Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Apr 4.
Published in final edited form as: Clin Chem. 2021 Jan 30;67(2):351–362. doi: 10.1093/clinchem/hvaa304

Forever Connected: The Lifelong Biological Consequences of Fetomaternal and Maternofetal Microchimerism

Diana W Bianchi a,*, Kiarash Khosrotehrani b, Sing Sing Way c, Tippi C MacKenzie d, Ingeborg Bajema e, Keelin O’Donoghue f
PMCID: PMC10072000  NIHMSID: NIHMS1885916  PMID: 33417673

Abstract

BACKGROUND:

Originally studied as a mechanism to understand eclampsia-related deaths during pregnancy, fetal cells in maternal blood have more recently garnered attention as a noninvasive source of fetal material for prenatal testing. In the 21st century, however, intact fetal cells have been largely supplanted by circulating cell-free placental DNA for aneuploidy screening. Instead, interest has pivoted to the ways in which fetal cells influence maternal biology. In parallel, an increasing appreciation of the consequences of maternal cells in the developing fetus has occurred.

CONTENT:

In this review, we highlight the potential clinical applications and functional consequences of the bidirectional trafficking of intact cells between a pregnant woman and her fetus. Fetal cells play a potential role in the pathogenesis of maternal disease and tissue repair. Maternal cells play an essential role in educating the fetal immune system and as a factor in transplant acceptance. Naturally occurring maternal microchimerism is also being explored as a source of hematopoietic stem cells for transplant in fetal hematopoietic disorders.

SUMMARY:

Future investigations in humans need to include complete pregnancy histories to understand maternal health and transplant success or failure. Animal models are useful to understand the mechanisms underlying fetal wound healing and/or repair associated with maternal injury and inflammation. The lifelong consequences of the exchange of cells between a mother and her child are profound and have many applications in development, health, and disease. This intricate exchange of genetically foreign cells creates a permanent connection that contributes to the survival of both individuals.


For well over a century it has been known in the medical literature that multi-nucleated syncytial giant cells could be demonstrated in the organs of women who died of eclampsia (1, 2). The renowned German pathologist, Christian Georg Schmorl, unexpectedly and consistently found them when performing autopsies on 17 women. While Schmorl’s focus was on understanding the pathophysiology of preeclampsia and eclampsia, his work established the foundation for a body of knowledge that has continuously evolved over time. In this review, we will discuss the potential clinical applications and functional consequences of bi-directional trafficking of intact cells between the pregnant woman and her fetus. Originally sought as a source of fetal material that could be noninvasively obtained for prenatal genetic diagnosis, the information presented here will highlight the potential role that fetal cells play in the pathogenesis of maternal disease and tissue repair, as well as the importance of maternal cells in educating the fetal immune system. This intricate interchange of genetically foreign cells creates a lifelong bond between a mother and her child that contributes to the survival of both individuals.

Fetomaternal Microchimerism

WHERE FETAL CELLS ARE FOUND IN THE MOTHER

Maternal blood.

Historically, the placenta was thought to be a barrier that separated the genetically distinct mother and fetus. It is now well established, however, that fetal cells pass into the maternal circulation during both human and rodent gestations (3, 4). Definitive proof of fetal cells in maternal blood occurred when XY metaphases in fetal lymphocytes were demonstrated in the peripheral blood of pregnant women carrying male fetuses (5). Even more convincing evidence came in the 1990s with the application of molecular genetic techniques such as polymerase chain reaction amplification and fluorescence in situ hybridization to detect unique amplified fetal DNA sequences from cellular components of the blood of pregnant women (4).

Fetal cells can be found in the peripheral blood of 100% of pregnant women and are detectable by 6 weeks of gestation, although the volume of fetal blood present in the maternal circulation is usually very small (6). Data from a review of fetomaternal hemorrhage show that the volume present in the maternal circulation at delivery is < 0.05 mL in 74% of women, < 1 mL in 96%, and < 30 mL in 99.67% (7). The principal mechanism is damage to placental villi with physical disruptions in the 5–45 mm barrier that separates the maternal and fetal circulations (Fig. 1). The number of fetal cells found in the maternal circulation is influenced by fetal and placental pathologies. Fetal cells can pass into maternal blood in significant amounts during or after spontaneous miscarriage (8, 9). Fetomaternal hemorrhage after first trimester termination of pregnancy results in an 80-fold increase in fetal cells in maternal blood (10). An increased number of fetal cells is also observed in maternal blood in cases of fetal aneuploidy, although this may reflect placental abnormalities rather than the underlying karyotype (11). More recently, cesarean delivery has been conclusively linked with higher detection and greater concentrations of microchimeric cells in maternal blood than vaginal delivery (12). Fetomaternal cell trafficking is increased in a range of common pregnancy problems, including hyperemesis gravidarum, preeclampsia, antepartum hemorrhage, and miscarriage (1319).

Fig. 1.

Fig. 1.

Male fetal chimeric cells in the uterus identified by in situ hybridization. (A) A single Y-chromosome positive cell in a healthy control myometrium; (B) A uterus with placenta increta. Large numbers of chimeric cells are present at sites of placental invasion (arrow); arrowheads show individual male cells. Figure courtesy of Marije Koopmans and Idske Kremer Hovinga.

Estimates during typical pregnancies suggest there are only 1 to 2 fetal cells per mL of maternal blood (11), with trophoblasts, monocytes, B and T lymphocytes, nucleated erythrocytes, and hematopoietic progenitors present (Table 1). In practice, stimulating fetal erythroid and hematopoietic progenitor cells in culture to provide a greater number for genetic analysis proved disappointing, because fetal and maternal cells were too similar, and amplification was unsuccessful (20).

Table 1.

Types of fetal cells found in maternal blood.

Trophoblasts Leukocytes NRBCs HSCs MSCs PAPCs
Specific cell markers No, although HLA-G has potential Yes Yes, improving with use of embryonic hemoglobins Yes Yes, although defined by absence of markers CD34+CD38+Endothelial
Similarity to maternal cells None Yes Yes, with exception of primitive fetal NRBCs Yes, but have higher proliferative potential None in blood Not known
Found in non-pregnant adult circulation No, cleared by pulmonary circulation Yes Yes Not typically No No
Persist in maternal tissues No Yes No, short-lived Yes Yes Yes
Other qualities Developmentally end-stage,
Multinucleated
Identified by HLA differences between mother and fetus Physiologic increase in maternal NRBCs occurs during pregnancy Rare, but can be amplified in vitro Rare, but can be amplified in vitro, may escape immune rejection.
Likely to adhere and engraft rapidly
Have capability of differentiation to repair maternal tissues

Abbreviations: NRBCs, nucleated erythrocytes; HSCs, hematopoietic stem cells; MSCs, mesenchymal stem cells; PAPCs, pregnancy-associated progenitor cells; HLA, human leukocyte antigen.

The identification of mesenchymal stem cells (MSC) in first trimester fetal blood (21) offered the prospect of targeting a unique fetal stem cell for noninvasive prenatal diagnosis. Fetal MSC were, however, found to circulate at a very low frequency in maternal blood (in 5% of post termination blood samples), making them unlikely to be useful clinically (22).

Subsequent work found that fetal CD34+ cells could be detected in the maternal intervillous blood space from term placental chorion, suggesting that their point of origin resided in the fetal villi; these cells were not hematopoietic but had endothelial lineage characteristics (23). Failure to routinely isolate the stem/progenitor cells in maternal blood was eventually and variously attributed to their low frequency, to limitations of the sensitivity of enrichment methods, and/or to their engraftment in maternal tissues soon after their transplacental passage (19, 24, 25). More recently, however, researchers identified, isolated and cultured fetal microchimeric stem cells from a pluripotent maternal stem cell niche 24–26 years after the delivery of a 45-year old woman’s sons (26). These cells expressed genes associated with a pluripotent stem cell phenotype and were able to be differentiated into osteocytes, adipocytes, and chondrocytes.

Maternal organs.

