Download as pdf or txt
Download as pdf or txt
You are on page 1of 85

Physiol Rev 95: 1025–1109, 2015

Published July 1, 2015; doi:10.1152/physrev.00028.2014

THE SICK AND THE WEAK: NEUROPATHIES/


MYOPATHIES IN THE CRITICALLY ILL
O. Friedrich, M. B. Reid, G. Van den Berghe, I. Vanhorebeek, G. Hermans, M. M. Rich,
and L. Larsson

Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-


University Erlangen-Nuremberg, Erlangen, Germany; College of Health and Human Performance, University
of Florida, Gainesville, Florida; Clinical Department and Laboratory of Intensive Care Medicine, Division of
Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; Department of Neuroscience, Cell Biology
and Physiology, Wright State University, Dayton, Ohio; and Department of Physiology and Pharmacology,
Department of Clinical Neuroscience, Clinical Neurophysiology, Karolinska Institutet, Stockholm, Sweden

Friedrich O, Reid MB, Van den Berghe G, Vanhorebeek I, Hermans G, Rich MM, and

L
Larsson L. The Sick and the Weak: Neuropathies/Myopathies in the Critically Ill. Physiol Rev
95: 1025–1109, 2015; Published July 1, 2015; doi:10.1152/physrev.00028.2014.—
Critical illness polyneuropathies (CIP) and myopathies (CIM) are common complica-
tions of critical illness. Several weakness syndromes are summarized under the
term intensive care unit-acquired weakness (ICUAW). We propose a classification of different
ICUAW forms (CIM, CIP, sepsis-induced, steroid-denervation myopathy) and pathophysiological
mechanisms from clinical and animal model data. Triggers include sepsis, mechanical ventila-
tion, muscle unloading, steroid treatment, or denervation. Some ICUAW forms require strin-
gent diagnostic features; CIM is marked by membrane hypoexcitability, severe atrophy, pref-
erential myosin loss, ultrastructural alterations, and inadequate autophagy activation while
myopathies in pure sepsis do not reproduce marked myosin loss. Reduced membrane excit-
ability results from depolarization and ion channel dysfunction. Mitochondrial dysfunction con-
tributes to energy-dependent processes. Ubiquitin proteasome and calpain activation trigger
muscle proteolysis and atrophy while protein synthesis is impaired. Myosin loss is more
pronounced than actin loss in CIM. Protein quality control is altered by inadequate autophagy.
Ca2⫹ dysregulation is present through altered Ca2⫹ homeostasis. We highlight clinical hall-
marks, trigger factors, and potential mechanisms from human studies and animal models that
allow separation of risk factors that may trigger distinct mechanisms contributing to weakness.
During critical illness, altered inflammatory (cytokines) and metabolic pathways deteriorate
muscle function. ICUAW prevention/treatment is limited, e.g., tight glycemic control, delaying
nutrition, and early mobilization. Future challenges include identification of primary/secondary
events during the time course of critical illness, the interplay between membrane excitability,
bioenergetic failure and differential proteolysis, and finding new therapeutic targets by help of
tailored animal models.

I. INTRODUCTION 1025 I. INTRODUCTION


II. HISTORY, TIME COURSE, AND... 1026
III. RISK FACTORS AND CLINICAL... 1028 “In adults the disease may attack persons in good health,
IV. INTRODUCTION TO... 1035 but who are overworked or ‘run down’ from any cause.
V. INFLAMMATORY CYTOKINES... 1036 Haemorrhage initiates the attack in a few cases. There may
VI. BIOMECHANICS, ... 1041 be repeated chills; the temperature is high, the pulse rapid,
VII. NEUROMUSCULAR TRANSMISSION... 1044 and the respirations are increased. The loss of flesh and
VIII. MEMBRANE AND ION CHANNEL... 1046 strength is very striking.” This description of the symptoms
IX. EXCITATION-CONTRACTION COUPLING,... 1053 in a condition of acute critical illness, in acute broncho-
X. METABOLIC EFFECTS IN CRITICAL... 1058 pneumonic phthisis, by Sir William Osler in his classical
XI. PROTEIN REGULATION IN MUSCLE... 1067 textbook on “principles and practice of medicine” (531) is
XII. ANIMAL AND HUMAN MODELS... 1072 one of the first analytical observations linking an aberrant
XIII. TREATMENT OF ICUAW: DRUG... 1082 systemic inflammatory response to a failure of the periph-
XIV. SUMMARY/OUTLOOK 1088 eral nervous system, i.e., muscle wasting and weakness.

0031-9333/15 Copyright © 2015 the American Physiological Society 1025


FRIEDRICH ET AL.

This condition can either affect the peripheral nerves (crit- the occurrence of polyneuropathies in coma patients fol-
ical illness polyneuropathy, CIP), skeletal muscle (critical lowing shock, cardiac arrest, and acute intoxication and in
illness myopathy, CIM) or both (critical illness polyneuro- burn patients (65). However, it took another century before
myopathy, CIPNM) (6, 402, 404, 747). These conditions this syndrome was described in more detail and was labeled
are the primary cause of muscle weakness and paralysis as CIP (67). Almost simultaneously, several authors re-
during and following critical illness (402). Weakness ap- ported the occurrence of profound flaccid and symmetrical
pears to be triggered by critical illness and the ICU course weakness after a severe disease (35, 66, 68, 134, 590, 812).
regardless of the underlying primary condition (382, 453, The weakness was accompanied by muscle wasting and
528). The chance of developing muscle weakness correlates often, but not necessarily, by reduction or loss of tendon
with the severity of critical illness, as assessed by several reflexes but with a relative sparing of the facial muscles.
organ dysfunction scores (149, 181, 370, 763), and repre- This typically results in grimacing of the facial muscles upon
sents a pathophysiological process that cannot be explained painful limb stimulation but with evoking only limited to
by immobilization alone (181). Mechanical ventilation and
absent limb movement. Distal muscles (781, 812) and lower
muscle immobilization, severe sepsis, and multiple organ
limbs (68, 206, 360, 406, 414) appeared most severely af-
dysfunction as well as neuro/myotoxic agents are among
fected. The predominant symptom pointing to the presence
the primary risk factors of ICU-acquired neuromyopathy
of CIP was often weaning failure, as respiratory muscles are
(215, 302, 747). With the advancements in modern inten-
sive care medicine, mortality of sepsis-related ICU condi- also involved (52, 145, 414, 781). Sensory loss, including
tions has dropped; however, at the cost of a growing inci- reduced sensitivity to pain, temperature, and vibration, was
dence of such intensive care unit (ICU)-acquired weak- variably present (414, 808), but sensory testing in these
nesses (ICUAW) (64, 332, 622). As sepsis ranks among the patients is often unreliable (52, 64, 808, 812) (TABLE 1).
top three causes of mortality in some Western countries Electrophysiological studies revealed reduced compound
(185), with incidences of weakness as high as 50 –100% in motor action potentials and sensory nerve action potentials,
critically ill patients (145, 361, 781), ICUAW is becoming a variably accompanied by the widespread presence of fibril-
major problem. Its prevalence not only influences patient lation potentials or positive sharp waves. In contrast to the
prognosis but also bears threats for secondary complica- Guillain-Barré syndrome where a demyelination of periph-
tions (infection, embolism, etc.), prolongs ICU treatment eral nerves results in delayed action potential conduction,
and rehabilitation, and greatly raises the costs of intensive nerve conduction velocity was found to be normal or near
care medicine worldwide (406, 458). As will be detailed normal (52, 68, 126, 414, 781, 808, 812). Biopsy and au-
later, the recently introduced term ICUAW (618) shall be topsy findings confirmed that the underlying problem was a
considered a syndromal umbrella term and is not synony- primary distal axonal degeneration of motor and sensory
mous with CIM or CIP “per se” but more pragmatically fibers (44, 67, 68, 193, 812). Later, it was shown that in
describes a syndromal complex of weakness in critically ill some patients with electrophysiological evidence of CIP,
patients who may be suffering from different pathophysio- nerve histology was normal (403), indicating that func-
logical entities. For most of these ICUAWs, specific thera- tional changes may precede structural changes. Consistent
pies do not yet exist, i.e., for CIP and CIM, because of our with this, rapidly reversible neuropathy was later described
patchy knowledge of their underlying pathological mecha- (515). Whether this concerns a distinct entity or a different
nisms. However, recent research has identified several inter- grade of severity is yet unresolved (TABLE 1). When consid-
playing pathways and candidates that might serve as ther- ering weaning failure, electrophysiological and autopsy
apy targets. An important issue in dissecting the different studies supported the involvement of the phrenic nerve,
entities of ICUAW and their pathophysiological mecha- diaphragm, as well as the intercostal and other accessory
nisms, the definition of proper animal models that as closely
muscles in the process of axonal degeneration and denerva-
as possible mimick the phenotype of muscle weakness seen
tion-associated muscle atrophy (450, 611, 781, 808, 812).
in ICU patients, is crucial. Therefore, in addition to sum-
Recovery from CIP first occurs in the upper and proximal
marizing and discussing the paucity of pathophysiological
lower limbs. This is then followed by recovery of the respi-
data, our review will also provide a special emphasis on
such animal models and compare the pros and cons in- ratory system and finally by the distal lower limbs (812).
volved.
In parallel with the description and characterization of CIP,
a case of acute quadriplegia developing in a patient treated
II. HISTORY, TIME COURSE, AND with corticosteroids and neuromuscular blocking agents
CLINICAL FEATURES OF ICU-RELATED was reported (448). Several other authors, by use of muscle
WEAKNESS biopsies, confirmed the occurrence of an acute myopathy in
the intensive care unit, associated with the use of cortico-
In the 19th century, patients with life-threatening infections steroids and neuromuscular blocking agents (37, 141, 165,
were reported to lose “a lot of flesh and strength” (531). 314, 382). These patients exhibited preferential myosin loss
During the next decades, occasional case reports indicated and also demonstrated flaccid weakness and failure to wean

1026 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

Table 1. Clinical and histological characteristics of CIP and CIM


Clinical Signs Spectrum of Histological Findings

Critical illness polyneuropathy


Flaccid, symmetrical atrophy, and weakness of the limbs Nerve:*
Distal⬎proximal Normal
Lower limbs⬎upper limbs Mildly reduced myelin fiber density with sporadic axonal
degeneration
Facial muscles mostly spared Marked fiber loss with abundant degenerative changes
Deep tendon reflexes mostly reduced to absent but may be Variable fiber regeneration
preserved
Variable distal sensory loss to pain, temperature, and vibration Muscle:
Weaning failure Denervation atrophy

Critical illness myopathy


Flaccid, symmetrical atrophy and weakness of the limbs and Nerve:
neck flexors Normal
Proximal⬎distal Muscle:
Fiber atrophy with abnormal variation in size of muscle fibers,
angulated fibers, fatty degeneration, fibrosis; mostly
affecting both fiber types, may be limited to type II fibers
Facial muscles mostly spared Focal or diffuse loss of thick myosin filaments
Deep tendon reflexes mostly reduced to absent but may be Scattered to more prominent myonecrosis, vacuolization, and
preserved phagocytosis of muscle fibers
No sensory loss
Weaning failure

*No evidence of primary demyelination or inflammatory infiltrates.

from the respirator. Terms used for this syndrome include Accumulating evidence from studies involving detailed elec-
acute quadriplegic myopathy, acute myopathy of the inten- trophysiological testing, including direct muscle stimula-
sive care, critical care myopathy, acute illness myopathy, tion, and muscle histology revealed that both neuropathy
and acute myopathy with selective loss of myosin filaments and myopathy may co-occur in patients (43, 147, 150, 360,
(69). As many critically ill patients, including those with 361). Interestingly, myopathy rather than neuropathy ap-
sepsis and multiple organ failure, appeared to have primary peared to be the most frequent cause of weakness (43, 125,
muscle involvement not secondary to muscle denervation 297, 370, 383, 403, 413, 577, 583, 614, 764). This led to
(125, 403, 812), the term critical illness myopathy was intro- the concept that CIP and CIM are part of a spectrum of
duced (383, 653, 811). Muscle histology studies (TABLE 1) neuromuscular diseases affecting critically ill patients,
further identified other myopathic changes, in addition to rather than being separate and distinct disorders (43). The
preferential myosin loss, which have been variably classified co-occurrence may imply a common pathogenic factor, af-
as subtypes of critical illness myopathy (69, 70, 87, 332, fecting both the nerves and the muscle in critically ill pa-
402, 535). In some patients muscle necrosis was present tients (112, 164, 361, 403). An alternative explanation may
(297, 331, 360, 383, 384, 812), accompanied by variable be that denervation due to CIP results in increased vulner-
elevation of creatine kinase levels (69, 150, 382, 383, 384). ability of muscles to several noxious stimuli (70, 145, 382).
Finally, in some myopathies, histological changes may be On the other hand, the diseased muscle might also signal
limited to overall atrophy (382, 383), which may be more back to affect innervation and the neuromuscular junction.
pronounced in fast type II (IIa and IIx in humans; IIa, IIx, Clinical examination usually cannot differentiate between
and IIb in rodents) fibers (59, 279) or even be absent in the CIP and CIM as key clinical features are similar and sensory
myopathy with purely functional impairment (402). The evaluation is typically unreliable (382, 383, 407). Conven-
generalized flaccid weakness in CIM may be more proxi- tional electrophysiological screening including nerve con-
mally pronounced (384). Tendon reflexes are generally at- duction studies and needle electromyography also does not
tenuated. Facial and extraocular muscles are generally distinguish between the two entities (403). Both in CIP and
spared but may occasionally be involved (100, 382, 529, CIM, reduced compound motor action potentials (CMAPs)
643). As in CIP, weaning failure is reported as a dominant are found. Abnormal spontaneous electrical activity pre-
symptom (382, 383). CIM itself does not account for senting as fibrillation potentials or positive sharp waves
changes in sensory nerve function. TABLE 1 lists clinical may be due to denervation in CIP but also may occur in
symptoms that may differentiate between CIM and CIP. CIM due to muscle sodium channel dysfunction (580, 581)

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1027


FRIEDRICH ET AL.

or secondary involvement of nerve endings in primary my- ity were found to be associated with CIP (166, 403, 415,
opathy (126, 403, 577, 742). Terms such as critical illness 416). Sepsis, as such, was identified as an independent risk
polyneuromyopathy or the acronym CRIMYNE subse- factor for neuromuscular complications (122, 300) as well
quently emerged (401, 405, 528). To emphasize the clinical as bacteremia (499, 738). The role of the presence and
problem of muscle weakness, regardless of its cause, de- persistence of SIRS and multiple organ failure (44, 147,
scriptive terminology such as ICU acquired paresis, critical 149) as well as of certain inflammatory markers (763), how-
illness neuromuscular abnormalities, acquired neuromus- ever, were confirmed in several studies. As some of these
cular disorders, or ICU-acquired (muscle) weakness was trials specifically focused on septic patients, and sepsis is a
launched (145, 618). Although encompassing both entities major cause of SIRS and organ failure, sepsis was likely to
is useful from a practical point of view, some evidence sug- be a crucial factor (146). Indirect evidence for the central
gests that for prognostic reasons, it may be relevant to dif- role of sepsis is also provided by the independent associa-
ferentiate between CIP, CIM, and other myopathies (381). tion of the use of catecholamines (735) as well as central
Early data indicated similar outcome for CIP and CIM neurological failure (231), which develops earlier during
(383), but recently it was shown that patients with CIM sepsis, with neuromuscular complications. Furthermore, it
may recover earlier and better than those with CIP (273). is confirmed that severity of illness independently predicts
The co-occurrence of CIP with CIM appeared to hamper the risk of neuromyopathy in critically ill individuals (44,
recovery (370) and was responsible for persisting disabili- 100, 149). As will be detailed later, animal models have
ties (344); however, the majority of patients recover within allowed the separation of sepsis and other confounding risk
the first 6 –12 mo (190, 504). factors, e.g., mechanical ventilation and muscle unloading,
which are usually present in combinations in ICU patients.
Although CIM and CIP are mostly described in association For instance, pure sepsis in animal models has not been able
with adult and aged ICU patient populations, they are also to reproduce hallmarks of the CIM phenotype (see below)
a problem in pediatric ICUs (608); however, occurrence of when animals were not mechanically ventilated, mechani-
ICU-related weakness in children has only occasionally cally unloaded, or subjected to a similar ICU-situation to
been reported. From the existing literature it is concluded
mimick the situation in patients. However, in septic ICU
that clinical and electrophysiological conditions are similar
patients, the CIM phenotype will most likely be seen be-
in adults and children (779), but there is still a lack of
cause critically ill septic patients will almost always also be
clinical and animal studies elucidating the significance and
found subjected to mechanical ventilation, muscle unload-
nature of weakness in critically ill children.
ing and other contributing factors.

III. RISK FACTORS AND CLINICAL For further discussion of risk factors for ICUAW, it is nec-
DIAGNOSTIC TOOLS FOR CIP/CIM essary to define the different syndromes causing weakness.
In particular, it is necessary to define what is meant by the
term CIM. Many critically ill patients presenting with CIM
A. Risk Factors for the Development of ICU- show several symptoms and alterations on the organ, cellu-
Acquired Neuromuscular Complications lar, and subcellular level of affected muscle, not all of which
may be detectable in each patient or at a given stage of
Several risk factors have been implicated in the development of investigation: electrical hypoexcitability of muscles; severe
neuromuscular complications in the ICU (FIGURE 1A). Several atrophy; preferential and significant myosin loss; disorga-
studies attempted to identify independent risk factors by per- nization of sarcomeres, ultrastructural abnormalities; and
forming multivariable analysis on data obtained during pro- impaired autophagy and altered protein turnover.
spective observational studies (44, 100, 147, 149, 231, 499,
763). Additional information on risk factors was based on a Whether severe sepsis and sepsis-induced organ failure are
number of randomized controlled trials (RCTs) intervening already on their own sufficient to create the full-blown
with a particular risk factor and reporting on neuromuscular pathological manifestations of CIM (in particular the pref-
complications, although in none of these neuromuscular out- erential myosin loss) remains unclear. There is no study to
come was the primary end point (TABLE 2). date showing preferential or selective loss of myosin sec-
ondary to sepsis in the absence of mechanical ventilation
Based on these data, several key players were suggested. and immobilization. In animal models (see sect. XII) where
First, the presence and persistence of sepsis, systemic in- sepsis and immobilization can be separated, sepsis does not
flammatory response syndrome (SIRS), and multiple organ trigger preferential loss of myosin pointing towards a dif-
failure appear to hold a central role. Sepsis was the main ferential pathophysiological process in pure sepsis.
suspect in the early days of the discovery of CIP, as it was a
common feature in the first case reports (67, 812), and the Immobilizing patients and mechanically ventilating them
incidence of CIP reported in patients with sepsis was very during the acute phase of critical illness is suggested to
high (52, 333). Sepsis, multiple organ failure, and its sever- contribute to muscle atrophy and weakness. By itself, the

1028 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

Steroids
Mechanical
Silencing
Prolonged Malnutrition
Respirator
Ventilation

Sepsis
? ICUAW

Hyperglycemia/
Insulin Resistance
Denervation,
Neuromuscular Blockade

B
ICUAW clinically descriptive
Intensive-Care Unit syndromal term
Acquired Weakness

CIP CIM SIM SDM


Critical Illness Critical Illness Sepsis-induced Steroid-Denervation
Polyneuropathy Myopathy Myopathy Myopathy

• mechanical ventilation • mechanical ventilation • muscle hypoexcitability • muscle immobilization


• muscle immobilization • muscle immobilization • atrophy • muscle hypoexcitability
• muscle excitable (direct • muscle hyopexcitability • no preferential • severe atrophy
muscle stimulation) • severe atrophy myosin proteolysis • preferential myosin
• atrophy • preferential myosin • disorganized sarcomeres proteolysis
proteolysis • mostly no ventilation in • disorganized sarcomeres
• disorganized sarcomeres animal models but in
patients with sepsis/severe
sepsis

Putatively distinct pathophysiologic processes separable in animal models that are


currently postulated to combine to produce weakness in critically ill patients
FIGURE 1. Risk factors contributing to the development of peripheral nervous system dysfunction seen as
muscle weakness in ICU patients and potential differences in disease entities. A: in intensive care unit (ICU)
patients, several conditions and risk factors have been associated with the development of muscle weakness
pointing to a failure in the peripheral nervous system. Clinically, in the past, the presence of “weakness” has
pragmatically been lumped together in the term ICU-acquired weakness (ICUAW), although this term neither
discriminates between primary neuropathy or primary myopathy, nor does it account for potentially different
mechanisms leading to neuropathy/myopathy. Apart from steroids, denervation, and sepsis, mechanical ventila-
tion and immobilization itself is a prominent risk factor that contributes to the development of ICU-related myopa-
thies. Critical illness myopathy (CIM) seen in critically ill patients is almost exclusively associated with altered muscle
excitability, severe atrophy, and a preferential myosin loss, usually seen in critically ill patients who are exposed to
several trigger factors at once. ICUAW, on the other hand, is a clinically pragmatic term that can be applied in the
absence of any further special electrophysiology or biopsy testing. B: the separation of the distinct disease entities
summarized under the term ICUAW as well as unraveling the specific differences of the underlying pathophysio-
logical mechanisms defining those separate disease entities is still a matter of active research, and results from
animal models suggest that some of those trigger factors on their own may be able to produce CIM while others
may not (e.g., pure sepsis without steroid/denervation and/or mechanical ventilation/immobilization). In ICU
patients suffering ICUAW, trigger factors usually combine, and septic ICU patients that are ususally subjected to
mechanical ventilation and muscle unloading will present with CIM rather than SIM.

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1029


1030
Table 2. Randomized trials intervening with presumed risk factors and reporting on neuromuscular outcome using electrophysiological or clinical evaluation
Effect of Intervention on Neuromyopathy Other Independent Risk Factors
Reference
No. Population Diagnosis Intervention Intervention Control P value Risk factors Relative risk (95%CI) P Value

735 SICU ⱖ7 days Electrophysiological IIT 46/181 (25%) 109/224 (49%) ⬍0.0001 Vasopressor support OR: 1.06 (1.02–1.11) 0.009
diagnosis (SEA)
738 Duration of ICU stay OR 1.05 (1.03–1.08) ⬍0.0001
RRT OR 1.9 (1.0–3.8) NA
Bacteremia OR 2.3 (1.3–4.1) NA
658 ARDS ⱖ7days Pragmatic diagnosis* Corticosteroids 26/88 (30%)⫹ 21/91 (22%)⫹ 0.2 NA NA NA
654 Septic shock Clinical diagnosis Corticosteroids 2/234 (1%) 4/232 (2%) NS NA NA NA
⬍72 h
309 MICU ⱖ7 days Electrophysiological IIT 81/208 (39%) 107/212 (51%) 0.02 Age OR 0.98 (0.96–0.99) 0.003
diagnosis (SEA)
Prolonged NMBA OR 2.01 (1.10–3.99) 0.02
CS (protective) OR 0.97 (0.94–0.99) 0.02
536 ARDS ⱕ48 h Clinical diagnosis MRC 48 h of NMBA 40/112 (36%) 28/89 (31%) 0.5 NA NA NA
sums score ⬍48
623 MV ⱕ72 h Clinical diagnosis MRC Early physical and 15/49 (31%) 27/55 (49%) 0.09 NA NA NA
sum ⬍48 occupational
therapy
300 MICU or SICU Clinical diagnosis MRC Late versus early 105/305 (34%) 127/295 (43%) 0.03 Age OR 1.03 (1.01–1.05) 0.001
ⱖ8 days and sum ⬍48 supplementation of
random set insufficient enteral
of nutrition in the
FRIEDRICH ET AL.

short-stayers first week in ICU


APACHE II OR 1.08 (1.04–1.11) ⬍0.001
Sepsis OR 2.20 (1.30–3.71) 0.003
Admission categories 0.03
(as compared with
medical)
Emergency surgical ICU OR 0.77 (0.42–1.43) 0.4
Elective surgical ICU OR 2.61 (0.77–8.88) 0.12
Cardiac surgery OR 1.8 (0.80–4.05) 0.15

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


Time to first MRC OR 1.05 (1.01–1.10) 0.01
measurement
CS OR 2.70 (1.73–4.22) ⬍0.001
NMBA OR 2.72 (1.65–4.51) ⬍0.001
New infection OR 2.75 (1.69–4.45) ⬍0.001

SICU, surgical intensive care unit; MICU, medical intensive care unit; ARDS, acute respiratory distress syndrome; SEA, spontaneous electrical activity; MRC, Medical Research Council;
IIT, intensive insulin therapy; MV, mechanical ventilation; OR, odds ratio; CI, confidence interval; RRT, renal replacement therapy; NMBA, neuromuscular blocking agents; CS,
corticosteroids; NS, not significant; NA, not available. *Presence of the terms “myopathy,” “myositis,” “neuropathy,” “paralysis,” or “unexplained weakness” in the medical record. ⫹The
first 88 patients were retrospectively evaluated.
NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

immobilization (pure bed rest) may not be sufficient to ex- infiltrates (150, 193), inflammatory processes are likely in-
plain the degree of muscle atrophy and weakness observed volved and corticosteroids could hypothetically downregu-
in acutely ill patients (146). “Mechanical silencing” (loss of late potential detrimental pathways. Corticosteroids also
weight bearing and internal strain caused by activation of induce hyperglycemia, another risk factor for neuromyopa-
contractile proteins) and mechanical ventilation in animal thy. Therefore, clinical effects of corticosteroids may be
models are sufficient to produce the preferential and signif- complex and the impact on the nerves and the muscles
icant myosin loss seen in patients (435, 517). Several clinical could be distinct. Effects may also depend on timing and
trials, however, found indicators of the duration of immo- dosing, as well as on controlling the resulting hyperglyce-
bilization to be independently related to the occurrence of mia (309).
neuromuscular complications in critical care (147, 735,
781) and the presence of weakness up to 2 yrs after acute The presumed harmful effects of neuromuscular blocking
respiratory distress syndrome (ARDS) (190). Immobiliza- agents (NMBAs) were extensively reported in many case
tion, therefore, is likely to contribute to CIM also in pa- reports (265). Nondepolarizing agents, often combined
tients. This provided the basis for the development of trials with corticosteroids, were considered to be detrimental
to examine the effects of interventions aimed at reducing the (165). Pharmacological denervation with neuromuscular
duration of immobilization on ICUAW (see sect. XIII). Sed- blocking agents may increase muscle susceptibility to corti-
ative and analgesic use, by association with time to awak- costeroids, as observed in anatomical denervation (466).
ening, can be expected to affect duration of immobilization, Despite this well founded hypothesis, again, data from sev-
but was not found to be independently related with the risk eral trials did not consistently show negative effects (44,
of neuromyopathy in several trials (147, 149, 763). 149, 150, 190, 306, 499, 735, 763). In three trials, NMBAs
were independently associated with neuromuscular compli-
Hyperglycemia was associated with neuromyopathy in ret- cations (231, 300, 309). Type and duration of exposure to
rospective (51, 306) and prospective studies (100, 147) and NMBAs could possibly explain differential findings. Recent
was also identified as an independent risk factor (499, 781) data from an RCT (48 h of NMBAs versus placebo in ARDS
(FIGURE 1A). These findings launched the hypothesis that patients) indeed did not find an increased incidence of
hyperglycemia might impair the microcirculation in the weakness in the intervention group (536). In addition, ear-
nerves, contributing to the development of CIP (64). In lier data may have been confounded by other treatment
addition, during critical illness, organs depending on pas- strategies associated with administration of NMBAs, such
sive uptake of glucose, such as the nervous system, may be as ventilator strategies and sedation regimens no longer
subjected to glucose overload resulting in acute neurotox- used in ICUs (562).
icity because of the hyperglycemia caused by insulin resis-
tance (308). Hyperglycemia may also contribute to nerve Two trials identified age as an independent predictor of
excitability changes or increased generation and deficient neuromuscular complications in the ICU (300, 309). The
scavenging of reactive oxygen species (303, 308). Such ob- role of nutrition has been debated since the first description
servations drove the exploration of restored neuromuscular of CIP. It was hypothesized that malnutrition was a major
function by normalizing hyperglycemia in critically ill pa- cause of ICUAW and that the catabolic process could be
tients (see sect. XIII). revoked by adequate parenteral nutrients (67). Some trials
evaluating the role of parenteral nutrition, however, could
Myopathy is a well-known adverse effect of glucocorticoid not confirm an association (309, 499, 781). In one trial,
treatment (542). Corticosteroids reduce protein synthesis parenteral nutrition was an independent risk factor for CIP
and increase protein breakdown, resulting in atrophy and (231), and in a recent RCT, early parenteral nutrition in-
weakness. Not surprisingly, after the first case report (448) creased the incidence of ICU-acquired weakness (300). Del-
many followed, suggesting corticosteroids as the culprit of eterious effects of refeeding, high amounts of polyunsatu-
myopathy occurring in critically ill subjects, especially in rated fats, or metabolic derangements contributing to dis-
patients treated with a combination of corticosteroids and turbances in the microcirculation were proposed as
neuromuscular blocking agents. Results of prospective tri- mechanisms (752). Autophagy is another pathway that may
als, however, were equivocal. Some found that corticoste- be involved in neuromuscular effects of nutrition in criti-
roids were independently associated with neuromyopathy cally ill individuals. Autophagy is a cellular housekeeping
(100, 147, 311), whereas most other trials could not con- system required for the elimination of damaged organelles
firm this (44, 51, 149, 231, 499, 654, 658, 735, 763). Sev- and large protein aggregates (122). The process is impor-
eral explanations are possible for these divergent findings. tant for maintaining muscle quality (465). Nutrients, espe-
First, in addition to the well-known catabolic effects, corti- cially amino acids, are powerful suppressors of autophagy
costeroids may improve outcome and shorten duration of (155, 247). Insufficient activation of autophagy was found
organ failure in certain ICU populations, which by itself is in muscle biopsies of fed critically ill patients (728), and
an important risk factor for neuromyopathy (21, 22, 473). higher protein delivery in the first week of critical illness
Although CIP and CIM are characterized by inflammatory was even associated with greater muscle wasting (563).

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1031


FRIEDRICH ET AL.

Therefore, muscle quality could be negatively affected by muscles more than distal muscles (147, 305, 413). The
nutrition as shown in a recent RCT (300) (see sect. X). MRC sum score was proposed as a routine and first line
screening tool in the ICU for patients at risk for developing
Several other risk factors were identified in single trials, but ICUAW (268, 402, 622, 661). As reported in various clin-
current evidence for a role of these factors is limited: low ical settings [mechanically ventilated patients with acute
serum albumin (781), hyperosmolality (231), female sex stroke (267), Guillain-Barré syndrome (369), stimulants
(147), aminoglycosides (499), and renal replacement ther- (189), post ICU (40, 325)], inter-rater reproducibility was
apy (738). found to be good in a large study of critically ill patients
(301), and one smaller series (10), but not in two other
small studies (129, 325). Methodological issues such as
B. Aspects of Clinical Evaluation and
judging the adequate awakening and cooperation of the
Diagnostics
patient required to perform the test may be very important.
There are several potential approaches for diagnosing the
Also, the use of a detailed and stringent protocol adapted
problem of intensive care unit acquired weakness as a syn-
for bedridden patients may be crucial (305). MRC sum
drome (ICUAW). In awake and cooperative patients, a bed-
score ⬍48 is associated with important clinical outcomes
side clinical approach can be used. Muscle strength can be
(TABLE 4) and is found to be an independent predictor of
quantified using the Medical Research Council (MRC) sum
score, a validated ordinal scale which grades muscle weak- prolonged weaning, ICU and hospital stay, as well as ICU,
ness in six bilateral muscle groups from upper and lower hospital, and 180 day mortality (TABLE 4). In addition,
limbs between 0 (no contraction) and 5 (normal strength) MRC sum score ⬍48 independently predicted 1 yr mortal-
(TABLE 3). Adding these subscores results in a maximum
ity (307) and pharyngeal dysfunction (487). The clinical
sum score of 60 (369). ICUAW was defined as MRC sum approach is easy and cheap. Drawbacks include the require-
score for muscle strength to be ⬍48, provided no other ment of adequate awakening and cooperation, which limits
cause of weakness could be identified (147). Since then, the number of critically ill patients actually evaluable. Re-
many other publications have used the MRC sum score to ported figures range from 25% (325) up to 90% (51) during
diagnose ICUAW (10, 44, 51, 143, 144, 149, 189, 273, ICU stay. Pain may also interfere with maximal effort. Due
305, 325, 413, 499, 536, 598, 623, 632) without referring to the ordinal character of the scale, subtle changes may not
to the precise underlying disease entity (FIGURE 1B). This be detected. The MRC sum score only points to weakness
clinical approach revealed that weakness affects proximal but does not provide information on the underlying patho-
physiological processes (FIGURE 1B). It is important to rule
out other known causes of weakness, in particular when the
clinical picture does not correspond to flaccid and symmet-
Table 3. MRC sum score rical weakness, facial muscle sparing, and reduced to absent
MRC Sum Score Criteria tendon reflexes. Finally, clinical muscle strength testing
does not allow differentiating between a neuropathic and
Evaluation of adequate awakening myopathic component of the weakness. Other clinical
Open/close your eyes methods of measuring muscle strength in critically ill pa-
Look at me tients have been evaluated. These include hand-grip
Open your mouth and put out your tongue strength measurement using a dynamometer. This proved
Nod your head to be a reliable tool in the ICU with good interobserver
Raise your eyebrows when I have counted up to 5 reliability (29, 301). Hand grip strength showed good test
Muscle groups evaluated performance in predicting MRC sum score ⬍48 and was
Wrist extension independently associated with mortality in a medical ICU
Elbow flexion (10). To avoid problems of maximal effort, use of electrical
Shoulder abduction (181) or magnetic (288) muscle stimulation of the adductor
Dorsiflexion of the foot pollicis has been described. Data on clinical impact of this
Knee extension kind of measurements are currently lacking. Respiratory
Hip flexion muscles are frequently involved in ICUAW. Evaluation of
Appointed scores respiratory muscle strength using maximal inspiratory pres-
0 no visible/palpable contraction sure was also suggested as a surrogate marker for ICUAW
1 visible/palpable contraction without movement of the because of good predictive value for MRC ⬍48 (722) and
limb
its association with duration of weaning (143, 722). Simi-
2 movement of the limb, but not against gravity
larly, bilateral anterior magnetic stimulation of the phrenic
3 movement against gravity
nerves, recording the resulting diaphragmatic contraction
4 movement against gravity and some resistance
using gastric and esophageal balloons, appears feasible and
5 normal force
safe, though complex and expensive and, therefore, not

1032 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


Table 4. Studies evaluating clinical outcomes associated with ICUAW determined using the MRC sum score
Reference Timing of Multivariate/Matched Relative Risk (95% CI) or
No. Population Incidence of ICUAW Measurement Univariate Analysis P Value Analysis Weak Versus Not Weak P Value

Prospective studies
144,147 MV ⱖ7 days 24/95 (25%) D7 after 1 Duration of MV ⬍0.001 Prolonged weaning HR 2.4 (1.4–4.2) 0.001
awakening
143,632 MV ⱖ7 days 75/115 (65%) At awakening Low MRCb delays successful 0.001 Low MRCb delays HR 1.96 (1.27–3.02) 0.002
extubation successful
extubation
1ICU mortality 0.006 1ICU mortality OR 7.99 (0.99–64.29) 0.05
1Hospital mortality 0.02 1Hospital mortality OR 2.02 (1.03–8.03) 0.048
499 ICU stay 50/474 (11%) At awakening 1ICU mortality ⬍0.05 NA NA NA
ⱖ24 h
ICU stay 44/185 (24%) 1ICU mortality ⬍0.05
⬎10 days
10 MV ⱖ5 days 35/136 (26%) At awakening 1Duration of MV ⬍0.001 2Hospital free days 41% (12–60%) 0.007
1ICU stay ⬍0.001 2ICU free days 54% (67–36%) 0.001
2ICU-free to day 30 ⬍0.001 1Hospital mortality OR 7.8 (2.4–25.3) 0.001
1Hospital stay ⬍0.001
2Hospital-free to day 60 ⬍0.001
1ICU readmission 0.01
1Discharged to other 0.01
location than home
1Hospital mortality ⬍0.001
82 SIRS and 13/39 (33%)a ICU 1Duration of MV 0.001 1Death before 180 NA 0.009
MV ⱖ48 discharge days
h
1ICU stay 0.005
2Functional outcome 0.01
(Barthel index) at D28
1Death before 180 days 0.03
1Resources needed 0.008
(cumulative TISS-28

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

scores)
722 ICU stay ⱖ7 17/33 (51%) At awakening Prolonged weaning ⬍0.05 NA NA NA
days
1ICU stay 0.001
2MIP ⬍0.001
410c SICU 44/95 (46%) At awakening 1ICU stay 0.002 1ICU stay IRR 0.98 (0.97–0.98) ⬍0.001
1Hospital stay 0.001 1Hospital stay IRR 0.97 (0.97–0.98) ⬍0.001
1Mortality 0.006 1Hospital mortality OR 0.95 (0.90–0.99) 0.04
1Duration of MV 0.01
304 ICU stay 227/415 (55%) At awakening 1Time to alive weaning ⬍0.001 1Time to alive 11d (7–22) versus 8d 0.009
ⱖ8days weaning (5–14)

Continued

1033
1034
Table 4. —Continued
Reference Timing of Multivariate/Matched Relative Risk (95% CI) or
No. Population Incidence of ICUAW Measurement Univariate Analysis P Value Analysis Weak Versus Not Weak P Value

1Time to alive ICU ⬍0.001 1Time to alive ICU 8d (2–14) versus 3d (0–8) 0.008
discharge discharge
1ICU mortality 0.02 1Time to alive 36d (16–83) versus 23d 0.007
hospital discharge (13–41)
1Time to alive hospital ⬍0.001 2 6 min WD 66m (0–207) versus 191m 0.01
discharge (90–270)
1Hospital mortality ⬍0.001 1Total billed 23,277€ (15,370–36,147) 0.04
hopsitalization versus 17,834€
costs (12,227–31,306)
26 min WD 0.002 11 yr mortality 30.6% versus 17.2% 0.014
1Total billed hopsitalization ⬍0.001
costs
11 yr mortality ⬍0.001
487 Long-term 20/30 (67%) Within 24 h Pharangeal dysfunction Pharangeal
ventilated of FEES dysfunction
(ⱖ10
days),
FRIEDRICH ET AL.

FEES
clinically
indicated
PAS ⬎1 0.038 PAS ⬎1 9 (1.3–61.14) 0.038
VPSR ⬎1 0.014 VPSR ⬎1 5.4 (1–28.5) 0.014
Symptomatic aspiration 0.003 Symptomatic 9.8 (1.6–60) 0.009
(retrospective analysis) aspiration
Retrospective studies

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


51 ARDS 27/45 (60%) At awakening 1ICU stay 0.001 NA NA NA
1Duration of MV (at 28d) 0.02

MV, mechanical ventilation; ICUAW, intensive care unit acquired weakness; MRC, medical research council; ICU, intensive care unit; NA, not available; CI, confidence interval; HR,
hazard ratio; OR, odds ratio; IRR, incidence rate ratio. All studies used MRC ⬍48 to diagnose ICUAW and to examine relationship with outcome unless explicitly stated: aankle
dorsiflexion not included, ICUAW defined as sum score ⬍35/50; bMRC sum score ⱕ41; cMRC sum score used as a continuous variable in the predictive models. Additional univariate
analysis with MRC cut-off of 48 showed significant association with days on MV, ICU, and hospital stay.
NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

routinely applicable in the ICU. Reduced diaphragmatic indication of prognosis. Different mechanisms that contrib-
muscle strength had been shown (299, 387, 760). A clear ute to weakness are likely to have a different prognosis for
relationship between respiratory muscle weakness using recovery. For example, it appears that loss of electrical ex-
this technique and worse outcome was recently demon- citability in muscle and nerve may be fully reversible and
strated (153, 677). Another way to get around the problem thus have a good prognosis for recovery (515, 583). Other
of consciousness and full cooperation is the use of electro- changes such as axon degeneration almost certainly have a
physiological testing, e.g., in combination with direct mus- poor prognosis. The prognosis of severe myosin loss and
cle stimulation (see below). severe muscle atrophy are less clear.

Mechanisms underlying weakness may vary with time.


C. Clinical Diagnostic Tools and Potential Within a few days of onset of critical illness, nerve conduc-
Contributing Mechanisms tions reveal reduced sensory and motor amplitudes (361,
698, 699). One likely contributor to early reduction in
Determining mechanisms contributing to weakness in the nerve conduction amplitudes is ion channelopathy (515).
acute setting is complicated by a number of factors. One As available evidence suggests reduction/loss of electrical
problem is that critically ill patients often cannot cooperate excitability is fully reversible (515, 583), it is unlikely to be
with motor and sensory examination due to sedation or a mechanism underlying chronic weakness. This suggests
encephalopathy. Because patients cannot cooperate, they that there may be an evolution of mechanisms contributing
are unable to voluntarily recruit motor units during EMG to weakness such that mechanisms underlying acute weak-
assessment. This prevents use of motor unit morphology ness (initially and within a few days) differ from those con-
and recruitment to distinguish between neuropathy and tributing to weakness during the ongoing course of critical
myopathy. Many studies have used the presence of sponta- illness (e.g., within weeks of critical illness). In particular,
neous activity on EMG as an indicator of denervation and there may also be phases of various pathophysiological
thus the presence of neuropathy. However, spontaneous mechanisms contributing to the development of subsequent
activity is also present in CIM, such that this has led to mechanisms, e.g., membrane dysfunction and depolariza-
over-diagnosis of neuropathy as the mechanism underlying tion triggering Ca2⫹ imbalance, metabolic dysregulation
weakness. To further complicate interpretation of motor which may lead to atrophy and preferential myosin loss
unit recruitment during EMG, a recent study in rats has (FIGURE 2). Determining whether mechanisms contributing
identified reduced motor neuron excitability as a possible to weakness evolve over time will require longitudinal use
contributor to reduced motor unit recruitment (500). Thus, of both electrophysiology and pathology studies and clearly
while classic nerve conduction/EMG can be used to serially represents a challenge for future clinical and animal studies.
follow development and resolution of neuropathy and my-
opathy, it should not be used in isolation to determine
mechanisms underlying weakness. IV. INTRODUCTION TO
PATHOPHYSIOLOGICAL MECHANISMS
To distinguish between neuropathy and myopathy, more
detailed electrophysiological tests, like direct muscle stimu- Multiple mechanisms contribute to development of weak-
lation to measure muscle fiber conduction velocity (12) and ness during critical illness. There seem to be numerous,
velocity recovery cycles of muscle action potentials (801), either independent or interacting, mechanisms that have
are needed. In nerve, specialized tests such as strength-du- been identified that contribute to muscular weakness trig-
ration time constant, threshold electrotonus, current- gered by critical illness (FIGURE 2) of which some are 1)
threshold relationship, and recovery cycles are necessary to reduced excitability of muscle and nerve, 2) altered Ca2⫹
identify reduced excitability (800). If nerve and muscle ex- homeostasis, 3) myosin loss following mechanical silencing,
citability are normal, but sensory and motor amplitudes are 4) atrophy of muscle, 5) death of axons, 6) death of muscle
reduced, it is likely that axon degeneration is present and fibers, 7) disturbed anabolism-to-catabolism ratio, 8) bio-
the patient has critical illness neuropathy. If small motor energetic failure, and 9) failure of neuromuscular transmis-
units are present that are recruited in a myopathic fashion, sion, to name only some that are explained in more detail in
it is probably safe to conclude myopathy is present. Muscle the subsequent sections. These will predominantly also fo-
biopsy can be used to study atrophy (myofibrillar protein cus on the myopathy aspect of critical illness. It is not
loss equally affecting actin and myosin without change in known whether myopathic mechanisms develop simultane-
acto-myosin ratios in pure atrophy) or preferential myosin ously or independently, and there may be distinct phases
loss (with significantly decreased myosin-to-actin ratios as underlying the clinical phenotype of “myopathy” that are
in CIM patients and animal models of CIM). Ultrasound inherent to different pathophysiological mechanisms (FIG-
might be used to follow muscle atrophy serially. Nerve bi- URE 2). To develop effective therapy, it may be necessary to
opsy is needed to study axon loss. Serum CK can give some prevent multiple contributors to weakness. If all the mech-
estimate of muscle fiber death. By focusing on mechanisms anisms underlying weakness were triggered by the same
that underlie ICUAW, it may be possible to get a better signal, development of therapy would be straightforward,

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1035


FRIEDRICH ET AL.

early phases late phases

motorprotein actomyosin
atrophy hyperproteolysis
muscle membrane dysfunction of myosin (‘ghost sarcomeres‘)
ICU stay Ca2+
critical illness hypoexcitability imbalance +
‘silent muscle‘ deficient autophagy, myofiber
bioenergetic vacuolization
failure etc.

1-2 5 >5-10 days


Immobilization, muscle unloading, patient mobilization,
mechanical ventilation respirator weaning

FIGURE 2. Specific pathophysiological mechanisms in ICUAW may either evolve in time independently or
trigger subsequent pathways to present with phases of myopathy during critical illness. Early phases may begin
as early as from 24 h of ICU treatment and are characterized by a dysfunction of membrane excitability
(hypoexcitability), followed by subcellular putative alterations in Ca2⫹ homeostasis, bioenergetics, and motor
protein function, whereas later phases after several days to 1 wk are marked by hyperproteolysis of myofibrillar
proteins (atrophy) and/or preferential myosin loss. Severe loss of sarcomeric proteins may also be responsible
for the structural fixation of the disease seen as altered cytoarchitecture and loss of contractile filaments that are
only slowly regenerated over months. Note that each mechanism is not limited to a particular phase, and
mechanisms may coexist at late stages (i.e., membrane hypoexcitability, myosin loss, impaired autophagy, etc.).

but if mechanisms underlying weakness were triggered in- plasma cytokine levels might correlate with illness severity
dependently by different signals, multiple interventions or mortality rate in septic children. They found that plasma
would be necessary. TNF-␣, IL-1, and IL-6 levels were elevated in patients. Cy-
tokine levels correlated with mortality rate; all three cyto-
kines were higher in nonsurvivors than in patients who
V. INFLAMMATORY CYTOKINES IN
survived. Damas et al. (140) measured TNF-␣, IL-1␤, and
CRITICAL ILLNESS
IL-6 levels in 40 critically ill surgical patients where IL-6
was a robust marker of illness, correlating with both
A. Cytokine Elevation in Critical Illness APACHE II score and mortality.

A half-century of research implicates proinflammatory Subsequent refinements in immunoassay technology have


cytokines as potential mediators of critical illness. This enabled broader documentation of cytokine profiles in the
concept originally emerged in animal studies designed to clinical setting. Sepsis severity, the evolution of organ fail-
characterize individual cytokines. For example, circulat- ure, and patient death were associated with distinct multi-
ing interferon (IFN) levels were markedly increased in cytokine profiles. Hranjec et al. (326) evaluated blood levels
mice following endotoxin administration or bacterial in- of IL-1, -2, -4, -5, -6, -8, -10, and -12, IFN-␥, TNF-␣ and
fection (663) or in guinea pigs subjected to burn injury granulocyte-macrophage colony-stimulating factor (GM-
(709). In discovering tumor necrosis factor-␣ (TNF-␣), CSF) within 48 h of admission to the ICU in a large patient
Carswell et al. (106) found that plasma TNF-␣ levels number. Patients were subclassified based on evidence of
were increased by administering endotoxin to mice, a infection or trauma. On admission to ICU, specific cytokine
finding later extended to rats and rabbits (521). Although profiles were associated with three patient subgroups: pa-
these early reports were limited in scope and narrowly tients who lacked infection or trauma (IL-6, -8, -10), in-
focused, they laid the foundation for an explosion of fected patients who died (IL-2, -8, -10, GM-CSF), and
basic science research in the field. It became clear that trauma patients who died (IL-4, -6, -8, TNF-␣). Elevation of
IFN, TNF-␣, members of the interleukin (IL) family, and individual cytokines predicted death in trauma patients
other cytokine mediators are highly sensitive to experi- (IL-4) and patients with neither trauma nor infection
mental conditions that are related to critical illness. A (IL-8) but not in patients with infection. It is interesting
partial listing with representative references includes sep- to note that in this study, ⬃10% of patients in all groups
sis (241, 328), mechanical trauma (246), surgery (670, were not mechanically ventilated. However, mechanical
802), pneumonia (71, 527), drug-induced organ failure ventilation was not separated as an independent variable
(660), thermal injury (484, 743), and peritonitis (36). in the analysis of cytokine profiles. A more targeted study
measured circulating cytokines during the first week after
Studies of critically ill patients provide more-specific infor- admission in 30 septic patients (480). They found that
mation on cytokine regulation in the clinical setting. An early elevation of IL-8 and monocyte chemoattractant
early study by Sullivan et al. (669) tested the hypothesis that protein (MCP-1) predicted mortality, whereas specific

1036 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

multi-cytokine profiles were associated with either organ proach has generated substantial evidence that cytokines
failure or shock. Proinflammatory cytokines are sug- can compromise skeletal muscle performance. The fol-
gested to promote muscle atrophy and weakness in crit- lowing sections highlight information on three cytokines
ically ill patients (780). Not all cytokines are implicated. that are best understood in this context.
Rather, a subset of cytokines has been linked to protein
loss or muscle dysfunction in critically ill patients. As B. TNF-␣, Muscle Atrophy, and Contractile
outlined in TABLE 5, these clinical associations were Dysfunction
strengthened by mechanistic studies using animal mod-
els, isolated tissue preparations, and cell culture systems. TNF-␣ is the proinflammatory cytokine that has been stud-
Over the past four decades, this vertically integrated ap- ied most extensively for effects in skeletal muscle. The pro-

Table 5. Inflammatory mediators associated with critical illness and effects in muscle
Inflammatory Mediator Molecular Mass, kDa Effects in Skeletal Muscle

TNF-␣ (cachectin) 26 (mature secreted form: Also expressed in type II muscle fibers (550)
17)
Depolarization of plasma membrane (712)
Na⫹-current inhibition, left shift of Na⫹ channel activation and inactivation
(274)
Activation of PKC-mediated Na⫹ channel phosphorylation (148)
Induces muscle proteolysis and atrophy (148)
Alters resting Ca2⫹ levels (decrease in myotubes) (741)
No change in tetanic Ca2⫹ amplitudes in muscle fibers (741)
Decreases Ca2⫹ transient amplitudes by 50% (in myotubes) (741)
Depresses tetanic force production in limb and diaphragm muscle (571)
Activates ubiquitin-proteasome pathway (227,428)
Increase major histocompatibility complex II surface expression (in
combination with IFN-␥) in human muscle cells (359)
Activation of NF-␬B (573)
Downregulation of MyoD inhibits myogenic differentiation (280)
Induces insulin resistance in muscle (248)
IL-1 13–17 (precursors: 33) Produced by activated macrophages; also expressed in muscle fibers in health
(619), following exercise (101), and in inflammatory myopathies (26)
IL-1␣: primarily Associates with RyR1, blocks SR Ca2⫹-release, and decreases SR Ca2⫹ leak
membrane-bound in muscle fibers (220)
IL-1␤: secreted form Upregulates iNOS expression (4)
Induces skeletal muscle proteolysis [IL-1␣ (785), but maybe not IL-1␤ (228)]
Stimulates IL-6 production in skeletal muscle cells (IL-1␤) (445)
Stimulates prostaglandin E2 (PGE2) production and proteolysis (250)
Ubiquitin gene expression 1 (437) or % (228)
Inhibition of protein synthesis (132)
IL-6 23–30 (glycosylation diversity) Produced by activated macrophages and T-cells, tumor cells, but also
produced by muscle fibers (type I ⬎ type II) (550)
No effect on ubiquitin expression (437)
Promotes infiltration of myocytes with prostaglandins (780)
Both pro- and anti-inflammatory actions in critical illness (24)
Induces skeletal muscle protein breakdown (in rats) (259)
Reduces insulin-stimulated glucose uptake in muscle (364)
IL-10 18 (unglycosylated in humans) Anti-inflammatory cytokine (780)
Prevents skeletal muscle from IL-6-induced defects in insulin action (364)
IFN-␥ 20–25 Produced by activated T-cells and natural killer cells but also in muscle fibers
following muscle injury (118)
Required for muscle regeneration (118)
Inhibits protein translation in muscle by stimulating NO synthase activity (223)
Modulator of TNF-␣ signaling in muscle (myotubes: downregulation of TNF-R2
and increased NF-␬B activity) (710)
Ubiquitin gene expression 1 (437)

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1037


FRIEDRICH ET AL.

tein is synthesized as a 26-kDa parent molecule that is states (418). These changes are associated with TNF-stim-
cleaved upon cellular release to form a 17-kDa polypeptide ulated increases in functional activity of the ubiquitin con-
that promotes anti-tumor and immune responses. This mol- jugating pathway. This is the major pathway for regulated
ecule was originally named “cachectin” to acknowledge its degradation of muscle protein via the 26S proteasome. In
association with catabolic states of cachexia. Circulating muscle cells, the rise in ubiquitin conjugating activity is
TNF-␣ concentrations are commonly elevated in critically abolished by selective inhibition of p38MAPK (418),
ill patients (see above) and in animal models of critical NF-␬B (421), or E2-20k (419), demonstrating that each of
illness, e.g., lipopolysaccharide administration (636), peri- these elements is essential for integrity of the response.
tonitis (507), trauma (539), heart failure (20), and burn Moreover, selective inhibition of NF-␬B prevents loss of
injury (799). Increases in TNF-␣ promote muscle weakness muscle protein stimulated by TNF (421), identifying NF-␬B
via two general mechanisms: 1) promotion of atrophy and as a critical regulator of catabolism in response to this cy-
2) induction of contractile dysfunction. tokine (see also sect. XI).

1. TNF-␣ promotes muscle atrophy 2. TNF-␣ and contractile dysfunction

This was first demonstrated in animal studies where sys- TNF-␣ also depresses the force of muscle contraction in the
temic administration of exogenous TNF-␣ decreased mus- absence of atrophy. This was first observed by Wilcox et al.
cle mass and muscle fiber cross-section (83, 226). Initially, (773) exposing fiber bundles isolated from hamster dia-
TNF-␣ was thought to be an indirect actor, e.g., via an- phragm to exogenous TNF-␣ in vitro. Isometric force nor-
orexia (485) or glucocorticoid elevation (472), since direct malized to fiber bundle cross-section (“specific force”) was
exposure to TNF-␣ for up to 3 h did not stimulate protein significantly depressed by TNF-␣. This was later confirmed
loss in excised muscle preparations (229, 251, 591). How- in fiber bundles from guinea pig diaphragm (14), mouse
ever, Li and colleagues (420, 421) later showed a direct diaphragm (423), and mouse flexor digitorum brevis (571).
catabolic action of TNF-␣ on differentiated muscle cells. Contractile dysfunction is not simply an artifact of TNF-␣
Incubation of cultured myotubes with a clinically relevant exposure in vitro. Li et al. (423) observed a similar decre-
TNF-␣ concentration caused progressive reductions in total ment in specific force of diaphragm fiber bundles isolated
protein content, muscle-specific protein levels, and myo- from transgenic mice engineered for cardiac-specific over-
tube diameter over several days. These changes mimic the expression of TNF-␣. In this case, the muscle was exposed
response of intact skeletal muscle to critical illness, support- to TNF-␣ in vivo (serum concentration 250 –350 pg/ml)
ing a potential role for TNF-␣. At the cellular level, TNF-␣ prior to excision; exogenous TNF-␣ was not used. Yet,
stimulates protein loss via receptor-mediated signaling specific force was approximately half the value of wild-type
events that alter muscle gene expression. Skeletal muscle muscle for twitch and maximum tetanus. The muscle was
constitutively expresses TNF-␣ receptor subtype 1 stimulated directly, eliminating neuromuscular activation
(TNFR1) and subtype 2 (TNFR2). Llovera and associates as a potential cause of weakness. There was no evidence of
(438, 439) have shown that TNF-␣ acts via TNFR1 (and muscle atrophy or damage. Body weight, the weights of
not TNFR2) to stimulate catabolism. In muscle, TNF-␣ representative trunk and limb muscles, and the ultrastruc-
exposure rapidly activates the transcription factor nuclear ture of limb and diaphragm muscle fibers were indistin-
factor-␬B (NF-␬B) which is essential for loss of muscle pro- guishable between transgenic and control animals. Thus
tein (421). The canonical pathway for NF-␬B activation is chronic exposure to low levels of circulating TNF-␣ can
mediated by the I-␬B kinase (IKK) complex (546) and is profoundly weaken muscle fibers without inducing detect-
redox sensitive during TNF-␣ stimulation. NF-␬B activa- able atrophy.
tion is positively modulated by a transient rise in muscle-
derived reactive oxygen species (ROS), which function as Several studies have addressed the cellular mechanism of
second messengers for TNF-␣ (421), and is negatively mod- TNF-induced dysfunction. Hardin et al. (286) used mice
ulated by the glutathione cycle (626). TNF-␣ also rapidly deficient in TNFR1 and TNFR2 to assess the receptor sub-
activates the mitogen-activated protein kinases (MAPKs) type by which TNF-␣ depresses specific force. One hour
including p38MAPK, extracellular signal-regulated kinases following intraperitoneal TNF-␣ by injection, diaphragm
1 and 2 (ERK1/2), and c-Jun NH2-terminal kinase (JNK) in fiber bundles showed marked depression of specific force.
differentiated myotubes (418). TNFR1 deficiency abolished this response, but TNFR2 de-
ficiency did not. Thus, like atrophy, contractile dysfunction
In muscle cells, these signaling events stimulate the expres- is mediated via TNFR1. Studies of post-receptor signaling
sion of genes that regulate the ubiquitin-proteasome path- have largely focused on muscle-derived oxidants as second
way. TNF-␣ signaling via NF-␬B increases mRNA levels for messengers. Within minutes, TNF-␣ exposure increases cy-
E2-20k, a ubiquitin carrier protein and murine homolog of tosolic oxidant activity (286, 423, 656), which can depress
human UbcH2 (419). In parallel, TNF/p38MAPK signaling specific force (570). Consistent with this model, preincubat-
increases the expression of atrogin1/MAFbx, a ubiquitin ing muscle fiber bundles with N-acetylcysteine (NAC), a
ligase that mediates muscle atrophy in a variety of catabolic reduced thiol donor, abolished the effects of exogenous

1038 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

TNF-␣ (423). NAC pretreatment prevented the rise in cy- C. IL-1 and Skeletal Muscle
tosolic oxidant activity and preserved specific force at con-
trol levels. Similarly, pretreating mice with Trolox, a water- IL-1 is commonly present at elevated concentrations in
soluble vitamin E analog, abolished the effects of TNF-␣ the serum of critically ill patients, a biomarker that often
injection on cytosolic oxidant activity and specific force correlates with clinical outcome. The cytokine exists as
(286). These studies reveal that muscle-derived oxidants two isoforms. IL-1␣ is primarily membrane-bound,
mediate TNF-␣ effects on contractile function. whereas IL-1␤ is the secretory form that mediates para-
crine and endocrine effects. IL-1 is synthesized both by
But which are the exact oxidants involved? Skeletal muscle activated macrophages and by nonimmune cells. These
fibers contain multiple intracellular sources of ROS includ- include skeletal muscle fibers which express IL-1␤ at low
ing mitochondria, NADPH oxidase, cyclooxygenase, and levels under basal conditions (619) and at higher levels
after exercise (101) and in inflammatory myopathies
xanthine oxidase. Also, muscle fibers constitutively express
(26). In critical illness, in particular in the context of
two enzymatic sources of nitric oxide (NO): the neuronal-
sepsis, the release pattern of IL-1 (alongside with other
type isoform of NO synthase (nNOS) and the endothelial-
proinflammatory cytokines) shows a distinct time course
type isoform (eNOS). Both ROS and NO derivatives are
that is summarized in FIGURE 3A.
continuously generated by skeletal muscle. Both redox cas-
cades participate in a variety of signal transduction path- IL-1 is a potential stimulus for the protein loss and muscle
ways. And in muscle, both cascades appear to be essential atrophy seen in critical illness. Elevated IL-1 levels clearly
for TNF-stimulated dysfunction. Alloati et al. (14) origi- promote muscle atrophy in experimental animals. For ex-
nally reported that TNF-␣ stimulates NO production by ample, Cooney et al. (132) showed that IL-1␣ infusion for 6
muscle and that pharmacological blockade of constitutive days decreases muscle weight and protein content of rat
NOS activity prevents the fall in specific force caused by gastrocnemius. The pathophysiological relevance is evident
TNF-␣. These findings were confirmed by Stasko et al. in animal models of sepsis where administration of an IL-1
(656) who identified the nNOS isoform as the molecular receptor antagonist can preserve muscle mass (131). Studies
source of NO signaling stimulated by TNF-␣. In this same of underlying mechanisms suggest that IL-1 influences both
study, the investigators also examined ROS involvement. protein degradation and protein synthesis. Baracos et al.
They found no evidence that TNF-␣ stimulates muscular (34) first reported that IL-1, then referred to as leukocytic
ROS production. However, selective depletion of ROS (su- pyrogen, acts directly on isolated skeletal muscle prepara-
peroxide anions, hydrogen peroxide) in the cytosolic milieu tions to increase intralysosomal proteolysis and net protein
abolished TNF-␣ effects on specific force without disrupt- degradation. This action was augmented at elevated muscle
ing TNF-␣/NO signaling. The authors concluded that temperatures, a model of fever, and appeared to be medi-
TNF-␣ stimulates NO production by nNOS but that endog- ated by a rise in prostaglandin E2 synthesis. IL-1 did not
enous ROS are obligate comediators of the signal transduc- alter protein synthesis in this preparation. The catabolic
tion pathway that depresses specific force. The mechanism actions of IL-1␣ were demonstrated in vivo by Zamir et al.
of NO/ROS interaction was not tested, but it was specu- (795) who found that total and myofibrillar protein break-
lated that peroxynitrite (a reaction product of NO and down rates were elevated in isolated limb muscles from rats
ROS) might be a downstream mediator required for con- injected with recombinant IL-1␣. Consistent with the asser-
tractile dysfunction (656). Two lines of evidence identify tion that IL-1 promotes proteolysis, intravenous adminis-
tration of this cytokine is reported to increase ubiquitin
myofibrillar proteins as the target of TNF-␣/NO/ROS sig-
expression in rodent muscle (437), although ubiquitin up-
naling. Reid et al. (571) tested TNF-␣ effects on contractile
regulation is not always observed (228). IL-1 effects on
regulation in intact single murine limb muscle fibers and
synthesis pathways also promote net loss of muscle protein.
found that TNF-␣ depressed specific force of tetanic con-
Cooney et al. (132) showed that IL-1␣ infusion depressed
tractions without altering the magnitude or duration of in-
protein synthesis rates in rat gastrocnemius, a muscle com-
tracellular calcium transients. They concluded that TNF-␣ posed primarily of fast fibers, whereas soleus muscle (pri-
signaling acted on an intracellular target downstream of the marily slow fibers) and the heart were unaffected. This find-
calcium transient, i.e., the myofibrillar lattice. This interpre- ing is consistent with earlier observations that amino acid
tation was confirmed by Hardin et al. (286) in permeabil- uptake was depressed in limb muscles of rats treated with
ized muscle fibers isolated from mice injected with either IL-1␣ (794).
TNF-␣ or saline. In “skinned” fibers from TNF-treated an-
imals, specific force was depressed at higher calcium con- Less is known about IL-1 effects on skeletal muscle contrac-
centrations, providing clear evidence of myofibrillar dys- tion. However, a recent report on IL-1␣ effects on calcium
function. Interestingly, TNF-␣ effects on the calcium-force release from the sarcoplasmic reticulum (SR) raises intrigu-
relationship closely resemble the effects of direct peroxyni- ing possibilities (220). With the use of permeabilized single
trite exposure (684), consistent with the postulate that per- fibers, spontaneous colocalization of exogenously applied
oxynitrite is a downstream mediator of TNF-␣ signaling. IL-1␣ with the RyR1 was found (220). IL-1␣ exerted revers-

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1039


FRIEDRICH ET AL.

