Abstract

P2X7 receptor activation induces the release of different cellular proteins, such as CD14, a glycosylphosphatidylinositol (GPI)-anchored protein to the plasma membrane important for LPS signaling via TLR4. Circulating CD14 has been found at elevated levels in sepsis, but the exact mechanism of CD14 release in sepsis has not been established. Here we show for first time that P2X7 receptor induces the release of CD14 in extracellular vesicles, resulting in a net reduction in macrophage plasma membrane CD14 that functionally affects LPS, but not monophosphoryl lipid A, pro-inflammatory cytokine production. Also, we found that during a murine model of sepsis, P2X7 receptor activity is important for maintaining elevated levels of CD14 in biological fluids and a decrease in its activity results in higher bacterial load and exacerbated organ damage, ultimately leading to premature deaths. Our data reveal that P2X7 is a key receptor for helping to clear sepsis because it maintains elevated concentrations of circulating CD14 during infection.

Data availability

All data generated or analysed during this study are included in the manuscript and provided as raw data as single values for all Figures.

Article and author information

Author details

  1. Cristina Alarcón-Vila

    Molecular Inflammation and Experimental Surgery, Biomedical Research Institute of Murcia IMIB-Arrixaca, Murcia, Spain
    Competing interests
    The authors declare that no competing interests exist.
  2. Alberto Baroja-Mazo

    Molecular Inflammation and Experimental Surgery, Biomedical Research Institute of Murcia IMIB-Arrixaca, Murcia, Spain
    Competing interests
    The authors declare that no competing interests exist.
  3. Carlos de Torre-Minguela

    Molecular Inflammation and Experimental Surgery, Biomedical Research Institute of Murcia IMIB-Arrixaca, Murcia, Spain
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0003-3314-3203
  4. Carlos M Martínez

    Pathology Platform, Biomedical Research Institute of Murcia IMIB-Arrixaca, Murcia, Spain
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0003-3307-1326
  5. Juan J Martínez-García

    Molecular Inflammation and Experimental Surgery, Biomedical Research Institute of Murcia IMIB-Arrixaca, Murcia, Spain
    Competing interests
    The authors declare that no competing interests exist.
  6. Helios Martínez-Banaclocha

    Molecular Inflammation and Experimental Surgery, Biomedical Research Institute of Murcia IMIB-Arrixaca, Murcia, Spain
    Competing interests
    The authors declare that no competing interests exist.
  7. Carlos García-Palenciano

    Molecular Inflammation and Experimental Surgery, Biomedical Research Institute of Murcia IMIB-Arrixaca, Murcia, Spain
    Competing interests
    The authors declare that no competing interests exist.
  8. Pablo Pelegrin

    Molecular Inflammation and Experimental Surgery, Biomedical Research Institute of Murcia IMIB-Arrixaca, Murcia, Spain
    For correspondence
    pablo.pelegrin@imib.es
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-9688-1804

Funding

European research council (614578)

  • Pablo Pelegrin

European Research council (899636)

  • Carlos García-Palenciano
  • Pablo Pelegrin

Ministerio de Economía y Competitividad (SAF2017-88276-R)

  • Pablo Pelegrin

Fundacion Seneca (20859/PI/18)

  • Carlos García-Palenciano
  • Pablo Pelegrin

Fundacion Seneca (21081/PDC/19)

  • Pablo Pelegrin

The funders had no role in study design, data collection and interpretation, or the decision to submit the work for publication.

Reviewing Editor

  1. Evangelos J Giamarellos-Bourboulis, Attikon University Hospital, Greece

Ethics

Animal experimentation: All experimental protocols for animal handling were refined and approved by the Animal Health Service of the General Directorate of Fishing and Farming of the Council of Murcia (Servicio de Sanidad Animal, Dirección General de Ganadería y Pesca, Consejería de Agricultura y Agua Región de Murcia, permit reference A1320140201). Mice were used in accordance with the Hospital Clínico Universitario Virgen Arrixaca animal experimentation guidelines (Permit Number 221116/1/PE), and Spanish national (Royal Decree 1201/2005 and Law 32/2007) and EU (86/609/EEC and 335 2010/63/EU) legislation. All surgery was performed under sodium pentobarbital anesthesia, and every effort was made to minimize suffering.

Human subjects: The samples and data from patients included in this study were provided by the Biobanco en Red de la Región de Murcia (PT13/0010/0018), which is integrated into the Spanish National Biobanks Network (B.000859) and approved by the clinical ethics committee of the Clinical University Hospital Virgen de la Arrixaca (reference numbers PI13/00174, 2019-9-4-HCUVA, 2019-12-15-HCUVA and 2019-12-471 14-HCUVA). All study procedures were conducted in accordance with the declaration of Helsinki. Whole peripheral blood samples were collected after receiving written informed consent from intraabdominal sepsis patients at the Surgical Critical Unit from the Clinical University Hospital Virgen de la Arrixaca.

