Pathological Mechanisms Underlying Myalgic Encephalomyelitis/Chronic Fatigue Syndrome
Abstract
:1. Introduction
2. Abnormal Metabolism and Mitochondrial Function
2.1. Dysregulated Amino Acid Metabolism and Impaired Provision of TCA Cycle Substrate
2.2. Inefficient ATP Synthesis and Abnormal Energy Stress Signaling
2.3. A Shift Towards Lipid Metabolism
3. Disturbed Immunity, Signaling, and Inflammatory Pathways
3.1. NK Cells
3.2. Calcium Signaling
3.3. Links to Mitochondrial Dysfunction
3.4. Inflammation
3.5. Autoimmunity
4. Implications of Altered Gut Microbiota and Physiology
4.1. The Gut Microbiota and Metabolism
4.2. Pathological Interactions between Gut Hyperpermeability and the Mitochondria
4.3. The Gut–Brain Axis: Autonomic and Hormonal Dysregulation
5. Multi-System Pathological Interactions
5.1. Exposure to Stressors and the Theoretical Homeostatic Perpetuation of a Disease State
5.2. Identifying Underlying Mechanisms
6. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Sharpe, M.C.; Archard, L.C.; Banatvala, J.E.; Borysiewicz, L.K.; Clare, A.W.; David, A.; Edwards, R.H.; Hawton, K.E.; Lambert, H.P.; Lane, R.J.; et al. A report—Chronic fatigue syndrome: Guidelines for research. J. R. Soc. Med. 1991, 84, 118–121. [Google Scholar] [CrossRef] [PubMed]
- Fukuda, K.; Straus, S.E.; Hickie, I.; Sharpe, M.C.; Dobbins, J.G.; Komaroff, A. The chronic fatigue syndrome: A comprehensive approach to its definition and study. International Chronic Fatigue Syndrome Study Group. Ann. Intern. Med. 1994, 121, 953–959. [Google Scholar] [CrossRef] [PubMed]
- Carruthers, B.M.; Jain, A.K.; De Meirleir, K.L.; Peterson, D.L.; Klimas, N.G.; Lerner, A.M.; Bested, A.C.; Flor-Henry, P.; Joshi, P.; Powles, A.C.; et al. Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: Clinical Working Case Definition, Diagnostic and Treatment Protocols. J. Chron. Fatigue Syndr. 2003, 11, 7–36. [Google Scholar] [CrossRef]
- Carruthers, B.M.; van de Sande, M.I.; De Meirleir, K.L.; Klimas, N.G.; Broderick, G.; Mitchell, T.; Staines, D.; Powles, A.C.; Speight, N.; Vallings, R.; et al. Myalgic encephalomyelitis: International Consensus Criteria. J. Intern. Med. 2011, 270, 327–338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lidbury, B.A.; Kita, B.; Lewis, D.P.; Hayward, S.; Ludlow, H.; Hedger, M.P.; de Kretser, D.M. 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. [Google Scholar] [CrossRef] [PubMed]
- Yamano, E.; Sugimoto, M.; Hirayama, A.; Kume, S.; Yamato, M.; Jin, G.; Tajima, S.; Goda, N.; Iwai, K.; Fukuda, S.; et al. Index markers of chronic fatigue syndrome with dysfunction of TCA and urea cycles. Sci. Rep. 2016, 6, 34990. [Google Scholar] [CrossRef] [PubMed]
- Naviaux, R.K.; Naviaux, J.C.; Li, K.; Bright, A.T.; Alaynick, W.A.; Wang, L.; Baxter, A.; Nathan, N.; Anderson, W.; Gordon, E. Metabolic features of chronic fatigue syndrome. Proc. Natl. Acad. Sci. USA 2016, 113, E5472–E5480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brenu, E.W.; Ashton, K.J.; van Driel, M.; Staines, D.R.; Peterson, D.; Atkinson, G.M.; Marshall-Gradisnik, S.M. Cytotoxic lymphocyte microRNAs as prospective biomarkers for Chronic Fatigue Syndrome/Myalgic Encephalomyelitis. J. Affect. Disord. 2012, 141, 261–269. [Google Scholar] [CrossRef]
- Myhill, S.; Booth, N.E.; McLaren-Howard, J. Chronic fatigue syndrome and mitochondrial dysfunction. Int. J. Clin. Exp. Med. 2009, 2, 1–16. [Google Scholar]
- Booth, N.E.; Myhill, S.; McLaren-Howard, J. Mitochondrial dysfunction and the pathophysiology of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS). Int. J. Clin. Exp. Med. 2012, 5, 208–220. [Google Scholar]
- Myhill, S.; Booth, N.E.; McLaren-Howard, J. Targeting mitochondrial dysfunction in the treatment of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS)-a clinical audit. Int. J. Clin. Exp. Med. 2013, 6, 1–15. [Google Scholar]
- Giloteaux, L.; Goodrich, J.K.; Walters, W.A.; Levine, S.M.; Ley, R.E.; Hanson, M.R. Reduced diversity and altered composition of the gut microbiome in individuals with myalgic encephalomyelitis/chronic fatigue syndrome. Microbiome 2016, 4, 30. [Google Scholar] [CrossRef]
- Germain, A.; Ruppert, D.; Levine, S.M.; Hanson, M.R. Metabolic profiling of a myalgic encephalomyelitis/chronic fatigue syndrome discovery cohort reveals disturbances in fatty acid and lipid metabolism. Mol. Biosyst. 2017, 13, 371–379. [Google Scholar] [CrossRef] [PubMed]
- Esfandyarpour, R.; Kashi, A.; Nemat-Gorgani, M.; Wilhelmy, J.; Davis, R.W. A nanoelectronics-blood-based diagnostic biomarker for myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). Proc. Natl. Acad. Sci. USA 2019. [Google Scholar] [CrossRef]
- Nacul, L.; de Barros, B.; Kingdon, C.C.; Cliff, J.M.; Clark, T.G.; Mudie, K.; Dockrell, H.M.; Lacerda, E.M. Evidence of Clinical Pathology Abnormalities in People with Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) from an Analytic Cross-Sectional Study. Diagnostics 2019, 9, 41. [Google Scholar] [CrossRef]
- Nacul, L.C.; Mudie, K.; Kingdon, C.C.; Clark, T.G.; Lacerda, E.M. Hand Grip Strength as a Clinical Biomarker for ME/CFS and Disease Severity. Front. Neurol. 2018, 9, 992. [Google Scholar] [CrossRef]
- DeLuca, J.; Johnson, S.K.; Ellis, S.P.; Natelson, B.H. Sudden vs gradual onset of chronic fatigue syndrome differentiates individuals on cognitive and psychiatric measures. J. Psychiatr. Res. 1997, 31, 83–90. [Google Scholar] [CrossRef]
- Lane, R.J.; Barrett, M.C.; Taylor, D.J.; Kemp, G.J.; Lodi, R. Heterogeneity in chronic fatigue syndrome: Evidence from magnetic resonance spectroscopy of muscle. Neuromuscul. Disord. 1998, 8, 204–209. [Google Scholar] [CrossRef]
- Afari, N.; Buchwald, D. Chronic fatigue syndrome: A review. Am. J. Psychiatry 2003, 160, 221–236. [Google Scholar] [CrossRef]
- Jason, L.A.; Corradi, K.; Torres-Harding, S.; Taylor, R.R.; King, C. Chronic fatigue syndrome: The need for subtypes. Neuropsychol. Rev. 2005, 15, 29–58. [Google Scholar] [CrossRef]
- Morris, G.; Maes, M. Case definitions and diagnostic criteria for Myalgic Encephalomyelitis and Chronic fatigue Syndrome: From clinical-consensus to evidence-based case definitions. Neuroendocrinol. Lett. 2013, 34, 185–199. [Google Scholar] [PubMed]
- Hardcastle, S.L.; Brenu, E.W.; Johnston, S.; Nguyen, T.; Huth, T.; Ramos, S.; Staines, D.; Marshall-Gradisnik, S. Longitudinal analysis of immune abnormalities in varying severities of Chronic Fatigue Syndrome/Myalgic Encephalomyelitis patients. J. Transl. Med. 2015, 13, 299. [Google Scholar] [CrossRef] [PubMed]
