439
Views
8
CrossRef citations to date
0
Altmetric
Article Addendum

Maternal microchimerism in patients with biliary atresia

Implications for allograft tolerance

, &
Pages 37-39 | Received 25 Mar 2012, Accepted 26 Mar 2012, Published online: 01 Apr 2012

Abstract

Maternal-fetal cellular trafficking during pregnancy results in bidirectional microchimerism with potentially long-term consequences for the mother and her fetus. Exposure of the fetus to maternal cells results in tolerance to non-inherited maternal antigens (NIMA) and may therefore impact transplant outcomes. We investigated the rates of graft failure and retransplantation after parental liver transplantation in pediatric recipients with biliary atresia (BA), a disease with high levels of maternal microchimerism. We observed significantly lower rates of graft failure and retransplantation in BA recipients of maternal livers compared with BA recipients of paternal livers. Importantly, recipients without BA had equivalent transplant outcomes with maternal and paternal organs, suggesting that increased maternal microchimerism in BA patients may be the underlying etiology for tolerance. These results support the concept that prenatal exposure to NIMA may have consequences for living-related organ transplantation.

This article refers to:

Maternal-fetal cellular trafficking (MFCT) is the bidirectional passage of cells between a mother and her fetus, resulting in long-lived maternal cells in the fetusCitation1 and of fetal cells in the mother.Citation2 It has been reported that the presence of maternal cells in the fetus (maternal microchimerism) promotes the formation of regulatory T cells that suppress the fetal immune response to non-inherited maternal antigens (NIMA)Citation3 and is therefore an important component of maternal-fetal tolerance. It is possible that this tolerance may be long-lived and have an impact on the success of organ transplantation when the mother serves as the donor. While intriguing, the results in several transplantation settings have been variable. A beneficial effect of NIMA exposure has been reported in bone marrow transplantation,Citation4,Citation5 but results in the setting of kidney transplantation have been mixed.Citation6-Citation8

Biliary atresia (BA) is a disease characterized by inflammation of the biliary tree resulting in obliteration of the bile ducts and neonatal liver failure, often requiring liver transplantation. The observation that patients with BA have an increased number of maternal cells in their liversCitation9-Citation11 led us to hypothesize that increased maternal microchimerism may lead to improved long-term allograft tolerance in this setting. We examined a national database of living related liver transplants in children between the ages of 0–6 y over a 15-year period, and found improved graft survival specifically among BA patients who received a maternal liver compared with those that received a paternal liver.Citation12 The beneficial effect of maternal liver transplantation was only seen when the underlying disease was BA and not for other pediatric liver diseases, suggesting that maternal microchimerism may impact transplant outcomes. Our findings were also independent of female donor gender, since BA patients receiving organs from female deceased donors did not have different outcomes. We observed consistent results when examining graft rejection episodes from patients treated at our institution: BA recipients of maternal livers had a lower rate of refractory graft rejection compared with recipients of paternal livers, whereas this difference was not observed among non-BA recipients.Citation12 These results indicate that BA patients have differential tolerance to maternal compared with paternal antigens and may be useful in patient counseling.

An alternative explanation for our findings is that paternal livers are more easily rejected in BA patients. The etiology of BA is unknown, and it has been speculated that maternal cells contribute to disease pathogenesis, especially since the predominant maternal cell type that is elevated in livers of BA patients is cytotoxic CD8+ T cells.Citation13 One potential model, therefore, is that maternal cells may reject paternal antigens on hepatocytes to cause BA and would therefore also reject those antigens on a paternal liver graft. However, this model would be incomplete since our study showed that 90% of paternal livers are not rejected in BA patients. In addition, it was recently reported that a high proportion of pediatric recipients of parental living donor liver transplants (the majority of whom have BA) could undergo withdrawal of their immunosuppression without experiencing rejection.Citation14 Ultimately, functional assays of isolated maternal cells from BA patients will be necessary to understand the link between the pathogenesis of BA and maternal microchimerism.

An interesting question raised by our study is whether the tolerogenic effect of NIMA exposure diminishes over time. It is possible that maternal microchimerism is highest in younger patients and therefore the tolerogenic effect of NIMA exposure should be most apparent in this age group. If so, the fact that BA patients are younger than non-BA patients at the time of transplantation may be an important confounding factor in our results. To address this issue directly, we performed subgroup analysis in various age groups (0–1 y, 0–2 y, 0–6 y and 0–18 y) and consistently observed improved outcomes with maternal transplantation only in BA patients, supporting our conclusion that there is altered tolerance to maternal antigens specifically in this disease.

Although we did not directly measure maternal microchimerism levels in our study, the observation that there are improved outcomes with maternal liver transplantation only in BA patients suggests that increased levels of maternal microchimerism in this disease impact tolerance. Since maternal microchimerism must also be present in non-BA patients, albeit at lower levels, this result suggests that there is a threshold level of chimerism required for tolerance induction. This idea is supported by the observation that in NIMA-exposed animals, there is a positive correlation between the level of maternal microchimerism and the suppression of an anti-NIMA immune response.Citation15 Importantly, a threshold level of maternal microchimerism correlates with high levels of regulation in NIMA-exposed animals.Citation15 A similar threshold concept has been reported for tolerance induction after in utero transplantation of hematopoietic cells.Citation16 These observations support the crucial need for a dedicated study of the levels of maternal cells in BA patients and subsequent transplant outcomes.

