Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Jun 8.
Published in final edited form as: Am J Reprod Immunol. 2013 Feb 8;69(4):304–314. doi: 10.1111/aji.12075

Minor Histocompatibility Antigens and the Maternal Immune Response to the Fetus During Pregnancy

Caitlin Linscheid 1, Margaret G Petroff 1,2
PMCID: PMC4048750  NIHMSID: NIHMS585417  PMID: 23398025

Abstract

The tolerance of the semiallogeneic fetus by the maternal immune system is an important area of research for understanding how the maternal and fetal systems interact during pregnancy to ensure a successful outcome. Several lines of research reveal that the maternal immune system can recognize and respond to fetal minor histocompatibility antigens during pregnancy. Reactions to these antigens arise because of allelic differences between the mother and fetus, and have been shown more broadly to play an important role in mediating transplantation outcomes. This review outlines the discovery of minor histocompatibility antigens and their importance in solid organ and hematopoietic stem cell transplantations, maternal T-cell responses to minor histocompatibility antigens during pregnancy, expression of minor histocompatibility antigens in the human placenta, and the potential involvement of minor histocompatibility antigens in the development and manifestation of pregnancy complications.

Keywords: Minor histocompatibility antigens, pregnancy, placenta

Introduction

Pregnancy provides researchers with a unique, naturally-occurring immunological model wherein the maternal immune system tolerates the semiallogeneic fetus. In human pregnancy, this tolerance is achieved both actively, via the expression of immuno-modulatory molecules on the surface of the placenta, and passively, through the restricted expression of classical MHC molecules on trophoblast cells1. Despite these adaptations, the maternal immune system is not naïve to the fetus. Rather, there is a robust and growing body of evidence indicating that, in both mice and humans, the maternal immune system actively responds to fetal antigens26.

Immunogenicity of Minor Histocompatibility Antigens

Fetal antigens include both the major histocompatibility complex (MHC) and minor histocompatibility antigens (mHAgs)3, 7. MHC molecules are responsible for the presentation of foreign peptides to immune cells, and also are mediators of transplant rejection. Minor histocompatibility antigens are derived from functional proteins and can elicit an immune response due to allelic differences between individuals7, typically single nucleotide polymorphisms (SNPs), insertions, deletions or presence of the antigen on the Y-chromosome (Tables I&II)14,2022,73113.

Table I.

Y-Chromosome Encoded Minor Histocompatibility Antigens

Minor Antigen Gene HLA Peptide Expression
(mRNA)1
Ref.
A1/HY DFFRY A*0101 IVDCLTEMY Ubiquitous 73
A2/HY KDM5D A*0201 FIDSYICQV Ubiquitous 74
B52/HY RPS4Y1 B*5201 TIRYPDPVI Ubiquitous 75
B60/HY UTY B*60 RESEEESVSL Ubiquitous 76
B7/HY KDM5D B*0702 SPSVDKARAEL Ubiquitous 77
B8/HY UTY B*8 LPHNHTDL Ubiquitous 78
TMSB4Y/A33 TMSB4Y A*3303 EVLLRPGLHFR Ubiquitous 79
UTY139–147 UTY A*2402 YYNAFHWAI Ubiquitous 80
DQ5/HY DDX3Y DQB1*05 HIENFSDIDMGE Ubiquitous 81
DRB1/HY DDX3Y DRB1*1501 SKGRYIPPHLR Ubiquitous 82
DRB3/HY RPS4Y1 DRB3*0301 VIKVNDTVQI Ubiquitous 83
1

Tissue expression information was obtained from biogps.org as well as the listed references.

Table II.

Autosomally Encoded Minor Histocompatibility Antigens

Minor Antigen Gene HLA Peptide2 Expression
(mRNA)1
Ref.
ACC-1 BCL2A1 A*2402 DYLQYVLQI, DYLQCVLQI Restricted 20, 84, 85
ACC-2 BCL2A1 B*4403 KEFEDDIINW Restricted 20, 84, 85
ACC-4 Cathepsin H A*3101 ATLPLLCAR Restricted 86
ACC-5 Cathepsin H A*3303 WATLPLLCAR Restricted 86
ACC-6 HMSD B*4402, B*4403 MEIFIEVFSHF Restricted 87
CD19 CD19 A1*05, B1*02, DQ WEGEPPCLP Restricted 88
HA3 Lbc/AKAP13 A*0101 VTEPGTAQY Ubiquitous 89
HA1 HMHA1 A*0201, A*0206,
B*60, B*40012
VLHDDLLEA,
KECVLHDDL
Restricted 14, 2022
HA2 MYO1G A*0201 YIGEVLVSV Restricted 9092
HA8 KIAA0020 A*0201 RTLDKVLEV Ubiquitous 20, 93
HB-1 HMHB1 B*4402, B*4403 EEKRGSLHVW,
EEKRGSLYVW
Restricted 9496
HEATR1 HEATR1 B*0801 ISKERAEAL Ubiquitous 97
HER2_1170 HER2 A*0201 GCC Restricted 98
LB-ADIR-1F ADIR/TOR3A A*0201 SVAPALALFPA Restricted 99
LB-APOBEC3B-1K APOBEC3B B*0702 KPQYHAEMCFL,
KPQYHAEMCF,
KPQYHAEMC
Restricted 100
LB-ARHGDIB-1R ARHGDIB B*0702 LPRACWREA,
LPRACWREAR,
LPRACWREART
Restricted 100
LB-BCAT2-1R BCAT2 B*0702 QPRRALLFVIL, QPRRALLFVI Ubiquitous 100
LB-EBI3-1I EBI3 B*0702 RPRARYYIQVA,
RPRARYYIQV,
RPRARYYIQ,
AVRPRARYYI
Restricted 100
LB-ECGF1-1 ECGF1 B*0702 RPHAIRRPLAL Restricted 101
LB-ERAP1-1R ERAP1 B*0702 HPRQEQIALLA Ubiquitous 100
LB-GEMIN4-1V GEMIN4 B*0702 FPALRFVEV Restricted 100
LB-NUP133-1R NUP133 B*4001 SEDLILCRL Restricted 102
LB-PDCD11-1F PDCD11 B*0702 GPDSSKTFLCL,
GPDSSKTFL
Ubiquitous 100
LB-PRCP-1D PRCP A*0201 FMWDVAEDLKA,
FMWDVAEDL
Ubiquitous 100
LB-SON-1R SON B*4001 SETKQRTVL Restricted 102
LB-SSR1-1S SSR1 A*0201 SLAVAQDLT Restricted 100
LB-SWAP70-1Q SWAP70 B*4001 MEQLEQLEL Restricted 102
LB-TRIP10-1EPC TRIP10 B*4001 GEPQDLCTL Ubiquitous 102
LB-WNK1-1I WNK1 A*0201 TLSPEIITV Ubiquitous 100
LRH-1 P2RX5 B*0702 TPNQRQNVC Restricted 103–105
PANE1 (CTL-7A7) CENPM A*0301 RVWDLPGVLK Restricted 106
SLC1A5 SLC1A5 B*4002 AEATANGGLAL Restricted 20, 107
SP110 SP110 A*0301 SLPRGTSTPK Restricted 108
UGT2B17 UGT2B17 A*2902, B*4403 AELLNIPFLY Ubiquitous 109, 110
UGTB17 A*0206 CVATMIFMI Ubiquitous 107
UTA2-1 C12orf35 A*0201 QLLNSVLTL Restricted 111
LAMA1 LAMA1 DRB1*0301 LLILRAIPKGIRDKGAK Ubiquitous 112
LB-LY75-1K LY75 DRB1*1301 GITYRNKSLM Restricted 113
ZNF544 ZNF544 DRB1*0301 KQNSAFINDEKNGADGK Ubiquitous 112
1

Tissue expression information was obtained from biogps.org as well as the listed references.

2

Immunogenic amino acid differences are shown in bold and underlined. Peptides generated by alternative splicing are shown in bold. Peptides generated by a frameshift mutation are shown in italics. Peptides generated a translational termination codon are shown in italics and underlined. Peptides generated by a deletion are underlined.

