Impact of Long-Term Cryopreservation on Blood Immune Cell Markers in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: Implications for Biomarker Discovery

Elisabet Gómez-Mora, Jorge Carrillo, Víctor Urrea, Josepa Rigau, José Alegre, Cecilia Cabrera, Elisa Oltra, Jesús Castro-Marrero, Julià Blanco, Elisabet Gómez-Mora, Jorge Carrillo, Víctor Urrea, Josepa Rigau, José Alegre, Cecilia Cabrera, Elisa Oltra, Jesús Castro-Marrero, Julià Blanco

Abstract

Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) is a complex neuroimmune disorder characterized by numerous symptoms of unknown etiology. The ME/CFS immune markers reported so far have failed to generate a clinical consensus, perhaps partly due to the limitations of biospecimen biobanking. To address this issue, we performed a comparative analysis of the impact of long-term biobanking on previously identified immune markers and also explored additional potential immune markers linked to infection in ME/CFS. A correlation analysis of marker cryostability across immune cell subsets based on flow cytometry immunophenotyping of fresh blood and frozen PBMC samples collected from individuals with ME/CFS (n = 18) and matched healthy controls (n = 18) was performed. The functionality of biobanked samples was assessed on the basis of cytokine production assay after stimulation of frozen PBMCs. T cell markers defining Treg subsets and the expression of surface glycoprotein CD56 in T cells and the frequency of the effector CD8 T cells, together with CD57 expression in NK cells, appeared unaltered by biobanking. By contrast, NK cell markers CD25 and CD69 were notably increased, and NKp46 expression markedly reduced, by long-term cryopreservation and thawing. Further exploration of Treg and NK cell subsets failed to identify significant differences between ME/CFS patients and healthy controls in terms of biobanked PBMCs. Our findings show that some of the previously identified immune markers in T and NK cell subsets become unstable after cell biobanking, thus limiting their use in further immunophenotyping studies for ME/CFS. These data are potentially relevant for future multisite intervention studies and cooperative projects for biomarker discovery using ME/CFS biobanked samples. Further studies are needed to develop novel tools for the assessment of biomarker stability in cryopreserved immune cells from people with ME/CFS.

Keywords: chronic fatigue syndrome; cryopreservation; freeze-thaw process; immune biomarkers; immunophenotyping; myalgic encephalomyelitis.

Conflict of interest statement

JR is an employee at the Rigau Private Clinic. JB is the founder and CEO and JC is also co-founder and CSO (AlbaJuna Therapeutics, Barcelona, Spain). The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2020 Gómez-Mora, Carrillo, Urrea, Rigau, Alegre, Cabrera, Oltra, Castro-Marrero and Blanco.

Figures

Figure 1
Figure 1
Analysis of T cell subset markers in fresh and frozen samples from ME/CFS patients (A–D). Frequencies of the analyzed T cell subsets in fresh (2013) and frozen (2016) PBMC samples from the same participants are shown. Complete identity between fresh and frozen cells is illustrated by blue dotted lines. Linear regression of data is illustrated by red lines. Spearman correlation coefficients and P-values are shown for each panel.
Figure 2
Figure 2
Analysis of T cell subset markers in frozen PBMC samples from individuals with ME/CFS and healthy controls. The indicated T cell subsets were assessed in frozen PBMC samples from 18 ME/CFS individuals (brown) and 18 healthy controls (green). Data is shown as median values with interquartile range (boxes), plus minimal and maximal observations (bars). P-values are indicated for each set of groups compared.
Figure 3
Figure 3
Analysis of NK cell subset markers in fresh and frozen samples from ME/CFS patients (A–D). Frequencies of the analyzed NK cell subsets in fresh (2013) and frozen (2016) blood samples from the same participants are shown. As in Figure 1, complete identity between fresh and frozen cells is illustrated by blue dotted lines. Linear regression of data is illustrated by red lines. Spearman correlation coefficients and P-values are shown for each panel.
Figure 4
Figure 4
Analysis of new Treg subset markers. (A) Gating strategy of Treg cells, first identified by high CD25+ and FOXP3+ expression (classic Treg) and defined as natural Tregs by the lack of CD127 expression. Coexpression of CD45RA, FOXP3 or co-expression of CD39 and CD73 as new potential Treg markers were assessed as indicated. (B, C) Show the frequencies of the indicated cell subsets obtained for healthy controls (n = 18; green) and ME/CFS patients (n = 18; brown). Data are presented as median with interquartile range (boxes) plus minimal and maximal values (bars). P-values are indicated for each set of groups compared.
Figure 5
Figure 5
Analysis of cytokine expression in frozen/thawed stimulated samples. (A) Representative example of cytokine production from ME/CFS PBMCs by gated CD4+ T cells. IFN-γ and IL-17 pro-inflammatory cytokines, and IL-4 and TFG-β1 anti-inflammatory cytokines were evaluated as indicated. (B) Percentages of CD4+ cytokine producing T cells are shown for healthy controls (n = 18; green) and ME/CFS patients (n = 18; brown). Data are shown as median with interquartile range (boxes) and minimal and maximal values (bars). Unstimulated conditions were included as negative controls in each experiment. P-values are indicated for each set compared.
Figure 6
Figure 6
Analysis of new NK cell markers. (A) Representative dot-plots for the co-expression of CD57 and either NKG2C or NKp46 in gated CD56+CD16+NK cells. (B) Correlation between CD57 and NKp46 expression in CD56+CD16+ NK cells from frozen PBMC samples (18 healthy controls in green and 18 ME/CFS patients in brown). Linear regression of data is illustrated by a red line. Spearman correlation coefficients and P-values are shown. (C, D) Marker coexpression for healthy controls (n = 18; green) and individuals with ME/CFS (n = 18; brown) are shown as indicated. Data are shown as median with interquartile ranges (boxes) plus minimal and maximal values (bars). P-values are indicated for each set compared.

References

    1. Institute of Medicine (IOM) Beyond myalgic encephalomyelitis/chronic fatigue syndrome: Redefining an illness. Washington, DC: The National Academies Press; (2015).
    1. Castro-Marrero J, Sáez-Francàs N, Santillo D, Alegre J. Treatment and management of chronic fatigue syndrome/myalgic encephalomyelitis: All roads lead to Rome. Br J Pharmacol (2017) 174:345–69. 10.1111/bph.13702
    1. Carruthers BM, van de Sande MI, De Meirleir KL, Klimas NG, Broderick G, Mitchell T, et al. Myalgic encephalomyelitis: International Consensus Criteria. J Intern Med (2011) 270:327–38. 10.1111/j.1365-2796.2011.02428.x
    1. Bansal AS, Bradley AS, Bishop KN, Kiani-Alikhan S, Ford B. Chronic fatigue syndrome, the immune system and viral infection. Brain Behav Immun (2012) 26:24–31. 10.1016/j.bbi.2011.06.016
    1. Klimas NG, Salvato FR, Morgan R, Fletcher MA. Immunologic abnormalities in chronic fatigue syndrome. J Clin Microbiol (1990) 28:1403–10. 10.1128/JCM.28.6.1403-1410.1990
    1. Brenu EW, van Driel ML, Staines DR, Ashton KJ, Hardcastle SL, Keane J, et al. Longitudinal investigation of natural killer cells and cytokines in chronic fatigue syndrome/myalgic encephalomyelitis. J Transl Med (2012) 10:88. 10.1186/1479-5876-10-88
    1. Curriu M, Carrillo J, Massanella M, Rigau J, Alegre J, Puig J, et al. B and T cell phenotype and function in patients suffering from Chronic Fatigue Syndrome. J Transl Med (2013) 11:68. 10.1186/1479-5876-11-68
    1. Brenu EW, Staines DR, Baskurt OK, Ashton KJ, Ramos SB, Christy RM, et al. Immune and hemorheological changes in chronic fatigue syndrome. J Transl Med (2010) 8:1. 10.1186/1479-5876-8-1
    1. Brenu EW, van Driel ML, Staines DR, Ashton KJ, Ramos SB, Keane J, et al. Immunological abnormalities as potential biomarkers in chronic fatigue syndrome/myalgic encephalomyelitis. J Transl Med (2011) 9:81. 10.1186/1479-5876-9-81
    1. Strawbridge R, Sartor M-L, Scott F, Cleare AJ. Inflammatory proteins are altered in chronic fatigue syndrome: A systematic review and meta-analysis. Neurosci Biobehav Rev (2019) 107:69–83. 10.1016/j.neubiorev.2019.08.011
    1. Irlbeck DM, Vernon SD, McCleary KK, Bateman L, Klimas NG, Lapp CW, et al. No association found between the detection of either xenotropic murine leukemia virus-related virus or polytropic murine leukemia virus and chronic fatigue syndrome in a blinded, multi-site, prospective study by the establishment and use of the SolveCFS Biobank. BMC Res Notes (2014) 7:461. 10.1186/1756-0500-7-461
    1. Lacerda EM, Mudie K, Kingdon CC, Butterworth JD, O’Boyle S, Nacul L. The UK ME/CFS biobank: a disease-specific biobank for advancing clinical research into myalgic encephalomyelitis/chronic fatigue syndrome. Front Neurol (2018) 9:1026. 10.3389/fneur.2018.01026
    1. Fukuda K, Straus SE, Hickie I, Sharpe MC, Dobbins JG, Komaroff A. The chronic fatigue syndrome: a comprehensive approach to its definition and study. International Chronic Fatigue Syndrome Study Group. Ann Intern Med (1994) 121:953–9. 10.7326/0003-4819-121-12-199412150-00009
    1. Fisk JD, Ritvo PG, Ross L, Haase DA, Marrie TJ, Schlech WF. Measuring the functional impact of fatigue: Initial validation of the fatigue impact scale. Clin Infect Dis (1994) 18(Suppl. 1):S79–83. 10.1093/clinids/18.supplement_1.s79
    1. Castro-Marrero J, Zaragozà MC, Domingo JC, Martinez-Martinez A, Alegre J, von Schacky C. Low omega-3 index and polyunsaturated fatty acid status in patients with chronic fatigue syndrome/myalgic encephalomyelitis. Prostaglandins Leukot Essent Fat Acids (2018) 139:20–4. 10.1016/j.plefa.2018.11.006
    1. Massanella M, Negredo E, Puig J, Puertas MC, Buzón MJ, Pérez-Álvarez N, et al. Raltegravir intensification shows differing effects on CD8 and CD4 T cells in HIV-infected HAART-suppressed individuals with poor CD4 T-cell recovery. AIDS (2012) 26:2285–93. 10.1097/QAD.0b013e328359f20f
    1. Sotzny F, Blanco J, Capelli E, Castro-Marrero J, Steiner S, Murovska M, et al. Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: evidence for an autoimmune disease. Autoimmun Rev (2018) 17:601–9. 10.1016/j.autrev.2018.01.009
    1. Cliff JM, King EC, Lee J-S, Sepúlveda N, Wolf A-S, Kingdon C, et al. Cellular immune function in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS). Front Immunol (2019) 10:796. 10.3389/fimmu.2019.00796
    1. Rivas JL, Palencia T, Fernández G, García M. Association of T and NK cell phenotype with the diagnosis of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS). Front Immunol (2018) 9:1028. 10.3389/fimmu.2018.01028
    1. Cuadrado E, van den Biggelaar M, de Kivit S, Chen Y-Y, Slot M, Doubal I, et al. Proteomic analyses of human regulatory T cells reveal adaptations in signaling pathways that protect cellular identity. Immunity (2018) 48:1046–59. 10.1016/j.immuni.2018.04.008
    1. Sugiyama D, Nishikawa H, Maeda Y, Nishioka M, Tanemura A, Katayama I, et al. Anti-CCR4 mAb selectively depletes effector-type FoxP3+CD4+ regulatory T cells, evoking antitumor immune responses in humans. Proc Natl Acad Sci (2013) 110:17945–50. 10.1073/pnas.1316796110
    1. Blanco J, Canela EI, Sayós J, Mallol J, Lluis C, Franco R. Adenine nucleotides and adenosine metabolism in pig kidney proximal tubule membranes. J Cell Physiol (1993) 157:77–83. 10.1002/jcp.1041570110
    1. Ohta A, Sitkovsky M. Extracellular adenosine-mediated modulation of regulatory T cells. Front Immunol (2014) 5:304. 10.3389/fimmu.2014.00304
    1. Brenu EW, Huth TK, Hardcastle SL, Fuller K, Kaur M, Johnston S, et al. Role of adaptive and innate immune cells in Chronic Fatigue Syndrome/Myalgic Encephalomyelitis. Int Immunol (2014) 26:233–42. 10.1093/intimm/dxt068
    1. Umansky V, Shevchenko I, Bazhin AV, Utikal J. Extracellular adenosine metabolism in immune cells in melanoma. Cancer Immunol Immunother (2014) 63:1073–80. 10.1007/s00262-014-1553-8
    1. Hornig M, Montoya JG, Klimas NG, Levine S, Felsenstein D, Bateman L, et al. Distinct plasma immune signatures in Mylagic Encephalomyelitis/Chronic Fatigue Syndrome are present early in the course of illness. Sci Adv (2015) 1:1400121. 10.1126/sciadv.1400121
    1. Broderick G, Fuite J, Kreitz A, Vernon SD, Klimas N, Fletcher MA. A formal analysis of cytokine networks in chronic fatigue syndrome. Brain Behav Immun (2010) 24:1209–17. 10.1016/j.bbi.2010.04.012
    1. Montoya JG, Holmes TH, Anderson JN, Maecker HT, Rosenberg-Hasson Y, Valencia IJ, et al. Cytokine signature associated with disease severity in chronic fatigue syndrome patients. Proc Natl Acad Sci U S A (2017) 114:e7150–8. 10.1073/pnas.1710519114
    1. Moneghetti KJ, Skhiri M, Contrepois K, Kobayashi Y, Maecker H, Davis M, et al. Value of circulating cytokine profiling during submaximal exercise testing in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. Sci Rep (2018) 8:2779. 10.1038/s41598-018-20941-w
    1. Zhang H-YY, Liu Z-DD, Hu C-JJ, Wang D-XX, Zhang Y-BB, Li Y-ZZ. Up-regulation of TGF-β1 mRNA expression in peripheral blood mononuclear cells of patients with chronic fatigue syndrome. J Formos Med Assoc (2011) 110:701–4. 10.1016/j.jfma.2011.09.006
    1. Björkström NK, Svensson A, Malmberg K-JJ, Eriksson K, Ljunggren H-GG. Characterization of natural killer cell phenotype and function during recurrent human HSV-2 infection. PLoS One (2011) 6:e27664. 10.1371/journal.pone.0027664
    1. Lauer FT, Denson JL, Burchiel SW. Isolation, cryopreservation, and immunophenotyping of human peripheral blood mononuclear cells. Curr Protoc Toxicol (2017) 74:18201–16. 10.1002/cptx.31
    1. Lemieux J, Jobin C, Simard C, Néron S. A global look into human T cell subsets before and after cryopreservation using multi-parametric flow cytometry and two-dimensional visualization analysis. J Immunol Methods (2016) 434:73–82. 10.1016/j.jim.2016.04.010
    1. Sattui S, de la Flor C, Sánchez C, Lewis D, López G, Rizo-Patrón E, et al. Cryopreservation modulates the detection of regulatory T cell markers. Cytom B Clin Cytom (2012) 82:54–8. 10.1002/cyto.b.20621
    1. Diks AM, Bonroy C, Teodosio C, Groenland RJ, de Mooij B, de Maertelaere E, et al. Impact of blood storage and sample handling on quality of high dimensional flow cytometric data in multicenter clinical research. J Immunol Methods (2019) 475:112616. 10.1016/j.jim.2019.06.007
    1. Metzger K, Frémont M, Roelant C, De Meirleir K. Lower frequency of IL-17F sequence variant (His161Arg) in chronic fatigue syndrome patients. Biochem Biophys Res Commun (2008) 376:231–3. 10.1016/j.bbrc.2008.08.135
    1. Moreno-Fernandez ME, Rueda CM, Rusie LK, Chougnet CA. Regulatory T cells control HIV replication in activated T cells through a cAMP-dependent mechanism. Blood (2011) 117:5372–80. 10.1182/blood-2010-12-323162
    1. Patel DD, Kuchroo VK. Th17 cell pathway in human immunity: Lessons from genetics and therapeutic interventions. Immunity (2015) 43:1040–51. 10.1016/j.immuni.2015.12.003
    1. Chege D, Sheth PM, Kain T, Kim CJ, Kovacs C, Loutfy M, et al. Sigmoid Th17 populations, the HIV latent reservoir, and microbial translocation in men on long-term antiretroviral therapy. AIDS (2011) 25:741–9. 10.1097/QAD.0b013e328344cefb
    1. Li Y, Wei C, Xu H, Jia J, Wei Z, Guo R, et al. The immunoregulation of Th17 in host against intracellular bacterial infection. Mediators Inflamm (2018) 2018:6587296. 10.1155/2018/6587296
    1. Giloteaux L, Goodrich JK, Walters WA, Levine SM, Ley RE, Hanson MR. Reduced diversity and altered composition of the gut microbiome in individuals with myalgic encephalomyelitis/chronic fatigue syndrome. Microbiome (2016) 4:30. 10.1186/s40168-016-0171-4
    1. Karhan E, Gunter CL, Ravanmehr V, Horne M, Kozhaya L, Renzullo S, et al. Perturbation of effect and regulatory T cell subsets in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. BioRxiv. 10.1101/2019.12.23.887505
    1. Esfandyarpour R, Kashi A, Nemat-Gorgani M, Wilhelmy J. Davis RW. A nanoelectronics-blood-based diagnostic biomarker for myalgic encephalomyelitis/chronic fatigue syndrome. Proc Natl Acad Sci U S A (2019) 116:10250–7. 10.1073/pnas.1901274116
    1. 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) 21:1142. 10.3390/ijms21031142
    1. Parsons MS, Tang C-CC, Jegaskanda S, Center RJ, Brooks AG, Stratov I, et al. Anti-HIV antibody-dependent activation of NK cells impairs NKp46 expression. J Immunol (2014) 192:308–15. 10.4049/jimmunol.1301247
    1. Veluchamy JP, Delso-Vallejo M, Kok N, Bohme F, Seggewiss-Bernhardt R, van der Vliet HJ, et al. Standardized and flexible eight colour flow cytometry panels harmonized between different laboratories to study human NK cell phenotype and function. Sci Rep (2017) 7:43873. 10.1038/srep43873
    1. Naviaux RK, Naviaux JC, Li K, Bright AT, Alaynick WA, Wang L, et al. Metabolic features of chronic fatigue syndrome. Proc Natl Acad Sci U S A (2016) 113:e5472–80. 10.1073/pnas.1607571113

Source: PubMed

3
Abonneren