exerts
hypothalamic concentrations of dopamine TRH infusion was capable of normalizing biological activity. Cortisol acts via the
or cortisol are candidate mediators (42–44). plasma T4 and T3 without an increase in glucocorticoid receptor, a 90-kD
An altered set point for feedback rT3 (53), which was explained by a GH- intracellular receptor protein virtually
inhibition within the hypothalamus could mediated suppressive effect on the thyroid expressed in all cells. Considering the fact
be due to locally increased type-2 hormone-inactivating D3 (Figure 4) (54). that glucocorticoid receptor protein levels
deiodinase (D2) expression and activity, This combined releasing factor treatment and glucocorticoid bioavailability are
which can elevate local thyroid hormone for 5 days also induced an anabolic largely similar in most cell types, the striking
availability (45, 46). Increased pituitary D2 response (Figure 4) that was not present cell-specific and sometimes even opposite
could also be possible (47) but was not with GHRP-2 alone. Hence, a causal effects of cortisol require a complex
confirmed in an experimental model (48). relationship between the central differential intracellular regulation (62).
In contrast with the apparent adaptive hypothyroidism and the impaired The molecular basis of cell-specific
nature of the low T3 syndrome present anabolism during prolonged critical illness glucocorticoid responsiveness involves
during acute illness, substantial evidence was suggested (53). A great advantage of glucocorticoid receptor splice variants and
from human and animal studies indicates treatment with hypothalamic releasing differential expression of coreceptor
that peripheral tissues of patients suffering factors is that the negative feedback proteins functioning as coactivators and
from prolonged critical illness show signs inhibition by thyroid hormones at the corepressors of transcription. Also,
of adaptation to the low production of pituitary level is maintained during critical differences in chromatin structure and
thyroid hormones by increasing thyroid illness, which prevents overstimulation of DNA methylation status of glucocorticoid
hormone transporters, local activation of the pituitary–thyroid axis (15, 53). receptor target genes determine variable
thyroid hormone, and increasing the cortisol effects in different tissues (63). The
expression of the active thyroid hormone physiological actions of cortisol range from
receptor isoform. For example, in skeletal The Pituitary–Adrenocortical the complex regulation of the immune
muscle and liver biopsies from prolonged Axis system and metabolism to effects on the
critically ill patients, the monocarboxylate cardiovascular and central nervous system.
transporter-8 was shown to be During health, cortisol is diurnally secreted
overexpressed (49). In an animal model, in pulses from the adrenal cortex, with The Pituitary–Adrenocortical Axis in
such up-regulation of the thyroid hormone the highest levels in the early morning hours the Acute Phase of Critical Illness
transporters in liver and kidney could be and lower levels at night. Cortisol release Circulating cortisol increases substantially
reversed with thyroid hormones (49, 50). is controlled by adrenocorticotropic in response to acute illness or trauma,
Furthermore, local activation of D2 in hormone (ACTH, corticotrophin) produced assumed to be driven by a several-fold
tissues such as skeletal muscle and lungs by the pituitary corticotrophes, in turn increased adrenocortical cortisol production
was documented, which could represent driven by hypothalamic corticotrophin- evoked by increased CRH and ACTH
an adaptive attempt to increase thyroid releasing hormone (CRH) and to a lesser release (55, 64, 65). Briefly, increased
hormone activity locally in these tissues (50, extent vasopressin (55, 56). ACTH plasma ACTH concentrations have indeed
51). And in liver biopsies of prolonged stimulates steroidogenesis by binding to been reported after trauma or sepsis (66).
critically ill patients, an inverse correlation the melanocortin-2 receptor in the Several cytokines can also directly modulate
was observed between on the one hand the adrenocortical cell membrane. ACTH up- cortisol production and the glucocorticoid
ratio of active thyroid hormone receptor regulates the expression of this receptor, receptor number and/or affinity (67). As
TRa1 over the ligand-insensitive isoform drives cholesterol release from the lipid albumin and corticosteroid-binding
TRa2, a surrogate marker of thyroid droplets, and increases the expression globulin levels are substantially decreased,
hormone sensitivity, and on the other hand of proteins required for cholesterol the latter in part due to elastase-induced
the ratio of T3 over rT3 (52). These data uptake (such as the low-density cleavage (68, 69), a proportionally much
suggest that in the chronic phase of critical lipoprotein–receptor and scavenger- higher increase in circulating free cortisol
illness, several tissues seem to require a receptor class B member-1) and for is present (70, 71). The diurnal variation
higher thyroid hormone availability. cholesterol synthesis (3-hydroxy-3- in cortisol secretion disappears with
In line with such an interpretation, low methylglutaryl-CoA reductase) (56–61). acute illnesses or trauma (55, 65). An
T3 levels have been found to correlate ACTH also increases expression of key appropriate activation of the
inversely with markers of muscle steroidogenic enzymes, such as hypothalamic–pituitary–adrenal axis and
breakdown and of bone loss in prolonged steroidogenic acute regulatory protein and cortisol response to acute critical illness is
critical illness (15). In itself, such an the cytochrome P450 cholesterol side-chain vital (72–77). The vital stress-induced
association cannot differentiate between cleavage enzyme CYP11A1 (56–61). hypercortisolism in critically ill patients
cause and consequence (16, 53), but the Besides this feed-forward activation of improves the hemodynamic status through
causality question was addressed by cortisol synthesis/secretion, feedback fluid retention and vasopressor
investigating the effect of TRH treatment. inhibition of CRH and ACTH by cortisol sensitization; provides energy via shifting
A continuous infusion of TRH given to tightly controls its own release. carbohydrate, fat, and protein metabolism;
prolonged critically ill patients was shown Ninety percent of circulating cortisol is and protects against excessive inflammation
to increase plasma T4 and T3, but also rT3 bound to binding proteins, predominantly by suppressing inflammation (1, 67).