Fetomaternal microchimerism is defined as low levels of intact fetal cells that persist in maternal blood and tissues for years after pregnancy (17, 19, 27, 28). Microchimerism is proposed as a state of balance between host versus graft and graft versus host reactions, leading to the acceptance of the allogenic fetus; it has been specifically suggested that fetal stem cells engraft in maternal bone marrow to maintain tolerance to the semi-allogeneic fetoplacental graft (29). Bianchi et al. (24) were the first to demonstrate that fetal cells could persist in maternal blood up to 27 years postpartum and later confirmed that a live birth was not required for a woman to become a chimera (10, 30, 31).

To understand more about the significance and pathophysiological role of microchimeric cells, the ‘where, when, and how’ is of extreme importance. The search for microchimerism can make use of several sources, such as normal or diseased tissues; the timepoints can be during health or disease, during reproductive ages or beyond, during pregnancy or not. Techniques to detect chimeric cells in tissues usually rely on the detection of Y-chromosome positive cells in women (Fig. 2), sometimes in combination with immunohistochemistry to detect the true nature of the cells, but this technique limits the focus to women who have had male fetuses. Fetal cell microchimerism occurs equally in both fetal sexes. When studying chimeric cells in tissues, a distinction can be made between tissue samples obtained during life or at autopsy. The latter provides an adequate amount of tissue for study, but usually suffers from degenerative changes. Studies on the presence and location of chimeric cells in tissues from women obtained at autopsy have shown that Y-chromosome positive chimeric cells were present in kidneys, livers, hearts, thyroids, lungs, skins, and lymph nodes unaffected by disease (Fig. 3) (32, 33). It is evident that a low level of ‘background’ chimerism is physiologic and that the number of chimeric cells increases in disease. Whether the role of chimeric cells lies mainly in the development of disease or in its repair phase is still an unresolved issue (34).

Fig. 2.

Fig. 2.

Microchimeric male fetal cells present in the appendix of a pregnant woman. Fluorescence in situ hybridization was performed with X and Y chromosomes labeled with SpectrumOrange and SpectrumGreen, respectively. A single male cell is seen in the center of the image.

Fig. 3.

Fig. 3.

Bidirectional trafficking of fetal and maternal cells during pregnancy. Fetal cells (red) become incorporated into maternal organs shown on the left. Maternal cells (blue) are found in fetal organs on the right. They play an important role in education of the fetal immune system.

Fetomaternal chimerism and maternal disease.

Microchimerism seems to be more common in affected tissues than in blood (35), in which the number of microchimeric cells is greater than in controls (31). Studies have confirmed that the increase in fetal microchimerism in autoimmune disease is derived from circulating cells and not from cell-free DNA. Fetomaternal microchimerism has been most strongly implicated in the pathogenesis of autoimmune diseases, which have a predilection for women after pregnancy (36) and clinically resemble graft versus host disease (28, 31, 37). Candidate autoimmune diseases with higher prevalence in women include systemic sclerosis (35, 3840), Sjogren’s syndrome (41), thyroid disorders (42), lupus nephritis (34), and systemic lupus erythematosus (43). Microchimerism has now been investigated in many of these diseases, with fetal cells demonstrated in skin lesions, tissues, and peripheral blood, and some results supporting a potential role in disease pathogenesis. Some authors suggest that an explanation for the conflicting results in studies relating fetal microchimerism and autoimmune disease is the migration of fetal cells preferentially into target organs of the disease rather than the circulation (41).

Both microchimeric cells and human leukocyte antigen relationships of host and non-host cells are most likely involved in the subsequent pathogenesis of autoimmune disease (37, 39). The finding of more male cells in women with older sons, as well as the increased amount of microchimerism remote from systemic lupus erythematosus diagnosis and pregnancy, lends support to the idea that it takes time for the fetal cells to repopulate and establish a cell line (43, 44).

During pregnancy, microchimeric fetal cells have been proven to invade maternal skin, and their presence has been associated with the development of otherwise unexplained inflammatory skin disorders (45). Melanomas that occur during pregnancy have also been shown to include microchimeric fetal cells that cluster around the tumor and appear to have mainly adopted an endothelial phenotype (46). Identification of male–presumed fetal cells in healed maternal cesarean scars after pregnancy suggests that, possibly in response to signals produced by maternal skin injury at surgery, fetal cells migrate to the site of damage to become involved in maternal tissue repair, or proliferate locally (47).

Research has also shown that fetal cells are present in maternal organs affected by non-autoimmune conditions, such as hepatitis C (30), cervical cancer (48), and breast cancer (49). Most investigations that have examined solid tumors convincingly show that fetal cells are preferentially present at tumor sites (28, 50). The function of these cells is unknown, but hypotheses proposed include promotion of tumorigenesis, immunosurveillance, and participation in tissue repair. It cannot be concluded that fetal cell microchimerism always results in a graft-versus-host phenomenon (27).

Furthermore, fetal microchimerism is not always found in association with maternal disease. Because fetomaternal cell trafficking occurs in all pregnancies, microchimerism is likely established in healthy women as well (19, 31, 51). If fetal progenitor/stem cell trafficking occurs in every pregnancy, a much greater incidence of autoimmune disease in postreproductive women would be expected. Therefore, differentiation of fetal stem/progenitor cells engrafted in maternal tissues must occur in response to pre-existing tissue injury such as autoimmunity, or the cells must differentiate into others able to trigger autoimmunity in response to activation events such as infectious disease, malignancy, exposure to chemicals, or tissue injury (19, 27).

Searching for microchimerism in tissues of pregnant women who died due to a variety of causes revealed that Y-chromosome-positive chimeric cells were present in practically all organs, and significantly more so than in organs of nonpregnant women, with the lungs being most chimeric (52). Chimeric cells appeared to be both parenchymal and hematopoietic cells (52). Most interestingly, the distribution of chimeric cells in various organs appears to be similar to those in a mouse model (53), with microchimerism most often present in the lungs (54). This raises the hypothesis of the lungs as a favorable microenvironment for chimerism, a site of passive entrapment, or a site where microchimerism occurs mostly because of the great percentage of cardiac output received in the pulmonary capillary bed. Interestingly, another autopsy study showed that even more chimeric cells in lungs were present in women with preeclampsia (55), a finding that dates back to Schmorl (1, 2) but with the additional evidence of syncytial aggregates containing the anti-angiogenic factor sFlt-1 that may contribute to the systemic endothelial dysfunction characteristic of preeclampsia.

Pregnancy-associated progenitor cells and significance for repair of maternal tissue and organs.

From the time that fetal microchimeric cells were identified in pregnant and postpartum women, investigators have explored a range of hypotheses regarding their role with a strong predilection around allo-immunity of these fetal cells (24, 39). An intriguing observation consisted of fetal cells persisting for decades in the maternal body and forming entire thyroid follicles (42). Similarly, when looking more systematically, fetal cells could be shown to adopt phenotypes across multiple lineages in a range of maternal injured tissues (56, 57). This introduced a new paradigm suggesting that fetal cells were stem or progenitors that would engraft in maternal tissues and participate in the naturally occurring injury repair process (58). These fetal stem cells were called pregnancy-associated progenitor cells (PAPCs) (56, 59). Many studies have attempted to understand the functional plasticity and potency of these cells. The initial study in the peripheral blood of pregnant women suggested that PAPCs had hematopoietic as well as lymphoid progenitor activities (CD34+CD38+ phenotype) (24).was subsequently functionally validated in murine T and B cell immunodeficient models in which fetal lymphoid progenitors could mature in the maternal thymus and bone marrow to form T cells and immunoglobulin-producing B cells (60). Although PAPCs did not demonstrate a significant myeloid potential (61), fetal granulocytes have been reported in maternal blood, suggesting that PAPCs also have myeloid capacity (62).

The multilineage potential of PAPCs supported the role and activity of an early progenitor cell acquired by the mother during the first trimester and early stages of pregnancy. Accordingly, fetal mesenchymal stem cells have been described to enter the circulation and engraft in the maternal bone marrow where they can persist for decades (22, 63). These fetal MSCs have extended plasticity towards mesenchymal lineages. In particular, contribution of PAPCs to mesenchymal lineages has been demonstrated when fibrosis occurs in the kidney, wounds, or the appendix (6466). Other studies have reported cardiomyocyte differentiation of fetal cells in maternal heart (67), however, muscle differentiation was not reproducible in mice with a maternal genetic defect or injury such as in models of Duchenne muscle dystrophy (68).

