Cargando…

Increased gut permeability and bacterial translocation are associated with fibromyalgia and myalgic encephalomyelitis/chronic fatigue syndrome: implications for disease-related biomarker discovery

BACKGROUND: There is growing evidence of the significance of gastrointestinal complaints in the impairment of the intestinal mucosal barrier function and inflammation in fibromyalgia (FM) and in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). However, data on intestinal permeability and...

Descripción completa

Detalles Bibliográficos
Autores principales: Martín, Franz, Blanco-Suárez, Manuel, Zambrano, Paola, Cáceres, Oscar, Almirall, Miriam, Alegre-Martín, José, Lobo, Beatriz, González-Castro, Ana Maria, Santos, Javier, Domingo, Joan Carles, Jurek, Joanna, Castro-Marrero, Jesús
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10512706/
https://www.ncbi.nlm.nih.gov/pubmed/37744357
http://dx.doi.org/10.3389/fimmu.2023.1253121
_version_ 1785108419615653888
author Martín, Franz
Blanco-Suárez, Manuel
Zambrano, Paola
Cáceres, Oscar
Almirall, Miriam
Alegre-Martín, José
Lobo, Beatriz
González-Castro, Ana Maria
Santos, Javier
Domingo, Joan Carles
Jurek, Joanna
Castro-Marrero, Jesús
author_facet Martín, Franz
Blanco-Suárez, Manuel
Zambrano, Paola
Cáceres, Oscar
Almirall, Miriam
Alegre-Martín, José
Lobo, Beatriz
González-Castro, Ana Maria
Santos, Javier
Domingo, Joan Carles
Jurek, Joanna
Castro-Marrero, Jesús
author_sort Martín, Franz
collection PubMed
description BACKGROUND: There is growing evidence of the significance of gastrointestinal complaints in the impairment of the intestinal mucosal barrier function and inflammation in fibromyalgia (FM) and in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). However, data on intestinal permeability and gut barrier dysfunction in FM and ME/CFS are still limited with conflicting results. This study aimed to assess circulating biomarkers potentially related to intestinal barrier dysfunction and bacterial translocation and their association with self-reported symptoms in these conditions. METHODS: A pilot multicenter, cross-sectional cohort study with consecutive enrolment of 22 patients with FM, 30 with ME/CFS and 26 matched healthy controls. Plasma levels of anti-beta-lactoglobulin antibodies (IgG anti-β-LGB), zonulin-1 (ZO-1), lipopolysaccharides (LPS), soluble CD14 (sCD14) and interleukin-1-beta (IL-1β) were assayed using ELISA. Demographic and clinical characteristics of the participants were recorded using validated self-reported outcome measures. The diagnostic accuracy of each biomarker was assessed using the receiver operating characteristic (ROC) curve analysis. RESULTS: FM patients had significantly higher levels of anti-β-LGB, ZO-1, LPS, and sCD14 than healthy controls (all P < 0.0001). In ME/CFS patients, levels of anti-β-LGB, ZO-1, LPS, and sCD14 were significantly higher than controls, but lower than in FM (all P < 0.01), while there was no significant difference in IL-1β level. In the FM and ME/CFS cohorts, both anti-β-LGB and ZO-1 correlated significantly with LPS and sCD14 (P < 0.001 for both). In the FM group, both anti-β-LGB and ZO-1 were correlated significantly with physical and mental health components on the SF-36 scale (P < 0.05); whereas IL-1β negatively correlated with the COMPASS-31 score (P < 0.05). In the ME/CFS cohort, ZO-1 was positively correlated with the COMPASS-31 score (P < 0.05). The ROC curve analysis indicated a strong ability of anti-β-LGB, ZO-1, LPS and sCD14 to predictively distinguish between FM and ME/CFS from healthy controls (P < 0.0001). CONCLUSION: Biomarkers of intestinal barrier function and inflammation were associated with autonomic dysfunction assessed by COMPASS-31 scores in FM and ME/CFS respectively. Anti-β-LGB antibodies, ZO-1, LPS, and sCD14 may be putative predictors of intestinal barrier dysfunction in these cohorts. Further studies are needed to assess whether these findings are causal and can therefore be applied in clinical practice.
