An Isolated Complex V Inefficiency and Dysregulated Mitochondrial Function in Immortalized Lymphocytes from ME/CFS Patients

Daniel Missailidis, Sarah J Annesley, Claire Y Allan, Oana Sanislav, Brett A Lidbury, Donald P Lewis, Paul R Fisher, Daniel Missailidis, Sarah J Annesley, Claire Y Allan, Oana Sanislav, Brett A Lidbury, Donald P Lewis, Paul R Fisher

Abstract

Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is an enigmatic condition characterized by exacerbation of symptoms after exertion (post-exertional malaise or "PEM"), and by fatigue whose severity and associated requirement for rest are excessive and disproportionate to the fatigue-inducing activity. There is no definitive molecular marker or known underlying pathological mechanism for the condition. Increasing evidence for aberrant energy metabolism suggests a role for mitochondrial dysfunction in ME/CFS. Our objective was therefore to measure mitochondrial function and cellular stress sensing in actively metabolizing patient blood cells. We immortalized lymphoblasts isolated from 51 ME/CFS patients diagnosed according to the Canadian Consensus Criteria and an age- and gender-matched control group. Parameters of mitochondrial function and energy stress sensing were assessed by Seahorse extracellular flux analysis, proteomics, and an array of additional biochemical assays. As a proportion of the basal oxygen consumption rate (OCR), the rate of ATP synthesis by Complex V was significantly reduced in ME/CFS lymphoblasts, while significant elevations were observed in Complex I OCR, maximum OCR, spare respiratory capacity, nonmitochondrial OCR and "proton leak" as a proportion of the basal OCR. This was accompanied by a reduction of mitochondrial membrane potential, chronically hyperactivated TOR Complex I stress signaling and upregulated expression of mitochondrial respiratory complexes, fatty acid transporters, and enzymes of the β-oxidation and TCA cycles. By contrast, mitochondrial mass and genome copy number, as well as glycolytic rates and steady state ATP levels were unchanged. Our results suggest a model in which ME/CFS lymphoblasts have a Complex V defect accompanied by compensatory upregulation of their respiratory capacity that includes the mitochondrial respiratory complexes, membrane transporters and enzymes involved in fatty acid β-oxidation. This homeostatically returns ATP synthesis and steady state levels to "normal" in the resting cells, but may leave them unable to adequately respond to acute increases in energy demand as the relevant homeostatic pathways are already activated.

Keywords: Complex V; TORC1; chronic fatigue syndrome; mitochondria; myalgic encephalomyelitis; seahorse respirometry.

Conflict of interest statement

The authors declare no conflict of interest.

Figures

Figure 1
Figure 1
Ex vivo lymphocytes are metabolically quiescent and myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) lymphocytes die more rapidly. Error bars are standard errors of the mean. (A) Basal oxygen consumption rates (OCR) was measured in lymphoblasts and lymphocytes from ME/CFS and control individuals. Lymphoblasts: each ME/CFS (n = 50) and control (n = 22) cell line was assayed over four replicates in at least three independent experiments. Lymphocytes: each ME/CFS (n = 14) and control (n = 9) cell line was assayed over four replicates once due to limited supply. The red arrows point to the same data magnified with a smaller Y axis scale. The low basal OCRs for lymphocytes match those previously reported [8]. (B) ME/CFS lymphocytes die more rapidly than healthy controls. Lymphocytes from ME/CFS patients (n = 35) and healthy controls (n = 14) were seeded at a density of 1 × 106 viable cells/mL in RPMI 1640 with 10% serum and kept in a humidified 5% CO2 incubator at 37 °C during the experiment. Each point represents the mean percentage of dead cells at the corresponding time point for ex vivo lymphocytes from ME/CFS patients and healthy controls. Stepwise multiple regression analysis was performed with dummy variables allowing both slopes and intercepts to differ between groups, with removal of least significant regression variables until only significant coefficients remained. The difference in the slopes (death rates) of the log-linear regressions between the ME/CFS and control group was statistically significant (t test).