A
Pro-inflammatory Anti-inflammatory

Sepsis: + Liver macrophages Serum


IL-6 Exercise:

Cytokine expression
TNF-α IL-6
IL-1α

Cytokine levels
IL-10
IL-1β
IL-1 TNF-α
sTNF-R TNF-α
IL-1RA IL-1

0 5 10 15 20 0 5 10 15 20
Time (hrs) Time (hrs)
Time
B
Non-muscle tissue Systemic Local (muscle)
(e.g., tissue macrophages, liver, Liver
adipocytes, glia cells, etc.) CRP, TNF-α,
IL-10, IFN-γ
Muscle
wasting ?
IL-1
IL-1 TNF-α IL-1
IL-6
TNF-α
IL-6

Macrophages IL-1RA
Monocytes –
Neutrophils
IL-10 +
IL-1 Muscle
Circulation TNF-α

Local Systemic Local


FIGURE 3. Pro- and anti-inflammatory cytokines and their role in sepsis/critical illness and the systemic
inflammation-muscle axis. A: time course of pro- and anti-inflammatory cytokines in the systemic circulation in
critical illness as best studied in the scenario of sepsis. During sepsis, pro-inflammatory IL-1 and TNF-␣ rise
early, followed by an increase in IL-1, which is then followed by a rise in anti-inflammatory mediators to initiate
the compensatory anti-inflammatory response syndrome (CARS). IL-6 both exerts pro- and anti-inflammatory
effects. For example, exercise lacks the pro-inflammatory response and is purely characterized by anti-
inflammatory response triggered by almost sole IL-6 release from exercising muscle (“myokine”; note: IL-6
amplitude in both sepsis and exercise scaled to maximum; absolute IL-6 amplitudes are much larger in
exercise). Individual time courses may vary depending on the model (e.g., CLP shown in the right panel) and
outcome. [Right panel according to data from Chensue et al. (119). Left panel modified from Petersen and
Pedersen (545).] B: systemic inflammation-muscle axis during sepsis and critical illness. Activated macro-
phages in inflamed tissue as well as monocytes and neutrophils in the blood secrete large amounts of
pro-inflammatory IL-1 and TNF-␣ that drive the pro-inflammatory systemic response. These mediators also
stimulate myokine production in skeletal muscle, of which IL-6 is the most predominant to initiate the systemic
CARS by inhibiting further IL-1/TNF-␣ release and to stimulate production of IL-10 and IL-1RA, for example.

ible effects on SR calcium release and specific force that myocardial contraction (321). Clearly, the effect of IL-1 on
were strongly dependent on Mg2⫹ concentration. Long- skeletal muscle contraction is a novel topic of potential
term IL-1␣ exposure of intact muscle fibers caused loss of importance in critical illness.
sarcolemmal integrity leading to intracellular deposition of
IL-1␣. These data suggest a novel mechanism of action by 1. The paradox of IL-6
which IL-1 might depress muscle contraction in critically ill
patients (see also sect. IX). This hypothesis has not been IL-6 is a pleiotropic cytokine with both pro- and anti-in-
tested but is consistent with several related observations: 1) flammatory properties. It is secreted by various cell types
specific force of skeletal muscle is depressed by secretory including T cells, macrophages, smooth muscle cells, osteo-
products of lipopolysaccharide-stimulated monocytes blasts, and skeletal muscle fibers. Among muscle-derived
(774), which are known to include IL-1; 2) similar to skel- cytokines, IL-6 has been studied extensively. IL-6 affects
etal muscle, IL-1 alters SR calcium release by cardiac myo- skeletal muscle myogenesis, lipid metabolism, glucose up-
cytes (170); and 3) in the heart, IL-1 depresses the force of take, protein synthesis, and protein degradation.

1040 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

IL-6 is commonly elevated in the serum of critically ill pa- IL-1 may contribute to proinflammatory feedback while,
tients (FIGURE 3A), suggesting a positive association with once muscle secretes large amounts of IL-6 into the circula-
the loss of muscle that occurs in these individuals. However, tion, this would shift the response towards an anti-inflam-
the experimental data paint a more nuanced picture: on the matory feedback. This hypothesis still awaits experimental
one hand, direct IL-6 exposure has not been shown to stim- clarification, but at least one recent study shows that in
ulate muscle proteolysis in vitro. Incubation of rat limb mechanically ventilated ICU patients who developed myop-
muscles with recombinant IL-6 in vitro does not alter the athy, the inflammation-induced acute phase response re-
rate of protein breakdown (230, 259), nor have cell culture sulted in a marked increase in IL-6 production in skeletal
studies detected an effect of exogenous IL-6 on the rate of muscle, and also in immobilized muscle (392). Although
proteolysis in rodent muscle-derived myotubes (179). In this increase in inflammation-induced IL-6 myokine pro-
contrast, IL-6 appears to promote protein degradation in duction is substantially smaller than in response to strenu-
vivo. High concentrations of circulating IL-6 stimulate ous exercise (⬃15- vs. 100-fold, respectively; Refs. 192,
muscle proteolysis in rats treated with the recombinant cy- 392), the results point towards a common denominator in a
tokine (259) and in transgenic mice engineered for stable “muscle-immune system” axis where skeletal muscle feeds
overexpression of IL-6 (718). Conversely, inhibition of cir- back to the proinflammatory response syndrome by tuning
culating IL-6 has been shown to lessen muscle atrophy in an the onset of the CARS reaction (FIGURE 3B). However, since
animal model of cancer cachexia (665) and in IL-6 trans- IL-6 also exerts potential detrimental effects on muscle such
genic mice (719). These divergent outcomes simply may be as muscle wasting, this may even become the more promi-
artifacts of experimental design, e.g., IL-6 concentration or nent effect in critical illness beyond a putative “tipping
the duration of exposure. However, Munoz-Canoves et al. point” of IL-6 production such as suggested in FIGURE 3B.
(497) have proposed a more interesting interpretation.
They suggest that IL-6 promotes muscle atrophy in the in- Apart from IL-6, IL-1 has recently also been shown to be
tact animal via indirect effects on insulin-like growth factor secreted by skeletal muscle in response to inflammasome
I (IGF-I) signaling. Normally, IGF-I promotes muscle upregulation (568), and this may be in part responsible for
growth. The authors reasoned that IL-6 reduces serum lev- detrimental effects of force production in critical illness
(220). However, the role of this “myokine” in regulating
els of IGF-I binding-protein-3 (497) and thereby promotes
systemic immune response on top of the already increased
degradation of serum IGF-I, lessening IGF-I effects on mus-
circulating systemic IL-1 is not known.
cle growth. To our knowledge, only one published report
has tested the direct effect of IL-6 on contractile function.
FIGURE 3B summarizes the local (muscle) and systemic in-
Janssen et al. (351) incubated diaphragm muscle fiber bun-
teraction of cytokines.
dles with recombinant IL-6, then measured specific force
and endurance capacity. IL-6 did not alter these parameters.
VI. BIOMECHANICS,
Apart from a systemic action of IL-6 on muscle, muscle MECHANOSENSATION, AND
itself has been recognized as an endocrine organ being able TENSEGRITY IN CRITICAL ILLNESS
to actively secrete several cytokines upon stimulation, e.g.,
in response to exercise or inflammation. These “myokines,” In this section, the mechanical activation of skeletal muscle
among which IL-6 seems to be most responsive to physical and its pathways that are significantly affected by the ICU
exercise (545) or electrical stimulation (89), are produced in condition are discussed in more detail. Mechanical external
an ATP- and Ca2⫹-dependent manner. IL-6 has sometimes stimuli may not only influence immediate signaling events,
been termed “a double-edged sword” exerting both protec- e.g., SR Ca2⫹ release, via mechanosensitive channels (697)
tive and deleterious effects on skeletal muscle (497). On the but also influence the shape and growth of all cells via direct
one hand, IL-6 production is stimulated by TNF-␣ and effects on protein synthesis and degradation. However, our
IL-1␤ and may contribute to the proinflammatory cascade knowledge of the mechanisms on how mechanical signals,
in critical illness and sepsis (706); on the other hand, stren- i.e., mechanosensation or “tensegrity” (339 –343), are
uous exercise induces a likewise marked increase in IL-6 transduced to influence transcriptional regulation and in-
plasma levels (up to 100-fold; Ref. 192) without the preced- tracellular biochemistry is far from complete. Tensegrity or
ing increase in proinflammatory TNF-␣ and IL-1 as seen in “tensional integrity,” a term derived from architecture, de-
sepsis (FIGURE 3A). The fact that the IL-6 peak is followed scribes the interaction between isolated components within
by rising levels of cytokine inhibitors, i.e., IL-1RA and sol- a system under mechanical compression or tension and re-
uble TNF-␣ receptors (sTNF-R) has cornered its role for sulting structure. It has been introduced to muscle biology
anti-inflammatory effect of exercise (532) but may also be to summarize cellular functional and structural responses to
important in initiating the compensatory anti-inflammatory mechanical stimuli (574). For example, mechanical signals
response syndrome (CARS) in sepsis and eventually, during may induce increased protein synthesis, satellite cell activa-
critical illness (533). It is attractive to speculate that sys- tion, and release of growth factors and may overlap with met-
temic IL-6 produced in nonmuscle tissues by TNF-␣ and abolic, action potential-induced, and oxidative signaling

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1041


FRIEDRICH ET AL.

(576). Different major signaling cascades such as PI-3K, different mechanosensors targeting specific intracellular
MAPKs, Ca2⫹-calmodulin-calcineurin-NFAT, glycogen signaling pathways with consequences for myofibrillar pro-
synthase kinase (GSK), and AMP activated kinase (AMPK) tein synthesis and degradation. It is accordingly not surpris-
are downstream effectors of mechanotransduction (86) ing that the effects of hindlimb unloading, denervation, and
(FIGURE 4). Mechanosensing in skeletal muscle involves immobilization will have different effects on myofibrillar
multiple components and spans from integrins, focal protein expression despite the fact that all are characterized
adhesion complexes (FAC), stretch-activated calcium- and by a decline in muscle mass and force. For instance, strict
sodium-permeable membrane ion channels, caveolin-3 and bed rest for 6 mo results in mild muscle fiber atrophy due to
the caveolin-3-associated nNOS, the sarcodystroglycan a general loss of contractile proteins that is larger than the
complex, intermediate filaments, and a number of mecha- loss in muscle fiber size, resulting in decreased single muscle
nosensitive sarcomeric proteins, as well as mechanosignal- fiber specific force (398). During muscle unloading by
ing via the IGF-I growth factor (76, 86, 346, 390, 391, 641, hindlimb suspension or microgravity, the loss of muscle
652, 686, 687, 754) signaling (FIGURE 4). In addition, ex- mass and function has been reported to be associated with a
ternal forces on surface membrane receptors, such as integ- preferential actin loss (203, 204, 585) and slow-oxidative
rins and cadherins, might act at distance and promote co- (type I) to fast-glycolytic (type II) fiber types switching
ordinated changes in nuclear structures via a hard-wired (493). In limb suspension, microgravity, and bed rest mod-
network including structural actin and specialized nuclear els, weight bearing has been removed, but there is still in-
anchoring structures (349, 754). This direct and fast mecha- ternal strain related to activation of contractile proteins. In
notransduction from the cell surface to the nucleus induces immobilization models with joint fixation by, e.g., plaster,
an ion flux through the nuclear membrane and the intranu- weight bearing is typically intact and activation of contrac-
clear acto-myosin complex may contribute to nuclear pre- tile proteins has not been eliminated, although contractions
stress and nuclear mechanosensation affecting gene regula- are mainly isometric. In denervation atrophy, muscles are
tion via various mechanisms (754) (FIGURE 4). still passively loaded due to weight bearing or activation of
agonists/antagonists. This also applies to spinal cord injury
The communication between transcription factors and where marked fiber atrophy and slow-to-fast fiber transi-
transcriptional modifiers in the sarcomere has recently been tion occurs with a larger extent in humans compared with
reviewed (76). Multiple mechanosensors are embedded in rodents (266). In deeply sedated mechanically ventilated
the sarcomere. In the Z-disk, several mechanosensitive ele- ICU patients, with or without neuromuscular blockade,
ments have been identified with direct transcriptional links both external (weight bearing or passive movement induced
to the nucleus, such as MYOZ2 (negative regulator of cal- by activation of agonists or antagonists) and internal me-
cineurin), MLP, and CLOCK, shuttling to the nucleus in chanical load (activation of contractile proteins) have been
response to mechanical strain, and CBF␤ (involved in removed. This complete “mechanical silencing” in ICU pa-
JUNB-mediated gene expression) (FIGURE 4). In the I-band, tients is, to our knowledge, unique for ICU patients with
different stretch-sensitive muscle ankyrin repeat proteins significant consequences for gene and protein expression
(MARPs) form complexes with titin and myopallidum/cal- and different from the other unloading conditions listed
pain-3 and have been suggested to sense passive stress on above.
the sarcomere (76). At the M-band, the titin kinase interacts
with a complex of atrogenes which during mechanical ar- The preferential loss of the molecular motor protein myosin
rest dissociates and translocates to the cytoplasm and nu- and myosin-associated proteins and maintained expression
cleus (NBR1, SQSTM1, and MuRFs) (391, 559). These of thin filament proteins are a hallmark of CIM in ICU
atrogenes are believed to act as transcriptional repressors patients (396, 397, 399, 400, 519, 662). Neuromuscular
amplifying the atrophy program affecting both protein syn- blockade, systemic administration of glucocorticosteroids,
thesis and different degradation pathways (see Ref. 76) (FIG- and sepsis have been forwarded as important factors trig-
URE 4). gering this type of acquired myopathy, but preferential my-
osin loss in ICU patients has been reported in the absence of
Different experimental and clinical models have been used any of these factors (399). Instead, all ICU patients who
to study the effects of immobilization on skeletal muscle develop CIM have been exposed to long-term mechanical
structure and function, such as hindlimb unloading, micro- ventilation and mechanical silencing (396, 519). Further-
gravity, bed rest, joint immobilization in plaster (or other more, both experimental and clinical studies have shown
types of fixation), denervation, etc. All these models are that long-term mechanical ventilation and “mechanical si-
associated with altered mechanosensing, but effects on pro- lencing” with or without neuromuscular blockade and
tein synthesis and degradation are dependent on type of without both sepsis and systemic steroid administration in-
mechanical load. The mechanical load induced by weight duce the preferential myosin loss observed in patients with
bearing, passive stretch, or shear stress in the sarcomere CIM (396, 435, 517, 519). Thus mechanosensation plays
caused by activation of contractile proteins and the level an integral role in the complex development of CIM in
and type of activation of contractile proteins will involve long-term immobilized and mechanically ventilated ICU

1042 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

SACT LIF IGF-I rp-vl Integrins

FAK DSG
ILK
Ca2+ Caveolae
Na+
Ca2+
PI3K
nNOS

AKT F-actin

Downstream effectors of
mechanotransduction:
p70S6K • PI-3K
mTOR • MAPKs
4EBP Nucleus
• Calmodulin
• Calcineurin
Telethonin • Glycogen synthase kinase
MLP • AMP activated kinase
CLOCK • AKT/mTOR
CBFβ MARPs Titin kinase:
MYOZ2 • SQSTMI
α-actinin • NBRI Protein synthesis/
abscurin • MuRF degradation
• Pb2
Titin Myosin Actin

Z-line M-band Z-line

FIGURE 4. Mechanosignaling in skeletal muscle. Mechanosignaling represents an emerging and dynamic


field in biomedical science, but the complexity of the different pathways involved, and how they are interrelated,
is not fully resolved. Different pathways involved in mechanosensing and tensegrity in skeletal muscle are briefly
summarized, and those reported to be altered in CIM are highlighted in red. However, mechanosignaling has
only recently been forwarded as a factor triggering CIM, and the mechanosensing pathways are most probably
far more complex than those indicated in red. Multiple signaling pathways influence protein synthesis and
degradation in the muscle fiber spanning from the muscle membrane and extracellular matrix to the M-band
in the center of the sarcomere. Insulin-like growth factor I (IGF-I) has been suggested to play an important role
for the muscle hypertrophy induced by mechanical overload, but a maintained hypertrophy response has been
reported in transgenic mice with an IGF-I receptor lacking the ability to bind IGF-I, suggesting multiple other
mechanosensitive pathways (651). Calcium- and sodium-permeable stretch-activated channels (SAC) respond
to mechanical stimuli and various intracellular signaling cascades. In addition, membrane invaginations, i.e.,
caveolae, respond to cell stress and stretch-induced signaling, and many different proteins involved in cell
signaling bind to caveolins, such as neural nitric oxide synthase (nNOS), G protein subunits, tyrosine kinases,
small GTPases, and growth receptors (240, 641). The cytokine leukemia inhibitory factor (LIF) plays an
important role in muscle hypertrophy in response to mechanical loading (651). Integrins spanning from the
extacellular matrix to the interior of the muscle cell, linked to cytoskeletal actin, directly connect to the nuclei
and mitochondria, thus allowing a “hard-wired” and rapid signal propagation to nuclear and mitochondrial DNA
(754). The subsarcolemmal dystrophin sarcoglycan complex (DSG) is involved in mechanosensing, and a
deficiency in this complex is a feature of muscular dystrophies with defects in the signaling related to
mechanical load, resulting in muscle degeneration (793). A number of sarcomeric proteins are involved in
mechanosensing, and there is emerging evidence of a very dynamic exchange of multiple sarcomeric proteins
to the cytoplasmic pool affecting muscle gene expression in response to mechanical load from the Z-line to the
center of the sarcomere in the M-band (76, 234, 390). Multiple major signaling cascades are downstream
effectors of mechanosensing, such as PI-3K, MAPKs, calmodulin, calcineurin, glycogen synthase kinase, AMP
activated kinase, and AKT/mTOR (86).

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1043


FRIEDRICH ET AL.

patients that distinguishes it from other myopathies under may explain why passive loading does not prevent all the
the ICUAW umbrella (FIGURE 1B). changes triggered by muscle inactivity. In particular, muscle
excitation significantly contributes to transcription in a pro-
In pioneering work by Griffiths et al. (269), it was demon- cess called excitation-transcription coupling (276, 724).
strated that passive mechanical loading 9 h/day for 7 days
had a significant positive effect on skeletal muscle in me- The significant positive effects of passive loading of skeletal
chanically ventilated ICU patients treated with nondepolar- muscle in experimental and clinical CIM intervention stud-
izing neuromuscular blocking agents. In an experimental ies support the early physical therapy and mobilization of
study using a novel rat ICU model where animals were deeply sedated or pharmacologically paralyzed mechani-
exposed to neuromuscular blockade and mechanical venti- cally ventilated ICU patients that has been shown to shorten
lation for durations varying from ⬍1 day to 14 days, it was ventilator- and ICU-days, to reduce health care costs as well
confirmed that unilateral loading 12 h/day had a positive as to improve prognosis and quality of life (88, 146, 495,
effect on skeletal muscle structure and function (574). That 505, 623).
is, both the loss of muscle fiber size and specific force at the
single muscle fiber level were significantly attenuated in re- VII. NEUROMUSCULAR TRANSMISSION IN
sponse to passive loading, resulting in a doubling of the CRITICAL ILLNESS
functional capacity on the loaded versus the unloaded side
(574). The mechanisms underlying the loading effects Nondepolarizing neuromuscular blocking agents are used
showed a complex temporal pattern involving oxidative to facilitate mechanical ventilation (536). In the 1980s, it
stress, posttranslational protein modifications, transcrip- was first reported that following prolonged use of neuro-
tional regulation of myosin synthesis, protein degradation muscular blocking agents, some patients developed persis-
tent weakness (529). Subsequent studies identified the
via the E3 ligase MuRF1, and extracellular matrix/cell ad-
mechanism underlying weakness as prolonged neuromus-
hesion proteins (574). Similar effects were observed in both
cular blockade (37, 386, 538, 596, 624, 625). Development
fast- and slow-twitch limb muscles, but the effects of load-
of prolonged weakness was due to slow clearance of block-
ing were typically more pronounced in slow- versus fast-
ing agents or their metabolites and was often associated
twitch muscles. In a follow-up clinical study, unilateral an-
with renal and/or hepatic failure (37, 386, 624, 625). Be-
kle joint flexion-extensions 10 h per day for 9 days in se-
cause of concerns regarding prolonged weakness, neuro-
dated, but not pharmacologically paralyzed, neurointensive
muscular blocking agents are now less frequently used and
care patients showed a significant positive effect on single
for as short a period as possible such that prolonged neuro-
muscle fiber force generation capacity (specific force; Ref.
muscular blockade is primarily of historical interest.
435). Furthermore, nNOS which is associated with the
mechanosensitive caveolin-3 in the muscle membrane was At the time of many of the initial reports of prolonged
translocated to the cytoplasm in response to the clinical ICU neuromuscular blockade, the role of myopathy as a contrib-
condition, i.e., in a similar fashion as reported in response utor to development of weakness in critical illness was not
to hindlimb suspension (687), and was suggested to play a yet well recognized. Thus studies generally did not perform
role in the posttranslational deamidation modifications of muscle biopsies or EMG analysis of motor unit recruitment
the myosin motor domain on both the loaded and unloaded to look for co-occurrence of myopathy. It was only later
side (435). A significant preferential myosin loss, the hall- proposed that use of neuromuscular blocking agents is a
mark of CIM, was observed in response to long-term im- risk factor for the development of ICUAW (141, 255, 256).
mobilization and mechanical ventilation in the ICU patients CIM was likely a contributor to weakness in some patients
in the absence of neuromuscular blockade, suggesting that reported to have weakness due to prolonged neuromuscular
the mechanical silencing may play a significantly more im- blockade (37, 255), but it was not clear whether neuromus-
portant etiological role in triggering CIM than neuromus- cular blockade itself would produce the full-blown picture
cular blockers (32, 435). In spite of the significant positive of CIM. With increased awareness of CIM as a cause of
effect of mechanical loading on skeletal muscle structure weakness in critically ill patients following the additional
and function, it did not abolish the negative effects of the use of neuromuscular blocking agents, and more cautious
ICU condition, suggesting that this mild type of mechanical use of neuromuscular blocking agents, most patients with
loading did not impact on all mechanosensitive signaling pure motor deficits have been found to have CIM rather
pathways or that other yet unidentified factors, in addition than prolonged neuromuscular block as the etiology of
to mechanosensation, may play an important etiological weakness, in particular since those patients were always
role. The latter statement is supported by the fact that most also mechanically ventilated and completely immobilized.
CIM patients recover from the muscle paralysis while still Some speculations for neuromuscular blocking agents to
mechanically ventilated and immobilized in the ICU. One trigger myopathy in ICU patients, and in particular CIM,
key difference between passive loading and normal activa- point towards their potential to induce functional denerva-
tion of muscle contraction is that during passive loading, tion and subsequent denervation atrophy during prolonged
muscle action potentials are not occurring. This difference usage (64, 625).

1044 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

A. Microcirculation-Induced Denervation particular, single-fiber electromyography (SFEMG) in several


patients with CIP, both before they developed spontaneous
Disturbances of the microcirculation are seen in sepsis and muscle electrical activity and during follow-up (621). In par-
critical illness. Transmigration of immune cells out of blood ticular, the latter technique allows assessing a measure for
vessels and cellular infiltration of the tissue may provide the neuromuscular transmission through quantification of the jit-
grounds for the specific aberrant effects of inflammatory ter between “just suprathreshold excitation” and action po-
mediators to both muscle and nerve (149, 193). Addition- tential recording, since a jitter ⬍10 ␮s usually reflects a direct
ally, tissue edema, seen in sepsis and critical illness, may stimulation of fibers (621, 655). Interestingly, patients who
also represent a nonspecific compression damage factor to developed spontaneous activity in muscles during the study
the muscle-nerve compartment, especially since the distal course also had significantly increased jitter time, indicating a
motor axons are located within the muscle fascia and can be delay in neuromuscular transmission.
prone to compression. In a histochemical and ultrastruc-
tural study of skeletal muscle biopsies from several patients
with sepsis and multiorgan failure (all being mechanically B. Neuromuscular Transmission in Sepsis
ventilated), five of which also presented with muscle weak- and Systemic Inflammation
ness, ultrastructure showed fiber atrophies and altered cap-
illaries with edema, infiltrates, and segmental necrosis As sepsis (501, 502, 508, 711) and nonseptic critical illness,
(158). A similar observation was noted in a porcine septic i.e., thermal injury and burns (180, 454, 462, 463), have
shock model where after 18 and 48 h of septic shock in- been reported to attenuate the action of nondepolarizing
duced by endotoxin injection (no mechanical ventilation or blocking agents, such as d-tubocurarine or rocuronium, a
immobilization of animals), endothelial cell damage with series of studies were performed to determine the underly-
endomysial edema and mitochondrial swelling in muscle ing mechanisms of this rather unexpected facilitation of
fibers were detected (293). Endomysial edema increased neuromuscular transmission in animal models of experi-
during the time course of septic shock, and plasma TNF-␣ mental sepsis, with sepsis as the only confounding condition
levels were significantly increased. However, whether the inducing muscle weakness. Direct exposure of frog sarto-
muscle damage seen was therefore primarily due to edema- rius muscle to Escherichia coli endotoxin (10 ␮g/ml) re-
induced damage or more directly due to either endotoxin or sulted in elevations of miniature-endplate potential (MEPP)
TNF-␣, or both, could not be assessed. Other studies frequencies without changing MEPP amplitudes. These
seemed to exclude a role for edema to induce muscle weak- changes were abrogated in Ca2⫹-free solutions and antag-
ness in septic patients. In a clinical study assessing muscle onized by elevating extracellular K⫹, indicating endotoxin-
force and edema in 18 septic ICU patients, tissue edema was induced alterations of the presynaptic terminal membrane
not associated with skeletal muscle weakness (245). More ion conductances, in particular for Ca2⫹ (544). In an at-
support to question the direct involvement of tissue edema tempt to better assign neuromuscular transmission changes
in the pathophysiological mechanism of ICUAW came from and neuromuscular blocking action to the stage of sepsis,
a rat CLP study where EDL muscle microcirculation was Narimatsu et al. (502) investigated phrenic nerve-induced
observed with intravital microscopy 6 – 48 h after sepsis twitch-tension responses in hemidiaphragm preparations
induction (549). In septic animals, the number of stopped- from rats with early (9 h post CLP) and late (18 h post CLP)
flow capillaries was significantly increased but not related acute panperitonitis sepsis. Those represent the bimodal
to an increase in tissue wet-to-dry weight ratio, indicating time course of the “hyperdynamic and hypermetabolic”
that extrinsic compression of capillaries by tissue edema and “hypodynamic and hypometabolic” phases of CLP-
was not a primary cause of interstitial hypoperfusion to induced sepsis in this model (117, 755). Indirect (nerve) and
induce damage to nerve and muscle. In a recent experimen- direct (muscle) twitch stimulation produced similar decre-
tal study on septic nonventilated rats, animals showed ments in isometric twitch-tension but much more pro-
marked signs of axonal neuropathy already from day 2 after nounced in late sepsis (⬃65% decrement) compared with
sepsis induction despite a normal morphology of nerve sec- early sepsis (⬃35% decrement). The dose-response curves
tions arguing against a major nerve compression by edema of twitch tension for rocuronium, pancuronium, and d-tu-
(515). Nevertheless, even without compression as the pri- bocurarine were consistently shifted towards higher con-
mary cause of tissue hypoperfusion, the latter may contrib- centrations by sepsis with an additionally marked increase
ute to the chronic membrane depolarization of terminal from early to late sepsis throughout (502). Since these re-
motor axons as concluded from a clinical study on patients sults were obtained under a low stimulation-frequency re-
with “electrophysiologically proven CIP” who showed re- gime (0.1 Hz), the NMBA-induced twitch depression pre-
duced nerve superexcitability and increased accommoda- dominantly reflects competitive blocking of postjunctional
tion to depolarizing and hyperpolarizing currents (800). acetylcholine receptors, and it was speculated that hyposen-
That the terminal motor axonopathy seen in critical illness- sitivity to neuromuscular blockade during sepsis may orig-
induced weakness also extends further to the neuromuscu- inate from upregulation of postjunctional nicotinic AChR
lar transmission was inferred from a clinical study perform- rather than a decrease in drug-receptor affinity (502), sim-
ing nerve conduction studies, electromyography and, in ilar as seen in a rat burn injury model (363). In a follow-up

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1045


FRIEDRICH ET AL.

study, Niiya et al. (508) focused on elucidating the exact tion is expected to increase expression of AChR of im-
mechanisms underlying changes in neuromuscular trans- mature isoforms (788). Immobilization, as a contributing
mission during “acute late” sepsis (⬃18 h post CLP). This correlate to ICUAW (214), either resulted in increased
was because results from chronic sepsis either in nonlethal (214, 461) or unaltered receptor expression (337) in the
panperitonitis CLP models or intraperitoneal endotoxin- immobilized limb. Finally, the condition “critical illness”
induced sepsis in animals were inconclusive, showing ei- also produced differential outcomes: upregulated (362,
ther hyper- or hyposensitivity to d-tubocurarine, or a 758) or unaltered (455) AChRs in thermal injury animal
decrease or no change at all in AChR number (313, 595, models, as well as in sepsis animal studies, depending on
720). Acute late sepsis (⬃18 h post CLP) induced a sig- whether a panperitonitis CLP model [concluded down-
nificant increase in endplate potential (EPP; postjunc- regulation at the neuromuscular endplate region (508);
tional potential induced by quantal release of acetylcho- no change in AChR (595)] or chronic intravenous E. coli
line from the motor nerve bouton in response to electric infection model (no upregulation of AChRs, Ref. 213)
stimulation) amplitudes and EPP quantal content, indi- was employed. The difference in the CLP studies may be
cating a facilitation of EPPs and excitability of muscle attributed to the stage of sepsis investigated, but this
(508). The authors also measured acetylcholine poten- requires further confirmation studies. It should be noted
tials (AChP; postjunctional potential induced by ionto- that none of those animal models involved to induce
phoretic acetylcholine application), electrotonic poten- myopathy in sepsis (“sepsis-induced myopathy”) con-
tials (EP; sequential muscle membrane potential changes ducted any kind of mechanical ventilation and/or immo-
to direct current applications), spontaneous miniature bilization and did not assess preferential myosin loss, and
endplate potentials (MEPP; spontaneous and quantal re- although being sometimes termed so in the literature,
lease of acetylcholine from the motor nerve bouton), and may not be considered as CIM per se (FIGURE 1B). Inter-
resting membrane potentials (RMP). With the use of this estingly, to date, there does not seem to be a single study
series of techniques, prejunctional effects of sepsis could that dissected the effects of sepsis and immobilization in
be dissected from postjunctional ones. Prejunctional ef- critical illness patients (which strictly means including
fects included an increase in quantal content of EPPs mechanical ventilation). This could experimentally be
overcome in future animal studies combining a pure sep-
through increased release probability (reflected by an in-
sis animal model with a continuous passive mechanical
creased MEPP frequency), probably mediated by a sepsis-
loading protocol, similar as reported in two clinical stud-
induced increase in intracellular Ca2⫹ concentrations to
ies where passive motion in mechanically ventilated ICU
facilitate transmitter release (508). Postjunctionally, ace-
patients without sepsis (435) was able to preserve specific
tylcholine-potential analysis revealed a significant de-
force (435) and prevent fiber atrophy (269).
crease in acetylcholine sensitivity during acute late sepsis
pointing towards a reduced function/density of AChR
Taken together, even though presynaptic facilitation of
and/or increased acetylcholinesterase activity (508).
transmission is a common finding in critical illness, the
Since previous studies reported an inhibition of the latter
overall loss-of-function aspect of muscle performance
in conjunction with inflammation (46, 142), thus expect-
seems to be represented by postsynaptic alterations from
ing an increase in acetylcholine sensitivity, it was con- the neuromuscular junction (this section) to downstream
cluded that decreased postjunctional nAChR density mechanisms (following sections). This ultimately outweighs
(720) explained the observed reduced acetylcholine sen- a facilitated presynaptic component of muscle activation in
sitivity by overcoming inhibited acetylcholinesterase ac- ICU-related myopathies. FIGURE 5 summarizes the mecha-
tivity in acute late sepsis (508). On the other side, also nisms discussed in this chapter.
acetylcholine sensitivity in extrajunctional areas might
need to be considered because sepsis was shown to induce
spread of AChRs along the sarcolemma (461), a condi- VIII. MEMBRANE AND ION CHANNEL
tion which would be expected to increase acetylcholine FUNCTION IN CRITICAL ILLNESS
sensitivity rather than decrease it.

The issue of altered AChR expression numbers as well as A. Sodium Channels Determine Membrane
shifts in expression profiles towards embryonic isoforms Excitability in Skeletal Muscle
(464) in myopathy in critical illness is still puzzling with
studies showing upregulation (214, 460, 639), down- Voltage-gated sodium channels in the adult skeletal mus-
regulation (508), or no change at all (213, 337), depend- cle (Nav1.4) determine the membrane excitability of the
ing on the experimental model of critical illness (464). muscle fiber sarcolemma (599) and are important to ini-
This is possibly due to the relative contribution of the tiate the upstroke phase of the action potential upon
isolated effects of systemic inflammation and sepsis, im- crossing a certain membrane potential threshold for
mobilization and denervation to the resulting overall channel opening during membrane depolarization. This
AChR expression in each model. For instance, denerva- potential threshold is mainly set by the steady-state inac-

1046 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

Extrajunctional
spread

ACh in
AChR
secretory
vesicles
ACh Motor nerve
terminal
AChR

CIM

Inflammation/
Denervation Immobilization
sepsis
Embryonic
Em
mbryoni
bryonic
y
ACh release AChR
AC
AChR
ChR
Acute
late
Embryonic AChR

? N le
Nucleus
e
AChR density

Acute Chronic
late
Extrajunctional spread

?
ACh sensitivity
Acute
late
FIGURE 5. Alterations at the level of neuromuscular transmission contributing to ICU-acquired weak-
ness. ICUAW is usually a mixed syndrome of either neuropathy- or myopathy-induced weakness during
critical illness either due to sepsis (or even sepsis-unrelated conditions, e.g., burns), immobilization, and
eventual denervation contributing to alterations to neuromuscular function. Typical effects on either
presynaptic (neuropathy) or postsynaptic (myopathy) functions are shown and, where available from
literature, dissected into the single confounding conditions where pure immobilization or denervation
studies were performed. See text for details. For pure denervation and immobilization, data were also
extracted from References 375, 668, and 782.

tivation of the channels (253, 270). Both changes in threshold. This is primarily achieved by K⫹ efflux
steady-state activation and inactivation modulate excit- through voltage-gated K⫹ channels. The resting mem-
ability and action potential properties in skeletal muscle brane potential in particular is set by the electrochemical
(219) and are thus of special interest in many diseases diffusion equilibrium of K⫹, and any changes towards
associated with channelopathies (e.g., Refs. 104, 380). the relative resting permeabilities of other ions (e.g.,
Nav1.4 channels are unevenly distributed over the muscle Na⫹) over K⫹ can markedly shift the resting membrane
fiber membrane with highest densities near the neuro- potential and impair action potential generation and
muscular junction and dropping towards the ends (15). spread. Furthermore, skeletal muscle has a severalfold
They are also located in high densities in the t-system to higher Cl⫺ permeability within the tubular membranes
warrant fast and even spread of action potentials from over the sarcolemma (168). This larger tubular conduc-
the surface membrane to the triad junctions to trigger tance is required to maintain excitability during bouts of
excitation-contraction coupling. Detailed information muscle activation where tubular K⫹ accumulation would
on normal Na⫹ channel function and membrane excita- otherwise depolarize the tubular potential to either facil-
tion is reviewed elsewhere (110, 111). Na⫹ channels, itate uncontrolled spontaneous myogenic contractions or
once opened and having undergone transition into their lock Na⫹ channels in an inactivated state, depending on
inactivated state, have to be reactivated by repolarizing the level of depolarization (173). From this, it is clear that
the membrane potential well below their activation any plasma ionic imbalance caused by critical illness

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1047


FRIEDRICH ET AL.

would have greater effects in the t-system over the sarco- lated during development (790). In experiments done in
lemma due to larger concentration gradients present in vitro, it has been found that the Nav1.5 isoform activates
this diffusion-restricted space. and inactivates at potentials 15–20 mV more hyperpolar-
ized than Nav1.4 (753, 804). Thus upregulation of Nav1.5
might cause a hyperpolarized shift in inactivation of the
B. “Sodium Channelopathy” in Muscle in total sodium current. There is marked upregulation of
Critical Illness Nav1.5 mRNA in steroid-denervated muscle beyond that
seen in either denervated or steroid-treated muscle alone
The first indication that there might be an ion channelopa- (578). However, when ␮-conotoxins (GIIIB) were used to
thy in critically ill patients came from studies showing that selectively block Nav1.4 sodium channels in steroid-dener-
muscle was electrically unexcitable in the acute phase of vated muscle, little sodium current remained, and the cur-
severe weakness (577, 583). Subsequently, there have been rent that remained was inactivated with similar voltage de-
a number of studies demonstrating reduced muscle excit- pendence as the total current (196). These data, as well as
ability that manifests as reduction of muscle fiber conduc- more recent protein measurements, indicate that Nav1.4 is
tion velocity, increased relative refractory period, and re- the primary sodium channel in both control and steroid-
duced excitability of fibers in response to direct muscle stim- denervated muscle (376). The conotoxin data further sug-
ulation (12, 59, 413, 613, 614, 764, 801). Reduction in gest that the voltage dependence of both Nav1.4 and Nav1.5
muscle fiber conduction velocity likely underlies prolonga- was shifted in the hyperpolarized direction in this rat model
tion of compound muscle action potential duration that is of steroid-denervation myopathy (SDM; FIGURE 1B) that
seen in patients with CIM (257).
seems to reproduce key symptoms found in CIM muscle
(196). Thus, despite in vitro data suggesting that Nav1.5
Studies of mechanisms underlying loss of muscle excitabil-
inactivates at more hyperpolarized potentials than Nav1.4,
ity in CIM have been performed in a rat model of the dis-
this may not be the case in vivo. These data suggested that
order. To simulate CIM in rats, denervation of muscle was
the primary cause of hypoexcitability of steroid-denervated
combined with corticosteroid treatment (see sect. XII). In
fibers was a hyperpolarized shift in the voltage dependence
steroid-treated, denervated rat muscle, it was found that a
of inactivation of Nav1.4 (196) (FIGURE 6).
combination of depolarization of the resting potential and a
hyperpolarized shift in the voltage dependence of sodium
A recent study of covalent modifications of Nav1.4 found
channel inactivation resulted in unexcitability of the major-
reduced glycosylation (376). However, as previous work
ity of muscle fibers (196, 580 –582). The voltage depen-
found that removal of sialic acid from sodium channels
dence of sodium channel inactivation was also found to be
shifted in the hyperpolarized direction by pure sepsis in the shifts inactivation gating in a depolarizing direction (47, 48,
rat (594). These data suggest that increased inactivation of 492), the reduction in glycosylation is likely to be compen-
sodium channels is a major contributor to reduced excit- satory rather than causative for the hyperpolarized shift in
ability. As the above studies were performed on muscle inactivation. One finding that may account for the hyper-
removed from rats and perfused in Ringer solution, the polarized shift in inactivation is increased association of
hyperpolarized shift cannot be accounted for by the pres- sodium channels with proteins of the dystrophin-associated
ence of a circulating factor acting on the sodium channel. protein complex (376). In particular, there is increased as-
However, there is a study suggesting that circulating factors sociation with nNOS. In mice lacking nNOS, denervation-
may contribute to modulation of sodium channel gating. induced loss of excitability was lessened (376). One way
When murine muscle fibers were exposed to serum fractions NO might trigger hyperpolarized inactivation is through
from septic patients for up to 80 min (1:10 dilutions in phosphorylation of Nav1.4; however, global levels of so-
standardized external solutions), there was a depolarized dium channel phosphorylation were unaltered following
shift in the voltage dependence of sodium channel inactiva- denervation and steroid treatment (376). The finding that
tion (218) (FIGURE 6). Thus, if a circulating factor contrib- nNOS is involved in regulation of excitability following
utes to altered membrane excitability, its acute effect is denervation adds to earlier findings that implicate NO in
opposite to the effect triggered by long-term exposure. the response of skeletal muscle to denervation. Previously, it
has been shown that inhibition of nNOS lessens muscle
One way in which the voltage dependence of sodium chan- atrophy triggered by denervation (687). Since dramatic
nel inactivation might be shifted in the hyperpolarized di- muscle atrophy occurs in CIM, this raised the possibility
rection is via expression of different sodium channel iso- that nNOS signaling plays a role in triggering both electrical
forms which inactivate at more hyperpolarized potentials. hypoexcitability and atrophy of fibers.
In innervated mature skeletal muscle, only the Nav1.4 so-
dium channel isoform is expressed. After denervation, ste- In an in vitro TNF-␣ challenge of isolated muscle fibers,
roid treatment, or sepsis, there is upregulation of the Nav1.5 TNF-␣ induced a reversible dose- and time-dependent inhi-
isoform (578, 594, 789). Normally, the Nav1.5 isoform is bition of Na⫹ currents of up to ⬃75% of control currents
expressed in embryonic skeletal muscle but is downregu- (275). Steady-state activation and inactivation curves were

1048 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

Steroid-denervation animal model


Nav+-channels:
Control RP Control inactivation
Control Excitable Inexcitable
• Reduced INa amplitudes 40 100 Excitable inactivation

Normalized current (%)


SD SD
(SD-model, chronic CLP-sepsis) Inexcitable inactivation
20 80
• Acute INa increase
0
(septic serum challenge) 60

mV
–20 13.5 mV
• Increased expression of Nav1.5 Excitable RP
–40 40
(embryonic isoform)
–60 20 Inexcitable RP
• Hyperpolarizing shift of steady-state inactivation
(chronic CLP-sepsis, SD-model) –80 10 ms
0
–100 –90 –80 –70 –60 –50
Voltage (mV)

DHPR Acute septic serum challenge


(heart) 100

Normalized current (%)


Nav1.4 Nav1.5 75

50
DHPR
25 CS 30 kDa
CIM 30 kDa

0
–100 –80 –60 –40
Pre-pulse potential [mV]
Chronic sepsis (CLP)
100

Normalized current (%)


INa
Other channels, membrane parameters: 75
• Membrane depolarization in all sepsis models
Septic –2.5 nA 50
• L-type channel function impaired Septic Control
• Cl– conductance reduced
25
Control –6.5 nA
0
–150 –100 –50 0 50
Pre-pulse potential (mV)
FIGURE 6. Alterations of ion channels in models of ICU-related myopathies. Graphical summary of docu-
mented mechanisms contributing to altered membrane excitability through ion channel dysfunction in ICU-
related myopathies. Most research performed points towards reduction in ion current densities, e.g., for Na⫹,
Ca2⫹, and chloride conductances. For voltage-gated Na⫹ channels, a hyperpolarizing shift of inactivation curves
is induced in chronic sepsis and steroid-denervation models of critical illness, thus reducing availability of Na⫹
channels for action potential generation at resting potentials, which is even worsened by additional membrane
depolarizations. Results shown are taken from References 218, 581, 594, and 696. [Top middle and right
panels from Rich and Pinter (580). Copyright John Wiley and Sons. Middle right panel from Friedrich et al.
(218). Copyright Springer Science ⫹ Business Media.]

left-shifted approximately ⫺20 mV towards more negative details whether and how patients were subjected to analgo-
potentials by TNF-␣ (TABLE 5). The time course of current sedation (i.e., use of fentanyl or related analgo-sedatives to
block was almost complete at its maximum potency from maintain intubation; Ref. 695) and mechanical ventilation
15 min post-exposure for larger concentrations tested (⬎10 and silencing that could further contribute to pro-inflam-
ng/ml) and even earlier for lower concentrations (⬃5 min matory cytokine production. Since interstitial TNF-␣ con-
for 2.5 ng/ml) and can be considered an acute effect during centrations might be substantially higher in LPS-stimulated
sepsis (274), probably also in nonseptic critical illness con- muscle (known to additionally secrete TNF-␣; Ref. 471), a
ditions associated with increase in pro-inflammatory mi- maximum block of INa of ⬃75% as assessed by Guillouet et
lieu. The lower concentrations for which ⬃5% Na⫹ current al. (274) might be a realistic scenario. Since the TNF-␣
(INa) block was observed have been correlated with serum effect was complete within minutes, a transcriptional mod-
plasma levels found in septic patients with multiple organ ification like channel numbers or channel isoform, espe-
failures (548) and lipopolysaccharide (LPS)-induced sepsis cially a shift towards the embryonic Nav1.5 isoform, seems
in rats (221), although vast variations can be found in the unlikely in the acute TNF-␣ challenge model. Moreover, the
literature with even 100-fold lower concentrations in sepsis authors observed a most intriguing effect of blocking PKC-
(249, 355). Another error variable may stem from the fact mediated phosphorylation by pretreatment of fibers with
that most studies on septic patients do not give further chelerythrine (274), a blocker of protein kinase C (761).

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1049


FRIEDRICH ET AL.

This suggests a specific PKC-mediated change in Na⫹ chan- membrane resistance and conductance changes, as well as
nel gating through phosphorylation, probably between do- energy depletion of ATP stocks that feed electrogenic
mains III and IV (45) by TNF-␣ in skeletal muscle, similar to pumps to maintain negative membrane potentials, i.e.,
what has been found in neurons (139). Whether TNF-␣ Na⫹-K⫹-ATPase. In early studies on skeletal muscles from
does so in muscle primarily by either the A2 phospholipase, primates with septic shock, the depolarized membrane po-
phospholipase C, or protein kinase A pathways or combi- tential was associated with increased intracellular sodium
nation of those, all of which stimulate PKC (593), remains chloride content and marked depletion of extracellular con-
to be determined. Interestingly, the hyperpolarizing shift of tent (717). In dogs and rabbits during E. coli-induced bac-
Nav1.4 inactivation of about ⫺20 mV is also reproduced in teremia (338, 635), a marked membrane depolarization of
human voltage-gated Nav1.4 channels expressed in human skeletal muscle was also noted. Cellular injury as a result of
embryonic kidney cells following acute application of lipo- cell swelling in E. coli-induced sepsis in a rabbit model was
polysaccharides at ⬎50 ng/ml (283), pointing towards an used to explain membrane depolarization in muscles before
acute acquired channelopathy by LPS or TNF-␣ similar to the onset of septic shock (338). The selective increase in
that seen in critical illness and sepsis (594) (FIGURE 6). Na⫹ and Cl⫺ concentrations in hindlimb adductor muscle
was also confirmed in a dog sepsis model 48 h after infusion
1. Transferability to patients of E. coli, without any change to K⫹ concentrations (635).
The increase in Na⫹ permeability of the membrane was
Although the muscle membrane hypoexcitability is a hallmark sufficient to explain the observed ⬃10 mV depolarization
in CIM patients, the presence of the same biophysical changes which was also supported by the significant decrease in ATP
to Na⫹ channels as detailed above in animal models also in concentration in those muscles (635). However, another
patients has never been proven to date and is clearly a chal- study in rats with subacute E. coli-induced sepsis showed a
lenge for future research directions. As detailed later in section different scenario: an increased Na⫹-K⫹-ATPase activity
XII, those animal models are adequate to reproduce the mus- and unaltered ATP and phosphocreatine levels. The pre-
cle phenotype of CIM patients; thus a similar Na⫹ channelo- served ATP content and increased pump activity were ex-
pathy in CIM patients is probable. Clearly, in the critically ill plained by sepsis-induced hyperlactatemia and increased
patients who succumbed to one of the varieties of the ICU muscle content of lactate, a known stimulator of Na⫹-K⫹-
syndrome, many systems will be altered, such as the ionic ATPase, that even led to decreased intracellular Na⫹ con-
balance of potassium, sodium and chloride in the plasma, cal- centrations (469). Unfortunately, membrane potentials
cium in the plasma, and the osmotic strength of the extracel- were not recorded in that study. Similar observations were
lular fluid bathing the muscle fibers. All of these will have a made in a rat CLP model (347). In another mouse LPS-
significant effect on the function of nerve and muscle. How- induced sepsis model, Liu et al. (432) observed a marked
ever, clinical data on this are extremely scarce. For example, depolarization in the diaphragm and increased input resis-
plasma Ca2⫹ concentration changes have been associated with
tance due to an endotoxin-induced shutdown of sarcolem-
CIPNM in ICU patients; however, this was true both for hypo-
mal chloride conductance while K⫹ conductance was unal-
and hypercalcemia (17). Studies on plasma Na⫹, K⫹, Cl⫺, and
tered. NO synthase (NOS) inhibitors reversed the observed
osmolarity in diagnosed CIM patients seem not to be available
effects arguing in favor of an NO-mediated alteration of
and are clearly needed.
chloride conductance that was also confirmed by the fact
that in vitro application of LPS by itself to diaphragm was
C. Membrane Depolarization in Muscle in not able to reproduce the membrane effects seen in the
Critical Illness septic mouse, whereas addition of an NO donor was (432).
In the CIM model of steroid denervation (SD), a downregu-
A consistent finding in skeletal muscle from animal models lation of Cl⫺ conductance was not seen with SD but only
and patients with critical illness conditions (sepsis, mechanical following denervation alone (582).
ventilation, immobilization, steroid-denervation, etc.) is a sub-
stantial membrane depolarization that limits excitability of In several studies, a circulating factor was postulated to be
muscle (138, 178). However, unlike for sepsis or steroid-de- responsible for fast membrane depolarization in the ap-
nervation, for pure mechanical ventilation and/or immobiliza- proximately minute range (178, 218). For instance, plasma
tion, membrane depolarization is much less well documented, samples from sheep or pigs in shock were assayed for depo-
both for patients and for animal models, except for one study larizing activity when applied to rat diaphragm in vitro
reporting slight depolarizations (⬃5 mV) in a rat limb muscle (90). The depolarizing effect of a so-called “circulating de-
immobilization model (surgical muscle shortening) (798). polarizing factor” (CDF) was similar as the membrane po-
tentials found in the muscles “in situ” in the original ani-
There are plenty potential mechanisms to explain depolar- mals in shock the plasma came from (90). Given that sepsis
ized membrane potentials, e.g., changes in Nernst poten- is associated with membrane depolarization, increased Na⫹
tials of respective ion species due to concentration changes permeability and Na⫹ content (121, 244, 614), one of the
(i.e., changes in plasma concentrations of Na⫹, K⫹, or Cl⫺), potential mechanisms that contribute to the maintenance of

1050 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

increased [Ca2⫹]i in septic muscle may be reflected by an illness in muscle. In the SD model of critical illness myop-
increased activity of the Na⫹/Ca2⫹ exchanger (NCX) to athy, Kraner et al. (377) observed an elevation of RyR1 and
import Ca2⫹ for the extrusion of subsarcolemmally ele- CaV1.1 from immunostaining of single fibers that especially
vated Na⫹. In a rat endotoxemia sepsis model, expression were severely atrophic and had disorganized sarcomeres. It
levels for NCX were found increased in the early course of was thus concluded that increased Ca2⫹ release from the SR
sepsis (4 h) in cardiomyocytes and then declined at later may contribute to the pathology in CIM. However, SR
stages (30). However, for skeletal muscle, such recordings Ca2⫹ release was not directly assessed, nor was dihydropyr-
are not yet available. The increased Ca2⫹ uptake as a re- idine receptor (DHPR) function. At least in the heart, stud-
sponse to enhanced depolarization is not only a mechanism ies have pointed towards an impaired SR Ca2⫹ release
in pathophysiology of septic myopathy but also visible in through the ryanodine receptor during the hypodynamic
exercising muscle where bouts of tetanic stimuli can cause phase of sepsis (162, 163), and a decrease in ryanodine
depolarization and Ca2⫹ uptake (447, 541). receptors was detected in endotoxin shock in the heart (430,
787). L-type Ca2⫹ currents recorded in single ventricular
Although much evidence is in favor of the sepsis-induced de- cardiomyocytes isolated from pigs with early phase hyper-
polarization hypothesis, isolated inflammatory cytokines, i.e., dynamic septic shock revealed a significant decrease in
TNF-␣, also produced controversial results. For instance, Ca2⫹ current densities (⬃25%) with a consistent shift of
while Tracey et al. (712) reported a depolarization in single both activation and inactivation curves ⬃5–10 mV towards
fibers during whole muscle impalements, another study more negative potentials (659). TNF-␣ did not induce any
showed a dose-dependent hyperpolarization of membrane po- further changes to the current properties (659). A similar
tentials by TNF-␣ in enzymatically isolated peroneus longus reduction of L-type Ca2⫹ currents in ventricular cardiomy-
fibers (274). However, since those authors used a vigorous ocytes from septic guinea pigs 4 h after E. coli LPS injection
collagenase isolation procedure (3 mg/ml collagenase for 2 h at was found that was, however, unrelated to any changes in
37°C), their control Em values were already somewhat depo- steady-state activation or inactivation (810). L-type chan-
larized (approximately ⫺60 mV) and repolarized by TNF-␣, nel function in skeletal muscle in conjunction with critical
presumably by stimulating the Na⫹-K⫹-ATPase (274). Yet, illness myopathy has so far only been addressed in one
another in vitro study in whole EDL muscles from guinea pigs study using the acute serum challenge from CIM patients to
treated with TNF-␣ (5 and 10 ng/l for 30 min) showed no normal muscle fibers (cf. sect. XII). Serum fractions from
effect at all on resting membrane potentials (14). septic and mechanically ventilated critically ill patients
acutely reduced Ca2⫹ current amplitudes when applied to
Although the relationship between sepsis, systemic inflamma- murine normal mouse single fibers without any shifts in
tion, and inflammatory cytokines on the one side and mem- steady-state activation and inactivation curves (217). This
brane depolarization in skeletal muscle seems well established, was interpreted as an acute reduction of the fraction of
it is 1) not yet established that this sepsis-induced myopathy functional L-type channels by a putative low-molecular-
(SIM) is equivalent to CIM since no study yet convincingly has weight factor (218, 290, 318, 759).
demonstrated the preferential myosin loss in long-term sepsis
[on top of the severe atrophy in experimental sepsis affecting In summary, ion channels other than Na⫹ channels still
both actin and myosin equally seen as a general myofilament have only scarcely been addressed but are certainly an in-
release, see below and Williams et al. (776)] and also 2) teresting field to highlight in future studies involving critical
whether membrane depolarization is indeed present in pa- illness myopathies.
tients with CIM. Although critically ill patients with clinically
diagnosed CIM show reduced muscle membrane excitability, 1. Are electrically active tissues other than skeletal
direct in vivo recordings of resting membrane potentials in muscle affected?
CIM patients are not available except for one early study that
showed substantial resting transmembrane potential depolar- One of the major problems triggered by critical illness is
ization in severely critically ill patients in vivo (138). Unfortu- multisystem organ failure. The mechanisms underlying or-
nately, details about whether these patients were mechanically gan failure remain poorly understood. The finding that so-
ventilated or septic were not reported. This limits our under- dium channel dysfunction contributes to failure in force
standing of membrane depolarization in CIM to direct proof generation in skeletal muscle demonstrates that a novel
from the SD rat animal models of CIM (377, 581) but not yet form of acquired channelopathy may be present during the
for mechanical ventilation and immobilization models (517). acute phase of critical illness. If channelopathy affects other
electrically active tissues, it could provide a mechanism to
account for failure of a number of different organ systems.
D. Membrane Ca2ⴙ Channels in ICU-Related
Myopathies It is well established that critical illness can induce neurop-
athy. The first indication that neuropathy in critically ill
Voltage-gated Ca2⫹ channels, surprisingly, have only been patients might not be entirely accounted for by axon degen-
very modestly studied in conjunction with sepsis and critical eration came from a study in which nerve and muscle biop-

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1051


FRIEDRICH ET AL.

sies were taken in patients with severe critical illness poly- quired sodium channelopathy in heart that affects the Nav1.5
neuropathy and myopathy. Nerve biopsy in many patients Na⫹ channel isoform. The presence of Na⫹ channelopathy in
with electrophysiological evidence of neuropathy was nor- heart may contribute to the reduced contractility of heart that
mal (403). This finding suggested there was a functional occurs during sepsis (481). Partial block of Na⫹ current in
deficit in nerves of affected patients that did not have a heart with the Na⫹ channel specific blocker tetrodotoxin
pathological correlate. More recently, it was found that mimicked changes in excitability that occurred during sepsis,
nerve excitability was reduced in patients with critical ill- i.e., reduced excitability and greatly reduced cardiac contrac-
ness polyneuropathy (800). The presence of a functional tility (371). These data suggest that reduction of Na⫹ current
deficit in the acute phase of critical illness polyneuropathy might contribute to reduction in both cardiac excitability and
raised the possibility of rapid recovery since restoration of cardiac contractility during sepsis.
excitability could occur more rapidly than regrowth of ax-
ons. Rapid improvement has been reported in some patients Motoneuron excitability is also significantly reduced within
with critical illness polyneuropathy (515). 24 h of sepsis induction in rats, in particular during repetitive
firing of action potentials (500). Any reduction in motoneuron
In studies of septic rats, peripheral nerves revealed reduced excitability will reduce motor unit recruitment and thus con-
excitability that could not be accounted for by changes in tribute to weakness. Motoneurons execute their role in mod-
input resistance or resting potential excitability (515). Instead, ulating force output by converting the synaptic current deliv-
restoration of normal excitability following brief hyperpolar- ered to the soma into a firing rate that determines muscle force
ization of axons suggested that a hyperpolarized shift in the output. The finding of reduced motoneuron excitability raises
voltage dependence of sodium channel inactivation was the the possibility that channelopathy within the central nervous
mechanism underlying reduced excitability (515). These data system also contributes to weakness.
suggest that a sodium channel dysfunction similar to the one in
skeletal muscle also occurs in peripheral nerve. To summarize, studies to date in skeletal muscle, peripheral
nerve, heart, and spinal cord are all suggestive of some
The presence of sodium channelopathy in both muscle and acquired channelopathy that is triggered by critical illness.
nerve might provide a mechanism to explain why critical No studies to date have examined excitability of neurons
illness myopathy and critical illness neuropathy often coex- within the brain. Septic encephalopathy is one of the most
ist (43, 44, 64, 147, 361, 403, 413, 528, 614). When pa- common and troubling complications of critical illness
tients with co-occurrence of neuropathy and myopathy are (564). Despite a number of studies, the mechanism under-
studied longitudinally, many evolve into either CIP or CIM lying septic encephalopathy remains a mystery. Presence of
(43, 273, 361). This raises the possibility that sodium chan- channelopathy in multiple tissues raises the possibility that
nelopathy is present at early stages. If reduced excitability of reduced excitability of neurons within the CNS might un-
peripheral nerve and muscle is due to sodium channelopa- derlie septic encephalopathy. If this was the case, a single
thy, the channelopathy would have to affect multiple so- therapy to improve excitability might treat failure of a num-
dium channel isoforms. In the rat steroid-denervation ber of electrically active tissues.
model of critical illness myopathy, the sodium channel iso-
forms present in skeletal muscle are Nav1.4 and Nav1.5 Why might sodium currents be reduced in multiple tissues
(578). In dorsal root axons, Nav 1.6 as well as other sodium during sepsis? One possibility is that Na⫹ currents are es-
channel isoforms are expressed (19, 92). pecially sensitive to cell sickness/injury. In studies in both
skeletal muscle and axons, it appears that damage triggers a
There is evidence consistent with sodium channelopathy in hyperpolarized shift in the voltage dependence of the Na⫹
the heart during sepsis. ECG amplitude is reduced during current activation and inactivation as well as a reduction in
the acute phase of sepsis (579). One interpretation of this is maximal current density (72, 195, 494). In injured cells
that there is reduced excitability of myocardium. To di- with a depolarized resting potential, a hyperpolarized shift
rectly measure myocardial excitability, a recent study in in the voltage dependence of inactivation would result in
CLP-induced sepsis in rats measured properties of papillary increased channel inactivation to reduce excitability. De-
muscle action potentials with microelectrodes. Although creased cell excitability might even serve to increase survival
resting potential was not altered, action potential amplitude of the injured cell by shutting down a Na⫹ “window” cur-
was reduced and threshold was elevated 24 h after sepsis rent that is triggered by depolarization of the resting poten-
induction (371). Reduction in the rate of action potential tial (547). The Na⫹ window current might further depolar-
rise suggested the etiology of reduced cardiac excitability ize the cell, to cause elevation of intracellular Ca2⫹ (252,
was a reduction in sodium current. The in vivo data fit well 547) and cell death. One way to prevent this cascade of
with an earlier in vitro study in which application of LPS to events is to shut down Na⫹ current. This has the cost of
mouse cardiomyocytes reduced excitability (785). Al- reducing excitability and, thus, temporarily worsening cell
though the mechanism underlying the reduction in sodium function, but might promote survival. Reduction of Na⫹
current is not known, these data are consistent with an ac- current might serve a similar purpose to the use of barbitu-

1052 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

rate coma in patients with acute neurologic injury: by de- flux both in septic and control animals, however, to much
creasing metabolic demand during a period of cell stress, it larger extent in the septic muscle group (57) (FIGURE 7B).
might increase cell survival to improve long-term outcome Treatment of normal muscle with the Ca2⫹ ionophore iono-
(319, 432, 604). mycin induced a similar increase in Ca2⫹ flux as seen in the
septic rat muscles. The increase in Ca2⫹ flux was paralleled
by an increase in tyrosine release linking increased Ca2⫹ to
IX. EXCITATION-CONTRACTION COUPLING,
Ca2ⴙ HOMEOSTASIS, AND MOTOR a higher proteolysis rate (57, 613). Similar results were
PROTEINS IN CRITICAL ILLNESS obtained after a single intraperitoneal E. coli injection to
male rats in soleus muscles 10 h after inoculation (771).
Another study using the CLP septic rat model also showed
A. The Normal Electromechanical Signaling an increased Ca2⫹ content by 50% in soleus muscle and by
Cascade in Muscle 10% in extensor digitorum longus (EDL) muscle when
bathed in different Ca2⫹ solutions for up to 120 min (49).
Force output in skeletal muscle is the result of a complex Since 45Ca flux measurements or total Ca2⫹ content do not
cascade of mechanisms that convey electrical stimulation of reflect free myoplasmic Ca2⫹ concentrations, Fischer et al.
the surface and tubular membranes to an intracellular (202) took the next step and measured approximately
chemical response (rise in myoplasmic Ca2⫹ concentration) threefold increased resting fura-2 Ca2⫹ levels in whole EDL
followed by a mechanical contractile response (50, 151, muscles from septic rats 16 h after CLP induction (⬃1 ␮M
477). Ca2⫹ ions are released from the sarcoplasmic reticu- vs. ⬃350 nM in untreated rats) (FIGURE 7C). Although these
lum (SR) in response to a tubular action potential (388) in a control values seem somewhat elevated compared with sin-
process called excitation-contraction coupling (EC cou- gle fiber Ca2⫹ determinations (40 –100 nM; Refs. 208, 296,
pling). Ca2⫹ ions then quickly diffuse within the myoplasm 777), they are similar to other studies using fura-2 calcium
(41, 151) to bind to troponin C that acts as a Ca2⫹ switch of fluorescence recordings in total muscle (447), probably re-
the thin filaments of the contractile apparatus (412, 671). flecting some inhomogeneity contributions from dye cap-
This initiates the cross-bridge cycle to recruit weakly or tured in the intercellular space or contributions from endo-
strongly bound cross-bridges, depending on the imposed thelial, vascular smooth muscle cells, or immune cells resid-
load (516a). This results in contractile power, either as iso- ing in the muscle tissue. Surprisingly, direct recordings of
metric, isotonic, or a mixed contraction. After the excita- resting Ca2⫹ levels in single muscle fibers following septic
tion ceases, Ca2⫹ ions are redistributed into the SR via a challenge are still missing. Specifically, most studies on al-
sequential buffering to low-affinity buffer proteins (e.g., tered Ca2⫹ homeostasis in sepsis models have been per-
parvalbumin, etc.) and the SR Ca2⫹-ATPase pump activity formed in cardiomyocytes because cardiomyopathy is also
(SERCA) (50, 354, 478). The release machinery at the level a complication in sepsis. In rat models of sepsis, either CLP-
of the SR ryanodine receptor (RyR1 in muscle) is a tightly (807) or LPS-induced (292), a decreased fraction of cardiac
controlled and diversely modulated molecular release gate shortening was explained by a significant reduction in Ca2⫹
(169, 281, 558, 813). The mechanical output of the motor transient amplitudes 48 h after CLP (807) or 4 h after LPS
proteins is mainly determined by the myosin heavy chain (292). Reduced SR Ca2⫹ content of ⬃380 nM (LPS) vs. 560
expression which varies from muscle to muscle and is also nM (control) was subsequently explained by a significantly
subject to a high degree of plasticity in health and disease compromised SR Ca2⫹ uptake and increased SR Ca2⫹ leak
(287). Additional determinants are also the light chain iso-
in SR vesicles (292) and from largely increased Ca2⫹ spark
forms and their respective phosphorylation (264) that can
frequencies in intact cardiomyocytes (807). Dantrolene, a
be affected downstream to many signaling pathways and
blocker of the ryanodine receptor, normalized the SR Ca2⫹
stimuli, e.g., hormones (422) or weight-bearing function
leak in SR cardiac vesicle preparations from LPS-treated
(398). It is apparent that disturbances at any step of the
rats (292) and significantly reduced elevated resting Ca2⫹
contractile cascade can induce or sustain some of the pa-
levels in LPS-treated guinea pig cardiomyocytes (⬃230 nM)
thology in all forms of ICU-related myopathies (FIGURE 1B).
to control values (⬃150 nM) (702). More importantly,
Thr-17 phosphorylation of phospholamban was 30% re-
B. Altered Global Muscle Ca2ⴙ Homeostasis duced, explaining the compromised SR Ca2⫹ uptake in sep-
in Critical Illness tic cardiomyocytes (292). Interestingly, the contractile ap-
paratus properties so far present a diverse picture. In an
Early studies in septic rats (bacterial peritonitis; no mechan- endotoxemia-induced myocardial dysfunction model in
ical ventilation) already pointed towards impaired intracel- guinea pigs, Ca2⫹ sensitivity of the myofilaments was unal-
lular Ca2⫹ regulation: a significant Ca2⫹ uptake was found tered 4 h after LPS induction in chemically skinned cardio-
in soleus muscle at days 1–3 of sepsis induction that was myocytes, although maximum CSA-normalized tension
abolished by the L-type calcium channel blocker diltiazem generated was significantly reduced, an effect that was re-
(57). 45Ca fluxes in septic muscles were ⬃25% larger com- voked in the presence of protease inhibitors (584). How-
pared with mock treated animals. Diltiazem reduced Ca2⫹ ever, another study in endotoxin-treated rabbits showed

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1053


FRIEDRICH ET AL.

