Next Article in Journal
Possible Roles for Purinergic Receptor P2RX4 in Breast and Prostate Cancers
Previous Article in Journal
TGFBI Facilitates Myogenesis and Limits Fibrosis in Mouse Skeletal Muscle Regeneration
Previous Article in Special Issue
Expanding the Spectrum of Selective IgM Deficiency: From Infections to Immune Dysregulation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Gulf War Illness, Fibromyalgia, Myalgic Encephalomyelitis/Chronic Fatigue Syndrome and Long COVID Overlap in Common Symptoms and Underlying Biological Mechanisms: Implications for Future Therapeutic Strategies

by
David Mantle
1,†,
Joan Carles Domingo
2,†,
Beatrice Alexandra Golomb
3 and
Jesús Castro-Marrero
2,4,*
1
Pharma Nord (UK) Ltd., Morpeth NE61 2DB, Northumberland, UK
2
Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, 08028 Barcelona, Spain
3
Department of Medicine, UC San Diego School of Medicine, La Jolla, CA 92093, USA
4
Research Unit in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome and Long COVID, Division of Rheumatology Research, Vall d’Hebron Hospital Research Institute, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2025, 26(18), 9044; https://doi.org/10.3390/ijms26189044
Submission received: 21 August 2025 / Revised: 7 September 2025 / Accepted: 15 September 2025 / Published: 17 September 2025
(This article belongs to the Special Issue New Insights into Immune Dysregulation Disorders)

Abstract

Although Gulf War Illness (GWI), fibromyalgia (FM), myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) and long COVID have distinct origins, in this article we have reviewed evidence that these disorders comprise a group of so-called low-energy associated disorders with overlapping common symptoms underlying pathology. In particular, evidence for mitochondrial dysfunction, oxidative stress, inflammation, immune dysregulation, neuroendocrine dysfunction, disrupted brain–gut-microbiome axis, apoptosis/ferroptosis and telomere shortening as common features in the pathogenesis of these disorders has been identified. Given the role of coenzyme Q10 (CoQ10) in promoting normal mitochondrial function, as an antioxidant, antiinflammatory and antiapoptotic and antiferroptotic agent, there is a rationale for supplementary CoQ10 in the management of these disorders. The reported benefits of supplementary CoQ10 administration in GWI, FM, ME/CFS and long COVID have been reviewed; the potential benefit of supplementary CoQ10 in reducing telomere shortening and improving the efficiency of stem cell transfer relevant has also been identified as promising therapeutic strategies in these disorders. This review advances beyond previous systematic reviews and consensus statements on overlapping similar symptoms and underlying biological pathomechanisms in these complex disorders.

1. Introduction

Gulf War Illness (GWI), fibromyalgia (FM), myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS), and long COVID are chronic, multifaceted, and poorly understood disabling disorders affecting millions of patients worldwide. The increasing prevalence of these disorders in the general population has been estimated as 2–6% for FM [1], 0.5–1% for ME/CFS [2], 6–10% for long COVID [3], predominantly more common in women than in men, with 25–35% of Gulf War veterans affected by GWI [4]. It should be noted that prevalence rates for these disorders vary somewhat between different epidemiological studies, and the figures quoted are estimates based on data from several sources.
These disorders share significant overlapping common symptoms, underlying biological pathomechanisms, and impacts on daily life, often resulting in similar management strategies [4,5,6]. These disorders present challenges in medical practice with regard to clinical diagnosis; whilst there are some specific diagnostic criteria for GWI (the Kansas case criteria, together with the less-restrictive Centers for Disease Control and Prevention (CDC) clinical definition), the diagnostic objectivity requires further improvement [7,8].
In this review we have compared what is known about the similarities and differences in shared symptoms but distinct origins and updated case criteria in GWI, FM, ME/CFS, and long COVID. Gulf War Illness, affecting veterans of the 1990–91 Gulf War, includes fatigue, pain, and cognitive issues linked to environmental exposure and genetic susceptibility [4]. Fibromyalgia, historically misunderstood, was formally recognized in 1990 with diagnostic criteria focusing on widespread pain and tender points, later revised to include fatigue and cognitive symptoms [9]. ME/CFS, dating back to the 19th century, gained prominence in the 1980s; its 2015 diagnostic criteria require post-exertional malaise, unrefreshing sleep, and cognitive or autonomic dysfunction [10]. Long COVID emerged in 2020 following acute SARS-CoV-2 infection, presenting with fatigue, brain fog, and multisystem symptoms lasting beyond four weeks, often resembling ME/CFS [11]. While all four disorders rely on clinical evaluation and symptom duration; their histories reflect evolving recognition, from initial skepticism to growing acceptance, with long COVID now driving research that may illuminate shared biological pathomechanisms across these neglected complex disorders.
In addition, there are no clear objective case criteria or specific biomarkers, nor any effective Food and Drug Administration (FDA)-approved treatments, with the exception of four drugs currently available for treating FM (duloxetine, pregabalin, milnacipran, and recently cyclobenzaprine). The above issues lead to variability in diagnosis, in monitoring illness severity, in predicting treatment response and in developing targeted therapies for these disorders, in turn highlighting a significant unmet medical need [4,5,6].
All of these disorders are characterized by common symptoms [4,11,12,13]; in particular persistent fatigue, post-exertional malaise (PEM) as a hallmark symptom that is not alleviated by rest, that often severely influences daily functioning. Other common symptoms include: (i) cognitive dysfunction (commonly referred to as brain fog), including problems with memory and concentration, and mental clarity, is prevalent in all four disorders; (ii) widespread musculoskeletal pain, defined as chronic pain especially in muscles and joints, is a hallmark symptom of FM and is commonly seen also in patients with GWI, ME/CFS and long COVID; (iii) sleep disturbance, including difficulty sleeping, unrefreshing sleep, and disrupted sleep patterns are seen in all these disorders; (iv) autonomic dysfunction occurs in many of these disorders, including dysregulation of the autonomic nervous system, leading to autonomic symptoms such as dizziness, light-headedness, orthostatic intolerance, and tachycardia; and (v) muscle weakness, particularly in FM, ME/CFS, and long COVID, is reported frequently, along with a general feeling of exhaustion that is disproportionate to physical and mental activity.
A growing body of literature has reported overlapping potential biological mechanisms in these disorders [14,15]. The pathophysiology of these disorders is still being explored, but recent research points to several shared biological mechanisms that may underlie the clinical symptoms. In terms of pathology, these disorders all show evidence of immune dysfunction, and at the cellular level there is evidence for an impaired mitochondrial function, increased oxidative stress, inflammation, apoptosis/ferroptosis and telomere shortening, as discussed in subsequent sections of this article [15,16]. Thus, GWI, FM, ME/CFS and long COVID comprise a group of so-called low-energy disorders with overlapping common symptoms and underlying pathophysiological mechanisms. Low-energy associated disorders are characterized by primary disabling fatigue that reduces the body’s capacity to perform daily activities, affecting both physical and mental functions.
Given the key role of CoQ10 in promoting normal mitochondrial function, as an antioxidant, antiinflammatory and antiapoptotic and antiferroptotic agent, in this article we have reviewed the potential role of supplemental CoQ10 in the therapeutic management of these disorders. A potential role for supplemental CoQ10 in reducing telomere shortening, and in improving the efficiency of stem cell therapy of relevance to these disorders has also been reviewed [17,18,19].

2. Common Symptoms in Low-Energy Associated Disorders

2.1. Fatigue and Post-Exertional Malaise

Debilitating fatigue and post-exertional malaise (PEM) are not the same, although they share some similarities. Debilitating fatigue is a broad term for extreme and severe tiredness that can be a symptom of many chronic illnesses, while PEM, also known as post-exertional symptom exacerbation (PESE) is a hallmark symptom of ME/CFS and sometimes GWI and long COVID, involving a worsening of symptoms following even minimal physical, and/or mental activity [6,20,21].
Post-exertional malaise can exacerbate a wide range of core symptoms, including fatigue, cognitive difficulties (brain fog), pain, sleep impairments and sensitivity to light and sound in these disorders. It can severely limit a person’s ability to participate in daily activities, making it difficult to work, attend school, or engage in social activities. The key is that the severity of symptoms is often disproportionate to the exertion that triggered it, and can be delayed, sometimes appearing 24–72 h after exertion, and may persist for months. Recovery from PEM can be slow and prolonged, potentially lasting for days, weeks, or even months [3,22].
Managing PEM often involves pacing, which is a strategy of balancing activity and rest to avoid triggering PEM episodes. PEM management is highly individualized, and what works for one person may not work for another [20,23].
In summary, PEM is a more specific symptom of fatigue, often described as a crash or flare that is a hallmark feature of GWI, ME/CFS and long COVID and is distinct from general fatigue. Effective management involves pacing and personalized specific symptom-based strategies to minimize its impact on daily life is urgently warranted.

2.2. Autonomic Dysfunction

Autonomic dysfunction (dysautonomia) results from damage to nerves of the autonomic nervous system, which controls automatic body functions. As noted in the Introduction, autonomic dysfunction has been increasingly recognized as a shared clinical feature across GWI, FM, ME/CFS, and long COVID [24,25,26].
While each of these disorders has a distinct origin, they converge in exhibiting abnormalities in autonomic nervous system regulation, particularly involving sympathetic and parasympathetic nervous system imbalance, orthostatic intolerance, heart rate variability, and blood pressure dysregulation [24,26]. In Gulf War veterans, autonomic dysfunction (particularly affecting cholinergic pathways) manifesting as dizziness, postural orthostatic tachycardia syndrome (POTS) and gastrointestinal dysfunction has also been reported [27,28,29].
Dysautonomia is a common feature of ME/CFS, particularly with regard to orthostatic intolerance, gastrointestinal problems, and body thermoregulation [30,31]. Patients with fibromyalgia also experience dysautonomia related symptoms of orthostatic intolerance, gastrointestinal dysfunction and problems with temperature regulation [32,33,34]. In long COVID patients, dysautonomia related symptoms including orthostatic intolerance, breathlessness and palpitations have been described, resulting from virus induced disruption of the autonomic nervous system [35,36,37]. In GWI and ME/CFS, autonomic dysfunction contributes to orthostatic hypotension syndrome and postural orthostatic tachycardia syndrome (POTS) [38]. It should be noted that in addition to autonomic dysfunction, symptoms such as palpitations, breathlessness, arrhythmia, and/or thermoregulation alterations (i.e., cold extremities) could also result from mitochondrial dysfunction in these disorders [39].

2.3. Cognitive Impairment

Cognitive impairment is a common feature of GWI, FM, ME/CFS and long COVID. Gulf War Illness is associated with cognitive impairments; including impaired memory and executive functioning (e.g., decision making, problem solving). Studies on GW veterans have demonstrated cognitive and mood impairments are linked to various adverse changes in neurons and neuroglial cells resulting in neuroinflammation [40,41,42,43,44,45].
There is also evidence for changes in brain structure and function associated with cognitive dysfunction in ME/CFS. For example, imaging studies in ME/CFS patients have found reduced gray matter density in certain brain regions associated with pain processing and cognitive function [46]. There is also evidence suggesting changes in brain activity in ME/CFS patients (and also long COVID), particularly in regions associated with memory, pain processing, and autonomic control [6]. In ME/CFS, elevated levels of pro-inflammatory cytokines/chemokines and growth factors have also been reported in patient blood samples, and there is evidence for neuroinflammation affecting the brain and spinal cord [47,48].
Fibromyalgia is often associated with cognitive impairment, commonly referred to as fibro fog [49]. This can manifest as difficulties with memory, concentration, attention, and overall mental clarity. While the exact cause of fibro fog is not fully understood, it is thought to be related to the chronic pain and fatigue associated with the disorder. In FM, evidence for elevated levels of pro-inflammatory cytokines/chemokines and growth factors, together with neuroinflammation, has also been reported [50]. The cognitive impairment may also be linked to changes in neurotransmitter and neural pathways involved in pain processing and cognitive function. While there is no cure for fibro fog, various strategies can help to manage symptoms such as some medications, cognitive behavioral therapy (CBT), lifestyle modifications, pacing activities, and support groups [51,52].
Cognitive impairment is a significant symptom of long COVID, with more than 50% of patients having cognitive slowing [53]. This cognitive impairment may result from neuroinflammation triggered by the immune response to the virus, or damage to blood vessels induced by the virus affecting blood flow to the brain [54]. Elevated markers of myeloid inflammation and complement activation have been identified in blood samples from long COVID patients [55].

2.4. Unrefreshing Sleep

Unrefreshing sleep is a prominent symptom of patients with GWI, FM, ME/CFS and long COVID. The exact cause of unrefreshing sleep is not fully understood in these disorders, although research suggests it may be related to disruptions in sleep architecture or alterations in autonomic nervous system function during sleep [56,57,58].
A significant percentage of GW veterans report experiencing unrefreshing sleep, along with other sleep-related issues such as difficulty falling asleep or staying asleep. Unrefreshing sleep is often accompanied by other symptoms that characterize GWI, including persistent fatigue, widespread pain, and cognitive difficulties [56]. This lack of restorative sleep can exacerbate other illness symptoms, affecting veterans’ overall health and well-being. Researchers are actively investigating the underlying mechanisms of GWI, including the role of sleep disturbance in its manifestation and progression. While there is no single cure for GWI, treatments like cognitive behavioral therapy for insomnia (CBT-I) have shown promise in improving sleep quality and reducing GWI symptoms [59].
FM is strongly associated with unrefreshing sleep, meaning that even after a full night’s sleep; individuals with FM often wake up feeling tired and not rested [57]. This is a core symptom, and it is often linked to other FM symptoms like disabling fatigue, chronic pain, and cognitive problems. People with FM often experience a disrupted sleep cycle/architecture, with more time spent in lighter stages of sleep (like stage 1) and less time in deep, restorative sleep (like slow-wave sleep) [60].
Several factors contribute to the sleep disturbances associated with FM. Individuals with FM tend to have more awakenings or arousals during the night, further disrupting the sleep cycle. Chronic pain can disrupt sleep, and poor sleep can exacerbate pain. This creates a vicious cycle where sleep problems worsen pain, and pain makes it harder to sleep [61]. Some research suggests that autonomic nervous system dysfunction might contribute to sleep problems in FM, leading to heightened arousal and difficulty relaxing into sleep [62]. By addressing sleep disturbances and chronic pain, individuals with FM can potentially improve their overall sleep quality and experience a greater sense of well-being.
Addressing sleep disturbance is crucial for managing FM. Treatment may involve improving sleep hygiene, establishing a regular sleep schedule, creating a relaxing bedtime routine, and optimizing the sleep environment, all of which are important [63]. Cognitive behavioral therapy for insomnia (CBT-I) helps individuals identify and change negative thoughts and behavior related to sleep [64]. In addition, sleep hygiene and conservative management is most appropriate for FM patients, along with ruling out or treating other confounding conditions such as obstructive sleep apnea, pain, anxiety, and depression [65].
Unrefreshing sleep is also a hallmark symptom of ME/CFS and long COVID [58,66]. This lack of restorative sleep significantly contributes to the overall fatigue and chronic pain and other common symptoms experienced by individuals with ME/CFS and long COVID. Many patients report not feeling rested; people with these disorders often wake up feeling just as tired, or even more tired, than when they went to bed. This persistent lack of restorative sleep contributes to the debilitating fatigue and widespread pain and other core symptoms associated with illness, affecting physical and mental well-being [67]. While objective sleep studies (like polysomnography) may not always show significant differences between patients with ME/CFS and long COVID and healthy individuals, the subjective experience of unrefreshing sleep is a major complaint and diagnostic criterion in these disorders [68,69].
In summary, unrefreshing sleep is a significant and troublesome symptom in these low-energy associated disorders, contributing to the overall burden of the illness and influencing the quality of life for those affected.

