Skip to main content

REVIEW article

Front. Med., 02 June 2023
Sec. Infectious Diseases: Pathogenesis and Therapy
Volume 10 - 2023 | https://doi.org/10.3389/fmed.2023.1187163

ME/CFS and Long COVID share similar symptoms and biological abnormalities: road map to the literature

Anthony L. Komaroff1* W. Ian Lipkin2
  • 1Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
  • 2Center for Infection and Immunity, Mailman School of Public Health, Vagelos College of Physicians and Surgeons of Columbia University, New York, NY, United States

Some patients remain unwell for months after “recovering” from acute COVID-19. They develop persistent fatigue, cognitive problems, headaches, disrupted sleep, myalgias and arthralgias, post-exertional malaise, orthostatic intolerance and other symptoms that greatly interfere with their ability to function and that can leave some people housebound and disabled. The illness (Long COVID) is similar to myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) as well as to persisting illnesses that can follow a wide variety of other infectious agents and following major traumatic injury. Together, these illnesses are projected to cost the U.S. trillions of dollars. In this review, we first compare the symptoms of ME/CFS and Long COVID, noting the considerable similarities and the few differences. We then compare in extensive detail the underlying pathophysiology of these two conditions, focusing on abnormalities of the central and autonomic nervous system, lungs, heart, vasculature, immune system, gut microbiome, energy metabolism and redox balance. This comparison highlights how strong the evidence is for each abnormality, in each illness, and helps to set priorities for future investigation. The review provides a current road map to the extensive literature on the underlying biology of both illnesses.

Introduction

Shortly after the onset of the COVID-19 pandemic, it became clear that some patients remained unwell for months to years after “recovering” from the acute infection. They suffered from fatigue, cognitive problems, headaches, disrupted sleep, myalgias and arthralgias, post-exertional malaise, orthostatic intolerance, tachyarrhythmias and gastrointestinal complaints, all of which greatly interfered with their ability to function at home and at work. In this post-COVID period, the most severely affected people were housebound and disabled. This was true even of people who had not been severely ill with acute COVID-19. The illness has been given several names, including post-acute SARS-CoV-2 sequelae (PASC), post-COVID-19 condition (PCC), post-acute COVID-19 syndrome (PACS), and “Long COVID”—the name we use in this review. We distinguish it from other post-COVID conditions in Table 1, below.

TABLE 1
www.frontiersin.org

Table 1. Post-acute sequelae of SARS-COV-2 infection (PASC).

In this review, we compare the symptoms of Long COVID and ME/CFS, noting considerable similarities and some differences. Early in the study of both illnesses, the lack of objective biomarkers led some to question whether the illnesses were “real”—whether people might be imagining or even fabricating their symptoms.

In this review we summarize the emerging evidence that, in fact, there are many underlying biological abnormalities reported in both illnesses, documented by multiple laboratories. Moreover, we show that the two illnesses share many of these underlying abnormalities, just as they share many symptoms. That is, both illnesses are “real,” and both share similar biological abnormalities.

Understanding the underlying biology of these illnesses is critically important, given the burden they are placing on all societies. The National Academy of Medicine and Centers for Disease Control and Prevention (CDC) estimate that, in the U.S., ME/CFS affects up to 2.5 million people and generates direct and indirect expenses of approximately $17–24 billion annually (1). It also has been reported in many countries around the world.

Post-COVID-19 conditions may affect 65 million people, globally (2). In the U.S., nearly 2% of the total civilian labor force is unable to work because of these illnesses, with foregone wages of $170–230 billion annually (3). Senior economists have estimated that the aggregate cost of medical care, lost productivity and disability may be $3.7 trillion over the next 5 years (4, 5). The most important, and unanswered, question regarding the economic impact of Long COVID is how long it will last. If, as with most people with ME/CFS, it lasts decades, the burden will be even greater than this projection. Because of the exceptional anticipated burden, the National Institutes of Health has dedicated over $1 billion in support of studies of the underlying biology and natural history of various post-COVID conditions.

In Tables 47 we try to efficiently summarize the findings from the large body of ME/CFS and post-COVID-19 literature. Our intention is to provide both an overview to general readers and a road map to a very large literature for investigators interested in pursuing the role of specific abnormalities in either or both illnesses. In summarizing what is known about biological abnormalities in both illnesses, we have cited both the “positive” studies that find an abnormality and the “negative” ones that do not. This allows readers to assess the strength (or weakness) of a reported finding.

As in any review, the studies we include are of varying size and quality. It is our hope that the road map we provide will help all interested readers judge for themselves how solid the evidence is for each of the underlying biological abnormalities, in each of the two illnesses, and where holes exist in our knowledge that need to be filled.

Brief summary of the illnesses

Overview of different conditions following acute COVID-19

Most people recover completely from acute COVID-19. However, others develop a variety of different post-acute sequelae of SARS-CoV-2 infection (PASC). The terminology for these different endotypes has not been standardized, in part because the detailed longitudinal studies necessary to generate the empirical data by which to define and distinguish different PASC endotypes have not yet been completed.

In Table 1, we propose three broad categories of PASC. Although we mention the first two categories for completeness, this review discusses only the third category—the condition called Long COVID. Some authorities refer to Long COVID as “PASC.” We think that is imprecise, since the tissue injury to multiple organs and the new onset of major diseases listed in Table 1 surely also are “sequelae” of COVID-19. In any event, the data summarized in Tables 37 of this review refer only to Long COVID.

The symptoms of Long COVID limit an individual’s ability to function at home or at work. The CDC states that in the “post-COVID condition” (which we take as equivalent to “Long COVID”) symptoms can be present four or more weeks after infection with SARS-CoV-2. The World Health Organization defines the condition as the continuation or development of new symptoms 3 months after the initial SARS-CoV-2 infection and lasting for at least 2 months with no other explanation.

Some evidence indicates that Long COVID is more likely to develop in: people who are sickest and have the greatest evidence of inflammation with acute COVID-19; were PCR positive; are female; and have a premorbid history of asthma, chronic obstructive pulmonary disease and depression. However, many people with Long COVID have none of these risk factors (68).

Long COVID

How frequently does Long COVID follow acute COVID-19? A wide range of values for the incidence of Long COVID following infection with SARS-CoV-2 has been reported, depending on how the investigators have defined Long COVID and how rigorously they have pursued alternative explanations for the persisting symptoms.

A meta-analysis of 57 studies involving over 250,000 people found that ongoing symptoms impairing functional mobility persisted in 43% of people with acute COVID-19 for at least 6 months after acute infection (511). A study of nearly 100,000 cases and matched never-infected controls found that over 40% of cases remained with persistent symptoms and impairment across all daily activities at 18 months (8). A study of six thousand hospitalized patients compared to uninfected matched controls found persistent symptoms in approximately 30% of cases—much more often than in uninfected controls—at least 2 years after acute infection (512). Among people with persistent, debilitating symptoms following acute COVID-19, an estimated 13–45% meet the National Academy of Medicine case definition for ME/CFS (447, 513515).

ME/CFS

Interest in ME/CFS (originally called “chronic fatigue syndrome”) surged in the mid-1980s, although a very similar illness has been described in the medical literature for several centuries. In the 19th and early 20th centuries the name “neurasthenia” was used to refer to a similar illness (516, 517). Since the 1980s, the names “chronic fatigue syndrome” and “systemic exertional intolerance disease” have been used. The illness also has several case definitions. We prefer the one proposed by the U.S. National Academy of Medicine (1), that is summarized in Table 2.

TABLE 2
www.frontiersin.org

Table 2. U.S. National Academy of Medicine case definition for ME/CFS.

Myalgic encephalomyelitis/CFS often, but not always, follows in the wake of an apparent “infectious-like” illness characterized by respiratory and gastrointestinal symptoms, fatigue, myalgias and other symptoms as well as fever and lymphadenopathy (518). This “infectious-like” illness often is little different, initially, from the common, transient infectious illnesses that most people experience throughout life. It is not standard medical practice to test for the responsible infectious agent in people with common and transient infectious illnesses. Thus, typically no testing has been done to determine the cause of the initial “infectious-like” illness that then becomes a chronic illness in subsequent months and years.

Although in this review we are describing cases of ME/CFS that occur sporadically, there also have been apparent epidemics of a similar illness described in communities (141, 519523) and in large hospitals over the past century (524). In one report, 6% of a community of 2,500 people—most young or middle-aged adults—became suddenly ill with an infectious-like illness followed by at least 5 months of a cyclic, debilitating illness characterized by fatigue, cognitive impairment (confirmed on objective testing) and pain (520). Details on symptoms, physical examination findings and laboratory test results in these studies are not sufficient to determine how similar these illnesses were to ME/CFS.

The severity of the symptoms, and the functional impairment, can range widely from one person to another. The functional impairment in people with ME/CFS may be even greater than in those with congestive heart failure and major depression (525, 526), and greater than in those with Long COVID (527). Some people remain able to fulfill their main responsibilities at work and at home, although hobbled. Others are bed-ridden or housebound, and unable to work. For most individuals with ME/CFS the symptoms are cyclic, with some relatively “good” days and frequent “bad” days. Several stressors—exercise, prolonged upright position, cognitive and emotional upset—typically produce a worsening of all of the symptoms of the illness. This condition, called post-exertional malaise, is a cardinal feature of the illness (1).

Attempts to identify a single and possibly novel infectious agent as the cause of most cases of ME/CFS have been unsuccessful. For example, claims that murine leukemia viruses cause ME/CFS have been refuted (528, 529), as have similar claims for Borna disease virus (530).

Several other conditions often are present in some people with ME/CFS: autonomic dysfunction (particularly postural orthostatic tachycardia syndrome), various rheumatologic conditions (particularly fibromyalgia), neurologic deficits (such as sensory hypersensitivity and small fiber neuropathy), secondary depression and anxiety, new or worsened allergic disorders, and endometriosis (531, 532).

Comparison of the symptoms

The symptoms reported by people with both ME/CFS and Long COVID have been integrated in a recent meta-analysis of 21 studies (17). In Table 3, we summarize the long list of symptoms shared by these two illnesses (and shared with other illnesses). The cardinal symptom of ME/CFS, post-exertional malaise, also is reported by the vast majority of people with Long COVID (533, 534). As seen in Table 3, most symptoms in the two illnesses are similar, although decreased smell and taste, rash and hair loss are frequent in Long COVID but only rarely reported by people with ME/CFS, and painful lymph nodes, chemical sensitivities and tinnitus are frequent in ME/CFS but rarely reported in Long COVID.

TABLE 3
www.frontiersin.org

Table 3. Comparison of symptoms, ME/CFS, and Long COVID.

Comparison of the underlying objective biological abnormalities

A wide range of objective abnormalities have been reported in both Long COVID and ME/CFS. Multi-organ magnetic resonance imaging (MRI) imaging of many Long COVID patients and non-symptomatic post-COVID control subjects has revealed abnormalities involving many organs up to 1 year following acute COVID-19, although it is unclear if the anatomic MRI abnormalities reflect defective organ function: correlation of MRI abnormalities with persistent symptoms was poor (535).

Probably with both illnesses, and certainly with ME/CFS, underlying biological abnormalities appear to change over time. Initially, there is activation of neuroendocrine axes and cytokine production, followed after several years by an apparent “exhaustion” of neuroendocrine and immunologic activity (260, 536).

The most compelling findings thus far have been described in the central and autonomic nervous system, immune system, infectious agents, metabolism, and the cardiopulmonary system. We will focus on those abnormalities in this review.

Central and autonomic nervous system

Table 4 summarizes the different abnormalities that have been identified in the central nervous system (CNS) and autonomic nervous system (ANS) in ME/CFS and in Long COVID. Most of them have been reported by multiple laboratories and are common to both illnesses. Often, the measured abnormality has been shown to be correlated with the presence and severity of symptoms. We comment in the text only on those abnormalities where the evidence is most extensive.

TABLE 4
www.frontiersin.org

Table 4. Neurologic/neuromuscular abnormalities.

Cognition

Psychometric testing has revealed cognitive deficits in both illnesses. In people with ME/CFS, testing typically has been performed years after the illness began: there have been few longitudinal studies of the trajectory of cognitive deficits. In contrast, because the inciting infectious agent is clear in Long COVID, and because longitudinal studies were launched shortly after the onset of the pandemic, it is clear that the initial cognitive deficits can persist for many months. One study found there may be some improvement by the end of the first year (537).

With both illnesses, the most consistently observed abnormalities have been impaired attention and information processing speed. Long-lasting objective cognitive impairment has been demonstrated even in people with mild acute COVID-19 (538, 539). One study estimated that the measured cognitive deficit was equivalent to 10 years of aging (540). Measured cognitive impairment correlated significantly with abnormalities seen with EEG and MRI studies (50, 51).

The most persuasive evidence that the persistent cognitive deficits in Long COVID reflect damage to the CNS comes from a large population-based longitudinal study in which MRI scans and cognitive testing were performed both before the pandemic and after. Compared to a matched group that did not develop COVID-19, those who developed COVID-19 (even those not requiring hospitalization) had greater reduction in gray matter thickness and in global brain size, and greater cognitive decline (541).

Neurovascular abnormalities

In both illnesses, there is a reduction in cerebral blood flow. This likely results from autonomic dysfunction (Table 4) and possibly from reduced blood volume (Table 7). It may contribute to some of the symptoms of both illnesses, including fatigue and impaired cognition; it may also reflect reduced physical activity and reduced oxygen consumption (hypometabolism).

Neuroendocrine abnormalities

The first neurological abnormalities to be documented in ME/CFS were a group of different neuroendocrine abnormalities. In particular, there appears to be down-regulation of activity in several hypothalamic-pituitary axes (Table 4) in ME/CFS, with some similar reports in Long COVID (542). These neuroendocrine changes could have bidirectional interactions with various immunologic and vascular abnormalities that also are seen in both illnesses (542).

Autonomic nervous system

Many of the common symptoms seen in people with both illnesses could reflect autonomic dysfunction. In addition, autonomic dysfunction could explain other underlying biological disturbances, such as the reported neurovascular abnormalities. The autonomic dysfunction, in turn, could be caused by other biological abnormalities seen in both illnesses, particularly antibodies directed at autonomic receptors (129) and biopsy-documented small-fiber neuropathy (72, 180182).

As summarized in Table 4, autonomic abnormalities have been well documented in both illnesses. Abnormalities of both the sympathetic and parasympathetic arms of the autonomic nervous system reflect “dyshomeostasis” (130): poor modulation of the balance between the two systems, with the imbalance favoring expression of the sympathetic system (72, 73, 131134). Objective autonomic dysfunction is common in the first 6–12 months following acute COVID-19, becoming less common by 24 months following infection, in comparison to matched comparison groups without COVID-19 (73).

Magnetic resonance imaging (MRI)

Magnetic resonance abnormalities involving both gray matter and white matter have been found in both illnesses and been shown to correlate positively with measured objective cognitive impairment (50, 51, 149, 150). Impaired responses to a variety of challenges have been revealed by functional MRI.

Immune system and infectious agents

Myalgic encephalomyelitis/CFS often follows in the wake of an “infectious-like” illness. Long COVID (by definition) follows in the wake of acute infection with SARS-CoV-2. As summarized in Table 5, a variety of immunological parameters distinguish people with ME/CFS from healthy control subjects of the same age and gender. The same is true of Long COVID: immune parameters distinguish patients from post-COVID patients who no longer have symptoms, and from non-infected controls subjects. Moreover, many of the same immune parameters distinguish patients with ME/CFS and with Long COVID from comparison/control groups.

TABLE 5
www.frontiersin.org

Table 5. Immunologic findings and infectious agents.

In addition, as summarized in Table 5, differences in the frequency of several reactivated herpesviruses, and differences in the gut microbiome, distinguish patients with these two illnesses from comparison/control groups.

Metabolic abnormalities

Beginning in the 1990s, evidence began to accumulate indicating that in people with ME/CFS who experienced a lack of “energy,” a contributing factor might be a cellular failure to generate and utilize adenosine triphosphate (ATP). As summarized in Table 6, considerable evidence has since emerged in support of that hypothesis. The ability to generate energy from multiple sources is impaired: from fatty acids, amino acids, glucose and oxygen. Moreover, similar evidence is emerging in people with Long COVID.

TABLE 6
www.frontiersin.org

Table 6. Metabolic abnormalities.

In addition to energy metabolic deficits, evidence of a systemic hypometabolic state (also manifest in the brain, Table 4), abnormalities in redox balance, and abnormalities in the kynurenine pathway have emerged in both illnesses, as summarized in Table 6. Later, we discuss briefly how abnormalities in redox balance may have bidirectional connections to abnormalities in the immune response and energy generation.

Cardiopulmonary and vascular abnormalities

Although less extensively studied than neurological, immunological, infectious or metabolic abnormalities, a growing number of cardiopulmonary abnormalities have been identified, as summarized in Table 7. The most well documented abnormalities are diminished exercise capacity on exercise testing, particularly when a second exercise test is performed 24 h after the first; reduced ventilatory efficiency; and endothelial dysfunction (particularly in Long COVID but also in ME/CFS). Some of the metabolic abnormalities noted in Table 6 are provoked by exercise, and thus there is some overlap between Tables 6, 7.

TABLE 7
www.frontiersin.org

Table 7. Cardiopulmonary and vascular abnormalities.

Discussion

Similarities and differences in symptoms

As summarized in Table 3, most of the symptoms reported with ME/CFS and Long COVID are similar. Decreased smell and taste, rash and hair loss are more likely in Long COVID than ME/CFS; this may reflect pathology induced specifically by SARS-CoV-2 (543).

Not only is a common core of symptoms shared by ME/CFS and Long COVID: these same symptoms also are also reported following multiple infectious illnesses (544) and major non-infectious injury such as post-critical illness syndrome or post-intensive care unit syndrome (545, 546), including heat stroke (547).

Similarities and differences in underlying biology

In this review we have compared what is known about the underlying biology of ME/CFS and Long COVID. Both clearly are systemic illnesses involving multiple organs and physiological systems. Long COVID is triggered by infection with SARS-CoV-2. ME/CFS often is triggered by an “infectious-like” illness. We think it is unlikely that ME/CFS is triggered by a single, novel infectious agent: more likely, it represents a dysfunctional response to infection with any of multiple agents, as recently described (544).

As summarized in this review, both illnesses share abnormalities involving the central and autonomic nervous systems, the immune system, reactivation of latent infectious agents (primarily herpesviruses), the gut microbiome, energy metabolism, a hypometabolic state, redox imbalance, and various cardiac, pulmonary and vascular abnormalities.

Many of these abnormalities bidirectionally influence each other. This creates the potential for multiple, self-reinforcing “vicious” pathophysiological cycles that could lead to persisting illness (548, 549). It also means that the precipitating event, which sets in motion those vicious cycles, may be different in one person with the illness from the precipitating event in another person.

In summarizing what is known about biological abnormalities in both illnesses, we have cited both the “positive” studies that find an abnormality and the “negative” ones that do not. These citations allow readers to assess the strength (or weakness) of a reported finding.

As shown by the tables, some reported findings are supported by robust evidence—confirmatory reports from multiple laboratories: even though there may be some dissenting reports, the preponderance of the evidence supports the finding. The tables also identify abnormalities that are not supported at this time by robust evidence and require future investigation.

Post-infection/post-injury syndrome

The similar symptoms and pathology of ME/CFS and Long COVID raise the question of whether these disorders represent just two examples of a broader illness in which symptoms occur because they are generated by a carefully orchestrated, stereotyped, multi-system response to infection and injury.

Why might such a carefully orchestrated group of symptoms be generated following infection or injury? We speculate, as have others (550552), that these symptoms are generated because they lead to metabolic reprogramming (553) as well as to behavioral changes that reduce non-essential, energy-consuming activities—thereby maximizing the amount of energy available to facilitate recovery. Fatigue, myalgia and orthostatic intolerance, for example, lead to reduced physical activity, redirecting energy stores to eradicate infection and heal tissue injury.

Such a carefully orchestrated response to vital threats exists throughout the animal kingdom. The best studied examples are hibernation and torpor in “higher” animals (554556), and the larval state of dauer in the worm C. elegans (557559).

Dauer, hibernation and torpor all involve “abnormal” innate immune responses, redox imbalance, increased glycolysis, decreased aerobic respiration, and possibly even alteration in the organisms’ microbiome. They also involve orchestration by the autonomic nervous system (and its counterparts in the nervous systems of more primitive organisms). Thus, these well-recognized “hunkering down” mechanisms all exhibit similarities to the emerging pathophysiology of ME/CFS and Long COVID, as summarized in this review.

Conclusion

The goal of this report is to provide a road map to the state of knowledge about the underlying biology of ME/CFS and Long COVID. The often-similar findings suggest that insights into each disorder will have implications for the other. They may also enhance our understanding of evolutionarily preserved biological responses that fight infection and heal injury. We urge that investigators studying the underlying biology of Long COVID take note of the robust findings in ME/CFS that have not yet been investigated in Long COVID: given the many similarities in the underlying biology of the two illnesses, it is likely that pursuing such abnormalities in Long COVID will prove instructive.

Research into the pathophysiology of these responses has the potential to lead to new strategies for reducing the morbidity of ME/CFS and Long COVID, and of similar illnesses that can follow a variety of infections and non-infectious traumatic injury.

Author contributions

AK and WL conceptualized and wrote the manuscript. Both authors contributed to the article and approved the submitted version.

Funding

The authors’ time in writing this review was supported by the US Public Health Service Grant (AI138370) to the Center for Solutions for ME/CFS at Columbia University.

Acknowledgments

We thank Jill Mazzetta for help in retrieving and cataloguing the literature.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Institute of Medicine. Beyond Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: Redefining an Illness. Washington, DC: The National Academies Press (2015).

Google Scholar

2. Davis H, McCorkell L, Vogel J, Topol E. Long COVID: major findings, mechanisms and recommendations. Nat Rev Microbiol. (2023) 21:133–46.

Google Scholar

3. Bach K. Is Long COVID Worsening the Labor Shortage?. Washington, D. C: Brookings Institution (2022).

Google Scholar

4. Cutler D, Summers L. The COVID-19 pandemic and the $16 trillion virus. JAMA. (2020) 324:1495–6.

Google Scholar

5. Cutler D. The costs of long COVID. JAMA Health Forum. (2022) 3:e221809.

Google Scholar

6. Sudre C, Murray B, Varsavsky T, Graham M, Penfold R, Bowyer R, et al. Attributes and predictors of long COVID. Nat Med. (2021) 27:626–31.

Google Scholar

7. Su Y, Yuan D, Chen D, Ng R, Wang K, Choi J, et al. Multiple early factors anticipate post-acute COVID-19 sequelae. Cell. (2022) 185:881–95.e20.

Google Scholar

8. Hastie C, Lowe D, McAuley A, Winter A, Mills N, Black C, et al. Outcomes among confirmed cases and a matched comparison group in the Long-COVID in Scotland study. Nat Commun. (2022) 13:5663.

Google Scholar

9. You J, Zhang L, Ni-Jia-Ti M, Zhang J, Hu F, Chen L, et al. Abnormal pulmonary function and residual CT abnormalities in rehabilitating COVID-19 patients after discharge. J Infect. (2020) 81:e150–2. doi: 10.1016/j.jinf.2020.06.003

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Lee M, Perl D, Nair G, Li W, Maric D, Murray H, et al. Microvascular injury in the brains of patients with Covid-19. N Engl J Med. (2021) 384:481–3.

Google Scholar

11. Stein S, Ramelli S, Grazioli A, Chung J, Singh M, Yinda C, et al. SARS-CoV-2 infection and persistence in the human body and brain at autopsy. Nature. (2022) 612:758–63. doi: 10.1038/s41586-022-05542-y

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Xie Y, Xu E, Bowe B, Al-Aly Z. Long-term cardiovascular outcomes of COVID-19. Nat Med. (2022) 28:583–90.

