Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Immunological implications of pregnancy-induced microchimerism

Key Points

  • The benefits of viviparity in placental mammals require dedicated immunological adaptations in mothers and offspring to avert maternal–fetal conflict during pregnancy. Given the dominant role that reproductive fitness has in driving positive refining selection, adaptations that enforce fetal tolerance and promote maternal well-being are likely to be engrained in mammalian reproduction.

  • Expanded systemic immune tolerance occurs in mothers, and allows the widespread seeding and persistence of genetically foreign fetal microchimeric cells in maternal tissues during pregnancy and after parturition.

  • Genetically foreign maternal cells, which express non-inherited maternal antigens, are vertically transferred into offspring during pregnancy. These maternal microchimeric cells persist throughout postnatal development into adulthood, and sustain a persistent immunological tolerance to non-inherited maternal antigens in the offspring.

  • The bidirectional transfer of genetically foreign cells between mothers and their offspring during pregnancy is probably not accidental. Instead, microchimeric cells that express familially relevant traits are purposefully retained to promote genetic fitness by improving the outcome of future pregnancies.

  • Expanded immune tolerance to genetically foreign antigens expressed by microchimeric cells (the 'microchiome') extends how the immunological identity of individuals is defined beyond classical models of binary 'self' versus 'non-self' antigen discrimination to include an expanded repertoire of familially relevant 'extended-self' antigens.

  • Despite a uniform agreement on the existence of microchimeric cells, little is currently known about their cellular identity, molecular phenotype and interactions with the immune system. Further study of the effects of microchimeric cells may not only reveal new approaches for improving the outcomes of pregnancy, but also for developing innovative therapeutic solutions to other immunological problems such as autoimmunity and transplantation.

Abstract

Immunological identity is traditionally defined by genetically encoded antigens, with equal maternal and paternal contributions as a result of Mendelian inheritance. However, vertically transferred maternal cells also persist in individuals at very low levels throughout postnatal development. Reciprocally, mothers are seeded during pregnancy with genetically foreign fetal cells that persist long after parturition. Recent findings suggest that these microchimeric cells expressing non-inherited, familially relevant antigenic traits are not accidental 'souvenirs' of pregnancy, but are purposefully retained within mothers and their offspring to promote genetic fitness by improving the outcome of future pregnancies. In this Review, we discuss the immunological implications, benefits and potential consequences of individuals being constitutively chimeric with a biologically active 'microchiome' of genetically foreign cells.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Pregnancy imprints expanded immune tolerance in mothers and offspring.
Figure 2: Familial sources of microchimeric cells that establish the 'microchiome'.
Figure 3: Potential pathways by which maternal microchimeric cells seeded in fetal tissues may influence immune system development in offspring.

Similar content being viewed by others

References

  1. Medzhitov, R. & Janeway, C. A. Jr. How does the immune system distinguish self from nonself? Semin. Immunol. 12, 185–188 (2000).

    CAS  PubMed  Google Scholar 

  2. Paul, W. E. Self/nonself-immune recognition and signaling: a new journal tackles a problem at the center of immunological science. Self Nonself 1, 2–3 (2010).

    PubMed  PubMed Central  Google Scholar 

  3. Owen, R. D. Immunogenetic consequences of vascular anastomoses between bovine twins. Science 102, 400–401 (1945). This study provides a pioneering description of expanded immune tolerance primed by early developmental exposure to genetically foreign antigens.

    CAS  PubMed  Google Scholar 

  4. Medawar, P. B. Some immunological and endocrinological problems raised by the evolution of viviparity in vertebrates. Symp. Soc. Exp. Biol. 7, 320–338 (1953).

    Google Scholar 

  5. Erlebacher, A. Mechanisms of T cell tolerance towards the allogeneic fetus. Nat. Rev. Immunol. 13, 23–33 (2013).

    CAS  PubMed  Google Scholar 

  6. Erlebacher, A. Immunology of the maternal–fetal interface. Annu. Rev. Immunol. 31, 387–411 (2013).

    CAS  PubMed  Google Scholar 

  7. Robertson, S. A., Petroff, M. G. & Hunt, J. in Physiology of Reproduction Ch. 41 (eds Plant, T. M. & Zeleznik, A. J.) 1835–1874 (Academic Press, 2015).

    Google Scholar 

  8. Arck, P. C. & Hecher, K. Fetomaternal immune cross-talk and its consequences for maternal and offspring's health. Nat. Med. 19, 548–556 (2013).

    CAS  PubMed  Google Scholar 

  9. Rijnink, E. C. et al. Tissue microchimerism is increased during pregnancy: a human autopsy study. Mol. Hum. Reprod. 21, 857–864 (2015).