Studies attempting to identify PAPCs in placental sections visualized fetal CD34+ in maternal decidual tissues. Surprisingly, most of these cells harbored endothelial markers such as CD31 or von Willebrand factor, suggesting a major contribution of fetal endothelial progenitors to PAPCs (23). Indeed, the endothelial potential of PAPCs has been shown in maternal inflammatory skin diseases (69), skin wounds (47, 70), myocardial infarction (71), melanoma (46), and more recently in stroke (72). In all of these examples, entire blood vessels have formed from fetal microchimeric cells and were connected to maternal vessels. The origin of these endothelial progenitors remains unclear, as some findings suggest that they can emanate from term placentas, and others support the role of earlier CDX2 positive progenitors (71). Evidence for chimeric cells in wound healing comes from a recent study in mice in which it was shown that fetal microchimeric cells with a CD11b+ CD34+ CD31+ phenotype and high expression of the C-C chemokine receptor 2 migrate to areas of maternal injury, where they participate in repair (73). These myeloid cells could be part of the myeloid angiogenic cells that promote wound vascularization via paracrine activities (74). It is suggested that these findings could lead to therapeutic strategies involving tissue repair through natural stem cell therapy.

Finally, beyond these classical mesodermal lineages, several studies have demonstrated the potential of PAPCs to have endodermal or ectodermal fates. Indeed, liver cells have been reported to originate from fetal cell microchimerism (75). Cells derived from multiple nervous system lineages including neurons have been found in postpartum maternal brains or spinal cord (76, 77). Intestinal or thyroid epithelium has been also reported (78). Although the functional significance of these findings is difficult to evaluate, these observations raise fundamental questions in the field of stem cell therapy. Indeed, a major question is whether PAPCs are a homogenous population acquired early during gestation that naturally has multilineage potential or if it is a mixed population including multiple cell types with diverse plasticity.

Maternofetal Microchimerism

MATERNAL CELLS IN OFFSPRING TISSUES

Transfer of cells between the pregnant woman and her fetus also occurs, resulting in persistence of maternal cells in offspring for many years after birth (7981). Maternal cells have been identified in a variety of tissues including the heart, liver, lung, and brain of neonates (Fig. 2) (82), older infants, and adults, with a variety of potential immunological implications (83, 84). For example, increased levels of maternal microchimerism have been described in many human autoimmune disorders including juvenile dermatomyositis (85), idiopathic inflammatory myopathies (86), and biliary atresia (8789).

Whether these genetically foreign maternal cells are harmful and instigate alloreactivity, or alternatively provide regenerative properties remains uncertain. Evidence suggesting the latter hypothesis includes the identification of female insulin-producing cells in the pancreatic islets of men with type 1 diabetes (90, 91), keratinocyte phenotypes amongst maternal microchimeric cells in patients with pityriasis lichenoides (92), and cardiac myocyte properties by maternal cells in infants with arrhythmias due to neonatal lupus 3). The rarity of maternal microchimeric cells (1 in 105 to 107 offspring cells) has precluded a more comprehensive systemic identification of their distribution and cellular phenotypes across multiple tissues. Likewise, the lack of tools for their experimental manipulation has prevented definitive identification of their functional roles.

A naturally occurring instance of maternal microchimerism that bypasses these limitations is the enriched accumulation of maternal immune cells in immune-deficient offspring. Graft-versus-host disease from transplacentally engrafted maternal lymphocytes is a well-described clinical manifestation of severe combined immune deficiency (93). On the other hand, expanded levels of circulating maternal T cells can also protect immune-deficient offspring by augmenting antimicrobial host defense against pathogens that cause opportunistic infections (94). In rodents, maternal microchimeric cells have been shown to produce IgG in B cell-deficient offspring (95), and interleukin-2 in the thymus and spleen of interleukin-2-deficient offspring (96).

The absence of defined fetal-neonatal immune cell subsets in these instances undoubtedly creates the physiological niche that allows for microchimerism of transplacentally engrafted maternal leukocytes. Whether microchimeric maternal cells provide similarly protective benefits in the immune components of offspring remains undefined. However, given the relatively naive and hyporesponsive state of fetal and neonatal adaptive immune components that promote increased vulnerability in these developmental windows (97, 98), and the increasingly established purposeful transfer of protective maternal antibodies (99, 100), it would not be surprising that vertically transferred intact maternal cells that persist in offspring also promote the maturation of neonatal immunity.

SIGNIFICANCE OF MATERNAL MICROCHIMERISM FOR THE EDUCATION OF FETAL IMMUNE SYSTEM

Exposure to maternal tissues and the presence of microchimeric maternal cells in the fetus throughout development have important implications for preventing fetal immune cells from rejecting noninherited maternal alloantigens. For example, it has been shown that in a healthy pregnancy, human fetuses develop regulatory T cells (Tregs) that suppress a fetal T cell response to non-inherited maternal antigens (101). The presence of microchimeric maternal cells may serve to educate fetal T cells toward tolerance, thus avoiding a fetal immune response against the mother. Conversely, this tolerance of the fetus to maternal antigens may be damaged in pregnancy complications such as preterm labor. The cord blood of infants born preterm (due to preterm premature rupture of membranes, a condition in which there is often a subclinical or clinical infection) (102), contains higher levels of maternal cell microchimerism, along with increased maturation of fetal dendritic and T cells (103). Interestingly, cord blood T cells from these neonates are sensitized against maternal antigens and produce tumor necrosis factor-α and interferon-γ, mediators that can contribute to the cascade of uterine contractions in preterm labor (104). These data are consistent with results obtained in mouse models in which there is increased maternal microchimerism after fetal stem cell transplantation (105) or infection with lipopolysaccharide to induce preterm labor (106) or miscarriage (107), and in fetuses undergoing surgery in the womb (108), which is often complicated by preterm labor. Thus, levels of maternal microchimerism may be “tunable” to enable fetal tolerance to maternal antigens at baseline, or sensitization against them during infection/inflammation, to contribute to the complex cascade that can prematurely end the pregnancy.

LONG-TERM IMMUNE CONSEQUENCES OF PREGNANCY/PARTURITION FOR FUTURE PREGNANCIES

Despite the similarities between maternal and fetal cell microchimerism, a critical distinction between them is how these genetically foreign cells establish persistence. This potentially relates to immunological maturity of each respective host. Fetal microchimeric cells enter pregnant women who have a functional repertoire of adaptive immune components. By contrast, maternal cells are found in fetal tissues beginning in the second trimester (108), prior to or during key milestones in human immunological development, such as development of the thymus and acquisition of effector function in peripheral T cells (109, 110).

Exposure to genetically foreign maternal cells in these early stages of immunological development parallels expanded immunological tolerance of offspring to non-inherited maternal antigens (NIMA) (79, 111). Immunological implications of NIMA-specific tolerance have classically been described in transplantation. Before the availability of recombinant erythropoietin, transfusion-dependent individuals broadly exposed to a wide repertoire of genetically foreign human leukocyte antigen alloantigens were found to selectively lack antibodies with NIMA specificity (112). Long-term survival of renal allografts is markedly improved between NIMA matched sibling donor–recipient pairings (113). The risk of graft versus host disease after hematopoietic stem cell transplantation is similarly reduced in NIMAmatched donor–recipient pairings (114116).

The bidirectional transfer during pregnancy, and persistence of microchimeric cells in mothers and offspring, is highly conserved across mammalian species. Considering the fetus as an allograft, and improved pregnancy outcomes with enforced fetal–allograft tolerance, further highlight potential teleological benefits driving expanded tolerance of offspring to NIMA. An example of these reproductive benefits is reduced susceptibility to fetal wastage in mice during pregnancies in which fetal alloantigens are matched with NIMAs (117). In other words, tolerance to NIMA likely has beneficial impacts in enforcing fetal tolerance during next-generation pregnancies that contain shared fetal alloantigens. Maternal microchimerism is required for these cross-generational reproductive benefits of NIMA-specific tolerance since selective depletion of microchimeric maternal cells in female mice prior to mating eliminates resiliency against fetal wastage in NIMA-fetal alloantigen matched pregnancies (117). This expanded tolerance in mice to non-inherited maternal MHC haplotype antigens parallels classical human observations of reduced sensitization to erythrocyte rhesus (Rh) antigen during pregnancy amongst Rh-negative women born to Rh-positive compared with Rh-negative mothers (118). In particular, pregnancy-induced Rh sensitization was shown to be significantly reduced amongst Rh-negative women with Rh-positive mothers (Rh is transformed into an NIMA) compared with Rh-negative women born to Rh-negative mothers (119). Thus, maternal microchimerism confers important immunological consequences in female offspring on the outcomes of next-generation, future pregnancies.