Our results also raise broader questions regarding the function of MFCT in human disease. In addition to BA,Citation11,Citation13 increased maternal microchimerism has been identified in numerous pediatric diseases including type I diabetes,Citation17,Citation18 neonatal lupus-syndrome congenital heart block,Citation19 myopathies,Citation20 dermatomyositis,Citation21,Citation22 Hirschsprung diseaseCitation23 and pityriasis lichenoides.Citation24 It is not known whether maternal cells contribute to disease or proliferate in response to tissue injury in any of these diseases. We have also recently reported increased maternal microchimerism after open fetal surgery in patients with myelomeningocele compared with patients who had postnatal repair, suggesting increased recruitment or increased proliferation of maternal cells after fetal intervention.Citation25 These results are consistent with our finding of increased maternal cells after fetal intervention in miceCitation26 and we are currently exploring whether such alterations in microchimerism impact maternal-fetal tolerance during pregnancy.

The role of MFCT in maternal and fetal health remains a fascinating yet unanswered question. Our results support the idea that maternal microchimerism correlates with long-term tolerance in the setting of liver transplantation for biliary atresia. However, alterations in microchimerism may have beneficial or harmful consequences depending on the disease setting. A better understanding of the cellular mechanisms that lead to increased microchimerism and improved tolerance in BA will be crucial to achieve further advances in this field.

