Findings From Critical Illness To Explain Myalgic EncephalomyelitisChronic Fatigue Syndrome

Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

PERSPECTIVE

published: 08 March 2022


doi: 10.3389/fmed.2022.818728

Perspective: Drawing on Findings


From Critical Illness to Explain
Myalgic Encephalomyelitis/Chronic
Fatigue Syndrome
Dominic Stanculescu 1 and Jonas Bergquist 2,3*
1
Independent Researcher, Sint-Martens-Latem, Belgium, 2 Division of Analytical Chemistry and Neurochemistry, Department
of Chemistry – Biomedical Center, Uppsala University, Uppsala, Sweden, 3 The Myalgic Encephalomyelitis/Chronic Fatigue
Syndrome (ME/CFS) Collaborative Research Centre at Uppsala University, Uppsala, Sweden

We propose an initial explanation for how myalgic encephalomyelitis / chronic fatigue


syndrome (ME/CFS) could originate and perpetuate by drawing on findings from critical
illness research. Specifically, we combine emerging findings regarding (a) hypoperfusion
and endotheliopathy, and (b) intestinal injury in these illnesses with our previously
published hypothesis about the role of (c) pituitary suppression, and (d) low thyroid
hormone function associated with redox imbalance in ME/CFS. Moreover, we describe
Edited by:
Juarez Antonio Simões Quaresma,
interlinkages between these pathophysiological mechanisms as well as “vicious cycles”
Universidade do Estado do involving cytokines and inflammation that may contribute to explain the chronic nature
Pará, Brazil
of these illnesses. This paper summarizes and expands on our previous publications
Reviewed by:
about the relevance of findings from critical illness for ME/CFS. New knowledge on
Anthony L. Komaroff,
Brigham and Women’s Hospital and diagnostics, prognostics and treatment strategies could be gained through active
Harvard Medical School, collaboration between critical illness and ME/CFS researchers, which could lead to
United States
Katherine Sylvia Rowe,
improved outcomes for both conditions.
Royal Children’s Hospital, Australia
Keywords: post-viral fatigue, hypoperfusion, endotheliopathy, gut permeability, endotoxemia, pituitary, non-
*Correspondence: thyroidal illness syndrome, myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS)
Jonas Bergquist
[email protected]
INTRODUCTION
Specialty section:
This article was submitted to Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a debilitating illness that affects
Pathology, millions of people worldwide (an estimated 800,000 to 2.5 million in the USA) (1, 2). Impaired
a section of the journal
function, post-exertional malaise, and unrefreshing sleep are core symptoms (1, 3, 4). At least one-
Frontiers in Medicine
quarter of ME/CFS patients are house- or bedbound at some point in their lives (1); the illness can
Received: 25 November 2021 be completely incapacitating (5). The etiology of the illness is unclear (6, 7) and peri-onset events
Accepted: 11 February 2022
include infection-related episodes, stressful incidents, and exposure to environmental toxins (8).
Published: 08 March 2022
Critical illness refers to the physiological response to virtually any severe injury or infection,
Citation: such as head injury, burns, cardiac surgery, SARS-CoV-2 infection and heat stroke (9). Researchers
Stanculescu D and Bergquist J (2022)
make a distinction between the acute phase of critical illness—in the first hours or days following
Perspective: Drawing on Findings
From Critical Illness to Explain Myalgic
severe trauma or infection; and the chronic or prolonged phase—in the case of patients who survive
Encephalomyelitis/Chronic Fatigue the acute phase but for unknown reasons do not start recovering and continue to require intensive
Syndrome. Front. Med. 9:818728. care (10–13). Regardless of the initial injury or infection, these “chronic Intensive Care Unit (ICU)
doi: 10.3389/fmed.2022.818728 patients” experience profound muscular weakness, cognitive impairment, pain, vulnerability to

Frontiers in Medicine | www.frontiersin.org 1 March 2022 | Volume 9 | Article 818728


Stanculescu and Bergquist Drawing on Critical Illness to Explain ME/CFS

infection, etc. (9, 11, 14). The treatment of prolonged critical lining (i.e., glycocalyx) are cytokines (31), inflammation,
illness is incomplete and remains an active area of research. exposure to oxidative stress (28, 32) and/or sympatho-adrenal
Moreover, cognitive and/or physical disability can last for months hyperactivation (33). Crucially, endothelial dysfunction during
or even years after treatment in ICUs (i.e., post intensive care critical illness has been associated with altered cerebral blood flow
syndrome, PICS) for as of yet unexplained reasons (15–17). (34, 35) and increased blood–brain barrier (BBB) permeability
Drawing on findings from critical illness, we here propose resulting in long-term cognitive impairment (36, 37). A leaky
an initial explanation for how ME/CFS could originate and BBB could also contribute to increased intracranial pressure
perpetuate. Specifically, we combine emerging findings regarding (38, 39). Finally, researchers have found that endotheliopathy
(a) hypoperfusion and endotheliopathy, and (b) intestinal injury and coagulation disorder bolster each other via inflammatory
in these illnesses with our previously published hypothesis about pathways (40). Coagulation abnormalities vary in critical illness,
the role of (c) pituitary suppression, and (d) low thyroid hormone but coagulopathy is associated with unfavorable outcomes
function associated with redox imbalance in ME/CFS. Moreover, in prolonged critical illness (i.e., length of ICU stay and
we describe interlinkages between these pathophysiological mortality) (41).
mechanisms as well as “vicious cycles” involving cytokines and We propose that similar alterations of the vascular system
inflammation that may contribute to explain the chronic nature in response to a physical, infectious and / or emotional
of these illnesses. This explanation summarizes and expands stressor (i.e., physiological insult) may also contribute to explain
on our previous publications about the relevance of findings the emergence of ME/CFS. This is consistent with recent
from critical illness for ME/CFS (18–20) and builds on the hypotheses describing vasoconstriction in muscle and brain
work by Nacul et al. (21). The general lack of large high-quality as a principal element of ME/CFS (42–46), and findings of
ME/CFS studies (a reflection of the lack of funding in this field) cerebral hypoperfusion (47–49) and intracranial hypertension
poses a challenge for the assessment of overlaps between the (50) in ME/CFS patients. It is also consistent with studies that
two conditions. have shown that endothelial function is impaired in ME/CFS
(51, 52), both in large vessels and in the microcirculation
(53, 54)—associated with redox imbalance (51). Finally, it is
PATHOPHYSIOLOGICAL MECHANISMS consistent with a new hypothesis for ME/CFS which suggests
In the following sections we describe four central that endothelial senescence underpins ME/CFS by disrupting the
pathophysiological mechanisms in critical illness, including intestinal barriers and BBBs (55), as well as with suggestions that
their relationship to inflammation. We also provide initial leakage from dysfunctional blood vessels could explain many of
arguments for suggesting that similar mechanisms may underlie the symptoms in ME/CFS (56).
ME/CFS. Readers are referred to our prior publications for
additional details about these mechanisms in critical illness
(including heat stroke) and possible lessons for understanding Intestinal Injury
ME/CFS (18–20). Critical illness researchers have found profound intestinal
alterations within hours following a physiological insult: a
dramatic shift in the composition and virulence of intestinal
Hypoperfusion and Endotheliopathy microbes (57–59), an erosion of the mucus barrier, an increase
It has long been suggested that inadequate oxygen circulation
in the permeability of the gut (i.e., “leaky gut”) (60–62), and a
is central to critical illness (22). Specifically, the redistribution
disruption in gut motility (63). This intestinal injury is thought
of blood away from the splanchnic area to critical tissues is
to be largely a consequence of local I/R and redox imbalance
considered an adaptive androgenic response to physiological
resulting from splanchnic hypoperfusion (58, 61, 64–67). Indeed,
stress (23, 24). However, the resulting ischemia / reperfusion
studies in the field of exercise immunology have shown that even
(I/R) can contribute to tissue injury driving sepsis and multi-
relatively low levels of splanchnic hypoperfusion during exercise
organ dysfunction (25, 26). The relative importance of reduced
result in intestinal injury (68).
blood flow, vasoconstriction (27), capillary flow disturbances
Critically, this intestinal injury may lead to bacterial
(28) and impaired cellular oxygen utilization (29, 30) in driving
translocation from the gut into circulation (i.e., endotoxemia)
critical illness continues to be debated.
and/or the formation of toxic gut-derived lymph (57, 60). This
Endothelial dysfunction appears to occur in parallel with
in turn can induce pro-inflammatory cytokines and systemic
circulation disturbances during critical illness. Probable drivers
inflammation (69, 70). Moreover, changes in the intestinal
of distortions in the structure and function of endothelial
microbiome or the mucus barrier may also impact the immune
Abbreviations: BBB, Blood–brain barrier; ACTH, Adrenocorticotropic hormone;
system directly (57). Thus, researchers have long considered the
GH, Growth hormone; GR, glucocorticoid receptors; HPA, hypothalamus- gut “the motor of critical illness” driving sepsis and distant organ
pituitary-adrenal axis: “Adreno-cortical axis”; HPS, Hypothalamic-pituitary- dysfunction (71). Some have suggested that a self-perpetuating
somatotropic axis: “Somatropic axis”; HPT, Hypothalamic-pituitary-thyroid: vicious inflammatory cycle centered around intestinal injury can
“Thyrotropic axis”; ICU, Intensive Care Unit; I/R, Ischemia/reperfusion; ME/CFS, hinder recovery from critical illness (61, 72).
Myalgic Encephalomyelitis/Chronic Fatigue Syndrome; NO, Nitrox oxide; NTIS,
Non-thyroidal illness syndrome; O&NS, oxidative and nitrosative stress; PACS,
We propose that the sequence during critical illness—from
Post-acute COVID-19 syndrome; PICS, Post-intensive care syndrome; TRH, splanchnic hypoperfusion to hypoxia, redox imbalance, altered
Thyrotropin-releasing hormone; TSH, Thyroid stimulating hormone. gut microbiome, intestinal injury, gut-related endotoxemia,

Frontiers in Medicine | www.frontiersin.org 2 March 2022 | Volume 9 | Article 818728