    CAS  PubMed  Google Scholar 

  10. Khosrotehrani, K., Johnson, K. L., Guegan, S., Stroh, H. & Bianchi, D. W. Natural history of fetal cell microchimerism during and following murine pregnancy. J. Reprod. Immunol. 66, 1–12 (2005).

    CAS  PubMed  Google Scholar 

  11. Jonsson, A. M., Uzunel, M., Gotherstrom, C., Papadogiannakis, N. & Westgren, M. Maternal microchimerism in human fetal tissues. Am. J. Obstet. Gynecol. 198, 325.e1–325.e6 (2008).

    Google Scholar 

  12. Mold, J. E. et al. Maternal alloantigens promote the development of tolerogenic fetal regulatory T cells in utero. Science 322, 1562–1565 (2008). This study provides evidence that fetal effector T cells are capable of alloreactivity, but are actively suppressed by fetal immune-suppressive T reg cells.

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Bianchi, D., Zickwolf, G., Weil, G., Sylvester, S. & DeMaria, M. Male fetal progenitor cells persist in maternal blood for as long as 27 years postpartum. Proc. Natl Acad. Sci. USA 93, 705–708 (1996). This study definitively demonstrates that genetically foreign male cells of presumed fetal origin can persist in mothers decades after parturition.

    CAS  PubMed  Google Scholar 

  14. Maloney, S. et al. Microchimerism of maternal origin persists into adult life. J. Clin. Invest. 104, 41–47 (1999). This study definitively demonstrates that maternal microchimeric cells persist in healthy offspring.

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Kinder, J. M. et al. Cross-generational reproductive fitness enforced by microchimeric maternal cells. Cell 162, 505–515 (2015). This study establishes the cross-generational reproductive benefits of maternal microchimeric cells retained in offspring by using tools for the selective in vivo depletion of these cells.

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Confavreux, C., Hutchinson, M., Hours, M. M., Cortinovis-Tourniaire, P. & Moreau, T. Rate of pregnancy-related relapse in multiple sclerosis. Pregnancy in Multiple Sclerosis Group. N. Engl. J. Med. 339, 285–291 (1998).

    CAS  PubMed  Google Scholar 

  17. Ostensen, M. & Villiger, P. M. The remission of rheumatoid arthritis during pregnancy. Semin. Immunopathol. 29, 185–191 (2007).

    PubMed  Google Scholar 

  18. Bischoff, A. L. et al. Altered response to A(H1N1)pnd09 vaccination in pregnant women: a single blinded randomized controlled trial. PLoS ONE 8, e56700 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Schlaudecker, E. P., McNeal, M. M., Dodd, C. N., Ranz, J. B. & Steinhoff, M. C. Pregnancy modifies the antibody response to trivalent influenza immunization. J. Infect. Dis. 206, 1670–1673 (2012).

    CAS  PubMed  Google Scholar 

  20. Herzenberg, L. A., Bianchi, D. W., Schroder, J., Cann, H. M. & Iverson, G. M. Fetal cells in the blood of pregnant women: detection and enrichment by fluorescence-activated cell sorting. Proc. Natl Acad. Sci. USA 76, 1453–1455 (1979).

    CAS  PubMed  Google Scholar 

  21. Ariga, H. et al. Kinetics of fetal cellular and cell-free DNA in the maternal circulation during and after pregnancy: implications for noninvasive prenatal diagnosis. Transfusion 41, 1524–1530 (2001).

    CAS  PubMed  Google Scholar 

  22. Krabchi, K. et al. Quantification of all fetal nucleated cells in maternal blood between the 18th and 22nd weeks of pregnancy using molecular cytogenetic techniques. Clin. Genet. 60, 145–150 (2001).

    CAS  PubMed  Google Scholar 

  23. Gammill, H. & Nelson, J. Naturally acquired microchimerism. Int. J. Dev. Biol. 54, 531–543 (2010).

    PubMed  PubMed Central  Google Scholar 

  24. Jimenez, D. F., Leapley, A. C., Lee, C. I., Ultsch, M. N. & Tarantal, A. F. Fetal CD34+ cells in the maternal circulation and long-term microchimerism in rhesus monkeys (Macaca mulatta). Transplantation 79, 142–146 (2005).

    PubMed  Google Scholar 

  25. Fujiki, Y., Johnson, K. L., Tighiouart, H., Peter, I. & Bianchi, D. W. Fetomaternal trafficking in the mouse increases as delivery approaches and is highest in the maternal lung. Biol. Reprod. 79, 841–848 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Jiang, T. T. et al. Regulatory T cells: new keys for further unlocking the enigma of fetal tolerance and pregnancy complications. J. Immunol. 192, 4949–4956 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Aluvihare, V., Kallikourdis, M. & Betz, A. Regulatory T cells mediate maternal tolerance to the fetus. Nat. Immunol. 5, 266–271 (2004).