A B C
- diltiazem + diltiazem

Ca2+ Flux: nmol/(g x 0.5hr)


250 (6) 1.5 - dantrolene
(22) P < 0.05
200 (10)
+ dantrolene

[Ca2+]i (μM)
(6) (8) (8)
(5) 1.0
(7) (6)
150 (15)

100
0.5
50

0 0
Control Sterile Septic Sterile Septic
control LPS sepsis
0 1 2
Days Post - Implantaion

D E F
diaphragm
25
M. soleus

membrane damage (%)


25 IL-1
20
depolarization (mV)

control
Muscle fibre
20 low Mg2+
15
15 low Mg2+
10 10

wash

0.5 (a.u.)
5

0.5 (a.u.)
5
out
0
10-1 100 101 102 103 0
25 s
endotoxin (mg/l) ctrl. LPS CLP 50 s 25 s

sepsis
FIGURE 7. Key findings involved in the reconstruction of the cellular pathology related to Ca2⫹ homeostasis,
EC coupling, and motor protein function in sepsis-related myopathies. A: macrophages (CD68-positive) within
muscle tissue in a biopsy from a CIPNM patient. [From De Letter et al. (150). Copyright 2000 Elsevier.] B:
Ca2⫹ flux into soleus muscles from septic (fecal pellet implantation) and sham-operated animals either
pretreated or not with diltiazem. [From Bhattacharyya et al. (57).] C: resting fura-2 Ca2⫹ levels in whole EDL
muscles from sham or CLP-septic rats (202). D: depolarization potency of plasma from a sheep in endotoxin
shock on rat diaphragm in vitro, compared with direct E. coli LPS depolarizing effect on rat diaphragm (90). E:
membrane integrity is compromised differentially in different muscles and sepsis models. Myofiber membrane
damage allows passage of extracellular cations and small molecules into the myoplasm (427). F: force
recordings in a single skinned fiber bathed in a low Mg2⫹ (10 ␮M) environment under control conditions (no
IL-1) and in the presence of 25 ng/l IL-1 (␣ isoform). As long as IL-1 is present, no force is produced. Washing
out IL-1 while still in low Mg2⫹ environment then restores a force transient indicative of IL-1 directly stabilizing
the inhibitory Mg2⫹ site on RyR1. [From Friedrich et al. (220). Reprinted with permission of the American
Thoracic Society. Copyright 2015 American Thoracic Society.]

higher Ca50 values for half-activation of papillary muscle control values (202), suggesting a potential involvement of
isometric force (1.78 vs. 1.53 ␮M) already 4 h after a 0.5 the SR and the RyR1 in the altered Ca2⫹ homeostasis dur-
mg/kg iv endotoxin treatment, indicating a reduced myofi- ing sepsis. The increased [Ca2⫹]i is furthermore expected to
brillar Ca2⫹ sensitivity. The latter even worsened after a activate calpains for the breakdown of desmin, ␣-actinin,
higher LPS dose (1 mg/kg: Ca50 ⫽ 2.08 ␮M) and with time and other Z-disk anchoring proteins (202, 776). However,
(2.12 ␮M after 24 h) but returned to normal after 5 days the above-mentioned CLP study did not claim to establish a
(692). Treatment of control papillary muscle strips with cause-effect relationship for dantrolene to exclusively act
isoproterenol produced a reduction in myofibrillar Ca2⫹ on muscle [Ca2⫹]i, since 1) only one time point was inves-
sensitivity similar to that seen in LPS-treated preparations, tigated (16 h post CLP or sham) and 2) dantrolene also
indicating a higher degree of protein kinase A-dependent significantly reduced plasma TNF-␣ and corticosteroids
phosphorylation as a cause for the changes in Ca2⫹ sensi- levels (202). Since TNF-␣ at 10 nM levels is known to
tivity seen in septic hearts (693). depolarize resting membrane potentials in whole EDL and
soleus muscles already after short incubation (712), it can
In skeletal muscle, elevated resting cytoplasmic [Ca2⫹]i also be speculated that the associated increase seen in [Ca2⫹]i in
seems to be a major trigger for subsequent downstream sepsis 16 h post-CLP may rather be an effect of increased
proteolysis and metabolic failure events. Strikingly, the plasma TNF-␣ because membrane depolarization has been
vastly increased [Ca2⫹]i found in whole EDL muscles 16 h shown to increase the membrane permeability towards
after CLP procedure in rats was dantrolene responsive, i.e., Ca2⫹ (120, 188, 771). Since the enhanced Ca2⫹ uptake in
dantrolene completely reversed the increased [Ca2⫹]i to K⫹ depolarized skeletal muscle from septic rats was blocked

1054 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

by diltiazem that blocks the dihydropyridine receptor this approach is considered qualitative, at best, and has to
(DHPR) (771), it is likely that the mechanism of this Ca2⫹ be interpreted with caution. For example, their calculated
entry in sepsis might be identical to the “excitation-coupled Ca2⫹ transient amplitudes in the general anesthesia group
Ca2⫹ entry” (ECCE) that requires a functional DHPR but is after 10 mM caffeine were close to 2 ␮M and ⬍1 ␮M in the
independent of store depletion (33, 334, 446). The upregu- CLP group (809). However, direct Ca2⫹ fluorescence re-
lation of the Cav1.1-ryanodine receptor complex in a rat cordings of Ca2⫹ transients in rat EDL muscles using Mag-
model of critical illness myopathy may also point towards fura-2 already gave twitch peak Ca2⫹ amplitudes of ⬃18
this direction (377). Therefore, it is tempting to speculate ␮M (41). Therefore, a mechanistic understanding of altered
that membrane depolarization might be a first event that Ca2⫹ homeostasis in experimental sepsis still awaits exper-
initiates increased membrane Ca2⫹ permeability and rise in imental clarification. In an isolated in vitro study in C2C12
[Ca2⫹]i followed by activation of Ca2⫹-dependent proteol- myotubes, TNF-␣ treatment (10 ng/ml) decreased electri-
ysis and, eventually, necrosis. cally evoked Ca2⫹ transient amplitudes significantly from
⬃0.4 to ⬃0.2 ␮M after 48 h (741). Resting Ca2⫹ levels were
The above-mentioned studies mostly relate to models of also ⬃30% reduced. A decreased rate of Ca2⫹ transient
sepsis-induced myopathy; strikingly, for CIM, no study on decay by TNF-␣, however, suggested that SR Ca2⫹ pump
Ca2⫹ homeostasis is available, neither for patient muscles function might have been compromised with less SR Ca2⫹
nor for animal models. available for release (741). In another in vitro study chal-
lenging single isolated flexor digitorum brevis fibers with a
C. Muscle Ca2ⴙ Stores and Ca2ⴙ Transients rather large dose of TNF-␣ (500 ng/ml), resting [Ca2⫹]i and
tetanic [Ca2⫹]i amplitudes were almost identical pre- and 4
in Critical Illness
h post-treatment (571). A very recent study in mouse EDL
skinned fibers challenged with IL-1 (␣ isoform) revealed a
Another mechanism for the Ca2⫹ entry in septic muscle may
rather unexpected mode of action in skeletal muscle: incu-
involve a dysregulation of the Ca2⫹ store with activation of
bation of skinned fiber bundles with IL-1 at concentrations
store-operated Ca2⫹ entry (SOCE). A reduced ryanodine-
to those found in septic patients (⬎10 ng/l) reversibly re-
induced contracture in diaphragm from septic mice (7.5
duced the SR Ca2⫹ release induced by relieving the Mg2⫹
mg/kg LPS ip) that was reversed by the LPS inhibitor poly-
inhibition on the RyR1 (220). Co-immunoprecipitation
myxin B confirmed the involvement of LPS signaling on the
and immunofluorescence colocalization experiments sug-
sarcoplasmic reticulum regulation (433). This was ex-
gested an association of IL-1␣ to RyR1 to putatively in-
plained by a similarly reduced SR Ca2⫹ release by LPS.
crease the Mg2⫹ inhibition on the RyR1. Assessment of the
Whether this is due to a decreased SR content of releasable
Ca2⫹ as a consequence of an increased SR leak similar to SR leak as the “Ca2⫹-retention index” (408) indicated a
cardiac muscle (e.g., Refs. 170, 292) has not yet been ad- reduction of SR Ca2⫹ leak by IL-1␣ (220). In contrast, in
dressed. If the SR content was indeed reduced in septic cardiac muscle, IL-1 (␤ isoform) increased SR Ca2⫹ leak
skeletal muscle, SOCE could in principle be activated (254, (170). Therefore, in skeletal muscle, this may point towards
806). However, in a steroid-denervation rat model of CIM, a novel mechanism where IL-1␣ associates with the RyR1
ORAI1 (Ca2⫹ release-activated Ca2⫹ channel protein 1) to interfere with SR Ca2⫹ release. Although the store con-
expression levels were found ⬃20% reduced compared tent was found increased (220), the increased blockage of
with controls 7 days post-op (377). SR function in skeletal Ca2⫹ release by IL-1␣ may be one explanation for weakness
muscle still has only been initially studied in a CLP murine in critically ill patients with increased pro-inflammatory
sepsis model by Zink et al. (809). The authors compared response. The question of how IL-1␣ would enter the mem-
“intracellular Ca2⫹ regulation” in skinned EDL muscle fi- brane barrier from outside was also addressed in the afore-
bers from CLP, sham-operated, and general-anaesthesia- mentioned study. Prolonged incubation of intact single fi-
only mice 2–7 days post-procedure, recording caffeine-in- bers with IL-1␣ dose-dependently impaired membrane in-
duced force transients. Relative force transients were de- tegrity, and IL-1␣ was subsequently found deposited within
creased in the surgical procedure groups from day two but the muscle cells, where it then could associate with RyR1 to
recovered in the sham group from day three while still de- impair EC coupling (220) (FIGURE 7F). In support of this,
clining in the CLP group to a minimum of ⬃40% at day experimental animal sepsis has been shown to induce mem-
three, followed by recovery to control values at day seven. brane damage in myofibers using membrane impermeant
SR reloading conditions were kept identical among groups, dyes. In diaphragm muscle, both CLP and LPS-induced sep-
though with a slightly overfilled SR compared with the usu- sis increased the percentage of myofibers with compromised
ally only one-third filling of fast-twitch muscles (554). Un- membrane integrity from ⬍1% (control) to between 15 and
fortunately, the study did not provide any mechanistic ex- 20%. In soleus muscle, LPS failed to compromise mem-
planation for the observed force reductions. A link to im- brane integrity but CLP-induced sepsis rose the percentage
paired Ca2⫹ homeostasis on the SR level was made from of damaged fibers to ⬃8% (427, 673). Alongside with
conversion of the force responses to “apparent Ca2⫹ tran- membrane damage, resting membrane potentials were sig-
sients,” using the steady-state pCa-force curves. However, nificantly depolarized in diaphragm muscle fibers (from

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1055


FRIEDRICH ET AL.

⫺79 to ⫺68 mV; Ref. 427) (FIGURE 7E). Thus sepsis “per cle cells stimulates neutrophils in a paracrine fashion to
se” or the combination of pro-inflammatory cytokines (like increase their production of cytokines, i.e., TNF-␣ (739).
TNF-␣ or IL-1) may compromise membrane integrity with
an additional entry pathway for cations and small-molecu- In both cardiac and skeletal muscle (diaphragm), a decrease
lar-weight factors to depolarize membrane potential, in- in myofibrillar Ca2⫹ sensitivity can be routinely detected,
crease resting [Ca2⫹]i, or exert specific effects like the puta- either in CLP- or LPS-induced sepsis models (94, 674). This
tive direct binding of IL-1 isoforms to RyR1. The mem- decrease in Ca2⫹ sensitivity seems to be a direct conse-
brane damage mechanism may be unspecific or involve quence of production of free radicals since prevention of
specific signaling pathways, e.g., free radicals that are up- peroxynitrite formation with superoxide scavengers (e.g.,
regulated in inflammatory myopathies and in response to pegylated superoxide dismutase; SOD) or NOS inhibition
cytokines (225, 679, 681, 683). Sepsis-induced membrane with L-NMMA not only rescued mitochondrial dysfunction
damage was substantially reduced by NG-monomethyl-L- in muscle (61, 95; see sect. X), but also pCa-force curves in
arginine (L-NMMA), an inhibitor of NOS activity, pointing diaphragm single fibers from endotoxin-treated rats co-ad-
towards a role of NO-mediated compromise of membrane ministered with PEG-SOD but not with denatured PEG-
integrity (427). As long as sufficient myofiber repair mech- SOD (94). The reason for the NO production may be seen
anisms were still active, such membrane damage would be in the appropriate cytokine profile during sepsis. In partic-
sufficiently repaired with fibers not necessarily undergoing ular, TNF-␣ and IL-1 are prime candidates that have been
necrosis (93). However, the damage would be sufficient shown to result in adjacent activation of NO production. In
enough to let Ca2⫹, other cations and small molecules pass skeletal muscle, the inducible isoform iNOS is upregulated
the membrane. in response to inflammation or other infectious conditions
(74, 394, 609). Upregulation of iNOS by combined incuba-
tion of IL-1␤ and IFN-␥ in rat skeletal myoblasts was asso-
D. Ca2ⴙ Regulation of the Contractile ciated with ERK1/ERK2 activation and was completely ab-
Apparatus in Critical Illness rogated by NF-␬B blockade (4). In skeletal muscle biopsies
of congestive heart failure patients, a linear correlation be-
Apart from the effects of increased [Ca2⫹]i in septic muscle tween IL-1␤ content and iNOS expression was detected (4).
on motor protein turnover and energy metabolism leading In C2C12 skeletal muscle myocytes, combinations of
to proteolysis (see sect. XI), it is of interest to highlight TNF-␣, IFN-␥, and IL-1␣, but not either cytokine alone,
regulatory consequences on the motor proteins with respect were found to substantially increase iNOS activity, and RT-
to Ca2⫹ activation during EC coupling. Studies in the 1980s PCR analysis revealed that the iNOS mRNA sequence
already showed an in vitro inhibition of myofibrillar Mg2⫹- showed exact homology with macrophage iNOS, indicat-
activated ATPase activity in myofibrillar preparations from ing stimulation of skeletal muscle NO production via induc-
both rabbit skeletal and dog heart muscles when incubated tion of the macrophage-type iNOS gene (778). A 4 h incu-
with E. coli endotoxin (⬃0.05– 0.3 mg/ml) (537). Already bation of isolated mouse limb and diaphragm muscle with
after 2 h, ATPase activity was reduced by 60% in skeletal, TNF-␣ markedly suppressed force production in response
and even more pronounced in cardiac myofibrils. Whether to field stimulations up to 150 Hz (571). This force depres-
those results can be of pathophysiological relevance in the sion was located downstream to Ca2⫹ signaling since no
pathogenesis of sepsis-induced dysregulation of myofibril- compromise of tetanic [Ca2⫹]i by TNF-␣ (500 ng/ml) was
lar activity, however, is unknown. It would require LPS detected (571). A later study by the same group confirmed a
itself to be translocated to the myoplasm of septic muscle to depression of field-stimulated myofibrillar force without
exert this direct effect. A mechanism similar to that found any changes in Ca2⫹ sensitivity of the contractile apparatus
for IL-1 in muscle (220) or in cerebro-microvascular endo- (286). Using TNFR1-deficient mice treated with TNF-␣, the
thelial cells (645) might be possible where endotoxin could decrease in myofibrillar force was prevented and was simi-
enter through local membrane defects, e.g., also induced by lar to wild-type animals treated with vehicles, pointing to-
NOS activation (427, 673). Probably more important are wards the absolute requirement of TNF-␣ signaling
subsequent inflammatory reactions induced by endotoxin through its TNFR1 receptor to suppress myofibrillar force,
in the tissue, i.e., immune activation of macrophages and probably indirectly through ROS and RNS production
release of pro-inflammatory cytokines that provide the (286). So far, the molecular proteins affected by TNF-␣ are
grounds for subsequent alterations of contractile perfor- not yet known (286). Apart from indirect phosphorylation
mance (692). During endotoxin shock, large amounts of of regulatory proteins by peroxynitrite, a direct interaction
NO, either released by endothelial cells but also by myo- of TNF-␣ with intracellular target proteins similar to as has
cytes, seem to be causative for a deregulation of thin fila- been suggested for IL-1␣ (220) may be possible but has not
ment troponins by raising cGMP and phosphorylation of yet been tested.
troponins by cGMP-dependent protein kinases, at least in
the heart (75, 629, 673, 693, 791). In a positive-feedback The literature reviewed above exclusively refers to the
loop, NO released by endothelial and vascular smooth mus- association between sepsis, pro-inflammatory mediators,

1056 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

and myopathic changes, indicative of sepsis-induced my- portant factor than intrinsic dysfunction in acute sepsis-
opathy. For CIM, surprisingly no published data seem at associated human muscle weakness. This finding, together
hand on Ca2⫹ regulation of the contractile apparatus in with the recent finding that there may be problems with
CIM patients (i.e., nonseptic). In a preliminary study, activation of lower motoneurons in septic patients and sep-
sequential vastus lateralis biopsies from 12 critically ill, tic rats (500), raises the possibility that difficulties with
septic, and mechanically ventilated ICU patients, of activation of motor units may contribute to weakness and
which six were clinically diagnosed with CIM, were pre- fatigue in critical illness.
pared for pCa-force recordings of skinned fiber bundles.
However, although there were some changes in myofi- Most mechanisms of muscle fatigue in human sepsis had
brillar Ca2⫹ sensitivity in two CIM patients between been tracked down to mitochondrial alterations and ATP
early (⬃2– 4 days) and late (7–14 days) biopsies, the depletion in cases where muscle biopsies for biochemistry
baseline sensitivity in early biopsies between CIM and assays were available (211), but other levels had not been
non-CIM patients was similar (Friedrich, unpublished looked at. Surprisingly, animal studies on sepsis and muscle
observations). Regarding animal models of CIM, no data
fatigue are likewise rare and on animal CIM models, not yet
on myofibrillar Ca2⫹ sensitivity are available so far from
available (but on their way; Larsson, unpublished results).
the steroid-denervation model. In the rat ICU model,
In chronic CLP-induced sepsis, soleus muscles of rats after 7
myofibrillar Ca2⫹ sensitivity was consistently found to
days of sepsis were stimulated via ventral root single motor
decrease over the time course of 14 days intensive care
management (NMBs, mechanical ventilation and muscle units to assess fatigue index which indicated increased fati-
silencing) (517) (see also sect. XII). gability (567). However, mechanisms inherent to impaired
nerve transmission were not dissected from fatigue mecha-
nisms within the muscle itself. Another study that also em-
E. Muscle Fatigue in Critical Illness ployed CLP-induced sepsis in rats used direct muscular
stimulation of isolated diaphragm 16 h post-CLP proce-
Another important yet rather unresolved symptom of ICU- dure. Trains of tetanic stimulations (50 Hz for 2 s every 10
acquired muscle weakness is “muscle fatigue.” Increased fati- s for 5 min) were applied. Times to half decrement of the
gability has been repeatedly documented in patients with ex- first peak tetanic tension (T50) as fatigability index revealed
acerbated chronic inflammatory conditions (311, 449), but an almost 50% shorter T50 indicative of massive fatigue
only very few studies have looked at patients with sepsis and (488). This fatigue was associated with a marked increase in
multiorgan failure (181). Interestingly, force decrement after myeloperoxidase activity (MPO) as an index of neutrophil
fatiguing tetanic stimulations was not different between pa- activation in septic muscle tissue. Since in septic patients
tients with sepsis and multiorgan failure and volunteers fol- cAMP stores were found depleted (54), the authors also
lowing 2 wk of immobilization, although the baseline tetanic speculated that phosphodiesterase inhibition would allevi-
force was markedly depressed already before the fatigue pro- ate fatigue patterns. Using the PDE inhibitor olprinone,
tocol in the sepsis/MOF group (181). Although it is suggestive both MPO activity and fatigue were restored similarly to-
that the patients in that study actually suffered from CIM (i.e., wards control levels, indicating a crucial involvement of
were septic, immobilized, and mechanically ventilated), the activated neutrophils or macrophages within septic tissue to
authors only assessed the muscle weakness and electrophysi- contribute to downstream muscle weakness and increased
ology parameters but no muscle biopsies for preferential my- fatigability, probably through their cytokine release pat-
osin loss confirmation. Thus, although the authors classify
terns of IL-1 and TNF-␣ or free radicals (94, 224, 488). In
their myopathy as “sepsis-induced myopathy,” patients might
chronic inflammatory conditions of polymyositis and der-
in fact have been CIM patients.
matomyositis, muscle weakness in patients was correlated
with a strong expression of IL-1␣ in patient muscle tissues
Muscle fatigue is a very complex phenomenon that involves
(516). An even more massive expression of several pro-
multiple cellular correlates, e.g., transmission failure of ac-
tion potentials, tubular excitation spread failure, voltage inflammatory cytokines and macrophage infiltration found
sensor dysregulation, SR Ca2⫹ release, metabolic impair- in muscle biopsies from critically ill patients with probable
ment through ATP depletion or reuptake alterations, or CIM (patients septic and mechanically ventilated; evidence
myofibrillar protein alterations (13, 28, 172, 417, 572, for CIM from electrophysiology plus immunohistochemis-
657). In critically ill patients, muscle fatigue is usually only try, but not assessed for preferential myosin loss) (150)
assessed via nerval routes either using voluntary muscle (FIGURE 7A) may already point to a common denominator
exercise (278) or nerve stimulation (181). In a study on the of such a hypothesis in several forms of myopathy-associ-
effect of acute infusion of LPS into healthy volunteers, it ated inflammatory conditions. This may also hold true for
was found that there was an acute onset of mild muscle conditions without primary presence of inflammatory cells
weakness within 3 h (471). Interestingly, there was no def- in primary inflammatory myopathies, where muscle fibers
icit in electrically stimulated muscle contractions. The au- have been found to intrinsically overexpress IL-1␣, TNF-␣,
thors concluded that “loss of volition” may be a more im- IL-2, and IFN-␥ (150, 701).

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1057


FRIEDRICH ET AL.

Sepsis, critical illness

Diapedesis, Vessel PDE-inhibition


Direct
tissue infiltration
action?
Activated neutrophil/
+
macrophage Myeloperoxidase
(MPO) activity

LPS? Cytokine release


– AP
IFN-γ TNF-α IL-1α/β Em NO
+ Na+
Ca2+ Cytokine receptor
Membrane +
integrity ATP K+ iNOS, NO
DHPR depletion
+ expression
Ca2+
uptake
L-NMMA RyR1
Ca2+
Ca2+
↑NO ↓cAMP, ↑cGMP
SR SERCA
NF-κB

Intracellular ATP ADP, Pi
[Ca2+]i IL-1, IFN-γ
expression Ca2+
T-tubule –
Myosin Actin ATP ATP ATP

Νυχλευσ

• Pro-inflammatory cytokines + ATP


• DHPR upregulation, NO production depletion
cGMP-dependent
• Ca2+ influx↑, resting [Ca2+]↑ Tnl Actin Troponin/Tm
phosphorylation
• Membrane damage (primary/NO-induced)
• Membrane depolarization (10–15 mV) TNF-α Ca2+ Tnl Fatigue
• SR Ca2+ leak↑ or ↓ (?) sensitivity HMM
• Myofibrillar Ca2+ sensitivity ↓ aor

• Fatigue↑

FIGURE 8. Cellular mechanisms that contribute to muscle dysfunction in the phase determined by altered
Ca2⫹ homeostasis, EC coupling, and motor protein function during critical illness. For details, see section IX.
CLP, cecal ligation and puncture; LPS, lipopolysaccharides; DHPR, dihydropyridine receptor; SR, sarcoplasmic
reticulum; RyR1, ryanodine receptor 1; SERCA, sarco/endoplasmic reticulum Ca2⫹-ATPase; L-NMMA: NG-
monomethyl-L-arginine; iNOS, inducible nitric oxide synthase.

FIGURE 8 summarizes most of the mechanisms found in septic ergy metabolism is disturbed in several organs including
muscle explained in this section. Again, it has to be kept in skeletal muscle. The mitochondria as power plants of the
mind that results from septic ICU patients cannot assess sepsis cell are responsible for the majority of the ATP production
as independent variable for CIM development since all those through oxidative phosphorylation (FIGURE 9A).
septic patients underwent mechanical ventilation and immo-
bilization simultaneously as part of the ICU treatment. For
potential separation of factors, refer to section XII. A. Disturbances in Energy-Rich Phosphates
in Critical Illness
X. METABOLIC EFFECTS IN CRITICAL
ILLNESS The process of oxidative phosphorylation is crucial for ad-
equate production of ATP for energy available in the steady
Critical illness is hallmarked by striking endocrine and meta- state, but takes some time to respond to alterations in ATP
bolic disturbances, the severity of which has been associated demands. Phosphocreatine (PCr), on the other hand, serves
with a high risk of morbidity and mortality (730, 732, 784). as a rapidly mobilizable short-term storage of high-energy
phosphates in skeletal muscle. Several studies on critical
Already several decades ago the hypothesis was raised that illness point towards a disturbance in the levels of these
during sepsis and other types of critical illness, cellular en- high-energy phosphates in skeletal muscle.

1058 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

A H+ H+ H+ H+
Intermembrane space

Cyt c IV
I e–
Q •–
e–
e– e– II III

O2 H2O V
FADH2 FAD FIGURE 9. Mechanisms of compro-
NADH NAD+ mised energy production in muscle during
critical illness. A: normal mitochondrial re-
Matrix spiratory chain function. Complexes I–V are
ADP + Pi ATP
shown. NAD⫹ and NADH, nicotinamide di-
nucleotide, oxidized and reduced form, re-
spectively; FAD and FADH2, flavine ade-
B NOS nine dinucleotide, oxidized and reduced
form, respectively; ATP, adenosine triphos-
phate; ADP, adenosine diphosphate; Pi, in-
NO• organic phosphate. B: disturbed mitochon-
Reversible inhibition
drial function in critical illness. A vicious
cycle of increased free radical generation
resulting from enhanced nitric oxide syn-
Cyt c IV thase and from increased escape of elec-
I e– trons from the respiratory chain with for-
Q •–
e– mation of superoxide anion radicals revers-
e– e– II III ibly or irreversibly inhibits several
mitochondrial respiratory chain enzyme
complexes. Consequently, the supply of en-
O2 H2O V ergy-rich phosphates is compromised.

e–
Irreversible inhibition –
ONOO– O2• – e– e
ADP + Pi ATP
NO• e–

H2O2
PCr

Damage to proteins, lipids, ... OH•

In limb muscle of critically ill patients, especially with sepsis, a charge was not altered after 1–3 wk of sepsis in another ex-
low-energy charge potential with reductions in ATP, ADP, perimental study (475). Interestingly, patients with severe sep-
and/or PCr and a rise in AMP and free creatine has been sis or septic shock (probably CIM, but not assessed for) who
described in the presence of high lactate levels and an increased would not survive their illness had lower ATP levels, a smaller
lactate-to-pyruvate ratio (210, 425, 426, 444). In rats with total adenine pool, but higher levels of AMP in vastus lateralis
hemorrhagic shock, ATP, ADP, and PCr levels already muscle biopsies taken within 24 h after ICU admission than
dropped after 1 h in diaphragm (continuously working mus- patients who would survive (77). Compromised ATP synthe-
cle) and after 2 h in soleus muscle (more sedentary or resting sis was described in muscle of septic rats 6 days post CLP
muscle) (115). These reductions were less severe than those in (691). Also, burn injury in mice reduced the rate of ATP syn-
vital organs as liver and kidney. 31P magnetic resonance spec- thesis, but did not affect high-energy phosphate levels apart
troscopy studies in rats after CLP sepsis showed a decrease in from a reduction in PCr (534).
the PCr/Pi ratio as measure of energy stores after 24 h but not
in energy available for immediate use, as reflected in the
ATP/Pi ratio (348). The increased Na⫹-K⫹-ATPase activity, B. Muscle Oxygenation in Critical Illness
decreased PCr/ATP, increased PCr breakdown, but unaltered
ATP levels and pH in this model were interpreted as increased Disturbances in cellular energy metabolism and consequent
ATP utilization to help maintain the ionic balance and/or sup- bioenergetic failure in trauma and sepsis in humans were
port other metabolic processes, whereas PCr stores are used to originally ascribed to inadequate tissue perfusion leading to
buffer the ATP concentration (347, 489). Muscle energy cellular hypoxia and, thus, compromised availability of the

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1059


FRIEDRICH ET AL.

final electron acceptor in the respiratory chain (350). This tive mitochondrial changes have been observed in skeletal
notion was based on the high lactate levels observed in these muscle of patients and experimental animal models in con-
conditions which are traditionally attributed to anaerobic ditions of ischemia-reperfusion which may persist for days
glycolysis as the result of inadequate oxygen delivery. How- to weeks after the insult (644, 708).
ever, studies on tissue oxygenation yielded ambiguous re-
sults. In a rat endotoxemia model, oxygen tension in resting Evidence for a functional correlate of the mitochondrial
muscle was reduced after 4 h in the presence of a normal morphological damage with an intrinsic dysfunction of the
microcirculatory perfusion and remained refractory to in- organelles during critical illness continuously increases, al-
creasing inspired oxygen (605). A 6-h rat peritonitis model though apparently conflicting results have been obtained
of sepsis-induced myopathy showed reduced oxygen ten- depending on the species, model, and severity and duration
sion in rectus femoris muscle, an increased lactate-to-pyru- of the insult. Nevertheless, mitochondrial function appears
vate ratio, and reduced levels of ATP and total adenine mostly depressed in models of longer duration and greater
nucleotides (23). These accompanying abnormalities were severity of illness (640). The majority of studies focused on
still observed when normal tissue oxygenation was main- sole sepsis as severe insult which, therefore, will be the focus
tained. After 36 – 42 h, no evidence of cellular hypoxia was here. Function of the mitochondrial respiratory chain is
detected in skeletal muscle or diaphragm in septic rats with most commonly assessed via the activities of its individual
peritonitis, based on infusion of a hypoxia marker, al- enzyme complexes (FIGURE 9A) or via oxygen consumption
though circulating lactate levels increased (324). Patient studies, with evaluation of state 3 and state 4 mitochondrial
studies suggested that muscle oxygenation is even increased respiration as well as the respiratory control ratio or index
in fluid-resuscitated, hemodynamically stable sepsis and re- and the ADP/O ratio.
lated to disease severity, unlike in patients with limited in-
fection, cardiogenic shock, or after cardiopulmonary by- Very early after an acute insult, mitochondrial function may
pass (62, 63, 606). Altogether, these data suggest that cel- be enhanced, as illustrated by the increased activities of
lular hypoxia is not the major driving force of energetic complex I and citrate synthase in vastus lateralis biopsies
disturbances in critical illness. Importantly, elevated lactate taken 2 h (but not at 4 h) after an endotoxin challenge in
levels in hemodynamically stable patients may well be the human healthy volunteers (209). This may be in line with
consequence of stimulated aerobic glycolysis rather than of
increased cytosolic Ca2⫹ in sepsis (see previous sections on
tissue hypoxia (350).
Ca2⫹ homeostasis). Elevated cytosolic Ca2⫹ has been
shown to stimulate mitochondrial biogenesis (524). How-
C. Intrinsic Mitochondrial Abnormalities in ever, such Ca2⫹-induced stimulation of biogenesis may
Muscle During Critical Illness: strongly depend on the level and duration of increased Ca2⫹
Morphological and Functional (524). In several (mostly rat) models of sepsis (endotoxin
Mitochondrial Damage administration, peritonitis induced by CLP, or injection of
fecal slurry or live bacteria), no consistent results have been
Recent studies infer a disturbance in oxygen utilization obtained regarding mitochondrial function in limb muscle
rather than in oxygen delivery, leaving us with an acquired up to 18 –24 h, with either no effect on individual enzyme
intrinsic derangement in cellular energy metabolism, which activities, state 3 and state 4 respiration, respiratory control
has been identified as “cytopathic hypoxia” (198, 640). index, and ADP/O ratio or either marked loss of respiratory
Mitochondrial ultrastructural damage had been described control (78, 96, 235, 436, 620). Early partial uncoupling
for hindlimb skeletal muscle of endotoxemic rats already has been described in the diaphragm but was not observed
more than 40 years ago with frequent distortion of inner in limb skeletal muscle (61, 436). The time point of 24 h
and outer mitochondrial membranes and the presence of may be a borderline at which mitochondrial functional al-
large vacuolar areas (620). These alterations were clearly terations develop in diaphragm and limb skeletal muscle of
present after 18 h but not yet after 6 or 12 h. In a porcine endotoxin models, if the administered endotoxin dose, and
model of endotoxemia, mitochondrial swelling was ob- thus severity of illness, are sufficiently high (96, 97, 552, 672,
served in skeletal muscle at 18- and 48-h time points (293). 716). The decrease in complex I activity in skeletal muscle
A study of serial skeletal muscle biopsies taken after 12 and from rats with fecal peritonitis also depended on the severity of
24 h and at the time of death showed that bacteremia in the disease (78). Importantly, mitochondrial functional alter-
baboons led to mitochondrial swelling and even membrane ations in vastus lateralis biopsies taken within 24 h after ICU
fragmentation with advanced injury (638, 769). Less admission were more severe in patients who would succumb
densely packed cristae were also observed in diaphragm to severe sepsis/septic shock than those who would survive
mitochondria of 48 h endotoxemic rats (99). Mitochondria (77). Unlike with shorter time windows of illness, mitochon-
in biopsies of vastus lateralis or intercostal muscles taken drial function appears uniformly depressed with a prolonged
after 2–22 days from patients with sepsis-induced multiple duration of illness beyond 24 h (78, 96 –98, 210, 672). There
organ failure showed no differences in morphology com- was no uncoupling of the mitochondria beyond this time point
pared with elective surgery patients (210). Early degenera- (96, 97, 672).

1060 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

Sepsis appears to most consistently affect the activity of diaphragm and/or limb muscle after prolonged sepsis (96,
complex I, but also other enzyme complex activities have 97, 99, 691), which may be related to decreased gene ex-
been found to be reduced (78, 97, 442, 716). The impact of pression and rate of mitochondrial protein synthesis (98,
sepsis on mitochondrial function may also differ according 592) or increased degradation secondary to side-group
to the substrate that is used to stimulate mitochondrial res- modification. It also has been suggested that a reduction in
piration. In that regard, pyruvate- or octanoylcarnitine-de- intramitochondrial calcium levels during endotoxemia may
pendent respiration, but not succinate-dependent respira- contribute to metabolic derangement, specifically through
tion, appeared to be reduced in endotoxin-treated rabbits its impact on calcium-dependent intramitochondrial en-
after 24 h (716). One or two weeks of bacteremia evoked a zymes (358). Interestingly, plasma from patients with sepsis
decrease in pyruvate-dependent respiration, whereas contained increased levels of extracellular mitochondrial
branched-chain keto-acid and ketone body utilization si- DNA and inflammatory cytokines and tended to reduce
multaneously increased in rat muscle (475). oxygen consumption by skeletal muscle mitochondria from
healthy volunteers (232).
Apart from inhibition of electron flow along the mitochon-
drial respiratory chain or uncoupling of oxidative phosphor- Apart from sepsis, other severe insults have been shown to
ylation, disturbances in other metabolic processes may impact impair mitochondrial function in skeletal muscle, such as
on cellular ATP generation. These include altered activities of severe burn injury (553). This is illustrated by reduced mi-
glycolytic or tricarboxylic acid cycle enzymes, as illustrated by tochondrial respiration and activities of individual mito-
the reduced expression and activity of diaphragmatic phos- chondrial enzymes after severe burn trauma in children and
phofructokinase 36 – 48 h after endotoxin administration downregulated expression of several genes involved in gly-
(98). Also, impaired ATP synthase activity and hampered colysis, tricarboxylic acid cycle, and oxidative phosphory-
transport of ADP/ATP in and out of mitochondria may com- lation in burn-injured mice (135, 136, 534). Furthermore,
promise cellular ATP generation. The decreased maximal res- burn injury elicited early and prolonged activation of apo-
piration after addition of a chemical uncoupling compound ptosis which was preceded by a disturbance in mitochon-
was, however, interpreted as ruling out alterations in ATP drial membrane potential (24, 411, 792) and evoked an
early transient rise in the expression of the uncoupling pro-
synthase activity and transport of ADP or ATP across the inner
tein UCP3 (803).
mitochondrial membrane (97, 672). Nevertheless, the activity
and expression of the mitochondrial creatine kinase (MtCK)
Unlike the abundance of data available on mitochondrial
shuttle appeared dramatically reduced in rat diaphragm 48 h
function from critically ill septic patients and animal models
after endotoxin injection (99). In most tissues, the adenine
of sepsis, no studies involving animal models on critical
nucleotide transporter system is the most important way by
illness to dissect sepsis from mechanical ventilation and
which ATP is transported from the mitochondrial matrix side
immobilization or steroid-denervation have yet been pub-
to the intermembrane space. From there, ATP goes through
lished but are on their way. Profound structural mitochon-
the outer mitochondrial voltage-dependent anion channel into
drial changes were observed early in rats subjected to me-
the cytosol. However, in organs requiring extremely high rates
chanical ventilation and immobilization in the absence of
of transmitochondrial ATP transport, a mitochondrial crea- any other insults (i.e., sepsis). Severe changes developed in
tine kinase isoform (MtCK) constitutes the major ATP the diaphragm already after 6 h of mechanical ventilation
transport system, such as the sarcomeric MtCK in striated and after 18 h in a distal hindlimb muscle (soleus) (Shalah
muscle. As the MtCK is also an important structural protein and Larsson, unpublished observations). Likewise, the con-
that stabilizes mitochondrial membrane architecture by nection between cytoplasmic and mitochondrial Ca2⫹ on
cross-linking of the inner and outer mitochondrial mem- the one hand and biogenesis on the other hand has not been
branes at contact sites, the mitochondrial morphological addressed yet in critical illness or sepsis models. Adequate
alterations with less densely packed cristae may be related ICU animal models, such as outlined in section XII, thus
to the loss of MtCK (99). The cytosolic muscle type creatine represent an important strategy for future studies on mito-
kinase (CK-MM) which is responsible for delivery of high- chondrial dysfunction in CIM.
energy phosphates to the myosin ATPase was not affected
by sepsis (99).
D. Mitochondrial Dysfunction, Oxidative/
Several biophysical mechanisms may contribute to mito- Nitrosative Stress, and Impaired Muscle
chondrial dysfunction among which are disruption of mi- Force During Critical Illness
tochondrial membrane integrity, modification of protein
side-groups, increased degradation or reduced synthesis of Endotoxin treatment has been shown to decrease force gen-
the pathway components, and dissegregation of the multi- eration over the entire range of pCa tested, as well as max-
protein complexes. Selective depletion of several respira- imal calcium-activated muscle force (Fmax) of skinned myo-
tory chain subunits and other mitochondrial proteins, in- fibers of diaphragm and limb muscle (94, 682). These ob-
cluding multiple cytochromes, has been described in rat servations point to altered intrinsic functional properties of

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1061


FRIEDRICH ET AL.

the contractile proteins. Interestingly, decreased muscle vert superoxide to molecular oxygen and hydrogen
force coincided with a selective depletion of several proteins peroxide which in the presence of transition metal ions can
and appeared preventable by treatment with a NOS inhib- give rise to the extremely reactive hydroxyl radical. Both
itor or superoxide scavenger (94). Thus increased free rad- catalase and glutathione peroxidase are able to detoxify
ical generation appears to play an important role in the hydrogen peroxide. Importantly, the reaction of NO with
establishment of sepsis-related skeletal muscle dysfunction. superoxide yields peroxynitrite (ONOO⫺), a potent nitrat-
Free radicals may impair mitochondrial function, but recip- ing agent that irreversibly inhibits the four complexes of the
rocally, disturbances in mitochondrial function may also mitochondrial respiratory chain and the ATP synthase (FIG-
enhance free radical production. URE 9B), as well as several other mitochondrial enzymes
such as aconitase and the mitochondrial superoxide dismu-
Oxidative stress increases in skeletal muscle early after the tase (MnSOD), and also oxidizes membrane lipids.
onset of sepsis induced by CLP in rats (436). NOS activity
rises within a few hours after the insult in this model as well Elevated nitrotyrosine levels as a footprint of ONOO⫺ have
as in endotoxemia and is sustained for several hours or been shown in parallel with peak iNOS protein levels in
days, especially in diaphragm, but is less unequivocally diaphragm of endotoxemic rats (182). Progressive increases
demonstrated or occurring at a later time point in limb or in nitration of mitochondrial proteins preceded the decrease
abdominal skeletal muscle (60, 61, 182, 233, 427, 607). In in muscle force, which could be reversed by administration
most of these studies, the rise in total NOS activity is as- of a NOS inhibitor (61). In another study, tyrosine nitrosy-
cribed to the increased expression and activity of the induc- lation of some mitochondrial proteins preceded the selec-
ible NOS isoform (iNOS), but also increased levels of the tive depletion of several mitochondrial proteins and com-
eNOS and nNOS isoforms have been reported (182). A study promised mitochondrial function which all could be pre-
that combined the evaluation of iNOS, muscle histology, and vented or attenuated by administration of a NOS inhibitor
muscle function in rat diaphragm over time detected iNOS or superoxide scavenger (96). Selective reductions in some
protein and activity first in inflammatory cells 6 h after the but not all mitochondrial proteins, including components
endotoxemia challenge in the presence of normal histology of the electron transfer chain, have been observed simulta-
and muscle force and fatigability (60). At 12 h, histology was neously with disturbed mitochondrial function in other
still normal and iNOS protein and activity were detected, co- studies but may also be partly related to decreased mRNA
inciding with a decreased muscle force but not fatigability. At expression (97, 98, 99). The rise in nitrotyrosine levels and
24 h, the inflammatory cells had disappeared but iNOS was decrease in muscle force could also be blunted by adminis-
still found in the myocytes and muscle force remained com- tration of a NADPH oxidase inhibitor, suggesting involve-
promised. By 48 h, iNOS was no longer detectable and muscle ment of this enzyme as well (684).
force was restored. In acute endotoxemia and subacute peri-
tonitis models, increased iNOS and macrophage staining were Soon after induction of sepsis, mitochondrial production of
associated with sarcolemmal injury in diaphragm and/or limb superoxide radicals and hydrogen peroxide rises, associated
skeletal muscle which correlated with membrane depolariza- with increased (mitochondrial) NOS activity, elevated (in-
tion (427). Muscle injury and decreases in contractile force tramitochondrial) levels of NO, hydrogen peroxide, and
were (at least partially) restored or prevented when the rise in ONOO⫺ and nitration of mitochondrial proteins (16, 61,
iNOS expression was prevented with dexamethasone admin- 436, 506). These responses appeared preventable with NOS
istration or when iNOS activity was inhibited (60, 61, 94, 182, inhibition (61). Several studies have implicated increased
233, 427). Later studies demonstrated increased mitochon- production of superoxide, hydrogen peroxide, and/or hy-
drial NOS activity in diaphragm and limb skeletal muscle of droxyl radicals in the decreased muscle contractility of re-
endotoxemic rats which was ascribed to an inducible (mti- spiratory and limb muscles in endotoxemia, in parallel with
NOS) rather than constitutive isoform (mtcNOS) of NOS, markers of oxidative stress-induced macromolecular dam-
based on the absence of this response in iNOS⫺/⫺ mice (16, age (increased levels of the lipid peroxidation markers mal-
186, 443). ondialdehyde, 8-isoprostane, and of protein carbonyls) (61,
634, 672, 681, 683).
NO reversibly inhibits the activity of the mitochondrial re-
spiratory chain complex IV by competing with molecular While ROS production is increased, the antioxidant defense
oxygen (FIGURE 9B). NO can also induce reversible inhibi- mechanisms are compromised in sepsis. A few hours of
tion of complex I by thiol group nitrosylation and has been endotoxemia increased the diaphragmatic activity of the
shown to decrease complex III activity by impairing elec- antioxidant enzyme MnSOD but did not affect catalase
tron flow at the cytochrome bc1 level. The impairment of activity (16). The activities of MnSOD, catalase, and gluta-
the electron transfer chain leads to a decreased mitochon- thione peroxidase, as well as mitochondrial function, were
drial membrane potential, leakage of electrons, and forma- decreased in limb muscle 12 h after CLP (436). Simultane-
tion of more reactive oxygen/nitrogen species, at first in- ously with remarkable inhibition of mitochondrial respira-
stance the superoxide anion radical (O2·⫺). SODs can con- tory chain activity and ATP production, intramitochondrial

1062 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

oxidative stress was observed after 24 h in diaphragm and etal muscle (e.g., liver) where activation of mitochondrial
limb muscle of septic mice with a decreased ratio of oxi- biogenesis preceded restoration of mitochondrial mass and
dized (GSSG) over reduced (GSH) glutathione levels, re- oxidative metabolism and appeared an important pro-sur-
duced total glutathione levels, higher activity of the GSH vival factor (137, 282, 667). In vastus lateralis muscle bi-
consuming glutathione peroxidase, and lower activity of opsies harvested within 1– 42 days after ICU admission
the GSH regenerating enzyme glutathione reductase (186, from patients with sepsis-induced multiple organ failure,
442, 443). These disturbances did not develop in iNOS⫺/⫺ most of whom survived their illness, mitochondrial biogen-
mice. Also, administration of melatonin, which scavenges esis may have been partially activated as illustrated by some
reactive oxygen and nitrogen species, counteracted mtNOS increased mitochondrial transcription factors, but never-
induction and respiratory chain failure, and restored the theless failed to maintain mitochondrial function (212). In
redox status. The degree of the decrease in GSH levels in another clinical study, an early mitochondrial biogenesis
limb muscle of a fecal peritonitis model was shown to de- response (within 1–2 days after ICU admission) was ob-
pend on the severity of illness (78). Interestingly, complex I served in muscle from surviving but not from nonsurviving
activity correlated inversely with NO production and di- patients with severe sepsis (105). This activation was
rectly with levels of GSH and ATP. Also, in skeletal muscle mainly seen upstream in the respective pathway and was
of severely burned rats, a remarkable decrease in mitochon- not accompanied by a clear difference in mitochondrial re-
drial GSH was observed (456). spiratory chain proteins. In a heterogeneous group of criti-
cally ill patients, upstream activation of the mitochondrial
biogenesis program was observed in postmortem biopsies
E. Mitochondrial Repair Mechanisms in
of rectus abdominis, but not in in vivo biopsies of vastus
Critical Illness lateralis taken after 2 wk in ICU, and this was irrespective of
future survival status (FIGURE 10) (727). In muscle taken
Persistent mitochondrial damage can be evoked by sus- within 10 days from children with severe burn injury,
tained direct mitochondrial damage and/or impaired or in- downregulated mitochondrial biogenesis has been sug-
sufficient activation of the mitochondrial repair mecha- gested (723). In experimentally denervated gastrocnemius
nisms. Damaged organelles in turn can perpetuate further muscle of rats and mice (a model of disuse), strong reduc-
damage, e.g., through production of more ROS. Several
tions in mitochondrial content coincided with a remarkable
repair mechanisms cooperate to remove or compensate for
downregulation of several key players of mitochondrial
mitochondrial damage. Mitochondria undergo continuous
biogenesis several weeks after the insult (5, 750). Several
cycles of fusion and fission. Mitochondrial fusion and fis-
components of the mitochondrial biogenesis pathway ap-
sion not only determine mitochondrial size and distribution
peared downregulated at gene expression level after 24 h of
in the cell but are also involved in the repair of mitochon-
endotoxemia, which was more pronounced in mouse tibia-
dria (116, 700). These processes allow the exchange of
lis anterior and soleus muscle than in diaphragm (490). On
damaged mitochondrial components and either dilution of
the other hand, freeze injury of murine gastrocnemius mus-
molecular damage over the daughter organelles or bundling
cle (a model of degeneration and regeneration) suggested
of dysfunctional structures by asymmetric fission into a sin-
that mitochondrial biogenesis plays a role in muscle regen-
gle, irreversibly damaged depolarized organelle, which is
eration because this pathway appeared activated early dur-
fusion-incompetent and is subsequently targeted for re-
ing muscle regeneration (751).
moval by mitochondrial autophagy or “mitophagy” (365,
700, 721). Autophagy is a crucial cellular quality control
mechanism to clear damaged organelles and potentially 2. Mitochondrial fusion and fission in critical illness
toxic protein aggregates (295, 379). Mitochondrial fusion
and fission are also important for mitochondrial biogenesis. Data on the impact of critical illness on mitochondrial fu-
This pathway generates new mitochondria when needed, sion and fission hardly exist. Both processes need to be
such as in response to mitochondrial damage and increased appropriately balanced, since both unopposed fusion and
energy demand (282, 615). Under normal physiological unopposed fission are detrimental for cellular function. The
conditions, the three processes are nicely balanced. FIGURE only study on mitochondrial fusion and fission in skeletal
10 shows a simplified presentation of these processes and muscle during critical illness found that key players in these
their interactions. Compared with mitochondrial damage, processes were upregulated in postmortem rectus abdomi-
much less attention has been paid to the mitochondrial nis biopsies, but not in in vivo vastus lateralis biopsies of a
repair mechanisms in critical illness. Actually, interest in heterogeneous group of critically ill patients (FIGURE 10)
these pathways is only recently emerging. (727). There was no relationship with eventual survival,
and patients were not investigated to classify for CIM. In a
1. Mitochondrial biogenesis in critical illness rabbit model of critical illness induced by severe burn in-
jury, key players of mitochondrial fusion and fission in liver
Initially, studies on mitochondrial repair rather focused on and kidney were also not significantly different between
mitochondrial biogenesis, mostly in tissues other than skel- survivors and nonsurvivors (277).

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1063


FRIEDRICH ET AL.

3. Autophagy in critical illness and development of a phenotype of myopathy (465). Myo-


fibers in skeletal muscle of these mice were smaller than in
An adequately balanced activation of autophagy appears wild-type mice and showed degenerative changes, including
crucial for maintenance of normal muscle homeostasis. Ini- vacuolization and central nuclei, damaged mitochondria,
tial studies on autophagy in conditions of muscle atrophy and accumulation of aberrant concentric membranous
implicated excessive autophagy in muscle breakdown, in structures. The lipidation of microtubule-associated protein
conjunction with the activation of the ubiquitin-protea- light chain-3 (LC3) with formation of LC3-II from LC3-I,
some system (42, 451, 756, 805). This conclusion was which is required for mature autophagosome formation,
based on the upregulation of several autophagy-related or was reduced. These abnormalities coincided with the accu-
-regulating genes in different atrophy models. However, mulation of p62 protein and aggregates positive for p62
selective genetic inactivation of autophagy in skeletal mus- and ubiquitin, which are normally cleared by autophagy. As
cle of mice resulted in a spontaneous induction of atrophy a functional correlate, muscle force was remarkably re-

Mitochondrial biogenesis
Post-mortem R. abdominis
in vivo V. lateralis PGC-1α
Diaphragm
RIP-140
NRF-1 NRF-2
Mitochondrial fusion and fission

Mitofusin TFAM Pol-γ mtSSB


mtDNA Replication/transcription
mtDNA copy number
Enzyme complex subunits
OPA1

Fis1 +
Drp1
Low ΔΨm
Low OPA1
Fusion-incompetent
Fis1 +
Drp1 Intact ΔΨm
Normal OPA1
Fusion-competent
Initiation
Autophagy
Phagophore Organelle,
(isolation cytoplasm
membrane)

Insufficiant activation:
Autophagy substrates
Upstream components
LC3-II/LC-3-I
Degenerative
myofiber changes Elongation

Autophagosome

Lysosome
Maturation

Autolysosome

1064 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

duced. Furthermore, inhibition of autophagy unexpectedly was not investigated in this study. In contrast, alterations in
promoted more severe muscle loss instead of protecting limb muscle were more pronounced than those in dia-
against atrophy in several experimental stress models of phragm of endotoxemic mice (490).
atrophy, including denervation and fasting (465). Hence, it
is clear that autophagy is required to preserve muscle mass, Activation of autophagy suggested by the scarce studies on
myofiber integrity, and function. This is in line with the this pathway in critical illness has been interpreted as a
crucial role of autophagy in cellular protein quality control, detrimental response which contributes to hypercatabolism
by removal of toxic proteins, aggregates, and dysfunctional and wasting. However, the hypothesis that autophagy
organelles that otherwise would amplify the damage. Over- could be protective for skeletal muscle and other organs
all, it remains controversial whether activation of au- during critical illness had initially been overlooked but war-
tophagy is actually detrimental in catabolic conditions by rants consideration in light of the lessons from mice with
contributing to atrophy or whether it is rather beneficial by tissue-selective autophagy (372, 465). In fact, the initial
clearance of damage and promotion of survival (612). studies did not investigate whether autophagy is sufficiently
activated to cope with the severe damage inflicted by critical
This controversy also holds true for critical illness and as- illness. However, a clinical study that considered conse-
sociated muscle pathology, in particular. Upregulated gene quences of insufficient autophagy confirmed similar
and/or protein expression of several components of the au- changes in skeletal muscle and liver of critically ill patients
tophagy-lysosome system and/or accumulation of au- as observed in autophagy-deficient mice (FIGURE 10) (154,
tophagic vacuoles has been observed in skeletal muscle of 728). These included the accumulation of ubiquitin aggre-
experimental models of acute and prolonged critical illness gates and other autophagy substrates, such as p62, de-
(55, 322, 434, 490, 616, 690). Muscle disuse may be an formed mitochondria, and aberrant concentric membra-
important factor since multiple (though not all) studies on nous structures. Likewise, an autophagy deficiency pheno-
denervation-induced atrophy (model of chronic muscle dis- type has been observed in skeletal muscle, liver, and kidney
use) also found such alterations, with one study addition- of a severe burn injury model of critical illness (155, 277).
ally showing an increase in autophagic flux (356, 525, 526, Signs of insufficient autophagy in muscle were also ob-
715). However, different models of disuse have suggested served several days after severe insults such as sepsis and
differential involvement of either the ubiquitin-proteasome denervation (31, 565). Fasting is the strongest physiological
system or autophagy in the associated atrophy (58). A hu- activator of autophagy, whereas feeding and insulin inhibit
man patient study suggested that diaphragm disuse induced this pathway (247). As in the latter studies patients and
by prolonged mechanical ventilation triggers autophagy, animals were artificially and continuously fed, this may in-
based again on the increased expression of several key play- dicate that early provision of nutrients during critical illness
ers in autophagy and an increased number of double-mem- may impair autophagy activation, thereby reducing damage
brane autophagosome vesicles (335). Unlike the dia- removal that is required for recovery. This constellation is
phragm, quadriceps muscle of these patients showed mostly supported by a study in critically ill rabbits demonstrating
no changes or even a decrease in these components in re- that early parenteral nutrition (especially when enriched in
sponse to mechanical ventilation. The development of CIM amino acids) reduced muscle catabolism at the expense of

FIGURE 10. Alterations in the mitochondrial repair mechanisms in muscle during critical illness. Mitochondrial repair mechanisms include
three major processes: mitochondrial fusion and fission, removal of damaged mitochondria by autophagy (mitophagy), and generation of new
mitochondria via mitochondrial biogenesis. Mitochondrial fusion and fission are important in determining the overall shape of these organelles,
have the potential to dilute damage over different organelles, and are also involved in mitochondrial biogenesis and autophagy. Mitofusins and
optic atrophy-1 (OPA1) are important in mitochondrial fusion, whereas dynamin-related protein-1 (Drp1) and Fission-1 (Fis1) are required for
mitochondrial fission. Asymmetric fission can generate uneven daughter organelles and, thus, can segregate the daughter unit that contains
most of the defects. Due to low membrane potential ⌬⌿m and low OPA1 content, this daughter organelle will have an impaired fusion capacity,
hence predisposing the organelle to removal by autophagy. Autophagy is a multiphase process by which portions of cytoplasm and/or organelles
are degraded, and this can be either selective or nonselective. In the initiation phase, a phagophore or isolation membrane is formed, which
elongates in the elongation phase to surround the material that is to be degraded. This phase ends with the formation of a double-membrane
structure, the autophagosome, which in the maturation phase fuses with a lysosome to form an autolysosome. This structure contains all the
enzymes that are needed to degrade the sequestered content. Microtubule-associated protein-1 light chain-3 (LC3) plays a central role in the
autophagy process. LC3 needs to be converted from its cytosolic precursor form LC3-I to its active mature form LC3-II by lipidation. LC3-II is
translocated to the growing autophagosomal membrane where it probably functions as a scaffold protein that supports membrane expansion
and is used as an autophagosomal marker. The LC3-II/LC3-I ratio is often used as a marker for autophagosome formation. Mitochondrial
biogenesis is controlled by both nuclear and mitochondrial gene expression. Receptor inhibitory protein-140 (RIP-140) is an inhibitor of this
process. In contrast, peroxisome proliferator-activated receptor-gamma coactivator-1␣ (PGC-1␣) stimulates mitochondrial biogenesis via
regulation of the nuclear respiratory factors NRF-1 and NRF-2. The NRFs control mitochondrial transcription factor-A (TFAM), mitochondrial
DNA polymerase-␥ (Pol-␥), and single-strand binding protein (SSB), which in turn stimulate mtDNA replication and transcription of several
mtDNA-encoded respiratory chain complex subunits. The NRFs also regulate other proteins, including several subunits of the respiratory chain
complexes that are encoded by the nuclear DNA. The colored signs indicate the reported impact of critical illness on these pathways in different
types of skeletal muscle during critical illness (increases indicated by the arrows, equal signs indicate no change).