2.5. Muscle and Joint Pain

Muscle and joint pain are cardinal symptoms common to GWI, FM, ME/CFS, and long COVID, although the presentation and pathophysiological mechanisms may differ from each other [3,70,71]. Whilst all these disorders can cause widespread pain, they also have unique characteristics and specific diagnostic case criteria.
GWI is characterized by a range of symptoms including muscle and joint pain resulting from exposure to environmental mitochondrial toxicants during the Gulf War [70]. Pain is common and widespread in GWI patients, and their health-related quality of life is poor [72].
FM is often described as a persistent painful aching sensation, and it may be more intense in specific areas or fluctuate in frequency and severity. Pain is often felt in muscles, ligaments, and tendons, particularly at the points where they attach to bones. There is no single test for FM, and diagnosis is often based on symptom assessment [71]. Muscle and joint pain are also significant common features of ME/CFS and long COVID [3]. Although widely variable, fatigue, muscle and joint pain, and brain fog are frequently reported. Recent research indicates that a significant percentage of individuals with long COVID meet also the case criteria for ME/CFS and FM [3].
While all these disorders involve muscle and joint pain, the location and intensity can vary among sufferers. FM is characterized by widespread pain with specific tender points, while GWI, ME/CFS and long COVID pain may be more generalized or specific to certain areas (i.e., tender points in GWI patients may worsen after exertion) [6].
In summary, muscle and joint pain are prominent features of GWI, FM, ME/CFS, and long COVID. While these disorders can be distinguished by their unique clinical characteristics and diagnostic case criteria, they can also overlap, particularly in their clinical presentation as shown in Figure 1.

3. Understanding the Biological Mechanisms Shared Between These Disorders

3.1. Mitochondrial Dysfunction, Oxidative Stress and Inflammation

Mitochondrial dysfunction, which affects cellular energy production, has been increasingly implicated in GWI, FM, ME/CFS, and long COVID [14]. There is some evidence suggesting that mitochondrial dysfunction may contribute to the profound fatigue and muscle weakness seen in these disorders. Previous studies have reported abnormalities in mitochondrial structure, impaired mitochondrial respiration, decreased ATP production, and abnormal mitochondrial DNA levels, as described below [73].
In GWI, exposure to chemical agents such as organophosphates or pyridostigmine bromide caused mitochondrial dysfunction in GW veterans. Chen et al. have identified damage to mitochondrial DNA [74], and Koslik et al. [75] and Golomb et al. [76,77,78] have reported impaired mitochondrial bioenergetic function in GWI veterans. In the latter studies, mitochondrial respiratory chain function was assessed in muscle biopsies from GWI patients, or via 31P-NMR spectroscopy in GWI patients; GWI severity was shown to be related to mitochondrial haplogroups. Elevated levels of oxidative stress biomarkers in serum from Gulf War veterans have been reported; however, there is limited evidence for increased oxidative stress in GWI [45]. In animal models of GWI, biomarkers of oxidative stress have been reported to be increased, both at the systemic level, and particularly in the hippocampus, a region of the brain important for memory and cognitive function [14,79]. GWI is associated with a persistent inflammatory state, particularly in the central nervous system; elevated levels of inflammatory markers (C-reactive protein, interleukin-6) have been identified in blood samples from Gulf War veterans [41]. It is of note that increased levels of inflammation biomarkers may represent a protective adaptation in GWI.
In FM, impaired oxidative phosphorylation, and reduced mitochondrial ATP production have also been reported, with the degree of mitochondrial dysfunction correlating with the severity of FM symptoms [80]. In FM, several studies have found evidence of increased oxidative stress in patient blood samples, including elevated levels of lipid peroxidation and decreased levels of antioxidants, including CoQ10, superoxide dismutase and catalase [14,81]. Evidence for elevated levels of pro-inflammatory cytokines/chemokines and growth factors, together with neuroinflammation, have been reported in FM [50].
Mitochondrial dysfunction in ME/CFS results in impaired ATP production and reduced mitochondrial bioenergetic reserve capacity, correlating with illness severity, which could explain the post-exertional malaise experienced by these patients [73]. Muscle biopsies have confirmed mitochondrial degeneration, deletions in mitochondrial DNA, and reduced mitochondrial activity in ME/CFS [82,83,84]. In ME/CFS, increased plasma levels of oxidative stress biomarkers have also been identified [14]. Elevated levels of pro-inflammatory cytokines/chemokines and growth factors have been reported in blood from ME/CFS patients, and there is evidence for neuroinflammation affecting the brain and spinal cord [47,48].
In long COVID, research has identified evidence of mitochondrial dysfunction in the blood immune cells of these patients. One of the hallmarks of long COVID is post-exertional malaise, which can be directly linked to mitochondrial dysfunction and compromised ATP production [85]. Evidence for structural and metabolic abnormalities in mitochondria from long COVID patients has been recently documented by Szögi et al. [86]. Increased levels of oxidative stress biomarkers and reduced antioxidant levels have been reported in long COVID patients [87]. In long COVID, elevated markers of myeloid inflammation and complement activation have also been identified [55].

3.2. Immune Dysregulation

Gulf War Illness, ME/CFS, and long COVID all show signs of immune system dysfunction, triggered by environmental toxins and/or viral persistence. Inflammation or altered immune responses, including elevated levels of pro-inflammatory cytokines/chemokines and growth factors, have been reported in these disorders [38,88]. Fibromyalgia may not show overt immune dysfunction, but is thought to involve central sensitization, where the brain becomes hypersensitive to centralized nociceptive and nociplastic pain signals [89].
Immune dysfunction in GWI is linked to exposure of neurotoxins, and other environmental stressors during military service; elevated levels of antibodies targeted against infectious agents found in veterans with GWI most likely results from aberrant immune function are linked with mitochondrial dysfunction [4]. Elevated plasma levels of pro-inflammatory cytokines, including interferon-gamma, interleukin (IL)-2, IL-6 and tumor necrosis factor-alpha (TNF-α), together with elevated levels of C-reactive protein have been found in Gulf War veterans. Elevated plasma levels of T- and B-cells, and reduced levels of natural killer (NK) cells, have also been reported in veterans with GWI [90,91,92]. Long COVID is associated with persistent immune cell activation, autoimmune antibody production and immune exhaustion/senescence [93,94,95].
ME/CFS can also be triggered by viral infection persistence (e.g., Epstein-Barr virus, cytomegalovirus, herpesvirus, etc.). In ME/CFS, elevated blood levels of pro-inflammatory cytokines/chemokines and growth factors, together with reduced functional ability of NK cells and B- or T-cell abnormalities have been described. In particular there is evidence that NK cells and cluster of differentiation CD8+ T cells in ME/CFS may exhibit signs of exhaustion, potentially limiting their ability to effectively fight persistent infection [96,97].
FM is sometimes thought to be triggered by infections or physical/emotional trauma, although this is less clear [98,99]. There is evidence for immune system dysfunction in FM, with altered levels of cytokines/chemokines and growth factors and other inflammatory biomarkers in the brain and spinal fluid indicating neuroinflammation may play a potential role [100,101]. A study by Goebel et al. found that transfer of antibodies present in the blood of fibromyalgia patients could induce fibromyalgia symptoms in mice, suggesting an autoimmune component [102].

3.3. Neuroendocrine Abnormalities

Gulf War Illness, FM, ME/CFS or long COVID are associated with hypothalamic–pituitary–adrenal (HPA) axis neuroendocrine abnormalities, characterized by an attenuated stress response and reduced HPA axis activity, potentially leading to lower cortisol levels and enhanced sensitivity to glucocorticoid feedback [103,104,105]. In Gulf War veterans, alterations in the HPA axis (specifically suppression of adrenocorticotropic hormone (ACTH) and cortisol) has been linked to environmental exposures, including chemical warfare agents, pesticides, and other environmental stressors, possibly contributing to long-term GWI symptoms such as disabling fatigue, post-exertional malaise, dysautonomia, sleep disturbances, and cognitive impairments [106].
Hypothalamic–pituitary–adrenal axis neuroendocrine dysregulation, characterized by low cortisol levels (hypocortisolism) and altered cortisol rhythms is particularly evident in individuals with ME/CFS, where a dysregulated stress response can lead to unexplainable fatigue, post-exertional malaise, autonomic dysfunction, and sleep disturbances, among other core symptoms [107]. There is also some evidence of altered hormone levels in FM and long COVID, although this is less well established [108,109].
However, the exact cause and effect association and heterogeneity of these findings are still areas of ongoing research. More research is needed to fully understand the role of HPA axis neuroendocrine alterations, their relationship to neuroimmune function, and their potential as surrogate biomarkers or therapeutic targets in these disorders.

3.4. Dysregulated Gut–Brain Axis

Recent research suggests a significant link between GWI and the gut–brain axis. Specifically, GWI is associated with gastrointestinal problems and neurological symptoms, and studies indicate that alterations in the gut microbiome (dysbiosis and impaired intestinal permeability) may play a crucial role in the development and persistence of symptoms [110,111]. Several studies have shown that GW veterans have distinct differences in their gut microbiome composition compared to matched healthy individuals [112]. A significant portion of GW veterans experience gastrointestinal problems, including abdominal pain, diarrhea and irritable bowel syndrome (IBS), which are closely linked to illness [113].
GWI also involves neurological symptoms including fatigue, cognitive deficits, and mood changes, suggesting a potential involvement of the gut–brain axis in these manifestations. The gut–brain axis is believed to be disrupted in GWI through various mechanisms, including changes in the gut microbiome, increased intestinal permeability (leaky gut), and altered enteric nervous system (ENS) function [114]. Understanding the role of the gut–brain axis in GWI opens avenues for developing novel potential therapeutic strategies, such as targeting the gut microbiome or modulating the ENS [115]. It is also of note that since Gulf War veterans have higher rates of infection, they may have received more antibiotics, which in turn will result in an altered gut microbiome, irrespective of whether this plays a causal role in gastrointestinal symptoms in GWI.
Studies using a mouse model of GWI have shown that the compound andrographolide can restore gut microbiome balance, improve gut barrier function, and reduce neuroinflammation, potentially alleviating GWI symptoms [116]. Exposure to pyridostigmine bromide, a medication used during the Gulf War, has been linked to gut dysbiosis and gastrointestinal neuroimmune disruption in animal models [117].
In summary, the gut–brain axis is a critical area of research in understanding the complexities of GWI, with evidence suggesting that gut microbiome alterations, gastrointestinal dysfunction, and neurological symptoms are interconnected. Further research into the gut–brain axis in GWI could lead to more effective treatments and management strategies for GW veterans.
Fibromyalgia is increasingly linked to disruptions in the gut–brain axis, often involving dysbiosis and altered gut permeability, contributing to the wide range of FM symptoms [118,119]. The gut–brain axis is a bidirectional communication system that connects the central nervous system with the enteric nervous system [120]. This communication occurs through various pathways, including the vagus nerve that plays a crucial role in relaying signals between the gut and the brain. The HPA axis, involved in stress response, can be impacted by gut health; the gut is a major site of immune activity, and immune cells can influence both gut and brain function [121].
The gut microbiome influences the production and regulation of neurotransmitters (such as imbalances of serotonin and dopamine), which play a potential role in mood, anxiety/depression, pain perception, and cognitive function. In addition, several studies have described how dysregulation of the gut–brain axis can contribute to central sensitization, a phenomenon where the nervous system becomes overly sensitive to pain signals [111,114,121].
Myalgic encephalomyelitis/chronic fatigue syndrome and long COVID exhibit disruptions in the gut–brain axis, which involve alterations in gut microbiota composition, immune responses, and neurotransmitter signaling, potentially contributing to the diverse and debilitating symptoms seen in both disorders [122,123]. Both disorders are associated with imbalances in the gut microbiome, including shifts in bacterial populations and reduced microbial diversity. For example, in ME/CFS, specific bacterial groups such as Alistipes may be increased, while Faecalibacterium is decreased [124,125,126]. In long COVID, prolonged fecal shedding of SARS-CoV-2 and associated inflammation could lead to dysbiosis [127,128,129,130,131].
Understanding the role of the gut–brain axis in these disorders opens up new avenues for treatment with probiotics and prebiotics that may help restore gut microbiome balance; dietary interventions to reduce inflammation and promote a healthy gut environment could also be beneficial. Vagus nerve stimulation, which can be achieved through various methods, may help modulate gut–brain communication. Further research into the intricate interplay between the gut and the brain is crucial for developing more effective and targeted therapies in these disorders [115,122,132].

3.5. Apoptosis and Ferroptosis

Apoptosis is a process of programmed cell death, which can occur after a number of triggering events, including mitochondrial dysfunction and oxidative stress [133]. Ferroptosis is a type of iron-dependent programmed cell death (distinct from apoptosis) characterized by the accumulation of free radical induced lipid peroxidation products, with key regulators including GPX4 and the SLC7A11/GSH system and FTH1, which counteract lipid peroxidation [134]. There is evidence that apoptosis and/or ferroptosis contribute to the pathogenesis of GWI, ME/CFS, FM and long COVID.
In GWI, serum from patients has been reported to increase apoptosis in cultured neuroblastoma cells, suggesting that apoptosis potentially contributes to the neurocognitive and mood dysfunction experienced by some veterans [135]. Such data related to apoptosis in GWI research have opened avenues for potential therapeutic interventions. For example, Tsilibary et al. investigated the role of the anthrax vaccine given to Gulf War veterans in inducing apoptosis in cultured neural cells, and the use of IgG isotype anti-protective antigen protein antibodies in neutralizing the adverse effects of serum on neural cultures, including reducing apoptosis [136]. This research suggests that immunotherapy targeting specific antigens or pathways involved in GWI-related apoptosis could be a viable approach for treating the illness.
In fibromyalgia, there is some evidence that increased apoptosis in nerve cells, particularly dorsal root ganglia, may contribute to nerve damage and altered pain processing, resulting in the pain and hypersensitivity associated with this disorder. Apoptosis-related genes are reportedly upregulated in dorsal root ganglia in models of neuropathic pain, suggesting a potential link to chronic pain disorders like FM [137]. Increased apoptosis markers and decreased cell viability have been observed in animal models of FM, factors that can be reduced following selenium administration [138]. Transient receptor potential (TRP) channels (like TRPM2 and TRPV1) play a role in calcium influx into nerve terminals, which can trigger apoptosis. Selenium treatment in animal models noted above reduces hyperalgesia, oxidative stress levels, and apoptosis by modulating these channels [138]. Elevated levels of anti-annexin antibodies, which are associated with apoptosis, have been reported in some FM patients, particularly those with Sjögren’s syndrome [139]. Similarly, there is evidence that ferroptosis, particularly in skeletal muscles, may play a role in the widespread pain and other symptoms associated with FM [140]. Specifically, studies have shown that magnesium hexacyanoferrate nanocatalysts, which act as ferroptosis inhibitors, can alleviate FM symptoms in mouse models by reducing oxidative stress and mitochondrial dysfunction [141].
Patients with ME/CFS have an increased rate of apoptosis in their neutrophils compared to healthy individuals; neutrophils are thus eliminated at a faster than normal rate, which may impair the immune system’s ability to respond effectively to pathogenic agents [142]. Cezar et al. found CD4+ T cells from participants who later presented long COVID were more apoptotic in culture than those of sequelae-free participants at month 1, suggesting that CD4+ T cell death during the acute phase of SARS-CoV-2 infection might pave the way for long COVID [143]. In particular with regard to the potential of ferroptosis and oxidative stress pathways in the pathogenesis of these disorders, Biamonte et al. reported a correlation between circulating iron status and hematological index indicators during the COVID-19 progression [144], and a low baseline level of a ferroptosis marker (SLC7A11) may indicate an increased risk for long COVID-19, and associated cardiovascular and neurological dysfunction [145,146,147].

