Cross‐decoration of dendritic cells by non‐inherited maternal antigen‐containing extracellular vesicles: Potential mechanism for PD‐L1‐based tolerance in cord blood and organ transplantation

Exposure to non‐inherited maternal antigens (NIMA) during the fetal period induces lifelong split tolerance to grafts expressing these allo‐antigens. In adult mice, the production of extracellular vesicles (EVs) from maternal microchimeric cells causes cross‐decoration (XD) of offspring dendritic cells (DC) with NIMA and upregulation of PD‐L1, contributing to NIMA tolerance. To see how this may apply to humans, we tested NIMA acquisition by fetal DCS in human cord blood. The average percentage of NIMA‐XD among total DCs was 2.6% for myeloid and 4.5% for Plasmacytoid DC. These cells showed higher PD‐L1 expression than their non‐XD counterparts (mDC: p = .0016; pDC: p = .024). We detected CD9+ EVs bearing NIMA and PD‐L1 in cord blood. To determine if this immune regulatory mechanism persists beyond the pregnancy, we analyzed NIMA‐expressing kidney and liver transplant recipients. We found donor antigen XD DCs in peripheral blood and graft‐infiltrating DCs. As in cord blood, the pattern of donor antigen expression was punctate, and PD‐L1 expression was upregulated, likely due to both protein and miRNA acquired from EV. Our findings support a mechanism for split tolerance to NIMAs that develops during pregnancy and is recapitulated in adult transplant recipients.

[1]  S. Mastoridis,et al.  Impact of donor extracellular vesicle release on recipient cell “cross‐dressing” following clinical liver and kidney transplantation , 2020, American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons.

[2]  Deepali V. Sawant,et al.  Treg-Cell-Derived IL-35-Coated Extracellular Vesicles Promote Infectious Tolerance. , 2020, Cell reports.

[3]  D. Landsittel,et al.  Cross-dressed dendritic cells sustain effector T cell responses in islet and kidney allografts. , 2019, The Journal of clinical investigation.

[4]  Rohit Kohli,et al.  Immunologic benefit of maternal donors in pediatric living donor liver transplantation , 2019, Pediatric transplantation.

[5]  F. Claas,et al.  Determining the extent of maternal-foetal chimerism in cord blood , 2019, Scientific Reports.

[6]  M. Westgren,et al.  Cellular Subsets of Maternal Microchimerism in Umbilical Cord Blood , 2018, Cell transplantation.

[7]  D. Stolz,et al.  Graft‐infiltrating PD‐L1hi cross‐dressed dendritic cells regulate antidonor T cell responses in mouse liver transplant tolerance , 2018, Hepatology.

[8]  T. Mohanakumar,et al.  Zbtb7a induction in alveolar macrophages is implicated in anti-HLA–mediated lung allograft rejection , 2017, Science Translational Medicine.

[9]  G. Rice,et al.  Review: Fetal-maternal communication via extracellular vesicles - Implications for complications of pregnancies. , 2017, Placenta.

[10]  I. Stelzer,et al.  Immunological implications of pregnancy-induced microchimerism , 2017, Nature Reviews Immunology.

[11]  S. D. De Rosa,et al.  Maternal microchimerism is prevalent in cord blood in memory T cells and other cell subsets, and persists post-transplant , 2017, Oncoimmunology.

[12]  F. Claas,et al.  Modification of host dendritic cells by microchimerism-derived extracellular vesicles generates split tolerance , 2017, Proceedings of the National Academy of Sciences.

[13]  G. Saade,et al.  Feto-Maternal Trafficking of Exosomes in Murine Pregnancy Models , 2016, Front. Pharmacol..

[14]  Simon C Watkins,et al.  Donor dendritic cell-derived exosomes promote allograft-targeting immune response. , 2016, The Journal of clinical investigation.

[15]  J. Markmann,et al.  Donor exosomes rather than passenger leukocytes initiate alloreactive T cell responses after transplantation , 2016, Science Immunology.

[16]  A. Naqvi,et al.  miR-24, miR-30b, and miR-142-3p Regulate Phagocytosis in Myeloid Inflammatory Cells , 2015, The Journal of Immunology.

[17]  P. Dutta,et al.  Correlation between post transplant maternal microchimerism and tolerance across MHC barriers in mice , 2011, Chimerism.

[18]  Marielle Martin,et al.  Cells from a vanished twin as a source of microchimerism 40 years later in a male with a scleroderma-like condition. , 2010, Chimerism.

[19]  Massimo Spada,et al.  High Levels of Exosomes Expressing CD63 and Caveolin-1 in Plasma of Melanoma Patients , 2009, PloS one.

[20]  Jeff E. Mold,et al.  Maternal Alloantigens Promote the Development of Tolerogenic Fetal Regulatory T Cells in Utero , 2008, Science.

[21]  D. Furst,et al.  Microchimerism of maternal origin persists into adult life. , 1999, The Journal of clinical investigation.

[22]  Tzong-Hae Lee,et al.  Survival of donor leukocyte subpopulations in immunocompetent transfusion recipients: frequent long-term microchimerism in severe trauma patients. , 1999, Blood.

[23]  G. Opelz The effect of tolerance to noninherited maternal HLA antigens on the survival of renal transplants from sibling donors. , 1999, The New England journal of medicine.

[24]  H. V. van Houwelingen,et al.  Do noninherited maternal antigens (NIMA) enhance renal graft survival? , 1998, Transplant international : official journal of the European Society for Organ Transplantation.

[25]  P. Lingenfelter,et al.  Detection of maternal cells in human umbilical cord blood using fluorescence in situ hybridization. , 1995, Blood.

[26]  P. Medawar,et al.  ‘Actively Acquired Tolerance’ of Foreign Cells , 1953, Nature.

[27]  R. Owen IMMUNOGENETIC CONSEQUENCES OF VASCULAR ANASTOMOSES BETWEEN BOVINE TWINS. , 1945, Science.

[28]  E. Carosella,et al.  Search for maternal cells in human umbilical cord blood by polymerase chain reaction amplification of two minisatellite sequences. , 1994, Blood.