Figure 2
Figure 2
ATP synthesis by Complex V is inefficient in ME/CFS lymphoblasts. Error bars are standard errors of the mean. (A) Basal OCR and OCR by ATP synthesis were unchanged while OCR by ATP synthesis as a% of basal OCR was reduced in ME/CFS lymphoblasts (independent t-test). Each ME/CFS (n = 50) and control (n = 22) cell line was assayed over four replicates in at least three independent experiments. (B) Intracellular ATP concentration (relative background-subtracted luciferase luminescence) is unchanged in ME/CFS lymphoblasts (independent t-test). Each ME/CFS (n = 49) and control (n = 22) cell line was assayed in duplicate within each of at least three independent experiments. Data is normalized to an internal control cell line.
Figure 3
Figure 3
ME/CFS lymphoblasts exhibit elevated respiratory capacity and expression of oxidative phosphorylation (OXPHOS) complexes. Error bars are standard errors of the mean. (A) Complex I OCR, maximum OCR and spare respiratory capacity are elevated in ME/CFS lymphoblasts (independent t-test). The OCR was measured in lymphoblasts from ME/CFS and control individuals by the Seahorse XFe24 Extracellular Flux Analyzer. Each ME/CFS (n = 50) and control (n = 22) cell line was assayed over four replicates in each of at least three independent experiments. (B) Relative expression levels of Complex I subunit NDUFB8, Complex II subunit SdhB and Complex IV subunit COXII were elevated in semiquantitative Western blots (independent t-test). Each ME/CFS (n = 48) and control (n = 17) cell line was assayed in at least three independent experiments. (C) Intracellular ROS levels (relative background-subtracted Deep Red fluorescence) are unchanged in ME/CFS lymphoblasts (independent t-test). Each ME/CFS (n = 49) and control (n = 22) cell line was assayed in duplicate within each of at least three independent experiments. (D) Proton leak as % of basal OCR and the nonmitochondrial OCR are elevated in ME/CFS lymphoblasts (independent t-test). Each ME/CFS (n = 50) and control (n = 22) cell line was assayed over four replicates per experiment in at least three independent experiments.
Figure 4
Figure 4
ME/CFS lymphoblasts exhibit elevated expression of Complexes I and V in their whole cell proteomes. (A) Complex I subunits NDUFB1, NDUFB10, and NDUFS5 were significantly upregulated (iBAQ) in whole cell proteomes (independent t-test). Each ME/CFS (n = 22) and control (n = 16) cell line was sampled once. (B) Complex V subunits ATP5A1, ATP5B, and ATP5H were significantly upregulated (iBAQ) in whole cell mass spectrometry proteomics experiments (independent t-test). Each ME/CFS (n = 22) and control (n = 16) cell line was sampled once. (C) Of the 44 known Complex I subunits, 31 were present amongst the 3700 proteins detected in the whole cell proteomes, 28 of these in both control and ME/CFS lymphoblast samples. Their relative abundances were compared and the majority were elevated in ME/CFS cells (fold change > 1) The background colors separate the two tested proportions in this data: blue corresponding to the samples with reduced abundance and pink for samples with elevated abundance. (D) 12 Complex V subunits were detected within the whole cell proteomes of ME/CFS and control lymphoblasts. Their relative abundances were compared to the controls and the majority were elevated in ME/CFS cells (fold change > 1). The background colors indicate the samples with reduced abundance (blue) and samples with elevated abundance (pink).
Figure 5
Figure 5
The respiratory shift in ME/CFS lymphoblasts is correlated with disease severity. ATP synthesis as a percentage of basal respiration and the elevated proton leak, maximum, nonmitochondrial and Complex I OCRs correlate with Weighted Standing Test score, a measure of disease severity in which lower values indicate more severe clinical presentation (Pearson correlation with indicated significance) (ME/CFS n = 45, control n = 17). Patients whose illness was so severe as to preclude them taking the test were not included. Lines fitted by linear least squares. Outliers falling outside 95% confidence limits were excluded.
