Skip to main content
Erschienen in: Journal of Translational Medicine 1/2013

Open Access 01.12.2013 | Research

Screening NK-, B- and T-cell phenotype and function in patients suffering from Chronic Fatigue Syndrome

verfasst von: Marta Curriu, Jorge Carrillo, Marta Massanella, Josepa Rigau, José Alegre, Jordi Puig, Ana M Garcia-Quintana, Jesus Castro-Marrero, Eugènia Negredo, Bonaventura Clotet, Cecilia Cabrera, Julià Blanco

Erschienen in: Journal of Translational Medicine | Ausgabe 1/2013

Abstract

Background

Chronic Fatigue Syndrome (CFS) is a debilitating neuro-immune disorder of unknown etiology diagnosed by an array of clinical manifestations. Although several immunological abnormalities have been described in CFS, their heterogeneity has limited diagnostic applicability.

Methods

Immunological features of CFS were screened in 22 CFS diagnosed individuals fulfilling Fukuda criteria and 30 control healthy individuals. Peripheral blood T, B and NK cell function and phenotype were analyzed by flow cytometry in both groups.

Results

CFS diagnosed individuals showed similar absolute numbers of T, B and NK cells, with minor differences in the percentage of CD4+ and CD8+ T cells. B cells showed similar subset frequencies and proliferative responses between groups. Conversely, significant differences were observed in T cell subsets. CFS individuals showed increased levels of T regulatory cells (CD25+/FOXP3+) CD4 T cells, and lower proliferative responses in vitro and in vivo. Moreover, CD8 T cells from the CFS group showed significantly lower activation and frequency of effector memory cells. No clear signs of T-cell immunosenescence were observed. NK cells from CFS individuals displayed higher expression of NKp46 and CD69 but lower expression of CD25 in all NK subsets defined. Overall, T cell and NK cell features clearly clustered CFS individuals.

Conclusions

Our findings suggest that alterations in T-cell phenotype and proliferative response along with the specific signature of NK cell phenotype may be useful to identify CFS individuals. The striking down modulation of T cell mediated immunity may help to understand intercurrent viral infections in CFS.
Begleitmaterial
Additional file 1: Table S1.: Main comorbidities identified in CFS affected individuals. Figure S1. Analysis of B-cell subsets and proliferation. Panel A. Gating strategy for the identification of B-cell subsets. IgA, IgG and IgD expressing cells were identified in the CD19+ gate, the expression of CD27 in each subset was analyzed. Plasma cells were identified by high CD38 and CD27 expression. Marginal Zone (MZ) B cells by high CD1c expression and transitional cells by CD5, CD10 and CD38 expression. Panel B. Fresh blood was stained with the antibody combinations described in Table I, lysed washed and acquired. B-cell subsets showed similar values in healthy donors (n = 27, HD, empty boxes) and CFS individuals (n = 19, CFS, solid boxes). Panel C. PBMC were stained with CFSE and cultured in the presence of the TLR9 agonist CpG2006 and the TLR7/8 agonist R848. Values of division index calculated using Flow Jo software from healthy donors (n = 5, HD, empty boxes) and CFS individuals (n = 9, CFS, solid boxes) are shown. In all cases, median values, interquartile ranges (boxes), 10-90 percentiles (bars) and p-values for nonparametric Mann-Whitney comparison are shown. Figure S2. Clustering CFS individuals according to NK cell phenotypic markers. A subset of 19 CFS (red labels) and 25 control individuals (green labels) was analyzed. Figure shows normalized centered data in yellow (for positive values, above median) and blue (for negative values, below median). NK cell parameters provided lower resolution than the combination of NK and T cell dta. However, CFS and healthy donors showed significant clustering (p = 3.1 × 10-7). Figure S3. Analysis of the effect of antioxidant intake on main biomarkers of CFS. 25 control individuals (HD) and 19 CFS individuals subgrouped according to antioxidant treatment were analyzed. Figure shows median and interquartile ranges for the 8 parameters defined in Figure 5. All figures show p-values for 1-way ANOVA analyses of the three groups (upper left corners) and p-values for Mann-Whitney comparisons between the CFS subgroups (right). (DOCX 3 MB)
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1479-5876-11-68) contains supplementary material, which is available to authorized users.
Marta Curriu, Jorge Carrillo, Marta Massanella contributed equally to this work.

Competing interest

The authors declare that they have no competing interests.

Authors’ contribution

MC, JC and MM performed experiments, analyzed data and wrote the manuscript. JR, JA AMG-Q and EN selected patients and edited the manuscript, JCM supervised clinical criteria and edited the manuscript, JP supervised sample extraction and processing, BC, CC and JB designed the study and wrote the manuscript. All authors read and approved the final manuscript.

Background

Chronic Fatigue Syndrome (CFS) is a complex clinical condition of unknown etiology, characterized by persistent or intermittent fatigue that is not the result of recent exertion and does not improve with rest, resulting in a significant reduction in the patient's previous normal activity[1]. Classical diagnostic criteria for CFS overlap with Myalgic Encephalomyelitis (ME) and require these symptoms to be present for at least six months and concomitant to at least four accompanying symptoms, among them: impaired memory, adenopathy, myalgia or polyartralgia[2]. The World Health Organization acknowledged ME/CFS as a disease of the nervous system (ICD G93.3)[3]. However, CFS is a multi-system disease, in which neurological disorders are accompanied by altered immune, musculoskeletal, endocrine and cardiovascular systems[4, 5]. Research efforts have recently provided a new International Consensus Criteria for ME[3]. However, the search for the etiology(ies) and the pathogenic mechanisms of CFS has unsuccessfully reviewed several viral hypotheses including herpesviruses or retroviruses as potential triggers of the disease[68]. The potential role of pathogens in the CFS field has boosted research in the immunological sides of the illness[911]. Indeed, the link between the immune system and CFS has been explored since the early 90’s[12], and is supported by the coincidence of the onset of symptoms with viral infections[13], the persistence of several infections in CFS individuals[10, 1417], the beneficial effect of treatment of human herpesvirus 6 and Epstein-Bar virus infection in CFS symptoms[18] and the reported role of autoimmunity[19, 20] Moreover, other features of ME/CFS, such as mild inflammation, oxidative/nitrosative stress, mitochondrial dysfunction and the presence of autoimmunne responses[4] may alter immune function and phenotype.
However, the characterization of the immune status of CFS individuals has frequently yield contradictory results. Early work associated CFS with a general status of immune activation assessed by CD38 or HLA-DR expression in CD8 T cells[12]. However, other authors found similar expression of these markers in CFS and healthy individuals[21], while lower expression of the activation markers CD69 or soluble CD26 has also been described as a feature of CFS[22, 23]. Similarly, B-cell function[24], B-cell mediated autoimmunity or unbalanced cytokine network have been linked to CFS[9, 2527] again with inconclusive results. Altered numbers of NK subsets, defined by CD56 or CD16 expression, and an impaired NK-cell lytic activity have been more consistently associated with CFS[24, 2832], although controversial data have been also reported[33] and a consensus in relevant NK cell markers is still lacking.
At least some of these immune features described in CFS may be related to active, poorly controlled viral infections, which differently modulate immune responses and may produce immune hyperactivation/exhaustion, as widely reported for HIV[34]; may contribute to immunosenescence, as postulated for CMV[35]; or may cause a status of immune anergy, as described for measles virus[36]. In this context, we sought to compare the phenotype and function of different immune cells between healthy and ME/CFS individuals. The study was designed in 2010 and could not include new definitions that currently allow for the separation of ME and CFS affected individuals[3]. However, our data point to an unaffected B-cell compartment, a biased NK-cell phenotype and a poorly responsive T-cell compartment as the main immune features of ME/CFS affected individuals.

Methods

Patients

A study to screen immunological features of patients suffering from CFS was designed and approved by the Ethics Committee of the Hospital Universitari Germans Trias i Pujol (Barcelona, Spain; EO-10-007). All procedures followed the Helsinki Declaration in 1975. The main objective was to compare phenotypic and functional alterations in immune cells between CFS patients and healthy donors. Therefore, a wide range of phenotypic features was analyzed in a limited number of CFS patients (N = 22) and controls (N = 30). CFS patients fulfilled the Fukuda criteria[1]. Individuals were selected from cohorts of CFS Clinical Units (CFS Unit, Tarragona, Spain and Vall d’Hebron University Hospital, Barcelona, Spain). General exclusion criteria from these cohorts were diabetes, hypertension, chronic obstructive pulmonary disease, inflammatory bowel or Crohn's disease, rheumatoid arthritis, Parkinson or Huntington disorders, schizophrenia, organic mental disorders, substance use disorders, multiple sclerosis and body mnass index BMI > 30 kg/m2. Eligible subjects were those with age over 18 years, confirmed diagnosis of CFS for more than two years and absence of current identified infections. Additional exclusion criteria were pregnancy and chemotherapy treatment. In order to mirror the potential heterogeneity of CFS-affected, other major comorbidities associated to CFS were evaluated but were not considered exclusion criteria. In particular, anxiety that is an common feature of CFS affected individuals was evaluated using HADS (Hospital Anxiety and Depression Scale)[37]. The severity of CFS was assessed using a national scale[38]. Informed consent was obtained from all participants in the study. Clinical and demographic data were collected from medical records.