1342 American Journal of Respiratory and Critical Care Medicine Volume 194 Number 11 | December 1 2016
CONCISE CLINICAL REVIEW

IGF-I (µg/L) T4 (nmol/L) T3 (nmol/L)

p < 0.0001 p < 0.0001 2.0 p < 0.0001


200

140
160 1.6

120 110
1.2

80
80 .8

40
50 .4
d0 d5 d10 d0 d5 d10 d0 d5 d10

GHRP-2 +TRH / placebo


placebo / GHRP-2 +TRH

Δ % OC over 5 days Δ % urea / creatinine ratio serum

100 p = 0.03

30 p < 0.0001

–80 –30
GHRP-2 placebo d0 d5
+ TRH
Figure 4. Effect of the combined infusion of thyrotropin-releasing hormone (TRH) and the growth hormone–releasing peptide-2 (GHRP-2) on the
“low T3 syndrome” during prolonged critical illness. A continuous infusion of TRH in combination with GHRP-2 in prolonged critically ill fed patients
reversed the hyposomatotropism and the low T3 syndrome, as evidenced via a crossover design with 5 days infusion of GHRP-2 1 TRH followed by
5 days placebo (red symbols) or vice-versa (blue symbols). Five days of GHRP-2 1 TRH infusion was shown to increase a marker of anabolism in bone
and to suppress ureagenesis, suggesting reduced catabolism in skeletal muscle. Adapted by permission from Reference 16. d = day; IGF-1 = insulin-like
growth factor I; OC = osteocalcin; T3 = triiodothyronine; T4 = thyroxin.

The Pituitary–Adrenocortical Axis in activation of the pituitary–adrenocortical ACTH expression levels were found to be
the Prolonged Phase of Critical Illness axis in response to such a high and suppressed in the more chronic phase
In most patients, hypercortisolism is still sustained level of physical stress. It remains of critical illness (82), which could be
present in the chronic phase of critical unclear what is driving the low ACTH evoked by nitric oxide or by suppressed
illness, but given that ACTH levels are now levels in the face of high plasma cortisol. orexin (81, 82). However, if such sepsis-
low, this must be brought about by Reduced ACTH mRNA levels have been induced suppression of pituitary ACTH
alternative, non–ACTH-mediated pathways reported in nine human pituitary glands expression was a sign of organ failure, this
(78, 79). Indeed, a steep decrease in harvested post mortem from patients who would inferentially cause abnormally low
plasma ACTH occurs after a short-lasting died after septic shock as compared with plasma cortisol concentrations, which in
elevation (Figure 5), whereas plasma patients who died suddenly from other patients is usually not the case. Although
cortisol concentrations remain high diseases, in the absence of a compensatory increased adrenocortical sensitivity to
(66, 80). This “ACTH-cortisol dissociation” increase in the expression of CRH or ACTH has been suggested as an alternative
of critical illness is not what one would vasopressin in the hypothalamus (81). Also, explanation (83), the cortisol response to
expect in the context of an ongoing in experimental models of sepsis, pituitary any given endogenous plasma ACTH level

Concise Clinical Review 1343


CONCISE CLINICAL REVIEW

HPA axis during CI HPA axis during recovery HPA axis during prolonged CI

Hypothalamus CRH ? CRH ? CRH ?

ACTH ↑→↓↓ ACTH ↑→nl ACTH ↓↓


Anterior
pituitary
Feedback inhibition

Feedback control
Adrenal cortex Cortisol availability ↑ Cortisol availability nl Cortisol availability low?
- Production nl / ±↑/ ±↓ - Production ↑→nl - Production ↓↓
due to adrenal
Cytokines, toxins, atrophy
low ACTH (loss of ACTH signaling)
Liver/Kidney
- Clearance ↓↓ - Clearance ↑→nl - Clearance ↓↓

A-ring reductases ↓↓
11β-HSD2 ↓
Circulation

Elevated plasma cortisol Normal plasma cortisol Risk of low plasma cortisol
Predominantly driven by
suppressed cortisol clearance
with suppressed ACTH
Figure 5. Schematic overview of the pituitary–adrenocortical axis changes during critical illness (CI) followed by either recovery or prolonged critical illness.
During critical illness, after an initial brief increase in plasma corticotrophin (ACTH), elevated plasma cortisol in the face of paradoxically low plasma ACTH is
explained predominantly by reduced cortisol breakdown in liver and kidney. When recovery follows after a short-lasting illness, the normalization of cortisol
breakdown allows for a rapid normalization of the circulating ACTH and cortisol levels within a tightly feedback-controlled system. When ACTH remains
suppressed for weeks during prolonged critical illness, there is a potential risk of adrenocortical atrophy. 11b-HSD2 = 11b-hydroxysteroid dehydrogenase 2;
CRH = corticotropin-releasing hormone; HPA = hypothalamic–pituitary–adrenocortical; Nl = normal.