The similarities between maternal microchimerism in children and fetal microchimerism in mothers raises the question as to whether fetal microchimeric cells retained in women also reinforce fetal tolerance during subsequent pregnancies. Human epidemiology studies consistently show pregnancy complications such as preeclampsia are much more common during first pregnancies (120). For example, an analysis of over 700 000 primiparous mothers showed that the overall 4.1% rate of preeclampsia in a first pregnancy was reduced to 1.7% in subsequent pregnancies (121). Importantly, these protective benefits are partner-specific since a change in paternity overrides the reduced risk of preeclampsia conferred by a prior pregnancy (122124). Although the necessity of fetal microchimeric cells in sustaining and enforcing fetal tolerance has not been demonstrated, immunological memory of prior pregnancies, similar to expanded tolerance of offspring to NIMA are each linked with persistent accumulation of immune suppressive regulatory CD4+ T cells with defined antigen-specificity (117, 125).

These reproductive benefits illustrate one aspect of nature’s intent in promoting the transfer and retention of microchimeric cells in mothers and their offspring, and also raise interesting questions with regard to potential immunological consequences of individuals being constitutively chimeric. A provocative hypothesis is that defective tolerance to microchimeric cells may drive the underlying pathogenesis of some autoimmune and/or autoinflammatory disorders (84). Important next steps will be to establish the origin, tissue distribution, and molecular properties of microchimeric cells that control whether tolerogenic or sensitization responses are primed to further investigate the role of microhimeric cells in the immune-pathogenesis of idiopathic autoimmune and/or autoinflammatory disorders (79).

SIGNIFICANCE OF MICROCHIMERISM FOR TRANSPLANTATION AND CURE OF GENETIC DISORDERS

Fetal tolerance to non-inherited maternal antigens is transient. Sustaining this tolerance after birth likely depends on the presence of sufficient exposure to maternal cells to enable continued education of T cells. There is one experiment of nature, biliary atresia, in which higher levels of maternal cells are found in the livers of affected children after birth (8789). Interestingly, there is improved survival of liver transplants from a maternal donor compared to a paternal donor in children with biliary atresia (126), suggesting that the continued presence of maternal microchimerism in this disease can sustain tolerance to the maternal donor.

One of the most exciting applications of fetal tolerance to maternal antigens is in the context of in utero stem cell transplantation. Infants with congenital hematopoietic disorders can be cured by hematopoietic stem cell (HSC) transplantation, but a suitable donor is often not available and there are severe complications of the conditioning and immune suppression regimens needed to prepare the recipient’s bone marrow for the transplant. However, these disorders are often diagnosed prenatally and could potentially benefit from in utero transplantation, a strategy that takes advantage of the unique immune milieu of the fetus to induce tolerance to the transplanted cells. Based on the observation that human fetuses are tolerant to NIMAs during pregnancy (101), and the understanding that the maternal immune system can mediate rejection of third-party cells transplanted into the fetus (104), there is now an ongoing phase 1 clinical trial to transplant maternal HSCs into fetuses with alpha thalassemia major (clinicaltrials.gov NCT02986698). These infants suffer from anemia before birth, requiring antenatal blood transfusions to survive. The HSC transplantation can be given at the same time as the transfusions. If this treatment is found to be safe and effective, it could be expanded to treat patients with similar, more common conditions such as sickle cell anemia. Thus, a better understanding of the biology that enables the trafficking of cells between the mother and the fetus could ultimately result in therapies to treat fetuses with genetic disorders.

SUMMARY AND FUTURE RESEARCH

Intentionally, the focus of this review was on the biological consequences of the exchange of intact cells between the pregnant woman and her fetus during pregnancy. Space limitations did not allow detailed mention of the maternal cells that pass through breast milk after birth (127), or exchange of cell-free nucleic acids, either through free floating fragments of DNA or RNA in the blood, or via structures such as exosomes. It is worth remembering, however, that the twentieth century interest in fetal cell microchimerism originated as a quest to develop noninvasive access to the fetus to facilitate prenatal diagnosis of chromosomal and single gene disorders (4, 5, 128, 129). Although the feasibility of using next generation sequencing in trophoblast cells isolated from maternal blood has been demonstrated using current technologies (130), this is still far from a trial-ready cell-based version of noninvasive prenatal diagnosis. Novel approaches suggested for manipulating first trimester fetal primitive erythroblasts may provide additional exploratory strategies for use of this cell type for noninvasive prenatal diagnosis (131) but improvements in enrichment and automation are necessary for any proposed clinical application (132). Reports using microfluidics to isolate fetal trophoblast cells and erythroblasts show promise (133), as do recent attempts to develop and utilize a single cell-based polymerase chain reaction system to analyze genomic DNA in fetal cells purified from maternal blood (134). In reality, however, it is the use of circulating cell-free fetal (cff) DNA for prenatal screening for aneuploidy that has progressed from an idea to widespread global clinical applications (135) in a very short time, mainly because of the significant quantity of fetal presumed-trophoblast DNA within maternal plasma (136) compared with the rarity of intact fetal cells within maternal blood, as well as their persistence after pregnancy (25). Prenatal screening technologies using cffDNA have limitations (137). Positive predictive values for aneuploidies other than trisomy 21 can be suboptimal based on the testing platform employed and are even lower for common microdeletions. In addition, reliable genome-wide fetal sequencing using cffDNA remains somewhat challenging due to the technological difficulties in detecting small chromosomal imbalances, issues of confined placental mosaicism, the presence of unexpected maternal chromosomal abnormalities, as well as the high proportion of maternal cell free DNA in plasma (137).

As for the biological consequences of fetal cells in maternal tissues and organs, future investigations in humans need to include complete pregnancy histories, including elective terminations and miscarriages. This is particularly true in the stem cell field, in which the presence of male donor cells in female recipients have been interpreted as transplant successes without mention or consideration of prior pregnancy histories (27). Animal models will continue to be very useful in establishing the mechanisms underlying fetal cell wound healing and/or repair associated with injury and inflammation in the mother. An additional important area for future investigation is whether ongoing stimulation by fetal microchimeric cells in a woman is required for protection against complications primed by prior pregnancy, such as preeclampsia.

The implications of maternal cell microchimerism for the development and education of the fetal immune system, as well as the significance of NIMAs for transplant acceptance, are becoming increasingly appreciated. The role of maternal cells in breast milk may play a role in maintaining NIMA-specific tolerance during childhood and adulthood. This is an area for further research. Microchimerism as a factor in multi-generational reproductive success is another area of interest for the future. Lastly, taking advantage of the priming of the fetal immune system by naturally occurring maternal cell microchimerism to provide lifesaving HSC transplants is an exciting and important clinical application.

In summary, the lifelong consequences of the exchange of cells between a mother and child are profound and have many applications in development, health, and disease. Whereas a mother and her child are undeniably forever connected in many ways, the studies presented here demonstrate that the connection exists at the most basic, granular, cellular level.

Nonstandard Abbreviations:

MSC

mesenchymal stem cell

PAPC

pregnancy-associated progenitor cell

NIMA

non-inherited maternal antigen

HSC

hematopoietic stem cell

cff

cell-free fetal

Footnotes

Authors’ Disclosures or Potential Conflicts of Interest: No authors declared any potential conflicts of interest.

