Skip to main content
medRxiv
  • Home
  • About
  • Submit
  • ALERTS / RSS
Advanced Search

PD-L1 mediated T cell inhibition by regulatory plasma cells induced after sepsis and COVID-19

Morgane Gossez, Clara Vigneron, Alexandra Vandermoeten, Margot Lepage, Louise Courcol, Remy Coudereau, Helena Paidassai, Laurent Jallades, Jonathan Lopez, Khalil Kandara, Marine Ortillon, Marine Mommert, Astrid Fabri, Estelle Peronnet, Clémence Grosjean, Marielle Buisson, Anne-Claire Lukaszewicz, Thomas Rimmelé, Laurent Argaud, Martin Cour, Bénédicte F Py, Olivier Thaunat, Thierry Defrance, Guillaume Monneret, View ORCID ProfileFabienne Venet, REALISM, RICO study groups
doi: https://doi.org/10.1101/2024.08.08.24311630
Morgane Gossez
1Immunology Laboratory, Edouard Herriot Hospital – Hospices Civils de Lyon, 69437 Lyon Cedex 3, France
2CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Clara Vigneron
2CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Alexandra Vandermoeten
3Service Commun des Animaleries de Rockefeller (SCAR) - Université Claude Bernard lyon1, Structure Fédérative de Recherche (SFR) Santé Lyon Est, Lyon, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Margot Lepage
1Immunology Laboratory, Edouard Herriot Hospital – Hospices Civils de Lyon, 69437 Lyon Cedex 3, France
2CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Louise Courcol
2CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Remy Coudereau
1Immunology Laboratory, Edouard Herriot Hospital – Hospices Civils de Lyon, 69437 Lyon Cedex 3, France
4EA 7426 “Pathophysiology of Injury-Induced Immunosuppression” (Université Claude Bernard Lyon 1, Hospices Civils de Lyon, bioMérieux), 69437 Lyon Cedex 3, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Helena Paidassai
2CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Laurent Jallades
2CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
5Hospices Civils de Lyon, Lyon Sud University Hospital, Hematology Laboratory – Pierre-Bénite France.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jonathan Lopez
6Biochemistry and Molecular Biology department, Lyon Est Faculty of Medicine, Université Claude Bernard Lyon 1, Université de Lyon, Lyon Sud University Hospital, Hospices Civils de Lyon, Lyon, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Khalil Kandara
1Immunology Laboratory, Edouard Herriot Hospital – Hospices Civils de Lyon, 69437 Lyon Cedex 3, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Marine Ortillon
1Immunology Laboratory, Edouard Herriot Hospital – Hospices Civils de Lyon, 69437 Lyon Cedex 3, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Marine Mommert
4EA 7426 “Pathophysiology of Injury-Induced Immunosuppression” (Université Claude Bernard Lyon 1, Hospices Civils de Lyon, bioMérieux), 69437 Lyon Cedex 3, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Astrid Fabri
1Immunology Laboratory, Edouard Herriot Hospital – Hospices Civils de Lyon, 69437 Lyon Cedex 3, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Estelle Peronnet
4EA 7426 “Pathophysiology of Injury-Induced Immunosuppression” (Université Claude Bernard Lyon 1, Hospices Civils de Lyon, bioMérieux), 69437 Lyon Cedex 3, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Clémence Grosjean
4EA 7426 “Pathophysiology of Injury-Induced Immunosuppression” (Université Claude Bernard Lyon 1, Hospices Civils de Lyon, bioMérieux), 69437 Lyon Cedex 3, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Marielle Buisson
7Centre d’Investigation Clinique de Lyon (CIC 1407 Inserm), Hospices Civils de Lyon, F-69677, Lyon, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Anne-Claire Lukaszewicz
4EA 7426 “Pathophysiology of Injury-Induced Immunosuppression” (Université Claude Bernard Lyon 1, Hospices Civils de Lyon, bioMérieux), 69437 Lyon Cedex 3, France
8Anesthesia and Critical Care Medicine Department, Hospices Civils de Lyon, Edouard Herriot Hospital, 69437 Lyon Cedex 3, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Thomas Rimmelé
4EA 7426 “Pathophysiology of Injury-Induced Immunosuppression” (Université Claude Bernard Lyon 1, Hospices Civils de Lyon, bioMérieux), 69437 Lyon Cedex 3, France
8Anesthesia and Critical Care Medicine Department, Hospices Civils de Lyon, Edouard Herriot Hospital, 69437 Lyon Cedex 3, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Laurent Argaud
9Medical Intensive Care Department, Hospices Civils de Lyon, Edouard Herriot Hospital, 69437 Lyon Cedex 3, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Martin Cour
9Medical Intensive Care Department, Hospices Civils de Lyon, Edouard Herriot Hospital, 69437 Lyon Cedex 3, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Bénédicte F Py
2CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Olivier Thaunat
2CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Thierry Defrance
2CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Guillaume Monneret
1Immunology Laboratory, Edouard Herriot Hospital – Hospices Civils de Lyon, 69437 Lyon Cedex 3, France
4EA 7426 “Pathophysiology of Injury-Induced Immunosuppression” (Université Claude Bernard Lyon 1, Hospices Civils de Lyon, bioMérieux), 69437 Lyon Cedex 3, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Fabienne Venet
1Immunology Laboratory, Edouard Herriot Hospital – Hospices Civils de Lyon, 69437 Lyon Cedex 3, France
2CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Fabienne Venet
  • For correspondence: fabienne.venet{at}chu-lyon.fr
  • Abstract
  • Full Text
  • Info/History
  • Metrics
  • Supplementary material
  • Preview PDF
Loading

Abstract

A better understanding of sepsis-induced immunosuppression pathophysiology is desirable for the development of novel therapeutic strategies to prevent and reduce the rates of secondary infections and their associated mortality. Here we demonstrate that PD-L1+CD44+B220LowCD138+IgM+ regulatory plasma cells (PCs) are induced in a murine model of sepsis-induced immune alterations and in critically ill patients with bacterial sepsis and COVID-19. This was revealed both by detailed analysis of their phenotypical features and gene expression profile and by functional explorations comparing capacity of purified B cells and PCs to suppress T cell proliferation and IFNɣ secretion ex vivo. Sepsis-induced regulatory PCs exerted their suppressive function on T cells through IL-10 production and increased PD-L1 expression independently of regulatory T cells. Our findings thus reveal a novel pathophysiological mechanism of sepsis-induced immunosuppression that involves regulatory PCs. As such, these PCs constitute valid therapeutic targets to improve immune cell functions impaired by sepsis.

INTRODUCTION

Sepsis, defined as a life-threatening organ dysfunction caused by a dysregulated host response to infection 1, is a highly frequent syndrome affecting around 50 million people worldwide leading to 11 million deaths annually 2. Recently, the severe cases of SARS-CoV-2 infection pandemic, which corresponds to a viral sepsis, has led to a major influx of patients to the ICU and will most likely increase these numbers. Sepsis thus represents a major healthcare priority worldwide as recently acknowledged by the WHO 3.

Sepsis initiates a complex immune response that varies over time, with the concomitant occurrence of both pro- and anti-inflammatory mechanisms. As a result, some patients may rapidly develop signs of profound immunosuppression with altered innate and adaptive immune responses associated with frequent viral reactivations, increased susceptibility to secondary infections (mostly opportunistic) and high mortality 4. Thus immune-adjuvant treatments targeting sepsis-induced immunosuppression are proposed in the most immunosuppressed patients as a novel therapeutic approach in sepsis 5,6.

Mechanisms leading to sepsis-induced immune alterations are still being explored. Among others, the expansion of immune cell subpopulations with immunoregulatory functions has been described 4. For example, the increased proportion of CD4+CD25+CD127lowFoxp3+ regulatory T cells has been well documented both in murine models of sepsis and in patients 7. Similarly, within the myeloid lineage, the induction of monocytic and granulocytic myeloid-derived suppressor cells (MDSCs) has been repeatedly observed 8.

Cells from the B cell lineage are also affected by sepsis. In particular, decreased circulating B cell numbers and altered effector functions have been described in patients and septic mice 4,9,10. In murine models of auto-immune diseases, cancer and infections, B cells with regulatory functions (termed Breg) have been described to emerge at different developmental stages from immature B cells to plasma cells (PCs) 11–13. Only few studies evaluated the induction of Bregs in murine models of sepsis and their reported phenotype and mechanisms of action appeared quite heterogeneous 14,15. Data are even scarcer in the human clinical setting, in particular for severe COVID-19, a condition in which no functional study has evaluated the induction of Bregs so far.

In the current study, we investigated whether B cells with regulatory functions may be induced in a murine model of sepsis-induced immune alterations and in 3 cohorts of patients diagnosed with either bacterial sepsis or a severe COVID-19. In addition, we explored the mechanisms whereby these regulatory B cell subsets may participate in sepsis-induced T lymphocyte alterations.

RESULTS

Murine model of resuscitated cecal ligation and puncture recapitulates features of sepsis-induced T lymphocyte dysfunctions

We developed a clinically relevant murine model of sepsis-induced immune alterations by submitting mice to a mild cecal ligation and puncture (CLP). Control group of Sham-treated animals were submitted to laparotomy without ligation nor puncture. Both groups received antibiotics, analgesics and a lactate Ringer solution.

Surgery induced weight loss in both groups, which was significantly more profound and persistent in CLP mice compared with Sham animals (Fig. 1A). CLP was associated with a significant increase over time of severity score whereas it remained low in Sham animals (Fig. 1B). At 48h after surgery, no mortality was observed in the group of Sham animals and mortality was low (< 10%) in CLP mice (Fig. 1C).

Figure 1.
  • Download figure
  • Open in new tab
Figure 1. Clinical and immunological features of the reanimated cecal ligation and puncture model.

(A) Percentage of weight change at day 1 and day 2 post-surgery compared to weight prior to surgery. (B) Clinical severity score measured every hour during 2 days post-surgery. (C) Kaplan-Meyer survival curves. Significance in survival analysis was determined by log-rank test (A-B-C: n = 72 Sham vs 109 CLP). (D-G) Absolute counts and percentages of splenocytes, CD4+ T, CD8+ T, CD4+CD25+CD127low regulatory T cell (n = 23 Sham and 25 CLP mice). (H-I) Splenocytes from Sham (n = 14) and CLP (n = 12) mice were cultured with anti-CD3/CD28 antibodies-coated beads. (H) T cell proliferation was measured among viable cells by flow cytometry. (I) IFNγ was measured in supernatant of splenocytes cultures from Sham (n = 5) and CLP (n = 5) mice. (J) Splenocytes from ovalbumin-immunized Sham (n = 9) and CLP (n = 9) mice were cultured with ovalbumin peptide. IFNγ was measured in supernatant. Red squares represent results in CLP mice and white circles in Sham animals. Geometric means with standard deviations (SD) and individual values are shown. Mann-Whitney tests were performed and only significant p values < 0.05 are shown.

After 48h, CLP was associated with a significant decrease in the number of total splenocytes (Fig 1D). Absolute counts and percentages of dendritic cells and NK cells but not of other myeloid cells subpopulations were significantly decreased after CLP (Fig S1A to E). Absolute counts (Fig 1E-F) but not percentages (Fig. S2A) of CD4+ and CD8+ T lymphocytes were simultaneously decreased compared with Sham animals. Among CD4+ T cells, absolute counts of naïve and effector T cells tended to decrease after CLP while percentages of central memory cells increased in CLP mice (Fig. S2B-C-D). Among CD8+ T cells, absolute count of effector T cells significantly decreased after CLP (Fig. S2G). Finally, the percentage of CD4+CD25+CD127low regulatory T cells (Treg) significantly increased while their absolute count was maintained in CLP mice (Fig. 1G).

CLP also induced functional alterations in remaining T lymphocytes as illustrated by the reduced proliferative response of total T cells and of both CD4+ and CD8+ T lymphocytes and reduced IFNɣ secretion after TCR stimulation in CLP mice as compared with Sham animals (Fig. 1H-I, Fig. S3A-B). Such altered functionality was also evidenced after non-specific mitogen stimulation (Fig. S3C) and after ex vivo antigen-specific re-stimulation of Ag-experienced T cells (Fig 1J).

In total, we designed a murine model of sepsis, which recapitulated T cell immune dysfunctions observed in patients 4.

Regulatory B lymphocytes are not induced in mice after CLP

As a putative mechanism of sepsis-induced immunosuppression, we next evaluated whether regulatory B cells were induced in the spleen after CLP and participated in sepsis-induced T cell dysfunctions.

Although a mild but significant decrease in B220+ B splenocyte counts was observed after CLP compared with Sham animals (Fig. 2A), the percentages of total B splenocytes and of CD1dHighCD5+ regulatory B10 cells 16,17 were not significantly modified (Fig. 2B-C). Expressions of CD40, CD86, PD-1, PD-L1, TIM-3 and LAG-3 were not changed on B220+ lymphocytes after sepsis (Fig. 2D-E). Only major histocompatibility complex class II (MHC class II) expression was significantly decreased on B splenocytes after CLP (Fig. 2D) in accordance with observations made in septic patients 9.

Figure 2.
  • Download figure
  • Open in new tab
Figure 2. Regulatory B lymphocytes are not induced in mice after CLP.

(A-B) B splenocytes counts and percentages in 19 Sham and 24 CLP mice. (C) Proportion of CD1dhighCD5+ B splenocytes in 5 Sham and 6 CLP mice. (D) CD40, CD86 and MHC class II expressions as median fluorescence intensity (MFI) on splenic B cells from 6 Sham and 10 CLP mice. (E) PD-1, PD-L1, TIM3 and LAG-3 expressions as median fluorescence intensity (MFI) on splenic B cells (n = 5 Sham and 7 CLP mice for all markers except for LAG-3 where n = 11 sham and 9 CLP). (F) Volcano plot showing fold changes of 540 mRNA expressions in splenic B cells from 6 Sham and 6 CLP mice. Significantly upregulated mRNA are shown in red and downregulated ones in blue. (G) Splenic T cells from Sham mice (n = 8) were co-cultured with or without Sham (n = 16) or CLP (n = 16) purified splenic B cells at 1:1 ratio for 72 h, in the presence or absence of anti-CD3/CD28 antibodies-coated beads. T cell proliferation was measured among viable cells by flow cytometry. Red squares represent results in CLP mice and white circles in Sham animals. (H) IFNγ was measured in culture supernatants of Sham T cells cultured either alone (n = 5) or with B cells from Sham (n = 7) or CLP (n = 12) mice. Geometric means with standard deviations (SD) and individual values are shown. Mann-Whitney tests were performed and only significant p values < 0.05 are shown.

Transcriptomic analysis of purified B220+ B splenocytes showed that, among the 540 mRNA detected in B cells, only 6 (< 2%) were significantly differentially expressed between B220+ B splenocytes from Sham and CLP animals (Fig. 2F).

Finally, co-culture experiments between T lymphocytes purified from Sham animals and B220+ B splenocytes purified either from Sham or CLP mice showed that B cells purified after sepsis did not significantly modulate T cell proliferative response or IFNɣ release after TCR stimulation (Fig. 2G-H).

Overall, these results argue against the induction of regulatory B cells in spleen in the current model of sepsis-induced immune alterations.

Regulatory CD138+IgM+ plasma cells are induced in spleen after CLP

In accordance with observations made in septic patients 9, our results revealed the induction of a population of B220lowCD138+ splenocytes with plasma cell (PCs) morphology after CLP (Fig. 3A-B). We confirmed that the percentages and absolute counts of these cells significantly increased in spleen after CLP as compared with Sham animals (Fig. 3C-D). We showed that these cells expressed high levels of Prdm1 (coding for Blimp1) and Irf4 mRNAs but low levels of pax5 and bcl6 mRNAs compared with B cells after CLP (Fig. 3E). In addition, intracellular BLIMP1 expression was significantly higher in B220lowCD138+TACI+ splenocytes compared with B cells from same CLP animals (Fig. 3F).

Figure 3.
  • Download figure
  • Open in new tab
Figure 3. Regulatory CD138+IgM+ plasma cells are induced in spleen after CLP.

(A) Representative examples of flow cytometry gating of plasma cells (PCs) in one Sham and one CLP animal. (B) Morphology of sorted CD138+ B220low splenic cells from a CLP mouse. (C-D) Proportions and absolute counts of PCs among splenocytes in 19 Sham and 24 CLP mice. (E) mRNA expressions of transcription factors bcl6, pax5, irf4 and prdm1 (coding for Blimp-1) measured in B cells and PCs purified from 6 CLP mice. (F) Blimp-1 expression as median fluorescence intensity (MFI) on splenic B cells and PCs in 7 CLP mice. (G) Percentages of cell surface IgM expressing PCs (among the entire PC population) in 8 Sham and 9 CLP mice. Representative examples of IgM intracellular expression profile in B220+ splenocytes from 2 CLP mice (H) Expressions on PCs in Sham and CLP mice of CD138 (n = 10 and 13), B220 (n = 10 and 13), CD86 (n = 6 and 10) and MHC II (n = 6 and 10) as median fluorescence intensities. (I) Heatmap of mRNA expressions of selected transcripts related to cell differentiation, BCR signaling or migration measured in PCs purified from 6 Sham and 6 CLP mice. The red and blue colors indicate over-and under-expressed markers respectively and the color intensity specifies the expression level. (J) Splenic T cells from Sham mice (n = 8) were co-cultured with Sham (n = 8) or CLP (n = 17) splenic PCs at 1:1 ratio for 72 h, with or without anti-CD3/CD28 antibodies-coated beads. T cell proliferation was measured among viable cells by flow cytometry. (K) IFNγ was measured in culture supernatants of Sham T cells cultured either alone (n = 5) or with PCs from Sham (n = 6) or CLP (n = 10) mice. Red squares represent results in CLP mice (either B cells or PCs) and white circles in Sham animals. Geometric means with standard deviations (SD) and individual values are shown. Mann-Whitney tests were performed and only significant p values < 0.05 are shown.

The majority of these sepsis-induced PCs expressed membrane and intracellular IgM (Fig. 3G) and were characterized by higher density of expression of B220 and MHC class II and reduced expression of CD138 as compared to Sham animals (Fig. 3H).

Transcriptomic analysis of PCs purified from CLP mice illustrated differences in gene expressions related to differentiation profile (i.e. decreased Cd28, Ms4a1 coding for CD20, Slamf7 Irf4, Prdm1 coding for Blimp-1 but increased Xbp1 mRNA levels) and BCR signaling (i.e. decreased Cd19 and Cd22 mRNA levels, compared with PCs from Sham animals, Fig. 3I). Interestingly, Ccr7, Cxcr5 and Cxcr4 mRNA levels were decreased in CD138+ PCs purified from CLP mice suggesting that these cells may possess decreased homing predispositions (Fig. 3I).

Finally, co-culture experiments showed that proliferation (Fig. 3J) and IFNɣ secretion (Fig 3K) induced by TCR stimulation of CD3+ T splenocytes purified from Sham animals were significantly decreased in the presence of CD138+ PCs purified from CLP mice but not of PCs purified from Sham animals.

These results demonstrate that B220LowCD138+IgM+HLA-DR+ PCs are induced in spleen after CLP and present with regulatory functions on T cell proliferation ex vivo.

In vivo depletion of CD138+ plasma cells improves T cell proliferation after CLP

Next, we aimed to confirm in vivo that PCs induced after CLP participated in altered T cell proliferation commonly observed after sepsis.

Thus we depleted CD138+ PCs in vivo through bortezomib administration after CLP (BZB, 0.5 mg/kg I.V.). BZB was administered 24h after CLP in order to preferentially deplete PCs induced after sepsis versus resident PCs in spleen. Injection of dimethylsufoxyde (DMSO, i.e. BZB vehicle) was used as control condition in both CLP and Sham animals.

We confirmed that BZB administration induced a modest but significant depletion of PCs in spleen after CLP (Fig. 4A-B) while the number of B220+ B splenocytes remained unmodified (Fig. 4C). BZB treatment was associated with restauration of the proliferative response of splenic T cells recovered from CLP mice as compared with that of splenic T cells originating from septic mice treated with DMSO (Fig. 4D). Finally, BZB administration led to a significant reduction in weight loss after 48h in CLP animals compared with untreated septic animals (Fig. 4E).

Figure 4.
  • Download figure
  • Open in new tab
Figure 4. In vivo depletion of CD138+ plasma cells improves T cell proliferation after CLP.

Animals were either treated with Bortezomib (n = 7 Sham and 8 CLP mice) or DMSO (control condition, n = 6 Sham and 8 CLP mice) 24h after surgery. (A) Proportions of plasma cells (PCs) among B cells and (B-C) absolute counts of PCs and B cells in spleen. (D) Splenic T cells from Sham and CLP mice were cultured with anti-CD3/CD28 antibodies-coated beads. T cell proliferation was measured among viable cells by flow cytometry. (E) Percentage of weight change at day 1 and day 2 post-surgery compared to weight prior to surgery. Red squares represent results in CLP mice and white circles in Sham animals. Geometric means with standard deviations (SD) and individual values are shown. Mann-Whitney tests were performed and only significant p values < 0.05 are shown.

Thus these results are in line with our hypothesis that CD138+ PCs induced after CLP participate in sepsis-induced impairment of T lymphocyte proliferative potential.

CD138+ plasma cells induced after CLP regulate T cell proliferation through cell-cell contact and IL-10 production

Next, we investigated putative pathways involved in regulatory mechanism of CD138+ PCs induced after sepsis.

Transwell experiments were performed in order to evaluate whether the suppressive effect exerted by CLP-induced PCs on T cell response relied upon direct cell-cell interactions. We observed that the absence of cell-cell contact between PCs and responder T cells was associated with a significantly increased T cell proliferation as compared with co-culturing both cell populations in the same well (Fig. 5A). However, in Transwell condition, T cell proliferation did not strictly return to control values thus suggesting that part of the immunoregulatory function of CLP-induced PCs could be mediated by the release of soluble factors.

Figure 5.
  • Download figure
  • Open in new tab
Figure 5. CD138+ plasma cells induced after CLP regulate T cell proliferation through cell-cell contact and IL-10 production.

(A) Splenic T cells from Sham mice (n = 4) were co-cultured with plasma cells (PCs) purified from CLP mice (n = 8) with or without HTS Transwell® inserts (T cells in the bottom of the well and PCs on the top of the membrane) at a 1:1 ratio for 72 h, and stimulated by anti-CD3/CD28 antibodies-coated beads. T cell proliferation was measured among viable cells by flow cytometry. Non-parametric Wilcoxon paired test was used to compare proliferation in presence of PCs with or without the inserts. (B) CD44 expression as median fluorescence intensity (MFI) on PCs measured in 15 Sham and 20 CLP mice. (C-D) Percentages among B cells and absolute counts of PCs in 4 CLP and 4 Sham Il10-eGFP-reporter VertX mice. (E) Representative examples of IL-10 expression profiles in B cells and PCs (red dots) in Sham and CLP VertX mice. (F) IL-10 expression as median fluorescence intensity (MFI) in B cells and PCs from 4 Sham and 4 CLP VertX mice. (G) IL-10 production measured in culture supernatants of: unstimulated Sham T cells (n = 4), Sham T cells stimulated by anti-CD3/CD28 antibodies-coated beads and co-cultured with or without CLP PCs (n= 4) or in supernatants of PCs purified from Sham (n = 4) or CLP mice (n = 3). Red squares represent results in CLP mice and white circles in Sham animals. Geometric means with standard deviations (SD) and individual values are shown. Mann-Whitney tests were performed and only significant p values < 0.05 are shown.

As the central role of IL-10 in Breg and PCreg functions has been well described 18, we monitored CD44 expression on PCs after CLP, which was documented as a marker of IL-10-producing B cell subpopulations 19. CD44 expression was significantly increased on CD138+ PCs after CLP (Fig. 5B) as observed by Matsumoto et al. in a model of experimental autoimmune encephalomyelitis 19. We also evaluated IL-10 production in PCs induced after CLP in a model with Il10-eGFP-reporter mice (VertX mice) 20. We first confirmed that CD138+ PCs absolute numbers and percentages were increased in VertX mice after CLP as they were in wild type animals (Fig. 5C-D). We next showed that IL-10-GFP expression was higher in PCs as compared with B cells both in Sham and CLP animals (Fig. 5E-F). In addition, IL-10 expression was significantly induced in CD138+ PCs after CLP compared with PCs from Sham animals (Fig. 5F). In accordance, IL-10 concentrations measured in the supernatants of CD138+ PCs purified from CLP mice were higher than in the supernatants of PCs purified from Sham animals irrespective of the presence of T cells in the culture (Fig. 5G).

In total, these results show that IL-10-producing CD44+B220LowCD138+IgM+ PCs are induced in spleen after CLP. Altogether, our data suggest that the immunoregulatory function of CLP-induced PCs is exerted both via cell/cell contacts and IL-10 release.

Increased PD-L1 expression on CLP-induced CD138+ plasma cells participates in their immunoregulatory functions

Next, we evaluated putative cell surface receptors involved in cell-cell contact regulatory mechanism of CD138+ PCs induced after sepsis.

Transcriptomic analysis of purified PCs showed that CLP induced a major transcriptional reprogramming of these cells with the differential regulation of 554 mRNAs compared with Sham animals (Fig. 6A). Seventeen pathways were significantly dysregulated in PCs after CLP (Fig. 6B). The majority of these pathways were downregulated (n = 16, 94%) after CLP such as the B cell receptor signaling or NF-kB signaling pathways.

Figure 6.
  • Download figure
  • Open in new tab
Figure 6. CLP-induced PD-L1 expression contributes to plasma cells regulatory function.

(A) Heatmap of 554 mRNAs expressed in plasma cells (PCs) purified from spleens of 6 Sham and 6 CLP mice. The red and blue colors indicate over- and under-expressed markers respectively and the color intensity specifies the expression level. Hierarchical clustering was performed using Euclidean distance method. (B) Ingenuity Pathway Analysis of pathways significantly differentially regulated in PCs from CLP (n = 6) compared with Sham mice (n = 6). Upregulated and downregulated pathways are shown in red and blue respectively. (C) PD-1 and PD-L1 expressions as median fluorescence intensity (MFI) on PCs measured in 12 Sham and 17 CLP mice. (D) Percentages of PD-1 and PD-L1 expressing PCs measured in 12 Sham and 17 CLP mice. (E-F) LAG-3 expression as MFI on PCs and percentages of PCs expressing LAG-3 among total PCs measured in 11 Sham and 9 CLP mice. (G) Splenic T cells from Sham mice (n = 4) were co-cultured with or without PCs purified from CLP animals (n = 10) at 1:1 ratio for 72 h with anti-PD-L1 blocking antibodies (10 µg/mL) or corresponding isotypic IgG (10 µg/mL), and stimulated or not by anti-CD3/CD28 antibodies-coated beads. Non parametric Wilcoxon paired test was used to compare results obtained in these last 2 conditions. T cell proliferation was measured among viable cells by flow cytometry. Red squares represent results in CLP mice and white circles in Sham animals. Geometric means with standard deviations (SD) and individual values are shown. Unless otherwise stated, Mann-Whitney tests were performed and only significant p values < 0.05 are shown.

Most importantly, only one pathway was up-regulated in CLP-induced CD138+ PCs: the PD-1 / PD-L1 immunoregulatory pathway (Fig. 6B). This change primarily corresponded to the upregulation of the PD-1 Ligand, as illustrated by the increased expression of PD-L1 on PCs from CLP animals shown by flow cytometry analysis (Fig. 6C). After CLP >90 % of plasma cells were positive for PD-L1 expression (Fig. 6D). Conversely, LAG3 expression, another immune checkpoint receptor expressed on PCs with regulatory functions 11, was not modified on CLP-induced PCs compared with Sham animals (Fig. 6E-F).

The functional consequences of upregulated PD-L1 expression on CLP-induced PCs were explored ex vivo by co-culturing splenic T lymphocytes with CLP-induced PCs, in the presence or absence of an anti-PD-L1 blocking antibody. As illustrated by Fig. 6G, addition of the anti-PD-L1 blocking antibody induced a significant recovery of T cell proliferation compared with addition of the corresponding isotypic IgG, thus suggesting that the PD-L1/PD-1 axis contributes to the immunosuppressive function of CLP-induced PCs.

Altogether, our results show that PD-L1+CD44+B220LowCD138+IgM+ PCs induced in spleen after CLP present with inhibitory functions on T cell proliferation involving both IL-10 production and upregulated expression of the checkpoint inhibitory ligand PD-L1.

PD-L1+CD138+ plasma cells are induced in critically ill patients with bacterial sepsis and severe COVID-19 in association with increased mortality and regulate T cell proliferation ex vivo

Last, we evaluated the induction of PCs in 3 cohorts of ICU patients: one retrospective cohort of patients with bacterial sepsis (REALISM cohort) 21,22 and two prospective cohorts of patients diagnosed respectively with bacterial sepsis (IMMUNOSEPSIS cohort) 23 and severe COVID-19 (RICO cohort) 24.

The induction of PCs in the blood of ICU patients with bacterial sepsis was first retrospectively evaluated by re-analyzing data from the REALISM study 21. Demographic and clinical data for this cohort are presented in Table S1. As CD138 and CD38 markers were not available in this cohort, flow cytometry stainings from the sub-cohort of septic patients and healthy donors were rea-analyzed to determine percentages and absolute counts of SSCIntCD19low lymphocytes, previously demonstrated as a surrogate phenotype for circulating PCs 9. The percentages of PCs were significantly increased in septic patients compared with healthy donors starting from D1 after ICU admission (Fig. 7A). Maximum value was observed one week (D5-D7) after ICU admission. PCs frequencies then progressively decreased over time until D60 without strictly returning to normal values. In this cohort, when comparing survivors and non-survivors at D14, we observed that the median PC frequency measured at D5 after ICU admission was significantly higher in non-survivors as compared with survivors (Fig. 7B).

Figure 7.
  • Download figure
  • Open in new tab
Figure 7. PD-L1+ regulatory plasma cells are induced in critically ill patients with bacterial sepsis and COVID-19.

(A) The percentages of SSCInt CD19low plasma cells (PCs) were retrospectively studied in septic patients (n = 107) and healthy volunteers (HV, n= 175) from REALISM cohort. Patients were sampled at D1 (n = 92), D3 (n = 87), D5 (n = 92), D14 (n = 61), D28 (n = 46) and D60 (n = 40) after intensive care unit admission. (B) Percentages of PCs were compared between day-14 survivors and non-survivors at D1 (n = 79 and 13 respectively), D3 (n = 78 and 9 respectively) and D5 (n = 87 and 5 respectively). (C) Percentages of CD38+CD138+ PCs were prospectively studied in septic patients from IMMUNOSEPSIS cohort (n = 67) and in critically ill COVID-19 patients (n = 33) from RICO cohort of COVID-19 critically ill patients compared with HVs (n = 33). In both cohorts, patients were sampled at D1 (n = 41 and 31 respectively), D3 (n = 61 and 20 respectively) and D5 (n = 40 and 11 respectively) after ICU admission. (D) PD-L1 expression as median fluorescence intensity (MFI) and percentage of PD-L1 expressing cells were measured on circulating B cells and PCs in 15 HVs and 17 septic patients at D3 (IMMUNOSEPSIS 4 cohort). (E) PD-L1 expression as MFI and percentage of PD-L1 expressing cells were measured on circulating B cells and PCs from 2 HVs and 21 critically ill COVID-19 patients (RICO cohort). (F) Representative CD138/IgM histograms gated on CD19+SSClow lymphocytes in 2 different septic patients are shown. IgM intracellular staining was detected in > 95% of circulating PCs (red dots). (G) Blimp-1 expression as MFI in circulating B cells and PCs from 8 septic patients. (H) Circulating T cells from HVs (n = 8; white squares) were cultured with or without purified PCs (n = 8; squares) or B cells (n = 6, dots) from septic patients (n = 7) or COVID-19 patient (n = 1) at 1:1 ratio, and stimulated by anti-CD2/CD3/CD28 antibodies-coated beads. After 72h, T cell proliferative response was evaluated by flow cytometry. (I) IFNγ was measured in culture supernatants of T cells from healthy volunteers cultured either alone (n = 11) or co-cultured with circulating B cells (n = 5) or PCs (n = 11) from septic patients. Results are shown as box plots and individual values or outliers. Mann-Whitney tests were performed to compare results obtained in patients versus HVs. Wilcoxon paired tests were performed to compare results of T cell proliferation and IFNγ concentration between culture conditions. Only significant p values < 0.05 are shown.

These results were then validated prospectively in observational studies including septic shock patients and critically ill patients with severe COVID-19 for which PCs were defined as CD138+CD38+CD19low lymphocytes. In addition, PD-L1 expression was monitored on circulating PCs. Demographic and clinical data for this cohort are presented in Table S1. In patients with bacterial sepsis, we confirmed the significant increase in the percentage of CD138+CD38+ PCs compared with HV with a peak value at the end of the first week (Fig. 7C). A similar increase was observed in COVID-19 ICU patients. However, maximum value was detected earlier (i.e. upon ICU admission, Fig. 7C). In both patients with bacterial sepsis and COVID-19, PCs presented with high PD-L1 expression (Fig. 7D-E), expressed intracellular IgM (Fig. 7F) and presented with elevated intracellular BLIMP1 expression (Fig. 7G). Thus, as observed in mice after CLP, PD-L1+ PCs are induced in ICU patients after bacterial sepsis and COVID-19.

Finally, the immunoregulatory function of circulating PCs was assessed ex vivo for 8 critically ill infected patients (7 ICU patients with bacterial sepsis and 1 COVID-19). We observed that addition of CD138+ PCs purified from septic patients significantly reduced the proliferation of total T cells, of both CD4+ and CD8+ T cells and IFNɣ secretion induced by TCR proliferation of responder T cells purified from healthy donors (Fig. 7H-I, Fig. S4A-B). By contrast, in 6 patients, addition of B lymphocytes purified from septic patients had no effect on T cell proliferative response and IFNɣ secretion in agreement with results observed in CLP mice (Fig. 7H-I).

Altogether, our data indicate that the induction of PCs with high PD-L1 expression and regulatory function on T cell proliferation was recapitulated in ICU patients with bacterial and viral sepsis (COVID-19) in association with increased mortality.

CD138+ plasma cells induction after sepsis correlates with that of regulatory T cells but their immunoregulatory functions may be independent

Last, we asked whether immunoregulatory PCs induced after sepsis may require Treg to inhibit T cell proliferation.

We observed that the absolute numbers of Treg and PCs were significantly correlated after sepsis both in patients (Fig. 8A) and in CLP mice (Figure 8B). This suggests that common induction mechanisms may lead to the differentiation / activation of both immunoregulatory cell types.

Figure 8.
  • Download figure
  • Open in new tab
Figure 8. CD138+ plasma cells and regulatory T cells after sepsis in mice and patients

(A) Correlation between absolute counts of regulatory T cells and plasma cells (PCs) in blood of patients at D5 after sepsis (n = 91). (B) Correlation between absolute counts of regulatory T cells and PCs in spleen of CLP mice (n = 32). Spearman correlation test was applied. R correlation coefficient and p values are shown. (C) Percentages of regulatory T cells among CD4+ splenocytes were measured in Sham (n = 7 and 6 respectively) and CLP (n = 7) mice treated with Bortezomib (BZB) or its control (DMSO). Red squares represent results in CLP mice and white circles in Sham animals. Geometric means with standard deviations (SD) and individual values are shown. Mann-Whitney tests were performed to compare proportions of regulatory T cells in mice. (D, E, F) Circulating T cells from healthy volunteers (n = 6), depleted or not for regulatory T cells, were co-cultured with circulating PCs purified from septic patients (n = 6) at 1:1 ratio for 72 h and stimulated by anti-CD2/CD3/CD28 antibodies-coated beads. Proliferation of CD3+ (D), CD4+ (E) and CD8+ (F) T cells was measured among viable cells by flow cytometry. Wilcoxon paired tests were used to compare results of T cell proliferation between culture conditions. Only significant p values < 0.05 are shown.

However, in BZB-treated mice, sepsis-induced PCs depletion and improvement in splenocytes proliferation was not associated with reduced Treg expansion (Fig. 8C).

Finally, ex vivo co-culture experiments showed that CD138+ PCs purified from septic patients were equally efficacious in decreasing T cell proliferation in the presence or not of Treg in the co-culture (Figure 8D-E-F).

In total, while these data suggest a common induction mechanism for Treg and PCs after sepsis, these immunoregulatory lymphocyte subsets may represent independent regulatory mechanisms to control T cell proliferation. Nevertheless, as we cannot definitively rule out a direct effect of BZB on Treg function, the confirmation of this last aspect would require a dedicated mouse model deficient for Treg.

DISCUSSION

A better understanding of the pathophysiology of sepsis-induced immunosuppression is desirable to lead to novel therapeutic strategies to prevent and reduce the rates of secondary infections and associated mortality. Expansion of cellular subsets with immunoregulatory functions has been proposed as one of the mechanisms involved in this process. This has been particularly well described for CD4+CD25+Foxp3+ regulatory T cells and more recently for myeloid-derived suppressor cells 4. However, contribution of the more recently described regulatory cells of the humoral arm of the adaptive immune system (B cells and PCs) remains elusive in this context.

“Regulatory B cells” is a generic term that encompasses all B cells that act to suppress immune responses. While the phenomenology of B cell-mediated suppression is well established; the identity of B cells mediating these activities in vivo remains incompletely defined 25. For example in murine models of sepsis and in septic patients, the increased frequency of CD1d+CD5+ Breg 14,15, CD19+CD24highCD38high Bregs (i. e. B cells with a transitional phenotype) 26 and of CD27+CD24high B10 cells 27,28 has been described. Thus, there was no consensus so far regarding the nature of Breg subtype involved in the immunosuppressive phase of sepsis. In the current study, we observed neither induction of CD1d+CD5+ Breg, nor any strong modification of the B cell transcriptional profile or phenotype, nor acquisition by B cells of a regulatory function towards T cells after sepsis. In addition, in septic patients, we did not observe any regulatory effect of peripheral B cells on T cell proliferation ex vivo. This argues against the induction of Breg in our model of sepsis-induced immunosuppression. Beside differences in models and cohorts, the discrepancies between our present results and the published literature could be explained by 1/ the possibly concomitant existence and/or induction of several different regulatory B cell subpopulations in sepsis; 2/ the absence of any consensual phenotype characterizing regulatory B cells subsets and 3/ the limits of studying regulatory B cells solely based on phenotypic characterization in the absence of any functional exploration.

More recently, Nascimento et al. showed the expansion of splenic CD39highCD138high PCs in CLP mice, which participated in immunosuppression through increased extracellular adenosine production and inhibition of macrophages anti-microbial functions 29. In their study, PCs expansion was established in a cohort of 21 septic patients. Here, we expand and complete these data by demonstrating that PD-L1+CD44+B220LowCD138+IgM+ regulatory PCs are induced in a murine model of sepsis-induced immune alterations and in critically ill septic patients. This was demonstrated not only by detailed analysis of their phenotypical features and gene expression profile but also by functional explorations comparing capacity of purified B cells and PCs to suppress T cell proliferation ex vivo. To our knowledge, this work is the first to strictly compare the regulatory potential of B cells and PCs in sepsis not only in mice but also in patients. In addition, our results extend the pathophysiologic role of PCreg to sepsis-induced immunosuppression as besides their previously described inhibitory role on innate immune response 29, the current study demonstrates that these regulatory cells also participate in sepsis-induced impairment of T cell functions.

Interestingly, we describe here that the regulatory function of sepsis-induced PCs is dual in that it involves both secretion of a suppressive cytokine (IL-10) and expression of a member of the checkpoint inhibitory ligands family (PD-L1). This observation is in accordance with studies placing CD138high PCs as the major source of B cell-derived IL-10 in autoimmune, infectious and malignant diseases 19,30–33. For example, Matsumoto et al described IL-10 producing CD138+CD44+HLA-DRhigh PCs that limit autoimmune inflammation in a murine model of multiple sclerosis 19. Thus, beside auto-immune diseases, the current results expand the immunoregulatory role of CD138+CD44+HLA-DRhigh PCs as IL-10 producers and regulator of T cell proliferation after sepsis as well. Our description of IL-10-producing PCs with a regulatory function is also coherent with the fact that IL-10 has been repeatedly described as a major mediator of sepsis-induced immunosuppression 34.

Besides IL-10, we demonstrated that the PD-1/PD-L1 axis is part of the PC regulatory armamentarium in sepsis inasmuch as PD-L1 expression is upregulated on sepsis-induced PCs and PD-L1 Abs partially alleviated PC inhibitory function ex vivo. LAG-3, another inhibitory checkpoint ligand was identified by Lino et al in a murine model of Salmonella typhimurium infection as a marker of a novel subset of regulatory PCs 11. In addition, the authors showed that these cells also expressed high levels of other immune checkpoints ligands such as PD-L1, PD-L2 and CD200. The authors suggested that such co-expression of several receptors implicated in the negative regulation of immunity may be important as these molecules may act synergistically. However, the contribution of these receptors to the immunosuppressive function of PCs was not evaluated. Our current study shows that the inhibitory function of sepsis-induced PCs towards T cells is not mediated through increased LAG-3 expression but involves activation of the PD-1 pathway. Such suppressive function of PD-L1/PD-L2 has also been documented for IgA+ PCs involved in intestinal immune homeostasis 32,35. Although Umakoshi et al. reported high levels of IL-10 and PD-L1 mRNA in CD1d+CD5+ Bregs from CLP mice 14, contribution of the PD-1/PD-L1 axis to the regulatory function of these cells was not addressed. However, treatments targeting the PD-1/PD-L1 axis are currently considered in septic patients 36, which might also help to lift part of the regulatory signals delivered by PCs in septic patients.

Finally, we studied the relationship between the two adaptive immunoregulatory lymphocyte subpopulations induced after sepsis, namely Treg and sepsis-induced immunoregulatory PCs. As observed in cancer patients 37,38, Treg and PCs were correlated and concomitantly induced both in patients and mice after sepsis suggesting a common induction mechanism. However, we suggest that these immunoregulatory lymphocyte subsets may constitute independent regulatory mechanisms to control T cell proliferation after sepsis. Indeed, we observed that Treg induction after CLP was maintained in BZB-treated mice. In addition, immunoregulatory function of PCs on T cell proliferation ex vivo was preserved in Treg-depleted samples. To note, in these last experiments, the improvement of T cell proliferation due to Treg depletion was close to statistical significance (p = 0.0625, non-parametric Wilcoxon paired test) as it was observed in all paired samples except for one. This is in agreement with previous data showing that different PC subpopulations produced IL-10 and IL-35 in a murine model of infection 32. In contrary, in some experimental settings of auto-immune diseases (uveitis and experimental autoimmune encephalomyelitis), regulatory B cell subpopulations were shown to induced Treg 32,39 via increased IL-35 production 40. Further work is now necessary to decipher the pathways induced after sepsis leading to induction/activation of these immunoregulatory cell subsets. Such study may have important impact in sepsis as results could help to identify treatable traits in patients impacting simultaneously several immunoregulatory mechanisms.

In line, our present study also brings two lines of evidence indicating that PCs are important contributors to sepsis-induced immunosuppression and therefore constitute valid therapeutic targets to improve immune cell functions impaired by sepsis. First, we showed that elevated PCs frequencies are associated with increased mortality in septic patients. Second, we found in the murine CLP model that in vivo PC depletion through bortezomib administration is associated with decreased features of sepsis-induced immune alterations and improvement of clinical endpoints. While further studies are now mandatory to confirm the clinical rationale for targeting PCs in sepsis, definition of biomarkers of PCs regulatory function will facilitate monitoring these cells in patients. These will be necessary not only to identify patients who could benefit from specific immunotherapies in an individualized targeted therapeutic approach but also to follow response to treatments.

Beyond bacterial sepsis, we describe here the induction of PCs with regulatory functions in clinical samples from ICU patients with severe COVID-19. Expansion of PCs has been repeatedly described in the most severely ill patients. Some studies even reported PC frequencies exceeding 30% of the total B cell pool 10,41–43. However, the putative regulatory function of these expanded PCs has never been evaluated so far. In addition, PCs expansion in COVID-19 was correlated with hallmarks of an extra-follicular B cells response activation 10. Extra-follicular B cell responses help to curb viral infection by allowing rapid production of IgM-expressing PCs 44. In accordance, PCs induced in our model presented with features of extrafollicular B cell response with high IgM expression and reduced Cxcr5 and Cxcr4 mRNA levels suggesting decreased follicular and bone-marrow homing predisposition. However, the link between extra-follicular B cell responses and the expansion of regulatory PCs in sepsis has yet to be determined.

The induction of PCs with regulatory functions in patients with both viral and bacterial sepsis suggests that common mechanisms may lead to their expansion in infected ICU patients. Ex vivo studies showed that several stimuli such as BCR signaling, CD40-CD40L interaction and TLR stimulation may be required to induce PCreg differentiation 45. In keeping with this, systemic inflammation and pro-inflammatory cytokines release induced by TLR ligands have been described as one mechanism of regulatory PCs expansion 46. Thus we postulate that systemic inflammation induced after bacterial and viral sepsis may participate in regulatory PCs differentiation. In agreement, we showed that PCs induction was transient after sepsis with a return to normal value one month after sepsis putatively in parallel with inflammation resolution in these patients 21. It is noteworthy that the kinetics of PCs induction in patients with bacterial and viral sepsis were different with a maximum value upon ICU admission in COVID-19 patients and 3 days after admission in patients with bacterial sepsis. This could be related to differential kinetics of inflammation, to higher viral/antigen burden or just be the reflection of different elapsed time after infection onset in these two clinical contexts (i.e. on average 10 days after date of first symptoms in COVID-19 patients and within 48h after diagnosis of infection in patients with bacterial sepsis). Dedicated research projects are now mandatory to fully explore mechanisms leading to PCs expansion and differentiation after viral and bacterial sepsis.

The current study presents with limitations. More specifically the link between extra-follicular B cell responses and the expansion of regulatory PCs in sepsis cannot be fully defined based on the current results. In addition, the independency between Treg and PCreg immunoregulatory functions must now be confirmed in dedicated Treg-deficient mouse models. Induction mechanisms leading to PCs expansion and differentiation after viral and bacterial sepsis should be explored. Finally biomarkers to specifically identify PCs with regulatory functions in clinic should be developed. These will be necessary to confirm the clinical rationale for targeting PCs in sepsis.

In conclusion, we have identified a novel pathophysiologic mechanism of sepsis-induced immunosuppression that involves regulatory PCs. We show that these cells are induced in a relevant murine model of sepsis and in clinical samples from patients and that they exert their suppressive function on T cells through IL-10 production and increased PD-L1 expression. These results open new fields of research not only regarding the pathophysiology of sepsis-induced immunosuppression but also the putative specific biomarkers of regulatory PCs and therapeutic potential of targeting these regulatory cells in patients.

METHODS

Experimental Design: mice and patients’ cohorts

Male C57BL/6J mice (Charles River, L’Arbresle, France) and VertX mice containing Il10-eGFP-reporter (The Jackson Laboratory, ME, USA), aged between 8 to 10 weeks and weighting from about 21 to 29 grams were used in this project. VertX mice presented with an internal ribosome entry site (IRES)-enhanced green fluorescent protein (eGFP) fusion protein placed downstream of exon 5 of the interleukin 10 (Il10) gene 20. Mice were housed one week before the experiments in a conventional animal facility (Service Commun des Animaleries de Rockefeller, Lyon, France and PBES specific pathogen-free rodent facility (ENS Lyon)). For each experiment, mice (total n = 266) from the same cage were randomly assigned to two groups: animals with polymicrobial sepsis induced by Cecal Ligation and Puncture (CLP, total n = 157) and control mice (Sham, total n = 109). They were kept together after surgery and until euthanasia. All the experiments were approved by the local animal ethical evaluation committee (#2016072616404065, and #45069-2023062011386592, CECCAPP, Lyon, France) in accordance with European Convention for the Protection of Vertebrate Animals used for Experimental and other Scientific Purposes.

Adult patients from intensive care units (ICU) of academic hospital (Hospices Civils de Lyon, Lyon, France) enrolled in three observational clinical cohorts were studied either prospectively (RICO = REA-IMMUNO-COVID and IMMUNOSEPSIS 4 cohorts) or retrospectively (REALISM = REAnimation Low Immune Status Markers cohort) 22. Samples in humans were collected under approved protocols registered under ClinicalTrials: NCT04392401 (RICO = REA-IMMUNO-COVID); NCT02638779 (REALISM cohort (REAnimation Low Immune Status Markers); NCT04067674 (IMMUNOSEPSIS 4).

RICO cohort included critically ill patients who presented with pulmonary infection with SARS-CoV-2 confirmed by RT-PCR testing. Exclusion criteria disqualified pregnant women and institutionalized patients. It was approved by ethics committee (“Comité de Protection des Personnes Ile de France 1”-N°IRB / IORG #: IORG0009918) under agreement number 2020-A01079-30. IMMUNOSEPSIS 4 included septic shock patients admitted into ICU identified according to the Third International Consensus Definitions for Sepsis and Septic shock (Sepsis-3)1: vasopressors administration started within the first 48 hours around inclusion, plasma lactate level above 2 mmol/L and diagnosed or suspected infection. The exclusion criteria were any condition modifying the immune status: immunosuppressive treatment (including > 10 mg equivalent prednisone per day or cumulative dose > 700mg), hematological disease treated within the 5 years, solid tumor treated with chemotherapy or in remission, number of circulating PNN < 500 / mm3, innate immune deficiency, extra-corporeal circulation with one month before inclusion (cardiac surgery or ECMO). The onset of septic shock was defined by the beginning of vasopressor therapy. It was approved by ethics committee (“Comité de Protection des Personnes Ouest II – Angers Sud-Est II”; RCB identification number: 2019-A000210-57, identification number SI/CPP: 19.01.23.71857), registered at the French Ministry of Research and Teaching (#DC-2008-509) and recorded at the Commission Nationale de l’Informatique et des Libertés. Since RICO and IMMUNOSEPSIS 4 studies were observational with low risk for the patients and no specific blood sampling procedure besides routine blood sampling required, there was no need for written informed consent, although oral information and non-opposition to inclusion in the study were mandatory and recorded in patients’ clinical files. Finally, REALISM included septic patients identified according to the Third International Consensus Definitions for Sepsis and Septic shock (Sepsis-3)1: vasopressors administration started within the first 48 hours after ICU admission, plasma lactate level above 2 mmol/L in case of septic shock, suspected infection for which microbiological sampling had been performed, along with the administration of antimicrobials. Exclusion criteria were aplasia or pre-existent immunosuppression, pregnancy, and individuals with no social security insurance, restricted liberty or under legal protection. It was approved by ethics committee (‘Comité de Protection des Personnes Sud-Est II’, Bron, France) and the French National Security agency for drugs and health-related products (Approval code: 69HCL15_0379, 30th November 2015).

In all three cohorts, EDTA and heparin anti-coagulated blood samples were collected in patients during routine procedure. First samples (i.e. D1) were obtained within the first 48h after inclusions in the studies (i.e. on average 10 days after date of first symptoms in COVID-19 patients and within 48h after diagnosis of infection for patients with bacterial sepsis). Clinical and biological parameters were collected, including demographic characteristics, date and cause of admission to ICU, vital status at day 28 after inclusion, type of infection, comorbidities (Charlson index) and severity scores (Simplified Acute Physiology Score II, Sepsis-related Organ Failure Assessment; McCabe classification). Concomitantly, EDTA and heparin anticoagulated blood samples were collected from healthy volunteers (HV) from the blood bank EFS of Lyon (Etablissement Français du Sang). According to EFS standardized procedures for blood donation, informed consent was obtained from healthy donors and personal data were anonymized at the time of blood donation and before the transfer of blood to our research lab. Every experiment was performed on fresh whole blood collected the same day. Samples were kept on ice during storage before analysis processing.

Murine model of sepsis-induced immune alterations

For each experiment, mice (total n = 266) were divided in two groups: animals with polymicrobial sepsis induced by Cecal Ligation and Puncture (CLP, total n = 157) as previously described 47 and control mice (Sham, total n = 109). Briefly, around 1 hour after a subcutaneous (SC) injection of 0.1 mg/kg buprenorphine (AXIENCE), mice were anaesthetized with isoflurane inhalation (induction 3%, maintenance 1.7-2%; FiO2 = 0.2). They were placed on a heating mat during the whole surgery procedure. Abdomen was shaved and disinfected. Local anesthesia was performed by SC injection of 100 µL of lidocaine chlorhydrate 1% (AGUETTANT). Paramedian laparotomy allowed caecum exteriorisation, which was then ligatured at its external third using 5.0 silk thread (Teleflex, DEKNATEL) and punctured twice (21-gauge needle) without crossing to the other side. A droplet of faeces was extruded from the puncture holes to ensure patency. Caecum was carefully replaced in the peritoneum to avoid any intestinal twist. Finally, incision was sutured in layers and animals were resuscitated with a SC injection of 1 ml of Ringer Lactate (VIAFLO). Mice were placed under heat lamp during recovery. Within the same conditions, Sham control mice underwent laparotomy with only exteriorisation of caecum from abdomen cavity, without ligation nor puncture. Mice were kept in the same cage after surgery, a heating mat placed under half of the cage and food softened with water. Six hours following surgery and then every 12 h for the next two days, all mice received injections of antibiotics (20 mg/kg/day intraperitoneal (IP) Metronidazole BRAUN; 85 mg/kg/day SC Cefoxitine PANPHARMA). Pain was controlled by SC injection of 0.1 mg/kg buprenorphine given 10 hours post-surgery, and twice daily for the next two days. 48 h after surgery, mice were anaesthetized with isoflurane before sacrifice by exsanguination from retro-orbital sinus (local anaesthesia by tetracaine 1% (THEA)) and cervical dislocation.

A severity score was evaluated every hour (except during the night) after surgery based on several clinical criteria: fur aspect, motor activity, posture, breathing, weight loss (Supplemental Table 2). Animals were euthanized if they reached a score ≥ 9 before the 2 days end-point.

In vivo depletion of plasma cells

0.5 mg/kg of Bortezomib (SIGMA-ALDRICH) or a control solution of 1/300 diluted Dimethylsulfoxyde (DMSO; SIGMA-ALDRICH), i.e. Bortezomib vehicle, was injected intravenously in both Sham and CLP mice 24h after surgery. This dose of Bortezomib was determined after dose-response experiment evaluating effectiveness of Bortezomib injection (0.5, 0.75 and 1 mg/kg) at depleting plasma cells without adversely affecting other cellular populations in spleens of CLP and Sham mice.

Cell purification

In mice, spleens were mechanically homogenized in phosphate buffer saline (PBS, EUROBIO) under sterile conditions to obtain a suspension of splenocytes. Red blood cell lysis was performed using hypotonic solution of NaCl and cells were then washed twice in PBS. Plasma cells were purified from splenocytes by positive selection with magnetic beads (EasySep™ Mouse CD138 Positive Selection Kit; STEMCELL), while B and T cells were purified by negative selection (EasySep™ Mouse B and T Cell Isolation Kits; STEMCELL). Purified cells were further used for proliferation assay or frozen at -80°C in 300µL RNA Lysis Buffer (ZYMO RESEARCH) for further RNA expression analysis.

In humans, healthy volunteers’ T cells were isolated from whole blood by immunodensity isolation (RosetteSep™ Human T Cell Enrichment Cocktail; STEMCELL). When necessary, regulatory T cells were depleted from T cells by magnetic cell separation (EasySep™ Human Pan-CD25 Positive Selection and Depletion Kit; STEMCELL), with a yield of <1% residual CD25+ CD4+ T cells. In septic patients, B cells were purified from whole blood by immunodensity isolation (RosetteSep™ Human B Cell Enrichment Cocktail; STEMCELL) and plasma cells by magnetic cell separation (MACSprep™ Multiple Myeloma CD138 MicroBeads; MILTENYI).

Analysis of cellular morphology by microscopy

Between 20,000 and 40,000 B cells and plasma cells were sorted by FACS (Aria II; BECTON DICKINSON) as B220+ CD138- and B220low CD138+ cells respectively. Cytocentrifuge was then used to concentrate cells onto a microscope slide so that they could be stained by May-Grünwald Giemsa and examined by a histologist blind of cell sorting populations. Images were acquired with a Nikon Eclipse 80i microscope at 60X magnification using a Basler camera and ICS Capture software (Tribvn, France).

Cell culture and proliferation assays

In mice, total splenocytes or splenic T cells were stained with Tag-it Violet™ Proliferation and Cell Tracking Dye (BIOLEGEND). T cell cultures (106 cells/mL) were set up in a culture medium consisting of RPMI (EUROBIO) supplemented with 10% AB human serum (Etablissement Français du Sang, Lyon, France), 200 mM L-glutamine (LONZA), 10 mg/mL penicillin-streptomycin (BIOLOGICAL INDRUSTRIES), 5 mg/mL fungizone (BRISTOL-MYERS SQUIBB) and 1% β-mercaptoethanol (SIGMA), in 96-well flat-bottom plates. Cells were stimulated by anti-CD3/CD28 antibodies-coated beads (T Cell Activation/Expansion Kit; MILTENYI) at a bead-to-cell ratio 2:1.5, or by phytohemagglutinin (4 ng/µL) (THERMOFISHER). In the coculture assays, splenic B cells or plasma cells were stained with CellTrace™ Far Red (THERMOFISHER) and added at 1:1 ratio to T cells in the stimulated condition. After 72h of culture, supernatant was frozen at -80°C and cells harvested, then stained with propidium iodide (SIGMA-ALDRICH) or Zombie Aqua™ Fixable Viability Kit (BIOLEGEND) and analysed on flow cytometer to determine proportion of viable proliferating T cells in each culture condition. To measure proliferation of CD4 and CD8 T lymphocytes, cells were also stained with CD45-APC-Fire750, CD3-PE, CD4-APC and CD8-PC7 prior to analysis on flow cytometer. To explore regulatory mechanisms of plasma cells, additional culture conditions were used. HTS Transwell® inserts (0.4 µm pore size polycarbonate membrane; CORNING) were set on 96-well plates, with T cells on the bottom of the well and plasma cells on the top of the membrane, at a cell-cell ratio 1:1 and a concentration of 106 cells/mL. Anti-PD-L1 (Ultra-LEAFeaf Purified anti-mouse CD274, clone 10F.9G2B7-H1; BIOLEGEND) blocking antibody or corresponding isotype control (Ultra-LEAF Purified Rat IgG2b kappa; BIOLEGEND) were also added in some cultures at 10 µg/mL.

In humans, T cell cultures (106 cells/mL) were set up in 96-well flat-bottom plates in a same culture medium than mouse splenic T cells, but without β-mercaptoethanol. Between 50,000 and 100,000 purified T cells from healthy volunteers (depleted or not of Tregs) were cultured in the absence or in the presence of stimulating anti-CD2/CD3/CD28 antibodies-coated beads (T Cell Activation/Expansion Kit; MILTENYI) at a bead-to-cell ratio of 1.25:1, either alone or with B cells or plasma cells from septic patients at 1:1 ratio in the stimulated condition. After 72h of culture, cells were harvested, stained with anti-CD3-APC-AF750, anti-CD4-APC and anti-CD8-KrO antibodies and T lymphocyte proliferative response was evaluated by flow cytometry in each condition measuring 5-ethynyl-2’deoxyuridine (EdU) incorporation in T cells as described elsewhere (Click-iT© EdU Flow cytometry assay kit, INVITROGEN)48.

Mice immunization with ovalbumin and evaluation of antigen-specific T lymphocyte response ex vivo

Mice were immunized with subcutaneous injection in each hip of 0.1 mL of chicken ovalbumin peptide OVA323-339 in complete Freund’s adjuvant (OVA323-339/CFA Emulsion at 1mg/mL, HOOKE LABORATORIES). Mice were submitted to CLP or Sham surgery fourteen days later. Mice were sacrificed 48h after surgery and spleens were harvested. Total splenic cells (106 cells/mL) were cultured in 96-well flat-bottom plates in complete culture medium in the absence or in the presence of OVA323-339 peptide (HOOKE LABORATORIES) at 3 concentrations (20 µg/mL, 5 µg/mL and 2 µg/mL). After 7 days of culture, supernatants were collected and frozen at -80°C for further IFNγ measurement.

Flow cytometry

In mice, total splenocytes absolute counts were determined using fluorescent microbeads (Flow-Count Fluorospheres, BECKMAN COULTER) and LDS 751 cell-permeant nucleic acid stain (INVITROGEN). For each immunophenotyping analysis, 100 µL of spleen cell suspension (i.e. 500,000 cells) were incubated with 10 µL of TruStain fcX™ antibody (clone 93; BIOLEGEND) for 10 min at 4°C prior to immunostaining. Without washing step, cells were then incubated with 3µL of each monoclonal antibodies (mAb) for 30 min in the dark at room temperature, then washed in PBS. List of reagents is provided in Supplementary Table 3. Supernatant was discarded and cell pellet resuspended in 300 µL of PBS and 1% formaldehyde (SIGMA-ALDRICH) before data acquisition on flow cytometer. Intracellular stainings for blimp-1 and IgM were performed using eBioscience™ FoxP3/transcription factor staining buffer set (THERMO FISHER SCIENTIFIC) as followed: after last washing step, cell pellet was resuspended in 50µL of fixation buffer and incubated for 30 min at 4°C in the dark. Cells were then centrifuge, supernatant discarded before a washing step with 50µL of permeabilization buffer. Cells were then incubated with monoclonal antibodies for 35 min at room temperature in the dark, then washed in perm buffer and finally resuspended in PBS before data acquisition on flow cytometer.

In humans, for cell surface stainings, 100 µL of whole blood were washed twice with PBS. Cell pellet was resuspended in 100 µL of AB human serum and incubated with mAb for 15 min in the dark at room temperature. Red blood cells were then lysed with 500 µL Optilyse C (BECKMAN COULTER) during 10 min in the dark at room temperature. Cells were finally washed with PBS and pellet resuspended with 300 µL PBS. Intracellular stainings for blimp-1 and IgM were performed using PerFix-nc Kit (BECKMAN COULTER) as followed: after 15 min incubation of 50 µL of whole blood with membrane antibodies, cells were washed and supernatant discarded. Cells were next incubated with 25 µL of R1 reagent in the dark at room temperature then washed. Intracellular antibodies were diluted in 300 µL of R2 reagent and added to cells. After 30 min incubation at room temperature in the dark, cells were washed by 3 mL of R3 reagent (1X) and finally resuspended in R3 reagent before data acquisition on flow cytometer. In retrospective study of REALISM data, plasma cells were identified as CD19low SSChigh circulating cells, while in RICO and IMMUNOSEPSIS prospective studies, they were defined as CD38+ CD138+ cells. We previously showed that these two phenotypes were well correlated and relevant for plasma cells analysis in human blood 9.

All cytometry experiments were performed on Navios cytometer (BECKMAN COULTER) or LSR II (BD BIOSCIENCES) the same day as sample collection. Calibration beads (Flow-Set and Flow-Check Fluorospheres; BECKMAN COULTER) were daily run to check for routine alignment, day-to-day and long-term performance validation. Fluorescence-minus-one (FMO) or negative cells controls were used to established positive windows when needed. FACS analysis used Kaluza software 2.1 (BECKMAN COULTER) and FlowJo 10.8.1 software (BD BIOSCIENCES). Depending on the marker or cell population analysed, percentage and/or median fluorescence intensity (MFI) were monitored. Representative histograms of flow cytometry stainings expressed as MFI are illustrated in Fig. S5, Fig S6, Fig S7.

RNA extraction and gene expression analysis

Minimum 50,000 purified splenic B and plasma cells from 6 Sham and 6 CLP mice (i.e. 24 samples in total) were thawed for total RNA extraction, using RNeasy Plus Mini Kit (QIAGEN) following manufacturer’s protocol. RNA concentration and integrity number (RIN) were measured with 2100 Bioanalyzer System (AGILENT). For each sample, 100 ng RNA presenting with RIN between 8.6 to 10 were processed for gene expression analysis. NANOSTRING nCounter assay, a hybridization-based multiplex assay, was performed for gene expression analysis at the hospital genomic platform (Plateforme BIOGENET Sud – Hospices Civils de Lyon, France). Total RNA were diluted with Nuclease-Free water (SIGMA-ALDRICH) at 20 ng/µl in the 12-strip provided by NANOSTRING, and were analysed using the nCounter® PanCancer Immune Profiling Panel according to manufacturer’s instructions. Eight negative probes and six serial concentrations of positive control probes were included in the multiplexed reaction, respectively allowing to determine the background signal and to perform a normalization to correct potential sources of variation associated with the technical platform, as described elsewhere 49. Then, to prevent effect of potential RNA input differences, we normalised the data according to the expression of two genes (Hdac3 and Cyld), identified with geNorm method 50 as stable between the four groups of samples studied. Finally, among 768 genes available in the nCounter® PanCancer Immune Profiling Panel, 110 turned out to have no expression in any of the 24 samples analysed, and we selected genes with RNA expression in > 75% of B cells or plasma cells samples for further analysis, which represented 540 genes for B cells and 554 genes for plasma cells.

IL-10 measurement

IL-10 was measured in undiluted culture supernatants using sandwich ELISA MAX™ Deluxe Set Mouse IL-10 (BIOLEGEND) according to manufacturer’s recommendations. Samples were analysed as duplicates when possible. Absorbance was read at 450 nm with Elx 808JU microplaque reader (BIOTECK). Gen5 software 3.04 (BIOTECK) was used to generate standard curve through regression analysis on the log of standard values and to calculate samples’ concentrations.

IFNγ measurement

In mice, IFNγ was measured in undiluted culture supernatants using Mouse IFN-gamma DuoSet ELISA (BIO-TECHNE) according to manufacturer’s recommendations. Samples were analysed as duplicates. In humans, IFNγ was measured in culture supernatants using Human IFN-gamma DuoSet ELISA (BIO-TECHNE) according to manufacturer’s recommendations. Samples were analysed undiluted for non-stimulated culture conditions or after 1/50 dilution for stimulated culture conditions, as duplicates when possible. Absorbance was read at 450 nm with ELx808 microplaque reader (BIOTECK). Gen5 software 3.04 (BIOTECK) was used to generate a standard curve through a four-parameter logistic curve-fit and to calculate samples’ concentrations.

Statistical Analyses

Experimental data were analysed by R (Version 3.6.2; Boston, MA, USA) and GraphPad Prism (Version 9.0.2; La Jolla, CA, USA). Statistical details of the experiments are provided in the respective figure legends. Bar plots present individual values as well as mean and standard deviation. Comparison of continuous variables was performed with non-parametric Mann– Whitney or Wilcoxon tests for matched samples. Kaplan–Meier survival analysis and log-rank test was used to compare mortality in Sham and CLP mice. RNA expression in purified B and plasma cells from mice was explored drawing volcano plots and heatmaps (pheatmap package in R), and using Ingenuity Pathway Analysis (QIAGEN). P values < 0.05 were considered statistically significant and are shown on plots.

DATA AVAILABILITY

Source data are provided with this paper.

AUTHORS CONTRIBUTIONS

Conceptualization: MG, TD, OT, GM, FV

Methodology: MG, CV, TD, OT, GM, FV, BP

Investigation: AV, RC, LJ, JL, KK, MO, HP, MM, AF, EP, CG, MB, ACL, TR, LA, MC, CV, ML, LC, BP

Visualization: MG, TD, LJ, JL, MM, OT, GM, FV, CV, ML

Funding acquisition: GM, FV

Project administration: MB, FV Supervision: TD, GM, FV

Writing – original draft: MG, FV

Writing – review & editing: MG, CV, TD, AV, ML, LC, ACL, RC, HP, LJ, JL, KK, MO, MM, AF, EP, CG, MB, TR, LA, MC, OT, GM, FV, BP

COMPETING INTERESTS STATEMENT

MM and EP are employees of bioMérieux SA, an in vitro diagnostic company. RC, MM, EP, CG, ACL, TR and GM work in a joint research unit, co funded by the Hospices Civils de Lyon and bioMérieux. This private company has no role in study design, results analysis or publication.

STUDY GROUPS

RICO study group

- Hospices Civils de Lyon, Lyon-Sud University Hospital, Immunology Laboratory: Remi Pescarmona, Lorna Garnier, Christine Lombard, Magali Perret, Marine Villard

- Joint Research Unit HCL-bioMérieux: Valérie Cheynet, Filippo Conti

- Centre d’Investigation Clinique de Lyon (CIC 1407 Inserm): Marie Groussaud, Marielle Buisson, Laetitia Itah, Inesse Boussaha

- Hospices Civils de Lyon, Edouard Herriot Hospital, Immunology Laboratory: Françoise Poitevin-Later, Christophe Malcus, Eleonore Micoud

- RICO clinical investigators: Florent Wallet, Marie-Charlotte Delignette, Frederic Dailler

- Hospices Civils de Lyon, Edouard Herriot Hospital, Medical intensive Care Department: Marie Simon, Auguste Dargent, Pierre-Jean Bertrand, Neven Stevic, Marion Provent

- Hospices Civils de Lyon, Edouard Herriot Hospital, Anesthesia and Critical Care Medicine Department: Laurie Bignet, Valérie Cerro

- Hospices Civils de Lyon, Croix-Rousse University Hospital, Medical intensive Care Department: Jean-Christophe Richard, Laurent Bitker, Mehdi Mezidi, Loredana Baboi

REALISM study group

- HCL: Sophie ARNAL, Caroline AUGRIS-MATHIEU, Frédérique BAYLE, Liana CARUSO, Charles-Eric BER, Asma BEN-AMOR, Anne-Sophie BELLOCQ, Farida BENATIR, Anne BERTIN-MAGHIT, Marc BERTIN-MAGHIT, André BOIBIEUX, Yves BOUFFARD, Jean-Christophe CEJKA, Valérie CERRO, Jullien CROZON-CLAUZEL, Julien DAVIDSON, Sophie DEBORD-PEGUET, Benjamin DELWARDE, Robert DELEAT-BESSON, Claire DELSUC, Bertrand DEVIGNE, Laure FAYOLLE-PIVOT, Alexandre FAURE, Bernard FLOCCARD, Julie GATEL, Charline GENIN, Thibaut GIRARDOT, Arnaud GREGOIRE, Baptiste HENGY, Laetitia HURIAUX, Catherine JADAUD, Alain LEPAPE, Véronique LERAY, Anne-Claire LUKASZEWICZ, Guillaume MARCOTTE, Olivier MARTIN, Marie MATRAY, Delphine MAUCORT-BOULCH, Pascal MEURET, Céline MONARD, Florent MORICEAU, Guillaume MONNERET, Nathalie PANEL, Najia RAHALI, Thomas RIMMELE, Cyrille TRUC, Thomas UBERTI, Hélène VALLIN, Fabienne VENET, Sylvie TISSOT, Abbès ZADAM

- bioMérieux: Sophie BLEIN, Karen BRENGEL-PESCE, Elisabeth CERRATO, Valérie CHEYNET, Emmanuelle GALLET-GORIUS, Audrey GUICHARD, Camille JOURDAN, Natacha KOENIG, François MALLET, Boris MEUNIER, Virginie MOUCADEL, Marine MOMMERT, Guy ORIOL, Alexandre PACHOT, Estelle PERONNET, Claire SCHREVEL, Olivier TABONE, Julien TEXTORIS, Javier YUGUEROS MARCOS

- BIOASTER: Jérémie BECKER, Frédéric BEQUET, Yacine BOUNAB, Florian BRAJON, Bertrand CANARD, Muriel COLLUS, Nathalie GARCON, Irène GORSE, Cyril GUYARD, Fabien LAVOCAT, Philippe LEISSNER, Karen LOUIS, Maxime MISTRETTA, Jeanne MORINIERE, Yoann MOUSCAZ, Laura NOAILLES, Magali PERRET, Frédéric REYNIER, Cindy RIFFAUD, Mary-Luz ROL, Nicolas SAPAY, Trang TRAN, Christophe VEDRINE

- SANOFI: Christophe CARRE, Pierre CORTEZ, Aymeric DE MONFORT, Karine FLORIN, Laurent FRAISSE, Isabelle FUGIER, Sandrine PAYRARD, Annick PELERAUX, Laurence QUEMENEUR

- ESPCI: Andrew GRIFFITHS, Stephanie TOETSCH

- GSK: Teri ASHTON, Peter J. GOUGH, Scott B. BERGER, David GARDINER, Iain GILLESPIE, Aidan MACNAMARA, Aparna RAYCHAUDHURI, Rob SMYLIE, Lionel TAN, Craig TIPPLE

ACKNOLEDGMENTS

The authors would like to thank (i) the clinical research assistants from intensive care units of Edouard Herriot Hospital (Valérie Cerro, Laurie Bignet, Marion Provent among others) for their great help in the screening and sampling of patients as well as in the preparation of case report forms, (ii) the technical staff from the Edouard Herriot Hospital Immunology Lab for help in sample management (iii) bioMérieux joint research unit for dedicated involvement in flow cytometry and transcriptomic data analysis (iv) Centre d’Investigation Clinique de Lyon (CIC 1407 Inserm) for help in RICO clinical study management. The authors would also like to acknowledge the important contribution and motivation of the medical and paramedical staff of the Edouard Herriot Hospital. Finally, the authors would like to thank the patients and their families for supporting this study.

REALISM study received funding from the Agence Nationale de la Recherche through a grant awarded to BIOASTER (Grant number #ANR-10-AIRT-03) and from bioMérieux, Sanofi and GSK. IMMUNOSEPSIS study was funded by Hospices civils de Lyon. RICO study was supported by funds from the Hospices Civils de Lyon, Fondation HCL and Claude Bernard Lyon 1 University/ Région Auvergne Rhône-Alpes. CV is a recipient of a Poste d’Accueil Inserm.

Footnotes

  • ↵# co-second to last authors

REFERENCES

  1. ↵
    Singer, M. et al. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA 315 (2016).
  2. ↵
    Rudd, K. E. et al. Global, regional, and national sepsis incidence and mortality, 1990-2017: analysis for the Global Burden of Disease Study. Lancet 395, 200–211 (2020).
    OpenUrlCrossRefPubMed
  3. ↵
    Reinhart, K. et al. Recognizing Sepsis as a Global Health Priority - A WHO Resolution. N Engl J Med 377, 414–417 (2017).
    OpenUrlCrossRefPubMed
  4. ↵
    Venet, F. & Monneret, G. Advances in the understanding and treatment of sepsis-induced immunosuppression. Nat Rev Nephrol 14, 121–137 (2018).
    OpenUrlCrossRefPubMed
  5. ↵
    Hotchkiss, R. S., Monneret, G. & Payen, D. Immunosuppression in sepsis: a novel understanding of the disorder and a new therapeutic approach. Lancet Infect Dis 13, 260–268 (2013).
    OpenUrlCrossRefPubMedWeb of Science
  6. ↵
    Leentjens, J., Kox, M., van der Hoeven, J. G., Netea, M. G. & Pickkers, P. Immunotherapy for the adjunctive treatment of sepsis: from immunosuppression to immunostimulation. Time for a paradigm change? Am J Respir Crit Care Med 187, 1287–1293 (2013).
    OpenUrlCrossRefPubMedWeb of Science
  7. ↵
    Venet, F. et al. Regulatory T cell populations in sepsis and trauma. J Leukoc Biol 83, 523–535 (2008).
    OpenUrlCrossRefPubMedWeb of Science
  8. ↵
    Venet, F., Demaret, J., Gossez, M. & Monneret, G. Myeloid cells in sepsis-acquired immunodeficiency. Ann N Y Acad Sci, 1499, 3–17 (2021).
  9. ↵
    Gustave, C. A. et al. Septic Shock Shapes B Cell Response toward an Exhausted-like/Immunoregulatory Profile in Patients. J Immunol 200, 2418–2425 (2018).
    OpenUrlAbstract/FREE Full Text
  10. ↵
    Woodruff, M. C. et al. Extrafollicular B cell responses correlate with neutralizing antibodies and morbidity in COVID-19. Nat Immunol 21, 1506–1516 (2020).
    OpenUrlCrossRefPubMed
  11. ↵
    Lino, A. C. et al. LAG-3 Inhibitory Receptor Expression Identifies Immunosuppressive Natural Regulatory Plasma Cells. Immunity 49, 120–133 (2018).
    OpenUrlPubMed
  12. Matsushita, T., Yanaba, K., Bouaziz, J. D., Fujimoto, M. & Tedder, T. F. Regulatory B cells inhibit EAE initiation in mice while other B cells promote disease progression. J Clin Invest 118, 3420–3430 (2008).
    OpenUrlCrossRefPubMedWeb of Science
  13. ↵
    Mauri, C. & Bosma, A. Immune regulatory function of B cells. Annu Rev Immunol 30, 221–241 (2012).
    OpenUrlCrossRefPubMedWeb of Science
  14. ↵
    Umakoshi, K. et al. B lymphocytopenia and Bregs in a not-to-die murine sepsis model. Biochem Biophys Res Commun 523, 202–207 (2020).
    OpenUrl
  15. ↵
    Aziz, M., Holodick, N. E., Rothstein, T. L. & Wang, P. B-1a Cells Protect Mice from Sepsis: Critical Role of CREB. J Immunol 199, 750–760 (2017).
    OpenUrlAbstract/FREE Full Text
  16. ↵
    Yanaba, K. et al. A regulatory B cell subset with a unique CD1dhiCD5+ phenotype controls T cell- dependent inflammatory responses. Immunity 28, 639–650 (2008).
    OpenUrlCrossRefPubMedWeb of Science
  17. ↵
    Candando, K. M., Lykken, J. M. & Tedder, T. F. B10 cell regulation of health and disease. Immunol Rev 259, 259–272 (2014).
    OpenUrlCrossRefPubMed
  18. ↵
    Cerqueira, C., Manfroi, B. & Fillatreau, S. IL-10-producing regulatory B cells and plasmocytes: Molecular mechanisms and disease relevance. Semin Immunol 44, 101323 (2019).
  19. ↵
    Matsumoto, M. et al. Interleukin-10-producing plasmablasts exert regulatory function in autoimmune inflammation. Immunity 41, 1040–1051 (2014).
    OpenUrlCrossRefPubMedWeb of Science
  20. ↵
    Madan, R. et al. Nonredundant roles for B cell-derived IL-10 in immune counter-regulation. J Immunol 183, 2312–2320 (2009).
    OpenUrlAbstract/FREE Full Text
  21. ↵
    Venet, F. et al. Immune Profiling Demonstrates a Common Immune Signature of Delayed Acquired Immunodeficiency in Patients With Various Etiologies of Severe Injury. Crit Care Med, 50,565–575 (2022).
    OpenUrlCrossRef
  22. ↵
    Rol, M. L. et al. The REAnimation Low Immune Status Markers (REALISM) project: a protocol for broad characterisation and follow-up of injury-induced immunosuppression in intensive care unit (ICU) critically ill patients. BMJ Open 7, e015734 (2017).
    OpenUrlAbstract/FREE Full Text
  23. ↵
    Leijte, G. P. et al. Monocytic HLA-DR expression kinetics in septic shock patients with different pathogens, sites of infection and adverse outcomes. Crit Care 24, 110 (2020).
  24. ↵
    Venet, F. et al. Longitudinal assessment of IFN-I activity and immune profile in critically ill COVID-19 patients with acute respiratory distress syndrome. Crit Care 25, 140 (2021).
  25. ↵
    Sanaei, M. J. et al. New insights into regulatory B cells biology in viral, bacterial, and parasitic infections. Infect Genet Evol 89, 104753 (2021).
  26. ↵
    Wang, C. et al. CD19(+)CD24(hi)CD38(hi) regulatory B cells deficiency revealed severity and poor prognosis in patients with sepsis. BMC Immunol 23, 54 (2022).
  27. ↵
    Cervantes-Diaz, R. et al. Circulating B10 regulatory cells are decreased in severe and critical COVID-19. J Leukoc Biol 112, 333–337 (2022).
    OpenUrl
  28. ↵
    Yanaba, K., Bouaziz, J. D., Matsushita, T., Tsubata, T. & Tedder, T. F. The development and function of regulatory B cells expressing IL-10 (B10 cells) requires antigen receptor diversity and TLR signals. J Immunol 182, 7459–7472 (2009).
    OpenUrlAbstract/FREE Full Text
  29. ↵
    Nascimento, D. C. et al. Sepsis expands a CD39(+) plasmablast population that promotes immunosuppression via adenosine-mediated inhibition of macrophage antimicrobial activity. Immunity 54, 2024–2041 (2021).
    OpenUrl
  30. ↵
    Neves, P. et al. Signaling via the MyD88 adaptor protein in B cells suppresses protective immunity during Salmonella typhimurium infection. Immunity 33, 777–790 (2010).
    OpenUrlCrossRefPubMedWeb of Science
  31. Shalapour, S. et al. Immunosuppressive plasma cells impede T-cell-dependent immunogenic chemotherapy. Nature 521, 94–98 (2015).
    OpenUrlCrossRefPubMed
  32. ↵
    Shen, P. et al. IL-35-producing B cells are critical regulators of immunity during autoimmune and infectious diseases. Nature 507, 366–370 (2014).
    OpenUrlCrossRefPubMedWeb of Science
  33. ↵
    Teichmann, L. L. et al. B cell-derived IL-10 does not regulate spontaneous systemic autoimmunity in MRL.Fas(lpr) mice. J Immunol 188, 678–685 (2012).
    OpenUrlAbstract/FREE Full Text
  34. ↵
    Steinhauser, M. L. et al. IL-10 is a major mediator of sepsis-induced impairment in lung antibacterial host defense. J Immunol 162, 392–399 (1999).
    OpenUrlAbstract/FREE Full Text
  35. ↵
    Doi, T. et al. IgA plasma cells express the negative regulatory co-stimulatory molecule programmed cell death 1 ligand and have a potential tolerogenic role in the intestine. Biochem Biophys Res Commun 425, 918–923 (2012).
    OpenUrlCrossRefPubMed
  36. ↵
    Hotchkiss, R. S., Monneret, G. & Payen, D. Sepsis-induced immunosuppression: from cellular dysfunctions to immunotherapy. Nat Rev Immunol 13, 862–874 (2013).
    OpenUrlCrossRefPubMed
  37. ↵
    Wang, K., Liu, J. & Li, J. IL-35-producing B cells in gastric cancer patients. Medicine (Baltimore) 97, e0710 (2018).
    OpenUrl
  38. ↵
    Wang, K. et al. Aberrant frequency of IL-35 producing B cells in colorectal cancer patients. Cytokine 102, 206–210 (2018).
    OpenUrl
  39. ↵
    Wang, R. X. et al. Interleukin-35 induces regulatory B cells that suppress autoimmune disease. Nat Med 20, 633–641 (2014).
    OpenUrlCrossRefPubMed
  40. ↵
    Collison, L. W. et al. The inhibitory cytokine IL-35 contributes to regulatory T-cell function. Nature 450, 566–569, (2007).
    OpenUrlCrossRefPubMedWeb of Science
  41. ↵
    Mathew, D. et al. Deep immune profiling of COVID-19 patients reveals distinct immunotypes with therapeutic implications. Science 369 (2020).
  42. Consortium., C.-M.-o. B. A. C. A blood atlas of COVID-19 defines hallmarks of disease severity and specificity. Cell 185, 916–938 (2022).
    OpenUrlCrossRef
  43. ↵
    Shuwa, H. A. et al. Alterations in T and B cell function persist in convalescent COVID-19 patients. Med (N Y) 2, 720–735 e724 (2021).
    OpenUrl
  44. ↵
    Elsner, R. A. & Shlomchik, M. J. Germinal Center and Extrafollicular B Cell Responses in Vaccination, Immunity, and Autoimmunity. Immunity 53, 1136–1150 (2020).
    OpenUrlCrossRefPubMed
  45. ↵
    Fillatreau, S. Novel regulatory functions for Toll-like receptor-activated B cells during intracellular bacterial infection. Immunol Rev 240, 52–71 (2011).
    OpenUrlCrossRefPubMed
  46. ↵
    Rosser, E. C. & Mauri, C. Regulatory B cells: origin, phenotype, and function. Immunity 42, 607–612 (2015).
    OpenUrlCrossRefPubMed
  47. ↵
    Restagno, D. et al. Mice Survival and Plasmatic Cytokine Secretion in a “Two Hit” Model of Sepsis Depend on Intratracheal Pseudomonas Aeruginosa Bacterial Load. PLoS One 11, e0162109 (2016).
    OpenUrl
  48. ↵
    Poujol, F. et al. Flow cytometric evaluation of lymphocyte transformation test based on 5-ethynyl- 2’deoxyuridine incorporation as a clinical alternative to tritiated thymidine uptake measurement. J Immunol Methods 415, 71–79 (2014).
    OpenUrl
  49. ↵
    Urrutia, A. et al. Standardized Whole-Blood Transcriptional Profiling Enables the Deconvolution of Complex Induced Immune Responses. Cell Rep 16, 2777–2791 (2016).
    OpenUrl
  50. ↵
    Vandesompele, J. et al. Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes. Genome Biol 3, RESEARCH0034 (2002).
Back to top
PreviousNext
Posted August 08, 2024.
Download PDF

Supplementary Material

Email

Thank you for your interest in spreading the word about medRxiv.

NOTE: Your email address is requested solely to identify you as the sender of this article.

Enter multiple addresses on separate lines or separate them with commas.
PD-L1 mediated T cell inhibition by regulatory plasma cells induced after sepsis and COVID-19
(Your Name) has forwarded a page to you from medRxiv
(Your Name) thought you would like to see this page from the medRxiv website.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Share
PD-L1 mediated T cell inhibition by regulatory plasma cells induced after sepsis and COVID-19
Morgane Gossez, Clara Vigneron, Alexandra Vandermoeten, Margot Lepage, Louise Courcol, Remy Coudereau, Helena Paidassai, Laurent Jallades, Jonathan Lopez, Khalil Kandara, Marine Ortillon, Marine Mommert, Astrid Fabri, Estelle Peronnet, Clémence Grosjean, Marielle Buisson, Anne-Claire Lukaszewicz, Thomas Rimmelé, Laurent Argaud, Martin Cour, Bénédicte F Py, Olivier Thaunat, Thierry Defrance, Guillaume Monneret, Fabienne Venet, REALISM, RICO study groups
medRxiv 2024.08.08.24311630; doi: https://doi.org/10.1101/2024.08.08.24311630
Twitter logo Facebook logo LinkedIn logo Mendeley logo
Citation Tools
PD-L1 mediated T cell inhibition by regulatory plasma cells induced after sepsis and COVID-19
Morgane Gossez, Clara Vigneron, Alexandra Vandermoeten, Margot Lepage, Louise Courcol, Remy Coudereau, Helena Paidassai, Laurent Jallades, Jonathan Lopez, Khalil Kandara, Marine Ortillon, Marine Mommert, Astrid Fabri, Estelle Peronnet, Clémence Grosjean, Marielle Buisson, Anne-Claire Lukaszewicz, Thomas Rimmelé, Laurent Argaud, Martin Cour, Bénédicte F Py, Olivier Thaunat, Thierry Defrance, Guillaume Monneret, Fabienne Venet, REALISM, RICO study groups
medRxiv 2024.08.08.24311630; doi: https://doi.org/10.1101/2024.08.08.24311630

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Subject Area

  • Intensive Care and Critical Care Medicine
Subject Areas
All Articles
  • Addiction Medicine (349)
  • Allergy and Immunology (668)
  • Allergy and Immunology (668)
  • Anesthesia (181)
  • Cardiovascular Medicine (2648)
  • Dentistry and Oral Medicine (316)
  • Dermatology (223)
  • Emergency Medicine (399)
  • Endocrinology (including Diabetes Mellitus and Metabolic Disease) (942)
  • Epidemiology (12228)
  • Forensic Medicine (10)
  • Gastroenterology (759)
  • Genetic and Genomic Medicine (4103)
  • Geriatric Medicine (387)
  • Health Economics (680)
  • Health Informatics (2657)
  • Health Policy (1005)
  • Health Systems and Quality Improvement (985)
  • Hematology (363)
  • HIV/AIDS (851)
  • Infectious Diseases (except HIV/AIDS) (13695)
  • Intensive Care and Critical Care Medicine (797)
  • Medical Education (399)
  • Medical Ethics (109)
  • Nephrology (436)
  • Neurology (3882)
  • Nursing (209)
  • Nutrition (577)
  • Obstetrics and Gynecology (739)
  • Occupational and Environmental Health (695)
  • Oncology (2030)
  • Ophthalmology (585)
  • Orthopedics (240)
  • Otolaryngology (306)
  • Pain Medicine (250)
  • Palliative Medicine (75)
  • Pathology (473)
  • Pediatrics (1115)
  • Pharmacology and Therapeutics (466)
  • Primary Care Research (452)
  • Psychiatry and Clinical Psychology (3432)
  • Public and Global Health (6527)
  • Radiology and Imaging (1403)
  • Rehabilitation Medicine and Physical Therapy (814)
  • Respiratory Medicine (871)
  • Rheumatology (409)
  • Sexual and Reproductive Health (410)
  • Sports Medicine (342)
  • Surgery (448)
  • Toxicology (53)
  • Transplantation (185)
  • Urology (165)