Stanculescu and Bergquist Drawing on Critical Illness to Explain ME/CFS

pro-inflammatory cytokines and systemic inflammatory— We propose that the sequence during critical illness—from
may also contribute to explain the emergence of ME/CFS increased release of pituitary hormones during the acute phase
following a physiological insult. Our proposal is in alignment to suppression of the pituitary gland’s pulsatile secretion in the
with others’ findings that intestinal injury and resulting prolonged phase—could also contribute to explain the emergence
inflammation are central to ME/CFS (73–81) and consistent of ME/CFS following a physiological insult. This proposal is
with findings linking the gut microbiome to inflammation consistent with descriptions of ME/CFS as a progression from
(82–85) and to fatigue symptoms in ME/CFS (86). If verified, a hypermetabolic to hypometabolic state (21). It also aligns
the existence of a vicious inflammatory cycle centered around with a recent hypothesis relating many of the symptoms in
intestinal injury could contribute to explain the perpetuation of severe ME/CFS to impaired pituitary function (111). Further
ME/CFS. Post-exertional malaise—a key symptom of ME/CFS— support for this proposal is provided by the many previous
could be the manifestation of an accentuation in intestinal ME/CFS studies that have documented dysfunctions in the
injury following exertion. Moreover, the translocation of gut hypothalamic–pituitary–somatotropic (HPS) axis (112–114), the
microbes or toxin from the intestines to the brain (55) might HPT axis (115–120), and the HPA axis (121–136)—notably
contribute to explain central nervous system inflammation associated with inflammation and oxidative & nitrosative stress
in ME/CFS (87–89). Finally, leaky gut is also associated with (O&NS) (137–140). Strikingly, models relating the persistence
auto-immunity (90, 91)—an important factor in ME/CFS of a suppressed HPA axis in ME/CFS to a change in central
pathology (92–94). GRs concentrations resemble the explanations provided for
pituitary suppression in critical illness (141–146). Moreover,
Pituitary Suppression suppression of ACTH release would explain why in a small study
Almost immediately after a physiological insult, endocrine axes ME/CFS patients were found to have 50% smaller adrenals than
experience profound alterations considered a vital response to controls (147), resembling adrenal atrophy in prolonged critical
severe stress or injury to allow for a shift in energy and resources illness. However, the relationship between the pituitary’s pulsatile
to essential organs and repair (95–97). Whereas, in critically secretions, physiological alterations and severity of illness—
ill patients who begin to recover, endocrine axes essentially which proved revelatory in understanding prolonged critical
normalize within 28 days of illness, in cases of prolonged critical illness—remains unexplored in ME/CFS.
illness the pituitary’s pulsatile secretion of tropic hormones
(unexpectedly) remains suppressed. Low Thyroid Hormone Function
Why and how this central suppression is maintained in Peripheral mechanisms involving cytokines lead to the rapid
prolonged critical illness continues to be debated. Inflammatory depression of thyroid hormone activity following a severe
pathways likely play a role irrespective of the nature of the physiological insult (148–152). This is termed “non-thyroidal
original injury or infection. For example, cytokines increase the illness syndrome” (NTIS), “euthyroid sick syndrome” or “low
abundance and affinity of glucocorticoid receptors (GR) at the T3 syndrome” and is thought to be an adaptive response
level of the hypothalamus / pituitary, thereby enhancing the to conserve energy resources during critical illness (152–154).
negative feedback loop of the hypothalamic-pituitary-adrenal The mechanisms involved include alterations in the half-life
(HPA) axis, and consequently suppressing pituitary release of thyroid hormone in circulation (155–157); modifications in
of adrenocorticotropic hormone (ACTH) (95, 98). Similarly, the uptake of thyroid hormone by cells (158, 159); down- and
cytokines up-regulate deiodinase enzymes in the hypothalamus up-regulation of deiodinase enzymes that convert the thyroid
resulting in higher local levels of the active thyroid hormone (T3), hormone into active and inactive forms respectively (156, 160);
thereby enhancing the hypothalamic-pituitary-thyroid (HPT) and alterations in sensitivity of cells to thyroid hormones (161–
axis’ negative feedback loop and consequently suppressing 163). These alterations can lead to important tissue-specific
pituitary secretion of thyroid stimulating hormone (TSH) depression in thyroid hormone function (164, 165) which is,
irrespective of circulating thyroid hormone concentrations (99– however, often missed altogether in clinical settings (166) because
101). Cytokines may also suppress the release of TSH by the most of the alterations do not translate into changes in the blood
pituitary directly (102, 103) contributing to a virtual complete concentrations of thyroid hormones (164, 167, 168). Indeed, the
loss of pulsatile TSH secretion (96). decrease in the ratio of the active form of thyroid hormone
The loss of pulsatile pituitary secretions has important (T3) relative to the inactivated thyroid hormone (rT3) (150, 152,
implications for the autonomic nervous system, metabolism, 169)—considered the most sensitive marker of NTIS—may be
and the immune system. Without sufficient pulsatile stimulation just the “tip of the iceberg” of the depressed thyroid hormone
by ACTH, adrenal glands begin to atrophy (104, 105), function in target tissues (120, 170).
compromising patients’ ability to cope with external stressors While NTIS may be beneficial in the acute phase of critical
and permitting excessive inflammatory responses. Erratic rather illness, it is increasingly seen as maladaptive and hampering
than pulsatile pituitary production of growth hormone (GH) the recovery of patients in the case of prolonged critical illness
leads to an imbalance between catabolic and anabolic hormones, (96, 101, 152, 169, 171–173). Low thyroid hormone function
resulting in loss of muscle and bone mass, muscle weakness, may hamper the function of organs (170) and the activity of
and changes in glucose and fat metabolism (106–108). Finally, immune cells, including natural killer cells (174–185). Immune
suppression of the HPT axis is associated with tiredness and other dysfunctions might in turn explain other pathologies, such as
hypothyroid-like symptoms (109, 110). viral reactivation observed in ICU patients (186–188). Some

Frontiers in Medicine | www.frontiersin.org 3 March 2022 | Volume 9 | Article 818728


Stanculescu and Bergquist Drawing on Critical Illness to Explain ME/CFS

critical illness researchers have proposed a model that describes solving ME/CFS given that “several of the main typical symptoms
how NTIS is maintained by reciprocal relationships between in severe ME/CFS, such as fatigue, myalgia, contractility, delaying
inflammation (notably pro-inflammatory cytokines), O&NS and muscle recovery and function, exertional malaise, neurocognitive
reduced thyroid hormone function, forming a “vicious cycle” dysfunction, and physical disability may be related to severe GH
(101, 173). This model can help to explain the perplexing failure deficiency” (111).
to recover of some critically ill patients in ICUs that survive their
initial severe illness or injury.
We propose that low thyroid hormone function could also
Linkages Between Pituitary Suppression
contribute to explain the emergence of ME/CFS following and Low Thyroid Hormone Function
a physiological insult. An immune-mediated loss of thyroid There are several pathways linking the activity of the pituitary
hormone function in ME/CFS has long been suspected (117). A with that of thyroid hormones. Firstly, GH secreted by the
recent study showed that the thyroid panel of ME/CFS patients pituitary co-regulates the activity of the deiodinase enzyme
resembles that of critical illness patients, including significantly (D3) responsible for the conversion of thyroid hormones into
lower ratio of T3 to rT3 hormones (120). Moreover, the other inactive forms (i.e., rT3 and inactivate forms of T2) (106, 201).
elements for a “vicious cycle” which researchers have suggested Researchers showed that normalization of the GH secretion in
perpetuate a hypometabolic and inflammatory state in critical prolonged critically ill patients is necessary to inhibit the increase
illness are also present in ME/CFS, including inflammation (140, in plasma rT3 concentrations (96, 108, 198). In other words,
189), increased O&NS (190–192) and altered cytokine profiles dampened GH release by the pituitary during prolonged critical
(193, 194). illness enables low thyroid hormone function. Secondly, the lack
of stimulation of the adrenals by ACTH could (by causing an
atrophy of adrenals) create the condition necessary for persistent
DISCUSSION inflammation which depresses the activity of thyroid hormones
during critical illness (148–152). In other words, dampened
Hypoperfusion and endotheliopathy, intestinal injury, pituitary
ACTH release by the pituitary during prolonged critical illness
suppression, and low thyroid hormone function are each central
might permit the vicious inflammatory cycles described above.
to prolonged critical illness regardless of the nature of the initial
Thirdly, there is evidence that thyroid hormone conversely also
severe injury or infection (101, 173, 195, 196). We propose that,
stimulates ACTH secretion (202, 203). In summary, the bi-
similarly, these mechanisms and their reciprocal relationships
directional relationships between the endocrine axes and thyroid
with inflammation could underlie ME/CFS regardless of the
hormone function (in addition to reciprocal relationships with
nature of the peri-onset event (i.e., infection, stressful incident,
inflammation) could contribute to explain the persistence of
exposure to environmental toxins or other) (Table 1). Moreover,
chronic ICU and ME/CFS.
the severity of ME/CFS may be a function of the strength of
these mechanisms.
However, each of these pathological mechanisms has largely
Linkages Between Low Thyroid Hormone
been studied in isolation and rarely have the linkages between Function and Endothelial Function
them been explored. Yet, the aggregate of these mechanisms Upon binding to specific receptors on endothelial cells, thyroid
is likely necessary to fully explain the perpetuation of critical hormones (T3 and T4) activate the endothelial nitric oxide
illness—and to inform the understanding of ME/CFS (Figure 1). synthase (eNOS) responsible for nitric oxide (NO) production
Additional areas for inquiry thus include the following: (204), which in turn impacts vasodilation and inflammation
(205–207). A further line of inquiry may thus be the role
of thyroid hormone function in endotheliopathy in ME/CFS,
Linkages Between Intestinal Injury and including as it relates to the new finding that plasma
Pituitary Suppression from ME/CFS patients inhibits eNOS and NO production in
Intestinal injury during critical illness results in decreased endothelial cells (208). Relatedly, critical illness researchers
secretion of gastrointestinal hormones including ghrelin (63, have found that serum from patients with NTIS inhibits the
197). Decreased stimulation of the pituitary and hypothalamus uptake of thyroid hormone (209, 210); the mechanisms remain
by ghrelin during prolonged critical illness in turn results in unresolved (165).
lower secretion of GH by the pituitary (199). Researchers have
found that the administration of an artificial ghrelin in chronic Linkages to Mitochondrial Function
ICU patients reactivated the pulsatile secretion of GH by the The impaired perfusion, redox imbalance, lower thyroid
pituitary and—when done in combination with thyrotropin- hormone function and inflammation appear to collectively affect
releasing hormones (TRH)—had beneficial metabolic effects mitochondrial activity in critical illness (via inhibition, damage,
(96, 108, 198). Similarly, the administration of ghrelin to and/or decreased turnover of new mitochondrial protein) (30,
the I/R rats “inhibited pro-inflammatory cytokine release, 211–213). Mitochondrial activity may be similarly affected in
reduced neutrophil infiltration, ameliorated intestinal barrier ME/CFS (190). Some have suggested that this down-regulation of
dysfunction, attenuated organ injury, and improved survival” mitochondrial activity (and oxygen utilization) in critical illness
(200). The sequence between intestinal injury, ghrelin secretion may be an adaptive form of “hibernation” to protect cells from
and GH release by the pituitary could be particularly relevant for death pathways (30, 213). This suggestion echoes the hypothesis

Frontiers in Medicine | www.frontiersin.org 4 March 2022 | Volume 9 | Article 818728


Stanculescu and Bergquist Drawing on Critical Illness to Explain ME/CFS

TABLE 1 | Central pathophysiological mechanisms in prolonged critical illness, probable drivers and implications, and initial evidence suggesting similar mechanisms in
ME/CFS.