- Maclachlan, L.; Watson, S.; Gallagher, P.; Finkelmeyer, A.; Jason, L.A.; Sunnquist, M.; Newton, J.L. Are current chronic fatigue syndrome criteria diagnosing different disease phenotypes? PLoS ONE 2017, 12, e0186885. [Google Scholar] [CrossRef] [PubMed]
- Kerr, J.R.; Petty, R.; Burke, B.; Gough, J.; Fear, D.; Sinclair, L.I.; Mattey, D.L.; Richards, S.C.; Montgomery, J.; Baldwin, D.A.; et al. Gene expression subtypes in patients with chronic fatigue syndrome/myalgic encephalomyelitis. J. Infect. Dis. 2008, 197, 1171–1184. [Google Scholar] [CrossRef] [PubMed]
- Kerr, J.R.; Burke, B.; Petty, R.; Gough, J.; Fear, D.; Mattey, D.L.; Axford, J.S.; Dalgleish, A.G.; Nutt, D.J. Seven genomic subtypes of chronic fatigue syndrome/myalgic encephalomyelitis: A detailed analysis of gene networks and clinical phenotypes. J. Clin. Pathol. 2008, 61, 730–739. [Google Scholar] [CrossRef] [PubMed]
- Zhang, L.; Gough, J.; Christmas, D.; Mattey, D.L.; Richards, S.C.; Main, J.; Enlander, D.; Honeybourne, D.; Ayres, J.G.; Nutt, D.J.; et al. Microbial infections in eight genomic subtypes of chronic fatigue syndrome/myalgic encephalomyelitis. J. Clin. Pathol. 2010, 63, 156–164. [Google Scholar] [CrossRef] [PubMed]
- Light, K.C.; Agarwal, N.; Iacob, E.; White, A.T.; Kinney, A.Y.; VanHaitsma, T.A.; Aizad, H.; Hughen, R.W.; Bateman, L.; Light, A.R. Differing leukocyte gene expression profiles associated with fatigue in patients with prostate cancer versus chronic fatigue syndrome. Psychoneuroendocrinology 2013, 38, 2983–2995. [Google Scholar] [CrossRef] [Green Version]
- De Vega, W.C.; Erdman, L.; Vernon, S.D.; Goldenberg, A.; McGowan, P.O. Integration of DNA methylation & health scores identifies subtypes in myalgic encephalomyelitis/chronic fatigue syndrome. Epigenomics 2018, 10, 539–557. [Google Scholar] [CrossRef]
- Jason, L.A.; Boulton, A.; Porter, N.S.; Jessen, T.; Njoku, M.G.; Friedberg, F. Classification of myalgic encephalomyelitis/chronic fatigue syndrome by types of fatigue. Behav. Med. 2010, 36, 24–31. [Google Scholar] [CrossRef]
- Maes, M.; Leunis, J.C.; Geffard, M.; Berk, M. Evidence for the existence of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) with and without abdominal discomfort (irritable bowel) syndrome. Neuroendocrinol. Lett. 2014, 35, 445–453. [Google Scholar]
- Nagy-Szakal, D.; Barupal, D.K.; Lee, B.; Che, X.; Williams, B.L.; Kahn, E.J.R.; Ukaigwe, J.E.; Bateman, L.; Klimas, N.G.; Komaroff, A.L.; et al. Insights into myalgic encephalomyelitis/chronic fatigue syndrome phenotypes through comprehensive metabolomics. Sci. Rep. 2018, 8, 10056. [Google Scholar] [CrossRef]
- Richardson, A.M.; Lewis, D.P.; Kita, B.; Ludlow, H.; Groome, N.P.; Hedger, M.P.; de Kretser, D.M.; Lidbury, B.A. Weighting of orthostatic intolerance time measurements with standing difficulty score stratifies ME/CFS symptom severity and analyte detection. J. Transl. Med. 2018, 16, 97. [Google Scholar] [CrossRef] [Green Version]
- Russell, L.; Broderick, G.; Taylor, R.; Fernandes, H.; Harvey, J.; Barnes, Z.; Smylie, A.; Collado, F.; Balbin, E.G.; Katz, B.Z.; et al. Illness progression in chronic fatigue syndrome: A shifting immune baseline. BMC Immunol. 2016, 17, 3. [Google Scholar] [CrossRef]
- Craddock, T.J.; Fritsch, P.; Rice, M.A., Jr.; del Rosario, R.M.; Miller, D.B.; Fletcher, M.A.; Klimas, N.G.; Broderick, G. A role for homeostatic drive in the perpetuation of complex chronic illness: Gulf War Illness and chronic fatigue syndrome. PLoS ONE 2014, 9, e84839. [Google Scholar] [CrossRef]
- Behan, W.M.; More, I.A.; Behan, P.O. Mitochondrial abnormalities in the postviral fatigue syndrome. Acta Neuropathol. 1991, 83, 61–65. [Google Scholar] [CrossRef]
- Barnes, P.R.; Taylor, D.J.; Kemp, G.J.; Radda, G.K. Skeletal muscle bioenergetics in the chronic fatigue syndrome. J. Neurol. Neurosurg. Psychiatry 1993, 56, 679–683. [Google Scholar] [CrossRef]
- McCully, K.K.; Natelson, B.H.; Iotti, S.; Sisto, S.; Leigh, J.S., Jr. Reduced oxidative muscle metabolism in chronic fatigue syndrome. Muscle Nerve 1996, 19, 621–625. [Google Scholar] [CrossRef]
- Armstrong, C.W.; McGregor, N.R.; Sheedy, J.R.; Buttfield, I.; Butt, H.L.; Gooley, P.R. NMR metabolic profiling of serum identifies amino acid disturbances in chronic fatigue syndrome. Clin. Chim. Acta 2012, 413, 1525–1531. [Google Scholar] [CrossRef]
- Armstrong, C.W.; McGregor, N.R.; Lewis, D.; Butt, H.; Gooley, P.R. Metabolic profiling reveals anomalous energy metabolism and oxidative stress pathways in chronic fatigue syndrome patients. Metabolomics 2015, 11, 1626–1639. [Google Scholar] [CrossRef]
- Fluge, O.; Mella, O.; Bruland, O.; Risa, K.; Dyrstad, S.E.; Alme, K.; Rekeland, I.G.; Sapkota, D.; Rosland, G.V.; Fossa, A.; et al. Metabolic profiling indicates impaired pyruvate dehydrogenase function in myalgic encephalopathy/chronic fatigue syndrome. JCI Insight 2016, 1, e89376. [Google Scholar] [CrossRef] [Green Version]
- Missailidis, D.; Annesley, S.J.; Allan, C.Y.; Sanislav, O.; Lidbury, B.A.; Lewis, D.P.; Fisher, P.R. An isolated Complex V defect and dysregulated mitochondrial function in immortalized lymphocytes from ME/CFS patients. 2019. submitted. [Google Scholar]
- Castro-Marrero, J.; Cordero, M.D.; Saez-Francas, N.; Jimenez-Gutierrez, C.; Aguilar-Montilla, F.J.; Aliste, L.; Alegre-Martin, J. Could mitochondrial dysfunction be a differentiating marker between chronic fatigue syndrome and fibromyalgia? Antioxid. Redox Signal. 2013, 19, 1855–1860. [Google Scholar] [CrossRef] [PubMed]
- Brown, A.E.; Dibnah, B.; Fisher, E.; Newton, J.L.; Walker, M. Pharmacological activation of AMPK and glucose uptake in cultured human skeletal muscle cells from patients with ME/CFS. Biosci. Rep. 2018, 38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lawson, N.; Hsieh, C.H.; March, D.; Wang, X. Elevated Energy Production in Chronic Fatigue Syndrome Patients. J. Nat. Sci. 2016, 2, e221. [Google Scholar]
- Tomas, C.; Brown, A.; Strassheim, V.; Elson, J.L.; Newton, J.; Manning, P. Cellular bioenergetics is impaired in patients with chronic fatigue syndrome. PLoS ONE 2017, 12, e0186802. [Google Scholar] [CrossRef] [PubMed]
- Hardie, D.G.; Carling, D. The AMP-activated protein kinase-fuel gauge of the mammalian cell? Eur. J. Biochem. 1997, 246, 259–273. [Google Scholar] [CrossRef]