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1065


FRIEDRICH ET AL.

suppressed autophagy, thus compromising myofiber integ- including the quality of glucose control likely played a major
rity as well as vital organ function compared with fasting role in the different outcomes (734). In fact, the discussion
(155). This was confirmed in a recent RCT (300), and this appears not to be about the need for glucose control but rather
may also explain the delay in recovery observed with early about which is the optimal level of glycemia to target for.
versus late initiation of parenteral nutrition to supplement
insufficient enteral nutrition in critically ill patients (107, Maintaining strict normoglycemia with insulin infusion
109, 300). Indeed, muscle biopsies of the patients showed a during critical illness in relation to a disturbed cellular en-
suppression of autophagy activation with early compared ergy metabolism was investigated in patients and animal
with late initiation of parenteral nutrition, in the absence of models. The intervention protected the endothelium in pa-
any effect on muscle atrophy markers (300). Importantly, tients where lowering of adhesion molecules prevented or-
the impact on autophagy was independently associated gan failure and death (393). In critically ill rabbits, glycemic
with a higher risk of clinically relevant muscle weakness. control improved endothelium-mediated relaxation of aor-
Adequate autophagy activation clearly appeared crucial to tic rings, but had no effect on tissue perfusion and oxygen
confer protection against mitochondrial dysfunction as well delivery to different organs, including skeletal muscle (183,
as cardiac dysfunction, hepatic injury, and kidney failure in 726, 729). Strict glycemic control with insulin protected
animal models of critical illness as shown by genetic inter- against severe mitochondrial morphological abnormalities
ference or pharmacological activation of autophagy (103, seen in liver from hyperglycemic critically ill patients (725).
277, 327). The importance of autophagy for muscle is fur- It also improved the activity of several mitochondrial respi-
ther underscored by studies in mouse models of muscular ratory chain enzymes. Mitochondrial protection in liver,
dystrophy. Skeletal muscles of Col6a1⫺/⫺ mice showed im- kidney, and myocardium was attributed to glucose control
paired activation of autophagy, but myofiber survival could rather than direct insulin effects, as shown in critically ill
be restored and the dystrophic phenotype could be amelio- rabbits (726, 729). Intriguingly, the intervention had no
rated by forced activation of autophagy with genetic, di- impact on the mitochondria in skeletal muscle of critically
etary, and pharmacological interventions (271). Potent ac- ill patients or rabbits (725, 726). Nevertheless, it attenuated
tivation of autophagy also restored the sensitivity of mito- iNOS expression in muscle and prevented excessive NO
chondria to calcium-induced permeability transition pore production (184, 725). The different mechanisms responsi-
opening in diaphragm of mdx mice, associated with im-
ble for glucose uptake in these organs were put forward as
proved histopathology and force-generating capacity (540).
a potential explanation for the observed organ specificity of
the mitochondrial effects. As such, cellular glucose overload
In summary, a critical balance between autophagy activa-
may develop in organs with insulin-independent glucose
tion and suppression is important for the maintenance of
transporters, whereas tissues that rely predominantly on
muscle mass, but more importantly, muscle quality. Most
insulin-dependent glucose uptake may be relatively well
studies (human and animal models) show insufficient au-
protected from glucose overload in view of the development
tophagy activation in critical illness, but the level of sup-
of insulin resistance. However, strict glycemic control did
pression of autophagy activation required to contribute to
improve skeletal muscle mitochondrial function in children
CIM is yet unknown. Also, whether impaired autophagy
with severe burn injury (207). Apart from any impact on the
will be considered as a diagnostic requirement for CIM is a
topic for future studies. mitochondrial compartment, strict glycemic control with
insulin may also affect the muscle by anticatabolic actions
in view of the catabolism-promoting property of hypergly-
F. Glucose Toxicity During Critical Illness cemia and anabolic properties of insulin (205, 261, 262). In
critically ill rabbits, increasing the amount of intravenously
The development of hyperglycemia as a consequence of insulin infused glucose calories was able to dose-dependently atten-
resistance and increased hepatic glucose production is a com- uate several proteolytic responses but only when hypergly-
mon complication of critical illness that has been associated cemia was simultaneously prevented (156). In the same
with morbidity and adverse outcome of patients irrespective of model, prevention of hyperglycemia better preserved au-
the underlying diagnosis that required admission to the ICU tophagy in liver and kidney which correlated with improved
(470). In three large randomized clinical studies performed in mitochondrial function and less organ damage (277). The
surgical, medical, and pediatric ICUs of Leuven, insulin infu- impact of glycemic control on autophagy in skeletal muscle
sion to the strict age-adjusted target of normal fasting blood has not been investigated yet. Several studies support pro-
glucose levels during the ICU stay improved survival and re- tection of the central and peripheral nervous system during
duced morbidity (736, 737, 738, 748). Some studies of other critical illness. Maintenance of normoglycemia during crit-
investigators have provided support for the beneficial effects of ical illness lowered intracranial pressure and seizures in pa-
this intervention, whereas others did not show any benefit or tients with isolated brain injury and reduced the incidence
even suggested harm (197, 731, 732). Discussing the contro- of critical illness polyneuropathy (309, 735, 736). Nor-
versy surrounding the efficacy and safety of this intervention is moglycemia also improved neurocognitive development of
beyond the scope of this review, but methodological issues critically ill children 4 yr after ICU admission (482). Fur-

1066 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

thermore, strict blood glucose control during critical illness, tein metabolism is outlined below. A central difference to
independently of glucose load, attenuated the neuropatho- pure atrophy usually seen in bedrest conditions, micrograv-
logical alterations at the level of neurons, astrocytes, and ity, or muscle unloading and sepsis-induced myopathy is
microglia in vulnerable areas of the brain as shown in hu- that critical illness myopathy is characterized by a preferen-
man and animal brain specimens (649, 650). The impact of tial myosinolysis that exceeds the mechanism of atrophy
this therapy on peripheral nerves remains to be investigated. (balanced proteolysis of sarcomeric proteins) by far and is
responsible for muscle weakness in CIM that is more severe
compared with sepsis alone. This is also highlighted in sec-
XI. PROTEIN REGULATION IN MUSCLE IN tion XII.
CRITICAL ILLNESS

Critical illness is commonly associated with loss of muscle A. The Ubiquitin-Proteasome Pathway in
protein and decrements in skeletal muscle mass (FIGURE Critical Illness
11A). The resulting muscle atrophy may contribute to
weakness, premature fatigue, and glucose intolerance (561) The ubiquitin-proteasome pathway is a major mechanism
and has been associated with morbidity and mortality in of regulated proteolysis. As reviewed elsewhere (498),
critically ill patients (10, 102, 147). At the cellular level, structural and regulatory proteins are selectively targeted
reductions in myofiber size reflect an imbalance between for degradation by covalent attachment of polyubiquitin
proteolysis and protein synthesis (FIGURE 12). This is regu- chains to lysine residues. This ATP-dependent process re-
lated by complex changes in signaling pathways and gene quires sequential interaction among three enzyme families:
products that regulate protein breakdown and accretion ubiquitin activating enzymes (E1 proteins), ubiquitin con-
(353, 575). The influence of critical illness on muscle pro- jugating enzymes (E2 proteins), and ubiquitin ligases (E3

A B C
Control

Patient

0.50
Respiratory Leg
pmol/μg protein x min

20S proteasome
0.40

0.30 MAFbx

0.20
MuRF1
0.10
Actin
0.00
Sepsis* Control Sepsis* Control

D E F Control
Tyrosine Release (nmol/g x 2h)

500 600 BN 82270 2000


0.35
Sepsis Control
Calpain activity (FU)

*
pmol/μg protein x min

400 500
Tyrosine Release
(nmol/g ww x 2h)

0.30
* 1500
0.25 400
300 *
0.20
300 *,+ 1000 *,+
0.15 200 * * +
200 +
0.10 500
100
0.05 100

0.00 0 0 0
Left* Right* Left* Right*

FIGURE 11. Critical illness and skeletal muscle proteolysis. A: primary data from critically ill patients
document decreased nitrogen balance and an increased rate of muscle protein breakdown in individuals with
sepsis; the rate of muscle protein synthesis was not different between septic and nonseptic individuals. [From
Klaude et al. (366).] B: Western blot analyses show higher levels of E3 proteins (MuRF1, MAFbx) and 20S
proteasome subunits in muscle biopsies from critically ill patients. [From Constantin et al. (130). Copyright
John Wiley and Sons.] C: immunocytochemical staining illustrates greater ubiquitin density in small vs. large
muscle fibers of ICU patients, consistent with fiber atrophy via the ubiquitin-proteasome pathway. [From
Helliwell et al. (298). Copyright John Wiley and Sons.] D: enzymatic assay data demonstrate greater 20S
proteasome activity in muscle of septic individuals. [From Klaude et al. (366).] E: proteasome inhibition lessens
degradation of muscle protein in the rat CLP model of sepsis. [From Hobler et al. (315).] F: calpain activity and
the rate of protein degradation are elevated in limb muscles of septic rats; proteolysis is partially inhibited by
pharmacologic calpain blockade. [From Fareed et al. (191).]

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1067


FRIEDRICH ET AL.

Protein synthesis Sepsis, inflammation, cytokines Protein degradation

Altered Increased Ubiquitin


Blunted anabolic Compensatory Disrupted
transcriptional Ca2+-dependent proteasome
signaling protein synthesis autophagy
regulation proteolysis activation

Kinase Pro-catabolic Amino acids Proteolytic Debris


Ubiquitination
deactivation signaling cleavage accumulation

Anabolic
ATP U ADP
tRNA Ca2+
pathway
Transcription E2, E3
U
PI3K factors U
U
MAPK U
DNA
NF-κB
AKT Muscle genes Protein
FOXO Proteasome
HDAC Proteasome
mRNA activation
GSK
Ribosome
Myofibrillar
mRNA
Polypeptides
mTOR
Amino acids
p70S6K S6
4E-BP1 eIF-4E Autophagosome

FIGURE 12. Muscle protein homeostasis in critical illness. Diagram depicts changes to protein metabolism
caused by pathophysiological processes in critical illness including sepsis, inflammation, and prolonged expo-
sure to pro-inflammatory cytokines. Upper light grey boxes denote altered regulation of major steps in protein
synthesis (left) and degradation (right). Blue boxes identify the downstream effects of altered pathway regula-
tion. PI3K, phosphatidylinositol-4,5-bisphosphate 3-kinase; AKT, also known as protein kinase B; GSK, glyco-
gen synthase kinase; mTOR, mechanistic target of rapamycin; p70S6K, p70S6 kinase; S6, ribosomal protein
S6; elF-4E, eukaryotic translation initiation factor-4E; 4E-BP1, elF-4E binding protein-1; MAPK, mitogen-
activated protein kinase; NF-␬B, nuclear factor-␬B; FOXO, forkhead box-O; HDAC, histone deacetylase; DNA,
deoxyribonucleic acid; RNA, ribonucleic acid; mRNA, messenger RNA; tRNA, transfer RNA; Ca2⫹, calcium
ions; E2, ubiquitin-conjugating enzyme; E3, ubiquitin ligase; U, ubiquitin; ATP, adenosine triphosphate; ADP,
adenosine diphosphate.

protein). E2/E3 protein pairs regulate ubiquitin attachment recent study in 63 critically ill ICU patients that demon-
to protein substrates. The composition of each E2/E3 pair strated increased catabolism in vastus lateralis muscle
confers substrate specificity, enabling selective tagging for showed the opposite: a downregulation of MuRF1 and
degradation via the 26S-proteasome complex. The 26S atrogin-1 (563). Helliwell et al. (298) found fiber atrophy
complex is composed of a 20S multienzyme core capped at and reductions in myosin ATPase activity in tibialis anterior
each end by two 19S regulatory subunits. The 19S subunits muscle biopsies from critically ill patients that were accom-
identify, bind, and unfold ubiquitinated proteins, cleaving panied by an increase in immunocytochemical staining of
and recycling the attached ubiquitin moieties. Peptidases ubiquitin conjugates (FIGURE 11C). At the functional level,
within the 20S core then degrade the unfolded protein to Klaude et al. (367) have shown that critical illness increases
generate small polypeptides. chymotrypsin-like peptidase activity of membrane-associ-
ated proteasomes isolated from human leg muscles. The
Critical illness appears to upregulate key components of the activity of membrane-associated proteasomes was elevated
ubiquitin-proteasome pathway in skeletal muscle. Constan- by 30% relative to healthy controls, whereas proteasome
tin et al. (130) found that muscle-specific E3 proteins activity in the soluble fraction was unaffected by critical
(MuRF1, MAFbx) and subunits of the proteasome were illness. A subsequent study by the same group (366) con-
elevated in vastus lateralis muscle of critically ill patients firmed and extended these findings in patients with sepsis in
(assessed by APACHE II scores, no information on mechan- the ICU. Proteasome activity was elevated by 45–55% in leg
ical ventilation given). These changes were evident at both muscle biopsies from septic patients (FIGURE 11D). Interest-
the mRNA and protein levels, arguing for their physiologi- ingly, proteasome activity was also elevated by 30% in
cal relevance (FIGURE 11B). On the other hand, another serratus anterior, a muscle of the rib cage. In rectus abdomi-

1068 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

nis muscle, a more than twofold increase in chymotrypsin- a rise in mRNA for E2-14k, an E2 protein that mediates
like activity of the 20S-proteasome has been described ubiquitin conjugation via the N-end rule pathway. Potential
(154). Such functional readouts are certainly more informa- involvement of E2-14k was reinforced by data from Hobler
tive and preferable over simple transcript or protein content et al. (316) who observed that E2-14k mRNA was elevated
analyses which may not account for the temporary nature in fast-twitch (but not slow-twitch) muscle of septic rats.
of mRNA levels, which can be as short-lived and fall Later studies confirmed upregulation of E2-14k in fast-
quickly during prolonged muscle proteolysis (409). twitch muscles of rats subjected to cecal ligation and punc-
ture (200) or endotoxemia (113). Fischer et al. (201) later
Rodent models of sepsis have confirmed observations in found that sepsis also increased mRNA levels for E3␣, an
humans and provided greater detail on the cellular and E3 ligase that partners with E2-14k. The importance of E3␣
molecular mechanisms by which the ubiquitin-proteasome is reinforced by the finding that elevated rates of ubiquitin
pathway is activated. Tiao et al. (704) first reported that conjugation can be suppressed by selectively inhibiting E3␣
sepsis stimulates nonlysosomal, energy-dependent (i.e., function (648). In aggregate, these findings strongly argue
proteasomal) proteolysis in skeletal muscles of rats as a for an involvement of the N-end rule pathway. Other E3
model for sepsis-induced myopathy. The process was proteins that regulate muscle proteolysis are also upregu-
shown to affect total and myofibrillar protein levels and to lated during sepsis. For example, mRNA for MuRF1 and
be associated with elevated ubiquitin mRNA. Subsequent atrogin-1/MAFbx were both elevated in extensor digitorum
work has established that the response is greater in fast- longus muscles of septic rats (786). Substrates of MuRF1
than in slow-twitch muscle (705) and that sepsis-stimulated include actin and the myosin heavy chains. Only MyoD and
proteolysis can be blocked using proteasome inhibitors, IF3-f are currently known substrates for atrogin-1 (25).
both in isolated muscle preparations (315) and intact ani- Both are involved in the control of protein synthesis, thus
mals (200) (FIGURE 11E). suggesting that atrogin-1 affects protein synthesis rather
than proteolysis. Such increases in MuRF1 and atrogin-1
Experimental sepsis appears to increase proteasomal activ- can be inhibited by pretreating septic animals with gluco-
ity, at least in part, via endogenous glucocorticoids. Tiao et corticoid receptor antagonists, either RU 38486 (786) or
al. (703) treated septic rats with RU 38486, a glucocorticoid RU 486 (222), identifying these E3 proteins as glucocorti-
coid-sensitive components of the sepsis response. Down-
receptor antagonist that inhibited the rise in total and myo-
stream events that appear to upregulate atrogin-1/MAFbx
fibrillar proteolysis and blunted increases in ubiquitin
include increased signaling via stress-activated protein ki-
mRNA, free ubiquitin, and ubiquitin conjugates. Con-
nases (123) and FOXO1 (646), complemented by less sig-
versely, dexamethasone administration to normal rats had
naling via PGC-1␤ (479) and HDAC (9). Alterations in
the reverse effects. The authors concluded that “ѧglucocor-
FOXO1 and PGC-1␤ signaling also appear to stimulate
ticoids regulate the energy-ubiquitin-dependent proteolytic
MuRF1 expression (479, 646).
pathway in skeletal muscle during sepsis.” Pro-inflamma-
tory cytokines have also been evaluated as downstream medi-
Proteasomal regulation is also altered by experimental sep-
ators. There was no evidence that IL-1␤ or IL-6 play a signif-
sis. Voisin et al. (749) demonstrated that mRNA for sub-
icant role in sepsis-associated muscle dysfunction (228, 775). units of the proteasome fluctuate in parallel with muscle
In contrast, Garcia-Martinez and co-workers found that in- proteolysis rates. Proteolytic activity of the proteasome is
creases in ubiquitin mRNA stimulated by sepsis could be rep- elevated by experimental sepsis (316), a response that in-
licated in healthy rats by systemic administration of recombi- volves both trypsin- and chymotrypsin-like peptidase activ-
nant TNF-␣ (227), and that this stimulus increases ubiquitina- ities (486).
tion of skeletal muscle protein (226). This suggested that the
elevated TNF-␣ levels observed in sepsis might be responsible
for ubiquitin upregulation. Schakman et al. (616) have evalu- B. Calpain Regulation in Critical Illness
ated the relative importance of glucocorticoids and TNF-␣
after systemic endotoxin administration. They found that the Calpains comprise a family of calcium-dependent, nonlyso-
glucocorticoid receptor antagonist RU 486 abolished activa- somal proteases that are constitutively expressed in skeletal
tion of the ubiquitin-proteasome pathway, whereas inhibitors muscle. Over two decades ago, Bhattacharyya et al. (56)
of TNF-␣ production and NF-␬B activation had no effect. reported that sepsis increases calcium-dependent calpain
Thus, in this LPS sepsis model, glucocorticoid production ap- activity in rat limb muscles. Subsequent rodent studies have
pears to play a more important role than TNF-␣/NF-␬B sig- generally confirmed this finding (FIGURE 11F). Greater cal-
naling. pain activity leads to Z-band disintegration and myofibril-
lar protein breakdown (202, 776), thereby depressing myo-
Experimental sepsis increases expression of gene products fibrillar force (674). Calpain inhibitors lessen sepsis effects
that regulate the ubiquitin-proteasome pathway. This is on muscle, reducing proteolysis (191, 767) and preserving
clearly true for regulatory proteins that control ubiquitin contractile function (675, 685). The mechanism by which
conjugation. In septic rats, Voisin et al. (749) first observed sepsis increases calpain activity is less clear. Tissue levels of

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1069


FRIEDRICH ET AL.

active calpain protein appear to be elevated (675, 685). This the rate of protein degradation altered by Ac-DEVD-CHO,
may reflect greater calpain activation by internal calcium a caspase-3 inhibitor. These investigators concluded that
stores since sepsis effects are opposed by dantrolene (202). caspase-3 was not involved in the catabolic response to
Several studies have detected increases in mRNA for m- CLP. However, later work showed that CLP increased
calpain, mu-calpain, and p94, a calpain thought to be mus- caspase-3 activity and active caspase-3 protein in rat dia-
cle specific (156, 202, 749, 776). Mu-calpain requires about phragm (685). CLP also decreased the specific force of dia-
10- to 100-fold lower Ca2⫹ concentrations for its half-max- phragm muscle fibers; this response was blunted by pre-
imum activation (in the low micromolar range) compared treating animals with zVAD-fmk, a caspase inhibitor. Sim-
with m-calpain, with the former, thus, being activated in the ilarly, endotoxin administration activates caspase-3 and
range achieved during mild to strenuous exercise and the increases procaspase-3 levels in porcine muscle (530). Mus-
latter in the range exceeded by normal muscle function cle caspase-3 mRNA levels were elevated in a rat ICU model
(hundreds of micro- to low millimolar range) (39). While of critical illness myopathy, but appeared relatively late and
muscle can usually fully recover from activation of mu-
were preceded by both the upregulation of the E2 ligases
calpain after strenuous exercise with a period of relative
(MuRF1 and atrogin-1) and the preferential myosin loss
muscle weakness within a few days, global activation of
(434). However, total caspase activity was unaltered in
m-calpains will more likely result in irreversible muscle
muscles from critically ill patients (368).
damage and cell death. Finally, apart from activation by
Ca2⫹, the rise in calpain activity may reflect lower activity
of calpastatin, the endogenous inhibitor of calpain (767). Caspase-8 has been identified as a key mediator of dia-
phragm weakness in endotoxin-treated mice. Supinski et al.
The importance of calpain activation in muscle of critically (679) originally reported that caspase-8 is activated in
ill patients is largely unstudied. To our knowledge, a recent C2C12 myotubes exposed to pro-inflammatory cytokines
report by Klaude et al. (368) on eight critically ill patients and in diaphragm of endotoxin-treated mice. Endotoxin
and seven healthy controls provides first information on also activated caspase-3 and depressed specific force of the
this topic. Patients had higher rates of protein degradation diaphragm. The latter responses were blocked by pretreat-
and higher activities of proteasomal and lysosomal path- ing animals with a caspase-8 inhibitor, identifying
ways. Calpain mRNA levels were elevated in muscle but caspase-8 as an upstream mediator of caspase-3 activation
calpain activity was not. These initial data provide very and diaphragm weakness. Subsequent studies by the same
limited information on calpain involvement and illustrate group have identified JNK, p38 MAPK, and double-
the need for more translational research on this topic. stranded RNA-dependent protein kinase (PKR) as up-
stream regulators of caspase-8 activation (674, 676, 680).
So far, evidence from experimental sepsis animal models These investigators proposed that pro-inflammatory cyto-
has not been able to point towards a significantly preferen- kines, such as TNF-␣, stimulate a postreceptor signaling
tial myosin loss, but only a general breakdown of myofi- cascade in which PKR is upstream of p38 MAPK, JNK, and
brillar proteins in septic muscle, i.e., similar proportions for other kinases. This cascade triggers cleavage of pro-
actin and myosin (776). This distinguishes the sepsis-in- caspase-8 to caspase-8. In turn, caspase-8 acts on pro-
duced myopathy from the CIM phenotyope seen in criti- caspase-3 to generate caspase-3 which degrades myofibril-
cally ill patients, and sepsis and ICU models in rodents. lar proteins, thereby depressing the specific force of muscle.

Other caspases have also been linked to muscle dysfunction


C. Caspase Involvement in Critical Illness
in sepsis or critical illness. Shao et al. (630) have shown that
caspase-1 cleaves proteins that regulate glycolysis including
The caspase family of cysteine proteases is well recognized
aldolase, triose-phosphate isomerase, glyceraldehyde-3-
as a regulator of apoptosis and immune function. In addi-
phosphate dehydrogenase, ␣-enolase, and pyruvate kinase.
tion, animal studies suggest that one or more members of
the caspase family may contribute to muscle proteolysis in In a murine model of sepsis, systemic lipopolysaccharide
critical illness or sepsis. Caspase-3 has been a primary focus. administration activated caspase-1 in diaphragm and inhib-
Du et al. (167) originally observed that recombinant ited glycolysis in muscle extracts. Glycolytic capacity was
caspase-3 degrades actomyosin (although degradation of preserved in caspase-1-deficient mice, identifying caspase-1
acto-myosin by caspase-3 is claimed throughout their work, as a mediator of sepsis effects on muscle metabolism. Llano-
blots were only probed for actin). They speculated that Diez et al. (434) found that caspase-1 mRNA was increased
caspase-3 activation might be an early step in muscle catab- in a rat model of critical illness myopathy (rats mechanically
olism that promotes proteolysis via the ubiquitin-protea- silenced and ventilated for up to 14 days), a late-phase
some pathway. Wei et al. (767) examined this possibility in response observed after 9 –14 days. In the same window of
limb muscles of rats with CLP-induced sepsis. They de- time, mRNAs for caspase-4 and caspase-12 were also ele-
tected no increases in caspase-3 mRNA, activated caspase-3 vated, suggesting potential roles for these more obscure
protein fragments, or caspase-3 enzyme activity. Nor was family members.

1070 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

D. Cathepsins in Critical Illness sults are intriguing, a partial glimpse into a complex prob-
lem.
Cathepsins are a family of proteolytic enzymes that have
long been recognized for their capacity to degrade proteins A number of studies have evaluated muscle protein synthe-
and polypeptides (53). In general, these proteases are active sis in critically ill patients. Essen et al. (187) measured the
in acidic environments, are localized to lysosomes, and are fractional rate of muscle protein synthesis in 15 ICU pa-
not regulated by calcium availability. Individual cathepsins tients among all of which the fractional synthesis rate for
are categorized as cysteine, serine, or aspartyl proteases skeletal muscle averaged 1.5%/day (over a 5-fold range
based on substrate specificity. individual variation). These values correlated with clinical
indices of metabolic status, notably arterial blood PO2 and
Two members of the cathepsin family, cathepsins B and L, pH and with illness severity. However, evaluating the dis-
have been evaluated extensively for their contributions to mus- tribution of protein synthesis rates among muscle and other
cle protein loss during sepsis. These studies began three de- tissues, the investigators identified no pattern that was char-
cades ago when Ruff and Secrist (600) first observed that skel- acteristic of critical illness. Gore and Wolfe (261) used iso-
etal muscle atrophy caused by experimental sepsis could be topic tracer techniques to measure the fractional rates of
prevented by pretreating rats with leupeptin, a cathepsin B muscle protein synthesis in six critically ill septic patients
inhibitor. Cathepsin B mRNA levels, activity as well as protein and six healthy controls. Under basal conditions, the frac-
degradation rates, were elevated in limb muscles isolated from tional synthesis rate in muscles of patients was more than
septic rats (284, 329, 330, 749). The clinical relevance of this twice the value measured in controls. A follow-up study
finding was reinforced by Helliwell et al. (298) who observed (263) with similar experimental design reached the same
greater immunolabeling for cathepsin B in muscle fibers of conclusion that critical illness significantly increases the
critically ill patients. Similarly, Deval et al. (157) reported that fractional synthesis rate of muscle protein. In contrast,
cathepsin L mRNA and protein levels were elevated in skeletal Klaude et al. (368) recently compared protein kinetics in leg
muscles of rodents with experimental sepsis. Increased expres- muscles of 16 ICU patients with sepsis or septic shock to
sion of cathepsin L has also been observed in muscles of criti- protein kinetics in 8 healthy control subjects using phenyl-
cally ill patients (130, 154), septic mice (569), and burn-in- alanine and 3-methylhistidine tracers. Arteriovenous differ-
jured rabbits (156). However, the putative roles of cathepsins ences were measured across the leg for each tracer. These
B and L are highly controversial. In muscles of septic rats, data were used to compute protein synthesis using both a
Bhattacharyya et al. (56) were unable to detect elevations in two-pool model and a three-pool model. Neither model
cathepsin B activity or cathepsin L activity, and Ahmad et al. detected any effect of critical illness. However, the variable
(7) found that cathepsin B protein levels were unaltered. In “sepsis” could not be separated from co-confounding vari-
critically ill patients, Klaude et al. (368) found no changes in ables of ICU treatment because all the patients studied suf-
the mRNA levels of either cathepsin B or cathepsin L. Inter-
fered from multiple organ failure and were analgo-sedated
ventional studies further challenge the importance of cathep-
and mechanically ventilated (368).
sins. In 1988, Hummel et al. (329) reported that leupeptin, a
cathepsin B inhibitor, only partially blunted the rise in protein
Of relevance to many ICU patients, Hammarqvist et al. (285)
degradation rate caused by experimental sepsis. They con-
examined the effect of surgery per se on markers of muscle
cluded that proteases other than cathepsin B might be in-
protein synthesis by evaluating ribosomal content in muscle
volved. Later studies (284) showed that leupeptin inhibited
biopsies of 22 patients before and 3 days after elective chole-
cathepsin B activity in muscles of septic rats but did not alter
cystectomy. After surgery, they observed decreases in 1) total
protein degradation rates. The authors concluded that cathep-
ribosomal concentration and 2) polyribosomes as a percent-
sin B was not a major regulator of accelerated muscle proteol-
age of total ribosomes, suggesting that surgery depresses mus-
ysis during sepsis. Therefore, there is no current consensus on
cle protein synthesis. Tjader et al. (707) tested this hypothesis
the role of cathepsins nor are they a compelling target for
therapeutic drug development. by measuring surgery effects on muscle protein fractional syn-
thesis rate. In 28 patients, measurements 90 min before sur-
gery were not different from measurements immediately after
E. Muscle Protein Synthesis in Critical surgery. Thus general anesthesia and surgical trauma did not
Illness immediately alter muscle protein synthesis. The apparently
disparate findings of these two studies may reflect differences
Net loss of muscle protein in critical illness requires that between the metabolic states of muscle immediately after sur-
protein is degraded faster than it is synthesized. Preceding gery versus 3 days later.
sections have outlined mechanisms responsible for in-
creased proteolysis. So, what of synthesis? Relatively few Signaling pathways that regulate muscle protein synthesis
studies have addressed this question directly. Such studies are sensitive to critical illness. As reviewed by Lang et al.
involved small, heterogeneous patient populations and ex- (389), animal studies suggest that signaling via the mamma-
perimental approaches that differed substantially. The re- lian target of rapamycin (mTOR) pathway is altered in

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1071


FRIEDRICH ET AL.

experimental models of sepsis, interfering with protein severely reduced for actin and myosin heavy chain I
translation and depressing protein synthesis. Data from (MyHC-I) and IIa (MyHC-IIa). These findings are consis-
critically ill patients are less clear. On the one hand, Con- tent with overall reductions in myofiber size that also were
stantin et al. (130) analyzed biopsies from vastus lateralis observed in these patients. At the protein level, myosin was
muscles of 10 patients and 10 age- and sex-matched healthy preferentially depleted by critical illness. The myosin-to-
control subjects. They measured changes in signaling pro- actin ratio was diminished both in limb and trunk muscles
teins that regulate the initiation of translation. These in- of patients relative to control values. Of those patients,
cluded v-akt murine thymoma oncogene homolog 1 (i.e., ⬎90% were mechanically ventilated and ⬎65% received
AKT1, PKB), glycogen synthase kinase 3 (GSK3), mTOR, corticosteroids while between 50 and 75% were septic so
70-kDa ribosomal protein S6 kinase 1 (p70S6K), and eu- that the preferential myosin loss reflects the consequences of
karyotic translation initiation factor 4E-binding protein 1 critical illness related to the mechanical ventilation and
(4E-BP1). Critical illness affected all of these signaling pro- analgo-sedation/steroid therapy and not primarily to sepsis.
teins similarly; in each case, protein phosphorylation was This molecular imbalance is predicted to alter myofibrillar
decreased. These findings suggest lower signaling activity in architecture and cross-bridge stoichiometry, compromising
the major pathways that promote protein translation. In specific force of skeletal muscle.
contrast, Jespersen et al. (353) used a similar experimental
design but obtained very different results analyzing AKT- In summary, the data in the literature related to protein
mTOR-S6K signaling in vastus lateralis biopsies from 12 imbalance in critical illness consistently point towards a
ICU patients and 12 age- and sex-matched healthy control general increase in protein catabolism and turnover result-
subjects. In patient muscles, they observed higher phospho- ing in severe atrophy. So far, no model of pure sepsis as only
rylated-to-total protein ratios for AKT (threonine 308), confounding variable in critical illness, whether in patients
mTOR (serine 2448), and S6K (threonine 389), whereas or in animal models has convincingly demonstrated prefer-
phosphorylated/total ratio for 4E-BP1 (threonine 37/46) ential myosin loss as in critically ill patients, mechanically
and GSK␤3 (serine 9) were indistinguishable from controls. ventilated, immobilized, and/or steroid-treated. Thus, while
sepsis seems to induce an atrophy-dominated phenotype of
It is difficult to reconcile these apparently contradictory
muscle weakness, critical illness myopathy in addition al-
data.
ters the quality of the atrophied muscle protein composition
towards preferential myosinolysis. More details are given in
In response to critical illness, skeletal muscle appears to
section XII. FIGURE 12 summarizes the protein synthesis
upregulate a variety of genes that regulate protein synthesis.
and degradation mechanisms discussed in this section.
Fredriksson et al. (212) analyzed vastus lateralis biopsies
from 17 ICU patients and 10 age-matched healthy controls.
Using microarray techniques, they detected increased XII. ANIMAL AND HUMAN MODELS OF
mRNA for 27 translation-related genes in muscles of pa- CRITICAL ILLNESS AND ICU-RELATED
tients, including proteins of the mitochondrial biogenesis MYOPATHIES
pathway and microRNA regulation. Subsequently, Con-
stantin et al. (130) used PCR techniques to document in-
creased mRNA for key signaling proteins including AKT1, A. The Problem of Choosing the Right
GSK3␣, GSK3␤, mTOR, p70S6K, and 4E-BP1 in muscles Animal Model: Matching Disease
of ICU patients. Less information is available about these Phenotype or Modeling Risk Factors?
molecules at the protein level, but Jespersen et al. (353)
observed higher amounts of AKT and marginally higher When trying to model a clinical endpoint, such as muscle
levels of 4E-BP1 in their patients. weakness, that can arise from a complex combination of
risk factors, one has to either 1) follow a trigger factor-
Critical illness also alters gene expression for key structural oriented approach of modeling risk factors and their impact
and functional elements in skeletal muscle. Fredriksson et on muscle function or 2) define specific pathological alter-
al. (212) found 1,457 unique genes/identifiers increased in ations found within the syndrome that define a specific dis-
muscles from ICU patients, including genes associated with ease entity seen in patients. Any trigger factor itself or in
catabolism, inflammation, and oxidative stress. Decreased combinations capable of reproducing the spectrum of path-
expression was observed in 525 genes including genes that ological changes is potentially causative for the disease. If
regulate mitochondrial biogenesis plus a variety of genes the trigger factor produces pathological alterations, but
that are skeletal muscle-specific. Importantly, myofibrillar they are distinct from the alterations present in patients,
proteins were among the gene products that were down- they likely trigger a different disease entity within the syn-
regulated. More recently, myofibrillar downregulation in drome ICUAW (FIGURE 1B). Potential causes of ICUAW in
critical illness was confirmed in muscle biopsies from 208 critically ill patients include, e.g., CIP, CIM, and combina-
ICU patients versus 35 healthy controls, the largest clinical tions. Acquired CIM is the most common cause underlying
study yet completed on this topic (154). mRNA levels were acquired paralysis in critically ill ICU patients, and it has

1072 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

been reported in 25–58% of the general ICU population 3) preferential and significant myosin loss,
and in 50 –100% of certain subgroups (52, 145, 216, 385, 4) eventual sarcomere disorganization,
415). For this reason, we will focus on animal models that 5) inadequate autophagy activation, and
have been used to study CIM rather than CIP. As detailed 6) altered protein turnover.
below, there is confusion in the literature about which as-
pect of CIM is being modeled by any given animal model, With regard to trigger factor-phenotype relationships, neu-
e.g., in the case of sepsis models, for which the proof of romuscular blockade (NMB), corticosteroids, and sepsis
producing preferential myosin loss is still elusive. In ICU have all been suggested to be important etiological factors,
patients suffering from CIM, the heterogeneity introduced but CIM has also been reported in the absence of one or two
by variable timing of disease onset, differences in underly- of these trigger factors in mechanically ventilated ICU pa-
ing diseases, age/gender differences, and pharmacological tients. However, all ICU patients with CIM have exposure
treatments make studies on mechanisms underlying CIM to long-term mechanical ventilation and immobilization in
extremely difficult. There is accordingly a strong need for common. Furthermore, the complete “mechanical silenc-
experimental models to explore 1) the relative importance ing,” unique for ICU patients, has been shown to play an
of different trigger factors; 2) the mechanisms underlying important role in the development of CIM in experimental
muscle specific differences between limb, respiratory, and and clinical studies (435, 517, 574). An additional factor
craniofacial muscles; and 3) the temporal sequence of that needs to be taken into account is the relatively long
events leading to the CIM phenotype with severe muscle delay between exposure to trigger factors and the pheno-
wasting and preferential loss of myosin in limb muscles. In type characterizing CIM, i.e., severe muscle wasting and
addition, there is a need for experimental mammalian in preferential myosin loss (399, 511, 610). The preferential
vivo models allowing time-resolved analyses from short to and significant myosin loss is the result of both a decreased
long durations in the search for specific intervention strat- synthesis at the transcriptional level and increased myofi-
egies targeting CIM, i.e., models allowing detailed studies brillar protein degradation (399, 434, 496, 513, 517). Thus
on early and late effects of the intervention on intracellular early effects via specific signaling pathways on protein syn-
signaling, protein synthesis/degradation, and regulation of thesis and degradation become manifest relatively late at
muscle contraction. In vitro models give important infor- the myofibrillar protein level, i.e., the preferential myosin
mation on specific signaling pathways, but CIM is not loss, due to the slow turnover rate of contractile proteins
caused by an isolated mechanism but is the consequence of with myosin having a 1–2% turnover rate per day (647).
a series of changes in a complex biological system, and in On the other hand, actin has been reported to have a turn-
vivo models mimicking the ICU condition are accordingly over rate twice as slow as myosin (457). Differences in
warranted. One might argue that the perfect animal model myosin and actin turnover rates and the preferential target-
would combine sepsis and treatment with antibiotics, im- ing of myosin by the E3 ligase MuRF1 for ubiquitination
mobilization and ventilator dependence, corticosteroid and degradation (127) offer a mechanism underlying the
treatment, and multiple organ failure as these are charac- preferential myosin loss in spite of a similar transcriptional
teristics shared by many ICU patients. Clearly, such a model downregulation of myosin and actin (513, 517). However,
would be expensive and labor-intensive, threatening the there is reason to believe that the mechanism underlying the
feasibility. The key issue for efficient use of animal models preferential myosin loss is more complex, involving other
of CIM is then which risk factors are relatively inexpensive mechanisms such as compromised protection by, e.g., small
to model and yet sufficient to trigger CIM, so they can be and large heat shock proteins. There is a need for experi-
studied efficiently. A second issue is how well the animal mental studies allowing time-resolved analyses with a high
model recreates the pathological phenotype of CIM or just temporal resolution to detect early changes in the complex
belongs to another form of myopathy under the “ICUAW intracellular signaling cascades and effects at the gene/pro-
umbrella” (FIGURE 1B). Pathological changes present in pa- tein levels leading to the CIM phenotype. Apart from the
tients are unique to CIM, suggesting the presence of a preferential myosin loss, it is important to reproduce the
unique disease. The phenotype defining CIM in patients reduced muscle membrane excitability in the animal model,
should therefore be the starting point of our considerations as a hallmark of early manifestation of CIM. In the litera-
when comparing the power and limitations of different an- ture, there have been three principal approaches employed
imal models. as potential models for CIM: sepsis models; ICU-porcine,
-rabbit, and -rat models; and pharmacological steroid-de-
The changes seen in patients with definite CIM that should nervation. In the following sections, we present the ratio-
be ideally reproduced by any animal model of CIM, on top nale behind those different models, evaluate their advan-
of severe muscle weakness, are as follows: tages and disadvantages, summarize prime findings and
1) electrical hypoexcitability of the muscle and poor exci- whether the respective model was able to reproduce all
tation-contraction coupling, phenotypic manifestations of CIM or whether they may
2) severe muscle atrophy (beyond that caused by inactivity point to a different disease entity within the ICUAW syn-
alone), drome. Finally, some in vitro models are discussed briefly.

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1073


FRIEDRICH ET AL.

Myosin loss, atrophy, disorganization of sarcomeres


Control CIM Control CIM

Control
H+E

CIM
Myosin
ATPase
Actin

Loss of electrical excitability Sodium channelopathy


Control Excitable Indeterm Inexcitable Hyperpolarized shift in inactivation
40 1.0

Normalized current
20 0.8
+ 0
0.6
mV
–20
Corticosteroids –40 0.4
–60 0.2
–80
10 ms 0.0
–100 –90 –80 –70 –60 –50
Voltage (mV)
FIGURE 13. Rat steroid-denervation model of CIM and phenotypic characterization. The steroid-denervation
(SD) model involves a combined surgical denervation (partial sciatic nerve removal) and a pharmacological
steroid treatment (e.g., dexamethasone 5 mg/kg ip daily). This model produces reduction/loss of electrical
excitability in limb muscle and Na⫹ channelopathy within a few days. In addition, severe atrophy (as seen in
reduced fiber cross-sectional areas), preferential myosin loss (as detected in gel electrophoresis), and sarco-
mere disorganization (as visualized by confocal microscopy) provide most of the phenotypic changes to model
CIM as seen in critically ill patients. [Top right panel from Kraner et al. (377). Left panel from Rich et al. (582).
Copyright John Wiley and Sons. Bottom middle panel from Rich and Pinter (580). Copyright John Wiley and
Sons.]

B. The Rat Steroid-Denervation Model potential activity in muscle is the primary factor inducing
denervation changes in muscle. In two papers, Lomo and
The issue of what aspect of muscle immobility triggers CIM colleagues (440, 441) found that stimulation of denervated
in patients is central to the validity of various animal mod- muscle prevented the upregulation of extrajunctional
els. There are ways to model different aspects of immobility AChRs and prevented depolarization of resting potential.
in patients. Immobility could trigger CIM due to either loss More recently, in mutant mice with hyperexcitable muscle
of muscle action potentials or loss of passive muscle move- due to a mutation in the muscle chloride channel ClC-1, it
ment, or a combination of both. Loss of muscle action po- was found that denervation of muscle did not prevent spon-
tentials can be easily modeled by cutting the sciatic nerve to taneous activity. The spontaneous activity was sufficient to
denervate leg muscles (FIGURE 13). A rat model of acute prevent both depolarization of the resting potential and
CIM using denervation was first developed over 20 yr ago development of extrajunctional ACh sensitivity following
(466, 597). The model involves pairing loss of muscle ac- denervation (757). These studies demonstrated that muscle
tivity (by denervation) with systemic administration of cor- activity was sufficient to prevent some well described den-
ticosteroids (FIGURE 13). The model was established to ervation-induced changes. By extension, these data suggest
mimic the situation in patients given systemic neuromuscu- that some properties of muscle are primarily regulated by
lar blocking agents in conjunction with high-dose cortico- action potentials rather than chemical trophic influences.
steroids to treat asthma and chronic obstructive pulmonary
disease (COPD). Those patients develop classical CIM. De- There are two main differences between the rat model and
nervation does not identically mimic the situation present in patients. In the rat model, trophic signaling from the nerve
immobilized patients as the intact nerve is still present in at the endplate is absent, whereas in patients, chemical
patients. The rat model using denervation assumes that loss trophic signaling should still be present. For example, neu-
of muscle activity due to denervation has the same effect on regulins are trophic signaling molecules from nerve that
muscle as loss of activity due to neuromuscular blockade or modulate muscle acetylcholine receptor expression (586).
immobilization due to sedation. The debate about whether Loss of neuregulin signaling due to denervation may medi-
trophic factors from nerve are primarily regulated by activ- ate the terminal Schwann cell response to denervation
ity or by chemical influences is a long-standing one. For an (586). A second issue is that in patients muscle is unloaded,
excellent early review on this issue, see McArdle (468). whereas in the rat leg following denervation by sciatic nerve
There are several studies which suggest that loss of action lesion, there is continued passive movement during gait. In

1074 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

an early study that directly compared unloading to dener- this porcine model, the same type of ventilators used in ICU
vation, it was found that unloading triggered atrophy sim- patients can be used in pigs. Previously, experiments with
ilar to denervation, but did not trigger upregulation of ex- the porcine model were typically for shorter durations, i.e.,
trajunctional AChRs (440). This suggests that unloading of a day or less. In pilot experiments, it was shown that me-
muscle does not trigger all of the changes triggered by de- chanically ventilated pigs exposed to NMB, systemic ad-
nervation. A recent study in patients examined the effects of ministration of corticosteroids, and sepsis for 5 days devel-
mild passive movement on muscle function in immobilized oped an electrophysiological phenotype resembling previ-
patients and found that, while it improved specific muscle ous observations in ICU patients with CIM, i.e., reduced
force, it did not improve either atrophy or myosin loss compound muscle action potential (CMAP) amplitudes
(435). Thus some defects in CIM are likely triggered by with maintained motor nerve conduction velocity and in-
unloading, but other defects are triggered by other aspects tact sensory nerve action potential amplitude and conduc-
of critical illness and immobility in the ICU. tion velocity (511, 517) (FIGURE 14C). This model was sub-
sequently used in a series of studies focusing on the relative
While there are differences between the risk factors present importance of different trigger agents such as neuromuscu-
in the rat model and patients, the rat steroid denervation lar blockade, sepsis, and steroids in the pathogenesis of
model does an excellent job of recreating pathological CIM, muscle specific differences, as well as the effects of a
changes present in patients with CIM. The features re- Ca2⫹ sensitizer on diaphragm muscle fiber function (1, 2,
created by the rat model include (FIGURE 13): 32, 511, 517, 520, 566, 686). Specific interest was focused
1) dramatic atrophy of muscle fibers (377, 466, 496, 597), on the early effects of NMB, systemic corticosteroid admin-
2) preferential/selective loss of myosin (377, 466, 496, 597), istration, and sepsis, separately or in combination, on mus-
3) disorganization of sarcomeres (377, 466), and cle fiber size, regulation of muscle contraction at the single
4) electrical hypoexcitability (580 –582). muscle fiber level in limb, respiratory and craniofacial mus-
cles, and electrophysiological properties (1, 2, 32, 517, 520,
Thus all the major changes in muscle found in CIM patients 686). Exposure to mechanical ventilation and sedation,
are shared by the rat model (it should be noted that inade- with or without NMB, did not have a significant effect on
quate autophagy and metabolic failure have not yet been
limb muscle fiber size or specific force during the 5-day
tested for in this rat steroid-denervation model). The sim-
observation period, but the addition of systemic adminis-
plest interpretation of these pathological data is that loss of
tration of corticosteroids, sepsis, or the combination of cor-
action potentials and steroid treatment cause myopathy in
ticosteroids and sepsis resulted in a decreased specific force
rats, which is very similar to CIM occurring in patients.
and maintained fiber size (517). The most dramatic decline
Major advantages of the model are that it is easy to create
in specific force was observed in pigs receiving the combi-
and studies are inexpensive. The primary disadvantage of
nation of mechanical ventilation, sedation, NMB, steroids,
the model is that although it reproduces the phenotypic
and sepsis where specific force declined almost 50% in spite
features of muscle in patients with CIM, the rats are not
of a maintained fiber size. In none of the groups were any
critically ill and thus lack many of the triggers that are
significant differences observed between muscle fibers ex-
involved in causing CIM in patients. It is important to note
that CIM can develop in critically patients and in experi- pressing different fast or slow myosin heavy chain isoforms
mental animal models that have not been exposed to exter- (517). Maximum velocity of unloaded shortening was not
nally applied corticosteroids. Thus, while this model is ex- affected by any of the different trigger factors in muscle
cellent in studying and treating pathological mechanisms, it fibers expressing the same myosin isoform, suggesting
should be used with caution for studies aimed at preventing maintained cross-bridge cycling kinetics in spite of the de-
CIM by treating triggers of CIM. creased force generation capacity (517). The CMAP ampli-
tude was reduced in all groups independent of the combi-
nation of trigger factors, suggesting that different mecha-
C. Porcine ICU Model nisms underlie the decline in muscle membrane excitability
and force generating capacity in response to the ICU con-
Unfortunately, there are very few experimental models dition (FIGURE 14C) (517). The sparing of both muscle fiber
where long-term effects of mechanical ventilation and im- size and force generation capacity in response to mechanical
mobilization on skeletal muscle have been studied in detail. ventilation with or without NMB were paralleled by a sig-
However, experimental porcine models have for many nificant upregulation of high- and low-molecular-weight
years been used to study the effects of sepsis on organ func- heat shock proteins (HSPs). In animals exposed to sepsis
tion (FIGURE 14) (11, 522) and in the development, treat- and corticosteroids in addition to mechanical ventilation
ment, and testing of diagnostic markers of malignant hyper- and NMB, the decline in force generation capacity was
thermia (476). The porcine model was, therefore, for- accompanied by decreased HSP levels (2, 32). In the dia-
warded as an ideal candidate for an animal model of CIM, phragm, 5 days of mechanical ventilation and sedation re-
due to the known similarity in metabolism between humans sulted in a severe decline in muscle fiber force without any
and pigs and extensive experience with this model (511). In structural remodeling at the cellular and motor protein lev-

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1075


FRIEDRICH ET AL.

A B D preferential myosin loss

Specific force (N.cm-2)


25

20
Myosin
15

10

5
muscle weakness
0
1800
1600
1400

CSA (μm2)
1200
1000
Actin
800
600
400
200 muscle atrophy
0 E ultrastructural disorganization

ys

ys

ys
da

da

da

da
control rat 10 d MV + IM rat
0

-4

-8

4
-1
2

5
0.

9
C 6 hypoexcitability

soleus
action potential (mV)
Compound muscle

* *
*

diaphragm
2
* *
rat/porcine ICU model 1

0
V

CS

is

L
BA

AL
M

ps
NM

V+

se
M

V+
V+

M
M

FIGURE 14. Rat and porcine ICU animal models of CIM. A: settings of the rodent (top) and porcine (bottom)
experimental ICU models. B: single muscle (EDL, soleus) cross-sectional area (CSA) and specific force
progressively declined in mechanically ventilated rats with duration of treatment and fell to ⬃50% after 2 wk
(518). The data show prominent muscle weakness beyond pure atrophy. C: electrical hypoexcitability of porcine
tibialis anterior muscle seen as marked drop in CMAP amplitudes 5 days following mechanical ventilation (MV)
alone and in combination with NMBAs, corticosteroids (CS), sepsis, or all triggering factors (517). D:
preferential myosin loss during mechanical ventilation, documented in Coomassie-stained 12% SDS-PAGE
stained gels from rat soleus muscle in a control animal (1) and in a rat mechanically ventilated and immobilized
for 10 days (2). The myosin-to-actin ratio was 2.1 in the control and 0.5 after 10 days of complete
immobilization. The myosin-to-actin ratio in the diaphragm (3) from a rat mechanically ventilated and immobi-
lized for 10 days was 2.0, i.e., similar to control values in both limb and respiratory muscles. E: electron
micrographs from soleus and diaphragm muscles from control rats and rats mechanically ventilated for 10
days. In the soleus muscle, 10 days immobilization and mechanical ventilation (MV ⫹ IM) resulted in a
disorganization of the A-band in the sarcomere, while an intact A-band is observed in the diaphragm after 10
days of mechanical ventilation, in accordance with the maintained stoichiometric relationship between myosin
and actin. In spite of a maintained myosin-to-actin ratio in the diaphragm, a slight general myofibrillar protein
loss was observed in response to 10 days of mechanical ventilation. In the diaphragm, mitochondria appeared
swollen with disorganized cristae after 10 days of mechanical ventilation (mitochondria are indicated by the
arrows). Horizontal bars denote 1 ␮m.

els. The addition of sepsis, corticosteroids, and NMB, sep- masseter muscle were maintained in response to the 5-day
arately or in combination, did not add any significant neg- exposure to mechanical ventilation, NMB, corticosteroids,
ative effects, in sharp contrast to observations in limb mus- and sepsis, in contrast to limb muscles (1). This is consistent
cles (520). The unaltered myosin and actin contents in the with the sparing of craniofacial muscles in ICU patients
diaphragm indicate that qualitative changes in contractile with CIM. Gene expression analyses revealed highly com-
proteins by posttranslational modifications may be the plex mechanisms underlying this muscle-specific mecha-
dominant mechanisms underlying the ventilator-induced nism and HSPs appear to play an important role in this
deterioration of diaphragm muscle function. Both muscle muscle specific difference (1). However, other factors, re-
fiber size and force generation capacity of the craniofacial lated to metalloproteinase inhibition, oxidative stress re-

1076 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

sponse elements, transcription, and growth factors are also come by studying adult mini-pigs who have approximately
involved in the muscle specific difference between limb and the same body weight as the piglets.
craniofacial muscles (1). The exact mechanism underlying
the relative sparing of craniofacial muscles versus limb mus-
cles, which is also reproduced in the porcine animal model, D. Rodent ICU Model
is not known, but intrinsic differences related to differences
in embryonic origin and inherent differences in membrane Experimental rodent ICU models offer a cost-efficient alter-
properties and myofibrillar protein expression may have a native to the porcine ICU model with significant logistic and
significant effect on muscle-specific differences also in administrative advantages. Mouse models allow efficient
mechanosensation and responsiveness to mechanical silenc- genetic engineering, but there is increasing evidence that the
rat is a better model for studies of muscle disease and aging
ing (1, 8).
(336, 628). However, long-term mechanical ventilation in
small rodents imposes substantial challenges, since com-
Both high- and low-molecular-weight HSPs are involved in
mercially available rodent ventilators are unable to main-
the protection of proteins in response to different types of
tain life support for longer than a couple of days. Most
stress (1). The high-molecular-weight HSPs include HSPs
rodent studies using commercially available ventilators are,
60, 70, 90, and 110, and the low-molecular-weight HSPs therefore, limited to observation periods shorter than 24 h,
include HSP 27 and ␣B-crystallin. HSP 27 and ␣B-crystallin limiting the interpretative value both by adding a significant
are translocated from the cytoplasm to the sarcomere where confounding factor when studying a failing preparation,
they protect sarcomeric proteins, including myosin, during and also that it takes significantly longer than 24 h to de-
stress. It was recently shown that low-molecular-weight velop CIM phenotypes.
HSPs also prevent aggregation of the giant sarcomeric pro-
tein titin during stress (374). In addition to the protective More than two decades ago, Dworkin and co-workers
role of HSPs, they are also involved in different physiolog- (174 –177) developed an experimental rat model to study
ical processes affecting muscle size and function. HSP 70 is blood pressure regulation in a pharmacologically paralyzed
involved in FOXO signaling, and HSP 27 is a negative reg- rat. A ventilator was designed allowing long-term con-
ulator of NF-␬B in skeletal muscle, and overexpression of trolled mechanical ventilation of the rat, and the longest
these HSPs reduces muscle atrophy during different muscle duration one rat has been exposed to NMB and mechanical
wasting conditions (160, 627). Thus results from the por- ventilation is 3 months (174 –177). Thus this model offers a
cine model indicate there is a loss in muscle function when unique possibility to conduct time-resolved analyses with a
HSP expression is compromised and that a muscle specific high temporal resolution on the effects of the ICU condition
difference in HSP expression is one factor underlying the on muscle, muscle specific differences, as well as in the
sparing of craniofacial muscles in critically ill ICU patients. design and evaluation of specific intervention strategies
(395). In accordance with clinical and experimental studies
The 5-day exposure to sepsis in mechanically ventilated pigs using the porcine ICU model, mechanical ventilation and
is, to our knowledge, significantly longer than in any other immobilization are required for durations longer than 5
porcine sepsis experiment, but still, this duration was too days to induce the CIM phenotype, i.e., muscle wasting and
short to induce the preferential myosin loss observed in ICU a preferential myosin loss in limb muscles, relative sparing
patients with CIM. This is consistent with clinical observa- of carniofacial muscles, and severe negative effects on reg-
tions, i.e., that it typically takes longer than 5 days to de- ulation of diaphragm muscle fiber function (8, 133, 517,
520). The CIM phenotype was observed in the rat model in
velop the CIM phenotype with the preferential myosin loss
the absence of both systemic corticosteriod hormone ad-
in mechanically ventilated ICU patients (6). Thus there is a
ministration and sepsis. Importantly, complete “mechani-
need for an experimental model allowing both short- and
cal silencing,” i.e., no external load related to weight bear-
long-term studies. In principle, this could be achieved with
ing and internal load related to activation of contractile
the porcine model, but it would be associated with signifi- proteins, was accordingly forwarded as an important factor
cant logistic problems and financial costs. The age of the underlying the preferential and significant myosin loss,
pigs is an additional problem, i.e., all experiments have so leading to impaired muscle function and persistent paralysis
far been performed in piglets weighing ⬃25 kg. These ani- (510 –512, 517, 574). Current studies raise the possibility
mals are in a significant growth phase with an expected that the complete mechanical silencing in the experimental
approximately fourfold increase in body weight within 6 –7 ICU models and in mechanically ventilated ICU patients is
mo, i.e., an anabolic situation significantly different from unique for the ICU condition and different from the immo-
what is observed in ICU patients with CIM and may more bilization during bed rest, joint fixation, hindlimb suspen-
reflect a model of CIM in pediatric patients. Furthermore, sion, microgravity, or peripheral denervation. Detailed
the incidence of CIM increases with patients’ age, and there time-resolved analyses of global protein synthesis rate, tran-
are only few reports of CIM in pediatric ICUs (608). How- scriptional regulation of myofibrillar protein synthesis, and
ever, these disadvantages could, at least in part, be over- activation of major proteolytic degradation pathways in

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1077


FRIEDRICH ET AL.

skeletal muscle revealed a unique and complex temporal 511, 517, 518). Interestingly, in intercostal muscles, there is
muscle-specific pattern in response to the ICU intervention. a similar decline in myosin-to-actin ratio as in the limb
Further studies directly comparing mechanical silencing in muscle (512). Thus it may be speculated that the passive
the ICU to other manipulations of muscle activity will be loading of the diaphragm by the mechanical ventilator (72
necesssary to test this hypothesis. The preferential myosin times/min in the rat model) had an impact on transcrip-
loss in limb muscles was paralleled by a strong transcrip- tional regulation of myofibrillar protein synthesis but not
tional downregulation of both actin and myosin, an early on the activation of protein degradation pathways. How-
activation of the ubiquitin-proteasome degradation path- ever, it cannot be ruled out that intrinsic muscle-specific
way preceding the muscle atrophy and the preferential my- differences underlie the differences in transcriptional regu-
osin loss. Calpain and lysosomal protein degradation path- lation of myofibrillar proteins between diaphragm and limb
ways, on the other hand, were preceded by the preferential muscles. While the preferential myosin loss plays an impor-
myosin loss. The MuRF1 and MuRF3 E3 ligases showed a tant role for the decreased specific force in limb muscle
biphasic spatial translocation in response to the mechanical fibers, other mechanisms are dominating in the diaphragm,
silencing with an early nuclear translocation and were fol- e.g., an early onset of oxidative stress, mitochondrial dys-
lowed by a translocation to the cytoplasm with a distinct function, intracellular lipid accumulation, and posttransla-
perinuclear accumulation after long-term immobilization tional protein modifications (PTMs) (133). Thus qualitative
and mechanical ventilation (517). Mild passive mechanical changes in contractile proteins resulting in the severely im-
loading of limb muscles ameliorates both the muscle fiber paired residual function appear to dominate in diaphragm,
atrophy and the loss of specific force in the rat ICU model. while quantitative changes in myosin and myosin-associ-
In the slow-twitch soleus, with a higher protein turnover ated proteins play a more important role for the decreased
rate than in fast-twitch muscles, the preferential myosin loss force-generating capacity in limb muscles.
was ameliorated together with lower levels of MuRF1
(574). Thus these insights offer molecular and cellular As discussed above, craniofacial muscles are typically
mechanisms underlying the positive effects of early mobili- spared or less affected than limb muscles in CIM patients.
zation in mechanically ventilated and immobilized ICU pa- The masticatory masseter muscle was, therefore, studied in
tients, as well as strongly supporting intense physical ther- the rat ICU model. In contrast to limb muscles, the masseter
apy in immobilized ICU patients. showed only a mild and delayed decline in myosin-to-actin
ratio, smaller reduction in fiber size, and no transcriptional
The major inspiratory muscle, the diaphragm, showed a downregulation of myofibrillar protein synthesis. In con-
different temporal pattern in response to the ICU condition trast to both limb muscles and the diaphragm, there was an
compared with limb muscles. A rapid early decline in max- increase in MuRF1 levels and more sustainable levels of
imum diaphragm muscle fiber force, preceding fiber atro- sarcomere protective HSPs and autophagy (8). Thus the
phy, was observed in response to mechanical ventilation. better preserved myofibrillar protein expression and muscle
Two weeks after controlled mechanical ventilation, resid- fiber size in the masseter muscle compared with limb mus-
ual function of diaphragm muscle to generate force was cles in response to the ICU condition were the result of
⬍15% of control values when combining the effect of both muscle-specific differences in both transcriptional regula-
atrophy and loss of specific force at the single muscle fiber tion of myofibrillar protein synthesis and protein degrada-
level (133). These measurements were performed directly tion pathways and coupled to a decreased activation of the
assessing contractile protein function and bypassing EC IGF-I/PI3K/Akt pathway as well as protective effects of HSP
coupling in permeabilized diaphragm muscle fibers; thus and autophagy machineries (8).
changes in membrane excitability and EC coupling may
accordingly add to the effects at the contractile protein level The rat ICU model offers the opportunity to investigate
in intact fibers. Furthermore, there was a progressive in- effects of different intervention strategies on muscle func-
crease in the number of no force generating fibers with tion. Passive mechanical loading has been discussed above,
increasing duration of the mechanical ventilation, adding to and the rat experimental ICU model is presently being used
the 85% reduction in diaphragm muscle function (133). In in different pharmacological intervention studies. Nutri-
accordance with observations in limb muscles, there was an tional status is another important factor in the treatment of
early activation of the ubiquitin proteasome, but in contrast critically ill ICU patients, but the role of nutrition in the
to limb muscles, myosin-to-actin ratios were not affected, pathogenesis of CIM remains unknown. In an attempt to
and transcriptional regulation of myosin isoforms did not study the effects of caloric intake in the pathogenesis of
show the dramatic and early downregulation as observed in CIM, limb muscles were compared between animals given
limb muscles. Although the exact underlying mechanisms low versus eucaloric parenteral feeding (523). However, the
for the diaphragm weakness are not known, these findings preferential myosin loss, decline in specific force, and mus-
may point towards an altered quality of acto-myosin inter- cle fiber atrophy did not differ between low and eucaloric
actions in the affected diaphragm where also the loading animals. Furthermore, passive mechanical loading had a
pattern would be changed during ICU management (133, sparing effect on muscle weight independent of nutritional

1078 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

status. Thus the mechanical silencing associated with the (602) and reliably produces a marked increase in myofibril-
ICU condition is forwarded as a stronger trigger factor than lar protein proteolysis from 6 h post-procedure (200, 291,
nutritional status in the pathogenesis of CIM (523). 317, 776) (FIGURE 15A). However, myofibrillar protein loss
always seems to affect actin and myosin likewise, at least in
The major disadvantages of the rat and porcine ICU models all studies investigating the time course of proteolysis up to
are the labor intense experiments with 24 h/day monitoring ⬃20 h (200, 290, 291, 776). It may be that a significant
of heart rate, respiratory function, blood pressure, EEG, preferential myosin loss does not develop until longer times
body temperature, PCO2, peep, urine output, peripheral per- post-CLP, but this has not been experimentally proven al-
fusion, and PO2. In addition, most of the equipment has to though CLP in mice has been followed up until day seven,
be custom-fabricated, continuously calibrated, and main- but not for protein contents in skeletal muscle (809). Thus
tained. Despite these disadvantages, the in vivo experimen- this sepsis model has so far not qualified yet to reproduce all
tal animal ICU models presented here (porcine and rat) are the pathological hallmarks seen in CIM, most importantly,
best at modeling triggers for CIM in patients. These models preferential myosin loss, and is thus considered as sepsis-
also do an excellent job at modeling pathological changes induced myopathy (SIM) under the ICUAW umbrella (FIG-
such as (FIGURE 14) URE 1B). Apart from this specific point, CLP in rodents has
1) severe atrophy (8, 133, 510 –512, 517), been criticized in several further points. First, while the
2) preferential and significant myosin loss (510 –512, 517, approach is well accepted to mimic the situation of perfo-
574), rated appendicitis or diverticulitis with peritoneal sepsis in
3) sarcomere disorganization (FIGURE 14), patients, this model only applies to a fraction of septic pa-
4) electrical hypoexcitability (511, 517), and tients, i.e., ⬍50% of septic patients show documented bac-
5) inadequate autophagy activation (8, 31). teremia (587). Also, CLP animals are often of young age
(although considered as adult; use of few weeks old animals
E. Rodent Sepsis Models (CLP and LPS may serve as models for human pediatric CIM in future)
Models) and without co-morbidities such as usually seen in older
patients with peritoneal sepsis. Moreover, the time window
of CLP sepsis in rodents is uncomparibly compressed to a
Rodent models most commonly used in sepsis research are
few days at most, unlike in patients (199). However, the
the cecal ligation and puncture (CLP) model and the LPS
most prominent argument excluding a simple transfer of the
challenge model. The former is most commonly performed
rodent procedure to the human situation of peritoneal sep-
in rats while the latter is preferred in rats or mice. Both
sis-induced critical illness is the lack of supportive measures
models have been extensively used to study muscle wasting
associated with sepsis and the underlying mechanisms in CLP-treated mice or rats, such as mechanical ventilation
(317). The various detailed findings associated with CLP- or complete immobilization shown to produce the CIM
and LPS-induced sepsis are given in the respective sections pathology (i.e., preferential and significant myosin loss,
of this review. This section focuses on distinct differences of Ref. 517). Last, probably similar to reasons explained be-
both techniques and their strengths and weaknesses, while low for the LPS model, differences between the immune
details on the procedures are given in several reviews else- system of mice and rats versus humans may preclude simple
where (317, 588). Although being used in abundance, sev- transfer of conclusions to humans (628). However, it is
eral lines of critique have recently emerged to the use of CLP important to raise general caution against overstating limi-
and LPS models to adequately reflect conclusions drawn for tations of animal models of human disease. Reasons many
sepsis in humans (199, 587, 628). animal studies fail to predict clinical success of therapy
include poorly designed experiments and small sample size
The CLP model has been used for over 30 yr as an animal (91, 345).
model of bacterial peritonitis in rodents (601, 772) and has
long been considered the “gold standard” in sepsis research In the LPS challenge model, endotoxins are injected into
(85, 588). It involves suture-ligation below the ileo-cecal animals either subcutaneously, intraperitoneally, or via
valve and a needle puncture, allowing bacterial contamina- intravenous routes. As a component of the bacterial wall
tion of the abdominal cavity, followed by peritonitis and of gram-negative bacteria, LPS activates the innate im-
septicemia (FIGURE 15A). The subsequent systemic inflam- mune response via binding to toll-like-4 receptors
matory response may then lead to septic shock, multiorgan (TLR-4) with a dose- and administration-dependent re-
failure, and death. The severity of sepsis can be modeled by sponse (79). Since continuous or repetitive injections may
procedure variables such as needle size, length of ligation, often result in tolerance, single-dose injections are most
and number of punctures (588). However, it is exactly this commonly employed (317). Depending on the applied
variability that is often not standardized in experimental dose mild, moderate (sublethal) and severe (lethal)
sepsis reports and which may complicate comparative re- courses of sepsis can be observed (FIGURE 15B). Sublethal
sults. The model adequately reproduces a hyperdynamic doses (0.5–5 mg/kg body wt) induce a transient increase
circulatory response and cytokine surge within a few hours in pro-inflammatory cytokines and febrile response with

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1079


FRIEDRICH ET AL.