is not elevated during critical illness (84), than doubled, in critically ill patients with predominantly the A-ring reductases
and in response to exogenous ACTH it hyperinflammation and unchanged in other in liver and 11b-hydroxysteroid
is quite often low. Furthermore, ACTH critically ill patients as compared with dehydrogenase-2 in kidney, as suggested by
signaling in human adrenal glands was the cortisol production rates of healthy urinary steroid ratios, tracer kinetics, and
found to be unaltered in the acute phase matched control subjects, in the face of assessment of human liver-biopsy samples
and substantially suppressed in the chronic several-fold higher plasma total and free (80). Reduced cortisol clearance was
phase of illness (85). Therefore, another cortisol levels in all patients (80). The stable present irrespective of type and severity
more plausible explanation for high plasma isotope technique also showed that cortisol of illness and irrespective of ICU stay
cortisol concentrations and low plasma plasma clearance was substantially and prognosis. This suggested that a
ACTH levels, with low ACTH-regulated suppressed in all patients irrespective of pronounced suppression of cortisol
gene expression in the adrenal cortex the degree of inflammation. With three breakdown may be the key mechanism that,
during critical illness, is negative feedback subsequent studies, this finding was further together with ongoing normal (or slightly
inhibition exerted by plasma cortisol that is explored (80). Cortisol half-life and plasma elevated) cortisol production, brings about
elevated through alternative, non–ACTH- clearance during critical illness was also the high plasma cortisol in prolonged
driven, pathways. quantified after the administration of a 100-mg critical illness.
Although clinicians always assumed bolus of hydrocortisone. Similar results The new insight that the plasma half-
that cortisol production rate must be about were obtained: plasma cortisol clearance life of cortisol is so much longer during
sixfold higher than normal to generate and reduced to 40% of that in matched control critical illness necessitated another detailed
maintain hypercortisolemia during critical subjects and a cortisol half-life that was a analysis of ACTH and cortisol secretion
illness, it was quantified in ICU patients only median fivefold longer in patients. This rates and their interaction via time series of
recently. Using stable isotopes, a study suppressed cortisol breakdown was due to plasma concentrations measured every
showed that morning cortisol production reduced expression and activity of the 10 minutes overnight in critically ill patients
rate was only moderately increased, less cortisol-metabolizing enzymes, and in healthy control subjects (84). With

1344 American Journal of Respiratory and Critical Care Medicine Volume 194 Number 11 | December 1 2016
CONCISE CLINICAL REVIEW

deconvolution analysis, which takes into above, offers a conceptual frame to better substantially reduced breakdown of steroids
account elimination half-life of the understand the results of RCTs that have during critical illness could explain why any
hormone, the plasma concentration time investigated endocrine and metabolic expected benefits may have been offset by
series were transformed into hormonal interventions and to improve the design of potential harm. In contrast, multicenter
secretion profiles (86). This technique future trials. RCTs performed in critically ill adults
unveiled that in the presence of high total First, a large multicenter RCT and children showed that withholding
and free plasma cortisol concentrations, investigated the impact on outcome of high parenteral nutrition during the first week of
both the nocturnal ACTH and cortisol doses of GH, started on ICU admission critical illness, and thereby accepting virtual
pulsatile secretion rates were reduced and continued into the chronic phase of fasting and its neuroendocrine responses,
rather than increased in critically ill critical illness (91). Instead of showing reduced organ failure, prevented weakness,
patients (87), but the normal feedback the anticipated improved outcome via and accelerated recovery (29–32, 34).
control was active. Thus, beyond the induction of anabolism, this treatment Accepting the fasting responses rather than
very acute phase of critical illness, high doubled mortality. Suppression of cell forcefully feeding patients early in the
nocturnal plasma cortisol concentrations damage removal and overdosing of GH in a course of illness was most beneficial for the
seem to be predominantly maintained by condition of recovered GH sensitivity in the sickest patients, who were at the highest
reduced cortisol breakdown. With only chronic phase of illness possibly played a risk. Such a benefit was at least partially
slightly increased daytime and suppressed role. Second, although only investigated explained by a better preservation of
nighttime cortisol secretion rates, it also in a few small studies, administration of autophagy during fasting, which is
seems that 24-hour cortisol production either substitution doses or high doses of important for an optimal innate immune
rates during critical illness are not, or are thyroid hormones also showed no clinical response and for cell damage removal in
only moderately, higher than during health. outcome benefit, and one was stopped early vital organs and in skeletal muscle (32).
When the adrenal cortex is deprived of for potential harm (92). Again, dosing The findings of these studies also shed new
adequate ACTH signaling for an extended issues of the peripherally active hormones light on the controversial issue of insulin
period of time, as with sustained low and timing of treatment initiation may have treatment during critical illness. Benefits
circulating ACTH that is present in the been important. Third, studies on the were initially shown with very strictly
prolonged phase of illness, its integrity potential impact of hydrocortisone in lowering blood glucose levels to the healthy
may become impaired (Figure 5). A study patients with sepsis, administered at a dose fasting ranges, both in adults and children,
of adrenal cortex biopsies harvested that represents six times the equivalent of an intervention that also accentuated the
immediately post mortem from acute and the cortisol production during critical acute fasting-like neuroendocrine responses
prolonged (>7 d) critically ill patients and illness, revealed conflicting results (67, 93, to illness in the context of early parenteral
from sudden out-of-hospital deaths 94). Such high doses in the context of a feeding (95–98), but risk of harm was
showed a very pronounced depletion
of cholesterol esters and suppressed
expression of ACTH-regulated genes in
adrenal glands from prolonged, but not
Hormone plasma concentration