- Ma, X.M.; Blenis, J. Molecular mechanisms of mTOR-mediated translational control. Nat. Rev. Mol. Cell Biol. 2009, 10, 307–318. [Google Scholar] [CrossRef]
- Brown, A.E.; Jones, D.E.; Walker, M.; Newton, J.L. Abnormalities of AMPK activation and glucose uptake in cultured skeletal muscle cells from individuals with chronic fatigue syndrome. PLoS ONE 2015, 10, e0122982. [Google Scholar] [CrossRef]
- Ciregia, F.; Kollipara, L.; Giusti, L.; Zahedi, R.P.; Giacomelli, C.; Mazzoni, M.R.; Giannaccini, G.; Scarpellini, P.; Urbani, A.; Sickmann, A.; et al. Bottom-up proteomics suggests an association between differential expression of mitochondrial proteins and chronic fatigue syndrome. Transl. Psychiatry 2016, 6, e904. [Google Scholar] [CrossRef]
- Kaushik, N.; Fear, D.; Richards, S.C.; McDermott, C.R.; Nuwaysir, E.F.; Kellam, P.; Harrison, T.J.; Wilkinson, R.J.; Tyrrell, D.A.; Holgate, S.T.; et al. Gene expression in peripheral blood mononuclear cells from patients with chronic fatigue syndrome. J. Clin. Pathol. 2005, 58, 826–832. [Google Scholar] [CrossRef]
- Vernon, S.D.; Whistler, T.; Cameron, B.; Hickie, I.B.; Reeves, W.C.; Lloyd, A. Preliminary evidence of mitochondrial dysfunction associated with post-infective fatigue after acute infection with Epstein Barr virus. BMC Infect. Dis. 2006, 6, 15. [Google Scholar] [CrossRef] [PubMed]
- Wallimann, T.; Wyss, M.; Brdiczka, D.; Nicolay, K.; Eppenberger, H.M. Intracellular compartmentation, structure and function of creatine kinase isoenzymes in tissues with high and fluctuating energy demands: The “phosphocreatine circuit” for cellular energy homeostasis. Biochem. J. 1992, 28, 21–40. [Google Scholar] [CrossRef] [PubMed]
- Nguyen, T.; Staines, D.; Johnston, S.; Marshall-Gradisnik, S. Reduced glycolytic reserve in isolated natural killer cells from Myalgic encephalomyelitis/chronic fatigue syndrome patients: A preliminary investigation. Asian Pac. J. Allergy Immunol. 2018. [Google Scholar] [CrossRef]
- Gardiner, C.M.; Finlay, D.K. What Fuels Natural Killers? Metabolism and NK Cell Responses. Front. Immunol. 2017, 8, 367. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Abdel-aleem, S.; Nada, M.A.; Sayed-Ahmed, M.; Hendrickson, S.C.; St Louis, J.; Walthall, H.P.; Lowe, J.E. Regulation of fatty acid oxidation by acetyl-CoA generated from glucose utilization in isolated myocytes. J. Mol. Cell. Cardiol. 1996, 28, 825–833. [Google Scholar] [CrossRef] [PubMed]
- Hardie, D.G.; Pan, D.A. Regulation of fatty acid synthesis and oxidation by the AMP-activated protein kinase. Biochem. Soc. Trans. 2002, 30, 1064–1070. [Google Scholar] [CrossRef] [PubMed]
- Laplante, M.; Sabatini, D.M. mTOR signaling in growth control and disease. Cell 2012, 149, 274–293. [Google Scholar] [CrossRef] [PubMed]
- Hindupur, S.K.; Gonzalez, A.; Hall, M.N. The opposing actions of target of rapamycin and AMP-activated protein kinase in cell growth control. Cold Spring Harb. Perspect. Biol. 2015, 7, a019141. [Google Scholar] [CrossRef]
- Dalle Pezze, P.; Ruf, S.; Sonntag, A.G.; Langelaar-Makkinje, M.; Hall, P.; Heberle, A.M.; Razquin Navas, P.; van Eunen, K.; Tolle, R.C.; Schwarz, J.J.; et al. A systems study reveals concurrent activation of AMPK and mTOR by amino acids. Nat. Commun. 2016, 7, 13254. [Google Scholar] [CrossRef]
- Horton, J.D.; Shah, N.A.; Warrington, J.A.; Anderson, N.N.; Park, S.W.; Brown, M.S.; Goldstein, J.L. Combined analysis of oligonucleotide microarray data from transgenic and knockout mice identifies direct SREBP target genes. Proc. Natl. Acad. Sci. USA 2003, 100, 12027–12032. [Google Scholar] [CrossRef] [Green Version]
- Winder, W.W.; Hardie, D.G. Inactivation of acetyl-CoA carboxylase and activation of AMP-activated protein kinase in muscle during exercise. Am. J. Physiol. 1996, 270, E299–E304. [Google Scholar] [CrossRef] [Green Version]
- Bartlett, K.; Eaton, S. Mitochondrial beta-oxidation. Eur. J. Biochem. 2004, 271, 462–469. [Google Scholar] [CrossRef] [PubMed]
- Liu, T.F.; Vachharajani, V.T.; Yoza, B.K.; McCall, C.E. NAD+-dependent sirtuin 1 and 6 proteins coordinate a switch from glucose to fatty acid oxidation during the acute inflammatory response. J. Biol. Chem. 2012, 287, 25758–25769. [Google Scholar] [CrossRef] [PubMed]
- Komaroff, A.L. Inflammation correlates with symptoms in chronic fatigue syndrome. Proc. Natl. Acad. Sci. USA 2017, 114, 8914–8916. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Maher, K.J.; Klimas, N.G.; Fletcher, M.A. Chronic fatigue syndrome is associated with diminished intracellular perforin. Clin. Exp. Immunol. 2005, 142, 505–511. [Google Scholar] [CrossRef]
- Fletcher, M.A.; Zeng, X.R.; Maher, K.; Levis, S.; Hurwitz, B.; Antoni, M.; Broderick, G.; Klimas, N.G. Biomarkers in chronic fatigue syndrome: Evaluation of natural killer cell function and dipeptidyl peptidase IV/CD26. PLoS ONE 2010, 5, e10817. [Google Scholar] [CrossRef] [PubMed]
- Marshall-Gradisnik, S.; Huth, T.; Chacko, A.; Johnston, S.; Smith, P.; Staines, D. Natural killer cells and single nucleotide polymorphisms of specific ion channels and receptor genes in myalgic encephalomyelitis/chronic fatigue syndrome. Appl. Clin. Genet. 2016, 9, 39–47. [Google Scholar] [CrossRef] [Green Version]
- Lorusso, L.; Mikhaylova, S.V.; Capelli, E.; Ferrari, D.; Ngonga, G.K.; Ricevuti, G. Immunological aspects of chronic fatigue syndrome. Autoimmun. Rev. 2009, 8, 287–291. [Google Scholar] [CrossRef]
- Barker, E.; Fujimura, S.F.; Fadem, M.B.; Landay, A.L.; Levy, J.A. Immunologic abnormalities associated with chronic fatigue syndrome. Clin. Infect. Dis. 1994, 18, S136–S141. [Google Scholar] [CrossRef]
- Tirelli, U.; Marotta, G.; Improta, S.; Pinto, A. Immunological abnormalities in patients with chronic fatigue syndrome. Scand. J. Immunol. 1994, 40, 601–608. [Google Scholar] [CrossRef]
- Brenu, E.W.; van Driel, M.L.; Staines, D.R.; Ashton, K.J.; Ramos, S.B.; Keane, J.; Klimas, N.G.; Marshall-Gradisnik, S.M. Immunological abnormalities as potential biomarkers in Chronic Fatigue Syndrome/Myalgic Encephalomyelitis. J. Transl. Med. 2011, 9, 81. [Google Scholar] [CrossRef]
- Klimas, N.G.; Salvato, F.R.; Morgan, R.; Fletcher, M.A. Immunologic abnormalities in chronic fatigue syndrome. J. Clin. Microbiol. 1990, 28, 1403–1410. [Google Scholar] [Green Version]
- Kawasaki, A.; Shinkai, Y.; Kuwana, Y.; Furuya, A.; Iigo, Y.; Hanai, N.; Itoh, S.; Yagita, H.; Okumura, K. Perforin, a pore-forming protein detectable by monoclonal antibodies, is a functional marker for killer cells. Int. Immunol. 1990, 2, 677–684. [Google Scholar] [CrossRef]