Google Scholar

13. Chan L, Chaudhary K, Saha A, Chauhan K, Vaid A, Zhao S, et al. AKI in hospitalized patients with COVID-19. J Am Soc Nephrol. (2021) 32:151–60.

Google Scholar

14. Xu E, Xie Y, Al-Aly Z. Long-term gastrointestinal outcomes of COVID-19. Nat Commun. (2023) 14:983.

Google Scholar

15. Al-Aly Z, Xie Y, Bowe B. High-dimensional characterization of post-acute sequelae of COVID-19. Nature. (2021) 594:259–64.

Google Scholar

16. Xie Y, Al-Aly Z. Risks and burdens of incident diabetes in long COVID: a cohort study. Lancet Diabetes Endocrinol. (2022) 10:311–21.

Google Scholar

17. Wong T, Weitzer D. Long COVID and myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS)—A systemic review and comparison of clinical presentation and symptomatology. Medicina (Kaunas). (2021) 57:418. doi: 10.3390/medicina57050418

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Sandman C, Barron J, Nackoul K, Goldstein J, Fidler F. Memory deficits associated with chronic fatigue immune dysfunction syndrome. Biol Psychiatry. (1993) 33:618–23.

Google Scholar

19. DeLuca J, Johnson S, Beldowicz D, Natelson B. Neuropsychological impairments in chronic fatigue syndrome, multiple sclerosis, and depression. J Neurol Neurosurg Psychiatry. (1995) 58:38–43.

Google Scholar

20. Marcel B, Komaroff A, Fagioli L, Kornish R, Albert M. Cognitive deficits in patients with chronic fatigue syndrome. Biol Psychiatry. (1996) 40:535–41.

Google Scholar

21. DeLuca J, Johnson S, Ellis S, Natelson B. Cognitive functioning is impaired in patients with chronic fatigue syndrome devoid of psychiatric disease. J Neurol Neurosurg Psychiatry. (1997) 62:151–5.

Google Scholar

22. Tiersky L, Johnson S, Lange G, Natelson B, DeLuca J. Neuropsychology of chronic fatigue syndrome: a critical review. J Clin Exp Neuropsychol. (1997) 19:560–86.

Google Scholar

23. Daly E, Komaroff A, Bloomingdale K, Wilson S, Albert M. Neuropsychological functioning in patients with chronic fatigue syndrome, multiple sclerosis and depression. App Neuropsychol. (2001) 8:12–22.

Google Scholar

24. Michiels V, Cluydts R. Neuropsychological functioning in chronic fatigue syndrome: a review. Acta Psychiatr Scand. (2001) 103:84–93.

Google Scholar

25. DeLuca J, Christodoulou C, Diamond B, Rosenstein, Kramer N, Natelson B. Working memory deficits in chronic fatigue syndrome: differentiating between speed and accuracy of information processing. J Int Neuropsychol Soc. (2004) 10:101–9. doi: 10.1017/S1355617704101124

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Dickson A, Toft A, O’Carroll R. Neuropsychological functioning, illness perception, mood and quality of life in chronic fatigue syndrome, autoimmune thyroid disease and healthy participants. Psychol Med. (2009) 39:1567–76. doi: 10.1017/S0033291708004960

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Cockshell S, Mathias J. Cognitive functioning in chronic fatigue syndrome: a meta-analysis. Psychol Med. (2010) 40:1253–67.

Google Scholar

28. Den Eede F, Moorkens G, Hulstijn W, Maas Y, Schrijvers D, Stevens S, et al. Psychomotor function and response inhibition in chronic fatigue syndrome. Pscyhiatry Res. (2011) 186:367–72. doi: 10.1016/j.psychres.2010.07.022

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Rasouli O, Gotaas M, Stensdotter A, Skovlund E, Landrø N, Dåstøl P, et al. Neuropsychological dysfunction in chronic fatigue syndrome and the relation between objective and subjective findings. Neuropsychology. (2019) 33:658–69.

Google Scholar

30. Murga I, Aranburu L, Gargiulo P, Gomez-Esteban J, Lafuente J. The maintained attention assessment in patients affected by myalgic encephalomyelitis/chronic fatigue syndrome: a reliable biomarker? J Transl Med. (2021) 19:494. doi: 10.1186/s12967-021-03153-1

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Aoun Sebaiti M, Hainselin M, Gounden Y, Sirbu C, Sekulic S, Lorusso L, et al. Systematic review and meta-analysis of cognitive impairment in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). Sci Rep. (2022) 12:2157.

Google Scholar

32. Fernandez-Quiros J, Lacasa-Cazcarra M, Alegre-Martin J, Sanmartin-Sentanes R, Almirall M, Launois-Obregon P, et al. The conners continuous performance test CPT3: is it a reliable marker to predict neurocognitive dysfunction in Myalgic encephalomyelitis/chronic fatigue syndrome? Front Psychol. (2023) 14:1127193. doi: 10.3389/fpsyg.2023.1127193

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Cook D, Nagelkirk P, Peckerman A, Poluri A, Mores J, Natelson B. Exercise and cognitive performance in chronic fatigue syndrome. Med Sci Sports Exerc. (2005) 37:1460–7.

Google Scholar

34. Schmaling K, DiClementi J, Cullum C, Jones J. Cognitive functioning in chronic fatigue syndrome and depression: a preliminary comparison. Psychosom Med. (1994) 56:383–8.

Google Scholar

35. Constant E, Adam S, Gillain B, Lambert M, Masquelier E, Seron X. Cognitive deficits in patients with chronic fatigue syndrope compared to those with major depressive disorder and healthy controls. Clin Neurol Neurosurg. (2011) 113:295–302.

Google Scholar

36. Lange G, Steffener J, Cook D, Bly B, Christodoulou C, Liu W, et al. Objective evidence of cognitive complaints in chronic fatigue syndrome: a BOLD fMRI study of verbal working memory. NeuroImage. (2005) 26:513–24. doi: 10.1016/j.neuroimage.2005.02.011

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Cook D, O’Connor P, Lange G, Steffener J. Functional neuroimaging correlates of mental fatigue induced by cognition among chronic fatigue syndrome patients and controls. NeuroImage. (2007) 36:108–22.

Google Scholar

38. Shan Z, Finegan K, Bhuta S, Ireland T, Staines D, Marshall-Gradisnik S, et al. Brain function characteristics of chronic fatigue syndrome: a task fMRI study. Neuroimage Clin. (2018) 19:279–86.

Google Scholar

39. Cook D, Light A, Light K, Broderick G, Shields M, Dougherty R, et al. Neural consequences of post-exertion malaise in myalgic encephalomyelitis/chronic fatigue syndrome. Brain Behav Immun. (2017) 62:87–99. doi: 10.1016/j.bbi.2017.02.009

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Santamarina-Perez P, Eiroa-Orosa F, Freniche V, Moreno-Mayos A, Alegre J, Saez N, et al. Length of illness does not predict cognitive dysfunction in chronic fatigue syndrome. Appl Neuropsychol. (2011) 18:216–22.

Google Scholar

41. Cockshell S, Mathias J. Test effort in persons with chronic fatigue syndrome when assessed using the validity indicator profile. J Clin Exp Neuropsychol. (2012) 34:679–87. doi: 10.1080/13803395.2012.668176

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Cope H, Pernet A, Kendall B, David A. Cognitive functioning and magnetic resonance imaging in chronic fatigue. Br J Psychiatry. (1995) 167:86–94.

Google Scholar

43. Mahurin R, Goldberg J, Claypoole K, Arguelles L, Ashton S, Buchwald D. Cognitive processing in monozygotic twins discordant for chronic fatigue syndrome. Neuropsychology. (2004) 18:232–9.

Google Scholar

44. Hosp J, Dressing A, Blazhenets G, Bormann T, Rau A, Schwabenland M, et al. Cognitive impairment and altered cerebral glucose metabolism in the subacute stage of COVID-19. Brain. (2021) 144:1263–76.

Google Scholar

45. Hampshire A, Chatfield D, Am M, Jolly A, Trender W, Hellyer P, et al. Multivariate profile and acute-phase correlates of cognitive deficits in a COVID-19 hospitalised cohort. EClinicalMedicine. (2022) 47:101417.

Google Scholar

46. Ollila H, Pihlaja R, Koskinen S, Tuulio-Henriksson A, Salmela V, Tiainen M, et al. Long-term cognitive functioning is impaired in ICU-treated COVID-19 patients: a comprehensive controlled neuropsychological study. Crit Care. (2022) 26:223.

Google Scholar

47. Jennings G, Monaghan A, Xue F, Duggan E, Romero-Ortuno R. Comprehensive clinical characterisation of brain fog in adults reporting long COVID symptoms. J Clin Med. (2022) 11:3440.

Google Scholar

48. Bertuccelli M, Ciringione L, Rubega M, Bisiacchi P, Masiero S, Del Felice A. Cognitive impairment in people with previous COVID-19 infection: a scoping review. Cortex. (2022) 154:212–30. doi: 10.1016/j.cortex.2022.06.002

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Azcue N, Gomez-Esteban J, Acera M, Tijero B, Fernandez T, Ayo-Mentxakatorre N, et al. Brain fog of post-COVID-19 condition and chronic fatigue syndrome, same medical disorder? J Transl Med. (2022) 20:569. doi: 10.1186/s12967-022-03764-2

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Cecchetti G, Agosta F, Canu E, Basaia S, Barbieri A, Cardamone R, et al. Cognitive, EEG, and MRI features of COVID-19 survivors: a 10-month study. J Neurol. (2022) 269:3400–12. doi: 10.1007/s00415-022-11047-5

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Andriuta D, Si-Ahmed C, Roussel M, Constans J, Makki M, Aarabi A, et al. Clinical and imaging determinants of neurocognitive disorders in post-acute COVID-19 patients with cognitive complaints. J Alzheimers Dis. (2022) 87:1239–50. doi: 10.3233/JAD-215506

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Samudyata, Oliveira AO, Malwade S, Rufino de Sousa N, Goparaju SK, Gracias J, et al. SARS-CoV-2 promotes microglial synapse elimination in human brain organoids. Mol Psychiatry. (2022) 27:3939–50. doi: 10.1038/s41380-022-01786-2

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Schwartz R, Komaroff A, Garada B, Gleit M, Doolittle T, Bates D, et al. SPECT imaging of the brain: comparison of findings in patients with chronic fatigue syndrome, AIDS dementia complex, and major unipolar depression. AJR Am J Roentgenol. (1994) 162:943–51. doi: 10.2214/ajr.162.4.8141022

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Tanaka H, Matsushima R, Tamai H, Kajimoto Y. Impaired postural cerebral hemodynamics in young patients with chronic fatigue with and without orthostatic intolerance. J Pediatr. (2002) 140:412–7.

Google Scholar

55. Schmaling K, Lewis D, Fiedelak J, Mahurin R, Buchwald D. Single-photon emission computerized tomography and neurocognitive function in patients with chronic fatigue syndrome. Psychosom Med. (2003) 65:129–36. doi: 10.1097/01.psy.0000038942.33335.9b

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Razumovsky A, DeBusk K, Calkins H, Snader S, Lucas K, Vyas P, et al. Cerebral and systemic hemodynamics changes during upright tilt in chronic fatigue syndrome. J Neuroimag. (2003) 13:57–67.

Google Scholar

57. Yoshiuchi K, Farkas J, Natelson B. Patients with chronic fatigue syndrome have reduced absolute cortical blood flow. Clin Physiol Funct Imaging. (2006) 26:83–6.

Google Scholar

58. Stewart J, Medow M, Messer Z, Baugham I, Terilli C, Ocon A. Postural neurocognitive and neuronal activated cerebral blood flow deficits in young chronic fatigue syndrome patients with postural tachycardia syndrome. Am J Physiol Heart Circ Physiol. (2012) 302:1185–94.

Google Scholar

59. Natelson B, Mao X, Stegner A, Lange G, Vu D, Blate M, et al. Multimodal and simultaneous assessments of brain and spinal fluid abnormalities in chronic fatigue syndrome and the effects of psychiatric comorbidity. J Neurol Sci. (2017) 375:411–6. doi: 10.1016/j.jns.2017.02.046

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Biswal B, Kunwar P, Natelson B. Cerebral blood flow is reduced in chronic fatigue syndrome as assessed by arterial spin labeling. J Neurol Sci. (2011) 301:9–11.

Google Scholar

61. Finkelmeyer A, He J, Maclachlan L, Blamire A, Newton J. Intracranial compliance is associated with symptoms of orthostatic intolerance in chronic fatigue syndrome. PLos One. (2018) 13:e0200068. doi: 10.1371/journal.pone.0200068

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Boissoneault J, Letzen J, Robinson M, Staud R. Cerebral blood flow and heart rate variability predict fatigue severity in patients with chronic fatigue syndrome. Brain Imaging Behav. (2018) 13:789–97. doi: 10.1007/s11682-018-9897-x

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Staud R, Boissoneault J, Craggs J, Lai S, Robinson M. Task related cerebral blood flow changes of patients with chronic fatigue syndrome: an arterial spin labeling study. Fatigue. (2018) 6:63–79.

Google Scholar

64. Li X, Julin P, Li T. Limbic perfusion is reduced in patients with myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). Tomography. (2021) 7:675–87. doi: 10.3390/tomography7040056

PubMed Abstract | CrossRef Full Text | Google Scholar

65. van Campen C, Verheugt F, Rowe P, Visser F. Cerebral blood flow is reduced in ME/CFS during head-up tilt testing even in the absence of hypotension or tachycardia: a quantitative, controlled study using Doppler echography. Clin Neurophysiol Pract. (2020) 5:50–8. doi: 10.1016/j.cnp.2020.01.003

PubMed Abstract | CrossRef Full Text | Google Scholar

66. van Campen C, Rowe P, Visser F. Cerebral blood flow is reduced in severe myalgic encephalomyelitis/chronic fatigue syndrome patients during mild orthostatic stress testing: an exploratory study at 20 degrees of head-up tilt testing. Healthcare (Basel). (2020) 8:169. doi: 10.3390/healthcare8020169

PubMed Abstract | CrossRef Full Text | Google Scholar

67. van Campen C, Visser F. Psychogenic pseudosyncope: real or imaginary? Results from a case-control study in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) patients. Medicina (Kaunas). (2022) 58:98.

Google Scholar

68. van Campen C, Rowe P, Visser F. Cerebral blood flow remains reduced after tilt testing in myalgic encephalomyelitis/chronic fatigue syndrome patients. Clin Neurophysiol Pract. (2021) 6:245–55. doi: 10.1016/j.cnp.2021.09.001

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Fischler B, D’Haenen H, Cluydts R, Michiels V, Demets K, Bossuyt A, et al. Comparison of 99mTc HMPAO SPECT scan between chronic fatigue syndrome, major depression and healthy controls: an exploratory study of clinical correlates of regional cerebral blood flow. Neuropsychobiology. (1996) 34:175–83. doi: 10.1159/000119307

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Malfliet A, Pas R, Brouns R, De Win J, Hatem S, Meeus M, et al. Cerebral blood flow and heart rate variability in chronic fatigue syndrome: a randomized cross-over study. Pain Physician. (2018) 21:E13–24.

PubMed Abstract | Google Scholar

71. Lewis D, Mayberg H, Fischer M, Goldberg J, Ashton S, Graham M, et al. Monozygotic twins discordant for chronic fatigue syndrome: regional cerebral blood flow SPECT. Radiology. (2001) 219:766–73.

Google Scholar

72. Novak P, Giannetti M, Weller E, Hamilton M, Mukerji S, Alabsi H, et al. Network autonomic analysis of post-acute sequelae of COVID-19 and postural tachycardia syndrome. Neurol Sci. (2022) 43:6627–38. doi: 10.1007/s10072-022-06423-y

PubMed Abstract | CrossRef Full Text | Google Scholar

73. van Campen C, Visser F. Long-haul COVID patients: prevalence of POTS are reduced but cerebral blood flow abnormalities remain abnormal with longer disease duration. Healthcare (Basel). (2022) 10:2105. doi: 10.3390/healthcare10102105

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Tian T, Wu J, Chen T, Li J, Yan S, Zhou Y, et al. Long-term follow-up of dynamic brain changes in patients recovered from COVID-19 without neurological manifestations. JCI Insight. (2022) 7:e155827. doi: 10.1172/jci.insight.155827

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Demitrack M, Dale J, Straus S, Laue L, Listwak S, Kruesi M, et al. Evidence for impaired activation of the hypothalamic-pituitary-adrenal axis in patients with chronic fatigue syndrome. J Clin Endocrinol Metab. (1991) 73:1224–34.

Google Scholar

76. Dinan T. Neuroendocrinology of chronic fatigue syndrome. J Chronic Fatigue Syndrome. (1996) 2:69–70.

Google Scholar

77. Cleare A. The HPA axis and the genesis of chronic fatigue syndrome. TRENDS Endocrin Metabol. (2004) 15:55–9. doi: 10.1016/j.tem.2003.12.002

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Roberts A, Wessely S, Chalder T, Papadopoulos A, Cleare A. Salivary cortisol response to awakening in chronic fatigue syndrome. Br J Psychiatry. (2004) 184:136–41.

Google Scholar

79. Jerjes W, Taylor N, Wood P, Cleare A. Enhanced feedback sensitivity to prednisolone in chronic fatigue syndrome. Psychoneuroendocrinology. (2007) 32:192–8. doi: 10.1016/j.psyneuen.2006.12.005

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Van Den Eede F, Moorkens G, Hulstijn W, van Houdenhove B, Cosyns P, Sabbe B, et al. Combined dexamethasone/corticotropin-releasing factor test in chronic fatigue syndrome. Psychol Med. (2008) 38:963–73.

Google Scholar

81. Wyller V, Vitelli V, Sulheim D, Fagermoen E, Winger A, Godang K, et al. Altered neuroendocrine control and association to clinical symptoms in adolescent chronic fatigue syndrome: a cross-sectional study. J Transl Med. (2016) 14:121.

Google Scholar

82. Morris G, Anderson G, Maes M. Hypothalamic-pituitary-adrenal hypofunction in myalgic encephalomyelitis (ME)/chronic fatigue syndrome (CFS) as a consequence of activated immune-inflammatory and oxidative and nitrosative pathways. Mol Neurobiol. (2017) 54:6806–19.

Google Scholar

83. Tak L, Cleare A, Ormel J, Manoharan A, Kok I, Wessely S, et al. Meta-analysis and meta-regression of hypothalamic-pituitary-adrenal axis activity in functional somatic disorders. Biol Psychol. (2011) 87:183–94.

Google Scholar

84. Powell D, Liossi C, Moss-Morris R, Schlotz W. Unstimulated cortisol secretory activity in everyday life and its relationship with fatigue and chronic fatigue syndrome: a systematic review and subset meta-analysis. Psychoneuroendocrinology. (2013) 38:2405–22. doi: 10.1016/j.psyneuen.2013.07.004

PubMed Abstract | CrossRef Full Text | Google Scholar

85. Meeus M, Nijs J, Van de Wauwer N, Toeback L, Truijen S. Diffuse noxious inhibitory control is delayed in chronic fatigue syndrome: an experimental study. Pain. (2008) 139:439–48. doi: 10.1016/j.pain.2008.05.018

PubMed Abstract | CrossRef Full Text | Google Scholar

86. Inder W, Prickett T, Mulder R. Normal opioid tone and hypothalamic-pituitary-adrenal axis function in chronic fatigue syndrome despite marked functional impairment. Clin Endocrinol. (2005) 62:343–8. doi: 10.1111/j.1365-2265.2005.02220.x

PubMed Abstract | CrossRef Full Text | Google Scholar

87. Urhan E, Karaca Z, Unuvar G, Gundogan K, Unluhizarci K. Investigation of pituitary functions after acute coronavirus disease 2019. Endocrine J. (2022) 69:649–58.

Google Scholar

88. Sunada N, Nakano Y, Otsuka Y, Tokumasu K, Honda H, Sakurada Y, et al. Characteristics of sleep disturbance in patients with long COVID: a retrospective observational study in Japan. J Clin Med. (2022) 11:7332. doi: 10.3390/jcm11247332

PubMed Abstract | CrossRef Full Text | Google Scholar

89. Sunada N, Honda H, Nakano Y, Yamamoto K, Tokumasu K, Sakurada Y, et al. Hormonal trends in patients suffering from long COVID symptoms. Endocrine J. (2022) 69:1173–81. doi: 10.1507/endocrj.EJ22-0093

PubMed Abstract | CrossRef Full Text | Google Scholar

90. Hamazaki K, Nishigaki T, Kuramoto N, Oh K, Konishi H. Secondary adrenal insufficiency after COVID-19 diagnosed by insulin tolerance test and corticotropin-releasing hormone test. Cureus. (2022) 14:e23021.

Google Scholar

91. Yavropoulou M, Tsokos G, Chrousos G, Sfikakis P. Protracted stress-induced hypocortisolemia may account for the clinical and immune manifestations of Long COVID. Clin Immunol. (2022) 245:109133. doi: 10.1016/j.clim.2022.109133

PubMed Abstract | CrossRef Full Text | Google Scholar

92. Clarke S, Phylactou M, Patel B, Mills E, Muzi B, Izzi-Engbeaya C, et al. Normal adrenal and thyroid function in patients who survive COVID-19 infection. J Clin Endocrinol Metab. (2021) 106:2208–20. doi: 10.1210/clinem/dgab349

PubMed Abstract | CrossRef Full Text | Google Scholar

93. Bakheit A, Behan P, Dinan T, Gray C, O’Keane V. Possible upregulation of hypothalamic 5-hydroxytryptamine receptors in patients with postviral fatigue syndrome. BMJ. (1992) 304:1010–2. doi: 10.1136/bmj.304.6833.1010

PubMed Abstract | CrossRef Full Text | Google Scholar

94. Cleare A, Bearn J, Allain T, McGregor A, Wessely S, Murray R, et al. Contrasting neuroendocrine responses in depression and chronic fatigue syndrome. J Affect Disord. (1995) 35:283–9. doi: 10.1016/0165-0327(95)00026-j

PubMed Abstract | CrossRef Full Text | Google Scholar

95. Bearn J, Allain T, Coskeran P, Munro N, Butler J, McGregor A, et al. Neuroendocrine responses to d-fenfluramine and insulin-induced hypoglycemia in chronic fatigue syndrome. Biol Psychiatry. (1995) 37:245–52. doi: 10.1016/0006-3223(94)00121-I

PubMed Abstract | CrossRef Full Text | Google Scholar

96. Yatham L, Morehouse R, Chisholm B, Haase D, MacDonald D, Marrie T. Neuroendocrine assessment of serotonin (5-HT) function in chronic fatigue syndrome. Can J Psychiatry. (1995) 40:93–6.

Google Scholar

97. Vassallo C, Feldman E, Peto T, Castell L, Sharpley A, Cowen P. Decreased tryptophan availability but normal post-synaptic 5-HT 2c receptor sensitivity in chronic fatigue syndrome. Psychol Med. (2001) 31:585–91. doi: 10.1017/s0033291701003580

PubMed Abstract | CrossRef Full Text | Google Scholar

98. Buchwald D, Umali J, Stene M. Insulin-like growth factor-I (somatomedin C) levels in chronic fatigue syndrome and fibromyalgia. J Rheumatol. (1996) 23:739–42.

Google Scholar

99. Allain T, Bearn J, Coskerman P, Jones J, Checkley A, Butler J, et al. Changes in growth hormone, insulin, insulinlike growth factors (IGFs), and IGF-binding protein-1 in chronic fatigue syndrome. Biol Psychiatry. (1997) 41:567–73. doi: 10.1016/s0006-3223(96)00074-1

PubMed Abstract | CrossRef Full Text | Google Scholar

100. Berwaerts J, Moorkens G, Abs R. Secretion of growth hormone in patients with chronic fatigue syndrome. Growth Horm IGF Res. (1998) 8:127–9.