cortisol
or hormone secretion rate

acute, critically ill patients (85). These


results are quite reminiscent of the
phenotype of proopiomelanocortin- ? Level
deficient mice and thus suggest that a observed
during health
sustained lack of ACTH effect could indeed GH, TSH, ACTH
predispose prolonged critically ill patients
to adrenal atrophy (88). Such risk of
adrenal atrophy in patients with a T3 (and T4), IGF-I
prolonged ICU stay also helps to explain
the reported 20-fold higher incidence of ACUTE PROLONGED RECOVERY (?)
symptomatic adrenal insufficiency in illness illness
critically ill patients being treated in the Figure 6. Simplified cartoon of anterior pituitary–dependent changes during the course of critical
intensive care unit for more than 14 days illness. In the acute phase of illness (first hours to few days after onset), the growth hormone (GH),
(89). It remains to be investigated whether thyrotrophin (TSH), and corticotrophin (ACTH) secretory activity (red) is amplified, whereas levels of
CRH or ACTH could offer a potential to active thyroid hormone (triiodothyronine, T3) and insulin-like growth factor-I (IGF-I) (green) are low,
prevent or treat adrenal failure in the and cortisol levels (yellow) are elevated. In prolonged critical illness (intensive care dependency for
prolonged phase of critical illness (90). weeks), pulsatile secretion of GH, TSH, and ACTH is uniformly suppressed, which drives the low
circulating levels of IGF-I and thyroid hormones. In the prolonged phase of critical illness, plasma
cortisol remains elevated in the presence of low ACTH, largely because of suppressed cortisol
breakdown. With sustained ACTH deficiency, however, adrenal atrophy may ensue, which may
Reconciliation with Results cause inappropriately low cortisol availability. The onset of recovery after acute illness is characterized
from Intervention Trials by increased TSH. Whether this is also the case for GH and ACTH currently remains unclear. Whether
recovery of the anterior pituitary axes after prolonged critical illness is complete in the long term or not
The biphasic nature of the neuroendocrine also remains to be investigated. Adapted by permission with new insights from figures in References
responses to critical illness, as described 1 and 2.

Concise Clinical Review 1345


CONCISE CLINICAL REVIEW

shown in subsequent pragmatic thus should not be interfered with. In respective hormonal axes, and thus
multicenter trials (99). The role of tight contrast, the uniform suppression of the toxic side effects of high peripheral
blood glucose control versus no blood neuroendocrine axes observed in the hormone levels, is avoided by intact
glucose control should thus be prolonged fed state of critical illness negative feed-back control and by
reinvestigated in the context of not giving might be harmful. Treatment with the ability to adaptively change
parenteral nutrition during the first week hypothalamic releasing factors in the peripheral hormonal metabolism
in the ICU. prolonged phase of illness can reactivate (15, 16, 53). Future intervention studies
the anterior pituitary, which may be more should focus more on patients with
effective and safer than administration of prolonged critical illness and investigate
Conclusions the potential of combined administration
the peripherally active hormones (GH,
of the hypothalamic releasing factors that
The hypothalamic–anterior pituitary thyroid hormones, hydrocortisone),
have been identified as suppressed or
axes respond biphasically to severe illness which have been shown to be either not
deficient. n
and trauma (Figure 6). The acute effective or even to cause harm. Indeed, a
adaptations represent at least partial particular advantage of releasing Author disclosures are available with the text
responses to fasting, which is beneficial and factors is that overstimulation of the of this article at www.atsjournals.org.

References 13. Defalque D, Brandt N, Ketelslegers JM, Thissen JP. GH insensitivity