    CAS  PubMed  Google Scholar 

  28. Rowe, J. H., Ertelt, J. M., Aguilera, M. N., Farrar, M. A. & Way, S. S. Foxp3+ regulatory T cell expansion required for sustaining pregnancy compromises host defense against prenatal bacterial pathogens. Cell Host Microbe 10, 54–64 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Bonney, E. A. & Brown, S. A. To drive or be driven: the path of a mouse model of recurrent pregnancy loss. Reproduction 147, R153–R167 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Rowe, J. H., Ertelt, J. M., Xin, L. & Way, S. S. Listeria monocytogenes cytoplasmic entry induces fetal wastage by disrupting maternal FoxP3+ regulatory cell-sustained fetal tolerance. PLoS Pathog. 8, e1002873 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Zenclussen, A. C. et al. Abnormal T-cell reactivity against paternal antigens in spontaneous abortion: adoptive transfer of pregnancy-induced CD4+CD25+ T regulatory cells prevents fetal rejection in a murine abortion model. Am. J. Pathol. 166, 811–822 (2005).

    PubMed  PubMed Central  Google Scholar 

  32. Kahn, D. & Baltimore, D. Pregnancy induces a fetal antigen-specific maternal T regulatory cell response that contributes to tolerance. Proc. Natl Acad. Sci. USA 107, 9299–9304 (2010).

    CAS  PubMed  Google Scholar 

  33. Chen, T. et al. Self-specific memory regulatory T cells protect embryos at implantation in mice. J. Immunol. 191, 2273–2281 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Feuerer, M. et al. Enhanced thymic selection of FoxP3+ regulatory T cells in the NOD mouse model of autoimmune diabetes. Proc. Natl Acad. Sci. USA 104, 18181–18186 (2007).

    CAS  PubMed  Google Scholar 

  35. Kuswanto, W. et al. Poor repair of skeletal muscle in aging mice reflects a defect in local, interleukin-33-dependent accumulation of regulatory T cells. Immunity 44, 355–367 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Rowe, J. H., Ertelt, J. M., Xin, L. & Way, S. S. Pregnancy imprints regulatory memory that sustains anergy to fetal antigen. Nature 490, 102–106 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Erlebacher, A., Vencato, D., Price, K., Zhang, D. & Glimcher, L. Constraints in antigen presentation severely restrict T cell recognition of allogeneic fetus. J. Clin. Invest. 117, 1399–1411 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Chaturvedi, V. et al. CXCR3 blockade protects against Listeria monocytogenes infection-induced fetal wastage. J. Clin. Invest. 125, 1713–1725 (2015).

    PubMed  PubMed Central  Google Scholar 

  39. Nancy, P. et al. Chemokine gene silencing in decidual stromal cells limits T cell access to maternal–fetal interface. Science 336, 1317–1321 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Xin, L. et al. Cutting edge: committed Th1 CD4+ T cell differentiation blocks pregnancy-induced Foxp3 expression with antigen-specific fetal loss. J. Immunol. 192, 2970–2974 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Samstein, R. M., Josefowicz, S. Z., Arvey, A., Treuting, P. M. & Rudensky, A. Y. Extrathymic generation of regulatory T cells in placental mammals mitigates maternal–fetal conflict. Cell 150, 29–38 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Mold, J. E. & McCune, J. M. Immunological tolerance during fetal development: from mouse to man. Adv. Immunol. 115, 73–111 (2012).

    CAS  PubMed  Google Scholar 

  43. Hall, J. M. et al. Detection of maternal cells in human umbilical cord blood using fluorescence in situ hybridization. Blood 86, 2829–2832 (1995).

    CAS  PubMed  Google Scholar 

  44. Stevens, A. M., Hermes, H. M., Kiefer, M. M., Rutledge, J. C. & Nelson, J. L. Chimeric maternal cells with tissue-specific antigen expression and morphology are common in infant tissues. Pediatr. Dev. Pathol. 12, 337–346 (2009).

    PubMed  PubMed Central  Google Scholar 

  45. Haynes, B. F. Phenotypic characterization and ontogeny of components of the human thymic microenvironment. Clin. Res. 32, 500–507 (1984).

    CAS  PubMed  Google Scholar 

  46. Andrassy, J. et al. Tolerance to noninherited maternal MHC antigens in mice. J. Immunol. 171, 5554–5561 (2003).

    CAS  PubMed  Google Scholar 

  47. Bakkour, S. et al. Analysis of maternal microchimerism in rhesus monkeys (Macaca mulatta) using real-time quantitative PCR amplification of MHC polymorphisms. Chimerism 5, 6–15 (2014).

