Extracellular Vesicles Transfer the Receptor Programmed Death-1 in Rheumatoid Arthritis

Introduction Extracellular vesicles (EVs) have been recognized as route of communication in the microenvironment. They transfer proteins and microRNAs (miRNAs) between cells, and possess immunoregulatory properties. However, their role in immune-mediated diseases remains to be elucidated. We hypothesized a role for EVs in the rheumatoid arthritis (RA) joint, potentially involving the development of T cell exhaustion and transfer of the co-inhibitory receptor programmed death 1 (PD-1). Methods Synovial fluid mononuclear cells (SFMCs) and peripheral blood mononuclear cells (PBMCs) from RA patients were investigated for PD-1 and other markers of T cell inhibition. EVs were isolated from RA plasma and synovial fluid. In addition, healthy control (HC) and RA PBMCs and SFMCs were cultured to produce EVs. These were isolated and investigated by immunogold electron microscopy (EM) and also co-cultured with lymphocytes and PD-1 negative cells to investigate their functions. Finally, the miRNA expression profiles were assessed in EVs isolated from RA and HC cell cultures. Results Cells from the RA joint expressed several T cell co-inhibitory receptors, including PD-1, TIM-3, and Tigit. ELISA demonstrated the presence of PD-1 in EVs from RA plasma and synovial fluid. Immunogold EM visualized PD-1 expression by EVs. Co-culturing lymphocytes and the PD-1 negative cell line, U937 with EVs resulted in an induction of PD-1 on these cells. Moreover, EVs from RA PBMCs increased proliferation in lymphocytes when co-cultured with these. All EVs contained miRNAs associated with PD-1 and other markers of T cell inhibition and the content was significantly lower in EVs from RA PBMCs than HC PBMCs. Stimulation of the cells increased the miRNA expression. However, EVs isolated from stimulated RA SFMCs did not change their miRNA expression profile to the same extend. Conclusion EVs carrying both the PD-1 receptor and miRNAs associated with T cell inhibition were present in RA cell cultures. Upon stimulation, these miRNAs failed to be upregulated in EVs from RA SFMCs. This was in line with increased expression of T cell co-inhibitory markers on SFMCs. In conclusion, we suggest EVs to play a significant role in the RA microenvironment, potentially favoring the progression of T cell exhaustion.

[1]  Cellular Immunology , 2020, Definitions.

[2]  V. Kuchroo,et al.  Lag-3, Tim-3, and TIGIT: Co-inhibitory Receptors with Specialized Functions in Immune Regulation. , 2016, Immunity.

[3]  C. Théry,et al.  Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes , 2016, Proceedings of the National Academy of Sciences.

[4]  P. Blanco,et al.  Role of extracellular vesicles in autoimmune diseases. , 2016, Autoimmunity reviews.

[5]  Elmar L. Gool,et al.  Recent developments in the nomenclature, presence, isolation, detection and clinical impact of extracellular vesicles , 2016, Journal of thrombosis and haemostasis : JTH.

[6]  Charles King,et al.  2015 American College of Rheumatology Guideline for the Treatment of Rheumatoid Arthritis , 2016, Arthritis & rheumatology.

[7]  Raveendhara R. Bannuru,et al.  2015 American College of Rheumatology Guideline for the Treatment of Rheumatoid Arthritis , 2016, Arthritis care & research.

[8]  E. Wherry,et al.  Molecular and cellular insights into T cell exhaustion , 2015, Nature Reviews Immunology.

[9]  Paul A. Lyons,et al.  T cell exhaustion, costimulation and clinical outcome in autoimmunity and infection , 2015, Nature.

[10]  B. Zhu,et al.  T-cell exhaustion in the tumor microenvironment , 2015, Cell Death and Disease.

[11]  E. Wherry,et al.  Overcoming T cell exhaustion in infection and cancer. , 2015, Trends in immunology.

[12]  R. Kaplan,et al.  4-1BB Costimulation Ameliorates T Cell Exhaustion Induced by Tonic Signaling of Chimeric Antigen Receptors , 2015, Nature Medicine.

[13]  X. Liu,et al.  T-cell exhaustion in chronic hepatitis B infection: current knowledge and clinical significance , 2015, Cell Death and Disease.

[14]  B. Bussolati,et al.  Extracellular vesicles in the urine: markers and mediators of tissue damage and regeneration , 2014, Clinical kidney journal.

[15]  J. Bijlsma,et al.  Synovial T cell hyporesponsiveness to myeloid dendritic cells is reversed by preventing PD-1/PD-L1 interactions , 2014, Arthritis Research & Therapy.

[16]  G. Silverman,et al.  Programmed death-1 pathway in cancer and autoimmunity. , 2014, Clinical immunology.

[17]  Lijuan Huang,et al.  The PD-1/PD-Ls pathway and autoimmune diseases. , 2014, Cellular immunology.