References

  • 1.Schmorl G Pathologisch-anatomische Untersuchungen ueber Puerperal Eklampsie. Leipzig, Vogel; 1893. [Google Scholar]
  • 2.Lapaire O, Holzgreve W, Oosterwijk JC, Brinkhaus R, Bianchi DW. Georg Schmorl on trophoblasts in the maternal circulation. Placenta 2007;28:1–5. [DOI] [PubMed] [Google Scholar]
  • 3.Bianchi DW, Lo YM. Fetomaternal cellular and plasma DNA trafficking. Ann N Y Acad Sci 2001;945:119–31. [DOI] [PubMed] [Google Scholar]
  • 4.Jackson L Fetal cells and DNA in maternal blood. Prenat Diagn 2003;23:837–46. [DOI] [PubMed] [Google Scholar]
  • 5.Walknowska J, Conte FA, Grumbach MM. Practical and theoretical implications of fetal-maternal lymphocyte transfer. Lancet 1969;293:1119–22. [DOI] [PubMed] [Google Scholar]
  • 6.Ariga H, Ohto H, Busch MP, Imamura S, Watson R, Reed W, Lee TH. Kinetics of fetal cellular and cell-free DNA in the maternal circulation during and after pregnancy: implications for noninvasive prenatal diagnosis. Transfusion 2001;41:1524–30. [DOI] [PubMed] [Google Scholar]
  • 7.Sebring ES, Polesky HF. Fetomaternal hemorrhage: incidence, risk factors, time of occurrence, and clinical effects. Transfusion 1990;30:344–57. [DOI] [PubMed] [Google Scholar]
  • 8.Khosrotehrani K, Johnson KL, Lau J, Dupuy A, Cha D, Bianchi DW. The influence of fetal loss on the presence of fetal cell microchimerism. Arthritis Rheum 2003;48: 3237–41. [DOI] [PubMed] [Google Scholar]
  • 9.Peterson SE, Nelson JL, Guthrie KA, Gadi VK, Aydelotte TM, Oyer DJ, et al. Prospective assessment of fetal-maternal cell transfer in miscarriage and pregnancy termination. Hum Reprod 2012; 27:2607–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Bianchi DW, Farina A, Weber W, Delli-Bovi LC, Deriso M, Williams JM, Klinger KW. Significant fetal-maternal hemorrhage after termination of pregnancy: implications for development of fetal cell microchimerism. Am J Obstet Gynecol 2001;184:703–6. [DOI] [PubMed] [Google Scholar]
  • 11.Bianchi DW, Williams JM, Sullivan LM, Hanson FW, Klinger KW, Shuber AP. PCR quantitation of fetal cells in maternal blood in normal and aneuploid pregnancies. Am J Hum Genet 1997;61:822–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Shree R, Harrington WE, Kanaan SB, Forsyth A, Cousin E, Lopez A, et al. Fetal microchimerism by mode of delivery: a prospective cohort study. BJOG 2019;126:24–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Hahn S, Huppertz B, Holzgreve W. Fetal cells and cell free fetal nucleic acids in maternal blood: new tools to study abnormal placentation? Placenta 2005;26: 515e526. [DOI] [PubMed] [Google Scholar]
  • 14.Gammill HS, Aydelotte TM, Guthrie KA, Nkwopara EC, Nelson JL. Cellular fetal microchimerism in preeclampsia. Hypertension 2013;62:1062–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Benirschke K, Kaufmann P. Pathology of the human placenta. 4th Ed. New York: Springer-Verlag, Inc. 2000. [Google Scholar]
  • 16.Charnock-Jones DS, Burton G. Placental vascular morphogenesis. Baillieres Best Pract Res Clin Obstetrics Gynaecol 2000;14:953–968. [DOI] [PubMed] [Google Scholar]
  • 17.Bianchi DW. Fetal cells in the mother: from genetic diagnosis to diseases associated with fetal cell microchimerism. Eur J Obstet Gynecol Reprod Biol 2000;92: 103–8. [DOI] [PubMed] [Google Scholar]
  • 18.Jansen MW, Korver-Hakkennes K, van Leenen D, Visser W, In ‘t Veld PA, de Groot CJ, Wladimiroff JW. Significantly higher number of fetal cells in the maternal circulation of women with preeclampsia. Prenat Diagn 2001;21:1022–6. [DOI] [PubMed] [Google Scholar]
  • 19.O’Donoghue K Implications of fetal stem cell trafficking in pregnancy. Rev Gynaecol Perinatal Pract 2006; 6:87–98. [Google Scholar]
  • 20.Campagnoli C, Roberts IA, Kumar S, Choolani M, Bennett PR, Letsky E, Fisk NM. Expandability of haemopoietic progenitors in first trimester fetal and maternal blood: implications for noninvasive prenatal diagnosis. Prenat Diagn 2002;22:463–9. [DOI] [PubMed] [Google Scholar]
  • 21.Campagnoli C, Roberts IA, Kumar S, Bennett PR, Bellantuono I, Fisk NM. Identification of mesenchymal stem/progenitor cells in human first-trimester fetal blood, liver, and bone marrow. Blood 2001;98: 2396–402. [DOI] [PubMed] [Google Scholar]
  • 22.O’Donoghue K, Choolani M, Chan J, de la Fuente J, Kumar S, Campagnoli C, et al. Identification of fetal mesenchymal stem cells in maternal blood: implications for non-invasive prenatal diagnosis. Mol Hum Reprod 2003;9:497–502. [DOI] [PubMed] [Google Scholar]
  • 23.Parant O, Dubernard G, Challier J-C, Oster M, Uzan S, Aractingi S, et al. CD34 cells in maternal placental blood are mainly fetal in origin and express endothelial markers. Lab Invest 2009;89:915–923. [DOI] [PubMed] [Google Scholar]
  • 24.Bianchi DW, Zickwolf GK, Weil GJ, Sylvester S, De Maria MA. Male fetal progenitor cells persist in maternal blood for as long as 27 years postpartum. Proc Nat Acad Sci USA 1996;93:705–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Bianchi DW, Simpson JL, Jackson LG, Elias S, Holzgreve W, Evans MI, et al. Fetal gender and aneuploidy detection using fetal cells in maternal blood: analysis of NIFTY I data. Prenat Diagn 2002;22: 609–15. [DOI] [PubMed] [Google Scholar]
  • 26.Cismaru CA, Soritau O, Jurj A-M, Lajos R, Pop B, Bocean C, Albzour B, et al. Isolation and characterization of a fetal-maternal microchimeric stem cell population in maternal hair follicles long after parturition. Stem Cell Rev Rep 2019;15:519–29. [DOI] [PubMed] [Google Scholar]
  • 27.Bianchi DW, Fisk NM. Fetomaternal cell trafficking and the stem cell debate: gender matters. Jama 2007;297: 289–91. [DOI] [PubMed] [Google Scholar]
  • 28.Lee ES, Bou-Gharios G, Seppanen E, Khosrotehrani K, Fisk NM. Fetal stem cell microchimerism: natural-born healers or killers? Mol Hum Reprod 2010;16:869–78. [DOI] [PubMed] [Google Scholar]
  • 29.Artlett CM. Pathophysiology of fetal microchimeric cells. Clin Chim Acta 2005;360:1–8. [DOI] [PubMed] [Google Scholar]
  • 30.Johnson KL, Samura O, Nelson JL, McDonnell MDWM, Bianchi DW. Significant fetal cell microchimerism in a nontransfused woman with hepatitis C: evidence of long-term survival and expansion. Hepatology 2002;36:1295–7. [DOI] [PubMed] [Google Scholar]
  • 31.Khosrotehrani K, Bianchi DW. Fetal cell microchimerism: helpful or harmful to the parous woman? Curr Opin Obstet Gynecol 2003;15:195–9. [DOI] [PubMed] [Google Scholar]
  • 32.Koopmans M, Hovinga ICLK, Baelde HJ, Fernandes RJ, de Heer E, Bruijn JA, et al. Chimerism in kidneys, livers and hearts of normal women: implications for transplantation studies. Am J Transplant 2005;5: 1495–1502. [DOI] [PubMed] [Google Scholar]
  • 33.Koopmans M, Kremer Hovinga ICL, Baelde HJ, Harvey MS, de Heer E, Bruijn JA, et al. Chimerism occurs in thyroid, lung, skin and lymph nodes of women with sons. J Reprod Immunol 2008;78:68–75. [DOI] [PubMed] [Google Scholar]
  • 34.Kremer Hovinga IC, Koopmans M, de Heer E, Bruijn JA, Bajema IM. Chimerism in systemic lupus erythematosus–three hypotheses. Rheumatology 2007;46: 200–8. [DOI] [PubMed] [Google Scholar]