Sample processing

A single blood sample was collected by venipuncture in EDTA vacutainer tubes (BD Biosciences) from all participants. An aliquot was used for immediate immunophenotype; remaining blood was processed for plasma and peripheral blood mononuclear cells (PBMC) preparation by standard methods as described[39]. PBMC were washed twice in PBS and resuspended in RPMI culture medium for immediate analysis of proliferation, NK cell activity or cell death assays. All samples were collected and freshly processed at the Hospital Germans Trias i Pujol. All samples were processed for immunophenotype and function on the same day of blood collection, no more than four hours were left between sample collection and immunophenotype staining, while no more than six hours were left for functional and cell death assays.

Determination of absolute counts of B, T and NK cells

Absolute counts of B, T and NK cells were analyzed by flow cytometry. First, the absolute lymphocyte count was determined using an anti-CD45–V450 antibody (BD Biosciences) in combination with perfect-count microspheres (Cytognos). Then, the percentage of the different lymphocyte subsets was determined using the following antibody combination: CD45–V450, CD19–AmCyan, CD3–APC-Cy7, CD4–APC, CD8–PerCP, CD56–PE and CD16–FITC (BD Biosciences). Absolute count of each cellular population was calculated as follows: (X*Y)/100, where X is the percentage of each subset and Y is the absolute count of lymphocytes.

Immunophenotype

Freshly obtained blood was incubated for 15 minutes at room temperature with the antibody combinations shown in Table 1 in order to characterize B, T and NK cell populations. All antibodies were from BD Biosciences unless indicated. Cells were then lysed for 15 minutes at room temperature in FACS Lysing solution (BD Biosciences), washed in PBS and fixed in PBS containing 1% formaldehyde (Sigma), before acquisition in an LSRII flow cytometer (BD Biosciences). For Ki67 and FOXp3 staining, two aliquots of fresh blood were incubated with extracellular antibodies (Table 1) as indicated above. After lysis, cells were washed and fixed/permeabilized using FOXp3 staining buffer set (eBiosciences) and incubated with anti Ki67 and FOXP3 antibodies or IgG isotype control antibodies (Table 1). Treg were defined by the presence of a separate population of CD25bright FOXP3+ cells.
Table 1
Antibody combinations used for the phenotypic characterization of T, NK, B cells and for the determination of absolute counts
Antibody combinations/
Fluorochrome
CELL TYPE
Phenotype
Tube #
V450/Pacific blue
V500/Am Cyan
FITC
PE
APC/Alexa Fluor 647
PerCP/PerCP-Cy5
PE-Cy7
APC-cy7
T cells
Thymic output/Exhaustion
T1
  
CD95
PD-1
CD4
HLA-DR
CD8
CD3
T2
  
CD45RA
CD31
CD4
CD38d
CD8
CD3
T3
    
CD4
 
CD8
CD3
CD8 activation
T4
CD4
 
HLA-DR
 
CD45RO
CD38d
CD8
CD3
T5
CD4
     
CD8
CD3
Main subsets/senescence
T6
CD45RA
CD8
CD57e
CD28
CD27
CD4
CCR7
CD3
T7
 
CD8
   
CD4
 
CD3
Treg/Prolif./Anergy
T8
CD4
CD8
KI67
FOXP3e
CD127c
CD5d
CD25
CD3
T9
CD4
CD8
IRR IgG
IRR IGG2ae
   
CD3
NK cells
Activation/receptor profile
NK1
CD69
CD3b/CD19b
CD107a
Nkp44
CD57e
CD25
CD56
CD16
NK2
NKp46a,ed
CD3b/CD19b
CD107a
NKG2De
Nkp30 c
NKG2Ae
CD56
CD16
NK3
 
CD3b/CD19b
    
CD56
CD16
B cells
Mature/Memory/Transitional
B1
  
CD1c
IgDe
IgM
CD38
CD27e
CD19
B2
  
CD23
IgDe
CD5
CD38
CD10e
CD19
B3
   
IgDe
   
CD19
Switched memory
B4
  
CD1ce
IgGe
IgA
CD38
CD27e
CD19
B5
   
IgGe
IgAe
  
CD19
a Pacific blue-coupled antibody.
b AmCyan-coupled antibody.
c Alexa Fluor 647-coupled antibody.
d PerCP-Cy5.5 coupled antibody.
e sources different from BD Biosciences (NKp46 and CD57 from BioLegend; NKG2A from R&D Systems, NKG2D and CD1c from Santa Cruz; FOXP3, IRR IGG2a, CD27 and CD10 from eBiosciences; IgG and IgA from Jackson ImmunoResearch, CD57 from Beckman Coulter).

B-cell and T-cell proliferation assays

Freshly obtained PBMC were stained with 0.33 μM CFSE (Invitrogen) for 5 minutes at room temperature. After extensive washes, cells were cultured in RPMI1640 medium supplemented with 10% of FBS (R10 medium) and different stimulus. The proliferation of T cells was assayed using 5 μg/mL of PHA (Sigma-Aldrich) plus 10 U/mL of IL-2 (Roche). For B cells, 3 μg/mL of endotoxin-free CpG2006 (InvivoGen) or 1 μg/mL of R848 (Alexis Biochemicals) were used alone or in combination with 5 μg/mL of F(ab)2 Goat anti-human Igs (Jackson Immunoresearch). Four days later, cellular proliferation was assessed by flow cytometry after staining T cells with anti CD2–PerCP-Cy5.5, CD5–APC, CD4–V450, CD8–APC-Cy7 and CD19–PE-Cy7 or B cells with CD3–APC-Cy7, CD19–PE-Cy7, CD14–PerCP (BD Biosciences) and IgD-APC (Miltenyi Biotec). Data analysis was performed with the Flowjo software (Tree Star, Inc.), calculating division and proliferation indexes for each condition using best fits provided by the software.

Cell death assays

Cell death was evaluated by culturing PBMC in of R10 medium at a density of 1 × 106 PBMC/ml for 24 hours[34, 40]. For T-cell death analysis, PBMC were incubated with 40 nM of the potentiometric mitochondrial probe DIOC6 (Invitrogen), 5 μg/mL propidium iodide (Sigma), and CD3–APC-Cy7, CD4–APC and CD8–PE-Cy7 antibodies. For B cell death analysis, PBMC were incubated with 40 nM of DIOC6, 0.3 μM of Sytox Blue (Invitrogen) and CD19–APC-Cy7, IgD–PE, CD38–PerCP-Cy5.5, CD5–APC (BD Biosciences) and CD27–PE-Cy7 (eBiosciences). Cells were acquired in an LSRII flow cytometer; dead cells were identified by their low DIOC6 staining[41].

NK activity assays

Lytic activity of NK cells was assessed as described[42] with slight modifications. Available samples of PBMC (n = 16) were extensively washed to remove any trace of EDTA from extraction tubes and monocytes/macrophages were removed by plastic adherence for 1 h at 37°C (5% CO2), this treatment removed 82.4% of monocytes from PBMC samples (n = 13). Exponentially growing eGFP-K562 cells å(obtained through the AIDS Research and Reference Reagent Program, from Dr. Kantakamalakul[43]) were used as target cells. Incubations were performed in duplicate in 96 well plates by seeding a fixed amount (10,000 target cells) alone or with increasing amounts of monocyte-depleted PBMC in R10 medium, covering a range from 80:1 to 2.5:1 effector:target cell ratios. Samples were incubated for 4 h at 37°C (5% CO2) stained for 10 min with 5 μg/mL Propidium Iodide (PI, Sigma) and acquired in a LSRII flow cytometer. The percentage of dead (PI+) eGFP-K652 cells in the gate of eGFP-K562 cells was calculated.

Clustering and statistical analyses

Continuous variables were expressed as the median (interquartile range) and compared using the Mann–Whitney non-parametric test. Discrete variables were described as percentages (number of patients) and the chi-square or Fisher exact test was used as appropriate. P values <0.05 were considered significant. Clustering of CFS and healthy individuals was performed using the Cluster 3.0 software. Data were normalized according to medians and clustered using non-parametric correlations. Treeview 1.1 software was used to generate and visualize dendrograms.