Pathophysiological mechanisms In prolonged critical illness (Probable drivers and In ME/CFS (Initial evidence)
implications)

Hypoperfusion Drivers: Initial evidence


• Redistribution of blood away from the splanchnic area • Vasoconstriction in muscle and brain (42–45)
to critical tissues (23, 24) • Cerebral hypoperfusion (47–49)
• Reduced blood flow, vasoconstriction (27) • Intracranial hypertension (50)
• Capillary flow disturbances (28)
• Additional: impaired cellular oxygen utilization (29, 30)
Implications:
• Ischemia / reperfusion (I/R)
• Tissue injury driving sepsis and multi-organ
dysfunction (25, 26)
Endotheliopathy Drivers: Initial evidence
• Cytokines (31), Inflammation, exposure to oxidative • Impaired endothelial function (51, 52), in large vessels
stress (28, 32) and microcirculation (53, 54)—associated with redox
• Sympatho-adrenal hyperactivation (33) imbalance (51)
Implications: • Endothelial senescence disrupting the intestinal barriers
• Altered cerebral blood flow (34, 35) and BBBs (55)
• Increased blood–brain barrier (BBB) permeability • Redox imbalance
(36, 37)
• Increased intracranial pressure (38, 39).
• (variable) Coagulation disorder (40)
Intestinal injury Drivers: Initial evidence
• Local I/R and redox imbalance resulting from • Intestinal injury and resulting inflammation (73–81)
splanchnic hypoperfusion (58, 61, 64–67) • Altered gut microbiome linked to inflammation (82–85).
• Disruption in gut motility (63) • Lack of beneficial gut bacteria linked to fatigue
• Shift in the composition and virulence of intestinal symptoms (86)
microbes (57–59) • Endothelial senescence disrupting the intestinal barriers
Implications: (55)
• Erosion of the mucus barrier, increase in the • Auto-immunity (92–94)
permeability of the gut (i.e., “leaky gut”) (60–62)
• Bacterial translocation from the gut into circulation
(i.e., endotoxemia) and/or the formation of toxic
gut-derived lymph (57, 60)
• Pro-inflammatory cytokines and systemic inflammation
(69, 70)
• Direct impacts on the immune system (57)
• Vicious inflammatory cycle centered around intestinal
injury (61, 72)
• Decreased secretion of gastrointestinal hormones
including ghrelin (63, 197) impacting pituitary activity
Suppression of pulsatile pituitary Drivers Initial evidence
function • Cytokines acting on abundance and affinity of • Progression from a hypermetabolic to hypometabolic
glucocorticoid receptors (GR) at central level (95, 98) state (21)
• Cytokines affecting deiodinase enzymes in the • Impaired pituitary function (hypothesis) (111)
hypothalamus (99–101) • Dysfunctions in HPS axis (112–114), HPT axis (115–
• Direct action of cytokines on TSH release by the 120) and HPA axis (121–136) – associated with
pituitary directly (102, 103) inflammation O&NS (137–140)
Implications • Changes in central GRs concentrations (models) (141–
• Loss of ACTH pulsatility: atrophy of adrenal glands 146)
(104, 105) compromising patients’ ability to cope with • Smaller adrenals (147)
external stressors and permitting excessive
inflammatory responses
• Loss of GH pulsatility: imbalance between catabolic
and anabolic hormones, resulting in loss of muscle
and bone mass, muscle weakness, and changes in
glucose and fat metabolism (106–108). Alterations in
deiodinase enzyme (D3) activity enabling low thyroid
hormone function (96, 108, 198)
• Loss of TSH pulsatility (109, 110)

(Continued)

Frontiers in Medicine | www.frontiersin.org 5 March 2022 | Volume 9 | Article 818728


Stanculescu and Bergquist Drawing on Critical Illness to Explain ME/CFS

TABLE 1 | Continued

Pathophysiological mechanisms In prolonged critical illness (Probable drivers and In ME/CFS (Initial evidence)
implications)

Low thyroid hormone function Drivers Initial evidence


• Alterations in the half-life of thyroid hormone in • Immune-mediated loss of thyroid hormone function in
circulation (155–157) ME/CFS (suspected) (117)
• Modifications in the uptake of thyroid hormone by cells • Significantly lower ratio of T3 to rT3 hormones (120)
(158, 159)
• Down- and up-regulation of deiodinase enzymes that
convert the thyroid hormone into active and inactive
forms, respectively (156, 160)
• Alternations in sensitivity of cells to thyroid hormones
(161–163)
Implications
• Tissue-specific depression in thyroid hormone function
(164–166)
• Hampered function of organs (170)
• Altered activity of immune cells, including natural killer
cells (174–185)
• Viral reactivation (186–188)
• Vicious inflammatory cycle (101, 173)

FIGURE 1 | Central pathophysiological mechanisms in critical illness including selected consequences and inter-linkages. Hypoperfusion and endotheliopathy,
intestinal injury, pituitary suppression, and low thyroid hormone function are each central to prolonged critical illness regardless of the nature of the initial severe injury
or infection. These pathophysiological mechanisms are in reciprocal relationships with inflammation; specifically, researchers have proposed vicious cycles involving
intestinal injury and low thyroid hormone function. Moreover, linkages have been described between these pathophysiological mechanisms, including (i) hypoperfusion
and intestinal injury (i.e., leaky gut resulting from ischemia/reperfusion, hypoxia and redox imbalance); (ii) intestinal injury and pituitary suppression (i.e., suppressed
growth hormone release resulting from reduced ghrelin secretion by the intestines); (iii) pituitary suppression and low thyroid hormone function (i.e., increased
inactivated thyroid hormone resulting from the upregulation of D3 deiodinase as a consequence of lower growth hormone); and (iv) low thyroid hormone function and
pituitary suppression (i.e., decreased ACTH secretion resulting from lower levels of activated thyroid hormone). We propose that these mechanisms and the linkages
between them—alongside reciprocal relationships with inflammation—could also underlie ME/CFS.

that ME/CFS is a form of “dauer” or “cell danger response” (214– Relevance of Critical Illness Treatment
216). Lower mitochondrial activity in turn affects the immune Trials for ME/CFS
system and the gut endothelial “such that the host’s immune Although prolonged critical illness remains unresolved, early
response and physical barriers to infection are simultaneously treatment trials—such as the reactivation of the pituitary, or
compromised” (29). interruption of the vicious inflammatory cycles centered around

Frontiers in Medicine | www.frontiersin.org 6 March 2022 | Volume 9 | Article 818728


Stanculescu and Bergquist Drawing on Critical Illness to Explain ME/CFS

either gut injury or low thyroid hormone function—may provide predisposing genetic or environmental factors, we propose that
therapeutic avenues for ME/CFS (19). Longitudinal studies of the pathological mechanisms—and the interlinkages between
(spontaneous) recovery from critical illness may also give clues them—that prevent recovery of some critically ill patients
about prerequisites for recovery from ME/CFS. Researchers have, may also underlie ME/CFS. This initial proposal is in line
for example, found that “supranormal TSH precedes onset of with and complements several existing hypotheses of ME/CFS
recovery” from prolonged critical illness (96) and that metabolic pathogenesis. If this hypothesis is validated, past treatment
rate rises > 50% above normal in the recovery phase (213). trials for critical illness may provide avenues for a cure for
ME/CFS. Certainly, given the similarities described above, active
Commonality With Other Illnesses collaboration between critical illness and ME/CFS researchers
Researchers have suggested commonality in the illnesses induced could lead to improved understanding of not only both
by physical, infectious, and / or emotional stressors (132, 217). conditions, but also PICS, long-COVID, PACS, and fibromyalgia.
These include heat stroke, fibromyalgia, ME/CFS, prolonged
critical illness, PICS, cancer-related fatigue, post-viral fatigue,
post-acute COVID-19 syndrome (PACS) and long-COVID.
DATA AVAILABILITY STATEMENT
Specifically, it is necessary to explore whether the pathological The original contributions presented in the study are included
mechanisms described above also underlie long COVID—a in the article/supplementary material, further inquiries can be
disease which resembles ME/CFS (218–228) and can arise even directed to the corresponding author/s.
after mild COVID-19 cases.

CONCLUSION AUTHOR CONTRIBUTIONS


DS wrote the first draft of the manuscript. All authors
Decades of research in the field of critical illness medicine
contributed to manuscript revision, read, and approved the
have demonstrated that in response to the stress of severe
submitted version.
infection or injury, the vascular system, intestines, endocrine axes
and thyroid hormone function experience profound alterations.
Self-reinforcing interlinkages between these pathophysiological FUNDING
mechanisms as well as “vicious cycles” involving cytokines
and inflammation may perpetuate illness irrespective of the The Open Medicine Foundation (JB) is acknowledged
initial severe infection or injury. Without excluding possible for support.

REFERENCES 10. Van den Berghe G. Novel insights into the neuroendocrinology of critical
illness. Eur J Endocrinol. (2000) 143:1–13. doi: 10.1530/eje.0.1430001
1. Institute of Medicine. Beyond Myalgic Encephalomyelitis/Chronic Fatigue 11. Nelson JE, Cox CE, Hope AA, Carson SS. Chronic critical illness. Am J Respir
Syndrome: Redefining an Illness. Washington, DC: The National Academies Crit Care Med. (2010) 182:446–44610espir C1164/rccm.201002-0210CI
Press (2015). 12. Van den Berghe GH. Acute and prolonged critical illness are two distinct
2. Jason LA, Mirin AA. Updating the National Academy of Medicine neuroendocrine paradigms. Verh K Acad Geneeskd Belg. (1998) 60:487–518;
ME/CFS prevalence and economic impact figures to account for discussion −20.
population growth and inflation. Fatigue Biomed Health Behav. (2021) 13. Vanhorebeek I, Van den Berghe G. The neuroendocrine response to
9:9–13. doi: 10.1080/21641846.2021.1878716 critical illness is a dynamic process. Crit Care Clin. (2006) 22:1–15,
3. Open Medicine Foundation. Symptoms of ME/CFS. (2020). Available online v. doi: 10.1016/j.ccc.2005.09.004
at: https://www.omf.ngo/symptoms-mecfs (accessed March 27, 2021). 14. Vanhorebeek I, Latronico N, Van den Berghe G. ICU-acquired weakness.
4. Nacul L, Authier FJ, Scheibenbogen C, Lorusso L, Helland IB, Martin JA, Intensive Care Med. (2020) 46:637–53. doi: 10.1007/s00134-020-05944-4
et al. European Network on Myalgic Encephalomyelitis/Chronic Fatigue 15. Van Aerde N, Van Dyck L, Vanhorebeek I, Van den Berghe G.
Syndrome (EUROMENE): expert consensus on the diagnosis, service Endocrinopathy of the Critically Ill. In: Preiser J-C, Herridge M, Azoulay
provision, and care of people with ME/CFS in Europe. Medicina. (2021) E, editors. Post-Intensive Care Syndrome. Cham: Springer International
57:510. doi: 10.3390/medicina57050510 Publishing (2020). p. 125–43.
5. Dafoe W. Extremely severe ME/CFSra personal account. Healthcare. (2021) 16. Rawal G, Yadav S, Kumar R. Post-intensive care syndrome: an overview. J
9:504. doi: 10.3390/healthcare9050504 Transl Int Med. (2017) 5:90–2. doi: 10.1515/jtim-2016-0016
6. Komaroff AL. Advances in understanding the pathophysiology of chronic 17. Smith S, Rahman O. Post Intensive Care Syndrome. Treasure Island, FL:
fatigue syndrome. JAMA. (2019) 322:499–500. doi: 10.1001/jama.2019.8312 StatPearls Publishing (2020).
7. Komaroff AL. Myalgic encephalomyelitis/chronic fatigue 18. Stanculescu D, Larsson L, Bergquist J. Hypothesis: mechanisms that
syndrome: when suffering is multiplied. Healthcare. (2021) prevent recovery in prolonged ICU patients also underlie Myalgic
9:919. doi: 10.3390/healthcare9070919 Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS). Front Med. (2021)
8. Chu L, Valencia IJ, Garvert DW, Montoya JG. Onset patterns and course of 8:628029. doi: 10.3389/fmed.2021.628029
myalgic encephalomyelitis/chronic fatigue syndrome. Front Pediatr. (2019) 19. Stanculescu D, Larsson L, Bergquist J. Theory: treatments for
7:12. doi: 10.3389/fped.2019.00012 prolonged ICU patients may provide new therapeutic avenues
9. Loss SH, Nunes DSL, Franzosi OS, Salazar GS, Teixeira C, Vieira SRR. for Myalgic Encephalomyelitis/Chronic Fatigue Syndrome
Chronic critical illness: are we saving patients or creating victims? Rev Bras (ME/CFS). Front Med. (2021) 8:672370. doi: 10.3389/fmed.202
Ter Intensiva. (2017) 29:87–95. doi: 10.5935/0103-507X.20170013 1.672370

Frontiers in Medicine | www.frontiersin.org 7 March 2022 | Volume 9 | Article 818728