Google Scholar

101. Moorkens G, Berwaerts J, Wynants H, Abs R. Characterization of pituitary function with emphasis on GH secretion in the chronic fatigue syndrome. Clin Endocrinol. (2000) 53:99–106. doi: 10.1046/j.1365-2265.2000.01049.x

PubMed Abstract | CrossRef Full Text | Google Scholar

102. Ruiz-Nunez B, Tarasse R, Vogelaar E, Janneke Dijck-Brouwer DA, Muskiet FAJ. Higher prevalence of “low T3 syndrome” in patients with chronic fatigue syndrome: a case-control study. Front Endocrinol. (2018) 9:97. doi: 10.3389/fendo.2018.00097

PubMed Abstract | CrossRef Full Text | Google Scholar

103. Mueller C, Lin J, Sheriff S, Maudsley A, Younger J. Evidence of widespread metabolite abnormalities in myalgic encephalomyelitis/chronic fatigue syndrome: assessment with whole-brain magnetic resonance spectroscopy. Brain Imaging Behav. (2019) 14:562–72.

Google Scholar

104. Mairal E, Barberon B, Laine N, Coulange M, Guedj E. Reversible widespread brain 18F-FDG PET hypometabolism in chronic fatigue syndrome treated by hyperbaric oxygen therapy. Eur J Nucl Med Mol Imaging. (2021) 48:1680–1.

Google Scholar

105. Guedj E, Campion J, Dudouet P, Kaphan E, Bregeon F, Tissot-Dupont H, et al. (18)F-FDG brain PET hypometabolism in patients with long COVID. Eur J Nucl Med Mol Imaging. (2021) 48:2823–33.

Google Scholar

106. Verger A, Kas A, Dudouet P, Goehringer F, Salmon-Ceron D, Guedj E. Visual interpretation of brain hypometabolism related to neurological long COVID: a French multicentric experience. Eur J Nucl Med Mol Imaging. (2022) 49:3197–202. doi: 10.1007/s00259-022-05753-5

PubMed Abstract | CrossRef Full Text | Google Scholar

107. Goehringer F, Bruyere A, Doyen M, Bevilacqua S, Charmillon A, Heyer S, et al. Brain (18)F-FDG PET imaging in outpatients with post-COVID-19 conditions: findings and associations with clinical characteristics. Eur J Nucl Med Mol Imaging. (2022) 50:1084–9.

Google Scholar

108. Ferrucci R, Cuffaro L, Capozza A, Rosci C, Maiorana N, Groppo E, et al. Brain positron emission tomography (PET) and cognitive abnormalities one year after COVID-19. J Neurol. (2023) 270:1823–34.

Google Scholar

109. Martini A, Carli G, Kiferle L, Piersanti P, Palumbo P, Morbelli S, et al. Time-dependent recovery of brain hypometabolism in neuro-COVID-19 patients. Eur J Nucl Med Mol Imaging. (2022) 50:90–102. doi: 10.1007/s00259-022-05942-2

PubMed Abstract | CrossRef Full Text | Google Scholar

110. Morand A, Campion J, Lepine A, Bosdure E, Luciani L, Cammilleri S, et al. Similar patterns of [(18)F]-FDG brain PET hypometabolism in paediatric and adult patients with long COVID: a paediatric case series. Eur J Nucl Med Mol Imaging. (2022) 49:913–20. doi: 10.1007/s00259-021-05528-4

PubMed Abstract | CrossRef Full Text | Google Scholar

111. Miskowiak K, Bech J, Henriksen A, Johnsen S, Podlekareva D, Marner L. Cerebral metabolic rate of glucose and cognitive tests in long COVID patients. Brain Sci. (2022) 13:23. doi: 10.3390/brainsci13010023

PubMed Abstract | CrossRef Full Text | Google Scholar

112. Rowe P, Bou-Holaigah I, Kan J, Calkins H. Is neurally mediated hypotension an unrecognised cause of chronic fatigue? Lancet. (1995) 345:623–4.

Google Scholar

113. Freeman R, Komaroff A. Does the chronic fatigue syndrome involve the autonomic nervous system? Am J Med. (1997) 102:357–62.

Google Scholar

114. Stewart J. Autonomic nervous system dysfunction in adolescents with postural orthostatic tachycardia syndrome and chronic fatigue syndrome is characterized by attenuated vagal baroreflex and potentiated sympathetic vasomotion. Pediatr Res. (2000) 48:218–26. doi: 10.1203/00006450-200008000-00016

PubMed Abstract | CrossRef Full Text | Google Scholar

115. Wyller V, Saul J, Amlie J, Thaulow E. Sympathetic predominance of cardiovascular regulation during mild orthostatic stress in adolescents with chronic fatigue. Clin Physiol Funct Imaging. (2007) 27:231–8.

Google Scholar

116. Wyller V, Barbieri R, Thaulow E, Saul J. Enhanced vagal withdrawal during mild orthostatic stress in adolescents with chronic fatigue. Ann Noninvasive Electrocardiol. (2008) 13:67–73. doi: 10.1111/j.1542-474X.2007.00202.x

PubMed Abstract | CrossRef Full Text | Google Scholar

117. Hoad A, Spickett G, Elliott J, Newton J. Postural orthostatic tachycardia syndrome is an under-recognized condition in chronic fatigue syndrome. QJM. (2008) 101:961–5. doi: 10.1093/qjmed/hcn123

PubMed Abstract | CrossRef Full Text | Google Scholar

118. Wyller V, Barbieri R, Saul J. Blood pressure variability and closed-loop baroreflex assessment in adolescent chronic fatigue syndrome during supine rest and orthostatic stress. Eur J Appl Physiol. (2011) 497:507. doi: 10.1007/s00421-010-1670-9

PubMed Abstract | CrossRef Full Text | Google Scholar

119. Van Cauwenbergh D, Nijs J, Kos D, Van Weijnen L, Struyf F, Meeus M. Malfunctioning of the autonomic nervous system in patients with chronic fatigue syndrome: a systematic literature review. Eur J Clin Invest. (2014) 44:516–26. doi: 10.1111/eci.12256

PubMed Abstract | CrossRef Full Text | Google Scholar

120. Barnden L, Kwiatek R, Crouch B, Burnet R, Del Fante P. Autonomic correlations with MRI are abnormal in the brainstem vasomotor centre in chronic fatigue syndrome. Neuroimage Clin. (2016) 11:530–7. doi: 10.1016/j.nicl.2016.03.017

PubMed Abstract | CrossRef Full Text | Google Scholar

121. Oosterwijck J, Marusic U, De Wandele I, Paul L, Meeus M, Moorkens G, et al. The role of autonomic function in exercise-induced endogenous analgesia: a case-control study in myalgic encephalomyelitis/chronic fatigue syndrome and healthy people. Pain Physician. (2017) 20:E389–99.

PubMed Abstract | Google Scholar

122. Nelson M, Bahl J, Buckley J, Thomson R, Davison K. Evidence of altered cardiac autonomic regulation in myalgic encephalomyelitis/chronic fatigue syndrome: a systematic review and meta-analysis. Medicine (Baltimore). (2019) 98:e17600. doi: 10.1097/MD.0000000000017600

PubMed Abstract | CrossRef Full Text | Google Scholar

123. Vuong Q, Allison J, Finkelmeyer A, Newton J, Durham J. Brain responses in CFS and TMD to autonomic challenges: an exploratory fMRI study. JDR Clin Trans Res. (2020) 5:224–32. doi: 10.1177/2380084419872135

PubMed Abstract | CrossRef Full Text | Google Scholar

124. Van Oosterwijck J, Marusic U, De Wandele I, Meeus M, Paul L, Lambrecht L, et al. Reduced parasympathetic reactivation during recovery from exercise in myalgic encephalomyelitis/chronic fatigue syndrome. J Clin Med. (2021) 10:4527.

Google Scholar

125. Natelson B, Lin J, Blate M, Khan S, Chen Y, Unger E. Physiological assessment of orthostatic intolerance in chronic fatigue syndrome. J Transl Med. (2022) 20:95.

Google Scholar

126. Vernon S, Funk S, Bateman L, Stoddard G, Hammer S, Sullivan K, et al. Orthostatic challenge causes distinctive symptomatic, hemodynamic and cognitive responses in long COVID and myalgic encephalomyelitis/chronic fatigue syndrome. Front Med. (2022) 9:917019. doi: 10.3389/fmed.2022.917019

PubMed Abstract | CrossRef Full Text | Google Scholar

127. Miwa K. Orthostatic intolerance and chronotropic incompetence in patients with myalgic encephalomyelitis or chronic fatigue syndrome. Circ Rep. (2023) 5:55–61.

Google Scholar

128. Jones J, Nicholson A, Nisenbaum R, Papanicolaou D, Solomon L, Boneva R, et al. Orthostatic instability in a population-based study of chronic fatigue syndrome. Am J Med. (2005) 118:1415.e19–e28.

Google Scholar

129. Dotan A, David P, Arnheim D, Shoenfeld Y. The autonomic aspects of the post-COVID19 syndrome. Autoimmun Rev. (2022) 21:103071.

Google Scholar

130. Goldstein D. The extended autonomic system, dyshomeostasis, and COVID-19. Clin Auton Res. (2020) 30:299–315. doi: 10.1007/s10286-020-00714-0

PubMed Abstract | CrossRef Full Text | Google Scholar

131. Barizien N, Le Guen M, Russel S, Touche P, Huang F, Vallee A. Clinical characterization of dysautonomia in long COVID-19 patients. Sci Rep. (2021) 11:14042.

Google Scholar

132. Aranyo J, Bazan V, Llados G, Dominguez M, Bisbal F, Massanella M, et al. Inappropriate sinus tachycardia in post-COVID-19 syndrome. Sci Rep. (2022) 12:298.

Google Scholar

133. Marques K, Silva C, Trindade S, Santos M, Rocha R, Vasconcelos P, et al. Reduction of cardiac autonomic modulation and increased sympathetic activity by heart rate variability in patients with long COVID. Front Cardiovasc Med. (2022) 9:862001. doi: 10.3389/fcvm.2022.862001

PubMed Abstract | CrossRef Full Text | Google Scholar

134. van Campen C, Visser F. Orthostatic intolerance in long-haul COVID after SARS-CoV-2: a case-control comparison with post-EBV and insidious-onset myalgic encephalomyelitis/chronic fatigue syndrome patients. Healthcare (Basel). (2022) 10:2058. doi: 10.3390/healthcare10102058

PubMed Abstract | CrossRef Full Text | Google Scholar

135. Novak P. Post COVID-19 syndrome associated with orthostatic cerebral hypoperfusion syndrome, small fiber neuropathy and benefit of immunotherapy: a case report. eNeurologicalSci. (2020) 21:100276. doi: 10.1016/j.ensci.2020.100276

PubMed Abstract | CrossRef Full Text | Google Scholar

136. Petracek L, Suskauer S, Vickers R, Patel N, Violand R, Swope R, et al. Adolescent and young adult ME/CFS after confirmed or probable COVID-19. Front Med. (2021) 8:668944. doi: 10.3389/fmed.2021.668944

PubMed Abstract | CrossRef Full Text | Google Scholar

137. Becker R. Autonomic dysfunction in SARS-COV-2 infection acute and long-term implications COVID-19 editor’s page series. J Thromb Thrombolysis. (2021) 52:692–707. doi: 10.1007/s11239-021-02549-6

PubMed Abstract | CrossRef Full Text | Google Scholar

138. Koren T, Yifa R, Amer M, Krot M, Boshnak N, Ben-Shaanan T, et al. Insular cortex neurons encode and retrieve specific immune responses. Cell. (2021) 184:5902–15.e17.

Google Scholar

139. Buoite Stella A, Furlanis G, Frezza N, Valentinotti R, Ajcevic M, Manganotti P. Autonomic dysfunction in post-COVID patients with and without neurological symptoms: a prospective multidomain observational study. J Neurol. (2022) 269:587–96. doi: 10.1007/s00415-021-10735-y

PubMed Abstract | CrossRef Full Text | Google Scholar

140. Steinsvik E, Hausken T, Fluge O, Mella O, Gilja O. Gastric dysmotility and gastrointestinal symptoms in myalgic encephalomyelitis/chronic fatigue syndrome. Scand J Gastroenterol. (2023). Online ahead of print. doi: 10.1080/00365521.2023.2173533

PubMed Abstract | CrossRef Full Text | Google Scholar

141. Buchwald D, Cheney P, Peterson D, Henry B, Wormsley S, Geiger A, et al. A chronic illness characterized by fatigue, neurologic and immunologic disorders, and active human herpesvirus type 6 infection. Ann Intern Med. (1992) 116:103–13. doi: 10.7326/0003-4819-116-2-103

PubMed Abstract | CrossRef Full Text | Google Scholar

142. Natelson B, Cohen J, Brassloff I, Lee HJ. A controlled study of brain magnetic resonance imaging in patients with the chronic fatigue syndrome. J Neurosurg Sci. (1993) 120:213–7.

Google Scholar

143. Schwartz R, Garada B, Komaroff A, Tice H, Gleit M, Jolesz F, et al. Detection of intracranial abnormalities in patients with chronic fatigue syndrome: comparison of MR imaging and SPECT. AJR Am J Roentgenol. (1994) 162:935–41. doi: 10.2214/ajr.162.4.8141020

PubMed Abstract | CrossRef Full Text | Google Scholar

144. Lange G, DeLuca J, Maldjian J, Lee H, Tiersky L, Natelson B. Brain MRI abnormalities exist in a subset of patients with chronic fatigue syndrome. J Neurosci. (1999) 171:3–7. doi: 10.1016/s0022-510x(99)00243-9

PubMed Abstract | CrossRef Full Text | Google Scholar

145. Barnden L, Crouch B, Kwiatek R, Burnet R, Mernone A, Chryssidis S, et al. A brain MRI study of chronic fatigue syndrome: evidence of brainstem dysfunction and altered homeostasis. NMR Biomed. (2011) 24:1302–12. doi: 10.1002/nbm.1692

PubMed Abstract | CrossRef Full Text | Google Scholar

146. Barnden L, Shan Z, Staines D, Marshall-Gradisnik S, Finegan K, Ireland T, et al. Hyperintense sensorimotor T1 spin echo MRI is associated with brainstem abnormality in chronic fatigue syndrome. Neuroimage Clin. (2018) 20:102–9. doi: 10.1016/j.nicl.2018.07.011

PubMed Abstract | CrossRef Full Text | Google Scholar

147. Cook D, Lange G, DeLuca J, Natelson B. Relationship of brain MRI abnormalities and physical functional status in chronic fatigue syndrome. Int J Neurosci. (2001) 107:1–6. doi: 10.3109/00207450109149754

PubMed Abstract | CrossRef Full Text | Google Scholar

148. Perrin R, Embleton K, Pentreath V, Jackson A. Longitudinal MRI shows no cerebral abnormality in chronic fatigue syndrome. Br J Radiol. (2010) 83:419–23.

Google Scholar

149. Benedetti F, Palladini M, Paolini M, Melloni E, Vai B, De Lorenzo R, et al. Brain correlates of depression, post-traumatic distress, and inflammatory biomarkers in COVID-19 survivors: a multimodal magnetic resonance imaging study. Brain Behav Immun Health. (2021) 18:100387. doi: 10.1016/j.bbih.2021.100387

PubMed Abstract | CrossRef Full Text | Google Scholar

150. Poletti S, Paolini M, Mazza M, Palladini M, Furlan R, Querini P, et al. Lower levels of glutathione in the anterior cingulate cortex associate with depressive symptoms and white matter hyperintensities in COVID-19 survivors. Eur Neuropsychopharmacol. (2022) 61:71–7. doi: 10.1016/j.euroneuro.2022.06.008

PubMed Abstract | CrossRef Full Text | Google Scholar

151. Heine J, Schwichtenberg K, Hartung T, Rekers S, Chien C, Boesl F, et al. Structural brain changes in patients with post-COVID fatigue: a prospective observational study. eClinicalMedicine. (2023) 58:101874. doi: 10.1016/j.eclinm.2023.101874

PubMed Abstract | CrossRef Full Text | Google Scholar

152. Zeineh M, Kang J, Atlas S, Raman M, Reiss A, Norris J, et al. Right arcuate fasciculus abnormality in chronic fatigue syndrome. Radiology. (2014) 274:517–26.

Google Scholar

153. Finkelmeyer A, He J, Maclachlan L, Watson S, Gallagher P, Newton J, et al. Grey and white matter differences in chronic fatigue syndrome - a voxel-based morphometry study. Neuroimage Clin. (2018) 17:24–30.

Google Scholar

154. Thapaliya K, Marshall-Gradisnik S, Staines D, Su J, Barnden L. Alteration of cortical volume and thickness in myalgic encephalomyelitis/chronic fatigue syndrome. Front Neurosci. (2022) 16:848730. doi: 10.3389/fnins.2022.848730

PubMed Abstract | CrossRef Full Text | Google Scholar

155. Shan Z, Kwiatek R, Burnet R, Del Fante P, Staines D, Marshall-Gradisnik S, et al. Progressive brain changes in patients with chronic fatigue syndrome: a longitudinal MRI study. J Magn Reson Imaging. (2016) 44:1301–11.

Google Scholar

156. Okada T, Tanaka M, Kuratsune H, Watanabe Y, Sadato N. Mechanisms underlying fatigue: a voxel-based morphometric study of chronic fatigue syndrome. BMC Neurol. (2004) 4:14. doi: 10.1186/1471-2377-4-14

PubMed Abstract | CrossRef Full Text | Google Scholar

157. de Lange F, Kalkman J, Bleijenberg G, Hagoort P, van der Meer J, Toni I. Gray matter volume reduction in the chronic fatigue syndrome. NeuroImage. (2005) 26:777–81.

Google Scholar

158. Putra H, Park K, Yamashita F, Nakagawa Y, Murai T. Cerebral gray matter volume correlates with fatigue and varies between desk workers and non-desk workers. Front Behav Neurosci. (2022) 16:951754. doi: 10.3389/fnbeh.2022.951754

PubMed Abstract | CrossRef Full Text | Google Scholar

159. Besteher B, Machnik M, Troll M, Toepffer A, Zerekidze A, Rocktaschel T, et al. Larger gray matter volumes in neuropsychiatric long-COVID syndrome. Psychiatry Res. (2022) 317:114836. doi: 10.1016/j.psychres.2022.114836

PubMed Abstract | CrossRef Full Text | Google Scholar

160. Hafiz R, Gandhi T, Mishra S, Prasad A, Mahajan V, Di X, et al. Higher limbic and basal ganglia volumes in surviving COVID-negative patients and the relations to fatigue. Neuroimage Rep. (2022) 2:100095.

Google Scholar

161. Rothstein T. Cortical grey matter volume depletion links to neurological sequelae in post COVID-19 “long haulers”. BMC Neurol. (2023) 23:22. doi: 10.1186/s12883-023-03049-1

PubMed Abstract | CrossRef Full Text | Google Scholar

162. Van Oosterwijck J, Nijs J, Meeus M, Lefever I, Huybrechts L, Lambrecht L, et al. Pain inhibition and postexertional malaise in myalgic encephalomyelitis/chronic fatigue syndrome: an experimental study. J Intern Med. (2010) 268:265–78.

Google Scholar

163. Meeus M, Roussel N, Truijen S, Nijs J. Reduced pressure pain thresholds in response to exercise in chronic fatigue syndrome but not in chronic low back pain: an experimental study. J Rehabil Med. (2010) 42:884–90. doi: 10.2340/16501977-0595

PubMed Abstract | CrossRef Full Text | Google Scholar

164. Meeus M, Nijs J, Huybrechts S, Truijen S. Evidence for generalized hyperalgesia in chronic fatigue syndrome. Clin Rheumatol. (2010) 29:393–8.

Google Scholar

165. Nijs J, Meeus M, Van Oosterwijck J, Ickmans K, Moorkens G, Hans G, et al. In the mind or in the brain? Scientific evidence for central sensitisation in chronic fatigue syndrome. Eur J Clin Invest. (2012) 42:203–12.

Google Scholar

166. Collin S, Nijs J, Meeus M, Polli A, Willekens B, Ickmans K. Endogenous pain facilitation rather than inhibition differs between people with chronic fatigue syndrome, multiple sclerosis, and controls: an observational study. Pain Physician. (2017) 20:E489–97.

PubMed Abstract | Google Scholar

167. Bourke J, Wodehouse T, Clark L, Constantinou E, Kidd B, Langford R, et al. Central sensitisation in chronic fatigue syndrome and fibromyalgia; a case control study. J Psychosom Res. (2021) 150:110624.

Google Scholar

168. van Campen C, Rowe P, Verheugt F, Visser F. Orthostatic stress testing in myalgic encephalomyelitis/chronic fatigue syndrome patients with or without concomitant fibromyalgia: effects on pressure pain thresholds and temporal summation. Clin Exp Rheumatol. (2021) 39 Suppl:39–47.

Google Scholar

169. Polli A, Van Oosterwijck J, Nijs J, Marusic U, De Wandele I, Paul L, et al. Relationship between exercise-induced oxidative stress changes and parasympathetic activity in chronic fatigue syndrome: an observational study in patients and healthy subjects. Clin Ther. (2019) 41:641–55.

Google Scholar

170. Vecchiet J, Cipollone F, Falasca K, Mezzetti A, Pizzigallo E, Bucciarelli T, et al. Relationship between musculoskeletal symptoms and blood markers of oxidative stress in patients with chronic fatigue syndrome. Neurosci Lett. (2003) 335:151–4.

Google Scholar

171. Al-Rawaf H, Alghadir A, Gabr S. MicroRNAs as biomarkers of pain intensity in patients with chronic fatigue syndrome. Pain Pract. (2019) 19:848–60. doi: 10.1111/papr.12817

PubMed Abstract | CrossRef Full Text | Google Scholar

172. Walitt B, Ceko M, Gracely J, Gracely R. Neuroimaging of central sensitivity syndromes: key insights from the scientific literature. Curr Rheumatol Rev. (2016) 12:55–87. doi: 10.2174/1573397112666151231111104

PubMed Abstract | CrossRef Full Text | Google Scholar

173. Pacho-Hernandez J, Fernandez-de-Las-Penas C, Fuensalida-Novo S, Jimenez-Antona C, Ortega-Santiago R, Cigaran-Mendez M. Sleep quality mediates the effect of sensitization-associated symptoms, anxiety, and depression on quality of life in individuals with post-COVID-19 pain. Brain Sci. (2022) 12:1363. doi: 10.3390/brainsci12101363

PubMed Abstract | CrossRef Full Text | Google Scholar

174. de Lange F, Kalkman J, Bleijenberg G, Hagoort P, van der Werf S, van der Meer J, et al. Neural correlates of the chronic fatigue syndrome–an fMRI study. Brain. (2004) 127:1948–57.