format Online
Article
Text
id pubmed-10512706
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-105127062023-09-22 Increased gut permeability and bacterial translocation are associated with fibromyalgia and myalgic encephalomyelitis/chronic fatigue syndrome: implications for disease-related biomarker discovery Martín, Franz Blanco-Suárez, Manuel Zambrano, Paola Cáceres, Oscar Almirall, Miriam Alegre-Martín, José Lobo, Beatriz González-Castro, Ana Maria Santos, Javier Domingo, Joan Carles Jurek, Joanna Castro-Marrero, Jesús Front Immunol Immunology BACKGROUND: There is growing evidence of the significance of gastrointestinal complaints in the impairment of the intestinal mucosal barrier function and inflammation in fibromyalgia (FM) and in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). However, data on intestinal permeability and gut barrier dysfunction in FM and ME/CFS are still limited with conflicting results. This study aimed to assess circulating biomarkers potentially related to intestinal barrier dysfunction and bacterial translocation and their association with self-reported symptoms in these conditions. METHODS: A pilot multicenter, cross-sectional cohort study with consecutive enrolment of 22 patients with FM, 30 with ME/CFS and 26 matched healthy controls. Plasma levels of anti-beta-lactoglobulin antibodies (IgG anti-β-LGB), zonulin-1 (ZO-1), lipopolysaccharides (LPS), soluble CD14 (sCD14) and interleukin-1-beta (IL-1β) were assayed using ELISA. Demographic and clinical characteristics of the participants were recorded using validated self-reported outcome measures. The diagnostic accuracy of each biomarker was assessed using the receiver operating characteristic (ROC) curve analysis. RESULTS: FM patients had significantly higher levels of anti-β-LGB, ZO-1, LPS, and sCD14 than healthy controls (all P < 0.0001). In ME/CFS patients, levels of anti-β-LGB, ZO-1, LPS, and sCD14 were significantly higher than controls, but lower than in FM (all P < 0.01), while there was no significant difference in IL-1β level. In the FM and ME/CFS cohorts, both anti-β-LGB and ZO-1 correlated significantly with LPS and sCD14 (P < 0.001 for both). In the FM group, both anti-β-LGB and ZO-1 were correlated significantly with physical and mental health components on the SF-36 scale (P < 0.05); whereas IL-1β negatively correlated with the COMPASS-31 score (P < 0.05). In the ME/CFS cohort, ZO-1 was positively correlated with the COMPASS-31 score (P < 0.05). The ROC curve analysis indicated a strong ability of anti-β-LGB, ZO-1, LPS and sCD14 to predictively distinguish between FM and ME/CFS from healthy controls (P < 0.0001). CONCLUSION: Biomarkers of intestinal barrier function and inflammation were associated with autonomic dysfunction assessed by COMPASS-31 scores in FM and ME/CFS respectively. Anti-β-LGB antibodies, ZO-1, LPS, and sCD14 may be putative predictors of intestinal barrier dysfunction in these cohorts. Further studies are needed to assess whether these findings are causal and can therefore be applied in clinical practice. Frontiers Media S.A. 2023-09-07 /pmc/articles/PMC10512706/ /pubmed/37744357 http://dx.doi.org/10.3389/fimmu.2023.1253121 Text en Copyright © 2023 Martín, Blanco-Suárez, Zambrano, Cáceres, Almirall, Alegre-Martín, Lobo, González-Castro, Santos, Domingo, Jurek and Castro-Marrero https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Immunology
Martín, Franz
Blanco-Suárez, Manuel
Zambrano, Paola
Cáceres, Oscar
Almirall, Miriam
Alegre-Martín, José
Lobo, Beatriz
González-Castro, Ana Maria
Santos, Javier
Domingo, Joan Carles
Jurek, Joanna
Castro-Marrero, Jesús
Increased gut permeability and bacterial translocation are associated with fibromyalgia and myalgic encephalomyelitis/chronic fatigue syndrome: implications for disease-related biomarker discovery