- Osinska, I.; Popko, K.; Demkow, U. Perforin: An important player in immune response. Cent. Eur. J. Immunol. 2014, 39, 109–115. [Google Scholar] [CrossRef]
- Theorell, J.; Bileviciute-Ljungar, I.; Tesi, B.; Schlums, H.; Johnsgaard, M.S.; Asadi-Azarbaijani, B.; Bolle Strand, E.; Bryceson, Y.T. Unperturbed Cytotoxic Lymphocyte Phenotype and Function in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome Patients. Front. Immunol. 2017, 8, 723. [Google Scholar] [CrossRef]
- Cliff, J.M.; King, E.C.; Lee, J.S.; Sepulveda, N.; Wolf, A.S.; Kingdon, C.; Bowman, E.; Dockrell, H.M.; Nacul, L.; Lacerda, E.; et al. Cellular Immune Function in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS). Front. Immunol. 2019, 10, 796. [Google Scholar] [CrossRef]
- Vig, M.; Kinet, J.-P. Calcium signalling in immune cells. Nat. Immunol. 2009, 10, 21–27. [Google Scholar] [CrossRef]
- Rizzuto, R.; De Stefani, D.; Raffaello, A.; Mammucari, C. Mitochondria as sensors and regulators of calcium signalling. Nat. Rev. Mol. Cell Biol. 2012, 13, 566–578. [Google Scholar] [CrossRef]
- Grimm, C.; Kraft, R.; Sauerbruch, S.; Schultz, G.; Harteneck, C. Molecular and functional characterization of the melastatin-related cation channel TRPM3. J. Biol. Chem. 2003, 278, 21493–21501. [Google Scholar] [CrossRef]
- Nguyen, T.; Johnston, S.; Clarke, L.; Smith, P.; Staines, D.; Marshall-Gradisnik, S. Impaired calcium mobilization in natural killer cells from chronic fatigue syndrome/myalgic encephalomyelitis patients is associated with transient receptor potential melastatin 3 ion channels. Clin. Exp. Immunol. 2017, 187, 284–293. [Google Scholar] [CrossRef]
- Cabanas, H.; Muraki, K.; Eaton, N.; Balinas, C.; Staines, D.; Marshall-Gradisnik, S. Loss of Transient Receptor Potential Melastatin 3 ion channel function in natural killer cells from Chronic Fatigue Syndrome/Myalgic Encephalomyelitis patients. Mol. Med. 2018, 24, 44. [Google Scholar] [CrossRef] [PubMed]
- Cabanas, H.; Muraki, K.; Balinas, C.; Eaton-Fitch, N.; Staines, D.; Marshall-Gradisnik, S. Validation of impaired Transient Receptor Potential Melastatin 3 ion channel activity in natural killer cells from Chronic Fatigue Syndrome/ Myalgic Encephalomyelitis patients. Mol. Med. 2019, 25, 14. [Google Scholar] [CrossRef] [Green Version]
- Cost, N.G.; Czyzyk-Krzeska, M.F. Regulation of autophagy by two products of one gene: TRPM3 and miR-204. Mol. Cell. Oncol. 2015, 2, e1002712. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Almenar-Perez, E.; Sanchez-Fito, T.; Ovejero, T.; Nathanson, L.; Oltra, E. Impact of Polypharmacy on Candidate Biomarker miRNomes for the Diagnosis of Fibromyalgia and Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: Striking Back on Treatments. Pharmaceutics 2019, 11, 126. [Google Scholar] [CrossRef]
- Tepikin, A.V. Mitochondrial junctions with cellular organelles: Ca2+ signalling perspective. Pflugers Arch. 2018, 470, 1181–1192. [Google Scholar] [CrossRef] [PubMed]
- Aich, A.; Shaha, C. Novel role of calmodulin in regulating protein transport to mitochondria in a unicellular eukaryote. Mol. Cell. Biol. 2013, 33, 4579–4593. [Google Scholar] [CrossRef] [PubMed]
- Przibilla, J.; Dembla, S.; Rizun, O.; Lis, A.; Jung, M.; Oberwinkler, J.; Beck, A.; Philipp, S.E. Ca2+-dependent regulation and binding of calmodulin to multiple sites of Transient Receptor Potential Melastatin 3 (TRPM3) ion channels. Cell Calcium 2018, 73, 40–52. [Google Scholar] [CrossRef] [PubMed]
- Szymanski, J.; Janikiewicz, J.; Michalska, B.; Patalas-Krawczyk, P.; Perrone, M.; Ziolkowski, W.; Duszynski, J.; Pinton, P.; Dobrzyn, A.; Wieckowski, M.R. Interaction of Mitochondria with the Endoplasmic Reticulum and Plasma Membrane in Calcium Homeostasis, Lipid Trafficking and Mitochondrial Structure. Int. J. Mol. Sci. 2017, 18, 1576. [Google Scholar] [CrossRef]
- Donnelly, R.P.; Loftus, R.M.; Keating, S.E.; Liou, K.T.; Biron, C.A.; Gardiner, C.M.; Finlay, D.K. mTORC1-dependent metabolic reprogramming is a prerequisite for NK cell effector function. J. Immunol. 2014, 193, 4477–4484. [Google Scholar] [CrossRef]
- Viel, S.; Marcais, A.; Guimaraes, F.S.; Loftus, R.; Rabilloud, J.; Grau, M.; Degouve, S.; Djebali, S.; Sanlaville, A.; Charrier, E.; et al. TGF-beta inhibits the activation and functions of NK cells by repressing the mTOR pathway. Sci. Signal. 2016, 9, ra19. [Google Scholar] [CrossRef]
- Salmond, R.J.; Mirchandani, A.S.; Besnard, A.G.; Bain, C.C.; Thomson, N.C.; Liew, F.Y. IL-33 induces innate lymphoid cell-mediated airway inflammation by activating mammalian target of rapamycin. J. Allergy Clin. Immunol. 2012, 130, 1159–1166.e1156. [Google Scholar] [CrossRef] [PubMed]
- Wagner, J.A.; Fehniger, T.A. Memory NK Cells Take Out the (Mitochondrial) Garbage. Immunity 2015, 43, 218–220. [Google Scholar] [CrossRef] [PubMed]
- Morris, G.; Maes, M. Oxidative and Nitrosative Stress and Immune-Inflammatory Pathways in Patients with Myalgic Encephalomyelitis (ME)/Chronic Fatigue Syndrome (CFS). Curr. Neuropharmacol. 2014, 12, 168–185. [Google Scholar] [CrossRef] [PubMed]
- Hornig, M.; Montoya, J.G.; Klimas, N.G.; Levine, S.; Felsenstein, D.; Bateman, L.; Peterson, D.L.; Gottschalk, C.G.; Schultz, A.F.; Che, X.; et al. Distinct plasma immune signatures in ME/CFS are present early in the course of illness. Sci. Adv. 2015, 1. [Google Scholar] [CrossRef] [PubMed]
- Maes, M.; Twisk, F.N.; Ringel, K. Inflammatory and cell-mediated immune biomarkers in myalgic encephalomyelitis/chronic fatigue syndrome and depression: Inflammatory markers are higher in myalgic encephalomyelitis/chronic fatigue syndrome than in depression. Psychother. Psychosom. 2012, 81, 286–295. [Google Scholar] [CrossRef] [PubMed]
- Milrad, S.F.; Hall, D.L.; Jutagir, D.R.; Lattie, E.G.; Ironson, G.H.; Wohlgemuth, W.; Nunez, M.V.; Garcia, L.; Czaja, S.J.; Perdomo, D.M.; et al. Poor sleep quality is associated with greater circulating pro-inflammatory cytokines and severity and frequency of chronic fatigue syndrome/myalgic encephalomyelitis (CFS/ME) symptoms in women. J. NeuroImmunol. 2017, 303, 43–50. [Google Scholar] [CrossRef] [PubMed]