Figure 6
Figure 6
Mitochondrial genome copy number and mass per cell are unchanged in ME/CFS lymphoblasts, but mitochondrial membrane potential is lowered. Error bars represent standard errors of the mean. (A) Mitochondrial mass (background-subtracted Mitotracker Green fluorescence normalized to an internal control cell line) and genome copy number (qPCR of two mitochondrial genes, mtND1 and mtND4, relative to nuclear β2-microglobulin gene) are unchanged in ME/CFS lymphoblasts (independent t-test). Each ME/CFS (n = 50) and control cell line (n = 22) was assayed in duplicate within each of at least three independent experiments. (B) Mitochondrial membrane potential is significantly reduced in ME/CFS lymphoblasts (independent t-test). The relative mitochondrial membrane potential (Δψm) was measured in lymphoblasts from ME/CFS and control individuals (ratio of MitoTracker® Red CMXRos (Δψm-dependent) to MitoTracker® Green (mitochondrial mass-dependent) fluorescence, normalized to an internal control cell line). Each ME/CFS (n = 50) and control (n = 22) cell line was assayed in duplicate within each of at least three independent experiments.
Figure 7
Figure 7
Glycolysis rates are unaffected in ME/CFS lymphoblasts, but levels of enzymes involved in fatty acid β-oxidation and the TCA cycle are elevated Error bars represent standard errors of the mean. (A) Glycolytic rate, capacity and reserve are unchanged in ME/CFS lymphoblasts (independent t-test). The ECAR was measured in lymphoblasts from ME/CFS and control individuals by the Seahorse XFe24 Extracellular Flux Analyzer. Each ME/CFS (n = 23) and control (n = 16) cell line was assayed over four replicates in at least three independent experiments. (B) Fatty acid carrier carnitine O-palmitoyltransferase 2 and β-oxidation enzyme enoyl-CoA delta isomerase 2 were significantly upregulated (iBAQ) in whole cell mass spectrometry proteomics experiments (independent t-test). Each ME/CFS (n = 22) and control (n = 16) cell line was sampled once. (C) 20 proteins involved in mitochondrial β-oxidation were detected within the whole cell proteomes of ME/CFS and control lymphoblasts. Their relative abundances were compared to the controls and the majority were elevated in ME/CFS cells (fold change > 1). The background colors separate the two tested proportions in this data: blue corresponding to the samples with reduced abundance and pink for samples with elevated abundance. (D) 19 proteins involved in the TCA cycle were detected within the whole cell proteomes of ME/CFS and control lymphoblasts. Their relative abundances were compared to the controls and the majority were elevated in ME/CFS cells (fold change > 1). The background colors indicate the samples with reduced abundance (blue) and samples with elevated abundance (pink).
Figure 8
Figure 8
Stress sensing pathways in ME/CFS are perturbed—TORC1 (Target of Rapamycin Complex I) is chronically hyperactivated. Error bars represent standard errors of the mean. TORC1 activity and response to TORIN2. inhibition is elevated in ME/CFS lymphoblasts (independent t-test). Each ME/CFS (n = 45) and control (n = 22) cell line was assayed over at least three independent experiments. Data is expressed in relative terms as each experiment is normalized to an internal control cell line.

References

    1. 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. doi: 10.1111/j.1365-2796.2011.02428.x.
    1. Afari N., Buchwald D. Chronic fatigue syndrome: A review. Am. J. Psychiatry. 2003;160:221–236. doi: 10.1176/appi.ajp.160.2.221.
    1. 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. doi: 10.1007/s11011-013-9435-x.