Stanculescu and Bergquist Drawing on Critical Illness to Explain ME/CFS

20. Stanculescu D, Sepulveda N, Lim CL, Bergquist J. Lessons from heat stroke 40. Vallet B, Wiel E. Endothelial cell dysfunction and coagulation. Crit Care Med.
for understanding Myalgic Encephalomyelitis / Chronic Fatigue Syndrome. (2001) 29:S36–41. doi: 10.1097/00003246-200107001-00015
Front Neurol. (2021) 12:789784. doi: 10.3389/fneur.2021.789784 41. Winer LK, Salyer C, Beckmann N, Caldwell CC, Nomellini V. Enigmatic
21. Nacul L, O’Boyle S, Palla L, Nacul FE, Mudie K, Kingdon CC, et al. role of coagulopathy among sepsis survivors: a review of coagulation
How Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) abnormalities and their possible link to chronic critical illness. Trauma Surg
progresses: the natural history of ME/CFS. Front Neurol. (2020) Acute Care Open. (2020) 5:e000462. doi: 10.1136/tsaco-2020-000462
11:826. doi: 10.3389/fneur.2020.00826 42. Wirth KJ, Scheibenbogen C. Pathophysiology of skeletal muscle disturbances
22. Broder G, Weil MH. Excess lactate: an index of reversibility in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS). J
of shock in human patients. Science. (1964) 143:1457– Transl Med. (2021) 19:162. doi: 10.1186/s12967-021-02833-2
9. doi: 10.1126/science.143.3613.1457 43. Wirth K, Scheibenbogen C, A. Unifying hypothesis of the
23. Halter JB, Pflug AE, Porte D. Jr. Mechanism of plasma catecholamine pathophysiology of Myalgic Encephalomyelitis/Chronic Fatigue
increases during surgical stress in man. J Clin Endocrinol Metab. (1977) Syndrome (ME/CFS): recognitions from the finding of autoantibodies
45:936–44. doi: 10.1210/jcem-45-5-936 against ßc-adrenergic receptors. Autoimmun Rev. (2020)
24. Zhang D, Li H, Li Y, Qu L. Gut rest strategy and trophic feeding in the 19:102527. doi: 10.1016/j.autrev.2020.102527
acute phase of critical illness with acute gastrointestinal injury. Nutr Res Rev. 44. Malato J, Sotzny F, Bauer S, Freitag H, Fonseca A, Grabowska AD, et
(2019) 32:176–82. doi: 10.1017/S0954422419000027 al. The SARS-CoV-2 receptor angiotensin-converting enzyme 2 (ACE2)
25. Rock P, Yao Z. Ischemia reperfusion injury, preconditioning in myalgic encephalomyelitis/chronic fatigue syndrome: a meta-analysis
and critical illness. Curr Opin Anaesthesiol. (2002) 15:139– of public DNA methylation and gene expression data. Heliyon. (2021)
46. doi: 10.1097/00001503-200204000-00001 7:e07665. doi: 10.1016/j.heliyon.2021.e07665
26. Schwarte L, Stevens M, Ince C. Splanchnic perfusion and 45. Fluge O, Tronstad KJ, Mella O. Pathomechanisms and possible interventions
oxygenation in critical illness. Intensive Care Med. (2006) in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). J Clin
627–40. doi: 10.1007/3-540-33396-7_58 Invest. (2021) 131:e150377. doi: 10.1172/JCI150377
27. Pastores SM, Katz DP, Kvetan V. Splanchnic ischemia and gut mucosal injury 46. Wirth KJ, Scheibenbogen C, Paul F. An attempt to explain the neurological
in sepsis and the multiple organ dysfunction syndrome. Am J Gastroenterol. symptoms of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. J
(1996) 91:1697–6979 Transl Med. (2021) 19:471. doi: 10.1186/s12967-021-03143-3
28. Ostergaard L, Granfeldt A, Secher N, Tietze A, Iversen NK, Jensen MS, et al. 47. Campen C, Rowe PC, Visser FC. Orthostatic symptoms and reductions
Microcirculatory dysfunction and tissue oxygenation in critical illness. Acta in cerebral blood flow in Long-Haul COVID-19 patients: similarities
Anaesthesiol Scand. (2015) 59:1246–59. doi: 10.1111/aas.12581 with myalgic encephalomyelitis/chronic fatigue syndrome. Medicina. (2021)
29. Crouser ED. Mitochondrial dysfunction in septic shock and 58:28. doi: 10.3390/medicina58010028
multiple organ dysfunction syndrome. Mitochondrion. (2004) 48. van Campen C, Visser FC. Psychogenic pseudosyncope: real
4:729–41. doi: 10.1016/j.mito.2004.07.023 or imaginary? Results from a case-control study in myalgic
30. Singer M. The role of mitochondrial dysfunction in sepsis-induced encephalomyelitis/chronic fatigue syndrome (ME/CFS) patients. Medicina.
multi-organ failure. Virulence. (2014) 5:66–72. doi: 10.4161/vir (2022) 58:98. doi: 10.3390/medicina58010098
u.26907 49. van Campen CMC, Verheugt FWA, Rowe PC, Visser FC. Cerebral
31. Kang S, Kishimoto T. Interplay between interleukin-6 signaling and the blood flow is reduced in ME/CFS during head-up tilt testing even in
vascular endothelium in cytokine storms. Exp Mol Med. (2021) 53:1116– the absence of hypotension or tachycardia: A quantitative, controlled
23. doi: 10.1038/s12276-021-00649-0 study using Doppler echography. Clinical Neurophysiol Pract. (2020) 5:50–
32. Cerny V, Astapenko D, Brettner F, Benes J, Hyspler R, Lehmann C, et al. 8. doi: 10.1016/j.cnp.2020.01.003
Targeting the endothelial glycocalyx in acute critical illness as a challenge 50. Bragee B, Michos A, Drum B, Fahlgren M, Szulkin R, Bertilson BC. Signs
for clinical and laboratory medicine. Crit Rev Clin Lab Sci. (2017) 54:343– of intracranial hypertension, hypermobility, and craniocervical obstructions
57. doi: 10.1080/10408363.2017.1379943 in patients with myalgic encephalomyelitis/chronic fatigue syndrome. Front
33. Johansson PI, Stensballe J, Ostrowski SR. Shock induced endotheliopathy Neurol. (2020) 11:828. doi: 10.3389/fneur.2020.00828
(SHINE) in acute critical illness - a unifying pathophysiologic mechanism. 51. Blauensteiner J, Bertinat R, Leon LE, Riederer M, Sepulveda N, Westermeier
Crit Care. (2017) 21:25. doi: 10.1186/s13054-017-1605-5 F. Altered endothelial dysfunction-related miRs in plasma from ME/CFS
34. Slessarev M, Mahmoud O, McIntyre CW, Ellis CG. Cerebral patients. Sci Rep. (2021) 11:10604. doi: 10.1038/s41598-021-89834-9
blood flow deviations in critically ill patients: potential insult 52. Scherbakov N, Szklarski M, Hartwig J, Sotzny F, Lorenz S, Meyer A, et
contributing to ischemic and hyperemic injury. Front Med. (2020) al. Peripheral endothelial dysfunction in myalgic encephalomyelitis/chronic
7:615318. doi: 10.3389/fmed.2020.615318 fatigue syndrome. ESC Heart Fail. (2020) 7:1064–71. doi: 10.1002/ehf2.12633
35. Bowton DL, Bertels NH, Prough DS, Stump DA. Cerebral blood flow is 53. Newton DJ, Kennedy G, Chan KK, Lang CC, Belch JJ, Khan F. Large and
reduced in patients with sepsis syndrome. Crit Care Med. (1989) 17:399– small artery endothelial dysfunction in chronic fatigue syndrome. Int J
39989are Medo1097/00003246-198905000-00004 Cardiol. (2012) 154:335–6. doi: 10.1016/j.ijcard.2011.10.030
36. Hughes CG, Patel MB, Brummel NE, Thompson JL, McNeil 54. Sorland K, Sandvik MK, Rekeland IG, Ribu L, Smastuen MC, Mella O,
JB, Pandharipande PP, et al. Relationships between markers of et al. Reduced endothelial function in myalgic encephalomyelitis/chronic
neurologic and endothelial injury during critical illness and long- fatigue syndrome-results from open-label cyclophosphamide intervention
term cognitive impairment and disability. Intensive Care Med. (2018) study. Front Med. (2021) 8:642710. doi: 10.3389/fmed.2021.642710
44:345–55. doi: 10.1007/s00134-018-5120-1 55. Sfera A, Osorio C, Zapata Martin Del Campo CM, Pereida S, Maurer
37. Hughes CG, Morandi A, Girard TD, Riedel B, Thompson JL, Shintani S, Maldonado JC, et al. Endothelial senescence and chronic fatigue
AK, et al. Association between endothelial dysfunction and acute syndrome, a COVID-19 based hypothesis. Front Cell Neurosci. (2021)
brain dysfunction during critical illness. Anesthesiology. (2013) 118:631– 15:673217. doi: 10.3389/fncel.2021.673217
9. doi: 10.1097/ALN.0b013e31827bd193 56. Lubell J. Letter: could endothelial dysfunction and vascular damage
38. Schizodimos T, Soulountsi V, Iasonidou C, Kapravelos N. An contribute to pain, inflammation and post-exertional malaise in individuals
overview of management of intracranial hypertension in the intensive with myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS)? J
care unit. J Anesth. (2020) 34:741–57. doi: 10.1007/s00540-02 Transl Med. (2022) 20:40. doi: 10.1186/s12967-022-03244-7
0-02795-7 57. Alverdy JC, Krezalek MA. Collapse of the Microbiome, Emergence of the
39. Naessens DMP, de Vos J, VanBavel E, Bakker E. Blood-brain and blood- Pathobiome, and the Immunopathology of Sepsis. Crit Care Med. (2017)
cerebrospinal fluid barrier permeability in spontaneously hypertensive 45:337–47. doi: 10.1097/CCM.0000000000002172
rats. Fluids Barriers CNS. (2018) 15:26. doi: 10.1186/s12987-01 58. Otani S, Coopersmith CM. Gut integrity in critical illness. J Intensive Care.
8-0112-7 (2019) 7:17. doi: 10.1186/s40560-019-0372-6

Frontiers in Medicine | www.frontiersin.org 8 March 2022 | Volume 9 | Article 818728