Septic
Sham
Stds
Cecum ligation & puncture (CLP)

Myofilament Release (%)


12

a b c 10 * ← Titin

8 * 204 ← Myosin
6
141 ← α-Actinin
4 ← Desmin
78
2 ← Actin
39
0 30.7
0 2 4 6 8 10 12 14 16 18
Hours
B
Lipopolysaccharide challenge (LPS) Rat edl muscle

Myofibrillar proteolytic rate (a.u.)


s.c. Low dose: <5mg/kg bw
Moderated dose: 0.5–5 mg/kg bw
High dose: >5 mg/kg bw Endotoxemia
(10 mg/kg bw i.p)

i.v.
Control group

2 6 12 24
i.p.
Time (hrs)
FIGURE 15. Rodent animal models of sepsis. Cecum ligation and puncture (CLP) (A) and lipopolysaccharide
challenge (LPS) (B) are the most commonly used animal models to mimic various degrees of sepsis from mild
to moderate sublethal sepsis and septic shock, depending on details of the techniques. In CLP, the prominent
rodent cecum is exposed after median laparatomy, ligated with suture, and punctured with a needle. After
gently squeezing the cecum to allow feces to penetrate, the cecum is relocated back into the abdomen and the
incision closed. [Photographs from Rittirsch et al. (588). Reprinted by permission from Macmillan Publishers
Ltd.] CLP aims to model human peritonitis sepsis. In muscle, myofibrillar protein loss (actin and myosin) is
detected from ⬃4 h post-CLP. [Data from Williams et al. (776), with permission from The FASEB Journal
(www.fasebj.org).] In LPS challenge, lipopolysaccharides from various bacterial sources (mostly E. coli) are
injected as single doses either intravenously, intraperitoneally, or subcutaneously. Depending on the doses, the
degree of sepsis can be roughly controlled. LPS-induced sepsis results in marked protein loss in skeletal muscle
as early as 2 h postinjection. [Modified according to data from Chai et al. (113).]

recovery of animals within 48 h (317), whereas higher wt, a million times greater dosage than required to induce
doses produce severe sepsis, multiorgan failure, and en- fever in humans (199, 666). Seok et al. (628) even con-
dotoxic shock that usually is lethal within 48 h, but there clude that “while the genomic responses to different
is substantial variation (317). Both moderate and higher acute inflammatory stresses are highly similar in humans,
doses alter muscle protein metabolism towards an in- these responses are not reproduced in current mouse
creased myofibrillar proteolysis and decreased protein models” (628). Therefore, while the finding of myofibril-
synthesis in many studies as early as 2 h post-LPS chal- lar proteolysis in LPS/CLP models might be attractive to
lenge (113) (FIGURE 15B). A selection of studies and their transfer to the situation in septic ICU patients, the impact
findings is given in Holecek (317). Although very com- of the sepsis response on muscle in rodent models might
monly used as a model mimicking endotoxin burden in be an inadequate correlate of muscle wasting in septic
septic patients, the LPS model in rodents has been quite patients. This is because whenever clinical studies in ICU
recently challenged regarding its transferability to human myopathies usually refer to septic ICU patients in the first
sepsis (628). In fact, the very transient increase in plasma place with the primary variable of sepsis, those patients
cytokine levels and rapid clearance of LPS from the sys- are almost always mechanically ventilated and/or immo-
temic circulation is not matching the situation in hu- bilized at the same time, at least in the initial phases.
mans. This may be due to circulating factors in the Thus, the hallmark of CIM in patients (immobilized
plasma of mice suppressing the production of pro-in- and/or mechanically ventilated regardless of whether
flammatory cytokines following LPS challenge, such as septic or not) is reflected by a preferential myosin loss
hemopexin (424). This may at least in part explain the that has so far not been confirmed in any pure CLP/LPS
vast difference in LPS sensitivity between mice and hu- model. It should be noted that almost all studies of sepsis
mans, with mice requiring an LD50 of 1–25 mg/kg body in rodents do not include mechanical ventilation and im-

1080 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

mobilization (exceptions see above). To more accurately or purified protein factors (cytokines, etc.) to skeletal mus-
mimic CIM in patients, these triggers need to be included. cle cells in vitro and studying protein contents and/or cell
functions thereafter. Usually, those muscle cells are of ani-
mal origin, either freshly isolated fibers, fiber bundles, or
F. Rabbit Burn Injury Model of Prolonged myotubes. While some laboratories could not confirm a
Critical Illness proteolytic effect induced by IL-1 or TNF-␣ in vitro (251,
258, 491), TNF-␣ injection in vivo into rats was effective in
A well-validated model of prolonged critical illness is that of accelerating muscle proteolysis, similar to endotoxin treat-
catheterized and fluid resuscitated rabbits suffering from ment, but unlike IL-1 injection which was ineffective (258).
hyperinflammation-induced critical illness evoked by third Other studies, however, were able to detect a marked stim-
degree burn injury (155). This model displays endocrine ulation of protein degradation by highly purified human
and metabolic alterations that are very similar to those in leukocytic pyrogen (IL-1) when applied to isolated rat mus-
ICU patients suffering from prolonged critical illness (766). cles kept at between 37 and 39°C (34). When the action of
Fasted critically ill rabbits lost weight and showed elevated purified IL-1 was compared with that of recombinant IL-1
mRNA expression and/or activity of several ubiquitin-pro- (␣ or ␤ isoform), recombinant factors failed to reproduce
teasome pathway components, calpain-1 and cathepsin-L, muscle proteolysis in vitro (251). From this finding, Gold-
as well as a shift towards smaller myofibers in skeletal mus- berg et al. (251) suggested an additional, still unidentified
cle (155, 156). Intravenous feeding largely counteracted product of activated monocytes to essentially regulate pro-
this response, provided hyperglycemia was avoided. Re- teolysis in systemic infections, and which would still be
sponses in diaphragm and heart were roughly similar. In present in IL-1 purifications but not in the recombinant
this model, it was clearly shown that fasting during critical
cytokine. Similar as for IL-1, IL-6 was able to induce muscle
illness also induces activation of autophagy in vital organs
proteolysis, but only when applied in vivo. In vitro incuba-
and skeletal muscle. Intravenous feeding abolished these
tion of isolated muscles with IL-6 failed to induce proteol-
responses, with most impact with amino acid-enriched nu-
ysis (258). Since these studies suggested additional factors
trition. Accumulation of p62 and ubiquitinated proteins in
present in the plasma during sepsis or systemic infection
muscle and liver, indicative of insufficient autophagy, oc-
distinct from the mentioned cytokines, Hasselgren et al.
curred with parenteral feeding enriched with amino acids
(289) collected plasma from septic rats 16 h after CLP and
and lipids. This was accompanied by fewer autophago-
produced ⬍30-kDa molecular weight fractions to be
somes, fewer intact mitochondria, suppressed respiratory
acutely applied to rat EDL and soleus muscles. Surprisingly,
chain activity, and an increase in markers of organ damage
no proteolysis of myofibrillar proteins was detected, further
in the liver. In muscle, early parenteral nutrition enriched
with amino acids or lipids aggravated vacuolization of myo- blurring the role of a putative circulating proteolytic factor
fibers. Hence, just as in human ICU patients, early paren- in sepsis. To look for specific mechanisms in muscle weak-
teral nutrition during critical illness in this model was ness in response to acute challenge with human sepsis serum
shown to evoke a phenotype of autophagy deficiency in rather than rodent serum, Friedrich et al. (218) collected
skeletal muscle which in human patients was causally re- serum samples from septic ICU patients with a clinical di-
lated to ICU-acquired weakness. From these initial studies, agnosis of CIM and applied various molecular weight frac-
the rabbit burn injury model of prolonged critical illness tions to single murine muscle fibers (217, 218). Acute serum
may be a model worth pursuing to further investigate key fraction applications were able to induce a membrane de-
features of CIM in affected muscles. polarization in time and also reduced caffeine-induced
Ca2⫹-activated force transients. The latter was only seen
with small-molecular-weight fractions ⬍10 kDa but was
G. “Human” Models of ICU-Related blunted for larger molecular weight fractions. This finding
Myopathies: The “Circulating Factor shows that it is important to dissect serum fractions to
Hypothesis” identify active factors that might be present in a biologically
inactivated form in full serum studies. In a later study, van
In a very touching foreword, J. E. Fischer (289) refers to Hees et al. (740) were able to demonstrate a proteolytic
George Clowe’s seminal work in 1983 concerning a prote- effect of plasma from patients with septic shock when ap-
olysis-inducing factor present in plasma from septic pa- plied to C2C12 myotubes.
tients (124). That study, for the first time, showed that
serum from septic patients contained a ⬃4-kDa low-molec- The in vitro septic serum challenge approach has the advan-
ular-weight protein that was able to induce muscle proteol- tage that any cell physiology parameter can be tested for
ysis in vitro (124). This proteolysis-inducing factor (PIF) using established laboratory techniques in a control versus
was later suggested to be a degradation product of IL-1␤ post-exposure setting similar to as in pharmacological ex-
(159). To assess the proteolytic potency of IL-1, its byprod- periments. The use of isolated cells removes many of the
ucts or other cytokines’ ability to induce muscle proteolysis, uncontrolled variables usually present in in vivo experi-
a series of studies aimed at applying serum, serum fractions, ments. However, major disadvantages involve the inability

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1081


FRIEDRICH ET AL.

to model specific trigger factors inherent to ICUAW, and electrophysiological findings were accompanied by a re-
results are often very inhomogeneous due to the diversity of duced need for prolonged mechanical ventilation. These
the underlying patient population, and even the time points results were confirmed in 420 long-stay medical patients
samples are retrieved from patients. In conjunction with the (309). Meta-analysis of both trials reported a relative risk
small number of patients usually involved, the statistical for developing neuromuscular complication with IIT of
power remains limited in most studies. 0.65 (304). Benefits were mainly attributed to glycemic con-
trol rather than insulin dose (735). In a post hoc analysis,
So far, some studies point towards an existing “myotoxic” the beneficial effects were only present in patients in whom
factor systemically circulating in sepsis and systemic inflam- actual normalization of glycemia was reached but not in the
mation. Whether this is also true for the controlled setting subgroup of patients reaching intermediate glycemic targets
of CIM, i.e., for pure mechanical ventilation and immobi- (110 –150 mg/dl) (736). Similar findings were made in a
lization, is currently not known. For this, the animal models retrospective study on the incidence of electrophysiological
described in detail above would need to serve as serum abnormalities before and after institution of IIT, reflecting
source (porcine/rodent ICU model) for subsequent in vitro daily care practice (306). Insulin resistance is well known to
testing. One hypothesis could be that such a circulating occur in critically ill patients and seems more pronounced in
factor was in fact produced by the lungs in the setting of patients with CIM (765). Insufficient GLUT-4 transloca-
mechanical ventilation. Whatever the source of such a fac- tion to the sarcolemma may result in decreased glucose
tor, the identity of it has not been resolved yet, even after supply in patients. Mechanistic studies based on muscle
more than 30 yr of its original proposition. biopsies from insulin trials indicated that despite improving
insulin resistance and skeletal muscle GLUT-4 expression
TABLE 6 compares the properties, the strengths, and the (483), IIT basically did not affect myofiber size, myofibrillar
weaknesses of each model presented in this section. protein synthesis capacity, or markers of muscle proteolysis
(154). This could indicate that benefits were mainly ex-
XIII. TREATMENT OF ICUAW: DRUG plained by neuroprotection, although no nerve histology
INTERVENTIONS AND CLINICAL data are available to confirm this. Generalizing treatment of
IMPLICATIONS critically ill patients towards reaching normoglycemia was
questioned by the NICE SUGAR trial. This multicenter
RCT pointed at the risks of broad implementation of IIT in
A. Intensified Insulin Treatment
critically ill patients, including higher incidence of hypogly-
cemia and even increased mortality (197). Various method-
Effective strategies to prevent and/or treat ICUAW (CIM,
ological differences between trials may be responsible for
sepsis-induced myopathies) are still scarce. These strategies
the distinct results (734). The discrepancy between results
mainly focused on avoiding or reducing risk factors (303).
As neuromuscular ICU complications are associated with of the large trials has come to the widespread acceptance
sepsis, hyperglycemia, prolonged immobilization, and du- that hyperglycemia should be avoided (152), but the actual
ration of mechanical ventilation as well as the use of corti- blood glucose target to aim for depends on local expertise
costeroids and neuromuscular blocking agents, avoiding and monitoring tools (733).
these is theoretically of interest (which is probably not fea-
sible for the mechanical ventilation unless external lung
B. Early Mobilization Strategies
assist devices are being considered). Also, strategies that
avert the catabolic state associated with critical illness are
appealing and could be beneficial. The strongest data cur- Reducing the duration of immobilization could also con-
rently available concern the treatment of hyperglycemia ceptually reduce the incidence of ICUAW, since bed rest and
that inevitably accompanies acute illness. Several RCTs on mechanical unloading induces a catabolic state, muscle at-
glycemic control in the ICU were recently performed (197, rophy, and loss of strength (114, 194, 373, 560). A first
557, 736, 738, 748). Two of these, comparing normaliza- logical approach to shorten the duration of immobilization
tion of glycemia using insulin (glycemic target 80 –110 mg/ is to apply strategies aimed at reducing sedation. During the
dl: intensive insulin therapy, IIT) in critically ill patients, last decade, multiple beneficial effects of such interventions,
versus tolerating hyperglycemia up to the renal threshold including reduced duration of mechanical ventilation and
(glycemia tolerated up to 215 mg/dl: conventional insulin ICU stay as well as less delirium, were described (38, 378,
therapy, CIT) also evaluated the neuromuscular effects in 633). Muscle function, however, was not within the pri-
the subgroup of patients staying in ICU for at least 1 wk. In mary or secondary outcomes tested. Shortening duration of
the 405 long-stay surgical patients, electrophysiological immobilization can also be pursued by applying early phys-
screening revealed that IIT reduced the incidence of critical iotherapy aiming at mobilizing limbs in the acute phase of
illness polyneuromyopathy, diagnosed by the presence of illness despite receiving life support treatments. This ap-
abundant spontaneous electrical activity, from 49 to 25% pears to be safe and feasible in the ICU (27, 429, 495, 551),
(735). Muscle strength data were not available, but the and a nonrandomized trial suggested beneficial effects on

1082 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

Table 6. Comparison of various animal models to study mechanisms in ICU-related myopathies


Model Manipulations Triggers Modeled Pathological Changes Replicated Strengths Weaknesses

Rat steroid Lesion to the sciatic Neuromuscular blockade Muscle atrophy (377) Easy to perform Does not model risk
denervation nerve, daily factors such as
administration of sepsis, loss of
corticosteroids movement
Corticosteroid treatment Preferential loss of myosin (377) Inexpensive
Immobilization not Disorganization of sarcomeres (377) Models all pathological
modeled changes seen in
CIM patients
Electrical hypo/inexcitability (582)
Rat ICU Infusion of Immobilization Muscle atrophy (518) Models atrophy and Labor intensive,
neuromuscular preferential myosin difficult to
blocking agents, loss perform
mechanical
ventilation
together with
treatments such
as LPS and/or
corticosteroids
Neuromuscular blockade Preferential loss of myosin (518) Muscle-specific
differences
observed in ICU
patients with CIM
Mechanical ventilation Disorganization of sarcomeres Suitable for
intervention studies
Sepsis Similar muscle-specific differences as
in ICU patients with CIM
Corticosteroid treatment
Porcine ICU Infusion of Immobility Muscle atrophy (517) Pigs as good model Labor intensive,
neuromuscular for human difficult to
blocking agents, physiology perform
mechanical
ventilation
Neuromuscular blockade Preferential loss of myosin (517) Models all triggers Expensive
Mechanical ventilation Decreased muscle membrane Reproduces atrophy,
excitability in mechanically preferential myosin
ventilated and immobilized animals loss and muscle
(independent of the addition of membrane
sepsis or systemic steroid hypoexcitability
administration) (517)
Corticosteroid treatment Similar muscle-specific differences as Muscle-specific
in ICU patients with CIM differences
observed in ICU
patients with CIM
Sepsis Suitable for
intervention studies
Cecum ligation and Median abdominal Various degrees of Muscle atrophy Simple to perform Animals not
puncture (CLP) laparotomy, sepsis (mild, immobilized
mobilization of moderate, severe),
cecum, suture- septic shock,
ligation of cecum depending on ligation
and needle length/level and
puncture, puncture number
relocation and
closure (588)
Myofibrillar protein loss (776) High reproducibility No mechanical
and grading of ventilation
sepsis severity
when sticking to
standardized
protocols (588)
Disorganization of sarcomeres (776) Systemic response Transferability to
and cytokine surge human
within 4–6 h problematic
postprocedure
Decrease in twitch/tetanic force
(595)

Continued

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1083


FRIEDRICH ET AL.

Table 6. Continued
Model Manipulations Triggers Modeled Pathological Changes Replicated Strengths Weaknesses

Muscular hypoexcitability (Na⫹


channel inactivation increased)
(594)
Mitochondrial dysfunction (543)
Lipopolysaccharide Single-dose Dose-dependent degrees Muscle atrophy (678) Simple to perform Animals not
challenge (LPS) injections of of sepsis immobilized
purified LPS from
gram-negative
bacteria (e.g., E.
coli) sc, ip, or iv
Mild ⬍0.5 mg/kg body Myofibrillar protein loss (317) Systemic response No mechanical
wt and cytokine surge ventilation
within 4–6 h
postprocedure
Moderate 0.5–5 mg/kg Decrease in twitch/tetanic force Use of genetically Transferability to
body wt (678) modified mice humans
enables study of problematic
LPS receptors and (628)
innate immune
activation
Severe (lethal) ⬎5–10 Muscular hypoexcitability (Na⫹ Easy to design studies Discrepancy
mg/kg body wt channel inactivation increased) for drug testing between LPS
(283) sensitivity of mice
and humans
Mitochondrial dysfunction (236) Systemic activation
of the innate
immune system
Rabbit burn injury Third degree burn Catheterization Muscle atrophy (156) Strongly mimics Labor intensive
model injury on 15–20% endocrine and
body surface metabolic
area alterations of
critically ill patients
(183,766)
Parenteral nutrition Ubiquitin proteasome activation High reproducibility No mechanical
(156,178) ventilation
Glucose Impaired autophagy activation with
monitoring/control (parenteral) feeding (156)
Hyperinflammation
Immobility due to
sickness
ICU patient in vitro Serum fractions Tests for a putatively Muscle atrophy (740) Easy to perform Does not model a
serum challenge from ICU patients myotoxic, systemically similar to drug single trigger
model are applied to circulating factor application factor
skeletal muscle experiments
cells (from
animals) and
acute or long-
term effects
studied in vitro
Preferential myosin loss (740) Defined in vitro Very
settings of single inhomogeneous
cells results profile
due to varying
patient cohorts
Decrease in chemically activated Controls (prior to Only studies with
force in skinned fibers (218) serum application) very small sample
well established sizes, patient
within labs samples not
readily available
Muscle membrane depolarization
(178, 218)
Ubiquitin proteasome activation
(740)

ICU and hospital length of stay (495). One RCT compared cluded patients who were expected to have an ICU stay for
20 min of daily bedside cycling exercise from the fifth day in another week. The incidence of weakness was not reported
the ICU in addition to standard physiotherapy with stan- in this study, but isometric quadriceps force as well as func-
dard physiotherapy alone in 90 patients (88). The trial in- tional status, measured by the 6-min walking distance at

1084 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

hospital discharge, was significantly higher in the interven- the ICU recently emerged. One trial in 140 patients reported
tion group. a decrease in incidence of ICUAW from 39.3 to 12.5%,
accompanied by a shorter time to weaning in the interven-
Combining both approaches of minimizing sedation with tion group (598). These data should be interpreted with
early mobilization was examined in a second RCT in criti- caution, since significant differences for baseline character-
cally ill patients who were on mechanical ventilation for istics, APACHE II, and certain comorbidities were present
⬍72 h (623). Early exercise and mobilization were com- between groups in the subset of actually evaluated patients
pared with standard care in 104 patients receiving daily (357). Another RCT in COPD patients in the ICU found no
sedative interruption. This trial showed that early exercise incidence of ICUAW in EMS-treated patients. Moreover,
and mobilization resulted in better functional outcome at quadriceps strength, measured using dynamometry, and
hospital discharge, shorter duration of delirium, and more 6-min walking distance were increased in the EMS group
ventilator-free days. There was no significant reduction in (3). EMS improved muscle strength and decreased number
the incidence of ICUAW and, therefore, the intervention of days needed to transfer from bed to chair in mechanically
may have helped patients to cope with weakness rather than ventilated COPD patients (797). No formal assessment for
improving muscle strength itself. These are promising find- ICUAW was made. Sixteen septic, mechanically ventilated
ings, but several barriers against the use of early rehabilita- patients were randomized to receive unilateral EMS versus
tion may impair widespread implementation of such prac- no EMS on the contralateral limbs. Muscle strength was
tice (410, 551, 796). Some of these, including high severity better on the stimulated side (589). In conclusion, data on
of illness, may not be modifiable. Others, such as depth of EMS are promising but remain scarce and, due to several
sedation (796) and mental attitude of caregivers (320), can methodological issues, such as small sample size and incom-
be addressed in training programs. Optimal implementa- plete outcomes, are prone to bias and should be confirmed
tion of rehabilitation programs and resources will require in larger trials.
confirmation of beneficial effects, identification of the pa-
tient population that benefits, and definition of appropriate
intensity of treatment. In a retrospective analysis of 49 pa- D. Sepsis Treatment
tients who failed to wean from mechanical ventilation for at
least 14 days, whole body rehabilitation and respiratory As a key factor, aggressive treatment of sepsis to avoid
muscle training showed to improve strength, weaning out- ICUAW is generally advocated. In addition to the general
come, and functional status (459). guidelines for sepsis treatment (152), modulating the in-
flammatory response as a potential mechanistical pathway
Finally, several beneficial effects of rehabilitation strategies may be of special interest with regard to the neuromuscular
following hospital discharge have been reported, although complications. Overall effects of anti-inflammatory and im-
based on limited data (128). It remains unclear to what mune-directed therapies in sepsis appeared disappointing
extent such strategies may promote recovery from ICUAW, (242, 745), although individualized approaches, including
as this question has not yet been addressed. optimal timing of such strategies, may be promising (243,
323). No trial has yet specifically addressed the effects of the
immune-modulatory treatments on neuromuscular compli-
C. Electrical Muscle Stimulation cations, and this should be considered in future studies.

Some patients may not be able to actively mobilize during


the acute phase, and electrical muscle stimulation (EMS) E. Corticosteroid Management
may therefore be another method to preserve muscle mass
and function in such a setting. Preliminary data obtained The use of corticosteroids, although also mentioned as a
from muscle biopsies of five patients at risk for ICUAW risk factor, is important to consider, as corticosteroids may
receiving unilateral EMS showed that EMS could improve reduce duration of organ dysfunction in a subgroup of pa-
AMPK activation, glucose utilization, and GLUT-4 trans- tients with septic shock (22), which by itself is related to the
location (765). The stimulated leg also showed larger cross- development of ICUAW. Moreover, when concomitant hy-
sectional area for type 2, but not type 1, fibers. In non-ICU perglycemia is treated, steroids might have beneficial neu-
patient populations, EMS has been shown to increase mus- romuscular effects in critically ill patients, possibly due to
cle strength and exercise tolerance (637). Preliminary re- anti-inflammatory properties (309). Several RCTs compar-
sults in small samples of critically ill patients were promis- ing corticosteroids versus placebo were performed in criti-
ing and found that EMS preserved muscle mass (237, 272, cally ill patients, but only few reported on neuromuscular
474), reduced muscle catabolism (73), and exerted short- complications. The Corticus trial found no effect of corti-
term systemic microcirculatory effects (238). In another costeroids on the incidence of polyneuropathy in 499 pa-
eight patients with septic shock however, 7 days of EMS did tients with septic shock (654). However, no diagnostic
not preserve muscle volume (555). Several randomized methods for polyneuropathy were provided, and the figures
studies examining the effects of EMS on muscle function in were unexpectedly low in both groups. Another RCT fo-

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1085


FRIEDRICH ET AL.

cused on 180 patients with persisting ARDS and compared risk for developing ICUAW, as well as in a random sample
treatment with corticosteroids (methylprednisolone) versus of short-stay patients (300). The evaluations were per-
placebo (658). There was no difference in the incidence of formed three times weekly from day eight until ICU dis-
neuromyopathy between both intervention groups. Meth- charge or death. The incidence of ICUAW at first evaluation
ylprednisolone increased the number of ventilator-free and was significantly lower in the late PN group compared with
shock-free days during the first 28 days and did not affect the early PN group. Muscle biopsies from 122 EPaNIC
mortality. Serious adverse events related to neuropathy or patients showed that this was possibly due to more efficient
myopathy, however, were reported in nine patients in the autophagic qualtity control of myofibers, whereas markers
steroid group versus none in the control group. In some of atrophy were not affected. Another large RCT focused
clinical situations, such as status asthmaticus and systemic on a specific subgroup of ICU patients, deemed to have
vasculitis, corticosteroids are potentially life-saving, and contraindications for enteral feeding (161). This trial ran-
possible side effects should not compromise their prompt domized 1,372 subjects to conventional treatment or early
institution. In other situations including septic shock (654) parenteral nutrition and indicated greater muscle wasting in
or ARDS (658), benefits are less clear and should be the standard care patients, but no assessment of muscle
weighed against possible side effects, including weakness. strength or functional outcome was made (161). The Eden
trial studied full enteral versus trophic feeding during the
first 6 days in 1,000 acute lung injury patients (503). No
F. Management of Neuromuscular Blockade strength measurements were provided at ICU or hospital
discharge, but at 6 and 12 mo follow-up, physical perfor-
Concerning the use of neuromuscular blocking agents, only mance was below the predicted one for a subset of 174
one RCT studied the effect on ICUAW (536). No difference survivors. No difference was found for MRC sum score,
in incidence of ICUAW was found in patients with early handgrip strength, maximal inspiratory pressure, 6-mo
ARDS receiving neuromuscular blocking agents for 48 h walking distance, and quality of life (504). Various supple-
versus controls. Neuromuscular blocking agents are still mental therapies may be of interest. Glutamine concentra-
being used, although much less frequently and more selec- tion in plasma and skeletal muscle are low in critical illness,
tively to date, for several indications such as urgent intuba- which independently predicts mortality. This led to the hy-
tion, patient-ventilator asynchrony, refractory respiratory pothesis that glutamine is a conditionally essential amino
failure, intracranial hypertension, and therapeutic hypo- acid and that supplementation may not only improve over-
thermia (265). Despite the lack of equivocal evidence of all outcome but also muscle function (87, 770). A meta-
negative neuromuscular effects, it is generally accepted to analysis reported beneficial effects of glutamine supplemen-
limit use of neuromuscular blocking agents (146) and when tation in critically ill patients (514), but this was not con-
used, to evaluate depth of neuromuscular blockade apply- firmed in two recent RCTs (18, 312). The latter even
ing monitoring techniques (744). showed increased mortality rates in critically ill patients
with multiple organ failure (312), and effects on muscle
function have not been studied yet. The use of other micro-
G. Nutritional Strategies nutrients against oxidative stress is also sensible from a
pathophysiological point of view. Two recent meta-analy-
Severe muscle atrophy due to the catabolic state in critically ill ses concluded that antioxidant micronutrients are of poten-
patients is generally considered to contribute to muscle weak- tial benefit for critically ill patients (452, 746). Again, ef-
ness. A nutritional deficit rapidly develops during critical ill- fects on muscle weakness were not studied, and the recent
ness due to dysfunction of the gastrointestinal tract. This is large RCT showed no beneficial effects from antioxidants in
most troublesome in patients who cannot be fed enterally be- critically ill patients with multiple organ failure (312). A
cause of contraindications. Postoperative esophagectomy or recent review concluded that there is no evidence for benefit
pancreato-duodenectomy patients received no enteral feeding with early nutrition, nor supplemental therapies in critically
in the first 6 days versus enteral feeding through a jejunos- ill patients (108). Preliminary data from animal experi-
tomy. No effect on grip strength or respiratory muscle ments showed that mitochondrial emission of ROS is a
strength was noted, and the fed patients tended to have less crucial element in atrophy development and contractile dys-
rapid recovery of mobility (762). The EPaNIC trial ran- function of the diaphragm occurring during mechanical
domized 4,640 patients for early parenteral supplementa- ventilation (556) which was prevented by a novel mito-
tion (early PN group) versus tolerating the caloric deficit chondria-targeted antioxidant.
during the first week in ICU (late PN group) (107). Overall,
accelerated recovery, shortened duration of mechanical
ventilation, and fewer complications were shown for the H. Hormone and Electrolyte Balance
late PN group compared with the early PN group. Addi-
tionally, systematic evaluation of muscle strength using the Several other hormones have a key role in the anabolic/
MRC sum score was performed in patients who stayed in catabolic balance of the muscle and could, therefore, possi-
ICU for at least 8 days and were considered to be at high bly be of interest in the prevention of ICUAW. One of the

1086 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

Table 7. Therapeutic strategies for ICU-related weakness


Presumed
Level of Evidence Target Presumed Target at Cellular/Molecular Level

Direct evidence for benefit


Tight glycemic control Nerve Improved microcirculation (E) (183, 393, 781)
Improved mitochondrial function (E) (735)
Attenuated loss of nerve fibers (E) (603)
Withholding parenteral nutrition during first Muscle Improved autophagic muscle quality control (A) (300)
week in ICU
Indirect evidence for benefit
Sedation sparing protocol Muscle Prevention of disuse atrophy caused by decreased protein
synthesis, anabolic resistance, and increased protein
degradation by the ubiquitin proteasome system (C, D) (81,
260)
Early limb mobilization Muscle Autophagy (C, D) (294, 689)
Potential evidence for harm: limit use unless evidence-based benefit
Corticosteroids Muscle Promoting atrophy by decreased protein synthesis and
increased proteolysis by the ubiquitin proteasome and
lysosomal system (A, B) (154, 616, 617, 703)
Neuromuscular blocking agents NMJ Prolonged neuromuscular blockade (A) (265)
Muscle Denervation atrophy (D)
Theoretical benefit
Careful electrolyte management (Na⫹, K⫹, Nerve Nerve membrane excitability (E)
P, Mg2⫹, Ca2⫹)
NMJ Neuromuscular transmission (E)
Muscle Muscle membrane excitability (E); excitation-contraction
coupling (E)
Ventilator weaning protocol Muscle Avoiding fatigue (E) (387)
Electrical muscle stimulation Muscle Prevention of atrophy by suppression of ubiquitin proteasome
system and calpain and increased anabolic pathways by
induction of growth factors (A, C, D) (73, 171, 467, 664,
765)
Autophagy (E)

The level of available evidence is graded as follows: A) human data from critically ill patients; B) experimental
data from ICU models; C) human data from noncritically ill or healthy volunteers; D) experimental data from
other animal models, including immobilization/denervation; E) hypothesis.

candidates to this effect was growth hormone, since resis- ally is not primarily determined by muscle function (768).
tance to growth hormone is a characteristic feature of crit- Also, dedicated weaning protocols might be indicated for
ical illness. Two large randomized trials showed clearly in- weak patients (80) to avoid muscle fatigue (387), although
creased mortality rates in patients treated with growth hor- no clinical data are available in this specific group of criti-
mone (688). Although the interest in the potential of growth cally ill patients.
hormone treatment in the recovery phase for selected
chronically ill patients is reviving (694), current recommen- In summary, data on effective strategies to prevent and/or
dations are clearly against its use. Low sex hormone con- treat muscle weakness in critical illness remain limited,
centration in aging may be a key factor in sarcopenia and mainly due to lack of well-designed large RCTs. Further
muscle weakness (642). ICUAW is also associated with hy- research is clearly needed. In particular, strategies to re-
pogonadism in men (631). Testosterone derivatives reduced duce hypoglycemic events during insulin treatment
length of stay and preserved lean body mass in burn patients should be pursued, as well as further exploring the po-
(352, 783). Data are not supportive for use in trauma (239) tential of early rehabilitation and electrical muscle stim-
or surgery (84) patients, although effects on ICUAW have ulation and other treatments should be addressed, sug-
not formally been addressed. Also, concerns about hepato- gested from targeting pathogenetically involved path-
toxicity with the use of oxandrolone, an oral synthetic tes- ways in this review. TABLE 7 summarizes current views
tosterone derivative, have been raised (352). Finally, elec- on strategies with direct, indirect, and potential evidence
trolyte disturbances could affect muscle function (768). The for therapeutic benefit as well as evidence for harmful
need for correction, rate, and degree to which electrolytes outcome and a list of some future challenges is provided
should be corrected depends on multiple factors and gener- at the end of the next chapter.

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1087


FRIEDRICH ET AL.

nerve pathology muscle pathology

Critical illness +
cytokine production

electrical microvascular metabolic bioenergetic electrical microvascular altered Ca2+ metabolic bioenergetic mechanical
alterations alterations alterations failure alterations alterations homeostasis alterations failure silencing

hyperglycemia

vasodialation + impairment ↑generation inactivation vasodialation + ↑activation ↑apoptosis relative ↑anabolic + antioxidant tensegrity
↑permability microcirculation ↓scavenging of Na+ ↑permability of glutamine ↓catabolic depletion impairment
of the ROS channels of the proteolytic deficiency hormones
microcirculation microcirculation (calpain +
ubiquitin +
lysosomal)
pathways

nerve passage endo- mitochondrial muscle change protein mitochondrial aberrant


extra- extra- mechano-
mem- neuro- vasation neural dysfunction membrane in the catabolism dysfunction
vasation transduction
brane toxic of edema + inexcit- of excitation-
depolar- factors activated hypox- ability activated contraction
ization leukocytes emia leukocytes coupling
+ local + local of the
cytokine cytokine SR
production production

CIP muscle denervation CIM

NMBA CS

FIGURE 16. Flow chart of proposed mechanisms and their interactions contributing to the development of
CIP and CIM. [Extended from Hermans et al. (302).] Summary of pathophysiological mechanisms in ICU-related
weakness affecting the nerve/muscle function, demonstrating the various pathways affected and mechanistic
explanations for the development of CIP and CIM.

XIV. SUMMARY/OUTLOOK may be used, but it is mandatory to perform a focused


search for features 3 and 5, in particular because the pres-
In this review, we have discussed the different entities of ence of preferential myosin loss seems, so far, to be the only
ICU-related weakness as well as potential mechanisms in- distinction between CIM and sepsis-induced myopathy,
herent to and differentiating between those entities. One and the presence of impaired autophagy may alter the clin-
important emphasis of the current consortium is to stress ical handling with an initial calorie restriction protocol.
the fact that inconsistencies exist in the literature regarding Strong symptoms arguing for the presence of CIM in most
the classification of weakness in general and myopathies in patients are reduced membrane excitability and preferential
particular, seen in critically ill patients. For critical illness myosin loss.
myopathy to be diagnosed appropriately, several patholog-
ical key features shall be present: 1) electrical hypoexcitabil- The choice of appropriate animal models mimicking the
ity of muscles, 2) severe atrophy, 3) preferential and signif- situation in ICU patients is crucial for current and future
icant myosin loss, 4) disorganization of sarcomeres and studies of missing links in the pathophysiological puzzle
ultrastructural abnormalities, and 5) impaired autophagy. and to test for treatment efficiencies. While immobilization
These findings may be present between patients in combi- is clearly a key risk factor for development of ICUAW and
nations of varying degree. Unless all of those features are CIM in particular, it is likely that there may also be a con-
confirmed, the definite diagnosis “CIM” does not hold and tribution of an ongoing inflammatory process. As detailed
the more pragmatic term “ICUAW” shall be used. How- in our review, the pathophysiological mechanisms found in
ever, it needs further clarification whether the finding of neuropathies and myopathies in the critically ill are very
impaired autophagy exclusively applies to CIM patients or diverse and affect all facets of neuron and muscle function
also to other subsets of ICUAW. If some of these features (FIGURE 16). One unique complication of critical illness
are present, e.g., most clinical routine diagnostics will be appears to be an acquired channelopathy that affects mul-
able to cover features 1, 2, and 4, the term probable CIM tiple electrically active tissues and may contribute to myop-

1088 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

athy, neuropathy, cardiac failure, and poor recruitment of weakness in the critically ill as well as test for novel poten-
motor units. However, whether this ion channel dysfunc- tial therapeutic interventions include the following.
tion is a primary pathology or simply a consequence sec-
ondary to alterations rendering the ion channel dysfunc- 1. Animal models and human experiments
tional, e.g., electrolyte disturbances, metabolic failure, pro-
teolytic activity in critical illness, etc., is not known and 1) Elucidating the exact time courses of individual phenotypic
represents a future field of studies where the animal models changes in CIM, e.g., beginning and duration of membrane
discussed in this review may become extremely valuable. hypoexcitability, Ca2⫹ imbalance, metabolic changes, atro-
Other pathological processes include impairments of the phy, preferential myosin loss in serial studies in patients and
neuromuscular transmission and postsynaptic membrane animal models (FIGURE 2);
depolarization, so muscle becomes electrically hypo- or in- 2) Using animal models at ages equivalent to human chil-
excitable. Aspects of critical illness related to intracellular dren as a model for pediatric ICU-acquired weakness;
Ca2⫹ homeostasis have only scarcely been investigated in 3) Using animal models with comorbidities (e.g., obese ICU
muscle, while not at all in nerve. Evidence points towards rat model, senescence models, etc.);
substantial alteration in Ca2⫹ handling mechanisms, at 4) Extending the porcine ICU model (as model closest to the
least documented for sepsis-associated weakness. Complete human) for long-term intensive care treatment (also in
mechanical silencing, i.e., no weight bearing and no internal adult/old pigs);
strain related to activation of contractile proteins, is unique 5) Exploiting multimodal readout systems (metabolic imag-
for mechanically ventilated, deeply sedated, or pharmaco- ing, metabolomics, multiplex RNA analyses, etc.) to pa-
logically paralyzed ICU patients and has been shown in tient samples in serial studies on ICU patients, septic or
experimental studies to play a very important role in the not, and septic patients with no mechanical ventilation
pathogenesis of CIM via different mechanosensitive path- or immobilization to map the pathophysiological mech-
ways, but other factors have additive/synergistic effects in anisms found in animal models to human models; and
the development of the CIM phenotype. In addition, the 6) Assessing the Na⫹ channelopathy defined in animal mod-
compromised inherent protective mechanisms of sarco- els in ICU patients. In particular, is the hypoexcitability
meric proteins and mitochondria by heat shock proteins seen in CIM patient muscle also reflected by the same
triggered by systemic corticosteroid hormone administra- changes in ion channel biophysics as seen in rat SD mus-
tion and sepsis, in combination with mechanical silencing, cle? Fresh muscle biopsies from CIM patients would be
play an important role for the impaired muscle function in required to be electrophysiologically characterized (volt-
CIM. Thus interventions targeting mechanosenstive path- age/patch clamp).
ways and heat shock protein expression offer novel venues
for future pharmacological treatments in mechanically ven- 2. Pathophysiological mechanisms
tilated critically ill ICU patients. To date, the only therapeu-
tic strategies with a proven positive outcome include inten- 1) Assessment of complete electrolyte and osmolarity pro-
sified insulin therapy to prevent hyperglycemia and avoid- files in patients from different ICUAW entities to differ-
ing early parenteral nutrition to allow autophagy activation entially explain muscle membrane hypoexcitability;
in muscle. Counteracting immobilization with early mobi- 2) Precise involvement of K⫹, Cl⫺, and Ca2⫹ channels in
lization may also help to reduce the severity of myopathy. muscle membrane excitability in critical illness and al-
tered Ca2⫹ homeostasis;
In summary, from a pathophysiological point of view, the 3) More evidence for the direct intracellular action of in-
dissection of contributing factors to the clinical picture of flammatory cytokines, e.g., direct binding of IL-1 or
ICU-related weakness is still a challenging venue for future TNF-␣ to regulatory proteins in muscle; and
research. Therefore, appropriate animal models such as the 4) Elucidation of the pathways for altered Ca2⫹ homeosta-
ones presented here with their strengths and weaknesses sis on mitochondrial biogenesis and mitochdonrial activ-
will become a major focus for critical illness-related re- ity on ATP outcome in the different animal models of
search and, hopefully, will become also a reliable tool to critical illness and criticially ill patients. Is there a myo-
more specifically test for therapeutic benefits of new treat- toxic circulating factor in serum collected from non-sep-
ment regimes. tic animal models of ICUAW?

3. Therapeutic concepts to test for


A. Future Research Topics (Basic
Mechanisms and Possible Therapeutic 1) Determination of the impact of glycemic control on au-
Interventions) tophagy in skeletal muscle;
2) Design of strategies for immune modulation in septic
Future research topics that will certainly represent an ad- patients and animal models to suppress overt systemic
vancement in our understaning of the nature of muscle inflammatory reaction and to avoid ICUAW;

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1089


FRIEDRICH ET AL.

3) Follow-up the concept of reduced membrane excitability 5. Adhihetty PJ, O’Leary MFN, Chabi B, Wicks KL, Hood DA. Effect of denervation on
mitochondrially mediated apoptosis in skeletal muscle. J Appl Physiol 102: 1143–1151,
to potentially increase survival of depolarized injured 2007.
cells (see sect. VIII).
6. Ahlbeck K, Fredriksson K, Rooyackers O, Maback G, Remahl S, Ansved T, Eriksson L,
Radell P. Signs of critical illness polyneuropathy and myopathy can be seen early in the
ACKNOWLEDGMENTS ICU course. Acta Anaesthesiol Scand 53: 717–723, 2009.

7. Ahmad S, Choudhry MA, Sayeed MM. Ca2⫹-dependent and Ca2⫹-independent pro-


Address for reprint requests and other correspondence: O. teinase contents in the skeletal muscle in septic rats. Shock 5: 247–250, 1996.
Friedrich, Institute of Medical Biotechnology, Department
8. Akkad H, Corpeno R, Larsson L. Masseter muscle myofibrillar protein synthesis and
of Chemical and Biological Engineering, Friedrich-Alexan- degradation in an experimental critical illness myopathy model. PLoS One 9: 92622,
der-University Erlangen-Nuremberg, Paul-Gordan-Str. 3, 2014.
91052 Erlangen, Germany (e-mail: oliver.friedrich@mbt.
9. Alamdari N, Smith IJ, Aversa Z, Hasselgren PO. Sepsis and glucocorticoids upregulate
uni-erlangen.de). p300 and downregulate HDAC6 expression and activity in skeletal muscle. Am J
Physiol Regul Integr Comp Physiol 299: R509 –R520, 2010.
GRANTS 10. Ali NA, O’Brien JM Jr, Hoffmann SP, Phillips G, Garland A, Finley JC, Almoosa K, Hejal
R, Wolf KM, Lemeshow S, Connors AF Jr, Marsh CB. Acquired weakness, handgrip
This work was supported by National Institutes of Health strength, and mortality in critically ill patients. Am J Respir Crit Care Med 178: 261–268,
2008.
Grant NS082354 (to M. Rich) and AR062083 (to M. B.
Reid); Research Foundation-Flanders (Fonds voor Weten- 11. Allaouchiche B, Duflo F, Debon R, Tournadre JP, Chassard D. Influence of sepsis on
minimum alveolar concentration of desflurane in a porcine model. Br J Anaesth 87:
schappelijk Onderzoek Vlaanderen), Belgium, Grants 280 –283, 2001.
G.0399.12 and G.0592.12 (to G. Hermans, I. Vanhore-
12. Allen DC, Arunachalam R, Mills KR. Critical illness myopathy: further evidence from
beek, and G. Van den Berghe); Postdoctoral Fellowship
muscle-fiber excitability studies of an acquired channelopathy. Muscle Nerve 37: 14 –
from the Clinical Research Fund (Klinische Onderzoeks- 22, 2008.
fonds, KOF) of the University Hospitals Leuven, Belgium
13. Allen DG, Lamg GD, Westerblad H. Skeletal muscle fatigue: cellular mechanisms. Phys
(to G. Hermans); structural research financing via the Rev 88: 287–332, 2008.
Methusalem program funded by the Flemish Government
14. Alloati G, Penna C, Mariano F, Camussi G. Role of NO and PAF in the impairment of
(METH08/07 to G. Van den Berghe and METH14/06 to G. skeletal muscle contractility induced by TNF-␣. Am J Physiol Regul Integr Comp Physiol
Van den Berghe and I. Vanhorebeek), via the University of 279: R2156 –R2163, 2000.
Leuven; and ERC Advanced Grant AdvG-2012-321670
15. Almers W, Stanfield PR, Stühmer W. Lateral distribution of sodium and potassium
from the Ideas Program of the EU FP7 (to G. Van den channels in frog skeletal muscle: measurements with a patch-clamp technique. J
Berghe). The Swedish Medical Research Council (8651), Physiol 336: 261–284, 1983.
the European Commission (MyoAge, EC Fp7 CT-223756, 16. Alvarez S, Boveris A. Mitochondrial nitric oxide metabolism in rat muscle during
and COST CM1001), the Swedish Foundation for Interna- endotoxemia. Free Rad Biol Med 37: 1472–1478, 2004.
tional Cooperation in Research and Higher Educatiuon 17. Anastasopoulos D, Kefaliakos A, Michalopoulos A. Is plasma calcium concentration
(STINT), and Uppsala University Hospital provided sup- implicated in the development of critical illness polyneuropathy and myopathy? Crit
port to L. Larsson. Care 15: R247, 2011.

18. Andrews PJ, Avenell A, Noble DW, Campbell MK, Croal BL, Simpson WG, Vale LD,
Battison CG, Jenkinson DJ, Cook JA. Randomised trial of glutamine, selenium, or both,
DISCLOSURES to supplement parenteral nutrition for critically ill patients. BMJ 342: d1542, 2011.

19. Angaut-Petit D, McArdle JJ, Mallart A, Bournaud R, Pincon-Raymond M, Rieger F.


No conflicts of interest, financial or otherwise, are declared Electrophysiological and morphological studies of a motor nerve in “motor endplate
by the authors. disease” of the mouse. Proc R Soc Lond B Biol Sci 215: 117–125, 1982.

20. Anker SD, Rauchaus M. Insights into the pathogenesis of chronic heart failure: immune
activation and cachexia. Curr Opin Cardiol 14: 211–216, 1999.
REFERENCES
21. Annane D, Bellissant E, Bollaert PE, Briegel J, Confalonieri M, De GR, Keh D, Kupfer
Y, Oppert M, Meduri GU. Corticosteroids in the treatment of severe sepsis and septic
1. Aare S, Ochala J, Norman HS, Radell P, Eriksson LI, Goransson H, Chen YW, Hoffman shock in adults: a systematic review. JAMA 301: 2362–2375, 2009.
EP, Larsson L. Mechanisms underlying the sparing of masticatory versus limb muscle
function in an experimental critical illness model. Physiol Genomics 43: 1334 –1350, 22. Annane D, Sebille V, Charpentier C, Bollaert PE, Francois B, Korach JM, Capellier G,
2011. Cohen Y, Azoulay E, Troche G, Chaumet-Riffaud P, Bellissant E. Effect of treatment
with low doses of hydrocortisone and fludrocortisone on mortality in patients with
2. Aare S, Radell P, Eriksson LI, Chen YW, Hoffman EP, Larsson L. The role of sepsis in septic shock. JAMA 288: 862– 871, 2002.
the development of limb muscle weakness in a porcine intensive care unit model.
Physiol Genomics 44: 865– 877, 2012. 23. Astiz M, Rackow EC, Weil MH, Schumer W. Early impairment of oxidative metabo-
lism and energy production in severe sepsis. Circ Shock 26: 311–320, 1988.
3. Abdellaoui A, Prefaut C, Gouzi F, Couillard A, Coisy-Quivy M, Hugon G, Molinari N,
Lafontaine T, Jonquet O, Laoudj-Chenivesse D, Hayot M. Skeletal muscle effects of 24. Astrakas LG, Goljer I, Yasahura S, Padfield KE, Zhang Q, Gopalan S, Mindrinos MN,
electrostimulation after COPD exacerbation: a pilot study. Eur Respir J 38: 781–788, Dai G, Yu YM, Martyn JAJ, Tompkins RG, Rahme LG, Tzika A. Proton NMR spectros-
2011. copy shows lipids accumulate in skeletal muscle in response to burn trauma-induced
apoptosis. FASEB J 19: 1431–1440, 2005.
4. Adams V, Nehrhoff B, Späte U, Linke A, Schulze PC, Baur A, Gielen S, Hambrecht R,
Chuler G. Induction of iNOS expression in skeletal muscle by IL-1beta and NFkappaB 25. Attaix D, Baracos VE. MAFbx/Atrogin-1 expression is a poor index of muscle prote-
activation. An in vitro and in vivo study. Cardiovasc Res 54: 95–104, 2002. olysis. Curr Opin Clin Nutr Metab Care 13: 223–224, 2010.

1090 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

26. Authier FJ, Mhiri C, Chazaud B, Christov C, Cherin P, Barlovatz-Meimon G, Gherardi 46. Bengoecchea Gonzalez ME, Perez Casas A, Alvarez Arenal A, Vega Alvarez JA, Tin-
RK. Interleukin-1 expression in inflammatory myopathies: evidence of marked immu- ture Eguren JC. Structure and acetylcholinesterase activity of the neuromuscular
noreactivity in sarcoid granulomas and muscle fibres showing ischaemic and regener- junction of rats in a state of septic shock. Rev Clin Esp 167: 359 –362, 1982.
ative changes. Neuropathol Appl Neurobiol 23: 132–140, 1997.
47. Bennett E, Urcan MS, Tinkle SS, Koszowski AG, Levinson SR. Contribution of sialic
27. Bailey P, Thomsen GE, Spuhler VJ, Blair R, Jewkes J, Bezdjian L, Veale K, Rodriquez L, acid to the voltage dependence of sodium channel gating. A possible electrostatic
Hopkins RO. Early activity is feasible and safe in respiratory failure patients. Crit Care mechanism. J Gen Physiol 109: 327–343, 1997.
Med 35: 139 –145, 2007.
48. Bennett ES. Isoform-specific effects of sialic acid on voltage-dependent Na⫹ channel
28. Baker AJ, Languemare MC, Brandes R, Weiner MW. Intracellular tetanic calcium gating: functional sialic acids are localized to the S5–S6 loop of domain I. J Physiol 538:
signals are reduced in fatigue of whole skeletal muscle. Am J Physiol Cell Physiol 264: 675– 690, 2002.
C577–C582, 1993.
49. Benson DW, Hasselgren PO, Hiyama DT, James JH, Li S, Rigel DF, Fischer JE. Effects
29. Baldwin CE, Paratz JD, Bersten AD. Muscle strength assessment in critically ill patients of sepsis on calcium uptake and content in skeletal muscle and regulation in vitro by
with handheld dynamometry: an investigation of reliability, minimal detectable calcium of total and myofibrillar protein breakdown in control and septic muscle:
change, and time to peak force generation. J Crit Care 28: 77– 86, 2013. results from a preliminary study. Surgery 106: 87–93, 1989.

30. Ballard-Croft C, Maass DL, Sikes PJ, Horton JW. Sepsis and burn complicated by 50. Berchtold MW, Brinkmeier H, Müntener M. Calcium ion in skeletal muscle: its crucial
sepsis alter cardiac transporter expression. Burns 33: 72– 80, 2007. role for muscle function, plasticity, and disease. Phys Rev 80: 1216 –1265, 2000.

31. Banduseela VC, Chen YW, Kultima HG, Norman HS, Aare Radell P S, Eriksson LI, 51. Bercker S, Weber-Carstens S, Deja M, Grimm C, Wolf S, Behse F, Busch T, Falke KJ,
Hoffman EP, Larsson L. Impaired autophagy, chaperone expression, and protein Kaisers U. Critical illness polyneuropathy and myopathy in patients with acute respi-
synthesis in response to critical illness interventions in porcine skeletal muscle. Physiol ratory distress syndrome. Crit Care Med 33: 711–715, 2005.
Genomics 45: 477– 486, 2013.
52. Berek K, Margreiter J, Willeit J, Berek A, Schmutzhard E, Mutz NJ. Polyneuropathies
32. Banduseela VC, Ochala J, Chen YW, Goransson H, Norman H, Radell P, Eriksson LI, in critically ill patients: a prospective evaluation. Intensive Care Med 22: 849 – 855,
Hoffman EP, Larsson L. Gene expression and muscle fiber function in a porcine ICU 1996.
model. Physiol Genomics 39: 141–159, 2009.
53. Bergmann M, Fruton JS. Regarding the general nature of catheptic enzymes. Science
2⫹
33. Bannister RA, Pessah IN, Beam KG. The skeletal L-type Ca current is a major 84: 89 –90, 1936.
contributor to excitation-coupled Ca2⫹ entry. J Gen Physiol 133: 79 –91, 2009.
54. Bernardin G, Strosberg AD, Bernard A, Mattei M, Marullo S. ␤-Adrenergic receptor-
34. Baracos V, Rodemann HP, Dinarello CA, Goldberg AL. Stimulation of muscle protein dependent and -independent stimulation of adenylate cyclase is impaired during se-
degradation and prostaglandin E2 release by leukocytic pyrogen (interleukin-1). A vere sepsis in humans. Intensive Care Med 24: 1315–1322, 1998.
mechanism for the increased degradation of muscle proteins during fever. N Engl J
55. Bertsch S, Lang CH, Vary TC. Inhibition of glycogen synthase kinase 3␤ activity with
Med 308: 553–558, 1983.
lithium in vitro attenuates sepsis-induced changes in muscle protein turnover. Shock
35: 266 –274, 2011.
35. Barat M, Brochet B, Vital C, Mazaux JM, Arne L. Polyneuropathies during prolonged
stays in resuscitation. Rev Neurol 143: 823– 831, 1987. 56. Bhattacharyya J, Thompson K, Sayeed MM. Calcium-dependent and calcium-inde-
pendent protease activities in skeletal muscle during sepsis. Circ Shock 35: 117–122,
36. Barnett RE, Keskey RC, Rao JM, Billeter AT, Kanaan Z, Cheadle WG. Poor outcome
1991.
in bacterial peritonitis is associated with dysregulated microRNAs and an increased
inflammatory response. Surgery 154: 521–527, 2013. 57. Bhattacharyya J, Thompson KD, Sayeed MM. Skeletal muscle Ca2⫹ flux and catabolic
response during sepsis. Am J Physiol Regul Integr Comp Physiol 265: R487–R493, 1993.
37. Barohn RJ, Jackson CE, Rogers SJ, Ridings LW, McVey AL. Prolonged paralysis due to
nondepolarizing neuromuscular blocking agents and corticosteroids. Muscle Nerve 17: 58. Bialek P, Morris C, Parkington J, St Andre M, Owens J, Yaworsky P, Seeherman H,
647– 654, 1994. Jelinsky SA. Distinct protein degradation profiles are induced by different disuse mod-
els of skeletal muscle atrophy. Physiol Genomics 43: 1075–1086, 2011.
38. Barr J, Fraser GL, Puntillo K, Ely EW, Gelinas C, Dasta JF, Davidson JE, Devlin JW,
Kress JP, Joffe AM, Coursin DB, Herr DL, Tung A, Robinson BR, Fontaine DK, Ramsay 59. Bierbrauer J, Koch S, Olbricht C, Hamati J, Lodka D, Schneider J, Luther-Schroder A,
MA, Riker RR, Sessler CN, Pun B, Skrobik Y, Jaeschke R. Clinical practice guidelines Kleber C, Faust K, Wiesener S, Spies CD, Spranger J, Spuler S, Fielitz J, Weber-
for the management of pain, agitation, and delirium in adult patients in the intensive Carstens S. Early type II fiber atrophy in intensive care unit patients with nonexcitable
care unit. Crit Care Med 41: 263–306, 2013. muscle membrane. Crit Care Med 40: 647– 650, 2012.

