1
|
Ludwig B, Hauer L, Böck M, Schillerwein-Kral C, Weyer L, Moser D, Zehetmayer S, Trimmel K, Seidel S. Assessing fatigue in myalgic encephalomyelitis/chronic fatigue syndrome patients before and after treatment with bright light therapy: A prospective randomized controlled crossover study. Sleep Med 2025; 129:369-374. [PMID: 40120538 DOI: 10.1016/j.sleep.2025.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/31/2025] [Accepted: 03/02/2025] [Indexed: 03/25/2025]
Abstract
OBJECTIVE The aim of the current study was to test the effectiveness of treatment with bright light therapy (BLT) on fatigue and cognitive function in patients with Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS). A randomized-controlled cross-over study design was chosen in order to provide all patients access to BLT treatment and account for placebo effects. METHODS In this study, a total of 36 outpatients with a diagnosis of ME/CFS according to the criteria of the Institute of Medicine (2015) were randomly assigned to a cross-over design starting out either with BLT or waitlist for the course of 2 weeks with a washout phase in between. Portable light boxes emitting full-spectrum visible light with a luminance intensity of 10,000 lux were used by the participants at home. Primary outcome of the study was fatigue as assessed by Chalder Fatigue Score (CFQ) and the secondary outcome variable was cognitive function assessed per standardized test battery (Test of Attentional Performance - TAP). RESULTS The primary outcome variable fatigue was not significantly improved after treatment with BLT compared to wait list in the full crossover design, although fatigue scores improved immediately after two weeks of BLT. Additionally, patients showed decreased reaction time after treatment with BLT in a subtest of TAP compared to wait list. Over 45 % of patients were diagnosed with postural tachycardia syndrome. CONCLUSION BLT for two weeks is not effective for the treatment of fatigue in ME/CFS, but it might have beneficial effects on attention in patients with ME/CFS. The clinical trial is registered with www. CLINICALTRIALS gov (NCT06635928).
Collapse
Affiliation(s)
- Birgit Ludwig
- Department of Neurology, Medical University Vienna, Vienna, Austria; Comprehensive Center for Clinical Neurosciences & Mental Health, Medical University of Vienna, Vienna, Austria
| | - Lea Hauer
- Department of Neurology, Medical University Vienna, Vienna, Austria; Comprehensive Center for Clinical Neurosciences & Mental Health, Medical University of Vienna, Vienna, Austria
| | - Marion Böck
- Department of Neurology, Medical University Vienna, Vienna, Austria; Comprehensive Center for Clinical Neurosciences & Mental Health, Medical University of Vienna, Vienna, Austria
| | - Cornelia Schillerwein-Kral
- Department of Neurology, Medical University Vienna, Vienna, Austria; Comprehensive Center for Clinical Neurosciences & Mental Health, Medical University of Vienna, Vienna, Austria
| | - Lena Weyer
- Center for Medical Data Science, Institute of Medical Statistics, Medical University Vienna, Vienna, Austria
| | - Doris Moser
- Department of Neurology, Medical University Vienna, Vienna, Austria; Comprehensive Center for Clinical Neurosciences & Mental Health, Medical University of Vienna, Vienna, Austria
| | - Sonja Zehetmayer
- Center for Medical Data Science, Institute of Medical Statistics, Medical University Vienna, Vienna, Austria
| | - Karin Trimmel
- Department of Neurology, Medical University Vienna, Vienna, Austria; Comprehensive Center for Clinical Neurosciences & Mental Health, Medical University of Vienna, Vienna, Austria
| | - Stefan Seidel
- Department of Neurology, Medical University Vienna, Vienna, Austria; Comprehensive Center for Clinical Neurosciences & Mental Health, Medical University of Vienna, Vienna, Austria; Rehabilitation Clinic Pirawarth, Bad Pirawarth, Austria.
| |
Collapse
|
2
|
Todhunter-Brown A, Campbell P, Broderick C, Cowie J, Davis B, Fenton C, Markham S, Sellers C, Thomson K. Recent research in myalgic encephalomyelitis/chronic fatigue syndrome: an evidence map. Health Technol Assess 2025:1-78. [PMID: 40162526 PMCID: PMC11973615 DOI: 10.3310/btbd8846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025] Open
Abstract
Background Myalgic encephalomyelitis/chronic fatigue syndrome is a chronic condition, classified by the World Health Organization as a nervous system disease, impacting around 17 million people worldwide. Presentation involves persistent fatigue and postexertional malaise (a worsening of symptoms after minimal exertion) and a wide range of other symptoms. Case definitions have historically varied; postexertional malaise is a core diagnostic criterion in current definitions. In 2022, a James Lind Alliance Priority Setting Partnership established research priorities relating to myalgic encephalomyelitis/chronic fatigue syndrome. Objective(s) We created a map of myalgic encephalomyelitis/chronic fatigue syndrome evidence (2018-23), showing the volume and key characteristics of recent research in this field. We considered diagnostic criteria and how current research maps against the James Lind Alliance Priority Setting Partnership research priorities. Methods Using a predefined protocol, we conducted a comprehensive search of Cochrane, MEDLINE, EMBASE and Cumulative Index to Nursing and Allied Health Literature. We included all English-language research studies published between January 2018 and May 2023. Two reviewers independently applied inclusion criteria with consensus involving additional reviewers. Studies including people diagnosed with myalgic encephalomyelitis/chronic fatigue syndrome using any criteria (including self-report), of any age and in any setting were eligible. Studies with < 10 myalgic encephalomyelitis/chronic fatigue syndrome participants were excluded. Data extraction, coding of topics (involving stakeholder consultation) and methodological quality assessment of systematic reviews (using A MeaSurement Tool to Assess systematic Reviews 2) was conducted independently by two reviewers, with disagreements resolved by a third reviewer. Studies were presented in an evidence map. Results Of the 11,278 identified studies, 742 met the selection criteria, but only 639 provided sufficient data for inclusion in the evidence map. These reported data from approximately 610,000 people with myalgic encephalomyelitis/chronic fatigue syndrome. There were 81 systematic reviews, 72 experimental studies, 423 observational studies and 63 studies with other designs. Most studies (94%) were from high-income countries. Reporting of participant details was poor; 16% did not report gender, 74% did not report ethnicity and 81% did not report the severity of myalgic encephalomyelitis/chronic fatigue syndrome. Forty-four per cent of studies used multiple diagnostic criteria, 16% did not specify criteria, 24% used a single criterion not requiring postexertional malaise and 10% used a single criterion requiring postexertional malaise. Most (89%) systematic reviews had a low methodological quality. Five main topics (37 subtopics) were included in the evidence map. Of the 639 studies; 53% addressed the topic 'what is the cause?'; 38% 'what is the problem?'; 26% 'what can we do about it?'; 15% 'diagnosis and assessment'; and 13% other topics, including 'living with myalgic encephalomyelitis/chronic fatigue syndrome'. Discussion Studies have been presented in an interactive evidence map according to topic, study design, diagnostic criteria and age. This evidence map should inform decisions about future myalgic encephalomyelitis/chronic fatigue syndrome research. Limitations An evidence map does not summarise what the evidence says. Our evidence map only includes studies published in 2018 or later and in English language. Inconsistent reporting and use of diagnostic criteria limit the interpretation of evidence. We assessed the methodological quality of systematic reviews, but not of primary studies. Conclusions We have produced an interactive evidence map, summarising myalgic encephalomyelitis/chronic fatigue syndrome research from 2018 to 2023. This evidence map can inform strategic plans for future research. We found some, often limited, evidence addressing every James Lind Alliance Priority Setting Partnership priority; high-quality systematic reviews should inform future studies. Funding This article presents independent research funded by the National Institute for Health and Care Research (NIHR) Evidence Synthesis programme as award number NIHR159926.
Collapse
Affiliation(s)
| | | | | | - Julie Cowie
- NESSIE, Glasgow Caledonian University, Glasgow, UK
| | | | - Candida Fenton
- NESSIE, Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Sarah Markham
- NESSIE Patient and public involvement member, UK
- Department of Biostatistics and Health Informatics, King's College London, London, UK
| | - Ceri Sellers
- NESSIE, Glasgow Caledonian University, Glasgow, UK
| | | |
Collapse
|
3
|
Van Campenhout J, Buntinx Y, Xiong HY, Wyns A, Polli A, Nijs J, Aerts JL, Laeremans T, Hendrix J. Unravelling the Connection Between Energy Metabolism and Immune Senescence/Exhaustion in Patients with Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. Biomolecules 2025; 15:357. [PMID: 40149893 PMCID: PMC11940106 DOI: 10.3390/biom15030357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 02/21/2025] [Accepted: 02/26/2025] [Indexed: 03/29/2025] Open
Abstract
Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) is a debilitating disease, characterized by a diverse array of symptoms including post-exertional malaise (PEM), severe fatigue, and cognitive impairments, all of which drastically diminish the patients' quality of life. Despite its impact, no curative treatments exist, largely due to the limited understanding of the disease's underlying pathophysiology. Mitochondrial dysfunction, leading to impaired energy production and utilization, is believed to play a key role in the onset of fatigue and PEM, positioning it as a potential key pathophysiological mechanism underlying ME/CFS. Additionally, the disorder shows similarities to chronic viral infections, with frequent reports of immune system alterations, suggesting a critical role for immune (dys)functioning. In particular, the roles of immune senescence and immune exhaustion-two fundamental immune states-remain poorly understood in ME/CFS. This state-of-the-art review explores how metabolic dysfunction and immune dysfunction may be interconnected in ME/CFS, proposing that energy deficits may directly impair immune function. By examining this metabolic-immune interplay, this review highlights potential pathways for developing innovative therapeutic strategies that target both energy metabolism and immune regulation, offering hope for improving patient outcomes.
Collapse
Affiliation(s)
- Jente Van Campenhout
- Pain in Motion Research Group (PAIN), Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education and Physiotherapy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium; (J.V.C.); (Y.B.); (H.-Y.X.); (A.W.); (J.N.); (J.H.)
| | - Yanthe Buntinx
- Pain in Motion Research Group (PAIN), Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education and Physiotherapy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium; (J.V.C.); (Y.B.); (H.-Y.X.); (A.W.); (J.N.); (J.H.)
| | - Huan-Yu Xiong
- Pain in Motion Research Group (PAIN), Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education and Physiotherapy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium; (J.V.C.); (Y.B.); (H.-Y.X.); (A.W.); (J.N.); (J.H.)
- Flanders Research Foundation-FWO, 1090 Brussels, Belgium
| | - Arne Wyns
- Pain in Motion Research Group (PAIN), Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education and Physiotherapy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium; (J.V.C.); (Y.B.); (H.-Y.X.); (A.W.); (J.N.); (J.H.)
- Department of Public Health and Primary Care, Centre for Environment & Health, KU Leuven, Kapucijnenvoer 35, 3000 Leuven, Belgium
| | - Andrea Polli
- Pain in Motion Research Group (PAIN), Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education and Physiotherapy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium; (J.V.C.); (Y.B.); (H.-Y.X.); (A.W.); (J.N.); (J.H.)
- Flanders Research Foundation-FWO, 1090 Brussels, Belgium
- Department of Public Health and Primary Care, Centre for Environment & Health, KU Leuven, Kapucijnenvoer 35, 3000 Leuven, Belgium
| | - Jo Nijs
- Pain in Motion Research Group (PAIN), Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education and Physiotherapy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium; (J.V.C.); (Y.B.); (H.-Y.X.); (A.W.); (J.N.); (J.H.)
- Unit of Physiotherapy, Department of Health and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, SE-405 30 Gothenburg, Sweden
- Department of Physical Medicine and Physiotherapy, University Hospital Brussels, 1090 Brussels, Belgium
| | - Joeri L. Aerts
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium; (J.L.A.); (T.L.)
| | - Thessa Laeremans
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium; (J.L.A.); (T.L.)
| | - Jolien Hendrix
- Pain in Motion Research Group (PAIN), Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education and Physiotherapy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium; (J.V.C.); (Y.B.); (H.-Y.X.); (A.W.); (J.N.); (J.H.)
- Flanders Research Foundation-FWO, 1090 Brussels, Belgium
- Department of Public Health and Primary Care, Centre for Environment & Health, KU Leuven, Kapucijnenvoer 35, 3000 Leuven, Belgium
| |
Collapse
|
4
|
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. [PMID: 39435656 PMCID: PMC11529985 DOI: 10.1172/jci.insight.183810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/28/2024] [Indexed: 10/23/2024] Open
Abstract
Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) and long COVID are debilitating multisystemic conditions sharing similarities in immune dysregulation and cellular signaling pathways contributing to the pathophysiology. In this study, immune exhaustion gene expression was investigated in participants with ME/CFS or long COVID concurrently. RNA was extracted from peripheral blood mononuclear cells isolated from participants with ME/CFS (n = 14), participants with long COVID (n = 15), and healthy controls (n = 18). Participants with ME/CFS were included according to Canadian Consensus Criteria. Participants with long COVID were eligible according to the case definition for "Post COVID-19 Condition" published by the World Health Organization. RNA was analyzed using the NanoString nCounter Immune Exhaustion gene expression panel. Differential gene expression analysis in ME/CFS revealed downregulated IFN signaling and immunoglobulin genes, and this suggested a state of immune suppression. Pathway analysis implicated dysregulated macrophage activation, cytokine production, and immunodeficiency signaling. Long COVID samples exhibited dysregulated expression of genes regarding antigen presentation, cytokine signaling, and immune activation. Differentially expressed genes were associated with antigen presentation, B cell development, macrophage activation, and cytokine signaling. This investigation elucidates the intricate role of both adaptive and innate immune dysregulation underlying ME/CFS and long COVID, emphasizing the potential importance of immune exhaustion in disease progression.
Collapse
Affiliation(s)
- Natalie Eaton-Fitch
- National Centre for Neuroimmunology and Emerging Diseases
- Consortium Health International for Myalgic Encephalomyelitis, and
| | - Penny Rudd
- Institute for Biomedicine and Glycomics, Griffith University, Gold Coast, Australia
| | - Teagan Er
- School of Human Sciences (Physiology), The University of Western Australia, Perth, Australia
| | - Livia Hool
- School of Human Sciences (Physiology), The University of Western Australia, Perth, Australia
- Victor Chang Cardiac Institute, Australia
| | - Lara Herrero
- Institute for Biomedicine and Glycomics, Griffith University, Gold Coast, Australia
| | - Sonya Marshall-Gradisnik
- National Centre for Neuroimmunology and Emerging Diseases
- Consortium Health International for Myalgic Encephalomyelitis, and
| |
Collapse
|
5
|
Arron HE, Marsh BD, Kell DB, Khan MA, Jaeger BR, Pretorius E. Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: the biology of a neglected disease. Front Immunol 2024; 15:1386607. [PMID: 38887284 PMCID: PMC11180809 DOI: 10.3389/fimmu.2024.1386607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 04/11/2024] [Indexed: 06/20/2024] Open
Abstract
Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) is a chronic, debilitating disease characterised by a wide range of symptoms that severely impact all aspects of life. Despite its significant prevalence, ME/CFS remains one of the most understudied and misunderstood conditions in modern medicine. ME/CFS lacks standardised diagnostic criteria owing to variations in both inclusion and exclusion criteria across different diagnostic guidelines, and furthermore, there are currently no effective treatments available. Moving beyond the traditional fragmented perspectives that have limited our understanding and management of the disease, our analysis of current information on ME/CFS represents a significant paradigm shift by synthesising the disease's multifactorial origins into a cohesive model. We discuss how ME/CFS emerges from an intricate web of genetic vulnerabilities and environmental triggers, notably viral infections, leading to a complex series of pathological responses including immune dysregulation, chronic inflammation, gut dysbiosis, and metabolic disturbances. This comprehensive model not only advances our understanding of ME/CFS's pathophysiology but also opens new avenues for research and potential therapeutic strategies. By integrating these disparate elements, our work emphasises the necessity of a holistic approach to diagnosing, researching, and treating ME/CFS, urging the scientific community to reconsider the disease's complexity and the multifaceted approach required for its study and management.
Collapse
Affiliation(s)
- Hayley E. Arron
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| | - Benjamin D. Marsh
- MRCPCH Consultant Paediatric Neurodisability, Exeter, Devon, United Kingdom
| | - Douglas B. Kell
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
- The Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| | - M. Asad Khan
- Directorate of Respiratory Medicine, Manchester University Hospitals, Wythenshawe Hospital, Manchester, United Kingdom
| | - Beate R. Jaeger
- Long COVID department, Clinic St Georg, Bad Aibling, Germany
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
6
|
Domingo JC, Battistini F, Cordobilla B, Zaragozá MC, Sanmartin-Sentañes 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. [PMID: 38600563 PMCID: PMC11005215 DOI: 10.1186/s12967-024-05148-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 03/30/2024] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND Accumulating evidence suggests that autonomic dysfunction and persistent systemic inflammation are common clinical features in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) and long COVID. However, there is limited knowledge regarding their potential association with circulating biomarkers and illness severity in these conditions. METHODS This single-site, prospective, cross-sectional, pilot cohort study aimed to distinguish between the two patient populations by using self-reported outcome measures and circulating biomarkers of endothelial function and systemic inflammation status. Thirty-one individuals with ME/CFS, 23 individuals with long COVID, and 31 matched sedentary healthy controls were included. All study participants underwent non-invasive cardiovascular hemodynamic challenge testing (10 min NASA lean test) for assessment of orthostatic intolerance. Regression analysis was used to examine associations between outcome measures and circulating biomarkers in the study participants. Classification across groups was based on principal component and discriminant analyses. RESULTS Four ME/CFS patients (13%), 1 with long COVID (4%), and 1 healthy control (3%) presented postural orthostatic tachycardia syndrome (POTS) using the 10-min NASA lean test. Compared with matched healthy controls, ME/CFS and long COVID subjects showed higher levels of ET-1 (p < 0.05) and VCAM-1 (p < 0.001), and lower levels of nitrites (NOx assessed as NO2- + NO3-) (p < 0.01). ME/CFS patients also showed higher levels of serpin E1 (PAI-1) and E-selectin than did both long COVID and matched control subjects (p < 0.01 in all cases). Long COVID patients had lower TSP-1 levels than did ME/CFS patients and matched sedentary healthy controls (p < 0.001). As for inflammation biomarkers, both long COVID and ME/CFS subjects had higher levels of TNF-α than did matched healthy controls (p < 0.01 in both comparisons). Compared with controls, ME/CFS patients had higher levels of IL-1β (p < 0.001), IL-4 (p < 0.001), IL-6 (p < 0.01), IL-10 (p < 0.001), IP-10 (p < 0.05), and leptin (p < 0.001). Principal component analysis supported differentiation between groups based on self-reported outcome measures and biomarkers of endothelial function and inflammatory status in the study population. CONCLUSIONS Our findings revealed that combining biomarkers of endothelial dysfunction and inflammation with outcome measures differentiate ME/CFS and Long COVID using robust discriminant analysis of principal components. Further research is needed to provide a more comprehensive characterization of these underlying pathomechanisms, which could be promising targets for therapeutic and preventive strategies in these conditions.
Collapse
Affiliation(s)
- Joan Carles Domingo
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, 08028, Spain
| | - Federica Battistini
- Molecular Modelling and Bioinformatics Group, Institute for Research in Biomedicine, Barcelona Institute of Science and Technology, Barcelona, 08028, Spain
| | - Begoña Cordobilla
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, 08028, Spain
| | | | - Ramón Sanmartin-Sentañes
- Division of Rheumatology, Clinical Unit in ME/CFS and Long COVID, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, 08035, Spain
- Division of Rheumatology, Research Unit in ME/CFS and Long COVID, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, 08035, Spain
| | - Jose Alegre-Martin
- Division of Rheumatology, Clinical Unit in ME/CFS and Long COVID, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, 08035, Spain
- Division of Rheumatology, Research Unit in ME/CFS and Long COVID, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, 08035, Spain
| | - Trinitat Cambras
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, 08028, Spain.
| | - Jesus Castro-Marrero
- Division of Rheumatology, Research Unit in ME/CFS and Long COVID, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, 08035, Spain.
| |
Collapse
|
7
|
Jahanbani F, Sing JC, Maynard RD, Jahanbani S, Dafoe J, Dafoe W, Jones N, Wallace KJ, Rastan A, Maecker HT, Röst HL, Snyder MP, Davis RW. Longitudinal cytokine and multi-modal health data of an extremely severe ME/CFS patient with HSD reveals insights into immunopathology, and disease severity. Front Immunol 2024; 15:1369295. [PMID: 38650940 PMCID: PMC11033372 DOI: 10.3389/fimmu.2024.1369295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 03/18/2024] [Indexed: 04/25/2024] Open
Abstract
Introduction Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) presents substantial challenges in patient care due to its intricate multisystem nature, comorbidities, and global prevalence. The heterogeneity among patient populations, coupled with the absence of FDA-approved diagnostics and therapeutics, further complicates research into disease etiology and patient managment. Integrating longitudinal multi-omics data with clinical, health,textual, pharmaceutical, and nutraceutical data offers a promising avenue to address these complexities, aiding in the identification of underlying causes and providing insights into effective therapeutics and diagnostic strategies. Methods This study focused on an exceptionally severe ME/CFS patient with hypermobility spectrum disorder (HSD) during a period of marginal symptom improvements. Longitudinal cytokine profiling was conducted alongside the collection of extensive multi-modal health data to explore the dynamic nature of symptoms, severity, triggers, and modifying factors. Additionally, an updated severity assessment platform and two applications, ME-CFSTrackerApp and LexiTime, were introduced to facilitate real-time symptom tracking and enhance patient-physician/researcher communication, and evaluate response to medical intervention. Results Longitudinal cytokine profiling revealed the significance of Th2-type cytokines and highlighted synergistic activities between mast cells and eosinophils, skewing Th1 toward Th2 immune responses in ME/CFS pathogenesis, particularly in cognitive impairment and sensorial intolerance. This suggests a potentially shared underlying mechanism with major ME/CFS comorbidities such as HSD, Mast cell activation syndrome, postural orthostatic tachycardia syndrome (POTS), and small fiber neuropathy. Additionally, the data identified potential roles of BCL6 and TP53 pathways in ME/CFS etiology and emphasized the importance of investigating adverse reactions to medication and supplements and drug interactions in ME/CFS severity and progression. Discussion Our study advocates for the integration of longitudinal multi-omics with multi-modal health data and artificial intelligence (AI) techniques to better understand ME/CFS and its major comorbidities. These findings highlight the significance of dysregulated Th2-type cytokines in patient stratification and precision medicine strategies. Additionally, our results suggest exploring the use of low-dose drugs with partial agonist activity as a potential avenue for ME/CFS treatment. This comprehensive approach emphasizes the importance of adopting a patient-centered care approach to improve ME/CFS healthcare management, disease severity assessment, and personalized medicine. Overall, these findings contribute to our understanding of ME/CFS and offer avenues for future research and clinical practice.
Collapse
Affiliation(s)
- Fereshteh Jahanbani
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, United States
| | - Justin Cyril Sing
- Department of Molecular Genetics, Donnelly Center, University of Toronto, Toronto, ON, Canada
| | - Rajan Douglas Maynard
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, United States
| | - Shaghayegh Jahanbani
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Veterans Affairs (VA) Palo Alto Health Care System, Palo Alto, CA, United States
| | - Janet Dafoe
- ME/CFS Collaborative Research Center at Stanford, Stanford Genome Technology Center, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Whitney Dafoe
- ME/CFS Collaborative Research Center at Stanford, Stanford Genome Technology Center, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Nathan Jones
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, United States
| | - Kelvin J. Wallace
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, United States
| | - Azuravesta Rastan
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, United States
| | - Holden T. Maecker
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Pulmonary and Critical Care Medicine, Institute of Immunity, Transplantation, and Infectious Diseases, Stanford University, Palo Alto, CA, United States
| | - Hannes L. Röst
- Department of Molecular Genetics, Donnelly Center, University of Toronto, Toronto, ON, Canada
| | - Michael P. Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, United States
| | - Ronald W. Davis
- ME/CFS Collaborative Research Center at Stanford, Stanford Genome Technology Center, Stanford University School of Medicine, Palo Alto, CA, United States
| |
Collapse
|
8
|
Raij T, Raij K. Association between fatigue, peripheral serotonin, and L-carnitine in hypothyroidism and in chronic fatigue syndrome. Front Endocrinol (Lausanne) 2024; 15:1358404. [PMID: 38505756 PMCID: PMC10948554 DOI: 10.3389/fendo.2024.1358404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 02/15/2024] [Indexed: 03/21/2024] Open
Abstract
Background Fatigue of unknown origin is a hallmark symptom in chronic fatigue syndrome (CFS) and is also found in 20% of hypothyroidism patients despite appropriate levothyroxine treatment. Here, we suggest that in these disorders, peripheral serotonin levels are low, and elevating them to normal range with L-carnitine is accompanied with reduced fatigue. Methods We conducted a retrospective analysis of follow-up clinical data (CFS N=12; hypothyroidism with fatigue N=40) where serum serotonin and fatigue levels were compared before vs. after 7 weeks of oral L-carnitine supplementation. Results After L-carnitine, serotonin increased (8-fold in CFS, Sig. = 0.002, 6-fold in hypothyroidism, Sig. < 0.001) whereas fatigue decreased (2-fold in both CFS and hypothyroidism, Sig. = 0.002 for CFS, Sig. < 0.001 for hypothyroidism). There was a negative correlation between serotonin level and fatigue (for CFS, rho = -0.49 before and -0.67 after L-carnitine; for hypothyroidism, rho = -0.24 before and -0.83 after L-carnitine). Conclusions These findings suggest a new link between low peripheral serotonin, L-carnitine, and fatigue.
Collapse
Affiliation(s)
- Tommi Raij
- Department of Physical Medicine and Rehabilitation, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Department Of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Chicago, IL, United States
- MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, MGH Department of Radiology, Boston, MA, United States
| | - Kari Raij
- Kruunuhaka Medical Center, Helsinki, Finland
| |
Collapse
|
9
|
Gramont B, Goutte J, Féasson L, Millet G, Hupin D, Cathébras P. [Chronic fatigue: What investigations? And what for?]. Rev Med Interne 2023; 44:662-669. [PMID: 37248110 DOI: 10.1016/j.revmed.2023.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 05/15/2023] [Indexed: 05/31/2023]
Abstract
Chronic fatigue is a frequent complaint, expressed at all levels of the healthcare system. It is perceived as disabling in a high proportion of cases, and internists are frequently called upon to find "the" cause. The etiological diagnostic approach of an unexplained state of fatigue relies on the careful search for more specific clues by questioning and clinical examination. It is necessary to recognize the limited place of complementary examinations apart from the basic biological parameters. Simple rating scales can be useful in the etiological and differential diagnosis of fatigue. Chronic fatigue syndrome (CFS), in the current state of knowledge, cannot be considered as a specific pathological entity distinct from idiopathic chronic fatigue states, and does not have validated biomarkers. It is important to know that a state of chronic asthenia often results from several intricated etiological factors (biological, psychological and social), to be classified as predisposing, precipitating and perpetuating. The metabolic and cardiorespiratory exercise test has a major place in the assessment and management of fatigue, as a prerequisite for personalized retraining or adapted physical activity (APA), which are the treatments of choice for chronic fatigue.
Collapse
Affiliation(s)
- B Gramont
- Service de médecine interne, CHU de Saint-Étienne, hôpital Nord, Saint-Étienne, France.
| | - J Goutte
- Service de médecine interne, CHU de Saint-Étienne, hôpital Nord, Saint-Étienne, France
| | - L Féasson
- Service de physiologie clinique et de l'exercice, IRMIS, CHU de Saint-Étienne, université Jean-Monnet, Saint-Étienne, France
| | - G Millet
- STAPS, IRMIS, université Jean-Monnet, Saint-Étienne, France
| | - D Hupin
- Service de physiologie clinique et de l'exercice, IRMIS, CHU de Saint-Étienne, université Jean-Monnet, Saint-Étienne, France
| | - P Cathébras
- Service de médecine interne, CHU de Saint-Étienne, hôpital Nord, Saint-Étienne, France
| |
Collapse
|
10
|
Banovic I, Scrima F, Fornasieri I, Beaugerie L, Coquart J, Fourgon C, Iodice P, Nion-Larmurier I, Savoye G, Sorin AL, Tourny C, Augustinova M. Psychometric validation of the French Multidimensional Chronic Asthenia Scale (MCAS) in a sample of 621 patients with chronic fatigue. BMC Psychol 2023; 11:324. [PMID: 37817287 PMCID: PMC10566142 DOI: 10.1186/s40359-023-01358-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 09/28/2023] [Indexed: 10/12/2023] Open
Abstract
BACKGROUND Psychometric validation of the Multidimensional Chronic Asthenia Scale (MCAS) was conducted in order to provide an effective tool for assessing the health-related quality of life of French-speaking patients with chronic asthenia (CA). METHODS Items resulting from the initial formulation of the self-reported MCAS (along with other materials) were completed by French-speaking volunteers with inactive or active inflammatory bowel disease (IBD-I vs. IBD-A) or chronic fatigue syndrome (CFS). Responses from 621 participants (180 patients with IBD-A, 172 with IBD-I, 269 with CFS) collected in a single online survey were divided into three subsamples to test the construct validity of the MCAS (Step 1, N = 240), to confirm its factorial structure (Step 2, N = 204) and to explore its convergent-discriminant validity with the Fatigue Symptoms Inventory (FSI) and revised Piper Fatigue Scale (r-PFS, Step 3, N = 177). RESULTS Steps 1 and 2 showed that, as expected, MCAS has four dimensions: feeling of constraint (FoC), physical (PC), life (LC) and interpersonal consequences (IC), which are also related to the duration of CA (i.e., the longer it lasts, the more the dimensions are impacted). The results further showed that the MCAS is sensitive enough to capture between-group differences, with the CFS group being the most impaired, followed by IBD-A and IBD-I. While convergent-discriminant validity between the 4 factors of MCAS and FSI and r-PFS, respectively, was satisfactory overall, Step 3 also pointed to some limitations that call for future research (e.g., shared variances between the PC and IC dimensions of MCAS and behavioral dimension of r-PFS). CONCLUSION Despite these limitations, the MCAS clearly constitutes a promising tool for measuring quantitative differences (i.e., severity/intensity) in CA associated with various diseases, but also, and importantly, the clinically important differences in domains of its expression (i.e., qualitative differences).
Collapse
Affiliation(s)
- Ingrid Banovic
- CRFDP Lab., University of Rouen Normandy, 76821, Mont-Saint-Aignan Cedex, France.
| | - Fabrizio Scrima
- CRFDP Lab., University of Rouen Normandy, 76821, Mont-Saint-Aignan Cedex, France
| | | | - Laurent Beaugerie
- Department of Gastroenterology, Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie Et de Santé Publique, AP-HP, Hôpital Saint-Antoine, 75012, Paris, France
| | - Jérémy Coquart
- CETAPS Lab., University of Rouen Normandy, Mont-Saint-Aignan, France
- Univ. Lille, Univ. Artois, Univ. Littoral Côte d'Opale, ULR - URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, 7369, Lille, France
| | - Chloé Fourgon
- CRFDP Lab., University of Rouen Normandy, 76821, Mont-Saint-Aignan Cedex, France
| | - Pierpaolo Iodice
- CETAPS Lab., University of Rouen Normandy, Mont-Saint-Aignan, France
| | - Isabelle Nion-Larmurier
- Department of Gastroenterology, Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie Et de Santé Publique, AP-HP, Hôpital Saint-Antoine, 75012, Paris, France
| | - Guillaume Savoye
- UMR 10173, Université de Rouen Normandie, Centre Hospitalier Universitaire Charles Nicolle, 76000, Rouen, France
| | - Anne-Laure Sorin
- CRFDP Lab., University of Rouen Normandy, 76821, Mont-Saint-Aignan Cedex, France
| | - Claire Tourny
- CETAPS Lab., University of Rouen Normandy, Mont-Saint-Aignan, France
| | - Maria Augustinova
- CRFDP Lab., University of Rouen Normandy, 76821, Mont-Saint-Aignan Cedex, France
| |
Collapse
|
11
|
Komaroff AL, Lipkin WI. ME/CFS and Long COVID share similar symptoms and biological abnormalities: road map to the literature. Front Med (Lausanne) 2023; 10:1187163. [PMID: 37342500 PMCID: PMC10278546 DOI: 10.3389/fmed.2023.1187163] [Citation(s) in RCA: 127] [Impact Index Per Article: 63.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 05/09/2023] [Indexed: 06/23/2023] Open
Abstract
Some patients remain unwell for months after "recovering" from acute COVID-19. They develop persistent fatigue, cognitive problems, headaches, disrupted sleep, myalgias and arthralgias, post-exertional malaise, orthostatic intolerance and other symptoms that greatly interfere with their ability to function and that can leave some people housebound and disabled. The illness (Long COVID) is similar to myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) as well as to persisting illnesses that can follow a wide variety of other infectious agents and following major traumatic injury. Together, these illnesses are projected to cost the U.S. trillions of dollars. In this review, we first compare the symptoms of ME/CFS and Long COVID, noting the considerable similarities and the few differences. We then compare in extensive detail the underlying pathophysiology of these two conditions, focusing on abnormalities of the central and autonomic nervous system, lungs, heart, vasculature, immune system, gut microbiome, energy metabolism and redox balance. This comparison highlights how strong the evidence is for each abnormality, in each illness, and helps to set priorities for future investigation. The review provides a current road map to the extensive literature on the underlying biology of both illnesses.
Collapse
Affiliation(s)
- Anthony L. Komaroff
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - W. Ian Lipkin
- Center for Infection and Immunity, Mailman School of Public Health, Vagelos College of Physicians and Surgeons of Columbia University, New York, NY, United States
| |
Collapse
|
12
|
Vassiliou AG, Vrettou CS, Keskinidou C, Dimopoulou I, Kotanidou A, Orfanos SE. Endotheliopathy in Acute COVID-19 and Long COVID. Int J Mol Sci 2023; 24:8237. [PMID: 37175942 PMCID: PMC10179170 DOI: 10.3390/ijms24098237] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 04/28/2023] [Accepted: 04/30/2023] [Indexed: 05/15/2023] Open
Abstract
The pulmonary endothelium is a highly regulated organ that performs a wide range of functions under physiological and pathological conditions. Since endothelial dysfunction has been demonstrated to play a direct role in sepsis and acute respiratory distress syndrome, its role in COVID-19 has also been extensively investigated. Indeed, apart from the COVID-19-associated coagulopathy biomarkers, new biomarkers were recognised early during the pandemic, including markers of endothelial cell activation or injury. We systematically searched the literature up to 10 March 2023 for studies examining the association between acute and long COVID-19 severity and outcomes and endothelial biomarkers.
Collapse
Affiliation(s)
- Alice G. Vassiliou
- First Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 106 76 Athens, Greece; (C.S.V.); (C.K.); (I.D.); (A.K.)
| | | | | | | | | | - Stylianos E. Orfanos
- First Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 106 76 Athens, Greece; (C.S.V.); (C.K.); (I.D.); (A.K.)
| |
Collapse
|
13
|
Kim J, Stechmiller J, Weaver M, Gibson DJ, Horgas A, Kelly DL, Lyon DE. The association of wound factors and symptoms of fatigue and pain with wound healing in chronic venous leg ulcers. Int Wound J 2023; 20:1098-1111. [PMID: 36181308 PMCID: PMC10031222 DOI: 10.1111/iwj.13966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 08/31/2022] [Accepted: 09/11/2022] [Indexed: 11/30/2022] Open
Abstract
The purpose of this study was: (1) to characterise the association of wound area, wound exudate C-reactive protein (CRP), broad-spectrum matrix metalloprotease protein (MMPs), and symptoms of fatigue and pain in individuals with chronic venous leg ulcers (CVLUs) over time and (2) to identify factors associated with the wound healing trajectory in CVLUs. Seventy four participants with CVLU who received weekly sharp debridement were recruited from a wound care clinic during the 8-week study period. To examine associations among wound CRP, MMPs, pain, fatigue, and wound healing trajectory over time, we calculated Bayes factors (BF) based on a linear mixed model. The mean age of participants was 71.8 (SD = 9.8) and the mean wound area was 2278 mm2 (SD = 7085 mm2 ) at baseline. Higher fatigue was strongly associated with higher MMPs (BF = 9, 95% HDI: [-.05, .43]), lower CRP (BF = 11, 95% HDI: [-.02, .002]), and large areas of wound (BF = 20, 95% HDI: [-.001, .01]). Higher CRP and MMPs activity in wound exudate and higher fatigue were associated with a larger wound area. To facilitate wound healing, clinicians need to utilise the multifactorial approach, which includes wound treatment and management of symptoms such as pain and fatigue, because of the molecular and psycho-behavioural factors involved in wound healing.
Collapse
Affiliation(s)
- Junglyun Kim
- College of Nursing, Chungnam National University College of Nursing, Daejeon, South Korea
- Department of Biobehavioral Nursing Science, University of Florida College of Nursing, Gainesville, Florida, USA
| | - Joyce Stechmiller
- Department of Biobehavioral Nursing Science, University of Florida College of Nursing, Gainesville, Florida, USA
| | - Michael Weaver
- Department of Biobehavioral Nursing Science, University of Florida College of Nursing, Gainesville, Florida, USA
| | - Daniel J Gibson
- University of Alabama Capstone College of Nursing, Tuscaloosa, Alabama, USA
| | - Ann Horgas
- Department of Biobehavioral Nursing Science, University of Florida College of Nursing, Gainesville, Florida, USA
| | - Debra L Kelly
- Department of Biobehavioral Nursing Science, University of Florida College of Nursing, Gainesville, Florida, USA
| | - Debra E Lyon
- Department of Biobehavioral Nursing Science, University of Florida College of Nursing, Gainesville, Florida, USA
| |
Collapse
|
14
|
Nepotchatykh E, Caraus I, Elremaly W, Leveau C, Elbakry M, Godbout C, Rostami-Afshari B, Petre D, Khatami N, Franco A, Moreau A. Circulating microRNA expression signatures accurately discriminate myalgic encephalomyelitis from fibromyalgia and comorbid conditions. Sci Rep 2023; 13:1896. [PMID: 36732593 PMCID: PMC9894933 DOI: 10.1038/s41598-023-28955-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS), and fibromyalgia (FM) are two chronic complex diseases with overlapping symptoms affecting multiple systems and organs over time. Due to the absence of validated biomarkers and similarity in symptoms, both disorders are misdiagnosed, and the comorbidity of the two is often unrecognized. Our study aimed to investigate the expression profiles of 11 circulating miRNAs previously associated with ME/CFS pathogenesis in FM patients and individuals with a comorbid diagnosis of FM associated with ME/CFS (ME/CFS + FM), and matched sedentary healthy controls. Whether these 11 circulating miRNAs expression can differentiate between the two disorders was also examined. Our results highlight differential circulating miRNAs expression signatures between ME/CFS, FM and ME/CFS + FM, which also correlate to symptom severity between ME/CFS and ME/CFS + FM groups. We provided a prediction model, by using a machine-learning approach based on 11 circulating miRNAs levels, which can be used to discriminate between patients suffering from ME/CFS, FM and ME/CFS + FM. These 11 miRNAs are proposed as potential biomarkers for discriminating ME/CFS from FM. The results of this study demonstrate that ME/CFS and FM are two distinct illnesses, and we highlight the comorbidity between the two conditions. Proper diagnosis of patients suffering from ME/CFS, FM or ME/CFS + FM is crucial to elucidate the pathophysiology of both diseases, determine preventive measures, and establish more effective treatments.
Collapse
Affiliation(s)
- Evguenia Nepotchatykh
- Viscogliosi Laboratory in Molecular Genetics of Musculoskeletal Diseases, Office 2.17.027, Sainte-Justine University Hospital Research Center, 3175 Cote-Ste-Catherine Road, Montreal, QC, H3T 1C5, Canada.,Molecular Biology PhD Program, Faculty of Medicine, Université de Montréal, 2900 Edouard Montpetit Blvd, Montreal, QC, H3T 1J4, Canada.,Open Medicine Foundation ME/CFS Collaborative Center at CHU Sainte-Justine/Université de Montréal, Montreal, Canada.,ICanCME Research Network, Sainte-Justine University Hospital Research Center, 3175 Cote-Ste-Catherine Road, Montreal, QC, H3T 1C5, Canada
| | - Iurie Caraus
- Viscogliosi Laboratory in Molecular Genetics of Musculoskeletal Diseases, Office 2.17.027, Sainte-Justine University Hospital Research Center, 3175 Cote-Ste-Catherine Road, Montreal, QC, H3T 1C5, Canada.,Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, 2900 Edouard Montpetit Blvd, Montreal, QC, H3T 1J4, Canada.,Open Medicine Foundation ME/CFS Collaborative Center at CHU Sainte-Justine/Université de Montréal, Montreal, Canada.,ICanCME Research Network, Sainte-Justine University Hospital Research Center, 3175 Cote-Ste-Catherine Road, Montreal, QC, H3T 1C5, Canada
| | - Wesam Elremaly
- Viscogliosi Laboratory in Molecular Genetics of Musculoskeletal Diseases, Office 2.17.027, Sainte-Justine University Hospital Research Center, 3175 Cote-Ste-Catherine Road, Montreal, QC, H3T 1C5, Canada.,Open Medicine Foundation ME/CFS Collaborative Center at CHU Sainte-Justine/Université de Montréal, Montreal, Canada.,ICanCME Research Network, Sainte-Justine University Hospital Research Center, 3175 Cote-Ste-Catherine Road, Montreal, QC, H3T 1C5, Canada
| | - Corinne Leveau
- Viscogliosi Laboratory in Molecular Genetics of Musculoskeletal Diseases, Office 2.17.027, Sainte-Justine University Hospital Research Center, 3175 Cote-Ste-Catherine Road, Montreal, QC, H3T 1C5, Canada.,Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, 2900 Edouard Montpetit Blvd, Montreal, QC, H3T 1J4, Canada.,Open Medicine Foundation ME/CFS Collaborative Center at CHU Sainte-Justine/Université de Montréal, Montreal, Canada.,ICanCME Research Network, Sainte-Justine University Hospital Research Center, 3175 Cote-Ste-Catherine Road, Montreal, QC, H3T 1C5, Canada
| | - Mohamed Elbakry
- Viscogliosi Laboratory in Molecular Genetics of Musculoskeletal Diseases, Office 2.17.027, Sainte-Justine University Hospital Research Center, 3175 Cote-Ste-Catherine Road, Montreal, QC, H3T 1C5, Canada.,Open Medicine Foundation ME/CFS Collaborative Center at CHU Sainte-Justine/Université de Montréal, Montreal, Canada.,ICanCME Research Network, Sainte-Justine University Hospital Research Center, 3175 Cote-Ste-Catherine Road, Montreal, QC, H3T 1C5, Canada.,Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, Egypt
| | - Christian Godbout
- Patient-Partner, ICanCME Research Network, Sainte-Justine University Hospital Research Center, 3175 Cote-Ste-Catherine Road, Montreal, QC, H3T 1C5, Canada
| | - Bita Rostami-Afshari
- Viscogliosi Laboratory in Molecular Genetics of Musculoskeletal Diseases, Office 2.17.027, Sainte-Justine University Hospital Research Center, 3175 Cote-Ste-Catherine Road, Montreal, QC, H3T 1C5, Canada.,Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, 2900 Edouard Montpetit Blvd, Montreal, QC, H3T 1J4, Canada.,Open Medicine Foundation ME/CFS Collaborative Center at CHU Sainte-Justine/Université de Montréal, Montreal, Canada.,ICanCME Research Network, Sainte-Justine University Hospital Research Center, 3175 Cote-Ste-Catherine Road, Montreal, QC, H3T 1C5, Canada
| | - Diana Petre
- Viscogliosi Laboratory in Molecular Genetics of Musculoskeletal Diseases, Office 2.17.027, Sainte-Justine University Hospital Research Center, 3175 Cote-Ste-Catherine Road, Montreal, QC, H3T 1C5, Canada.,Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, 2900 Edouard Montpetit Blvd, Montreal, QC, H3T 1J4, Canada.,Open Medicine Foundation ME/CFS Collaborative Center at CHU Sainte-Justine/Université de Montréal, Montreal, Canada.,ICanCME Research Network, Sainte-Justine University Hospital Research Center, 3175 Cote-Ste-Catherine Road, Montreal, QC, H3T 1C5, Canada
| | - Nasrin Khatami
- Viscogliosi Laboratory in Molecular Genetics of Musculoskeletal Diseases, Office 2.17.027, Sainte-Justine University Hospital Research Center, 3175 Cote-Ste-Catherine Road, Montreal, QC, H3T 1C5, Canada.,Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, 2900 Edouard Montpetit Blvd, Montreal, QC, H3T 1J4, Canada
| | - Anita Franco
- Viscogliosi Laboratory in Molecular Genetics of Musculoskeletal Diseases, Office 2.17.027, Sainte-Justine University Hospital Research Center, 3175 Cote-Ste-Catherine Road, Montreal, QC, H3T 1C5, Canada.,Open Medicine Foundation ME/CFS Collaborative Center at CHU Sainte-Justine/Université de Montréal, Montreal, Canada.,ICanCME Research Network, Sainte-Justine University Hospital Research Center, 3175 Cote-Ste-Catherine Road, Montreal, QC, H3T 1C5, Canada
| | - Alain Moreau
- Viscogliosi Laboratory in Molecular Genetics of Musculoskeletal Diseases, Office 2.17.027, Sainte-Justine University Hospital Research Center, 3175 Cote-Ste-Catherine Road, Montreal, QC, H3T 1C5, Canada. .,Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, 2900 Edouard Montpetit Blvd, Montreal, QC, H3T 1J4, Canada. .,Open Medicine Foundation ME/CFS Collaborative Center at CHU Sainte-Justine/Université de Montréal, Montreal, Canada. .,ICanCME Research Network, Sainte-Justine University Hospital Research Center, 3175 Cote-Ste-Catherine Road, Montreal, QC, H3T 1C5, Canada. .,Department of Stomatology, Faculty of Dentistry, Université de Montréal, 2900 Edouard Montpetit Blvd, Montreal, QC, H3T 1J4, Canada.
| |
Collapse
|
15
|
Sukocheva OA, Maksoud R, Beeraka NM, Madhunapantula SV, Sinelnikov M, Nikolenko VN, Neganova ME, Klochkov SG, Amjad Kamal M, Staines DR, Marshall-Gradisnik S. Analysis of post COVID-19 condition and its overlap with myalgic encephalomyelitis/chronic fatigue syndrome. J Adv Res 2022; 40:179-196. [PMID: 36100326 PMCID: PMC8619886 DOI: 10.1016/j.jare.2021.11.013] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/02/2021] [Accepted: 11/22/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) disease (COVID-19) triggers the development of numerous pathologies and infection-linked complications and exacerbates existing pathologies in nearly all body systems. Aside from the primarily targeted respiratory organs, adverse SARS-CoV-2 effects were observed in nervous, cardiovascular, gastrointestinal/metabolic, immune, and other systems in COVID-19 survivors. Long-term effects of this viral infection have been recently observed and represent distressing sequelae recognised by the World Health Organisation (WHO) as a distinct clinical entity defined as post-COVID-19 condition. Considering the pandemic is still ongoing, more time is required to confirm post COVID-19 condition diagnosis in the COVID-19 infected cohorts, although many reported post COVID-19 symptoms overlap with myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). AIMS OF REVIEW In this study, COVID-19 clinical presentation and associated post-infection sequelae (post-COVID-19 condition) were reviewed and compared with ME/CFS symptomatology. KEY SCIENTIFIC CONCEPTS OF REVIEW The onset, progression, and symptom profile of post COVID-19 condition patients have considerable overlap with ME/CFS. Considering the large scope and range of pro-inflammatory effects of this virus, it is reasonable to expect development of post COVID-19 clinical complications in a proportion of the affected population. There are reports of a later debilitating syndrome onset three months post COVID-19 infection (often described as long-COVID-19), marked by the presence of fatigue, headache, cognitive dysfunction, post-exertional malaise, orthostatic intolerance, and dyspnoea. Acute inflammation, oxidative stress, and increased levels of interleukin-6 (IL-6) and tumor necrosis factor α (TNFα), have been reported in SARS-CoV-2 infected patients. Longitudinal monitoring of post COVID-19 patients is warranted to understand the long-term effects of SARS-CoV-2 infection and the pathomechanism of post COVID-19 condition.
Collapse
Affiliation(s)
- Olga A Sukocheva
- College of Nursing and Health Sciences, Flinders University of South Australia, Bedford Park 5042, SA, Australia; The National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia.
| | - Rebekah Maksoud
- The National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia; Consortium Health International for Myalgic Encephalomyelitis, National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia
| | - Narasimha M Beeraka
- Department of Biochemistry, Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), JSS Academy of Higher Education & Research (JSS AHER), Mysore, India
| | - SabbaRao V Madhunapantula
- Department of Biochemistry, Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), JSS Academy of Higher Education & Research (JSS AHER), Mysore, India; Special Interest Group in Cancer Biology and Cancer Stem Cells (SIG-CBCSC), JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysore, India
| | - Mikhail Sinelnikov
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Mohovaya 11c10, Moscow, Russia
| | - Vladimir N Nikolenko
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Mohovaya 11c10, Moscow, Russia
| | - Margarita E Neganova
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka 142432, Russia
| | - Sergey G Klochkov
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka 142432, Russia
| | - Mohammad Amjad Kamal
- King Fahd Medical Research Center, King Abdulaziz University, P. O. Box 80216, Jeddah 21589, Saudi Arabia; Enzymoics, 7 Peterlee Place, Novel Global Community Educational Foundation, Hebersham, NSW 2770, Australia
| | - Donald R Staines
- The National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia; Consortium Health International for Myalgic Encephalomyelitis, National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia
| | - Sonya Marshall-Gradisnik
- The National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia; Consortium Health International for Myalgic Encephalomyelitis, National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia
| |
Collapse
|
16
|
Serum of Post-COVID-19 Syndrome Patients with or without ME/CFS Differentially Affects Endothelial Cell Function In Vitro. Cells 2022; 11:cells11152376. [PMID: 35954219 PMCID: PMC9367589 DOI: 10.3390/cells11152376] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 12/24/2022] Open
Abstract
A proportion of COVID-19 reconvalescent patients develop post-COVID-19 syndrome (PCS) including a subgroup fulfilling diagnostic criteria of Myalgic encephalomyelitis/Chronic Fatigue Syndrome (PCS/CFS). Recently, endothelial dysfunction (ED) has been demonstrated in these patients, but the mechanisms remain elusive. Therefore, we investigated the effects of patients’ sera on endothelia cells (ECs) in vitro. PCS (n = 17), PCS/CFS (n = 13), and healthy controls (HC, n = 14) were screened for serum anti-endothelial cell autoantibodies (AECAs) and dysregulated cytokines. Serum-treated ECs were analysed for the induction of activation markers and the release of small molecules by flow cytometry. Moreover, the angiogenic potential of sera was measured in a tube formation assay. While only marginal differences between patient groups were observed for serum cytokines, AECA binding to ECs was significantly increased in PCS/CFS patients. Surprisingly, PCS and PCS/CFS sera reduced surface levels of several EC activation markers. PCS sera enhanced the release of molecules associated with vascular remodelling and significantly promoted angiogenesis in vitro compared to the PCS/CFS and HC groups. Additionally, sera from both patient cohorts induced the release of molecules involved in inhibition of nitric oxide-mediated endothelial relaxation. Overall, PCS and PCS/CFS patients′ sera differed in their AECA content and their functional effects on ECs, i.e., secretion profiles and angiogenic potential. We hypothesise a pro-angiogenic effect of PCS sera as a compensatory mechanism to ED which is absent in PCS/CFS patients.
Collapse
|
17
|
Joseph P, Pari R, Miller S, Warren A, Stovall MC, Squires J, Chang CJ, Xiao W, Waxman AB, Systrom DM. Neurovascular Dysregulation and Acute Exercise Intolerance in ME/CFS: A Randomized, Placebo-Controlled Trial of Pyridostigmine. Chest 2022; 162:1116-1126. [PMID: 35526605 DOI: 10.1016/j.chest.2022.04.146] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/22/2022] [Accepted: 04/22/2022] [Indexed: 10/18/2022] Open
Abstract
BACKGROUND Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is characterized by intractable fatigue, postexertional malaise, and orthostatic intolerance, but its pathophysiology is poorly understood. Pharmacologic cholinergic stimulation was used to test the hypothesis that neurovascular dysregulation underlies exercise intolerance in ME/CFS. RESEARCH QUESTION Does neurovascular dysregulation contribute to exercise intolerance in ME/CFS, and can its treatment improve exercise capacity? STUDY DESIGN AND METHODS Forty-five subjects with ME/CFS were enrolled in a single-center, randomized, double-blind, placebo-controlled trial. Subjects were assigned in a 1:1 ratio to receive a 60-mg dose of oral pyridostigmine or placebo after an invasive cardiopulmonary exercise test (iCPET). A second iCPET was performed 50 min later. The primary end point was the difference in peak exercise oxygen uptake (Vo2). Secondary end points included exercise pulmonary and systemic hemodynamics and gas exchange. RESULTS Twenty-three subjects were assigned to receive pyridostigmine and 22 to receive placebo. The peak Vo2 increased after pyridostigmine but decreased after placebo (13.3 ± 13.4 mL/min vs -40.2 ± 21.3 mL/min; P < .05). The treatment effect of pyridostigmine was 53.6 mL/min (95% CI, -105.2 to -2.0). Peak vs rest Vo2 (25.9 ± 15.3 mL/min vs -60.8 ± 25.6 mL/min; P < .01), cardiac output (-0.2 ± 0.6 L/min vs -1.9 ± 0.6 L/min; P < .05), and right atrial pressure (1.0 ± 0.5 mm Hg vs -0.6 ± 0.5 mm Hg; P < .05) were greater in the pyridostigmine group compared with placebo. INTERPRETATION Pyridostigmine improves peak Vo2 in ME/CFS by increasing cardiac output and right ventricular filling pressures. Worsening peak exercise Vo2, cardiac output, and right atrial pressure following placebo may signal the onset of postexertional malaise. We suggest that treatable neurovascular dysregulation underlies acute exercise intolerance in ME/CFS. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov; No.: NCT03674541; URL: www. CLINICALTRIALS gov.
Collapse
Affiliation(s)
- Phillip Joseph
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Yale-New Haven Hospital, Yale University, New Haven, CT
| | - Rosa Pari
- Department of Medicine, University of Rochester Medical Center, Rochester, NY
| | - Sarah Miller
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Arabella Warren
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Mary Catherine Stovall
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Johanna Squires
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Chia-Jung Chang
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Wenzhong Xiao
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Aaron B Waxman
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - David M Systrom
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
18
|
Campagne J, Fornasieri I, Andreani B, Eginard M, de Korwin JD. Separating Patients with SEID from Those with CFS in the French ME/CFS Association, with Some Thoughts on Nomenclature. Diagnostics (Basel) 2022; 12:1095. [PMID: 35626248 PMCID: PMC9139646 DOI: 10.3390/diagnostics12051095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/17/2022] [Accepted: 04/22/2022] [Indexed: 12/04/2022] Open
Abstract
In 2015, the American Institute of Medicine, now called the National Academy of Medicine, (IOM/NAM) proposed new diagnostic criteria for both Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) and a new label: Systemic Exertion Intolerance Disease (SEID). This study aimed to evaluate the SEID criteria among members of the French Association of ME/CFS (ASFC) and their opinion about this new name. We sent an anonymous questionnaire to 494 ASFC members, using French-translated questions derived from the IOM/NAM tool kit. Among the 178/231 responding subjects who reported ME/CFS diagnosis, 150 (84%) met the criteria of SEID. For each set of questions, we identified some of them that significantly distinguished SEID from non-SEID patients concerning unrefreshing sleep, cognitive disorders, and orthostatic intolerance items. Forty-six percent of the respondents considered the "SEID" terminology as more appropriate than "CFS", 39% considered it inappropriate, and 15% had no opinion. Some questions better identified the SEID criteria. The IOM/NAM SEID criteria captured a large part of ASFC members suffering from ME/CFS. However, this new SEID label was not well accepted by the subjects, nor were the other denominations, suggesting that a better term should be found. Pending development of specific markers, further work with patient communities is needed to find a more suitable label.
Collapse
Affiliation(s)
- Julien Campagne
- Internal Medicine Department, University of Lorraine, 34, Cours Leopold, CS 25233, CEDEX, 54052 Nancy, France;
- Internal Medicine Department, University Hospital of Nancy, Rue du Morvan, CEDEX, 54511 Vandœuvre-Lès-Nancy, France
| | - Isabelle Fornasieri
- Faculty of Psychology, University of Strasbourg, 12, Rue Goethe, 67000 Strasbourg, France;
- French Association for Chronic Fatigue Syndrome (ASFC), Maison des Associations Nice Centre, 3 bis, rue Guigonis, 06300 Nice, France
| | - Barbara Andreani
- Regional Center for Scientific Documentation and Clinical Research, Legouest Army Instruction Hospital, 27, Avenue de Plantières, 57077 Metz, France;
| | - Monique Eginard
- French Association for Chronic Fatigue Syndrome (ASFC), 25, Impasse des Lavandes, 13710 Fuveau, France;
| | - Jean-Dominique de Korwin
- Internal Medicine Department, University of Lorraine, 34, Cours Leopold, CS 25233, CEDEX, 54052 Nancy, France;
- Internal Medicine Department, University Hospital of Nancy, Rue du Morvan, CEDEX, 54511 Vandœuvre-Lès-Nancy, France
| |
Collapse
|
19
|
Labus JS, Mayer EA, Tillisch K, Aagaard KM, Stains J, Broniowska K, Van Remortel C, Tun G, Rapkin A. Dysregulation in Sphingolipid Signaling Pathways is Associated With Symptoms and Functional Connectivity of Pain Processing Brain Regions in Provoked Vestibulodynia. THE JOURNAL OF PAIN 2021; 22:1586-1605. [PMID: 34029688 PMCID: PMC10460622 DOI: 10.1016/j.jpain.2021.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/27/2021] [Accepted: 04/29/2021] [Indexed: 10/21/2022]
Abstract
Provoked vestibulodynia (PVD) is a chronic pain disorder characterized by local hypersensitivity and severe pain with pressure localized to the vulvar vestibule. Despite decades of study, the lack of identified biomarkers has slowed the development of effective therapies. The primary aim of this study was to use metabolomics to identify novel biochemical mechanisms in vagina and blood underlying brain biomarkers and symptoms in PVD, thereby closing this knowledge gap. Using a cross-sectional case-control observational study design, untargeted and unbiased metabolomic profiling of vaginal fluid and plasma was performed in women with PVD compared to healthy controls. In women with PVD, we also obtained assessments of vulvar pain, vestibular and vaginal muscle tenderness, and 24-hour symptom intensity alongside resting-state brain functional connectivity of brain regions involved in pain processing and modulation. Compared to healthy controls, women with PVD demonstrated differences primarily in vaginal (but not plasma) concentrations of metabolites of the sphingolipid signaling pathways, suggesting localized effects in vagina and vulvar vestibule rather than systemic effects. Our findings reveal that dysregulation of sphingolipid metabolism in PVD is associated with increased vulvar pain and muscle tenderness, sexual dysfunction, and decreased functional connectivity strength in pain processing/modulatory brain regions. This data collectively suggests that alterations in sphingolipid signaling pathways are likely an important molecular biomarker in PVD that could lead to new targets for therapeutic intervention. PERSPECTIVE: This manuscript presents the results of a robust, unbiased molecular assessment of plasma and vaginal fluid samples in women with provoked vestibulodynia compared to healthy controls. The findings suggest that alterations in sphingolipid signaling pathways are associated with symptoms and brain biomarkers and may be an important molecular marker that could provide new targets for therapeutic intervention.
Collapse
Affiliation(s)
- Jennifer S Labus
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, California; Brain Research Institute UCLA, Gonda (Goldschmied) Neuroscience and Genetics Research Center, Los Angeles, California.
| | - Emeran A Mayer
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, California
| | - Kirsten Tillisch
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, California
| | - Kjersti M Aagaard
- Division of Maternal-Fetal Medicine, Departments of Obstetrics and Gynecology, Baylor College of Medicine and Texas Children's Hospital, Houston, Texas; Department of Molecular and Human Genetics, Bioinformatics Research Laboratory, Baylor College of Medicine, Houston, Texas; Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, Texas
| | - Jean Stains
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, California
| | | | - Charlotte Van Remortel
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, California
| | - Guistinna Tun
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, California
| | - Andrea Rapkin
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, California; Department of Obstetrics and Gynecology, David Geffen School of Medicine at the University of California, Los Angeles, California
| |
Collapse
|
20
|
Simonato M, Dall’Acqua S, Zilli C, Sut S, Tenconi R, Gallo N, Sfriso P, Sartori L, Cavallin F, Fiocco U, Cogo P, Agostinis P, Aldovini A, Bruttomesso D, Marcolongo R, Comai S, Baritussio A. Tryptophan Metabolites, Cytokines, and Fatty Acid Binding Protein 2 in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. Biomedicines 2021; 9:biomedicines9111724. [PMID: 34829952 PMCID: PMC8615774 DOI: 10.3390/biomedicines9111724] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/14/2021] [Accepted: 11/15/2021] [Indexed: 02/04/2023] Open
Abstract
Patients with Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) differ for triggers, mode of start, associated symptoms, evolution, and biochemical traits. Therefore, serious attempts are underway to partition them into subgroups useful for a personalized medicine approach to the disease. Here, we investigated clinical and biochemical traits in 40 ME/CFS patients and 40 sex- and age-matched healthy controls. Particularly, we analyzed serum levels of some cytokines, Fatty Acid Binding Protein 2 (FAPB-2), tryptophan, and some of its metabolites via serotonin and kynurenine. ME/CFS patients were heterogeneous for genetic background, trigger, start mode, symptoms, and evolution. ME/CFS patients had higher levels of IL-17A (p = 0.018), FABP-2 (p = 0.002), and 3-hydroxykynurenine (p = 0.037) and lower levels of kynurenine (p = 0.012) and serotonin (p = 0.045) than controls. Changes in kynurenine and 3-hydroxykynurenine were associated with increased kynurenic acid/kynurenine and 3-hydroxykynurenine/kynurenine ratios, indirect measures of kynurenine aminotransferases and kynurenine 3-monooxygenase enzymatic activities, respectively. No correlation was found among cytokines, FABP-2, and tryptophan metabolites, suggesting that inflammation, anomalies of the intestinal barrier, and changes of tryptophan metabolism may be independently associated with the pathogenesis of the disease. Interestingly, patients with the start of the disease after infection showed lower levels of kynurenine (p = 0.034) than those not starting after an infection. Changes in tryptophan metabolites and increased IL-17A levels in ME/CFS could both be compatible with anomalies in the sphere of energy metabolism. Overall, clinical traits together with serum biomarkers related to inflammation, intestine function, and tryptophan metabolism deserve to be further considered for the development of personalized medicine strategies for ME/CFS.
Collapse
Affiliation(s)
- Manuela Simonato
- PCare Laboratory, Fondazione Istituto di Ricerca Pediatrica, Citta’ della Speranza, 35127 Padova, Italy;
| | - Stefano Dall’Acqua
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, 35131 Padua, Italy; (S.D.); (S.S.)
| | | | - Stefania Sut
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, 35131 Padua, Italy; (S.D.); (S.S.)
| | - Romano Tenconi
- Department of Medicine, University of Padova, 35128 Padova, Italy; (R.T.); (P.S.); (L.S.); (U.F.); (D.B.); (R.M.); (A.B.)
| | - Nicoletta Gallo
- Department of Laboratory Medicine, Policlinico Azienda Ospedaliera di Padova, 35128 Padova, Italy;
| | - Paolo Sfriso
- Department of Medicine, University of Padova, 35128 Padova, Italy; (R.T.); (P.S.); (L.S.); (U.F.); (D.B.); (R.M.); (A.B.)
| | - Leonardo Sartori
- Department of Medicine, University of Padova, 35128 Padova, Italy; (R.T.); (P.S.); (L.S.); (U.F.); (D.B.); (R.M.); (A.B.)
| | | | - Ugo Fiocco
- Department of Medicine, University of Padova, 35128 Padova, Italy; (R.T.); (P.S.); (L.S.); (U.F.); (D.B.); (R.M.); (A.B.)
| | - Paola Cogo
- Department of Medicine, University Hospital Santa Maria della Misericordia, University of Udine, 33100 Udine, Italy;
| | - Paolo Agostinis
- Department of Medicine, Ospedale Sant’Antonio Abate, Azienda Sanitaria del Friuli Centrale, 33100 Udine, Italy;
| | - Anna Aldovini
- Department of Medicine, Boston Children’s Hospital, Boston, MA 02115, USA;
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Daniela Bruttomesso
- Department of Medicine, University of Padova, 35128 Padova, Italy; (R.T.); (P.S.); (L.S.); (U.F.); (D.B.); (R.M.); (A.B.)
| | - Renzo Marcolongo
- Department of Medicine, University of Padova, 35128 Padova, Italy; (R.T.); (P.S.); (L.S.); (U.F.); (D.B.); (R.M.); (A.B.)
| | - Stefano Comai
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, 35131 Padua, Italy; (S.D.); (S.S.)
- Department of Biomedical Sciences, University of Padua, 35121 Padua, Italy
- Department of Psychiatry, McGill University, Montreal, QC H4H 1R3, Canada
- Division of Neuroscience, IRCSS San Raffaele Scientific Institute, 20132 Milan, Italy
- Correspondence: ; Tel.: +39-049-827-5098
| | - Aldo Baritussio
- Department of Medicine, University of Padova, 35128 Padova, Italy; (R.T.); (P.S.); (L.S.); (U.F.); (D.B.); (R.M.); (A.B.)
| |
Collapse
|
21
|
Ruiz-Pablos M, Paiva B, Montero-Mateo R, Garcia N, Zabaleta A. Epstein-Barr Virus and the Origin of Myalgic Encephalomyelitis or Chronic Fatigue Syndrome. Front Immunol 2021; 12:656797. [PMID: 34867935 PMCID: PMC8634673 DOI: 10.3389/fimmu.2021.656797] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 10/19/2021] [Indexed: 01/04/2023] Open
Abstract
Myalgic encephalomyelitis or chronic fatigue syndrome (ME/CFS) affects approximately 1% of the general population. It is a chronic, disabling, multi-system disease for which there is no effective treatment. This is probably related to the limited knowledge about its origin. Here, we summarized the current knowledge about the pathogenesis of ME/CFS and revisit the immunopathobiology of Epstein-Barr virus (EBV) infection. Given the similarities between EBV-associated autoimmune diseases and cancer in terms of poor T cell surveillance of cells with EBV latency, expanded EBV-infected cells in peripheral blood and increased antibodies against EBV, we hypothesize that there could be a common etiology generated by cells with EBV latency that escape immune surveillance. Albeit inconclusive, multiple studies in patients with ME/CFS have suggested an altered cellular immunity and augmented Th2 response that could result from mechanisms of evasion to some pathogens such as EBV, which has been identified as a risk factor in a subset of ME/CFS patients. Namely, cells with latency may evade the immune system in individuals with genetic predisposition to develop ME/CFS and in consequence, there could be poor CD4 T cell immunity to mitogens and other specific antigens, as it has been described in some individuals. Ultimately, we hypothesize that within ME/CFS there is a subgroup of patients with DRB1 and DQB1 alleles that could confer greater susceptibility to EBV, where immune evasion mechanisms generated by cells with latency induce immunodeficiency. Accordingly, we propose new endeavors to investigate if anti-EBV therapies could be effective in selected ME/CFS patients.
Collapse
Affiliation(s)
| | - Bruno Paiva
- Clinica Universidad de Navarra, Centro de Investigación Medica Aplicada (CIMA), IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | | | - Nicolas Garcia
- Clinica Universidad de Navarra, Centro de Investigación Medica Aplicada (CIMA), IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Aintzane Zabaleta
- Clinica Universidad de Navarra, Centro de Investigación Medica Aplicada (CIMA), IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| |
Collapse
|
22
|
Song JH, Won SK, Eom GH, Lee DS, Park BJ, Lee JS, Son CG, Park JY. Improvement Effects of Myelophil on Symptoms of Chronic Fatigue Syndrome in a Reserpine-Induced Mouse Model. Int J Mol Sci 2021; 22:10199. [PMID: 34638540 PMCID: PMC8508381 DOI: 10.3390/ijms221910199] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/10/2021] [Accepted: 09/13/2021] [Indexed: 01/15/2023] Open
Abstract
Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is associated with various symptoms, such as depression, pain, and fatigue. To date, the pathological mechanisms and therapeutics remain uncertain. The purpose of this study was to investigate the effect of myelophil (MYP), composed of Astragali Radix and Salviaemiltiorrhizae Radix, on depression, pain, and fatigue behaviors and its underlying mechanisms. Reserpine (2 mg/kg for 10 days, intraperitoneally) induced depression, pain, and fatigue behaviors in mice. MYP treatment (100 mg/kg for 10 days, intragastrically) significantly improved depression behaviors, mechanical and thermal hypersensitivity, and fatigue behavior. MYP treatment regulated the expression of c-Fos, 5-HT1A/B receptors, and transforming growth factor β (TGF-β) in the brain, especially in the motor cortex, hippocampus, and nucleus of the solitary tract. MYP treatment decreased ionized calcium binding adapter molecule 1 (Iba1) expression in the hippocampus and increased tyrosine hydroxylase (TH) expression and the levels of dopamine and serotonin in the striatum. MYP treatment altered inflammatory and anti-oxidative-related mRNA expression in the spleen and liver. In conclusion, MYP was effective in recovering major symptoms of ME/CFS and was associated with the regulation of dopaminergic and serotonergic pathways and TGF-β expression in the brain, as well as anti-inflammatory and anti-oxidant mechanisms in internal organs.
Collapse
Affiliation(s)
- Ji-Hye Song
- Department of Korean Medicine, College of Korean Medicine, Daejeon University, Daejeon 34520, Korea; (J.-H.S.); (S.-K.W.); (G.-H.E.); (D.-S.L.); (B.-J.P.); (J.-S.L.); (C.-G.S.)
- Institute of Bioscience & Integrative Medicine, Daejeon University, Daejeon 34520, Korea
| | - Seul-Ki Won
- Department of Korean Medicine, College of Korean Medicine, Daejeon University, Daejeon 34520, Korea; (J.-H.S.); (S.-K.W.); (G.-H.E.); (D.-S.L.); (B.-J.P.); (J.-S.L.); (C.-G.S.)
- Institute of Bioscience & Integrative Medicine, Daejeon University, Daejeon 34520, Korea
| | - Geun-Hyang Eom
- Department of Korean Medicine, College of Korean Medicine, Daejeon University, Daejeon 34520, Korea; (J.-H.S.); (S.-K.W.); (G.-H.E.); (D.-S.L.); (B.-J.P.); (J.-S.L.); (C.-G.S.)
- Institute of Bioscience & Integrative Medicine, Daejeon University, Daejeon 34520, Korea
| | - Da-Som Lee
- Department of Korean Medicine, College of Korean Medicine, Daejeon University, Daejeon 34520, Korea; (J.-H.S.); (S.-K.W.); (G.-H.E.); (D.-S.L.); (B.-J.P.); (J.-S.L.); (C.-G.S.)
- Institute of Bioscience & Integrative Medicine, Daejeon University, Daejeon 34520, Korea
| | - Byung-Jin Park
- Department of Korean Medicine, College of Korean Medicine, Daejeon University, Daejeon 34520, Korea; (J.-H.S.); (S.-K.W.); (G.-H.E.); (D.-S.L.); (B.-J.P.); (J.-S.L.); (C.-G.S.)
| | - Jin-Seok Lee
- Department of Korean Medicine, College of Korean Medicine, Daejeon University, Daejeon 34520, Korea; (J.-H.S.); (S.-K.W.); (G.-H.E.); (D.-S.L.); (B.-J.P.); (J.-S.L.); (C.-G.S.)
- Institute of Bioscience & Integrative Medicine, Daejeon University, Daejeon 34520, Korea
| | - Chang-Gue Son
- Department of Korean Medicine, College of Korean Medicine, Daejeon University, Daejeon 34520, Korea; (J.-H.S.); (S.-K.W.); (G.-H.E.); (D.-S.L.); (B.-J.P.); (J.-S.L.); (C.-G.S.)
- Institute of Bioscience & Integrative Medicine, Daejeon University, Daejeon 34520, Korea
| | - Ji-Yeun Park
- Department of Korean Medicine, College of Korean Medicine, Daejeon University, Daejeon 34520, Korea; (J.-H.S.); (S.-K.W.); (G.-H.E.); (D.-S.L.); (B.-J.P.); (J.-S.L.); (C.-G.S.)
- Institute of Bioscience & Integrative Medicine, Daejeon University, Daejeon 34520, Korea
| |
Collapse
|
23
|
Noor N, Urits I, Degueure A, Rando L, Kata V, Cornett EM, Kaye AD, Imani F, Narimani-Zamanabadi M, Varrassi G, Viswanath O. A Comprehensive Update of the Current Understanding of Chronic Fatigue Syndrome. Anesth Pain Med 2021; 11:e113629. [PMID: 34540633 PMCID: PMC8438707 DOI: 10.5812/aapm.113629] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 05/31/2021] [Accepted: 06/03/2021] [Indexed: 02/07/2023] Open
Abstract
This is a comprehensive literature review of chronic fatigue syndrome (CFS). We provide a description of the background, etiology, pathogenesis, diagnosis, and management regarding CFS. CFS is a multifaceted illness that has many symptoms and a wide array of clinical presentations. As of recent, CFS has been merged with myalgic encephalomyelitis (ME). Much of the difficulty in its management has stemmed from a lack of a concrete understanding of its etiology and pathogenesis. There is a potential association between dysfunction of the autoimmune, neuroendocrine, or autonomic nervous systems and the development of CFS. Possible triggering events, such as infections followed by an immune dysregulation resulting have also been proposed. In fact, ME/CFS was first described following Epstein Barr virus (EBV) infections, but it was later determined that it was not always preceded by EBV infection. Patient diagnosed with CFS have shown a noticeably earlier activation of anaerobic metabolism as a source of energy, which is suggestive of impaired oxygen consumption. The differential diagnoses range from tick-borne illnesses to psychiatric disorders to thyroid gland dysfunction. Given the many overlapping symptoms of CFS with other illnesses makes diagnosing it far from an easy task. The Centers for Disease Control and Prevention (CDC) considers it a diagnosing of exclusion, stating that self-reported fatigue for at minimum of six months and four of the following symptoms are necessary for a proper diagnosis: memory problems, sore throat, post-exertion malaise, tender cervical or axillary lymph nodes, myalgia, multi-joint pain, headaches, and troubled sleep. In turn, management of CFS is just as difficult. Treatment ranges from conservative, such as cognitive behavioral therapy (CBT) and antidepressants, to minimally invasive management. Minimally invasive management involving ranscutaneous electrical acupoint stimulation of target points has demonstrated significant improvement in fatigue and associated symptoms in a 2017 randomized controlled study. The understanding of CFS is evolving before us as we continue to learn more about it. As further reliable studies are conducted, providing a better grasp of what the syndrome encompasses, we will be able to improve our diagnosis and management of it.