title Increased gut permeability and bacterial translocation are associated with fibromyalgia and myalgic encephalomyelitis/chronic fatigue syndrome: implications for disease-related biomarker discovery
title_full Increased gut permeability and bacterial translocation are associated with fibromyalgia and myalgic encephalomyelitis/chronic fatigue syndrome: implications for disease-related biomarker discovery
title_fullStr Increased gut permeability and bacterial translocation are associated with fibromyalgia and myalgic encephalomyelitis/chronic fatigue syndrome: implications for disease-related biomarker discovery
title_full_unstemmed Increased gut permeability and bacterial translocation are associated with fibromyalgia and myalgic encephalomyelitis/chronic fatigue syndrome: implications for disease-related biomarker discovery
title_short Increased gut permeability and bacterial translocation are associated with fibromyalgia and myalgic encephalomyelitis/chronic fatigue syndrome: implications for disease-related biomarker discovery
title_sort increased gut permeability and bacterial translocation are associated with fibromyalgia and myalgic encephalomyelitis/chronic fatigue syndrome: implications for disease-related biomarker discovery
topic Immunology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10512706/
https://www.ncbi.nlm.nih.gov/pubmed/37744357
http://dx.doi.org/10.3389/fimmu.2023.1253121
work_keys_str_mv AT martinfranz increasedgutpermeabilityandbacterialtranslocationareassociatedwithfibromyalgiaandmyalgicencephalomyelitischronicfatiguesyndromeimplicationsfordiseaserelatedbiomarkerdiscovery
AT blancosuarezmanuel increasedgutpermeabilityandbacterialtranslocationareassociatedwithfibromyalgiaandmyalgicencephalomyelitischronicfatiguesyndromeimplicationsfordiseaserelatedbiomarkerdiscovery
AT zambranopaola increasedgutpermeabilityandbacterialtranslocationareassociatedwithfibromyalgiaandmyalgicencephalomyelitischronicfatiguesyndromeimplicationsfordiseaserelatedbiomarkerdiscovery
AT caceresoscar increasedgutpermeabilityandbacterialtranslocationareassociatedwithfibromyalgiaandmyalgicencephalomyelitischronicfatiguesyndromeimplicationsfordiseaserelatedbiomarkerdiscovery
AT almirallmiriam increasedgutpermeabilityandbacterialtranslocationareassociatedwithfibromyalgiaandmyalgicencephalomyelitischronicfatiguesyndromeimplicationsfordiseaserelatedbiomarkerdiscovery
AT alegremartinjose increasedgutpermeabilityandbacterialtranslocationareassociatedwithfibromyalgiaandmyalgicencephalomyelitischronicfatiguesyndromeimplicationsfordiseaserelatedbiomarkerdiscovery
AT lobobeatriz increasedgutpermeabilityandbacterialtranslocationareassociatedwithfibromyalgiaandmyalgicencephalomyelitischronicfatiguesyndromeimplicationsfordiseaserelatedbiomarkerdiscovery
AT gonzalezcastroanamaria increasedgutpermeabilityandbacterialtranslocationareassociatedwithfibromyalgiaandmyalgicencephalomyelitischronicfatiguesyndromeimplicationsfordiseaserelatedbiomarkerdiscovery
AT santosjavier increasedgutpermeabilityandbacterialtranslocationareassociatedwithfibromyalgiaandmyalgicencephalomyelitischronicfatiguesyndromeimplicationsfordiseaserelatedbiomarkerdiscovery
AT domingojoancarles increasedgutpermeabilityandbacterialtranslocationareassociatedwithfibromyalgiaandmyalgicencephalomyelitischronicfatiguesyndromeimplicationsfordiseaserelatedbiomarkerdiscovery
AT jurekjoanna increasedgutpermeabilityandbacterialtranslocationareassociatedwithfibromyalgiaandmyalgicencephalomyelitischronicfatiguesyndromeimplicationsfordiseaserelatedbiomarkerdiscovery
AT castromarrerojesus increasedgutpermeabilityandbacterialtranslocationareassociatedwithfibromyalgiaandmyalgicencephalomyelitischronicfatiguesyndromeimplicationsfordiseaserelatedbiomarkerdiscovery