Stanculescu and Bergquist Drawing on Critical Illness to Explain ME/CFS

59. Ojima M, Motooka D, Shimizu K, Gotoh K, Shintani A, Yoshiya K, et al. 79. Missailidis D, Annesley SJ, Fisher PR. Pathological mechanisms underlying
Metagenomic analysis reveals dynamic changes of whole gut microbiota in myalgic encephalomyelitis/chronic fatigue syndrome. Diagnostics. (2019)
the acute phase of intensive care unit patients. Dig Dis Sci. (2016) 61:1628– 9:80. doi: 10.20944/preprints201907.0196.v1
34. doi: 10.1007/s10620-015-4011-3 80. Anderson G, Maes M. Mitochondria and immunity in chronic
60. Mittal R, Coopersmith CM. Redefining the gut as the motor of critical illness. fatigue syndrome. Prog Neuropsychopharmacol Biol Psychiatry. (2020)
Trends Mol Med. (2014) 20:214–23. doi: 10.1016/j.molmed.2013.08.004 103:109976. doi: 10.1016/j.pnpbp.2020.109976
61. Sertaridou E, Papaioannou V, Kolios G, Pneumatikos I. Gut failure in critical 81. Shukla SK, Cook D, Meyer J, Vernon SD, Le T, Clevidence D, et al. Changes
care: old school versus new school. Ann Gastroenterol. (2015) 28:309–22. in gut and plasma microbiome following exercise challenge in Myalgic
62. Doig CJ, Sutherland LR, Sandham JD, Fick GH, Verhoef M, Meddings Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS). PLoS ONE. (2015)
JB. Increased intestinal permeability is associated with the development of 10:e0145453. doi: 10.1371/journal.pone.0145453
multiple organ dysfunction syndrome in critically ill ICU patients. Am J 82. Giloteaux L, Goodrich JK, Walters WA, Levine SM, Ley RE, Hanson
Respir Crit Care Med. (1998) 158:444–51. doi: 10.1164/ajrccm.158.2.9710092 MR. Reduced diversity and altered composition of the gut microbiome
63. Martinez EE, Fasano A, Mehta NM. Gastrointestinal function in critical in individuals with myalgic encephalomyelitis/chronic fatigue syndrome.
illness—a complex interplay between the nervous and enteroendocrine Microbiome. (2016) 4:30. doi: 10.1186/s40168-016-0171-4
systems. Pediatr Med. (2020) 3:26. doi: 10.21037/pm-20-74 83. Lakhan SE, Kirchgessner A. Gut inflammation in chronic fatigue syndrome.
64. Bischoff SC, Barbara G, Buurman W, Ockhuizen T, Schulzke JD, Serino M, et Nutr Metab. (2010) 7:79. doi: 10.1186/1743-7075-7-79
al. Intestinal permeability–a new target for disease prevention and therapy. 84. Varesi A, Deumer U-S, Ananth S, Ricevuti G. The emerging role of
BMC Gastroenterol. (2014) 14:189. doi: 10.1186/s12876-014-0189-7 gut microbiota in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome
65. Aranow J, Fink M. Determinants of intestinal barrier failure in critical illness. (ME/CFS): current evidence and potential therapeutic applications. J Clin
Br J Anaesth. (1996) 77:71–81. doi: 10.1093/bja/77.1.71 Med. (2021) 10:5077. doi: 10.3390/jcm10215077
66. Jakob SM. Clinical review: splanchnic ischaemia. Crit Care. (2002) 6:306– 85. König RS, Albrich WC, Kahlert CR, Bahr LS, Löber U, Vernazza
12. doi: 10.1186/cc1515 P, et al. the gut microbiome in Myalgic Encephalomyelitis
67. Stechmiller JK, Treloar D, Allen N. Gut dysfunction in critically ill (ME)/Chronic Fatigue Syndrome (CFS). Front Immunol. (2022)
patients: a review of the literature. Am J Crit Care. (1997) 6:204– 12:628741. doi: 10.3389/fimmu.2021.628741
9. doi: 10.4037/ajcc1997.6.3.204 86. Guo C, Che X, Briese T, Allicock O, Yates RA, Cheng A, et al.
68. van Wijck K, Lenaerts K, van Loon LJ, Peters WH, Buurman WA, Dejong Deficient butyrate-producing capacity in the gut microbiome of Myalgic
CH. Exercise-induced splanchnic hypoperfusion results in gut dysfunction in Encephalomyelitis/Chronic Fatigue Syndrome patients is associated with
healthy men. PLoS ONE. (2011) 6:e22366. doi: 10.1371/journal.pone.0022366 fatigue symptoms. [Preprint] medRxiv. (2021) Available at: https://www.
69. Fink MP, Delude RL. Epithelial barrier dysfunction: a unifying theme to medrxiv.org/content/10.1101/2021.10.27.21265575v1 (accessed February 21,
explain the pathogenesis of multiple organ dysfunction at the cellular level. 2021).
Crit Care Clin. (2005) 21:177–96. doi: 10.1016/j.ccc.2005.01.005 87. Nakatomi Y, Kuratsune H, Watanabe Y. [Neuroinflammation in the brain
70. Holland J, Carey M, Hughes N, Sweeney K, Byrne PJ, Healy M, et al. of patients with myalgic encephalomyelitis/chronic fatigue syndrome]. Brain
Intraoperative splanchnic hypoperfusion, increased intestinal permeability, Nerve. (2018) 70:19–25. doi: 10.11477/mf.1416200945
down-regulation of monocyte class II major histocompatibility complex 88. Nakatomi Y, Mizuno K, Ishii A, Wada Y, Tanaka M, Tazawa S, et al.
expression, exaggerated acute phase response, and sepsis. Am J Surg. (2005) Neuroinflammation in patients with chronic fatigue syndrome/myalgic
190:393–400. doi: 10.1016/j.amjsurg.2005.03.038 encephalomyelitis: an (1)(1)C-(R)-PK11195 PET study. J Nucl Med. (2014)
71. Meakins J, Marshall J. Multi-organ-failure syndrome. The 55:945–50. doi: 10.2967/jnumed.113.131045
gastrointestinal tract: the “motor” of MOF. Arch Surg. (1986) 89. Mueller C, Lin JC, Sheriff S, Maudsley AA, Younger JW.
121:196–208. doi: 10.1001/archsurg.1986.01400020082010 Evidence of widespread metabolite abnormalities in Myalgic
72. Deitch EA. Gut-origin sepsis: evolution of a concept. Surgeon. (2012) 10:350– encephalomyelitis/chronic fatigue syndrome: assessment with whole-
6. doi: 10.1016/j.surge.2012.03.003 brain magnetic resonance spectroscopy. Brain Imaging Behav. (2020)
73. Maes M, Coucke F, Leunis JC. Normalization of the increased translocation 14:562–72. doi: 10.1007/s11682-018-0029-4
of endotoxin from gram negative enterobacteria (leaky gut) is accompanied 90. Mu Q, Kirby J, Reilly CM, Luo XM. Leaky gut as a danger
by a remission of chronic fatigue syndrome. Neuro Endocrinol Lett. signal for autoimmune diseases. Front Immunol. (2017)
(2007) 28:739–44. 8:598. doi: 10.3389/fimmu.2017.00598
74. Morris G, Berk M, Carvalho AF, Caso JR, Sanz Y, Maes M. 91. Fasano A. Leaky gut and autoimmune diseases. Clin Rev Allergy Immunol.
The role of microbiota and intestinal permeability in the (2012) 42:71–71 doi: 10.1007/s12016-011-8291-x
pathophysiology of autoimmune and neuroimmune processes 92. Sotzny F, Blanco J, Capelli E, Castro-Marrero J, Steiner S, Murovska
with an emphasis on inflammatory bowel disease type 1 M, et al. Myalgic encephalomyelitis/chronic fatigue syndrome –
diabetes and chronic fatigue syndrome. Curr Pharm Des. (2016) evidence for an autoimmune disease. Autoimmun Rev. (2018)
22:6058–75. doi: 10.2174/1381612822666160914182822 17:601–9. doi: 10.1016/j.autrev.2018.01.009
75. Morris G, Maes M, Berk M, Puri BK. Myalgic encephalomyelitis or chronic 93. Morris G, Berk M, Galecki P, Maes M. The emerging role of autoimmunity
fatigue syndrome: how could the illness develop? Metab Brain Dis. (2019) in myalgic encephalomyelitis/chronic fatigue syndrome (ME/cfs). Mol
34:385–415. doi: 10.1007/s11011-019-0388-6 Neurobiol. (2014) 49:741–56. doi: 10.1007/s12035-013-8553-0
76. Maes M, Mihaylova I, Leunis JC. Increased serum IgA and IgM against 94. Blomberg J, Gottfries CG, Elfaitouri A, Rizwan M, Rosén A. Infection
LPS of enterobacteria in chronic fatigue syndrome (CFS): indication for elicited autoimmunity and myalgic encephalomyelitis/chronic
the involvement of gram-negative enterobacteria in the etiology of CFS and fatigue syndrome: an explanatory model. Front Immunol. (2018)
for the presence of an increased gut-intestinal permeability. J Affect Disord. 9:229. doi: 10.3389/fimmu.2018.00229
(2007) 99:237–40. doi: 10.1016/j.jad.2006.08.021 95. Boonen E, Bornstein SR, Van den Berghe G. New insights into the
77. Zhang ZT, Du XM, Ma XJ, Zong Y, Chen JK Yu CL, et al. Activation controversy of adrenal function during critical illness. Lancet Diabetes
of the NLRP3 inflammasome in lipopolysaccharide-induced mouse fatigue Endocrinol. (2015) 3:805–15. doi: 10.1016/S2213-8587(15)00224-7
and its relevance to chronic fatigue syndrome. J Neuroinflammation. (2016) 96. Van den Berghe G. On the neuroendocrinopathy of critical
13:71. doi: 10.1186/s12974-016-0539-1 illness. Perspectives for feeding and novel treatments. Am J Respir
78. Maes M, Leunis JC. Normalization of leaky gut in chronic fatigue syndrome Crit Care Med. (2016) 194:1337–48. doi: 10.1164/rccm.20160
(CFS) is accompanied by a clinical improvement: effects of age, duration 7-1516CI
of illness and the translocation of LPS from gram-negative bacteria. Neuro 97. Bergquist M, Huss F, Fredén F, Hedenstierna G, Hästbacka J, Rockwood
Endocrinol Lett. (2008) 29:902–10. AL, et al. Altered adrenal and gonadal steroids biosynthesis in patients

Frontiers in Medicine | www.frontiersin.org 9 March 2022 | Volume 9 | Article 818728