Google Scholar

175. Tanaka M, Sadato N, Okada T, Mizuno K, Sasabe T, Tanabe H, et al. Reduced responsiveness is an essential feature of chronic fatigue syndrome: a fMRI study. BMC Neurol. (2006) 6:9. doi: 10.1186/1471-2377-6-9

PubMed Abstract | CrossRef Full Text | Google Scholar

176. Caseras X, Mataix-Cols D, Giampietro V, Rimes K, Brammer M, Zelaya F, et al. Probing the working memory system in chronic fatigue syndrome: a functional magnetic resonance imaging study using the n-back task. Psychosom Med. (2006) 68:947–55. doi: 10.1097/01.psy.0000242770.50979.5f

PubMed Abstract | CrossRef Full Text | Google Scholar

177. Mizuno K, Kawatani J, Tajima K, Sasaki A, Yoneda T, Komi M, et al. Low putamen activity associated with poor reward sensitivity in childhood chronic fatigue syndrome. Neuroimage Clin. (2016) 12:600–6. doi: 10.1016/j.nicl.2016.09.016

PubMed Abstract | CrossRef Full Text | Google Scholar

178. Wortinger L, Endestad T, Melinder A, Oie M, Sulheim D, Fagermoen E, et al. Emotional conflict processing in adolescent chronic fatigue syndrome: a pilot study using functional magnetic resonance imaging. J Clin Exp Neuropsychol. (2017) 39:355–68. doi: 10.1080/13803395.2016.1230180

PubMed Abstract | CrossRef Full Text | Google Scholar

179. Almutairi B, Langley C, Crawley E, Thai N. Using structural and functional MRI as a neuroimaging technique to investigate chronic fatigue syndrome/myalgic encephalopathy: a systematic review. BMJ Open. (2020) 10:e031672.

Google Scholar

180. Joseph P, Arevalo C, Oliveira R, Faria-Urbina M, Felsenstein D, Oaklander A, et al. Insights from invasive cardiopulmonary exercise testing of patients with myalgic encephalomyelitis/chronic fatigue syndrome. Chest. (2021) 160:642–51.

Google Scholar

181. Oaklander A, Mills A, Kelley M, Toran L, Smith B, Dalakas M, et al. Peripheral neuropathy evaluations of patients with prolonged long COVID. Neurol Neuroimmunol Neuroinflamm. (2022) 9:e1146.

Google Scholar

182. Novak P, Mukerji S, Alabsi H, Systrom D, Marciano S, Felsenstein D, et al. Multisystem involvement in post-acute sequelae of coronavirus disease 19. Ann Neurol. (2022) 91:367–79.

Google Scholar

183. Abrams R, Simpson D, Navis A, Jette N, Zhou L, Shin S. Small fiber neuropathy associated with SARS-CoV-2 infection. Muscle Nerve. (2022) 65:440–3.

Google Scholar

184. Bitirgen G, Korkmaz C, Zamani A, Ozkagnici A, Zengin N, Ponirakis G, et al. Corneal confocal microscopy identifies corneal nerve fibre loss and increased dendritic cells in patients with long COVID. Br J Ophthalmol. (2022) 106:1635–41. doi: 10.1136/bjophthalmol-2021-319450

PubMed Abstract | CrossRef Full Text | Google Scholar

185. Duffy F, McAnulty G, McCreary M, Cuchural G, Komaroff ALEEG. spectral coherence data distinguish chronic fatigue syndrome patients from healthy controls and depressed patients - A case control study. BMC Neurol. (2011) 11:82. doi: 10.1186/1471-2377-11-82

PubMed Abstract | CrossRef Full Text | Google Scholar

186. Rubega M, Ciringione L, Bertuccelli M, Paramento M, Sparacino G, Vianello A, et al. High-density EEG sleep correlates of cognitive and affective impairment at 12-month follow-up after COVID-19. Clin Neurophysiol. (2022) 140:126–35. doi: 10.1016/j.clinph.2022.05.017

PubMed Abstract | CrossRef Full Text | Google Scholar

187. Unger E, Nisenbaum R, Moldofsky H, Cesta A, Sammut C, Reyes M, et al. Sleep assessment in a population-based study of chronic fatigue syndrome. BMC Neurol. (2004) 4:6. doi: 10.1186/1471-2377-4-6

PubMed Abstract | CrossRef Full Text | Google Scholar

188. Maksoud R, Eaton-Fitch N, Matula M, Cabanas H, Staines D, Marshall-Gradisnik S. Systematic review of sleep characteristics in myalgic encephalomyelitis/chronic fatigue syndrome. Healthcare (Basel). (2021) 9:568.

Google Scholar

189. McCarthy M. Circadian rhythm disruption in myalgic encephalomyelitis/chronic fatigue syndrome: implications for the post-acute sequelae of COVID-19. Brain Behav Immun Health. (2022) 20:100412. doi: 10.1016/j.bbih.2022.100412

PubMed Abstract | CrossRef Full Text | Google Scholar

190. Mohamed A, Andersen T, Radovic S, Del Fante P, Kwiatek R, Calhoun V, et al. Objective sleep measures in chronic fatigue syndrome patients: a systematic review and meta-analysis. Sleep Med Rev. (2023) 69:101771.

Google Scholar

191. Moura A, Oliveira D, Torres D, Tavares-Junior J, Nobrega P, Braga-Neto P, et al. Central hypersomnia and chronic insomnia: expanding the spectrum of sleep disorders in long COVID syndrome - a prospective cohort study. BMC Neurol. (2022) 22:417. doi: 10.1186/s12883-022-02940-7

PubMed Abstract | CrossRef Full Text | Google Scholar

192. Mahmoudi H, Saffari M, Movahedi M, Sanaeinasab H, Rashidi-Jahan H, Pourgholami M, et al. A mediating role for mental health in associations between COVID-19-related self-stigma, PTSD, quality of life, and insomnia among patients recovered from COVID-19. Brain Behav. (2021) 11:e02138. doi: 10.1002/brb3.2138

PubMed Abstract | CrossRef Full Text | Google Scholar

193. Alzueta E, Perrin P, Yuksel D, Ramos-Usuga D, Kiss O, Iacovides S, et al. An international study of post-COVID sleep health. Sleep Health. (2022) 8:684–90. doi: 10.1016/j.sleh.2022.06.011

PubMed Abstract | CrossRef Full Text | Google Scholar

194. Merikanto I, Dauvilliers Y, Chung F, Wing Y, De Gennaro L, Holzinger B, et al. Sleep symptoms are essential features of long-COVID - comparing healthy controls with COVID-19 cases of different severity in the international COVID sleep study (ICOSS-II). J Sleep Res. (2022):e13754. doi: 10.1111/jsr.13754

PubMed Abstract | CrossRef Full Text | Google Scholar

195. Goldstein C, Kagan D, Rizvydeen M, Warshaw S, Troost J, Burgess H. The possibility of circadian rhythm disruption in long COVID. Brain Behav Immun Health. (2022) 23:100476. doi: 10.1016/j.bbih.2022.100476

PubMed Abstract | CrossRef Full Text | Google Scholar

196. Badenoch J, Rengasamy E, Watson C, Jansen K, Chakraborty S, Sundaram R, et al. Persistent neuropsychiatric symptoms after COVID-19: a systematic review and meta-analysis. Brain Commun. (2022) 4:fcab297.

Google Scholar

197. Rutherford G, Manning P, Newton J. Understanding muscle dysfunction in chronic fatigue syndrome. J Aging Res. (2016) 2016:2497348.

Google Scholar

198. Retornaz F, Rebaudet S, Stavris C, Jammes Y. Long-term neuromuscular consequences of SARS-Cov-2 and their similarities with myalgic encephalomyelitis/chronic fatigue syndrome: results of the retrospective CoLGEM study. J Transl Med. (2022) 20:429.

Google Scholar

199. Klaver-Krol E, Hermens H, Vermeulen R, Klaver M, Luyten H, Henriquez N, et al. Chronic fatigue syndrome: abnormally fast muscle fiber conduction in the membranes of motor units at low static force load. Clin Neurophysiol. (2021) 132:967–74.

Google Scholar

200. Wirth K, Scheibenbogen C. Pathophysiology of skeletal muscle disturbances in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). J Transl Med. (2021) 19:162. doi: 10.1186/s12967-021-02833-2

PubMed Abstract | CrossRef Full Text | Google Scholar

201. Petter E, Scheibenbogen C, Linz P, Stehning C, Wirth K, Kuehne T, et al. Muscle sodium content in patients with myalgic encephalomyelitis/chronic fatigue syndrome. J Transl Med. (2022) 20:580.

Google Scholar

202. Nakatomi Y, Mizuno K, Ishii A, Wada Y, Tanaka M, Tazawa S, et al. Neuroinflammation in patients with chronic fatigue syndrome/myalgic encephalomyelitis: an11C-(R)-PK11195 PET study. J Nucl Med. (2014) 55:945–50. doi: 10.2967/jnumed.113.131045

PubMed Abstract | CrossRef Full Text | Google Scholar

203. Renz-Polster H, Tremblay M, Bienzle D, Fischer J. The pathobiology of myalgic encephalomyelitis/chronic fatigue syndrome: the case for neuroglial failure. Front Cell Neurosci. (2022) 16:888232. doi: 10.3389/fncel.2022.888232

PubMed Abstract | CrossRef Full Text | Google Scholar

204. Godlewska B, Williams S, Emir U, Chen C, Sharpley A, Goncalves A, et al. Neurochemical abnormalities in chronic fatigue syndrome: a pilot magnetic resonance spectroscopy study at 7 Tesla. Psychopharmacology (Berl). (2022) 239:163–71. doi: 10.1007/s00213-021-05986-6

PubMed Abstract | CrossRef Full Text | Google Scholar

205. Raijmakers R, Roerink M, Keijmel S, Joosten L, Netea M, van der Meer J, et al. No signs of neuroinflammation in women with chronic fatigue syndrome or Q fever fatigue syndrome using the TSPO ligand [(11)C]-PK11195. Neurol Neuroimmunol Neuroinflamm. (2022) 9:e1113.

Google Scholar

206. Kiatkittikul P, Promteangtrong C, Kunawudhi A, Siripongsatian D, Siripongboonsitti T, Ruckpanich P, et al. Abnormality pattern of f-18 FDG PET whole body with functional MRI brain in post-acute COVID-19. Nucl Med Mol Imaging. (2022) 56:29–41. doi: 10.1007/s13139-021-00730-6

PubMed Abstract | CrossRef Full Text | Google Scholar

207. Fernandez-Castaneda A, Lu P, Geraghty A, Song E, Lee M, Wood J, et al. Mild respiratory COVID can cause multi-lineage neural cell and myelin dysregulation. Cell. (2022) 185:2452–68.e16. doi: 10.1016/j.cell.2022.06.008

PubMed Abstract | CrossRef Full Text | Google Scholar

208. Klein R, Berg P. High incidence of antibodies to 5-hydroxytryptamine, gangliosides and phospholipids in patients with chronic fatigue and fibromyalgia syndrome and their relatives: evidence for a clinical entity of both disorders. Eur J Med Res. (1995) 1:21–6.

PubMed Abstract | Google Scholar

209. Tanaka S, Kuratsune H, Hidaka Y, Hakariya Y, Tatsumi K, Takano T, et al. Autoantibodies against muscarinic cholinergic receptor in chronic fatigue syndrome. Int J Mol Med. (2003) 12:225–30.

Google Scholar

210. Yamamoto S, Ouchi Y, Nakatsuka D, Tahara T, Mizuno K, Tajima S, et al. Reduction of [11C](+)3-MPB binding in brain of chronic fatigue syndrome with serum autoantibody against muscarinic cholinergic receptor. PLos One. (2012) 7:e51515. doi: 10.1371/journal.pone.0051515

PubMed Abstract | CrossRef Full Text | Google Scholar

211. Loebel M, Grabowski P, Heidecke H, Bauer S, Hanitsch L, Wittke K, et al. Antibodies to beta adrenergic and muscarinic cholinergic receptors in patients with chronic fatigue syndrome. Brain Behav Immun. (2016) 52:32–9.

Google Scholar

212. Sotzny F, Blanco J, Capelli E, Castro-Marrero J, Steiner S, Murovska M, et al. Myalgic encephalomyelitis/chronic fatigue syndrome – evidence for an autoimmune disease. Autoimmun Rev. (2018) 17:601–9.

Google Scholar

213. Shoenfeld Y, Ryabkova V, Scheibenbogen C, Brinth L, Martinez-Lavin M, Ikeda S, et al. Complex syndromes of chronic pain, fatigue and cognitive impairment linked to autoimmune dysautonomia and small fiber neuropathy. Clin Immunol. (2020) 214:108384. doi: 10.1016/j.clim.2020.108384

PubMed Abstract | CrossRef Full Text | Google Scholar

214. Fujii H, Sato W, Kimura Y, Matsuda H, Ota M, Maikusa N, et al. Altered structural brain networks related to adrenergic/muscarinic receptor autoantibodies in chronic fatigue syndrome. J Neuroimaging. (2020) 30:822–7. doi: 10.1111/jon.12751

PubMed Abstract | CrossRef Full Text | Google Scholar

215. Wirth K, Scheibenbogen C. A unifying hypothesis of the pathophysiology of myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS): recognitions from the finding of autoantibodies against ss2-adrenergic receptors. Autoimmun Rev. (2020) 19:102527. doi: 10.1016/j.autrev.2020.102527

PubMed Abstract | CrossRef Full Text | Google Scholar

216. Szklarski M, Freitag H, Lorenz S, Becker S, Sotzny F, Bauer S, et al. Delineating the association between soluble CD26 and autoantibodies against G-protein coupled receptors, immunological and cardiovascular parameters identifies distinct patterns in post-infectious vs. non-infection-triggered myalgic encephalomyelitis/chronic fatigue syndrome. Front Immunol. (2021) 12:644548. doi: 10.3389/fimmu.2021.644548

PubMed Abstract | CrossRef Full Text | Google Scholar

217. De Bellis A, Bellastella G, Pernice V, Cirillo P, Longo M, Maio A, et al. Hypothalamic-pituitary autoimmunity and related impairment of hormone secretions in chronic fatigue syndrome. J Clin Endocrinol Metab. (2021) 106:e5147–55. doi: 10.1210/clinem/dgab429

PubMed Abstract | CrossRef Full Text | Google Scholar

218. Freitag H, Szklarski M, Lorenz S, Sotzny F, Bauer S, Philippe A, et al. Autoantibodies to vasoregulative G-protein-coupled receptors correlate with symptom severity, autonomic dysfunction and disability in myalgic encephalomyelitis/chronic fatigue syndrome. J Clin Med. (2021) 10:3675. doi: 10.3390/jcm10163675

PubMed Abstract | CrossRef Full Text | Google Scholar

219. Szewczykowski C, Mardin C, Lucio M, Wallukat G, Hoffmanns J, Schroder T, et al. Long COVID: association of functional autoantibodies against G-protein-coupled receptors with an impaired retinal microcirculation. Int J Mol Sci. (2022) 23:7209. doi: 10.3390/ijms23137209

PubMed Abstract | CrossRef Full Text | Google Scholar

220. Kim B, Namkoong K, Kim J, Lee S, Yoon K, Choi M, et al. Altered resting-state functional connectivity in women with chronic fatigue syndrome. Psychiatry Res. (2015) 234:292–7.

Google Scholar

221. Wortinger L, Endestad T, Melinder A, Oie M, Sevenius A, Bruun Wyller V. Aberrant resting-state functional connectivity in the salience network of adolescent chronic fatigue syndrome. PLos One. (2016) 11:e0159351. doi: 10.1371/journal.pone.0159351

PubMed Abstract | CrossRef Full Text | Google Scholar

222. Gay C, Robinson M, Lai S, O’Shea A, Craggs J, Price D, et al. Abnormal resting-state functional connectivity in patients with chronic fatigue syndrome: results of seed and data-driven analyses. Brain Connect. (2016) 6:48–56. doi: 10.1089/brain.2015.0366

PubMed Abstract | CrossRef Full Text | Google Scholar

223. Zinn M, Zinn M, Jason L. Intrinsic functional hypoconnectivity in core neurocognitive networks suggests central nervous system pathology in patients with myalgic encephalomyelitis: a pilot study. Appl Psychophysiol Biofeedback. (2016) 41:283–300. doi: 10.1007/s10484-016-9331-3

PubMed Abstract | CrossRef Full Text | Google Scholar

224. Boissoneault J, Letzen J, Lai S, O’Shea A, Craggs J, Robinson M, et al. Abnormal resting state functional connectivity in patients with chronic fatigue syndrome: an arterial spin-labeling fMRI study. Magn Reson Imaging. (2016) 34:603–8.

Google Scholar

225. Wortinger L, Glenne Oie M, Endestad T, Bruun Wyller V. Altered right anterior insular connectivity and loss of associated functions in adolescent chronic fatigue syndrome. PLos One. (2017) 12:e0184325. doi: 10.1371/journal.pone.0184325

PubMed Abstract | CrossRef Full Text | Google Scholar

226. Shan Z, Finegan K, Bhuta S, Ireland T, Staines D, Marshall-Gradisnik S, et al. Decreased connectivity and increased blood oxygenation level dependent complexity in the default mode network in individuals with chronic fatigue syndrome. Brain Connect. (2018) 8:33–9.

Google Scholar

227. Barnden L, Shan Z, Staines D, Marshall-Gradisnik S, Finegan K, Ireland T, et al. Intra brainstem connectivity is impaired in chronic fatigue syndrome. Neuroimage Clin. (2019) 24:102045. doi: 10.1016/j.nicl.2019.102045

PubMed Abstract | CrossRef Full Text | Google Scholar

228. Zinn M, Jason L. Cortical autonomic network connectivity predicts symptoms in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). Int J Psychophysiol. (2021) 170:89–101.

Google Scholar

229. Nelson T, Zhang L, Guo H, Nacul L, Song X. Brainstem abnormalities in myalgic encephalomyelitis/chronic fatigue syndrome: a scoping review and evaluation of magnetic resonance imaging findings. Front Neurol. (2021) 12:769511. doi: 10.3389/fneur.2021.769511

PubMed Abstract | CrossRef Full Text | Google Scholar

230. Josev E, Malpas C, Seal M, Scheinberg A, Lubitz L, Rowe K, et al. Resting-state functional connectivity, cognition, and fatigue in response to cognitive exertion: a novel study in adolescents with chronic fatigue syndrome. Brain Imaging Behav. (2019) 14:1815–30. doi: 10.1007/s11682-019-00119-2

PubMed Abstract | CrossRef Full Text | Google Scholar

231. Baraniuk J, Casado B, Maibach H, Clauw D, Pannell L, Hess S. A chronic fatigue syndrome-related proteome in human cerebrospinal fluid. BMC Neurol. (2005) 5:22. doi: 10.1186/1471-2377-5-22

PubMed Abstract | CrossRef Full Text | Google Scholar

232. Schutzer S, Angel T, Liu T, Schepmoes A, Clauss T, Adkins J, et al. Distinct cerebrospinal fluid proteomes differentiate post-treatment lyme disease from chronic fatigue syndrome. PLos One. (2011) 6:e17287. doi: 10.1371/journal.pone.0017287

PubMed Abstract | CrossRef Full Text | Google Scholar

233. Jarius S, Pache F, Kortvelyessy P, Jelcic I, Stettner M, Franciotta D, et al. Cerebrospinal fluid findings in COVID-19: a multicenter study of 150 lumbar punctures in 127 patients. J Neuroinflamm. (2022) 19:19. doi: 10.1186/s12974-021-02339-0

PubMed Abstract | CrossRef Full Text | Google Scholar

234. Schweitzer F, Goereci Y, Franke C, Silling S, Bosl F, Maier F, et al. Cerebrospinal fluid analysis post-COVID-19 is not suggestive of persistent central nervous system infection. Ann Neurol. (2022) 91:150–7.

Google Scholar

235. Du Preez S, Eaton-Fitch N, Cabanas H, Staines D, Marshall-Gradisnik S. Characterization of IL-2 stimulation and TRPM7 pharmacomodulation in NK cell cytotoxicity and channel co-localization with PIP2 in myalgic encephalomyelitis/chronic fatigue syndrome patients. Int J Environ Res Public Health. (2021) 18:11879. doi: 10.3390/ijerph182211879

PubMed Abstract | CrossRef Full Text | Google Scholar

236. Du Preez S, Cabanas H, Staines D, Marshall-Gradisnik S. Potential implications of mammalian transient receptor potential melastatin 7 in the pathophysiology of myalgic encephalomyelitis/chronic fatigue syndrome: a review. Int J Environ Res Public Health. (2021) 18:10708. doi: 10.3390/ijerph182010708

PubMed Abstract | CrossRef Full Text | Google Scholar

237. Sasso E, Muraki K, Eaton-Fitch N, Smith P, Lesslar O, Deed G, et al. Transient receptor potential melastatin 3 dysfunction in post COVID-19 condition and myalgic encephalomyelitis/chronic fatigue syndrome patients. Mol Med. (2022) 28:98. doi: 10.1186/s10020-022-00528-y

PubMed Abstract | CrossRef Full Text | Google Scholar

238. Baraniuk J. Review of the midbrain ascending arousal network nuclei and implications for myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS), Gulf War Illness (GWI) and postexertional malaise (PEM). Brain Sci. (2022) 12:132. doi: 10.3390/brainsci12020132

PubMed Abstract | CrossRef Full Text | Google Scholar

239. Thapaliya K, Marshall-Gradisnik S, Barth M, Eaton-Fitch N, Barnden L. Brainstem volume changes in myalgic encephalomyelitis/chronic fatigue syndrome and long COVID patients. Front Neurosci. (2023) 17:1125208. doi: 10.3389/fnins.2023.1125208

PubMed Abstract | CrossRef Full Text | Google Scholar

240. Yong S. Persistent brainstem dysfunction in long-COVID: a hypothesis. ACS Chem Neurosci. (2021) 12:573–80.

Google Scholar

241. Peluso M, Sans H, Forman C, Nylander A, Ho H, Lu S, et al. Plasma markers of neurologic injury and inflammation in people with self-reported neurologic postacute sequelae of SARS-CoV-2 infection. Neurol Neuroimmunol Neuroinflamm. (2022) 9:e200003. doi: 10.1212/NXI.0000000000200003

PubMed Abstract | CrossRef Full Text | Google Scholar

242. Caligiuri M, Murray C, Buchwald D, Levine H, Cheney P, Peterson D, et al. Phenotypic and functional deficiency of natural killer cells in patients with chronic fatigue syndrome. J Immunol. (1987) 139:3306–13.

Google Scholar

243. Klimas N, Salvato F, Morgan R, Fletcher M. Immunologic abnormalities in chronic fatigue syndrome. J Clin Microbiol. (1990) 28:1403–10.

Google Scholar

244. Aoki T, Miyakoshi H, Usuda Y, Herberman R. Low NK syndrome and its relationship to chronic fatigue syndrome. Clin Immunol Immunopathol. (1993) 69:253–65.

Google Scholar

245. Vojdani A, Thrasher J. Cellular and humoral immune abnormalities in Gulf War veterans. Environ Health Perspect. (2004) 112:840–6.

Google Scholar

246. Brenu E, van Driel M, Straines D, Ashton K, Hardcastle S, Keane J, et al. Longitudinal investigation of natural killer cells and cytokines in chronic fatigue syndrome/myalgic encephalomyelitis. J Transl Med. (2012) 10:88. doi: 10.1186/1479-5876-10-88

PubMed Abstract | CrossRef Full Text | Google Scholar

247. Brenu E, Huth T, Hardcastle S, Fuller K, Kaur M, Johnston S, et al. Role of adaptive and innate immune cells in chronic fatigue syndrome/myalgic encephalomyelitis. Int Immunol. (2014) 26:233–42.

Google Scholar

248. Hardcastle S, Brenu E, Johnston S, Nguyen T, Huth T, Kaur M, et al. Analysis of the relationship between immune dysfunction and symptom severity in patients with chronic fatigue syndrome/myalgic encephalomyelitis (CFS/ME). J Clin Cell Immunol. (2014) 5:1000190.

Google Scholar

249. Chacko A, Staines D, Johnston S, Marshall-Gradisnik S. Dysregulation of protein kinase gene expression in NK cells from chronic fatigue syndrome/myalgic encephalomyelitis patients. Gene Regul Syst Bio. (2016) 10:85–93.

Google Scholar

250. Huth T, Brenu E, Ramos S, Nguyen T, Broadley S, Staines D, et al. Pilot study of natural killer cells in chronic fatigue syndrome/myalgic encephalomyelitis and multiple sclerosis. Scand J Immunol. (2016) 83:44–51.