Results

Patient characteristics

The main characteristics of the individuals recruited for the study are summarized in Table 2. Both Control and CFS groups showed similar median age values (38 and 44 years, respectively, p = ns, Mann–Whitney test) and were mostly composed by females (55% and 73% respectively, p = ns, Fisher exact test). Patients suffering from CFS showed a median [IQR] time from certified diagnosis of 3[35] years, with 53% of them reporting onset of symptoms with viral infections. The median grade of CFS assessed by a national scale[38] was 3, while only three recruited individuals showed grade 2. Grade 3 was comparable to mild level of severity (an approximate 50% reduction in pre-illness activity level) according to the newest consensus criteria for ME[3]. Grade 2 is assignable to a moderate severity. No severe cases were included in the study. Three individuals in the CFS group were excluded from the analysis, one due to a B cell lymphocytosis (B cells represented 24% of lymphocytes showing a total B cell count of 435 cells/μL, with more than 85% of cells showing a IgD+IgMCD23+CD27+CD5+CD38 phenotype), one due to an IgA deficiency and a third due to sample unavailability.
Table 2
Main characteristics of individuals recruited in the study
 
SFC
HD
P-value
(N = 22)
(N = 30)
(Mann–Whitney)
Age (years, Median, IQR)
44 [40–50]
38 [33–52]
ns
Gender (% of female)
73
55
ns
Time from diagnosis (Years, Median, IQR)
3 [3–5]
-
 
Grade of Fatigue (Median, IQR)
3 [2–3]
-
 
Reported onset with viral infection (%)
53
-
 
Additional factors that may modulate immune function such as comorbidities and polypharmacy associated with CFS were also evaluated. Polypharmacy is described in Table 3. The main potential interference of pharmacy was homeopathy (52% of patients), antioxidants (52%) and analgesics (42%, mainly paracetamol), which were separately distributed among participants.. Regarding comorbidities, anxiety was the most prevalent; myalgia or tendinopathy along with reported multiple chemical sensitivity were also relevant (Additional file1: Table S1).
Table 3
Self-reported polypharmacy in CFS affected individuals
 
Analgesics
Antidepressants Anxiolytics Benzodiazepines
Homeopa Thy
Antioxidants
Patient #
Paracetamol
Ibuprofen
Tramadol
Duloxetine
Pregabalin
Diazepam
CFS1
-
-
-
-
-
-
  
CFS2
-
-
-
-
-
-
+
+
CFS3
-
-
-
+
-
-
+
+
CFS4
-
-
-
-
-
+
+
+
CFS5
-
-
-
-
-
-
+
+
CFS6
-
-
-
-
-
-
+
+
CFS7
-
-
-
-
-
-
+
+
CFS8
-
-
-
-
-
-
+
+
CFS9
-
-
-
-
-
-
+
+
CFS10
-
-
-
-
-
+
+
+
CFS11
-
-
-
-
-
-
-
+
CFS12
+
-
+
-
+
-
-
-
CFS13
-
-
-
-
-
-
-
-
CFS14
+
-
-
-
-
-
-
-
CFS15
+
+
-
+
+
-
-
-
CFS16
+
+
-
+
+
-
-
-
CFS17
+
-
-
-
-
+
-
-
CFS18
+
+
-
+
+
+
-
-
CFS19
-
-
-
-
-
-
-
-
Daily antioxidant doses were: 500 mg omega-3 fatty acids (Docosahexaenoic acid DHA/Eicosapentaenoic acid EPA), 150 mg Methylsulfonylmethane, 150 mg alpha lipoic àcid, 75 mg Superoxide dismutase (SOD), 50 mg N-Acetyl cisteine (NAC), 3,5 mg Zn2+ and 0.5 mg Cu2 + .

Quantification of main lymphocyte subsets

The absolute numbers and percentages of B cells (CD19+), NK cells (CD3-CD56+CD16+), T cells (CD3+), CD4 T cells (CD3+CD4+) and CD8 T cells (CD3+CD8+) in the CD45+ lymphocyte gate were similar between CFS individuals and healthy donors (data not shown and Figure 1). However, significantly lower frequency of CD3+CD56+ lymphocytes was observed in the CFS group. Moreover, a slightly unbalanced composition of the CD3+ cell subset was also observed, showing a higher CD4 T-cell representation in the CFS group (Figure 1).

B-cell phenotype and function

The potential role of B cells on CFS has been reinforced by recent data on the clinical benefit of Rituximab treatment[19, 20]. Thus, we characterized circulating B cells using the antibody panels shown in Table 1. No significant differences between groups were observed in the percentage of IgD+, IgG+, IgA+ or CD27+ B cells, indicating a similar memory compartment in CFS and control individuals. Moreover, the levels of transitional (CD19+IgD+CD38highCD10+CD5+), plasma-plasmablastic (CD19+CD27highCD38high) or marginal zone B cells (CD19+IgD+IgM+CD27+CD1c+), were comparable in both groups (Additional file1: Figure S1). Consistently, functional assessment of B-cell responses showed similar proliferation to different stimuli targeting TLR9 and TLR7/8, and similar level of ex vivo cell death (Additional file1: Figure S1 and data not shown). Thus, no major perturbations on the phenotype and function of circulating B cells could be identified.

NK-cell phenotype and function

NK-cell alterations have been classically associated with CFS, showing decreased numbers and function[9, 44]. Therefore, we evaluated the phenotype of NK cells using the antibody panel shown in Table 1. The three main NK-cell subsets identified in our gating strategy CD56highCD16, CD56+CD16+ and CD16+CD56 cells (Figure 2A) and most of the markers analyzed were comparable between groups (data not shown). However, the expression of CD69 and NKp46 was significantly higher in CFS individuals, while the expression of CD25, was significantly lower (Figure 2B).
A phenotypic feature of NK cells from CFS individuals is the low expression of CD57[45]. Figure 2C shows that in our study, the percentage of CD57 expressing NK cells is similar among groups, although the intensity of CD57 staining showed significantly lower values in CFS individuals. Despite alterations in phenotype, NK cell cytotoxic activity did not reach significant differences between groups in a subset of samples (9 CFS and 7 HD) available for functional assays. Furthermore, no differences in sensitivity of NK cells to ex vivo cell death could be detected between groups (data not shown).

T-cell phenotype and function

Several authors have pointed to a general status of T-cell activation in CFS[12] that may be consistent with intercurrent viral infections. A similar scenario has been described for HIV infection, in which chronic viral infection alters the balance of naïve, central and effector memory cells[46], and increases hyperactivation, immunosenescence and apoptosis[34, 47, 48]. In our cohort, CD4 T cells from CFS individuals and controls showed similar levels of naïve (CD45RA+CCR7+CD27+CD28+), central (CD45RACCR7+CD27+CD28+), transitional (CD45RACCR7-CD27+CD28+), effector (CD45RACCR7-CD27+CD28-) and terminally differentiated memory (CD45RA-CCR7-CD27-CD28-) cells (Figure 3A and B). Most CD8 T cell subsets were also similar in both groups, although CFS affected individuals showed lower frequency of cells with an effector phenotype (defined in CD8 T cells by the following markers CD45RA-CCR7-CD27-CD28+, Figure 3B). This observation maybe related with the lower level of CD56 expression in CD3+ cells (Figure 1), although the heterogeneity of this latter population impedes a proper interpretation of these data. We also analyzed several markers of immunosenescence or immune exhaustion, with discordant results: CFS and control individuals had similar expression of the T-cell immunosenescence marker CD57[46] in CD4 and CD8 T cells, while differences were observed in the expression of exhaustion markers PD-1 and CD95[46] in CD4 and CD8 T cells, respectively (Figure 3C).
We assessed the frequency of T regulatory cells (Treg) and several proliferation/activation markers (Figure 4A). Treg cells defined as CD4+CD25++FOXP3+ or CD4+CD25++FOXP3+CD127 showed significantly higher percentages in CFS individuals (Figure 4B and data not shown), concomitant with lower levels of Ki67+ cells in CD4 T cells (Figure 4B). In contrast, CD8 T cells did not show differences among groups in Ki67 positivity, although CFS individuals displayed higher expression of CD5 (Figure 4B), a marker associated with impaired T-cell responses[49, 50]. CFS group also showed lower levels of the activation marker CD38 in total and the memory (CD45RO+) CD8 T cells (not shown and Figure 4B). Despite these small differences in T-cell subsets, ex vivo proliferative responses of CD4 T cells were significantly lower in CFS individuals although showed no differences among groups for CD8 T cells (Figure 4C). Finally, no differences in cell death were noticed for CD4 and CD8 T cells among groups (Figure 4D).
As a whole, these data suggest an impaired T-cell response in CFS individuals associated with increased Treg numbers and with some specific markers in both CD4 and CD8 T cells, but not directly caused by a general status of immunosenescence.

Analysis of T-cell and NK-cell phenotype as markers for CFS

In order to evaluate the potential utility of the described immunological markers for the identification of CFS cases, an unsupervised clustering analysis was performed. While unrestricted selection of markers provided poor ability to separate CFS individuals and HD, marker selection according to their selectivity yielded a combination of 8 NK and T-cell phenotypic parameters that showed the best resolution in classifying CFS and healthy individuals (p = 3.3×10-8, Figure 5). The NK markers: CD25, CD69, NKp46 (expressed as percentage of positive cells) and CD57 (as fluorescence intensity) in combination with the T-cell subsets: regulatory, proliferating Ki67+CD4+, effector CD8+ and CD56+ T cells, showed a high sensitivity (100%) but a moderate false positive rate (5/24, specificity 79%, Figure 5). A more restrictive choice of parameters, including exclusively NK cell markers showed similar false positive rates (5/23, specificity 78%) but lower sensitivity (95%) in the detection of CFS cases (Additional file1: Figure S2).