Stanculescu and Bergquist Drawing on Critical Illness to Explain ME/CFS

with burn injury. Clin Mass Spectr. (2016) 1:19–26. doi: 10.1016/j.clinms.201 118. Holtorf K. Thyroid hormone transport into cellular tissue. J Restor Med.
6.10.002 (2014) 3:53–68. doi: 10.14200/jrm.2014.3.0104
98. Marik PE. Mechanisms and clinical consequences of critical illness 119. Holtorf K. Peripheral thyroid hormone conversion and its
associated adrenal insufficiency. Curr Opin Crit Care. (2007) 13:363– impact on TSH and metabolic activity. J Restor Med. (2014)
9. doi: 10.1097/MCC.0b013e32818a6d74 3:30–52. doi: 10.14200/jrm.2014.3.0103
99. Boelen A, Kwakkel J, Thijssen-Timmer DC, Alkemade A, Fliers E, Wiersinga 120. Ruiz-Núñez B, Tarasse R, Vogelaar EF, Janneke Dijck-Brouwer DA,
WM. Simultaneous changes in central and peripheral components of the Muskiet FAJ. Higher prevalence of “low T3 syndrome” in patients with
hypothalamus-pituitary-thyroid axis in lipopolysaccharide-induced acute chronic fatigue syndrome: a case–control study. Front Endocrinol. (2018)
illness in mice. J Endocrinol. (2004) 182:315–23. doi: 10.1677/joe.0.1820315 9:97. doi: 10.3389/fendo.2018.00097
100. Joseph-Bravo P, Jaimes-Hoy L, Charli JL. Regulation of TRH neurons 121. Poteliakhoff A. Adrenocortical activity and some clinical
and energy homeostasis-related signals under stress. J Endocrinol. (2015) findings in acute and chronic fatigue. J Psychosom Res. (1981)
224:R139–59. doi: 10.1530/JOE-14-0593 25:91–5. doi: 10.1016/0022-3999(81)90095-7
101. Chatzitomaris A, Hoermann R, Midgley JE, Hering S, Urban A, Dietrich B, 122. Demitrack MA, Dale JK, Straus SE, Laue L, Listwak SJ, Kruesi MJ, et al.
et al. Thyroid allostasis–adaptive responses of thyrotropic feedback control Evidence for impaired activation of the hypothalamic-pituitary-adrenal axis
to conditions of strain, stress, and developmental programming. Front in patients with chronic fatigue syndrome. J Clin Endocrinol Metab. (1991)
Endocrinol. (2017) 8:163. doi: 10.3389/fendo.2017.00163 73:1224–34. doi: 10.1210/jcem-73-6-1224
102. Harel G, Shamoun DS, Kane JP, Magner JA, Szabo M. Prolonged effects of 123. Scott LV, Medbak S, Dinan TG. Blunted adrenocorticotropin and
tumor necrosis factor-alpha on anterior pituitary hormone release. Peptides. cortisol responses to corticotropin-releasing hormone stimulation
(1995) 16:641–5. doi: 10.1016/0196-9781(95)00019-G in chronic fatigue syndrome. Acta Psychiatr Scand. (1998)
103. Wassen FW, Moerings EP, Van Toor H, De Vrey EA, Hennemann G, 97:450–7. doi: 10.1111/j.1600-0447.1998.tb10030.x
Everts ME. Effects of interleukin-1 beta on thyrotropin secretion and thyroid 124. De Becker P, De Meirleir K, Joos E, Campine I, Van Steenberge E,
hormone uptake in cultured rat anterior pituitary cells. Endocrinology. (1996) Smitz J, et al. Dehydroepiandrosterone (DHEA) response to iv ACTH in
137:1591–8. doi: 10.1210/endo.137.5.8612490 patients with chronic fatigue syndrome. Horm Metab Res. (1999) 31:18–
104. Boonen E, Langouche L, Janssens T, Meersseman P, Vervenne H, De 21. doi: 10.1055/s-2007-978690
Samblanx E, et al. Impact of Duration of Critical Illness on the Adrenal 125. Cleare AJ, Miell J, Heap E, Sookdeo S, Young L, Malhi GS, et
Glands of Human Intensive Care Patients. J Clin Endocrinol Metab. (2014) al. Hypothalamo-pituitary-adrenal axis dysfunction in chronic fatigue
99:4214–22. doi: 10.1210/jc.2014-2429 syndrome, and the effects of low-dose hydrocortisone therapy. J Clin
105. Téblick A, Peeters B, Langouche L, Van den Berghe G. Adrenal function Endocrinol Metab. (2001) 86:3545–54. doi: 10.1210/jcem.86.8.7735
and dysfunction in critically ill patients. Nat Rev Endocrinol. (2019) 15:417– 126. Gaab J, Huster D, Peisen R, Engert V, Heitz V, Schad T, et al. Hypothalamic-
27. doi: 10.1038/s41574-019-0185-7 pituitary-adrenal axis reactivity in chronic fatigue syndrome and health
106. Weekers F, Van den Berghe G. Endocrine modifications and under psychological, physiological, and pharmacological stimulation.
interventions during critical illness. Proc Nutr Soc. (2004) Psychosom Med. (2002) 64:951–62. doi: 10.1097/00006842-200211000-00012
63:443–50. doi: 10.1079/PNS2004373 127. Jerjes WK, Cleare AJ, Wessely S, Wood PJ, Taylor NF. Diurnal patterns of
107. Baxter RC. Changes in the IGF–IGFBP axis in critical illness. Best Pract Res salivary cortisol and cortisone output in chronic fatigue syndrome. J Affect
Clin Endocrinol Metab. (2001) 15:421–34. doi: 10.1053/beem.2001.0161 Disord. (2005) 87:299–304. doi: 10.1016/j.jad.2005.03.013
108. Van den Berghe G, Wouters P, Weekers F, Mohan S, Baxter RC, Veldhuis JD, 128. Segal TY, Hindmarsh PC, Viner RM. Disturbed adrenal function in
et al. Reactivation of pituitary hormone release and metabolic improvement adolescents with chronic fatigue syndrome. J Pediatr Endocrinol Metab.
by infusion of growth hormone-releasing peptide and thyrotropin-releasing (2005) 18:295–301. doi: 10.1515/JPEM.2005.18.3.295
hormone in patients with protracted critical illness. J Clin Endocrinol Metab. 129. Van Den Eede F, Moorkens G, Hulstijn W, Van Houdenhove B, Cosyns
(1999) 84:1311–23. doi: 10.1210/jc.84.4.1311 P, Sabbe BG, et al. Combined dexamethasone/corticotropin-releasing
109. Litin SC. Mayo Clinic Family Health Book 5th Edition: Completely Revised factor test in chronic fatigue syndrome. Psychol Med. (2008) 38:963–
and Updated. Rochester, MN: Mayo Clinic Press (2018). 73. doi: 10.1017/S0033291707001444
110. Hertoghe T. Atlas of Endocrinology for Hormone Therapy. Luxembourg: 130. Van Den Eede F, Moorkens G, Van Houdenhove B, Cosyns P, Claes SJ.
International Medical Books (2010). Hypothalamic-pituitary-adrenal axis function in chronic fatigue syndrome.
111. De Bellis A, Bellastella G, Pernice V, Cirillo P, Longo M, Maio A, et al. Neuropsychobiology. (2007) 55:112–20. doi: 10.1159/000104468
Hypothalamic-Pituitary autoimmunity and related impairment of hormone 131. Papadopoulos AS, Cleare AJ. Hypothalamic-pituitary-adrenal axis
secretions in chronic fatigue syndrome. J Clin Endocrinol Metab. (2021) dysfunction in chronic fatigue syndrome. Nat Rev Endocrinol. (2011)
106:e5147–55. doi: 10.1210/clinem/dgab429 8:22–32. doi: 10.1038/nrendo.2011.153
112. Berwaerts J, Moorkens G, Abs R. Secretion of growth hormone in patients 132. Craddock TJ, Fritsch P, Rice MA Jr., del Rosario RM, Miller DB, et al.
with chronic fatigue syndrome. Growth Horm IGF Res. (1998) 8(Suppl. A role for homeostatic drive in the perpetuation of complex chronic
B):127–9. doi: 10.1016/S1096-6374(98)80036-1 illness: Gulf War Illness and chronic fatigue syndrome. PLoS ONE. (2014)
113. Moorkens G, Berwaerts J, Wynants H, Abs R. Characterization of pituitary 9:e84839. doi: 10.1371/journal.pone.0084839
function with emphasis on GH secretion in the chronic fatigue syndrome. 133. Gaab J, Engert V, Heitz V, Schad T, Schurmeyer TH, Ehlert U. Associations
Clin Endocrinol. (2000) 53:99–106. doi: 10.1046/j.1365-2265.2000.01049.x between neuroendocrine responses to the Insulin Tolerance Test and patient
114. Cleare AJ, Sookdeo SS, Jones J, O’Keane V, Miell JP. Integrity of the characteristics in chronic fatigue syndrome. J Psychosom Res. (2004) 56:419–
growth hormone/insulin-like growth factor system is maintained in patients 24. doi: 10.1016/S0022-3999(03)00625-1
with chronic fatigue syndrome. J Clin Endocrinol Metab. (2000) 85:1433– 134. Tomas C, Newton J, Watson S, A. review of hypothalamic-pituitary-
9. doi: 10.1210/jc.85.4.1433 adrenal axis function in chronic fatigue syndrome. ISRN Neurosci. (2013)
115. Teitelbaum J, Bird B. Effective Treatment of Severe Chronic Fatigue: A 2013:784520. doi: 10.1155/2013/784520
Report of a Series of 64 Patients. J Musculoskelet Pain. (1995) 3:91– 135. Di Giorgio A, Hudson M, Jerjes W, Cleare AJ. 24-hour pituitary
110. doi: 10.1300/J094v03n04_11 and adrenal hormone profiles in chronic fatigue syndrome.
116. Holtorf K. Diagnosis and treatment of Hypothalamic-Pituitary-Adrenal Psychosom Med. (2005) 67:433–40. doi: 10.1097/01.psy.0000161206.55
(HPA) axis dysfunction in patients with chronic Fatigue Syndrome 324.8a
(CFS) and Fibromyalgia (FM). J Chronic Fatigue Syndr. (2008) 14:59– 136. Pednekar DD, Amin MR, Fekri Azgomi H, Aschbacher K, Crofford LJ,
88. doi: 10.1300/J092v14n03_06 Faghih RT. Characterization of cortisol dysregulation in fibromyalgia
117. Fuite J, Vernon SD, Broderick G. Neuroendocrine and immune network re- and chronic fatigue syndromes: a state-space approach. IEEE
modeling in chronic fatigue syndrome: An exploratory analysis. Genomics. Trans Biomed Eng. (2020) 67:3163–72. doi: 10.1109/TBME.2020.2
(2008) 92:393–9. doi: 10.1016/j.ygeno.2008.08.008 978801

Frontiers in Medicine | www.frontiersin.org 10 March 2022 | Volume 9 | Article 818728