Google Scholar

251. Nguyen T, Johnston S, Clarke L, Smith P, Staines D, Marshall-Gradisnik S. Impaired calcium mobilization in natural killer cells from chronic fatigue syndrome/myalgic encephalomyelitis patients is associated with transient receptor potential melastatin 3 ion channels. Clin Exp Immunol. (2017) 187:284–93.

Google Scholar

252. Rivas J, Palencia T, Fernandez G, Garcia M. Association of T and NK cell phenotype with the diagnosis of myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). Front Immunol. (2018) 9:1028. doi: 10.3389/fimmu.2018.01028

PubMed Abstract | CrossRef Full Text | Google Scholar

253. Nguyen T, Staines D, Johnston S, Marshall-Gradisnik S. Reduced glycolytic reserve in isolated natural killer cells from myalgic encephalomyelitis/chronic fatigue syndrome patients: a preliminary investigation. Asian Pac J Allergy Immunol. (2019) 37:102–8.

Google Scholar

254. Eaton-Fitch N, du Preez S, Cabanas H, Staines D, Marshall-Gradisnik S. A systematic review of natural killer cells profile and cytotoxic function in myalgic encephalomyelitis/chronic fatigue syndrome. Syst Rev. (2019) 8:279.

Google Scholar

255. Sung A, Tang J, Guglielmo M, Smith-Gagen J, Bateman L, Navarrete-Galvan L, et al. Antibody-dependent cell-mediated cytotoxicity (ADCC) in familial myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). Fatigue. (2021) 8:226–44.

Google Scholar

256. Theorell J, Bileviciute-Ljungar I, Tesi B, Schlums H, Johnsgaard M, Asadi-Azarbaijani B, et al. Unperturbed cytotoxic lymphocyte phenotype and function in myalgic encephalomyelitis/chronic fatigue syndrome patients. Front Immunol. (2017) 8:723. doi: 10.3389/fimmu.2017.00723

PubMed Abstract | CrossRef Full Text | Google Scholar

257. Cliff J, King E, Lee J, Sepulveda N, Wolf A, Kingdon C, et al. Cellular immune function in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). Front Immunol. (2019) 10:796. doi: 10.3389/fimmu.2019.00796

PubMed Abstract | CrossRef Full Text | Google Scholar

258. Galan M, Vigon L, Fuertes D, Murciano-Anton M, Casado-Fernandez G, Dominguez-Mateos S, et al. Persistent overactive cytotoxic immune response in a Spanish cohort of individuals with long-COVID: identification of diagnostic biomarkers. Front Immunol. (2022) 13:848886. doi: 10.3389/fimmu.2022.848886

PubMed Abstract | CrossRef Full Text | Google Scholar

259. Ryan F, Hope C, Masavuli M, Lynn M, Mekonnen Z, Yeow A, et al. Long-term perturbation of the peripheral immune system months after SARS-CoV-2 infection. BMC Med. (2022) 20:26. doi: 10.1186/s12916-021-02228-6

PubMed Abstract | CrossRef Full Text | Google Scholar

260. Hornig M, Montoya J, Klimas N, Levine S, Felsenstein D, Bateman L, et al. Distinct plasma immune signatures in ME/CFS are present early in the course of illness. Sci Adv. (2015) 1:e1400121. doi: 10.1126/sciadv.1400121

PubMed Abstract | CrossRef Full Text | Google Scholar

261. Bennett A, Chao C, Hu S, Buchwald D, Fagioli L, Schur P, et al. Elevation of bioactive transforming growth factor-B in serum from patients with chronic fatigue syndrome. J Clin Immunol. (1997) 17:160–6.

Google Scholar

262. Moss R, Mercandetti A, Vojdani AT. NF-α and chronic fatigue syndrome. J Clin Immunol. (1999) 19:314–6.

Google Scholar

263. Kerr J, Barah F, Mattey D, Laing I, Hopkins S, Hutchinson I, et al. Circulating tumour necrosis factor-α and interferon- g are detectable during acute and convalescent parvovirus B19 infection and are associated with prolonged and chronic fatigue. J Gen Virol. (2001) 82:3011–9.

Google Scholar

264. Patarca R. Cytokines and chronic fatigue syndrome. Ann NY Acad Sci. (2001) 933:185–200.

Google Scholar

265. Fletcher M, Zeng X, Barnes Z, Levis S, Klimas N. Plasma cytokines in women with chronic fatigue syndrome. J Transl Med. (2009) 7:96.

Google Scholar

266. Broderick G, Fuite J, Kreitz A, Vernon S, Klimas N, Fletcher MA. A formal analysis of cytokine networks in chronic fatigue syndrome. Brain Behav Immun. (2010) 24:1209–17.

Google Scholar

267. Maes M, Twisk F, Ringel K. Inflammatory and cell-mediated immune biomarkers in myalgic encephalomyelitis/chronic fatigue syndrome and depression: inflammatory markers are higher in myalgic encephalomyelitis/chronic fatigue syndrome than in depression. Psychother Psychosom. (2012) 81:286–95. doi: 10.1159/000336803

PubMed Abstract | CrossRef Full Text | Google Scholar

268. Stringer E, Baker K, Carroll I, Montoya J, Chu L, Maecker H, et al. Daily cytokine fluctuations, driven by leptin, are associated with fatigue severity in chronic fatigue syndrome: evidence of inflammatory pathology. J Transl Med. (2013) 11:93. doi: 10.1186/1479-5876-11-93

PubMed Abstract | CrossRef Full Text | Google Scholar

269. Khaiboullina S, DeMeirleir K, Rawat S, Berk G, Gaynor-Berk R, Mijatovic T, et al. Cytokine expression provides clues to the pathophysiology of Gulf War illness and myalgic encephalomyelitis. Cytokine. (2015) 72:1–8. doi: 10.1016/j.cyto.2014.11.019

PubMed Abstract | CrossRef Full Text | Google Scholar

270. Roerink M, Knoop H, Bronkhorst E, Mouthaan H, Hawinkels L, Joosten L, et al. Cytokine signatures in chronic fatigue syndrome patients: a case control study and the effect of anakinra treatment. J Transl Med. (2017) 15:267. doi: 10.1186/s12967-017-1371-9

PubMed Abstract | CrossRef Full Text | Google Scholar

271. Montoya J, Holmes T, Anderson J, Maecker H, Rosenberg-Hasson Y, Valencia I, et al. Cytokine signature associated with disease severity in chronic fatigue syndrome patients. Proc Natl Acad Sci USA. (2017) 114:E7150–8.

Google Scholar

272. Strawbridge R, Sartor M, Scott F, Cleare A. Inflammatory proteins are altered in chronic fatigue syndrome-A systematic review and meta-analysis. Neurosci Biobehav Rev. (2019) 107:69–83. doi: 10.1016/j.neubiorev.2019.08.011

PubMed Abstract | CrossRef Full Text | Google Scholar

273. Jonsjö M, Olsson G, Wicksell R, Alving K, Holmström L, Andreasson A. The role of low-grade inflammation in ME/CFS (myalgic encephalomyelitis/chronic fatigue syndrome) - associations with symptoms. Psychoneuroendocrinology. (2020) 113:104578.

Google Scholar

274. Mandarano A, Maya J, Giloteaux L, Peterson D, Maynard M, Gottschalk C, et al. Myalgic encephalomyelitis/chronic fatigue syndrome patients exhibit altered T cell metabolism and cytokine associations. J Clin Invest. (2020) 130:1491–505. doi: 10.1172/JCI132185

PubMed Abstract | CrossRef Full Text | Google Scholar

275. Raanes E, Stiles T. Associations between psychological and immunological variables in chronic fatigue syndrome/myalgic encephalomyelitis: a systematic review. Front Psychiatry. (2021) 12:716320. doi: 10.3389/fpsyt.2021.716320

PubMed Abstract | CrossRef Full Text | Google Scholar

276. Hornig M, Gottschalk G, Peterson D, Knox K, Schultz A, Eddy M, et al. Cytokine network analysis of cerebrospinal fluid in myalgic encephalomyelitis/chronic fatigue syndrome. Mol Psychiatry. (2016) 21:261–9.

Google Scholar

277. Nakamura T, Schwander S, Donnelly R, Ortega F, Togo F, Broderick G, et al. Cytokines across the night in chronic fatigue syndrome with and without fibromyalgia. Clin Vaccine Immunol. (2010) 17:582–7. doi: 10.1128/CVI.00379-09

PubMed Abstract | CrossRef Full Text | Google Scholar

278. Clark L, Buckland M, Murphy G, Taylor N, Vleck V, Mein C, et al. Cytokine responses to exercise and activity in patients with chronic fatigue syndrome: case-control study. Clin Exp Immunol. (2017) 190:360–71.

Google Scholar

279. Blundell S, Ray K, Buckland M, White P. Chronic fatigue syndrome and circulating cytokines: a systematic review. Brain Behav Immun. (2015) 50:186–95.

Google Scholar

280. Wyller V, Sorensen O, Sulheim D, Fagermoen E, Ueland T, Mollnes T. Plasma cytokine expression in adolescent chronic fatigue syndrome. Brain Behav Immun. (2015) 46:80–6.

Google Scholar

281. Kashipaz M, Swinden D, Todd I, Powell R. Normal production of inflammatory cytokines in chronic fatigue and fibromyalgia syndromes determined by intracellular cytokine staining in short-term cultured blood mononuclear cells. Clin Exp Immunol. (2003) 132:360–5. doi: 10.1046/j.1365-2249.2003.02149.x

PubMed Abstract | CrossRef Full Text | Google Scholar

282. VanElzakker M, Brumfield S, Lara Mejia P. Neuroinflammation and cytokines in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS): a critical review of research methods. Front Neurol. (2019) 9:1033. doi: 10.3389/fneur.2018.01033

PubMed Abstract | CrossRef Full Text | Google Scholar

283. Corbitt M, Eaton-Fitch N, Staines D, Cabanas H, Marshall-Gradisnik S. A systematic review of cytokines in chronic fatigue syndrome/myalgic encephalomyelitis/systemic exertion intolerance disease (CFS/ME/SEID). BMC Neurol. (2019) 19:207. doi: 10.1186/s12883-019-1433-0

PubMed Abstract | CrossRef Full Text | Google Scholar

284. Patterson B, Guevara-Coto J, Yogendra R, Francisco E, Long E, Pise A, et al. Immune-based prediction of COVID-19 severity and chronicity decoded using machine learning. Front Immunol. (2021) 12:700782. doi: 10.3389/fimmu.2021.700782

PubMed Abstract | CrossRef Full Text | Google Scholar

285. Schultheiss C, Willscher E, Paschold L, Gottschick C, Klee B, Henkes S, et al. The IL-1beta, IL-6, and TNF cytokine triad is associated with post-acute sequelae of COVID-19. Cell Rep Med. (2022) 3:100663.

Google Scholar

286. Shuwa H, Shaw T, Knight S, Wemyss K, McClure F, Pearmain L, et al. Alterations in T and B cell function persist in convalescent COVID-19 patients. Med (N Y). (2021) 2:720–35.e4. doi: 10.1016/j.medj.2021.03.013

PubMed Abstract | CrossRef Full Text | Google Scholar

287. Ruenjaiman V, Sodsai P, Kueanjinda P, Bunrasmee W, Klinchanhom S, Reantragoon R, et al. Impact of SARS-CoV-2 infection on the profiles and responses of innate immune cells after recovery. J Microbiol Immunol Infect. (2022) 55(6 Pt 1):993–1004.

Google Scholar

288. Acosta-Ampudia Y, Monsalve D, Rojas M, Rodriguez Y, Zapata E, Ramirez-Santana C, et al. Persistent autoimmune activation and proinflammatory state in post-coronavirus disease 2019 syndrome. J Infect Dis. (2022) 225:2155–62. doi: 10.1093/infdis/jiac017

PubMed Abstract | CrossRef Full Text | Google Scholar

289. Bellan M, Apostolo D, Albe A, Crevola M, Errica N, Ratano G, et al. Determinants of long COVID among adults hospitalized for SARS-CoV-2 infection: a prospective cohort study. Front Immunol. (2022) 13:1038227. doi: 10.3389/fimmu.2022.1038227

PubMed Abstract | CrossRef Full Text | Google Scholar

290. Frere J, Serafini R, Pryce K, Zazhytska M, Oishi K, Golynker I, et al. SARS-CoV-2 infection in hamsters and humans results in lasting and unique systemic perturbations post recovery. Sci Trans Med. (2022) 14:eabq3059. doi: 10.1126/scitranslmed.abq3059

PubMed Abstract | CrossRef Full Text | Google Scholar

291. Phetsouphanh C, Darley D, Wilson D, Howe A, Munier C, Patel S, et al. Immunological dysfunction persists for 8 months following initial mild-to-moderate SARS-CoV-2 infection. Nat Immunol. (2022) 23:210–6. doi: 10.1038/s41590-021-01113-x

PubMed Abstract | CrossRef Full Text | Google Scholar

292. Low R, Low R, Akrami A. A review of cytokine-based pathophysiology of Long COVID symptoms. Front Med. (2023) 10:1011936. doi: 10.3389/fmed.2023.1011936

PubMed Abstract | CrossRef Full Text | Google Scholar

293. Kovarik J, Bileck A, Hagn G, Meier-Menches S, Frey T, Kaempf A, et al. A multi-omics based anti-inflammatory immune signature characterizes long COVID-19 syndrome. iScience. (2023) 26:105717. doi: 10.1016/j.isci.2022.105717

PubMed Abstract | CrossRef Full Text | Google Scholar

294. Clough E, Inigo J, Chandra D, Chaves L, Reynolds J, Aalinkeel R, et al. Mitochondrial dynamics in SARS-COV2 spike protein treated human microglia: implications for neuro-COVID. J Neuroimmune Pharmacol. (2021) 16:770–84.

Google Scholar

295. Sommen S, Havdal L, Selvakumar J, Einvik G, Leegaard T, Lund-Johansen F, et al. Inflammatory markers and pulmonary function in adolescents and young adults 6 months after mild COVID-19. Front Immunol. (2023) 13:1081718. doi: 10.3389/fimmu.2022.1081718

PubMed Abstract | CrossRef Full Text | Google Scholar

296. Son K, Jamil R, Chowdhury A, Mukherjee M, Venegas C, Miyasaki K, et al. Circulating anti-nuclear autoantibodies in COVID-19 survivors predict long COVID symptoms. Eur Respir J. (2023) 61:2200970. doi: 10.1183/13993003.00970-2022

PubMed Abstract | CrossRef Full Text | Google Scholar

297. Bates D, Buchwald D, Lee J, Doolittle T, Kornish J, Rutherford C, et al. Laboratory abnormalities in patients with chronic fatigue syndrome. Clin Infect Dis. (1994) 18(Suppl 1):S86–S.

Google Scholar

298. Clark M, Katon W, Russo J, Kith P, Sintay M, Buchwald D. Chronic fatigue: risk factors for symptom persistence in a 2 1/2-year follow-up study. Am J Med. (1995) 88:187–95. doi: 10.1016/S0002-9343(99)80403-3

PubMed Abstract | CrossRef Full Text | Google Scholar

299. Landay A, Jessop C, Lennette E, Levy J. Chronic fatigue syndrome: clinical condition associated with immune activation. Lancet. (1991) 338:707–12.

Google Scholar

300. Barker E, Fujimura S, Fadem M, Landay A, Levy J. Immunologic abnormalities associated with chronic fatigue syndrome. Clin Infect Dis. (1994) 18(Suppl. 1):S136–41.

Google Scholar

301. Nijs J, de Becker P, de Meirleir K, Demanet C, Vincken W, Schuermans D, et al. Associations between bronchial hyperresponsiveness and immune cell parameters in patients with chronic fatigue syndrome. Chest. (2003) 123:998–1007. doi: 10.1378/chest.123.4.998

PubMed Abstract | CrossRef Full Text | Google Scholar

302. Maya J, Leddy S, Gottschalk C, Peterson D, Hanson M. Altered fatty acid oxidation in lymphocyte populations of myalgic encephalomyelitis/chronic fatigue syndrome. Int J Mol Sci. (2023) 24:2010.

Google Scholar

303. Tirelli U, Marotta G, Improta S, Pinto A. Immmunological abnormalities in patients with chronic fatigue syndrome. Scand J Immunol. (1994) 40:601–8.

Google Scholar

304. Mensah F, Bansal A, Berkovitz S, Sharma A, Reddy V, Leandro M, et al. Extended B cell phenotype in patients with myalgic encephalomyelitis/chronic fatigue syndrome: a cross-sectional study. Clin Exp Immunol. (2016) 184:237–47.

Google Scholar

305. Mensah F, Armstrong C, Reddy V, Bansal A, Berkovitz S, Leandro M, et al. CD24 expression and B cell maturation shows a novel link with energy metabolism: potential implications for patients with myalgic encephalomyelitis/chronic fatigue syndrome. Front Immunol. (2018) 9:2421. doi: 10.3389/fimmu.2018.02421

PubMed Abstract | CrossRef Full Text | Google Scholar

306. Nguyen C, Alsoe L, Lindvall J, Sulheim D, Fagermoen E, Winger A, et al. Whole blood gene expression in adolescent chronic fatigue syndrome: an exploratory cross-sectional study suggesting altered B cell differentiation and survival. J Transl Med. (2017) 15:102. doi: 10.1186/s12967-017-1201-0

PubMed Abstract | CrossRef Full Text | Google Scholar

307. Lunde S, Kristoffersen E, Sapkota D, Risa K, Dahl O, Bruland O, et al. Serum BAFF and APRIL Levels, T-lymphocyte subsets, and immunoglobulins after B-cell depletion using the monoclonal anti-CD20 antibody rituximab in myalgic encephalopathy/chronic fatigue syndrome. PLos One. (2016) 11:e0161226. doi: 10.1371/journal.pone.0161226

PubMed Abstract | CrossRef Full Text | Google Scholar

308. Smith J, Fritz E, Kerr J, Cleare A, Wessely S, Mattey D. Association of chronic fatigue syndrome with human leucocyte antigen class II alleles. J Clin Pathol. (2005) 58:860–3.

Google Scholar

309. Carlo-Stella N, Bozzini S, De Silvestri A, Sbarsi I, Pizzochero C, Lorusso L, et al. Molecular study of receptor for advanced glycation endproduct gene promoter and identification of specific HLA haplotypes possibly involved in chronic fatigue syndrome. Int J Immunopathol Pharmacol. (2009) 22:745–54. doi: 10.1177/039463200902200320

PubMed Abstract | CrossRef Full Text | Google Scholar

310. Pasi A, Bozzini S, Carlo-Stella N, Martinetti M, Bombardieri S, De Silvestri A, et al. Excess of activating killer cell immunoglobulin-like receptors and lack of HLA-Bw4 ligands: a two-edged weapon in chronic fatigue syndrome. Mol Med Rep. (2011) 4:535–40. doi: 10.3892/mmr.2011.447

PubMed Abstract | CrossRef Full Text | Google Scholar

311. Milivojevic M, Che X, Bateman L, Cheng A, Garcia B, Hornig M, et al. Plasma proteomic profiling suggests an association between antigen driven clonal B cell expansion and ME/CFS. PLos One. (2020) 15:e0236148. doi: 10.1371/journal.pone.0236148

PubMed Abstract | CrossRef Full Text | Google Scholar

312. Sato W, Ono H, Matsutani T, Nakamura M, Shin I, Amano K, et al. Skewing of the B cell receptor repertoire in myalgic encephalomyelitis/chronic fatigue syndrome. Brain Behav Immun. (2021) 95:245–55. doi: 10.1016/j.bbi.2021.03.023

PubMed Abstract | CrossRef Full Text | Google Scholar

313. Steiner S, Becker S, Hartwig J, Sotzny F, Lorenz S, Bauer S, et al. Autoimmunity-related risk variants in PTPN22 and CTLA4 are associated with ME/CFS with infectious onset. Front Immunol. (2020) 11:578. doi: 10.3389/fimmu.2020.00578

PubMed Abstract | CrossRef Full Text | Google Scholar

314. Konstantinov K, von Mikecz A, Buchwald D, Jones J, Gerace L, Tan E. Autoantibodies to nuclear envelope antigens in chronic fatigue syndrome. J Clin Invest. (1996) 98:1888–96.

Google Scholar

315. Nishikai M. Antinuclear antibodies in patients with chronic fatigue syndrome. Nippon Rinsho. (2007) 65:1067–70.

Google Scholar

316. Hokama Y, Empey-Campora C, Hara C, Higa N, Siu N, Lau R, et al. Acute phase phospholipids related to the cardiolipin of mitochondria in the sera of patients with chronic fatigue syndrome (CFS), chronic ciguatera fish poisoning (CCFP), and other diseases attributed to chemicals, Gulf War and marine toxins. J Clin Lab Anal. (2008) 22:99–105. doi: 10.1002/jcla.20217

PubMed Abstract | CrossRef Full Text | Google Scholar

317. Hokama Y, Campora C, Hara C, Kuribayashi T, Huynh D, Yabusaki K. Anticardiolipin antibodies in the sera of patients with diagnosed chronic fatigue syndrome. J Clin Lab Anal. (2009) 23:210–2.

Google Scholar

318. Buchwald D, Wener M, Komaroff A. Anti-neuronal antibody levels in chronic fatigue syndrome patients with neurologic abnormalities. Arthritis Rheum. (1991) 34:1485–6.

Google Scholar

319. Ryabkova V, Gavrilova N, Poletaeva A, Pukhalenko A, Koshkina I, Churilov L, et al. Autoantibody correlation signatures in fibromyalgia and myalgic encephalomyelitis/chronic fatigue syndrome: association with symptom severity. Biomedicines. (2023) 11:257.

Google Scholar

320. Wang E, Mao T, Klein J, Dai Y, Huck J, Jaycox J, et al. Diverse functional autoantibodies in patients with COVID-19. Nature. (2021) 595:283–8.

Google Scholar

321. Wallukat G, Hohberger B, Wenzel K, Furst J, Schulze-Rothe S, Wallukat A, et al. Functional autoantibodies against G-protein coupled receptors in patients with persistent Long-COVID-19 symptoms. J Transl Autoimmun. (2021) 4:100100. doi: 10.1016/j.jtauto.2021.100100

PubMed Abstract | CrossRef Full Text | Google Scholar

322. L’Huillier A, Pagano S, Baggio S, Meyer B, Andrey D, Nehme M, et al. Autoantibodies against apolipoprotein A-1 after COVID-19 predict symptoms persistence. Eur J Clin Invest. (2022) 52:e13818. doi: 10.1111/eci.13818

PubMed Abstract | CrossRef Full Text | Google Scholar

323. Pisareva E, Badiou S, Mihalovicova L, Mirandola A, Pastor B, Kudriavtsev A, et al. Persistence of neutrophil extracellular traps and anticardiolipin auto-antibodies in post-acute phase COVID-19 patients. J Med Virol. (2023) 95:e28209. doi: 10.1002/jmv.28209

PubMed Abstract | CrossRef Full Text | Google Scholar

324. Sotzny F, Filgueiras I, Kedor C, Freitag H, Wittke K, Bauer S, et al. Dysregulated autoantibodies targeting vaso- and immunoregulatory receptors in Post COVID Syndrome correlate with symptom severity. Front Immunol. (2022) 13:981532. doi: 10.3389/fimmu.2022.981532

PubMed Abstract | CrossRef Full Text | Google Scholar

325. Seessle J, Waterboer T, Hippchen T, Simon J, Kirchner M, Lim A, et al. Persistent symptoms in adult patients 1 year after coronavirus disease 2019 (COVID-19): a prospective cohort study. Clin Infect Dis. (2022) 74:1191–8.