- Montoya, J.G.; Holmes, T.H.; Anderson, J.N.; Maecker, H.T.; Rosenberg-Hasson, Y.; Valencia, I.J.; Chu, L.; Younger, J.W.; Tato, C.M.; Davis, M.M. Cytokine signature associated with disease severity in chronic fatigue syndrome patients. Proc. Natl. Acad. Sci. USA 2017, 114, E7150–E7158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Peterson, D.; Brenu, E.W.; Gottschalk, G.; Ramos, S.; Nguyen, T.; Staines, D.; Marshall-Gradisnik, S. Cytokines in the cerebrospinal fluids of patients with chronic fatigue syndrome/myalgic encephalomyelitis. Mediat. Inflamm. 2015, 2015, 929720. [Google Scholar] [CrossRef]
- Maes, M.; Twisk, F.N.; Kubera, M.; Ringel, K. Evidence for inflammation and activation of cell-mediated immunity in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS): Increased interleukin-1, tumor necrosis factor-alpha, PMN-elastase, lysozyme and neopterin. J. Affect. Disord. 2012, 136, 933–939. [Google Scholar] [CrossRef]
- Tomoda, A.; Joudoi, T.; Rabab, E.M.; Matsumoto, T.; Park, T.H.; Miike, T. Cytokine production and modulation: Comparison of patients with chronic fatigue syndrome and normal controls. Psychiatry Res. 2005, 134, 101–104. [Google Scholar] [CrossRef]
- Fletcher, M.A.; Zeng, X.R.; Barnes, Z.; Levis, S.; Klimas, N.G. Plasma cytokines in women with chronic fatigue syndrome. J. Transl. Med. 2009, 7, 96. [Google Scholar] [CrossRef]
- Mensah, F.K.F.; Bansal, A.S.; Ford, B.; Cambridge, G. Chronic fatigue syndrome and the immune system: Where are we now? Neurophysiol. Clin. 2017, 47, 131–138. [Google Scholar] [CrossRef] [Green Version]
- Maes, M.; Coucke, F.; Leunis, J.C. Normalization of the increased translocation of endotoxin from gram negative enterobacteria (leaky gut) is accompanied by a remission of chronic fatigue syndrome. Neuroendocrinol. Lett. 2007, 28, 739–744. [Google Scholar]
- Maes, M.; Leunis, J.C. Normalization of leaky gut in chronic fatigue syndrome (CFS) is accompanied by a clinical improvement: Effects of age, duration of illness and the translocation of LPS from gram-negative bacteria. Neuroendocrinol. Lett. 2008, 29, 902–910. [Google Scholar]
- Blomberg, J.; Gottfries, C.G.; Elfaitouri, A.; Rizwan, M.; Rosen, A. Infection Elicited Autoimmunity and Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: An Explanatory Model. Front. Immunol. 2018, 9, 229. [Google Scholar] [CrossRef] [Green Version]
- Fluge, O.; Risa, K.; Lunde, S.; Alme, K.; Rekeland, I.G.; Sapkota, D.; Kristoffersen, E.K.; Sorland, K.; Bruland, O.; Dahl, O.; et al. B-Lymphocyte Depletion in Myalgic Encephalopathy/ Chronic Fatigue Syndrome. An Open-Label Phase II Study with Rituximab Maintenance Treatment. PLoS ONE 2015, 10, e0129898. [Google Scholar] [CrossRef]
- Fluge, O.; Mella, O. Clinical impact of B-cell depletion with the anti-CD20 antibody rituximab in chronic fatigue syndrome: A preliminary case series. BMC Neurol. 2009, 9, 28. [Google Scholar] [CrossRef]
- Bradley, A.S.; Ford, B.; Bansal, A.S. Altered functional B cell subset populations in patients with chronic fatigue syndrome compared to healthy controls. Clin. Exp. Immunol. 2013, 172, 73–80. [Google Scholar] [CrossRef]
- Fluge, O.; Rekeland, I.G.; Lien, K.; Thurmer, H.; Borchgrevink, P.C.; Schafer, C.; Sorland, K.; Assmus, J.; Ktoridou-Valen, I.; Herder, I.; et al. B-Lymphocyte Depletion in Patients with Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: A Randomized, Double-Blind, Placebo-Controlled Trial. Ann. Intern. Med. 2019. [Google Scholar] [CrossRef]
- Rekeland, I.G.; Fluge, O.; Alme, K.; Risa, K.; Sorland, K.; Mella, O.; de Vries, A.; Schjott, J. Rituximab Serum Concentrations and Anti-Rituximab Antibodies During B-Cell Depletion Therapy for Myalgic Encephalopathy/Chronic Fatigue Syndrome. Clin. Ther. 2018. [Google Scholar] [CrossRef]
- Loebel, M.; Grabowski, P.; Heidecke, H.; Bauer, S.; Hanitsch, L.G.; Wittke, K.; Meisel, C.; Reinke, P.; Volk, H.D.; Fluge, O.; et al. Antibodies to beta adrenergic and muscarinic cholinergic receptors in patients with Chronic Fatigue Syndrome. Brain Behav. Immun. 2016, 52, 32–39. [Google Scholar] [CrossRef]
- Scheibenbogen, C.; Loebel, M.; Freitag, H.; Krueger, A.; Bauer, S.; Antelmann, M.; Doehner, W.; Scherbakov, N.; Heidecke, H.; Reinke, P.; et al. Immunoadsorption to remove ss2 adrenergic receptor antibodies in Chronic Fatigue Syndrome CFS/ME. PLoS ONE 2018, 13, e0193672. [Google Scholar] [CrossRef]
- Elfaitouri, A.; Herrmann, B.; Bolin-Wiener, A.; Wang, Y.; Gottfries, C.G.; Zachrisson, O.; Pipkorn, R.; Ronnblom, L.; Blomberg, J. Epitopes of microbial and human heat shock protein 60 and their recognition in myalgic encephalomyelitis. PLoS ONE 2013, 8, e81155. [Google Scholar] [CrossRef]
- Maes, M.; Mihaylova, I.; Leunis, J.C. Increased serum IgM antibodies directed against phosphatidyl inositol (Pi) in chronic fatigue syndrome (CFS) and major depression: Evidence that an IgM-mediated immune response against Pi is one factor underpinning the comorbidity between both CFS and depression. Neuroendocrinol. Lett. 2007, 28, 861–867. [Google Scholar]
- Butt, H.L.; Dunstan, R.; McGregor, N.R.; Roberts, T.K. Bacterial colonosis in patients with persistent fatigue. In Proceedings of the AHMF International Clinical and Scientific Conference, Sydney, Australia, 1–2 December 2001. [Google Scholar]
- Sheedy, J.R.; Wettenhall, R.E.; Scanlon, D.; Gooley, P.R.; Lewis, D.P.; McGregor, N.; Stapleton, D.I.; Butt, H.L.; de Meirleir, K.L. Increased d-lactic Acid intestinal bacteria in patients with chronic fatigue syndrome. In Vivo 2009, 23, 621–628. [Google Scholar]
- Jackson, M.L.; Butt, H.; Ball, M.; Lewis, D.P.; Bruck, D. Sleep quality and the treatment of intestinal microbiota imbalance in Chronic Fatigue Syndrome: A pilot study. Sleep Sci. 2015, 8, 124–133. [Google Scholar] [CrossRef] [Green Version]
- Armstrong, C.W.; McGregor, N.R.; Lewis, D.P.; Butt, H.L.; Gooley, P.R. The association of fecal microbiota and fecal, blood serum and urine metabolites in myalgic encephalomyelitis/chronic fatigue syndrome. Metabolomics 2017, 13, 8. [Google Scholar] [CrossRef]
- Fremont, M.; Coomans, D.; Massart, S.; De Meirleir, K. High-throughput 16S rRNA gene sequencing reveals alterations of intestinal microbiota in myalgic encephalomyelitis/chronic fatigue syndrome patients. Anaerobe 2013, 22, 50–56. [Google Scholar] [CrossRef] [Green Version]
- Shukla, S.K.; Cook, D.; Meyer, J.; Vernon, S.D.; Le, T.; Clevidence, D.; Robertson, C.E.; Schrodi, S.J.; Yale, S.; Frank, D.N. Changes in Gut and Plasma Microbiome following Exercise Challenge in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS). PLoS ONE 2015, 10, e0145453. [Google Scholar] [CrossRef]