Discussion

The immunological status of individuals suffering from CFS has yielded heterogeneous results[12, 17, 31, 51, 52]. Clear examples are data from different CFS cohorts analyzed in a single laboratory[32]. These observations may be the consequence of an intrinsic heterogeneity in the classification of CFS affected individuals[3], or could be related to the presence of intercurrent infections in the individuals recruited in the different studies. In particular, the prevalence of infections by herpesviruses or enteroviruses[15], which have been described more frequently in CFS individuals[18, 53], are known to modulate immune phenotype[46]. We have performed a wide screening of the phenotype and function of B, NK and T cells in CFS. In contrast to other studies, our main inclusion criterion was focused on the lack of active infections, rather than on the CFS-related comorbidities. Although this could be a limitation, as comorbidities may also affect immune status, we believed that these criteria might provide a more homogeneous immune profile of CFS.
Our data suggest that most differences between CFS and healthy controls were observed in NK and T cells; while the B-cell compartment showed similar composition in both groups. Importantly, these differences could not be associated to polypharmacy or comorbidities (Additional file1: Figure S3 and data not shown), although the sample size of our study limited the statistical power of these analyses. The lack of B cell alterations observed in our study contrasts with the active role of B cells and autoimmune responses in CFS that have been highlighted by the recent clinical use of Rituximab[19]. The possibility that B-cell alterations are restricted to tissue B cells may explain this apparent contradiction. However, it should be noted that in contrast to NK and T cell markers described herein, parameters of B cell phenotype showed heterogeneous values and seem to be more affected by antioxidant/analgesic treatments. This could be a second limitation of our study; thus, a more detailed analysis of B cell phenotype and function using larger cohorts will be required to fully understand the role of these cells in CFS.
A different scenario is observed in NK cells. In this case, several markers showed consistent alterations in CFS individuals and illustrate a skewed NK cell population with high CD69 and low CD25 expression, a paradoxical phenotype that has been described in acute influenza infection or vaccination[54] and that is in clear conflict with recent data reporting low CD69 expression in NK and T cells from CFS affected individuals[22, 23]. Of note, this latter work measured CD69 expression done after in vitro stimulation while our data were obtained using freshly obtained unstimulated cells. Thus, although apparently in conflict, both data may reflect different aspects of a deregulated CD69 expression in CFS. We have also observed increased levels of NKp46 expression in CFS individuals. Interestingly the expression of this receptor has been recently linked to T-cell responses in murine models[55]; however, no clear association could be found in our cohort. The phenotype of NK cells is controlled by genetic, epigenetic and environmental factors and probably summarizes the infectious history of an individual in a poorly-understood form of immunological memory[56]. Specific phenotypes have been associated to infectious agents, such as CMV or influenza[54, 57]. Although it could be tempting to speculate on a common infective history in our cohort of CFS individuals, the analysis of their serological status for some viruses (CMV, EBV, parvoviruses) did not show a clear link between past infections and current NK-cell phenotype. Probably, wider studies are required to answer this relevant question and to confirm the association of NK-cell phenotype with impaired lytic activity, which has been described in larger cohorts but failed to reach significant differences in our limited analysis, probably due to the small subset of samples analyzed or to the use of EDTA as anticoagulant. NK function decreases with age, a phenomenon associated with NK immunosenescence that can be evidenced by the increased expression of CD57, present in terminally differentiated NK cells[58]. However, there is no consensus on the role of CD57 in immunosenescence of NK cells and CFS has been associated with low expression of CD57[59]; an observation partly confirmed in our cohort.
The most unanticipated data in our cohort of CFS individuals is related to T-cell phenotype and function, which could be defined as a general hyporesponsiveness. These data contrast with descriptions of high T-cell activation in CFS[12], but are consistent with other reports describing reduced CD8 cytotoxic activity[9]. Again, the presence of active viral infections at the sampling time may be a source of heterogeneity. Alternatively, the leakage of bacterial products from gut may also determine T-cell activation[60, 61]. To explore this possibility, we analyzed plasma levels of sCD14 In healthy and CFS individuals, showing similar median levels (4.5 and 4.7 μg/ml, respectively, p = 0.44, data not shown), suggesting that gut leakage is not a major contributor to immune alterations in our cohort.
Our data may suggest a general default of T-cell function that can be observed using different makers in both CD4 and CD8 subsets. While CD4 T cells show lower Ki67 staining and ex vivo proliferation, CD8 T cells showed no proliferative differences, but lower levels of CD56 expression, effector (CCR7CD45RA) cells, CD38+ cells and higher expression of CD5, a marker of anergy associated to continuous antigen exposure[50]. Furthermore, this general status of T-cell hyporesponsiveness seems to be unrelated to immunosenescence, since no differences in CD57 or no increased levels of CD27- or CD28- cells were observed between groups. A key factor in the control of T cell responses is the function of Treg cells[62], which are significantly increased in our cohort of CFS individuals confirming data from another recent study[9]. Although no clear correlation was observed between Treg frequency and other markers of NK cell or T-cell phenotype; an active role of Treg could be supported by reported data on key mediators of Treg action, such as TGF-β[63], that seem to be also upregulated in CFS individuals[64, 65].
The potential use of the immunological markers identified in this study was explored in cluster analyses. While the best resolution required both T and NK cell markers, we also identified a robust combination of NK cell markers that may be useful for diagnosis. This combination includes CD25, CD69 and NKp46 expression in CD56 + CD16+ cells and CD56 expression in CD3+ cells, suggesting that a five-color flow cytometry strategy including these markers may be useful for diagnostic purposes. However, a wider range of parameters including FOXP3 and Ki67 expression in CD4 T cells improved specificity. The use of these potential combinations as diagnostic tools requires validation in further studies, including both larger cohorts and a wider range of CFS clinical status.
In conclusion, CFS individuals analyzed in this study show no differences in B-cell compartment, skewed NK cells and poorly responsive T cells. The observed immunological defaults do not provide any causative link to the illness, but could explain some of the symptoms and in particular the poor control of viral infections reported in these individuals[15, 53]. However, some of these markers along with other previously described, such as NK function or DPPIV[22, 66] may be useful for the characterization of CFS. However, major roadblocks still exist to reach a reliable combination of immune biomarkers for CFS. First, the potential different etiologies or comorbidities of CFS and second the clear identification of the target population (ME, CFS or both). In addition, immunological markers may reveal only a part or the complex pathogenic spectrum of ME/CFS. Most likely, immune features in combination with a detailed analysis of intercurrent infections and previously described[9, 22, 25, 29, 66, 67]neurological and metabolic disorders may provide the clues to define a full set of markers helpful for our knowledge of CFS pathogenesis and for its clinical management.

Acknowledgements

We are grateful to Manel Juan and Alan Landay for critical reading of the manuscript. We are also grateful to Rafaela Ayén, Tania González and Eulalia Grau for sample processing. We wish to thank all volunteers that have participated or have shown their support to this study.
This work was supported by the IrsiCaixa, FLS and Catalan Fatigue Foundations. JB and CC are researchers from Fundació Institut de Recerca en Ciències de la Salut Germans Trias i Pujol supported by the ISCIII and the Health Department of the Catalan Government (Generalitat de Catalunya). JC is supported by a “Sara Borrell” grant from the Spanish Health Institute “ISCIII”. M.M is supported by a predoctoral grant from Generalitat de Catalunya and European Social Fund.
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interest

The authors declare that they have no competing interests.

Authors’ contribution

MC, JC and MM performed experiments, analyzed data and wrote the manuscript. JR, JA AMG-Q and EN selected patients and edited the manuscript, JCM supervised clinical criteria and edited the manuscript, JP supervised sample extraction and processing, BC, CC and JB designed the study and wrote the manuscript. All authors read and approved the final manuscript.
Anhänge