39. Barrett MJ, Goll DE, Thompson VF. Effect of substrate on Ca2⫹ concentration re- 60. Boczkowski J, Lanone S, Ungureanu-Longrois D, Danialou G, Fournier T, Aubier M.
quired for activity of the Ca2⫹-dependent proteinases, mu- and m-calpain. Life Sci 48: Induction of diaphragmatic nitric oxide synthase after endotoxin administration in
1659 –1669, 1991. rats: role on diaphragmatic contractile dysfunction. J Clin Invest 98: 1550 –1559, 1996.

40. Bartlett RH, Roloff DW, Custer JR, Younger JG, Hirschl RB. Extracorporeal life sup- 61. Boczkowski J, Lisdero CL, Lanone S, Samb A, Carreras MC, Boveris A, Aubier M,
port: the University of Michigan experience. JAMA 283: 904 –908, 2000. Poderoso JJ. Endogenous peroxynitrite mediates mitochondrial dysfunction in rat
diaphragm during endotoxemia. FASEB J 13: 1637–1646, 1999.
41. Baylor SM, Hollingworth S. Sarcoplasmic reticulum calcium release compared in
slow-twitch and fast-twitch fibers of mouse muscle. J Physiol 551: 125–138, 2003. 62. Boekstegers P, Weidenhöfer S, Kapsner T, Werdan K. Skeletal muscle partial pres-
sure of oxygen in patients with sepsis. Crit Care Med 22: 640 – 650, 1994.
42. Bechet D, Tassa A, Taillandier D, Combaret L, Attaix D. Lysosomal proteolysis in
skeletal muscle. Int J Biochem Cell Biol 37: 2098 –2114, 2005. 63. Boekstegers P, Weidenhöfer S, Pilz G, Werdan K. Peripheral oxygen availability within
skeletal muscle in sepsis and septic shock: comparison to limited infection and car-
43. Bednarik J, Lukas Z, Vondracek P. Critical illness polyneuromyopathy: the electro- diogenic shock. Infection 19: 317–323, 1991.
physiological components of a complex entity. Intensive Care Med 29: 1505–1514,
2003. 64. Bolton CF. Neuromuscular manifestations of critical illness. Muscle Nerve 32: 140 –
163, 2005.
44. Bednarik J, Vondracek P, Dusek L, Moravcova E, Cundrle I. Risk factors for critical
illness polyneuromyopathy. J Neurol 252: 343–351, 2005. 65. Bolton CF. The discovery of critical illness polyneuropathy: a memoir. Can J Neurol Sci
37: 431– 438, 2010.
45. Bendahhou S, Cummins TR, Potts JF, Tong J, Agnew WS. Serine-1321-independent
regulation of the mu1 adult skeletal muscle Na⫹ channel by protein kinase C. Proc Natl 66. Bolton CF, Brown JD, Sibbald WA. The electrophysiologic investigation of respiratory
Acad Sci USA 92: 12003–12007, 1995. paralysis in critically ill patients. Neurology 33: 186, 1983.

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1091


FRIEDRICH ET AL.

67. Bolton CF, Gilbert JJ, Hahn AF, Sibbald WJ. Polyneuropathy in critically ill patients. J 89. Bustamante M, Fernandez-Verdejo R, Jaimovich E, Buvinic S. Electrical stimulation
Neurol Neurosurg Psychiatry 47: 1223–1231, 1984. induces interleukin-6 in skeletal muscle through extracellular AT, by activating Ca2⫹
signals and an interleukin-6 autocrine loop. Am J Physiol Endocrinol Metab 306: E869 –
68. Bolton CF, Laverty DA, Brown JD, Witt NJ, Hahn AF, Sibbald WJ. Critically ill poly- E882, 2014.
neuropathy: electrophysiological studies and differentiation from Guillain-Barre syn-
drome. J Neurol Neurosurg Psychiatry 49: 563–573, 1986. 90. Button B, Baker RD, Vertrees RA, Allen SE, Brodwick MS, Kramer GC. Quantitative
assessment of a circulating depolarizing factor in shock. Shock 15: 239 –244, 2001.
69. Bolton CF, Young GB. Critical illness polyneuropathy. Curr Treat Options Neurol 2:
489 – 498, 2000. 91. Button KS, Ioannidis JP, Mokrysz C, Nosek BA, Flint J, Robinson ES, Munafo MR.
Power failure: why small sample size undermines the reliability of neuroscience. Nat
70. Bolton CF, Young GB, Zochodne DW. The neurological complications of sepsis. Ann Rev Neurosci 14: 365–376, 2013.
Neurol 33: 94 –100, 1993.
92. Caffrey JM, Eng DL, Black JA, Waxman SG, Kocsis JD. Three types of sodium channels
71. Botelho FM, Rangel-Moreno J, Fritz D, Randall TD, Xing Z. Pulmonary expression of in adult rat dorsal root ganglion neurons. Brain Res 592: 283–297, 1992.
oncostatin M (OSM) promotes inducible BALT formation independently of IL-6, de-
spite a role for IL-6 in OSM-driven pulmonary inflammation. J Immunol 191: 1453– 93. Cai C, Weisleder N, Ko JK, Komazaki S, Sunada Y, Nishi M, Takeshima H, Ma J.
1464, 2013. Membrane repair defects in muscular dystrophy are linked to altered interaction
between MG53, caveolin-3, and dysferlin. J Biol Chem 284: 15894 –15902, 2009.
72. Boucher PA, Joos B, Morris CE. Coupled left-shift of Nav channels: modeling the
Na⫹-loading and dysfunctional excitability of damaged axons. J Comput Neurosci 33: 94. Callahan LA, Nethery D, Stofan D, DiMarco A, Supinski GS. Free radical-induced
301–319, 2012. contractile protein dysfunction in endotoxin-induced sepsis. Am J Respir Cell Mol Biol
24: 210 –217, 2001.
73. Bouletreau P, Patricot MC, Saudin F, Guiraud M, Mathian B. Effects of intermittent
electrical stimulations on muscle catabolism in intensive care patients. JPEN J Parenter 95. Callahan LA, She ZW, Nosek TM. Superoxide, hydroxyl radical, and hydrogen per-
Enteral Nutr 11: 552–555, 1987. oxide effects on single-diaphragm fiber contractile apparatus. J Appl Physiol 90: 45–54,
2001.
74. Boveris A, Alvarez S, Navarro A. The role of mitochondrial nitric oxide synthase in
inflammation and septic shock. Free Radic Biol Med 33: 1186 –1193, 2002. 96. Callahan LA, Stofan DA, Szweda LI, Nethery DE, Supinski GS. Free radicals alter
maximal diaphragmatic mitochondrial oxygen consumption in endotoxin-induced
75. Brady AJB, Poole-Wilson PA, Harding SE, Warren JB. Nitric oxide production within sepsis. Free Radic Biol Med 30: 129 –138, 2001.
cardiac myocytes reduces their contractility in endotoxemia. Am J Physiol Heart Circ
Physiol 263: H1963–H1966, 1992. 97. Callahan LA, Supinski GS. Sepsis induces diaphragm electron transport chain dysfunc-
tion and protein depletion. Am J Resp Crit Care Med 172: 861– 868, 2005.
76. Braun T, Gautel M. Transcriptional mechanisms regulating skeletal muscle differenti-
ation, growth and homeostasis. Nat Rev Mol Cell Biol 12: 349 –361, 2011. 98. Callahan LA, Supinski GS. Downregulation of diaphragm electron transport chain and
glycolytic enzyme gene expression in sepsis. J Appl Physiol 99: 1120 –1126, 2005.
77. Brealey D, Brand M, Hargreaves I, Heales S, Land J, Smolenski R, Davies NA, Cooper
CE, Singer M. Association between mitochondrial dysfunction and severity and out- 99. Callahan LA, Supinski GS. Diaphragm and cardiac mitochondrial creatine kinases are
come of septic shock. Lancet 360: 219 –223, 2002. impaired in sepsis. J Appl Physiol 102: 44 –53, 2007.

78. Brealey D, Karyampudi S, Jacques TS, Novelli M, Stidwill R, Taylor V, Smolenski RT, 100. Campellone JV, Lacomis D, Kramer DJ, Van Cott AC, Giuliani MJ. Acute myopathy
Singer M. Mitochondrial dysfunction in a long-term rodent model of sepsis and organ after liver transplantation. Neurology 50: 46 –53, 1998.
failure. Am J Physiol Regul Integr Comp Physiol 286: R491–R497, 2004.
101. Cannon JG, Fielding RA, Fiatarone MA, Orencole SF, Dinarello CA, Evans WJ. In-
79. Breuille D, Voisin L, Contrepois M, Arnal M, Rose F, Obled C. A sustained rat model creased interleukin1 beta in human skeletal muscle after exercise. Am J Physiol Regul
for studying the long-lasting catabolic state of sepsis. Infect Immun 67: 1079 –1085, Integr Comp Physiol 257: R451–R455, 1989.
1999.
102. Caporossi FS, Caporossi C, Borges Dock-Nascimento D, de Aguilar-Nascimento JE.
80. Brochard L, Thille AW. What is the proper approach to liberating the weak from Measurement of the thickness of the adductor pollicis muscle as a predictor of out-
mechanical ventilation? Crit Care Med 37: -S410 – 415, 2009. come in critically ill patients. Nutr Hosp 27: 490 – 495, 2012.

81. Brooks NE, Myburgh KH. Skeletal muscle wasting with disuse atrophy is multi-dimen- 103. Carchman EH, Rao J, Loughran PA, Rosengart MR, Zuckerbraun BS. Heme oxygen-
sional: the response and interaction of myonuclei, satellite cells and signaling path- ase-1-mediated autophagy protects against hepatocyte cell death and hepatic injury
ways. Front Physiol 5: 99, 2014. from infection/sepsis in mice. Hepatology 53: 2053–2062, 2011.

82. Brunello AG, Haenggi M, Wigger O, Porta F, Takala J, Jakob SM. Usefulness of a clinical 104. Carle T, Fournier E, Sternberg D, Fontaine B, Tabti N. Cold-induced disruption of
diagnosis of ICU-acquired paresis to predict outcome in patients with SIRS and acute Na⫹ channel slow inactivation underlies paralysis in highly thermosensitive paramyo-
respiratory failure. Intensive Care Med 36: 66 –74, 2010. tonia. J Physiol 587: 1705–1714, 2009.

83. Buck M, Chojkier M. Muscle wasting and dedifferentiation induced by oxidative stress 105. Carré JE, Orban JC, Re L, Felsmann K, Iffert W, Bauer M, Suliman HB, Piantadosi CA,
in a murine model of cachexia is prevented by inhibitors of nitric oxide synthesis and Mayhew TM, Breen P, Stotz M, Singer M. Survival in critical illness is associated with
antioxidants. EMBO J 15: 1753–1765, 1996. early activation of mitochondrial biogenesis. Am J Respir Crit Care Med 182: 745–751,
2010.
84. Bulger EM, Jurkovich GJ, Farver CL, Klotz P, Maier RV. Oxandrolone does not im-
prove outcome of ventilator dependent surgical patients. Ann Surg 240: 472– 478, 106. Carswell EA, Old LJ, Kassel RL, Green S, Fiore N, Williamson B. An endotoxin-
2004. induced serum factor that causes necrosis of tumors. Proc Natl Acad Sci USA 72:
3666 –3670, 1975.
85. Buras JA, Holzmann B, Sitkovsky M. Animal models of sepsis: setting the stage. Nat Rev
Drug Discov 4: 854 – 865, 2005. 107. Casaer MP, Mesotten D, Hermans G, Wouters PJ, Schetz M, Meyfroidt G, Van CS,
Ingels C, Meersseman P, Muller J, Vlasselaers D, Debaveye Y, Desmet L, Dubois J, Van
86. Burkholder TJ. Mechanotransduction in skeletal muscle. Front Biosci 12: 174 –191, AA, Vanderheyden S, Wilmer A, Van den Berghe G. Early versus late parenteral
2007. nutrition in critically ill adults. N Engl J Med 365: 506 –517, 2011.

87. Burnham EL, Moss M, Ziegler TR. Myopathies in critical illness: characterization and 108. Casaer MP, Van den Berghe G. Nutrition in the acute phase of critical illness. N Engl J
nutritional aspects. J Nutr 135: 1818S–1823S, 2005. Med 370: 1227–1236, 2014.

88. Burtin C, Clerckx B, Robbeets C, Ferdinande P, Langer D, Troosters T, Hermans G, 109. Casaer MP, Wilmer A, Hermans G, Wouters PJ, Mesotten D, Van den Berghe G. Role
Decramer M, Gosselink R. Early exercise in critically ill patients enhances short-term of disease and macronutrient dose in the randomized controlled EPaNIC trial: a post
functional recovery. Crit Care Med 37: 2499 –2505, 2009. hoc analysis. Am J Respir Crit Care Med 187: 247–255, 2013.

1092 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

110. Catterall WA. Signaling complexes of voltage-gated sodium and calcium channels. 132. Cooney RN, Maish GO 3rd, Gilpin T, Shumate ML, Lang CH, Vary TC. Mechanism of
Neurosci Lett 486: 107–116, 2010. IL-1 induced inhibition of protein synthesis in skeletal muscle. Shock 11: 235–241,
1999.
111. Catterall WA, Goldin AL, Waxman SG. International Union of Pharmacology. XLVII.
Nomenclature and structure-function relationships of voltage-gated sodium channels. 133. Corpeno R, Dworkin B, Cacciani N, Shalah H, Bergman H, Rvara B, Vitadello M,
Pharmacol Rev 57: 397– 409, 2005. Gorza L, Gystafson AM, Hedström Y, Peterson J, Feng HZ, Jin JP, Iwamoto H, Yagi N,
Artemenko K, Bergquist J, Larsson L. Time-course analysis of mechanical ventilation-
112. Chad DA, Lacomis D. Critically ill patients with newly acquired weakness: the clini- induced diaphragm contractile muscle dysfunction. J Physiol 592: 3859 –3880, 2014.
copathological spectrum. Ann Neurol 35: 257–259, 1994.
134. Couturier JC, Robert D, Monier P. Polynevrites compliquant des sejours prolonges en
113. Chai J, Wu Y, Sheng ZZ. Role of ubiquitin-proteasome pathway in skeletal muscle reanimation. Lyon Medical 252: 247–249, 1984.
wasting in rats with endotoxemia. Crit Care Med 31: 1802–1807, 2003.
135. Cree MG, Fram RY, Herndon DN, Qian T, Angel C, Greem JM, Mlcak R, Aarsland A,
114. Chambers MA, Moylan JS, Reid MB. Physical inactivity and muscle weakness in the Wolfe RR. Human mitochondrial oxidative capacity is acutely impaired after burn
critically ill. Crit Care Med 37: S337–S346, 2009. trauma. Am J Surg 196: 234 –239, 2008.

115. Chaudry IH, Sayeed MM, Baue AE. Alterations in high-energy phosphates in hemor- 136. Cree MG, Zwetsloot JJ, Herndon DN, Qian T, Morio B, Fram R, Sanford AP, Aarsland
rhagic shock as related to tissue and organ function. Surgery 79: 666 – 668, 1976. A, Wolfe RR. Insulin sensitivity and mitochondrial function are improved in children
with burn injury during a randomized controlled trial of fenofibrate. Ann Surg 245:
116. Chen H, Chan DC. Emerging functions of mammalian mitochondrial fusion and fis- 214 –221, 2007.
sion. Hum Mol Genet 14: R283–R289, 2005.
137. Crouser ED, Julian MW, Huff JE, Struck J, Cook CH. Carbamoyl phosphate syn-
117. Chen SJ, Wu CC, Yen MH. Alterations of ex vivo vascular reactivity in intraperitoneal thase-1: a marker of mitochondrial damage and depletion in the liver during sepsis.
sepsis. J Cardiovasc Pharmacol 24: 786 –793, 1994. Crit Care Med 34: 2439 –2446, 2006.

118. Cheng M, Nguyen MH, Fantuzzi G, Koh TJ. Endogeneous interferon-gamma is re- 138. Cunningham JN, Carter NW Jr, Rector FC Jr, Seldin DW. Resting transmembrane
quired for efficient muscle regeneration. Am J Physiol Cell Physiol 294: C1183–C1191, potential difference of skeletal muscle in nirmal subjects and severely ill patients. J Clin
2008. Invest 50: 49 –59, 1971.

119. Chensue SW, Terebuh PD, Remick DG, Scales WE, Kunkel SL. In vivo biologic and 139. Czeschik JC, Hagenacker T, Schafers M, Busselberg D. TNF-alpha differentially mod-
immunohistochemical analysis of interleukin-1 alpha, beta and tumor necrosis factor ulates ion channels of nociceptive neurons. Neurosci Lett 434: 293–298, 2008.
during experimental endotoxemia. Am J Pathol 138: 395– 402, 1991.
140. Damas P, Ledoux D, Nys M, Vrindts Y, De Groote D, Franchimont P, Lamy M.
120. Cherednichenko G, Hurne AM, Fessebden JD, Lee EH, Allen PD, Beam KG, Pessah Cytokine serum level during severe sepsis in human IL-6 as a marker of severity. Ann
IN. Conformational activation of Ca2⫹ entry by depolarization of skeletal myotubes. Surg 215: 356 –362, 1992.
Proc Natl Acad Sci USA 101: 15793–15798, 2004.
141. Danon MJ, Carpenter S. Myopathy with thick filament (myosin) loss following pro-
121. Chiao JJ, Jones WG 2nd, Shires GT 3rd, Barber AE, Shires GT. Effect of sepsis on longed paralysis with vecuronium during steroid treatment. Muscle Nerve 14: 1131–
intracellular sodium activity, sodium concentration, and water content in thermal 1139, 1991.
injury rat. Circ Shock 38: 42– 49, 1992.
142. Davis KA, Masella J, Blennerhassett MG. Acetylcholine metabolism in the inflamed rat
122. Choi AM, Ryter SW, Levine B. Autophagy in human health and disease. N Engl J Med intestine. Exp Neurol 152: 251–258, 1998.
368: 651– 662, 2013.
143. De Jonghe B, Bastuji-Garin S, Durand MC, Malissin I, Rodrigues P, Cerf C, Outin H,
123. Clavel S, Siffroi-Fernandez S, Coldefy AS, Boulukos K, Pisani DF, Derijard B. Regula- Sharshar T. Respiratory weakness is associated with limb weakness and delayed
tion of the intracellular localization of Foxo3a by stress-activated protein kinase sig- weaning in critical illness. Crit Care Med 35: 2007–2015, 2007.
naling pathways in skeletal muscle cells. Mol Cell Biol 30: 470 – 480, 2010. 144. De Jonghe B, Bastuji-Garin S, Sharshar T, Outin H, Brochard L. Does ICU-acquired
paresis lengthen weaning from mechanical ventilation? Intensive Care Med 30: 1117–
124. Clowes GH Jr, George BC, Villee CA Jr, Saravis CA. Muscle proteolysis induced by a
1121, 2004.
circulating peptide in patients with sepsis and trauma. N Engl J Med 308: 545–552,
1983. 145. De Jonghe B, Cook D, Sharshar T, Lefaucheur JP, Carlet J, Outin H. Acquired neuro-
muscular disorders in critically ill patients: a systematic review. Groupe de Reflexion
125. Coakley JH, Nagendran K, Honavar M, Hinds CJ. Preliminary observations on the
et d’Etude sur les Neuromyopathies en Reanimation. Intensive Care Med 24: 1242–
neuromuscular abnormalities in patients with organ failure and sepsis. Intensive Care
1250, 1998.
Med 19: 323–328, 1993.
146. De Jonghe B, Lacherade JC, Sharshar T, Outin H. Intensive care unit-acquired weak-
126. Coakley JH, Nagendran K, Yarwood GD, Honavar M, Hinds CJ. Patterns of neuro-
ness: risk factors and prevention. Crit Care Med 37: S309 –S315, 2009.
physiological abnormality in prolonged critical illness. Intensive Care Med 24: 801– 807,
1998. 147. De Jonghe B, Sharshar T, Lefaucheur JP, Authier FJ, Durand-Zaleski I, Boussarsar M,
Cerf C, Renaud E, Mesrati F, Carlet J, Raphael JC, Outin H, Bastuji-Garin S. Paresis
127. Cohen S, Brault JJ, Gygi SP, Glass DJ, Valenzuela DM, Gartner C, Latres E, Goldberg acquired in the intensive care unit: a prospective multicenter study. JAMA 288: 2859 –
AL. During muscle atrophy, thick, but not thin, filament components are degraded by 2867, 2002.
MuRF1-dependent ubiquitylation. J Cell Biol 185: 1083–1095, 2009.
148. De Larichaudy J, Zufferli A, Serra F, Isidori AM, Naro F, Dessalle K, Desgeorges M,
128. Connolly B, Denehy L, Brett S, Elliott D, Hart N. Exercise rehabilitation following Piraud M, Cheillan D, Vidal H, Lefai E, Nemoz G. TNF-␣- and tumor-induced skeletal
hospital discharge in survivors of critical illness: an integrative review. Crit Care 16: muscle atrophy involves sphingolipid metabolism. Skelet Muscle 2: 2, 2012.
226, 2012.
149. De Letter MA, Schmitz PI, Visser LH, Verheul FA, Schellens RL, Op de Coul DA, van
129. Connolly BA, Jones GD, Curtis AA, Murphy PB, Douiri A, Hopkinson NS, Polkey MI, der Meche FG. Risk factors for the development of polyneuropathy and myopathy in
Moxham J, Hart N. Clinical predictive value of manual muscle strength testing during critically ill patients. Crit Care Med 29: 2281–2286, 2001.
critical illness: an observational cohort study. Crit Care 17: R229, 2013.
150. De Letter MACJ, van Doorn PA, Savelkoul HFJ, Laman JD, Schmitz PIM, Op de Coul
130. Constantin D, McCullough J, Mahajan RP, Greenhaff PL. Novel events in the molec- AAW, Visser LH, Kros JM, Teepen JLJM, van der Meche FGA. Critical illness polyneu-
ular regulation of muscle mass in critically ill patients. J Physiol 589: 3883–3895, 2011. ropathy and myopathy (CIPNM): evidence for local immune activation by cytokine-
expression in the muscle tissue. J Neuroimmunol 106: 206 –213, 2000.
131. Cooney R, Owens E, Jurasinski C, Gray K, Vannice J, Vary T. Interleukin-1 receptor
antagonist prevents sepsis-induced inhibition of protein synthesis. Am J Physiol Endo- 151. Delbono O, Stefani E. Calcium transients in mammalian skeletal muscle fibers. J Physiol
crinol Metab 267: E636 –E641, 1994. 463: 689 –707, 1993.

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1093


FRIEDRICH ET AL.

152. Dellinger RP, Levy MM, Rhodes A, Annane D, Gerlach H, Opal SM, Sevransky JE, 170. Duncan DJ, Yang Z, Hopkins PM, Steele DS, Harrison SM. TNF-alpha and IL-1beta
Sprung CL, Douglas IS, Jaeschke R, Osborn TM, Nunnally ME, Townsend SR, Reinhart increase Ca2⫹ leak from the sarcoplasmic reticulum and susceptibility to arrhythmia in
K, Kleinpell RM, Angus DC, Deutschman CS, Machado FR, Rubenfeld GD, Webb S, rat ventricular myocytes. Cell Calcium 47: 378 –386, 2010.
Beale RJ, Vincent JL, Moreno R. Surviving Sepsis Campaign: international guidelines for
management of severe sepsis and septic shock, 2012. Intensive Care Med 39: 165–228, 171. Durigan JL, Peviani SM, Delfino GB, de Souza Jose RJ, Parra T, Salvini TF. Neuromus-
2013. cular electrical stimulation induces beneficial adaptations in the extracellular matrix of
quadriceps muscle after anterior cruciate ligament transection of rats. Am J Phys Med
153. Demoule A, Jung B, Prodanovic H, Molinari N, Chanques G, Coirault C, Matecki S, Rehabil 93: 948 –961, 2014.
Duguet A, Similowski T, Jaber S. Diaphragm dysfunction on admission to the intensive
172. Dutka TL, Lamb GD. Transverse tubular system depolarization reduces tetanic force
care unit. Prevalence, risk factors, and prognostic impact–a prospective study. Am J
in rat skeletal muscle fibers by impairing action potential repriming. Am J Physiol Cell
Respir Crit Care Med 188: 213–219, 2013.
Physiol 292: C2112–C2121, 2007.
154. Derde S, Hermans G, Derese I, Guiza F, Hedstrom Y, Wouters PJ, Bruyninckx F,
173. Dutka TL, Murphy RM, Stephenson DG, Lamb GD. Chloride conductance in the
D’Hoore A, Larsson L, Van den Berghe G, Vanhorebeek I. Muscle atrophy and pref-
transverse tubular system of rat skeletal muscle fibres: importance in excitation-
erential loss of myosin in prolonged critically ill patients. Crit Care Med 40: 79 – 89,
contraction coupling and fatigue. J Physiol 586: 875– 887, 2008.
2012.
174. Dworkin BR, Dworkin S. Learning of physiological responses. I. Habituation, sensiti-
155. Derde S, Vanhorebeek I, Guiza F, Derese I, Gunst J, Fahrenkrog B, Martinet W,
zation, and classical conditioning. Behav Neurosci 104: 298 –319, 1990.
Vervenne H, Ververs EJ, Larsson L, Van den Berghe G. Early parenteral nutrition
evokes a phenotype of autophagy deficiency in liver and skeletal muscle of critically ill 175. Dworkin BR, Dworkin S. Baroreflexes of the rat. III. Open-loop gain and electroen-
rabbits. Endocrinology 153: 2267–2276, 2012. cephalographic arousal. Am J Physiol Regul Integr Comp Physiol 286: R597–R605, 2004.

156. Derde S, Vanhorebeek I, Ververs EJ, Vanhees I, Darras VM, Van Herck E, Larsson L, 176. Dworkin BR, Dworkin S, Tang X. Carotid and aortic baroreflexes of the rat. I. Open-
Van den Berghe G. Increasing intravenous glucose load in the presence of normogly- loop steady-state properties and blood pressure variability. Am J Physiol Regul Integr
cemia: effect on outcome and metabolism in critically ill rabbits. Crit Care Med 38: Comp Physiol 279: R1910 –R1921, 2000.
602– 611, 2010.
177. Dworkin BR, Tang X, Snyder AJ, Dworkin S. Carotid and aortic baroreflexes of the
157. Deval C, Mordier S, Obled C, Bechet D, Combaret L, Attaix D, Ferrara M. Identifi- rat. II. Open-loop frequency response and the blood pressure spectrum. Am J Physiol
cation of cathepsin L as a differentially expressed message associated with skeletal Regul Integr Comp Physiol 279: R1922–R1933, 2000.
muscle wasting. Biochem J 15: 143–150, 2001.
178. Eastridge BJ, Darlington DN, Evans JA, Gann DS. A circulating shock protein depo-
158. Diaz NL, Finol HJ, Torres SH, Zambrano CI, Adjounian H. Histochemical and ultra- larizes cells in hemorrhage and sepsis. Ann Surg 219: 298 –305, 1994.
structural study of skeletal muscle in patients with sepsis and multiorgan failure syn-
179. Ebisui C, Tsujinaka T, Morimoto T, Kan K, Iijima S, Yano M, Kominami E, Tanaka K,
drome (MOFS). Histol Histopathol 13: 121–128, 1998.
Monden M. Interleukin-6 induces proteolysis by activating intracellular proteases
159. Dinarello CA, Clowes GH Jr, Gordon AH, Saravis CA, Wolff SM. Cleavage of human (cathepsins B and L, proteasome) in C2C12 myotubes. Clin Sci 89: 431– 439, 1995.
interleukin-1: isolation of a peptide fragment from plasma of febrile humans and
180. Edwards JP, Hatton PA, Little RA, Pennington RA, Wareham AC. Increased quantal
activated monocytes. J Immunol 133: 1332–1338, 1984.
release of acetylcholine at the neuromuscular junction following scald injury in the rat.
160. Dodd S, Hain B, Judge A. Hsp70 prevents disuse muscle atrophy in senescent rats. Muscle Nerve 22: 1660 –1666, 1999.
Biogerontology 10: 605– 611, 2009.
181. Eikermann M, Koch G, Gerwig M, Ochterbeck C, Beiderlinden M, Koeppen S,
161. Doig GS, Simpson F, Sweetman EA, Finfer SR, Cooper DJ, Heighes PT, Davies AR, Neuhäuser M, Peters J. Muscle force and fatigue in patients with sepsis and multiorgan
O’Leary M, Solano T, Peake S. Early parenteral nutrition in critically ill patients with failure. Intensive Care Med 32: 251–259, 2006.
short-term relative contraindications to early enteral nutrition: a randomized con-
182. El-Dwairi Q, Comtois A, Guo Y, Hussain SN. Endotoxin-induced skeletal muscle
trolled trial. JAMA 309: 2130 –2138, 2013. contractile dysfunction: contribution of nitric oxide synthases. Am J Physiol Cell Physiol
274: C770 –C779, 1998.
162. Dong LW, Ji Y, Wang XQ, Wu LL, Tang CS. Altered ryanodine receptor of rat cardiac
sarcoplasmic reticulum and its underlying mechanism during septic shock. Sheng Li 183. Ellger B, Debaveye Y, Vanhorebeek I, Langouche L, Giulietti A, Van Etten E, Herijgers
Xue Bao 47: 349 –356, 1995. P, Mathieu C, Van den Berghe G. Survival benefits of intensive insulin therapy in critical
illness: impact of maintaining normoglycemia versus glycemia-independent actions of
163. Dong LW, Wu LL, Ji Y, Liu MS. Impairment of the ryanodine-sensitive calcium release
insulin. Diabetes 55: 1096 –1105, 2006.
channels in the cardiac sarcoplasmic reticulum and its underlying mechanism during
the hypodynamic phase of sepsis. Shock 16: 33–39, 2001. 184. Ellger B, Langouche L, Richir M, Debaveye Y, Vanhorebeek I, Teerlink T, Van Leeu-
wen PA, Van den Berghe G. Modultaion of regional nitric oxide metabolism: blood
164. Dos Santos CC, Batt J. ICU-acquired weakness: mechanisms of disability. Curr Opin
glucose control or insulin? Intensive Care Med 34: 1525–1533, 2008.
Crit Care 18: 509 –517, 2012.
185. Engel C, Brunkhorst FM, Bone HG, Brunkhorst R, Gerlach H, Grond S, Gruendling M,
165. Douglass JA, Tuxen DV, Horne M, Scheinkestel CD, Weinmann M, Czarny D, Bowes Huhle G, Jaschinski U, John S, Mayer K, Oppert M, Olthoff D, Quintel M, Ragaller M,
G. Myopathy in severe asthma. Am Rev Respir Dis 146: 517–519, 1992. Rossaint R, Stuber F, Weiler N, Welte T, Bogatsch H, Hartog C, Loeffler M, Reinhart
K. Epidemiology of sepsis in Germany: results from a national prospective multicenter
166. Druschky A, Herkert M, Radespiel-Troger M, Druschky K, Hund E, Becker CM, Hilz
study. Intensive Care Med 33: 606 – 618, 2007.
MJ, Erbguth F, Neundorfer B. Critical illness polyneuropathy: clinical findings and cell
culture assay of neurotoxicity assessed by a prospective study. Intensive Care Med 27: 186. Escames G, Lopez LC, Tapias V, Utrilla P, Reiter RJ, Hitos AB, Leon J, Rodriguez MI,
686 – 693, 2001. Acuna-Castroviejo D. Melatonin counteracts inducible mitochondrial nitric oxide syn-
thase-dependent mitochondrial dysfunction in skeletal muscle of septic mice. J Pineal
167. Du J, Wang X, Miereles C, Bailey JL, Debigare R, Zheng B, Price SR, Mitch WE.
Res 40: 71–78, 2006.
Activation of caspase-3 is an initial step triggering accelerated muscle proteolysis in
catabolic conditions. J Clin Invest 113: 115–123, 2004. 187. Essen P, McNurlan MA, Gamrin L, Hunter K, Calder G, Garlick PJ, Wernerman J.
Tissue protein synthesis rates in critically ill patients. Crit Care Med 26: 92–100, 1998.
168. Dulhunty AF. Distribution of potassium and chloride permeability over the surface
and T-tubule membranes of mammalian skeletal muscle. J Membr Biol 45: 293–310, 188. Everts ME, Clausen T. Effects of thyroid hormones on calcium contents and 45Ca
1979. exchange in rat skeletal muscle. Am J Physiol Endocrinol Metab 251: E258 –E265, 1986.

169. Dulhunty AF, Derek RL, Gallant EM, casarotto MG, Pace SM, Curtis S. Activation and 189. Fan E, Ciesla ND, Truong AD, Bhoopathi V, Zeger SL, Needham DM. Inter-rater
inhibition of skeletal RyR channels by a part of the skeletal DHPR II-III loop: effects of reliability of manual muscle strength testing in ICU survivors and simulated patients.
DHPR Ser687 and FKBP12. Biophys J 77: 189 –203, 1999. Intensive Care Med 36: 1038 –1043, 2010.

1094 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

190. Fan E, Dowdy DW, Colantuoni E, Mendez-Tellez PA, Sevransky JE, Shanholtz C, muscle of critically ill patients with sepsis-induced multiple organ failure. Am J Physiol
Himmelfarb CR, Desai SV, Ciesla N, Herridge MS, Pronovost PJ, Needham DM. Endocrinol Metab 291: E1044 –E1050, 2006.
Physical complications in acute lung injury survivors: a two-year longitudinal prospec-
tive study. Crit Care Med 42: 849 – 859, 2014. 211. Fredriksson K, Rooyackers O. Mitochondrial function in sepsis: respiratory vs. leg
muscle. Crit Care Med 35 Suppl: S449 –S453, 2007.
191. Fareed MU, Evenson AR, Wei W, Menconi M, Poylin V, Petkova V, Pignol B, Hassel-
gren PO. Treatment of rats with calpain inhibitors prevents sepsis-induced muscle 212. Fredriksson K, Tjäder I, Keller P, Petrovic N, Ahlman B, Schéele C, Wernerman J,
proteolysis independent of atrogin-1/MAFbx and MuRF1 expression. Am J Physiol Timmons JA, Rooyackers O. Dysregulation of mitochondrial dynamics and the muscle
Regul Integr Comp Physiol 290: R1589 –R1597, 2006. transcriptome in ICU patients suffering from sepsis induced multiple organ failure.
PloS One 3: e3686, 2008.
192. Febbraio MA, Pedersen BK. Muscle-derived interleukin-6: mechanisms for activation
and possible biological roles. FASEB J 16: 1335–1347, 2002. 213. Frick CG, Fink H, Gordan ML, Eckel B, Martyn JA, Blobner M. Chronic Escherischia
coli infection induces muscle wasting without changing acetylcholine receptor num-
193. Fenzi F, Latronico N, Refatti N, Rizzuto N. Enhanced expression of E-selectin on the bers. Intensive Care Med 34: 561–567, 2008.
vascular endothelium of peripheral nerve in critically ill patients with neuromuscular
disorders. Acta Neuropathol 106: 75– 82, 2003. 214. Frick CG, Helming M, Martyn JA, Blobner M, Fink H. Continuous administration of
pyridostigmine improves immobilization-induced neuromuscular weakness. Crit Care
194. Ferrando AA, Lane HW, Stuart CA, vis-Street J, Wolfe RR. Prolonged bed rest de- Med 38: 922–927, 2010.
creases skeletal muscle and whole body protein synthesis. Am J Physiol Endocrinol
Metab 270: E627–E633, 1996. 215. Friedrich O. Critical illness myopathy: what is happening? Curr Opin Clin Nutr Metab
Care 9: 403– 409, 2006.
195. Filatov GN, Pinter MJ, Rich MM. Role of Ca2⫹ in injury-induced changes in sodium
current in rat skeletal muscle. Am J Physiol Cell Physiol 297: C352–C359, 2009. 216. Friedrich O, Fink RHA, Hund E. Understanding critical illness myopathy: approaching
the pathomechanism. J Nutr 135: 1813S–1817S, 2005.
196. Filatov GN, Rich MM. Hyperpolarized shifts in the voltage dependence of fast inacti-
vation of Nav1.4 and Nav15 in a rat model of critical illness myopathy. J Physiol 559: 217. Friedrich O, Hund E, von Wegner F. Enhanced muscle shortening and impaired Ca2⫹
813– 820, 2004. channel function in an acute septic myopathy model. J Neurol 257: 546 –555, 2010.

197. Finfer S, Chittock DR, Su SY, Blair D, Foster D, Dhingra V, Bellomo R, Cook D, Dodek 218. Friedrich O, Hund E, Weber C, Hacke W, Fink RH. Critical illness myopathy serum
P, Henderson WR, Hebert PC, Heritier S, Heyland DK, McArthur C, McDonald E, fractions affect membrane excitability and intracellular calcium release in mammalian
Mitchell I, Myburgh JA, Norton R, Potter J, Robinson BG, Ronco JJ. Intensive versus skeletal muscle. J Neurol 251: 53– 65, 2004.
conventional glucose control in critically ill patients. N Engl J Med 360: 1283–1297,
219. Friedrich O, v Wegner F, Wink M, Fink RH. Na⫹- and K⫹-channels as molecular
2009.
targets of the alkaloid ajmaline in skleletal muscle fibres. Br J Pharmacol 151: 63–74,
198. Fink MP. Cytopathic hypoxia: mitochondrial dysfunction as a mechanism contributing 2007.
to organ dysfunction in sepsis. Crit Care Clin 17: 219 –237, 2001.
220. Friedrich O, Yi B, Edwards JN, Reischl B, Wirth-Hücking A, Buttgereit A, Lang R,
199. Fink MP. Animal models of sepsis. Virulence 5: -1543–153, 2014. Weber C, Polyak F, Liu I, von Wegner F, Cully TR, Lee A, Most P, Völkers M.
Interleukin-1␣ reversibly inhibits skeletal muscle ryanodine receptor: a novel mech-
200. Fischer D, Gang G, Pritts T, Hasselgren PO. Sepsis-induced proteolysis is prevented anism for critical illness myopathy? Am J Respir Cell Mol Biol 50: 1096 –1106, 2014.
by a proteasome inhibitor in vivo. Biochem Biophys Res Commun 270: 215–221, 2000.
221. Frolkis I, Klein Y, Locker C, Nimrod A, Dahan E, Uretzsky G, Shapira I, Sorkine P.
201. Fischer D, Sun X, Gang G, Pritts T, Hasselgren PO. The gene expression of ubiquitin Vipera aspis venom reduces lethality and down-regulates tumor necrosis factor-␣ in a
ligase E3alpha is upregulated in skeletal muscle during sepsis in rats: potential role of rat model of LPS-induced sepsis. Cytokine 3: 319 –324, 2010.
glucocorticoids. Biochem Biophys Res Commun 267: 504 –508, 2000.
222. Frost RA, Nystrom GJ, Jefferson LS, Lang CH. Hormone, cytokine, and nutritional
202. Fischer DR, Sun X, Williams AB, Gang G, Pritts TA, James JH, Molloy M, Fischer JE, regulation of sepsis-induced increases in atrogin-1 and MuRF1 in skeletal muscle. Am
Paul RJ, Hasselgren PO. Dantrolene reduces serum TNF␣ and corticosterone levels J Physiol Endocrinol Metab 292: E501–E512, 2007.
and muscle calcium, calpain gene expression, and protein breakdown in septic rats.
Shock 15: 200 –207, 2001. 223. Frost RA, Nystrom GJ, Lang CH. Endotoxin and interferon-gamma inhibit translation
in skeletal muscle cells by stimulating nitric oxide synthase activity. Shock 32: 416 –
203. Fitts RH, Riley DR, Widrick JJ. Physiology of a microgravity environment invited 426, 2009.
review: microgravity and skeletal muscle. J Appl Physiol 89: 823– 839, 2000.
224. Fujimura N, Sumitha Aimono M S, Masuda Y, Shichinohe Y, Narimatsu E, Namiki A.
204. Fitts RH, Riley DR, Widrick JJ. Functional and structural adaptations of skeletal muscle Effect of free radical scavengers on diaphragmatic contractility in septic peritonitis. Am
to microgravity. J Exp Biol 204: 3201–3208, 2001. J Respir Crit Care Med 162: 2159 –2165, 2000.

205. Flakoll PJ, Hill JO, Abumrad NN. Acute hyperglycemia enhances proteolysis in normal 225. Garcia YR, May JJ, Green AM, Krolick KA. Acetylcholine receptor-reactive antibody
man. Am J Physiol Endocrinol Metab 265: E715–E721, 1993. induces nitric oxide production by a rat skeletal muscle cell line: influence of cytokine
environment. J Neuroimmunol 120: 103–111, 2001.
206. Fletcher SN, Kennedy DD, Ghosh IR, Misra VP, Kiff K, Coakley JH, Hinds CJ. Persis-
tent neuromuscular and neurophysiologic abnormalities in long-term survivors of 226. Garcia-Martinez C, Agell N, Llovera M, Lopez-Soriano FJ, Argiles JM. Tumor necrosis
prolonged critical illness. Crit Care Med 31: 1012–1016, 2003. factor-alpha increases the ubiquitinization of rat skeletal muscle proteins. FEBS Lett
323: 211–214, 1993.
207. Fram RY, Cree MG, Wolfe RR, Mlcak RP, Qian T, Chinkes DL, Herndon DN. Intensive
insulin therapy improves insulin sensitivity and mitochondrial function in severely 227. Garcia-Martinez C, Llovera M, Agell N, Lopez-Soriano FJ, Argiles JM. Ubiquitin gene
burned children. Crit Care Med 38: 1475–1483, 2010. expression in skeletal muscle is increased by tumour necrosis factor-alpha. Biochem
Biophys Res Commun 201: 682– 686, 1994.
208. Fraysse B, Desaphy JF, Rolland JF, Pierno S, Liantonio A, Giannuzzi V, Camerino C,
Didonna MP, Cocchi D, De Luca A, Conte Camerino D. Fiber type-related changes in 228. Garcia-Martinez C, Llovera M, Agnell N, Lopez-Soriano FJ, Argiles JM. Ubiquitin gene
rat skeletal muscle calcium homeostasis during aging and restoration by growth hor- expression in skeletal muscle is increased during sepsis: involvement of TNF-alpha but
mone. Neurobiol Dis 21: 372–380, 2006. not IL-1. Biochem Biophys Res Commun 217: 839 – 844, 1995.

209. Fredriksson K, Fläring U, Guillet C, Wernerman J, Rooyackers O. Muscle mitochon- 229. Garcia-Martinez C, Lopez-Soriano FJ, Argiles JM. Acute treatment with tumor necro-
drial activity increases rapidly after an endotoxin challenge in human volunteers. Acta sis factor-alpha induces changes in protein metabolism in rat skeletal muscle. Mol Cell
Anaesthesiol Scand 53: 299 –304, 2009. Biochem 125: 11–18, 1993.

210. Fredriksson K, Hammarqvist F, Strigard K, Hultenby K, Ljungqvist O, Wernerman J, 230. Garcia-Martinez C, Lopez-Soriano FJ, Argiles JM. Interleukin-6 does not activate
Rooyackers O. Derangements in mitochondrial metabolism in intercostal and leg protein breakdown in rat skeletal muscle. Cancer Lett 76: 1– 4, 1994.

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1095


FRIEDRICH ET AL.

231. Garnacho-Montero J, Madrazo-Osuna J, Garcia-Garmendia JL, Ortiz-Leyba C, Jime- 252. Goldhaber JI, Philipson KD. Cardiac sodium-calcium exchange and efficient excita-
nez-Jimenez FJ, Barrero-Almodovar A, Garnacho-Montero MC, Moyano-Del-Estad tion-contraction coupling: implications for heart disease. Adv Exp Med Biol 961: 355–
MR. Critical illness polyneuropathy: risk factors and clinical consequences. A cohort 364, 2013.
study in septic patients. Intensive Care Med 27: 1288 –1296, 2001.
253. Gonoi T, Hille B. Gating of Na channels. Inactivation modifiers discriminate among
232. Garrabou G, Moren C, Lopez S, Tobias E, Cardellach F, Miro O, Casademont J. The models. J Gen Physiol 89: 253–274, 1987.
effects of sepsis on mitochondria. J Infect Dis 205: 392– 400, 2012.
254. Gonzalez Narvaez AA, Castillo A. Ca2⫹ store determines gating of store-operated
233. Gath I, Closs EI, Gödtel-Armbrust U, Schmitt S, Nakane M, Wessler I, Förstermann U. calcium entry in mammalian skeletal muscle. J Muscle Res Cell Motil 28: 105–113,
Inducible NO synthase II and neuronal NO synthase I are constitutively expressed in 2007.
different structures of guinea pig skeletal muscle: implications for contractile function.
255. Gooch JL. AAEM case report #29: prolonged paralysis after neuromuscular blockade.
FASEB J 10: 1614 –1620, 1996.
Muscle Nerve 18: 937–942, 1995.
234. Gautel M. Cytoskeletal protein kinases: titin and its relations in mechanosensing.
256. Gooch JL, Moore MH, Ryser DK. Prolonged paralysis after neuromuscular junction
Pflügers Arch 462: 119 –134, 2011.
blockade: case reports and electrodiagnostic findings. Arch Phys Med Rehabil 74: 1007–
235. Geller ER, Jankauskas S, Kirkpatrick J. Mitochondrial death in sepsis: a failed concept. 1011, 1993.
J Surg Res 40: 514 –517, 1986.
257. Goodman BP, Harper CM, Boon AJ. Prolonged compound muscle action potential
236. Gellerich FN, Trumbeckaite S, Opalka JR, Gellerich JF, Chen Y, Neuhof C, Redl H, duration in critical illness myopathy. Muscle Nerve 40: 1040 –1042, 2009.
Werdan K, Zierz S. Mitochondrial dysfunction in sepsis: evidence from bacteraemic 258. Goodman MN. Tumor necrosis factor induces skeletal muscle protein breakdown in
baboons and endotoxaemic rabbits. Biosci Rep 22: 99 –113, 2002. rats. Am J Physiol Endocrinol Metab 260: E727–E730, 1991.
237. Gerovasili V, Stefanidis K, Vitzilaios K, Karatzanos E, Politis P, Koroneos A, Chatzimi- 259. Goodman MN. Interleukin-6 induces skeletal muscle protein breakdown in rats. Proc
chail A, Routsi C, Roussos C, Nanas S. Electrical muscle stimulation preserves the Soc Exp Biol Med 205: 182–185, 1994.
muscle mass of critically ill patients: a randomized study. Crit Care 13: R161, 2009.
260. Gordon BS, Kelleher AR, Kimball SR. Regulation of muscle protein synthesis and the
238. Gerovasili V, Tripodaki E, Karatzanos E, Pitsolis T, Markaki V, Zervakis D, Routsi C, effects of catabolic states. Int J Biochem Cell Biol 45: 2147–2157, 2013.
Roussos C, Nanas S. Short-term systemic effect of electrical muscle stimulation in
critically ill patients. Chest 136: 1249 –1256, 2009. 261. Gore DC, Chinkes DL, Hart DW, Wolf SE, Herndon DN, Sanford AP. Hyperglycemia
exacerbates muscle protein catabolism in burn-injured patients. Crit Care Med 30:
239. Gervasio JM, Dickerson RN, Swearingen J, Yates ME, Yuen C, Fabian TC, Croce MA, 2438 –2442, 2002.
Brown RO. Oxandrolone in trauma patients. Pharmacotherapy 20: 1328 –1334, 2000.
262. Gore DC, Wolfe RR. Glutamine supplementation fails to affect muscle protein kinetics
240. Gervasio OL, Phillips WD, Cole L, Allen DG. Caveolae respond to cell stretch and in critically ill patients. JPEN J Parenter Enteral Nutr 26: 342–349, 2002.
contribute to stretch-induced signaling. J Cell Sci 124: 3581–3590, 2011.
263. Gore DC, Wolfe RR. Metabolic response of muscle to alanine, glutamine, and valine
241. Giai C, Gonzalez C, Ledo C, Garofalo A, De Genaro MS, Sordelli DO, Gomez MI. supplementation during severe illness. JPEN J Parenter Enteral Nutr 27: 307–314, 2003.
Shedding of tumor necrosis factor receptor 1 induced by protein A decreases tumor
necrosis factor alpha availability and inflammation during systemic Staphylococcus 264. Greenberg MJ, Mealy TR, Jones M, Szczesna-Cordary D, Moore JR. The direct mo-
aureus infection. Infect Immunol 81: 4200 – 4207, 2013. lecular effects of fatigue and myosin regulatory light chain phosphorylation on the
actomyosin contractile apparatus. Am J Physiol Regul Integr Comp Physiol 298: R989 –
242. Giamarellos-Bourboulis EJ. Immunomodulatory therapies for sepsis: unexpected ef- R996, 2010.
fects with macrolides. Int J Antimicrob Agents 32 Suppl 1: S39 –S43, 2008.
265. Greenberg SB, Vender J. The use of neuromuscular blocking agents in the ICU: where
243. Giamarellos-Bourboulis EJ. The failure of biologics in sepsis: where do we stand? Int J are we now? Crit Care Med 41: 1332–1344, 2013.
Antimicrob Agents 42 Suppl: S45–S47, 2013.
266. Gregory CM, Vandenborne K, Castro MJ, Dudley GA. Human and rat skeletal muscle
244. Gibson WH, Cook JJ, Gatipon G, Moses ME. Effect of endotoxin shock on skeletal adaptations to spinal cord injury. Can J Appl Physiol 28: 491–500, 2003.
muscle cell membrane potential. Surgery 81: 571–577, 1977.
267. Gregson JM, Leathley MJ, Moore AP, Smith TL, Sharma AK, Watkins CL. Reliability of
245. Ginz HF, Bandschapp O, Urwyler A, Girard T, Iaizzo PA. Tissue oedema is not measurements of muscle tone and muscle power in stroke patients. Age Ageing 29:
associated with skeletal muscle weakness in septic patients. Acta Anaesthesiol Scand 223–228, 2000.
54: 904, 2010.
268. Griffiths RD, Hall JB. Intensive care unit-acquired weakness. Crit Care Med 38: 779 –
246. Glabinski AR, Balasingam V, Tani M, Kunkel SL, Strieter RM, Yong VW, Ransohoff RM. 787, 2010.
Chemokine monocyte chemoattractant protein-1 is expressed by astrocytes after
269. Griffiths RD, Palmer TE, Helliwell T, MacLennan P, MacMillan RR. Effect of passive
mechanical injury to the brain. J Immunol 156: 4363– 4368, 1996.
stretching on the wasting of muscle in the critically ill. Nutrition 11: 428 – 432, 1995.
247. Glick D, Barth S, Macleod KF. Autophagy: cellular and molecular mechanisms. J Pathol 270. Groome JR, Lehmann-Horn F, Holzherr BR. Open- and closed-state fast inactivation
221: 3–12, 2010. in sodium channels. Channels 5: 65–78, 2011.
248. Görgens SW, Eckardt K, Elsen M, Tennagels N, Eckel J. Chitinase-3-like protein 1 271. Grumati P, Coletto L, Sabatelli P, Cescon M, Angelin A, Bertaggia E, Blaauw B,
protects skeletal muscle from TNF␣-induced inflammation and insulin resistance. Urciuolo A, Tiepolo T, Merlini L, Maraldi NM, Bernardi P, Sandri M, Bonaldo P.
Biochem J 459: 479 – 488, 2014. Autphagy is defective in collagen VI muscular dystrophies, and its reactivation rescues
myofiber degeneration. Nat Med 16: 1313–1320, 2010.
249. Gogos C, Drosou E, Bassaris HP, Skoutelis A. Pro- versus anti-inflammatory cytokine
profile in patients with severe sepsis: a marker for prognosis and future therapeutic 272. Gruther W, Kainberger F, Fialka-Moser V, Paternostro-Sluga T, Quittan M, Spiss C,
options. J Infect Dis 181: 176 –180, 2000. Crevenna R. Effects of neuromuscular electrical stimulation on muscle layer thickness
of knee extensor muscles in intensive care unit patients: a pilot study. J Rehabil Med 42:
250. Goldberg AL, Baracos V, Rodemann P, Waxman L, Dinarello C. Control of protein
593–597, 2010.
degradation in muscle by prostaglandins, Ca2⫹, and leukocytic pyrogen (interleukin
1). Federation Proc 43: 1301–1306, 1984. 273. Guarneri B, Bertolini G, Latronico N. Long-term outcome in patients with critical
illness myopathy or neuropathy: the Italian multicentre CRIMYNE study. J Neurol
251. Goldberg AL, Kettelhut IC, Furuno K, Fagen JM, Barascos V. Activation of protein Neurosurg Psychiatry 79: 838 – 841, 2008.
breakdown and prostaglandin E2 production in rat skeletal muscle in fever is signaled
by a macrophage product distinct from interleukin-1 or other known monokines. J 274. Guillouet M, Gueret G, Rannou F, Giroux-Metges MA, Gioux M, Arvieux C, Pennec
Clin Invest 81: 1378 –1383, 1988. JP. TNF␣ increases resting potential in isolated fibers from rat peroneus longus by a

1096 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

PKC mediated mechanism: involvement in ICU acquired polyneuromyopathy. Cyto- Scherer PE, Levine B. Exercise-induced BCL2-regulated autophagy is required for
kine 56: 149 –152, 2011. muscle glucose homeostasis. Nature 481: 511–515, 2012.

275. Guillouet M, Gueret G, Rannou F, Giroux-Metges MA, Gioux M, Arvieux C, Pennec 295. He C, Klionsky DJ. Regulation mechanisms and signaling pathways of autophagy. Annu
JP. Tumor necrosis factor-␣ downregulates sodium current in skeletal muscle by Rev Genet 43: 67–93, 2009.
protein kinase C activation: involvement in critical illness polyneuromyopathy. Am J
Physiol Cell Physiol 301: C1057–C1063, 2011. 296. Head SI. Membrane potential, resting calcium and calcium transients in isolated mus-
cle fibers from normal and dystrophic mice. J Physiol 469: 11–19, 1993.
276. Gundersen K. Excitation-transcription coupling in skeletal muscle: the molecular
pathways of exercise. Biol Rev Camp Philos Soc 86: 564 – 600, 2011. 297. Helliwell TR, Coakley JH, Wagenmakers AJ, Griffiths RD, Campbell IT, Green CJ,
McClelland P, Bone JM. Necrotizing myopathy in critically-ill patients. J Pathol 164:
277. Gunst J, Derese I, Aertgeerts A, Ververs EJ, Wauters A, Van den Berghe G, Vanhore- 307–314, 1991.
beek I. Insufficient autophagy contributes to mitochondrial dysfunction, organ failure,
and adverse outcome in an animal model of critical illness. Crit Care Med 41: 182–194, 298. Helliwell TR, Wilkinson A, Griffiths RD, McClelland P, Palmer TE, Bone JM. Muscle
2013. fibre atrophy in critically ill patients is associated with the loss of myosin filaments and
the presence of lysosomal enzymes and ubiquitin. Neuropathol Appl Neurobiol 24:
278. Gupta R, Rajani R, Primrose JN, Johnson CD. Body composition, physiological func- 507–517, 1998.
tion and psychological changes in patients with predicted severe acute pancreatitis.
Pancreatology 1: 58 – 62, 2001. 299. Hermans G, Agten A, Testelmans D, Decramer M, Gayan-Ramirez G. Increased
duration of mechanical ventilation is associated with decreased diaphragmatic force: a
279. Gutmann L, Blumenthal D, Gutmann L, Schochet SS. Acute type II myofiber atrophy prospective observational study. Crit Care 14: R127, 2010.
in critical illness. Neurology 46: 819 – 821, 1996.
300. Hermans G, Casaer MP, Clerckx B, Güiza F, Vanhullebusch T, Derde S, Meerseman
280. Guttridge DC, Mayo MW, Madrid LV, Wang CY, Baldwin AS Jr. NF-kappaB-induced P, Derese I, Mesotten D, Wouters PJ, Van Cromphaut S, Debaveye S, Gosselink R,
loss of MyoD messenger RNA: possible role in muscle decay and cachexia. Science Gunst J, Wilmer A, Van den Berghe G, Vanhorebeek I. Effect of tolerating macronu-
289: 2363–2366, 2000. trient deficit on the development of intensive-care unit acquired weakness: a suba-
nalysis of the EPaNic trial. Lancet Resp Med 1: 621– 629, 2013.
281. Haarmann CS, Dulhunty AF, Laver DR. Regulation of skeletal muscle ryanodine re-
ceptors by dihydropyridine receptor II-III loop C-region peptides: relief of Mg2⫹ 301. Hermans G, Clerckx B, Vanhullebusch T, Segers J, Vanpee G, Robbeets C, Casaer MP,
inhibition. Biochem J 387: 429 – 436, 2005. Wouters P, Gosselink R, Van den Berghe G. Interobserver agreement of Medical
Research Council sum score and handgrip strength in the intensive care unit. Muscle
282. Haden DW, Suliman HB, Carraway MS, Welty-Wolf KE, Ali AS, Shitara H, Yonekawa
Nerve 45: 18 –25, 2012.
H, Piantadosi CA. Mitochondrial biogenesis restores oxidative metabolism during
Staphylococcus aureus sepsis. Am J Respir Crit Care Med 176: 768 –777, 2007. 302. Hermans G, De Jonghe B, Bruyninckx F, Van den Berghe G. Clinical review: critical
illness polyneuropathy and myopathy. Crit Care 12: 238, 2008.
283. Haeseler G, Foadi N, Wiegand E, Ahrens J, Krampfl K, Dengler R, Leuwer M. Endo-
toxin reduces availability of voltage-gated human skeletal muscle sodium channels at 303. Hermans G, De Jonghe B, Bruyninckx F, Van den Berghe G. Interventions for pre-
depolarized membrane potentials. Crit Care Med 36: 1239 –1247, 2008. venting critical illness polyneuropathy and critical illness myopathy. Cochrane Database
Syst Rev 1: CD006832, 2009.
284. Hall-Angeras M, Hasselgren PO, Dimlich RV, Fischer JE. Myofibrillar proteinase, ca-
thepsin B, and protein breakdown rates in skeletal muscle from septic rats. Metabo- 304. Hermans G, De Jonghe B, Bruyninckx F, Van den Berghe G. Interventions for pre-
lism 40: 302–306, 1991. venting critical illness polyneuropathy and critical illness myopathy. Cochrane Database
Syst Rev 1: CD006832, 2014.
285. Hammarqvist F, Wernerman J, Ali R, von der Decken A, Vinnars E. Addition of
glutamine to total parenteral nutrition after elective abdominal surgery spares free 305. Hermans G, Gosselink R. Should we abandon manual muscle strength testing in the
glutamine in muscle, counteracts the fall in muscle protein synthesis, and improves ICU? Crit Care 15: 127, 2011.
nitrogen balance. Ann Surg 209: -455– 61, 1989.
306. Hermans G, Schrooten M, Van Damme P, Berends N, Bouckaert B, De Vooght W,
286. Hardin BJ, Campbell KS, Smith JD, Arbogast S, Smith J, Moylan JS, Reid MB. TNF-␣ Robberecht W, Van den Berghe G. Benefits of intensive insulin therapy on neuromus-
acts via TNFR1 and muscle-derived oxidants to depress myofibrillar force in murine cular complications in routine daily critical care practice: a retrospective study. Crit
skeletal muscle. J Appl Physiol 104: 694 – 699, 2008. Care 13: R5, 2009.
287. Harridge SD. Plasticity of human skeletal muscle: gene expression to in vivo function. 307. Hermans G, Van Mechelen H, Clerckx B, Vanhullebusch T, Mesotten D, Wilmer A,
Exp Physiol 92: 783–797, 2007. Casaer MP, Meersseman P, Debaveye Y, Van Cromphaut S, Wouters PJ, Gosselink R,
Van den Berghe G. Acute outcomes and 1-year mortality of ICU-acquired weakness:
288. Harris ML, Luo YM, Watson AC, Rafferty GF, Polkey MI, Green M, Moxham J.
a cohort study and propensity matched analysis. Am J Respir Crit Care Med 190:
Adductor pollicis twitch tension assessed by magnetic stimulation of the ulnar nerve.
410 – 420, 2014.
Am J Respir Crit Care Med 162: 240 –245, 2000.