Version history

  1. Received: July 8, 2020
  2. Accepted: October 31, 2020
  3. Accepted Manuscript published: November 2, 2020 (version 1)
  4. Version of Record published: November 26, 2020 (version 2)

Copyright

© 2020, Alarcón-Vila et al.

This article is distributed under the terms of the Creative Commons Attribution License permitting unrestricted use and redistribution provided that the original author and source are credited.

Metrics

  • 1,281
    views
  • 283
    downloads
  • 27
    citations

Views, downloads and citations are aggregated across all versions of this paper published by eLife.

Download links

A two-part list of links to download the article, or parts of the article, in various formats.

Downloads (link to download the article as PDF)

Open citations (links to open the citations from this article in various online reference manager services)

Cite this article (links to download the citations from this article in formats compatible with various reference manager tools)

  1. Cristina Alarcón-Vila
  2. Alberto Baroja-Mazo
  3. Carlos de Torre-Minguela
  4. Carlos M Martínez
  5. Juan J Martínez-García
  6. Helios Martínez-Banaclocha
  7. Carlos García-Palenciano
  8. Pablo Pelegrin
(2020)
CD14 release induced by P2X7 receptor restrict inflammation and increases survival during sepsis
eLife 9:e60849.
https://doi.org/10.7554/eLife.60849

Share this article

https://doi.org/10.7554/eLife.60849

Further reading

    1. Evolutionary Biology
    2. Immunology and Inflammation
    Mark S Lee, Peter J Tuohy ... Michael S Kuhns
    Research Advance

    CD4+ T cell activation is driven by five-module receptor complexes. The T cell receptor (TCR) is the receptor module that binds composite surfaces of peptide antigens embedded within MHCII molecules (pMHCII). It associates with three signaling modules (CD3γε, CD3δε, and CD3ζζ) to form TCR-CD3 complexes. CD4 is the coreceptor module. It reciprocally associates with TCR-CD3-pMHCII assemblies on the outside of a CD4+ T cells and with the Src kinase, LCK, on the inside. Previously, we reported that the CD4 transmembrane GGXXG and cytoplasmic juxtamembrane (C/F)CV+C motifs found in eutherian (placental mammal) CD4 have constituent residues that evolved under purifying selection (Lee et al., 2022). Expressing mutants of these motifs together in T cell hybridomas increased CD4-LCK association but reduced CD3ζ, ZAP70, and PLCγ1 phosphorylation levels, as well as IL-2 production, in response to agonist pMHCII. Because these mutants preferentially localized CD4-LCK pairs to non-raft membrane fractions, one explanation for our results was that they impaired proximal signaling by sequestering LCK away from TCR-CD3. An alternative hypothesis is that the mutations directly impacted signaling because the motifs normally play an LCK-independent role in signaling. The goal of this study was to discriminate between these possibilities. Using T cell hybridomas, our results indicate that: intracellular CD4-LCK interactions are not necessary for pMHCII-specific signal initiation; the GGXXG and (C/F)CV+C motifs are key determinants of CD4-mediated pMHCII-specific signal amplification; the GGXXG and (C/F)CV+C motifs exert their functions independently of direct CD4-LCK association. These data provide a mechanistic explanation for why residues within these motifs are under purifying selection in jawed vertebrates. The results are also important to consider for biomimetic engineering of synthetic receptors.

    1. Genetics and Genomics
    2. Immunology and Inflammation
    Jean-David Larouche, Céline M Laumont ... Claude Perreault
    Research Article

    Transposable elements (TEs) are repetitive sequences representing ~45% of the human and mouse genomes and are highly expressed by medullary thymic epithelial cells (mTECs). In this study, we investigated the role of TEs on T-cell development in the thymus. We performed multiomic analyses of TEs in human and mouse thymic cells to elucidate their role in T-cell development. We report that TE expression in the human thymus is high and shows extensive age- and cell lineage-related variations. TE expression correlates with multiple transcription factors in all cell types of the human thymus. Two cell types express particularly broad TE repertoires: mTECs and plasmacytoid dendritic cells (pDCs). In mTECs, transcriptomic data suggest that TEs interact with transcription factors essential for mTEC development and function (e.g., PAX1 and REL), and immunopeptidomic data showed that TEs generate MHC-I-associated peptides implicated in thymocyte education. Notably, AIRE, FEZF2, and CHD4 regulate small yet non-redundant sets of TEs in murine mTECs. Human thymic pDCs homogenously express large numbers of TEs that likely form dsRNA, which can activate innate immune receptors, potentially explaining why thymic pDCs constitutively secrete IFN ɑ/β. This study highlights the diversity of interactions between TEs and the adaptive immune system. TEs are genetic parasites, and the two thymic cell types most affected by TEs (mTEcs and pDCs) are essential to establishing central T-cell tolerance. Therefore, we propose that orchestrating TE expression in thymic cells is critical to prevent autoimmunity in vertebrates.