Electronic supplementary material

Additional file 1: Table S1.: Main comorbidities identified in CFS affected individuals. Figure S1. Analysis of B-cell subsets and proliferation. Panel A. Gating strategy for the identification of B-cell subsets. IgA, IgG and IgD expressing cells were identified in the CD19+ gate, the expression of CD27 in each subset was analyzed. Plasma cells were identified by high CD38 and CD27 expression. Marginal Zone (MZ) B cells by high CD1c expression and transitional cells by CD5, CD10 and CD38 expression. Panel B. Fresh blood was stained with the antibody combinations described in Table I, lysed washed and acquired. B-cell subsets showed similar values in healthy donors (n = 27, HD, empty boxes) and CFS individuals (n = 19, CFS, solid boxes). Panel C. PBMC were stained with CFSE and cultured in the presence of the TLR9 agonist CpG2006 and the TLR7/8 agonist R848. Values of division index calculated using Flow Jo software from healthy donors (n = 5, HD, empty boxes) and CFS individuals (n = 9, CFS, solid boxes) are shown. In all cases, median values, interquartile ranges (boxes), 10-90 percentiles (bars) and p-values for nonparametric Mann-Whitney comparison are shown. Figure S2. Clustering CFS individuals according to NK cell phenotypic markers. A subset of 19 CFS (red labels) and 25 control individuals (green labels) was analyzed. Figure shows normalized centered data in yellow (for positive values, above median) and blue (for negative values, below median). NK cell parameters provided lower resolution than the combination of NK and T cell dta. However, CFS and healthy donors showed significant clustering (p = 3.1 × 10-7). Figure S3. Analysis of the effect of antioxidant intake on main biomarkers of CFS. 25 control individuals (HD) and 19 CFS individuals subgrouped according to antioxidant treatment were analyzed. Figure shows median and interquartile ranges for the 8 parameters defined in Figure 5. All figures show p-values for 1-way ANOVA analyses of the three groups (upper left corners) and p-values for Mann-Whitney comparisons between the CFS subgroups (right). (DOCX 3 MB)
Literatur
1.
Zurück zum Zitat 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: 953-959.CrossRefPubMed 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: 953-959.CrossRefPubMed
2.
Zurück zum Zitat Avellaneda Fernández A, Pérez Martín A, Izquierdo Martínez M, Arruti Bustillo M, Barbado Hernández FJ, de la Cruz Labrado J: Chronic fatigue syndrome: aetiology, diagnosis and treatment. BMC Psychiatry. 2009, 9 (Suppl 1): S1-10.1186/1471-244X-9-S1-S1.PubMedCentralCrossRefPubMed Avellaneda Fernández A, Pérez Martín A, Izquierdo Martínez M, Arruti Bustillo M, Barbado Hernández FJ, de la Cruz Labrado J: Chronic fatigue syndrome: aetiology, diagnosis and treatment. BMC Psychiatry. 2009, 9 (Suppl 1): S1-10.1186/1471-244X-9-S1-S1.PubMedCentralCrossRefPubMed
3.
Zurück zum Zitat Carruthers BM, van de Sande MI, De Meirleir KL, Klimas NG, Broderick G, Mitchell T: Myalgic encephalomyelitis: International Consensus Criteria. J Intern Med. 2011, 270: 327-338. 10.1111/j.1365-2796.2011.02428.x.PubMedCentralCrossRefPubMed Carruthers BM, van de Sande MI, De Meirleir KL, Klimas NG, Broderick G, Mitchell T: Myalgic encephalomyelitis: International Consensus Criteria. J Intern Med. 2011, 270: 327-338. 10.1111/j.1365-2796.2011.02428.x.PubMedCentralCrossRefPubMed
4.
Zurück zum Zitat Morris G, Maes M: A neuro-immune model of Myalgic Encephalomyelitis/Chronic fatigue syndrome. Metab Brain Dis. 2012, 10.1007/s11011-012-9324-8. Morris G, Maes M: A neuro-immune model of Myalgic Encephalomyelitis/Chronic fatigue syndrome. Metab Brain Dis. 2012, 10.1007/s11011-012-9324-8.
5.
Zurück zum Zitat Jason LA, Corradi K, Torres-Harding S, Taylor RR, King C: Chronic fatigue syndrome: the need for subtypes. Neuropsychol Rev. 2005, 15: 29-58. 10.1007/s11065-005-3588-2.CrossRefPubMed Jason LA, Corradi K, Torres-Harding S, Taylor RR, King C: Chronic fatigue syndrome: the need for subtypes. Neuropsychol Rev. 2005, 15: 29-58. 10.1007/s11065-005-3588-2.CrossRefPubMed
6.
Zurück zum Zitat Papadopoulos AS, Cleare AJ: Hypothalamic-pituitary-adrenal axis dysfunction in chronic fatigue syndrome. Nat Rev Endocrinol. 2012, 8: 22-32.CrossRef Papadopoulos AS, Cleare AJ: Hypothalamic-pituitary-adrenal axis dysfunction in chronic fatigue syndrome. Nat Rev Endocrinol. 2012, 8: 22-32.CrossRef
7.
Zurück zum Zitat Lombardi VC, Ruscetti FW, Das Gupta J, Pfost MA, Hagen KS, Peterson DL: Detection of an infectious retrovirus, XMRV, in blood cells of patients with chronic fatigue syndrome. Science. 2009, 326: 585-589. 10.1126/science.1179052.CrossRefPubMed Lombardi VC, Ruscetti FW, Das Gupta J, Pfost MA, Hagen KS, Peterson DL: Detection of an infectious retrovirus, XMRV, in blood cells of patients with chronic fatigue syndrome. Science. 2009, 326: 585-589. 10.1126/science.1179052.CrossRefPubMed
8.
Zurück zum Zitat Silverman RH, Das Gupta J, Lombardi VC, Ruscetti FW, Pfost MA, Hagen KS: Partial retraction. Detection of an infectious retrovirus, XMRV, in blood cells of patients with chronic fatigue syndrome. Science. 2011, 334: 176-CrossRefPubMed Silverman RH, Das Gupta J, Lombardi VC, Ruscetti FW, Pfost MA, Hagen KS: Partial retraction. Detection of an infectious retrovirus, XMRV, in blood cells of patients with chronic fatigue syndrome. Science. 2011, 334: 176-CrossRefPubMed
9.
Zurück zum Zitat Brenu EW, van Driel ML, Staines DR, Ashton KJ, Ramos SB, Keane J: Immunological abnormalities as potential biomarkers in Chronic Fatigue Syndrome/Myalgic Encephalomyelitis. J Transl Med. 2011, 9: 81-10.1186/1479-5876-9-81.PubMedCentralCrossRefPubMed Brenu EW, van Driel ML, Staines DR, Ashton KJ, Ramos SB, Keane J: Immunological abnormalities as potential biomarkers in Chronic Fatigue Syndrome/Myalgic Encephalomyelitis. J Transl Med. 2011, 9: 81-10.1186/1479-5876-9-81.PubMedCentralCrossRefPubMed
10.
Zurück zum Zitat 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.CrossRefPubMed 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.CrossRefPubMed
11.
Zurück zum Zitat Nijs J, Meeus M, Van Oosterwijck J, Ickmans K, Moorkens G, Hans G: In the mind or in the brain? Scientific evidence for central sensitisation in chronic fatigue syndrome. Eur J Clin Invest. 2012, 42: 203-212. 10.1111/j.1365-2362.2011.02575.x.CrossRefPubMed Nijs J, Meeus M, Van Oosterwijck J, Ickmans K, Moorkens G, Hans G: In the mind or in the brain? Scientific evidence for central sensitisation in chronic fatigue syndrome. Eur J Clin Invest. 2012, 42: 203-212. 10.1111/j.1365-2362.2011.02575.x.CrossRefPubMed
12.
Zurück zum Zitat Landay AL, Jessop C, Lennette ET, Levy JA: Chronic fatigue syndrome: clinical condition associated with immune activation. Lancet. 1991, 338: 707-712. 10.1016/0140-6736(91)91440-6.CrossRefPubMed Landay AL, Jessop C, Lennette ET, Levy JA: Chronic fatigue syndrome: clinical condition associated with immune activation. Lancet. 1991, 338: 707-712. 10.1016/0140-6736(91)91440-6.CrossRefPubMed
13.
Zurück zum Zitat Galbraith S, Cameron B, Li H, Lau D, Vollmer-Conna U, Lloyd AR: Peripheral blood gene expression in postinfective fatigue syndrome following from three different triggering infections. J Infect Dis. 2011, 204: 1632-1640. 10.1093/infdis/jir612.CrossRefPubMed Galbraith S, Cameron B, Li H, Lau D, Vollmer-Conna U, Lloyd AR: Peripheral blood gene expression in postinfective fatigue syndrome following from three different triggering infections. J Infect Dis. 2011, 204: 1632-1640. 10.1093/infdis/jir612.CrossRefPubMed
14.
Zurück zum Zitat Chia J, Chia A, Voeller M, Lee T, Chang R: Acute enterovirus infection followed by myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) and viral persistence. J Clin Pathol. 2010, 63: 165-168. 10.1136/jcp.2009.070466.CrossRefPubMed Chia J, Chia A, Voeller M, Lee T, Chang R: Acute enterovirus infection followed by myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) and viral persistence. J Clin Pathol. 2010, 63: 165-168. 10.1136/jcp.2009.070466.CrossRefPubMed