289. Hasselgren PO, James JH, Benson DW, Li S, Fischer JE. Is there a circulating proteol- 308. Hermans G, Vanhorebeek I, Derde S, Van den Berghe G. Metabolic aspects of critical
ysis-inducing factor during sepsis? Arch Surg 125: 510 –514, 1990. illness polyneuromyopathy. Crit Care Med 37: S391–S397, 2009.

290. Hasselgren PO, James JH, Warner BW, Ogle C, Takehara H, Fischer JE. Reduced 309. Hermans G, Wilmer A, Meersseman W, Milants I, Wouters PJ, Bobbaers H, Bruyn-
muscle amino acid uptake in sepsis and the effects in vitro of septic plasma and inckx F, Van den Berghe G. Impact of intensive insulin therapy on neuromuscular
interleukin-1. Surgery 100: 222–228, 1986. complications and ventilator-dependency in MICU. Am J Respir Crit Care Med 175:
480 – 489, 2007.
291. Hasselgren PO, Talamini M, James H, Fischer JE. Protein metabolism in different types
of skeletal muscle during early and late sepsis in rats. Arch Surg 121: 918 –923, 1986. 311. Herridge MS, Cheung AM, Tansey CM, Matte-Martyn A, Diaz-Granados N, Al-Saidi F,
Cooper AB, Guest CB, Mehta S, Stewart TE, Barr A, Cook D, Slutsky AS, Canadian
292. Hassoun SM, Marechal X, Montaigne D, Bouazza Y, Decoster B, Lancel S, Neviere R. Critical Care Trials Group. One-year outcomes in survivors of the acute respiratory
Prevention of endotoxin-induced sarcoplasmic reticulum calcium leak improves mi- distress syndrome. N Engl J Med 348: 683– 693, 2003.
tochondrial and myocardial dysfunction. Crit Care Med 36: 2590 –2596, 2008.
312. Heyland D, Muscedere J, Wischmeyer PE, Cook D, Jones G, Albert M, Elke G, Berger
293. Hauptmann S, Klosterhalfen B, Weis J, Mittermayer C, Kirkpatrick CJ. Skeletal muscle MM, Day AG. A randomized trial of glutamine and antioxidants in critically ill patients.
oedema and muscle fiber necrosis during septic shock. Observations with a porcine N Engl J Med 368: 1489 –1497, 2013.
septic shock model. Virchows Arch 424: 653– 659, 1994.
313. Hinohara H, Morita T, Okano N, Kuhimoto F, Goto F. Chronic intraperitoneal endo-
294. He C, Bassik MC, Moresi V, Sun K, Wei Y, Zou Z, An Z, Loh J, Fisher J, Sun Q, toxin treatment in rats induces resistance to d-tubocurarine but does not produce
Korsmeyer S, Packer M, May HI, Hill JA, Virgin HW, Gilpin C, Xiao G, Bassel-Duby R, up-regulation of acetylcholine receptors. Acta Anaesthesiol Scand 47: 335–341, 2003.

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1097


FRIEDRICH ET AL.

314. Hirano M, Ott BR, Raps EC, Minetti C, Lennihan L, Libbey NP, Bonilla E, Hays AP. Goldberg P. Mechanical ventilation-induced diaphragm disuse in humans triggers au-
Acute quadriplegic myopathy: a complication of treatment with steroids, nondepo- tophagy. Am J Respir Crit Care Med 182: 1377–1386, 2010.
larizing blocking agents, or both. Neurology 42: 2082–2087, 1992.
336. Ibebunjo C, Chick JM, Kendall T, Eash JK, Li C, Zhang Y, Vickers C, Wu Z, Clarke BA,
315. Hobler SC, Tiao G, Fischer JE, Monaco J, Hasselgren PO. Sepsis-induced increase in Shi J, Cruz J, Fournier B, Brachat S, Gutzwiller S, Ma Q, Markovits J, Broome M,
muscle proteolysis is blocked by specific proteasome inhibitors. Am J Physiol Regul Steinkrauss M, Skuba E, Galarneau JR, Gygi SP, Glass DJ. Genomic and proteomic
Integr Comp Physiol 274: R30 –R37, 1998. profiling reveals reduced mitochondrial function and disruption of the neuromuscular
junction driving rat sarcopenia. Mol Cell Biol 33: 194 –212, 2013.
316. Hobler SC, Williams A, Fischer D, Wang JJ, Sun X, Fischer JE, Monaco JJ, Hasselgren
PO. Activity and expression of the 20S proteasome are increased in skeletal muscle 337. Ibebunjo C, Martyn JA. Fiber atrophy, but not changes in acetylcholine receptor
during sepsis. Am J Physiol Regul Integr Comp Physiol 277: R434 –R440, 1999. expression, contributes to the muscle dysfunction after immobilization. Crit Care Med
27: 275–285, 1999.
317. Holecek M. Muscle wasting in animal models of severe illness. Int J Exp Pathol 93:
157–171, 2012. 338. Illner HP, Shires GT. Membrane defect and energy status of rabbit skeletal muscle
318. Hollenberg SM, Cunnion RE, Parillo JE. Effect of septic serum on vascular smooth cells in sepsis and septic shock. Arch Surg 116: 1302–1305, 1981.
muscle: in vitro studies using rat aortic rings. Crit Care Med 20: 993–998, 1992.
339. Ingber DE. Control of capillary growth and differentiation by extracellular matrix. Use
319. Holzer M. Targeted temperature management for comatose survivors of cardiac of a tensegrity (tensional integrity) mechanism for signal processing. Chest 99 Suppl:
arrest. N Engl J Med 363: 1256 –1264, 2010. 34S– 40S, 1991.

320. Hopkins RO, Spuhler VJ, Thomsen GE. Transforming ICU culture to facilitate early 340. Ingber DE. Cellular tensegrity: defining new rules of biological design that govern the
mobility. Crit Care Clin 23: 81–96, 2007. cytoskeleton. J Cell Sci 104: 613– 627, 1993.

321. Hosenpud JD, Campbell SM, Mendelson DJ. Interleukin-1-induced myocardial de- 341. Ingber DE. Integrins, tensegrity, mechanotransduction. Gravit Space Biol Bull 10: 49 –
pression in an isolated beating heart. J Heart Transplant 8: 460 – 464, 1989. 55, 1997.

322. Hosokawa S, Koseki H, Nagashima M, Maeyama Y, Yomogida K, Mehr C, Rutledge M, 342. Ingber DE. Tensegrity: the architectural basis of cellular mechanotransduction. Annu
Greenfeld H, Kaneki M, Tompkins RG, Martyn JA, Yasuhara SE. Title efficacy of Rev Physiol 59: 575–599, 1997.
phosphodiesterase 5 inhibitor on distant burn-induced muscle autophagy, microcir-
culation, and survival rate. Am J Physiol Endocrinol Metab 304: E922–E933, 2013. 343. Ingber DE. Mechanical signaling and the cellular response to extracellular matrix in
angiogenesis and cardiovascular physiology. Circ Res 91: 877– 887, 2002.
323. Hotchkiss RS, Monneret G, Payen D. Immunosuppression in sepsis: a novel under-
standing of the disorder and a new therapeutic approach. Lancet Infect Dis 13: 260 – 344. Intiso D, Amoruso L, Zarrelli M, Pazienza L, Basciani M, Grimaldi G, Iarossi A, Di RF.
268, 2013. Long-term functional outcome and health status of patients with critical illness poly-
neuromyopathy. Acta Neurol Scand 123: 211–219, 2011.
324. Hotchkiss RS, Rust RS, Dence CS, Wasserman TH, Song SK, Hwang DR, Karl IE,
Welch MJ. Evaluation of the role of cellular hypoxia in sepsis by the hypoxic marker 345. Ioannidis JP. Extrapolating from animals to humans. Sci Transl Med 4: 151ps115, 2012.
[18F]fluoromisonidazole. Am J Physiol Regul Integr Comp Physiol 261: R965–R972, 1991.
346. Ito N, Ruegg UT, Kudo A, Miyagoe-Suzuki S, Takeda Y. Activation of calcium signaling
325. Hough CL, Lieu BK, Caldwell ES. Manual muscle strength testing of critically ill pa- through Trpv1 by nNOS and peroxynitrite as a key trigger of skeletal muscle hyper-
tients: feasibility and interobserver agreement. Crit Care 15: R43, 2011. trophy. Nat Med 19: 101–106, 2013.

326. Hranjec T, Swenson BR, Dossett LA, Metzger R, Flohr TR, Popovsky KA, Bonatti HJ, 347. Jacobs DO, Kobayashi T, Imaqire J, Grant C, Kesselly B, Wilmore DW. Sepsis alters
May AK, Sawyer RG. Diagnosis-dependent relationships between cytokine levels and skeletal muscle energetic and membrane function. Surgery 110: 318 –325, 1991.
survival in patients admitted for surgical critical care. J Am Coll Surg 210: 833– 846,
2010. 348. Jacobs DO, Maris J, Fried R, Settle RG, Rolandelli RR, Koruda MJ, Chance B, Rombeau
JL. In vivo phosphorus 31 magnetic resonance spectroscopy of rat hind limb skeletal
327. Hsieh CH, Pai PY, Hsueh HW, Yuan SS, Hsieh YC. Complete induction of autophagy muscle during sepsis. Arch Surg 123: 1425–1428, 1988.
is essential for cardioprotection in sepsis. Ann Surg 253: 1190 –1200, 2011.
349. Jain N, Iyer KV, Kumar A, Shivashankar GV. Cell geometric constraints induce mod-
328. Hsieh PF, Liu YF, Pan YJ, Wu MC, Lin TL, Huang YT, Wang JT. Klebsiella pneumoniae ular gene-expression patterns via redistribution of HDAC3 regulated by actomyosin
peptidoglycan-associated lipoprotein and murein liporprotein contribute to serum contractility. Proc Natl Acad Sci USA 110: 11349 –11354, 2013.
resistance, antiphagocytosis, and proinflammatory cytokine stimulation. J Infect Dis
208: 1580 –1589, 2013. 350. James JH, Luchette FA, McCarter FD, Fischer JE. Lactate is an unreliable predictor of
tissue hypoxia in injury or sepsis. Lancet 354: 505–508, 1999.
329. Hummel RP 3rd, James JH, Warner BW, Hasselgren PO, Fischer JE. Evidence that
cathepsin B contributes to skeletal muscle protein breakdown during sepsis. Arch Surg 351. Janssen SP, Gayan-Ramirez G, Van den Bergh A, Herijgers P, Maes K, Verbeken E,
123: 221–224, 1988. Decramer M. Interleukin-6 causes myocardial failure and skeletal muscle atrophy in
rats. Circulation 111: 996 –1005, 2005.
330. Hummel RP 3rd, Warner BW, James JH, Hasselgren PO, Fischer JE. Effects of indo-
methacin and leupeptin on muscle cathepsin B activity and protein degradation during 352. Jeschke MG, Finnerty CC, Suman OE, Kulp G, Mlcak RP, Herndon DN. The effect of
sepsis. J Surg Res 45: 140 –144, 1988. oxandrolone on the endocrinologic, inflammatory, and hypermetabolic responses
during the acute phase postburn. Ann Surg 246: 351–360, 2007.
331. Hund E. Myopathy in critically ill patients. Crit Care Med 27: 2544 –2547, 1999.

332. Hund E. Neurological complications of sepsis: critical illness polyneuropathy and 353. Jespersen JG, Nedergaard A, Reitelseder S, Mikkelsen UR, Dideriksen KJ, Agergaard
myopathy. J Neurol 248: 929 –934, 2001. J, Kreiner F, Pott FC, Schjerling P, Kjaer M. Activated protein synthesis and sup-
pressed protein breakdown signaling in skeletal muscle of critically-ill patients. PLoS
333. Hund E, Grenzwürker H, Böhrer H, Jakob H, Thiele R, Hacke W. Predominant One 31: e18090, 2011.
involvement of motor fibres in patients with critical illness polyneuropathy. Br J An-
aesth 78: 274 –278, 1997. 354. Johnson JD, Jiang Y, Rall JA. Intracellular EDTA mimics parvalbumin in the promotion
of skeletal muscle relaxation. Biophys J 76: 1514 –1522, 1999.
334. Hurne AM, O’Brien JJ, Wingrove D, Cherednichenko G, Allen PD, Beam KG, Pessah
IN. Ryanodine receptor type 1 (RyR1) mutations C4958S and C4961S reveal excita- 355. Joulin O, Petillot P, Labalette M, Lancel S, Neviere R. Cytokine profile of human septic
tion-coupled calcium entry (ECCE) is independent of sarcoplasmic reticulum store shock serum inducing cardiomyocyte contractile dysfunction. Physiol Res 56: 291–
depletion. J Biol Chem 280: 36994 –37004, 2005. 297, 2007.

335. Hussain SN, Mofarrahi M, Sigala I, Kim HC, Vassilakopoulos T, Maltais F, Bellenis I, 356. Ju JS, Varadhachary AS, Miller SE, Weihl CC. Quantitation of “autophagic flux” in
Chaturvedi R, Gottfried SB, Metrakos P, Danialou G, Matecki S, Jaber S, Petrof BJ, mature skeletal muscle. Autophagy 6: 929 –935, 2010.

1098 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

357. Karatzanos E, Gerovasili V, Zervakis D, Tripodaki ES, Apostolou K, Vasileiadis I, 378. Kress JP, Pohlman AS, O’Connor MF, Hall JB. Daily interruption of sedative infusions
Papadopoulos E, Mitsiou G, Tsimpouki D, Routsi C, Nanas S. Electrical muscle stim- in critically ill patients undergoing mechanical ventilation. N Engl J Med 342: 1471–
ulation: an effective form of exercise and early mobilization to preserve muscle 1477, 2000.
strength in critically ill patients. Crit Care Res Pract 2012: 432752, 2012.
379. Kroemer G, Marin G, Levine B. Autophagy and the integrated stress response. Mol
358. Kasden SE, Kirkpatrick JR. Endotoxin and calcium effect on metabolism. Am Surg 56: Cell 40: 280 –293, 2010.
707–714, 1990.
380. Kuzmenkin A, Jurkat-Rott K, Lehmann-Horn F, Mitrovic N. Impaired slow inactivation
359. Keller CW, Fokken C, Turville SG, Lünemann A, Schmidt J, Münz C, Lünemann JD. due to a polymorphism and substitutions of Ser-906 in the II-III loop of the human
TNF-alpha induces macroautophagy and regulates MHC II class expression in human Nav1.4 channel. Pflügers Arch 447: 71–77, 2003.
skeletal muscle cells. J Biol Chem 286: 3970 –3980, 2011.
381. Lacomis D. Electrophysiology of neuromuscular disorders in critical illness. Muscle
360. Kerbaul F, Brousse M, Collart F, Pellissier JF, Planche D, Fernandez C, Gouin F, Nerve 47: 452– 463, 2013.
Guidon C. Combination of histopathological and electromyographic patterns can help
382. Lacomis D, Giuliani MJ, Van Gott A, Kramer DJ. Acute myopathy of intensive care:
to evaluate functional outcome of critical ill patients with neuromuscular weakness
clinical, electromyographic, and pathological aspects. Ann Neurol 40: 645– 654, 1996.
syndromes. Crit Care 8: R358 –R366, 2004.
383. Lacomis D, Petrella JT, Giuliani MJ. Causes of neuromuscular weakness in the inten-
361. Khan J, Harrison TB, Rich MM, Moss M. Early development of critical illness myopathy
sive care unit: a study of ninety-two patients. Muscle Nerve 21: 610 – 617, 1998.
and neuropathy in patients with severe sepsis. Neurology 67: 1421–1425, 2006.
384. Lacomis D, Smith TW, Chad DA. Acute myopathy and neuropathy in status asthmati-
362. Kim C, Fuke N, Martyn JAJ. Burn injury to rat increases nicotinic acetylcholine recep-
cus: case report and literature review. Muscle Nerve 16: 84 –90, 1993.
tors in the diaphragm. Anesthesiology 68: 401– 406, 1988.
385. Lacomis D, Zochodne DW, Bird SJ. Critical illness myopathy. Muscle Nerve 23: 1785–
363. Kim C, Martyn J, Fuke N. Burn injury to trunk of rat causes denervation-like responses
1788, 2000.
in the gastrocnemius muscle. J Appl Physiol 65: 1745–1751, 1988.
386. Lagasse RS, Katz RI, Petersen M, Jacobson MJ, Poppers PJ. Prolonged neuromuscular
364. Kim HJ, Higashimori T, Park SY, Choi H, Dong J, Kim YJ, Noh HL, Cho YR, Cline G,
blockade following vecuronium infusion. J Clin Anesth 2: 269 –271, 1990.
Kim YB, Kim JK. Differential effects of interleukin-6 and -10 on skeletal muscle and
liver insulin action in vivo. Diabetes 53: 1060 –1067, 2004. 387. Laghi F, Cattapan SE, Jubran A, Parthasarathy S, Warshawsky P, Choi YS, Tobin MJ. Is
weaning failure caused by low-frequency fatigue of the diaphragm? Am J Respir Crit
365. Kim I, Rodriguez-Enriquez S, Lemasters JJ. Selective degradation of mitochondria by
Care Med 167: 120 –127, 2003.
mitophagy. Arch Biochem Biophys 462: 245–253, 2007.
388. Lamb GD, Cellini MA, Stephenson DG. Different Ca2⫹ releasing action of caffeine and
366. Klaude M, Fredriksson K, Tjäder I, Hammaqvist F, Ahlman B, Rooyackers O, Wer-
depolarisation in skeletal muscle fibers of the rat. J Physiol 531.3: 715–728, 2001.
nerman J. Proteasome proteolytic activity in skeletal muscle is increased in patients
with sepsis. Clin Sci 112: 499 –506, 2007. 389. Lang CH, Frost RA, Vary TC. Regulation of muscle protein synthesis during sepsis and
inflammation. Am J Physiol Endocrinol Metab 293: E453–E459, 2007.
367. Klaude M, Hammarqvist F, Wemerman J. An assay of microsomal membrane-associ-
ated proteasomes demonstrates increased proteolytic activity in skeletal muscle of 390. Lange S, Ehler E, Gautel M. From A to Z and back? Multicompartment proteins in the
intensive care unit patients. Clin Nutr 24: 259 –265, 2005. sarcomere. Trends Cell Biol 16: 11–18, 2006.

368. Klaude M, Mori M, Tjader I, Gustafsson T, Wernerman J, Rooyackers O. Protein 391. Lange S, Xiang F, Yakovenko A, Vihola A, Hackman P, Rostkova E, Kristensen J,
metabolism and gene expression in skeletal muscle of critically ill patients with sepsis. Brandmeier B, Franzen G, Hedberg B, Gunnarsson LG, Hughes SM, Marchand S,
Clin Sci 122: 133–142, 2012. Sejersen T, Richard I, Edstrom L, Ehler E, Udd B, Gautel M. The kinase domain of titin
controls muscle gene expression and protein turnover. Science 308: 1599 –1603,
369. Kleyweg RP, van der Meche FG, Schmitz PI. Interobserver agreement in the assess- 2005.
ment of muscle strength and functional abilities in Guillain-Barre syndrome. Muscle
Nerve 14: 1103–1109, 1991. 392. Langhans C, Weber-Carstens S, Schmidt F, Hamati J, Kny M, Zhu X, Wollersheim T,
Koch S, Krebs M, Schulz H, Lodka D, Saar K, Labeit S, Spies C, Hubner N, Spranger
370. Koch S, Spuler S, Deja M, Bierbrauer J, Dimroth A, Behse F, Spies CD, Wernecke KD, J, Spuler S, Boschmann M, Dittmar G, Butler-Browne G, Mouly V, Fielitz J. Inflamma-
Weber-Carstens S. Critical illness myopathy is frequent: accompanying neuropathy tion-induced acute phase response in skeletal muscle and critical illness myopathy.
protracts ICU discharge. J Neurol Neurosurg Psychiatry 82: 287–293, 2011. PLoS One 9: e92048, 2014.

371. Koesters A, Engisch KL, Rich MM. Decreased cardiac excitability secondary to reduc- 393. Langouche L, Vanhorebeek I, Vlasselaers D, Vander Perre S, Wouters PJ, Skogstrand
tion of sodium current may be a significant contributor to reduced contractility in a rat K, Hansen TK, Van den Berghe G. Intensive insulin therapy protects the endothelium
model of sepsis. Crit Care 18: R54, 2014. of critically ill patients. J Clin Invest 115: 2277–2286, 2005.

372. Komatsu M, Waguri S, Ueno T, Iwata J, Murata S, Tanida I, Ezaki J, Mizushima N, 394. Lanone S, Mebazza A, Heymes C, Henin D, Poderoso JJ, Panis Y, Zedda C, Billiar T,
Ohsumi Y, Uchiyama Y, Kominami E, Tanaka K, Chiba T. Impairment of starvation- Payen D, Aubier M, Boczkowski J. Muscular contractile failure in septic patients: role
induced and constitutive autophagy in Atg7-deficient mice. J Cell Biol 169: 425– 434, of the inducible nitric oxide synthase pathway. Am J Respir Crit Care Med 162: 2308 –
2005. 2315, 2000.

373. Kortebein P, Ferrando A, Lombeida J, Wolfe R, Evans WJ. Effect of 10 days of bed rest 395. Larsson L. Experimental animal models of muscle wasting in intensive care unit pa-
on skeletal muscle in healthy older adults. JAMA 297: 1772–1774, 2007. tients. Crit Care Med 35 Suppl: -S484 – 487, 2007.

374. Kotter S, Unger A, Hamdani N, Lang P, Vorgerd M, Nagel-Steger L, Linke WA. 396. Larsson L. Acute quadriplegic myopathy: an acquired “myosinopathy.” Adv Exp Med
Human myocytes are protected from titin aggregation-induced stiffening by small Biol 642: 92–98, 2008.
heat shock proteins. J Cell Biol 204: 187–202, 2014.
397. Larsson L. Acute Quadriplegic Myopathy, an acquired “myosinopathy.” In: The Sarco-
375. Kouyoumdjian JA. Neuromuscular abnormalities in disues, ageing and cachexia. Arq mere and Skeletal Muscle Diseases, edited by Laing NG. New York: Springer, 2008,
Neuropsiquiatr 51: 299 –306, 1993. vol. 642, chapt. 8, p. 92–98.

376. Kraner SD, Novak KR, Wang Q, Peng J, Rich MM. Altered sodium channel-protein 398. Larsson L, Li X, Berg HE, Frontera WR. Effects of removal of weight-bearing function
associations in critical illness myopathy. Skelet Muscle 2: 17, 2012. on contractility and myosin isoform composition in single human skeletal muscle cells.
Pflügers Arch 432: 320 –328, 1996.
377. Kraner SD, Wang Q, Novak KR, Cheng D, Cool DR, Peng J, Rich MM. Upregulation of
the CaV 1.1-ryanodine receptor complex in a rat model of critical illness myopathy. 399. Larsson L, Li X, Edstrom L, Eriksson LI, Zackrisson H, Argentini C, Schiaffino S. Acute
Am J Physiol Regul Integr Comp Physiol 300: R1384 –R1391, 2011. quadriplegia and loss of muscle myosin in patients treated with nondepolarizing neu-

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1099


FRIEDRICH ET AL.

romuscular blocking agents and corticosteroids: mechanisms at the cellular and mo- 420. Li YP, Schwartz RJ, Waddell ID, Holloway BR, Reid MB. Skeletal muscle myocytes
lecular levels. Crit Care Med 28: 34 – 45, 2000. undergo protein loss and reactive oxygen-mediated NF-␬B activation in response to
tumor necrosis factor-␣. FASEB J 12: 871– 880, 1998.
400. Larsson L, Roland A. Drug induced tetraparesis and loss of myosin. Mild types are
probably overlooked. Lakartidningen 93: 2249 –2254, 1996. 421. Li YP, Reid MB. NF-kappaB mediates the protein loss induced by TNF-alpha in
differentiated skeletal muscle myotubes. Am J Physiol Regul Integr Comp Physiol 279:
401. Latronico N, Bertolini G, Guarneri B, Botteri M, Peli E, Andreoletti S, Bera P, Luciani R1165–R1170, 2000.
D, Nardella A, Vittorielli E, Simini B, Candiani A. Simplified electrophysiological eval-
uation of peripheral nerves in critically ill patients: the Italian multi-centre CRIMYNE 422. Li X, Hughes SM, Salviati G, Teresi A, Larsson L. Thyroid hormone effects on con-
study. Crit Care 11: R11, 2007. tractility and myosin composition of soleus muscle and single fibers from young and
old rats. J Physiol 494: 555–567, 1996.
402. Latronico N, Bolton CF. Critical illness polyneuropathy and myopathy: a major cause
of muscle weakness and paralysis. Lancet Neurol 10: 931–941, 2011. 423. Li X, Moody MR, Engel D, Walker S, Clubb FJ Jr, Sivasubramanian N, Mann DL, Reid
MB. Cardiac-specific overexpression of tumor necrosis factor-alpha causes oxidative
403. Latronico N, Fenzi F, Recupero D, Guarneri B, Tomelleri G, Tonin P, De Maria G, stress and contractile dysfunction in mouse diaphragm. Circulation 102: 1690 –1696,
Antonini L, Rizzuto N, Candiani A. Critical illness myopathy and neuropathy. Lancet 2000.
347: 1579 –1582, 1996.
424. Liang X, Lin T, Sun G, Beasley-Topliffe L, Cavaillon JM, Warren HS. Hemopexin
404. Latronico N, Guarneri B. Critical illness myopathy and neuropathy. Minerva Anesthe- down-regulates LPS-induced proinflammatory cytokines from macrophages. J Leukoc
siol 74: 319 –323, 2008. Biol 86: 229 –235, 2009.

405. Latronico N, Guarneri B, Alongi S, Bussi G, Candiani A. Acute neuromuscular respi- 425. Liaw KY. Effect of injury, sepsis and parenteral nutrition on high-energy phosphates in
ratory failure after ICU discharge. Report of five patients. Intensive Care Med 25: human liver and muscle. J Parenter Enteral Nutr 9: 28 –33, 1985.
1302–1306, 1999.
426. Liaw KY, Wei TC, Hsu SC, Lin JK. Effects of severe injury and critical illness on
406. Latronico N, Shehu I, Seghelini E. Neuromuscular sequelae of critical illness. Curr Opin high-energy phosphates in human liver and muscle. J Trauma 25: 628 – 633, 1985.
Crit Care 11: 381–390, 2005.
427. Lin MC, Ebihara S, El Dwairi Q, Hussain SNA, Yang L, Gottfried SB, Comtois A, Petrof
407. Latronico N, Tomelleri G, Filosto M. Critical illness myopathy. Curr Opin Rheumatol BJ. Diaphragm sarcolemmal injury is induced by sepsis and alleviated by nitric oxide
24: 616 – 622, 2012. synthase inhibition. Am J Respir Crit Care Med 158: 1656 –1663, 1998.

408. Launikonis BS, Stephenson DG. Effect of saponin treatment on the sarcoplasmic 428. Lin SY, Chen WY, Lee FY, Huang CJ, Sheu WH. Activation of ubiquitin-proteasome
resticulum of rat, cane toad and crustacean (yabby) skeletal muscle. J Physiol 504.2: pathway is involved in skeletal muscle wasting in a rat model with biliary cirrhosis:
425– 437, 1997. potential role of TNF-alpha. Am J Physiol Endocrinol Metab 288: E493–E501, 2005.

409. Lecker SH, Solomon V, Mitch WE, Goldberg AL. Muscle protein breakdown and the 429. Lipshutz AK, Gropper MA. Acquired neuromuscular weakness and early mobilization
critical role of the ubiquitin proteasome pathway in normal and disease states. J Nutr in the intensive care unit. Anesthesiology 118: 202–215, 2013.
129: 227S–237S, 1999.
430. Liu MS, Wu LL. Reduction in the Ca2⫹-induced Ca2⫹ release from canine cardiac
410. Lee CM, Fan E. ICU-acquired weakness: what is preventing its rehabilitation in criti- sarcoplasmic reticulum following endotoxin administration. Biochem Biophys Res Com-
cally ill patients? BMC Med 10: 115, 2012. mun 174: 1248 –1254, 1991.

411. Lee HY, Kaneki M, Andreas J, Tompkins RG, Martyn JA. Novel mitochondria-targeted 431. Liu S, Chen JF. Strategies for therapeutic hypometabothermia. J Exp Stroke Transl Med
antioxidant peptide ameliorates burn-induced apoptosis and endoplasmic reticulum 5: 31– 42, 2012.
stress in the skeletal muscle of mice. Shock 36: 580 –585, 2011.
432. Liu SH, Lai JL, Lin RH, Lin MJ, Lin-Shiau SY. Involvement of nitric oxide in the in vivo
412. Lee RS, Tikunova SB, Kilne KP, Zot HG, Hasbun JE, Minh NV, Swartz DR, Rall JA, effects of lipopolysaccharides oon the contractile and electrical properties of mouse
Davis JP. Effect of Ca2⫹ binding properties of troponin C on rate of skeletal diaphragm. Naunyn-Schmiedebergs Arch Pharmacol 356: 500 –504, 1997.
muscle force redevelopment. Am J Physiol Cell Physiol 299: C1091–C1099,
433. Liu SH, Lai JL, Yang RS, Lin-Shiau SY. Nitric oxide is not involved in the endotoxemia-
2010. induced alterations in Ca2⫹ and ryanodine responses in mouse diaphragms. Naunyn-
Schmiedeberg Arch Pharmacol 366: 327–334, 2002.
413. Lefaucheur JP, Nordine T, Rodriguez P, Brochard L. Origin of ICU acquired paresis
determined by direct muscle stimulation. J Neurol Neurosurg Psychiatry 77: 500 –506, 434. Llano-Diez M, Gustafson AM, Olsson C, Goransson H, Larsson L. Muscle wasting and
2006. the temporal gene expression pattern in a novel rat intensive care unit model. BMC
Genomics 12: 602, 2011.
414. Leijten FS, de Weerd AW. Critical illness polyneuropathy. A review of the literature,
definition and pathophysiology. Clin Neurol Neurosurg 96: 10 –19, 1994. 435. Llano-Diez M, Renaud G, Anderson M, Marrero HG, Cacciani N, Enqquist H, Cor-
peno R, Artemenko K, Berqquist J, Larsson L. Mechanisms underlying ICU muscle
415. Leijten FS, de Weerd AW, Poortvliet DC, De Ridder VA, Ulrich C, Harink-De Weerd
wasting and effects of passive mechanical loading. Crit Care 16: R209, 2012.
JE. Critical illness polyneuropathy in multiple organ dysfunction syndrome and wean-
ing from the ventilator. Intensive Care Med 22: 856 – 861, 1996. 436. Llesuy S, Evelson P, Gonzalez-Flecha B, Peralta J, Carreras MC, Poderoso JJ, Boveris
A. Oxidative stress in muscle and liver of rats with septic syndrome. Free Rad Biol Med
416. Leijten FS, Harinck-de Weerd JE, Poortvliet DC, de Weerd AW. The role of polyneu-
16: 445– 451, 1994.
ropathy in motor convalescence after prolonged mechanical ventilation. JAMA 274:
1221–1225, 1995. 437. Llovera M, Carbo N, Lopez-Soriano J, Garcia-Martinez C, Busquets S, Alvarez B, Agell
N, Costelli P, Lopez-Soriano FJ, Celada A, Argiles JM. Different cytokines modulate
417. Leppik JA, Aughey RJ, Medved I, Fairweather I, Carey MF, McKenna MJ. Prolonged
ubiquitin gene expression in rat skeletal muscle. Cancer Lett 133: 83– 87, 1998.
exercise to fatigue in humans impairs skeletal muscle Na⫹-K⫹-ATPase activity, sar-
coplasmic reticulum Ca2⫹ regulation, Ca2⫹ uptake. J Appl Physiol 97: 1414 –1423, 438. Llovera M, Garcia-Martinez C, Lopez-Soriano J, Agell M, Lopez-Soriano FJ, Garcia I,
2004. Argiles JM. Protein turnover in skeletal muscle of tumour-bearing mice overexpress-
ing the soluble TNF receptor-1. Cancer Lett 130: 19 –27, 1998.
418. Li YP, Chen Y, John J, Moylan J, Jin B, Mann DL, Reid MB. TNF-alpha acts via p38
MAPK to stimulate expression of the ubiquitin ligase atrogin1/MAFbx in skeletal mus- 439. Llovera M, Garcia-Martinez C, Lopez-Soriano J, Carbo N, Agell M, Lopez-Soriano FJ,
cle. FASEB J 19: 362–370, 2005. Argiles JM. Role of TNF receptor 1 in protein turnover during cancer cachexia using
gene knockout mice. Mol Cell Endocrionl 142: 183–189, 1998.
419. Li YP, Lecker SH, Chen Y, Waddell ID, Goldberg AL. TNF-alpha increases ubiquitin-
conjugating activity in skeletal muscle by upregulating UbcH2/E220k. FASEB J 17: 440. Lomo T, Rosenthal J. Control of ACh sensitivity by muscle activity in the rat. J Physiol
1048 –1057, 2003. 221: 493–513, 1972.

1100 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

441. Lomo T, Westgaard RH. Further studies on the control of ACh sensitivity by muscle 463. Martyn JAJ, Tomera JF. Acetylcholine receptor density and acetylcholinesterase en-
activity in the rat. J Physiol 252: 603– 626, 1975. zyme activity in skeletal muscle of rats following thermal injury. Anesthesiology 71:
625– 627, 1989.
442. Lopez LC, Escames G, Ortiz F, Ros E, Acuna-Castroviejo D. Melatonin restores the
mitochondrial production of ATP in septic mice. Neuro Endocrinol Lett 27: 623– 630, 464. Martyn JA, White DA, Gronert GA, Jaffe RS, Ward JM. Up-and-down regulation of
2006. skeletal muscle acetylcholine receptors. Effects on neuromuscular blockers. Anesthe-
siology 76: 822– 843, 1992.
443. Lopez LC, Escames G, Tapias V, Utrilla P, Leon J, Acuna-Castroviejo D. Identification
of an inducible nitric oxide synthase in diaphragm mitochondria from septic mice: its 465. Masiero E, Agatea L, Mammucari C, Blaauw B, Loro E, Komatsu M, Metzger D,
relation with mitochondrial dysfunction and prevention by melatonin. Int J Biochem Reggiani C, Schiaffino S, Sandri M. Autophagy is required to maintain muscle mass. Cell
Cell Biol 38: 267–278, 2006. Metab 10: 507–515, 2009.

444. Loven L, Larsson J, Lennquist S, Liljedahl SO. Hypophosphatemia and muscle phos- 466. Massa R, Carpenter S, Holland P, Karpati G. Loss and renewal of thick myofilaments
phate metabolism in severely injured patients. Acta Chir Scand 149: 743–749, 1983. in glucocorticoid-treated rat soleus after denervation and reinnervation. Muscle Nerve
15: 1290 –1298, 1992.
445. Luo G, Hershko DD, Robb BW, Wray CJ, Hasselgren PO. IL-1beta stimulates IL-6
production in cultured skeletal muscle cells through activation of MAP kinase signaling 467. Matsumoto A, Fujita N, Arakawa T, Fujino H, Miki A. Influence of electrical stimula-
pathway and NF-kappaB. Am J Physiol Regul Integr Comp Physiol 284: R1249 –R1254, tion on calpain and ubiquitin-proteasome systems in the denervated and unloaded rat
2003. tibialis anterior muscles. Acta Histochem 116: 936 –942, 2014.

446. Lyfenko AD, Dirksen RT. Differential dependence of store-operated and excitation- 468. McArdle JJ. Molecular aspects of the trophic influence of nerve on muscle. Prog
coupled Ca2⫹ entry in skeletal muscle on STIM1 and ORAI1. J Physiol 586: 4815– Neurobiol 21: 135–198, 1983.
4824, 2008.
469. McCarter FD, Nierman SR, James JH, Wang L, Friedn LA, Fischer JE. Role of skeletal
447. Lynch GS, Fary CJ, Williams DA. Quantitative measurement of resting skeletal muscle muscle Na⫹K⫹ ATPase activity in increased lactate production in sub-acute sepsis.
[Ca2⫹]i following acute and long-term downhill running exercise in mice. Cell Calcium Life Sci 70: 1875–1888, 2002.
22: 373–383, 1997.
470. McCowen KC, Malhotra A, Bistrian BR. Stress-induced hyperglycemia. Crit Care Clin
448. MacFarlane IA, Rosenthal FD. Severe myopathy after status asthmaticus. Lancet 2: 17: 107–124, 2001.
615, 1977.
471. McNicol FJ, Hoyland JA, Cooper RG, Carslon GL. Skeletal muscle contractile prop-
449. Mador MJ, Deniz O, Aggarwal A, Kufel TJ. Quadriceps fatigability after single muscle erties and proinflammatory cytokine gene expression in human endotoxaemia. Br J
exercise in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Surg 97: 434 – 442, 2010.
Med 168: 102–108, 2003.
472. Mealy K, van Lanschott JJ, Robinson BG, Rounds J, Wilmore DW. Are the catabolic
450. Maher J, Rutledge F, Remtulla H, Parkes A, Bernardi L, Bolton CF. Neuromuscular effects of tumor necrosis factor mediated by glucocorticoids? Arch Surg 125: 42– 48,
disorders associated with failure to wean from the ventilator. Intensive Care Med 21: 1990.
737–743, 1995.
473. Meduri GU, Headley AS, Golden E, Carson SJ, Umberger RA, Kelso T, Tolley EA.
451. Mammucari C, Milan G, Romanello V, Masiero E, Rudolf R, Del Piccolo P, Burden SJ, Effect of prolonged methylprednisolone therapy in unresolving acute respiratory dis-
Di Lisi R, Sandri C, Zhao J, Goldberg AL, Schiaffino S, Sandri M. FoxO3 controls tress syndrome: a randomized controlled trial. JAMA 280: 159 –165, 1998.
autophagy in skeletal muscle in vivo. Cell Metab 6: 458 – 471, 2007.
474. Meesen RL, Dendale P, Cuypers K, Berger J, Hermans A, Thijs H, Levin O. Neuro-
452. Manzanares W, Dhaliwal R, Jiang X, Murch L, Heyland DK. Antioxidant micronutri- muscular electrical stimulation as a possible means to prevent muscle tissue wasting in
ents in the critically ill: a systematic review and meta-analysis. Crit Care 16: R66, 2012. artificially ventilated and sedated patients in the intensive care unit: a pilot study.
Neuromodulation 13: 315–320, 2010.
453. Maramattom B, Wijdicks EFM, Sundt TM, Cassivi SD. Flaccid quadriplegia due to
necrotizing myopathy following lung transplantation. Transplant Proc 36: 2830 –2833, 475. Mela-Riker L, Bartos D, Vlessis AA, Widener L, Muller P, Trunkey DD. Chronic
2004. hyperdynamic sepsis in the rat. Characterization of liver and energy metabolism. Circ
Shock 36: 83–92, 1992.
454. Marathe PH, Dwersteg JF, Pavlin EG, Haschke RH, Heimbach DM, Slattery JT. Effect
of thermal injury on the pharmacokinetics and pharmacodynamics of atracurium in 476. Melzer W, Dietze B. Malignant hyperthermia and excitation-contraction coupling.
humans. Anesthesiology 70: 752–755, 1989. Acta Physiol Scand 171: 367–378, 2001.

455. Marathe PH, Haschke RH, Slattery JT, Zucker JR, Pavlin EG. Acetylcholine receptor 477. Melzer W, Herrmann-Frank A, Lüttgau Ch. The role of Ca2⫹ ions in excitation-
density and acetylcholine esterase activity in skeletal muscle of rats following thermal contraction coupling of skeletal muscle fibers. Biochim Biophys Acta 1241: 59 –116,
injury. Anesthesiology 70: 654 – 659, 1989. 1995.

456. Martensson J, Goodwin CW, Blake R. Mitochondrial glutathione in hypermetabolic 478. Melzer W, Rios E, Schneider MF. A general procedure for determining the rate of
rats following burn injury and thyroid hormone administration: evidence of a selective calcium release from the sarcoplasmic reticulum in skeletal muscle fibers. Biophys J 51:
effect on brain glutathione by burn injury. Metabolism 41: 273–277, 1992. 849 – 863, 1987.

457. Martin AF. Turnover of cardiac troponin subunits. Kinetic evidence for a precursor 479. Menconi MJ, Arany ZP, Alamdari N, Aversa Z, Gonnella P, O’Neal P, Smith IJ, Tizio S,
pool of troponin-I. J Biol Chem 256: 964 –968, 1981. Hasselgren PO. Sepsis and glucocorticoids downregulate the expression of the nu-
clear cofactor PGC-1beta in skeletal muscle. Am J Physiol Endocrinol Metab 299: E533–
458. Martin CM, Hill AD, Burns K, Chen LM. Characteristics and outcomes for critically ill E543, 2010.
patients with prolonged intensive care unit stays. Crit Care Med 33: 1922–1927, 2005.
480. Mera S, Tatulescu D, Cismaru C, Bondor C, Slavcvici A, Zanc V, Carstina D, Oltean M.
459. Martin UJ, Hincapie L, Nimchuk M, Gaughan J, Criner GJ. Impact of whole-body Multiplex cytokine profiling in patients with sepsis. APMIS 119: 155–163, 2011.
rehabilitation in patients receiving chronic mechanical ventilation. Crit Care Med 33:
2259 –2265, 2005. 481. Merx MW, Weber C. Sepsis and the heart. Circulation 116: 793– 802, 2007.

460. Martyn JA, Fagerlund MJ, Eriksson LI. Basic principles of neuromuscular transmission. 482. Mesotten D, Gielen M, Sterken C, Claessens K, Hermans G, Vlasselaers D, Lemiere J,
Anaesthesia 64 Suppl 1: 1–9, 2009. Lagae L, Gewillig M, Eyskens B, Vanhorebeek I, Wouters PJ, Van den Berghe G.
Neurocognitive development of children 4 years after critical illness and treatment
461. Martyn JA, Richtsfeld M. Succinylcholine-induced hyperkalemia in acquired pathologic with tight glucose control: a randomized controlled trial. JAMA 308: 1641–1650, 2012.
states. Anesthesiology 104: 158 –169, 2006.
483. Mesotten D, Swinnen JV, Vanderhoydonc F, Wouters PJ, Van den Berghe G. Contri-
462. Martyn JAJ, Szfelbein SK, Ali HH, Matteo RS, Savarese JJ. Increased d-tubocurarine bution of circulating lipids to the improved outcome of critical illness by glycemic
requirements following major thermal injury. Anesthesiology 52: 352–355, 1980. control with intensive insulin therapy. J Clin Endocrinol Metab 89: 219 –226, 2004.

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1101


FRIEDRICH ET AL.

484. Mester M, Carter EA, Tompkins RG, Gelfand JA, Dinarello CA, Burke JF, Clark BD. Morris A, Dong N, Rock P. Initial trophic vs full enteral feeding in patients with acute
Thermal injury induces very early production of interleukin-1 alpha in the rat by lung injury: the EDEN randomized trial. JAMA 307: 795– 803, 2012.
mechanisms other than endotoxemia. Surgery 115: 588 –596, 1994.
504. Needham DM, Dinglas VD, Bienvenu OJ, Colantuoni E, Wozniak AW, Rice TW,
485. Michie HR, Sherman ML, Spriggs DR, Rounds J, Christie M, Wilmore DW. Chronic Hopkins RO. One year outcomes in patients with acute lung injury randomised to
TNF infusion causes anorexia but not accelerated nitrogen loss. Ann Surg 209: 19 –24, initial trophic or full enteral feeding: prospective follow-up of EDEN randomised trial.
1989. BMJ 346: f1532, 2013.

486. Minnaard R, Wagenmakers AJ, Combaret L, Attaix D, Drost MR, van Kranenburg GP, 505. Needham DM, Korupolu R, Zanni JM, Pradhan P, Colantuoni E, Palmer JB, Brower
Schaart G, Hesselink MK. Ubiquitin-proteasome-dependent proteolytic activity re- RG, Fan E. Early physical medicine and rehabilitation for patients with acute respira-
mains elevated after zymosan-induced sepsis in rats while muscle mass recovers. Int J tory failure: a quality improvement project. Arch Phys Med Rehabil 91: 536 –542, 2010.
Biochem Cell Biol 37: 2217–2225, 2005.
506. Nethery D, Callahan LA, Stofan D, Mattera R, Di Marco A, Supinski G. PLA2 depen-
487. Mirzakhani H, Williams JN, Mello J, Joseph S, Meyer MJ, Waak K, Schmidt U, Kelly E, dence of diaphragm mitochondrial formation of reactive oxygen species. J Appl Physiol
Eikermann M. Muscle weakness predicts pharyngeal dysfunction and symptomatic 89: 72– 80, 2000.
aspiration in long-term ventilated patients. Anesthesiology 119: 389 –397, 2013.
507. Newham P, Ross D, Ceuppens P, Das S, Yates JW, Betts C, Reens J, Randall KJ, Knight
488. Miyakawa H, Oishi K, Hagiwara S, Kira S, Kitano T, Iwasaka H, Noguchi T. Olprinone
R, McKay JS. Determination of the safety and efficacy of therapeutic neutralization of
improves diaphragmatic contractility and fatigability during abdominal sepsis in a rat
tumor necrosis factor-␣ (TNF-␣) using AZD9773, an anti-TNF-␣ immune Fab, in
model. Acta Anaesthesiol Scand 48: 637– 641, 2004.
murine CLP sepsis. Inflamm Res 63: 149 –160, 2014.
489. Mizobata Y, Prechek D, Rounds JD, Robinson V, Wilmore DW, Jacobs DO. The
508. Niiya T, Narimatsu E, Maiki A. Acute late sepsis attenuates effects of a nondepolar-
duration of infection modifies mitochondrial oxidative capacity in rat skeletal muscle.
izing neuromuscular blocker, rocuronium, by facilitation of endplate potential and
J Surg Res 59: 165–173, 1995.
enhancement of membrane excitability in vitro. Anesthesiology 105: 968 –975, 2006.
490. Mofarrahi M, Sigala I, Guo Y, Godin R, Davis EC, Petrof B, Sandri M, Buruelle Y,
510. Nordquist J, Hoglund AS, Norman H, Tang X, Dworkin B, Larsson L. Transcription
Hussain SN. Autophagy and skeletal muscles in sepsis. PLoS One 7: e47265, 2012.
factors in muscle atrophy caused by blocked neuromuscular transmission and muscle
491. Moldawer LL, Swaninger G, Gelin J, Lundholm K. Interleukin 1 (alpha or beta) and unloading in rats. Mol Med 13: 461– 470, 2007.
tumor necrosis factor-alpha do not regulate protein balance in skeletal muscle. Am J
Physiol Cell Physiol 253: C766 –C773, 1987. 511. Norman H, Kandala K, Kolluri R, Zackrisson H, Nordquist J, Walther S, Eriksson LI,
Larsson L. A porcine model of acute quadriplegic myopathy: a feasibility study. Acta
492. Montpetit ML, Stocker PJ, Schwetz TA, Harper JM, Norring SA, Schaffer L, North SJ, Anaesthesiol Scand 50: 1058 –1067, 2006.
Jang-Lee J, Gilmartin T, Head SR, Haslam SM, Dell A, Marth JD, Bennett ES. Regulated
and aberrant glycosylation modulate cardiac electrical signaling. Proc Natl Acad Sci USA 512. Norman H, Nordquist J, Andersson P, Ansved T, Tang X, Dworkin B, Larsson L.
106: 16517–16522, 2009. Impact of post-synaptic block of neuromuscular transmission, muscle unloading and
mechanical ventilation on skeletal muscle protein and mRNA expression. Pflügers Arch
493. Moriggi M, Cassano P, Vasso M, Capitanio D, Fania C, Musicco C, Pesce V, Gadaleta 453: 53– 66, 2006.
MN, Gelfi C. A DIGE approach for the assessment of rat soleus muscle changes during
unloading: effect of acetyl-L-carnitine aupplementation. Proteomics 8: 3588 –3604, 513. Norman H, Zackrisson H, Hedstrom Y, Andersson P, Nordquist J, Eriksson LI, Libe-
2008. lius R, Larsson L. Myofibrillar protein and gene expression in acute quadriplegic my-
opathy. J Neurol Sci 285: 28 –38, 2009.
494. Morris CE, Boucher PA, Joos B. Left-shifted nav channels in injured bilayer: primary
targets for neuroprotective nav antagonists? Front Pharmacol 3: 19, 2012. 514. Novak F, Heyland DK, Avenell A, Drover JW, Su X. Glutamine supplementation in
serious illness: a systematic review of the evidence. Crit Care Med 30: 2022–2029,
495. Morris PE, Goad A, Thompson C, Taylor K, Harry B, Passmore L, Ross A, Anderson 2002.
L, Baker S, Sanchez M, Penley L, Howard A, Dixon L, Leach S, Small R, Hite RD,
Haponik E. Early intensive care unit mobility therapy in the treatment of acute respi- 515. Novak KR, Nardelli P, Cope TC, Filatov G, Glass JD, Khan J, Rich MM. Inactivation of
ratory failure. Crit Care Med 36: 2238 –2243, 2008. sodium channels underlies reversible neuropathy during critical illness in rats. J Clin
Invest 119: 1150 –1158, 2009.
496. Mozaffar T, Haddad F, Zeng M, Zhang LY, Adams GR, Baldwin KM. Molecular and
cellular defects of skeletal muscle in an animal model of acute quadriplegic myopathy. 516. Nyberg P, Wikman AL, Nennesmo I, Lundberg I. Increased expression of interleukin
Muscle Nerve 35: 55– 65, 2007. 1alpha and MHC class I in muscle tissue of patients with chronic, inactive polymyositis
and dermatomyositis. J Rheumatol 27: 940 –948, 2000.
497. Munoz-Canoves P, Scheele C, Pedersen BK, Serrano AL. Interleukin-6 myokine sig-
naling in skeletal muscle: a double-edged sword? FEBS J 280: 4131– 4148, 2013. 516a.Nyitrai M, Rossi R, Adamek N, Pellegrino MA, Bottinelli R, Geeves MA. What limits the
velocity of fast-skeletal muscle contraction in mammals? J Mol Biol 355: 432– 442,
498. Murton AJ, Constantin D, Greenhaff PL. The involvement of the ubiquitin proteasome
2006.
system in human skeletal muscle remodeling and atrophy. Biochim Biophys Acta 1782:
730 –743, 2008. 517. Ochala J, Ahlbeck K, Radell PJ, Eriksson LI, Larsson L. Factors underlying the early limb
muscle weakness in acute quadriplegic myopathy using an experimental ICU porcine
499. Nanas S, Kritikos K, Angelopoulos E, Siafaka A, Tsikriki S, Poriazi M, Kanaloupiti D,
Kontogeorgi M, Pratikaki M, Zervakis D, Routsi C, Roussos C. Predisposing factors for model. PLoS One 6: e20876, 2011.
critical illness polyneuromyopathy in a multidisciplinary intensive care unit. Acta Neu-
518. Ochala J, Gustafson AM, Diez ML, Renaud G, Li M, Aare S, Qaisar R, Banduseela VC,
rol Scand 118: 175–181, 2008.
Hedstrom Y, Tang X, Dworkin B, Ford GC, Nair KS, Perera S, Gautel M, Larsson L.
500. Nardelli P, Khan J, Powers R, Cope TC, Rich MM. Reduced motor neuron excitability Preferential skeletal muscle myosin loss in response to mechanical silencing in a novel
in a rat model of sepsis. J Neurophysiol 109: 1775–1781, 2013. rat intensive care unit model: underlying mechanisms. J Physiol 589: 2007–2026, 2011.

501. Narimatsu E, Nakayama Y, Sumita S, Iwasaki H, Fujimura N, Satoh K, Namiki A. Sepsis 519. Ochala J, Larsson L. Acquired and hereditary sarcomeric protein diseases. In: Muscle:
attenuates the intensity of the neuromuscular blocking effect of d-tubocurarine and Fundamental Biology and Mechanisms and Disease, edited by Joseph JTS, Hill A,
the antagonistic actions of neostigmine and edrophonium accompanying depression Sweeney HL, Olson EN. London: Academic, 2012, p. 1023–1030.
of muscle contractility of the diaphragm. Acta Anaesthesiol Scand 43: 196 –201, 1999.
520. Ochala J, Renaud G, Llano Diez M, Banduseela VC, Aare S, Ahlbeck K, Radell PJ,
502. Narimatsu E, Niiya T, Kawamata M, Namiki A. Sepsis stage dependently and differ- Eriksson LI, Larsson L. Diaphragm muscle weakness in an experimental porcine in-
entially attenuates the effects of nondepolarizing neuromuscular blockers on the rat tensive care unit model. PLoS One 6: e20558, 2011.
diaphragm in vitro. Anesth Analg 100: 823– 829, 2005.
521. Oettgen HF, Carswell EA, Kassel RL, Fiore N, Williamson B, Hoffmann MK, Haranaka
503. National Heart, Lung, and Blood Institute Acute Respiratory Distress Syndrome Clin- K, Old LJ. Endotoxin-induced tumor necrosis factor. Recent results. Cancer Res 75:
ical Trials, Rice TW, Wheeler AP, Thompson BT, Steingrub J, Hite RD, Moss M, 207–212, 1980.

1102 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

522. Offner PJ, Ogura H, Jordan BS, Pruitt BA Jr, Cioffi WG. Effects of inhaled nitric oxide 543. Peruchi BB, Petronilho F, Rojas HA, Constantino L, Mina F, Vuolo F, Cardoso MR,
on right ventricular function in endotoxin shock. J Trauma 39: 179 –186, 1995. Goncalves CL, Rezin GT, Streck EL, Dal-Pizzol F. Skeletal muscle electron transport
chain dysfunction after sepsis in rats. J Surg Res 167: e333– e338, 2011.
523. Ogilvie H, Larsson L. The effect of nutritional status in the pathogenesis of critical
illness myopathy (CIM). Biology 3: 368 –382, 2014. 544. Person RJ. Endotoxin alters spontaneous transmitter release at the frog neuromus-
cular junction. J Neurosci Res 3: 63–72, 1977.
524. Ojuka EO, Jones TE, Han DH, Chen M, Wamhoff BR, Sturek M, Holloszy JO. Inter-
mittent increases in cytosolic Ca2⫹ stimulate mitochondrial biogenesis in muscle cells. 545. Petersen AMW, Pedersen BK. The anti-inflammatory effect of exercise. J Appl Physiol
Am J Physiol Endocrinol Metab 283: E1040 –E1045, 2002. 98: 1154 –1162, 2005.

525. O’Leary MFN, Hood DA. Effect of prior chronic contractile activity on mitochondrial 546. Peterson JM, Bakkar N, Guttridge DC. NF-␬B signaling in skeletal muscle health and
function and apoptotic protein expression in denervated muscle. J Appl Physiol 105: disease. Curr Top Dev Biol 96: 85–119, 2011.
114 –120, 2008.
547. Pinet C, Le Grand B, John GW, Coulombe A. Thrombin facilitation of voltage-gated
526. O’Leary MFN, Hood DA. Denervation-induced oxidative stress and autophagy sig- sodium channel activation in human cardiomyocytes: implications for ischemic sodium
naling in muscle. Autophagy 5: 230 –231, 2009. loading. Circulation 106: 2098 –2103, 2002.

527. Opata MM, Ye Z, Hollifield M, Garvy BA. B cell production of tumor necrosis factor in 548. Pinsky MR, Vincent JL, Deviere J, Alegre M, Kahn RJ, Duport E. Serum cytokine levels
response to Pneumocystis murina infection in mice. Infect Immun 81: 4252– 4260, in human septic shock. Relation to multiple-system organ failure and mortality. Chest
2013. 103: 565–575, 1993.

528. Op de Coul AA, Verheul GA, Leyten AC, Schellens RL, Teepen JL. Critical illness 549. Piper RD, Pitt-Hyde M, Li F, Sibbald WJ, Potter RF. Microcirculatory changes in rat
polyneuromyopathy after artificial respiration. Clin Neurol Neurosurg 96: 261–263, skeletal muscle in sepsis. Am J Respir Crit Care Med 154: 931–937, 1996.
1991.
550. Plomguaard P, Penkowa M, Pedersen BK. Fiber type specific expression of TNF-
529. Op de Coul AA, Lambregts PC, Koeman J, van Puyenbroek MJ, Ter Laak HJ, alpha, IL-6 and IL-18 in human skeletal muscle. Exerc Immunol Rev 11: 53– 63, 2005.
Gabreels-Festen AA. Neuromuscular complications in patients given Pavulon
(pancuronium bromide) during artificial ventilation. Clin Neurol Neurosurg 87: 551. Pohlman MC, Schweickert WD, Pohlman AS, Nigos C, Pawlik AJ, Esbrook CL, Spears
17–22, 1985. L, Miller M, Franczyk M, Deprizio D, Schmidt GA, Bowman A, Barr R, McCallister K,
Hall JB, Kress JP. Feasibility of physical and occupational therapy beginning from
530. Orellana RA, Suryawan A, Wilson FA, Gazzaneo MC, Fiorotto ML, Nguyen HV, Davis initiation of mechanical ventilation. Crit Care Med 38: 2089 –2094, 2010.
TA. Development aggravates the severity of skeletal muscle catabolism induced by
endotoxemia in neonatal pigs. Am J Physiol Regul Integr Comp Physiol 302: R682–R690, 552. Porta F, Takala J, Weikert C, Bracht H, Kolarova A, Lauterburg BH, Borotto E, Jakob
2012. SM. Effects of prolonged endotoxemia on liver, skeletal muscle and kidney mitochon-
drial function. Crit Care 10: R118, 2006.
531. Osler W. The Principles and Practice of Medicine. New York: Appleton, 1892.
553. Porter C, Herndon DN, Sidossis LS, Borsheim E. The impact of severe burns on
532. Ostrowski K, Schjerling P, Pedersen BK. Physical activity and plasma interleukin-6 in skeletal muscle mitochondrial function. Burns 39: 1039 –1047, 2013.
humans: effect of intensity of exercise. Eur J Appl Physiol 83: 512–515, 2000.
554. Posterino GS, Lamb GD. Effect of sarcoplasmic reticulum Ca2⫹ content on action
533. Osuchowski MF, Welch K, Siddiqui J, Remich DG. Circulating cytokine/inhibitor pro- potential-induced Ca2⫹ release in rat skeletal muscle fibers. J Physiol 551.1: 219 –237,
files reshape the understanding of the SIRS/CARS continuum in sepsis and predict 2003.
mortality. J Immunol 177: 1967–1974, 2006.
555. Poulsen JB, Moller K, Jensen CV, Weisdorf S, Kehlet H, Perner A. Effect of transcu-
534. Padfield KE, Astrakas LG, Zhang Q, Gopalan S, Dai G, Mindrinos MN, Tompkins RG, taneous electrical muscle stimulation on muscle volume in patients with septic shock.
Rhame LG, Tzika AA. Burn injury causes mitochondrial dysfunction in skeletal muscle. Crit Care Med 39: 456 – 461, 2011.
Proc Natl Acad Sci USA 102: 5368 –5373, 2005.
556. Powers SK, Hudson MB, Nelson WB, Talbert EE, Min K, Szeto HH, Kavazis AN,
535. Pandit L, Agrawal A. Neuromuscular disorders in critical illness. Clin Neurol Neurosurg Smuder AJ. Mitochondria-targeted antioxidants protect against mechanical ventila-
108: 433– 439, 2006. tion-induced diaphragm weakness. Crit Care Med 39: 1749 –1759, 2011.

536. Papazian L, Forel JM, Gacouin A, Penot-Ragon C, Perrin G, Loundou A, Jaber S, Arnal 557. Preiser JC, Devos P, Ruiz-Santana S, Melot C, Annane D, Groeneveld J, Iapichino G,
JM, Perez D, Seghboyan JM, Constantin JM, Courant P, Lefrant JY, Guerin C, Prat G, Leverve X, Nitenberg G, Singer P, Wernerman J, Joannidis M, Stecher A, Chiolero R.
Morange S, Roch A. Neuromuscular blockers in early acute respiratory distress syn- A prospective randomised multi-centre controlled trial on tight glucose control by
drome. N Engl J Med 363: 1107–1116, 2010. intensive insulin therapy in adult intensive care units: the Glucontrol study. Intensive
Care Med 35: 1738 –1748, 2009.
537. Parker CJ, Sardesai VM. The effect of E. coli endotoxin on cardiac and skeletal muscle
myofibrillar ATPase. Biochem Med 8: 11–17, 1973. 558. Prosser BL, Hernandez-Ochoa EO, Schneider MF. S100A1 and calmodulin regulation
of ryanodine receptor in striated muscle. Cell Calcium 50: 323–331, 2011.
538. Partridge BL, Abrams JH, Bazemore C, Rubin R. Prolonged neuromuscular blockade
after long-term infusion of vecuronium bromide in the intensive care unit. Crit Care 559. Puchner EM, Alexandrovich A, Kho AL, Hensen U, Schafer LV, Brandmeier B, Grater
Med 18: 1177–1179, 1990. F, Grubmuller H, Gaub HE, Gautel M. Mechanoenzymatics of titin kinase. Proc Natl
Acad Sci USA 105: 13385–13390, 2008.
539. Paterniti I, Impellizzeri D, Di Paola R, Esposito E, Gladman S, Yip P, Priestly JV,
Michael-Titus AT, Cuzzocrea S. Docosahexaenoic acid attenuates the early inflam- 560. Puthucheary Z, Harridge S, Hart N. Skeletal muscle dysfunction in critical care: wast-
matory response following spinal cord injury in mice: in-vivo and in-vitro studies. J ing, weakness, and rehabilitation strategies. Crit Care Med 38: S676 –S682, 2010.
Neuroinflammation 11: 6, 2014.
561. Puthucheary Z, Montgomery H, Moxham J, Harridge S, Hart N. Structure to function:
540. Pauly M, Daussin F, Burelle Y, Li T, Godin R, Fauconnier J, Koechlin-Ramonatxo C, muscle failure in critically ill patients. J Physiol 588: 4641– 4648, 2010.
Hugon G, Lacampagne A, Coisy-Quivy M, Liang F, Hussain S, Matecki S, Petrof BJ.
AMPK activation stimulates autophagy and ameliorates muscular dystrophy in the 562. Puthucheary Z, Rawal J, Ratnayake G, Harridge S, Montgomery H, Hart N. Neuro-
mdx mouse diaphragm. Am J Pathol 181: 583–592, 2012. muscular blockade and skeletal muscle weakness in critically ill patients: time to
rethink the evidence? Am J Respir Crit Care Med 185: 911–917, 2012.
541. Pedersen TH, de Paoli F, Nielsen OB. Increased excitability of acidified skeletal mus-
cle: role of chloride conductance. J Gen Physiol 125: 237–246, 2005. 563. Puthucheary ZA, Rawal J, McPhail M, Connolly B, Ratnayake G, Chan P, Hopkinson
NS, Phadke R, Dew T, Sidhu PS, Velloso C, Seymour J, Agley CC, Selba A, Limb M,
542. Pereira RM, Freire de CJ. Glucocorticoid-induced myopathy. Joint Bone Spine 78: Edwards LM, Smith K, Rowlerson A, Rennie MJ, Moxham J, Harridge SD, Hart N,
41– 44, 2011. Montgomery HE. Acute muscle wasting in critical illness. JAMA 310: 1591–1600, 2013.