References

  • Maloney S, Smith A, Furst DE, Myerson D, Rupert K, Evans PC, et al. Microchimerism of maternal origin persists into adult life. J Clin Invest 1999; 104:41 - 7; http://dx.doi.org/10.1172/JCI6611; PMID: 10393697
  • Bianchi DW, Zickwolf GK, Weil GJ, Sylvester S, DeMaria MA. Male fetal progenitor cells persist in maternal blood for as long as 27 years postpartum. Proc Natl Acad Sci U S A 1996; 93:705 - 8; http://dx.doi.org/10.1073/pnas.93.2.705; PMID: 8570620
  • Mold JE, Michaëlsson J, Burt TD, Muench MO, Beckerman KP, Busch MP, et al. Maternal alloantigens promote the development of tolerogenic fetal regulatory T cells in utero. Science 2008; 322:1562 - 5; http://dx.doi.org/10.1126/science.1164511; PMID: 19056990
  • Stern M, Ruggeri L, Mancusi A, Bernardo ME, de Angelis C, Bucher C, et al. Survival after T cell-depleted haploidentical stem cell transplantation is improved using the mother as donor. Blood 2008; 112:2990 - 5; http://dx.doi.org/10.1182/blood-2008-01-135285; PMID: 18492955
  • van Rood JJ, Loberiza FR Jr., Zhang MJ, Oudshoorn M, Claas F, Cairo MS, 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 2002; 99:1572 - 7; http://dx.doi.org/10.1182/blood.V99.5.1572; PMID: 11861270
  • Burlingham WJ, Grailer AP, Heisey DM, Claas FH, Norman D, Mohanakumar T, et al. The effect of tolerance to noninherited maternal HLA antigens on the survival of renal transplants from sibling donors. N Engl J Med 1998; 339:1657 - 64; http://dx.doi.org/10.1056/NEJM199812033392302; PMID: 9834302
  • Miles CD, Schaubel DE, Liu D, Port FK, Rao PS. The role of donor-recipient relationship in long-term outcomes of living donor renal transplantation. Transplantation 2008; 85:1483 - 8; http://dx.doi.org/10.1097/TP.0b013e3181705a0f; PMID: 18497690
  • Neu AM, Stablein DM, Zachary A, Furth SL, Fivush BA. Effect of parental donor sex on rejection in pediatric renal transplantation: a report of the North American Pediatric Renal Transplant Cooperative Study. Pediatr Transplant 1998; 2:309 - 12; PMID: 10084735
  • Hayashida M, Nishimoto Y, Matsuura T, Takahashi Y, Kohashi K, Souzaki R, et al. The evidence of maternal microchimerism in biliary atresia using fluorescent in situ hybridization. J Pediatr Surg 2007; 42:2097 - 101; http://dx.doi.org/10.1016/j.jpedsurg.2007.08.039; PMID: 18082716
  • Kobayashi H, Tamatani T, Tamura T, Kusafuka J, Yamataka A, Lane GJ, et al. Maternal microchimerism in biliary atresia. J Pediatr Surg 2007; 42:987 - 91, discussion 991; http://dx.doi.org/10.1016/j.jpedsurg.2007.01.051; PMID: 17560207
  • Suskind DL, Rosenthal P, Heyman MB, Kong D, Magrane G, Baxter-Lowe LA, et al. Maternal microchimerism in the livers of patients with biliary atresia. BMC Gastroenterol 2004; 4:14; http://dx.doi.org/10.1186/1471-230X-4-14; PMID: 15285784
  • Nijagal A, Fleck S, Hills NK, Feng S, Tang Q, Kang SM, et al. Decreased risk of graft failure with maternal liver transplantation in patients with biliary atresia. Am J Transplant 2012; 12:409 - 19; http://dx.doi.org/10.1111/j.1600-6143.2011.03895.x; PMID: 22221561
  • Muraji T, Hosaka N, Irie N, Yoshida M, Imai Y, Tanaka K, et al. Maternal microchimerism in underlying pathogenesis of biliary atresia: quantification and phenotypes of maternal cells in the liver. Pediatrics 2008; 121:517 - 21; http://dx.doi.org/10.1542/peds.2007-0568; PMID: 18310200
  • Feng S, Ekong UD, Lobritto SJ, Demetris AJ, Roberts JP, Rosenthal P, et al. Complete immunosuppression withdrawal and subsequent allograft function among pediatric recipients of parental living donor liver transplants. JAMA 2012; 307:283 - 93; http://dx.doi.org/10.1001/jama.2011.2014; PMID: 22253395
  • Dutta P, Molitor-Dart M, Bobadilla JL, Roenneburg DA, Yan Z, Torrealba JR, et al. Microchimerism is strongly correlated with tolerance to noninherited maternal antigens in mice. Blood 2009; 114:3578 - 87; http://dx.doi.org/10.1182/blood-2009-03-213561; PMID: 19700665
  • Durkin ET, Jones KA, Rajesh D, Shaaban AF. Early chimerism threshold predicts sustained engraftment and NK-cell tolerance in prenatal allogeneic chimeras. Blood 2008; 112:5245 - 53; http://dx.doi.org/10.1182/blood-2007-12-128116; PMID: 18796629
  • Nelson JL, Gillespie KM, Lambert NC, Stevens AM, Loubiere LS, Rutledge JC, et al. Maternal microchimerism in peripheral blood in type 1 diabetes and pancreatic islet beta cell microchimerism. Proc Natl Acad Sci U S A 2007; 104:1637 - 42; http://dx.doi.org/10.1073/pnas.0606169104; PMID: 17244711
  • Vanzyl B, Planas R, Ye Y, Foulis A, de Krijger RR, Vives-Pi M, et al. Why are levels of maternal microchimerism higher in type 1 diabetes pancreas?. Chimerism 2010; 1:45 - 50; http://dx.doi.org/10.4161/chim.1.2.13891; PMID: 21327046
  • Stevens AM, Hermes HM, Rutledge JC, Buyon JP, Nelson JL. Myocardial-tissue-specific phenotype of maternal microchimerism in neonatal lupus congenital heart block. Lancet 2003; 362:1617 - 23; http://dx.doi.org/10.1016/S0140-6736(03)14795-2; PMID: 14630442
  • Artlett CM, Ramos R, Jiminez SA, Patterson K, Miller FW, Rider LG, Childhood Myositis Heterogeneity Collaborative Group. Chimeric cells of maternal origin in juvenile idiopathic inflammatory myopathies. Lancet 2000; 356:2155 - 6; http://dx.doi.org/10.1016/S0140-6736(00)03499-1; PMID: 11191545
  • Reed AM, McNallan K, Wettstein P, Vehe R, Ober C. Does HLA-dependent chimerism underlie the pathogenesis of juvenile dermatomyositis?. J Immunol 2004; 172:5041 - 6; PMID: 15067086
  • Reed AM, Picornell YJ, Harwood A, Kredich DW. Chimerism in children with juvenile dermatomyositis. Lancet 2000; 356:2156 - 7; http://dx.doi.org/10.1016/S0140-6736(00)03500-5; PMID: 11191546
  • Kiefer AS, Lang TR, Hein MS, McNallan KT, Moir CR, Reed AM. Maternal microchimerism in Hirschsprung’s disease. Am J Perinatol 2012; 29:71 - 8; http://dx.doi.org/10.1055/s-0031-1295645; PMID: 22105432
  • Khosrotehrani K, Guegan S, Fraitag S, Oster M, de Prost Y, Bodemer C, et al. Presence of chimeric maternally derived keratinocytes in cutaneous inflammatory diseases of children: the example of pityriasis lichenoides. J Invest Dermatol 2006; 126:345 - 8; http://dx.doi.org/10.1038/sj.jid.5700060; PMID: 16374466
  • Saadai P, Lee T, Bautista G, Gonzales KD, Nijagal A, Busch MP, et al. Alterations in maternal-fetal cellular trafficking after open fetal surgery. J Pediatr Surg 2012; 47:1089 - 94; PMID: 22703775
  • Nijagal A, Wegorzewska M, Jarvis E, Le T, Tang Q, MacKenzie TC. Maternal T cells limit engraftment after in utero hematopoietic cell transplantation in mice. J Clin Invest 2011; 121:582 - 92; http://dx.doi.org/10.1172/JCI44907; PMID: 21245575

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.