15.
Zurück zum Zitat Chia JKS, Chia AY: Chronic fatigue syndrome is associated with chronic enterovirus infection of the stomach. J Clin Pathol. 2008, 61: 43-48.CrossRefPubMed Chia JKS, Chia AY: Chronic fatigue syndrome is associated with chronic enterovirus infection of the stomach. J Clin Pathol. 2008, 61: 43-48.CrossRefPubMed
16.
Zurück zum Zitat Kerr JR, Gough J, Richards SCM, Main J, Enlander D, McCreary M: Antibody to parvovirus B19 nonstructural protein is associated with chronic arthralgia in patients with chronic fatigue syndrome/myalgic encephalomyelitis. J Gen Virol. 2010, 91: 893-897. 10.1099/vir.0.017590-0.CrossRefPubMed Kerr JR, Gough J, Richards SCM, Main J, Enlander D, McCreary M: Antibody to parvovirus B19 nonstructural protein is associated with chronic arthralgia in patients with chronic fatigue syndrome/myalgic encephalomyelitis. J Gen Virol. 2010, 91: 893-897. 10.1099/vir.0.017590-0.CrossRefPubMed
17.
Zurück zum Zitat Koelle DM, Barcy S, Huang M-L, Ashley RL, Corey L, Zeh J: Markers of viral infection in monozygotic twins discordant for chronic fatigue syndrome. Clin Infect Dis. 2002, 35: 518-525. 10.1086/341774.CrossRefPubMed Koelle DM, Barcy S, Huang M-L, Ashley RL, Corey L, Zeh J: Markers of viral infection in monozygotic twins discordant for chronic fatigue syndrome. Clin Infect Dis. 2002, 35: 518-525. 10.1086/341774.CrossRefPubMed
18.
Zurück zum Zitat Watt T, Oberfoell S, Balise R, Lunn MR, Kar AK, Merrihew L: Response to valganciclovir in chronic fatigue syndrome patients with human herpesvirus 6 and Epstein-Barr virus IgG antibody titers. J Med Virol. 2012, 84: 1967-1974. 10.1002/jmv.23411.CrossRefPubMed Watt T, Oberfoell S, Balise R, Lunn MR, Kar AK, Merrihew L: Response to valganciclovir in chronic fatigue syndrome patients with human herpesvirus 6 and Epstein-Barr virus IgG antibody titers. J Med Virol. 2012, 84: 1967-1974. 10.1002/jmv.23411.CrossRefPubMed
19.
Zurück zum Zitat Fluge O, Bruland O, Risa K, Storstein A, Kristoffersen EK, Sapkota D: Benefit from B-Lymphocyte Depletion Using the Anti-CD20 Antibody Rituximab in Chronic Fatigue Syndrome. A Double-Blind and Placebo-Controlled Study. PLoS One. 2011, 6: e26358-10.1371/journal.pone.0026358.PubMedCentralCrossRefPubMed Fluge O, Bruland O, Risa K, Storstein A, Kristoffersen EK, Sapkota D: Benefit from B-Lymphocyte Depletion Using the Anti-CD20 Antibody Rituximab in Chronic Fatigue Syndrome. A Double-Blind and Placebo-Controlled Study. PLoS One. 2011, 6: e26358-10.1371/journal.pone.0026358.PubMedCentralCrossRefPubMed
20.
Zurück zum Zitat Fluge , 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: 28-10.1186/1471-2377-9-28.PubMedCentralCrossRefPubMed Fluge , 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: 28-10.1186/1471-2377-9-28.PubMedCentralCrossRefPubMed
21.
Zurück zum Zitat Swanink CM, Vercoulen JH, Galama JM, Roos MT, Meyaard L, van der Ven-Jongekrijg J: Lymphocyte subsets, apoptosis, and cytokines in patients with chronic fatigue syndrome. J Infect Dis. 1996, 173: 460-463. 10.1093/infdis/173.2.460.CrossRefPubMed Swanink CM, Vercoulen JH, Galama JM, Roos MT, Meyaard L, van der Ven-Jongekrijg J: Lymphocyte subsets, apoptosis, and cytokines in patients with chronic fatigue syndrome. J Infect Dis. 1996, 173: 460-463. 10.1093/infdis/173.2.460.CrossRefPubMed
22.
Zurück zum Zitat Fletcher MA, Zeng XR, Maher K, Levis S, Hurwitz B, Antoni M: Biomarkers in chronic fatigue syndrome: evaluation of natural killer cell function and dipeptidyl peptidase IV/CD26. PLoS One. 2010, 5: e10817-10.1371/journal.pone.0010817.PubMedCentralCrossRefPubMed Fletcher MA, Zeng XR, Maher K, Levis S, Hurwitz B, Antoni M: Biomarkers in chronic fatigue syndrome: evaluation of natural killer cell function and dipeptidyl peptidase IV/CD26. PLoS One. 2010, 5: e10817-10.1371/journal.pone.0010817.PubMedCentralCrossRefPubMed
23.
Zurück zum Zitat Mihaylova I, DeRuyter M, Rummens J-L, Bosmans E, Maes M: Decreased expression of CD69 in chronic fatigue syndrome in relation to inflammatory markers: evidence for a severe disorder in the early activation of T lymphocytes and natural killer cells. Neuro Endocrinol Lett. 2007, 28: 477-483.PubMed Mihaylova I, DeRuyter M, Rummens J-L, Bosmans E, Maes M: Decreased expression of CD69 in chronic fatigue syndrome in relation to inflammatory markers: evidence for a severe disorder in the early activation of T lymphocytes and natural killer cells. Neuro Endocrinol Lett. 2007, 28: 477-483.PubMed
24.
Zurück zum Zitat Buchwald D, Komaroff AL: Review of Laboratory Findings for Patients with Chronic Fatigue Syndrome. Rev Infect Dis. 1991, 13: S12-S18. 10.1093/clinids/13.Supplement_1.S12.CrossRefPubMed Buchwald D, Komaroff AL: Review of Laboratory Findings for Patients with Chronic Fatigue Syndrome. Rev Infect Dis. 1991, 13: S12-S18. 10.1093/clinids/13.Supplement_1.S12.CrossRefPubMed
25.
Zurück zum Zitat Fletcher MA, Zeng XR, Barnes Z, Levis S, Klimas NG: Plasma cytokines in women with chronic fatigue syndrome. J Transl Med. 2009, 7: 96-10.1186/1479-5876-7-96.PubMedCentralCrossRefPubMed Fletcher MA, Zeng XR, Barnes Z, Levis S, Klimas NG: Plasma cytokines in women with chronic fatigue syndrome. J Transl Med. 2009, 7: 96-10.1186/1479-5876-7-96.PubMedCentralCrossRefPubMed
26.
Zurück zum Zitat 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: 1209-1217. 10.1016/j.bbi.2010.04.012.PubMedCentralCrossRefPubMed 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: 1209-1217. 10.1016/j.bbi.2010.04.012.PubMedCentralCrossRefPubMed
27.
Zurück zum Zitat Lombardi VC, Hagen KS, Hunter KW, Diamond JW, Smith-Gagen J, Yang W: Xenotropic murine leukemia virus-related virus-associated chronic fatigue syndrome reveals a distinct inflammatory signature. In Vivo. 2011, 25: 307-314.PubMed Lombardi VC, Hagen KS, Hunter KW, Diamond JW, Smith-Gagen J, Yang W: Xenotropic murine leukemia virus-related virus-associated chronic fatigue syndrome reveals a distinct inflammatory signature. In Vivo. 2011, 25: 307-314.PubMed
28.
Zurück zum Zitat Hanevik K, Kristoffersen EK, Sørnes S, Mørch K, Næss H, Rivenes AC: Immunophenotyping in post-giardiasis functional gastrointestinal disease and chronic fatigue syndrome. BMC Infect Dis. 2012, 12: 258-10.1186/1471-2334-12-258.PubMedCentralCrossRefPubMed Hanevik K, Kristoffersen EK, Sørnes S, Mørch K, Næss H, Rivenes AC: Immunophenotyping in post-giardiasis functional gastrointestinal disease and chronic fatigue syndrome. BMC Infect Dis. 2012, 12: 258-10.1186/1471-2334-12-258.PubMedCentralCrossRefPubMed
29.
Zurück zum Zitat Brenu EW, van Driel ML, Staines DR, Ashton KJ, Hardcastle SL, Keane J: 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.PubMedCentralCrossRefPubMed Brenu EW, van Driel ML, Staines DR, Ashton KJ, Hardcastle SL, Keane J: 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.PubMedCentralCrossRefPubMed
30.
Zurück zum Zitat Morrison LJ, Behan WH, Behan PO: Changes in natural killer cell phenotype in patients with post-viral fatigue syndrome. Clin Exp Immunol. 1991, 83: 441-446.PubMedCentralCrossRefPubMed Morrison LJ, Behan WH, Behan PO: Changes in natural killer cell phenotype in patients with post-viral fatigue syndrome. Clin Exp Immunol. 1991, 83: 441-446.PubMedCentralCrossRefPubMed
31.
Zurück zum Zitat Barker E, Fujimura SF, Fadem MB, Landay AL, Levy JA: Immunologic abnormalities associated with chronic fatigue syndrome. Clin Infect Dis. 1994, 18 (Suppl 1): S136-S141.CrossRefPubMed Barker E, Fujimura SF, Fadem MB, Landay AL, Levy JA: Immunologic abnormalities associated with chronic fatigue syndrome. Clin Infect Dis. 1994, 18 (Suppl 1): S136-S141.CrossRefPubMed
32.
Zurück zum Zitat Stewart CC, Cookfair DL, Hovey KM, Wende KE, Bell DS, Warner CL: Predictive immunophenotypes: disease-related profile in chronic fatigue syndrome. Cytometry B Clin Cytom. 2003, 53: 26-33.CrossRefPubMed Stewart CC, Cookfair DL, Hovey KM, Wende KE, Bell DS, Warner CL: Predictive immunophenotypes: disease-related profile in chronic fatigue syndrome. Cytometry B Clin Cytom. 2003, 53: 26-33.CrossRefPubMed
