Abstract

BackgroundQ fever fatigue syndrome (QFS) is characterised by a state of prolonged fatigue that is seen in 20% of acute Q fever infections and has major health-related consequences. The molecular mechanisms underlying QFS are largely unclear. In order to better understand its pathogenesis, we applied a multi-omics approach to study the patterns of the gut microbiome, blood metabolome, and inflammatory proteome of QFS patients, and compared these with those of chronic fatigue syndrome (CFS) patients and healthy controls (HC).MethodsThe study population consisted of 31 QFS patients, 50 CFS patients, and 72 HC. All subjects were matched for age, gender, and general geographical region (South-East part of the Netherlands). The gut microbiome composition was assessed by Metagenomic sequencing using the Illumina HiSeq platform. A total of 92 circulating inflammatory markers were measured using Proximity Extension Essay and 1607 metabolic features were assessed with a high-throughput non-targeted metabolomics approach.ResultsInflammatory markers, including 4E-BP1 (P = 9.60–16 and 1.41–7) and MMP-1 (P = 7.09–9 and 3.51–9), are significantly more expressed in both QFS and CFS patients compared to HC. Blood metabolite profiles show significant differences when comparing QFS (319 metabolites) and CFS (441 metabolites) patients to HC, and are significantly enriched in pathways like sphingolipid (P = 0.0256 and 0.0033) metabolism. When comparing QFS to CFS patients, almost no significant differences in metabolome were found. Comparison of microbiome taxonomy of QFS and CFS patients with that of HC, shows both in- and decreases in abundancies in Bacteroidetes (with emphasis on Bacteroides and Alistiples spp.), and Firmicutes and Actinobacteria (with emphasis on Ruminococcus and Bifidobacterium spp.). When we compare QFS patients to CFS patients, there is a striking resemblance and hardly any significant differences in microbiome taxonomy are found.ConclusionsWe show that QFS and CFS patients are similar across three different omics layers and 4E-BP1 and MMP-1 have the potential to distinguish QFS and CFS patients from HC.

Highlights

  • Q fever fatigue syndrome (QFS) is characterised by a state of prolonged fatigue that is seen in 20% of acute Q fever infections and has major health-related consequences

  • Mean fatigue severity scores were significantly higher for chronic fatigue syndrome (CFS) patients compared to QFS patients (Student’s t test, P = 0.0034)

  • The following variables proved most important in prediction when comparing QFS with healthy controls (HC); 4E binding protein 1 (4E-BP1), CD40, AXIN1, CCL11, CD244, IL-8, OPG, CCL4, TRAIL, and CD8A, CFS with HC; 4E-BP1, CDCP1, AXIN1, Matrix metalloproteinase (MMP)-10, CSF-1, TNFB, NT-3, FGF-23, IL-12B, and IL-8, and QFS and CFS with HC; 4E-BP1, AXIN1, CD40, CDCP1, CSF1, IL-8, FGF-23, CCL4, ADA, and MMP-10 (Additional file 3: Figure S3)

Read more

Summary

Introduction

Q fever fatigue syndrome (QFS) is characterised by a state of prolonged fatigue that is seen in 20% of acute Q fever infections and has major health-related consequences. Q fever fatigue syndrome (QFS) is characterised as a state of prolonged fatigue following acute Q fever infection [1]. Complaints of QFS are similar to those of chronic fatigue syndrome (CFS) [4, 5], and the pathogenesis of these syndromes remains largely unclear. Acute Q fever often is a self-limiting disease that usually presents as a flu-like illness that may be accompanied by pneumonia or hepatitis [7]. Of all those who become infected with C. burnetii, both symptomatic and asymptomatic, around 1–5% develop a persistent infection with the bacterium, called chronic Q fever or persistent focalised infection [9]. Of all those who develop acute Q fever, i.e., symptomatic, infection, around 20% will develop QFS [1]

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call