Collapse
Affiliation(s)
- Nazir Noor
- Mount Sinai Medical Center, Department of Anesthesiology, Miami Beach, FL, USA
| | - Ivan Urits
- Louisiana State University Health Shreveport, Department of Anesthesiology, Shreveport, LA, USA
- Southcoast Health, Southcoast Physician Group Pain Medicine, MA, USA
| | - Arielle Degueure
- Louisiana State University Health Shreveport School of Medicine, Shreveport, LA, USA
| | - Lauren Rando
- Louisiana State University Health Shreveport School of Medicine, Shreveport, LA, USA
| | - Vijay Kata
- Louisiana State University Health Shreveport School of Medicine, Shreveport, LA, USA
| | - Elyse M. Cornett
- Louisiana State University Health Shreveport, Department of Anesthesiology, Shreveport, LA, USA
| | - Alan D. Kaye
- Louisiana State University Health Shreveport, Department of Anesthesiology, Shreveport, LA, USA
| | - Farnad Imani
- Pain Research Center, Department of Anesthesiology and Pain Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | | | - Omar Viswanath
- Louisiana State University Health Shreveport, Department of Anesthesiology, Shreveport, LA, USA
- Valley Anesthesiology and Pain Consultants – Envision Physician Services, Phoenix, AZ, USA
- Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE, USA
- University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| |
Collapse
|
24
|
Maksoud R, Eaton-Fitch N, Matula M, Cabanas H, Staines D, Marshall-Gradisnik S. Systematic Review of Sleep Characteristics in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. Healthcare (Basel) 2021; 9:healthcare9050568. [PMID: 34065013 PMCID: PMC8150292 DOI: 10.3390/healthcare9050568] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/01/2021] [Accepted: 05/07/2021] [Indexed: 02/04/2023] Open
Abstract
(1) Background—Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) is a multifaceted illness characterized by profound and persistent fatigue unrelieved by rest along with a range of other debilitating symptoms. Experiences of unrefreshing and disturbed sleep are frequently described by ME/CFS patients. This is the first systematic review assessing sleep characteristics in ME/CFS. The aim of this review is to determine whether there are clinical characteristics of sleep in ME/CFS patients compared to healthy controls using objective measures such as polysomnography and multiple sleep latency testing. (2) Methods—the following databases—Pubmed, Embase, Medline (EBSCO host) and Web of Science, were systematically searched for journal articles published between January 1994 to 19 February 2021. Articles that referred to polysomnography or multiple sleep latency testing and ME/CFS patients were selected, and further refined through use of specific inclusion and exclusion criteria. Quality and bias were measured using the Joanna Briggs Institute checklist. (3) Results—twenty observational studies were included in this review. The studies investigated objective measures of sleep quality in ME/CFS. Subjective measures including perceived sleep quality and other quality of life factors were also described. (4) Conclusions—Many of the parameters measured including slow- wave sleep, apnea- hypopnea index, spectral activity and multiple sleep latency testing were inconsistent across the studies. The available research on sleep quality in ME/CFS was also limited by recruitment decisions, confounding factors, small sample sizes and non-replicated findings. Future well-designed studies are required to understand sleep quality in ME/CFS patients.
Collapse
Affiliation(s)
- Rebekah Maksoud
- National Centre for Neuroimmunology and Emerging Diseases (NCNED), Menzies Health Institute Queensland, Griffith University, Gold Coast 4222, Australia; (N.E.-F.); (M.M.); (H.C.); (D.S.); (S.M.-G.)
- Consortium Health International for Myalgic Encephalomyelitis, Griffith University, Gold Coast 4222, Australia
- Correspondence:
| | - Natalie Eaton-Fitch
- National Centre for Neuroimmunology and Emerging Diseases (NCNED), Menzies Health Institute Queensland, Griffith University, Gold Coast 4222, Australia; (N.E.-F.); (M.M.); (H.C.); (D.S.); (S.M.-G.)
- Consortium Health International for Myalgic Encephalomyelitis, Griffith University, Gold Coast 4222, Australia
- School of Medical Science, Griffith University, Gold Coast 4222, Australia
| | - Michael Matula
- National Centre for Neuroimmunology and Emerging Diseases (NCNED), Menzies Health Institute Queensland, Griffith University, Gold Coast 4222, Australia; (N.E.-F.); (M.M.); (H.C.); (D.S.); (S.M.-G.)
- Consortium Health International for Myalgic Encephalomyelitis, Griffith University, Gold Coast 4222, Australia
| | - Hélène Cabanas
- National Centre for Neuroimmunology and Emerging Diseases (NCNED), Menzies Health Institute Queensland, Griffith University, Gold Coast 4222, Australia; (N.E.-F.); (M.M.); (H.C.); (D.S.); (S.M.-G.)
- Consortium Health International for Myalgic Encephalomyelitis, Griffith University, Gold Coast 4222, Australia
| | - Donald Staines
- National Centre for Neuroimmunology and Emerging Diseases (NCNED), Menzies Health Institute Queensland, Griffith University, Gold Coast 4222, Australia; (N.E.-F.); (M.M.); (H.C.); (D.S.); (S.M.-G.)
- Consortium Health International for Myalgic Encephalomyelitis, Griffith University, Gold Coast 4222, Australia
| | - Sonya Marshall-Gradisnik
- National Centre for Neuroimmunology and Emerging Diseases (NCNED), Menzies Health Institute Queensland, Griffith University, Gold Coast 4222, Australia; (N.E.-F.); (M.M.); (H.C.); (D.S.); (S.M.-G.)
- Consortium Health International for Myalgic Encephalomyelitis, Griffith University, Gold Coast 4222, Australia
| |
Collapse
|
25
|
Muthanna FMS, Karuppannan M, Hassan BAR, Mohammed AH. Impact of fatigue on quality of life among breast cancer patients receiving chemotherapy. Osong Public Health Res Perspect 2021; 12:115-125. [PMID: 33980002 PMCID: PMC8102880 DOI: 10.24171/j.phrp.2021.12.2.09] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/27/2021] [Indexed: 02/07/2023] Open
Abstract
Objectives Fatigue is the most frequently reported symptom experienced by cancer patients and has a profound effect on their quality of life (QOL). The study aimed to determine the impact of fatigue on QOL among breast cancer patients receiving chemotherapy and to identify the risk factors associated with severe fatigue incidence. Methods This was an observational prospective study carried out at multiple centers. In total, 172 breast cancer patients were included. The Functional Assessment of Chronic Illness Therapy-Fatigue Questionnaire was used to measure QOL, while the Brief Fatigue Inventory (BFI) was used to assess the severity of fatigue. Results The total average mean and standard deviation of QOL were 84.58±18.07 and 4.65±1.14 for BFI scores, respectively. A significant association between fatigue and QOL was found in linear and multiple regression analyses. The relationships between fatigue severity and cancer stage, chemotherapy dose delay, dose reduction, chemotherapy regimen, and ethnicity were determined using binary logistic regression analysis. Conclusion The findings of this study are believed to be useful for helping oncologists effectively evaluate, monitor, and treat fatigue related to QOL changes.
Collapse
Affiliation(s)
- Fares Mohammed Saeed Muthanna
- Department of Pharmacy Practice, Faculty of Pharmacy, Universiti Teknologi MARA, Cawangan Selangor, Kampus Puncak Alam, Selangor, Malaysia
| | - Mahmathi Karuppannan
- Department of Pharmacy Practice, Faculty of Pharmacy, Universiti Teknologi MARA, Cawangan Selangor, Kampus Puncak Alam, Selangor, Malaysia
| | | | - Ali Haider Mohammed
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Selangor, Malaysia
| |
Collapse
|
26
|
Kuo CF, Shi L, Lin CL, Yao WC, Chen HT, Lio CF, Wang YTT, Su CH, Hsu NW, Tsai SY. How peptic ulcer disease could potentially lead to the lifelong, debilitating effects of chronic fatigue syndrome: an insight. Sci Rep 2021; 11:7520. [PMID: 33824394 PMCID: PMC8024330 DOI: 10.1038/s41598-021-87018-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 03/22/2021] [Indexed: 02/08/2023] Open
Abstract
Chronic Fatigue Syndrome (CFS) has been defined as unexplained relapsing or persistent fatigue for at least 6 consecutive months. Immuno-inflammatory pathway, bacterial infection, and other causes play essential roles in CFS. Helicobacter pylori infection is one of the most common causes of foregut inflammation, leading to peptic ulcer disease (PUD). This study aimed to analyze the risk of CFS development between patients with and without PUD. Other related factors were also analyzed. We performed a retrospective, nationwide cohort study identifying patients with or without PUD respectively by analyzing the Longitudinal Health Insurance Database 2000 (LHID2000), Taiwan. The overall incidence of CFS was higher in the PUD cohort than in the non- PUD cohort (HR = 2.01, 95% CI = 1.75-2.30), with the same adjusted HR (aHR) when adjusting for age, sex, and comorbidities. The sex-specific PUD cohort to the non-PUD cohort relative risk of CFS was significant in both genders. The age-specific incidence of CFS showed incidence density increasing with age in both cohorts. There is an increased risk of developing CFS following PUD, especially in females and the aging population. Hopefully, these findings can prevent common infections from progressing to debilitating, chronic conditions such as CFS.
Collapse
Affiliation(s)
- Chien-Feng Kuo
- Department of Medicine, Graduate Institute of Long-Term Care, Graduate Institute of Biomedical Sciences, Mackay Medical College, New Taipei City, Taiwan
- Department of Cosmetic Applications and Management, MacKay Junior College of Medicine, Nursing and Management, New Taipei City, Taiwan
- Division of Infectious Diseases, Department of Internal Medicine, Mackay Memorial Hospital, Taipei, Taiwan
| | - Leiyu Shi
- Department of Health Policy and Management, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Cheng-Li Lin
- College of Medicine, China Medical University, Taichung City, Taiwan
- Management Office for Health Data, China Medical University Hospital, Taichung City, Taiwan
| | - Wei-Cheng Yao
- Department of Anesthesiology and Pain Medicine, Min-Sheng General Hospital, Tao-Yuan, 330, Taiwan
| | - Hsiang-Ting Chen
- Department of Laboratory Medicine, Mackay Memorial Hospital, Taipei, Taiwan
| | - Chon-Fu Lio
- Department of Laboratory Medicine, Mackay Memorial Hospital, Taipei, Taiwan
| | - Yu-Ting Tina Wang
- Department of Laboratory Medicine, Mackay Memorial Hospital, Taipei, Taiwan
| | - Ching-Huang Su
- Department of Laboratory Medicine, Mackay Memorial Hospital, Taipei, Taiwan
| | - Nai-Wei Hsu
- Department of Medicine, Graduate Institute of Long-Term Care, Graduate Institute of Biomedical Sciences, Mackay Medical College, New Taipei City, Taiwan
- Department of Laboratory Medicine, Mackay Memorial Hospital, Taipei, Taiwan
| | - Shin-Yi Tsai
- Department of Medicine, Graduate Institute of Long-Term Care, Graduate Institute of Biomedical Sciences, Mackay Medical College, New Taipei City, Taiwan.
- Department of Cosmetic Applications and Management, MacKay Junior College of Medicine, Nursing and Management, New Taipei City, Taiwan.
- Department of Laboratory Medicine, Mackay Memorial Hospital, Taipei, Taiwan.
| |
Collapse
|
27
|
Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: The Human Herpesviruses Are Back! Biomolecules 2021; 11:biom11020185. [PMID: 33572802 PMCID: PMC7912523 DOI: 10.3390/biom11020185] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/04/2021] [Accepted: 01/23/2021] [Indexed: 12/20/2022] Open
Abstract
Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) or Systemic Exertion Intolerance Disease (SEID) is a chronic multisystem illness of unconfirmed etiology. There are currently no biomarkers and/or signatures available to assist in the diagnosis of the syndrome and while numerous mechanisms have been hypothesized to explain the pathology of ME/CFS, the triggers and/or drivers remain unknown. Initial studies suggested a potential role of the human herpesviruses especially Epstein-Barr virus (EBV) in the disease process but inconsistent and conflicting data led to the erroneous suggestion that these viruses had no role in the syndrome. New studies using more advanced approaches have now demonstrated that specific proteins encoded by EBV could contribute to the immune and neurological abnormalities exhibited by a subgroup of patients with ME/CFS. Elucidating the role of these herpesvirus proteins in ME/CFS may lead to the identification of specific biomarkers and the development of novel therapeutics.
Collapse
|
28
|
Poenaru S, Abdallah SJ, Corrales-Medina V, Cowan J. COVID-19 and post-infectious myalgic encephalomyelitis/chronic fatigue syndrome: a narrative review. Ther Adv Infect Dis 2021; 8:20499361211009385. [PMID: 33959278 PMCID: PMC8060761 DOI: 10.1177/20499361211009385] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 03/13/2021] [Indexed: 12/11/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a viral infection which can cause a variety of respiratory, gastrointestinal, and vascular symptoms. The acute illness phase generally lasts no more than 2-3 weeks. However, there is increasing evidence that a proportion of COVID-19 patients experience a prolonged convalescence and continue to have symptoms lasting several months after the initial infection. A variety of chronic symptoms have been reported including fatigue, dyspnea, myalgia, exercise intolerance, sleep disturbances, difficulty concentrating, anxiety, fever, headache, malaise, and vertigo. These symptoms are similar to those seen in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS), a chronic multi-system illness characterized by profound fatigue, sleep disturbances, neurocognitive changes, orthostatic intolerance, and post-exertional malaise. ME/CFS symptoms are exacerbated by exercise or stress and occur in the absence of any significant clinical or laboratory findings. The pathology of ME/CFS is not known: it is thought to be multifactorial, resulting from the dysregulation of multiple systems in response to a particular trigger. Although not exclusively considered a post-infectious entity, ME/CFS has been associated with several infectious agents including Epstein-Barr Virus, Q fever, influenza, and other coronaviruses. There are important similarities between post-acute COVID-19 symptoms and ME/CFS. However, there is currently insufficient evidence to establish COVID-19 as an infectious trigger for ME/CFS. Further research is required to determine the natural history of this condition, as well as to define risk factors, prevalence, and possible interventional strategies.
Collapse
Affiliation(s)
- Sonia Poenaru
- Department of Medicine, The Ottawa Hospital,
General Campus, 501 Smyth Road, Box 206, Ottawa, Ontario, K1H 8L6,
Canada
| | - Sara J. Abdallah
- Clinical Epidemiology Program, The Ottawa
Hospital Research Institute, Ottawa, Ontario, Canada
| | - Vicente Corrales-Medina
- Clinical Epidemiology Program, The Ottawa
Hospital Research Institute, Ottawa, Ontario, Canada
- Division of Infectious Diseases, Department of
Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Juthaporn Cowan
- Clinical Epidemiology Program, The Ottawa
Hospital Research Institute, Ottawa, Ontario, Canada
- Division of Infectious Diseases, Department of
Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
29
|
Abstract
Owing to the broad differential diagnoses that can present as fatigue, a rational approach to diagnosis is paramount. Performance of a battery of diagnostic tests is unlikely to assist with diagnosis, highlighting the importance of a thorough history and physical examination. Fatigue can be a sequela of an underlying medical disease or exists as a primary condition. Management of secondary fatigue largely depends on treatment of the underlying condition. There are no FDA-approved medications for primary fatigue, now known as system exertion intolerance disease. Treatment is focused on individualized exercise therapy and cognitive behavioral therapy.
Collapse
Affiliation(s)
- Jason C Dukes
- Internal Medicine Department, Eastern Virginia Medical School, 825 Fairfax Avenue, Suite 565, Norfolk, VA 23507, USA.
| | - Matthew Chakan
- Internal Medicine Department, Eastern Virginia Medical School, 825 Fairfax Avenue, Suite 481, Norfolk, VA 23507, USA
| | - Aaron Mills
- Internal Medicine Department, Eastern Virginia Medical School, 825 Fairfax Avenue, Suite 483, Norfolk, VA 23507, USA
| | - Maurice Marcaurd
- Internal Medicine Department, Eastern Virginia Medical School, 825 Fairfax Avenue, Suite 572, Norfolk, VA 23507, USA
| |
Collapse
|
30
|
Muller AE, Tveito K, Bakken IJ, Flottorp SA, Mjaaland S, Larun L. Potential causal factors of CFS/ME: a concise and systematic scoping review of factors researched. J Transl Med 2020; 18:484. [PMID: 33317576 PMCID: PMC7734915 DOI: 10.1186/s12967-020-02665-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 12/04/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Chronic fatigue syndrome/myalgic encephalomyelitis (CFS/ME) is understood as a complex condition, likely triggered and sustained by an interplay of biological, psychological, and social factors. Little oversight exists of the field of causal research. This systematic scoping review explores potential causal factors of CFS/ME as researched by primary studies. METHODS We searched eight databases for primary studies that examined potential causal factors of CFS/ME. Based on title/abstract review, two researchers independently sorted each study's factors into nine main categories and 71 subordinate categories, using a system developed with input given during a 2018 ME conference, specialists and representatives from a ME patient advocacy group, and using BMJ Best Practice's description of CFS/ME etiology. We also extracted data related to study design, size, diagnostic criteria and comparison groups. RESULTS We included 1161 primary studies published between January 1979 and June 2019. Based on title/abstract analysis, no single causal factor dominated in these studies, and studies reported a mean of 2.73 factors. The four most common factors were: immunological (297 studies), psychological (243), infections (198), and neuroendocrinal (198). The most frequent study designs were case-control studies (894 studies) comparing CFS/ME patients with healthy participants. More than half of the studies (that reported study size in the title/abstract) included 100 or fewer participants. CONCLUSION The field of causal hypotheses of CFS/ME is diverse, and we found that the studies examined all the main categories of possible factors that we had defined a priori. Most studies were not designed to adequately explore causality, rather to establish hypotheses. We need larger studies with stronger study designs to gain better knowledge of causal factors of CFS/ME.
Collapse
Affiliation(s)
| | - Kari Tveito
- Journal of the Norwegian Medical Association, Sentrum, PO Box 1152, 0107, Oslo, Norway
| | | | - Signe A Flottorp
- Norwegian Institute of Public Health, Skøyen, PO Box 222, 0213, Oslo, Norway
- University of Oslo, Oslo, Norway
| | - Siri Mjaaland
- Norwegian Institute of Public Health, Skøyen, PO Box 222, 0213, Oslo, Norway
| | - Lillebeth Larun
- Norwegian Institute of Public Health, Skøyen, PO Box 222, 0213, Oslo, Norway
| |
Collapse
|
31
|
Musker M, McArthur A, Munn Z, Wong ML. Circulating leptin levels in patients with myalgic encephalomyelitis, chronic fatigue syndrome or fibromyalgia: a systematic review protocol. JBI Evid Synth 2020; 19:695-701. [PMID: 33136710 DOI: 10.11124/jbies-20-00125] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
OBJECTIVE The objective of the review is to evaluate circulating levels of leptin in people diagnosed with myalgic encephalomyelitis chronic fatigue syndrome or fibromyalgia syndrome and to investigate the differences compared with healthy controls. INTRODUCTION Myalgic encephalomyelitis chronic fatigue syndrome is a condition that has major symptoms, including self-reported fatigue, post-exertional malaise, and unexplained pain across the body. The widespread pain is measured in a systematic way and is often referred to as fibromyalgia. The two disorders have many similarities, but their association with leptin has indicated that leptin may affect the role of pro-inflammatory cytokines and symptom severity. INCLUSION CRITERIA This review will consider observational studies of varying study designs including prospective and retrospective cohort studies, case-control studies, time-series, and analytical cross-sectional studies that include both cases and healthy comparators. Cases will include a diagnosis of myalgic encephalomyelitis, chronic fatigue syndrome, and/or fibromyalgia. Controls are people without this diagnosis, usually healthy participants. Only studies published in English will be included due to limited resources for translation. METHODS This protocol will be reported based on the Preferred Reporting Items for Systematic Reviews and Meta-analysis (PRISMA) checklist and will follow the JBI methodology for systematic reviews of etiology and risk. A comprehensive search strategy will include PubMed, Embase, Scopus, Science Direct, and PsycINFO. Two reviewers will screen, critically appraise eligible articles, and extract data using a standardized data extraction tool informed by JBI SUMARI. The authors will complete a quantitative analysis that synthesizes findings across studies using pooled effect sizes and confidence intervals of the measures provided. SYSTEMATIC REVIEW REGISTRATION NUMBER PROSPERO CRD42020169903.
Collapse
Affiliation(s)
- Michael Musker
- Mental Health and Wellbeing, South Australian Health and Medical Research Institute, Adelaide, SA, Australia.,The Centre for Evidence-based Practice South Australia (CEPSA): A JBI Centre of Excellence, Adelaide, SA, Australia.,Department of Psychiatry, Flinders University, Adelaide, SA, Australia
| | - Alexa McArthur
- The Centre for Evidence-based Practice South Australia (CEPSA): A JBI Centre of Excellence, Adelaide, SA, Australia
| | - Zachary Munn
- The Centre for Evidence-based Practice South Australia (CEPSA): A JBI Centre of Excellence, Adelaide, SA, Australia
| | - Ma-Li Wong
- Mental Health and Wellbeing, South Australian Health and Medical Research Institute, Adelaide, SA, Australia.,Department of Psychiatry, Flinders University, Adelaide, SA, Australia.,Department of Psychiatry, Upstate University, New York, NY, USA
| |
Collapse
|
32
|
Giloteaux L, O'Neal A, Castro-Marrero J, Levine SM, Hanson MR. Cytokine profiling of extracellular vesicles isolated from plasma in myalgic encephalomyelitis/chronic fatigue syndrome: a pilot study. J Transl Med 2020; 18:387. [PMID: 33046133 PMCID: PMC7552484 DOI: 10.1186/s12967-020-02560-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 10/03/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) is a debilitating disease of unknown etiology lasting for a minimum of 6 months but usually for many years, with features including fatigue, cognitive impairment, myalgias, post-exertional malaise, and immune system dysfunction. Dysregulation of cytokine signaling could give rise to many of these symptoms. Cytokines are present in both plasma and extracellular vesicles, but little investigation of EVs in ME/CFS has been reported. Therefore, we aimed to characterize the content of extracellular vesicles (EVs) isolated from plasma (including circulating cytokine/chemokine profiling) from individuals with ME/CFS and healthy controls. METHODS We included 35 ME/CFS patients and 35 controls matched for age, sex and BMI. EVs were enriched from plasma by using a polymer-based precipitation method and characterized by Nanoparticle Tracking Analysis (NTA), Transmission Electron Microscopy (TEM) and immunoblotting. A 45-plex immunoassay was used to determine cytokine levels in both plasma and isolated EVs from a subset of 19 patients and controls. Linear regression, principal component analysis and inter-cytokine correlations were analyzed. RESULTS ME/CFS individuals had significantly higher levels of EVs that ranged from 30 to 130 nm in size as compared to controls, but the mean size for total extracellular vesicles did not differ between groups. The enrichment of typical EV markers CD63, CD81, TSG101 and HSP70 was confirmed by Western blot analysis and the morphology assessed by TEM showed a homogeneous population of vesicles in both groups. Comparison of cytokine concentrations in plasma and isolated EVs of cases and controls yielded no significant differences. Cytokine-cytokine correlations in plasma revealed a significant higher number of interactions in ME/CFS cases along with 13 inverse correlations that were mainly driven by the Interferon gamma-induced protein 10 (IP-10), whereas in the plasma of controls, no inverse relationships were found across any of the cytokines. Network analysis in EVs from controls showed 2.5 times more significant inter-cytokine interactions than in the ME/CFS group, and both groups presented a unique negative association. CONCLUSIONS Elevated levels of 30-130 nm EVs were found in plasma from ME/CFS patients and inter-cytokine correlations revealed unusual regulatory relationships among cytokines in the ME/CFS group that were different from the control group in both plasma and EVs. These disturbances in cytokine networks are further evidence of immune dysregulation in ME/CFS.
Collapse
Affiliation(s)
- Ludovic Giloteaux
- Department of Molecular Biology and Genetics, Cornell University, 323 Biotechnology Building, 526 Campus Road, Ithaca, NY, 14853, USA
| | - Adam O'Neal
- Department of Molecular Biology and Genetics, Cornell University, 323 Biotechnology Building, 526 Campus Road, Ithaca, NY, 14853, USA
| | - Jesús Castro-Marrero
- Department of Molecular Biology and Genetics, Cornell University, 323 Biotechnology Building, 526 Campus Road, Ithaca, NY, 14853, USA
- CFS/ME Unit, Division of Rheumatology, Vall d'Hebron University Hospital Research Institute, Universitat Autònoma de Barcelona, Barcelona, 08035, Spain
| | | | - Maureen R Hanson
- Department of Molecular Biology and Genetics, Cornell University, 323 Biotechnology Building, 526 Campus Road, Ithaca, NY, 14853, USA.
| |
Collapse
|