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

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.

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.

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.

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.

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.

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.

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.

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.

留言 (0)

沒有登入
gif