- Rao, A.V.; Bested, A.C.; Beaulne, T.M.; Katzman, M.A.; Iorio, C.; Berardi, J.M.; Logan, A.C. A randomized, double-blind, placebo-controlled pilot study of a probiotic in emotional symptoms of chronic fatigue syndrome. Gut Pathog. 2009, 1, 6. [Google Scholar] [CrossRef]
- Burnet, R.B.; Chatterton, B.E. Gastric emptying is slow in chronic fatigue syndrome. BMC Gastroenterol. 2004, 4, 32. [Google Scholar] [CrossRef]
- Morris, G.; Berk, M.; Carvalho, A.F.; Caso, J.R.; Sanz, Y.; Maes, M. The Role of Microbiota and Intestinal Permeability in the Pathophysiology of Autoimmune and Neuroimmune Processes with an Emphasis on Inflammatory Bowel Disease Type 1 Diabetes and Chronic Fatigue Syndrome. Curr. Pharm. Des. 2016, 22, 6058–6075. [Google Scholar] [CrossRef]
- Maes, M.; Mihaylova, I.; Leunis, J.C. Increased serum IgA and IgM against LPS of enterobacteria in chronic fatigue syndrome (CFS): Indication for the involvement of gram-negative enterobacteria in the etiology of CFS and for the presence of an increased gut-intestinal permeability. J. Affect. Disord. 2007, 99, 237–240. [Google Scholar] [CrossRef]
- Aaron, L.A.; Burke, M.M.; Buchwald, D. Overlapping conditions among patients with chronic fatigue syndrome, fibromyalgia, and temporomandibular disorder. Arch. Intern. Med. 2000, 160, 221–227. [Google Scholar] [CrossRef]
- Tsai, S.Y.; Chen, H.J.; Lio, C.F.; Kuo, C.F.; Kao, A.C.; Wang, W.S.; Yao, W.C.; Chen, C.; Yang, T.Y. Increased risk of chronic fatigue syndrome in patients with inflammatory bowel disease: A population-based retrospective cohort study. J. Transl. Med. 2019, 17, 55. [Google Scholar] [CrossRef]
- Grainger, J.; Daw, R.; Wemyss, K. Systemic instruction of cell-mediated immunity by the intestinal microbiome. F1000Research 2018, 7. [Google Scholar] [CrossRef]
- Brown, R.L.; Clarke, T.B. The regulation of host defences to infection by the microbiota. Immunology 2017, 150, 1–6. [Google Scholar] [CrossRef]
- Neumann, C.; Blume, J.; Roy, U.; Teh, P.P.; Vasanthakumar, A.; Beller, A.; Liao, Y.; Heinrich, F.; Arenzana, T.L.; Hackney, J.A.; et al. c-Maf-dependent Treg cell control of intestinal TH17 cells and IgA establishes host-microbiota homeostasis. Nat. Immunol. 2019, 20, 471–481. [Google Scholar] [CrossRef]
- Nakajima, A.; Vogelzang, A.; Maruya, M.; Miyajima, M.; Murata, M.; Son, A.; Kuwahara, T.; Tsuruyama, T.; Yamada, S.; Matsuura, M.; et al. IgA regulates the composition and metabolic function of gut microbiota by promoting symbiosis between bacteria. J. Exp. Med. 2018, 215, 2019–2034. [Google Scholar] [CrossRef] [Green Version]
- Kaliannan, K.; Wang, B.; Li, X.Y.; Kim, K.J.; Kang, J.X. A host-microbiome interaction mediates the opposing effects of omega-6 and omega-3 fatty acids on metabolic endotoxemia. Sci. Rep. 2015, 5, 11276. [Google Scholar] [CrossRef]
- Clark, A.; Mach, N. The Crosstalk between the Gut Microbiota and Mitochondria during Exercise. Front. Physiol. 2017, 8, 319. [Google Scholar] [CrossRef]
- Bretin, A.; Gewirtz, A.T.; Chassaing, B. Microbiota and metabolism: what’s new in 2018? Am. J. Physiol. Endocrinol. Metab 2018, 315, E1–E6. [Google Scholar] [CrossRef]
- Janssen, A.W.; Kersten, S. The role of the gut microbiota in metabolic health. FASEB J. 2015, 29, 3111–3123. [Google Scholar] [CrossRef] [Green Version]
- Chambers, E.S.; Preston, T.; Frost, G.; Morrison, D.J. Role of Gut Microbiota-Generated Short-Chain Fatty Acids in Metabolic and Cardiovascular Health. Curr. Nutr. Rep. 2018, 7, 198–206. [Google Scholar] [CrossRef] [Green Version]
- Saint-Georges-Chaumet, Y.; Edeas, M. Microbiota-mitochondria inter-talk: Consequence for microbiota-host interaction. Pathog. Dis. 2016, 74, ftv096. [Google Scholar] [CrossRef]
- Henriksson, A.E.; Tagesson, C.; Uribe, A.; Uvnas-Moberg, K.; Nord, C.E.; Gullberg, R.; Johansson, C. Effects of prostaglandin E2 on disease activity, gastric secretion and intestinal permeability, and morphology in patients with rheumatoid arthritis. Ann. Rheum. Dis. 1988, 47, 620–627. [Google Scholar] [CrossRef]
- Mathew, S.J.; Mao, X.; Keegan, K.A.; Levine, S.M.; Smith, E.L.; Heier, L.A.; Otcheretko, V.; Coplan, J.D.; Shungu, D.C. Ventricular cerebrospinal fluid lactate is increased in chronic fatigue syndrome compared with generalized anxiety disorder: An in vivo 3.0 T 1H MRS imaging study. NMR Biomed. 2009, 22, 251–258. [Google Scholar] [CrossRef]
- Murrough, J.W.; Mao, X.; Collins, K.A.; Kelly, C.; Andrade, G.; Nestadt, P.; Levine, S.M.; Mathew, S.J.; Shungu, D.C. Increased ventricular lactate in chronic fatigue syndrome measured by 1H MRS imaging at 3.0 T. II: Comparison with major depressive disorder. NMR Biomed. 2010, 23, 643–650. [Google Scholar] [CrossRef]
- Shungu, D.C.; Weiduschat, N.; Murrough, J.W.; Mao, X.; Pillemer, S.; Dyke, J.P.; Medow, M.S.; Natelson, B.H.; Stewart, J.M.; Mathew, S.J. Increased ventricular lactate in chronic fatigue syndrome. III. Relationships to cortical glutathione and clinical symptoms implicate oxidative stress in disorder pathophysiology. NMR Biomed. 2012, 25, 1073–1087. [Google Scholar] [CrossRef] [Green Version]
- Rutherford, G.; Manning, P.; Newton, J.L. Understanding Muscle Dysfunction in Chronic Fatigue Syndrome. J. Aging Res. 2016, 2016, 2497348. [Google Scholar] [CrossRef]
- Maes, M.; Twisk, F.N.; Kubera, M.; Ringel, K.; Leunis, J.C.; Geffard, M. Increased IgA responses to the LPS of commensal bacteria is associated with inflammation and activation of cell-mediated immunity in chronic fatigue syndrome. J. Affect. Disord. 2012, 136, 909–917. [Google Scholar] [CrossRef] [PubMed]
- Cadenas, E.; Davies, K.J. Mitochondrial free radical generation, oxidative stress, and aging. Free Radic. Biol. Med. 2000, 29, 222–230. [Google Scholar] [CrossRef]
- Naviaux, R.K. Oxidative shielding or oxidative stress? J. Pharmacol. Exp. Ther. 2012, 342, 608–618. [Google Scholar] [CrossRef] [PubMed]
- Ghosh, S.; Dai, C.; Brown, K.; Rajendiran, E.; Makarenko, S.; Baker, J.; Ma, C.; Halder, S.; Montero, M.; Ionescu, V.A.; et al. Colonic microbiota alters host susceptibility to infectious colitis by modulating inflammation, redox status, and ion transporter gene expression. Am. J. Physiol. Gastrointest. Liver Physiol. 2011, 301, G39–G49. [Google Scholar] [CrossRef]
- Abuaita, B.H.; Schultz, T.L.; O’Riordan, M.X. Mitochondria-Derived Vesicles Deliver Antimicrobial Reactive Oxygen Species to Control Phagosome-Localized Staphylococcus aureus. Cell Host Microbe 2018, 24, 625–636. [Google Scholar] [CrossRef]