    PubMed  PubMed Central  Google Scholar 

  48. Marleau, A. M., Greenwood, J. D., Wei, Q., Singh, B. & Croy, B. A. Chimerism of murine fetal bone marrow by maternal cells occurs in late gestation and persists into adulthood. Lab. Invest. 83, 673–681 (2003).

    PubMed  Google Scholar 

  49. Piotrowski, P. & Croy, B. A. Maternal cells are widely distributed in murine fetuses in utero. Biol. Reprod. 54, 1103–1110 (1996). This study reports a pioneering immunohistochemical analysis that shows the presence and widespread distribution of maternal microchimeric cells in fetal tissues.

    CAS  PubMed  Google Scholar 

  50. Owen, R. D., Wood, H. R., Foord, A. G., Sturgeon, P. & Baldwin, L. G. Evidence for actively acquired tolerance to Rh antigens. Proc. Natl Acad. Sci. USA 40, 420–424 (1954). This classical study shows that developmental exposure to genetically foreign maternal antigens confers long-lasting tolerance through reduced sensitization to the erythrocyte Rh antigen.

    CAS  PubMed  Google Scholar 

  51. Claas, F. H., Gijbels, Y., van der Velden- de Munck, J. & van Rood, J. J. Induction of B cell unresponsiveness to noninherited maternal HLA antigens during fetal life. Science 241, 1815–1817 (1988). This study shows that developmental exposure to genetically foreign non-inherited maternal HLA confers long-lasting functional tolerance in humans, as indicated by the diminished priming of HLA-specific antibodies.

    CAS  PubMed  Google Scholar 

  52. Burlingham, W. J. et al. The effect of tolerance to noninherited maternal HLA antigens on the survival of renal transplants from sibling donors. N. Engl. J. Med. 339, 1657–1664 (1998). This study shows that developmental exposure to genetically foreign non-inherited maternal HLA confers long-lasting functional tolerance in humans, as indicated by prolonged renal allograft survival.

    CAS  PubMed  Google Scholar 

  53. Ichinohe, T. et al. Feasibility of HLA-haploidentical hematopoietic stem cell transplantation between noninherited maternal antigen (NIMA)-mismatched family members linked with long-term fetomaternal microchimerism. Blood 104, 3821–3828 (2004).

    CAS  PubMed  Google Scholar 

  54. van Rood, J. J. et al. Effect of tolerance to noninherited maternal antigens on the occurrence of graft-versus-host disease after bone marrow transplantation from a parent or an HLA-haploidentical sibling. Blood 99, 1572–1577 (2002). This study shows that developmental exposure to genetically foreign non-inherited maternal HLA confers long-lasting functional tolerance in humans, as indicated by diminished rates of severe GVHD.

    PubMed  Google Scholar 

  55. Matsuoka, K. et al. Fetal tolerance to maternal antigens improves the outcome of allogeneic bone marrow transplantation by a CD4+ CD25+ T-cell-dependent mechanism. Blood 107, 404–409 (2006).

    CAS  PubMed  Google Scholar 

  56. Campbell, D. A. Jr et al. Breast feeding and maternal-donor renal allografts. Possibly the original donor-specific transfusion. Transplantation 37, 340–344 (1984).

    PubMed  Google Scholar 

  57. Molitor, M. L., Haynes, L. D., Jankowska-Gan, E., Mulder, A. & Burlingham, W. J. HLA class I noninherited maternal antigens in cord blood and breast milk. Hum. Immunol. 65, 231–239 (2004).

    CAS  PubMed  Google Scholar 

  58. Zhou, L. et al. Two independent pathways of maternal cell transmission to offspring: through placenta during pregnancy and by breast-feeding after birth. Immunology 101, 570–580 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Dutta, P. et al. Microchimerism is strongly correlated with tolerance to noninherited maternal antigens in mice. Blood 114, 3578–3587 (2009). This study shows that maternal microchimeric cells are widely distributed in the tissues of adult offspring, and that exposure to NIMAs during lactation is essential for persisting tolerance to maternal alloantigens.

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Stelzer, I. A., Thiele, K. & Solano, M. E. Maternal microchimerism: lessons learned from murine models. J. Reprod. Immunol. 108, 12–25 (2015).

    PubMed  Google Scholar 

  61. Molitor-Dart, M. L. et al. Developmental exposure to noninherited maternal antigens induces CD4+ T regulatory cells: relevance to mechanism of heart allograft tolerance. J. Immunol. 179, 6749–6761 (2007).