Stanculescu and Bergquist Drawing on Critical Illness to Explain ME/CFS

137. Morris G, Anderson G, Maes M. Hypothalamic-pituitary-adrenal 156. Bartalena L, Bogazzi F, Brogioni S, Grasso L, Martino E. Role of cytokines
hypofunction in Myalgic Encephalomyelitis (ME)/Chronic Fatigue in the pathogenesis of the euthyroid sick syndrome. Eur J Endocrinol. (1998)
Syndrome (CFS) as a consequence of activated immune-inflammatory 138 6:603–14. doi: 10.1530/eje.0.1380603
and oxidative and nitrosative pathways. Mol Neurobiol. (2017) 157. Afandi B, Vera R, Schussler GC, Yap MG. Concordant decreases of thyroxine
54:6806–19. doi: 10.1007/s12035-016-0170-2 and thyroxine binding protein concentrations during sepsis. Metabolism.
138. Hatziagelaki E, Adamaki M, Tsilioni I, Dimitriadis G, Theoharides TC. (2000) 49:753–4. doi: 10.1053/meta.2000.6239
Myalgic encephalomyelitis/chronic fatigue syndrome-metabolic disease or 158. Bartalena L, Robbins J. Variations in thyroid hormone transport
disturbed homeostasis due to focal inflammation in the hypothalamus? J proteins and their clinical implications. Thyroid. (1992)
Pharmacol Exp Ther. (2018) 367:155–67. doi: 10.1124/jpet.118.250845 2:237–45. doi: 10.1089/thy.1992.2.237
139. Jason LA, Porter N, Herrington J, Sorenson M, Kubow S. Kindling and 159. Mebis L, Paletta D, Debaveye Y, Ellger B, Langouche L, D’Hoore A, et al.
oxidative stress as contributors to myalgic encephalomyelitis/chronic fatigue Expression of thyroid hormone transporters during critical illness. Eur J
syndrome. J Behav Neurosci Res. (2009) 7:1–17. Endocrinol. (2009) 161:243. doi: 10.1530/EJE-09-0290
140. Morris G, Maes M. Oxidative and nitrosative stress and immune- 160. Huang SA, Mulcahey MA, Crescenzi A, Chung M, Kim BW, Barnes C, et
inflammatory pathways in patients with Myalgic Encephalomyelitis al. Transforming growth factor-beta promotes inactivation of extracellular
(ME)/Chronic Fatigue Syndrome (CFS). Curr Neuropharmacol. (2014) thyroid hormones via transcriptional stimulation of type 3 iodothyronine
12:168–85. doi: 10.2174/1570159X11666131120224653 deiodinase. Mol Endocrinol. (2005) 19:3126–36. doi: 10.1210/me.2005-0173
141. Gupta S, Aslakson E, Gurbaxani BM, Vernon SD. Inclusion of 161. Kwakkel J, Wiersinga WM, Boelen A. Interleukin-1beta modulates
the glucocorticoid receptor in a hypothalamic pituitary adrenal endogenous thyroid hormone receptor alpha gene transcription in liver cells.
axis model reveals bistability. Theor Biol Med Model. (2007) J Endocrinol. (2007) 194:257–65. doi: 10.1677/JOE-06-0177
4:8. doi: 10.1186/1742-4682-4-8 162. Rodriguez-Perez A, Palos-Paz F, Kaptein E, Visser TJ, Dominguez-Gerpe
142. Ben-Zvi A, Vernon SD, Broderick G. Model-based therapeutic correction of L, Alvarez-Escudero J, et al. Identification of molecular mechanisms
hypothalamic-pituitary-adrenal axis dysfunction. PLoS Comput Biol. (2009) related to nonthyroidal illness syndrome in skeletal muscle and adipose
5:e1000273. doi: 10.1371/journal.pcbi.1000273 tissue from patients with septic shock. Clin Endocrinol. (2008) 68:821–
143. Sedghamiz H, Morris M, Craddock TJA, Whitley D, Broderick G. High- 7. doi: 10.1111/j.1365-2265.2007.03102.x
fidelity discrete modeling of the HPA axis: a study of regulatory plasticity 163. Lado-Abeal J, Romero A, Castro-Piedras I, Rodriguez-Perez A, Alvarez-
in biology. BMC Syst Biol. (2018) 12:76. doi: 10.1186/s12918-018-0599-1 Escudero J. Thyroid hormone receptors are down-regulated in skeletal
144. Hosseinichimeh N, Rahmandad H, Wittenborn AK. Modeling the muscle of patients with non-thyroidal illness syndrome secondary to non-
hypothalamus-pituitary-adrenal axis: A review and extension. Math Biosci. septic shock. Eur J Endocrinol. (2010) 163:765–73. doi: 10.1530/EJE-10-0376
(2015) 268:52–65. doi: 10.1016/j.mbs.2015.08.004 164. Gereben B, Zavacki AM, Ribich S, Kim BW, Huang SA, Simonides WS, et
145. Craddock TJ, Del Rosario RR, Rice M, Zysman JP, Fletcher MA, al. Cellular and molecular basis of deiodinase-regulated thyroid hormone
Klimas NG, et al. Achieving remission in gulf war illness: a signaling. Endocr Rev. (2008) 29:898–938. doi: 10.1210/er.2008-0019
simulation-based approach to treatment design. PLoS ONE. (2015) 165. De Groot LJ. The non-thyroidal illness syndrome. In: Feingold KR, Anawalt
10:e0132774. doi: 10.1371/journal.pone.0132774 B, Boyce A, Chrousos G, de Herder WW, Dungan K, et al. editors. Endotext.
146. Morris MC, Cooney KE, Sedghamiz H, Abreu M, Collado F, South Dartmouth, MA: MDText.com, Inc. (2000).
Balbin EG, et al. Leveraging prior knowledge of endocrine immune 166. Dietrich JW, Landgrafe-Mende G, Wiora E, Chatzitomaris A, Klein HH,
regulation in the therapeutically relevant phenotyping of women Midgley JE, et al. Calculated parameters of thyroid homeostasis: emerging
with chronic fatigue syndrome. Clin Ther. (2019) 41:656–74 tools for differential diagnosis and clinical research. Front Endocrinol. (2016)
e4. doi: 10.1016/j.clinthera.2019.03.002 7:57. doi: 10.3389/fendo.2016.00057
147. Scott LV, Teh J, Reznek R, Martin A, Sohaib A, Dinan TG. 167. Mendoza A, Hollenberg AN. New insights into thyroid hormone action.
Small adrenal glands in chronic fatigue syndrome: a preliminary Pharmacol Ther. (2017) 173:135–45. doi: 10.1016/j.pharmthera.2017.02.012
computer tomography study. Psychoneuroendocrinology. (1999) 168. Cicatiello AG, Di Girolamo D, Dentice M. Metabolic effects of the
24:759–68. doi: 10.1016/S0306-4530(99)00028-1 intracellular regulation of thyroid hormone: old players, new concepts. Front
148. Boelen A, Platvoet-Ter Schiphorst MC, Wiersinga WM. Association Endocrinol. (2018) 9:474. doi: 10.3389/fendo.2018.00474
between serum interleukin-6 and serum 3,5,3’-triiodothyronine 169. De Groot LJ. Dangerous dogmas in medicine: the nonthyroidal
in nonthyroidal illness. J Clin Endocrinol Metab. (1993) illness syndrome. J Clin Endocrinol Metab. (1999) 84:151–
77:1695–9. doi: 10.1210/jcem.77.6.8263160 64. doi: 10.1210/jcem.84.1.5364
149. Davies PH, Black EG, Sheppard MC, Franklyn JA. Relation between 170. Donzelli R, Colligiani D, Kusmic C, Sabatini M, Lorenzini L, Accorroni A, et
serum interleukin-6 and thyroid hormone concentrations in 270 hospital al. Effect of hypothyroidism and hyperthyroidism on tissue thyroid hormone
in-patients with non-thyroidal illness. Clin Endocrinol. (1996) 44:199– concentrations in rat. Eur Thyroid J. (2016) 5:27–34. doi: 10.1159/000443523
205. doi: 10.1046/j.1365-2265.1996.668489.x 171. Plikat K, Langgartner J, Buettner R, Bollheimer LC, Woenckhaus U,
150. Warner MH, Beckett GJ. Mechanisms behind the non- Scholmerich J, et al. Frequency and outcome of patients with nonthyroidal
thyroidal illness syndrome: an update. J Endocrinol. (2010) illness syndrome in a medical intensive care unit. Metabolism. (2007) 56:239–
205:1–13. doi: 10.1677/JOE-09-0412 44. doi: 10.1016/j.metabol.2006.09.020
151. Wajner SM, Goemann IM, Bueno AL, Larsen PR, Maia AL. IL-6 promotes 172. Boelen A, Kwakkel J, Fliers E. Beyond low plasma T3: local thyroid hormone
nonthyroidal illness syndrome by blocking thyroxine activation while metabolism during inflammation and infection. Endocr Rev. (2011) 32:670–
promoting thyroid hormone inactivation in human cells. J Clin Invest. (2011) 93. doi: 10.1210/er.2011-0007
121:1834–45. doi: 10.1172/JCI44678 173. Mancini A, Di Segni C, Raimondo S, Olivieri G, Silvestrini A, Meucci E, et al.
152. Wajner SM, Maia AL. New insights toward the acute non-thyroidal illness Thyroid hormones, oxidative stress, and inflammation. Mediators Inflamm.
syndrome. Front Endocrinol. (2012) 3:8. doi: 10.3389/fendo.2012.00008 (2016) 2016:6757154. doi: 10.1155/2016/6757154
153. Carter JN, Eastman CJ, Corcoran JM, Lazarus L. Effect of 174. Balazs C, Leovey A, Szabo M, Bako G. Stimulating effect of triiodothyronine
severe, chronic illness on thyroid function. Lancet. (1974) on cell-mediated immunity. Eur J Clin Pharmacol. (1980) 17:19–
2:971–4. doi: 10.1016/S0140-6736(74)92070-4 23. doi: 10.1007/BF00561672
154. Van den Berghe G. Novel insights in the HPA-axis during critical illness. Acta 175. Pillay K. Congenital hypothyroidism and immunodeficiency: evidence for an
Clin Belg. (2014) 69:397–406. doi: 10.1179/2295333714Y.0000000093 endocrine-immune interaction. J Pediatr Endocrinol Metab. (1998) 11:757–
155. Bartalena L, Farsetti A, Flink IL, Robbins J. Effects of interleukin-6 on the 61. doi: 10.1515/JPEM.1998.11.6.757
expression of thyroid hormone-binding protein genes in cultured human 176. Klecha AJ, Genaro AM, Gorelik G, Barreiro Arcos ML, Silberman
hepatoblastoma-derived (Hep G2) cells. Mol Endocrinol. (1992) 6:935– DM, Schuman M, et al. Integrative study of hypothalamus-pituitary-
42. doi: 10.1210/mend.6.6.1323058 thyroid-immune system interaction: thyroid hormone-mediated

Frontiers in Medicine | www.frontiersin.org 11 March 2022 | Volume 9 | Article 818728


Stanculescu and Bergquist Drawing on Critical Illness to Explain ME/CFS

modulation of lymphocyte activity through the protein kinase C 194. Hornig M, Montoya JG, Klimas NG, Levine S, Felsenstein D, Bateman L, et
signaling pathway. J Endocrinol. (2006) 189:45–55. doi: 10.1677/jo al. Distinct plasma immune signatures in ME/CFS are present early in the
e.1.06137 course of illness. Sci Adv. (2015) 1:e1400121. doi: 10.1126/sciadv.1400121
177. Klein JR. The immune system as a regulator of thyroid hormone activity. Exp 195. Langouche L, Van den Berghe G. Hypothalamic-pituitary hormones during
Biol Med. (2006) 231:229–36. doi: 10.1177/153537020623100301 critical illness: a dynamic neuroendocrine response. Handb Clin Neurol.
178. Hans VH, Lenzlinger PM, Joller-Jemelka HI, Morganti-Kossmann MC, (2014) 124:115–26. doi: 10.1016/B978-0-444-59602-4.00008-3
Kossmann T. Low T3 syndrome in head-injured patients is associated 196. Boonen E, Van den Berghe G. Endocrine responses to critical illness:
with prolonged suppression of markers of cell-mediated immune novel insights and therapeutic implications. J Clin Endocrinol Metab. (2014)
response. Eur J Trauma. (2005) 31:359–68. doi: 10.1007/s00068-00 99:1569–82. doi: 10.1210/jc.2013-4115
5-2068-y 197. Deane A, Chapman MJ, Fraser RJ, Horowitz M. Bench-to-bedside review:
179. Hodkinson CF, Simpson EE, Beattie JH, O’Connor JM, Campbell DJ, Strain the gut as an endocrine organ in the critically ill. Crit Care. (2010)
JJ, et al. Preliminary evidence of immune function modulation by thyroid 14:228. doi: 10.1186/cc9039
hormones in healthy men and women aged 55-70 years. J Endocrinol. (2009) 198. Van den Berghe G, de Zegher F, Baxter RC, Veldhuis JD, Wouters P,
202:55–63. doi: 10.1677/JOE-08-0488 Schetz M, et al. Neuroendocrinology of prolonged critical illness: effects
180. Straub RH, Cutolo M, Buttgereit F, Pongratz G. Energy regulation and of exogenous thyrotropin-releasing hormone and its combination with
neuroendocrine-immune control in chronic inflammatory diseases. J Intern growth hormone secretagogues. J Clin Endocrinol Metab. (1998) 83:309–
Med. (2010) 267:543–60. doi: 10.1111/j.1365-2796.2010.02218.x 19. doi: 10.1210/jc.83.2.309
181. Jara EL, Munoz-Durango N, Llanos C, Fardella C, Gonzalez PA, 199. Mesotten D, Van den Berghe G. Changes within the growth
Bueno SM, et al. Modulating the function of the immune system hormone/insulin-like growth factor I/IGF binding protein axis during
by thyroid hormones and thyrotropin. Immunol Lett. (2017) 184:76– critical illness. Endocrinol Metab Clin North Am. (2006) 35:793–805,
83. doi: 10.1016/j.imlet.2017.02.010 ix–x. doi: 10.1016/j.ecl.2006.09.010
182. Bilal MY, Dambaeva S, Kwak-Kim J, Gilman-Sachs A, Beaman KD. A role 200. Wu R, Dong W, Ji Y, Zhou M, Marini CP, Ravikumar TS, et al.
for iodide and thyroglobulin in modulating the function of human immune Orexigenic hormone ghrelin attenuates local and remote organ
cells. Front Immunol. (2017) 8:1573. doi: 10.3389/fimmu.2017.01573 injury after intestinal ischemia-reperfusion. PLoS ONE. (2008)
183. van der Spek AH, Surovtseva OV, Jim KK, van Oudenaren A, Brouwer MC, 3:e2026. doi: 10.1371/journal.pone.0002026
Vandenbroucke-Grauls C, et al. Regulation of intracellular triiodothyronine 201. Debaveye Y, Ellger B, Mebis L, Darras VM, Van den Berghe G. Regulation
is essential for optimal macrophage function. Endocrinology. (2018) of tissue iodothyronine deiodinase activity in a model of prolonged critical
159:2241–52. doi: 10.1210/en.2018-00053 illness. Thyroid. (2008) 18:551–60. doi: 10.1089/thy.2007.0287
184. De Vito P, Incerpi S, Pedersen JZ, Luly P, Davis FB, Davis PJ. Thyroid 202. Lizcano F, Rodríguez JS. Thyroid hormone therapy modulates
hormones as modulators of immune activities at the cellular level. Thyroid. hypothalamo-pituitary-adrenal axis. Endocr J. (2011) 58:137–
(2011) 21:879–90. doi: 10.1089/thy.2010.0429 42. doi: 10.1507/endocrj.K10E-369
185. De Luca R, Davis PJ, Lin HY, Gionfra F, Percario ZA, Affabris E, et al. 203. Sánchez-Franco F, Fernández L, Fernández G, Cacicedo L. Thyroid
Thyroid hormones interaction with immune response, inflammation hormone action on ACTH secretion. Horm Metab Res. (1989) 21:550–
and non-thyroidal illness syndrome. Front Cell Dev Biol. (2020) 2. doi: 10.1055/s-2007-1009285
8:614030. doi: 10.3389/fcell.2020.614030 204. Hiroi Y, Kim HH, Ying H, Furuya F, Huang Z, Simoncini T, et al. Rapid
186. Textoris J, Mallet F. Immunosuppression and herpes viral reactivation in nongenomic actions of thyroid hormone. Proc Natl Acad Sci USA. (2006)
intensive care unit patients: one size does not fit all. Crit Care. (2017) 103:14104–9. doi: 10.1073/pnas.0601600103
21:230. doi: 10.1186/s13054-017-1803-1 205. Sharma JN, Al-Omran A, Parvathy SS. Role of nitric oxide
187. Coşkun O, Yazici E, Sahiner F, Karakaş A, Kiliç S, Tekin M, et in inflammatory diseases. Inflammopharmacology. (2007)
al. Cytomegalovirus and Epstein–Barr virus reactivation in the 15:252–9. doi: 10.1007/s10787-007-0013-x
intensive care unit. Med Klin Intensivmed Notfallmed. (2017) 206. Handa O, Stephen J, Cepinskas G. Role of endothelial nitric oxide synthase-
112:239–45. doi: 10.1007/s00063-016-0198-0 derived nitric oxide in activation and dysfunction of cerebrovascular
188. Walton AH, Muenzer JT, Rasche D, Boomer JS, Sato B, Brownstein BH, et endothelial cells during early onsets of sepsis. Am J Physiol Heart Circ Physiol.
al. Reactivation of multiple viruses in patients with sepsis. PLoS ONE. (2014) (2008) 295:H1712–9. doi: 10.1152/ajpheart.00476.2008
9:e98819. doi: 10.1371/journal.pone.0098819 207. Gluvic ZM, Obradovic MM, Sudar-Milovanovic EM, Zafirovic
189. Pall M. The NO/ONOO-cycle mechanism as the cause of chronic fatigue SS, Radak DJ, Essack MM, et al. Regulation of nitric oxide
syndrome/myalgia encephalomyelitis. In: Svoboda E, Zelenjcik K, editors. production in hypothyroidism. Biomed Pharmacother. (2020)
Chronic Fatigue Syndrome: Symptoms, Causes and Prevention. Hauppauge, 124:109881. doi: 10.1016/j.biopha.2020.109881
NY: Nova Publishers (2009). p. 27–56 208. Bertinat R, Villalobos-Labra R, Hofmann L, Blauensteiner J, Sepúlveda
190. Morris G, Maes M. Mitochondrial dysfunctions in myalgic N, Westermeier F. Decreased NO production in endothelial cells
encephalomyelitis/chronic fatigue syndrome explained by activated exposed to plasma from ME/CFS patients. Vascul Pharmacol. (2022)
immuno-inflammatory, oxidative and nitrosative stress pathways. Metab 148:106953. doi: 10.1016/j.vph.2022.106953
Brain Dis. (2014) 29:19–36. doi: 10.1007/s11011-013-9435-x 209. Lim CF, Docter R, Visser TJ, Krenning EP, Bernard B, van Toor H,
191. Armstrong CW, McGregor NR, Lewis DP, Butt HL, Gooley PR. Metabolic et al. Inhibition of thyroxine transport into cultured rat hepatocytes
profiling reveals anomalous energy metabolism and oxidative stress by serum of nonuremic critically ill patients: effects of bilirubin and
pathways in chronic fatigue syndrome patients. Metabolomics. (2015) nonesterified fatty acids. J Clin Endocrinol Metab. (1993) 76:1165–
11:1626–39. doi: 10.1007/s11306-015-0816-5 72. doi: 10.1210/jcem.76.5.8496307
192. Shungu DC, Weiduschat N, Murrough JW, Mao X, Pillemer S, Dyke 210. Vos RA, De Jong M, Bernard BF, Docter R, Krenning EP, Hennemann G.
JP, et al. Increased ventricular lactate in chronic fatigue syndrome. III Impaired thyroxine and 3,5,3’-triiodothyronine handling by rat hepatocytes
Relationships to cortical glutathione and clinical symptoms implicate in the presence of serum of patients with nonthyroidal illness. J Clin
oxidative stress in disorder pathophysiology. NMR Biomed. (2012) 25:1073– Endocrinol Metab. (1995) 80:2364–70. doi: 10.1210/jcem.80.8.7629231
87. doi: 10.1002/nbm.2772 211. Preiser JC, Ichai C, Orban JC, Groeneveld AB. Metabolic
193. Montoya JG, Holmes TH, Anderson JN, Maecker HT, Rosenberg- response to the stress of critical illness. Br J Anaesth. (2014)
Hasson Y, Valencia IJ, et al. Cytokine signature associated with 113:945–54. doi: 10.1093/bja/aeu187
disease severity in chronic fatigue syndrome patients. Proc Natl 212. McBride MA, Owen AM, Stothers CL, Hernandez A, Luan L, Burelbach
Acad Sci USA. (2017) 114:E7150–8. doi: 10.1073/pnas.17105 KR, et al. The metabolic basis of immune dysfunction following sepsis and
19114 trauma. Front Immunol. (2020) 11:1043. doi: 10.3389/fimmu.2020.01043

Frontiers in Medicine | www.frontiersin.org 12 March 2022 | Volume 9 | Article 818728


Stanculescu and Bergquist Drawing on Critical Illness to Explain ME/CFS

213. Singer M. Critical illness and flat batteries. Crit Care. (2017) may contribute to persistent symptoms. Front Microbiol. (2021)
21:309. doi: 10.1186/s13054-017-1913-9 12:698169. doi: 10.3389/fmicb.2021.698169
214. Naviaux RK, Naviaux JC Li K, Bright AT, Alaynick WA, Wang L, et al. 225. Mackay A, A. Paradigm for post-Covid-19 fatigue syndrome analogous
Metabolic features of chronic fatigue syndrome. Proc Natl Acad Sci USA. to ME/CFS. Front Neurol. (2021) 12:701419. doi: 10.3389/fneur.2021.7
(2016) 113:E5472–80. doi: 10.1073/pnas.1607571113 01419
215. Naviaux RK. Perspective: Cell danger response Biology—The new 226. Komaroff AL, Lipkin WI. Insights from myalgic encephalomyelitis/chronic
science that connects environmental health with mitochondria fatigue syndrome may help unravel the pathogenesis of
and the rising tide of chronic illness. Mitochondrion. (2020) postacute COVID-19 syndrome. Trends Mol Med. (2021)
51:40–5. doi: 10.1016/j.mito.2019.12.005 27:895–906. doi: 10.1016/j.molmed.2021.06.002
216. Naviaux RK. Metabolic features and regulation of the healing 227. Comella PH, Gonzalez-Kozlova E, Kosoy R, Charney AW, Peradejordi IF,
cycle-a new model for chronic disease pathogenesis and Chandrasekar S, et al. A Molecular network approach reveals shared cellular
treatment. Mitochondrion. (2019) 46:278–97. doi: 10.1016/j.mito.201 and molecular signatures between chronic fatigue syndrome and other
8.08.001 fatiguing illnesses. [Preprint] medRxiv. (2021). Available at: https://www.
217. Arnett SV, Clark IA. Inflammatory fatigue and sickness behaviour - lessons medrxiv.org/content/10.1101/2021.01.29.21250755v1 (accessed February 21,
for the diagnosis and management of chronic fatigue syndrome. J Affect 2021).
Disord. (2012) 141:130–42. doi: 10.1016/j.jad.2012.04.004 228. Paul BD, Lemle MD, Komaroff AL, Snyder SH. Redox imbalance
218. Greenhalgh T, Knight M, A’Court C, Buxton M, Husain L. links COVID-19 and myalgic encephalomyelitis/chronic fatigue syndrome.
Management of post-acute covid-19 in primary care. BMJ. (2020) Proc Natl Acad Sci USA. (2021) 118:e2024358118. doi: 10.1073/pnas.202
370:m3026. doi: 10.1136/bmj.m3026 4358118
219. Dani M, Dirksen A, Taraborrelli P, Torocastro M, Panagopoulos
D, Sutton R, et al. Autonomic dysfunction in ’long COVID’:
rationale, physiology and management strategies. Clin Med. (2020) Conflict of Interest: The authors declare that the research was conducted in the
21:e63–7. doi: 10.7861/clinmed.2020-0896 absence of any commercial or financial relationships that could be construed as a
220. Huang C, Huang L, Wang Y, Li X, Ren L, Gu X, et al. 6-month consequences potential conflict of interest.
of COVID-19 in patients discharged from hospital: a cohort study. Lancet.
(2021) 397:220–32. doi: 10.1016/S0140-6736(20)32656-8
Publisher’s Note: All claims expressed in this article are solely those of the authors
221. Townsend L, Dyer AH, Jones K, Dunne J, Mooney A, Gaffney F,
and do not necessarily represent those of their affiliated organizations, or those of
et al. Persistent fatigue following SARS-CoV-2 infection is common
and independent of severity of initial infection. PLoS ONE. (2020) the publisher, the editors and the reviewers. Any product that may be evaluated in
15:e0240784. doi: 10.1371/journal.pone.0240784 this article, or claim that may be made by its manufacturer, is not guaranteed or
222. Komaroff AL, Bateman L. Will COVID-19 lead to myalgic endorsed by the publisher.
encephalomyelitis/chronic fatigue syndrome? Front Med. (2021)
7:606824. doi: 10.3389/fmed.2020.606824 Copyright © 2022 Stanculescu and Bergquist. This is an open-access article
223. Wildwing T, Holt N. The neurological symptoms of COVID-19: a systematic distributed under the terms of the Creative Commons Attribution License (CC BY).
overview of systematic reviews, comparison with other neurological The use, distribution or reproduction in other forums is permitted, provided the
conditions and implications for healthcare services. Ther Adv Chronic Dis. original author(s) and the copyright owner(s) are credited and that the original
(2021) 12. doi: 10.1177/2040622320976979 publication in this journal is cited, in accordance with accepted academic practice.
224. Proal AD, VanElzakker MB. Long COVID or post-acute sequelae No use, distribution or reproduction is permitted which does not comply with these
of COVID-19 (PASC): an overview of biological factors that terms.

Frontiers in Medicine | www.frontiersin.org 13 March 2022 | Volume 9 | Article 818728

You might also like