Google Scholar

326. Lui D, Lee C, Chow W, Lee A, Tam A, Pang P, et al. Long COVID in patients with mild to moderate disease: do thyroid function and autoimmunity play a role? Endocr Pract. (2021) 27:894–902. doi: 10.1016/j.eprac.2021.06.016

PubMed Abstract | CrossRef Full Text | Google Scholar

327. Moody R, Sonda S, Johnston F, Smith K, Stephens N, McPherson M, et al. Antibodies against Spike protein correlate with broad autoantigen recognition 8 months post SARS-CoV-2 exposure, and anti-calprotectin autoantibodies associated with better clinical outcomes. Front Immunol. (2022) 13:945021. doi: 10.3389/fimmu.2022.945021

PubMed Abstract | CrossRef Full Text | Google Scholar

328. Peluso M, Mitchell A, Wang C, Takahashi S, Hoh R, Tai V, et al. Low prevalence of interferon-α autoantibodies in people experiencing long COVID symptoms. J Infect Dis. (2023) 227:246–50. doi: 10.1093/infdis/jiac372

PubMed Abstract | CrossRef Full Text | Google Scholar

329. Vojdani A, Ghoneum M, Choppa P, Magtoto L, Lapp C. Elevated apoptotic cell population in patients with chronic fatigue syndrome: the pivotal role of protein kinase RNA. J Intern Med. (1997) 242:465–78. doi: 10.1111/j.1365-2796.1997.tb00019.x

PubMed Abstract | CrossRef Full Text | Google Scholar

330. Kennedy G, Spence V, Underwood C, Belch J. Increased neutrophil apoptosis in chronic fatigue syndrome. J Clin Pathol. (2004) 57:891–3.

Google Scholar

331. Kennedy G, Khan F, Hill A, Underwood C, Belch J. Biochemical and vascular aspects of pediatric chronic fatigue syndrome. Arch Pediatr Adolesc Med. (2010) 164:817–23. doi: 10.1001/archpediatrics.2010.157

PubMed Abstract | CrossRef Full Text | Google Scholar

332. Fang H, Xie Q, Boneva R, Fostel J, Perkins R, Tong W. Gene expression profile exploration of a large dataset on chronic fatigue syndrome. Pharmacogenomics. (2006) 7:429–40. doi: 10.2217/14622416.7.3.429

PubMed Abstract | CrossRef Full Text | Google Scholar

333. Gow J, Hagan S, Herzyk P, Cannon C, Behan P, Chaudhuri A. A gene signature for post-infectious chronic fatigue syndrome. BMC Med Genomics. (2009) 2:38. doi: 10.1186/1755-8794-2-38

PubMed Abstract | CrossRef Full Text | Google Scholar

334. Brenu E, Ashton K, van Driel M, Staines D, Peterson D, Atkinson G, et al. Cytotoxic lymphocyte microRNAs as prospective biomarkers for chronic fatigue syndrome/myalgic encephalomyelitis. J Affect Disord. (2012) 141:261–9. doi: 10.1016/j.jad.2012.03.037

PubMed Abstract | CrossRef Full Text | Google Scholar

335. Sweetman E, Kleffmann T, Edgar C, de Lange M, Vallings R, Tate W. A SWATH-MS analysis of myalgic encephalomyelitis/chronic fatigue syndrome peripheral blood mononuclear cell proteomes reveals mitochondrial dysfunction. J Transl Med. (2020) 18:365. doi: 10.1186/s12967-020-02533-3

PubMed Abstract | CrossRef Full Text | Google Scholar

336. Jahanbani F, Maynard R, Sing J, Jahanbani S, Perrino J, Spacek D, et al. Phenotypic characteristics of peripheral immune cells of myalgic encephalomyelitis/chronic fatigue syndrome via transmission electron microscopy: a pilot study. PLos One. (2022) 17:e0272703. doi: 10.1371/journal.pone.0272703

PubMed Abstract | CrossRef Full Text | Google Scholar

337. Swanink C, Vercoulen J, Galama J, Roos M, Meyaard L, van der Ven-Jongekrijg J, et al. Lymphocyte subsets, apoptosis, and cytokines in patients with chronic fatigue syndrome. J Infect Dis. (1996) 173:460–3.

Google Scholar

338. Hassan I, Bannister B, Akbar A, Weir W, Bofill M. A study of the immunology of the chronic fatigue syndrome: correlation of immunologic parameters to health dysfunction. Clin Immunol Immunopathol. (1998) 87:60–7.

Google Scholar

339. Schacterle R, Milford E, Komaroff A. The frequency of HLA class II antigens in chronic fatigue syndrome. J Chron Fatigue Synd. (2003) 11:33–42.

Google Scholar

340. Hajdarevic R, Lande A, Rekeland I, Rydland A, Strand E, Sosa D, et al. Fine mapping of the major histocompatibility complex (MHC) in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) suggests involvement of both HLA class I and class II loci. Brain Behav Immun. (2021) 98:101–9. doi: 10.1016/j.bbi.2021.08.219

PubMed Abstract | CrossRef Full Text | Google Scholar

341. Underhill J, Mahalingam M, Peakman M, Wessely S. Lack of association between HLA genotype and chronic fatigue syndrome. Eur J Immunogenet. (2001) 28:425–8. doi: 10.1046/j.1365-2370.2001.00235.x

PubMed Abstract | CrossRef Full Text | Google Scholar

342. Ramos S, Brenu E, Broadley S, Kwiatek R, Ng J, Nguyen T, et al. Regulatory T, natural killer T and gammadelta T cells in multiple sclerosis and chronic fatigue syndrome/myalgic encephalomyelitis: a comparison. Asian Pac J Allergy Immunol. (2016) 34:300–5. doi: 10.12932/AP0733

PubMed Abstract | CrossRef Full Text | Google Scholar

343. Curriu M, Carillo J, Massanella M, Rigau J, Alegre J, Puig J, et al. Screening NK-, B- and T-cell phenotype and function in patients suffering from chronic fatigue syndrome. J Transl Med. (2013) 11:68. doi: 10.1186/1479-5876-11-68

PubMed Abstract | CrossRef Full Text | Google Scholar

344. Nguyen T, Johnston S, Chacko A, Gibson D, Cepon J, Smith P, et al. Novel characterisation of mast cell phenotypes from peripheral blood mononuclear cells in chronic fatigue syndrome/myalgic encephalomyelitis patients. Asian Pac J Allergy Immunol. (2017) 35:75–81. doi: 10.12932/ap0771

PubMed Abstract | CrossRef Full Text | Google Scholar

345. Balinas C, Nguyen T, Johnston S, Smith P, Staines D, Marshall-Gradisnik S. Investigation of mast cell toll-like receptor 3 in chronic fatigue syndrome/myalgic encephalomyelitis and systemic mastocytosis using the novel application of autoMACS magnetic separation and flow cytometry. Asian Pac J Allergy Immunol. (2018) 36:257–64. doi: 10.12932/AP-200517-0086

PubMed Abstract | CrossRef Full Text | Google Scholar

346. Nepotchatykh E, Caraus I, Elremaly W, Leveau C, Elbakry M, Godbout C, et al. Circulating microRNA expression signatures accurately discriminate myalgic encephalomyelitis from fibromyalgia and comorbid conditions. Sci Rep. (2023) 13:1896.

Google Scholar

347. Helliwell A, Stockwell P, Edgar C, Chatterjee A, Tate W. Dynamic epigenetic changes during a relapse and recovery cycle in myalgic encephalomyelitis/chronic fatigue syndrome. Int J Mol Sci. (2022) 23:11852.

Google Scholar

348. Zuo T, Liu Q, Zhang F, Lui G, Tso E, Yeoh Y, et al. Depicting SARS-CoV-2 faecal viral activity in association with gut microbiota composition in patients with COVID-19. Gut. (2021) 70:276–84. doi: 10.1136/gutjnl-2020-322294

PubMed Abstract | CrossRef Full Text | Google Scholar

349. Gaebler C, Wang Z, Lorenzi J, Muecksch F, Finkin S, Tokuyama M, et al. Evolution of antibody immunity to SARS-CoV-2. Nature. (2021) 591:639–44.

Google Scholar

350. Swank Z, Senussi Y, Manickas-Hill Z, Yu X, Li J, Alter G, et al. Persistent circulating SARS-CoV-2 spike is associated with post-acute COVID-19 sequelae. Clin Infect Dis. (2022) 76:e487–e490.

Google Scholar

351. Cheung C, Goh D, Lim X, Tien T, Lim J, Lee J, et al. Residual SARS-CoV-2 viral antigens detected in GI and hepatic tissues from five recovered patients with COVID-19. Gut. (2022) 71:226–9.

Google Scholar

352. Natarajan A, Zlitni S, Brooks E, Vance S, Dahlen A, Hedlin H, et al. Gastrointestinal symptoms and fecal shedding of SARS-CoV-2 RNA suggest prolonged gastrointestinal infection. Med (N Y). (2022) 3:371–87.e9.

Google Scholar

353. Tejerina F, Catalan P, Rodriguez-Grande C, Adan J, Rodriguez-Gonzalez C, Munoz P, et al. Post-COVID-19 syndrome. SARS-CoV-2 RNA detection in plasma, stool, and urine in patients with persistent symptoms after COVID-19. BMC Infect Dis. (2022) 22:211. doi: 10.1186/s12879-022-07153-4

PubMed Abstract | CrossRef Full Text | Google Scholar

354. Straus S, Tosato G, Armstrong G, Lawley T, Preble O, Henle W, et al. Persisting illness and fatigue in adults with evidence of Epstein-Barr virus infection. Ann Intern Med. (1985) 102:7–16.

Google Scholar

355. Jones J, Ray C, Minnich L, Hicks M, Kibler R, Lucas D. Evidence for active Epstein-Barr virus infection in patients with persistent, unexplained illnesses: elevated anti-early antigen antibodies. Ann Intern Med. (1985) 102:1–7. doi: 10.7326/0003-4819-102-1-

PubMed Abstract | CrossRef Full Text | Google Scholar

356. Wray B, Gaughf C, Chandler F Jr., Berry S, Latham J, Wood L, et al. Detection of Epstein-Barr virus and cytomegalovirus in patients with chronic fatigue. Ann Allergy. (1993) 71:223–6.

Google Scholar

357. Di Luca D, Zorzenon M, Mirandola P, Colle R, Botta G, Cassai E. Human herpesvirus 6 and human herpesvirus 7 in chronic fatigue syndrome. J Clin Microbiol. (1995) 33:1660–1.

Google Scholar

358. Patnaik M, Komaroff A, Conley E, Ojo-Amaize E, Peter J. Prevalence of IgM antibodies to human herpesvirus 6 early antigen (p41/38) in patients with chronic fatigue syndrome. J Infect Dis. (1995) 172:1364–7. doi: 10.1093/infdis/172.5.1364

PubMed Abstract | CrossRef Full Text | Google Scholar

359. Komaroff A. Is human herpesvirus-6 a trigger for chronic fatigue syndrome? J Clin Virol. (2006) 37(Suppl. 1):S39–46. doi: 10.1016/S1386-6532(06)70010-5

PubMed Abstract | CrossRef Full Text | Google Scholar

360. Koelle D, Barcy S, Huang M, Ashley R, Corey L, Zeh J, et al. Markers of viral infection in monozygotic twins discordant for chronic fatigue syndrome. Clin Infect Dis. (2002) 35:518–25. doi: 10.1086/341774

PubMed Abstract | CrossRef Full Text | Google Scholar

361. Simonnet A, Engelmann I, Moreau A, Garcia B, Six S, El Kalioubie A, et al. High incidence of Epstein-Barr virus, cytomegalovirus, and human-herpes virus-6 reactivations in critically ill patients with COVID-19. Infect Dis Now. (2021) 51:296–9. doi: 10.1016/j.idnow.2021.01.005

PubMed Abstract | CrossRef Full Text | Google Scholar

362. Brooks B, Tancredi C, Song Y, Mogus A, Huang M, Zhu H, et al. Epstein-Barr virus and human herpesvirus-6 reactivation in acute COVID-19 patients. Viruses. (2022) 14:1872.

Google Scholar

363. Shafiee A, Teymouri Athar M, Amini M, Hajishah H, Siahvoshi S, Jalali M, et al. Reactivation of herpesviruses during COVID-19: a systematic review and meta-analysis. Rev Med Virol. (2023) 33:e2437.

Google Scholar

364. Carneiro V, Alves-Leon S, Sarmento D, Coelho W, Moreira O, Salvio A, et al. Herpesvirus and neurological manifestations in patients with severe coronavirus disease. Virol J. (2022) 19:101.

Google Scholar

365. Gold J, Okyay R, Licht W, Hurley D. Investigation of long COVID prevalence and its relationship to Epstein-Barr virus reactivation. Pathogens. (2021) 10:763.

Google Scholar

366. Peluso M, Deveau T, Munter S, Ryder D, Buck A, Beck-Engeser G, et al. Chronic viral coinfections differentially affect the likelihood of developing long COVID. J Clin Invest. (2023) 133:e163669.

Google Scholar

367. Giloteaux L, Goodrich J, Walters W, Levine S, Ley R, Hanson M. Reduced diversity and altered composition of the gut microbiome in individuals with myalgic encephalomyelitis/chronic fatigue syndrome. Microbiome. (2016) 4:30. doi: 10.1186/s40168-016-0171-4

PubMed Abstract | CrossRef Full Text | Google Scholar

368. Nagy-Szakal D, Williams B, Mishra N, Che X, Lee B, Bateman L, et al. Fecal metagenomic profiles in subgroups of patients with myalgic encephalomyelitis/chronic fatigue syndrome. Microbiome. (2017) 5:44.

Google Scholar

369. Guo C, Che X, Briese T, Ranjan A, Allicock O, Yates R, et al. Deficient butyrate-producing capacity in the gut microbiome is associated with bacterial network disturbances and fatigue symptoms in ME/CFS. Cell Host Microbe. (2023) 31:288–304.e8. doi: 10.1016/j.chom.2023.01.004

PubMed Abstract | CrossRef Full Text | Google Scholar

370. Xiong R, Gunter C, Fleming E, Vernon S, Bateman L, Unutmaz D, et al. Multi-’omics of gut microbiome-host interactions in short- and long-term myalgic encephalomyelitis/chronic fatigue syndrome patients. Cell Host Microbe. (2023) 31:273–87.e5.

Google Scholar

371. Yeoh Y, Zuo T, Lui G, Zhang F, Liu Q, Li A, et al. Gut microbiota composition reflects disease severity and dysfunctional immune responses in patients with COVID-19. Gut. (2021) 70:698–706. doi: 10.1136/gutjnl-2020-323020

PubMed Abstract | CrossRef Full Text | Google Scholar

372. Haran J, Bradley E, Zeamer A, Cincotta L, Salive M, Dutta P, et al. Inflammation-type dysbiosis of the oral microbiome associates with the duration of COVID-19 symptoms and long COVID. JCI Insight. (2021) 6:e152346.

Google Scholar

373. Liu Q, Mak J, Su Q, Yeoh Y, Lui G, Ng S, et al. Gut microbiota dynamics in a prospective cohort of patients with post-acute COVID-19 syndrome. Gut. (2022) 71:544–52.

Google Scholar

374. Rajeevan M, Murray J, Oakley L, Lin J, Unger E. Association of chronic fatigue syndrome with premature telomere attrition. J Transl Med. (2018) 16:44. doi: 10.1186/s12967-018-1414-x

PubMed Abstract | CrossRef Full Text | Google Scholar

375. Sfera A, Osorio C, Zapata Martin Del Campo CM, Pereida S, Maurer S, Maldonado JC, et al. Endothelial senescence and chronic fatigue syndrome, a COVID-19 based hypothesis. Front Cell Neurosci. (2021) 15:673217. doi: 10.3389/fncel.2021.673217

PubMed Abstract | CrossRef Full Text | Google Scholar

376. Lynch S, Guo G, Gibson D, Bjourson A, Rai T. Role of senescence and aging in SARS-CoV-2 infection and COVID-19 Disease. Cells. (2021) 10:3367.

Google Scholar

377. Kuo C, Pilling L, Atkins J, Masoli J, Delgado J, Tignanelli C, et al. Biological aging predicts vulnerability to COVID-19 severity in UK Biobank participants. J Gerontol A Biol Sci Med Sci. (2021) 76:e133–41. doi: 10.1093/gerona/glab060

PubMed Abstract | CrossRef Full Text | Google Scholar

378. Kuratsune H, Yamaguti K, Takahashi M, Misaki H, Tagawa S, Kitani T. Acylcarnitine deficiency in chronic fatigue syndrome. Clin Infect Dis. (1994) 18(Suppl. 1):S62–7.

Google Scholar

379. Majeed T, de Simone C, Famularo G, Marcellini S, Behan P. Abnormalities of carnitine metabolism in chronic fatigue syndrome. Eur J Neurol. (1995) 2:425–8.

Google Scholar

380. Kuratsune H, Yamaguti K, Lindh G, Evengard B, Takahashi H, Machii T, et al. Low levels of serum acylcarnitine in chronic fatigue syndrome and chronic hepatitis type C, but not seen in other diseases. Int J Mol Med. (1998) 2:51–6. doi: 10.3892/ijmm.2.1.51

PubMed Abstract | CrossRef Full Text | Google Scholar

381. Jones M, Goodwin C, Amjad S, Chalmers R. Plasma and urinary carnitine and acylcarnitines in chronic fatigue syndrome. Clin Chim Acta. (2005) 360:173–7.

Google Scholar

382. Reuter S, Evans A. Long-chain acylcarnitine deficiency in patients with chronic fatigue syndrome. Potential involvement of altered carnitine palmitoyltransferase-l activity. J Intern Med. (2011) 270:76–84.

Google Scholar

383. Armstrong C, McGregor N, Lewis D, Butt H, Gooley P. The association of fecal microbiota and fecal, blood serum and urine metabolites in myalgic encephalomyelitis/chronic fatigue syndrome. Metabolomics. (2017) 13:1–13.

Google Scholar

384. Naviaux R, Naviaux J, Li K, Bright A, Alaynick W, Wang L, et al. Metabolic features of chronic fatigue syndrome. Proc Natl Acad Sci USA. (2016) 113:E5472–80.

Google Scholar

385. Germain A, Ruppert D, Levine S, Hanson M. Metabolic profiling of a myalgic encephalomyelitis/chronic fatigue syndrome discovery cohort reveals disturbances in fatty acid and lipid metabolism. Mol Biosyst. (2017) 13:371–9.

Google Scholar

386. Nagy-Szakal D, Barupal D, Lee B, Che X, Williams B, Kahn E, et al. Insights into myalgic encephalomyelitis/chronic fatigue syndrome phenotypes through comprehensive metabolomics. Sci Rep. (2018) 8:10056. doi: 10.1038/s41598-018-28477-9

PubMed Abstract | CrossRef Full Text | Google Scholar

387. Germain A, Barupal D, Levine S, Hanson M. Comprehensive circulatory metabolomics in ME/CFS reveals disrupted metabolism of acyl lipids and steroids. Metabolites. (2020) 10:34. doi: 10.3390/metabo10010034

PubMed Abstract | CrossRef Full Text | Google Scholar

388. Hoel F, Hoel A, Pettersen I, Rekeland I, Risa K, Alme K, et al. A map of metabolic phenotypes in patients with myalgic encephalomyelitis/chronic fatigue syndrome. JCI Insight. (2021) 6:e149217.

Google Scholar

389. Nkiliza A, Parks M, Cseresznye A, Oberlin S, Evans J, Darcey T, et al. Sex-specific plasma lipid profiles of ME/CFS patients and their association with pain, fatigue, and cognitive symptoms. J Transl Med. (2021) 19:370. doi: 10.1186/s12967-021-03035-6

PubMed Abstract | CrossRef Full Text | Google Scholar

390. Glass K, Germain A, Huang Y, Hanson M. Urine metabolomics exposes anomalous recovery after maximal exertion in female ME/CFS patients. Int J Mol Sci. (2023) 24:3685. doi: 10.3390/ijms24043685

PubMed Abstract | CrossRef Full Text | Google Scholar

391. de Boer E, Petrache I, Goldstein N, Olin J, Keith R, Modena B, et al. Decreased fatty acid oxidation and altered lactate production during exercise in patients with post-acute COVID-19 syndrome. Am J Respir Crit Care Med. (2022) 205:126–9.

Google Scholar

392. Guntur V, Nemkov T, de Boer E, Mohning M, Baraghoshi D, Cendali F, et al. Signatures of mitochondrial dysfunction and impaired fatty acid metabolism in plasma of patients with post-acute sequelae of COVID-19 (PASC). Metabolites. (2022) 12:1026. doi: 10.3390/metabo12111026

PubMed Abstract | CrossRef Full Text | Google Scholar

393. Stromberg S, Baxter B, Dooley G, LaVergne S, Gallichotte E, Dutt T, et al. Relationships between plasma fatty acids in adults with mild, moderate, or severe COVID-19 and the development of post-acute sequelae. Front Nutr. (2022) 9:960409. doi: 10.3389/fnut.2022.960409

PubMed Abstract | CrossRef Full Text | Google Scholar

394. Yang C, Chang C, Yang C, Pariante C, Su K. Long COVID and long chain fatty acids (LCFAs): Psychoneuroimmunity implication of omega-3 LCFAs in delayed consequences of COVID-19. Brain Behav Immun. (2022) 103:19–27.

Google Scholar

395. Jones M, Cooper E, Amjad S, Goodwin C, Barron J, Chalmers R. Urinary and plasma organic acids and amino acids in chronic fatigue syndrome. Clin Chim Acta. (2005) 361:150–8.

Google Scholar

396. Armstrong C, McGregor N, Sheedy J, Buttfield I, Butt H, Gooley PR. NMR metabolic profiling of serum identifies amino acid disturbances in chronic fatigue syndrome. Clin Chim Acta. (2012) 413:1525–31. doi: 10.1016/j.cca.2012.06.022

PubMed Abstract | CrossRef Full Text | Google Scholar

397. Armstrong C, McGregor N, Butt H, Gooley P. Metabolism in chronic fatigue syndrome. In: Makowski G editor. Advances in Clinical Chemistry. Waltham, MA: Elsevier (2014). p. 121–72.

Google Scholar

398. Armstrong C, McGregor N, Lewis D, Butt H, Gooley P. Metabolic profiling reveals anomalous energy metabolism and oxidative stress pathways in chronic fatigue syndrome patients. Metabolomics. (2015) 11:1626–39.

Google Scholar

399. Fluge O, Mella O, Bruland O, Risa K, Dyrstad S, Alme K, et al. Metabolic profiling indicates impaired pyruvate dehydrogenase function in myalgic encephalopathy/chronic fatigue syndrome. JCI Insight. (2016) 1:e89376.

Google Scholar

400. Yamano E, Sugimoto M, Hirayama A, Kume S, Yamato M, Jin G, et al. Index markers of chronic fatigue syndrome with dysfunction of TCA and urea cycles. Sci Rep. (2016) 6:34990. doi: 10.1038/srep34990

PubMed Abstract | CrossRef Full Text | Google Scholar

401. Huth T, Eaton-Fitch N, Staines D, Marshall-Gradisnik S. A systematic review of metabolomic dysregulation in chronic fatigue syndrome/myalgic encephalomyelitis/systemic exertion intolerance disease (CFS/ME/SEID). J Transl Med. (2020) 18:198.

Google Scholar

402. Groven N, Reitan S, Fors E, Guzey I. Kynurenine metabolites and ratios differ between chronic fatigue syndrome, fibromyalgia, and healthy controls. Psychoneuroendocrinology. (2021) 131:105287. doi: 10.1016/j.psyneuen.2021.105287

PubMed Abstract | CrossRef Full Text | Google Scholar

403. Simonato M, Dall’Acqua S, Zilli C, Sut S, Tenconi R, Gallo N, et al. Tryptophan metabolites, cytokines, and fatty acid binding protein 2 in myalgic encephalomyelitis/chronic fatigue syndrome. Biomedicines. (2021) 9:1724. doi: 10.3390/biomedicines9111724

PubMed Abstract | CrossRef Full Text | Google Scholar

404. Holmes E, Wist J, Masuda R, Lodge S, Nitschke P, Kimhofer T, et al. Incomplete systemic recovery and metabolic phenoreversion in post-acute-phase nonhospitalized COVID-19 patients: implications for assessment of post-acute COVID-19 syndrome. J Proteome Res. (2021) 20:3315–29. doi: 10.1021/acs.jproteome.1c00224

PubMed Abstract | CrossRef Full Text | Google Scholar

405. Nataf S, Pays L. Molecular insights into SARS-CoV2-induced alterations of the gut/brain axis. Int J Mol Sci. (2021) 22:10440. doi: 10.3390/ijms221910440

PubMed Abstract | CrossRef Full Text | Google Scholar

406. Ciregia F, Kollipara L, Giusti L, Zahedi R, Giacomelli C, Mazzoni M, et al. Bottom-up proteomics suggests an association between differential expression of mitochondrial proteins and chronic fatigue syndrome. Transl Psychiatry. (2016) 6:e904. doi: 10.1038/tp.2016.184

PubMed Abstract | CrossRef Full Text | Google Scholar

407. Germain A, Ruppert D, Levine S, Hanson M. Prospective biomarkers from plasma metabolomics of myalgic encephalomyelitis/chronic fatigue syndrome implicate redox imbalance in disease symptomatology. Metabolites. (2018) 8:90. doi: 10.3390/metabo8040090

PubMed Abstract | CrossRef Full Text | Google Scholar

408. Jason L, Conroy K, Furst J, Vasan K, Katz B. Pre-illness data reveals differences in multiple metabolites and metabolic pathways in those who do and do not recover from infectious mononucleosis. Mol Omics. (2022) 18:662–5. doi: 10.1039/d2mo00124a

PubMed Abstract | CrossRef Full Text | Google Scholar

409. McGregor N, Armstrong C, Lewis D, Gooley P. Post-exertional malaise is associated with hypermetabolism, hypoacetylation and purine metabolism deregulation in ME/CFS cases. Diagnostics (Basel). (2019) 9:E70. doi: 10.3390/diagnostics9030070

PubMed Abstract | CrossRef Full Text | Google Scholar

410. Lawson N, Hsieh C, March D, Wang X. Elevated energy production in chronic fatigue syndrome patients. J Nat Sci. (2016) 2:e221.

Google Scholar

411. Tomas C, Brown A, Strassheim V, Elson J, Newton J, Manning P. Cellular bioenergetics is impaired in patients with chronic fatigue syndrome. PLos One. (2017) 12:e0186802. doi: 10.1371/journal.pone.0186802

PubMed Abstract | CrossRef Full Text | Google Scholar

412. Tomas C, Brown A, Newton J, Elson J. Mitochondrial complex activity in permeabilised cells of chronic fatigue syndrome patients using two cell types. PeerJ. (2019) 7:e6500. doi: 10.7717/peerj.6500

PubMed Abstract | CrossRef Full Text | Google Scholar

413. Tomas C, Elson J, Newton J, Walker M. Substrate utilisation of cultured skeletal muscle cells in patients with CFS. Sci Rep. (2020) 10:18232.

Google Scholar

414. Tomas C, Elson J, Strassheim V, Newton J, Walker M. The effect of myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) severity on cellular bioenergetic function. PLos One. (2020) 15:e0231136. doi: 10.1371/journal.pone.0231136

PubMed Abstract | CrossRef Full Text | Google Scholar

415. Missailidis D, Annesley S, Allan C, Sanislav O, Lidbury B, Lewis D, et al. An isolated Complex V inefficiency and dysregulated mitochondrial function in immortalized lymphocytes from ME/CFS patients. Int J Mol Sci. (2020) 21:1074. doi: 10.3390/ijms21031074

PubMed Abstract | CrossRef Full Text | Google Scholar

416. Missailidis D, Sanislav O, Allan C, Smith P, Annesley S, Fisher P. Dysregulated provision of oxidisable substrates to the mitochondria in ME/CFS lymphoblasts. Int J Mol Sci. (2021) 22:2046.

Google Scholar

417. Maes M, Kubera M, Uytterhoeven M, Vrydags N, Bosmans E. Increased plasma peroxides as a marker of oxidative stress in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). Med Sci Monit. (2011) 17:SC11–5.

Google Scholar

418. Robinson M, Gray S, Watson M, Kennedy G, Hill A, Belch J, et al. Plasma IL-6, its soluble receptors and F2-isoprostanes at rest and during exercise in chronic fatigue syndrome. Scand J Med Sci Sports. (2010) 20:282–90. doi: 10.1111/j.1600-0838.2009.00895.x

PubMed Abstract | CrossRef Full Text | Google Scholar

419. Kennedy G, Spence V, McLaren M, Hill A, Underwood C, Belch J. Oxidative stress levels are raised in chronic fatigue syndrome and are associated with clinical symptoms. Free Radic Biol Med. (2005) 39:584–9.

Google Scholar

420. Block T, Kuo J. Rationale for nicotinamide adenine dinucleotide (n.d.) metabolome disruption as a pathogenic mechanism of post-acute COVID-19 syndrome. Clin Pathol (2022) 15:2632010X221106986.

Google Scholar

421. Al-Hakeim H, Al-Rubaye H, Al-Hadrawi D, Almulla A, Maes M. Long-COVID post-viral chronic fatigue and affective symptoms are associated with oxidative damage, lowered antioxidant defenses and inflammation: a proof of concept and mechanism study. Mol Psychiatry. (2023) 28:564–78.

Google Scholar

422. Vollbracht C, Kraft K. Feasibility of vitamin C in the treatment of post viral fatigue with focus on long COVID, based on a systematic review of IV vitamin C on fatigue. Nutrients. (2021) 13:1154.

Google Scholar

423. Izzo R, Trimarco V, Mone P, Aloe T, Capra Marzani M, Diana A, et al. Combining L-arginine with vitamin C improves long-COVID symptoms: the LINCOLN Survey. Pharmacol Res. (2022) 183:106360. doi: 10.1016/j.phrs.2022.106360

PubMed Abstract | CrossRef Full Text | Google Scholar

424. Miwa K, Fujita M. Increased oxidative stress suggested by low serum vitamin E concentrations in patients with chronic fatigue syndrome. Int J Cardiol. (2009) 136:238–9.

Google Scholar

425. Jammes Y, Guillaume Steinberg J, Guieu R, Delliaux S. Chronic fatigue syndrome with history of severe infection combined altered blood oxidant status, and reduced potassium efflux and muscle excitability at exercise. Open J Int Med. (2013) 3:98–105.

Google Scholar

426. Fenouillet E, Vigouroux A, Steinberg J, Chagvardieff A, Retornaz F, Guieu R, et al. Association of biomarkers with health-related quality of life and history of stressors in myalgic encephalomyelitis/chronic fatigue syndrome patients. J Transl Med. (2016) 14:251. doi: 10.1186/s12967-016-1010-x

PubMed Abstract | CrossRef Full Text | Google Scholar

427. Maes M, Mihaylova I, Leunis J. Chronic fatigue syndrome is accompanied by an IgM-related immune response directed against neoepitopes formed by oxidative or nitrosative damage to lipids and proteins. Neuro Endocrinol Lett. (2006) 27:615–21.

Google Scholar

428. Morris G, Berk M, Klein H, Walder K, Galecki P, Maes M. Nitrosative stress, hypernitrosylation, and autoimmune responses to nitrosylated proteins: new pathways in neuroprogressive disorders including depression and chronic fatigue syndrome. Mol Neurobiol. (2017) 54:4271–91. doi: 10.1007/s12035-016-9975-2

PubMed Abstract | CrossRef Full Text | Google Scholar

429. Monro J, Puri BK. A molecular neurobiological approach to understanding the aetiology of chronic fatigue syndrome (myalgic encephalomyelitis or systemic exertion intolerance disease) with treatment implications. Mol Neurobiol. (2018) 55:7377–88. doi: 10.1007/s12035-018-0928-9

PubMed Abstract | CrossRef Full Text | Google Scholar

430. Suárez A, Guillamó E, Roig T, Blázquez A, Alegre J, Bermúdez J, et al. Nitric oxide metabolite production during exercise in chronic fatigue syndrome: a case-control study. J Womens Health. (2010) 19:1073–7. doi: 10.1089/jwh.2008.1255

PubMed Abstract | CrossRef Full Text | Google Scholar

431. Morris G, Maes M. Oxidative and nitrosative stress and immune-inflammatory pathways in patients with myalgic encephalomyelitis (ME)/chronic fatigue syndrome (CFS). Curr Neuropharmacol. (2014) 12:168–85.

Google Scholar

432. Gottschalk G, Peterson D, Knox K, Maynard M, Whelan R, Roy A. Elevated ATG13 in serum of patients with ME/CFS stimulates oxidative stress response in microglial cells via activation of receptor for advanced glycation end products (RAGE). Mol Cell Neurosci. (2022) 120:103731. doi: 10.1016/j.mcn.2022.103731

PubMed Abstract | CrossRef Full Text | Google Scholar

433. Shungu D, Weiduschat N, Murrough J, Mao X, Pillemer S, Dyke J, et al. Increased ventricular lactate in chronic fatigue syndrome. III. relationships to cortical glutathione and clinical symptoms implicate oxidative stress in disorder pathophysiology. NMR Biomed. (2012) 25:1073–87. doi: 10.1002/nbm.2772

PubMed Abstract | CrossRef Full Text | Google Scholar

434. Natelson B, Vu D, Coplan J, Mao X, Blate M, Kang G, et al. Elevations of ventricular lactate levels occur in both chronic fatigue syndrome and fibromyalgia. Fatigue. (2017) 5:15–20.

Google Scholar

435. Georgiades E, Behan W, Kilduff L, Hadjicharalambous M, Mackie E, Wilson J, et al. Chronic fatigue syndrome: new evidence for a central fatigue disorder. Clin Sci. (2003) 105:213–8.

Google Scholar

436. Badawy A, Morgan C, Llewelyn M, Albuquerque S, Farmer A. Heterogeneity of serum tryptophan concentration and availability to the brain in patients with chronic fatigue syndrome. J Psychopharm. (2005) 19:385–91.

Google Scholar

437. Thomas T, Stefanoni D, Reisz J, Nemkov T, Bertolone L, Francis R, et al. COVID-19 infection alters kynurenine and fatty acid metabolism, correlating with IL-6 levels and renal status. JCI Insight. (2020) 5:e140327.

Google Scholar

438. Ruffieux H, Hanson A, Lodge S, Lawler N, Whiley L, Gray N, et al. A patient-centric modeling framework captures recovery from SARS-CoV-2 infection. Nat Immunol. (2023) 24:349–58. doi: 10.1038/s41590-022-01380-2

PubMed Abstract | CrossRef Full Text | Google Scholar

439. Russell A, Hepgul N, Nikkheslat N, Borsini A, Zajkowska Z, Moll N, et al. Persistent fatigue induced by interferon-alpha: a novel, inflammation-based, proxy model of chronic fatigue syndrome. Psychoneuroendocrinology. (2019) 100:276–85. doi: 10.1016/j.psyneuen.2018.11.032

PubMed Abstract | CrossRef Full Text | Google Scholar

440. van Campen C, Visser F. Comparing idiopathic chronic fatigue and myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) in males: response to two-day cardiopulmonary exercise testing protocol. Healthcare (Basel). (2021) 9:683.

Google Scholar

441. van Campen C, Visser F. Female patients with myalgic encephalomyelitis/chronic fatigue syndrome or idiopathic chronic fatigue: comparison of responses to a two-day cardiopulmonary exercise testing protocol. Healthcare (Basel). (2021) 9:682.

Google Scholar

442. Franklin J, Atkinson G, Atkinson J, Batterham A. Peak oxygen uptake in chronic fatigue syndrome/myalgic encephalomyelitis: a meta-analysis. Int J Sports Med. (2018) 40:77–87.

Google Scholar

443. Lim E, Kang E, Jang E, Son C. The prospects of the two-day cardiopulmonary exercise test (CPET) in ME/CFS patients: a meta-analysis. J Clin Med. (2020) 9:4040.

Google Scholar

444. Davenport T, Stevens S, Stevens J, Snell C, Van Ness J, Mooney A. Properties of measurements obtained during cardiopulmonary exercise testing in individuals with myalgic encephalomyelitis/chronic fatigue syndrome. Work. (2020) 66:247–56.

Google Scholar

445. Cook D, Nagelkirk P, Poluri A, Mores J, Natelson B. The influence of aerobic fitness and fibromyalgia on cardiorespiratory and perceptual responses to exercise in patients with chronic fatigue syndrome. Arthritis Rheum. (2006) 54:3351–62. doi: 10.1002/art.22124

PubMed Abstract | CrossRef Full Text | Google Scholar

446. Cook D, VanRiper S, Dougherty R, Lindheimer J, Falvo M, Chen Y, et al. Cardiopulmonary, metabolic, and perceptual responses during exercise in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS): a multi-site clinical assessment of ME/CFS (MCAM) sub-study. PLos One. (2022) 17:e0265315. doi: 10.1371/journal.pone.0265315

PubMed Abstract | CrossRef Full Text | Google Scholar

447. Mancini D, Brunjes D, Lala A, Trivieri M, Contreras J, Natelson B. Use of cardiopulmonary stress testing for patients with unexplained dyspnea post-coronavirus disease. JACC Heart Fail. (2021) 9:927–37. doi: 10.1016/j.jchf.2021.10.002

PubMed Abstract | CrossRef Full Text | Google Scholar

448. Clavario P, De Marzo V, Lotti R, Barbara C, Porcile A, Russo C, et al. Cardiopulmonary exercise testing in COVID-19 patients at 3 months follow-up. Int J Cardiol. (2021) 340:113–8.

Google Scholar

449. Vonbank K, Lehmann A, Bernitzky D, Gysan M, Simon S, Schrott A, et al. Predictors of prolonged cardiopulmonary exercise impairment after COVID-19 infection: a prospective observational study. Front Med. (2021) 8:773788. doi: 10.3389/fmed.2021.773788

PubMed Abstract | CrossRef Full Text | Google Scholar

450. Vannini L, Quijada-Fumero A, Martin M, Pina N, Afonso J. Cardiopulmonary exercise test with stress echocardiography in COVID-19 survivors at 6 months follow-up. Eur J Intern Med. (2021) 94:101–4. doi: 10.1016/j.ejim.2021.10.004

PubMed Abstract | CrossRef Full Text | Google Scholar

451. Szekely Y, Lichter Y, Sadon S, Lupu L, Taieb P, Banai A, et al. Cardiorespiratory abnormalities in patients recovering from coronavirus disease 2019. J Am Soc Echocardiogr. (2021) 34:1273–84.e9.

Google Scholar

452. Skjorten I, Ankerstjerne O, Trebinjac D, Bronstad E, Rasch-Halvorsen O, Einvik G, et al. Cardiopulmonary exercise capacity and limitations 3 months after COVID-19 hospitalisation. Eur Respir J. (2021) 58:2100996.

Google Scholar

453. Rinaldo R, Mondoni M, Parazzini E, Baccelli A, Pitari F, Brambilla E, et al. Severity does not impact on exercise capacity in COVID-19 survivors. Respir Med. (2021) 187:106577.

Google Scholar

454. Rinaldo R, Mondoni M, Parazzini E, Pitari F, Brambilla E, Luraschi S, et al. Deconditioning as main mechanism of impaired exercise response in COVID-19 survivors. Eur Respir J. (2021) 58:2100870.

Google Scholar

455. Motiejunaite J, Balagny P, Arnoult F, Mangin L, Bancal C, Vidal-Petiot E, et al. Hyperventilation as one of the mechanisms of persistent dyspnoea in SARS-CoV-2 survivors. Eur Respir J. (2021) 58:2101578.

Google Scholar

456. Mohr A, Dannerbeck L, Lange T, Pfeifer M, Blaas S, Salzberger B, et al. Cardiopulmonary exercise pattern in patients with persistent dyspnoea after recovery from COVID-19. Multidisc Resp Med. (2021) 16:732. doi: 10.4081/mrm.2021.732

PubMed Abstract | CrossRef Full Text | Google Scholar

457. Debeaumont D, Boujibar F, Ferrand-Devouge E, Artaud-Macari E, Tamion F, Gravier F, et al. Cardiopulmonary exercise testing to assess persistent symptoms at 6 months in people with COVID-19 who survived hospitalization: a pilot study. Phys Ther. (2021) 101:1–9. doi: 10.1093/ptj/pzab099

PubMed Abstract | CrossRef Full Text | Google Scholar

458. Cassar M, Tunnicliffe E, Petousi N, Lewandowski A, Xie C, Mahmod M, et al. Symptom persistence despite improvement in cardiopulmonary health - Insights from longitudinal CMR, CPET and lung function testing post-COVID-19. EClinicalMedicine. (2021) 41:101159. doi: 10.1016/j.eclinm.2021.101159

PubMed Abstract | CrossRef Full Text | Google Scholar

459. Aparisi A, Ladron R, Ybarra-Falcon C, Tobar J, San Roman J. Exercise intolerance in post-acute sequelae of COVID-19 and the value of cardiopulmonary exercise testing- a mini-review. Front Med. (2022) 9:924819. doi: 10.3389/fmed.2022.924819

PubMed Abstract | CrossRef Full Text | Google Scholar

460. Margalit I, Yelin D, Sagi M, Rahat M, Sheena L, Mizrahi N, et al. Risk factors and multidimensional assessment of long coronavirus disease fatigue: a nested case-control study. Clin Infect Dis. (2022) 75:1688–97. doi: 10.1093/cid/ciac283

PubMed Abstract | CrossRef Full Text | Google Scholar

461. Longobardi I, Prado D, Goessler K, Meletti M, de Oliveira Junior G, de Andrade D, et al. Oxygen uptake kinetics and chronotropic responses to exercise are impaired in survivors of severe COVID-19. Am J Physiol Heart Circ Physiol. (2022) 323:H569–76. doi: 10.1152/ajpheart.00291.2022

PubMed Abstract | CrossRef Full Text | Google Scholar

462. Ladlow P, O’Sullivan O, Bennett A, Barker-Davies R, Houston A, Chamley R, et al. The effect of medium-term recovery status after COVID-19 illness on cardiopulmonary exercise capacity in a physically active adult population. J Appl Physiol. (2022) 132:1525–35. doi: 10.1152/japplphysiol.00138.2022

PubMed Abstract | CrossRef Full Text | Google Scholar

463. Ladlow P, O’Sullivan O, Houston A, Barker-Davies R, May S, Mills D, et al. Dysautonomia following COVID-19 is not associated with subjective limitations or symptoms but is associated with objective functional limitations. Heart Rhythm. (2022) 19:613–20.

Google Scholar

464. Singh I, Joseph P, Heerdt P, Cullinan M, Lutchmansingh D, Gulati M, et al. Persistent exertional intolerance after COVID-19. insights from invasive cardiopulmonary exercise testing. Chest. (2022) 161:54–63.

Google Scholar

465. Kimmig L, Rako Z, Ziegler S, Richter M, Ashkan Tolou GS, Roller F, et al. Long-term comprehensive cardiopulmonary phenotyping of COVID-19. Respir Res. (2022) 23:263.

Google Scholar

466. Alba G, Ziehr D, Rouvina J, Hariri L, Knipe R, Medoff B, et al. Exercise performance in patients with post-acute sequelae of SARS-CoV-2 infection compared to patients with unexplained dyspnea. EClinicalMedicine. (2021) 39:101066.

Google Scholar

467. Wood G, Kirkevang T, Agergaard J, Leth S, Hansen E, Laustsen C, et al. Cardiac performance and cardiopulmonary fitness after infection with SARS-CoV-2. Front Cardiovasc Med. (2022) 9:871603. doi: 10.3389/fcvm.2022.871603

PubMed Abstract | CrossRef Full Text | Google Scholar

468. van Voorthuizen E, van Helvoort H, Peters J, van den Heuvel M, van den Borst B. Persistent exertional dyspnea and perceived exercise intolerance after mild COVID-19: a critical role for breathing dysregulation? Phys Ther. (2022) 102:zac105.

Google Scholar

469. Kersten J, Hoyo L, Wolf A, Hull E, Nunn S, Tadic M, et al. Cardiopulmonary exercise testing distinguishes between post-COVID-19 as a dysfunctional syndrome and organ pathologies. Int J Environ Res Public Health. (2022) 19:11421.

Google Scholar

470. Beaudry R, Brotto A, Varughese R, de Waal S, Fuhr D, Damant R, et al. Persistent dyspnea after COVID-19 is not related to cardiopulmonary impairment; a cross-sectional study of persistently dyspneic COVID-19, non-dyspneic COVID-19 and controls. Front Physiol. (2022) 13:917886. doi: 10.3389/fphys.2022.917886

PubMed Abstract | CrossRef Full Text | Google Scholar

471. Durstenfeld M, Sun K, Tahir P, Peluso M, Deeks S, Aras M, et al. Use of cardiopulmonary exercise testing to evaluate long COVID-19 symptoms in adults: a systematic review and meta-analysis. JAMA Netw Open. (2022) 5:e2236057.

Google Scholar

472. Jones D, Hollingsworth K, Jakovljevic D, Fattakhova G, Pairman J, Blamire A, et al. Loss of capacity to recover from acidosis on repeat exercise in chronic fatigue syndrome: a case-control study. Eur J Clin Invest. (2012) 42:186–94. doi: 10.1111/j.1365-2362.2011.02567.x

PubMed Abstract | CrossRef Full Text | Google Scholar

473. Lien K, Johansen B, Veierod M, Haslestad A, Bohn S, Melsom M, et al. Abnormal blood lactate accumulation during repeated exercise testing in myalgic encephalomyelitis/chronic fatigue syndrome. Physiol Rep. (2019) 7:e14138.

Google Scholar

474. Germain A, Giloteaux L, Moore G, Levine S, Chia J, Keller B, et al. Plasma metabolomics reveals disrupted response and recovery following maximal exercise in myalgic encephalomyelitis/chronic fatigue syndrome. JCI Insight. (2022) 7:e157621.

Google Scholar

475. Van Booven D, Gamer J, Joseph A, Perez M, Zarnowski O, Pandya M, et al. Stress-induced transcriptomic changes in females with myalgic encephalomyelitis/chronic fatigue syndrome reveal disrupted immune signatures. Int J Mol Sci. (2023) 24:2698.

Google Scholar

476. Tsilioni I, Natelson B, Theoharides T. Exosome-associated mitochondrial DNA from patients with myalgic encephalomyelitis/chronic fatigue syndrome stimulates human microglia to release IL-1beta. Eur J Neurosci. (2022) 56:5784–94. doi: 10.1111/ejn.15828

PubMed Abstract | CrossRef Full Text | Google Scholar

477. Shukla S, Cook D, Meyer J, Vernon S, Le T, Clevidence D, et al. Changes in gut and plasma microbiome following exercise challenge in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). PLos One. (2015) 10:e0145453. doi: 10.1371/journal.pone.0145453

PubMed Abstract | CrossRef Full Text | Google Scholar

478. Joseph P, Pari R, Miller S, Warren A, Stovall M, Squires J, et al. Neurovascular dysregulation and acute exercise intolerance in myalgic encephalomyelitis/chronic fatigue syndrome: a randomized, placebo-controlled trial of pyridostigmine. Chest. (2022) 162:1116–26. doi: 10.1016/j.chest.2022.04.146

PubMed Abstract | CrossRef Full Text | Google Scholar

479. Streeten D, Thomas D, Bell D. The roles of orthostatic hypotension, orthostatic tachycardia, and subnormal erythrocyte volume in the pathogenesis of the chronic fatigue syndrome. Am J Med Sci. (2000) 320:1–8. doi: 10.1097/00000441-200007000-00001

PubMed Abstract | CrossRef Full Text | Google Scholar

480. Farquhar W, Hunt B, Taylor J, Darling S, Freeman R. Blood volume and its relation to peak O2 consumption and physical activity in patients with chronic fatigue. Am J Physiol Heart Circ Physiol. (2002) 282:H66–71. doi: 10.1152/ajpheart.2002.282.1.H66

PubMed Abstract | CrossRef Full Text | Google Scholar

481. Stewart J. Chronic fatigue syndrome: comments on deconditioning, blood volume and resulting cardiac function. Clin Sci. (2009) 118:121–3.

Google Scholar

482. Hurwitz B, Coryell V, Parker M, Martin P, Laperriere A, Klimas N, et al. Chronic fatigue syndrome: illness severity, sedentary lifestyle, blood volume and evidence of diminished cardiac function. Clin Sci (Lond). (2010) 118:125–35.

Google Scholar

483. Newton J, Finkelmeyer A, Petrides G, Frith J, Hodgson T, Maclachlan L, et al. Reduced cardiac volumes in chronic fatigue syndrome associate with plasma volume but not length of disease: a cohort study. Open Heart. (2016) 3:e000381.

Google Scholar

484. van Campen C, Rowe P, Visser F. Blood volume status in ME/CFS correlates with the presence or absence of orthostatic symptoms: preliminary results. Front Pediatr. (2018) 6:352. doi: 10.3389/fped.2018.00352

PubMed Abstract | CrossRef Full Text | Google Scholar

485. Moore Y, Serafimova T, Anderson N, King H, Richards A, Brigden A, et al. Recovery from chronic fatigue syndrome: a systematic review-heterogeneity of definition limits study comparison. Arch Dis Child. (2021) 106:1087–94. doi: 10.1136/archdischild-2020-320196

PubMed Abstract | CrossRef Full Text | Google Scholar

486. Newton D, Kennedy G, Chan K, Lang C, Belch J, Khan F. Large and small artery endothelial dysfunction in chronic fatigue syndrome. Int J Cardiol. (2012) 154:335–6.

Google Scholar

487. Scherbakov N, Szklarski M, Hartwig J, Sotzny F, Lorenz S, Meyer A, et al. Peripheral endothelial dysfunction in myalgic encephalomyelitis/chronic fatigue syndrome. ESC Heart Fail. (2020) 7:1064–71.

Google Scholar

488. Fluge Ø, Tronstad KJ, Mella O. Pathomechanisms and possible interventions in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). J Clin Invest (2021) 131:e150377.

Google Scholar

489. Blauensteiner J, Bertinat R, León L, Riederer M, Sepúlveda N, Westermeier F. Altered endothelial dysfunction-related miRs in plasma from ME/CFS patients. Sci Rep. (2021) 11:10604.

Google Scholar

490. Bond J, Nielsen T, Hodges L. Effects of post-exertional malaise on markers of arterial stiffness in individuals with myalgic encephalomyelitis/chronic fatigue syndrome. Int J Environ Res Public Health. (2021) 18:2366. doi: 10.3390/ijerph18052366

PubMed Abstract | CrossRef Full Text | Google Scholar

491. Sørland K, Sandvik M, Rekeland I, Ribu L, Småstuen M, Mella O, et al. Reduced endothelial function in myalgic encephalomyelitis/chronic fatigue syndrome–results from open-label cyclophosphamide intervention study. Front Med. (2021) 8:642710. doi: 10.3389/fmed.2021.642710

PubMed Abstract | CrossRef Full Text | Google Scholar

492. Bertinat R, Villalobos-Labra R, Hofmann L, Blauensteiner J, Sepulveda N, Westermeier F. Decreased NO production in endothelial cells exposed to plasma from ME/CFS patients. Vascul Pharmacol. (2022) 143:106953. doi: 10.1016/j.vph.2022.106953

PubMed Abstract | CrossRef Full Text | Google Scholar

493. Lubell J. Letter: could endothelial dysfunction and vascular damage contribute to pain, inflammation and post-exertional malaise in individuals with myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS)? J Transl Med. (2022) 20:40. doi: 10.1186/s12967-022-03244-7

PubMed Abstract | CrossRef Full Text | Google Scholar

494. Nunes J, Kruger A, Proal A, Kell D, Pretorius E. The occurrence of hyperactivated platelets and fibrinaloid microclots in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). Pharmaceuticals (Basel). (2022) 15:931. doi: 10.3390/ph15080931

PubMed Abstract | CrossRef Full Text | Google Scholar

495. Sandvik M, Sorland K, Leirgul E, Rekeland I, Stavland C, Mella O, et al. Endothelial dysfunction in ME/CFS patients. PLos One. (2023) 18:e0280942. doi: 10.1371/journal.pone.0280942

PubMed Abstract | CrossRef Full Text | Google Scholar

496. Libby P, Lüscher T. COVID-19 is, in the end, an endothelial disease. Eur Heart J. (2020) 41:3038–44.

Google Scholar

497. Rutkai I, Mayer M, Hellmers L, Ning B, Huang Z, Monjure C, et al. Neuropathology and virus in brain of SARS-CoV-2 infected non-human primates. Nat Commun. (2022) 13:1745.

Google Scholar

498. Oikonomou E, Souvaliotis N, Lampsas S, Siasos G, Poulakou G, Theofilis P, et al. Endothelial dysfunction in acute and long standing COVID-19: a prospective cohort study. Vascul Pharmacol. (2022) 144:106975.

Google Scholar

499. Fogarty H, Townsend L, Morrin H, Ahmad A, Comerford C, Karampini E, et al. Persistent endotheliopathy in the pathogenesis of long COVID syndrome. J Thromb Haemost. (2021) 19:2546–53.

Google Scholar

500. Ambrosino P, Calcaterra I, Molino A, Moretta P, Lupoli R, Spedicato G, et al. Persistent endothelial dysfunction in post-acute COVID-19 syndrome: a case-control study. Biomedicines. (2021) 9:957. doi: 10.3390/biomedicines9080957

PubMed Abstract | CrossRef Full Text | Google Scholar

501. Santoro L, Falsetti L, Zaccone V, Nesci A, Tosato M, Giupponi B, et al. Impaired endothelial function in convalescent phase of COVID-19: a 3 month follow up observational prospective study. J Clin Med. (2022) 11:1774. doi: 10.3390/jcm11071774

PubMed Abstract | CrossRef Full Text | Google Scholar

502. Patel M, Knauer M, Nicholson M, Daley M, Van Nynatten L, Martin C, et al. Elevated vascular transformation blood biomarkers in long-COVID indicate angiogenesis as a key pathophysiological mechanism. Mol Med. (2022) 28:122. doi: 10.1186/s10020-022-00548-8

PubMed Abstract | CrossRef Full Text | Google Scholar

503. Flaskamp L, Roubal C, Uddin S, Sotzny F, Kedor C, Bauer S, et al. Serum of post-COVID-19 syndrome patients with or without ME/CFS differentially affects endothelial cell function in vitro. Cells. (2022) 11:2376.

Google Scholar

504. Kruger A, Vlok M, Turner S, Venter C, Laubscher G, Kell D, et al. Proteomics of fibrin amyloid microclots in long COVID/post-acute sequelae of COVID-19 (PASC) shows many entrapped pro-inflammatory molecules that may also contribute to a failed fibrinolytic system. Cardiovasc Diabetol. (2022) 21:190.

Google Scholar

505. Beltran-Camacho L, Eslava-Alcon S, Rojas-Torres M, Sanchez-Morillo D, Martinez-Nicolas M, Martin-Bermejo V, et al. The serum of COVID-19 asymptomatic patients up-regulates proteins related to endothelial dysfunction and viral response in circulating angiogenic cells ex-vivo. Mol Med. (2022) 28:40.

Google Scholar

506. Haffke M, Freitag H, Rudolf G, Seifert M, Doehner W, Scherbakov N, et al. Endothelial dysfunction and altered endothelial biomarkers in patients with post-COVID-19 syndrome and chronic fatigue syndrome (ME/CFS). J Transl Med. (2022) 20:138. doi: 10.1186/s12967-022-03346-2

PubMed Abstract | CrossRef Full Text | Google Scholar

507. Marcic M, Marcic L, Lovric Kojundzic S, Marinovic Guic M, Marcic B, Caljkusic K. Chronic endothelial dysfunction after COVID-19 infection shown by transcranial color-coded Doppler: a cross-sectional study. Biomedicines. (2022) 10:2550. doi: 10.3390/biomedicines10102550

PubMed Abstract | CrossRef Full Text | Google Scholar

508. Tosato M, Calvani R, Picca A, Ciciarello F, Galluzzo V, Coelho-Junior H, et al. Effects of L-arginine plus vitamin C supplementation on physical performance, endothelial function, and persistent fatigue in adults with long COVID: a single-blind randomized controlled trial. Nutrients. (2022) 14:4984. doi: 10.3390/nu14234984

PubMed Abstract | CrossRef Full Text | Google Scholar

509. Kell D, Laubscher G, Pretorius E. A central role for amyloid fibrin microclots in long COVID/PASC: origins and therapeutic implications. Biochem J. (2022) 479:537–59. doi: 10.1042/BCJ20220016

PubMed Abstract | CrossRef Full Text | Google Scholar

510. Grobbelaar L, Kruger A, Venter C, Burger E, Laubscher G, Maponga T, et al. Relative hypercoagulopathy of the SARS-CoV-2 beta and delta variants when compared to the less severe omicron variants is related to TEG parameters, the extent of fibrin amyloid microclots, and the severity of clinical illness. Semin Thromb Hemost. (2022) 48:858–68. doi: 10.1055/s-0042-1756306

PubMed Abstract | CrossRef Full Text | Google Scholar

511. Groff D, Sun A, Ssentongo A, Ba D, Parsons N, Poudel G, et al. Short-term and long-term rates of postacute sequelae of SARS-CoV-2 infection: a systematic review. JAMA Netw Open. (2021) 4:e2128568.

Google Scholar

512. Huang L, Li X, Gu X, Zhang H, Ren L, Guo L, et al. Health outcomes in people 2 years after surviving hospitalisation with COVID-19: a longitudinal cohort study. Lancet Respiratory Med. (2022) 10:863–76.

Google Scholar

513. González-Hermosillo J, Martínez-López J, Carrillo-Lampón S, Ruiz-Ojeda D, Herrera-Ramírez S, Amezcua-Guerra L, et al. Post-acute COVID-19 symptoms, a potential link with myalgic encephalomyelitis/chronic fatigue syndrome: a 6-month survey in a Mexican cohort. Brain Sci. (2021) 11:760. doi: 10.3390/brainsci11060760

PubMed Abstract | CrossRef Full Text | Google Scholar

514. Mirfazeli F, Sarabi-Jamab A, Pereira-Sanchez V, Kordi A, Shariati B, Shariat S, et al. Chronic fatigue syndrome and cognitive deficit are associated with acute-phase neuropsychiatric manifestations of COVID-19: a 9-month follow-up study. Neurol Sci. (2022) 43:2231–9.

Google Scholar

515. Bonilla H, Quach T, Tiwari A, Bonilla A, Miglis M, Yang P, et al. Myalgic encephalomyelitis/chronic fatigue syndrome is common in post-acute sequelae of SARS-CoV-2 infection (PASC): results from a post-COVID-19 multidisciplinary clinic. Front Neurol. (2023) 14:1090747. doi: 10.3389/fneur.2023.1090747

PubMed Abstract | CrossRef Full Text | Google Scholar

516. Wessely S. History of postviral fatigue syndrome. Br Med Bull. (1991) 47: 919–41.

Google Scholar

517. Paul O. DaCosta’s syndrome or neurocirculatory asthenia. Br Heart J. (1987) 58:306–15.

Google Scholar

518. Komaroff A, Buchwald D. Symptoms and signs of chronic fatigue syndrome. Rev Infect Dis. (1991) 13:S8–11.

Google Scholar

519. Shelokov A, Habel K, Verder E, Welsh W. Epidemic neuromyasthenia: an outbreak of poliomyelitislike illness in student nurses. N Engl J Med. (1957) 257:345–55. doi: 10.1056/NEJM195708222570801

PubMed Abstract | CrossRef Full Text | Google Scholar

520. Poskanzer D, Henderson D, Kunkle E, Kalter S, Clement W, Bond J. Epidemic neuromyasthenia: an outbreak in Punta Gorda. Florida. N Engl J Med. (1957) 257: 356–64.

Google Scholar

521. Acheson ED. The clinical syndrome variously called benign myalgic encephalomyelitis, Iceland disease and epidemic neuromyasthenia. Am J Med. (1959) 4:569–95. doi: 10.1016/0002-9343(59)90280-3

PubMed Abstract | CrossRef Full Text | Google Scholar

522. Henderson D, Shelokov A. Epidemic neuromyasthenia – clinical syndrome. N Engl J Med. (1959) 260:757–64.

Google Scholar

523. Levine P, Jacobson S, Pocinki A, Cheney P, Peterson D, Connelly R, et al. Clinical, epidemiologic, and virologic studies in four clusters of the chronic fatigue syndrome. Arch Intern Med. (1992) 152:1611–6.

Google Scholar

524. Medical staff of the Royal Free Hospital. An outbreak of encephalomyelitis in the Royal Free Hospital Group, London, in l955. BMJ. (1957) 2:895–904.

Google Scholar

525. Komaroff A, Fagioli L, Doolittle T, Gandek B, Gleit M, Guerriero R, et al. Health status in patients with chronic fatigue syndrome and in general population and disease comparison groups. Am J Med. (1996) 101:281–90.

Google Scholar

526. Buchwald D, Pearlman T, Umali J, Schmaling K, Katon W. Functional status in patients with chronic fatigue syndrome, other fatiguing illnesses, and healthy individuals. Am J Med. (1996) 171:364–70.

Google Scholar

527. Raman B, Cassar M, Tunnicliffe E, Filippini N, Griffanti L, Alfaro-Almagro F, et al. Medium-term effects of SARS-CoV-2 infection on multiple vital organs, exercise capacity, cognition, quality of life and mental health, post-hospital discharge. EClinicalMedicine. (2021) 31:100683.

Google Scholar

528. Simmons G, Glynn S, Komaroff A, Mikovits J, Tobler L, Hackett J Jr., et al. Failure to confirm XMRV/MLVs in the blood of patients with chronic fatigue syndrome: a multi-laboratory study. Science. (2011) 334:814–7.

Google Scholar

529. Alter H, Mikovits J, Switzer W, Ruscetti F, Lo S, Klimas N, et al. A multicenter blinded analysis indicates no association between chronic fatigue syndrome/myalgic encephalomyelitis and either xenotropic murine leukemia virus-related virus or polytropic murine leukemia virus. mBio. (2012) 3:e266–212.

Google Scholar

530. Evengård B, Briese T, Lindh G, Lee S, Lipkin W. Absence of evidence of Borna disease virus infection in Swedish patients with chronic fatigue syndrome. J Neurovirol. (1999) 5:495–9.

Google Scholar

531. Dansie E, Furberg H, Afari N, Buchwald D, Edwards K, Goldberg J, et al. Conditions comorbid with chronic fatigue in a population-based sample. Psychosomatics. (2012) 53:44–50. doi: 10.1016/j.psym.2011.04.001

PubMed Abstract | CrossRef Full Text | Google Scholar

532. Bateman L, Bested A, Bonilla H, Chheda B, Chu L, Curtin J, et al. Myalgic encephalomyelitis/chronic fatigue syndrome: essentials of diagnosis and management. Mayo Clin Proc. (2021) 96:2861–78.

Google Scholar

533. Davis H, Assaf G, McCorkell L, Wei H, Low R, Re’em Y, et al. Characterizing long COVID in an international cohort: 7 months of symptoms and their impact. EClinicalMedicine. (2021) 38:101019. doi: 10.1016/j.eclinm.2021.101019

PubMed Abstract | CrossRef Full Text | Google Scholar

534. Vernon S, Hartle M, Sullivan K, Bell J, Abbaszadeh S, Unutmaz D, et al. Post-exertional malaise among people with long COVID compared to myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). Work. (2023) 74:1179–86. doi: 10.3233/WOR-220581

PubMed Abstract | CrossRef Full Text | Google Scholar

535. Dennis A, Cuthbertson D, Wootton D, Crooks M, Gabbay M, Eichert N, et al. Multi-organ impairment and long COVID: a 1-year prospective, longitudinal cohort study. J R Soc Med. (2023) 116:97–112. doi: 10.1177/01410768231154703

PubMed Abstract | CrossRef Full Text | Google Scholar

536. Nacul L, O’Boyle S, Palla L, Nacul F, Mudie K, Kingdon C, et al. How myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) progresses: the natural history of ME/CFS. Front Neurol. (2020) 11:826. doi: 10.3389/fneur.2020.00826

PubMed Abstract | CrossRef Full Text | Google Scholar

537. Ferrucci R, Dini M, Rosci C, Capozza A, Groppo E, Reitano M, et al. One-year cognitive follow-up of COVID-19 hospitalized patients. Eur J Neurol. (2022) 29:2006–14.

Google Scholar

538. Wild C, Norton L, Menon D, Ripsman D, Swartz R, Owen A. Disentangling the cognitive, physical, and mental health sequelae of COVID-19. Cell Rep Med. (2022) 3:100750. doi: 10.1016/j.xcrm.2022.100750

PubMed Abstract | CrossRef Full Text | Google Scholar

539. Ortelli P, Benso F, Ferrazzoli D, Scarano I, Saltuari L, Sebastianelli L, et al. Global slowness and increased intra-individual variability are key features of attentional deficits and cognitive fluctuations in post COVID-19 patients. Sci Rep. (2022) 12:13123. doi: 10.1038/s41598-022-17463-x

PubMed Abstract | CrossRef Full Text | Google Scholar

540. Holdsworth D, Chamley R, Barker-Davies R, O’Sullivan O, Ladlow P, Mitchell J, et al. Comprehensive clinical assessment identifies specific neurocognitive deficits in working-age patients with long-COVID. PLos One. (2022) 17:e0267392. doi: 10.1371/journal.pone.0267392

PubMed Abstract | CrossRef Full Text | Google Scholar

541. Douaud G, Lee S, Alfaro-Almagro F, Arthofer C, Wang C, McCarthy P, et al. SARS-CoV-2 is associated with changes in brain structure in UK Biobank. Nature. (2022) 604:697–707.

Google Scholar

542. Stanculescu D, Bergquist J. Perspective: drawing on findings from critical illness to explain myalgic encephalomyelitis/chronic fatigue syndrome. Front Med. (2022) 9:818728. doi: 10.3389/fmed.2022.818728

PubMed Abstract | CrossRef Full Text | Google Scholar

543. Meinhardt J, Radke J, Dittmayer C, Franz J, Thomas C, Mothes R, et al. Olfactory transmucosal SARS-CoV-2 invasion as a port of central nervous system entry in individuals with COVID-19. Nat Neurosci. (2020) 24:168–75.

Google Scholar

544. Choutka J, Jansari V, Hornig M, Iwasaki A. Unexplained post-acute infection syndromes. Nat Med. (2022) 28:911–23.

Google Scholar

545. Preiser J, Ichai C, Orban J, Groeneveld A. Metabolic response to the stress of critical illness. Br J Anaesth. (2014) 113:945–54.

Google Scholar

546. Stanculescu D, Larsson L, Bergquist J. Hypothesis: mechanisms that prevent recovery in prolonged ICU patients also underlie myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). Front Med. (2021) 8:628029. doi: 10.3389/fmed.2021.628029

PubMed Abstract | CrossRef Full Text | Google Scholar

547. Stanculescu D, Sepulveda N, Lim C, Bergquist J. Lessons from heat stroke for understanding myalgic encephalomyelitis/chronic fatigue syndrome. Front Neurol. (2021) 12:789784. doi: 10.3389/fneur.2021.789784

PubMed Abstract | CrossRef Full Text | Google Scholar

548. Komaroff A, Lipkin W. Insights from myalgic encephalomyelitis/chronic fatigue syndrome may help unravel the pathogenesis of postacute COVID-19 syndrome. Trends Mol Med. (2021) 27:895–906.

Google Scholar

549. Paul B, Lemle M, Komaroff A, Snyder S. Redox imbalance links COVID-19 and myalgic encephalomyelitis/chronic fatigue syndrome. Proc Natl Acad Sci USA. (2021) 118:e2024358118.

Google Scholar

550. Dantzer R, Heijnen C, Kavelaars A, Laye S, Capuron L. The neuroimmune basis of fatigue. Trends Neurosci. (2014) 37:39–46.

Google Scholar

551. Miller A, Haroon E, Felger J. The immunology of behavior-Exploring the role of the immune system in brain health and illness. Neuropsychopharmacology. (2017) 42:1–4.

Google Scholar

552. Raizen D, Mullington J, Anaclet C, Clarke G, Critchley H, Dantzer R, et al. Beyond the symptom: the biology of fatigue. Sleep (2023) (in press).

Google Scholar

553. Wang T, Cao Y, Zhang H, Wang Z, Man C, Yang Y, et al. COVID-19 metabolism: mechanisms and therapeutic targets. MedComm (2020). (2022) 3:e157.

Google Scholar

554. Storey K, Storey J. Metabolic rate depression: the biochemistry of mammalian hibernation. Adv Clin Chem. (2010) 52:77–108.

Google Scholar

555. Tessier S, Storey K. Transitioning between entry and exit from mammalian torpor: the involvement of signal transduction pathways. Temperature (Austin). (2014) 1:92–3. doi: 10.4161/temp.29972

PubMed Abstract | CrossRef Full Text | Google Scholar

556. Drew K, Buck C, Barnes B, Christian S, Rasley B, Harris M. Central nervous system regulation of mammalian hibernation: implications for metabolic suppression and ischemia tolerance. J Neurochem. (2007) 102:1713–26.

Google Scholar

557. Bargmann C, Horvitz H. Control of larval development by chemosensory neurons in Caenorhabditis elegans. Science. (1991) 251:1243–6. doi: 10.1126/science.2006412

PubMed Abstract | CrossRef Full Text | Google Scholar

558. Wang Y, Hou X. Dauer in nematodes as a way to persist or obviate. Nematropica. (2015) 45:128–37.

Google Scholar

559. Penkov S, Kaptan D, Erkut C, Sarov M, Mende F, Kurzchalia T. Integration of carbohydrate metabolism and redox state controls dauer larva formation in Caenorhabditis elegans. Nat Commun. (2015) 6:8060. doi: 10.1038/ncomms9060

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: chronic fatigue syndrome (CFS), myalgic encephalomyelitis (ME), myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS), Long COVID, post-acute sequelae of SARS-CoV-2 infection, post-infectious fatigue syndrome, post-intensive care unit syndrome

Citation: Komaroff AL and Lipkin WI (2023) ME/CFS and Long COVID share similar symptoms and biological abnormalities: road map to the literature. Front. Med. 10:1187163. doi: 10.3389/fmed.2023.1187163

Received: 15 March 2023; Accepted: 09 May 2023;
Published: 02 June 2023.

Edited by:

Ramcés Falfán-Valencia, National Institute of Respiratory Diseases-Mexico (INER), Mexico

Reviewed by:

Jeremy S. Rossman, Research-Aid Networks, United States
Johannes Fessler, Medical University of Graz, Austria

Copyright © 2023 Komaroff and Lipkin. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Anthony L. Komaroff, komaroff@hms.harvard.edu

Download