    CAS  PubMed  Google Scholar 

  62. Eikmans, M. et al. Naturally acquired microchimerism: implications for transplantation outcome and novel methodologies for detection. Chimerism 5, 24–39 (2014).

    PubMed  PubMed Central  Google Scholar 

  63. Nelson, J. L. The otherness of self: microchimerism in health and disease. Trends Immunol. 33, 421–427 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Axiak-Bechtel, S. M., Kumar, S. R., Hansen, S. A. & Bryan, J. N. Y-Chromosome DNA is present in the blood of female dogs suggesting the presence of fetal microchimerism. PLoS ONE 8, e68114 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Campbell, D., MacGillivray, I. & Carr-Hill, R. Pre-eclampsia in second pregnancy. Br. J. Obstet. Gynaecol. 92, 131–140 (1985).

    CAS  PubMed  Google Scholar 

  66. Li, D. K. & Wi, S. Changing paternity and the risk of preeclampsia/eclampsia in the subsequent pregnancy. Am. J. Epidemiol. 151, 57–62 (2000).

    CAS  PubMed  Google Scholar 

  67. Boddy, A. M., Fortunato, A., Wilson Sayres, M. & Aktipis, A. Fetal microchimerism and maternal health: a review and evolutionary analysis of cooperation and conflict beyond the womb. Bioessays 37, 1106–1118 (2015).

    PubMed  PubMed Central  Google Scholar 

  68. Haig, D. Does microchimerism mediate kin conflicts? Chimerism 5, 53–55 (2014).

    PubMed  PubMed Central  Google Scholar 

  69. Skjaerven, R., Wilcox, A. J. & Lie, R. T. The interval between pregnancies and the risk of preeclampsia. N. Engl. J. Med. 346, 33–38 (2002).

    PubMed  Google Scholar 

  70. Tandberg, A., Klungsoyr, K., Romundstad, L. B. & Skjaerven, R. Pre-eclampsia and assisted reproductive technologies: consequences of advanced maternal age, interbirth intervals, new partner and smoking habits. BJOG 122, 915–922 (2015).

    CAS  PubMed  Google Scholar 

  71. Masson, E. et al. Incidence and risk factors of anti-HLA immunization after pregnancy. Hum. Immunol. 74, 946–951 (2013).

    CAS  PubMed  Google Scholar 

  72. Vilches, M. & Nieto, A. Analysis of pregnancy-induced anti-HLA antibodies using Luminex platform. Transplant. Proc. 47, 2608–2610 (2015).

    CAS  PubMed  Google Scholar 

  73. Lynch, R. J. & Platt, J. L. Accommodation in organ transplantation. Curr. Opin. Organ. Transplant. 13, 165–170 (2008).

    PubMed  PubMed Central  Google Scholar 

  74. Morris, P. J. Suppression of rejection of organ allografts by alloantibody. Immunol. Rev. 49, 93–125 (1980).

    CAS  PubMed  Google Scholar 

  75. Maynard, C. L., Elson, C. O., Hatton, R. D. & Weaver, C. T. Reciprocal interactions of the intestinal microbiota and immune system. Nature 489, 231–241 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Rooks, M. G. & Garrett, W. S. Gut microbiota, metabolites and host immunity. Nat. Rev. Immunol. 16, 341–352 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Gammill, H. S., Guthrie, K. A., Aydelotte, T. M., Adams Waldorf, K. M. & Nelson, J. L. Effect of parity on fetal and maternal microchimerism: interaction of grafts within a host? Blood 116, 2706–2712 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Klein, S. L. & Flanagan, K. L. Sex differences in immune responses. Nat. Rev. Immunol. 16, 626–638 (2016).

    CAS  PubMed  Google Scholar 

  79. Muller, A. C. et al. Microchimerism of male origin in a cohort of Danish girls. Chimerism 6, 65–71 (2015).

    PubMed  Google Scholar 

  80. Bucher, C. et al. Role of primacy of birth in HLA-identical sibling transplantation. Blood 110, 468–469 (2007).

    CAS  PubMed  Google Scholar 

  81. Dobbelstein, C. et al. Birth order and transplantation outcome in HLA-identical sibling stem cell transplantation: an analysis on behalf of the Center for International Blood and Marrow Transplantation. Biol. Blood Marrow Transplant. 19, 741–745 (2013).

    PubMed  Google Scholar 

  82. Gratwohl, A. et al. Birth order and outcome after HLA-identical sibling donor transplantation. Blood 114, 5569–5570 (2009).

    CAS  PubMed  Google Scholar 

  83. Mancusi, A. et al. Haploidentical hematopoietic transplantation from KIR ligand-mismatched donors with activating KIRs reduces nonrelapse mortality. Blood 125, 3173–3182 (2015).