  • 35.Johnson KL, Nelson JL, Furst DE, McSweeney PA, Roberts DJ, Zhen DK, Bianchi DW. Fetal cell microchimerism in tissue from multiple sites in women with systemic sclerosis. Arthritis Rheum 2001;44: 1848–54. [DOI] [PubMed] [Google Scholar]
  • 36.Khashan AS, Kenny LC, Laursen TM, Mahmood U, Mortensen PB, Henriksen TB, O’Donoghue K. Pregnancy and the risk of autoimmune disease. PLoS One 2011;6:e19658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Nelson JL. Pregnancy and microchimerism in autoimmune disease: protector or insurgent? Arthritis Rheum 2002;46:291–7. [DOI] [PubMed] [Google Scholar]
  • 38.Artlett CM, Smith JB, Jimenez SA. Identification of fetal DNA and cells in skin lesions from women with systemic sclerosis. N Engl J Med 1998;338:1186–91. [DOI] [PubMed] [Google Scholar]
  • 39.Nelson JL, Furst DE, Maloney S, Gooley T, Evans PC, Smith A, et al. Microchimerism and HLA-compatible relationships of pregnancy in scleroderma. Lancet 1998;351:559–62. [DOI] [PubMed] [Google Scholar]
  • 40.Evans PC, Lambert NC, 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: 2033–7. [PubMed] [Google Scholar]
  • 41.Toda I, Kuwana M, Tsubota K, Kawakami Y. Lack of evidence for an increased microchimerism in the circulation of patients with Sjogren’s syndrome. Ann Rheum Dis 2001;60:248–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Srivatsa B, Srivatsa S, Johnson KL, Samura O, Lee SL, Bianchi DW. Microchimerism of presumed fetal origin in thyroid specimens from women: a case-control study. Lancet 2001;358:2034–8. [DOI] [PubMed] [Google Scholar]
  • 43.Abbud Filho M, Pavarino-Bertelli EC, Alvarenga MPS, Fernandes IMM, Toledo RA, Tajara EH, et al. Systemic lupus erythematosus and microchimerism in autoimmunity. Transplant Proc 2002;34:2951–2. [DOI] [PubMed] [Google Scholar]
  • 44.da Silva Florim GM, Caldas HC, Pavarino EC, Bertollo EMG, Fernandes IMM, Abbud-Filho M. Variables associated to fetal microchimerism in systemic lupus erythematosus patients. Clin Rheumatol 2016;35:107–11. [DOI] [PubMed] [Google Scholar]
  • 45.Aractingi S, Berkane N, Bertheau P, Le Goué C, Dausset J, Uzan S, Carosella ED. Fetal DNA in skin of polymorphic eruptions of pregnancy. Lancet 1998;352: 1898–901. [DOI] [PubMed] [Google Scholar]
  • 46.Nguyen Huu S, Oster M, Avril M-F, Boitier F, Mortier L, Richard M-A, et al. Fetal microchimeric cells participate in tumor angiogenesis in melanomas occurring during pregnancy. Am J Pathol 2009;174:630–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Mahmood U, O’Donoghue K. Microchimeric fetal cells play a role in maternal wound healing after pregnancy. Chimerism 2014;5:40–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Cha D, Khosrotehrani K, Kim Y, Stroh H, Bianchi DW, Johnson KL. Cervical cancer and microchimerism. Obstet Gynecol 2003;102:774–81. [DOI] [PubMed] [Google Scholar]
  • 49.Gadi VR, Nelson JL. Fetal microchimerism in women with breast cancer. Cancer Res 2007;67:9035–8. [DOI] [PubMed] [Google Scholar]
  • 50.Kallenbach LR, Johnson KL, Bianchi DW. Fetal cell microchimerism and cancer: a nexus of reproduction, immunology, and tumor biology. Cancer Res 2011;71: 8–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Gannag M, Amoura Z, Lantz O, Piette J-C, Caillat-Zucman S. Feto-maternal microchimerism in connective tissue diseases. Eur J Immunol 2002;32:3405–13. [DOI] [PubMed] [Google Scholar]
  • 52.Rijnink EC, Penning ME, Wolterbeek R, Wilhelmus S, Zandbergen M, van Duinen SG, et al. Tissue microchimerism is increased during pregnancy: a human autopsy study. Mol Hum Reprod 2015;21:857–64. [DOI] [PubMed] [Google Scholar]
  • 53.Fujiki Y, Johnson KL, Tighiouart H, Peter I, Bianchi DW. Fetomaternal trafficking in the mouse increases as delivery approaches and is highest in the maternal lung. Biol Reprod 2008;79:841–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Pritchard S, Hoffman AM, Johnson KL, Bianchi DW. Pregnancy-associated progenitor cells: an under-recognized potential source of stem cells in maternal lung. Placenta 2011;32:S298–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Buurma AJ, Penning ME, Prins F, Schutte JM, Bruijn JA, Wilhelmus S, et al. Preeclampsia is associated with the presence of transcriptionally active placental fragments in the maternal lung. Hypertension 2013;62: 608–13. [DOI] [PubMed] [Google Scholar]
  • 56.Khosrotehrani K, Johnson KL, Cha DH, Salomon RN, Bianchi DW. Transfer of fetal cells with multilineage potential to maternal tissue. JAMA 2004;292:75–80. [DOI] [PubMed] [Google Scholar]
  • 57.Khosrotehrani K, Reyes RR, Johnson KL, Freeman RB, Salomon RN, Peter I, et al. Fetal cells participate over time in the response to specific types of murine maternal hepatic injury. Hum Reprod 2007;22:654–61. [DOI] [PubMed] [Google Scholar]
  • 58.Khosrotehrani K, Bianchi DW. Multi-lineage potential of fetal cells in maternal tissue: a legacy in reverse. J Cell Sci 2005;118:1559–63. [DOI] [PubMed] [Google Scholar]
  • 59.Seppanen E, Fisk NM, Khosrotehrani K. Pregnancy-acquired fetal progenitor cells. J Reprod Immunol 2013; 97:27–35. [DOI] [PubMed] [Google Scholar]
  • 60.Khosrotehrani K, Leduc M, Bachy V, Nguyen HS, Oster M, Abbas A, et al. Pregnancy allows the transfer and differentiation of fetal lymphoid progenitors into functional T and B cells in mothers. J Immunol 2008;180:889–97. [DOI] [PubMed] [Google Scholar]
  • 61.Leduc M, Guegan S, Roy E, Oster M, Aractingi S, Khosrotehrani K. Limited functional capacity of microchimeric fetal hematopoietic progenitors acquired by mothers during pregnancy. Exp Hematol 2010;38: 852–3. [DOI] [PubMed] [Google Scholar]
  • 62.Sunku CC, Gadi VK, de Laval de Lacoste B, Guthrie KA, Nelson JL. Maternal and fetal microchimerism in granulocytes. Chimerism 2010;1:11–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.O’Donoghue K, Chan J, de la Fuente J, Kennea N, Sandison A, Anderson JR, et al. Microchimerism in female bone marrow and bone decades after fetal mesenchymal stem cell trafficking pregnancy. Lancet 2004; 364:179–82. [DOI] [PubMed] [Google Scholar]
  • 64.Santos MA, O’Donoghue K, Wyatt-Ashmead J, Fisk NM. Fetal cells in the maternal appendix: a marker of inflammation or fetal tissue repair? Hum Reprod 2008; 23:2319–25. [DOI] [PubMed] [Google Scholar]
  • 65.Roy E, Seppanen E, Ellis R, Lee ES, Khosroterani K, Fisk NM, Bou-Gharios G. Biphasic recruitment of microchimeric fetal mesenchymal cells in fibrosis following acute kidney injury. Kidney Int 2014;85:600–10. [DOI] [PubMed] [Google Scholar]
  • 66.Seppanen E, Roy E, Ellis R, Bou-Gharios G, Fisk NM, Khosrotehrani K. Distant mesenchymal progenitors contribute to skin wound healing and produce collagen: evidence from a murine fetal microchimerism model. PLoS One 2013;8:e62662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Bayes-Genis A, Bellosillo B, de la Calle O, Salido M, Roura S, Ristol FS, et al. Identification of male cardiomyocytes of extracardiac origin in the hearts of women with male progeny: male fetal cell microchimerism of the heart. J Heart Lung Transplant 2005;24:2179–83. [DOI] [PubMed] [Google Scholar]