33.
Zurück zum Zitat Robertson MJ, Schacterle RS, Mackin GA, Wilson SN, Bloomingdale KL, Ritz J: Lymphocyte subset differences in patients with chronic fatigue syndrome, multiple sclerosis and major depression. Clin Exp Immunol. 2005, 141: 326-332. 10.1111/j.1365-2249.2005.02833.x.PubMedCentralCrossRefPubMed Robertson MJ, Schacterle RS, Mackin GA, Wilson SN, Bloomingdale KL, Ritz J: Lymphocyte subset differences in patients with chronic fatigue syndrome, multiple sclerosis and major depression. Clin Exp Immunol. 2005, 141: 326-332. 10.1111/j.1365-2249.2005.02833.x.PubMedCentralCrossRefPubMed
34.
Zurück zum Zitat Massanella M, Negredo E, Pérez-Alvarez N, Puig J, Ruiz-Hernández R, Bofill M: CD4 T-cell hyperactivation and susceptibility to cell death determine poor CD4 T-cell recovery during suppressive HAART. AIDS. 2010, 24: 959-968. 10.1097/QAD.0b013e328337b957.CrossRefPubMed Massanella M, Negredo E, Pérez-Alvarez N, Puig J, Ruiz-Hernández R, Bofill M: CD4 T-cell hyperactivation and susceptibility to cell death determine poor CD4 T-cell recovery during suppressive HAART. AIDS. 2010, 24: 959-968. 10.1097/QAD.0b013e328337b957.CrossRefPubMed
35.
Zurück zum Zitat Gress RE, Deeks SG: Reduced thymus activity and infection prematurely age the immune system. J Clin Invest. 2009, 119: 2884-2887. 10.1172/JCI40855.PubMedCentralCrossRefPubMed Gress RE, Deeks SG: Reduced thymus activity and infection prematurely age the immune system. J Clin Invest. 2009, 119: 2884-2887. 10.1172/JCI40855.PubMedCentralCrossRefPubMed
36.
37.
Zurück zum Zitat Zigmond AS, Snaith RP: The hospital anxiety and depression scale. Acta Psychiatr Scand. 1983, 67: 361-370. 10.1111/j.1600-0447.1983.tb09716.x.CrossRefPubMed Zigmond AS, Snaith RP: The hospital anxiety and depression scale. Acta Psychiatr Scand. 1983, 67: 361-370. 10.1111/j.1600-0447.1983.tb09716.x.CrossRefPubMed
38.
Zurück zum Zitat Alijotas J, Alegre J, Fernández-Solà J, Cots JM, Panisello J, Peri JM: Consensus report on the diagnosis and treatment of chronique fatigue syndrome in Catalonia. Med Clin (Barc). 2002, 118: 73-76.CrossRef Alijotas J, Alegre J, Fernández-Solà J, Cots JM, Panisello J, Peri JM: Consensus report on the diagnosis and treatment of chronique fatigue syndrome in Catalonia. Med Clin (Barc). 2002, 118: 73-76.CrossRef
39.
Zurück zum Zitat Barretina J, Blanco J, Bonjoch A, Llano A, Clotet B, Esté JA: Immunological and virological study of enfuvirtide-treated HIV-positive patients. AIDS. 2004, 18: 1673-1682. 10.1097/01.aids.0000131350.22032.b5.CrossRefPubMed Barretina J, Blanco J, Bonjoch A, Llano A, Clotet B, Esté JA: Immunological and virological study of enfuvirtide-treated HIV-positive patients. AIDS. 2004, 18: 1673-1682. 10.1097/01.aids.0000131350.22032.b5.CrossRefPubMed
40.
Zurück zum Zitat Negredo E, Massanella M, Puig J, Pérez-Alvarez N, Gallego-Escuredo JM, Villarroya J: Nadir CD4 T cell count as predictor and high CD4 T cell intrinsic apoptosis as final mechanism of poor CD4 T cell recovery in virologically suppressed HIV-infected patients: clinical implications. Clin Infect Dis. 2010, 50: 1300-1308. 10.1086/651689.CrossRefPubMed Negredo E, Massanella M, Puig J, Pérez-Alvarez N, Gallego-Escuredo JM, Villarroya J: Nadir CD4 T cell count as predictor and high CD4 T cell intrinsic apoptosis as final mechanism of poor CD4 T cell recovery in virologically suppressed HIV-infected patients: clinical implications. Clin Infect Dis. 2010, 50: 1300-1308. 10.1086/651689.CrossRefPubMed
41.
Zurück zum Zitat Blanco J, Barretina J, Clotet B, Esté JA: R5 HIV gp120-mediated cellular contacts induce the death of single CCR5-expressing CD4 T cells by a gp41-dependent mechanism. J Leukoc Biol. 2004, 76: 804-811. 10.1189/jlb.0204100.CrossRefPubMed Blanco J, Barretina J, Clotet B, Esté JA: R5 HIV gp120-mediated cellular contacts induce the death of single CCR5-expressing CD4 T cells by a gp41-dependent mechanism. J Leukoc Biol. 2004, 76: 804-811. 10.1189/jlb.0204100.CrossRefPubMed
42.
Zurück zum Zitat Allegra S, Deleine C, Michael-Jubely R, Gryson C, Boirie Y, Kantakamalakul W: Implementation of the EGFP-K562 flow cytometric NK test: determination of NK cytotoxic activity in healthy elderly volunteers before and after feeding. Cytometry A. 2006, 69: 992-998.CrossRefPubMed Allegra S, Deleine C, Michael-Jubely R, Gryson C, Boirie Y, Kantakamalakul W: Implementation of the EGFP-K562 flow cytometric NK test: determination of NK cytotoxic activity in healthy elderly volunteers before and after feeding. Cytometry A. 2006, 69: 992-998.CrossRefPubMed
43.
Zurück zum Zitat Kantakamalakul W, Jaroenpool J, Pattanapanyasat K: A novel enhanced green fluorescent protein (EGFP)-K562 flow cytometric method for measuring natural killer (NK) cell cytotoxic activity. J Immunol Methods. 2003, 272: 189-197. 10.1016/S0022-1759(02)00505-7.CrossRefPubMed Kantakamalakul W, Jaroenpool J, Pattanapanyasat K: A novel enhanced green fluorescent protein (EGFP)-K562 flow cytometric method for measuring natural killer (NK) cell cytotoxic activity. J Immunol Methods. 2003, 272: 189-197. 10.1016/S0022-1759(02)00505-7.CrossRefPubMed
44.
Zurück zum Zitat Lorusso L, Mikhaylova SV, Capelli E, Ferrari D, Ngonga GK, Ricevuti G: Immunological aspects of chronic fatigue syndrome. Autoimmun Rev. 2009, 8: 287-291. 10.1016/j.autrev.2008.08.003.CrossRefPubMed Lorusso L, Mikhaylova SV, Capelli E, Ferrari D, Ngonga GK, Ricevuti G: Immunological aspects of chronic fatigue syndrome. Autoimmun Rev. 2009, 8: 287-291. 10.1016/j.autrev.2008.08.003.CrossRefPubMed
45.
Zurück zum Zitat Stricker RB, Winger EE: Decreased CD57 lymphocyte subset in patients with chronic Lyme disease. Immunol Lett. 2001, 76: 43-48. 10.1016/S0165-2478(00)00316-3.CrossRefPubMed Stricker RB, Winger EE: Decreased CD57 lymphocyte subset in patients with chronic Lyme disease. Immunol Lett. 2001, 76: 43-48. 10.1016/S0165-2478(00)00316-3.CrossRefPubMed
46.
Zurück zum Zitat Appay V, van Lier RAW, Sallusto F, Roederer M: Phenotype and function of human T lymphocyte subsets: consensus and issues. Cytometry A. 2008, 73: 975-983.CrossRefPubMed Appay V, van Lier RAW, Sallusto F, Roederer M: Phenotype and function of human T lymphocyte subsets: consensus and issues. Cytometry A. 2008, 73: 975-983.CrossRefPubMed
47.
Zurück zum Zitat Appay V, Fastenackels S, Katlama C, Ait-Mohand H, Schneider L, Guihot A: Old age and anti-CMV immunity are associated with altered T cell reconstitution in HIV-1 infected patients. AIDS. 2011, 25: 1813-1822. 10.1097/QAD.0b013e32834640e6.CrossRefPubMed Appay V, Fastenackels S, Katlama C, Ait-Mohand H, Schneider L, Guihot A: Old age and anti-CMV immunity are associated with altered T cell reconstitution in HIV-1 infected patients. AIDS. 2011, 25: 1813-1822. 10.1097/QAD.0b013e32834640e6.CrossRefPubMed
48.
49.
Zurück zum Zitat Hawiger D, Masilamani RF, Bettelli E, Kuchroo VK, Nussenzweig MC: Immunological unresponsiveness characterized by increased expression of CD5 on peripheral T cells induced by dendritic cells in vivo. Immunity. 2004, 20: 695-705. 10.1016/j.immuni.2004.05.002.CrossRefPubMed Hawiger D, Masilamani RF, Bettelli E, Kuchroo VK, Nussenzweig MC: Immunological unresponsiveness characterized by increased expression of CD5 on peripheral T cells induced by dendritic cells in vivo. Immunity. 2004, 20: 695-705. 10.1016/j.immuni.2004.05.002.CrossRefPubMed
50.
Zurück zum Zitat Stamou P, de Jersey J, Carmignac D, Mamalaki C, Kioussis D, Stockinger B: Chronic exposure to low levels of antigen in the periphery causes reversible functional impairment correlating with changes in CD5 levels in monoclonal CD8 T cells. J Immunol. 2003, 171: 1278-1284.CrossRefPubMed Stamou P, de Jersey J, Carmignac D, Mamalaki C, Kioussis D, Stockinger B: Chronic exposure to low levels of antigen in the periphery causes reversible functional impairment correlating with changes in CD5 levels in monoclonal CD8 T cells. J Immunol. 2003, 171: 1278-1284.CrossRefPubMed
51.
Zurück zum Zitat Tirelli U, Marotta G, Improta S, Pinto A: Immunological abnormalities in patients with chronic fatigue syndrome. Scand J Immunol. 1994, 40: 601-608. 10.1111/j.1365-3083.1994.tb03511.x.CrossRefPubMed Tirelli U, Marotta G, Improta S, Pinto A: Immunological abnormalities in patients with chronic fatigue syndrome. Scand J Immunol. 1994, 40: 601-608. 10.1111/j.1365-3083.1994.tb03511.x.CrossRefPubMed
52.
Zurück zum Zitat Russell IJ, Vipraio GA, Michalek JE, Craig FE, Kang YK, Richards AB: Lymphocyte markers and natural killer cell activity in fibromyalgia syndrome: effects of low-dose, sublingual use of human interferon-alpha. J Interferon Cytokine Res. 1999, 19: 969-978. 10.1089/107999099313523.CrossRefPubMed Russell IJ, Vipraio GA, Michalek JE, Craig FE, Kang YK, Richards AB: Lymphocyte markers and natural killer cell activity in fibromyalgia syndrome: effects of low-dose, sublingual use of human interferon-alpha. J Interferon Cytokine Res. 1999, 19: 969-978. 10.1089/107999099313523.CrossRefPubMed
53.
Zurück zum Zitat Soto NE, Straus SE: Chronic Fatigue Syndrome and Herpesviruses: the Fading Evidence. Herpes. 2000, 7: 46-50.PubMed Soto NE, Straus SE: Chronic Fatigue Syndrome and Herpesviruses: the Fading Evidence. Herpes. 2000, 7: 46-50.PubMed
54.
Zurück zum Zitat Jost S, Quillay H, Reardon J, Peterson E, Simmons RP, Parry BA: Changes in cytokine levels and NK cell activation associated with influenza. PLoS One. 2011, 6: e25060-10.1371/journal.pone.0025060.PubMedCentralCrossRefPubMed Jost S, Quillay H, Reardon J, Peterson E, Simmons RP, Parry BA: Changes in cytokine levels and NK cell activation associated with influenza. PLoS One. 2011, 6: e25060-10.1371/journal.pone.0025060.PubMedCentralCrossRefPubMed
55.
Zurück zum Zitat Narni-Mancinelli E, Jaeger BN, Bernat C, Fenis A, Kung S, De Gassart A: Tuning of natural killer cell reactivity by NKp46 and Helios calibrates T cell responses. Science. 2012, 335: 344-348. 10.1126/science.1215621.CrossRefPubMed Narni-Mancinelli E, Jaeger BN, Bernat C, Fenis A, Kung S, De Gassart A: Tuning of natural killer cell reactivity by NKp46 and Helios calibrates T cell responses. Science. 2012, 335: 344-348. 10.1126/science.1215621.CrossRefPubMed
56.
Zurück zum Zitat Sun JC, Beilke JN, Lanier LL: Immune memory redefined: characterizing the longevity of natural killer cells. Immunol Rev. 2010, 236: 83-94. 10.1111/j.1600-065X.2010.00900.x.PubMedCentralCrossRefPubMed Sun JC, Beilke JN, Lanier LL: Immune memory redefined: characterizing the longevity of natural killer cells. Immunol Rev. 2010, 236: 83-94. 10.1111/j.1600-065X.2010.00900.x.PubMedCentralCrossRefPubMed
57.
Zurück zum Zitat Gumá M, Cabrera C, Erkizia I, Bofill M, Clotet B, Ruiz L: Human cytomegalovirus infection is associated with increased proportions of NK cells that express the CD94/NKG2C receptor in aviremic HIV-1-positive patients. J Infect Dis. 2006, 194: 38-41. 10.1086/504719.CrossRefPubMed Gumá M, Cabrera C, Erkizia I, Bofill M, Clotet B, Ruiz L: Human cytomegalovirus infection is associated with increased proportions of NK cells that express the CD94/NKG2C receptor in aviremic HIV-1-positive patients. J Infect Dis. 2006, 194: 38-41. 10.1086/504719.CrossRefPubMed
58.
Zurück zum Zitat Gayoso I, Sanchez-Correa B, Campos C, Alonso C, Pera A, Casado JG: Immunosenescence of human natural killer cells. J Innate Immun. 2011, 3: 337-343. 10.1159/000328005.CrossRefPubMed Gayoso I, Sanchez-Correa B, Campos C, Alonso C, Pera A, Casado JG: Immunosenescence of human natural killer cells. J Innate Immun. 2011, 3: 337-343. 10.1159/000328005.CrossRefPubMed
59.
Zurück zum Zitat Gupta S, Vayuvegula B: A comprehensive immunological analysis in chronic fatigue syndrome. Scand J Immunol. 1991, 33: 319-327. 10.1111/j.1365-3083.1991.tb01777.x.CrossRefPubMed Gupta S, Vayuvegula B: A comprehensive immunological analysis in chronic fatigue syndrome. Scand J Immunol. 1991, 33: 319-327. 10.1111/j.1365-3083.1991.tb01777.x.CrossRefPubMed
60.
Zurück zum Zitat Brenchley JM, Price DA, Schacker TW, Asher TE, Silvestri G, Rao S: Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat Med. 2006, 12: 1365-1371.CrossRefPubMed Brenchley JM, Price DA, Schacker TW, Asher TE, Silvestri G, Rao S: Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat Med. 2006, 12: 1365-1371.CrossRefPubMed
61.
Zurück zum Zitat 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. Neuro Endocrinol Lett. 2008, 29: 902-910.PubMed 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. Neuro Endocrinol Lett. 2008, 29: 902-910.PubMed
62.
Zurück zum Zitat Josefowicz SZ, Lu L-F, Rudensky AY: Regulatory T cells: mechanisms of differentiation and function. Annu Rev Immunol. 2012, 30: 531-564. 10.1146/annurev.immunol.25.022106.141623.CrossRefPubMed Josefowicz SZ, Lu L-F, Rudensky AY: Regulatory T cells: mechanisms of differentiation and function. Annu Rev Immunol. 2012, 30: 531-564. 10.1146/annurev.immunol.25.022106.141623.CrossRefPubMed
63.
Zurück zum Zitat Hall BM, Verma ND, Tran GT, Hodgkinson SJ: Distinct regulatory CD4 + T cell subsets; differences between naïve and antigen specific T regulatory cells. Curr Opin Immunol. 2011, 23: 641-647. 10.1016/j.coi.2011.07.012.CrossRefPubMed Hall BM, Verma ND, Tran GT, Hodgkinson SJ: Distinct regulatory CD4 + T cell subsets; differences between naïve and antigen specific T regulatory cells. Curr Opin Immunol. 2011, 23: 641-647. 10.1016/j.coi.2011.07.012.CrossRefPubMed
64.
Zurück zum Zitat Bennett AL, Chao CC, Hu S, Buchwald D, Fagioli LR, Schur PH: Elevation of bioactive transforming growth factor-beta in serum from patients with chronic fatigue syndrome. J Clin Immunol. 1997, 17: 160-166. 10.1023/A:1027330616073.CrossRefPubMed Bennett AL, Chao CC, Hu S, Buchwald D, Fagioli LR, Schur PH: Elevation of bioactive transforming growth factor-beta in serum from patients with chronic fatigue syndrome. J Clin Immunol. 1997, 17: 160-166. 10.1023/A:1027330616073.CrossRefPubMed
65.
Zurück zum Zitat Zhang H-Y, Liu Z-D, Hu C-J, Wang D-X, Zhang Y-B, Li Y-Z: Up-regulation of TGF-β1 mRNA expression in peripheral blood mononuclear cells of patients with chronic fatigue syndrome. J Formos Med Assoc. 2011, 110: 701-704. 10.1016/j.jfma.2011.09.006.CrossRefPubMed Zhang H-Y, Liu Z-D, Hu C-J, Wang D-X, Zhang Y-B, Li Y-Z: Up-regulation of TGF-β1 mRNA expression in peripheral blood mononuclear cells of patients with chronic fatigue syndrome. J Formos Med Assoc. 2011, 110: 701-704. 10.1016/j.jfma.2011.09.006.CrossRefPubMed
66.
Zurück zum Zitat Fletcher MA, Rosenthal M, Antoni M, Ironson G, Zeng XR, Barnes Z: Plasma neuropeptide Y: a biomarker for symptom severity in chronic fatigue syndrome. Behav Brain Funct. 2010, 6: 76-10.1186/1744-9081-6-76.PubMedCentralCrossRefPubMed Fletcher MA, Rosenthal M, Antoni M, Ironson G, Zeng XR, Barnes Z: Plasma neuropeptide Y: a biomarker for symptom severity in chronic fatigue syndrome. Behav Brain Funct. 2010, 6: 76-10.1186/1744-9081-6-76.PubMedCentralCrossRefPubMed
67.
Zurück zum Zitat Brenu EW, Staines DR, Baskurt OK, Ashton KJ, Ramos SB, Christy RM: Immune and hemorheological changes in chronic fatigue syndrome. J Transl Med. 2010, 8: 1-10.1186/1479-5876-8-1.PubMedCentralCrossRefPubMed Brenu EW, Staines DR, Baskurt OK, Ashton KJ, Ramos SB, Christy RM: Immune and hemorheological changes in chronic fatigue syndrome. J Transl Med. 2010, 8: 1-10.1186/1479-5876-8-1.PubMedCentralCrossRefPubMed
Metadaten
Titel
Screening NK-, B- and T-cell phenotype and function in patients suffering from Chronic Fatigue Syndrome
verfasst von
Marta Curriu
Jorge Carrillo
Marta Massanella
Josepa Rigau
José Alegre
Jordi Puig
Ana M Garcia-Quintana
Jesus Castro-Marrero
Eugènia Negredo
Bonaventura Clotet
Cecilia Cabrera
Julià Blanco
Publikationsdatum
01.12.2013
Verlag
BioMed Central
Erschienen in
Journal of Translational Medicine / Ausgabe 1/2013
Elektronische ISSN: 1479-5876
DOI
https://doi.org/10.1186/1479-5876-11-68

Weitere Artikel der Ausgabe 1/2013

Journal of Translational Medicine 1/2013 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.