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1103


FRIEDRICH ET AL.

564. Pytel P, Alexander JJ. Pathogenesis of septic encephalopathy. Curr Opin Neurol 22: 587. Rittirsch D, Hoesel LM, Ward PA. The disconnect between animal models of sepsis
283–287, 2009. and human sepsis. J Leukoc Biol 81: 137–143, 2007.

565. Quy PN, Kuma A, Piere P, Mizushima N. Proteasome-dependent activation of mam- 588. Rittirsch D, Huber-Lang MS, Flierl MA, Ward PA. Immunodesign of experimental
malian target of rapamycin complex 1 (mTOR1) is essential for autophagy suppression sepsis by cecal ligation and puncture. Nature Protocols 4: 31–36, 2009.
and muscle remodeling following denervation. J Biol Chem 288: 1125–1134, 2013.
589. Rodriguez PO, Setten M, Maskin LP, Bonelli I, Vidomlansky SR, Attie S, Frosiani SL,
566. Radell PJ, Remahl S, Nichols DG, Eriksson LI. Effects of prolonged mechanical venti- Kozima S, Valentini R. Muscle weakness in septic patients requiring mechanical ven-
lation and inactivity on piglet diaphragm function. Intensive Care Med 28: 358 –364, tilation: protective effect of transcutaneous neuromuscular electrical stimulation. J
2002. Crit Care 27: 319 –328, 2012.

567. Rannou F, Pennec JP, Rossignol B, Morel J, Dorange G, Arvieux C, Gioux M, Giroux- 590. Roelofs RI, Cerra F, Bielka N, Rosenberg L, Delaney J. Prolonged respiratory
Metges MA. Effects of chronic sepsis on rat motor units: experimental study of critical insufficiency due to acute motor neuropathy: a new syndrome? Neurology 33: 240,
illness polyneuromyopathy. Exp Neurol 204: 741–747, 2007. 1983.

568. Rawat R, Cohen TV, Ampong B, Francia D, Henriques-Pons A, Hoffman EP, Nagaraju 591. Rofe AM, Conyers RAJ, Bais R, Gamble JR, Vadas MA. The effects of recombinant
K. Inflammasome up-regulation and activation in dysferlin-deficient skeletal muscle. tumor necrosis factor (cachectin) on metabolism in isolated rat adipocyte, hepato-
Am J Pathol 176: 2891–2900, 2010. cyte, and muscle preparations. Biochem J 247: 789 –792, 1987.

569. Reed SA, Sandesara PB, Senf SM, Judge AR. Inhibition of FoxO transcriptional activity 592. Rooyackers OE, Kersten AH, Wagenmakers AJ. Mitochondrial protein content
prevents muscle fiber atrophy during cachexia and induces hypertrophy. FASEB J 26: and in vivo synthesis rates in skeletal muscle from critically ill rats. Clin Sci 91:
987–1000, 2012. 475– 481, 1996.

570. Reid MB, Khawli FA, Moody MR. Reactive oxygen species in skeletal muscle. III. 593. Rossie S. Regulation of voltage-sensitive sodium and calcium channels by phosphory-
lation. Adv Second Messenger Phosphoprotein Res 33: 23– 48, 1999.
Contractility of unfatigued muscle. J Appl Physiol 75: 1081–1087, 1992.
594. Rossignol B, Gueret G, Pennec JP, Morel J, Giroux-Metges MA, Talarmin H, Arvieux
571. Reid MB, Lännergren J, Westerblad H. Respiratory and limb muscle weakness induced
CC. Effects of chronic sepsis on the voltage-gated sodium channel in isolated rat
by tumor necrosis factor-␣. Involvement of muscle myofilaments. Am J Respir Crit Care
muscle fibers. Crit Care Med 35: 351–357, 2007.
Med 166: 479 – 484, 2002.
595. Rossignol B, Gueret G, Pennec JP, Morel J, Rannou F, Giroux-Metges MA, Talarmin H,
572. Reid MB, Stokic DS, Koch SM, Khawli FA, Leis AA. N-acetyl-cysteine inhibits muscle
Gioux M, Arvieux C. Effects of chronic sepsis on contractile properties of fast twitch
fatigue in humans. J Clin Invest 94: 2468 –2474, 1994.
muscle in an experimental model of critical illness neuromyopathy in the rat. Crit Care
573. Remels AH, Gosker HR, Bakker J, Guttridge DC, Schols AM, Langen RC. Regulation Med 36: 1855–1863, 2008.
of skeletal muscle oxidative phenotype by classical NF-␬B signalling. Biochem Biophys
596. Rossiter A, Souney PF, McGowan S, Carvajal P. Pancuronium-induced prolonged
Acta 1832: 1313–1325, 2013.
neuromuscular blockade. Crit Care Med 19: 1583–1587, 1991.
574. Renaud G, Llano-Diez M, Ravara B, Gorza L, Feng HZ, Jin JP, Cacciani N, Gustafson
597. Rouleau G, Karpati G, Carpenter S, Soza M, Prescott S, Holland P. Glucocorticoid
AM, Ochala J, Corpeno R, Li M, Hedstrom Y, Ford GC, Nair KS, Larsson L. Sparing of excess induces preferential depletion of myosin in denervated skeletal muscle fibers.
muscle mass and function by passive loading in an experimental intensive care unit Muscle Nerve 10: 428 – 438, 1987.
model. J Physiol 591: 1385–1402, 2013.
598. Routsi C, Gerovasili V, Vasileiadis I, Karatzanos E, Pitsolis T, Tripodaki ES, Markaki V,
575. Rennie MJ. Anabolic resistance in critically ill patients. Crit Care Med 37: S398 –S399, Zervakis D, Nanas S. Electrical muscle stimulation prevents critical illness polyneuro-
2009. myopathy: a randomized parallel intervention trial. Crit Care 14: R74, 2010.
576. Rennie MJ, Wackerhage H, Spangenburg EE, Booth FW. Control of the size of the 599. Ruff RL. Sodium channel regulation of skeletal muscle membrane excitability. Ann NY
human muscle mass. Annu Rev Physiol 66: 799 – 828, 2004. Acad Sci 835: 64 –76, 1997.

577. Rich MM, Bird SJ, Raps EC, McCluskey LF, Teener JW. Direct muscle stimulation in 600. Ruff RL, Secrist D. Inhibitors of prostaglandin synthesis or cathepsin B prevent muscle
acute quadriplegic myopathy. Muscle Nerve 20: 665– 673, 1997. wasting due to sepsis in the rat. J Clin Invest 73: 1483–1486, 1984.

578. Rich MM, Kraner SD, Barchi RL. Altered gene expression in steroid-treated dener- 601. Ryan NT, Blackburn GL, Clowes GHA. Differential tissue sensitivity to elevated en-
vated muscle. Neurobiol Dis 6: 515–522, 1999. dogenous insulin levels during experimental peritonitis in rats. Metabolism 23: 1081,
1974.
579. Rich MM, McGarvey ML, Teener JW, Frame LH. ECG changes during septic shock.
Cardiology 97: 187–196, 2002. 602. Safranek R, Ishibashi N, Oka Y, Ozaa H, Shirouzu K, Holecek M. Modulation of
inflammatory response in sepsis by proteasome inhibition. Int J Exp Pathol 87: 369 –
580. Rich MM, Pinter MJ. Sodium channel inactivation in an animal model of acute quadri- 372, 2006.
plegic myopathy. Ann Neurol 50: 26 –33, 2001.
603. Said G, Goulon-Goeau C, Slama G, Tchobroutsky G. Severe early-onset polyneurop-
581. Rich MM, Pinter MJ. Crucial role of sodium channel fast inactivation in muscle fiber athy in insulin-dependent diabetes mellitus. A clinical and pathological study. N Engl J
inexcitability in a rat model of critical illness myopathy. J Physiol 547: 555–566, 2003. Med 326: 1257–1263, 1992.
582. Rich MM, Pinter MJ, Kraner SD, Barchi RL. Loss of electrical excitability in an animal 604. Sagalyn E, Band RA, Gaieski DF, Abella BS. Therapeutic hypothermia after cardiac
model of acute quadriplegic myopathy. Ann Neurol 43: 171–179, 1998. arrest in clinical practice: review and compilation of recent experiences. Crit Care Med
37: S223–S226, 2009.
583. Rich MM, Teener JW, Raps EC, Schotland DL, Bird SJ. Muscle is electrically inexcitable
in acute quadriplegic myopathy. Neurology 46: 731–736, 1996. 605. Sair M, Etherington PJ, Curzen NP, Winlove CP, Evans TW. Tissue oxygenation and
perfusion in endotoxemia. Am J Physiol Heart Circ Physiol 271: H1620 –H1625, 1996.
584. Rigby SL, Hofmann PA, Zhong J, Adams HR, Rubin LJ. Endotoxemia-induced myocar-
dial dysfunction is not associated with changes in myofilament Ca2⫹ responsiveness. 606. Sair M, Etherington PJ, Winlove P, Evans TW. Tissue oxygenation and perfusion in
Am J Physiol Heart Circ Physiol 274: H580 –H590, 1998. patients with systemic sepsis. Crit Care Med 29: 1343–1349, 2001.

585. Riley DA, Bain JL, Thompson JL, Fitts RH, Widrick JJ, Trappe SW, Trappe TA, Costill 607. Salter RG, Knowles M, Moncada S. Widespread tissue distribution, species distribu-
DL. Decreased thin filament density and length in human atrophic soleus muscle fibers tion and changes in activity of Ca2⫹-dependent and Ca2⫹-independent nitric oxide
after spaceflight. J Appl Physiol 88: 567–572, 2000. synthases. FEBS Lett 291: 145–149, 1991.

586. Rimer M. Neuregulins at the neuromuscular synapse: past, present, and future. J 608. Salviati L, Laverda AM, Zancan L, Fanin M, Angelini C, Meznaric-Petrusa M. Acute
Neurosci Res 85: 1827–1833, 2007. quadriplegic myopathy in a 17-month-old boy. J Child Neurol 15: 63– 66, 2000.

1104 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

609. Sambe A, Ungureanu-Longrois D, Danialou G, Lanone S, Benessiano J, Aubier M, 629. Shah AM, Spurgeon HA, Sollott S, Talo A, Lakatta EG. 8-Bromo cGMP reduces the
Boczkowski J. Role of nitric oxide on diaphragmatic contractile failure in Escherischia myofilament response to Ca2⫹ in intact cardiac myocytes. Circ Res 74: 970 –978,
coli endotoxemic rats. Comp Biochem Physiol A 119: 167–175, 1998. 1994.

610. Samuelsson J, Zackrisson H, Tokics L, Ljungman P, Lidbrink E, Pircher P, Larsson L. 630. Shao W, Yeretssian G, Doiron K, Hussain SN, Saleh M. The caspase-1 digestome
Acute quadriplegic myopathy following autologous peripheral blood stem cell trans- identifies the glycolysis pathway as a target during infection and septic shock. J Biol
plantation for breast cancer. Bone Marrow Transplant 23: 835– 837, 1999. Chem 282: 36321–36329, 2007.

611. Sander HW, Saadeh PB, Chandswang N, Greenbaum D, Chokroverty S. Diaphrag- 631. Sharshar T, Bastuji-Garin S, De JB, Stevens RD, Polito A, Maxime V, Rodriguez P, Cerf
matic denervation in intensive care unit patients. Electromyogr Clin Neurophysiol 39: C, Outin H, Touraine P, Laborde K. Hormonal status and ICU-acquired paresis in
3–5, 1999. critically ill patients. Intensive Care Med 36: 1318 –1326, 2010.

612. Sandri M. Involvement of autophagy in muscle atrophy. Am J Physiol Cell Physiol 298: 632. Sharshar T, Bastuji-Garin S, Stevens RD, Durand MC, Malissin I, Rodriguez P, Cerf C,
C1291–C1299, 2010. Outin H, De JB. Presence and severity of intensive care unit-acquired paresis at time
of awakening are associated with increased intensive care unit and hospital mortality.
613. Sayeed MM. Signaling mechanisms of altered cellular responses in trauma, burn and
Crit Care Med 37: 3047–3053, 2009.
sepsis. Arch Surg 135: 1432–1442, 2000.
633. Shehabi Y, Bellomo R, Mehta S, Riker R, Takala J. Intensive care sedation: the past,
614. Sayeed MM. Ion transport in circulatory and/or septic shock. Am J Physiol Regul Integr
present and the future. Crit Care 17: 322, 2013.
Comp Physiol 252: R809 –R821, 1987.
634. Shindoh C, Di Marco A, Nethery D, Supinski G. Effect of PEG-superoxide dismutase
615. Scarpulla RC. Transcriptional paradigms in mammalian mitochondrial biogenesis and
on the diaphragmatic response to endotoxin. Am Rev Respir Dis 145: 1350 –1354,
function. Physiol Rev 88: 611– 638, 2008.
1992.
616. Schakman O, Dehoux M, Bouchuari S, Delaere S, Lause P, Decroly N, Shoelson SE,
635. Shiono S, Fantini GA, Roberts JP, Chiao J, Shires GT. Assessment of the early cellular
Thissen JP. Role of IGFI, the TNF␣/NFkB pathway, in the induction of muscle atro-
membrane response to live Escherichia coli bacteremia. J Surg Res 46: 9 –15, 1989.
genes by acute inflammation. Am J Physiol Endocrinol Metab 303: E729 –E739, 2012.
636. Siegfried A, Berchtold S, Manncke B, Deuschle E, Reber J, Ott T, Weber M, Kalinke U,
617. Schakman O, Gilson H, Kalista S, Thissen JP. Mechanisms of muscle atrophy induced
Hofer MJ, Hatesuer B, Schughart K, Gailus-Durner V, Fuchs H, Hrabe de Angelis M,
by glucocorticoids. Horm Res 72 Suppl 1: 36 – 41, 2009.
Weber F, Hornef MW, Autenrieth IB, Bohn E. IFIT2 is an effector protein of type I
618. Schefold JC, Bierbrauer J, Weber-Carstens S. Intensive care unit-acquired weakness IFN-mediated amplification of lipopolysaccharide (LPS)-induced TNF-␣ secretion
(ICUAW) and muscle wasting in critically ill patients with severe sepsis and septic and LPS-induced endotoxin shock. J Immunol 191: 3913–3921, 2013.
shock. J Cachexia Sarcopenia Muscle 1: 147–157, 2010.
637. Sillen MJ, Speksnijder CM, Eterman RM, Janssen PP, Wagers SS, Wouters EF, Uszko-
619. Schubert W, Kontozis L, Sticker G, Schwan H, Haraldsen G, Jerusalem F. Immuno- Lencer NH, Spruit MA. Effects of neuromuscular electrical stimulation of muscles of
fluorescent evidence for presence of interleukin-1 in normal and diseased human ambulation in patients with chronic heart failure or COPD: a systematic review of the
skeletal muscle. Muscle Nerve 11: 890 – 892, 1988. English-language literature. Chest 136: 44 – 61, 2009.

620. Schumer W, Erve PR, Obernolte RP. Endotoxemic effect on cardiac and skeletal 638. Simonson SG, Welty-Wolf K, Huang YT, Griebel JA, Caplan MS, Fracica PJ, Piantadosi
muscle mitochondria. Surg Gynecol Obstet 133: 433– 436, 1971. CA. Altered mitochondrial redox responses in gram negative septic shock in primates.
Circ Shock 43: 34 – 43, 1994.
621. Schwarz J, Planck J, Briegel J, Straube A. Single-fiber electromyography, nerve con-
duction studies, and conventional electromyography in patients with critical-illness 639. Simpson LL. The effects of acute and chronic botulinum toxin treatment on receptor
polyneuropathy: evidence for a lesion of terminal motor axons. Muscle Nerve 20: number, receptor distribution and tissue sensitivity in rat diaphragm. J Pharmacol Exp
696 –701, 1997. Ther 200: 343–351, 1977.

622. Schweickert WD, Hall J. ICU-acquired weakness. Chest 131: 1541–1549, 2007. 640. Singer M, Brealey D. Mitochondrial dysfunction in sepsis. Biochem Soc Symp 66: 149 –
166, 1999.
623. Schweickert WD, Pohlman MC, Pohlman AS, Nigos C, Pawlik AJ, Esbrook CL, Spears
L, Miller M, Franczyk M, Deprizio D, Schmidt GA, Bowman A, Barr R, McCallister KE, 641. Sinha B, Koster D, Ruez R, Gonnord P, Bastiani M, Abankwa D, Stan RV, Butler-
Hall JB, Kress JP. Early physical and occupational therapy in mechanically ventilated, Browne G, Vedie B, Johannes L, Morone N, Parton RG, Raposo G, Sens P, Lamaze C,
critically ill patients: a randomised controlled trial. Lancet 373: 1874 –1882, 2009. Nassoy P. Cells respond to mechanical stress by rapid disassembly of caveolae. Cell
144: 402– 413, 2011.
624. Segredo V, Matthay MA, Sharma ML, Gruenke LD, Caldwell JE, Miller RD. Prolonged
neuromuscular blockade after long-term administration of vecuronium in two criti- 642. Sipila S, Narici M, Kjaer M, Pollanen E, Atkinson RA, Hansen M, Kovanen V. Sex
cally ill patients. Anesthesiology 72: 566 –570, 1990. hormones and skeletal muscle weakness. Biogerontology 14: 231–245, 2013.

625. Segredo V, Caldwell JE, Matthay MA, Sharma ML, Gruenke LD, Miller RD. Persistent 643. Sitwell LD, Weinshenker BG, Monpetit V, Reid D. Complete ophthalmoplegia as a
paralysis in critically ill patients after long-term administration of vecuronium. N Engl complication of acute corticosteroid- and pancuronium-associated myopathy. Neu-
J Med 327: 524 –528, 1992. rology 41: 921–922, 1991.
626. Sen CK, Khanna S, Resznick AZ, Roy S, Packer L. Glutathione regulation of tumor 644. Sjöström M, Neglén P, Fridén J, Eklöf B. Human skeletal muscle metabolism and
necrosis factor-␣-induced NF-␬B activation in skeletal muscle-derived L6 cells. morphology after temporary incomplete ischemia. Eur J Clin Invest 12: 69 –79, 1982.
Biochem Biophys Res Commun 237: 645– 649, 1997.
645. Skinner RA, Gibson RM, Rothwell NJ, Pinteaux E, Penny JI. Transport of interleukin-1
627. Senf SM, Dodd SL, McClung JM, Judge AR. Hsp70 overexpression inhibits NF-kappaB across cerebromicrovascular endothelial cells. Br J Pharmacol 156: 1115–1123, 2009.
and Foxo3a transcriptional activities and prevents skeletal muscle atrophy. FASEB J 22:
3836 –3845, 2008. 646. Smith IJ, Alamdari N, O’Neal P, Gonnella P, Aversa Z, Hasselgren PO. Sepsis increases
the expression and activity of the transcription factor Forkhead Box O 1 (FOXO1) in
628. Seok J, Warren HS, Cuenca AG, Mindrinos MN, Baker HV, Xu W, Richards DW, skeletal muscle by a glucocorticoid-dependent mechanism. Int J Biochem Cell Biol 42:
McDonald-Smith GP, Gao H, Hennessy L, Finnerty CC, Lopez CM, Honari S, Moore 701–711, 2010.
EE, Minei JP, Cuschieri J, Bankey PE, Johnson JL, Sperry J, Nathens AB, Billiar TR, West
MA, Jeschke MG, Klein MB, Gamelli RL, Gibran NS, Brownstein BH, Miller-Graziano 647. Smith K, Rennie MJ. The measurement of tissue protein turnover. Baillieres Clin
C, Calvano SE, Mason PH, Cobb JP, Rahme LG, Lowry SF, Maier RV, Moldawer LL, Endocrinol Metab 10: 469 – 495, 1996.
Herndon DN, Davis RW, Xiao W, Tompkins RG. Inflammation, and Host Response to
Injury, Large Scale Collaborative Research Program. Genomic responses in mouse 648. Solomon V, Baracos V, Sarraf P, Goldberg AL. Rates of ubiquitin conjugation increase
models poorly mimic human inflammatory diseases. Proc Natl Acad Sci USA 110: when muscles atrophy, largely through activation of the N-end rule pathway. Proc Natl
3507–3512, 2013. Acad Sci USA 95: 12602–12607, 1998.

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1105


FRIEDRICH ET AL.

649. Sonneville R, den Hertog HM, Derde S, Güiza F, Derese I, Van den Berghe G, 669. Sullivan JS, Kilpatrick L, Costarino AT Jr, Lee SC, Harris MC. Correlation of plasma
Vanhorebeek I. Increasing glucose load while maintaining normoglycemia does not cytokine elevations with mortality rate in children with sepsis. J Pediatr 120: 510 –515,
evoke neuronal damage in prolonged critically ill rabbits. Clin Nutr 32: 1077–1080, 1992.
2013.
670. Sultan SS. Paravertebral block can attenuate cytokine response when it replaces
650. Sonneville R, den Hertog HM, Güiza F, Gunst J, Derese I, Wouters PJ, Brouland JP, general anesthesia for cancer breast surgeries. Saudi J Anaesth 7: 373–377, 2013.
Polito A, Gray F, Chrétien F, Charlier P, Annane D, Sharshar T, Van den Berghe G,
Vanhorebeek I. Impact of hyperglycemia on neuropathological alterations during crit- 671. Sun YB, Brandmeier B, Irving M. Structural changes in troponin in response to Ca2⫹
ical illness. J Clin Endocrinol Metab 97: 2113–2123, 2012. and myosin binding to thin filaments during activation of skeletal muscle. Proc Natl
Acad Sci USA 103: 17771–17776, 2006.
651. Spangenburg EE. Changes in muscle mass with mechanical load: possible cellular
mechanisms. Appl Physiol Nutr Metab 34: 328 –335, 2009. 672. Supinski GS, Callahan LA. Hemin prevents cardiac and diaphragm mitochondrial dys-
function in sepsis. Free Rad Biol Med 40: 127–137, 2006.
652. Spangenburg EE, Brown DA, Johnson MS, Moore RL. Alterations in peroxisome
proliferator-activated receptor mRNA expression in skeletal muscle after acute and 673. Supinski GS, Callahan LA. Free radical-mediated skeletal muscle dysfunction in inflam-
repeated bouts of exercise. Mol Cell Biochem 332: 225–231, 2009. matory conditions. J Appl Physiol 102: 2056 –2063, 2007.

653. Spitzer AR, Giancarlo T, Maher L, Awerbuch G, Bowles A. Neuromuscular causes of 674. Supinski GS, Callahan LA. The JNK MAP kinase pathway contributes to the develop-
prolonged ventilator dependency. Muscle Nerve 15: 682– 686, 1992. ment of endotoxin-induced diaphragm caspase activation. Am J Physiol Regul Integr
Comp Physiol 297: R825–R834, 2009.
654. Sprung CL, Annane D, Keh D, Moreno R, Singer M, Freivogel K, Weiss YG, Benben-
ishty J, Kalenka A, Forst H, Laterre PF, Reinhart K, Cuthbertson BH, Payen D, Briegel 675. Supinski GS, Callahan LA. Calpain activation contributes to endotoxin-induced dia-
J. Hydrocortisone therapy for patients with septic shock. N Engl J Med 358: 111–124, phragm dysfunction. Am J Respir Cell Mol Biol 42: 80 – 87, 2010.
2008.
676. Supinski GS, Callahan LA. Double-stranded RNA-dependent protein kinase activation
655. Sta˚lberg E, Ekstedt J, Broman A. Neuromuscular transmission in myasthenia gravis modulates endotoxin-induced diaphragm weakness. J Appl Physiol 110: 199 –205,
studied with single fibre electromyography. J Neurol Neurosurg Psychiatry 37: 540 – 2011.
547, 1974.
677. Supinski GS, Callahan LA. Diaphragm weakness in mechanically ventilated critically ill
656. Stasko SA, Hardin BJ, Smith JD, Moylan JS, Reid MB. TNF signals via neuronal-type
patients. Crit Care 17: R120, 2013.
nitric oxide synthase and reactive oxygen species to depress specific force of skeletal
muscle. J Appl Physiol 114: 1629 –1636, 2013. 678. Supinski GS, Vanags J, Callahan LA. Effect of proteasome inhibitors on endotoxin-
induced diaphragm dysfunction. Am J Physiol Lung Cell Mol Physiol 296: L994 –L1001,
657. Steele DS, Duke AM. Metabolic factors contributing to altered Ca2⫹ regulation in
2009.
skeletal muscle fatigue. Acta Physiol Scand 179: 39 – 48, 2003.
679. Supinski GS, Ji X, Wang W, Callahan LA. The extrinsic caspase pathway modulates
658. Steinberg KP, Hudson LD, Goodman RB, Hough CL, Lanken PN, Hyzy R, Thompson
endotoxin-induced diaphragm contractile dysfunction. J Appl Physiol 102: 1649 –1657,
BT, Ancukiewicz M. Efficacy and safety of corticosteroids for persistent acute respi-
2007.
ratory distress syndrome. N Engl J Med 354: 1671–1684, 2006.

659. Stengl M, Bartak F, Sykora R, Chvojka J, Benes J, Krouzecky A, Novak I, Sviglerova J, 680. Supinski GS, Ji XY, Callahan LA. p38 mitogen-activated protein kinase modulates
Kuncova J, Matejovic M. Reduced L-type calcium current in ventricular myocytes from endotoxin-induced diaphragm caspase activation. Am J Respir Cell Mol Biol 43: 121–
pigs with hyperdynamic septic shock. Crit Care Med 38: 579 –587, 2010. 127, 2010.

660. Steuerwald NM, Foureau DM, Norton HJ, Zhou J, Parsons JC, Chalasani N, Fontana 681. Supinski GS, Nethery D, DiMarco A. Effect of free radical scavengers on endotoxin-
RJ, Watkins PB, Lee WM, Reddy KR, Stolz A, Talwalkar J, Davern T, Saha D, Bell LN, induced respiratory muscle dysfunction. Am Rev Respir Dis 148: 1318 –1324, 1993.
Barnhart H, Gu J, Serrano J, Bonkovsky HL. Profiles of serum cytokines in acute
682. Supinski GS, Nethery D, Nosek TM, Callahan LA, Stofan D, Di Marco A. Endotoxin
drug-induced liver injury and their prognostic significance. PLoS One 8: e81974, 2013.
administration alters the force vs pCa relationship of skeletal muscle fibers. Am J
661. Stevens RD, Marshall SA, Cornblath DR, Hoke A, Needham DM, De JB, Ali NA, Physiol Regul Integr Comp Physiol 278: R891–R896, 2000.
Sharshar T. A framework for diagnosing and classifying intensive care unit-acquired
weakness. Crit Care Med 37: S299 –S308, 2009. 683. Supinski GS, Nethery D, Stofan D, DiMarco A. Comparison of the effects of endo-
toxin on limb, respiratory, and cardiac muscles. J Appl Physiol 81: 1370 –1378, 1996.
662. Stibler H, Edstrom L, Ahlbeck K, Remahl S, Ansved T. Electrophoretic determination
of the myosin/actin ratio in the diagnosis of critical illness myopathy. Intensive Care 684. Supinski G, Stofan D, Nethery D, Szweda L, Di Marco A. Apocynin improves dia-
Med 29: 1515–1527, 2003. phragmatic function after endotoxin administration. J Appl Physiol 87: 776 –782, 1999.

663. Stinebring WR, Youngner JS. Patterns of interferon appearance in mice infected with 685. Supinski GS, Wang W, Callahan LA. Caspase and calpain activation both contribute to
bacteria or bacterial endotoxin. Nature 204: 712, 1964. sepsis-induced diaphragmatic weakness. J Appl Physiol 107: 1389 –1396, 2009.

664. Strasser EM, Stattner S, Karner J, Klimpfinger M, Freynhofer M, Zaller V, Graf A, 686. Suzuki N, Mizuno H, Warita H, Takeda S, Itoyama Y, Aoki M. Neuronal NOS is
Wessner B, Bachl N, Roth E, Quittan M. Neuromuscular electrical stimulation re- dislocated during muscle atrophy in amyotrophic lateral sclerosis. J Neurol Sci 294:
duces skeletal muscle protein degradation and stimulates insulin-like growth factors in 95–101, 2010.
an age- and current-dependent manner: a randomized, controlled clinical trial in
major abdominal surgical patients. Ann Surg 249: 738 –743, 2009. 687. Suzuki N, Motohashi N, Uezumi A, Fukada S, Yoshimura T, Itoyama Y, Aoki M,
Miyagoe-Suzuki Y, Takeda S. NO production results in suspension-induced muscle
665. Strassmann G, Fong M, Kenney JS, Jacob CO. Evidence for the involvement of inter- atrophy through dislocation of neuronal NOS. J Clin Invest 117: 2468 –2476, 2007.
leukin 6 in experimental cancer cachexia. J Clin Invest 89: 1681–1684, 1992.
688. Takala J, Ruokonen E, Webster NR, Nielsen MS, Zandstra DF, Vundelinckx G, Hinds
666. Suffredini AF, Reda D, Banks SM, Tropea M, Agosti JM, Miller R. Effects of recombi- CJ. Increased mortality associated with growth hormone treatment in critically ill
nant TNF receptor on human inflammatory responses following intravenous endo- adults. N Engl J Med 341: 785–792, 1999.
toxin administration. J Immunol 155: 5038 –5045, 1995.
689. Tam BT, Siu PM. Autophagic cellular responses to physical exercise in skeletal muscle.
667. Suliman HB, Carraway MS, Welty-Wolf KE, Whorton AR, Piantadosi CA. Lipopoly- Sports Med 44: 625– 640, 2014.
saccharide stimulates mitochondrial biogenesis via activation of nuclear respiratory
factor-1. J Biol Chem 278: 41510 – 41518, 2003. 690. Tang H, Lee M, Khuong A, Wright E, Shrager JB. Diaphragm muscle atrophy in the
mouse after long-term mechanical ventilation. Muscle Nerve 48: 272–278, 2013.
668. Suliman IA, Lindgren JU, Gillberg PG, Diab KM, Adem A. Effect of immobilization on
skeletal muscle nicotinic cholinergic receptors in the rat. NeuroReport 8: 2821–2824, 691. Tavakoli H, Mela L. Alterations of mitochondrial metabolism and protein concentra-
1997. tions in subacute septicemia. Infect Immun 38: 536 –541, 1982.

1106 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

692. Tavernier B, Garrique D, Boulle C, Vallet B, Adnet P. Myofilament calcium sensitivity 713. Trojaborg W. Electrophysiologic techniques in critical illness-associated weakness. J
is decreased in skinned cardiac fibers of endotoxin-treated rabbits. Cardiovasc Res 38: Neurol Sci 242: 83– 85, 2006.
472– 479, 1998.
714. Trojaborg W, Weimer LH, Hays AP. Electrophysiologic studies in critical illness asso-
693. Tavernier B, Mebazaa A, Mateo P, Sys S, Ventura-Clapier R, Veksler V. Phosphoryla- ciated weakness: myopathy or neuropathy–a reappraisal. Clin Neurophysiol 112:
tion-dependent alteration in myofilament Ca2⫹ sensitivity but normal mitochondrial 1586 –1593, 2001.
function in septic heart. Am J Respir Crit Care Med 163: 362–367, 2001.
715. Trout JJ, Stauber WT, Schottelius BA. Degeneration and regeneration in denervated
694. Taylor BE, Buchman TG. Is there a role for growth hormone therapy in refractory tonic and phasic skeletal muscle: morphology and acid phosphatase cytochemistry.
critical illness? Curr Opin Crit Care 14: 438 – 444, 2008. Virchows Arch B Cell Pathol Incl Mol Pathol 38: 67–76, 1981.

695. Tedders KM, McNorton KN, Edwin SB. Efficacy and safety of analgosedation with 716. Trumbeckaite S, Opalka JR, Neuhof C, Zierz S, Gellerich FN. Different sensitivity of
fentanyl compared with traditional sedation with propofol. Pharmacotherapy 34: 643– rabbit heart and skeletal muscle to endotoxin-induced impairment of mitochondrial
647, 2014. function. Eur J Biochem 268: 1422–1429, 2001.

696. Teener JW, Rich MM. Dysregulation of sodium channel gating in critical illness myop- 717. Trunkey DD, Illner H, Wagner Y, Shires GT. The effect of septic shock on skeletal
athy. J Muscle Res Cell Motil 27: 291–296, 2006. muscle action potentials in the primate. Surgery 85: 638 – 643, 1979.

697. Teichmann MD, Wegner FV, Fink RH, Chamberlain JS, Launikonis BS, Martinac B, 718. Tsujinaka T, Ebisui C, Fujita J, Kishibuchi M, Morimoto T, Ogawa A, Katsume A,
Friedrich O. Inhibitory control over Ca2⫹ sparks via mechanosensitive channels is Ohsugi Y, Kominami E, Monden M. Muscle undergoes atrophy in association with
disrupted in dystrophin deficient muscle but restored by mini-dystrophin expression. increase of lysosomal cathepsin activity in interleukin-6 transgenic mouse. Biochem
Biophys Res Commun 207: 168 –174, 1995.
PLoS One 3: e3644, 2008.
719. Tsujinaka T, Fujita J, Ebisui C, Yano M, Kominami E, Suzuki K, Tanaka K, Katsume A,
698. Tennila A, Salmi T, Pettila V, Roine RO, Varpula T, Takkunen O. Early signs of critical
Ohsugi Y, Shiozaki H, Monden M. Interleukin 6 receptor antibody inhibits muscle
illness polyneuropathy in ICU patients with systemic inflammatory response syn-
atrophy and modulates proteolytic systems in interleukin 6 transgenic mice. J Clin
drome or sepsis. Intensive Care Med 26: 1360 –1363, 2000.
Invest 97: 244 –249, 1996.
699. Tepper M, Rakic S, Haas JA, Woittiez AJ. Incidence and onset of critical illness poly-
720. Tsukagoshi H, Morita T, Takahashi K, Kunimoto F, Goto F. Cecal ligation and punc-
neuropathy in patients with septic shock. Neth J Med 56: 211–214, 2000.
ture peritonitis model shows decreased nicotinic acetylcholine receptor numbers in
700. Terman A, Kurz T, Navratil M, Arriaga EA, Brunk UT. Mitochondrial turnover and rat muscle. Anesthesiology 891: 448 – 460, 1999.
aging of long-lived postmitotic cells: the mitochondrial-lysosomal axis theory of aging.
721. Twig G, Elorza A, Molina AJA, Mohamed H, Wikstrom JD, Walzer G, Stiles L, Haigh
Antioxid Redox Signal 12: 503–535, 2010.
SE, Katz S, Las G, Alroy J, Wu M, Py BF, Yuan J, Deeney JT, Corkey BE, Shirihai OS.
Fusion and fission govern mitochondrial segregation and elimination by autophagy.
701. Tews DS, Goebel HH. Cytokine expression profile in idiopathic inflammatory myop-
EMBO J 27: 433– 446, 2008.
athies. J Neuropathol Exp Neurol 55: 342–347, 1996.
722. Tzanis G, Vasileiadis I, Zervakis D, Karatzanos E, Dimopoulos S, Pitsolis T, Tripodaki
702. Thompson M, Kliewer A, Maass D, Becker L, White DJ, Bryant D, Arteaga G, Horton
E, Gerovasili V, Routsi C, Nanas S. Maximum inspiratory pressure, a surrogate pa-
J, Giroir BP. Increased cardiomyocyte intracellular calcium during endotoxin-induced
rameter for the assessment of ICU-acquired weakness. BMC Anesthesiol 11: 14, 2011.
cardiac dysfunction in guinea pigs. Pediatr Res 47: 669 – 676, 2000.
723. Tzika AA, Mintzopoulos D, Mindrinos M, Zhang J, Rahme LG, Tompkins RG. Microar-
703. Tiao G, Fagan J, Roegner V, Lieberman M, Wang JJ, Fischer JE, Hasselgren PO.
ray analysis suggests that burn injury results in mitochondrial dysfunction in human
Energy-ubiquitin-dependent muscle proteolysis during sepsis in rats is regulated by
skeletal muscle. Int J Mol Med 24: 387–392, 2009.
glucocorticoids. J Clin Invest 97: 339 –348, 1996.
724. Valdes JA, Gaggero E, Hidalgo J, Leal N, Jaimovich E, Carrasco MA. NFAT activation
704. Tiao G, Fagan JM, Samuels N, James JH, Hudson K, Lieberman M, Fischer JE, Hassel-
by membrane potential follows a calcium pathway distinct from other activity-related
gren PO. Sepsis stimulates nonlysosomal, energy-dependent proteolysis and in- transcription factors in skeletal muscle cells. Am J Physiol Cell Physiol 294: C715–C725,
creases ubiquitin mRNA levels in rat skeletal muscle. J Clin Invest 94: 2255–2264, 2008.
1994.
725. Vanhorebeek I, De Vos R, Mesotten D, Wouters PJ, De Wolf-Peeters C, Van den
705. Tiao G, Lieberman M, Fischer JE, Hasselgren PO. Intracellular regulation of protein Berghe G. Protection of hepatocyte mitochondrial ultrastructure and function by
degradation during sepsis is different in fast- and slow-twitch muscle. Am J Physiol Regul strict blood glucose control with insulin in critically ill patients. Lancet 365: 53–59,
Integr Comp Physiol 272: R849 –R856, 1997. 2005.

706. Tilg H, Dinarello CA, Mier JW. IL-6 and APPs: anti-inflammatory and immunosuppres- 726. Vanhorebeek I, Ellger B, De Vos R, Boussemaere M, Debaveye Y, Vander Perre S,
sive mediators. Immunol Today 18: 428 – 432, 1997. Rabbani N, Thornalley PJ, Van den Berghe G. Tissue-specific glucose toxicity induces
mitochondrial damage in a burn injury model of critical illness. Crit Care Med 37:
707. Tjader I, Essen P, Garlick PG, McMnurlan MA, Rooyackers O, Wernerman J. Impact of
1355–1364, 2009.
surgical trauma on human skeletal muscle protein synthesis. Clin Sci 107: 601– 607,
2004. 727. Vanhorebeek I, Gunst J, Derde S, Derese I, Boussemaere M, D’Hoore A, Wouters PJ,
Van den Berghe G. Mitochondrial fusion, fission, and biogenesis in prolonged critically
708. Tömböl T, Pataki G, Németh A, Hamar J. Ultrastructural changes of the neuromus- ill patients. J Clin Endocrinol Metab 97: E59 –E64, 2012.
cular junction in reperfusion injury. Cells Tissues Organs 170: 139 –150, 2002.
728. Vanhorebeek I, Gunst J, Derde S, Derese I, Boussemaere M, Guiza F, Martinet W,
709. Tokumaru T. The effect of trauma on production of herpes interferon in guinea pigs. Timmermans JP, D’Hoore A, Wouters PJ, Van den Berghe G. Insufficient activation of
Arch Gesamte Virusforsch 21: 61–70, 1967. autophagy allows cellular damage to accumulate in critically ill patients. J Clin Endocri-
nol Metab 96: E633–E645, 2011.
710. Tolosa L, Morla M, Iglesias A, Busquets X, Llado J, Olmos G. INF-gamma prevents
TNF-alpha-induced apoptosis in C2C12 myotubes through down-regulation of 729. Vanhorebeek I, Gunst J, Ellger B, Boussemaere M, Lerut E, Debaveye Y, Rabbani N,
TFN-R2 and increased NF-kappaB activity. Cell Signal 17: 1333–1342, 2005. Thornalley PJ, Schetz M, Van den Berghe G. Hyperglycemic kidney damage in an
animal model of prolonged critical illness. Kidney Int 76: 512–520, 2009.
711. Tomera JF, Martyn J. Intraperitoneal endotoxin but not protein malnutrition shifts
d-tubocurarine dose-repsonse curves in mouse gastrocnemius muscle. J Pharmacol 730. Vanhorebeek I, Langouche L, Van den Berghe G. Endocrine aspects of acute and
Exp Ther 250: 216 –220, 1989. prolonged critical illness. Nat Clin Pract Endocrinol Metab 2: 20 –31, 2006.

712. Tracey KJ, Lowry SF, Beutler B, Cerami A, Albert JD, Shires GT. Cachectin/tumor 731. Vanhorebeek I, Langouche L. Molecular mechanisms behind clinical benefits of inten-
necrosis factor mediates changes of skeletal muscle plasma membrane potential. J Exp sive insulin therapy during critical illness: glucose versus insulin. Best Pract Res Clin
Med 164: 1368 –1373, 1986. Anaesthesiol 23: 449 – 459, 2009.

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1107


FRIEDRICH ET AL.

732. Vanhorebeek I, Langouche L, Van den Berghe G. Tight blood glucose control with 754. Wang N, Tytell JD, Ingber DE. Mechanotransduction at a distance: mechanically
insulin in the ICU: facts and controversies. Chest 132: 268 –278, 2007. coupling the extracellular matrix with the nucleus. Nat Rev Mol Cell Biol 10: 75– 82,
2009.
733. Van den Berghe G. Intensive insulin therapy in the ICU–reconciling the evidence. Nat
Rev Endocrinol 8: 374 –378, 2012. 755. Wang P, Zhou M, Cioffi WG, Bland KI, Ba ZF, Chaudry IH. Is prostacyclin responsible
for producing the hyperdynamic response during early sepsis? Crit Care Med 28:
734. Van den Berghe G, Schetz M, Vlasselaers D, Hermans G, Wilmer A, Bouillon R, 1534 –1539, 2000.
Mesotten D. Clinical review: intensive insulin therapy in critically ill patients: NICE-
SUGAR or Leuven blood glucose target? J Clin Endocrinol Metab 94: 3163–3170, 2009. 756. Wang X, Blagden C, Fan J, Nowak SJ, Taniuchi I, Littman DR, Burden SJ. Runx1
prevents wasting, myofibrillar disorganization, and autophagy of skeletal muscle.
735. Van den Berghe G, Schoonheydt K, Becx P, Bruyninckx F, Wouters PJ. Insulin therapy Genes Dev 19: 1715–1722, 2005.
protects the central and peripheral nervous system of intensive care patients. Neu-
rology 64: 1348 –1353, 2005. 757. Wang X, Li Y, Engisch KL, Nakanishi ST, Dodson SE, Miller GW, Cope TC, Pinter MJ,
Rich MM. Activity-dependent presynaptic regulation of quantal size at the mammalian
736. Van den Berghe G, Wilmer A, Hermans G, Meersseman W, Wouters PJ, Milants I, Van neuromuscular junction in vivo. J Neurosci 25: 343–351, 2005.
Wijngaerden E, Bobbaers H, Bouillon R. Intensive insulin therapy in the medical ICU.
N Engl J Med 354: 449 – 461, 2006. 758. Ward JM, Martyn JAJ. Burn injury-induced nicotinic acetylcholine receptor changes on
muscle membrane. Muscle Nerve 16: 348 –354, 1993.
737. Van den Berghe G, Wilmer A, Milants I, Wouters PJ, Bouckaert B, Bruyninckx F,
Bouillon R, Schetz M. Intensive insulin therapy in mixed medical/surgical intensive care 759. Warner BW, Hasselgren PO, James JH, Hummel RP 3rd, Rigel DF, Fischer JE. Re-
units: benefit versus harm. Diabetes 55: 3151–3159, 2006. duced amino acid transport in skeletal muscle caused by a circulating factor during
endotoxemia. Ann Surg 211: 323–328, 1990.
738. Van den Berghe G, Wouters P, Weekers F, Verwaest C, Bruyninckx F, Schetz M,
Vlasselaers D, Ferdinande P, Lauwers P, Bouillon R. Intensive insulin therapy in the 760. Watson AC, Hughes PD, Louise HM, Hart N, Ware RJ, Wendon J, Green M, Moxham
critically ill patients. N Engl J Med 345: 1359 –1367, 2001. J. Measurement of twitch transdiaphragmatic, esophageal, and endotracheal tube
pressure with bilateral anterolateral magnetic phrenic nerve stimulation in patients in
739. Van Dervort AL, Yan L, Madara PJ, Cobb JP, Wesley RA, Corriveau CC, Tropea MM, the intensive care unit. Crit Care Med 29: 1325–1331, 2001.
Danner RL. Nitric oxide regulates endotoxin-induced TNF-alpha production in hu-
man neutrophils. J Immunol 152: 4102– 4109, 1994. 761. Watson CL, Gold MR. Modulation of Na⫹ current inactivation by stimulation of
protein kinase C in cardiac cells. Circ Res 81: 380 –386, 1997.
740. Van Hees HWH, Schellekens WJM, Linkels M, Leenders F, Zoll J, Donders R, Dekhui-
jzen PNR, van der Hoeven J, Heunks LMA. Plasma from septic shock patients induces 762. Watters JM, Kirkpatrick SM, Norris SB, Shamji FM, Wells GA. Immediate postoper-
loss of muscle proteins. Crit Care 15: R233, 2011. ative enteral feeding results in impaired respiratory mechanics and decreased mobil-
ity. Ann Surg 226: 369 –377, 1997.
741. Van Kann LN, Bakker AJ. Effect of tumor necrosis factor ␣ on electrically induced
calcium transients elicited in C2C12 skeletal myotubes. Muscle Nerve 35: 251–253, 763. Weber-Carstens S, Deja M, Koch S, Spranger J, Bubser F, Wernecke KD, Spies CD,
2007. Spuler S, Keh D. Risk factors in critical illness myopathy during the early course of
critical illness: a prospective observational study. Crit Care 14: R119, 2010.
742. Van Mook WN, Hulsewe-Evers RP. Critical illness polyneuropathy. Curr Opin Crit Care
8: 302–310, 2002. 764. Weber-Carstens S, Koch S, Spuler S, Spies CD, Bubser F, Wernecke KD, Deja M.
Nonexcitable muscle membrane predicts intensive care unit-acquired paresis in me-
743. Vega GL, Baxter CR. Tumor necrosis factor mediates hypertriglyceridemia during chanically ventilated, sedated patients. Crit Care Med 37: 2632–2637, 2009.
thermal injury in mice genetically susceptible to lipopolysaccharides. J Burn Care Re-
habil 12: -4637, 1991. 765. Weber-Carstens S, Schneider J, Wollersheim T, Assmann A, Bierbrauer J, Marg A, Al
Hasani H, Chadt A, Wenzel K, Koch S, Fielitz J, Kleber C, Faust K, Mai K, Spies CD,
744. Vender JS, Szokol JW, Murphy GS, Nitsun M. Sedation, analgesia, and neuromuscular Luft FC, Boschmann M, Spranger J, Spuler S. Critical illness myopathy and GLUT4:
blockade in sepsis: an evidence-based review. Crit Care Med 32: S554 –S561, 2004. significance of insulin and muscle contraction. Am J Respir Crit Care Med 187: 387–396,
2013.
745. Vincent JL. The rise and fall of drotrecogin alfa (activated). Lancet Infect Dis 12:
649 – 651, 2012. 766. Weekers F, Van Herck E, Coopmans W, Michalaki M, Bowers CY, Veldhuis JD, Van
den Berghe G. A novel in vivo rabbit model of hypercatabolic critical illness reveals a
746. Visser J, Labadarios D, Blaauw R. Micronutrient supplementation for critically ill biphasic neuroendocrine stress response. Endocrinology 143: 764 –774, 2002.
adults: a systematic review and meta-analysis. Nutrition 27: 745–758, 2011.
767. Wei W, Fareed MU, Evenson A, Menconi MJ, Yang H, Petkova V, Hasselgren PO.
747. Visser LH. Critical illness polyneuropathy and myopathy: clinical features, risk factors Sepsis stimulates calpain activity in skeletal muscle by decreasing calpastatin activity
and prognosis. Eur J Neurol 13: 1203–1212, 2006. but does not activate caspase-3. Am J Physiol Regul Integr Comp Physiol 288: R580 –
R590, 2005.
748. Vlasselaers D, Milants I, Desmet L, Wouters PJ, Vanhorebeek I, van den Heuvel I,
Mesotten D, Casaer MP, Meyfroidt G, Ingels C, Muller J, Van Cromphaut S, Schetz M, 768. Weiss-Guillet EM, Takala J, Jakob SM. Diagnosis and management of electrolyte emer-
Van den Berghe G. Intensive insulin therapy for patients in paediatric intensive care: a gencies. Best Pract Res Clin Endocrinol Metab 17: 623– 651, 2003.
prospective, randomised controlled study. Lancet 373: 547–556, 2009.
769. Welty-Wolf KE, Simonson SG, Huang YC, Fracica PJ, Patterson JW, Piantadosi CA.
749. Voisin L, Breuille D, Combaret L, Pouyet C, Taillandier D, Aurousseau E, Obled C, Ultrastructural changes in skeletal muscle mitochondria in gram-negative sepsis.
Attaix D. Muscle wasting in a rat model of long-lasting sepsis results from the activa- Shock 5: 378 –384, 1996.
tion of lysosomal, Ca2⫹-activated, and ubiquitin-proteasome proteolytic pathways. J
Clin Invest 97: 1610 –1617, 1996. 770. Wernerman J. Glutamine supplementation. Ann Intensive Care 1: 25, 2011.

750. Wagatsuma A, Kotake N, Mabuchi K, Yamada S. Expression of nuclear-encoded genes 771. Westfall MV, Sayeed MM. Skeletal muscle calcium uptake in bacteremic rats. Am J
involved in mitochondrial biogenesis and dynamics in experimentally denervated mus- Physiol Regul Integr Comp Physiol 256: R201–R206, 1989.
cle. J Physiol Biochem 67: 359 –370, 2011.
772. Wichterman KA, Baue AE, Chaudry IH. Sepsis and septic shock: a review of laboratory
751. Wagatsuma A, Kotake N, Yamada S. Muscle regeneration occurs to coincide with models and a proposal. J Surg Res 29: 189 –201, 1980.
mitochondrial biogenesis. Mol Cell Biochem 349: 139 –147, 2011.
773. Wilcox P, Miliken C, Bressler B. High-dose tumor necrosis factor alpha produces an
752. Waldhausen E, Mingers B, Lippers P, Keser G. Critical illness polyneuropathy due to impairment of hamster diaphragm contractility. Attenuation with a prostaglandin in-
parenteral nutrition. Intensive Care Med 23: 922–923, 1997. hibitor. Am J Respir Crit Care Med 153: 1611–1615, 1996.

753. Wang DW, George AL Jr, Bennett PB. Comparison of heterologously expressed 774. Wilcox P, Osborne S, Bressler B. Monocyte inflammatory mediators impair in vitro
human cardiac and skeletal muscle sodium channels. Biophys J 70: 238 –245, 1996. hamster diaphragm contractility. Am Rev Resp Dis 146: 462– 466, 1992.

1108 Physiol Rev • VOL 95 • JULY 2015 • www.prv.org


NEUROMUSCULAR FAILURE IN THE CRITICALLY ILL

775. Williams A, Wang JJ, Wang L, Sun X, Fischer JE, Hasselgren PO. Sepsis in mice 794. Zamir O, Hasselgren PO, James H, Higashiguchi T, Fischer JE. Effect of tumor necro-
stimulates muscle proteolysis in the absence of IL-6. Am J Physiol Regul Integr Comp sis factor or interleukin-1 on muscle amino acid uptake and the role of glucocorticoids.
Physiol 275: R1983–R1991, 1998. Surg Gynecol Obstet 177: 27–32, 1993.

776. Williams AB, deCourten-Myers GM, Fischer JE, Luo G, Sun X, Hasselgren PO. Sepsis 795. Zamir O, Hasselgren PO, von Allmen D, Fischer JE. The effect of interleukin-1 alpha
stimulates release of myofilaments in skeletal muscle by a calcium-dependent mech- and the glucocorticoid receptor blocker RU38486 on total and myofibrillar break-
anism. FASEB J 13: 1435–1443, 1999. down in skeletal muscle. J Surg Res 50: 579 –583, 1991.

777. Williams DA, Head SI, Bakker AJ, Stephenson DG. Resting calcium concentrations in 796. Zanni JM, Korupolu R, Fan E, Pradhan P, Janjua K, Palmer JB, Brower RG, Needham
isolated skeletal muscle fibers of dystrophic mice. J Physiol 428: 243–256, 1990. DM. Rehabilitation therapy and outcomes in acute respiratory failure: an observa-
tional pilot project. J Crit Care 25: 254 –262, 2010.
778. Williams G, Brown T, Becker L, Prager M, Giroir BP. Cytokine-induced expression of
nitric oxide synthase in C2C12 skeletal muscle myocytes. Am J Physiol Regul Integr 797. Zanotti E, Felicetti G, Maini M, Fracchia C. Peripheral muscle strength training in
Comp Physiol 267: R1020 –R1025, 1994. bed-bound patients with COPD receiving mechanical ventilation: effect of electrical
stimulation. Chest 124: 292–296, 2003.
779. Williams S, Horrocks IA, Ouvrier RA, Gillis J, Ryan MM. Critical illness polyneuropathy
and myopathy in prediatric intensive care: a review. Pediatr Crit Care Med 8: 18 –22, 798. Zemkova H, Teisinger J, Almon RR, Veisada R, Hnik P, Vyskocil F. Immobilization
2007. atrophy and membrane properties in rat skeletal muscle fibres. Pflügers Arch 416:
126 –129, 1990.
780. Winkelmann C. Inactivity and inflammation. Selected cytokines as biologic mediators
in muscle dysfunction during critical illness. AACN Clin Issues 15: 74 – 82, 2004. 799. Zeng J, Lin X, Fan H, Li C. Hydrogen sulfide attenuates the inflammatory response in
a mouse burn injury model. Mol Med Rep 8: 1204 –1208, 2013.
781. Witt NJ, Zochodne DW, Bolton CF, Grand’Maison F, Wells G, Young GB, Sibbald WJ.
Peripheral nerve function in sepsis and multiple organ failure. Chest 99: 176 –184, 800. Z’Graggen WJ, Lin CS, Howard RS, Beale RJ, Bostock H. Nerve excitability changes in
1991. critical illness polyneuropathy. Brain 129: 2461–2470, 2006.

782. Witzemann V, Brenner HR, Sakmann B. Neural factors regulate AChR subunit mR- 801. Z’Graggen WJ, Brander L, Tuchscherer D, Scheidegger O, Takala J, Bostock H.
NAs at rat neuromuscular synapses. J Cell Biol 114: 125–141, 1991. Muscle membrane dysfunction in critical illness myopathy assessed by velocity recov-
ery cycles. Clin Neurophysiol 122: 834 – 841, 2011.
783. Wolf SE, Edelman LS, Kemalyan N, Donison L, Cross J, Underwood M, Spence RJ,
Noppenberger D, Palmieri TL, Greenhalgh DG, Lawless M, Voigt D, Edwards P, 802. Zhang J, Jiang W, Zuo Z. Pyrrolidine dithiocarbamate attenuates surgery-induced
Warner P, Kagan R, Hatfield S, Jeng J, Crean D, Hunt J, Purdue G, Burris A, Cairns B, neuroinflammation and cognitive dysfunction possibly via inhibition of nuclear factor
Kessler M, Klein RL, Baker R, Yowler C, Tutulo W, Foster K, Caruso D, Hildebrand B, ␬B. Neuroscience S0306-4522: 01057-01059, 2013.
Benjamin W, Villarreal C, Sanford AP, Saffle J. Effects of oxandrolone on outcome
803. Zhang Q, Cao H, Astrakas LG, Mintzopoulos D, Mindrinos MN, Schulz IIIJ, Tompkins
measures in the severely burned: a multicenter prospective randomized double-blind
RG, Rahme LG, Tzika AA. Uncoupling protein 3 expression and intramyocellular lipid
trial. J Burn Care Res 27: 131–139, 2006.
accumulation by NMR following local burn trauma. Int J Mol Med 18: 1223–1229,
2006.
784. Wolfe RR, Martini WZ. Changes in intermediary metabolism in severe surgical illness.
World J Surg 24: 639 – 647, 2000. 804. Zhang Y, Hartmann HA, Satin J. Glycosylation influences voltage-dependent gating
of cardiac and skeletal muscle sodium channels. J Membr Biol 171: 195–207, 1999.
785. Wondergem R, Graves BM, Li C, Williams DL. Lipopolysaccharide prolongs action
potential duration in HL-1 mouse cardiomyocytes. Am J Physiol Cell Physiol 303: C825– 805. Zhao J, Brault JJ, Schild A, Cao P, Sandri M, Schiaffino S, Lecker SH, Goldberg AL.
C833, 2012. FoxO3 coordinately activates protein degradation by the autophagic/lysosomal
and proteasomal pathways in atrophying muscle cells. Cell Metab 6: 472– 483,
786. Wray CJ, Mammen JM, Hershko DD, Hasselgren PO. Sepsis upregulates the gene
2007.
expression of multiple ubiquitin ligases in skeletal muscle. Int J Biochem Cell Biol 35:
698 –705, 2003. 806. Zhao X, Min CK, Ko JK, Parness J, Kim do H, Weisleder N, Ma J. Increased store-
operated Ca2⫹ entry in skeletal muscle with reduced calsequestrin-1 expression.
787. Wu LL, Liu MS. Altered ryanodine receptor of canine cardiac sarcoplasmic reticulum
Biophys J 99: 1556 –1564, 2010.
and its underlying mechanism in endotoxin shock. J Surg Res 53: 82–90, 1992.
807. Zhu X, Bernecker OY, Haijar RJ, Hellman J, Ichinose F, Valdivia HH, Schmidt U.
788. Yang B, Jiang JH, Zhou YC, Zhang Y, Li ST. Denervation stage differentially influences
Increased leakage of sarcoplasmic reticulum Ca2⫹ contributes to abnormal myocyte
reistance to neuromuscular blockers in rat gastrocnemius. J Surg Res 180: 266 –273,
Ca2⫹ handling and shortening in sepsis. Crit Care Med 33: 598 – 604, 2005.
2013.
808. Zifko UA, Zipko HT, Bolton CF. Clinical and electrophysiological findings in critical
789. Yang JS, Sladky JT, Kallen RG, Barchi RL. TTX-sensitive and TTX-insensitive sodium
illness polyneuropathy. J Neurol Sci 159: 186 –193, 1998.
channel mRNA transcripts are independently regulated in adult skeletal muscle after
denervation. Neuron 7: 421– 427, 1991. 809. Zink W, Kaess M, Hofer S, Plachky J, Zausig YA, Sinner B, Weigand MA, Fink RHA,
Graf BM. Alterations in intracellular Ca2⫹-homeostasis of skeletal muscle fibers dur-
790. Yang JS, Bennett PB, Makita N, George AL, Barchi RL. Expression of the sodium ing sepsis. Crit Care Med 36: 1559 –1563, 2008.
channel beta 1 subunit in rat skeletal muscle is selectively associated with the tetro-
dotoxin-sensitive alpha subunit isoform. Neuron 11: 915–922, 1993. 810. Zhong J, Hwang TC, Adams HR, Rubin LJ. Reduced L-type calcium current in ventric-
ular myocytes from endotoxemic guinea pigs. Am J Physiol Heart Circ Physiol 273:
791. Yasuda S, Lew W. Lipopolysaccharide depresses cardiac contractility and ␤-adrener- H2312–H2324, 1997.
gic contractile response by decreasing myofilament response to Ca2⫹ in cardiac
myocytes. Circ Res 81: 1011–1020, 1997. 811. Zochodne DW, Bolton CF, Thompson RT, Driedger AA, Hahn AF, Gilbert JJ. Myop-
athy in critical illness. Muscle Nerve 9: 652, 1986.
792. Yasuhara S, Perez ME, Kanakubo E, Yasuhara Y, Shin YS, Kaneki M, Fujita T, Martyn
JAJ. Skeletal muscle apoptosis after burns is associated with activation of proapoptotic 812. Zochodne DW, Bolton CF, Wells GA, Gilbert JJ, Hahn AF, Brown JD, Sibbald WA.
signals. Am J Physiol Endocrinol Metab 279: E1114 –E1121, 2000. Critical illness polyneuropathy. A complication of sepsis and multiple organ failure.
Brain 110: 819 – 841, 1987.
793. Yoshida M, Hama H, Ishikawa-Sakurai M, Imamura M, Mizuno Y, Araishi K, Waka-
bayashi-Takai E, Noguchi S, Sasaoka T, Ozawa E. Biochemical evidence for associa- 813. Zucchi R, Ronca-Testoni S. The sarcoplasmic reticulum Ca2⫹ channel/ryanodine re-
tion of dystrobrevin with the sarcoglycan complex as a basis for understanding sar- ceptor: modulation by endogenous effectors, drugs and disease states. Pharmacol Rev
coglycanopathy. Hum Mol Genet 9: 1033–1040, 2000. 49: 1–51, 1997.

Physiol Rev • VOL 95 • JULY 2015 • www.prv.org 1109

You might also like