  • 68.Seppanen EJ, Hodgson SS, Khosrotehrani K, Bou-Gharios G, Fisk NM. Fetal microchimeric cells in a fetustreats-its-mother paradigm do not contribute to dystrophin production in serially parous mdx females. Stem Cells Dev 2012;21:2809–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Nguyen HS, Oster M, Uzan S, Chareyre F, Aractingi S, Khosrotehrani K. Maternal neoangiogenesis during pregnancy partly derives from fetal endothelial progenitor cells. Proc Natl Acad Sci USA 2007;104:1871–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Nassar D, Droitcourt C, Mathieu-d’Argent E, Kim MJ, Khosrotehrani K, Aractingi S. Fetal progenitor cells naturally transferred through pregnancy participate in inflammation and angiogenesis during wound healing. FASEB J 2012;26:149–57. [DOI] [PubMed] [Google Scholar]
  • 71.Kara RJ, Bolli P, Karakikes I, Matsunaga I, Tripodi J, Tanweer O, et al. Fetal cells traffic to injured maternal myocardium andundergo cardiac differentiation. Circ Res 2012;110:82–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Ritzel RM, Patel AR, Spychala M, Verma R, Crapser J, Koellhoffer EC, et al. Multiparity improves outcomes after cerebral ischemia in female mice despite features of increased metabovascular risk. Proc Natl Acad Sci USA 2017;114:E5673–E5682. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Castela M, Nassar D, Sbeih M, Jachiet M, Wang Z, Aractingi S. Ccl2/Ccr2 signalling recruits a distinct fetal microchimeric population that rescues delayed maternal wound healing. Nat Commun 2017;8:15463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Medina RJ, Barber CL, Sabatier F, Dignat-George F, Melero-Martin JM, Khosrotehrani K, et al. Endothelial progenitors: a consensus statement on nomenclature. Stem Cells Transl Med 2017;6:1316–1320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Stevens AM, McDonnell WM, Mullarkey ME, Pang JM, Leisenring W, Nelson JL. Liver biopsies from human females contain male hepatocytes in the absence of transplantation. Lab Invest 2004;84:1603–9. [DOI] [PubMed] [Google Scholar]
  • 76.Tan XW, Liao H, Sun L, Okabe M, Xiao ZC, Dawe GS. Fetal microchimerism in the maternal mouse brain: a novel population of fetal progenitor or stem cells able to cross the blood-brain barrier? Stem Cells 2005;23: 1443–52. [DOI] [PubMed] [Google Scholar]
  • 77.Zhang G, Zhao Y, Li ZM, Kong J. Fetal cell microchimerism in the maternal mouse spinal cord. Neurosci Bull 2014;30:81–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Cirello V, Rizzo R, Crippa M, Campi I, Bortolotti D, Bolzani S, et al. Fetal cell microchimerism: a protective role in autoimmune thyroid diseases. Eur J Endocrinol 2015;173:111–8. [DOI] [PubMed] [Google Scholar]
  • 79.Kinder JM, Stelzer IA, Arck PC, Way SS. Immunological implications of pregnancy-induced microchimerism. Nat Rev Immunol 2017;17:483–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Lo YM, Lo ES, Watson N, Noakes L, Sargent IL, Thilaganathan B, Wainscoat JS. Two-way cell traffic between mother and fetus: biologic and clinical implications. Blood 1996;88:4390–95. [PubMed] [Google Scholar]
  • 81.Maloney S, Smith A, Furst DE, Myerson D, Rupert K, Evans PC, Nelson JL. Microchimerism of maternal origin persists into adult life. J Clin Invest 1999;104: 41–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Srivatsa B, Srivatsa S, Johnson KL, Bianchi DW. Maternal cell microchimerism in newborn tissues. J Pediatr 2003;142:31–5. [DOI] [PubMed] [Google Scholar]
  • 83.Gammill HS, Nelson JL. Naturally acquired microchimerism. Int J Dev Biol 2010;54:531–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Nelson JL. The otherness of self: microchimerism in health and disease. Trends Immunol 2012;33:421–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Reed AM, Picornell YJ, Harwood A, Kredich DW. Chimerism in children with juvenile dermatomyositis. Lancet 2000;356:2156–7. [DOI] [PubMed] [Google Scholar]
  • 86.Artlett CM, Ramos R, Jiminez SA, Patterson K, Miller FW, Rider LG. Chimeric cells of maternal origin in juvenile idiopathic inflammatory myopathies. Childhood Myositis Heterogeneity Collaborative Group. Lancet 2000;356:2155–6. [DOI] [PubMed] [Google Scholar]
  • 87.Hayashida M, Nishimoto Y, Matsuura T, Takahashi Y, Kohashi K, Souzaki R, Taguchi T. The evidence of maternal microchimerism in biliary atresia using fluorescent in situ hybridization. J Pediatr Surg 2007;42: 2097–2101. [DOI] [PubMed] [Google Scholar]
  • 88.Kobayashi H, Tamatani T, Tamura T, Kusafuka J, Yamataka A, Lane GJ, et al. Maternal microchimerism in biliary atresia. J Pediatr Surg 2007;42:98–91. [DOI] [PubMed] [Google Scholar]
  • 89.Suskind DL, Rosenthal P, Heyman MB, Kong D, Magrane G, Baxter-Lowe LA, Muench MO. Maternal microchimerism in the livers of patients with biliary atresia. BMC Gastroenterol 2004;4:14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Ye J, Vives-Pi M, Gillespie KM. 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 2014;9: e86985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Khosrotehrani K, Guegan S, Fraitag S, Oster M, de Prost Y, Bodemer C, Aractingi S. Presence of chimeric maternally derived keratinocytes in cutaneous inflammatory diseases of children: the example of pityriasis lichenoides. J Invest Dermatol 2006;126:345–8. [DOI] [PubMed] [Google Scholar]
  • 93.Müller SM, Ege M, Pottharst A, Schulz AS, Schwarz K, Friedrich W. Transplacentally acquired maternal T lymphocytes in severe combined immunodeficiency: a study of 121 patients. Blood 2001;98:1847–51. [DOI] [PubMed] [Google Scholar]
  • 94.Touzot F, Dal-Cortivo L, Verkarre V, Lim A, Crucis-Armengaud A, Moshous D, et al. Massive expansion of maternal T cells in response to EBV infection in a patient with SCID-Xl. Blood 2012;120:1957–9. [DOI] [PubMed] [Google Scholar]
  • 95.Arvola M, Gustafsson E, Svensson L, Jansson L, Holmdahl R, Heyman B, et al. Immunoglobulin-secreting cells of maternal origin can be detected in B cell-deficient mice. Biol Reprod 2000;63:1817–1824. [DOI] [PubMed] [Google Scholar]
  • 96.Wrenshall LE, Stevens ET, Smith DR, Miller JD. 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 2007;245:80–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Basha S, Surendran N, Pichichero M. Immune responses in neonates. Expert Rev Clin Immunol 2014;10:1171–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Kollmann TR, Kampmann B, Mazmanian SK, Marchant A, Levy O. Protecting the newborn and young infant from infectious diseases: lessons from immune ontogeny. Immunity 2017;46:350–63. [DOI] [PubMed] [Google Scholar]
  • 99.Jennewein MF, Abu-Raya B, Jiang Y, Alter G, Marchant A. Transfer of maternal immunity and programming of the newborn immune system. Semin Immunopathol 2017;39:605–13. [DOI] [PubMed] [Google Scholar]
  • 100.Jennewein MF, Goldfarb I, Dolatshahi S, Cosgrove C, Noelette FJ, Krykbaeva M, et al. Fc glycan-mediated regulation of placental antibody transfer. Cell 2019; 178:202–15. E14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Mold JE, Michaelsson 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Goldenberg RL, Culhane JF, Iams JD, Romero R. Epidemiology and causes of preterm birth. Lancet 2008;371:75–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Frascoli M, Coniglio L, Witt R, Jeanty C, Fleck-Derderian S, Myers DE, et al. Alloreactive fetal T cells promote uterine contractility in preterm labor via IFN-g and TNF-a. Sci Transl Med 2018;10:eaan2263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Nijagal A, Wegorzewska M, Jarvis E, Le T, Tang Q,TC MacKenzie TC. Maternal T cells limit engraftment after in utero hematopoietic cell transplantation in mice. J Clin Invest 2011;121:582–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Wegorzewska M, Le T, Tang Q, MacKenzie TC. Increased maternal T cell microchimerism in the allogeneic fetus during LPS-induced preterm labor in mice. Chimerism 2015;16:1–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Johnson KL, Tao K, Stroh H, Kallenbach L, Peter I, Richey L, et al. Increased fetal cell trafficking in murine lung following complete pregnancy loss from exposure to lipopolysaccharide. Fertil Steril 2010;93:1718–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Saadai P, Lee T-H, Bautista G, Gonzales KD, NIjagal A, Busch MP, et al. Alterations in maternal-fetal cellular trafficking after fetal surgery. J Pediatr Surg 2012;47: 1089–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Jonsson AM, Uzunel M, Gotherstrom C, Papadogiannakis N, Westgren M. Maternal microchimerism in human fetal tissues. Am J Obstet Gynecol 2008;198:325.e1–6–325.e6. [DOI] [PubMed] [Google Scholar]
  • 109.Haynes BF. Phenotypic characterization and ontogeny of components of the human thymic microenvironment. Clin Res 1984;32:500–507. [PubMed] [Google Scholar]
  • 110.von Hoegen P, Sarin S, Krowka JF. Deficiency in T cell responses of human fetal lymph node cells: a lack of accessory cells. Immunol Cell Biol 1995;73:353–61. [DOI] [PubMed] [Google Scholar]
  • 111.Burlingham WJ, Benichou G. Bidirectional alloreactivity: A proposed microchimerism-based solution to the NIMA paradox. Chimerism 2012;3:29–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Claas FH, Gijbels Y, van der Velden-de Munck J, van Rood JJ. Induction of B cell unresponsiveness to noninherited maternal HLA antigens during fetal life. Science 1988;241:1815–17. [DOI] [PubMed] [Google Scholar]
  • 113.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. [DOI] [PubMed] [Google Scholar]
  • 114.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. [DOI] [PubMed] [Google Scholar]
  • 115.Ichinohe T, Uchiyama T, Shimazaki C, Matsuo K, Tamaki S, Hino M, 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 2004;104:3821–8. [DOI] [PubMed] [Google Scholar]
  • 116.Kanda J, Ichinohe T, Shimazaki C, Hamaguchi M, Watanabe A, Ishida H, et al. Long-term survival after HLA-haploidentical SCT from non-inherited maternal antigen-mismatched family donors: impact of chronic GVHD. Bone Marrow Transplant 2009;44:327–9. [DOI] [PubMed] [Google Scholar]
  • 117.Kinder JM, Jiang TT, Ertelt JM, Xin L, Strong BS, Shaaban AF, Way SS. Cross-generational reproductive fitness enforced by microchimeric maternal cells. Cell 2015;162:505–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Kinder JM, Jiang TT, Ertelt JM, Xin L, Strong BS, Shaaban AF, Way SS. Tolerance to noninherited maternal antigens, reproductive microchimerism and regulatory T cell memory: 60 years after ‘Evidence for actively acquired tolerance to Rh antigens’. Chimerism 2015;6:8–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Owen RD, Wood HR, Foord AG, Sturgeon P, Baldwin LG. Evidence for actively acquired tolerance to rh antigens. Proc Natl Acad Sci USA 1954;40:420–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Deshmukh H, Way SS. Immunological basis for recurrent fetal loss and pregnancy complications. Annu Rev Pathol Mech Dis 2019;14:185–210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Hernandez-Diaz S, Toh S, Cnattingius S. Risk of preeclampsia in first and subsequent pregnancies: prospective cohort study. BMJ 2009;338:b2255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Li DK, Wi S. Changing paternity and the risk of preeclampsia/eclampsia in the subsequent pregnancy. Am J Epidemiol 2000;151:57–62. [DOI] [PubMed] [Google Scholar]
  • 123.Trogstad LI, Eskild A, Magnus P, Samuelsen SO, Nesheim BI. Changing paternity and time since last pregnancy; the impact on preeclampsia risk. A study of 547 238 women with and without previous preeclampsia. Int J Epidemiol 2001;30:1317–22. [DOI] [PubMed] [Google Scholar]
  • 124.Trupin LS, Simon LP, Eskenazi B. Change in paternity: a risk factor for preeclampsia in multiparas. Epidemiology 1996;7:240–4. [DOI] [PubMed] [Google Scholar]
  • 125.Rowe JH, Ertelt JM, Xin L, Way SS. Pregnancy imprints regulatory memory that sustains anergy to fetal antigen. Nature 2012;490:102–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.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. [DOI] [PubMed] [Google Scholar]
  • 127.Molès J-P, Tuaillon E, Kankasa C, Bedin A-S, Nagot N, Marchant A, et al. Breastmilk cell trafficking induces microchimerism-mediated immune system maturation in the infant. Pediatr Allergy Immunol 2018;29:133–43. [DOI] [PubMed] [Google Scholar]
  • 128.Lo YM, Patel P, Sampietro M, Gillmer MD, Fleming KA, Wainscoat JS. Detection of single-copy fetal DNA sequence from maternal blood. Lancet 1990;335: 1463–4. [DOI] [PubMed] [Google Scholar]
  • 129.Bianchi DW. The inadvertent discovery of human fetal cell microchimerism. Clin Chem 2018;64:1400–1. [DOI] [PubMed] [Google Scholar]
  • 130.Breman AM, Chow JC, U’Ren L, Normand EA, Qdaisat S, Zhao L, et al. Evidence for feasibility of fetal trophoblastic cell-based noninvasive prenatal testing. Prenat Diagn 2016;36:1009–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Huang Z, Fong CY, Gauthaman K, Sukumar P, Choolani M, Bongso A. Novel approaches to manipulating foetal cells in the maternal circulation for non-invasive prenatal diagnosis of the unborn child. J Cell Biochem 2011;112:1475–85. [DOI] [PubMed] [Google Scholar]
  • 132.Choolani M, Mahyuddin AP, Hahn S. The promise of fetal cells in maternal blood. Best Pract Res Clin Obstet Gynaecol 2012;26:655–67. [DOI] [PubMed] [Google Scholar]
  • 133.Huang CE, Ma GC, Jou HJ, Lin WH, Lee DJ, Lin YS, et al. Noninvasive prenatal diagnosis of fetal aneuploidy by circulating fetal nucleated red blood cells and extravillous trophoblasts using silicon-based nanostructured microfluidics. Mol Cytogenet 2017;10:44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Sato T, Ito Y, Samura O, Aoki H, Uchiyama T, Okamoto A, Hata K. Direct assessment of single-cell DNA using crudely purified live cells: a proof of concept for noninvasive prenatal definitive diagnosis. J Mol Diagn 2020; 22:132–40. [DOI] [PubMed] [Google Scholar]
  • 135.Bianchi DW, Chiu RW. Sequencing of circulating cell-free DNA during pregnancy. N Engl J Med 2018;379: 464–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Lo YM, Tein MS, Lau TK, Haines CJ, Leung T, Poon PM, et al. Quantitative analysis of fetal DNA in maternal plasma and serum: implications for non-invasive prenatal diagnosis. Am J Hum Genet 1998;62:768–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Rezaei M, Winter M, Zander-Fox D, Whitehead C, Liebelt J, Warkiani ME, et al. A reappraisal of circulating fetal cell noninvasive prenatal testing. Trends Biotechnol 2019;37:632–44. [DOI] [PubMed] [Google Scholar]

RESOURCES