3.6. Telomere Shortening

Telomeres are repetitive DNA sequences at the ends of chromosomes that protect the chromosomes from damage and degradation. During DNA replication DNA polymerase, the enzyme that copies DNA, cannot fully replicate the very ends of the chromosomes, leading to a small loss of telomere length with each cell division; this shortening of the telomeres eventually leads to cellular senescence and apoptosis [148]. Whilst telomere shortening is a natural part of the aging process, a number of factors can accelerate this process, including lifestyle factors and exposure to toxins [149]. At the cellular level, telomere shortening is linked to mitochondrial dysfunction, oxidative stress, and inflammation [150,151,152]. Telomere shortening leads to premature cellular aging and increased risk of age-related diseases [153].
Elevated mitochondrial ROS levels directly damage telomeric DNA by oxidizing guanine bases, creating lesions such as 8-oxoguanine [150,151,152]. Telomeric 8-oxoguanine lesions can disrupt the binding of essential telomere-binding proteins (TRF1 and TRF2) as part of the shelterin complex, which are crucial for protecting telomere stability and maintenance [150,151,152].
There is evidence for telomere shortening in military veterans with GWI, FM, ME/CFS and long COVID. Military veterans are reported to have shorter telomeres compared to their civilian peers [154]. Gulf War veterans were exposed to a variety of environmental toxins, including pesticides, pyridostigmine bromide, and smoke from oil well fires [155]. Such exposures are linked to mitochondrial dysfunction, oxidative stress and inflammation [156], thereby accelerating telomere shortening. Veterans with GWI may therefore have shorter telomeres compared to their peers without the illness, although this requires confirmation. Such accelerated telomere shortening could be a contributing factor to the development of various health problems associated with GWI, including cardiovascular and neurodegenerative disorders [154,156].
There is evidence that individuals with ME/CFS experience a faster rate of telomere shortening compared to healthy individuals, indicating premature aging. A study by Rajeevan et al. found that the association between ME/CFS and shorter telomeres was largely moderated by females under 45 years old [157]. The observed telomere shortening in ME/CFS could be a marker of premature immunosenescence (premature aging of the immune system) and may contribute to the development of other age-related diseases.
Fibromyalgia is also associated with telomere shortening, with the degree of shortening correlating with the frequency/severity of pain [158]. When FM is accompanied by anxiety/depression, the effects on telomere length can be even more pronounced in these patients. These data suggest that chronic pain in FM may contribute to premature cellular aging, affecting overall health and potentially increasing the risk of age-related diseases [158].
Research suggests that acute SARS-CoV-2 infection can accelerate telomere shortening in blood cells, potentially contributing to the persistent symptoms associated with long COVID [159,160,161]. The telomere shortening observed in long COVID appears to be independent of the patient’s chronological age, suggesting a direct impact of the virus on telomere length [161]. Patients with shorter telomeres in the immune cells (leukocytes) have been linked to worse prognoses in COVID-19, and this shortening may be independent of chronological age [159].
In summary, Figure 2 displays shared biological abnormalities in the development of low-energy associated disorders.

4. Implications for Current and Promising Therapeutic Strategies

4.1. Current Therapeutic Approaches

There are no specific cures for GWI, FM, ME/CFS and long COVID but current treatments are focusing on improving quality of life through symptom management and non-pharmacological approaches like pacing (activity management), cognitive-behavioral therapy (CBT), graded exercise therapy (GET), relaxation, yoga, Tai-Chi, and acupuncture [65,162,163,164,165]. For symptoms like anxiety/depression, some antidepressants, including tricyclic and heterocyclic antidepressants, monoamine oxidase inhibitors (MAOIs) and selective serotonin reuptake inhibitors (SSRIs) may be used amongst disorders [65,162]. The treatments are individualized, with a focus on active management and coping strategies, and are often coordinated with health coaching and care management. Mindfulness-based therapies and CoQ10 supplementation have shown potential benefits for managing symptoms of sleep disturbances, chronic pain and fatigue in these chronic multisymptom disorders [65,162,163,164,165]. In summary, there are ongoing research efforts to better understand these complex disorders and develop new diagnostic criteria, potential biomarkers and targeted treatments.

4.2. Stem Cell Therapy

Stem cells can differentiate into various cell types, promoting tissue regeneration and potentially repairing damaged organs. Based on their ability to differentiate into other cell types, stem cells are classified as either pluripotent, multipotent, omnipotent, totipotent, oligopotent, or unipotent. Depending on where they originated from, stem cells may be classified as embryonic, adult, fetal, or induced pluripotent stem cells (iPSCs) [166]. Stem cell therapy involves transplanting healthy stem cells to replace damaged or diseased stem cells, or using them to stimulate the body’s own repair mechanisms, for example, by secreting growth factors or recruiting other cell types necessary for tissue repair [167].
Stem cells can transfer their healthy mitochondria to cells with damaged or dysfunctional mitochondria, thereby restoring cellular energy levels and reducing oxidative stress; this transfer can occur through various mechanisms, including tunneling nanotubes, gap junctions, and extracellular vesicles [168]. In addition to reducing oxidative stress by improving mitochondrial function, stem cells per se have antioxidant action, directly scavenging free radicals or stimulating production of antioxidant enzymes like superoxide dismutase, catalase, and glutathione peroxidase in other cells [169]. Stem cells can also engulf and degrade dysfunctional mitochondria from damaged cells, promoting cellular repair and proliferation; the engulfment of damaged mitochondria can trigger the production of cytoprotective enzymes like heme-oxygenase-1 (HO-1), which can further enhance the stem cell’s ability to rescue damaged cells [170]. Additionally, stem cells can release factors that stimulate mitochondrial biogenesis in recipient cells. Stem cells can also help to regulate the immune system, reducing inflammation via shifting the balance of immune cell activity from a pro-inflammatory to an antiinflammatory state [171].
Stem cell therapy is of relevance to the treatment of GWI, FM, ME/CFS, and long COVID, although research in each of these areas is still at an early stage. Mesenchymal stem cells may be particularly beneficial in treating patients with ME/CFS; improvements in sleep quality, levels of energy and pain, and cognitive function have been reported following stem cell therapy. Several randomized controlled clinical trials have reported the beneficial effects of stem cell therapy in patients with severe COVID-19 infection [172,173,174]. Mokhemer et al. reported symptomatic improvement in a rat model of FM following stem cell therapy [175]. Using a cell culture based system; Tsilibary et al., identified the potential of stem cell therapy to protect cells against toxic factors present in serum from veterans with GWI [19].

4.3. Coenzyme Q10 Supplementation

Supplementation with CoQ10 has been shown to be beneficial in GWI, FM, ME/CFS and long COVID, resulting from improved mitochondrial function, reductions in oxidative stress, inflammation and apoptosis/ferroptosis, and telomere shortening, as described below.
In GWI, supplementary CoQ10 improved physical function and self-reported health, as well as fatigue, pain and muscle strength [176]. Several studies have shown that CoQ10 supplementation can reduce fatigue and improve quality of life in individuals with ME/CFS. Castro-Marrero et al. reported supplementation with CoQ10 and nicotinamide adenine dinucleotide (NADH) improved fatigue in ME/CFS [177]. Supplementation with CoQ10 for symptoms such as fatigue, pain, and cognitive dysfunction in ME/CFS has been suggested in a consensus report from the European network on ME/CFS (EUROMENE consortium) [178].
Fibromyalgia patients have depleted CoQ10 levels in tissues (typically 40–50% of the normal level) [179]. Hence, supplementation with CoQ10 can help reduce chronic pain, fatigue, and improve overall quality of life in FM patients. A randomized, double blind, placebo-controlled study found that 300 mg/day of CoQ10 for 40 days significantly reduced chronic pain and fatigue in FM patients, with a corresponding improvement in mitochondrial energy production [179]. Supplemental CoQ10 may be particularly helpful in reducing pain and fatigue in those with FM who are also taking pregabalin [180].
Clinical studies supplementing CoQ10 in COVID-19 patients (and in patients with long COVID) have reported mixed outcomes. In a prospective observational study, 116 patients with long COVID were supplemented with 200 mg of CoQ10 and 200 mg of alpha-lipoic acid per day for 2 months versus 58 long COVID patients who received placebo; the fatigue severity assessed using self-reported questionnaire was substantially reduced in the treated patients compared to placebo [181]. However, a randomized controlled intervention study comprising 121 long COVID patients supplemented with 500 mg of CoQ10 per day for 6 weeks reported no significant benefit on illness symptoms [182].
There is evidence that supplementation with CoQ10 may help protect telomeres from damage caused by oxidative stress and inflammation. A study involving older adults with low selenium levels showed that supplementation with CoQ10 and selenium resulted in less telomere shortening compared to placebo. This study also found a correlation between longer telomeres and reduced cardiovascular mortality [183].
There is also evidence that supplementary CoQ10 could improve the efficiency of stem cell therapy when this procedure is applied in these disorders. A number of studies have demonstrated beneficial effects of CoQ10 on stem cell metabolism, of relevance to improving the efficiency of stem cell therapy when applied in the above disorders. Coenzyme Q10 has been shown to protect stem cells from damage caused by factors like hypoxia, oxidative stress, and aging [184].
For example, CoQ10 has been shown to protect bone marrow-derived mesenchymal stem cells (BMSCs) from H2O2-induced oxidative stress and cell death. It has also been shown promise in reducing oxidative stress and potentially enhancing the therapeutic efficiency of BMSC transplantation for spinal cord injury treatment [185,186]. Coenzyme Q10 can enhance the regenerative potential of stem cells by promoting cell differentiation, angiogenesis, and tissue repair. It can also help stem cells migrate to the site of injury and integrate into the damaged tissue. Coenzyme Q10 can be combined with stem cells in various ways, such as co-administration or loading stem cells with CoQ10. For example, CoQ10-loaded exosomes have shown promise in delivering CoQ10 to target cells and enhancing their therapeutic effects [187,188,189].
Coenzyme Q10 has shown promise in enhancing stem cell therapy for conditions like lung fibrosis, nerve injury, and intervertebral disk degeneration [190]. Maruo et al. described the activation of mitochondria in human mesenchymal stem cells using encapsulated CoQ10 [191]. Li et al. identified impaired mitochondrial function in mesenchymal stem cells of aged mice compared to young mice, and suggested that this area of metabolism as a potential therapeutic target (i.e., via CoQ10 supplementation) to enhance the regenerative function of these cells [192]. Studies to investigate the effect of CoQ10 in reducing stem cell senescence, improving the survival rate and activity of stem cells against toxic agents, and increasing the efficiency of transplanted stem cells have been summarized in Table 1.
It is important to note that individual responses to CoQ10 supplementation may vary, and further research is needed to fully understand its therapeutic implications for these disorders.

5. Conclusions and Future Perspectives

Although GWI, FM, ME/CFS and long COVID have distinct origins, it should be noted that a significant proportion of GWI veterans meet case criteria for ME/CFS and FM. This review explores the evidence that these disorders comprise a group of so-called low-energy associated disorders with overlapping common symptoms and underlying to shared pathophysiological mechanisms. In particular, evidence for the roles of mitochondrial dysfunction, oxidative stress, inflammation, apoptosis/ferroptosis, immune dysregulation, autonomic dysfunction, disrupted brain–gut–microbiome axis and telomere shortening as common features in the pathogenesis of these disorders has been identified.
In this review, we have explored the overlapping shared symptoms and pathological features that are common to these disorders. One might therefore question why the above have been classified as four distinct disorders, rather than a single disorder. Given that there are lack of specific biomarkers and accurate objective case criteria to differentiate these disorders; particularly stems from their inherent multifactorial nature, heterogeneity, and overlapping common symptoms and underlying biological pathomechanisms. All the above leads to subjective clinical diagnosis, difficulties in comparing research findings, a higher risk of patients receiving ineffective interventions, and an inability to identify specific patient subgroup for developing targeted therapies. While some progress has been made with biomarkers for these disorders, significant challenges remain in discovery, validation, stratification, and translating findings into reliable clinical tools due to complexities in underlying pathology and intra- and inter-patient sample variations.
Given the potential role of CoQ10 in promoting normal mitochondrial function, as an antioxidant, antiinflammatory and antiapoptotic/antiferroptotic agent, there is a rationale for the role of supplementary CoQ10 in the management of these disorders. The reported benefits of supplementary CoQ10 administration in reducing telomere shortening and improving the efficiency of stem cell transfer has also been identified as promising therapeutic strategies in these disorders.
Although previously published reports have explored shared common symptoms and biological abnormalities in ME/CFS and long COVID, to our knowledge this is the first study to summarize overlapping common symptoms and specific pathophysiological mechanisms in GWI, FM, ME/CFS and long COVID, and how these might be addressed via CoQ10 supplementation and stem cell therapy.
While promising, further research and rigorous large-scale randomized controlled trials are needed to fully elucidate the underlying biological pathomechanisms by which CoQ10 can mitigate the common symptoms, and to establish efficacy and dosing protocols in these disorders. Understanding these issues, connections based on a personalized medicine approach may provide new targets for therapeutic interventions and improve outcomes for individuals with these complex disorders.

Author Contributions

Conceptualization, D.M. and J.C.-M.; methodology, D.M., J.C.D., B.A.G. and J.C.-M.; investigation, D.M., J.C.D., B.A.G. and J.C.-M.; writing—original draft preparation, D.M., J.C.D., B.A.G. and J.C.-M.; writing—review and editing, D.M., J.C.D., B.A.G. and J.C.-M. All authors have read and agreed to the published version of the manuscript.