    CAS  PubMed  Google Scholar 

  84. Spalding, K. L., Bhardwaj, R. D., Buchholz, B. A., Druid, H. & Frisen, J. Retrospective birth dating of cells in humans. Cell 122, 133–143 (2005).

    CAS  PubMed  Google Scholar 

  85. Nelson, J. L. et al. Microchimerism and HLA-compatible relationships of pregnancy in scleroderma. Lancet 351, 559–562 (1998).

    CAS  PubMed  Google Scholar 

  86. Lambert, N. C. et al. Cutting edge: persistent fetal microchimerism in T lymphocytes is associated with HLA-DQA1*0501: implications in autoimmunity. J. Immunol. 164, 5545–5548 (2000).

    CAS  PubMed  Google Scholar 

  87. Ponsonby, A. L. et al. Offspring number, pregnancy, and risk of a first clinical demyelinating event: the AusImmune Study. Neurology 78, 867–874 (2012).

    PubMed  Google Scholar 

  88. Guthrie, K. A., Dugowson, C. E., Voigt, L. F., Koepsell, T. D. & Nelson, J. L. Does pregnancy provide vaccine-like protection against rheumatoid arthritis? Arthritis Rheum. 62, 1842–1848 (2010).

    PubMed  PubMed Central  Google Scholar 

  89. Hazes, J. M., Dijkmans, B. A., Vandenbroucke, J. P., de Vries, R. R. & Cats, A. Pregnancy and the risk of developing rheumatoid arthritis. Arthritis Rheum. 33, 1770–1775 (1990).

    CAS  PubMed  Google Scholar 

  90. Lambe, M., Bjornadal, L., Neregard, P., Nyren, O. & Cooper, G. S. Childbearing and the risk of scleroderma: a population-based study in Sweden. Am. J. Epidemiol. 159, 162–166 (2004).

    PubMed  Google Scholar 

  91. Masera, S. et al. Parity is associated with a longer time to reach irreversible disability milestones in women with multiple sclerosis. Mult. Scler. 21, 1291–1297 (2015).

    CAS  PubMed  Google Scholar 

  92. Pisa, F. E. et al. Reproductive factors and the risk of scleroderma: an Italian case-control study. Arthritis Rheum. 46, 451–456 (2002).

    PubMed  Google Scholar 

  93. Patas, K., Engler, J. B., Friese, M. A. & Gold, S. M. Pregnancy and multiple sclerosis: feto-maternal immune cross talk and its implications for disease activity. J. Reprod. Immunol. 97, 140–146 (2013).

    CAS  PubMed  Google Scholar 

  94. Straub, R. H., Buttgereit, F. & Cutolo, M. Benefit of pregnancy in inflammatory arthritis. Ann. Rheum. Dis. 64, 801–803 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Voskuhl, R. R. et al. Estriol combined with glatiramer acetate for women with relapsing-remitting multiple sclerosis: a randomised, placebo-controlled, phase 2 trial. Lancet Neurol. 15, 35–46 (2016).

    CAS  PubMed  Google Scholar 

  96. Engler, J. B. et al. Glucocorticoid receptor in T cells mediates protection from autoimmunity in pregnancy. Proc. Natl Acad. Sci. USA 114, E181–E190 (2017).

    CAS  PubMed  Google Scholar 

  97. Sunami, R., Komuro, M., Yuminamochi, T., Hoshi, K. & Hirata, S. Fetal cell microchimerism develops through the migration of fetus-derived cells to the maternal organs early after implantation. J. Reprod. Immunol. 84, 117–123 (2010).

    CAS  PubMed  Google Scholar 

  98. Mahmood, U. & O'Donoghue, K. Microchimeric fetal cells play a role in maternal wound healing after pregnancy. Chimerism 5, 40–52 (2014).

    PubMed  PubMed Central  Google Scholar 

  99. Kara, R. J. et al. Fetal cells traffic to injured maternal myocardium and undergo cardiac differentiation. Circ. Res. 110, 82–93 (2012).

    CAS  PubMed  Google Scholar 

  100. Roy, E. et al. Biphasic recruitment of microchimeric fetal mesenchymal cells in fibrosis following acute kidney injury. Kidney Int. 85, 600–610 (2014).

    CAS  PubMed  Google Scholar 

  101. Santos, M. A., O'Donoghue, K., Wyatt-Ashmead, J. & Fisk, N. M. Fetal cells in the maternal appendix: a marker of inflammation or fetal tissue repair? Hum. Reprod. 23, 2319–2325 (2008).