    1. Smits B., van den Heuvel L., Knoop H., Kusters B., Janssen A., Borm G., Bleijenberg G., Rodenburg R., van Engelen B. Mitochondrial enzymes discriminate between mitochondrial disorders and chronic fatigue syndrome. Mitochondrion. 2011;11:735–738. doi: 10.1016/j.mito.2011.05.005.
    1. O’Neill H.M., Maarbjerg S.J., Crane J.D., Jeppesen J., Jorgensen S.B., Schertzer J.D., Shyroka O., Kiens B., van Denderen B.J., Tarnopolsky M.A., et al. AMP-activated protein kinase (AMPK) beta1beta2 muscle null mice reveal an essential role for AMPK in maintaining mitochondrial content and glucose uptake during exercise. Proc. Natl. Acad. Sci. USA. 2011;108:16092–16097. doi: 10.1073/pnas.1105062108.
    1. Myhill S., Booth N.E., McLaren-Howard J. Chronic fatigue syndrome and mitochondrial dysfunction. Int. J. Clin. Exp. Med. 2009;2:1–16.
    1. 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.
    1. 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. doi: 10.1371/journal.pone.0186802.
    1. Tomas C., Lodge T.A., Potter M., Elson J.L., Newton J.L., Morten K.J. Assessing cellular energy dysfunction in CFS/ME using a commercially available laboratory test. Sci. Rep. 2019;9:11464. doi: 10.1038/s41598-019-47966-z.
    1. Vermeulen R.C., Kurk R.M., Visser F.C., Sluiter W., Scholte H.R. Patients with chronic fatigue syndrome performed worse than controls in a controlled repeated exercise study despite a normal oxidative phosphorylation capacity. J. Transl. Med. 2010;8:93. doi: 10.1186/1479-5876-8-93.
    1. Lawson N., Hsieh C.H., March D., Wang X. Elevated Energy Production in Chronic Fatigue Syndrome Patients. J. Nat. Sci. 2016;2
    1. 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. doi: 10.1136/jcp.2005.025718.
    1. 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. doi: 10.1038/tp.2016.184.
    1. Nelson C., Ambros V., Baehrecke E.H. miR-14 regulates autophagy during developmental cell death by targeting ip3-kinase 2. Mol. Cell. 2014;56:376–388. doi: 10.1016/j.molcel.2014.09.011.
    1. Mandarano A.H., Maya J., Giloteaux L., Peterson D.L., Maynard M., Gottschalk C.G., Hanson M.R. Myalgic encephalomyelitis/chronic fatigue syndrome patients exhibit altered T cell metabolism and cytokine associations. J. Clin. Invest. 2019 doi: 10.1172/JCI132185.
    1. 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. doi: 10.1007/s11306-015-0816-5.
    1. 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. doi: 10.1172/jci.insight.89376.
    1. Loewith R., Hall M.N. Target of rapamycin (TOR) in nutrient signaling and growth control. Genetics. 2011;189:1177–1201. doi: 10.1534/genetics.111.133363.
    1. Cunningham J.T., Rodgers J.T., Arlow D.H., Vazquez F., Mootha V.K., Puigserver P. mTOR controls mitochondrial oxidative function through a YY1-PGC-1alpha transcriptional complex. Nature. 2007;450:736–740. doi: 10.1038/nature06322.
    1. Morita M., Gravel S.P., Hulea L., Larsson O., Pollak M., St-Pierre J., Topisirovic I. mTOR coordinates protein synthesis, mitochondrial activity and proliferation. Cell Cycle. 2015;14:473–480. doi: 10.4161/15384101.2014.991572.
    1. Annesley S.J., Lay S.T., De Piazza S.W., Sanislav O., Hammersley E., Allan C.Y., Francione L.M., Bui M.Q., Chen Z.P., Ngoei K.R., et al. Immortalized Parkinson’s disease lymphocytes have enhanced mitochondrial respiratory activity. Dis. Model. Mech. 2016;9:1295–1305. doi: 10.1242/dmm.025684.
    1. Loesch D.Z., Annesley S.J., Trost N., Bui M.Q., Lay S.T., Storey E., De Piazza S.W., Sanislav O., Francione L.M., Hammersley E.M., et al. Novel Blood Biomarkers Are Associated with White Matter Lesions in Fragile X- Associated Tremor/Ataxia Syndrome. Neurodegener. Dis. 2017;17:22–30. doi: 10.1159/000446803.
    1. 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. doi: 10.1186/1471-2334-6-15.
    1. 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. doi: 10.1186/s12967-018-1473-z.
    1. Bereiter-Hahn J., Voth M. Dynamics of mitochondria in living cells: Shape changes, dislocations, fusion, and fission of mitochondria. Microsc. Res. Tech. 1994;27:198–219. doi: 10.1002/jemt.1070270303.
    1. 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 doi: 10.12932/AP-011117-0188.
    1. 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. doi: 10.1038/s41598-018-28477-9.
    1. 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. doi: 10.1039/C6MB00600K.
    1. 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. doi: 10.1073/pnas.1607571113.
    1. Carling D., Mayer F.V., Sanders M.J., Gamblin S.J. AMP-activated protein kinase: Nature’s energy sensor. Nat. Chem. Biol. 2011;7:512–518. doi: 10.1038/nchembio.610.
    1. 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. doi: 10.1038/ncomms13254.
    1. Hardie D.G. Adenosine monophosphate-activated protein kinase: A central regulator of metabolism with roles in diabetes, cancer, and viral infection. Cold Spring Harb. Symp. Quant. Biol. 2011;76:155–164. doi: 10.1101/sqb.2011.76.010819.
    1. 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. doi: 10.1101/cshperspect.a019141.
    1. Reznick R.M., Shulman G.I. The role of AMP-activated protein kinase in mitochondrial biogenesis. J. Physiol. 2006;574:33–39. doi: 10.1113/jphysiol.2006.109512.
    1. Zong H., Ren J.M., Young L.H., Pypaert M., Mu J., Birnbaum M.J., Shulman G.I. AMP kinase is required for mitochondrial biogenesis in skeletal muscle in response to chronic energy deprivation. Proc. Natl. Acad. Sci. USA. 2002;99:15983–15987. doi: 10.1073/pnas.252625599.
    1. Ma X.M., Blenis J. Molecular mechanisms of mTOR-mediated translational control. Nat Rev. Mol. Cell Biol. 2009;10:307–318. doi: 10.1038/nrm2672.
    1. Dowling R.J., Topisirovic I., Alain T., Bidinosti M., Fonseca B.D., Petroulakis E., Wang X., Larsson O., Selvaraj A., Liu Y., et al. mTORC1-mediated cell proliferation, but not cell growth, controlled by the 4E-BPs. Science. 2010;328:1172–1176. doi: 10.1126/science.1187532.
    1. Hsieh A.C., Costa M., Zollo O., Davis C., Feldman M.E., Testa J.R., Meyuhas O., Shokat K.M., Ruggero D. Genetic dissection of the oncogenic mTOR pathway reveals druggable addiction to translational control via 4EBP-eIF4E. Cancer Cell. 2010;17:249–261. doi: 10.1016/j.ccr.2010.01.021.
    1. She Q.B., Halilovic E., Ye Q., Zhen W., Shirasawa S., Sasazuki T., Solit D.B., Rosen N. 4E-BP1 is a key effector of the oncogenic activation of the AKT and ERK signaling pathways that integrates their function in tumors. Cancer Cell. 2010;18:39–51. doi: 10.1016/j.ccr.2010.05.023.
    1. Qin X., Jiang B., Zhang Y. 4E-BP1, a multifactor regulated multifunctional protein. Cell Cycle. 2016;15:781–786. doi: 10.1080/15384101.2016.1151581.
    1. Tomas C., Brown A.E., Newton J.L., Elson J.L. Mitochondrial complex activity in permeabilised cells of chronic fatigue syndrome patients using two cell types. PeerJ. 2019;7:e6500. doi: 10.7717/peerj.6500.
    1. Wolvetang E.J., Johnson K.L., Krauer K., Ralph S.J., Linnane A.W. Mitochondrial respiratory chain inhibitors induce apoptosis. FEBS Lett. 1994;339:40–44. doi: 10.1016/0014-5793(94)80380-3.
    1. Ransohoff R.M., Schafer D., Vincent A., Blachere N.E., Bar-Or A. Neuroinflammation: Ways in Which the Immune System Affects the Brain. Neurotherapeutics. 2015;12:896–909. doi: 10.1007/s13311-015-0385-3.
    1. Hussain T., Mulherkar R. Lymphoblastoid Cell lines: A Continuous in Vitro Source of Cells to Study Carcinogen Sensitivity and DNA Repair. Int. J. Mol. Cell Med. 2012;1:75–87.
    1. Sie L., Loong S., Tan E.K. Utility of lymphoblastoid cell lines. J. Neurosci. Res. 2009;87:1953–1959. doi: 10.1002/jnr.22000.
    1. Genomes Project C., Auton A., Brooks L.D., Durbin R.M., Garrison E.P., Kang H.M., Korbel J.O., Marchini J.L., McCarthy S., McVean G.A., et al. A global reference for human genetic variation. Nature. 2015;526:68–74. doi: 10.1038/nature15393.
    1. Cortes Rivera M., Mastronardi C., Silva-Aldana C.T., Arcos-Burgos M., Lidbury B.A. Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: A Comprehensive Review. Diagnostics (Basel) 2019;9:91. doi: 10.3390/diagnostics9030091.
    1. Hernando H., Shannon-Lowe C., Islam A.B., Al-Shahrour F., Rodriguez-Ubreva J., Rodriguez-Cortez V.C., Javierre B.M., Mangas C., Fernandez A.F., Parra M., et al. The B cell transcription program mediates hypomethylation and overexpression of key genes in Epstein-Barr virus-associated proliferative conversion. Genome Biol. 2013;14:R3. doi: 10.1186/gb-2013-14-1-r3.
    1. Bigot A., Jacquemin V., Debacq-Chainiaux F., Butler-Browne G.S., Toussaint O., Furling D., Mouly V. Replicative aging down-regulates the myogenic regulatory factors in human myoblasts. Biol. Cell. 2008;100:189–199. doi: 10.1042/BC20070085.
    1. Huttenlocher A., Lakonishok M., Kinder M., Wu S., Truong T., Knudsen K.A., Horwitz A.F. Integrin and cadherin synergy regulates contact inhibition of migration and motile activity. J. Cell Biol. 1998;141:515–526. doi: 10.1083/jcb.141.2.515.
    1. Schlauch K.A., Khaiboullina S.F., De Meirleir K.L., Rawat S., Petereit J., Rizvanov A.A., Blatt N., Mijatovic T., Kulick D., Palotas A., et al. Genome-wide association analysis identifies genetic variations in subjects with myalgic encephalomyelitis/chronic fatigue syndrome. Transl. Psychiatry. 2016;6:e730. doi: 10.1038/tp.2015.208.
    1. Billing-Ross P., Germain A., Ye K., Keinan A., Gu Z., Hanson M.R. Mitochondrial DNA variants correlate with symptoms in myalgic encephalomyelitis/chronic fatigue syndrome. J. Transl. Med. 2016;14:19. doi: 10.1186/s12967-016-0771-6.
    1. Campanella M., Seraphim A., Abeti R., Casswell E., Echave P., Duchen M.R. IF1, the endogenous regulator of the F(1)F(o)-ATPsynthase, defines mitochondrial volume fraction in HeLa cells by regulating autophagy. Biochim. Biophys. Acta. 2009;1787:393–401. doi: 10.1016/j.bbabio.2009.02.023.
    1. Garcia-Bermudez J., Cuezva J.M. The ATPase Inhibitory Factor 1 (IF1): A master regulator of energy metabolism and of cell survival. Biochim. Biophys. Acta. 2016;1857:1167–1182. doi: 10.1016/j.bbabio.2016.02.004.
    1. Gilardini Montani M.S., Santarelli R., Granato M., Gonnella R., Torrisi M.R., Faggioni A., Cirone M. EBV reduces autophagy, intracellular ROS and mitochondria to impair monocyte survival and differentiation. Autophagy. 2019;15:652–667. doi: 10.1080/15548627.2018.1536530.
    1. Anand S.K., Tikoo S.K. Viruses as modulators of mitochondrial functions. Adv. Virol. 2013;2013:738794. doi: 10.1155/2013/738794.
    1. Woods A., Dickerson K., Heath R., Hong S.P., Momcilovic M., Johnstone S.R., Carlson M., Carling D. Ca2+/calmodulin-dependent protein kinase kinase-beta acts upstream of AMP-activated protein kinase in mammalian cells. Cell Metab. 2005;2:21–33. doi: 10.1016/j.cmet.2005.06.005.
    1. Hawley S.A., Pan D.A., Mustard K.J., Ross L., Bain J., Edelman A.M., Frenguelli B.G., Hardie D.G. Calmodulin-dependent protein kinase kinase-beta is an alternative upstream kinase for AMP-activated protein kinase. Cell Metab. 2005;2:9–19. doi: 10.1016/j.cmet.2005.05.009.
    1. Hardie D.G., Carling D. The AMP-activated protein kinase--fuel gauge of the mammalian cell? Eur. J. Biochem. 1997;246:259–273. doi: 10.1111/j.1432-1033.1997.00259.x.
    1. Bokko P.B., Francione L., Bandala-Sanchez E., Ahmed A.U., Annesley S.J., Huang X., Khurana T., Kimmel A.R., Fisher P.R. Diverse cytopathologies in mitochondrial disease are caused by AMP-activated protein kinase signaling. Mol. Biol. Cell. 2007;18:1874–1886. doi: 10.1091/mbc.e06-09-0881.
    1. 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. doi: 10.1042/bst0301064.
    1. 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. doi: 10.1371/journal.pone.0122982.
    1. 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. Chronic Fatigue Syndr. 2003;11:7–36. doi: 10.1300/J092v11n01_02.
    1. Pendergrass W., Wolf N., Poot M. Efficacy of MitoTracker Green and CMXrosamine to measure changes in mitochondrial membrane potentials in living cells and tissues. Cytometry A. 2004;61:162–169. doi: 10.1002/cyto.a.20033.
    1. Lay M.L., Lucas R.M., Ratnamohan M., Taylor J., Ponsonby A.L., Dwyer D.E., Ausimmune Investigator G. Measurement of Epstein-Barr virus DNA load using a novel quantification standard containing two EBV DNA targets and SYBR Green I dye. Virol. J. 2010;7:252. doi: 10.1186/1743-422X-7-252.
    1. Fox J. The R commander: A basic-statistics graphical user interface to R. J. Stat. Softw. 2005;14:1–42. doi: 10.18637/jss.v014.i09.
    1. Kanda Y. Investigation of the freely available easy-to-use software ‘EZR’ for medical statistics. Bone Marrow Transplant. 2013;48:452–458. doi: 10.1038/bmt.2012.244.
    1. Harris M.A., Clark J., Ireland A., Lomax J., Ashburner M., Foulger R., Eilbeck K., Lewis S., Marshall B., Mungall C., et al. The Gene Ontology (GO) database and informatics resource. Nucleic Acids Res. 2004;32:D258–D261. doi: 10.1093/nar/gkh036.
    1. 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 doi: 10.1042/BSR20180242.

Source: PubMed

3
Prenumerera