[18]  S. Mornet,et al.  Extracellular vesicles from blood plasma: determination of their morphology, size, phenotype and concentration , 2014, Journal of thrombosis and haemostasis : JTH.

[19]  K. Stengaard-Pedersen,et al.  Increased soluble programmed death-1 (sPD-1) is associated with disease activity and radiographic progression in early rheumatoid arthritis , 2014, Scandinavian journal of rheumatology.

[20]  Burton E. Barnett,et al.  Molecular and transcriptional basis of CD4⁺ T cell dysfunction during chronic infection. , 2014, Immunity.

[21]  P. Greenberg,et al.  Tolerance and exhaustion: defining mechanisms of T cell dysfunction. , 2014, Trends in immunology.

[22]  D. Kaplan,et al.  Peripheral CD27-CD21- B-cells represent an exhausted lymphocyte population in hepatitis C cirrhosis. , 2014, Clinical immunology.

[23]  Yan-fang Jiang,et al.  Peripheral B-cell activation and exhaustion markers in patients with ankylosing spondylitis. , 2013, Life sciences.

[24]  Winston Patrick Kuo,et al.  Current methods for the isolation of extracellular vesicles , 2013, Biological chemistry.

[25]  Lieping Chen,et al.  Molecular mechanisms of T cell co-stimulation and co-inhibition , 2013, Nature Reviews Immunology.

[26]  Graça Raposo,et al.  Extracellular vesicles: Exosomes, microvesicles, and friends , 2013, The Journal of cell biology.

[27]  Burton E. Barnett,et al.  Progenitor and Terminal Subsets of CD8+ T Cells Cooperate to Contain Chronic Viral Infection , 2012, Science.

[28]  Fátima Sánchez-Cabo,et al.  Unidirectional transfer of microRNA-loaded exosomes from T cells to antigen-presenting cells , 2011, Nature communications.

[29]  D. Unutmaz,et al.  Revisiting Immune Exhaustion During HIV Infection , 2011, Current HIV/AIDS reports.

[30]  A. Zajac,et al.  T‐cell exhaustion: characteristics, causes and conversion , 2010, Immunology.

[31]  X. Mariette,et al.  Increased levels of circulating microparticles in primary Sjögren's syndrome, systemic lupus erythematosus and rheumatoid arthritis and relation with disease activity , 2009, Arthritis research & therapy.

[32]  C. Théry,et al.  Membrane vesicles as conveyors of immune responses , 2009, Nature Reviews Immunology.

[33]  Kaleb M. Pauley,et al.  MicroRNA in autoimmunity and autoimmune diseases. , 2009, Journal of autoimmunity.

[34]  A. Sawaguchi,et al.  Leukocytapheresis (LCAP) decreases the level of platelet-derived microparticles (MPs) and increases the level of granulocytes-derived MPs: a possible connection with the effect of LCAP on rheumatoid arthritis , 2009, Modern rheumatology.

[35]  Lianbo Yu,et al.  Detection of microRNA Expression in Human Peripheral Blood Microvesicles , 2008, PloS one.

[36]  G. Freeman,et al.  PD-1 Regulates Self-Reactive CD8+ T Cell Responses to Antigen in Lymph Nodes and Tissues1 , 2007, The Journal of Immunology.

[37]  R. Ahmed,et al.  Restoring function in exhausted CD8 T cells during chronic viral infection , 2006, Nature.

[38]  D. Bartel MicroRNAs Genomics, Biogenesis, Mechanism, and Function , 2004, Cell.

[39]  F. Breedveld,et al.  Cell-derived microparticles in synovial fluid from inflamed arthritic joints support coagulation exclusively via a factor VII-dependent mechanism. , 2002, Arthritis and rheumatism.

[40]  M. A. van de Laar,et al.  Elevated levels of platelet microparticles are associated with disease activity in rheumatoid arthritis. , 2002, Arthritis and rheumatism.

[41]  A. Régnault,et al.  TCR Activation of Human T Cells Induces the Production of Exosomes Bearing the TCR/CD3/ζ Complex1 , 2002, The Journal of Immunology.

[42]  C. Dinarello,et al.  Basis for defective responses of rheumatoid arthritis synovial fluid lymphocytes to anti-CD3 (T3) antibodies. , 1986, The Journal of clinical investigation.

[43]  Raveendhara R. Bannuru,et al.  American College of Rheumatology Guideline for the Treatment of Rheumatoid Arthritis , 2015 .

[44]  M. Logozzi,et al.  Exosomes: the ideal nanovectors for biodelivery , 2013, Biological chemistry.

[45]  Wei Wu MicroRNA and Cancer , 2011, Methods in Molecular Biology.

[46]  R. Smeenk,et al.  Functional analysis of synovial fluid and peripheral blood T cells from patients with rheumatoid arthritis , 2004, Rheumatology International.