Funding

Author B.A.G.’s contribution to this manuscript was supported by the DOD CDMRP (Award W81XWH20-0523) and the Krupp Endowment Research Fund Award. The authors have reviewed and edited the output and take full responsibility for the content of this publication.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

David Mantle has been involved as a consultant and expert witness in Pharma Nord Company (UK) Ltd. All other authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
ANSAutonomic nervous system
ATPAdenosine triphosphate
BMSCBone marrow-derived mesenchymal stem cells
CBT-ICognitive behavior therapy-insomnia
CDCCenters for Disease Control and Prevention
CoQ10Coenzyme Q10
ENSEnteric nervous system
FMFibromyalgia
FDAFood and Drug Administration
FTH1Ferritin heavy chain 1
GETGraded exercise therapy
GPX4Glutathione peroxidase 4
GWIGulf War Illness
HRVHeart rate variability
HPAHypothalamic–pituitary–adrenal axis
HO-1Heme-oxygenase-1
IBSIrritable bowel syndrome
ILInterleukin
MAOIMonoamine oxidase inhibitors
ME/CFS Myalgic encephalomyelitis/chronic fatigue syndrome
MPP+1-methyl-4-phenylpyridinium
NADHNicotinamide adenine dinucleotide
NKNatural killer cells
PEMPost-exertional malaise
PESEPost-exertional symptom exacerbation
PI3KPhosphatidylinositol-3-kinase
POTSPostural orthostatic tachycardia syndrome
iPSCInduced-pluripotent stem cells
RCTRandomized controlled trial
ROSReactive oxygen species
SARS-CoV-2Severe acute respiratory syndrome coronavirus 2
SLC7A11Solute carrier family 7 member 11
SSRISelective serotonin reuptake inhibitors
TRPV1Transient receptor potential vanilloid 1
TRPM2Transient receptor potential melastatin 2
TRF1Telomeric repeat-binding factor 1
TRF2Telomeric repeat-binding factor 2

References

  1. Queiroz, L.P. Worldwide epidemiology of fibromyalgia. Curr. Pain Headache Rep. 2013, 17, 356. [Google Scholar] [CrossRef]
  2. Lim, E.J.; Ahn, Y.C.; Jang, E.S.; Lee, S.W.; Lee, S.H.; Son, C.G. Systematic review and meta-analysis of the prevalence of chronic fatigue syndrome/myalgic encephalomyelitis (CFS/ME). J. Transl. Med. 2020, 18, 100. [Google Scholar] [CrossRef]
  3. Dehlia, A.; Guthridge, M.A. The persistence of myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) after SARS-CoV-2 infection: A systematic review and meta-analysis. J. Infect. 2024, 89, 106297. [Google Scholar] [CrossRef]
  4. Bast, E.; Jester, D.J.; Palacio, A.; Krengel, M.; Reinhard, M.; Ashford, J.W. Gulf War Illness: A Historical Review and Considerations of a Post-Viral Syndrome. Mil. Med. 2025, usaf092. [Google Scholar] [CrossRef]
  5. Annesley, S.J.; Missailidis, D.; Heng, B.; Josev, E.K.; Armstrong, C.W. Unravelling shared mechanisms: Insights from recent ME/CFS research to illuminate long COVID pathologies. Trends Mol. Med. 2024, 30, 443–458. [Google Scholar] [CrossRef]
  6. Goldenberg, D.L. The pivotal role of central sensitization in long COVID, fibromyalgia and myalgic encephalomyelitis/chronic fatigue syndrome. Expert Rev. Neurother. 2025, 25, 973–989. [Google Scholar] [CrossRef] [PubMed]
  7. Fukuda, K.; Nisenbaum, R.; Stewart, G.; Thompson, W.W.; Robin, L.; Washko, R.M.; Noah, D.L.; Barrett, D.H.; Randall, B.; Herwaldt, B.L.; et al. Chronic multisymptom illness affecting Air Force veterans of the Gulf War. Jama 1998, 280, 981–988. [Google Scholar] [CrossRef] [PubMed]
  8. Steele, L. Prevalence and patterns of Gulf War illness in Kansas veterans: Association of symptoms with characteristics of person, place, and time of military service. Am. J. Epidemiol. 2000, 152, 992–1002. [Google Scholar] [CrossRef] [PubMed]
  9. Clauw, D.J.; D’Arcy, Y.; Gebke, K.; Semel, D.; Pauer, L.; Jones, K.D. Normalizing fibromyalgia as a chronic illness. Postgrad. Med. 2018, 130, 9–18. [Google Scholar] [CrossRef] [PubMed]
  10. Singh, A.P. A Historical Review of Chronic Fatigue Syndrome. J. Humanit. Soc. Sci. 2018, 23, 23–28. [Google Scholar]
  11. Komaroff, A.L.; Lipkin, W.I. ME/CFS and Long COVID share similar symptoms and biological abnormalities: Road map to the literature. Front. Med. 2023, 10, 1187163. [Google Scholar] [CrossRef] [PubMed]
  12. Goldenberg, D.L. How to understand the overlap of long COVID, chronic fatigue syndrome/myalgic encephalomyelitis, fibromyalgia and irritable bowel syndromes. Semin. Arthritis Rheum. 2024, 67, 152455. [Google Scholar] [CrossRef] [PubMed]
  13. Khakban, I.; Jain, S.; Gallab, J.; Dharmaraj, B.; Zhou, F.; Lokker, C.; Abdelkader, W.; Zeraatkar, D.; Busse, J.W. Impact of the COVID-19 Pandemic and the 2021 National Institute for Health and Care Excellence Guidelines on Public Perspectives Toward Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: Thematic and Sentiment Analysis on Twitter (Rebranded as X). J. Med. Internet Res. 2025, 27, e65087. [Google Scholar] [CrossRef]
  14. Davis, L.; Higgs, M.; Snaith, A.; Lodge, T.A.; Strong, J.; Espejo-Oltra, J.A.; Kujawski, S.; Zalewski, P.; Pretorius, E.; Hoerger, M.; et al. Dysregulation of lipid metabolism, energy production, and oxidative stress in myalgic encephalomyelitis/chronic fatigue syndrome, Gulf War Syndrome and fibromyalgia. Front. Neurosci. 2025, 19, 1498981. [Google Scholar] [CrossRef]
  15. Mantle, D.; Hargreaves, I.P.; Domingo, J.C.; Castro-Marrero, J. Mitochondrial Dysfunction and Coenzyme Q10 Supplementation in Post-Viral Fatigue Syndrome: An Overview. Int. J. Mol. Sci. 2024, 25, 574. [Google Scholar] [CrossRef]
  16. James, L.M.; Georgopoulos, A.P. At the Root of 3 “Long” Diseases: Persistent Antigens Inflicting Chronic Damage on the Brain and Other Organs in Gulf War Illness, Long-COVID-19, and Chronic Fatigue Syndrome. Neurosci. Insights 2022, 17, 26331055221114817. [Google Scholar] [CrossRef]
  17. León-Moreno, L.C.; Reza-Zaldívar, E.E.; Hernández-Sapiéns, M.A.; Villafaña-Estarrón, E.; García-Martin, M.; Ojeda-Hernández, D.D.; Matias-Guiu, J.A.; Gomez-Pinedo, U.; Matias-Guiu, J.; Canales-Aguirre, A.A. Mesenchymal Stem Cell-Based Therapies in the Post-Acute Neurological COVID Syndrome: Current Landscape and Opportunities. Biomolecules 2023, 14, 8. [Google Scholar] [CrossRef]
  18. Padda, J.; Khalid, K.; Zubair, U.; Al Hennawi, H.; Yadav, J.; Almanie, A.H.; Mehta, K.A.; Tasnim, F.; Cooper, A.C.; Jean-Charles, G. Stem Cell Therapy and Its Significance in Pain Management. Cureus 2021, 13, e17258. [Google Scholar] [CrossRef]
  19. Tsilibary, E.C.; Carlson, D.; Georgopoulos, A.P. Protective Effect of Stem Cells from Toxicity Induced by Gulf War Illness (GWI) Serum in N2A Neuroblastoma Cells. J. Neurol. Neuromedicine 2021, 6, 26–31. [Google Scholar] [CrossRef] [PubMed]
  20. Hodges, L. Repeated Cardiopulmonary Exercise Testing of ME/CFS Patients. Methods Mol. Biol. 2025, 2920, 163–172. [Google Scholar] [CrossRef]
  21. Risbano, M.G. From fatigue to physiology: Submaximal 2-day cardiopulmonary exercise test and emerging standards in long COVID. Exp. Physiol. 2025. [Google Scholar] [CrossRef]
  22. Frank, J.; Tehrani, L.; Gamer, J.; Van Booven, D.J.; Ballarin, S.; Rossman, R.; Edelstein, A.; Uppalati, S.; Reuthebuck, A.; Collado, F.; et al. Gulf War Illness Induced Sex-Specific Transcriptional Differences Under Stressful Conditions. Int. J. Mol. Sci. 2025, 26, 3610. [Google Scholar] [CrossRef]
  23. Vink, M.; Partyka-Vink, K. The PACE Trial’s GET Manual for Therapists Exposes the Fixed Incremental Nature of Graded Exercise Therapy for ME/CFS. Life 2025, 15, 584. [Google Scholar] [CrossRef]
  24. Boruch, A.; Branchaw, G.; O’Connor, P.J.; Cook, D.B. Physical Activity and Fatigue Symptoms: Neurotypical Adults and People with Chronic Multisymptom Illnesses. Curr. Top. Behav. Neurosci. 2024, 67, 281–308. [Google Scholar] [CrossRef]
  25. Di Bari, A.; Demo, G.; Patron, E. Unravelling the relationship between anxiety, autonomic nervous system dysfunction and fibromyalgia: A systematic review. Clin. Exp. Rheumatol. 2025, 43, 1136–1145. [Google Scholar] [CrossRef]
  26. Escorihuela, R.M.; Capdevila, L.; Castro, J.R.; Zaragozà, M.C.; Maurel, S.; Alegre, J.; Castro-Marrero, J. Reduced heart rate variability predicts fatigue severity in individuals with chronic fatigue syndrome/myalgic encephalomyelitis. J. Transl. Med. 2020, 18, 4. [Google Scholar] [CrossRef]
  27. Avery, T.J.; Mathersul, D.C.; Schulz-Heik, R.J.; Mahoney, L.; Bayley, P.J. Self-Reported Autonomic Dysregulation in Gulf War Illness. Mil. Med. 2023, 188, e1462–e1467. [Google Scholar] [CrossRef]
  28. Garner, R.S.; Rayhan, R.U.; Baraniuk, J.N. Verification of exercise-induced transient postural tachycardia phenotype in Gulf War Illness. Am. J. Transl. Res. 2018, 10, 3254–3264. [Google Scholar] [PubMed]
  29. Verne, Z.T.; Fields, J.Z.; Zhang, B.B.; Zhou, Q. Autonomic dysfunction and gastroparesis in Gulf War veterans. J. Investig. Med. 2023, 71, 7–10. [Google Scholar] [CrossRef] [PubMed]
  30. Kemp, J.; Sunnquist, M.; Jason, L.A.; Newton, J.L. Autonomic dysfunction in myalgic encephalomyelitis and chronic fatigue syndrome: Comparing self-report and objective measures. Clin. Auton. Res. 2019, 29, 475–477. [Google Scholar] [CrossRef] [PubMed]
  31. Ziaja, C.P.; Young, S.; Stark, M. Dysregulation of the autonomic nervous system in ME/CFS and post-COVID syndrome: Insights from 48-h heart rate variability monitoring. J. Neurol. Sci. 2023, 455, 121241. [Google Scholar] [CrossRef]
  32. El-Sawy, N.; El-Tantawi, G.; Achmawi, G.A.H.; Sultan, H.; Younis, S. Autonomic changes in fibromyalgia: Clinical and electrophysiological study. Alexandria J. Med. 2012, 48, 215–222. [Google Scholar] [CrossRef][Green Version]
  33. Kingsley, J.D. Autonomic dysfunction in women with fibromyalgia. Arthritis Res. Ther. 2012, 14, 103. [Google Scholar] [CrossRef]
  34. Staud, R. Autonomic dysfunction in fibromyalgia syndrome: Postural orthostatic tachycardia. Curr. Rheumatol. Rep. 2008, 10, 463–466. [Google Scholar] [CrossRef]
  35. Dani, M.; Dirksen, A.; Taraborrelli, P.; Torocastro, M.; Panagopoulos, D.; Sutton, R.; Lim, P.B. Autonomic dysfunction in ‘long COVID’: Rationale, physiology and management strategies. Clin. Med. 2021, 21, e63–e67. [Google Scholar] [CrossRef] [PubMed]
  36. Fedorowski, A.; Sutton, R. Autonomic dysfunction and postural orthostatic tachycardia syndrome in post-acute COVID-19 syndrome. Nat. Rev. Cardiol. 2023, 20, 281–282. [Google Scholar] [CrossRef]
  37. Giunta, S.; Giordani, C.; De Luca, M.; Olivieri, F. Long-COVID-19 autonomic dysfunction: An integrated view in the framework of inflammaging. Mech. Ageing Dev. 2024, 218, 111915. [Google Scholar] [CrossRef] [PubMed]
  38. Domingo, J.C.; Battistini, F.; Cordobilla, B.; Zaragoza, M.C.; Sanmartin-Sentanes, R.; Alegre-Martin, J.; Cambras, T.; Castro-Marrero, J. Association of circulating biomarkers with illness severity measures differentiates myalgic encephalomyelitis/chronic fatigue syndrome and post-COVID-19 condition: A prospective pilot cohort study. J. Transl. Med. 2024, 22, 343. [Google Scholar] [CrossRef] [PubMed]
  39. Golomb, B.A. Oxidative Stress and Mitochondrial Injury in Chronic Multisymptom Conditions: From Gulf War Illness to Autism Spectrum Disorder. Nat. Prec. 2012. [Google Scholar] [CrossRef]
  40. Alshelh, Z.; Albrecht, D.S.; Bergan, C.; Akeju, O.; Clauw, D.J.; Conboy, L.; Edwards, R.R.; Kim, M.; Lee, Y.C.; Protsenko, E.; et al. In-vivo imaging of neuroinflammation in veterans with Gulf War illness. Brain Behav. Immun. 2020, 87, 498–507. [Google Scholar] [CrossRef]
  41. Butterick, T.A.; Trembley, J.H.; Hocum Stone, L.L.; Muller, C.J.; Rudquist, R.R.; Bach, R.R. Gulf War Illness-associated increases in blood levels of interleukin 6 and C-reactive protein: Biomarker evidence of inflammation. BMC Res. Notes 2019, 12, 816. [Google Scholar] [CrossRef]
  42. Chao, L.L.; Sullivan, K.; Krengel, M.H.; Killiany, R.J.; Steele, L.; Klimas, N.G.; Koo, B.B. The prevalence of mild cognitive impairment in Gulf War veterans: A follow-up study. Front. Neurosci. 2023, 17, 1301066. [Google Scholar] [CrossRef]
  43. Cheng, C.H.; Alshelh, Z.; Guan, Y.; Sullivan, K.; Loggia, M.L.; Koo, B.B. Association of the tissue microstructural diffusivity and translocator protein PET in Gulf War Illness. Brain Behav. Immun. Health 2021, 18, 100364. [Google Scholar] [CrossRef]
  44. Cheng, C.H.; Koo, B.B.; Calderazzo, S.; Quinn, E.; Aenlle, K.; Steele, L.; Klimas, N.; Krengel, M.; Janulewicz, P.; Toomey, R.; et al. Alterations in high-order diffusion imaging in veterans with Gulf War Illness is associated with chemical weapons exposure and mild traumatic brain injury. Brain Behav. Immun. 2020, 89, 281–290. [Google Scholar] [CrossRef]
  45. Dickey, B.; Madhu, L.N.; Shetty, A.K. Gulf War Illness: Mechanisms Underlying Brain Dysfunction and Promising Therapeutic Strategies. Pharmacol. Ther. 2021, 220, 107716. [Google Scholar] [CrossRef]
  46. Godlewska, B.R.; Sylvester, A.L.; Emir, U.E.; Sharpley, A.L.; Clarke, W.T.; Williams, S.R.; Gonçalves, A.J.; Raman, B.; Valkovič, L.; Cowen, P.J. Brain and muscle chemistry in myalgic encephalitis/chronic fatigue syndrome (ME/CFS) and long COVID: A 7T magnetic resonance spectroscopy study. Mol. Psychiatry 2025, 1–12. [Google Scholar] [CrossRef] [PubMed]
  47. Jonsjö, M.A.; Olsson, G.L.; Wicksell, R.K.; 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] [CrossRef] [PubMed]
  48. Lee, J.S.; Sato, W.; Son, C.G. Brain-regional characteristics and neuroinflammation in ME/CFS patients from neuroimaging: A systematic review and meta-analysis. Autoimmun. Rev. 2024, 23, 103484. [Google Scholar] [CrossRef] [PubMed]
  49. Dass, R.; Kalia, M.; Harris, J.; Packham, T. Understanding the Experience and Impacts of Brain Fog in Chronic Pain: A Scoping Review. Can. J. Pain 2023, 7, 2217865. [Google Scholar] [CrossRef]
  50. García-Domínguez, M. Fibromyalgia and Inflammation: Unrevealing the Connection. Cells 2025, 14, 271. [Google Scholar] [CrossRef]
  51. Ferrés, S.; Serrat, M.; Auer, W.; Royuela-Colomer, E.; Almirall, M.; Lizama-Lefno, A.; Nijs, J.; Maes, M.; Luciano, J.V.; Borràs, X.; et al. Immune-inflammatory effects of the multicomponent intervention FIBROWALK in outdoor and online formats for patients with fibromyalgia. Brain Behav. Immun. 2025, 125, 184–197. [Google Scholar] [CrossRef]
  52. Zhang, C.; Zhang, P.; Zhao, Y.; Liu, Y.; Hu, Y.; Zhu, Z.; Xiao, H. The Effect of a Remote Network Technology Supervised Exercise Program Combined With Drug Treatment for Fibromyalgia: Randomized, Single-Blind, Controlled Trial. J. Med. Internet Res. 2025, 27, e71624. [Google Scholar] [CrossRef]
  53. Zhao, S.; Martin, E.M.; Reuken, P.A.; Scholcz, A.; Ganse-Dumrath, A.; Srowig, A.; Utech, I.; Kozik, V.; Radscheidt, M.; Brodoehl, S.; et al. Long COVID is associated with severe cognitive slowing: A multicentre cross-sectional study. EClinicalMedicine 2024, 68, 102434. [Google Scholar] [CrossRef] [PubMed]
  54. Wang, W.; Cui, R.; Leng, L.; Wang, G.; Peng, G. Cognitive Impairment in the Post-Acute Phases of COVID-19 and Mechanisms: An Introduction and Narrative Review. J. Alzheimers Dis. Rep. 2024, 8, 647–658. [Google Scholar] [CrossRef]
  55. Liew, F.; Efstathiou, C.; Fontanella, S.; Richardson, M.; Saunders, R.; Swieboda, D.; Sidhu, J.K.; Ascough, S.; Moore, S.C.; Mohamed, N.; et al. Large-scale phenotyping of patients with long COVID post-hospitalization reveals mechanistic subtypes of disease. Nat. Immunol. 2024, 25, 607–621. [Google Scholar] [CrossRef] [PubMed]
  56. Lipsky, R.K.; Helmer, D.A.; Beckham, J.C.; Calhoun, P.S.; Pugh, M.J.; Kimbrel, N.A. The association between Gulf War Illness and suicidal thoughts and behaviors among Gulf War Era Veterans. J. Psychiatr. Res. 2025, 183, 302–307. [Google Scholar] [CrossRef]
  57. Van Wilgen, C.P.; Ucles-Juarez, R.; Krutko, D.; Li, Y.; Polli, A.; Syed, A.; Zampese, S.; Reis, F.J.J.; de Zeeuw, J. Knowledge on cause, clinical manifestation and treatment for fibromyalgia among medical doctors: A worldwide survey. Pain Pract. 2024, 24, 620–626. [Google Scholar] [CrossRef] [PubMed]
  58. Zeron-Rugerio, M.F.; Zaragoza, M.C.; Domingo, J.C.; Sanmartin-Sentanes, R.; Alegre-Martin, J.; Castro-Marrero, J.; Cambras, T. Sleep and circadian rhythm alterations in myalgic encephalomyelitis/chronic fatigue syndrome and post-COVID fatigue syndrome and its association with cardiovascular risk factors: A prospective cohort study. Chronobiol. Int. 2024, 41, 1104–1115. [Google Scholar] [CrossRef]
  59. Winograd, D.M.; Sullivan, N.L.; Thien, S.R.; Pigeon, W.R.; Litke, D.R.; Helmer, D.A.; Rath, J.F.; Lu, S.E.; McAndrew, L.M. Veterans with Gulf War Illness perceptions of management strategies. Life Sci. 2021, 279, 119219. [Google Scholar] [CrossRef]
  60. Karabey Aksalli, I.; Baygin, N.; Hagiwara, Y.; Paul, J.K.; Iype, T.; Barua, P.D.; Koh, J.E.W.; Baygin, M.; Dogan, S.; Tuncer, T.; et al. Automated characterization and detection of fibromyalgia using slow wave sleep EEG signals with glucose pattern and D’hondt pooling technique. Cogn. Neurodyn. 2024, 18, 383–404. [Google Scholar] [CrossRef]
  61. Lederman, S.; Arnold, L.M.; Vaughn, B.; Engels, J.M.; Kelley, M.; Sullivan, G.M. Pain Relief by Targeting Nonrestorative Sleep in Fibromyalgia: A Phase 3 Randomized Trial of Bedtime Sublingual Cyclobenzaprine. Pain Med. 2025, pnaf089. [Google Scholar] [CrossRef]
  62. Malluru, N.; Abdullah, Y.; Hackshaw, K.V. Early diagnostics of fibromyalgia: An overview of the challenges and opportunities. Expert Rev. Mol. Diagn. 2025, 25, 21–31. [Google Scholar] [CrossRef] [PubMed]
  63. Benjamin, N.Z.Y.; Parekh, R.S.; Inban, P.; Sakthi, S.; Tekuru, Y.; Prajjwal, P.; John, J.; Sharma, R. Fibromyalgia: Advances in pathophysiology, diagnostic biomarkers, genetic insights, multisystemic involvement, and treatment updates and multidisciplinary interventions. Dis. Mon. 2025, 71, 101965. [Google Scholar] [CrossRef] [PubMed]
  64. Climent-Sanz, C.; Valenzuela-Pascual, F.; Martínez-Navarro, O.; Blanco-Blanco, J.; Rubí-Carnacea, F.; García-Martínez, E.; Soler-González, J.; Barallat-Gimeno, E.; Gea-Sánchez, M. Cognitive behavioral therapy for insomnia (CBT-i) in patients with fibromyalgia: A systematic review and meta-analysis. Disabil. Rehabil. 2022, 44, 5770–5783. [Google Scholar] [CrossRef]
  65. Imamura, M.; Robertson, C.; Hudson, J.; Whibley, D.; Aucott, L.; Gillies, K.; Beasley, M.; Stevens, M.J.; Manson, P.; Dulake, D.; et al. Effects of pharmacological and non-pharmacological interventions for the management of sleep problems in people with fibromyalgia: A multi-methods evidence synthesis. Health Technol. Assess. 2025, 29, 1–228. [Google Scholar] [CrossRef]
  66. Castro-Marrero, J.; Zaragozá, M.C.; González-Garcia, S.; Aliste, L.; Sáez-Francàs, N.; Romero, O.; Ferré, A.; Fernández de Sevilla, T.; Alegre, J. Poor self-reported sleep quality and health-related quality of life in patients with chronic fatigue syndrome/myalgic encephalomyelitis. J. Sleep Res. 2018, 27, e12703. [Google Scholar] [CrossRef] [PubMed]
  67. Weigel, B.; Eaton-Fitch, N.; Thapaliya, K.; Marshall-Gradisnik, S. Illness presentation and quality of life in myalgic encephalomyelitis/chronic fatigue syndrome and post COVID-19 condition: A pilot Australian cross-sectional study. Qual. Life Res. 2024, 33, 2489–2507. [Google Scholar] [CrossRef]
  68. Mohamed, A.Z.; Andersen, T.; Radovic, S.; Del Fante, P.; Kwiatek, R.; Calhoun, V.; Bhuta, S.; Hermens, D.F.; Lagopoulos, J.; Shan, Z.Y. Objective sleep measures in chronic fatigue syndrome patients: A systematic review and meta-analysis. Sleep Med. Rev. 2023, 69, 101771. [Google Scholar] [CrossRef]
  69. Yin, J.; Xu, J.; Ren, T.L. Recent Progress in Long-Term Sleep Monitoring Technology. Biosensors 2023, 13, 395. [Google Scholar] [CrossRef]
  70. Chao, L.L. Examining the current health of Gulf War veterans with the veterans affairs frailty index. Front. Neurosci. 2023, 17, 1245811. [Google Scholar] [CrossRef]
  71. Pettersen, P.S.; Haugmark, T.; Berg, I.J.; Hammer, H.B.; Neogi, T.; Zangi, H.; Haugen, I.K.; Provan, S.A. Pain sensitization in fibromyalgia. Cross-sectional associations between quantitative sensory testing of pain sensitization and fibromyalgia disease burden. Eur. J. Pain 2025, 29, e4771. [Google Scholar] [CrossRef]
  72. Escalante, A.; Fischbach, M. Musculoskeletal manifestations, pain, and quality of life in Persian Gulf War veterans referred for rheumatologic evaluation. J. Rheumatol. 1998, 25, 2228–2235. [Google Scholar]
  73. Castro-Marrero, J.; Cordero, M.D.; Sáez-Francas, N.; Jimenez-Gutierrez, C.; Aguilar-Montilla, F.J.; Aliste, L.; Alegre-Martin, J. Could mitochondrial dysfunction be a differentiating marker between chronic fatigue syndrome and fibromyalgia? Antioxid. Redox Signal. 2013, 19, 1855–1860. [Google Scholar] [CrossRef] [PubMed]
  74. Chen, Y.; Meyer, J.N.; Hill, H.Z.; Lange, G.; Condon, M.R.; Klein, J.C.; Ndirangu, D.; Falvo, M.J. Role of mitochondrial DNA damage and dysfunction in veterans with Gulf War Illness. PLoS ONE 2017, 12, e0184832. [Google Scholar] [CrossRef]
  75. Koslik, H.J.; Hamilton, G.; Golomb, B.A. Mitochondrial dysfunction in Gulf War illness revealed by 31Phosphorus Magnetic Resonance Spectroscopy: A case-control study. PLoS ONE 2014, 9, e92887. [Google Scholar] [CrossRef] [PubMed]
  76. Golomb, B.A.; Han, J.H.; Fung, A.; Berg, B.K.; Miller, B.J.; Hamilton, G. Bioenergetic impairment in Gulf War illness assessed via (31)P-MRS. Sci. Rep. 2024, 14, 7418. [Google Scholar] [CrossRef]
  77. Golomb, B.A.; Kelley, R.I.; Han, J.H.; Miller, B.; Bui, L. Gulf war illness: A tale of two genomes. BMC Res. Notes 2024, 17, 230. [Google Scholar] [CrossRef] [PubMed]
  78. Golomb, B.A.; Sanchez Baez, R.; Schilling, J.M.; Dhanani, M.; Fannon, M.J.; Berg, B.K.; Miller, B.J.; Taub, P.R.; Patel, H.H. Mitochondrial impairment but not peripheral inflammation predicts greater Gulf War illness severity. Sci. Rep. 2023, 13, 10739. [Google Scholar] [CrossRef]
  79. Shetty, G.A.; Hattiangady, B.; Upadhya, D.; Bates, A.; Attaluri, S.; Shuai, B.; Kodali, M.; Shetty, A.K. Chronic Oxidative Stress, Mitochondrial Dysfunction, Nrf2 Activation and Inflammation in the Hippocampus Accompany Heightened Systemic Inflammation and Oxidative Stress in an Animal Model of Gulf War Illness. Front. Mol. Neurosci. 2017, 10, 182. [Google Scholar] [CrossRef]
  80. Cordero, M.D.; de Miguel, M.; Carmona-López, I.; Bonal, P.; Campa, F.; Moreno-Fernández, A.M. Oxidative stress and mitochondrial dysfunction in fibromyalgia. Neuro Endocrinol. Lett. 2010, 31, 169–173. [Google Scholar]
  81. Assavarittirong, C.; Samborski, W.; Grygiel-Górniak, B. Oxidative Stress in Fibromyalgia: From Pathology to Treatment. Oxid. Med. Cell. Longev. 2022, 2022, 1582432. [Google Scholar] [CrossRef]
  82. Holden, S.; Maksoud, R.; Eaton-Fitch, N.; Cabanas, H.; Staines, D.; Marshall-Gradisnik, S. A systematic review of mitochondrial abnormalities in myalgic encephalomyelitis/chronic fatigue syndrome/systemic exertion intolerance disease. J. Transl. Med. 2020, 18, 290. [Google Scholar] [CrossRef]
  83. Smits, B.; van den Heuvel, L.; Knoop, H.; Küsters, B.; Janssen, A.; Borm, G.; Bleijenberg, G.; Rodenburg, R.; van Engelen, B. Mitochondrial enzymes discriminate between mitochondrial disorders and chronic fatigue syndrome. Mitochondrion 2011, 11, 735–738. [Google Scholar] [CrossRef] [PubMed]
  84. Tomas, C.; Brown, A.; Strassheim, V.; Elson, J.L.; Newton, J.; Manning, P. Cellular bioenergetics is impaired in patients with chronic fatigue syndrome. PLoS ONE 2017, 12, e0186802. [Google Scholar] [CrossRef]
  85. Molnar, T.; Lehoczki, A.; Fekete, M.; Varnai, R.; Zavori, L.; Erdo-Bonyar, S.; Simon, D.; Berki, T.; Csecsei, P.; Ezer, E. Mitochondrial dysfunction in long COVID: Mechanisms, consequences, and potential therapeutic approaches. Geroscience 2024, 46, 5267–5286. [Google Scholar] [CrossRef]
  86. Szögi, T.; Borsos, B.N.; Masic, D.; Radics, B.; Bella, Z.; Bánfi, A.; Ördög, N.; Zsiros, C.; Kiricsi, Á.; Pankotai-Bodó, G.; et al. Novel biomarkers of mitochondrial dysfunction in Long COVID patients. Geroscience 2025, 47, 2245–2261. [Google Scholar] [CrossRef] [PubMed]
  87. Stufano, A.; Isgrò, C.; Palese, L.L.; Caretta, P.; De Maria, L.; Lovreglio, P.; Sardanelli, A.M. Oxidative Damage and Post-COVID Syndrome: A Cross-Sectional Study in a Cohort of Italian Workers. Int. J. Mol. Sci. 2023, 24, 7445. [Google Scholar] [CrossRef]
  88. Cox, B.; Goolkasian, P.; Palomo, I.M.; Williams, M.V.; Maloney, S.R.; Ariza, M.E. Reactivation of Latent Herpesviruses and a Faulty Antiviral Response may Contribute to Chronic Multi-Symptom and Multi-System Illnesses in U.S. Military Veterans. J. Med. Virol. 2025, 97, e70400. [Google Scholar] [CrossRef]
  89. Mettler, J.; Ming-Azevedo, P.; Hügle, T. Fibromyalgia with concomitant immune-mediated rheumatic diseases: An evaluation of clinical characteristics, diagnostic criteria and multimodal treatment outcomes. Adv. Rheumatol. 2025, 65, 27. [Google Scholar] [CrossRef] [PubMed]
  90. Burzynski, H.E.; Reagan, L.P. Exposing the latent phenotype of Gulf War Illness: Examination of the mechanistic mediators of cognitive dysfunction. Front. Immunol. 2024, 15, 1403574. [Google Scholar] [CrossRef]
  91. Trageser, K.J.; Sebastian-Valverde, M.; Naughton, S.X.; Pasinetti, G.M. The Innate Immune System and Inflammatory Priming: Potential Mechanistic Factors in Mood Disorders and Gulf War Illness. Front. Psychiatry 2020, 11, 704. [Google Scholar] [CrossRef]
  92. Whistler, T.; Fletcher, M.A.; Lonergan, W.; Zeng, X.R.; Lin, J.M.; Laperriere, A.; Vernon, S.D.; Klimas, N.G. Impaired immune function in Gulf War Illness. BMC Med. Genom. 2009, 2, 12. [Google Scholar] [CrossRef]
  93. Klein, J.; Wood, J.; Jaycox, J.R.; Dhodapkar, R.M.; Lu, P.; Gehlhausen, J.R.; Tabachnikova, A.; Greene, K.; Tabacof, L.; Malik, A.A.; et al. Distinguishing features of long COVID identified through immune profiling. Nature 2023, 623, 139–148. [Google Scholar] [CrossRef]
  94. Ruf, W. Immune damage in Long COVID. Science 2024, 383, 262–263. [Google Scholar] [CrossRef] [PubMed]
  95. Yin, K.; Peluso, M.J.; Luo, X.; Thomas, R.; Shin, M.G.; Neidleman, J.; Andrew, A.; Young, K.C.; Ma, T.; Hoh, R.; et al. Long COVID manifests with T cell dysregulation, inflammation and an uncoordinated adaptive immune response to SARS-CoV-2. Nat. Immunol. 2024, 25, 218–225. [Google Scholar] [CrossRef]
  96. Eaton-Fitch, N.; Rudd, P.; Er, T.; Hool, L.; Herrero, L.; Marshall-Gradisnik, S. Immune exhaustion in ME/CFS and long COVID. JCI Insight 2024, 9, e183810. [Google Scholar] [CrossRef] [PubMed]
  97. Lutz, L.; Rohrhofer, J.; Zehetmayer, S.; Stingl, M.; Untersmayr, E. Evaluation of Immune Dysregulation in an Austrian Patient Cohort Suffering from Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. Biomolecules 2021, 11, 1359. [Google Scholar] [CrossRef] [PubMed]
  98. O’Neal, A.J.; Hanson, M.R. The Enterovirus Theory of Disease Etiology in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: A Critical Review. Front. Med. 2021, 8, 688486. [Google Scholar] [CrossRef]
  99. Vinker-Shuster, M.; Magen, E.; Green, I.; Merzon, E.; Golan-Cohen, A.; Israel, A. Increased Rates of Infectious Diseases in Fibromyalgia Patients: A Population-Based Case-Control Study. Biomedicines 2024, 12, 2821. [Google Scholar] [CrossRef]
  100. Clauw, D.; Sarzi-Puttini, P.; Pellegrino, G.; Shoenfeld, Y. Is fibromyalgia an autoimmune disorder? Autoimmun. Rev. 2024, 23, 103424. [Google Scholar] [CrossRef]
  101. Findeisen, K.; Guymer, E.; Littlejohn, G. Neuroinflammatory and Immunological Aspects of Fibromyalgia. Brain Sci. 2025, 15, 206. [Google Scholar] [CrossRef] [PubMed]
  102. Goebel, A.; Krock, E.; Gentry, C.; Israel, M.R.; Jurczak, A.; Urbina, C.M.; Sandor, K.; Vastani, N.; Maurer, M.; Cuhadar, U.; et al. Passive transfer of fibromyalgia symptoms from patients to mice. J. Clin. Investig. 2021, 131, e144201. [Google Scholar] [CrossRef] [PubMed]
  103. Camici, M.; Del Duca, G.; Brita, A.C.; Antinori, A. Connecting dots of long COVID-19 pathogenesis: A vagus nerve- hypothalamic-pituitary- adrenal-mitochondrial axis dysfunction. Front. Cell. Infect. Microbiol. 2024, 14, 1501949. [Google Scholar] [CrossRef]
  104. Tak, L.M.; Cleare, A.J.; Ormel, J.; Manoharan, A.; Kok, I.C.; Wessely, S.; Rosmalen, J.G. Meta-analysis and meta-regression of hypothalamic-pituitary-adrenal axis activity in functional somatic disorders. Biol. Psychol. 2011, 87, 183–194. [Google Scholar] [CrossRef]
  105. O’Callaghan, J.P.; Miller, D.B. Neuroinflammation disorders exacerbated by environmental stressors. Metabolism 2019, 100, 153951. [Google Scholar] [CrossRef]
  106. Golier, J.A.; Caramanica, K.; Yehuda, R. Neuroendocrine response to CRF stimulation in veterans with and without PTSD in consideration of war zone era. Psychoneuroendocrinology 2012, 37, 350–357. [Google Scholar] [CrossRef]
  107. Tomic, S.; Brkic, S.; Lendak, D.; Maric, D.; Medic Stojanoska, M.; Novakov Mikic, A. Neuroendocrine disorder in chronic fatigue syndrome. Turk. J. Med. Sci. 2017, 47, 1097–1103. [Google Scholar] [CrossRef]
  108. Demori, I.; Losacco, S.; Giordano, G.; Mucci, V.; Blanchini, F.; Burlando, B. Fibromyalgia pathogenesis explained by a neuroendocrine multistable model. PLoS ONE 2024, 19, e0303573. [Google Scholar] [CrossRef] [PubMed]
  109. Sasikumar, S.; Unniappan, S. SARS-CoV-2 Infection and the Neuroendocrine System. Neuroendocrinology 2024, 114, 1158–1175. [Google Scholar] [CrossRef]
  110. Kimono, D.A. Gastrointestinal problems, mechanisms and possible therapeutic directions in Gulf war illness: A mini review. Mil. Med. Res. 2021, 8, 50. [Google Scholar] [CrossRef]
  111. Trivedi, A.; Bose, D.; Moffat, K.; Pearson, E.; Walsh, D.; Cohen, D.; Skupsky, J.; Chao, L.; Golier, J.; Janulewicz, P.; et al. Gulf War Illness Is Associated with Host Gut Microbiome Dysbiosis and Is Linked to Altered Species Abundance in Veterans from the BBRAIN Cohort. Int. J. Environ. Res. Public Health 2024, 21, 1102. [Google Scholar] [CrossRef]
  112. Keating, J.A.; Shaughnessy, C.; Baubie, K.; Kates, A.E.; Putman-Buehler, N.; Watson, L.; Dominguez, N.; Watson, K.; Cook, D.B.; Rabago, D.; et al. Characterising the gut microbiome in veterans with Gulf War Illness: A protocol for a longitudinal, prospective cohort study. BMJ Open 2019, 9, e031114. [Google Scholar] [CrossRef]
  113. Nono Djotsa, A.B.S.; Nguyen Wenker, T.H.; Ahmed, S.T.; Ghosh, S.; Malhotra, D.; Boyle, S.H.; Gifford, E.J.; Sims, K.J.; White, D.L.; Steele, L.; et al. Irritable Bowel Syndrome in Veterans With Gulf War Illness Evaluated at VA’s War-Related Illness and Injury Study Center. Mil. Med. 2024, 189, e2644–e2654. [Google Scholar] [CrossRef]
  114. Malhotra, D.; Boyle, S.H.; Gifford, E.J.; Sullivan, B.A.; Nguyen Wenker, T.H.; Abs, N.D.; Ahmed, S.T.; Upchurch, J.; Vahey, J.; Stafford, C.; et al. Self-reported gastrointestinal disorders among veterans with gulf war illness with and without posttraumatic stress disorder. Neurogastroenterol Motil. 2023, 35, e14548. [Google Scholar] [CrossRef]
  115. Chatterjee, S.; Bose, D.; Seth, R. Host gut microbiome and potential therapeutics in Gulf War Illness: A short review. Life Sci. 2021, 280, 119717. [Google Scholar] [CrossRef] [PubMed]
  116. Saha, P.; Skidmore, P.T.; Holland, L.A.; Mondal, A.; Bose, D.; Seth, R.K.; Sullivan, K.; Janulewicz, P.A.; Horner, R.; Klimas, N.; et al. Andrographolide Attenuates Gut-Brain-Axis Associated Pathology in Gulf War Illness by Modulating Bacteriome-Virome Associated Inflammation and Microglia-Neuron Proinflammatory Crosstalk. Brain Sci. 2021, 11, 905. [Google Scholar] [CrossRef] [PubMed]
  117. Collier, C.A.; Salikhova, A.; Sabir, S.; Raghavan, S.A. Persistent enteric neuroinflammation chronically impairs colonic motility in a pyridostigmine bromide-induced mouse model of Gulf War illness. Biol. Open 2025, 14, bio061867. [Google Scholar] [CrossRef]
  118. Martín, F.; Blanco-Suárez, M.; Zambrano, P.; Cáceres, O.; Almirall, M.; Alegre-Martín, J.; Lobo, B.; González-Castro, A.M.; Santos, J.; Domingo, J.C.; et al. Increased gut permeability and bacterial translocation are associated with fibromyalgia and myalgic encephalomyelitis/chronic fatigue syndrome: Implications for disease-related biomarker discovery. Front. Immunol. 2023, 14, 1253121. [Google Scholar] [CrossRef]
  119. Shtrozberg, S.; Bazzichi, L.; Sarzi-Puttini, P.; Aloush, V.; Ablin, J.N. Is the gut microbiome of importance in fibromyalgia? A critical review of emerging evidence. Clin. Exp. Rheumatol. 2025, 43, 990–998. [Google Scholar] [CrossRef] [PubMed]
  120. Waterman, A.; Doumas, S.A.; Fischer, M.; Mattar, M.; Charbel, S.; Jennings, J.; Doman, D.B. Uncovering the Hidden Link Between the Aberrant Intestinal Microbiome and Fibromyalgia. Gastroenterol. Hepatol. 2025, 21, 111–121. [Google Scholar]
  121. Iannuccelli, C.; Favretti, M.; Dolcini, G.; Di Carlo, M.; Pellegrino, G.; Bazzichi, L.; Atzeni, F.; Lucini, D.; Varassi, G.; Leoni, M.L.G.; et al. Fibromyalgia: One year in review 2025. Clin. Exp. Rheumatol. 2025, 43, 957–969. [Google Scholar] [CrossRef]
  122. Jurek, J.M.; Castro-Marrero, J. A Narrative Review on Gut Microbiome Disturbances and Microbial Preparations in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: Implications for Long COVID. Nutrients 2024, 16, 1545. [Google Scholar] [CrossRef] [PubMed]
  123. Trautmann, A. Core features and inherent diversity of post-acute infection syndromes. Front. Immunol. 2025, 16, 1509131. [Google Scholar] [CrossRef]
  124. Guo, C.; Che, X.; Briese, T.; Ranjan, A.; Allicock, O.; Yates, R.A.; Cheng, A.; March, D.; Hornig, M.; Komaroff, A.L.; 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.e288. [Google Scholar] [CrossRef] [PubMed]
  125. Tian, H.; Wang, L.; Aiken, E.; Ortega, R.J.V.; Hardy, R.; Placek, L.; Kozhaya, L.; Unutmaz, D.; Oh, J.; Yao, X. Fast Targeted Metabolomics for Analyzing Metabolic Diversity of Bacterial Indole Derivatives in ME/CFS Gut Microbiome. bioRxiv 2024. [Google Scholar] [CrossRef] [PubMed]
  126. Xiong, R.; Gunter, C.; Fleming, E.; Vernon, S.D.; Bateman, L.; Unutmaz, D.; Oh, J. Multi-’omics of gut microbiome-host interactions in short- and long-term myalgic encephalomyelitis/chronic fatigue syndrome patients. Cell Host Microbe 2023, 31, 273–287.e275. [Google Scholar] [CrossRef]
  127. Lau, R.I.; Su, Q.; Ng, S.C. Long COVID and gut microbiome: Insights into pathogenesis and therapeutics. Gut Microbes 2025, 17, 2457495. [Google Scholar] [CrossRef]
  128. Oh, S.; An, S.; Park, K.; Lee, S.; Han, Y.M.; Koh, S.J.; Lee, J.; Gim, H.; Kim, D.; Seo, H. Gut Microbial Signatures in Long COVID: Potential Biomarkers and Therapeutic Targets. Infect. Dis. Ther. 2025, 14, 1461–1475. [Google Scholar] [CrossRef]
  129. Smail, S.W.; Albarzinji, N.; Salih, R.H.; Taha, K.O.; Hirmiz, S.M.; Ismael, H.M.; Noori, M.F.; Azeez, S.S.; Janson, C. Microbiome dysbiosis in SARS-CoV-2 infection: Implication for pathophysiology and management strategies of COVID-19. Front. Cell. Infect. Microbiol. 2025, 15, 1537456. [Google Scholar] [CrossRef]
  130. Yao, L.; Devotta, H.; Li, J.; Lunjani, N.; Sadlier, C.; Lavelle, A.; Albrich, W.C.; Walter, J.; O’Toole, P.W.; O’Mahony, L. Dysrupted microbial tryptophan metabolism associates with SARS-CoV-2 acute inflammatory responses and long COVID. Gut Microbes 2024, 16, 2429754. [Google Scholar] [CrossRef]
  131. Yu, L.C. Gastrointestinal pathophysiology in long COVID: Exploring roles of microbiota dysbiosis and serotonin dysregulation in post-infectious bowel symptoms. Life Sci. 2024, 358, 123153. [Google Scholar] [CrossRef] [PubMed]
  132. El-Sehrawy, A.; Ayoub, I.I.; Uthirapathy, S.; Ballal, S.; Gabble, B.C.; Singh, A.; V, K.; Panigrahi, R.; Kamali, M.; Khosravi, M. The microbiota-gut-brain axis in myalgic encephalomyelitis/chronic fatigue syndrome: A narrative review of an emerging field. Eur. J. Transl. Myol. 2025, 35, 13690. [Google Scholar] [CrossRef]
  133. Elmore, S. Apoptosis: A review of programmed cell death. Toxicol. Pathol. 2007, 35, 495–516. [Google Scholar] [CrossRef] [PubMed]
  134. Li, J.; Kang, R.; Tang, D. Monitoring autophagy-dependent ferroptosis. Methods Cell Biol. 2021, 165, 163–176. [Google Scholar] [CrossRef] [PubMed]
  135. Georgopoulos, A.P.; James, L.M.; Carpenter, A.F.; Engdahl, B.E.; Leuthold, A.C.; Lewis, S.M. Gulf War illness (GWI) as a neuroimmune disease. Exp. Brain Res. 2017, 235, 3217–3225. [Google Scholar] [CrossRef]
  136. Tsilibary, E.C.; Souto, E.P.; Kratzke, M.; James, L.M.; Engdahl, B.E.; Georgopoulos, A.P. Anthrax Protective Antigen 63 (PA63): Toxic Effects in Neural Cultures and Role in Gulf War Illness (GWI). Neurosci. Insights 2020, 15, 2633105520931966. [Google Scholar] [CrossRef] [PubMed]
  137. Martínez-Lavín, M. Dorsal root ganglia: Fibromyalgia pain factory? Clin. Rheumatol. 2021, 40, 783–787. [Google Scholar] [CrossRef]
  138. Yüksel, E.; Nazıroğlu, M.; Şahin, M.; Çiğ, B. Involvement of TRPM2 and TRPV1 channels on hyperalgesia, apoptosis and oxidative stress in rat fibromyalgia model: Protective role of selenium. Sci. Rep. 2017, 7, 17543. [Google Scholar] [CrossRef]
  139. Gheita, T.A.; El Sisi, R.W.; Raafat, H.A.; Khalil, H.M. Anti-annexin V antibodies in primary fibromyalgia syndrome: Relation to associated Sjögren’s syndrome. J. Clin. Immunol. 2013, 33, 311–312. [Google Scholar] [CrossRef]
  140. Wang, Y.; Zhang, Z.; Jiao, W.; Wang, Y.; Wang, X.; Zhao, Y.; Fan, X.; Tian, L.; Li, X.; Mi, J. Ferroptosis and its role in skeletal muscle diseases. Front. Mol. Biosci. 2022, 9, 1051866. [Google Scholar] [CrossRef]
  141. Jiang, L.; Zhu, Z.; Han, K.; Chen, D.; Huang, L.; Hu, N.; Du, J.; Zhang, B.; Su, Y.; Li, T.; et al. Magnesium hexacyanoferrate nanocatalysts alleviates fibromyalgia syndrome by reversing cellular ferroptosis. J. Chem. Eng. 2024, 498, 155019. [Google Scholar] [CrossRef]
  142. Kennedy, G.; Khan, F.; Hill, A.; Underwood, C.; Belch, J.J. Biochemical and vascular aspects of pediatric chronic fatigue syndrome. Arch. Pediatr. Adolesc. Med. 2010, 164, 817–823. [Google Scholar] [CrossRef]
  143. Cezar, R.; Kundura, L.; André, S.; Lozano, C.; Vincent, T.; Muller, L.; Lefrant, J.Y.; Roger, C.; Claret, P.G.; Duvnjak, S.; et al. T4 apoptosis in the acute phase of SARS-CoV-2 infection predicts long COVID. Front. Immunol. 2023, 14, 1335352. [Google Scholar] [CrossRef]
  144. Biamonte, F.; Botta, C.; Mazzitelli, M.; Rotundo, S.; Trecarichi, E.M.; Foti, D.; Torti, C.; Viglietto, G.; Torella, D.; Costanzo, F. Combined lymphocyte/monocyte count, D-dimer and iron status predict COVID-19 course and outcome in a long-term care facility. J. Transl. Med. 2021, 19, 79. [Google Scholar] [CrossRef]
  145. Sousa, R.A.L.; Yehia, A.; Abulseoud, O.A. Attenuation of ferroptosis as a potential therapeutic target for neuropsychiatric manifestations of post-COVID syndrome. Front. Neurosci. 2023, 17, 1237153. [Google Scholar] [CrossRef]
  146. Yang, L.; Wu, Y.; Jin, W.; Mo, N.; Ye, G.; Su, Z.; Tang, L.; Wang, Y.; Li, Y.; Du, J. The potential role of ferroptosis in COVID-19-related cardiovascular injury. Biomed. Pharmacother. 2023, 168, 115637. [Google Scholar] [CrossRef] [PubMed]
  147. Zhao, W.; Wang, S.; Han, Y.; Zhang, H.; Cao, J.; Dong, S.; Li, D.; Lei, M.; Liu, C.; Gao, Y. Role of Ferroptosis in the Progression of COVID-19 and the Development of Long COVID. Curr. Med. Chem. 2024, 32, 4324–4342. [Google Scholar] [CrossRef] [PubMed]
  148. Smith, E.M.; Pendlebury, D.F.; Nandakumar, J. Structural biology of telomeres and telomerase. Cell. Mol. Life Sci. 2020, 77, 61–79. [Google Scholar] [CrossRef] [PubMed]
  149. Starkweather, A.R.; Alhaeeri, A.A.; Montpetit, A.; Brumelle, J.; Filler, K.; Montpetit, M.; Mohanraj, L.; Lyon, D.E.; Jackson-Cook, C.K. An integrative review of factors associated with telomere length and implications for biobehavioral research. Nurs. Res. 2014, 63, 36–50. [Google Scholar] [CrossRef]
  150. Gao, X.; Yu, X.; Zhang, C.; Wang, Y.; Sun, Y.; Sun, H.; Zhang, H.; Shi, Y.; He, X. Telomeres and Mitochondrial Metabolism: Implications for Cellular Senescence and Age-related Diseases. Stem Cell Rev. Rep. 2022, 18, 2315–2327. [Google Scholar] [CrossRef]
  151. Nassour, J.; Przetocka, S.; Karlseder, J. Telomeres as hotspots for innate immunity and inflammation. DNA Repair 2024, 133, 103591. [Google Scholar] [CrossRef]
  152. Opresko, P.L.; Sanford, S.L.; De Rosa, M. Oxidative Stress and DNA Damage at Telomeres. Cold Spring Harb. Perspect. Biol. 2025, 17, a041707. [Google Scholar] [CrossRef] [PubMed]
  153. Armanios, M. The Role of Telomeres in Human Disease. Annu. Rev. Genom. Hum. Genet. 2022, 23, 363–381. [Google Scholar] [CrossRef] [PubMed]
  154. Howard, J.T.; Janak, J.C.; Santos-Lozada, A.R.; McEvilla, S.; Ansley, S.D.; Walker, L.E.; Spiro, A.; Stewart, I.J. Telomere Shortening and Accelerated Aging in US Military Veterans. Int. J. Environ. Res. Public Health 2021, 18, 1743. [Google Scholar] [CrossRef]
  155. Charatan, F. Nerve gas antidote a possible cause of gulf war illness. BMJ 1999, 319, 1154. [Google Scholar] [CrossRef]
  156. Mantle, D.; Golomb, B.A. Coenzyme Q10 and Xenobiotic Metabolism: An Overview. Int. J. Mol. Sci. 2025, 26, 5788. [Google Scholar] [CrossRef]
  157. Rajeevan, M.S.; Murray, J.; Oakley, L.; Lin, J.S.; Unger, E.R. Association of chronic fatigue syndrome with premature telomere attrition. J. Transl. Med. 2018, 16, 44. [Google Scholar] [CrossRef]
  158. Hassett, A.L.; Epel, E.; Clauw, D.J.; Harris, R.E.; Harte, S.E.; Kairys, A.; Buyske, S.; Williams, D.A. Pain is associated with short leukocyte telomere length in women with fibromyalgia. J. Pain 2012, 13, 959–969. [Google Scholar] [CrossRef]
  159. Dos Reis, G.G.; Silvestre, R.T.; Alves, G.; Delmonico, L.; Chantre-Justino, M.; Moreira, A.D.S.; Müller, B.L.A.; do Nascimento, C.R.; da Silva, D.L.P.; Dos Santos, L.S.; et al. Leukocyte telomere length and telomerase activity in Long COVID patients from Rio de Janeiro, Brazil. Mem. Inst. Oswaldo Cruz 2025, 120, e240129. [Google Scholar] [CrossRef]
  160. Mongelli, A.; Barbi, V.; Gottardi Zamperla, M.; Atlante, S.; Forleo, L.; Nesta, M.; Massetti, M.; Pontecorvi, A.; Nanni, S.; Farsetti, A.; et al. Evidence for Biological Age Acceleration and Telomere Shortening in COVID-19 Survivors. Int. J. Mol. Sci. 2021, 22, 6151. [Google Scholar] [CrossRef] [PubMed]
  161. Polli, A.; Godderis, L.; Martens, D.S.; Patil, M.S.; Hendrix, J.; Wyns, A.; Van Campenhout, J.; Richter, E.; Fanning, L.; Vandekerckhove, O.; et al. Exploring DNA methylation, telomere length, mitochondrial DNA, and immune function in patients with Long-COVID. BMC Med. 2025, 23, 60. [Google Scholar] [CrossRef]
  162. Castro-Marrero, J.; Sáez-Francàs, N.; Santillo, D.; Alegre, J. Treatment and management of chronic fatigue syndrome/myalgic encephalomyelitis: All roads lead to Rome. Br. J. Pharmacol. 2017, 174, 345–369. [Google Scholar] [CrossRef] [PubMed]
  163. Chandan, J.S.; Brown, K.R.; Simms-Williams, N.; Bashir, N.Z.; Camaradou, J.; Heining, D.; Turner, G.M.; Rivera, S.C.; Hotham, R.; Minhas, S.; et al. Non-Pharmacological Therapies for Post-Viral Syndromes, Including Long COVID: A Systematic Review. Int. J. Environ. Res. Public Health 2023, 20, 3477. [Google Scholar] [CrossRef] [PubMed]
  164. Mengshoel, A.M.; Helland, I.B.; Meeus, M.; Castro-Marrero, J.; Pheby, D.; Bolle Strand, E. Patients’ experiences and effects of non-pharmacological treatment for myalgic encephalomyelitis/chronic fatigue syndrome—A scoping mixed methods review. Int. J. Qual. Stud. Health Well-Being 2020, 15, 1764830. [Google Scholar] [CrossRef] [PubMed]
  165. Nugent, S.M.; Freeman, M.; Ayers, C.K.; Winchell, K.A.; Press, A.M.; O’Neil, M.E.; Paynter, R.; Kansagara, D. A Systematic Review of Therapeutic Interventions and Management Strategies for Gulf War Illness. Mil. Med. 2021, 186, e169–e178. [Google Scholar] [CrossRef]
  166. Hetta, H.F.; Elsaghir, A.; Sijercic, V.C.; Ahmed, A.K.; Gad, S.A.; Zeleke, M.S.; Alanazi, F.E.; Ramadan, Y.N. Clinical Progress in Mesenchymal Stem Cell Therapy: A Focus on Rheumatic Diseases. Immun. Inflamm. Dis. 2025, 13, e70189. [Google Scholar] [CrossRef]
  167. Safina, I.; Embree, M.C. Biomaterials for recruiting and activating endogenous stem cells in situ tissue regeneration. Acta Biomater. 2022, 143, 26–38. [Google Scholar] [CrossRef]
  168. Liu, K.; Zhou, Z.; Pan, M.; Zhang, L. Stem cell-derived mitochondria transplantation: A promising therapy for mitochondrial encephalomyopathy. CNS Neurosci. Ther. 2021, 27, 733–742. [Google Scholar] [CrossRef]
  169. Xu, Z.; Zhang, L.; Fei, X.; Yi, X.; Li, W.; Wang, Q. The miR-29b-Sirt1 axis regulates self-renewal of mouse embryonic stem cells in response to reactive oxygen species. Cell. Signal. 2014, 26, 1500–1505. [Google Scholar] [CrossRef]
  170. Mahrouf-Yorgov, M.; Augeul, L.; Da Silva, C.C.; Jourdan, M.; Rigolet, M.; Manin, S.; Ferrera, R.; Ovize, M.; Henry, A.; Guguin, A.; et al. Mesenchymal stem cells sense mitochondria released from damaged cells as danger signals to activate their rescue properties. Cell Death Differ. 2017, 24, 1224–1238. [Google Scholar] [CrossRef]
  171. Ennis, W.J.; Sui, A.; Bartholomew, A. Stem Cells and Healing: Impact on Inflammation. Adv. Wound Care 2013, 2, 369–378. [Google Scholar] [CrossRef] [PubMed]
  172. Rebelatto, C.L.K.; Boldrini-Leite, L.M.; Daga, D.R.; Marsaro, D.B.; Vaz, I.M.; Jamur, V.R.; de Aguiar, A.M.; Vieira, T.B.; Furman, B.P.; Aguiar, C.O.; et al. Quality Control Optimization for Minimizing Security Risks Associated with Mesenchymal Stromal Cell-Based Product Development. Int. J. Mol. Sci. 2023, 24, 12955. [Google Scholar] [CrossRef] [PubMed]
  173. Shi, L.; Huang, H.; Lu, X.; Yan, X.; Jiang, X.; Xu, R.; Wang, S.; Zhang, C.; Yuan, X.; Xu, Z.; et al. Effect of human umbilical cord-derived mesenchymal stem cells on lung damage in severe COVID-19 patients: A randomized, double-blind, placebo-controlled phase 2 trial. Signal Transduct. Target Ther. 2021, 6, 58. [Google Scholar] [CrossRef]
  174. Yuan, M.Q.; Song, L.; Wang, Z.R.; Zhang, Z.Y.; Shi, M.; He, J.; Mo, Q.; Zheng, N.; Yao, W.Q.; Zhang, Y.; et al. Long-term outcomes of mesenchymal stem cell therapy in severe COVID-19 patients: 3-year follow-up of a randomized, double-blind, placebo-controlled trial. Stem Cell Res. Ther. 2025, 16, 94. [Google Scholar] [CrossRef]
  175. Mokhemer, S.A.; Desouky, M.K.; Abdelghany, A.K.; Ibrahim, M.F.G. Stem cells therapeutic effect in a reserpine-induced fibromyalgia rat model: A possible NLRP3 inflammasome modulation with neurogenesis promotion in the cerebral cortex. Life Sci. 2023, 325, 121784. [Google Scholar] [CrossRef]
  176. Golomb, B.A.; Allison, M.; Koperski, S.; Koslik, H.J.; Devaraj, S.; Ritchie, J.B. Coenzyme Q10 benefits symptoms in Gulf War veterans: Results of a randomized double-blind study. Neural Comput 2014, 26, 2594–2651. [Google Scholar] [CrossRef]
  177. Castro-Marrero, J.; Segundo, M.J.; Lacasa, M.; Martinez-Martinez, A.; Sentañes, R.S.; Alegre-Martin, J. Effect of Dietary Coenzyme Q10 Plus NADH Supplementation on Fatigue Perception and Health-Related Quality of Life in Individuals with Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: A Prospective, Randomized, Double-Blind, Placebo-Controlled Trial. Nutrients 2021, 13, 2658. [Google Scholar] [CrossRef]
  178. Nacul, L.; Authier, F.J.; Scheibenbogen, C.; Lorusso, L.; Helland, I.B.; Martin, J.A.; Sirbu, C.A.; Mengshoel, A.M.; Polo, O.; Behrends, U.; et al. European Network on Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (EUROMENE): Expert Consensus on the Diagnosis, Service Provision, and Care of People with ME/CFS in Europe. Medicina 2021, 57, 510. [Google Scholar] [CrossRef]
  179. Cordero, M.D.; Alcocer-Gómez, E.; de Miguel, M.; Culic, O.; Carrión, A.M.; Alvarez-Suarez, J.M.; Bullón, P.; Battino, M.; Fernández-Rodríguez, A.; Sánchez-Alcazar, J.A. Can coenzyme q10 improve clinical and molecular parameters in fibromyalgia? Antioxid. Redox Signal. 2013, 19, 1356–1361. [Google Scholar] [CrossRef]
  180. Sawaddiruk, P.; Apaijai, N.; Paiboonworachat, S.; Kaewchur, T.; Kasitanon, N.; Jaiwongkam, T.; Kerdphoo, S.; Chattipakorn, N.; Chattipakorn, S.C. Coenzyme Q10 supplementation alleviates pain in pregabalin-treated fibromyalgia patients via reducing brain activity and mitochondrial dysfunction. Free Radic Res. 2019, 53, 901–909. [Google Scholar] [CrossRef] [PubMed]
  181. Barletta, M.A.; Marino, G.; Spagnolo, B.; Bianchi, F.P.; Falappone, P.C.F.; Spagnolo, L.; Gatti, P. Coenzyme Q10 + alpha lipoic acid for chronic COVID syndrome. Clin. Exp. Med. 2023, 23, 667–678. [Google Scholar] [CrossRef] [PubMed]
  182. Hansen, K.S.; Mogensen, T.H.; Agergaard, J.; Schiøttz-Christensen, B.; Østergaard, L.; Vibholm, L.K.; Leth, S. High-dose coenzyme Q10 therapy versus placebo in patients with post COVID-19 condition: A randomized, phase 2, crossover trial. Lancet Reg. Health Eur. 2023, 24, 100539. [Google Scholar] [CrossRef] [PubMed]
  183. Opstad, T.B.; Alexander, J.; Aaseth, J.O.; Larsson, A.; Seljeflot, I.; Alehagen, U. Selenium and Coenzyme Q(10) Intervention Prevents Telomere Attrition, with Association to Reduced Cardiovascular Mortality-Sub-Study of a Randomized Clinical Trial. Nutrients 2022, 14, 3346. [Google Scholar] [CrossRef]
  184. Zhang, D.; Yan, B.; Yu, S.; Zhang, C.; Wang, B.; Wang, Y.; Wang, J.; Yuan, Z.; Zhang, L.; Pan, J. Coenzyme Q10 inhibits the aging of mesenchymal stem cells induced by D-galactose through Akt/mTOR signaling. Oxid. Med. Cell. Longev. 2015, 2015, 867293. [Google Scholar] [CrossRef]
  185. Li, X.; Zhan, J.; Hou, Y.; Hou, Y.; Chen, S.; Luo, D.; Luan, J.; Wang, L.; Lin, D. Coenzyme Q10 Regulation of Apoptosis and Oxidative Stress in H2O2 Induced BMSC Death by Modulating the Nrf-2/NQO-1 Signaling Pathway and Its Application in a Model of Spinal Cord Injury. Oxid. Med. Cell. Longev. 2019, 2019, 6493081. [Google Scholar] [CrossRef]
  186. Li, Z.; Zhao, T.; Ding, J.; Gu, H.; Wang, Q.; Wang, Y.; Zhang, D.; Gao, C. A reactive oxygen species-responsive hydrogel encapsulated with bone marrow derived stem cells promotes repair and regeneration of spinal cord injury. Bioact. Mater. 2023, 19, 550–568. [Google Scholar] [CrossRef]
  187. Li, T.; Li, X.; Han, G.; Liang, M.; Yang, Z.; Zhang, C.; Huang, S.; Tai, S.; Yu, S. The Therapeutic Potential and Clinical Significance of Exosomes as Carriers of Drug Delivery System. Pharmaceutics 2022, 15, 21. [Google Scholar] [CrossRef]
  188. Saka, O.M.; Dora, D.D.; Kibar, G.; Tevlek, A. Expanding the role of exosomes in drug, biomolecule, and nanoparticle delivery. Life Sci. 2025, 368, 123499. [Google Scholar] [CrossRef]
  189. Sheykhhasan, M.; Amini, R.; Soleimani Asl, S.; Saidijam, M.; Hashemi, S.M.; Najafi, R. Neuroprotective effects of coenzyme Q10-loaded exosomes obtained from adipose-derived stem cells in a rat model of Alzheimer’s disease. Biomed. Pharmacother. 2022, 152, 113224. [Google Scholar] [CrossRef]
  190. Sun, J.; Yang, F.; Wang, L.; Yu, H.; Yang, Z.; Wei, J.; Vasilev, K.; Zhang, X.; Liu, X.; Zhao, Y. Delivery of coenzyme Q10 loaded micelle targets mitochondrial ROS and enhances efficiency of mesenchymal stem cell therapy in intervertebral disc degeneration. Bioact. Mater. 2023, 23, 247–260, Erratum in Bioact. Mater. 2023, 24, 171–173. [Google Scholar] [CrossRef] [PubMed]
  191. Maruo, Y.; Shiraishi, M.; Hibino, M.; Abe, J.; Takeda, A.; Yamada, Y. Activation of Mitochondria in Mesenchymal Stem Cells by Mitochondrial Delivery of Coenzyme Q(10). Biol. Pharm. Bull. 2024, 47, 1415–1421. [Google Scholar] [CrossRef]
  192. Li, X.; Wang, X.; Zhang, C.; Wang, J.; Wang, S.; Hu, L. Dysfunction of metabolic activity of bone marrow mesenchymal stem cells in aged mice. Cell Prolif. 2022, 55, e13191. [Google Scholar] [CrossRef]
  193. Park, J.; Park, H.H.; Choi, H.; Kim, Y.S.; Yu, H.J.; Lee, K.Y.; Lee, Y.J.; Kim, S.H.; Koh, S.H. Coenzyme Q10 protects neural stem cells against hypoxia by enhancing survival signals. Brain Res. 2012, 1478, 64–73. [Google Scholar] [CrossRef]
  194. Choi, H.; Park, H.H.; Lee, K.Y.; Choi, N.Y.; Yu, H.J.; Lee, Y.J.; Park, J.; Huh, Y.M.; Lee, S.H.; Koh, S.H. Coenzyme Q10 restores amyloid beta-inhibited proliferation of neural stem cells by activating the PI3K pathway. Stem Cells Dev. 2013, 22, 2112–2120. [Google Scholar] [CrossRef]
  195. Lee, H.J.; Park, M.J.; Joo, B.S.; Joo, J.K.; Kim, Y.H.; Yang, S.W.; Kim, C.W.; Kim, K.H. Effects of coenzyme Q10 on ovarian surface epithelium-derived ovarian stem cells and ovarian function in a 4-vinylcyclohexene diepoxide-induced murine model of ovarian failure. Reprod. Biol. Endocrinol. 2021, 19, 59. [Google Scholar] [CrossRef]
  196. Velichkovska, M.; Surnar, B.; Nair, M.; Dhar, S.; Toborek, M. Targeted Mitochondrial COQ(10) Delivery Attenuates Antiretroviral-Drug-Induced Senescence of Neural Progenitor Cells. Mol. Pharm. 2019, 16, 724–736. [Google Scholar] [CrossRef]
  197. Liu, H.; Liu, S.; Jiang, J.; Zhang, Y.; Luo, Y.; Zhao, J.; Xu, J.; Xie, Y.; Liao, W.; Wang, W.; et al. CoQ10 enhances the efficacy of airway basal stem cell transplantation on bleomycin-induced idiopathic pulmonary fibrosis in mice. Respir. Res. 2022, 23, 39. [Google Scholar] [CrossRef] [PubMed]
  198. Hernández-Pérez, O.R.; Juárez-Navarro, K.J.; Diaz, N.F.; Padilla-Camberos, E.; Beltran-Garcia, M.J.; Cardenas-Castrejon, D.; Corona-Perez, H.; Hernández-Jiménez, C.; Díaz-Martínez, N.E. Biomolecules resveratrol + coenzyme Q10 recover the cell state of human mesenchymal stem cells after 1-methyl-4-phenylpyridinium-induced damage and improve proliferation and neural differentiation. Front. Neurosci. 2022, 16, 929590. [Google Scholar] [CrossRef] [PubMed]
  199. Zheng, Z.; Wang, X.; Ouyang, L.; Chen, W.; Zhang, L.; Cao, Y. Antioxidants Improve the Proliferation and Efficacy of hUC-MSCs against H2O2-Induced Senescence. Antioxidants 2023, 12, 1334. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic representation shows the most common symptoms among low-energy associated disorders. The figure highlights shared clinical features as well as those that are unique to each disorder. GWI, Gulf War Illness; FM, fibromyalgia; ME/CFS, myalgic encephalomyelitis/chronic fatigue syndrome; long COVID, post-COVID syndrome; PTSD, post-traumatic stress disorder.
Figure 1. Schematic representation shows the most common symptoms among low-energy associated disorders. The figure highlights shared clinical features as well as those that are unique to each disorder. GWI, Gulf War Illness; FM, fibromyalgia; ME/CFS, myalgic encephalomyelitis/chronic fatigue syndrome; long COVID, post-COVID syndrome; PTSD, post-traumatic stress disorder.
Ijms 26 09044 g001
Figure 2. Potential shared pathophysiological mechanisms underlying low-energy associated disorders. The schematic overview highlights how pathologies with different origins can converge on common pathophysiological mechanisms, illustrating a complex interplay that leads to overlapping symptoms observed in these disorders. GWI, Gulf War Illness; FM, fibromyalgia; ME/CFS, myalgic encephalomyelitis/chronic fatigue syndrome; long COVID, post-COVID syndrome. Created with BioRender (BioRender software version 04. https://app.biorender.com; accessed on 4 August 2025).
Figure 2. Potential shared pathophysiological mechanisms underlying low-energy associated disorders. The schematic overview highlights how pathologies with different origins can converge on common pathophysiological mechanisms, illustrating a complex interplay that leads to overlapping symptoms observed in these disorders. GWI, Gulf War Illness; FM, fibromyalgia; ME/CFS, myalgic encephalomyelitis/chronic fatigue syndrome; long COVID, post-COVID syndrome. Created with BioRender (BioRender software version 04. https://app.biorender.com; accessed on 4 August 2025).
Ijms 26 09044 g002
Table 1. Summary of studies supplementing Coenzyme Q10 in stem cell models.
Table 1. Summary of studies supplementing Coenzyme Q10 in stem cell models.
Study Refs.ModelsOutcomes
Park et al. (2012) [193] Neural stem cells (rat)Improved cell viability and intracellular signaling proteins during hypoxia-reperfusion
Choi et al. (2013) [194] Neural stem cells (mouse)CoQ10 restored amyloid beta-inhibited proliferation by activating the PI3K pathway
Zhang et al. (2015) [184] Mesenchymal stem cells (rat)Inhibition of oxidative stress and cell aging induced by D-galactose
Lee et al. (2021) [195] Ovarian stem cells (mouse)Improved stem cell function in vinylcyclohexene-diepoxide induced model of ovarian failure
Velichkovska et al. (2019) [196] Neural progenitor cells (mouse)Mitochondrial dysfunction induced by anti-retroviral drugs (tenofovir and emtricitabine) improved
Liu et al. (2022) [197] Airway basal stem cells
(human)
Reduced oxidative stress induced by hydrogen peroxide; improved efficiency of transplanted cells in bleomycin-induced model of pulmonary fibrosis
Sun et al. (2023) [190] Mesenchymal stem cells (rat)Improved efficiency of transplanted cells in rat model of intervertebral disk degeneration
Hernández-Perez et al. (2022) [198] Mesenchymal stem cells
(human)
Reduced oxidative stress, improved cell viability and proliferation following exposure to MPP+
Zheng et al. (2023) [199] Umbilical cord mesenchymal stem cells (human)Reduced oxidative stress induced by hydrogen peroxide, cell senescence reduced and proliferation capacity improved
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Mantle, D.; Domingo, J.C.; Golomb, B.A.; Castro-Marrero, J. Gulf War Illness, Fibromyalgia, Myalgic Encephalomyelitis/Chronic Fatigue Syndrome and Long COVID Overlap in Common Symptoms and Underlying Biological Mechanisms: Implications for Future Therapeutic Strategies. Int. J. Mol. Sci. 2025, 26, 9044. https://doi.org/10.3390/ijms26189044