- Hurst, J.K.; Lymar, S.V. Toxicity of peroxynitrite and related reactive nitrogen species toward Escherichia coli. Chem. Res. Toxicol. 1997, 10, 802–810. [Google Scholar] [CrossRef]
- Vecchiet, L.; Montanari, G.; Pizzigallo, E.; Iezzi, S.; de Bigontina, P.; Dragani, L.; Vecchiet, J.; Giamberardino, M.A. Sensory characterization of somatic parietal tissues in humans with chronic fatigue syndrome. Neurosci. Lett. 1996, 208, 117–120. [Google Scholar] [CrossRef]
- Vecchiet, J.; Cipollone, F.; Falasca, K.; Mezzetti, A.; Pizzigallo, E.; Bucciarelli, T.; De Laurentis, S.; Affaitati, G.; De Cesare, D.; Giamberardino, M.A. Relationship between musculoskeletal symptoms and blood markers of oxidative stress in patients with chronic fatigue syndrome. Neurosci. Lett. 2003, 335, 151–154. [Google Scholar] [CrossRef]
- Kennedy, G.; Spence, V.A.; McLaren, M.; Hill, A.; Underwood, C.; Belch, J.J. Oxidative stress levels are raised in chronic fatigue syndrome and are associated with clinical symptoms. Free Radic. Biol. Med. 2005, 39, 584–589. [Google Scholar] [CrossRef]
- Jammes, Y.; Steinberg, J.G.; Mambrini, O.; Bregeon, F.; Delliaux, S. Chronic fatigue syndrome: Assessment of increased oxidative stress and altered muscle excitability in response to incremental exercise. J. Intern. Med. 2005, 257, 299–310. [Google Scholar] [CrossRef] [PubMed]
- Jammes, Y.; Steinberg, J.G.; Delliaux, S. Chronic fatigue syndrome: Acute infection and history of physical activity affect resting levels and response to exercise of plasma oxidant/antioxidant status and heat shock proteins. J. Intern. Med. 2012, 272, 74–84. [Google Scholar] [CrossRef] [PubMed]
- Polli, A.; Van Oosterwijck, J.; Nijs, J.; Marusic, U.; De Wandele, I.; Paul, L.; Meeus, M.; Moorkens, G.; Lambrecht, L.; Ickmans, K. Relationship Between Exercise-induced Oxidative Stress Changes and Parasympathetic Activity in Chronic Fatigue Syndrome: An Observational Study in Patients and Healthy Subjects. Clin. Ther. 2019. [Google Scholar] [CrossRef] [PubMed]
- Papatheodorou, P.; Domanska, G.; Oxle, M.; Mathieu, J.; Selchow, O.; Kenny, B.; Rassow, J. The enteropathogenic Escherichia coli (EPEC) Map effector is imported into the mitochondrial matrix by the TOM/Hsp70 system and alters organelle morphology. Cell. Microbiol. 2006, 8, 677–689. [Google Scholar] [CrossRef] [PubMed]
- Crane, J.K.; McNamara, B.P.; Donnenberg, M.S. Role of EspF in host cell death induced by enteropathogenic Escherichia coli. Cell. MicroBiol. 2001, 3, 197–211. [Google Scholar] [CrossRef] [PubMed]
- Swidsinski, A.; Ladhoff, A.; Pernthaler, A.; Swidsinski, S.; Loening-Baucke, V.; Ortner, M.; Weber, J.; Hoffmann, U.; Schreiber, S.; Dietel, M.; et al. Mucosal flora in inflammatory bowel disease. Gastroenterology 2002, 122, 44–54. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nater, U.M.; Maloney, E.; Heim, C.; Reeves, W.C. Cumulative life stress in chronic fatigue syndrome. Psychiatry Res. 2011, 189, 318–320. [Google Scholar] [CrossRef] [PubMed]
- Lakhan, S.E.; Kirchgessner, A. Gut inflammation in chronic fatigue syndrome. Nutr. Metab. 2010, 7, 79. [Google Scholar] [CrossRef] [PubMed]
- Gaab, J.; Rohleder, N.; Heitz, V.; Engert, V.; Schad, T.; Schurmeyer, T.H.; Ehlert, U. Stress-induced changes in LPS-induced pro-inflammatory cytokine production in chronic fatigue syndrome. Psychoneuroendocrinology 2005, 30, 188–198. [Google Scholar] [CrossRef] [PubMed]
- Cani, P.D.; Possemiers, S.; Van de Wiele, T.; Guiot, Y.; Everard, A.; Rottier, O.; Geurts, L.; Naslain, D.; Neyrinck, A.; Lambert, D.M.; et al. Changes in gut microbiota control inflammation in obese mice through a mechanism involving GLP-2-driven improvement of gut permeability. Gut 2009, 58, 1091–1103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Martich, G.D.; Boujoukos, A.J.; Suffredini, A.F. Response of man to endotoxin. Immunobiology 1993, 187, 403–416. [Google Scholar] [CrossRef]
- Nordgreen, J.; Munsterhjelm, C.; Aae, F.; Popova, A.; Boysen, P.; Ranheim, B.; Heinonen, M.; Raszplewicz, J.; Piepponen, P.; Lervik, A.; et al. The effect of lipopolysaccharide (LPS) on inflammatory markers in blood and brain and on Behav. ior in individually-housed pigs. Physiol. Behav. 2018, 195, 98–111. [Google Scholar] [CrossRef] [PubMed]
- Carabotti, M.; Scirocco, A.; Maselli, M.A.; Severi, C. The gut-brain axis: Interactions between enteric microbiota, central and enteric nervous systems. Ann. Gastroenterol. 2015, 28, 203–209. [Google Scholar] [PubMed]
- Furness, J.B. The enteric nervous system: Normal functions and enteric neuropathies. Neurogastroenterol. Motil. 2008, 20 (Suppl. 1), 32–38. [Google Scholar] [CrossRef] [PubMed]
- Barnden, L.R.; Kwiatek, R.; Crouch, B.; Burnet, R.; Del Fante, P. Autonomic correlations with MRI are abnormal in the brainstem vasomotor centre in Chronic Fatigue Syndrome. Neuroimage Clin. 2016, 11, 530–537. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Maes, M.; Mihaylova, I.; Kubera, M.; Uytterhoeven, M.; Vrydags, N.; Bosmans, E. Coenzyme Q10 deficiency in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is related to fatigue, autonomic and neurocognitive symptoms and is another risk factor explaining the early mortality in ME/CFS due to cardiovascular disorder. NeuroEndocrinol. Lett. 2009, 30, 470–476. [Google Scholar] [PubMed]
- Freitas, J.; Santos, R.; Azevedo, E.; Costa, O.; Carvalho, M.; de Freitas, A.F. Reversible sympathetic vasomotor dysfunction in POTS patients. Rev. Port. Cardiol. 2000, 19, 1163–1170. [Google Scholar] [PubMed]
- Tsigos, C.; Chrousos, G.P. Hypothalamic-pituitary-adrenal axis, neuroendocrine factors and stress. J. Psychosom. Res. 2002, 53, 865–871. [Google Scholar] [CrossRef]
- Tak, L.M.; Cleare, A.J.; Ormel, J.; Manoharan, A.; Kok, I.C.; Wessely, S.; Rosmalen, J.G. Meta-analysis and meta-regression of hypothalamic-pituitary-adrenal axis activity in functional somatic disorders. Biol. Psychol. 2011, 87, 183–194. [Google Scholar] [CrossRef]
- Poteliakhoff, A. Adrenocortical activity and some clinical findings in acute and chronic fatigue. J. Psychosom. Res. 1981, 25, 91–95. [Google Scholar] [CrossRef]
- Tomas, C.; Newton, J.; Watson, S. A review of hypothalamic-pituitary-adrenal axis function in chronic fatigue syndrome. ISRN Neurosci. 2013, 2013, 784520. [Google Scholar] [CrossRef]
- Naviaux, R.K. Metabolic features of the cell danger response. Mitochondrion 2014, 16, 7–17. [Google Scholar] [CrossRef] [PubMed]
- Rasa, S.; Nora-Krukle, Z.; Henning, N.; Eliassen, E.; Shikova, E.; Harrer, T.; Scheibenbogen, C.; Murovska, M.; Prusty, B.K.; the European Network on ME/CFS. Chronic viral infections in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). J. Transl. Med. 2018, 16, 268. [Google Scholar] [CrossRef] [PubMed]
- Lacourt, T.E.; Vichaya, E.G.; Chiu, G.S.; Dantzer, R.; Heijnen, C.J. The High Costs of Low-Grade Inflammation: Persistent Fatigue as a Consequence of Reduced Cellular-Energy Availability and Non-adaptive Energy Expenditure. Front. Behav. NeuroSci. 2018, 12, 78. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Morris, G.; Maes, M. Mitochondrial dysfunctions in myalgic encephalomyelitis/chronic fatigue syndrome explained by activated immuno-inflammatory, oxidative and nitrosative stress pathways. Metab. Brain Dis. 2014, 29, 19–36. [Google Scholar] [CrossRef] [PubMed]
- Meeus, M.; Nijs, J.; Hermans, L.; Goubert, D.; Calders, P. The role of mitochondrial dysfunctions due to oxidative and nitrosative stress in the chronic pain or chronic fatigue syndromes and fibromyalgia patients: Peripheral and central mechanisms as therapeutic targets? Expert Opin. Ther. Targets 2013, 17, 1081–1089. [Google Scholar] [CrossRef] [PubMed]
- Van Horssen, J.; van Schaik, P.; Witte, M. Inflammation and mitochondrial dysfunction: A vicious circle in neurodegenerative disorders? Neurosci. Lett. 2017. [Google Scholar] [CrossRef] [PubMed]
- Kolmychkova, K.I.; Zhelankin, A.V.; Karagodin, V.P.; Orekhov, A.N. Mitochondria and inflammation. Patol. Fiziol. Eksp. Ter. 2016, 60, 114–121. [Google Scholar] [PubMed]
- Meyer, L.E.; Machado, L.B.; Santiago, A.P.; da-Silva, W.S.; De Felice, F.G.; Holub, O.; Oliveira, M.F.; Galina, A. Mitochondrial creatine kinase activity prevents reactive oxygen species generation: Antioxidant role of mitochondrial kinase-dependent ADP re-cycling activity. J. Biol. Chem. 2006, 281, 37361–37371. [Google Scholar] [CrossRef]
- Armstrong, C.W.; McGregor, N.R.; Butt, H.L.; Gooley, P.R. Metabolism in chronic fatigue syndrome. Adv. Clin. Chem. 2014, 66, 121–172. [Google Scholar]
- Vermeulen, R.C.; Vermeulen van Eck, I.W. Decreased oxygen extraction during cardiopulmonary exercise test in patients with chronic fatigue syndrome. J. Transl. Med. 2014, 12, 20. [Google Scholar] [CrossRef]
- Blundell, S.; Ray, K.K.; Buckland, M.; White, P.D. Chronic fatigue syndrome and circulating cytokines: A systematic review. Brain Behav. Immun. 2015, 50, 186–195. [Google Scholar] [CrossRef] [PubMed]
- Mensah, F.F.K.; Armstrong, C.W.; Reddy, V.; Bansal, A.S.; Berkovitz, S.; Leandro, M.J.; Cambridge, G. CD24 Expression and B Cell Maturation Shows a Novel Link with Energy Metabolism: Potential Implications for Patients with Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. Front. Immunol. 2018, 9, 2421. [Google Scholar] [CrossRef] [PubMed]
- Mandarano, A.H.; Giloteaux, L.; Keller, B.A.; Levine, S.M.; Hanson, M.R. Eukaryotes in the gut microbiota in myalgic encephalomyelitis/chronic fatigue syndrome. PeerJ 2018, 6, e4282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Allen, J.; Murray, A.; Di Maria, C.; Newton, J.L. Chronic fatigue syndrome and impaired peripheral pulse characteristics on orthostasis—A new potential diagnostic biomarker. Physiol. Meas. 2012, 33, 231–241. [Google Scholar] [CrossRef] [PubMed]
- Demitrack, M.A.; Dale, J.K.; Straus, S.E.; Laue, L.; Listwak, S.J.; Kruesi, M.J.; Chrousos, G.P.; Gold, P.W. Evidence for impaired activation of the hypothalamic-pituitary-adrenal axis in patients with chronic fatigue syndrome. J. Clin. Endocrinol. Metab. 1991, 73, 1224–1234. [Google Scholar] [CrossRef] [PubMed]
- Papadopoulos, A.S.; Cleare, A.J. Hypothalamic-pituitary-adrenal axis dysfunction in chronic fatigue syndrome. Nat. Rev. Endocrinol. 2011, 8, 22–32. [Google Scholar] [CrossRef] [PubMed]
- Berger, E.; Rath, E.; Yuan, D.; Waldschmitt, N.; Khaloian, S.; Allgauer, M.; Staszewski, O.; Lobner, E.M.; Schottl, T.; Giesbertz, P.; et al. Mitochondrial function controls intestinal epithelial stemness and proliferation. Nat. Commun. 2016, 7, 13171. [Google Scholar] [CrossRef]
- Novak, E.A.; Mollen, K.P. Mitochondrial dysfunction in inflammatory bowel disease. Front. Cell Dev. Biol. 2015, 3, 62. [Google Scholar] [CrossRef] [Green Version]
- Madsen, K.L.; Yanchar, N.L.; Sigalet, D.L.; Reigel, T.; Fedorak, R.N. FK506 increases permeability in rat intestine by inhibiting mitochondrial function. Gastroenterology 1995, 109, 107–114. [Google Scholar] [CrossRef]
- Green, D.R.; Galluzzi, L.; Kroemer, G. Mitochondria and the autophagy-inflammation-cell death axis in organismal aging. Science 2011, 333, 1109–1112. [Google Scholar] [CrossRef]
- Walker, M.A.; Volpi, S.; Sims, K.B.; Walter, J.E.; Traggiai, E. Powering the immune system: Mitochondria in immune function and deficiency. J. Immunol. Res. 2014, 2014, 164309. [Google Scholar] [CrossRef]
- Belkaid, Y.; Hand, T.W. Role of the microbiota in immunity and inflammation. Cell 2014, 157, 121–141. [Google Scholar] [CrossRef]
- Wu, H.J.; Wu, E. The role of gut microbiota in immune homeostasis and autoimmunity. Gut Microbes 2012, 3, 4–14. [Google Scholar] [CrossRef] [Green Version]
- Kamada, N.; Nunez, G. Role of the gut microbiota in the development and function of lymphoid cells. J. Immunol. 2013, 190, 1389–1395. [Google Scholar] [CrossRef]
Area of Study | Brief Summary of Key Reports in ME/CFS |
---|---|
Metabolomics |
|
Mitochondrial function |
|
Muscle activity | |
Natural killer cells | |
Calcium signaling | |
Inflammation and cytokines | |
Autoimmunity |
|
B cells | |
Gut microbiota and physiology | |
Autonomic and hormonal dysregulation |
© 2019 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Missailidis, D.; Annesley, S.J.; Fisher, P.R. Pathological Mechanisms Underlying Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. Diagnostics 2019, 9, 80. https://doi.org/10.3390/diagnostics9030080
Missailidis D, Annesley SJ, Fisher PR. Pathological Mechanisms Underlying Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. Diagnostics. 2019; 9(3):80. https://doi.org/10.3390/diagnostics9030080
Chicago/Turabian StyleMissailidis, Daniel, Sarah J. Annesley, and Paul R. Fisher. 2019. "Pathological Mechanisms Underlying Myalgic Encephalomyelitis/Chronic Fatigue Syndrome" Diagnostics 9, no. 3: 80. https://doi.org/10.3390/diagnostics9030080