1
|
Rosenström AHC, Ahmed AS, Farinotti AB, Kultima K, Berg S, Bjurström MF, Svensson CI, Kosek E. CCL25 in the cerebrospinal fluid is negatively correlated with fatigue in chronic pain patients. Brain Behav Immun 2025; 128:54-64. [PMID: 40158641 DOI: 10.1016/j.bbi.2025.03.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 03/22/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND AND AIM Chronic pain is often accompanied by other symptoms such as fatigue and sleep disturbance, and these symptoms all correlate with neuroimmune activation. However, their relation to one another on a neuroimmune axis remains elusive. Based on a recent review, cytokines in the cerebrospinal fluid (CSF) seem to be generally upregulated in patients with chronic pain compared to controls, disregarding pain type. Some of these have the possilibity of altering blood-brain barrier (BBB) permeability. Therefore, cytokine levels in serum and CSF, as well as BBB permeability, were measured in a cohort of patients suffering from either degenerative disc disease (DDD), lumbar disc herniation (LDH) or osteoarthritis (OA). In this exploratory study, we were interested in whether cytokines in the serum or CSF are associated with sleep disturbance or fatigue, with special consideration of the effect of BBB permeability, and whether functional clusters can be found among these cytokines. METHOD One-hundred-twenty patients with DDD/LDH/OA, all awaiting surgery, were included. Blood and CSF were collected on the day of surgery. Pain was measured with a visual analog scale 0-100 mm, sleep disturbance was assessed using Pittsburgh Sleep Quality Index (PSQI), and fatigue was measured using the Multidimensional Fatigue Inventory (MFI). A 92-protein multiplex panel (OLINK, Sweden) was used to analyze cytokine expression in serum and CSF, respectively. CSF-serum albumin quotient was measured using ELISA. Non-parametric statistics were used for univariate analyses, and a false discovery rate (FDR) < 0.10 was considered statistically significant. Bonferroni correction was applied to all multivariable protein analyses to obtain conservative effect estimates. MAIN RESULTS There was an association between BBB permeability and serum-CSF dynamics: thirty-one cytokines showed significant CSF-serum correlation, and BBB permeability was significantly correlated to the quotients of 35 cytokines and to the CSF levels of 11 cytokines. Several cytokines were negatively correlated to both pain at rest and general fatigue. No correlations were found between sleep disturbance and cytokines. Network analyses of serum and CSF cytokines that were correlated with fatigue revealed functional clusters in both compartments. Anxiety, depression, and pain during rest were important regressors for sleep disturbance with an R2 = 0.41. In addition to depression and pain during rest, CSF levels of CCL25 was a significant regressor regarding general fatigue, with an R2 = 0.47. DISCUSSION AND CONCLUSION In this exploratory study of immune profiles in chronic pain cohorts awaiting surgery, the importance of BBB dynamics on serum-CSF cytokine dynamics, and to a lesser extent on central levels of cytokines, is highlighted. Surprisingly, there were no associations between any cytokines in serum or CSF and sleep disturbance, despite a high prevalence of clinically significantly disturbed sleep. In contrast, several associations between general fatigue and cytokine levels in both serum and CSF were found. Cytokines of note are CXCL11 and CCL25 in the CSF, especially because of their direct functional association. CXCL11 has been found to exert neuroprotective effects in animal models, while CCL25 is known as a proinflammatory cytokine and is the only cytokine to fall out as a significant negative contributor to degree of fatigue. While causality cannot be addressed, the negative correlation between CCL25 and fatigue in both univariate and multivariate analyses implies that neuroimmune activity might have an ameliorating effect on the degree of fatigue. This study further adds to existing evidence that centrally acting cytokines are associated with severity of symptomatology, and highlights that pain and fatigue seem to have slightly different cytokine profiles. Sleep disturbance needs to be further addressed, ideally using both subjective and objective assessment methods. CCL25 and CXCL11 are interesting biomarkers for future research on pain and associated symptoms.
Collapse
Affiliation(s)
- Alexander H C Rosenström
- Department of Surgical Sciences, Clinical Pain Research, Uppsala University, Akademiska sjukhuset, ingång 70, 1tr, 751 85 Uppsala, Sweden.
| | - Aisha Siddiqah Ahmed
- Department of Molecular Medicine and Surgery, Karolinska Institute, Karolinska University Hospital, Solna (L8:03), 171 76 Stockholm, Sweden.
| | - Alex Bersellini Farinotti
- Department of Physiology and Pharmacology, Karolinska Institute, Karolinska Institutet, Dept. of Physiology and Pharmacology, 171 77 Stockholm, Sweden.
| | - Kim Kultima
- Department of Physiology and Pharmacology, Karolinska Institute, Karolinska Institutet, Dept. of Physiology and Pharmacology, 171 77 Stockholm, Sweden; Department of Medical Sciences, Uppsala University, Akademiska sjukhuset, Ingång 40, 5 tr, 751 85 Uppsala, Sweden.
| | - Svante Berg
- Department of Molecular Medicine and Surgery, Karolinska Institute, Karolinska University Hospital, Solna (L8:03), 171 76 Stockholm, Sweden
| | - Martin F Bjurström
- Department of Surgical Sciences, Clinical Pain Research, Uppsala University, Akademiska sjukhuset, ingång 70, 1tr, 751 85 Uppsala, Sweden.
| | - Camilla I Svensson
- Department of Physiology and Pharmacology, Karolinska Institute, Karolinska Institutet, Dept. of Physiology and Pharmacology, 171 77 Stockholm, Sweden.
| | - Eva Kosek
- Department of Surgical Sciences, Clinical Pain Research, Uppsala University, Akademiska sjukhuset, ingång 70, 1tr, 751 85 Uppsala, Sweden; Department of Clinical Neuroscience, Karolinska Institute, Karolinska Institutet, Nobels väg 9, 171 77 Stockholm, Sweden.
| |
Collapse
|
2
|
Niu M, Pan J, Wang K, Zhang L, Lin Z, Zhang F. Circulating Inflammatory Factors and Bidirectional Mendelian Randomization Analysis in Patients with Kawasaki Disease. Vasc Health Risk Manag 2025; 21:99-108. [PMID: 40092571 PMCID: PMC11910938 DOI: 10.2147/vhrm.s509753] [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: 12/02/2024] [Accepted: 03/02/2025] [Indexed: 03/19/2025] Open
Abstract
Background Kawasaki disease (KD), also known as mucocutaneous lymph node syndrome, is a systemic immune vasculitis with an unclear etiology. It is often complicated by coronary artery disease. This study uses bidirectional Mendelian randomization (MR) to investigate the interaction between KD and circulating inflammatory factors, providing insights into their causal relationships. Methods We conducted a two-way pooled MR analysis to examine the causal links between 41 circulating inflammatory regulators and the risk of KD. Genetic data related to inflammation were sourced from three genome-wide association studies (GWASs) involving CRP, PCT, and cytokines, while KD data were derived from other studies. Inverse-variance weighting (IVW) was the primary MR method, with sensitivity analyses performed using MR‒Egger, weighted median, weighted mode, and MR-PRESSO to ensure robustness. Results Forward MR analyses showed no significant relationship between inflammatory factors and KD outcomes. In contrast, reverse MR, with KD as the exposure factor, revealed that interleukin-2 (IL-2) and interleukin-8 (IL-8) were significantly associated with KD (IL-2: OR=1.0085, P=0.037; IL-8: OR=1.0099, P=0.014). Borderline significant associations were observed for factors such as B_NGF, EOTAXIN, HGF, and IL_12_P70 in MR‒Egger and weighted median analyses. Conclusion This bidirectional MR study highlights the role of circulating inflammatory modulators in KD risk, offering insights into KD pathogenesis and potential therapeutic targets.
Collapse
Affiliation(s)
- Muqing Niu
- The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang, People’s Republic of China
| | - Jinyong Pan
- Department of Pediatrics, The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang, People’s Republic of China
- Clinical Medical Research Center for Children’s Diseases in the Xinjiang Production and Construction Corps, Shihezi, Xinjiang, People’s Republic of China
| | - Kui Wang
- Shihezi University School of Medicine, Shihezi, Xinjiang, People’s Republic of China
| | - Li Zhang
- Department of Pediatrics, The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang, People’s Republic of China
| | - Zhaotang Lin
- Department of Pediatrics, The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang, People’s Republic of China
| | - Fengling Zhang
- The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang, People’s Republic of China
- Department of Pediatrics, The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang, People’s Republic of China
| |
Collapse
|
3
|
Soffritti I, Gravelsina S, D'Accolti M, Bini F, Mazziga E, Vilmane A, Rasa-Dzelzkaleja S, Nora-Krukle Z, Krumina A, Murovska M, Caselli E. Circulating miRNAs Expression in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. Int J Mol Sci 2023; 24:10582. [PMID: 37445763 DOI: 10.3390/ijms241310582] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/15/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a complex multifactorial disease that causes increasing morbidity worldwide, and many individuals with ME/CFS symptoms remain undiagnosed due to the lack of diagnostic biomarkers. Its etiology is still unknown, but increasing evidence supports a role of herpesviruses (including HHV-6A and HHV-6B) as potential triggers. Interestingly, the infection by these viruses has been reported to impact the expression of microRNAs (miRNAs), short non-coding RNA sequences which have been suggested to be epigenetic factors modulating ME/CFS pathogenic mechanisms. Notably, the presence of circulating miRNAs in plasma has raised the possibility to use them as valuable biomarkers for distinguishing ME/CFS patients from healthy controls. Thus, this study aimed at determining the role of eight miRNAs, which were selected for their previous association with ME/CFS, as potential circulating biomarkers of the disease. Their presence was quantitatively evaluated in plasma from 40 ME/CFS patients and 20 healthy controls by specific Taqman assays, and the results showed that six out of the eight of the selected miRNAs were differently expressed in patients compared to controls; more specifically, five miRNAs were significantly upregulated (miR-127-3p, miR-142-5p, miR-143-3p, miR-150-5p, and miR-448), and one was downmodulated (miR-140-5p). MiRNA levels directly correlated with disease severity, whereas no significant correlations were observed with the plasma levels of seven pro-inflammatory cytokines or with the presence/load of HHV-6A/6B genome, as judged by specific PCR amplification. The results may open the way for further validation of miRNAs as new potential biomarkers in ME/CFS and increase the knowledge of the complex pathways involved in the ME/CFS development.
Collapse
Affiliation(s)
- Irene Soffritti
- Department of Chemical, Pharmaceutical and Agricultural Sciences, and LTTA, University of Ferrara, 44121 Ferrara, Italy
| | - Sabine Gravelsina
- Institute of Microbiology and Virology, Rīga Stradiņš University, LV-1067 Riga, Latvia
| | - Maria D'Accolti
- Department of Chemical, Pharmaceutical and Agricultural Sciences, and LTTA, University of Ferrara, 44121 Ferrara, Italy
| | - Francesca Bini
- Department of Chemical, Pharmaceutical and Agricultural Sciences, and LTTA, University of Ferrara, 44121 Ferrara, Italy
| | - Eleonora Mazziga
- Department of Chemical, Pharmaceutical and Agricultural Sciences, and LTTA, University of Ferrara, 44121 Ferrara, Italy
| | - Anda Vilmane
- Institute of Microbiology and Virology, Rīga Stradiņš University, LV-1067 Riga, Latvia
| | | | - Zaiga Nora-Krukle
- Institute of Microbiology and Virology, Rīga Stradiņš University, LV-1067 Riga, Latvia
| | - Angelika Krumina
- Faculty of Medicine, Department of Infectology, Rīga Stradiņš University, LV-1006 Riga, Latvia
| | - Modra Murovska
- Institute of Microbiology and Virology, Rīga Stradiņš University, LV-1067 Riga, Latvia
| | - Elisabetta Caselli
- Department of Chemical, Pharmaceutical and Agricultural Sciences, and LTTA, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
4
|
Giloteaux L, Li J, Hornig M, Lipkin WI, Ruppert D, Hanson MR. Proteomics and cytokine analyses distinguish myalgic encephalomyelitis/chronic fatigue syndrome cases from controls. J Transl Med 2023; 21:322. [PMID: 37179299 PMCID: PMC10182359 DOI: 10.1186/s12967-023-04179-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
BACKGROUND Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a complex, heterogenous disease characterized by unexplained persistent fatigue and other features including cognitive impairment, myalgias, post-exertional malaise, and immune system dysfunction. Cytokines are present in plasma and encapsulated in extracellular vesicles (EVs), but there have been only a few reports of EV characteristics and cargo in ME/CFS. Several small studies have previously described plasma proteins or protein pathways that are associated with ME/CFS. METHODS We prepared extracellular vesicles (EVs) from frozen plasma samples from a cohort of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) cases and controls with prior published plasma cytokine and plasma proteomics data. The cytokine content of the plasma-derived extracellular vesicles was determined by a multiplex assay and differences between patients and controls were assessed. We then performed multi-omic statistical analyses that considered not only this new data, but extensive clinical data describing the health of the subjects. RESULTS ME/CFS cases exhibited greater size and concentration of EVs in plasma. Assays of cytokine content in EVs revealed IL2 was significantly higher in cases. We observed numerous correlations among EV cytokines, among plasma cytokines, and among plasma proteins from mass spectrometry proteomics. Significant correlations between clinical data and protein levels suggest roles of particular proteins and pathways in the disease. For example, higher levels of the pro-inflammatory cytokines Granulocyte-Monocyte Colony-Stimulating Factor (CSF2) and Tumor Necrosis Factor (TNFα) were correlated with greater physical and fatigue symptoms in ME/CFS cases. Higher serine protease SERPINA5, which is involved in hemostasis, was correlated with higher SF-36 general health scores in ME/CFS. Machine learning classifiers were able to identify a list of 20 proteins that could discriminate between cases and controls, with XGBoost providing the best classification with 86.1% accuracy and a cross-validated AUROC value of 0.947. Random Forest distinguished cases from controls with 79.1% accuracy and an AUROC value of 0.891 using only 7 proteins. CONCLUSIONS These findings add to the substantial number of objective differences in biomolecules that have been identified in individuals with ME/CFS. The observed correlations of proteins important in immune responses and hemostasis with clinical data further implicates a disturbance of these functions 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
| | - Jiayin Li
- Department of Statistics and Data Science, Cornell University, Ithaca, NY, USA
| | - Mady Hornig
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, NY, USA
| | - W Ian Lipkin
- Center for Infection and Immunity, Columbia University Mailman School of Public Health, New York, NY, USA
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, NY, USA
- Departments of Neurology and Pathology, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - David Ruppert
- Department of Statistics and Data Science, Cornell University, Ithaca, NY, USA
- School of Operations Research and Information Engineering, Cornell University, Ithaca, NY, USA
| | - Maureen R Hanson
- Department of Molecular Biology and Genetics, Cornell University, 323 Biotechnology Building, 526 Campus Road, Ithaca, NY, 14853, USA.
| |
Collapse
|
5
|
LI BB, FENG CW, QU YY, SUN ZR, CHEN T, WANG YL, WANG QY, LU J, SHAO YY, YANG TS. Research progress on central mechanism of acupuncture treatment for chronic fatigue syndrome 针刺调控慢性疲劳综合征中枢机制研究进展. WORLD JOURNAL OF ACUPUNCTURE-MOXIBUSTION 2023. [DOI: 10.1016/j.wjam.2023.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
|
6
|
Dolcetti E, Bruno A, Azzolini F, Gilio L, Pavone L, Iezzi E, Galifi G, Gambardella S, Ferese R, Buttari F, De Vito F, Colantuono P, Furlan R, Finardi A, Musella A, Mandolesi G, Centonze D, Stampanoni Bassi M. Genetic regulation of IL-8 influences disease presentation of multiple sclerosis. Mult Scler 2023; 29:512-520. [PMID: 36803228 DOI: 10.1177/13524585231155049] [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] [Indexed: 02/22/2023]
Abstract
BACKGROUND Individual genetic variability may influence the course of multiple sclerosis (MS). The interleukin (IL)-8C>T rs2227306 single nucleotide polymorphism (SNP) regulates IL-8 activity in other clinical conditions; however, its role in MS has never been investigated. OBJECTIVES To explore the association between IL-8 SNP rs2227306, cerebrospinal fluid (CSF) IL-8 concentrations, clinical, and radiological characteristics in a group of newly diagnosed MS patients. METHODS In 141 relapsing-remitting (RR)-MS patients, rs2227306 polymorphism, CSF levels of IL-8, clinical, and demographical characteristics were determined. In 50 patients, structural magnetic resonance imaging (MRI) measures were also assessed. RESULTS An association between CSF IL-8 and Expanded Disability Status Scale (EDSS) at diagnosis was found in our set of patients (r = 0.207, p = 0.014). CSF IL-8 concentrations were significantly higher in patients carrying the T variant of rs2227306 (p = 0.004). In the same group, a positive correlation emerged between IL-8 and EDSS (r = 0.273, p = 0.019). Finally, a negative correlation between CSF levels of IL-8 and cortical thickness emerged in rs2227306T carriers (r = -0.498, p = 0.005). CONCLUSION We describe for the first time a role of SNP rs2227306 of IL-8 gene in regulating the expression and the activity of this inflammatory cytokine in MS.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Stefano Gambardella
- IRCSS Neuromed, Pozzilli, Italy/Department of Biomolecular Sciences, University of Urbino "Carlo Bo," Urbino, Italy
| | | | | | | | - Paola Colantuono
- Department of Medicine and Health Sciences "Vincenzo Tiberio," Unimol, Campobasso, Italy
| | - Roberto Furlan
- Clinical Neuroimmunology Unit, Institute of Experimental Neurology (INSpe), Division of Neuroscience, San Raffaele Hospital, Milan, Italy
| | - Annamaria Finardi
- Clinical Neuroimmunology Unit, Institute of Experimental Neurology (INSpe), Division of Neuroscience, San Raffaele Hospital, Milan, Italy
| | - Alessandra Musella
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Roma, Italy/Department of Human Sciences and Quality of Life Promotion, University of Rome San Raffaele, Rome, Italy
| | - Georgia Mandolesi
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Roma, Italy/Department of Human Sciences and Quality of Life Promotion, University of Rome San Raffaele, Rome, Italy
| | - Diego Centonze
- IRCSS Neuromed, Pozzilli, Italy/Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | | |
Collapse
|
7
|
Gottschalk CG, Peterson D, Armstrong J, Knox K, Roy A. Potential molecular mechanisms of chronic fatigue in long haul COVID and other viral diseases. Infect Agent Cancer 2023; 18:7. [PMID: 36750846 PMCID: PMC9902840 DOI: 10.1186/s13027-023-00485-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 02/01/2023] [Indexed: 02/09/2023] Open
Abstract
Historically, COVID-19 emerges as one of the most devastating diseases of humankind, which creates an unmanageable health crisis worldwide. Until now, this disease costs millions of lives and continues to paralyze human civilization's economy and social growth, leaving an enduring damage that will take an exceptionally long time to repair. While a majority of infected patients survive after mild to moderate reactions after two to six weeks, a growing population of patients suffers for months with severe and prolonged symptoms of fatigue, depression, and anxiety. These patients are no less than 10% of total COVID-19 infected individuals with distinctive chronic clinical symptomatology, collectively termed post-acute sequelae of COVID-19 (PASC) or more commonly long-haul COVID. Interestingly, Long-haul COVID and many debilitating viral diseases display a similar range of clinical symptoms of muscle fatigue, dizziness, depression, and chronic inflammation. In our current hypothesis-driven review article, we attempt to discuss the molecular mechanism of muscle fatigue in long-haul COVID, and other viral diseases as caused by HHV6, Powassan, Epstein-Barr virus (EBV), and HIV. We also discuss the pathological resemblance of virus-triggered muscle fatigue with myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS).
Collapse
Affiliation(s)
- Carl Gunnar Gottschalk
- Simmaron Research INC, 948 Incline Way, Incline Village, NV 89451 USA ,grid.267468.90000 0001 0695 7223Research and Development Laboratory, Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI 53211 USA ,Coppe Laboratories, W229N1870 Westwood Dr, Waukesha, WI 53186 USA
| | - Daniel Peterson
- Simmaron Research INC, 948 Incline Way, Incline Village, NV 89451 USA ,Coppe Laboratories, W229N1870 Westwood Dr, Waukesha, WI 53186 USA
| | - Jan Armstrong
- Simmaron Research INC, 948 Incline Way, Incline Village, NV 89451 USA ,Coppe Laboratories, W229N1870 Westwood Dr, Waukesha, WI 53186 USA
| | - Konstance Knox
- grid.267468.90000 0001 0695 7223Research and Development Laboratory, Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI 53211 USA ,Coppe Laboratories, W229N1870 Westwood Dr, Waukesha, WI 53186 USA
| | - Avik Roy
- Simmaron Research INC, 948 Incline Way, Incline Village, NV, 89451, USA. .,Research and Development Laboratory, Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI, 53211, USA. .,Coppe Laboratories, W229N1870 Westwood Dr, Waukesha, WI, 53186, USA.
| |
Collapse
|
8
|
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
|
9
|
Rasa-Dzelzkaleja S, Krumina A, Capenko S, Nora-Krukle Z, Gravelsina S, Vilmane A, Ievina L, Shoenfeld Y, Murovska M. The persistent viral infections in the development and severity of myalgic encephalomyelitis/chronic fatigue syndrome. J Transl Med 2023; 21:33. [PMID: 36653846 PMCID: PMC9847171 DOI: 10.1186/s12967-023-03887-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 01/11/2023] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a multifactorial disease with an unexplained aetiology in which viral infections are possible trigger factors. The aim of this study was to determine the involvement of human herpesvirus (HHV)-6A/B, HHV-7, and parvovirus B19 (B19V) in the etiopathogenesis of ME/CFS. METHODS 200 patients with clinically diagnosed ME/CFS and 150 apparently healthy individuals were enrolled in this study. Single-round, nested, and quantitative real-time polymerase chain reactions (PCR) were used to detect the presence and load of HHV-6A/B, HHV-7, and B19V. HHV-6A and HHV-6B were distinguished by PCR and restriction analysis. Immunoenzymatic assays were applied to estimate the presence of virus-specific antibodies and the level of cytokines. RESULTS HHV-6A/B, HHV-7, and B19V specific antibodies were detected among patients and healthy individuals in 92.1% and 76.7%, 84.6% and 93.8%, and 78% and 67.4% of cases. HHV-6B had 99% of HHV-6 positive patients. Latent HHV-6A/B, HHV-7, and B19V infection/co-infection was observed in 51.5% of the patients and 76.7% of the healthy individuals, whereas active-45% of the ME/CFS patients and 8.7% of healthy individuals. HHV-6A/B load in patients with a persistent infection/co-infection in a latent and active phase was 262 and 653.2 copies/106 cells, whereas HHV-7 load was 166.5 and 248.5 copies/106 cells, and B19V-96.8 and 250.8 copies/106 cells, respectively. ME/CFS patients with persistent infection in an active phase had a higher level of pro-inflammatory cytokines (interleukin(IL)-6, tumor necrosis factor-alpha(TNF-α) and IL-12) and anti-inflammatory (IL-10) than with a persistent infection in a latent phase. A significant difference was revealed in the levels of TNF-α, IL-12, and IL-10 among the patient groups without infection, with latent infection/co-infection, active single, double and triple co-infection. The levels of TNF-α, IL-12, and IL-10 are significantly higher in patients with severe compared with a moderate course of ME/CFS. CONCLUSIONS Significantly more persistent HHV-6A/B, HHV-7, and B19V infection/co-infection in an active phase with a higher viral load and elevated levels of pro- and anti-inflammatory cytokines among patients with ME/CFS than healthy individuals indicate the importance of these infections/co-infections in ME/CFS development. The presence of these infections/co-infections influences the ME/CFS clinical course severity.
Collapse
Affiliation(s)
- Santa Rasa-Dzelzkaleja
- grid.17330.360000 0001 2173 9398Institute of Microbiology and Virology, Rīga Stradiņš University, Riga, Latvia
| | - Angelika Krumina
- grid.17330.360000 0001 2173 9398Faculty of Medicine, Department of Infectology, Rīga Stradiņš University, Riga, Latvia
| | - Svetlana Capenko
- grid.17330.360000 0001 2173 9398Institute of Microbiology and Virology, Rīga Stradiņš University, Riga, Latvia
| | - Zaiga Nora-Krukle
- grid.17330.360000 0001 2173 9398Institute of Microbiology and Virology, Rīga Stradiņš University, Riga, Latvia
| | - Sabine Gravelsina
- grid.17330.360000 0001 2173 9398Institute of Microbiology and Virology, Rīga Stradiņš University, Riga, Latvia
| | - Anda Vilmane
- grid.17330.360000 0001 2173 9398Institute of Microbiology and Virology, Rīga Stradiņš University, Riga, Latvia
| | - Lauma Ievina
- grid.17330.360000 0001 2173 9398Institute of Microbiology and Virology, Rīga Stradiņš University, Riga, Latvia
| | - Yehuda Shoenfeld
- grid.413795.d0000 0001 2107 2845Zabludowicz Center for Autoimmune Diseases, Sheba Medical Centre, Ramat Gan, Israel
| | - Modra Murovska
- grid.17330.360000 0001 2173 9398Institute of Microbiology and Virology, Rīga Stradiņš University, Riga, Latvia
| | | |
Collapse
|
10
|
Jegal KH, Yoon J, Kim S, Jang S, Jin YH, Lee JH, Choi SM, Kim TH, Kwon S. Herbal Medicines for Post-Acute Sequelae (Fatigue or Cognitive Dysfunction) of SARS-CoV-2 Infection: A Phase 2 Pilot Clinical Study Protocol. Healthcare (Basel) 2022; 10:healthcare10101839. [PMID: 36292286 PMCID: PMC9601660 DOI: 10.3390/healthcare10101839] [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: 08/10/2022] [Revised: 09/13/2022] [Accepted: 09/18/2022] [Indexed: 11/19/2022] Open
Abstract
Long-term sequelae refer to persistent symptoms or signs for >6 months after SARS-CoV-2 infection. The most common symptoms of sequelae are fatigue and neuropsychiatric symptoms (concentration difficulty, amnesia, cognitive dysfunction, anxiety, and depression). However, approved treatments have not been fully established. Herbal medicines are administered for 12 weeks to patients who continuously complain of fatigue or cognitive dysfunction for >4 weeks that only occurred after COVID-19 diagnoses. Based on the Korean Medicine syndrome differentiation diagnosis, patients with fatigue will be administered Bojungikgi-tang or Kyungok-go, whereas those with cognitive dysfunction will be administered Cheonwangbosim-dan. Results could support evidence that herbal medicines may mitigate fatigue and cognitive dysfunction caused by COVID-19. Furthermore, by investigating the effects of herbal medicines on changes in metabolite and immune response due to COVID-19, which may be responsible for sequelae, the potential of herbal medicines as one of the therapeutic interventions for post-acute sequelae of SARS-CoV-2 infection can be evaluated. Therefore, the effects of herbal medicine on fatigue and cognitive dysfunction sequelae due to COVID-19 will be elucidated in this study to provide an insight into the preparation of medical management for the post-acute sequelae of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Kyung Hwan Jegal
- Digital Health Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Korea
- College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Korea
| | - Jiwon Yoon
- Korean Medicine Data Division, Korea Institute of Oriental Medicine, Daejeon 34054, Korea
| | - Sanghyun Kim
- Korean Medicine Data Division, Korea Institute of Oriental Medicine, Daejeon 34054, Korea
| | - Soobin Jang
- College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Korea
| | - Young-Hee Jin
- Korean Medicine Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Korea
| | - Jun-Hwan Lee
- Korean Medicine Science Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Korea
- UST KIOM School, University of Science and Technology (UST), Daejeon 34113, Korea
| | - Sun-Mi Choi
- Korean Medicine Data Division, Korea Institute of Oriental Medicine, Daejeon 34054, Korea
- UST KIOM School, University of Science and Technology (UST), Daejeon 34113, Korea
| | - Tae Hun Kim
- Korean Medicine Clinical Trial Center, Korean Medicine Hospital, Kyung Hee University, Seoul 02447, Korea
- Correspondence: (T.H.K.); (S.K.); Tel.: +82-2-958-9088 (T.H.K.); +82-42-868-9675 (S.K.)
| | - Sunoh Kwon
- UST KIOM School, University of Science and Technology (UST), Daejeon 34113, Korea
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Korea
- Correspondence: (T.H.K.); (S.K.); Tel.: +82-2-958-9088 (T.H.K.); +82-42-868-9675 (S.K.)
| |
Collapse
|
11
|
Fatigue in Multiple Sclerosis Is Associated with Reduced Expression of Interleukin-10 and Worse Prospective Disease Activity. Biomedicines 2022; 10:biomedicines10092058. [PMID: 36140159 PMCID: PMC9495727 DOI: 10.3390/biomedicines10092058] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 08/03/2022] [Accepted: 08/08/2022] [Indexed: 11/21/2022] Open
Abstract
In multiple sclerosis (MS), fatigue is a frequent symptom that negatively affects quality of life. The pathogenesis of fatigue is multifactorial and inflammation may play a specific role. To explore the association between fatigue, central inflammation and disease course in MS in 106 relapsing-remitting (RR)-MS patients, clinical characteristics, including fatigue and mood, were explored at the time of diagnosis. NEDA (no evidence of disease activity)-3 status after one-year follow up was calculated. Cerebrospinal fluid (CSF) levels of a set of proinflammatory and anti-inflammatory molecules and peripheral blood markers of inflammation were also analyzed. MRI structural measures were explored in 35 patients. A significant negative correlation was found at diagnosis between fatigue measured with the Modified Fatigue Impact Scale (MFIS) and the CSF levels of interleukin (IL)-10. Conversely, no significant associations were found with peripheral markers of inflammation. Higher MFIS scores were associated with reduced probability to reach NEDA-3 status after 1-year follow up. Finally, T2 lesion load showed a positive correlation with MFIS scores and a negative correlation with CSF IL-10 levels at diagnosis. CSF inflammation, and particularly the reduced expression of the anti-inflammatory molecule IL-10, may exacerbate fatigue. Fatigue in MS may reflect subclinical CSF inflammation, predisposing to greater disease activity.
Collapse
|
12
|
Filippi M, Krähenmann R, Fissler P. The Link Between Energy-Related Sensations and Metabolism: Implications for Treating Fatigue. Front Psychol 2022; 13:920556. [PMID: 35800955 PMCID: PMC9255916 DOI: 10.3389/fpsyg.2022.920556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Energy-related sensations include sensation of energy and fatigue as well as subjective energizability and fatigability. First, we introduce interdisciplinary useful definitions of all constructs and review findings regarding the question of whether sensations of fatigue and energy are two separate constructs or two ends of a single dimension. Second, we describe different components of the bodily energy metabolism system (e.g., mitochondria; autonomic nervous system). Third, we review the link between sensation of fatigue and different components of energy metabolism. Finally, we present an overview of different treatments shown to affect both energy-related sensations and metabolism before outlining future research perspectives.
Collapse
Affiliation(s)
- Marco Filippi
- Psychiatric Services Thurgau, Münsterlingen, Switzerland
- University Hospital for Psychiatry and Psychotherapy, Paracelsus Medical University, Salzburg, Austria
| | - Rainer Krähenmann
- Psychiatric Services Thurgau, Münsterlingen, Switzerland
- University Hospital for Psychiatry and Psychotherapy, Paracelsus Medical University, Salzburg, Austria
- Department of Psychiatry, Psychotherapy and Psychosomatics, University of Zürich, Zürich, Switzerland
- *Correspondence: Rainer Krähenmann,
| | - Patrick Fissler
- Psychiatric Services Thurgau, Münsterlingen, Switzerland
- University Hospital for Psychiatry and Psychotherapy, Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
13
|
Renz-Polster H, Tremblay ME, Bienzle D, Fischer JE. The Pathobiology of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: The Case for Neuroglial Failure. Front Cell Neurosci 2022; 16:888232. [PMID: 35614970 PMCID: PMC9124899 DOI: 10.3389/fncel.2022.888232] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/13/2022] [Indexed: 12/20/2022] Open
Abstract
Although myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) has a specific and distinctive profile of clinical features, the disease remains an enigma because causal explanation of the pathobiological matrix is lacking. Several potential disease mechanisms have been identified, including immune abnormalities, inflammatory activation, mitochondrial alterations, endothelial and muscular disturbances, cardiovascular anomalies, and dysfunction of the peripheral and central nervous systems. Yet, it remains unclear whether and how these pathways may be related and orchestrated. Here we explore the hypothesis that a common denominator of the pathobiological processes in ME/CFS may be central nervous system dysfunction due to impaired or pathologically reactive neuroglia (astrocytes, microglia and oligodendrocytes). We will test this hypothesis by reviewing, in reference to the current literature, the two most salient and widely accepted features of ME/CFS, and by investigating how these might be linked to dysfunctional neuroglia. From this review we conclude that the multifaceted pathobiology of ME/CFS may be attributable in a unifying manner to neuroglial dysfunction. Because the two key features - post exertional malaise and decreased cerebral blood flow - are also recognized in a subset of patients with post-acute sequelae COVID, we suggest that our findings may also be pertinent to this entity.
Collapse
Affiliation(s)
- Herbert Renz-Polster
- Division of General Medicine, Center for Preventive Medicine and Digital Health Baden-Württemberg (CPD-BW), University Medicine Mannheim, Heidelberg University, Mannheim, Germany
| | - Marie-Eve Tremblay
- Axe Neurosciences, Centre de recherche du CHU de Québec, Université Laval, Quebec, QC, Canada
- Département de Médecine Moléculaire, Université Laval, Quebec, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Center for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Dorothee Bienzle
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Joachim E. Fischer
- Division of General Medicine, Center for Preventive Medicine and Digital Health Baden-Württemberg (CPD-BW), University Medicine Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
14
|
Zhu Z, Zhao X, OuYang Q, Wang Y, Xiong Y, Cong S, Zhou M, Zhang M, Luo X, Cheng M. Waterfall Forest Environment Regulates Chronic Stress via the NOX4/ROS/NF-κB Signaling Pathway. Front Neurol 2021; 12:619728. [PMID: 33868142 PMCID: PMC8044934 DOI: 10.3389/fneur.2021.619728] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 01/25/2021] [Indexed: 01/25/2023] Open
Abstract
Background: Forest therapy has been proven to have beneficial effects on people with depression and anxiety. However, it remains unknown whether the waterfall forest environment (WF) affects the physical and psychological health of patients with chronic fatigue and how the WF regulates chronic stress. Methods: Twenty-four patients with chronic fatigue were randomly divided into two groups: the WF group and the urban (U) group. Scores on the Hamilton Anxiety Scale (HAMA), Hamilton Depression Scale (HAMD), and Fatigue Scale-14 (FS-14) were evaluated before and after environmental intervention. Detection of physiological indexes and inflammatory factor levels and immunological analysis were also performed. In addition, the chronic stress rat model was constructed, and the effects of the WF on hopelessness and liver damage of rats were investigated. Results: Patients with chronic fatigue in the WF group showed a significant decrease in FS-14, HAMA, and HAMD scores compared with the U group. The expression levels of glutathione peroxidase and superoxide dismutase were remarkably higher in the WF group than in the U group. However, the expression levels of malondialdehyde and inflammatory factors (IL-1β, TNF-α, IL-6, and IL-10) were remarkably decreased after the intervention of the WF. In addition, animal experiments confirmed that the WF improved hopelessness, liver damage, and excitability of neurons of chronic stress rats. Mechanistically, the WF reduced the liver damage caused by chronic stress in rats by inhibiting the NOX4/ROS/NF-κB signaling pathway. Conclusions: Collectively, the WF had a positive effect on immune enhancement and physical and psychological health in patients with chronic fatigue and might inhibit chronic stress by regulating the NOX4/ROS/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Zixin Zhu
- Department of Pathophysiology, Guizhou Medical University, Guiyang, China
- Department of Infectious Diseases, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xueke Zhao
- Department of Infectious Diseases, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Qiuyue OuYang
- Department of Infectious Diseases, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yinghui Wang
- Department of Clinical Laboratory, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yan Xiong
- Department of Infectious Diseases, Guizhou Provincial People's Hospital, Guiyang, China
| | - Shuo Cong
- Department of Infectious Diseases, Guizhou Provincial People's Hospital, Guiyang, China
| | - Mingyu Zhou
- Department of Infectious Diseases, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Manman Zhang
- Department of Gastroenterology, Guizhou Provincial People's Hospital, Guiyang, China
| | - Xinhua Luo
- Department of Infectious Diseases, Guizhou Provincial People's Hospital, Guiyang, China
| | - Mingliang Cheng
- Department of Infectious Diseases, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
15
|
Mueller C, Lin JC, Sheriff S, Maudsley AA, Younger JW. Evidence of widespread metabolite abnormalities in Myalgic encephalomyelitis/chronic fatigue syndrome: assessment with whole-brain magnetic resonance spectroscopy. Brain Imaging Behav 2021; 14:562-572. [PMID: 30617782 DOI: 10.1007/s11682-018-0029-4] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Previous neuroimaging studies have detected markers of neuroinflammation in patients with Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS). Magnetic Resonance Spectroscopy (MRS) is suitable for measuring brain metabolites linked to inflammation, but has only been applied to discrete regions of interest in ME/CFS. We extended the MRS analysis of ME/CFS by capturing multi-voxel information across the entire brain. Additionally, we tested whether MRS-derived brain temperature is elevated in ME/CFS patients. Fifteen women with ME/CFS and 15 age- and gender-matched healthy controls completed fatigue and mood symptom questionnaires and whole-brain echo-planar spectroscopic imaging (EPSI). Choline (CHO), myo-inositol (MI), lactate (LAC), and N-acetylaspartate (NAA) were quantified in 47 regions, expressed as ratios over creatine (CR), and compared between ME/CFS patients and controls using independent-samples t-tests. Brain temperature was similarly tested between groups. Significant between-group differences were detected in several regions, most notably elevated CHO/CR in the left anterior cingulate (p < 0.001). Metabolite ratios in seven regions were correlated with fatigue (p < 0.05). ME/CFS patients had increased temperature in the right insula, putamen, frontal cortex, thalamus, and the cerebellum (all p < 0.05), which was not attributable to increased body temperature or differences in cerebral perfusion. Brain temperature increases converged with elevated LAC/CR in the right insula, right thalamus, and cerebellum (all p < 0.05). We report metabolite and temperature abnormalities in ME/CFS patients in widely distributed regions. Our findings may indicate that ME/CFS involves neuroinflammation.
Collapse
Affiliation(s)
- Christina Mueller
- Department of Psychology, The University of Alabama at Birmingham, 1720 2nd Ave S, Birmingham, AL, 35294, USA
| | - Joanne C Lin
- Department of Psychology, The University of Alabama at Birmingham, 1720 2nd Ave S, Birmingham, AL, 35294, USA
| | - Sulaiman Sheriff
- Department of Radiology, Miller School of Medicine, University of Miami, 1600 NW 10th Ave #1140, Miami, FL, 33136, USA
| | - Andrew A Maudsley
- Department of Radiology, Miller School of Medicine, University of Miami, 1600 NW 10th Ave #1140, Miami, FL, 33136, USA
| | - Jarred W Younger
- Department of Psychology, The University of Alabama at Birmingham, 1720 2nd Ave S, Birmingham, AL, 35294, USA.
| |
Collapse
|
16
|
Anderson G, Maes M. Mitochondria and immunity in chronic fatigue syndrome. Prog Neuropsychopharmacol Biol Psychiatry 2020; 103:109976. [PMID: 32470498 DOI: 10.1016/j.pnpbp.2020.109976] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 05/19/2020] [Indexed: 02/07/2023]
Abstract
It is widely accepted that the pathophysiology and treatment of myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) could be considerably improved. The heterogeneity of ME/CFS and the confusion over its classification have undoubtedly contributed to this, although this would seem a consequence of the complexity of the array of ME/CFS presentations and high levels of diverse comorbidities. This article reviews the biological underpinnings of ME/CFS presentations, including the interacting roles of the gut microbiome/permeability, endogenous opioidergic system, immune cell mitochondria, autonomic nervous system, microRNA-155, viral infection/re-awakening and leptin as well as melatonin and the circadian rhythm. This details not only relevant pathophysiological processes and treatment options, but also highlights future research directions. Due to the complexity of interacting systems in ME/CFS pathophysiology, clarification as to its biological underpinnings is likely to considerably contribute to the understanding and treatment of other complex and poorly managed conditions, including fibromyalgia, depression, migraine, and dementia. The gut and immune cell mitochondria are proposed to be two important hubs that interact with the circadian rhythm in driving ME/CFS pathophysiology.
Collapse
Affiliation(s)
- G Anderson
- CRC Scotland & London, Eccleston Square, London, UK.
| | - M Maes
- Dept Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Dept Psychiatry, Medical University Plovdiv, Plovdiv, Bulgaria.; IMPACT Research Center, Deakin University, Geelong, Australia
| |
Collapse
|
17
|
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: 27] [Impact Index Per Article: 5.4] [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
|
18
|
Hildebrandt H, Stachowiak R, Heber I, Schlake HP, Eling P. Relation between cognitive fatigue and circadian or stress related cortisol levels in MS patients. Mult Scler Relat Disord 2020; 45:102440. [DOI: 10.1016/j.msard.2020.102440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 07/28/2020] [Accepted: 08/02/2020] [Indexed: 10/23/2022]
|
19
|
Missailidis D, Sanislav O, Allan CY, Annesley SJ, Fisher PR. Cell-Based Blood Biomarkers for Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. Int J Mol Sci 2020; 21:ijms21031142. [PMID: 32046336 PMCID: PMC7037777 DOI: 10.3390/ijms21031142] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/02/2020] [Accepted: 02/04/2020] [Indexed: 12/14/2022] Open
Abstract
Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a devastating illness whose biomedical basis is now beginning to be elucidated. We reported previously that, after recovery from frozen storage, lymphocytes (peripheral blood mononuclear cells, PBMCs) from ME/CFS patients die faster in culture medium than those from healthy controls. We also found that lymphoblastoid cell lines (lymphoblasts) derived from these PBMCs exhibit multiple abnormalities in mitochondrial respiratory function and signalling activity by the cellular stress-sensing kinase Target Of Rapamycin Complex 1 (TORC1). These differences were correlated with disease severity, as measured by the Richardson and Lidbury weighted standing test. The clarity of the differences between these cells derived from ME/CFS patient blood and those from healthy controls suggested that they may provide useful biomarkers for ME/CFS. Here, we report a preliminary investigation into that possibility using a variety of analytical classification tools, including linear discriminant analysis, logistic regression and receiver operating characteristic (ROC) curve analysis. We found that results from three different tests—lymphocyte death rate, mitochondrial respiratory function and TORC1 activity—could each individually serve as a biomarker with better than 90% sensitivity but only modest specificity vís a vís healthy controls. However, in combination, they provided a cell-based biomarker with sensitivity and specificity approaching 100% in our sample. This level of sensitivity and specificity was almost equalled by a suggested protocol in which the frozen lymphocyte death rate was used as a highly sensitive test to triage positive samples to the more time consuming and expensive tests measuring lymphoblast respiratory function and TORC1 activity. This protocol provides a promising biomarker that could assist in more rapid and accurate diagnosis of ME/CFS.
Collapse
|
20
|
Joung JY, Lee JS, Cho JH, Lee DS, Ahn YC, Son CG. The Efficacy and Safety of Myelophil, an Ethanol Extract Mixture of Astragali Radix and Salviae Radix, for Chronic Fatigue Syndrome: A Randomized Clinical Trial. Front Pharmacol 2019; 10:991. [PMID: 31551788 PMCID: PMC6746924 DOI: 10.3389/fphar.2019.00991] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 08/05/2019] [Indexed: 12/12/2022] Open
Abstract
Background: There is a strong demand for therapeutics to treat chronic fatigue syndrome (CFS), although there are limitations. Myelophil, which is a combination of extracts from Astragali Radix and Salviae Miltiorrhizae Radix, has been clinically used to treat fatigue-related disorders in South Korea. We conducted a randomized controlled clinical trial of Myelophil in patients with CFS and evaluated its efficacy and safety in two hospitals. Methods: We enrolled 98 participants (M: 38, F: 60) with CFS in a phase 2 trial of oral Myelophil (2 g daily) or placebo for 12 weeks. The primary end point was a change in the Chalder fatigue scale, as scored by a numeric rating scale (NRS). The secondary end points included changes in the visual analogue scale, fatigue severity scale (FSS), and 36-item short-form health survey (SF-36). Biomarkers of oxidative stress and cytokines were evaluated by blood tests. Results: Ninety-seven participants (48 in the Myelophil group and 49 in the placebo group) completed the trial. An analysis of all participants showed that Myelophil slightly improved fatigue symptoms compared with those of the placebo, but this effect was not statistically significant (p > 0.05 for the NRS, VAS, FSS, and SF-36). By contrast, an analysis of the subpopulation (53 participants, M: 24, F: 29) with severe symptoms (≥63, median NRS value of total participants) showed a statistically significant improvement in fatigue symptoms in the Myelophil group compared with the placebo (p < 0.05 for NRS, FSS, and SF-36). There were no significant changes in the biomarkers for oxidative stress and cytokines before or after the treatment. No Myelophil-related adverse response was observed during the trial. Conclusion: These results support the hypothesis that Myelophil can be a therapeutic candidate to manage CFS and provide the rationale for its progression to a phase 3 clinical trial. Clinical Trial Registration: www.ClinicalTrials.gov, identifier KCT0002317.
Collapse
Affiliation(s)
- Jin-Yong Joung
- Liver and Immunology Research Center, Oriental Medical Collage of Daejeon University, Daejeon, South Korea
| | - Jin-Seok Lee
- Liver and Immunology Research Center, Oriental Medical Collage of Daejeon University, Daejeon, South Korea
| | - Jung-Hyo Cho
- Liver and Immunology Research Center, Oriental Medical Collage of Daejeon University, Daejeon, South Korea
| | - Dong-Soo Lee
- Department of Internal Medicine, Daejeon St. Mary’s Hospital of Catholic University, Daejeon, South Korea
| | - Yo-Chan Ahn
- Department of Health Service Management, Daejeon University, Daejeon, South Korea
| | - Chang-Gue Son
- Liver and Immunology Research Center, Oriental Medical Collage of Daejeon University, Daejeon, South Korea
| |
Collapse
|
21
|
Myalgic encephalomyelitis/chronic fatigue syndrome: From pathophysiological insights to novel therapeutic opportunities. Pharmacol Res 2019; 148:104450. [PMID: 31509764 DOI: 10.1016/j.phrs.2019.104450] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 08/26/2019] [Accepted: 09/06/2019] [Indexed: 12/12/2022]
Abstract
Myalgic encephalomyelitis (ME) or chronic fatigue syndrome (CFS) is a common and disabling condition with a paucity of effective and evidence-based therapies, reflecting a major unmet need. Cognitive behavioural therapy and graded exercise are of modest benefit for only some ME/CFS patients, and many sufferers report aggravation of symptoms of fatigue with exercise. The presence of a multiplicity of pathophysiological abnormalities in at least the subgroup of people with ME/CFS diagnosed with the current international consensus "Fukuda" criteria, points to numerous potential therapeutic targets. Such abnormalities include extensive data showing that at least a subgroup has a pro-inflammatory state, increased oxidative and nitrosative stress, disruption of gut mucosal barriers and mitochondrial dysfunction together with dysregulated bioenergetics. In this paper, these pathways are summarised, and data regarding promising therapeutic options that target these pathways are highlighted; they include coenzyme Q10, melatonin, curcumin, molecular hydrogen and N-acetylcysteine. These data are promising yet preliminary, suggesting hopeful avenues to address this major unmet burden of illness.
Collapse
|
22
|
Corbitt M, Eaton-Fitch N, Staines D, Cabanas H, Marshall-Gradisnik S. A systematic review of cytokines in chronic fatigue syndrome/myalgic encephalomyelitis/systemic exertion intolerance disease (CFS/ME/SEID). BMC Neurol 2019; 19:207. [PMID: 31445522 PMCID: PMC6708220 DOI: 10.1186/s12883-019-1433-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 08/18/2019] [Indexed: 02/13/2023] Open
Abstract
Background Cytokines in Chronic Fatigue Syndrome/Myalgic Encephalomyelitis/Systemic Exertion Intolerance Disease (CFS/ME/SEID) patients compared with healthy controls have been extensively studied. However, the evidence regarding whether a baseline difference between CFS/ME/SEID patients and the normal population remains unclear. The aim of this study was to conduct a systematic review of the literature regarding cytokines in CFS/ME/SEID and whether there is a significant difference in cytokine levels between this patient group and the normal population. Methods Pubmed, Scopus, Medline (EBSCOHost), and EMBASE databases were searched to source relevant studies for CFS/ME/SEID. The review included any studies examining cytokines in CFS/ME/SEID patients compared with healthy controls. Results of the literature search were summarised according to aspects of their study design and outcome measures, namely, cytokines. Quality assessment was also completed to summarise the level of evidence available. Results A total of 16,702 publications were returned using our search terms. After screening of papers according to our inclusion and exclusion criteria, 15 studies were included in the review. All the included studies were observational case control studies. Ten of the studies identified measured serum cytokines in CFS/ME/SEID patients, and four measured cytokines in other physiological fluids of CFS/ME/SEID patients. The overall quality assessment revealed most papers included in this systematic review to be consistent. Conclusions Despite the availability of moderate quality studies, the findings of this review are inconclusive as to whether cytokines play any definitive role in CFS/ME/SEID, and consequently, they would not serve as reliable biomarkers. Therefore, in light of these results, it is recommended that further efforts toward a diagnostic test and treatment for CFS/ME/SEID continue to be developed in a range of research fields. Electronic supplementary material The online version of this article (10.1186/s12883-019-1433-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Matthew Corbitt
- National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute, Griffith University, Gold Coast, Australia.
| | - Natalie Eaton-Fitch
- National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute, Griffith University, Gold Coast, Australia.,School of Medical Science, Griffith University, Gold Coast, Australia.,Consortium Health International for Myalgic Encephalomyelitis, National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia
| | - Donald Staines
- National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute, Griffith University, Gold Coast, Australia.,School of Medical Science, Griffith University, Gold Coast, Australia.,Consortium Health International for Myalgic Encephalomyelitis, National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia
| | - Hélène Cabanas
- National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute, Griffith University, Gold Coast, Australia.,School of Medical Science, Griffith University, Gold Coast, Australia.,Consortium Health International for Myalgic Encephalomyelitis, National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia
| | - Sonya Marshall-Gradisnik
- National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute, Griffith University, Gold Coast, Australia.,School of Medical Science, Griffith University, Gold Coast, Australia.,Consortium Health International for Myalgic Encephalomyelitis, National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia
| |
Collapse
|
23
|
Missailidis D, Annesley SJ, Fisher PR. Pathological Mechanisms Underlying Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. Diagnostics (Basel) 2019; 9:E80. [PMID: 31330791 PMCID: PMC6787592 DOI: 10.3390/diagnostics9030080] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 07/15/2019] [Accepted: 07/19/2019] [Indexed: 02/06/2023] Open
Abstract
The underlying molecular basis of myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is not well understood. Characterized by chronic, unexplained fatigue, a disabling payback following exertion ("post-exertional malaise"), and variably presenting multi-system symptoms, ME/CFS is a complex disease, which demands a concerted biomedical investigation from disparate fields of expertise. ME/CFS research and patient treatment have been challenged by the lack of diagnostic biomarkers and finding these is a prominent direction of current work. Despite these challenges, modern research demonstrates a tangible biomedical basis for the disorder across many body systems. This evidence is mostly comprised of disturbances to immunological and inflammatory pathways, autonomic and neurological dysfunction, abnormalities in muscle and mitochondrial function, shifts in metabolism, and gut physiology or gut microbiota disturbances. It is possible that these threads are together entangled as parts of an underlying molecular pathology reflecting a far-reaching homeostatic shift. Due to the variability of non-overlapping symptom presentation or precipitating events, such as infection or other bodily stresses, the initiation of body-wide pathological cascades with similar outcomes stemming from different causes may be implicated in the condition. Patient stratification to account for this heterogeneity is therefore one important consideration during exploration of potential diagnostic developments.
Collapse
Affiliation(s)
- Daniel Missailidis
- Department of Physiology Anatomy and Microbiology, La Trobe University, VIC 3086, Australia
| | - Sarah J Annesley
- Department of Physiology Anatomy and Microbiology, La Trobe University, VIC 3086, Australia
| | - Paul R Fisher
- Department of Physiology Anatomy and Microbiology, La Trobe University, VIC 3086, Australia.
| |
Collapse
|
24
|
Rethinking ME/CFS Diagnostic Reference Intervals via Machine Learning, and the Utility of Activin B for Defining Symptom Severity. Diagnostics (Basel) 2019; 9:diagnostics9030079. [PMID: 31331036 PMCID: PMC6787626 DOI: 10.3390/diagnostics9030079] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/09/2019] [Accepted: 07/15/2019] [Indexed: 12/17/2022] Open
Abstract
Biomarker discovery applied to myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS), a disabling disease of inconclusive aetiology, has identified several cytokines to potentially fulfil a role as a quantitative blood/serum marker for laboratory diagnosis, with activin B a recent addition. We explored further the potential of serum activin B as a ME/CFS biomarker, alone and in combination with a range of routine test results obtained from pathology laboratories. Previous pilot study results showed that activin B was significantly elevated for the ME/CFS participants compared to healthy (control) participants. All the participants were recruited via CFS Discovery and assessed via the Canadian/International Consensus Criteria. A significant difference for serum activin B was also detected for ME/CFS and control cohorts recruited for this study, but median levels were significantly lower for the ME/CFS cohort. Random Forest (RF) modelling identified five routine pathology blood test markers that collectively predicted ME/CFS at ≥62% when compared via weighted standing time (WST) severity classes. A closer analysis revealed that the inclusion of activin B to the panel of pathology markers improved the prediction of mild to moderate ME/CFS cases. Applying correct WST class prediction from RFA modelling, new reference intervals were calculated for activin B and associated pathology markers, where 24-h urinary creatinine clearance, serum urea and serum activin B showed the best potential as diagnostic markers. While the serum activin B results remained statistically significant for the new participant cohorts, activin B was found to also have utility in enhancing the prediction of symptom severity, as represented by WST class.
Collapse
|
25
|
Yang T, Yang Y, Wang D, Li C, Qu Y, Guo J, Shi T, Bo W, Sun Z, Asakawa T. The clinical value of cytokines in chronic fatigue syndrome. J Transl Med 2019; 17:213. [PMID: 31253154 PMCID: PMC6599310 DOI: 10.1186/s12967-019-1948-6] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 06/05/2019] [Indexed: 02/07/2023] Open
Abstract
Chronic fatigue syndrome (CFS) is a heterogeneous disorder with uncertain pathogenesis. Without effective therapy, CFS is characterized by disabling fatigue, depression, memory loss, and somatic discomfort. This comprehensive and impartial review aimed to assess the available evidence and examined the potential clinical value of using cytokines for the monitoring of CFS and as targets for the treatment of CFS. Inflammatory reactions and immune modulation are considered to contribute to the pathophysiology of CFS, and it is well documented that cytokines present in both blood and cerebrospinal fluid (CSF) are closely associated with the progression and severity of CFS. However, pathophysiological and methodological limitations prevent using circulating cytokines as independent diagnostic indices. Moreover, there is no evidence to support the use of CSF cytokines as independent diagnostic indices. Nevertheless, a comprehensive evaluation of changes in circulating and CSF cytokines may improve clinical understanding of the pathophysiology of patients with CFS, aiding in the establishment of an appropriate diagnosis. Importantly, the available evidence does not support the value of cytokines as therapeutic targets. We believe that an improved understanding of cytokine-related mechanisms will be helpful to explore new cytokine-related therapeutic targets.
Collapse
Affiliation(s)
- Tiansong Yang
- First Affiliated Hospital, Heilongjiang University of Chinese Medicine, 26 Heping Road, Harbin, China
| | - Yan Yang
- Heilongjiang University of Chinese Medicine, 24 Heping Road, Harbin, China
| | - Delong Wang
- Heilongjiang University of Chinese Medicine, 24 Heping Road, Harbin, China
| | - Chaoran Li
- Heilongjiang University of Chinese Medicine, 24 Heping Road, Harbin, China
| | - Yuanyuan Qu
- Heilongjiang University of Chinese Medicine, 24 Heping Road, Harbin, China
| | - Jing Guo
- Heilongjiang University of Chinese Medicine, 24 Heping Road, Harbin, China
| | - Tianyu Shi
- Heilongjiang University of Chinese Medicine, 24 Heping Road, Harbin, China
| | - Wang Bo
- Heilongjiang University of Chinese Medicine, 24 Heping Road, Harbin, China
| | - Zhongren Sun
- Heilongjiang University of Chinese Medicine, 24 Heping Road, Harbin, China.
| | - Tetsuya Asakawa
- Department of Neurosurgery, Hamamatsu University School of Medicine, Handayama, 1-20-1, Higashi-ku, Hamamatsu, Shizuoka, 431-3192, Japan. .,Research Base of Traditional Chinese Medicine Syndrome, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China.
| |
Collapse
|
26
|
Chaves-Filho AJM, Macedo DS, de Lucena DF, Maes M. Shared microglial mechanisms underpinning depression and chronic fatigue syndrome and their comorbidities. Behav Brain Res 2019; 372:111975. [PMID: 31136774 DOI: 10.1016/j.bbr.2019.111975] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 04/09/2019] [Accepted: 05/23/2019] [Indexed: 12/24/2022]
Abstract
In 2011, it was reviewed that a) there is a strong co-occurrence between major depression and chronic fatigue syndrome (CFS), with fatigue and physio-somatic symptoms being key symptoms of depression, and depressive symptoms appearing during the course of CFS; and b) the comorbidity between both disorders may in part be explained by activated immune-inflammatory pathways, including increased translocation of Gram-negative bacteria and increased levels of pro-inflammatory cytokines, such as interleukin (IL)-1. Nevertheless, the possible involvement of activated microglia in this comorbidity has remained unclear. This paper aims to review microglial disturbances in major depression, CFS and their comorbidity. A comprehensive literature search was conducted using the PubMed / MEDLINE database to identify studies, which are relevant to this current review. Depressed patients present neuroinflammatory alterations, probably related to microglial activation, while animal models show that a microglial response to immune challenges including lipopolysaccharides is accompanied by depressive-like behaviors. Recent evidence from preclinical studies indicates that activated microglia have a key role in the onset of fatigue. In chronic inflammatory conditions, such as infections and senescence, microglia orchestrate an inflammatory microenvironment thereby causing fatigue. In conclusion, based on our review we may posit that shared immune-inflammatory pathways and especially activated microglia underpin comorbid depression and CFS. As such, microglial activation and neuro-inflammation may be promising targets to treat the overlapping manifestations of both depression and CFS.
Collapse
Affiliation(s)
- Adriano José Maia Chaves-Filho
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil.
| | - Danielle S Macedo
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil; National Institute for Translational Medicine (INCT-TM, CNPq), Ribeirão Preto, Brazil.
| | - David Freitas de Lucena
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil.
| | - Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; IMPACT Strategic Research Center, Deakin University, Geelong, Australia.
| |
Collapse
|
27
|
Larssen E, Brede C, Hjelle A, Tjensvoll AB, Norheim KB, Bårdsen K, Jonsdottir K, Ruoff P, Omdal R, Nilsen MM. Fatigue in primary Sjögren's syndrome: A proteomic pilot study of cerebrospinal fluid. SAGE Open Med 2019; 7:2050312119850390. [PMID: 31205695 PMCID: PMC6537061 DOI: 10.1177/2050312119850390] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 04/22/2019] [Indexed: 12/16/2022] Open
Abstract
Objectives: Fatigue is a frequent and often disabling phenomenon that occurs in patients
with chronic inflammatory and immunological diseases, and the underlying
biological mechanisms are largely unknown. Because fatigue is generated in
the brain, we aimed to investigate cerebrospinal fluid and search for
molecules that participate in the pathophysiology of fatigue processes. Methods: A label-free shotgun proteomics approach was applied to analyze the
cerebrospinal fluid proteome of 20 patients with primary Sjögren’s syndrome.
Fatigue was measured with the fatigue visual analog scale. Results: A total of 828 proteins were identified and the 15 top discriminatory
proteins between patients with high and low fatigue were selected. Among
these were apolipoprotein A4, hemopexin, pigment epithelium-derived factor,
secretogranin-1, secretogranin-3, selenium-binding protein 1, and complement
factor B. Conclusion: Most of the discriminatory proteins have important roles in regulation of
innate immunity, cellular stress defense, and/or functions in the central
nervous system. These proteins and their interacting protein networks may
therefore have central roles in the generation and regulation of fatigue,
and the findings contribute with evidence to the concept of fatigue as a
biological phenomenon signaled through specific molecular pathways.
Collapse
Affiliation(s)
- Eivind Larssen
- Research Department, Stavanger University Hospital, Stavanger, Norway.,Norwegian Research Centre AS (NORCE), Stavanger, Norway
| | - Cato Brede
- Department of Medical Biochemistry, Stavanger University Hospital, Stavanger, Norway
| | - Anne Hjelle
- Research Department, Stavanger University Hospital, Stavanger, Norway
| | | | - Katrine Brække Norheim
- Clinical Immunology Unit, Department of Internal Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Kjetil Bårdsen
- Research Department, Stavanger University Hospital, Stavanger, Norway
| | - Kristin Jonsdottir
- Department of Pathology, Stavanger University Hospital, Stavanger, Norway
| | - Peter Ruoff
- Centre for Organelle Research (CORE), University of Stavanger, Stavanger, Norway
| | - Roald Omdal
- Clinical Immunology Unit, Department of Internal Medicine, Stavanger University Hospital, Stavanger, Norway.,Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Mari Mæland Nilsen
- Research Department, Stavanger University Hospital, Stavanger, Norway.,Norwegian Research Centre AS (NORCE), Stavanger, Norway
| |
Collapse
|
28
|
Morris G, Maes M, Berk M, Puri BK. Myalgic encephalomyelitis or chronic fatigue syndrome: how could the illness develop? Metab Brain Dis 2019; 34:385-415. [PMID: 30758706 PMCID: PMC6428797 DOI: 10.1007/s11011-019-0388-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 01/23/2019] [Indexed: 12/19/2022]
Abstract
A model of the development and progression of chronic fatigue syndrome (myalgic encephalomyelitis), the aetiology of which is currently unknown, is put forward, starting with a consideration of the post-infection role of damage-associated molecular patterns and the development of chronic inflammatory, oxidative and nitrosative stress in genetically predisposed individuals. The consequences are detailed, including the role of increased intestinal permeability and the translocation of commensal antigens into the circulation, and the development of dysautonomia, neuroinflammation, and neurocognitive and neuroimaging abnormalities. Increasing levels of such stress and the switch to immune and metabolic downregulation are detailed next in relation to the advent of hypernitrosylation, impaired mitochondrial performance, immune suppression, cellular hibernation, endotoxin tolerance and sirtuin 1 activation. The role of chronic stress and the development of endotoxin tolerance via indoleamine 2,3-dioxygenase upregulation and the characteristics of neutrophils, monocytes, macrophages and T cells, including regulatory T cells, in endotoxin tolerance are detailed next. Finally, it is shown how the immune and metabolic abnormalities of chronic fatigue syndrome can be explained by endotoxin tolerance, thus completing the model.
Collapse
Affiliation(s)
- Gerwyn Morris
- IMPACT Strategic Research Centre, School of Medicine, Barwon Health, Deakin University, Geelong, Victoria, Australia
| | - Michael Maes
- IMPACT Strategic Research Centre, School of Medicine, Barwon Health, Deakin University, Geelong, Victoria, Australia
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Michael Berk
- IMPACT Strategic Research Centre, School of Medicine, Barwon Health, Deakin University, Geelong, Victoria, Australia
- Department of Psychiatry, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
- Orygen, The National Centre of Excellence in Youth Mental Health, Parkville, Victoria, Australia
| | - Basant K Puri
- Department of Medicine, Imperial College London, Hammersmith Hospital, London, England, W12 0HS, UK.
| |
Collapse
|
29
|
VanElzakker MB, Brumfield SA, Lara Mejia PS. Neuroinflammation and Cytokines in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS): A Critical Review of Research Methods. Front Neurol 2019; 9:1033. [PMID: 30687207 PMCID: PMC6335565 DOI: 10.3389/fneur.2018.01033] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Accepted: 11/16/2018] [Indexed: 01/18/2023] Open
Abstract
Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is the label given to a syndrome that can include long-term flu-like symptoms, profound fatigue, trouble concentrating, and autonomic problems, all of which worsen after exertion. It is unclear how many individuals with this diagnosis are suffering from the same condition or have the same underlying pathophysiology, and the discovery of biomarkers would be clarifying. The name "myalgic encephalomyelitis" essentially means "muscle pain related to central nervous system inflammation" and many efforts to find diagnostic biomarkers have focused on one or more aspects of neuroinflammation, from periphery to brain. As the field uncovers the relationship between the symptoms of this condition and neuroinflammation, attention must be paid to the biological mechanisms of neuroinflammation and issues with its potential measurement. The current review focuses on three methods used to study putative neuroinflammation in ME/CFS: (1) positron emission tomography (PET) neuroimaging using translocator protein (TSPO) binding radioligand (2) magnetic resonance spectroscopy (MRS) neuroimaging and (3) assays of cytokines circulating in blood and cerebrospinal fluid. PET scanning using TSPO-binding radioligand is a promising option for studies of neuroinflammation. However, methodological difficulties that exist both in this particular technique and across the ME/CFS neuroimaging literature must be addressed for any results to be interpretable. We argue that the vast majority of ME/CFS neuroimaging has failed to use optimal techniques for studying brainstem, despite its probable centrality to any neuroinflammatory causes or autonomic effects. MRS is discussed as a less informative but more widely available, less invasive, and less expensive option for imaging neuroinflammation, and existing studies using MRS neuroimaging are reviewed. Studies seeking to find a peripheral circulating cytokine "profile" for ME/CFS are reviewed, with attention paid to the biological and methodological reasons for lack of replication among these studies. We argue that both the biological mechanisms of cytokines and the innumerable sources of potential variance in their measurement make it unlikely that a consistent and replicable diagnostic cytokine profile will ever be discovered.
Collapse
Affiliation(s)
- Michael B. VanElzakker
- Division of Neurotherapeutics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | | | | |
Collapse
|
30
|
Hatziagelaki E, Adamaki M, Tsilioni I, Dimitriadis G, Theoharides TC. Myalgic Encephalomyelitis/Chronic Fatigue Syndrome-Metabolic Disease or Disturbed Homeostasis due to Focal Inflammation in the Hypothalamus? J Pharmacol Exp Ther 2018; 367:155-167. [PMID: 30076265 DOI: 10.1124/jpet.118.250845] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 08/01/2018] [Indexed: 12/13/2022] Open
Abstract
Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a complex disease characterized by debilitating fatigue, lasting for at least 6 months, with associated malaise, headaches, sleep disturbance, and cognitive impairment, which severely impacts quality of life. A significant percentage of ME/CFS patients remain undiagnosed, mainly due to the complexity of the disease and the lack of reliable objective biomarkers. ME/CFS patients display decreased metabolism and the severity of symptoms appears to be directly correlated to the degree of metabolic reduction that may be unique to each individual patient. However, the precise pathogenesis is still unknown, preventing the development of effective treatments. The ME/CFS phenotype has been associated with abnormalities in energy metabolism, which are apparently due to mitochondrial dysfunction in the absence of mitochondrial diseases, resulting in reduced oxidative metabolism. Such mitochondria may be further contributing to the ME/CFS symptomatology by extracellular secretion of mitochondrial DNA, which could act as an innate pathogen and create an autoinflammatory state in the hypothalamus. We propose that stimulation of hypothalamic mast cells by environmental, neuroimmune, pathogenic and stress triggers activates microglia, leading to focal inflammation in the brain and disturbed homeostasis. This process could be targeted for the development of novel effective treatments.
Collapse
Affiliation(s)
- Erifili Hatziagelaki
- Second Department of Internal Medicine, Attikon General Hospital, Athens Medical School, Athens, Greece (E.H., M.A., G.D.); Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology (I.T., T.C.T.) and Sackler School of Graduate Biomedical Sciences (T.C.T.), Tufts University School of Medicine, Boston, Massachusetts; and Departments of Internal Medicine and Psychiatry, Tufts University School of Medicine and Tufts Medical Center, Boston, Massachusetts (T.C.T.)
| | - Maria Adamaki
- Second Department of Internal Medicine, Attikon General Hospital, Athens Medical School, Athens, Greece (E.H., M.A., G.D.); Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology (I.T., T.C.T.) and Sackler School of Graduate Biomedical Sciences (T.C.T.), Tufts University School of Medicine, Boston, Massachusetts; and Departments of Internal Medicine and Psychiatry, Tufts University School of Medicine and Tufts Medical Center, Boston, Massachusetts (T.C.T.)
| | - Irene Tsilioni
- Second Department of Internal Medicine, Attikon General Hospital, Athens Medical School, Athens, Greece (E.H., M.A., G.D.); Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology (I.T., T.C.T.) and Sackler School of Graduate Biomedical Sciences (T.C.T.), Tufts University School of Medicine, Boston, Massachusetts; and Departments of Internal Medicine and Psychiatry, Tufts University School of Medicine and Tufts Medical Center, Boston, Massachusetts (T.C.T.)
| | - George Dimitriadis
- Second Department of Internal Medicine, Attikon General Hospital, Athens Medical School, Athens, Greece (E.H., M.A., G.D.); Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology (I.T., T.C.T.) and Sackler School of Graduate Biomedical Sciences (T.C.T.), Tufts University School of Medicine, Boston, Massachusetts; and Departments of Internal Medicine and Psychiatry, Tufts University School of Medicine and Tufts Medical Center, Boston, Massachusetts (T.C.T.)
| | - Theoharis C Theoharides
- Second Department of Internal Medicine, Attikon General Hospital, Athens Medical School, Athens, Greece (E.H., M.A., G.D.); Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology (I.T., T.C.T.) and Sackler School of Graduate Biomedical Sciences (T.C.T.), Tufts University School of Medicine, Boston, Massachusetts; and Departments of Internal Medicine and Psychiatry, Tufts University School of Medicine and Tufts Medical Center, Boston, Massachusetts (T.C.T.)
| |
Collapse
|
31
|
Blomberg J, Gottfries CG, Elfaitouri A, Rizwan M, Rosén A. Infection Elicited Autoimmunity and Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: An Explanatory Model. Front Immunol 2018; 9:229. [PMID: 29497420 PMCID: PMC5818468 DOI: 10.3389/fimmu.2018.00229] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 01/26/2018] [Indexed: 12/13/2022] Open
Abstract
Myalgic encephalomyelitis (ME) often also called chronic fatigue syndrome (ME/CFS) is a common, debilitating, disease of unknown origin. Although a subject of controversy and a considerable scientific literature, we think that a solid understanding of ME/CFS pathogenesis is emerging. In this study, we compiled recent findings and placed them in the context of the clinical picture and natural history of the disease. A pattern emerged, giving rise to an explanatory model. ME/CFS often starts after or during an infection. A logical explanation is that the infection initiates an autoreactive process, which affects several functions, including brain and energy metabolism. According to our model for ME/CFS pathogenesis, patients with a genetic predisposition and dysbiosis experience a gradual development of B cell clones prone to autoreactivity. Under normal circumstances these B cell offsprings would have led to tolerance. Subsequent exogenous microbial exposition (triggering) can lead to comorbidities such as fibromyalgia, thyroid disorder, and orthostatic hypotension. A decisive infectious trigger may then lead to immunization against autoantigens involved in aerobic energy production and/or hormone receptors and ion channel proteins, producing postexertional malaise and ME/CFS, affecting both muscle and brain. In principle, cloning and sequencing of immunoglobulin variable domains could reveal the evolution of pathogenic clones. Although evidence consistent with the model accumulated in recent years, there are several missing links in it. Hopefully, the hypothesis generates testable propositions that can augment the understanding of the pathogenesis of ME/CFS.
Collapse
Affiliation(s)
- Jonas Blomberg
- Department of Medical Sciences, Uppsala University, Clinical Microbiology, Academic Hospital, Uppsala, Sweden
| | | | - Amal Elfaitouri
- Department of Infectious Disease and Tropical Medicine, Faculty of Public Health, Benghazi University, Benghazi, Libya
| | - Muhammad Rizwan
- Department of Medical Sciences, Uppsala University, Clinical Microbiology, Academic Hospital, Uppsala, Sweden
| | - Anders Rosén
- Department of Clinical and Experimental Medicine, Division of Cell Biology, Linköping University, Linköping, Sweden
| |
Collapse
|
32
|
Depression, evening salivary cortisol and inflammation in chronic fatigue syndrome: A psychoneuroendocrinological structural regression model. Int J Psychophysiol 2017; 131:124-130. [PMID: 28918107 DOI: 10.1016/j.ijpsycho.2017.09.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 09/05/2017] [Accepted: 09/12/2017] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Chronic Fatigue Syndrome (CFS) is a poorly understood illness that is characterized by diverse somatic symptoms, hypothalamic pituitary adrenal (HPA) axis dysfunction and heightened inflammatory indicators. These symptoms are often exacerbated and accompanied by psychological distress states and depression. Since depression is known to be associated with HPA axis dysfunction and greater inflammation, a psychoneuroendocrinological (PNE) model of inflammation was examined in persons diagnosed with CFS in order to uncover underlying biopsychosocial mechanisms in this poorly understood chronic illness. METHODS Baseline data were drawn from two randomized controlled trials testing the efficacy of different forms of psychosocial intervention, and included psychological questionnaires, di-urnal salivary cortisol, and blood samples. Data were analyzed with structural equation modeling (SEM). RESULTS The sample (N=265) was mostly middle-aged (Mage=49.36±10.9, range=20-73years), Caucasian (67.7%), female (81.7%), highly educated (85.5% completed some college, college, or graduate program), and depressed (CES-D M=23.87±12.02, range 2-57). The SEM supporting a psychoneuroendocrinological model of immune dysregulation in CFS fit the data χ2 (12)=17.725, p=0.1243, RMSEA=0.043, CFI=0.935, SRMR=0.036. Depression was directly related to evening salivary cortisol and inflammation, such that higher evening cortisol predicted greater depressive symptoms (β=0.215, p<0.01) and higher pro-inflammatory cytokines (interleukin-2 [IL-2], IL-6, and tumor necrosis factor-alpha [TNF-α] levels (β=0.185, p<0.05), when controlling for covariates. DISCUSSION Results highlight the role of depression, cortisol and inflammation in possible biological mechanisms involved in the pathophysiology of CFS. Time-lagged, longitudinal analyses are needed to fully explore these relationships.
Collapse
|
33
|
Li Y, Li H, Wen B, Zhang J, Li S, Xie Y, Lv X, Qu X, Huang R, Liu W. Cytokine expression profile in hospitalized children with herpes simplex virus-1 infection. Future Virol 2017. [DOI: 10.2217/fvl-2017-0010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Aim: We aim to demonstrate the cytokine expression profile of children with herpes simplex virus-1 (HSV-1) infection. Methods: One hundred eighty three children with acute encephalitis were enrolled in this study. Out of those with HSV-1, multiple cytokines were measured. Results: Cytokine analysis of 13 HSV-1-infected patients revealed that IL-8 and granulocyte macrophage colony stimulating factor accumulated at significantly higher levels than macrophage inflammatory protein-1β in the cerebrospinal fluid. Additionally, IFN-γ in serum and cerebrospinal fluid was positively correlated with course of illness, while IL-10 in cerebrospinal fluid was negatively correlated with course of illness. Conclusion: Granulocyte macrophage colony stimulating factor, IL-8, macrophage inflammatory protein-1β, IFN-γ and IL-10 are involved in the HSV-1 immune response and pathogenesis.
Collapse
Affiliation(s)
- Yuanyuan Li
- Department of Neurology, The Third People's Hospital of Yancheng, Yancheng, Jiangsu, China
| | - Haipeng Li
- Department of Neurology, The First People's Hospital of Chenzhou, Chenzhou, Hunan 423000, China
| | - Bo Wen
- Translational Medicine Institute, The First People's Hospital of Chenzhou, University of South China, Chenzhou, Hunan, China
| | - Jian Zhang
- Translational Medicine Institute, The First People's Hospital of Chenzhou, University of South China, Chenzhou, Hunan, China
| | - Shuochi Li
- Translational Medicine Institute, The First People's Hospital of Chenzhou, University of South China, Chenzhou, Hunan, China
| | - Yuee Xie
- Translational Medicine Institute, The First People's Hospital of Chenzhou, University of South China, Chenzhou, Hunan, China
| | - Xinjun Lv
- National Institute of Viral Disease Control & Prevention, China CDC, Beijing, China
| | - Xiaowang Qu
- Translational Medicine Institute, The First People's Hospital of Chenzhou, University of South China, Chenzhou, Hunan, China
| | - Renbin Huang
- Department of Neurology, The First People's Hospital of Chenzhou, Chenzhou, Hunan 423000, China
| | - Wenpei Liu
- Translational Medicine Institute, The First People's Hospital of Chenzhou, University of South China, Chenzhou, Hunan, China
| |
Collapse
|
34
|
Cytokine signature associated with disease severity in chronic fatigue syndrome patients. Proc Natl Acad Sci U S A 2017; 114:E7150-E7158. [PMID: 28760971 DOI: 10.1073/pnas.1710519114] [Citation(s) in RCA: 241] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Although some signs of inflammation have been reported previously in patients with myalgic encephalomyelitis or chronic fatigue syndrome (ME/CFS), the data are limited and contradictory. High-throughput methods now allow us to interrogate the human immune system for multiple markers of inflammation at a scale that was not previously possible. To determine whether a signature of serum cytokines could be associated with ME/CFS and correlated with disease severity and fatigue duration, cytokines of 192 ME/CFS patients and 392 healthy controls were measured using a 51-multiplex array on a Luminex system. Each cytokine's preprocessed data were regressed on ME/CFS severity plus covariates for age, sex, race, and an assay property of newly discovered importance: nonspecific binding. On average, TGF-β was elevated (P = 0.0052) and resistin was lower (P = 0.0052) in patients compared with controls. Seventeen cytokines had a statistically significant upward linear trend that correlated with ME/CFS severity: CCL11 (Eotaxin-1), CXCL1 (GROα), CXCL10 (IP-10), IFN-γ, IL-4, IL-5, IL-7, IL-12p70, IL-13, IL-17F, leptin, G-CSF, GM-CSF, LIF, NGF, SCF, and TGF-α. Of the 17 cytokines that correlated with severity, 13 are proinflammatory, likely contributing to many of the symptoms experienced by patients and establishing a strong immune system component of the disease. Only CXCL9 (MIG) inversely correlated with fatigue duration.
Collapse
|
35
|
Mensah FKF, Bansal AS, Ford B, Cambridge G. Chronic fatigue syndrome and the immune system: Where are we now? Neurophysiol Clin 2017; 47:131-138. [PMID: 28410877 DOI: 10.1016/j.neucli.2017.02.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Accepted: 02/07/2017] [Indexed: 10/19/2022] Open
Abstract
Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is characterised by multiple symptoms including fatigue, headaches and cognitive impairment, which have a significantly adverse effect on the normal functioning and well-being of the individual. These symptoms are often triggered or worsened following physical or mental exertion. ME/CFS has long been thought of as having a significant immunological component, but reports describing changes in immune function are often inconsistent between study groups. Although the wide range of physical, neurocognitive and autonomic symptoms reported have seriously hampered attempts to understand pathophysiological pathways, investment in biomedical research in ME/CFS is finally increasing with a number of novel and promising investigations being published. The onset of ME/CFS may often be linked to (viral) infections which would be consistent with a variety of alterations in natural killer (NK) cell function as described by a number of different groups. Consistency in cytokine data has been lacking so far, although recently more sophisticated approaches have led to more robust data from large patient cohorts. New hope has also been given to sufferers with the possibility that therapies that deplete B cells can result in clinical improvement. To understand the pathogenic mechanism in this complex condition, it is important to consider repeated analysis in different cohorts. In this review, we will discuss the potential of different components of the immune system to be involved in the pathogenesis of ME/CFS.
Collapse
Affiliation(s)
- Fane Kojo Fosu Mensah
- Department of Rheumatology Research, Division of Medicine, University College of London, Rayne Building, 5, University Street, WC1E 6JF London, United Kingdom.
| | - Amolak Singh Bansal
- Department of Immunology, Epsom and St-Helier University Hospitals NHS Trust, London, United Kingdom
| | - Brian Ford
- Department of Immunology, Epsom and St-Helier University Hospitals NHS Trust, London, United Kingdom
| | - Geraldine Cambridge
- Department of Rheumatology Research, Division of Medicine, University College of London, Rayne Building, 5, University Street, WC1E 6JF London, United Kingdom
| |
Collapse
|
36
|
Lidbury BA, Kita B, Lewis DP, Hayward S, Ludlow H, Hedger MP, de Kretser DM. Activin B is a novel biomarker for chronic fatigue syndrome/myalgic encephalomyelitis (CFS/ME) diagnosis: a cross sectional study. J Transl Med 2017; 15:60. [PMID: 28302133 PMCID: PMC5353946 DOI: 10.1186/s12967-017-1161-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 03/10/2017] [Indexed: 12/15/2022] Open
Abstract
Background Investigations of activin family proteins as serum biomarkers for chronic fatigue syndrome/myalgic encephalomyelitis (CFS/ME). CFS/ME is a disease with complex, wide-ranging symptoms, featuring persistent fatigue of 6 months or longer, particularly post exertion. No definitive biomarkers are available. Methods A cross-sectional, observational study of CFS/ME patients fulfilling the 2003 Canadian Consensus Criteria, in parallel with healthy non-fatigued controls, was conducted. Comparisons with a previously defined activin reference population were also performed. For the total study cohort the age range was 18–65 years with a female: male participant ratio of greater than 3:1. All participants were assessed via a primary care community clinic. Blood samples were collected for pathology testing after physical examination and orthostatic intolerance assessment. Cytokines, activin A, activin B and follistatin were also measured in sera from these samples. All data were compared between the CFS/ME and control cohorts, with the activins and follistatin also compared with previously defined reference intervals. Results Serum activin B levels for CFS/ME participants were significantly elevated when compared to the study controls, as well as the established reference interval. Serum activin A and follistatin were within their normal ranges. All routine and special pathology markers were within the normal laboratory reference intervals for the total study cohort, with no significant differences detected between CFS/ME and control groups. Also, no significant differences were detected for IL-2, IL-4, IL-6, IL-10, IL-17A, TNF or IFN-gamma. Conclusion Elevated activin B levels together with normal activin A levels identified patients with the diagnostic symptoms of CFS/ME, thus providing a novel serum based test. The activins have multiple physiological roles and capture the diverse array of symptoms experienced by CFS/ME patients.
Collapse
Affiliation(s)
- Brett A Lidbury
- Pattern Recognition and Pathology, Department of Genome Sciences, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, 2601, Australia. .,The National Centre for Epidemiology and Public Health, The Research School of Population Health, ANU, Canberra, ACT, 2601, Australia.
| | - Badia Kita
- Paranta Biosciences Limited, Caulfield North, VIC, 3161, Australia
| | - Donald P Lewis
- CFS Discovery, Donvale Medical Specialist Centre, Donvale, VIC, 3111, Australia
| | - Susan Hayward
- The Hudson Medical Research Institute, Monash University, Clayton, VIC, 3168, Australia
| | - Helen Ludlow
- Centre for Proteins and Peptides, School of Life Sciences, Oxford Brookes University, Headington, Oxford, OX3 0BP, UK
| | - Mark P Hedger
- The Hudson Medical Research Institute, Monash University, Clayton, VIC, 3168, Australia
| | - David M de Kretser
- The Hudson Medical Research Institute, Monash University, Clayton, VIC, 3168, Australia.,Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 3168, Australia
| |
Collapse
|
37
|
Roerink ME, van der Schaaf ME, Dinarello CA, Knoop H, van der Meer JWM. Interleukin-1 as a mediator of fatigue in disease: a narrative review. J Neuroinflammation 2017; 14:16. [PMID: 28109186 PMCID: PMC5251329 DOI: 10.1186/s12974-017-0796-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 01/12/2017] [Indexed: 01/17/2023] Open
Abstract
Fatigue is commonly reported in a variety of illnesses, and it has major impact on quality of life. Previously, it was thought that fatigue originates in the skeletal muscles, leading to cessation of activity. However, more recently, it has become clear that the brain is the central regulator of fatigue perception. It has been suggested that pro-inflammatory cytokines, especially interleukin-1 alpha (IL-1α) and interleukin-1 beta (IL-1β), play a prominent role in the development of central fatigue, and several studies have been performed to elucidate the connection between inflammation and these central processes.In this narrative review, mechanisms of action of IL-1 are described, with special attention to its effect on the central nervous system. In addition, we present a summary of studies that (i) investigated the relationship between circulating IL-1α and IL-1β and fatigue severity and/or (ii) evaluated the effect of inhibiting IL-1 on fatigue. We aim to improve the understanding of fatigue in both inflammatory and non-inflammatory illnesses, which could help develop strategies to treat fatigue more effectively.Reviewing the studies that have been performed, it appears that there is a limited value of measuring circulating IL-1. However, inhibiting IL-1 has a positive effect on severe fatigue in most studies that have been conducted.
Collapse
Affiliation(s)
- Megan E Roerink
- Department of Internal Medicine, Radboud University Medical Centre, Geert Grooteplein Zuid 8, 6500HB, Nijmegen, The Netherlands.
| | - Marieke E van der Schaaf
- Expert Centre for Chronic Fatigue, Radboud University Medical Centre, Reinier Postlaan 4, 6525GC, Nijmegen, The Netherlands
| | - Charles A Dinarello
- Department of Internal Medicine, Radboud University Medical Centre, Geert Grooteplein Zuid 8, 6500HB, Nijmegen, The Netherlands.,Department of Medicine, University of Colorado Denver, 12700 E. 19th Avenue Box B168, Aurora, CO, 80045, USA
| | - Hans Knoop
- Expert Centre for Chronic Fatigue, Radboud University Medical Centre, Reinier Postlaan 4, 6525GC, Nijmegen, The Netherlands.,Department of Medical Psychology, Academic Medical Centre (AMC), University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Jos W M van der Meer
- Department of Internal Medicine, Radboud University Medical Centre, Geert Grooteplein Zuid 8, 6500HB, Nijmegen, The Netherlands
| |
Collapse
|
38
|
Muluye RA, Bian Y, Wang L, Alemu PN, Cui H, Peng X, Li S. Placenta Peptide Can Protect Mitochondrial Dysfunction through Inhibiting ROS and TNF-α Generation, by Maintaining Mitochondrial Dynamic Network and by Increasing IL-6 Level during Chronic Fatigue. Front Pharmacol 2016; 7:328. [PMID: 27729861 PMCID: PMC5037131 DOI: 10.3389/fphar.2016.00328] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 09/06/2016] [Indexed: 12/04/2022] Open
Abstract
Background: Level of fatigue is related to the metabolic energy available to tissues and cells, mainly through mitochondrial respiration, as well fatigue is the most common symptom of poorly functioning mitochondria. Hence, dysfunction of these organelles may be the cause of the fatigue seen in Chronic fatigue (CF). Placenta has been used for treatment of fatigue and various disease, moreover peptides has known protect mitochondrial viability, and alleviate fatigue. These properties of placenta and peptides may link with its effect on mitochondria; therefore, it is highly important to investigate the effectiveness of placenta peptide on fatigue and mitochondrial dysfunction. Methods: After administration of sheep placenta peptide (SPP) for 1 month, mice's were forced to swim till exhaustion for 90 min to induce chronic fatigue. Electron microscopic examination of skeletal muscle mitochondrial structure, tissue Malondialdehyde (MDA), mitochondrial SOD and serum inflammatory cytokines level were investigated in order to determine the potential effect of SPP on mitochondria during CF. Rat skeletal muscle (L6 cell) were also treated with different concentration of SPP to determine the effect of SPP on cell viability using Thiazoyl blue tetrazolium assay. Results: Our finding revealed that forced swimming induced fatigue model can cause mitochondrial damage through Reactive oxygen species (ROS) mediated lipid peroxidation and Tumor Necrosis factor alpha (TNF-α) elevation. Whereas SPP protected fatigue induced mitochondrial dysfunction through preventing ROS and TNF-α generation, by maintaining mitochondrial dynamic network and by increasing serum IL-6 level. Conclusion: SPP can protect damage in mitochondrial components which will allow proper functioning of mitochondria that will in turn inhibit progression of chronic fatigue. Therefore, SPP may represent a novel therapeutic advantage for preventing mitochondrial dysfunction in patients with chronic fatigue.
Collapse
Affiliation(s)
- Rekik A. Muluye
- Tianjin University of Traditional Chinese MedicineTianjin, China
- Ethiopian Public Health Institute Addis Ababa, Ethiopia
| | - Yuhong Bian
- Tianjin University of Traditional Chinese MedicineTianjin, China
| | - Li Wang
- Tianjin Second People’s HospitalTianjin, China
| | - Paulos N. Alemu
- Tianjin University of Traditional Chinese MedicineTianjin, China
- Ethiopian Public Health Institute Addis Ababa, Ethiopia
| | - Huantian Cui
- Tianjin University of Traditional Chinese MedicineTianjin, China
| | - Xiaofei Peng
- Tianjin University of Traditional Chinese MedicineTianjin, China
| | - Shanshan Li
- Tianjin University of Traditional Chinese MedicineTianjin, China
| |
Collapse
|
39
|
Chacko A, Staines DR, Johnston SC, Marshall-Gradisnik SM. Dysregulation of Protein Kinase Gene Expression in NK Cells from Chronic Fatigue Syndrome/Myalgic Encephalomyelitis Patients. GENE REGULATION AND SYSTEMS BIOLOGY 2016; 10:85-93. [PMID: 27594784 PMCID: PMC5003121 DOI: 10.4137/grsb.s40036] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 06/26/2016] [Accepted: 06/28/2016] [Indexed: 01/10/2023]
Abstract
BACKGROUND The etiology and pathomechanism of chronic fatigue syndrome/myalgic encephalomyelitis (CFS/ME) are unknown. However, natural killer (NK) cell dysfunction, in particular reduced NK cytotoxic activity, is a consistent finding in CFS/ME patients. Previous research has reported significant changes in intracellular mitogen-activated protein kinase pathways from isolated NK cells. The purpose of this present investigation was to examine whether protein kinase genes have a role in abnormal NK cell intracellular signaling in CFS/ME. METHOD Messenger RNA (mRNA) expression of 528 protein kinase genes in isolated NK cells was analyzed (nCounter GX Human Kinase Kit v2 (XT); NanoString Technologies) from moderate (n = 11; age, 54.9 ± 10.3 years) and severe (n = 12; age, 47.5 ± 8.0 years) CFS/ME patients (classified by the 2011 International Consensus Criteria) and nonfatigued controls (n = 11; age, 50.0 ± 12.3 years). RESULTS The expression of 92 protein kinase genes was significantly different in the severe CFS/ME group compared with nonfatigued controls. Among these, 37 genes were significantly upregulated and 55 genes were significantly downregulated in severe CFS/ME patients compared with nonfatigued controls. CONCLUSIONS In severe CFS/ME patients, dysfunction in protein kinase genes may contribute to impairments in NK cell intracellular signaling and effector function. Similar changes in protein kinase genes may be present in other cells, potentially contributing to the pathomechanism of this illness.
Collapse
Affiliation(s)
- Anu Chacko
- School of Medical Science, Griffith University, QLD, Australia.; The National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, QLD, Australia
| | - Donald R Staines
- The National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, QLD, Australia
| | - Samantha C Johnston
- School of Medical Science, Griffith University, QLD, Australia.; The National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, QLD, Australia
| | - Sonya M Marshall-Gradisnik
- School of Medical Science, Griffith University, QLD, Australia.; The National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, QLD, Australia
| |
Collapse
|
40
|
Hajighasemi F, Mirshafiey A. In Vitro Effects of Propranolol on T Helper Type 1 Cytokine Profile in Human Leukemic T Cells. Int J Hematol Oncol Stem Cell Res 2016; 10:99-105. [PMID: 27252810 PMCID: PMC4888155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
INTRODUCTION Cytokines are a large group of proteins play a key role in inflammation. Down-regulation of pro-inflammatory cytokines has beneficial effect on heart function. Propranolol, as a non selective beta-adrenergic blocker, has been extensively used for treatment of many cardiovascular problems such as arrhythmias and heart malfunction. In addition anti-inflammatory effects of propranolol have been revealed. In this study the propranolol effect on T helper type 1 cytokine profile in human leukemic T cells has been assessed in vitro. MATERIALS AND METHODS Human leukemic T cells (Molt-4 and Jurkat) were cultured in complete RPMI medium. The cells were then incubated with different concentrations of propranolol (0.03- 30 µM) in the presence or absence of PHA (10 µg/ml) for 48 hours. The supernatants of cell culture media were collected and used for cytokines assay. RESULTS Propranolol significantly decreased the T helper type 1 cytokine profile [Interleukin-2 (IL-2) and Interferon- γ (IFN-γ)] production in PHA stimulated Molt-4 and Jurkat cells, after 48 hour of incubation time, dose-dependently compared to untreated control cells. CONCLUSION Our data showed a dose dependent inhibitory effect of propranolol on the IL-2 and IFN-γ production in human leukemic Molt-4 and Jurkat cells. The anti- inflammatory effect of propranolol reported by other investigators may be in part due to its suppressive effect on production of inflammatory cytokines such as IL-2 and IFN-γ. So, propranolol along with its chronic long-term usage in cardiovascular problems may have potential implication in treatment of inflammatory-based disorders.
Collapse
Affiliation(s)
- Fatemeh Hajighasemi
- Department of Immunology, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Abbas Mirshafiey
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
41
|
Russell L, Broderick G, Taylor R, Fernandes H, Harvey J, Barnes Z, Smylie A, Collado F, Balbin EG, Katz BZ, Klimas NG, Fletcher MA. Illness progression in chronic fatigue syndrome: a shifting immune baseline. BMC Immunol 2016; 17:3. [PMID: 26965484 PMCID: PMC4785654 DOI: 10.1186/s12865-016-0142-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 02/29/2016] [Indexed: 01/04/2023] Open
Abstract
Background Validation of biomarkers for myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) across data sets has proven disappointing. As immune signature may be affected by many factors, our objective was to explore the shift in discriminatory cytokines across ME/CFS subjects separated by duration of illness. Methods Cytokine expression collected at rest across multiple studies for female ME/CFS subjects (i) 18 years or younger, ill for 2 years or less (n = 18), (ii) 18–50 years of age, ill for 7 years (n = 22), and (iii) age 50 years or older (n = 28), ill for 11 years on average. Control subjects were matched for age and body mass index (BMI). Data describing the levels of 16 cytokines using a chemiluminescent assay was used to support the identification of separate linear classification models for each subgroup. In order to isolate the effects of duration of illness alone, cytokines that changed significantly with age in the healthy control subjects were excluded a priori. Results Optimal selection of cytokines in each group resulted in subsets of IL-1α, 6, 8, 15 and TNFα. Common to any 2 of 3 groups were IL-1α, 6 and 8. Setting these 3 markers as a triple screen and adjusting their contribution according to illness duration sub-groups produced ME/CFS classification accuracies of 75–88 %. The contribution of IL-1α, higher in recently ill adolescent ME/CFS subjects was progressively less important with duration. While high levels of IL-8 screened positive for ME/CFS in the recently afflicted, the opposite was true for subjects ill for more than 2 years. Similarly, while low levels of IL-6 suggested early ME/CFS, the reverse was true in subjects over 18 years of age ill for more than 2 years. Conclusions These preliminary results suggest that IL-1α, 6 and 8 adjusted for illness duration may serve as robust biomarkers, independent of age, in screening for ME/CFS. Electronic supplementary material The online version of this article (doi:10.1186/s12865-016-0142-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lindsey Russell
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Gordon Broderick
- Department of Medicine, University of Alberta, Edmonton, AB, Canada. .,Miami Veterans Affairs Medical Center, Miami, FL, USA. .,Institute for Neuro-immune Medicine, Nova Southeastern University, Suite 3440 University Park Plaza, 3424 South University Drive, Fort Lauderdale, FL, 33328, USA.
| | - Renee Taylor
- Department of Occupational Therapy, University of Illinois at Chicago, Chicago, IL, USA
| | | | - Jeanna Harvey
- Department of Medicine, University of Miami, Miami, FL, USA.,Miami Veterans Affairs Medical Center, Miami, FL, USA
| | - Zachary Barnes
- Department of Medicine, University of Miami, Miami, FL, USA.,Miami Veterans Affairs Medical Center, Miami, FL, USA.,Institute for Neuro-immune Medicine, Nova Southeastern University, Suite 3440 University Park Plaza, 3424 South University Drive, Fort Lauderdale, FL, 33328, USA
| | - AnneLiese Smylie
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Fanny Collado
- Miami Veterans Affairs Medical Center, Miami, FL, USA
| | | | - Ben Z Katz
- Division of Infectious Diseases, Ann & Robert H Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Nancy G Klimas
- Miami Veterans Affairs Medical Center, Miami, FL, USA.,Institute for Neuro-immune Medicine, Nova Southeastern University, Suite 3440 University Park Plaza, 3424 South University Drive, Fort Lauderdale, FL, 33328, USA
| | - Mary Ann Fletcher
- Miami Veterans Affairs Medical Center, Miami, FL, USA.,Institute for Neuro-immune Medicine, Nova Southeastern University, Suite 3440 University Park Plaza, 3424 South University Drive, Fort Lauderdale, FL, 33328, USA
| |
Collapse
|
42
|
Mensah F, Bansal A, Berkovitz S, Sharma A, Reddy V, Leandro MJ, Cambridge G. Extended B cell phenotype in patients with myalgic encephalomyelitis/chronic fatigue syndrome: a cross-sectional study. Clin Exp Immunol 2016; 184:237-47. [PMID: 26646713 DOI: 10.1111/cei.12749] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2015] [Indexed: 12/21/2022] Open
Abstract
Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a heterogeneous condition of unknown aetiology characterized by multiple symptoms including fatigue, post-exertional malaise and cognitive impairment, lasting for at least 6 months. Recently, two clinical trials of B cell depletion therapy with rituximab (anti-CD20) reported convincing improvement in symptoms. A possible but undefined role for B cells has therefore been proposed. Studies of the relative percentages of B cell subsets in patients with ME/CFS have not revealed any reproducible differences from healthy controls (HC). In order to explore whether more subtle alterations in B cell subsets related to B cell differentiation exist in ME/CFS patients we used flow cytometry to immunophenotype CD19⁺ B cells. The panel utilized immunoglobulin (Ig)D, CD27 and CD38 (classical B cell subsets) together with additional markers. A total of 38 patients fulfilling Canadian, Centre for Disease Control and Fukuda ME/CFS criteria and 32 age- and sex-matched HC were included. We found no difference in percentages of classical subsets between ME/CFS patients and HC. However, we observed an increase in frequency (P < 0·01) and expression (MFI; P = 0·03) of CD24 on total B cells, confined to IgD⁺ subsets. Within memory subsets, a higher frequency of CD21⁺ CD38⁻ B cells (> 20%) was associated with the presence of ME/CFS [odds ratio: 3·47 (1·15-10·46); P = 0·03] compared with HC, and there was a negative correlation with disease duration. In conclusion, we identified possible changes in B cell phenotype in patients with ME/CFS. These may reflect altered B cell function and, if confirmed in other patient cohorts, could provide a platform for studies based on clinical course or responsiveness to rituximab therapy.
Collapse
Affiliation(s)
- F Mensah
- Department of Rheumatology Research, Division of Medicine, University College of London
| | - A Bansal
- Department of Immunology, Epsom and St Helier University Hospitals NHS Trust
| | - S Berkovitz
- Department of Neurology, Royal London Hospital of Integrated Medicine, London, UK
| | - A Sharma
- Department of Rheumatology Research, Division of Medicine, University College of London
| | - V Reddy
- Department of Rheumatology Research, Division of Medicine, University College of London
| | - M J Leandro
- Department of Rheumatology Research, Division of Medicine, University College of London
| | - G Cambridge
- Department of Rheumatology Research, Division of Medicine, University College of London
| |
Collapse
|
43
|
Landi A, Broadhurst D, Vernon SD, Tyrrell DLJ, Houghton M. Reductions in circulating levels of IL-16, IL-7 and VEGF-A in myalgic encephalomyelitis/chronic fatigue syndrome. Cytokine 2015; 78:27-36. [PMID: 26615570 DOI: 10.1016/j.cyto.2015.11.018] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 11/02/2015] [Accepted: 11/15/2015] [Indexed: 01/17/2023]
Abstract
Recently, differences in the levels of various chemokines and cytokines were reported in patients with myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) as compared with controls. Moreover, the analyte profile differed between chronic ME/CFS patients of long duration versus patients with disease of less than 3years. In the current study, we measured the plasma levels of 34 cytokines, chemokines and growth factors in 100 chronic ME/CFS patients of long duration and in 79 gender and age-matched controls. We observed highly significant reductions in the concentration of circulating interleukin (IL)-16, IL-7, and Vascular Endothelial Growth Factor A (VEGF-A) in ME/CFS patients. All three biomarkers were significantly correlated in a multivariate cluster analysis. In addition, we identified significant reductions in the concentrations of fractalkine (CX3CL1) and monokine-induced-by-IFN-γ (MIG; CXCL9) along with increases in the concentrations of eotaxin 2 (CCL24) in ME/CFS patients. Our data recapitulates previous data from another USA ME/CFS cohort in which circulating levels of IL-7 were reduced. Also, a reduced level of VEGF-A was reported previously in sera of patients with Gulf War Illness as well as in cerebral spinal fluid samples from a different cohort of USA ME/CFS patients. To our knowledge, we are the first to test for levels of IL-16 in ME/CFS patients. In combination with previous data, our work suggests that the clustered reduction of IL-7, IL-16 and VEGF-A may have physiological relevance to ME/CFS disease. This profile is ME/CFS-specific since measurement of the same analytes present in chronic infectious and autoimmune liver diseases, where persistent fatigue is also a major symptom, failed to demonstrate the same changes. Further studies of other ME/CFS and overlapping disease cohorts are warranted in future.
Collapse
Affiliation(s)
- Abdolamir Landi
- Li Ka Shing Institute of Virology, Department of Medical Microbiology and Immunology, Canada.
| | - David Broadhurst
- Department of Medicine, Katz Group Centre for Pharmacy & Health, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Suzanne D Vernon
- Bateman Horne Center, 1002 E. South Temple, Suite 408, Salt Lake City, UT 84102, USA
| | - D Lorne J Tyrrell
- Li Ka Shing Institute of Virology, Department of Medical Microbiology and Immunology, Canada
| | - Michael Houghton
- Li Ka Shing Institute of Virology, Department of Medical Microbiology and Immunology, Canada.
| |
Collapse
|