AMA Style

Mantle D, Domingo JC, Golomb BA, Castro-Marrero J. Gulf War Illness, Fibromyalgia, Myalgic Encephalomyelitis/Chronic Fatigue Syndrome and Long COVID Overlap in Common Symptoms and Underlying Biological Mechanisms: Implications for Future Therapeutic Strategies. International Journal of Molecular Sciences. 2025; 26(18):9044. https://doi.org/10.3390/ijms26189044

Chicago/Turabian Style

Mantle, David, Joan Carles Domingo, Beatrice Alexandra Golomb, and Jesús Castro-Marrero. 2025. "Gulf War Illness, Fibromyalgia, Myalgic Encephalomyelitis/Chronic Fatigue Syndrome and Long COVID Overlap in Common Symptoms and Underlying Biological Mechanisms: Implications for Future Therapeutic Strategies" International Journal of Molecular Sciences 26, no. 18: 9044. https://doi.org/10.3390/ijms26189044

APA Style

Mantle, D., Domingo, J. C., Golomb, B. A., & Castro-Marrero, J. (2025). Gulf War Illness, Fibromyalgia, Myalgic Encephalomyelitis/Chronic Fatigue Syndrome and Long COVID Overlap in Common Symptoms and Underlying Biological Mechanisms: Implications for Future Therapeutic Strategies. International Journal of Molecular Sciences, 26(18), 9044. https://doi.org/10.3390/ijms26189044

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop