Skip to main content

Advertisement

Log in

Reduced peripheral blood dendritic cell and monocyte subsets in MDS patients with systemic inflammatory or dysimmune diseases

  • Original Article
  • Published:
Clinical and Experimental Medicine Aims and scope Submit manuscript

Abstract

Background

Systemic inflammatory and autoimmune diseases (SIADs) occur in 10–20% of patients with myelodysplastic syndrome (MDS). Recently identified VEXAS (Vacuoles, E1 enzyme, X-linked, Autoinflammatory, Somatic) syndrome, associated with somatic mutations in UBA1 (Ubiquitin-like modifier-activating enzyme 1), encompasses a range of severe inflammatory conditions along with hematological abnormalities, including MDS. The pathophysiological mechanisms underlying the association between MDS and SIADs remain largely unknown, especially the roles of different myeloid immune cell subsets. The aim of this study was to quantitatively evaluate peripheral blood myeloid immune cells (dendritic cells (DC) and monocytes) by flow cytometry in MDS patients with associated SIAD (n = 14, most often including relapsing polychondritis or neutrophilic dermatoses) and to compare their distribution in MDS patients without SIAD (n = 23) and healthy controls (n = 7). Most MDS and MDS/SIAD patients had low-risk MDS. Eight of 14 (57%) MDS/SIAD patients carried UBA1 somatic mutations, defining VEXAS syndrome.Compared with MDS patients, most DC and monocyte subsets were significantly decreased in MDS/SIAD patients, especially in MDS patients with VEXAS syndrome. Our study provides the first overview of the peripheral blood immune myeloid cell distribution in MDS patients with associated SIADs and raises several hypotheses: possible redistribution to inflammation sites, increased apoptosis, or impaired development in the bone marrow.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

Availability of data and materials

Available under request.

Abbreviations

AML:

Acute myeloid leukemia

ANSM:

Agence Nationale de Sécurité du Médicament

AZA:

Azacitidine

CD:

Cluster of differentiation

cDC:

Conventional myeloid dendritic cells

cDC1:

Conventional myeloid dendritic cells 1

cDC2:

Conventional myeloid dendritic cells 2

CMML:

Chronic myelomonocytic leukemia

cMo:

Classical monocytes

DC:

Dendritic cells

EDTA:

Ethylenediaminetetraacetic acid

PBMC:

Peripheral blood mononuclear cells

GFM:

Groupe Francophone des Myélodysplasies

IPSS:

International prognostic scoring system

MDS:

Myelodysplastic syndrome

MINHEMON:

French Network of dysimmune disorders associated with hemopathies

Mo:

Monocytes

ncMo:

Nonclassical monocytes

iMo:

Intermediate monocytes

pDC:

Plasmacytoid dendritic cells

R-IPSS:

Revised international prognostic scoring system

SD:

Standard deviation

SIAD:

Systemic inflammatory and autoimmune disease

slan Mo:

Slan + nonclassical monocytes.

slanDC:

6-Sulfo LacNAc dendritic cells

SLE:

Systemic lupus erythematosus

UBA1:

Ubiquitin-like modifier-activating enzyme 1

UBA1-WT:

Wild-type UBA1

VEXAS:

Vacuoles, E1 enzyme, X-linked, autoinflammatory, somatic

References

  1. Adès L, Itzykson R, Fenaux P. Myelodysplastic syndromes. Lancet. 2014;383:2239–52.

    PubMed  Google Scholar 

  2. Kordasti SY, et al. IL-17-producing CD4+T cells, pro-inflammatory cytokines and apoptosis are increased in low risk myelodysplastic syndrome. Br J Haematol. 2009;145:64–72.

    CAS  PubMed  Google Scholar 

  3. Bouchliou I, et al. Th17 and Foxp3+ T regulatory cell dynamics and distribution in myelodysplastic syndromes. Clin Immunol. 2011;139:350–9.

    CAS  PubMed  Google Scholar 

  4. Kordasti SY, et al. CD4+CD25high Foxp3+ regulatory T cells in myelodysplastic syndrome (MDS). Blood. 2007;110:847–50.

    CAS  PubMed  Google Scholar 

  5. Kotsianidis I, et al. Kinetics, function and bone marrow trafficking of CD4+CD25+FOXP3+ regulatory T cells in myelodysplastic syndromes (MDS). Leukemia. 2009;23:510–8.

    CAS  PubMed  Google Scholar 

  6. Kittang AO, et al. Expansion of myeloid derived suppressor cells correlates with number of T regulatory cells and disease progression in myelodysplastic syndrome. Oncoimmunology. 2016;5: e1062208.

    PubMed  Google Scholar 

  7. Grignano E, et al. Autoimmune manifestations associated with myelodysplastic syndromes. Ann Hematol. 2018;97:2015–23.

    CAS  PubMed  Google Scholar 

  8. Beck DB, et al. Somatic mutations in UBA1 and severe adult-onset autoinflammatory disease. N Engl J Med. 2020;383:2628–38.

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Georgin-Lavialle S, et al. Further characterization of clinical and laboratory features in VEXAS syndrome: large-scale analysis of a multicentre case series of 116 French patients. Br J Dermatol. 2021. https://doi.org/10.1111/bjd.20805.

    Article  PubMed  Google Scholar 

  10. Breton G, et al. Human dendritic cells (DCs) are derived from distinct circulating precursors that are precommitted to become CD1c+ or CD141+ DCs. J Exp Med. 2016;213:2861–70.

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Breton G, et al. Circulating precursors of human CD1c+ and CD141+ dendritic cells. J Exp Med. 2015;212:401–13.

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Schlitzer A, et al. Identification of cDC1- and cDC2-committed DC progenitors reveals early lineage priming at the common DC progenitor stage in the bone marrow. Nat Immunol. 2015;16:718–28.

    CAS  PubMed  Google Scholar 

  13. Dzionek A, et al. BDCA-2, BDCA-3, and BDCA-4: three markers for distinct subsets of dendritic cells in human peripheral blood. J Immunol. 2000;165:6037–46.

    CAS  PubMed  Google Scholar 

  14. MacDonald KPA, et al. Characterization of human blood dendritic cell subsets. Blood. 2002;100:4512–20.

    CAS  PubMed  Google Scholar 

  15. Robbins SH, et al. Novel insights into the relationships between dendritic cell subsets in human and mouse revealed by genome-wide expression profiling. Genome Biol. 2008;9:R17.

    PubMed  PubMed Central  Google Scholar 

  16. Ziegler-Heitbrock L, et al. Nomenclature of monocytes and dendritic cells in blood. Blood. 2010;116:e74–80.

    CAS  PubMed  Google Scholar 

  17. Cros J, et al. Human CD14dim monocytes patrol and sense nucleic acids and viruses via TLR7 and TLR8 receptors. Immunity. 2010;33:375–86.

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Wong KL, et al. Gene expression profiling reveals the defining features of the classical, intermediate, and nonclassical human monocyte subsets. Blood. 2011;118:e16–31.

    CAS  PubMed  Google Scholar 

  19. Zawada AM, et al. SuperSAGE evidence for CD14++CD16+ monocytes as a third monocyte subset. Blood. 2011;118:e50–61.

    CAS  PubMed  Google Scholar 

  20. Schäkel K, et al. Human 6-Sulfo LacNAc-expressing dendritic cells are principal producers of early interleukin-12 and are controlled by erythrocytes. Immunity. 2006;24:767–77.

    PubMed  Google Scholar 

  21. Schäkel K, et al. A novel dendritic cell population in human blood: one-step immunomagnetic isolation by a specific mAb (M-DC8) and in vitro priming of cytotoxic T lymphocytes. Eur J Immunol. 1998;28:4084–93.

    PubMed  Google Scholar 

  22. Schäkel K, et al. 6-Sulfo LacNAc, a novel carbohydrate modification of PSGL-1, defines an inflammatory type of human dendritic cells. Immunity. 2002;17:289–301.

    PubMed  Google Scholar 

  23. van Leeuwen-Kerkhoff N, et al. Human bone marrow-derived myeloid dendritic cells show an immature transcriptional and functional profile compared to their peripheral blood counterparts and separate from slan+ non-classical monocytes. Front Immunol. 2018;9:1619.

    PubMed  PubMed Central  Google Scholar 

  24. van Leeuwen-Kerkhoff N, et al. Transcriptional profiling reveals functional dichotomy between human Slan+non-classical monocytes and myeloid dendritic cells. J Leukoc Biol. 2017;102:1055–68.

    PubMed  Google Scholar 

  25. Hofer TP, et al. slan-defined subsets of CD16-positive monocytes: impact of granulomatous inflammation and M-CSF receptor mutation. Blood. 2015;126:2601–10.

    CAS  PubMed  Google Scholar 

  26. Braun T, Fenaux P. Myelodysplastic syndromes (MDS) and autoimmune disorders (AD): cause or consequence? Best Pract Res Clin Haematol. 2013;26:327–36.

    CAS  PubMed  Google Scholar 

  27. Sallman DA, List A. The central role of inflammatory signaling in the pathogenesis of myelodysplastic syndromes. Blood. 2019;133:1039–48.

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Zhao LP, et al. Genomic landscape of MDS/CMML associated with systemic inflammatory and autoimmune disease. Leukemia. 2021;35:2720–4.

    PubMed  Google Scholar 

  29. Van Leeuwen-Kerkhoff N, et al. Reduced frequencies and functional impairment of dendritic cell subsets and non-classical monocytes in myelodysplastic syndromes. Haematologica. 2021. https://doi.org/10.3324/haematol.2020.268136.

    Article  PubMed Central  Google Scholar 

  30. Gill MA, et al. Blood dendritic cells and DC-poietins in systemic lupus erythematosus. Hum Immunol. 2002;63:1172–80.

    CAS  PubMed  Google Scholar 

  31. Robak E, Smolewski P, Woźniacka A, Sysa-Jedrzejowska A, Robak T. Clinical significance of circulating dendritic cells in patients with systemic lupus erythematosus. Mediat Inflamm. 2004;13:171–80.

    CAS  Google Scholar 

  32. Scheinecker C, Zwlfer B, Kller M, Mnner G, Smolen JS. Alterations of dendritic cells in systemic lupus erythematosus: phenotypic and functional deficiencies. Arthritis Rheum. 2001;44:856–65.

    CAS  PubMed  Google Scholar 

  33. Migita K, et al. Reduced blood BDCA-2+ (lymphoid) and CD11c+ (myeloid) dendritic cells in systemic lupus erythematosus. Clin Exp Immunol. 2005;142:84–91.

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Fiore N, et al. Immature myeloid and plasmacytoid dendritic cells infiltrate renal tubulointerstitium in patients with lupus nephritis. Mol Immunol. 2008;45:259–65.

    CAS  PubMed  Google Scholar 

  35. Jin O, et al. Systemic lupus erythematosus patients have increased number of circulating plasmacytoid dendritic cells, but decreased myeloid dendritic cells with deficient CD83 expression. Lupus. 2008;17:654–62.

    CAS  PubMed  Google Scholar 

  36. Henriques A, et al. Functional characterization of peripheral blood dendritic cells and monocytes in systemic lupus erythematosus. Rheumatol Int. 2012;32:863–9.

    CAS  PubMed  Google Scholar 

  37. Gerl V, et al. Blood dendritic cells in systemic lupus erythematosus exhibit altered activation state and chemokine receptor function. Ann Rheum Dis. 2010;69:1370–7.

    CAS  PubMed  Google Scholar 

  38. Tucci M, et al. Glomerular accumulation of plasmacytoid dendritic cells in active lupus nephritis: role of interleukin-18. Arthritis Rheum. 2008;58:251–62.

    CAS  PubMed  Google Scholar 

  39. Olaru F, et al. Intracapillary immune complexes recruit and activate slan-expressing CD16+ monocytes in human lupus nephritis. JCI Insight. 2018;3: e96492.

    PubMed  PubMed Central  Google Scholar 

  40. Zakine E, et al. UBA1 variations in neutrophilic dermatosis skin lesions of patients with VEXAS syndrome. JAMA Dermatol. 2021;157:1349–54.

    PubMed  Google Scholar 

  41. Mékinian A, et al. A phase II study of the efficacy and tolerance of azacytidine (AZA) in steroid dependent/refractory systemic autoimmune and inflammatory disorders (SAID) associated with MDS or CMML (GFM- AZA-SAID -trial). Blood. 2021;138:3697.

    Google Scholar 

  42. Arber DA, et al. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood. 2016;127:2391–405.

    CAS  PubMed  Google Scholar 

  43. Greenberg PL, et al. Revised international prognostic scoring system for myelodysplastic syndromes. Blood. 2012;120:2454–65.

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Pfeilstöcker M, et al. Time-dependent changes in mortality and transformation risk in MDS. Blood. 2016;128:902–10.

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank Frédéric de Vassoigne for his help in collecting patient samples (Tumorothèque Saint-Antoine, APHP, Hôpital Saint-Antoine, Paris, France).

Funding

There was no funding for this research.

Author information

Authors and Affiliations

Authors

Consortia

Contributions

All coauthors were involved in study design, sample collection, conducting experiments, analyzing results, drafting the manuscript, and approving the final manuscript prior to submission.

Corresponding author

Correspondence to Vincent Jachiet.

Ethics declarations

Conflict of interest

AM is investigator of CELGENE, ROCHE, CHUGAI founded trials with APHP and Hopital 15–20 promotion; AM received several fees for congress travels and experts’ use from LFB, SANOFI, SHIRE, and CELGENE. The other authors declare no conflict of interest.

Ethics approval and consent to participate

All participants provided written informed consent. The study was approved by the local institutional review board (EudraCT 2016-000918-30, ANSM (Agence Nationale de Sécurité du Médicament) 160580A-11, registration date 26-AUG-2016) and in accordance with the Declaration of Helsinki.

Consent for publication

Not applicable.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Below is the link to the electronic supplementary material.

Supplementary file1 (TIF 30629 kb)

Rights and permissions

Springer Nature or its licensor holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jachiet, V., Ricard, L., Hirsch, P. et al. Reduced peripheral blood dendritic cell and monocyte subsets in MDS patients with systemic inflammatory or dysimmune diseases. Clin Exp Med 23, 803–813 (2023). https://doi.org/10.1007/s10238-022-00866-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10238-022-00866-5

Keywords

Navigation