    PubMed  Google Scholar 

  102. Seppanen, E., Fisk, N. M. & Khosrotehrani, K. Pregnancy-acquired fetal progenitor cells. J. Reprod. Immunol. 97, 27–35 (2013).

    CAS  PubMed  Google Scholar 

  103. Zeng, X. X. et al. Pregnancy-associated progenitor cells differentiate and mature into neurons in the maternal brain. Stem Cells Dev. 19, 1819–1830 (2010).

    PubMed  Google Scholar 

  104. Nassar, D. et al. Fetal progenitor cells naturally transferred through pregnancy participate in inflammation and angiogenesis during wound healing. FASEB J. 26, 149–157 (2012).

    CAS  PubMed  Google Scholar 

  105. Nguyen Huu, S. et al. Maternal neoangiogenesis during pregnancy partly derives from fetal endothelial progenitor cells. Proc. Natl Acad. Sci. USA 104, 1871–1876 (2007).

    PubMed  Google Scholar 

  106. Roy, E. et al. Specific maternal microchimeric T cells targeting fetal antigens in β cells predispose to auto-immune diabetes in the child. J. Autoimmun. 36, 253–262 (2011).

    CAS  PubMed  Google Scholar 

  107. Leveque, L. et al. Selective organ specific inflammation in offspring harbouring microchimerism from strongly alloreactive mothers. J. Autoimmun. 50, 51–58 (2014).

    CAS  PubMed  Google Scholar 

  108. Nelson, J. L. et al. Maternal microchimerism in peripheral blood in type 1 diabetes and pancreatic islet β cell microchimerism. Proc. Natl Acad. Sci. USA 104, 1637–1642 (2007).

    CAS  PubMed  Google Scholar 

  109. Ye, J., Vives-Pi, M. & Gillespie, K. M. Maternal microchimerism: increased in the insulin positive compartment of type 1 diabetes pancreas but not in infiltrating immune cells or replicating islet cells. PLoS ONE 9, e86985 (2014).

    PubMed  PubMed Central  Google Scholar 

  110. Khosrotehrani, K. et al. Presence of chimeric maternally derived keratinocytes in cutaneous inflammatory diseases of children: the example of pityriasis lichenoides. J. Invest. Dermatol. 126, 345–348 (2006).

    CAS  PubMed  Google Scholar 

  111. Stevens, A. M., Hermes, H. M., Rutledge, J. C., Buyon, J. P. & Nelson, J. L. Myocardial-tissue-specific phenotype of maternal microchimerism in neonatal lupus congenital heart block. Lancet 362, 1617–1623 (2003).

    PubMed  Google Scholar 

  112. von Hoegen, P., Sarin, S. & Krowka, J. F. Deficiency in T cell responses of human fetal lymph node cells: a lack of accessory cells. Immunol. Cell Biol. 73, 353–361 (1995).

    CAS  PubMed  Google Scholar 

  113. Petit, T. et al. Detection of maternal cells in human fetal blood during the third trimester of pregnancy using allele-specific PCR amplification. Br. J. Haematol. 98, 767–771 (1997).

    CAS  PubMed  Google Scholar 

  114. Srivatsa, B., Srivatsa, S., Johnson, K. L. & Bianchi, D. W. Maternal cell microchimerism in newborn tissues. J. Pediatr. 142, 31–35 (2003).

    PubMed  Google Scholar 

  115. Touzot, F. et al. Massive expansion of maternal T cells in response to EBV infection in a patient with SCID-Xl. Blood 120, 1957–1959 (2012).

    CAS  PubMed  Google Scholar 

  116. Arvola, M. et al. Immunoglobulin-secreting cells of maternal origin can be detected in B cell-deficient mice. Biol. Reprod. 63, 1817–1824 (2000).

    CAS  PubMed  Google Scholar 

  117. Wrenshall, L. E., Stevens, E. T., Smith, D. R. & Miller, J. D. Maternal microchimerism leads to the presence of interleukin-2 in interleukin-2 knock out mice: implications for the role of interleukin-2 in thymic function. Cell. Immunol. 245, 80–90 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. von Mutius, E. The microbial environment and its influence on asthma prevention in early life. J. Allergy Clin. Immunol. 137, 680–689 (2016).

    PubMed  Google Scholar 

  119. Elahi, S. et al. Immunosuppressive CD71+ erythroid cells compromise neonatal host defence against infection. Nature 504, 158–162 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Tian, Y., Kuo, C. F., Akbari, O. & Ou, J. H. Hepatitis B virus persistence in offspring after vertical transmission is driven by macrophages that are altered by virus e antigen in mother. Immunity 44, 1204–1214 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Berry, S. M. et al. Association of maternal histocompatibility at class II HLA loci with maternal microchimerism in the fetus. Pediatr. Res. 56, 73–78 (2004).