induced by endotoxin injection is associated with decreased liver GH
1. Van den Berghe G, de Zegher F, Bouillon R. Clinical review 95: acute and receptors. Am J Physiol 1999;276:E565–E572.
prolonged critical illness as different neuroendocrine paradigms. J Clin 14. Van den Berghe G, de Zegher F, Veldhuis JD, Wouters P, Awouters M,
Endocrinol Metab 1998;83:1827–1834. Verbruggen W, Schetz M, Verwaest C, Lauwers P, Bouillon R, et al.
2. Vanhorebeek I, Langouche L, Van den Berghe G. Endocrine aspects of The somatotropic axis in critical illness: effect of continuous growth
acute and prolonged critical illness. Nat Clin Pract Endocrinol Metab hormone (GH)-releasing hormone and GH-releasing peptide-2
2006;2:20–31. infusion. J Clin Endocrinol Metab 1997;82:590–599.
3. Thorner MO, Vance ML, Laws ER, Horvath E, Kovacs K. The anterior 15. Van den Berghe G, de Zegher F, Baxter RC, Veldhuis JD, Wouters P,
pituitary. In: Wilson JD, Foster D, Kronenberg HM, Larsen PR, editors. Schetz M, Verwaest C, Van der Vorst E, Lauwers P, Bouillon R, et al.
Williams’ textbook of endocrinology, 9th ed. Philadelphia: WB Neuroendocrinology of prolonged critical illness: effects of
Saunders; 1998. pp. 249–340. exogenous thyrotropin-releasing hormone and its combination with
growth hormone secretagogues. J Clin Endocrinol Metab 1998;83:
4. Kojima M, Hosoda H, Date Y, Nakazato M, Matsuo H, Kangawa K.
309–319.
Ghrelin is a growth-hormone-releasing acylated peptide from
16. Van den Berghe G, Wouters P, Weekers F, Mohan S, Baxter RC,
stomach. Nature 1999;402:656–660.
Veldhuis JD, Bowers CY, Bouillon R. Reactivation of pituitary
5. Cowley MA, Smith RG, Diano S, Tschöp M, Pronchuk N, Grove KL,
hormone release and metabolic improvement by infusion of growth
Strasburger CJ, Bidlingmaier M, Esterman M, Heiman ML, et al. The
hormone-releasing peptide and thyrotropin-releasing hormone in
distribution and mechanism of action of ghrelin in the CNS
patients with protracted critical illness. J Clin Endocrinol Metab 1999;
demonstrates a novel hypothalamic circuit regulating energy
84:1311–1323.
homeostasis. Neuron 2003;37:649–661.
17. Yen PM. Physiological and molecular basis of thyroid hormone action.
6. Bowers CY, Momany FA, Reynolds GA, Hong A. On the in vitro and
Physiol Rev 2001;81:1097–1142.
in vivo activity of a new synthetic hexapeptide that acts on the
18. Bianco AC, Salvatore D, Gereben B, Berry MJ, Larsen PR.
pituitary to specifically release growth hormone. Endocrinology 1984;
Biochemistry, cellular and molecular biology, and physiological roles
114:1537–1545.
of the iodothyronine selenodeiodinases. Endocr Rev 2002;23:38–89.
7. Howard AD, Feighner SD, Cully DF, Arena JP, Liberator PA, 19. Friesema EC, Jansen J, Visser TJ. Thyroid hormone transporters.
Rosenblum CI, Hamelin M, Hreniuk DL, Palyha OC, Anderson J, et al. Biochem Soc Trans 2005;33:228–232.
A receptor in pituitary and hypothalamus that functions in growth 20. Chopra IJ, Huang TS, Beredo A, Solomon DH, Chua Teco GN, Mead JF.
hormone release. Science 1996;273:974–977. Evidence for an inhibitor of extrathyroidal conversion of thyroxine to
8. Baxter RC. Changes in the IGF-IGFBP axis in critical illness. Best Pract 3,5,39-triiodothyronine in sera of patients with nonthyroidal illnesses.
Res Clin Endocrinol Metab 2001;15:421–434. J Clin Endocrinol Metab 1985;60:666–672.
9. Van den Berghe G, Baxter RC, Weekers F, Wouters P, 21. Peeters RP, Wouters PJ, Kaptein E, van Toor H, Visser TJ, Van den
Bowers CY, Veldhuis JD. A paradoxical gender dissociation Berghe G. Reduced activation and increased inactivation of thyroid
within the growth hormone/insulin-like growth factor I axis hormone in tissues of critically ill patients. J Clin Endocrinol Metab
during protracted critical illness. J Clin Endocrinol Metab 2000; 2003;88:3202–3211.
85:183–192. 22. Michalaki M, Vagenakis AG, Makri M, Kalfarentzos F, Kyriazopoulou V.
10. Ross R, Miell J, Freeman E, Jones J, Matthews D, Preece M, Buchanan C. Dissociation of the early decline in serum T(3) concentration and
Critically ill patients have high basal growth hormone levels with serum IL-6 rise and TNFalpha in nonthyroidal illness syndrome
attenuated oscillatory activity associated with low levels of insulin-like induced by abdominal surgery. J Clin Endocrinol Metab 2001;86:
growth factor-I. Clin Endocrinol (Oxf) 1991;35:47–54. 4198–4205.
11. Baxter RC, Hawker FH, To C, Stewart PM, Holman SR. Thirty- 23. Van den Berghe G. Novel insights into the neuroendocrinology of
day monitoring of insulin-like growth factors and their binding critical illness. Eur J Endocrinol 2000;143:1–13.
proteins in intensive care unit patients. Growth Horm IGF Res 24. Peeters RP, Wouters PJ, van Toor H, Kaptein E, Visser TJ, Van den
1998;8:455–463. Berghe G. Serum 3,39,59-triiodothyronine (rT3) and 3,5,39-
12. Hermansson M, Wickelgren RB, Hammarqvist F, Bjarnason R, triiodothyronine/rT3 are prognostic markers in critically ill patients
Wennström I, Wernerman J, Carlsson B, Carlsson LM. Measurement and are associated with postmortem tissue deiodinase activities.
of human growth hormone receptor messenger ribonucleic acid J Clin Endocrinol Metab 2005;90:4559–4565.
by a quantitative polymerase chain reaction-based assay: 25. van der Poll T, Van Zee KJ, Endert E, Coyle SM, Stiles DM, Pribble JP,
demonstration of reduced expression after elective surgery. J Clin Catalano MA, Moldawer LL, Lowry SF. Interleukin-1 receptor
Endocrinol Metab 1997;82:421–428. blockade does not affect endotoxin-induced changes in plasma

1346 American Journal of Respiratory and Critical Care Medicine Volume 194 Number 11 | December 1 2016
CONCISE CLINICAL REVIEW

thyroid hormone and thyrotropin concentrations in man. J Clin 43. Van den Berghe G, de Zegher F, Lauwers P. Dopamine and the sick
Endocrinol Metab 1995;80:1341–1346. euthyroid syndrome in critical illness. Clin Endocrinol (Oxf) 1994;41:
26. Lim CF, Docter R, Visser TJ, Krenning EP, Bernard B, van Toor H, 731–737.
de Jong M, Hennemann G. Inhibition of thyroxine transport into 44. Van den Berghe G, de Zegher F, Lauwers P. Dopamine suppresses
cultured rat hepatocytes by serum of nonuremic critically ill patients: pituitary function in infants and children. Crit Care Med 1994;22:
effects of bilirubin and nonesterified fatty acids. J Clin Endocrinol 1747–1753.
Metab 1993;76:1165–1172. 45. Boelen A, Kwakkel J, Thijssen-Timmer DC, Alkemade A, Fliers E,
27. Gardner DF, Kaplan MM, Stanley CA, Utiger RD. Effect of Wiersinga WM. Simultaneous changes in central and peripheral
tri-iodothyronine replacement on the metabolic and pituitary components of the hypothalamus-pituitary-thyroid axis in
responses to starvation. N Engl J Med 1979;300:579–584. lipopolysaccharide-induced acute illness in mice. J Endocrinol 2004;
28. Moshang T Jr, Parks JS, Baker L, Vaidya V, Utiger RD, Bongiovanni AM, 182:315–323.
Snyder PJ. Low serum triiodothyronine in patients with anorexia 46. Mebis L, Debaveye Y, Ellger B, Derde S, Ververs EJ, Langouche L,
nervosa. J Clin Endocrinol Metab 1975;40:470–473. Darras VM, Fliers E, Visser TJ, Van den Berghe G. Changes in the
29. Casaer MP, Mesotten D, Hermans G, Wouters PJ, Schetz M, Meyfroidt G, central component of the hypothalamus-pituitary-thyroid axis in a
Van Cromphaut S, Ingels C, Meersseman P, Muller J, et al. Early versus rabbit model of prolonged critical illness. Crit Care 2009;13:R147.
late parenteral nutrition in critically ill adults. N Engl J Med 2011;365: 47. Alkemade A, Friesema EC, Kuiper GG, Wiersinga WM, Swaab DF,
506–517. Visser TJ, Fliers E. Novel neuroanatomical pathways for thyroid
30. Fivez T, Kerklaan D, Mesotten D, Verbruggen S, Wouters PJ, hormone action in the human anterior pituitary. Eur J Endocrinol
Vanhorebeek I, Debaveye Y, Vlasselaers D, Desmet L, Casaer MP, 2006;154:491–500.
et al. Early versus late parenteral nutrition in critically ill children. 48. Langouche L, Princen L, Gunst J, Güiza F, Derde S, Van den Berghe G.
N Engl J Med 2016;374:1111–1122. Anterior pituitary morphology and hormone production during
31. Casaer MP, Wilmer A, Hermans G, Wouters PJ, Mesotten D, Van den sustained critical illness in a rabbit model. Horm Metab Res 2013;45:
Berghe G. Role of disease and macronutrient dose in the randomized 277–282.
controlled EPaNIC trial: a post hoc analysis. Am J Respir Crit Care 49. Mebis L, Paletta D, Debaveye Y, Ellger B, Langouche L, D’Hoore A,
Med 2013;187:247–255. Darras VM, Visser TJ, Van den Berghe G. Expression of thyroid
32. Hermans G, Casaer MP, Clerckx B, Güiza F, Vanhullebusch T, Derde S, hormone transporters during critical illness. Eur J Endocrinol 2009;
Meersseman P, Derese I, Mesotten D, Wouters PJ, et al. Effect of 161:243–250.
tolerating macronutrient deficit on the development of intensive-care 50. Mebis L, Langouche L, Visser TJ, Van den Berghe G. The type II
unit acquired weakness: a subanalysis of the EPaNIC trial. Lancet iodothyronine deiodinase is up-regulated in skeletal muscle during
Respir Med 2013;1:621–629. prolonged critical illness. J Clin Endocrinol Metab 2007;92:
33. Gunst J, Vanhorebeek I, Casaer MP, Hermans G, Wouters PJ, Dubois J, 3330–3333.
Claes K, Schetz M, Van den Berghe G. Impact of early parenteral 51. Ma SF, Xie L, Pino-Yanes M, Sammani S, Wade MS, Letsiou E, Siegler J,
nutrition on metabolism and kidney injury. J Am Soc Nephrol 2013;24: Wang T, Infusino G, Kittles RA, et al. Type 2 deiodinase and host
995–1005. responses of sepsis and acute lung injury. Am J Respir Cell Mol Biol
2011;45:1203–1211.
34. Langouche L, Vander Perre S, Marques M, Boelen A, Wouters PJ,
52. Thijssen-Timmer DC, Peeters RP, Wouters P, Weekers F, Visser TJ,
Casaer MP, Van den Berghe G. Impact of early nutrient restriction
Fliers E, Wiersinga WM, Bakker O, Van Den Berghe G. Thyroid
during critical illness on the nonthyroidal illness syndrome and its
hormone receptor isoform expression in livers of critically ill patients.
relation with outcome: a randomized, controlled clinical study. J Clin
Thyroid 2007;17:105–112.
Endocrinol Metab 2013;98:1006–1013.
53. Van den Berghe G, Baxter RC, Weekers F, Wouters P, Bowers CY,
35. Mebis L, Eerdekens A, Güiza F, Princen L, Derde S, Vanwijngaerden YM,
Iranmanesh A, Veldhuis JD, Bouillon R. The combined administration
Vanhorebeek I, Darras VM, Van den Berghe G, Langouche L.
of GH-releasing peptide-2 (GHRP-2), TRH and GnRH to men with
Contribution of nutritional deficit to the pathogenesis of the
prolonged critical illness evokes superior endocrine and metabolic
nonthyroidal illness syndrome in critical illness: a rabbit model study.
effects compared to treatment with GHRP-2 alone. Clin Endocrinol
Endocrinology 2012;153:973–984.
(Oxf) 2002;56:655–669.
36. Boelen A, Kwakkel J, Fliers E. Beyond low plasma T3: local thyroid
54. Weekers F, Michalaki M, Coopmans W, Van Herck E, Veldhuis JD,
hormone metabolism during inflammation and infection. Endocr Rev
Darras VM, Van den Berghe G. Endocrine and metabolic
2011;32:670–693.
effects of growth hormone (GH) compared with GH-releasing
37. Boelen A, Boorsma J, Kwakkel J, Wieland CW, Renckens R, Visser TJ, peptide, thyrotropin-releasing hormone, and insulin infusion in a
Fliers E, Wiersinga WM. Type 3 deiodinase is highly expressed in rabbit model of prolonged critical illness. Endocrinology 2004;145:
infiltrating neutrophilic granulocytes in response to acute bacterial 205–213.
infection. Thyroid 2008;18:1095–1103. 55. Cooper MS, Stewart PM. Corticosteroid insufficiency in acutely ill
38. Van den Berghe G, de Zegher F, Veldhuis JD, Wouters P, Gouwy S, patients. N Engl J Med 2003;348:727–734.
Stockman W, Weekers F, Schetz M, Lauwers P, Bouillon R, et al. 56. Lebrethon MC, Naville D, Begeot M, Saez JM. Regulation of
Thyrotrophin and prolactin release in prolonged critical illness: corticotropin receptor number and messenger RNA in cultured
dynamics of spontaneous secretion and effects of growth hormone- human adrenocortical cells by corticotropin and angiotensin II. J Clin
secretagogues. Clin Endocrinol (Oxf) 1997;47:599–612. Invest 1994;93:1828–1833.
39. Fliers E, Guldenaar SE, Wiersinga WM, Swaab DF. Decreased 57. Lehoux JG, Lefebvre A, Bélisle S, Bellabarba D. Hormonal regulation of
hypothalamic thyrotropin-releasing hormone gene expression in 3-hydroxy-3-methylglutaryl coenzyme A reductase mRNA in the rat
patients with nonthyroidal illness. J Clin Endocrinol Metab 1997;82: adrenal gland. J Steroid Biochem 1989;34:379–384.
4032–4036. 58. Lehoux JG, Fleury A, Ducharme L. The acute and chronic effects of
40. Bacci V, Schussler GC, Kaplan TB. The relationship between serum adrenocorticotropin on the levels of messenger ribonucleic acid and
triiodothyronine and thyrotropin during systemic illness. J Clin protein of steroidogenic enzymes in rat adrenal in vivo.
Endocrinol Metab 1982;54:1229–1235. Endocrinology 1998;139:3913–3922.
41. Damas P, Reuter A, Gysen P, Demonty J, Lamy M, Franchimont P. 59. Liu J, Heikkilä P, Meng QH, Kahri AI, Tikkanen MJ, Voutilainen R.
Tumor necrosis factor and interleukin-1 serum levels Expression of low and high density lipoprotein receptor genes in
during severe sepsis in humans. Crit Care Med 1989;17: human adrenals. Eur J Endocrinol 2000;142:677–682.
975–978. 60. Simpson ER, Waterman MR. Regulation by ACTH of steroid hormone
42. Faglia G, Ferrari C, Beck-Peccoz P, Spada A, Travaglini P, Ambrosi B. biosynthesis in the adrenal cortex. Can J Biochem Cell Biol 1983;61:
Reduced plasma thyrotropin response to thyrotropin releasing 692–707.
hormone after dexamethasone administration in normal subjects. 61. Stocco DM, Clark BJ. Regulation of the acute production of steroids in
Horm Metab Res 1973;5:289–292. steroidogenic cells. Endocr Rev 1996;17:221–244.

Concise Clinical Review 1347


CONCISE CLINICAL REVIEW

62. Vandevyver S, Dejager L, Tuckermann J, Libert C. New insights into the 82. Deutschman CS, Raj NR, McGuire EO, Kelz MB. Orexinergic activity
anti-inflammatory mechanisms of glucocorticoids: an emerging role modulates altered vital signs and pituitary hormone secretion in
for glucocorticoid-receptor-mediated transactivation. Endocrinology experimental sepsis. Crit Care Med 2013;41:e368–e375.
2013;154:993–1007. 83. Gibbison B, Angelini GD, Lightman SL. Dynamic output and control of
63. Miesfeld RL. Glucocorticoid action: biochemistry. In: DeGroot LJ, the hypothalamic-pituitary-adrenal axis in critical illness and major
Jameson JL, editors. Endocrinology. Philadelphia: WB Saunders surgery. Br J Anaesth 2013;111:347–360.
Company; 2001. pp. 1647–1654. 84. Boonen E, Meersseman P, Vervenne H, Meyfroidt G, Guı̈za F, Wouters
64. Rivier C, Vale W. Modulation of stress-induced ACTH release by PJ, Veldhuis JD, Van den Berghe G. Reduced nocturnal ACTH-driven
corticotropin-releasing factor, catecholamines and vasopressin. cortisol secretion during critical illness. Am J Physiol Endocrinol
Nature 1983;305:325–327. Metab 2014;306:E883–E892.
65. Burchard K. A review of the adrenal cortex and severe inflammation: 85. Boonen E, Langouche L, Janssens T, Meersseman P, Vervenne H,
quest of the “eucorticoid” state. J Trauma 2001;51:800–814. De Samblanx E, Pironet Z, Van Dijck L, Vander Perre S, Derese I, et al.
66. Vermes I, Beishuizen A, Hampsink RM, Haanen C. Dissociation of Impact of duration of critical illness on the adrenal glands of human
plasma adrenocorticotropin and cortisol levels in critically ill patients: intensive care patients. J Clin Endocrinol Metab 2014;99:4214–4222.
possible role of endothelin and atrial natriuretic hormone. J Clin 86. Veldhuis JD, Keenan DM, Pincus SM. Motivations and methods for
Endocrinol Metab 1995;80:1238–1242. analyzing pulsatile hormone secretion. Endocr Rev 2008;29:
67. Marik PE, Zaloga GP. Adrenal insufficiency in the critically ill: a new look 823–864.
at an old problem. Chest 2002;122:1784–1796. 87. Waterhouse R. A case of suprarenal apoplexy. Lancet 1911;1:577–578.
68. Pemberton PA, Stein PE, Pepys MB, Potter JM, Carrell RW. Hormone 88. Karpac J, Czyzewska K, Kern A, Brush RS, Anderson RE,
binding globulins undergo serpin conformational change in Hochgeschwender U. Failure of adrenal corticosterone production in
inflammation. Nature 1988;336:257–258. POMC-deficient mice results from lack of integrated effects of
69. Hammond GL, Smith CL, Paterson NA, Sibbald WJ. A role for POMC peptides on multiple factors. Am J Physiol Endocrinol Metab
corticosteroid-binding globulin in delivery of cortisol to activated 2008;295:E446–E455.
neutrophils. J Clin Endocrinol Metab 1990;71:34–39. 89. Barquist E, Kirton O. Adrenal insufficiency in the surgical intensive care
70. Beishuizen A, Thijs LG, Vermes I. Patterns of corticosteroid-binding unit patient. J Trauma 1997;42:27–31.
globulin and the free cortisol index during septic shock and
90. Boonen E, Van den Berghe G. Endocrine responses to critical illness:
multitrauma. Intensive Care Med 2001;27:1584–1591.
novel insights and therapeutic implications. J Clin Endocrinol Metab
71. Hamrahian AH, Oseni TS, Arafah BM. Measurements of serum free
2014;99:1569–1582.
cortisol in critically ill patients. N Engl J Med 2004;350:1629–1638.
91. Takala J, Ruokonen E, Webster NR, Nielsen MS, Zandstra DF,
72. Finlay WE, McKee JI. Serum cortisol levels in severely stressed
Vundelinckx G, Hinds CJ. Increased mortality associated with growth
patients. Lancet 1982;1:1414–1415.
hormone treatment in critically ill adults. N Engl J Med 1999;341:
73. Rothwell PM, Udwadia ZF, Lawler PG. Cortisol response to
785–792.
corticotropin and survival in septic shock. Lancet 1991;337:582–583.
92. Van den Berghe G. Non-thyroidal illness in the ICU: a syndrome with
74. Span LF, Hermus AR, Bartelink AK, Hoitsma AJ, Gimbrère JS, Smals
different faces. Thyroid 2014;24:1456–1465.
AG, Kloppenborg PW. Adrenocortical function: an indicator of
severity of disease and survival in chronic critically ill patients. 93. Sprung CL, Annane D, Keh D, Moreno R, Singer M, Freivogel K, Weiss YG,
Intensive Care Med 1992;18:93–96. Benbenishty J, Kalenka A, Forst H, et al.; CORTICUS Study Group.
75. Annane D, Sébille V, Troché G, Raphaël JC, Gajdos P, Bellissant E. Hydrocortisone therapy for patients with septic shock. N Engl J Med
A 3-level prognostic classification in septic shock based on cortisol 2008;358:111–124.
levels and cortisol response to corticotropin. JAMA 2000;283: 94. Mesotten D, Vanhorebeek I, Van den Berghe G. The altered adrenal
1038–1045. axis and treatment with glucocorticoids during critical illness. Nat
76. Annane D, Sébille V, Charpentier C, Bollaert PE, François B, Korach JM, Clin Pract Endocrinol Metab 2008;4:496–505.
Capellier G, Cohen Y, Azoulay E, Troché G, et al. Effect of treatment 95. van den Berghe G, Wouters P, Weekers F, Verwaest C, Bruyninckx F,
with low doses of hydrocortisone and fludrocortisone on mortality in Schetz M, Vlasselaers D, Ferdinande P, Lauwers P, Bouillon R.
patients with septic shock. JAMA 2002;288:862–871. Intensive insulin therapy in critically ill patients. N Engl J Med 2001;
77. Sam S, Corbridge TC, Mokhlesi B, Comellas AP, Molitch ME. Cortisol 345:1359–1367.
levels and mortality in severe sepsis. Clin Endocrinol (Oxf) 2004;60: 96. Van den Berghe G, Wilmer A, Hermans G, Meersseman W, Wouters PJ,
29–35. Milants I, Van Wijngaerden E, Bobbaers H, Bouillon R. Intensive
78. Bornstein SR, Chrousos GP. Clinical review 104: adrenocorticotropin insulin therapy in the medical ICU. N Engl J Med 2006;354:449–461.
(ACTH)- and non-ACTH-mediated regulation of the adrenal cortex: 97. Vlasselaers D, Milants I, Desmet L, Wouters PJ, Vanhorebeek I, van den
neural and immune inputs. J Clin Endocrinol Metab 1999;84: Heuvel I, Mesotten D, Casaer MP, Meyfroidt G, Ingels C, et al.
1729–1736. Intensive insulin therapy for patients in paediatric intensive care: a
79. Vermes I, Beishuizen A. The hypothalamic-pituitary-adrenal response prospective, randomised controlled study. Lancet 2009;373:
to critical illness. Best Pract Res Clin Endocrinol Metab 2001;15: 547–556.
495–511. 98. Gielen M, Mesotten D, Wouters PJ, Desmet L, Vlasselaers D,
80. Boonen E, Vervenne H, Meersseman P, Andrew R, Mortier L, Declercq PE, Vanhorebeek I, Langouche L, Van den Berghe G. Effect of tight
Vanwijngaerden YM, Spriet I, Wouters PJ, Vander Perre S, et al. glucose control with insulin on the thyroid axis of critically ill children
Reduced cortisol metabolism during critical illness. N Engl J Med 2013; and its relation with outcome. J Clin Endocrinol Metab 2012;97:
368:1477–1488. 3569–3576.
81. Polito A, Sonneville R, Guidoux C, Barrett L, Viltart O, Mattot V, Siami S, 99. Finfer S, Chittock DR, Su SY, Blair D, Foster D, Dhingra V, Bellomo R,
Lorin de la Grandmaison G, Chrétien F, Singer M, et al. Changes in Cook D, Dodek P, Henderson WR, et al.; NICE-SUGAR Study
CRH and ACTH synthesis during experimental and human septic Investigators. Intensive versus conventional glucose control in
shock. Plos One 2011;6:e25905. critically ill patients. N Engl J Med 2009;360:1283–1297.

1348 American Journal of Respiratory and Critical Care Medicine Volume 194 Number 11 | December 1 2016

You might also like