In order for mHAgs to be recognized by T-cells, the antigens must be presented to T-cells in the context of specific MHC molecules, belonging to either Class I or II (Tables I&II)14,2022,73113. MHC Class II molecules bind to CD4+ T-cells and are primarily located on the surface of antigen presenting cells, most notably dendritic cells, macrophages and B cells. MHC Class I molecules bind to CD8+ T-cells and are present on the surface of most nucleated cells where they can present endogenous antigens and facilitate self vs. non-self discrimination by the immune system. MHC Class I is also critical for the process of cross-presentation whereby antigen presenting cells, typically dendritic cells, phagocytose exogenous material and process it for presentation on the MHC Class I molecule on the surface of the dendritic cell810. This allows for CD8+ T-cell recognition of antigens coming from tissues that do not express MHC Class I, including placental trophoblast1. Thus, placental debris containing fetal mHAgs could be released into the maternal blood stream, phagocytosed and processed by maternal dendritic cells and presented to maternal CD8+ T-cells, thus eliciting a maternal immune response to the fetus11. In addition to encountering and binding to the appropriate MHC for a specific peptide, responding T-cells must recognize the immunogenic peptide as foreign (i.e. non-self) and thus must come from an individual lacking the immunogenic peptide.

Histocompatibility Antigen 1 (HA1) is a widely studied mHAg that has been found on Hofbauer and trophoblast cells in the human placenta12, and has been shown to be important for bone marrow transplantation outcomes13. The antigenic peptide that arises from HA1 results from a single nucleotide difference between the non-immunogenic peptide (KECVLRDDLLEA) and the immunogenic peptide (KECVLHDDLLEA)14. The immunogenic peptide can be presented in the context of at least four different Class I MHCs, including HLA-A*0201 (Table II)14,2022,84113. As a result of the immunogenic SNP, the binding affinity of the HA1H peptide to the HLA-A*0201 peptide binding groove on antigen presenting cells (APCs) is increased15, thus leading to an immunogenic peptide that can be recognized by HLA-A*0201 restricted T-cells.

Recognition of the HA1 antigen can occur in the pathological situations of graft-versus-host disease and graft rejection, as well as in the physiological situation of pregnancy. In graft-versus-host disease, donor HLA-A*0201-restircted T-cells can recognize the immunogenic peptide in the antigen binding groove, thus eliciting an immune response targeting the recipients’ tissues13, 16. Graft rejection occurs when recipient HLA-A*0201-restricted T-cells respond to and target the immunogenic peptide on the graft itself17. In the case of pregnancy, maternal HLA-A*0201 restricted T-cells can recognize fetal immunogenic HA1, as evidenced by the presence of HA1-specific T-cells in maternal blood following pregnancy18. The source of fetal HA1 could be either fetal cells that cross the placenta and enter the maternal blood stream and organs (microchimerism), or cells and vesicles released from the placenta. In each of the above situations, the T cells responding to the antigen are both HLA-A*0201 restricted, and are derived from an individual lacking the immunogenic HA1H allele.

The probability of this interaction varies depending on the population frequency of both mHAg alleles and the MHC(s) restricting the immunogenic peptide. In the case of HA1, the population frequency of HA1H in a North American Caucasian population is approximately 44%19 and the likelihood that an individual will possess one of the four MHCs capable of presenting HA1R (A*0201, A*0206, B*60 or B*40012)14, 2022 is at least 48.1%23. Consequently, the possibility that two individuals will be discordant for HA1 and that the individual with the non-immunogenic form of HA1 will have the correct MHC to present the immunogenic peptide is approximately 11.9%:

  • P(HA1H) × P(HA1R) × P(MHC) = 44% × 56% × 48.1% = 11.9%

Thus, histoincompatibility between individuals is far from a rare event: given the large number of mHAgs discovered so far, it is very likely that for a given pregnancy, there will be at least one if not many fetal mHAgs that could be recognized by the mother’s immune system.

The Discovery of Minor Histocompatibility Antigens

The role of mHAgs in eliciting an immune response has been clearly demonstrated by transplantation studies. mHAgs were first discovered due to their role in modulating graft rejection and graft-versus-host disease in HLA-matched transplant recipients13, 24, 25. The first mHAg was discovered by Goulmy et. al. following the rejection of transplanted HLA-matched bone marrow cells from a male donor by a female recipient25. It was shown that cytotoxic T-lymphocytes (CTLs) isolated from the recipient’s blood had the capacity to lyse HLA-matched male cells, indicating that the target was located on the Y-chromosome26 and belonged to the HY family of mHAgs. Shortly after this discovery, the same group of investigators found that mHAgs could contribute to graft-versus-host disease, as donor CTLs can target and lyse recipient cells expressing a Y-chromosome-encoded antigen24.

Since these original discoveries, fifty unique mHAgs derived from forty-three genes have been found (Tables I & II)14,2022,73113. These mHAgs arise from SNPs, presence on the Y-chromosome, deletions, insertions, frameshift mutations, nonsense mutations and splice variants. mHAgs are encoded on the Y-chromosome (Table I)7383 and on many autosomes (Table II)14,2022,84113. Expression of some mHAgs is restricted to hematopoietic cells or a select group of tissues, whereas other mHAgs are expressed ubiquitously. Given the numerous potential HLA combinations as well as the vast number of SNPs present in the population, it is likely that many more mHAgs exist that have not yet been identified.

One of the major constraints on research involving mHAg-specific CD8+ T-cells is their relative scarcity in both peripheral blood and at their target sites. Research using multimeric MHC reagents (MHC multimers) has estimated the prevalence of HY-specific T-cells in peripheral blood following multiple pregnancies with male babies at 0.0001% to 0.03% of the total CD8+ T-cell population5. MHC multimers are complexes comprised of 2–10 or more linked peptide-MHC ligands that can bind T-cells through the T cell receptor in an antigen-specific manner27, 28. This allows for identification and quantification of T-cells specific for a particular antigen. However, in order to identify and characterize mHAg-specific T-cells, it is often necessary to expand the ex vivo population using cytokines and antigen, thus potentially altering the functionality of these cells both as a consequence of antigen/cytokine exposure and as a result of multimer-binding itself2931. Therefore, caution should be used when assessing the functional significance of these cells in vivo.

Minor Histocompatibility Antigens and Transplantation Outcomes

Immune responses to mHAgs in the context of transplantation can have both beneficial and detrimental consequences for the patient. mHAgs appear to be the primary mediator of graft-versus-host disease in HLA-matched transplantations13, 2426, leading to the need for increased immunosuppression as well as other negative health outcomes including dermatitis, kidney failure and even death. However, mHAg specific donor T-cells can play an important role in mediating graft-versus-leukemia effects or graft-versus-tumor effects32, 33. These effects can significantly prolong the lives of patients who receive transplantations to treat various types of cancers and can help ensure longer periods of disease-free survival.

Recently researchers have proposed that modulation of T-cells specific for mHAgs may provide a unique opportunity for augmenting graft-versus-leukemia and graft-versus-tumor effects34, 35, thus providing a potential avenue for increasing disease-free survival while subsequently reducing the need for immunosuppressive drugs. In this paradigm, donor T-cells specific for a particular mHAg whose expression is restricted to leukemic/tumor cells are isolated from the recipient’s blood following the initial transplant and expanded ex vivo34, 35. These T-cells are then re-infused into the recipient following a disease relapse. A preliminary clinical trial targeting the mHAg HA1 showed some success in treating disease relapse, although a high percentage of the participants experienced serious side effects35.

Minor Histocompatibility-Specific T-cells in Pregnancy

The role of mHAgs in pregnancy was first considered due to a finding by a number of researchers that parous female donors are more likely to elicit graft-versus-host disease in transplant recipients than non-parous or male donors3641. These researchers hypothesized that this was due to the formation of mHAg-specific T-cells in the mother during or immediately following pregnancy.

Studies in mice have found that CD4+ and CD8+ T-cells can develop in response to the endogenous fetal antigen, HY42. Other mouse studies have demonstrated that the presentation of fetal antigens to maternal T-cells can begin as early as copulation and that paternal antigens can be found in the seminal fluid11, 43. In women, a number of studies have found T-cells specific for at least three mHAgs, HA1, HA2 and HY, following pregnancy5, 18, 44. These T-cells have been found up to twenty-two years following delivery of the baby, suggesting that at least a small cohort of these cells can persist for long periods of time18. It is thought that during normal pregnancy these cells are prevented from attacking the placenta and fetus via numerous tolerogenic mechanisms, thus allowing for a successful pregnancy. A disruption of this tolerance could have significant effects on clinical outcome, as is evidenced by the links between mHAg expression, recognition by the maternal immune system and secondary recurrent miscarriage4547.

A recent study demonstrated that HY-specific CD8+ T-cells are elicited in maternal blood during human pregnancy with a male fetus6. These cells retained their proliferative capacity as well as their ability to lyse target cells and produce IFN-. The authors proposed that this indicates that fetus-specific T-cells are not completely deleted during pregnancy, as previously suggested3, 48, 49. As to why these T-cells do not cause the rejection of the fetus during normal pregnancies, there are at least three possibilities. The first is that the T-cells are incompletely activated during pregnancy, and thus lack effector function in vivo. The second hypothesis is that other cell types at the maternal-fetal interface, most notably regulatory CD4+ T-cells, prevent rejection of the fetus by promoting a tolerogenic environment as is indicated by the fact that regulatory T-cells are required for the success of allogeneic pregnancies in a mouse model50, 51. The third hypothesis is that the fetal-specific T-cells are unable to traffic to the maternal-fetal interface, and thus cannot mediate direct rejection of the fetus. This hypothesis is supported by recent work that showed an inability of maternal T-cells to traffic into the decidua during pregnancy as a result of epigenetic modifications leading to the loss of expression of specific chemokine genes by the decidual stromal cells52. Further to these mechanisms, the highly restricted expression of class I MHC molecules by placental trophoblast1 most likely renders antigen-specific T-cells wholly or largely unable to target the placenta directly.

Minor Histocompatibility Antigens are Expressed in the Placenta

There are at least two likely sources of fetal mHAgs during pregnancy: the placenta and fetal cells trafficking from the fetus into the maternal blood supply (microchimerism)5356. We have shown that at least six fetal mHAgs are expressed in human placental lysate, fetal cord blood and, most significantly, purified trophoblast cells. These findings provide strong evidence that the human placenta is one likely source of maternal immune exposure to fetal antigens during pregnancy (Table III)12. The close physical relationship between the syncytiotrophoblast, which covers the outer surface of the placenta, and the maternal blood supply, which surrounds the placenta, provides a likely avenue for maternal immune exposure to fetally-derived mHAgs57, 58. In addition to this close physical proximity, the syncytiotrophoblast undergoes a continual renewal process wherein the underlying cytotrophoblast fuses to give rise to the multinucleated syncytiotrophoblast and the excess, dead or damaged syncytiotrophoblast debris is released into the maternal blood space. This results in a large volume of fetally-derived placental debris being released into the maternal blood supply during pregnancy. In addition to the larger, shed debris, there is growing evidence that microvesicles/nanoparticles and exosomes are actively secreted from the surface of the placenta into the maternal blood space during pregnancy5961. The total volume of this deported material has been estimated at 1g/day for the term placenta. Most of this placental material is easily cleared by the maternal system during normal pregnancy, but large, multinucleated syncytiotrophoblast debris has been found in the lungs of women who died of eclampsia6265. Given the robust expression of fetal mHAgs in the syncytiotrophoblast and the large amount of syncytiotrophoblast debris that is released into the maternal system, it seems highly likely that the placenta serves as a source of fetal antigens during pregnancy.

Table III.

Minor Histocompatibility Antigens Expressed in the Human Placenta

Minor Antigen Gene Function Placental Expression3 Ref.
B52/HY RPS4Y1 Ribosomal Protein S4 STB, EVTs, Macrophages, CTBs 12, 75
DRB3/HY RPS4Y1 Ribosomal Protein S4 STB, EVTs, Macrophages, CTBs 12, 83
A2/HY KDM5D Protein containing zinc finger domains Whole placental lysate, fetal cord blood, CTBs (mRNA only) 12, 74
B7/HY KDM5D Protein containing zinc finger domains Whole placental lysate, fetal cord blood, CTBs (mRNA only) 12, 77
DQ5/HY DDX3Y RNA helicase, involved in spermatogenesis Whole placental lysate, fetal cord blood, CTBs (mRNA only) 12, 81
DRB1/HY DDX3Y RNA helicase, involved in spermatogenesis Whole placental lysate, fetal cord blood, CTBs (mRNA only) 12, 82
HA8 KIAA0020 Protein containing 6 PUF domains STBs, EVT, CTBs 12, 20, 93
HA1 HMHA1 GTPase activating protein (GAP) STB (first trimester only), EVTs, maternal and fetal leukocytes, macrophages, CTBs 12, 14, 2022
ACC-1 BCL2A1 Anti-apoptotic factor STB, EVTs, macrophages, CTBs 12, 84, 85
ACC-2 BCL2A1 Anti-apoptotic factor STB, EVTs, macrophages, CTBs 12, 84, 85
3

STB=syncytiotrophoblast, EVT=extravillous trophoblast, CTB=cytotrophoblast

Clinical Implications

Of particular interest is the role that the mHAgs expressed in syncytiotrophoblast debris may play in the development or manifestation of numerous pregnancy complications. Recent work by our lab has shown that expression of at least one mHAg, HA1, is upregulated in preeclamptic placentas as compared to normotensive control placentas (Linscheid C and Petroff MG, unpublished data). We hypothesize that this increase in HA1 expression may contribute to disruption of the overall immunologic balance by increasing the antigenic load encountered by the maternal immune system in the context of increased proinflammatory cytokine release, a feature of preeclampsia and other pregnancy complications66. Specifically, the increased release of placental debris that is characteristic of preeclampsia6265, 6769, compounded by the upregulation of HA1 expression in the deported syncytiotrophoblast, combined with an increase in inflammatory cytokines, most notably TNF-α and IL-666, 70, could alter the phenotype of both the dendritic cells that are presenting antigen to maternal T-cells as well as the T-cells themselves to promote anti-fetal immune responses, either during the current pregnancy or during subsequent pregnancies.

Dysfunction of the maternal immune system has been implicated in a number of other pregnancy complications, including secondary recurrent miscarriage. Epidemiologic evidence suggests that the recognition of fetal mHAgs may play an important role in secondary recurrent miscarriage, particularly if the preceding live birth was a male baby4547. Specifically, Christiansen et. al. have found that women who experience secondary recurrent miscarriage are more likely than the general population to possess the appropriate Class II MHC to present Y-chromosome-encoded mHAgs and that secondary recurrent miscarriage is more likely to occur in women who have previously given birth to a male baby45, 46, thus presenting the possibility that the development of secondary recurrent miscarriage is related to the generation of a maternal immune response specific for Y-chromosome-encoded antigens during the preceding, successful pregnancy. One theory regarding this phenomenon is that pregnancy complications late in the first pregnancy may disrupt the tolerogenic environment required for maintaining maternal immune tolerance towards the fetus and that this disruption contributes to the failure of subsequent pregnancies, especially those with a male fetus71. It is important to note that disruption of the tolerogenic environment and maternal T-cell recognition of both autosomal and Y-chromosome encoded fetal mHAgs may contribute to many cases of idiopathic infertility as well as numerous pregnancy complications.

A recent paper by Rowe et. al. (2012) found that in mice there is a substantial expansion of fetal-specific regulatory T-cells (T-regs) during primary and subsequent pregnancies72. The authors also found that resorption rates were significantly decreased in second pregnancies, due to a rapid expansion of a memory population of fetal-antigen specific T-regs that were formed during the first pregnancy. The authors propose that this mechanism may help explain why preeclampsia is more common in primiparous women as well as providing some insights as to why preeclampsia risk increases with interpregnancy interval. The role of these fetus-specific T-regs in secondary recurrent miscarriage is unclear, but it seems possible that a failed expansion of fetal-specific T-regs during the first pregnancy or a loss of fetal-specific T-regs between the first and second pregnancies could contribute to the manifestation of secondary recurrent miscarriage.

Conclusions and Future Directions

Pregnancy presents numerous challenges to the maternal immune system, which must simultaneously tolerate the semiallogeneic fetus and protect both the mother and fetus from potentially life-threatening infections. The mechanisms by which this is achieved are varied and include the recognition of fetal antigens by maternal T-cells. There are a number of studies to suggest that this process has important implications for both maternal and fetal health. Developing a better understanding of the cellular interactions that mediate this tolerance may contribute to both the development of more successful transplantation protocols as well as the prevention and/or treatment of common pregnancy complications.

Future work should seek to determine the role of the maternal environment in modulating fetus-specific T-cell responses. Specifically, understanding how increased inflammatory cytokines and the production of reactive oxygen species, both of which occur in preeclampsia, affect mHAg-specific T-cell responses to placental and fetal tissues could provide important insights into the manifestation of the clinical symptoms of preeclampsia and other pro-inflammatory pregnancy complications. In addition, work designed to better understand the persistence and function of fetus-specific T-cells following parturition could provide important insights into the role of mHAg-specific T-cells in determining transplant success or failure. By better understanding the development and function of mHAg-specific T-cells during and following pregnancy, we can generate insights important for understanding immune tolerance in general and, more specifically, how maternal tolerance of the fetus is achieved during normal pregnancy and what happens when this tolerance is disrupted.

References

  • 1.Petroff MG. Immune interactions at the maternal-fetal interface. J Reprod Immunol. 2005;68:1–13. doi: 10.1016/j.jri.2005.08.003. [DOI] [PubMed] [Google Scholar]
  • 2.Aluvihare VR, Kallikourdis M, Betz AG. Regulatory T cells mediate maternal tolerance to the fetus. Nat Immunol. 2004;5:266–271. doi: 10.1038/ni1037. [DOI] [PubMed] [Google Scholar]
  • 3.Tafuri A, Alferink J, Moller P, Hammerling GJ, Arnold B. T cell awareness of paternal alloantigens during pregnancy. Science. 1995;270:630–633. doi: 10.1126/science.270.5236.630. [DOI] [PubMed] [Google Scholar]
  • 4.Zhou M, Mellor AL. Expanded cohorts of maternal CD8+ T-cells specific for paternal MHC class I accumulate during pregnancy. J Reprod Immunol. 1998;40:47–62. doi: 10.1016/s0165-0378(98)00030-8. [DOI] [PubMed] [Google Scholar]
  • 5.Piper KP, McLarnon A, Arrazi J, Horlock C, Ainsworth J, Kilby MD, Martin WL, Moss PA. Functional HY-specific CD8+ T cells are found in a high proportion of women following pregnancy with a male fetus. Biol Reprod. 2007;76:96–101. doi: 10.1095/biolreprod.106.055426. [DOI] [PubMed] [Google Scholar]
  • 6.Lissauer D, Piper K, Goodyear O, Kilby MD, Moss PA. Fetal-specific CD8+ cytotoxic T cell responses develop during normal human pregnancy and exhibit broad functional capacity. J Immunol. 2012;189:1072–1080. doi: 10.4049/jimmunol.1200544. [DOI] [PubMed] [Google Scholar]
  • 7.Goulmy E. Minor histocompatibility antigens: from transplantation problems to therapy of cancer. Hum Immunol. 2006;67:433–438. doi: 10.1016/j.humimm.2006.03.012. [DOI] [PubMed] [Google Scholar]
  • 8.Bevan MJ. Minor H antigens introduced on H-2 different stimulating cells cross-react at the cytotoxic T cell level during in vivo priming. J Immunol. 1976;117:2233–2238. [PubMed] [Google Scholar]
  • 9.Bevan MJ. Cross-priming for a secondary cytotoxic response to minor H antigens with H-2 congenic cells which do not cross-react in the cytotoxic assay. J Exp Med. 1976;143:1283–1288. doi: 10.1084/jem.143.5.1283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Rodriguez A, Regnault A, Kleijmeer M, Ricciardi-Castagnoli P, Amigorena S. Selective transport of internalized antigens to the cytosol for MHC class I presentation in dendritic cells. Nat Cell Biol. 1999;1:362–368. doi: 10.1038/14058. [DOI] [PubMed] [Google Scholar]
  • 11.Erlebacher A, Vencato D, Price KA, Zhang D, Glimcher LH. Constraints in antigen presentation severely restrict T cell recognition of the allogeneic fetus. J Clin Invest. 2007;117:1399–1411. doi: 10.1172/JCI28214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Holland OJ, Linscheid C, Hodes HC, Nauser TL, Gilliam M, Stone P, Chamley LW, Petroff MG. Minor histocompatibility antigens are expressed in syncytiotrophoblast and trophoblast debris: implications for maternal alloreactivity to the fetus. Am J Pathol. 2012;180:256–266. doi: 10.1016/j.ajpath.2011.09.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Goulmy E, Schipper R, Pool J, Blokland E, Falkenburg JH, Vossen J, Gratwohl A, Vogelsang GB, van Houwelingen HC, van Rood JJ. Mismatches of minor histocompatibility antigens between HLA-identical donors and recipients and the development of graft-versus-host disease after bone marrow transplantation. N Engl J Med. 1996;334:281–285. doi: 10.1056/NEJM199602013340501. [DOI] [PubMed] [Google Scholar]
  • 14.den Haan JM, Meadows LM, Wang W, Pool J, Blokland E, Bishop TL, Reinhardus C, Shabanowitz J, Offringa R, Hunt DF, Engelhard VH, Goulmy E. The minor histocompatibility antigen HA-1: a diallelic gene with a single amino acid polymorphism. Science. 1998;279:1054–1057. doi: 10.1126/science.279.5353.1054. [DOI] [PubMed] [Google Scholar]
  • 15.Nicholls S, Piper KP, Mohammed F, Dafforn TR, Tenzer S, Salim M, Mahendra P, Craddock C, van Endert P, Schild H, Cobbold M, Engelhard VH, Moss PA, Willcox BE. Secondary anchor polymorphism in the HA-1 minor histocompatibility antigen critically affects MHC stability and TCR recognition. Proc Natl Acad Sci U S A. 2009;106:3889–3894. doi: 10.1073/pnas.0900411106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Mutis T, Gillespie G, Schrama E, Falkenburg JH, Moss P, Goulmy E. Tetrameric HLA class I-minor histocompatibility antigen peptide complexes demonstrate minor histocompatibility antigen-specific cytotoxic T lymphocytes in patients with graft-versus-host disease. Nat Med. 1999;5:839–842. doi: 10.1038/10563. [DOI] [PubMed] [Google Scholar]
  • 17.Krishnan NS, Higgins RM, Lam FT, Kashi H, Jobson S, Ramaiyan K, Rahman M, Morris A. HA-1 mismatch has significant effect in chronic allograft nephropathy in clinical renal transplantation. Transplant Proc. 2007;39:1439–1445. doi: 10.1016/j.transproceed.2007.02.066. [DOI] [PubMed] [Google Scholar]
  • 18.Verdijk RM, Kloosterman A, Pool J, van de Keur M, Naipal AM, van Halteren AG, Brand A, Mutis T, Goulmy E. Pregnancy induces minor histocompatibility antigen-specific cytotoxic T cells: implications for stem cell transplantation and immunotherapy. Blood. 2004;103:1961–1964. doi: 10.1182/blood-2003-05-1625. [DOI] [PubMed] [Google Scholar]
  • 19.Tseng LH, Lin MT, Martin PJ, Pei J, Smith AG, Hansen JA. Definition of the gene encoding the minor histocompatibility antigen HA-1 and typing for HA-1 from genomic DNA. Tissue Antigens. 1998;52:305–311. doi: 10.1111/j.1399-0039.1998.tb03052.x. [DOI] [PubMed] [Google Scholar]
  • 20.Bleakley M, Riddell SR. Exploiting T cells specific for human minor histocompatibility antigens for therapy of leukemia. Immunol Cell Biol. 2011;89:396–407. doi: 10.1038/icb.2010.124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Mommaas B, Kamp J, Drijfhout JW, Beekman N, Ossendorp F, Van Veelen P, Den Haan J, Goulmy E, Mutis T. Identification of a novel HLA-B60-restricted T cell epitope of the minor histocompatibility antigen HA-1 locus. J Immunol. 2002;169:3131–3136. doi: 10.4049/jimmunol.169.6.3131. [DOI] [PubMed] [Google Scholar]
  • 22.Torikai H, Akatsuka Y, Miyauchi H, Terakura S, Onizuka M, Tsujimura K, Miyamura K, Morishima Y, Kodera Y, Kuzushima K, Takahashi T. The HLA-A*0201-restricted minor histocompatibility antigen HA-1H peptide can also be presented by another HLA-A2 subtype, A*0206. Bone Marrow Transplant. 2007;40:165–174. doi: 10.1038/sj.bmt.1705689. [DOI] [PubMed] [Google Scholar]
  • 23.Gonzalez-Galarza FF, Christmas S, Middleton D, Jones AR. Allele frequency net: a database and online repository for immune gene frequencies in worldwide populations. Nucleic Acids Res. 2011;39:D913–D919. doi: 10.1093/nar/gkq1128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Goulmy E, Gratama JW, Blokland E, Zwaan FE, van Rood JJ. A minor transplantation antigen detected by MHC-restricted cytotoxic T lymphocytes during graft-versus-host disease. Nature. 1983;302:159–161. doi: 10.1038/302159a0. [DOI] [PubMed] [Google Scholar]
  • 25.Goulmy E, Termijtelen A, Bradley BA, van Rood JJ. Alloimmunity to human H-Y. Lancet. 1976;2:1206. doi: 10.1016/s0140-6736(76)91727-x. [DOI] [PubMed] [Google Scholar]
  • 26.Goulmy E, Termijtelen A, Bradley BA, van Rood JJ. Y-antigen killing by T cells of women is restricted by HLA. Nature. 1977;266:544–545. doi: 10.1038/266544a0. [DOI] [PubMed] [Google Scholar]
  • 27.Altman JD, Moss PA, Goulder PJ, Barouch DH, McHeyzer-Williams MG, Bell JI, McMichael AJ, Davis MM. Phenotypic analysis of antigen-specific T lymphocytes. Science. 1996;274:94–96. [PubMed] [Google Scholar]
  • 28.Batard P, Peterson DA, Devevre E, Guillaume P, Cerottini JC, Rimoldi D, Speiser DE, Winther L, Romero P. Dextramers: new generation of fluorescent MHC class I/peptide multimers for visualization of antigen-specific CD8+ T cells. J Immunol Methods. 2006;310:136–148. doi: 10.1016/j.jim.2006.01.006. [DOI] [PubMed] [Google Scholar]
  • 29.Chattopadhyay PK, Melenhorst JJ, Ladell K, Gostick E, Scheinberg P, Barrett AJ, Wooldridge L, Roederer M, Sewell AK, Price DA. Techniques to improve the direct ex vivo detection of low frequency antigen-specific CD8+ T cells with peptide-major histocompatibility complex class I tetramers. Cytometry A. 2008;73:1001–1009. doi: 10.1002/cyto.a.20642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Lyons GE, Roszkowski JJ, Man S, Yee C, Kast WM, Nishimura MI. T-cell receptor tetramer binding or the lack there of does not necessitate antigen reactivity in T-cell receptor transduced T cells. Cancer Immunol Immunother. 2006;55:1142–1150. doi: 10.1007/s00262-005-0103-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Laugel B, van den Berg HA, Gostick E, Cole DK, Wooldridge L, Boulter J, Milicic A, Price DA, Sewell AK. Different T cell receptor affinity thresholds and CD8 coreceptor dependence govern cytotoxic T lymphocyte activation and tetramer binding properties. J Biol Chem. 2007;282:23799–23810. doi: 10.1074/jbc.M700976200. [DOI] [PubMed] [Google Scholar]
  • 32.Marijt WA, Heemskerk MH, Kloosterboer FM, Goulmy E, Kester MG, van der Hoorn MA, van Luxemburg-Heys SA, Hoogeboom M, Mutis T, Drijfhout JW, van Rood JJ, Willemze R, Falkenburg JH. Hematopoiesis-restricted minor histocompatibility antigens HA-1- or HA-2-specific T cells can induce complete remissions of relapsed leukemia. Proc Natl Acad Sci U S A. 2003;100:2742–2747. doi: 10.1073/pnas.0530192100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Goulmy E. Minor histocompatibility antigens: allo target molecules for tumor-specific immunotherapy. Cancer J. 2004;10:1–7. doi: 10.1097/00130404-200401000-00001. [DOI] [PubMed] [Google Scholar]
  • 34.Kircher B, Stevanovic S, Urbanek M, Mitterschiffthaler A, Rammensee HG, Grunewald K, Gastl G, Nachbaur D. Induction of HA-1-specific cytotoxic T-cell clones parallels the therapeutic effect of donor lymphocyte infusion. Br J Haematol. 2002;117:935–939. doi: 10.1046/j.1365-2141.2002.03536.x. [DOI] [PubMed] [Google Scholar]
  • 35.Warren EH, Fujii N, Akatsuka Y, Chaney CN, Mito JK, Loeb KR, Gooley TA, Brown ML, Koo KK, Rosinski KV, Ogawa S, Matsubara A, Appelbaum FR, Riddell SR. Therapy of relapsed leukemia after allogeneic hematopoietic cell transplantation with T cells specific for minor histocompatibility antigens. Blood. 2010;115:3869–3878. doi: 10.1182/blood-2009-10-248997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Flowers ME, Pepe MS, Longton G, Doney KC, Monroe D, Witherspoon RP, Sullivan KM, Storb R. Previous donor pregnancy as a risk factor for acute graft-versus-host disease in patients with aplastic anaemia treated by allogeneic marrow transplantation. Br J Haematol. 1990;74:492–496. doi: 10.1111/j.1365-2141.1990.tb06340.x. [DOI] [PubMed] [Google Scholar]
  • 37.Gratwohl A, Hermans J, Niederwieser D, van Biezen A, van Houwelingen HC, Apperley J. Female donors influence transplant-related mortality and relapse incidence in male recipients of sibling blood and marrow transplants. Hematol J. 2001;2:363–370. doi: 10.1038/sj.thj.6200117. [DOI] [PubMed] [Google Scholar]
  • 38.Kollman C, Howe CW, Anasetti C, Antin JH, Davies SM, Filipovich AH, Hegland J, Kamani N, Kernan NA, King R, Ratanatharathorn V, Weisdorf D, Confer DL. Donor characteristics as risk factors in recipients after transplantation of bone marrow from unrelated donors: the effect of donor age. Blood. 2001;98:2043–2051. doi: 10.1182/blood.v98.7.2043. [DOI] [PubMed] [Google Scholar]
  • 39.Randolph SS, Gooley TA, Warren EH, Appelbaum FR, Riddell SR. Female donors contribute to a selective graft-versus-leukemia effect in male recipients of HLA-matched, related hematopoietic stem cell transplants. Blood. 2004;103:347–352. doi: 10.1182/blood-2003-07-2603. [DOI] [PubMed] [Google Scholar]
  • 40.Loren AW, Bunin GR, Boudreau C, Champlin RE, Cnaan A, Horowitz MM, Loberiza FR, Porter DL. Impact of donor and recipient sex and parity on outcomes of HLA-identical sibling allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2006;12:758–769. doi: 10.1016/j.bbmt.2006.03.015. [DOI] [PubMed] [Google Scholar]
  • 41.Takami A, Sugimori C, Feng X, Yachie A, Kondo Y, Nishimura R, Kuzushima K, Kotani T, Asakura H, Shiobara S, Nakao S. Expansion and activation of minor histocompatibility antigen HY-specific T cells associated with graft-versus-leukemia response. Bone Marrow Transplant. 2004;34:703–709. doi: 10.1038/sj.bmt.1704583. [DOI] [PubMed] [Google Scholar]
  • 42.James E, Chai JG, Dewchand H, Macchiarulo E, Dazzi F, Simpson E. Multiparity induces priming to male-specific minor histocompatibility antigen, HY, in mice and humans. Blood. 2003;102:388–393. doi: 10.1182/blood-2002-10-3170. [DOI] [PubMed] [Google Scholar]
  • 43.Moldenhauer LM, Diener KR, Thring DM, Brown MP, Hayball JD, Robertson SA. Cross-presentation of male seminal fluid antigens elicits T cell activation to initiate the female immune response to pregnancy. J Immunol. 2009;182:8080–8093. doi: 10.4049/jimmunol.0804018. [DOI] [PubMed] [Google Scholar]
  • 44.van Halteren AG, Jankowska-Gan E, Joosten A, Blokland E, Pool J, Brand A, Burlingham WJ, Goulmy E. Naturally acquired tolerance and sensitization to minor histocompatibility antigens in healthy family members. Blood. 2009;114:2263–2272. doi: 10.1182/blood-2009-01-200410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Nielsen HS, Steffensen R, Varming K, Van Halteren AG, Spierings E, Ryder LP, Goulmy E, Christiansen OB. Association of HY-restricting HLA class II alleles with pregnancy outcome in patients with recurrent miscarriage subsequent to a firstborn boy. Hum Mol Genet. 2009;18:1684–1691. doi: 10.1093/hmg/ddp077. [DOI] [PubMed] [Google Scholar]
  • 46.Christiansen OB, Steffensen R, Nielsen HS. The impact of anti-HY responses on outcome in current and subsequent pregnancies of patients with recurrent pregnancy losses. J Reprod Immunol. 2010;85:9–14. doi: 10.1016/j.jri.2009.12.008. [DOI] [PubMed] [Google Scholar]
  • 47.Ooi PV, Russell N, O'Donoghue K. Secondary recurrent miscarriage is associated with previous male birth. J Reprod Immunol. 2011;88:38–41. doi: 10.1016/j.jri.2010.10.004. [DOI] [PubMed] [Google Scholar]
  • 48.Jiang SP, Vacchio MS. Multiple mechanisms of peripheral T cell tolerance to the fetal "allograft". J Immunol. 1998;160:3086–3090. [PubMed] [Google Scholar]
  • 49.Vacchio MS, Hodes RJ. CD28 costimulation is required for in vivo induction of peripheral tolerance in CD8 T cells. J Exp Med. 2003;197:19–26. doi: 10.1084/jem.20021429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Kahn DA, Baltimore D. Pregnancy induces a fetal antigen-specific maternal T regulatory cell response that contributes to tolerance. Proc Natl Acad Sci U S A. 2010;107:9299–9304. doi: 10.1073/pnas.1003909107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Shima T, Sasaki Y, Itoh M, Nakashima A, Ishii N, Sugamura K, Saito S. Regulatory T cells are necessary for implantation and maintenance of early pregnancy but not late pregnancy in allogeneic mice. J Reprod Immunol. 2010;85:121–129. doi: 10.1016/j.jri.2010.02.006. [DOI] [PubMed] [Google Scholar]
  • 52.Nancy P, Tagliani E, Tay CS, Asp P, Levy DE, Erlebacher A. Chemokine gene silencing in decidual stromal cells limits T cell access to the maternal-fetal interface. Science. 2012;336:1317–1321. doi: 10.1126/science.1220030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Adams KM, Lambert NC, Heimfeld S, Tylee TS, Pang JM, Erickson TD, Nelson JL. Male DNA in female donor apheresis and CD34-enriched products. Blood. 2003;102:3845–3847. doi: 10.1182/blood-2003-05-1570. [DOI] [PubMed] [Google Scholar]
  • 54.Evans PC, Lambert N, Maloney S, Furst DE, Moore JM, Nelson JL. Long-term fetal microchimerism in peripheral blood mononuclear cell subsets in healthy women and women with scleroderma. Blood. 1999;93:2033–2037. [PubMed] [Google Scholar]
  • 55.Khostrotehrani K, Bianchi DW. Multi-lineage potential of fetal cells in maternal tissue: a legacy in reverse. J Cell Sci. 2005;118:1558–1563. doi: 10.1242/jcs.02332. [DOI] [PubMed] [Google Scholar]
  • 56.Nelson JL. Your cells are my cells. Sci Am. 2008;298:64–71. [PubMed] [Google Scholar]
  • 57.Petroff MG. Review: Fetal antigens--identity, origins, and influences on the maternal immune system. Placenta. 2011;32(Suppl 2):S176–S181. doi: 10.1016/j.placenta.2010.12.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Chamley LW, Chen Q, Ding J, Stone PR, Abumaree M. Trophoblast deportation: just a waste disposal system or antigen sharing? J Reprod Immunol. 2011;88:99–105. doi: 10.1016/j.jri.2011.01.002. [DOI] [PubMed] [Google Scholar]
  • 59.Taylor DD, Akyol S, Gercel-Taylor C. Pregnancy-associated exosomes and their modulation of T cell signaling. J Immunol. 2006;176:1534–1542. doi: 10.4049/jimmunol.176.3.1534. [DOI] [PubMed] [Google Scholar]
  • 60.Redman CW, Sargent IL. Microparticles and immunomodulation in pregnancy and pre-eclampsia. J Reprod Immunol. 2007;76:61–67. doi: 10.1016/j.jri.2007.03.008. [DOI] [PubMed] [Google Scholar]
  • 61.Burton GJ, Jones CJ. Syncytial knots, sprouts, apoptosis, and trophoblast deportation from the human placenta. Taiwan J Obstet Gynecol. 2009;48:28–37. doi: 10.1016/S1028-4559(09)60032-2. [DOI] [PubMed] [Google Scholar]
  • 62.Huppertz B, Frank HG, Kingdom JC, Reister F, Kaufmann P. Villous cytotrophoblast regulation of the syncytial apoptotic cascade in the human placenta. Histochem Cell Biol. 1998;110:495–508. doi: 10.1007/s004180050311. [DOI] [PubMed] [Google Scholar]
  • 63.Huppertz B, Kaufmann P, Kingdom J. Trophoblast turnover in health and disease. Fetal Maternal Med Rev. 2002;13:103–118. [Google Scholar]
  • 64.Attwood HD, Park WW. Embolism to the lungs by trophoblast. J Obstet Gynaecol Br Commonw. 1961;68:611–617. doi: 10.1111/j.1471-0528.1961.tb02778.x. [DOI] [PubMed] [Google Scholar]
  • 65.Ilke FA. Dissemination of syncytiotrophoblastic cells in the maternal blood stream during pregnancy. Bull Schweiz Akad Med. 1964;20:62–72. [PubMed] [Google Scholar]
  • 66.Kronborg CS, Gjedsted J, Vittinghus E, Hansen TK, Allen J, Knudsen UB. Longitudinal measurement of cytokines in pre-eclamptic and normotensive pregnancies. Acta Obstet Gynecol Scand. 2011;90:791–796. doi: 10.1111/j.1600-0412.2011.01134.x. [DOI] [PubMed] [Google Scholar]
  • 67.Chen Y, Huang Y, Jiang R, Teng Y. Syncytiotrophoblast-derived microparticle shedding in early-onset and late-onset severe pre-eclampsia. Int J Gynaecol Obstet. 2012;119:234–238. doi: 10.1016/j.ijgo.2012.07.010. [DOI] [PubMed] [Google Scholar]
  • 68.Goswami D, Tannetta DS, Magee LA, Fuchisawa A, Redman CW, Sargent IL, von Dadelszen P. Excess syncytiotrophoblast microparticle shedding is a feature of early-onset pre-eclampsia, but not normotensive intrauterine growth restriction. Placenta. 2006;27:56–61. doi: 10.1016/j.placenta.2004.11.007. [DOI] [PubMed] [Google Scholar]
  • 69.Knight M, Redman CW, Linton EA, Sargent IL. Shedding of syncytiotrophoblast microvilli into the maternal circulation in pre-eclamptic pregnancies. Br J Obstet Gynaecol. 1998;105:632–640. doi: 10.1111/j.1471-0528.1998.tb10178.x. [DOI] [PubMed] [Google Scholar]
  • 70.Lee SM, Romero R, Lee YJ, Park IS, Park CW, Yoon BH. Systemic inflammatory stimulation by microparticles derived from hypoxic trophoblast as a model for inflammatory response in preeclampsia. Am J Obstet Gynecol. 2012;207:337 e331–338. doi: 10.1016/j.ajog.2012.06.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Christiansen OB, Steffensen R, Nielsen HS. Anti-HY responses in pregnancy disorders. Am J Reprod Immunol. 2011;66(Suppl 1):93–100. doi: 10.1111/j.1600-0897.2011.01038.x. [DOI] [PubMed] [Google Scholar]
  • 72.Rowe JH, Ertelt JM, Xin L, Way SS. Pregnancy imprints regulatory memory that sustains anergy to fetal antigen. Nature. 2012;490:102–106. doi: 10.1038/nature11462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Pierce RA, Field ED, den Haan JM, Caldwell JA, White FM, Marto JA, Wang W, Frost LM, Blokland E, Reinhardus C, Shabanowitz J, Hunt DF, Goulmy E, Engelhard VH. Cutting edge: the HLA-A*0101-restricted HY minor histocompatibility antigen originates from DFFRY and contains a cysteinylated cysteine residue as identified by a novel mass spectrometric technique. J Immunol. 1999;163:6360–6364. [PubMed] [Google Scholar]
  • 74.Ofran Y, Kim HT, Brusic V, Blake L, Mandrell M, Wu CJ, Sarantopoulos S, Bellucci R, Keskin DB, Soiffer RJ, Antin JH, Ritz J. Diverse patterns of T-cell response against multiple newly identified human Y chromosome-encoded minor histocompatibility epitopes. Clin Cancer Res. 2010;16:1642–1651. doi: 10.1158/1078-0432.CCR-09-2701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Ivanov R, Aarts T, Hol S, Doornenbal A, Hagenbeek A, Petersen E, Ebeling S. Identification of a 40S ribosomal protein S4-derived H-Y epitope able to elicit a lymphoblast-specific cytotoxic T lymphocyte response. Clin Cancer Res. 2005;11:1694–1703. doi: 10.1158/1078-0432.CCR-04-1772. [DOI] [PubMed] [Google Scholar]
  • 76.Vogt MH, Goulmy E, Kloosterboer FM, Blokland E, de Paus RA, Willemze R, Falkenburg JH. UTY gene codes for an HLA-B60-restricted human male-specific minor histocompatibility antigen involved in stem cell graft rejection: characterization of the critical polymorphic amino acid residues for T-cell recognition. Blood. 2000;96:3126–3132. [PubMed] [Google Scholar]
  • 77.Wang W, Meadows LR, den Haan JM, Sherman NE, Chen Y, Blokland E, Shabanowitz J, Agulnik AI, Hendrickson RC, Bishop CE, Hunt DF, Goulmy E, Engelhard VH. Human H-Y: a male-specific histocompatibility antigen derived from the SMCY protein. Science. 1995;269:1588–1590. doi: 10.1126/science.7667640. [DOI] [PubMed] [Google Scholar]
  • 78.Warren EH, Gavin MA, Simpson E, Chandler P, Page DC, Disteche C, Stankey KA, Greenberg PD, Riddell SR. The human UTY gene encodes a novel HLA-B8-restricted H-Y antigen. J Immunol. 2000;164:2807–2814. doi: 10.4049/jimmunol.164.5.2807. [DOI] [PubMed] [Google Scholar]
  • 79.Torikai H, Akatsuka Y, Miyazaki M, Warren EH, 3rd, Oba T, Tsujimura K, Motoyoshi K, Morishima Y, Kodera Y, Kuzushima K, Takahashi T. A novel HLA-A*3303-restricted minor histocompatibility antigen encoded by an unconventional open reading frame of human TMSB4Y gene. J Immunol. 2004;173:7046–7054. doi: 10.4049/jimmunol.173.11.7046. [DOI] [PubMed] [Google Scholar]
  • 80.Mortensen BK, Rasmussen AH, Larsen ME, Larsen MV, Lund O, Braendstrup P, Harndahl M, Rasmussen M, Buus S, Stryhn A, Vindelov L. Identification of a novel UTY-encoded minor histocompatibility antigen. Scand J Immunol. 2012;76:141–150. doi: 10.1111/j.1365-3083.2012.02708.x. [DOI] [PubMed] [Google Scholar]
  • 81.Vogt MH, van den Muijsenberg JW, Goulmy E, Spierings E, Kluck P, Kester MG, van Soest RA, Drijfhout JW, Willemze R, Falkenburg JH. The DBY gene codes for an HLA-DQ5-restricted human male-specific minor histocompatibility antigen involved in graft-versus-host disease. Blood. 2002;99:3027–3032. doi: 10.1182/blood.v99.8.3027. [DOI] [PubMed] [Google Scholar]
  • 82.Zorn E, Miklos DB, Floyd BH, Mattes-Ritz A, Guo L, Soiffer RJ, Antin JH, Ritz J. Minor histocompatibility antigen DBY elicits a coordinated B and T cell response after allogeneic stem cell transplantation. J Exp Med. 2004;199:1133–1142. doi: 10.1084/jem.20031560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Spierings E, Vermeulen CJ, Vogt MH, Doerner LE, Falkenburg JH, Mutis T, Goulmy E. Identification of HLA class II-restricted H-Y-specific T-helper epitope evoking CD4+ T-helper cells in H-Y-mismatched transplantation. Lancet. 2003;362:610–615. doi: 10.1016/S0140-6736(03)14191-8. [DOI] [PubMed] [Google Scholar]
  • 84.Kawase T, Nannya Y, Torikai H, Yamamoto G, Onizuka M, Morishima S, Tsujimura K, Miyamura K, Kodera Y, Morishima Y, Takahashi T, Kuzushima K, Ogawa S, Akatsuka Y. Identification of human minor histocompatibility antigens based on genetic association with highly parallel genotyping of pooled DNA. Blood. 2008;111:3286–3294. doi: 10.1182/blood-2007-10-118950. [DOI] [PubMed] [Google Scholar]
  • 85.Akatsuka Y, Nishida T, Kondo E, Miyazaki M, Taji H, Iida H, Tsujimura K, Yazaki M, Naoe T, Morishima Y, Kodera Y, Kuzushima K, Takahashi T. Identification of a polymorphic gene, BCL2A1, encoding two novel hematopoietic lineage-specific minor histocompatibility antigens. J Exp Med. 2003;197:1489–1500. doi: 10.1084/jem.20021925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Torikai H, Akatsuka Y, Miyazaki M, Tsujimura A, Yatabe Y, Kawase T, Nakao Y, Tsujimura K, Motoyoshi K, Morishima Y, Kodera Y, Kuzushima K, Takahashi T. The human cathepsin H gene encodes two novel minor histocompatibility antigen epitopes restricted by HLA-A*3101 and -A*3303. Br J Haematol. 2006;134:406–416. doi: 10.1111/j.1365-2141.2006.06205.x. [DOI] [PubMed] [Google Scholar]
  • 87.Kawase T, Akatsuka Y, Torikai H, Morishima S, Oka A, Tsujimura A, Miyazaki M, Tsujimura K, Miyamura K, Ogawa S, Inoko H, Morishima Y, Kodera Y, Kuzushima K, Takahashi T. Alternative splicing due to an intronic SNP in HMSD generates a novel minor histocompatibility antigen. Blood. 2007;110:1055–1063. doi: 10.1182/blood-2007-02-075911. [DOI] [PubMed] [Google Scholar]
  • 88.Spaapen RM, Lokhorst HM, van den Oudenalder K, Otterud BE, Dolstra H, Leppert MF, Minnema MC, Bloem AC, Mutis T. Toward targeting B cell cancers with CD4+ CTLs: identification of a CD19-encoded minor histocompatibility antigen using a novel genome-wide analysis. J Exp Med. 2008;205:2863–2872. doi: 10.1084/jem.20080713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Spierings E, Brickner AG, Caldwell JA, Zegveld S, Tatsis N, Blokland E, Pool J, Pierce RA, Mollah S, Shabanowitz J, Eisenlohr LC, van Veelen P, Ossendorp F, Hunt DF, Goulmy E, Engelhard VH. The minor histocompatibility antigen HA-3 arises from differential proteasome-mediated cleavage of the lymphoid blast crisis (Lbc) oncoprotein. Blood. 2003;102:621–629. doi: 10.1182/blood-2003-01-0260. [DOI] [PubMed] [Google Scholar]
  • 90.de Bueger M, Bakker A, Van Rood JJ, Van der Woude F, Goulmy E. Tissue distribution of human minor histocompatibility antigens. Ubiquitous versus restricted tissue distribution indicates heterogeneity among human cytotoxic T lymphocyte-defined non-MHC antigens. J Immunol. 1992;149:1788–1794. [PubMed] [Google Scholar]
  • 91.den Haan JM, Sherman NE, Blokland E, Huczko E, Koning F, Drijfhout JW, Skipper J, Shabanowitz J, Hunt DF, Engelhard VH, Goulmy E. Identification of a graft versus host disease-associated human minor histocompatibility antigen. Science. 1995;268:1476–1480. doi: 10.1126/science.7539551. [DOI] [PubMed] [Google Scholar]
  • 92.Pierce RA, Field ED, Mutis T, Golovina TN, Von Kap-Herr C, Wilke M, Pool J, Shabanowitz J, Pettenati MJ, Eisenlohr LC, Hunt DF, Goulmy E, Engelhard VH. The HA-2 minor histocompatibility antigen is derived from a diallelic gene encoding a novel human class I myosin protein. J Immunol. 2001;167:3223–3230. doi: 10.4049/jimmunol.167.6.3223. [DOI] [PubMed] [Google Scholar]
  • 93.Brickner AG, Warren EH, Caldwell JA, Akatsuka Y, Golovina TN, Zarling AL, Shabanowitz J, Eisenlohr LC, Hunt DF, Engelhard VH, Riddell SR. The immunogenicity of a new human minor histocompatibility antigen results from differential antigen processing. J Exp Med. 2001;193:195–206. doi: 10.1084/jem.193.2.195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Dolstra H, Fredrix H, Preijers F, Goulmy E, Figdor CG, de Witte TM, van de Wiel-van Kemenade E. Recognition of a B cell leukemia-associated minor histocompatibility antigen by CTL. J Immunol. 1997;158:560–565. [PubMed] [Google Scholar]
  • 95.Dolstra H, Fredrix H, Maas F, Coulie PG, Brasseur F, Mensink E, Adema GJ, de Witte TM, Figdor CG, van de Wiel-van Kemenade E. A human minor histocompatibility antigen specific for B cell acute lymphoblastic leukemia. J Exp Med. 1999;189:301–308. doi: 10.1084/jem.189.2.301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Dolstra H, de Rijke B, Fredrix H, Balas A, Maas F, Scherpen F, Aviles MJ, Vicario JL, Beekman NJ, Ossendorp F, de Witte TM, van de Wiel-van Kemenade E. Bi-directional allelic recognition of the human minor histocompatibility antigen HB-1 by cytotoxic T lymphocytes. Eur J Immunol. 2002;32:2748–2758. doi: 10.1002/1521-4141(2002010)32:10<2748::AID-IMMU2748>3.0.CO;2-T. [DOI] [PubMed] [Google Scholar]
  • 97.Bleakley M, Otterud BE, Richardt JL, Mollerup AD, Hudecek M, Nishida T, Chaney CN, Warren EH, Leppert MF, Riddell SR. Leukemia-associated minor histocompatibility antigen discovery using T-cell clones isolated by in vitro stimulation of naive CD8+ T cells. Blood. 2010;115:4923–4933. doi: 10.1182/blood-2009-12-260539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Wenandy L, Kollgaard T, Letsch A, Andersen RS, Stather D, Seremet T, Svane IM, Vindelov L, Andersen MH, thor Straten P. The 1170 A–P single-nucleotide polymorphism (SNP) in the Her-2/neu protein (HER2) as a minor histocompatibility antigen (mHag) Leukemia. 2009;23:1926–1929. doi: 10.1038/leu.2009.112. [DOI] [PubMed] [Google Scholar]
  • 99.van Bergen CA, Kester MG, Jedema I, Heemskerk MH, van Luxemburg-Heijs SA, Kloosterboer FM, Marijt WA, de Ru AH, Schaafsma MR, Willemze R, van Veelen PA, Falkenburg JH. Multiple myeloma-reactive T cells recognize an activation-induced minor histocompatibility antigen encoded by the ATP-dependent interferon-responsive (ADIR) gene. Blood. 2007;109:4089–4096. doi: 10.1182/blood-2006-08-043935. [DOI] [PubMed] [Google Scholar]
  • 100.Van Bergen CA, Rutten CE, Van Der Meijden ED, Van Luxemburg-Heijs SA, Lurvink EG, Houwing-Duistermaat JJ, Kester MG, Mulder A, Willemze R, Falkenburg JH, Griffioen M. High-throughput characterization of 10 new minor histocompatibility antigens by whole genome association scanning. Cancer Res. 2010;70:9073–9083. doi: 10.1158/0008-5472.CAN-10-1832. [DOI] [PubMed] [Google Scholar]
  • 101.Slager EH, Honders MW, van der Meijden ED, van Luxemburg-Heijs SA, Kloosterboer FM, Kester MG, Jedema I, Marijt WA, Schaafsma MR, Willemze R, Falkenburg JH. Identification of the angiogenic endothelial-cell growth factor-1/thymidine phosphorylase as a potential target for immunotherapy of cancer. Blood. 2006;107:4954–4960. doi: 10.1182/blood-2005-09-3883. [DOI] [PubMed] [Google Scholar]
  • 102.Griffioen M, Honders MW, van der Meijden ED, van Luxemburg-Heijs SA, Lurvink EG, Kester MG, van Bergen CA, Falkenburg JH. Identification of 4 novel HLA-B*40:01 restricted minor histocompatibility antigens and their potential as targets for graft-versus-leukemia reactivity. Haematologica. 2012;97:1196–1204. doi: 10.3324/haematol.2011.049478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.de Rijke B, van Horssen-Zoetbrood A, Beekman JM, Otterud B, Maas F, Woestenenk R, Kester M, Leppert M, Schattenberg AV, de Witte T, van de Wiel-van Kemenade E, Dolstra H. A frameshift polymorphism in P2×5 elicits an allogeneic cytotoxic T lymphocyte response associated with remission of chronic myeloid leukemia. J Clin Invest. 2005;115:3506–3516. doi: 10.1172/JCI24832. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Overes IM, de Rijke B, van Horssen-Zoetbrood A, Fredrix H, de Graaf AO, Jansen JH, van Krieken JH, Raymakers RA, van der Voort R, de Witte TM, Dolstra H. Expression of P2×5 in lymphoid malignancies results in LRH-1-specific cytotoxic T-cell-mediated lysis. Br J Haematol. 2008;141:799–807. doi: 10.1111/j.1365-2141.2008.07125.x. [DOI] [PubMed] [Google Scholar]
  • 105.Norde WJ, Overes IM, Maas F, Fredrix H, Vos JC, Kester MG, van der Voort R, Jedema I, Falkenburg JH, Schattenberg AV, de Witte TM, Dolstra H. Myeloid leukemic progenitor cells can be specifically targeted by minor histocompatibility antigen LRH-1-reactive cytotoxic T cells. Blood. 2009;113:2312–2323. doi: 10.1182/blood-2008-04-153825. [DOI] [PubMed] [Google Scholar]
  • 106.Brickner AG, Evans AM, Mito JK, Xuereb SM, Feng X, Nishida T, Fairfull L, Ferrell RE, Foon KA, Hunt DF, Shabanowitz J, Engelhard VH, Riddell SR, Warren EH. The PANE1 gene encodes a novel human minor histocompatibility antigen that is selectively expressed in B-lymphoid cells and B-CLL. Blood. 2006;107:3779–3786. doi: 10.1182/blood-2005-08-3501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Kamei M, Nannya Y, Torikai H, Kawase T, Taura K, Inamoto Y, Takahashi T, Yazaki M, Morishima S, Tsujimura K, Miyamura K, Ito T, Togari H, Riddell SR, Kodera Y, Morishima Y, Kuzushima K, Ogawa S, Akatsuka Y. HapMap scanning of novel human minor histocompatibility antigens. Blood. 2009;113:5041–5048. doi: 10.1182/blood-2008-07-171678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Warren EH, Vigneron NJ, Gavin MA, Coulie PG, Stroobant V, Dalet A, Tykodi SS, Xuereb SM, Mito JK, Riddell SR, Van den Eynde BJ. An antigen produced by splicing of noncontiguous peptides in the reverse order. Science. 2006;313:1444–1447. doi: 10.1126/science.1130660. [DOI] [PubMed] [Google Scholar]
  • 109.Murata M, Warren EH, Riddell SR. A human minor histocompatibility antigen resulting from differential expression due to a gene deletion. J Exp Med. 2003;197:1279–1289. doi: 10.1084/jem.20030044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Terakura S, Murata M, Warren EH, Sette A, Sidney J, Naoe T, Riddell SR. A single minor histocompatibility antigen encoded by UGT2B17 and presented by human leukocyte antigen-A*2902 and -B*4403. Transplantation. 2007;83:1242–1248. doi: 10.1097/01.tp.0000259931.72622.d1. [DOI] [PubMed] [Google Scholar]
  • 111.Oostvogels R, Minnema MC, van Elk M, Spaapen RM, Te Raa GD, Giovannone B, Buijs A, van Baarle D, Kater AP, Griffioen M, Spierings E, Lokhorst HM, Mutis T. Towards effective and safe immunotherapy after allogeneic stem cell transplantation: identification of hematopoietic-specific minor histocompatibility antigen UTA2-1. Leukemia. 2012:1–8. doi: 10.1038/leu.2012.277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Stone B, Rieck M, Rawlings CA, Kas A, Shendure J, Jones H, Buckner JH. Identification of novel HLA class II target epitopes for generation of donor-specific T regulatory cells. Clin Immunol. 2012;145:153–160. doi: 10.1016/j.clim.2012.09.003. [DOI] [PubMed] [Google Scholar]
  • 113.Stumpf AN, van der Meijden ED, van Bergen CA, Willemze R, Falkenburg JH, Griffioen M. Identification of 4 new HLA-DR-restricted minor histocompatibility antigens as hematopoietic targets in antitumor immunity. Blood. 2009;114:3684–3692. doi: 10.1182/blood-2009-03-208017. [DOI] [PubMed] [Google Scholar]

RESOURCES