    CAS  PubMed  Google Scholar 

  122. Kaplan, J. & Land, S. Influence of maternal–fetal histocompatibility and MHC zygosity on maternal microchimerism. J. Immunol. 174, 7123–7128 (2005).

    CAS  PubMed  Google Scholar 

  123. Wienecke, J. et al. Pro-inflammatory effector Th cells transmigrate through anti-inflammatory environments into the murine fetus. Placenta 33, 39–46 (2012).

    CAS  PubMed  Google Scholar 

  124. Nijagal, A. et al. Maternal T cells limit engraftment after in utero hematopoietic cell transplantation in mice. J. Clin. Invest. 121, 582–592 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Saadai, P. & MacKenzie, T. C. Increased maternal microchimerism after open fetal surgery. Chimerism 3, 1–3 (2012).

    PubMed  Google Scholar 

  126. Delassus, S. & Cumano, A. Circulation of hematopoietic progenitors in the mouse embryo. Immunity 4, 97–106 (1996).

    CAS  PubMed  Google Scholar 

  127. Mikkola, H. K. & Orkin, S. H. The journey of developing hematopoietic stem cells. Development 133, 3733–3744 (2006).

    CAS  PubMed  Google Scholar 

  128. Gibbons, D. et al. Interleukin-8 (CXCL8) production is a signatory T cell effector function of human newborn infants. Nat. Med. 20, 1206–1210 (2014).

    CAS  PubMed  Google Scholar 

  129. Kinder, J. M. et al. Tolerance to noninherited maternal antigens, reproductive microchimerism and regulatory T cell memory: 60 years after 'evidence for actively acquired tolerance to Rh antigens'. Chimerism 6, 8–20 (2015).

    PubMed  PubMed Central  Google Scholar 

  130. Stevens, A. M. Maternal microchimerism in health and disease. Best Pract. Res. Clin. Obstet. Gynaecol. 31, 121–130 (2016).

    PubMed  Google Scholar 

  131. Leveque, L. & Khosrotehrani, K. Feto-maternal allo-immunity, regulatory T cells and predisposition to auto-immunity: does it all start in utero? Chimerism 5, 59–62 (2014).

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The writing of this Review and reference to the authors' own work were made possible through funding by Cusanuswerk-Studienförderung (to I.A.S.); Deutsche Forschungsgemeinschaft (AR232/25-1 in KFO296 and AR232/27-1 to P.C.A.); the US National Institutes of Health, Office of the Director (DP1AI131080 to S.S.W.); the US National Institute of Allergy and Infectious Disease (R01AI100934 and R01AI120202 to S.S.W.); and the March of Dimes Foundation (FY15-254 to S.S.W.). S.S.W. is a Burroughs Wellcome Fund Investigator in the pathogenesis of infectious disease and is a Howard Hughes Medical Institute Faculty Scholar.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Sing Sing Way.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary information S1 (table)

Cellular phenotype of maternal and fetal microchimeric cells during pregnancy and after parturition (DOC 521 kb)

PowerPoint slides

Glossary

Immunological identity

The signature of distinct protein antigens that is encoded by the unique DNA of each individual, which includes MHC haplotype alleles and other alloantigens.

Viviparity

Development of offspring inside the body of the parent that results in the birth of live offspring capable of independent existence.

Fetal tolerance

The processes that allow fetal cells and tissues that express genetically foreign paternal antigens to avoid immune rejection and coexist in harmony inside expecting mothers during pregnancy.

Microchimeric cells

Rare cells found in one individual that originate from another individual and are genetically distinct from the host individual.

Non-inherited maternal antigens

(NIMAs). The half of genetically encoded maternal antigens that are not transmitted to an offspring by classical Mendelian inheritance.

Allogeneic pregnancies

Pregnancies that occur as the result of mating between individuals that are genetically distinct. For genetically identical, inbred animal strains, this refers to matings between unique male and female strains that have discordant MHC haplotypes, a setting that recapitulates the natural diversity of MHC alleles among individuals in outbred populations.

Peripherally induced Treg cells

CD4+ T cells that are induced to express forkhead box protein P3 and acquire immunosuppressive properties by cognate antigen stimulation in extrathymic peripheral tissues.

Maternal–fetal histocompatibility

The degree of similarity between genetically encoded MHC alleles in each mother–child pair.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kinder, J., Stelzer, I., Arck, P. et al. Immunological implications of pregnancy-induced microchimerism. Nat Rev Immunol 17, 483–494 (2017). https://doi.org/10.1038/nri.2017.38

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nri.2017.38

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing