6,131
Views
16
CrossRef citations to date
0
Altmetric
Editorial

Recurrent miscarriages: What is the role of cytokines?

&
Pages 663-668 | Received 11 May 2008, Accepted 20 May 2008, Published online: 07 Jul 2009

Abstract

Recurrent miscarriage is defined as three or more consecutive miscarriages before 20 weeks of gestation. This condition is a frequent reproductive problem worldwide, affecting up to 1% of couples. Immune effector cell dysfunction has been implicated in the pathogenesis of early pregnancy loss. This dysfunction may involve defects in cytokines, growth factors and immunosuppressive factors at the maternofetal interface. This is a growing research field, especially the role of cytokines in recurrent miscarriage.

Introduction

Recurrent miscarriage is defined as three or more consecutive miscarriages before 20 weeks of gestation Citation[1],Citation[2]. This condition is a frequent reproductive problem worldwide, affecting up to 1% of couples Citation[3],Citation[4]. This incidence is greater than one would expect by chance alone. If up to 15% of all clinically recognized pregnancies end in miscarriage, then the theoretical risk of having three consecutive miscarriages is 0.34%Citation[5]. Therefore, there will be a persistent underlying cause for the pregnancy loss in a proportion of women suffering from recurrent miscarriage Citation[6].

The pathophysiology is complex, and involves both maternal and fetal factors, with the possibility of more than one underlying contributory factor Citation[6]. The two main independent risk factors for recurrent miscarriage are advanced maternal age and greater number of previous miscarriages Citation[2]. Maternal underlying mechanisms include uterine pathology, endocrine dysfunction, thrombophilias and antiphospholipid syndrome Citation[7]. Genetic or developmental abnormalities in the fetus can also contribute to recurrent miscarriage Citation[1]. However, up to 50% of cases are idiopathic Citation[1].

Immune effector cell dysfunction has been implicated in the pathogenesis of early pregnancy loss Citation[8]. This dysfunction may involve defects in cytokines, growth factors and immunosuppressive factors at the maternofetal interface Citation[6]. This is a growing research field, especially the role of cytokines in recurrent miscarriage.

What are cytokines?

Cytokines are soluble proteins or glycoproteins that are produced by leukocytes and in many cases also by other cell types. They act as chemical communicators between cells, but are not effector molecules in their own right. A feature that is common among most cytokines is that they are regulators of host defense against pathogens and/or the inflammatory response. Most cytokines are secreted, but some can also be expressed on the cell membrane, while others are held in reservoirs in the extracellular matrix Citation[9].

Cytokines bind to specific receptors on the surface of target cells. These receptors are coupled to intracellular signal transduction and second messenger pathways. Most cytokines are growth and/or differentiation factors. They generally act on cells within the hematopoietic system Citation[9].

There are four key features displayed by most cytokines.

  1. Pleiotropy. Most cytokines have more than one action. This is because their receptors are expressed on multiple cell types. Consequently, the signaling pathways activated will increase gene expression specific for that cell type.

  2. Redundancy. Most cytokines have biological effects also observed in other cytokines. This is because of similar signals generated by different cytokines due to similarities in the amino acid sequences within cytokine receptors.

  3. Potency. Most cytokines act in the nanomolar to femtomolar range. This is because the cytokine receptors have a very high affinity for their ligands.

  4. Action as part of a network or cascade. Most cytokines are part of a cascade of cytokines released in succession. They often act in synergy, and, in most cases, are counter-regulated by inhibitory cytokines or soluble receptors.

Cytokines are grouped into families, depending on the structure of their receptors, which have highly conserved structural features. This provides an explanation for the redundancy in biological effects among cytokines.

There are six major families of cytokine receptors (). Each family is defined by similar sequences in their cytosolic domains. Within each family, there are homologies in receptor sequence both intracellularly and extracellularly. However, there are also differences, particularly extracellularly, where additional domains can be found.

Table I.  The six different families of cytokine receptors (modified from Citation[9])

In general, the functions of cytokines fitting into each family are also conserved Citation[10]. But since cytokines are so pleiotropic, it is difficult to classify them according to function. General principles on the functioning of cytokines within families have emerged. This can provide a framework from which to approach the complexities involved when studying the function of cytokines.

What is the immunology of pregnancy?

Recent advances in reproductive immunology have aimed to elucidate ways in which the fetus evades specific immunological attack. The maternal immune system in pregnancy adapts in such a way that there is a degree of suppression to avoid rejection of fetal (paternal) tissues, while maintaining immune competence to fight infection. There are, however, other potential functions of immune cells and cytokines, which include aiding fetal–maternal communication and maternal recognition of pregnancy, so that appropriate physiological changes can be made, e.g. in the clotting system and vascular endothelium. In fact, some cytokines also act as local growth factors and encourage fetal growth Citation[11].

Medawar had observed that rejection of skin grafts could be prevented by the administration of cortisone and, because he was aware that steroid levels were elevated in pregnancy, he made the ‘fetal allograft’ hypothesis. He postulated that the anatomical separation of the fetus from its mother, the antigenic immaturity of the fetus and the immunological inertness of the mother led to the survival of the fetus Citation[12]. However, an alternative model of pregnancy was also postulated, leading to the Medawar–Shwartzman paradox.

The localized Shwartzman reaction of hemorrhagic necrosis is induced in some animals by a priming injection of endotoxin followed by a sublethal intravenous endotoxin injection 24 h later Citation[13]. Identical pathological lesions are observed in pregnant animals after just a single intravenous injection of endotoxin, without requiring a priming injection Citation[14]. Pregnant women may be similarly prone to exaggerated responses to endotoxin during pregnancy. The ‘priming agent’ may be cortisone itself, through non-specific mechanisms involving monocytes and natural killer (NK) cells Citation[15]. Tumor necrosis factor-α (TNF-α), interferon-γ (IFN-γ) and interleukin-2 (IL-2) have been identified as key cytokines for the generalized Shwartzman reaction Citation[16]. Thus the maternal immune system is suppressed in one model, but primed to respond in the other. The explanation lies in the differential modulation of the two main arms of the immune system: the innate (non-specific) and adaptive (specific), both of which have cellular and humoral components Citation[11].

The innate system is the first line of defense, leading to ‘signal 1’. It also instigates the adaptive system, which is effective at eliminating infection. The antigen is displayed on the cell surface in association with major histocompatibility complex proteins. This triggers the activation of CD4+ helper T-cells (Th cells) which produce cytokines, and in turn coordinate the effector response (‘signal 2’) Citation[17]. In ‘signal 2’, the innate system appears to be able to distinguish between dangerous and non-dangerous Citation[18].

T-helper 1 versus T-helper 2 response

Activated Th cells can differentiate into Th1 and Th2. These have different immune responses via different patterns of cytokine production Citation[19],Citation[20]. Studies on murine and human pregnancy show a strong association between maternal Th1-type immunity and pregnancy loss on one hand and, on the other, Th2-type immunity, together with transforming growth factor-β (TGF-β) secreted by Th3 cells, and successful pregnancy.

Th1 cells secrete TNF-α, TNF-β, IFN-γ and IL-2. Low levels of TNF-α are also derived from some Th2 cells and cytotoxic T-cells. However, since much higher levels of TNF-α are produced during Th1 responses and due to its cytolytic effects contributing to cellular immunity, this cytokine can be regarded as exclusively associated with a Th1 or Th1-like response. Th1-associated cytokines activate macrophages and cell-mediated reactions involved in resisting infections due to intracellular pathogens and in cytotoxic and delayed-type hypersensitivity reactions. This type of reaction is also known as a type 1 or cell-mediated immune response Citation[20],Citation[21].

The Th2 cells stimulate B-cells and eosinophils. The Th2-type cytokines are the following interleukins: IL-4, IL-5, IL-6, IL-10 and IL-13. These are associated with strong antibody responses to infections with extracellular organisms. Such immune responses are also known as type 2 or humoral reactions Citation[20],Citation[21].

Th3 cells are a unique subset of CD4+ helper T-cells usually found in mucosa and are involved in the downregulation of Th1 and other immune cells. In addition, these two responses are known to be antagonistic to each other. In fact, IL-10 acts on the antigen-presenting cells thus inhibiting development of Th1 cells. Similarly, IFN-γ prevents the activation of Th2 cells.

It is hypothesized that during pregnancy, there is a subtle immunological shift to the Th2-type cytokine responses that would suppress the potential harmful effects of the cell-mediated (Th1-type) immune system Citation[22]. Animal studies have shown that powerful Th2-promoting factors such as progesterone Citation[23] and prostaglandin E2 Citation[24],Citation[25] are present in high concentrations at the maternal–fetal interface. In humans, there is a wide variety of cytokines (both Th1 and Th2 types) at the maternal–fetal interface Citation[26],Citation[27]. Cell-specific methods have shown preferential production by trophoblast cells of IL-4 (Th2-type cytokine) rather than IFN-γ, IL-2 or TNF-α (Th1-type) Citation[28].

Thus, pregnancy has been labeled a ‘Th2 phenomenon’. This is substantiated by the evidence that pregnancy tends to ameliorate pre-existing autoimmune cell-mediated conditions Citation[22]. In normal pregnant women, there is significantly higher IL-10 and other Th2 cytokine production by mitogen-activated peripheral blood mononuclear cells Citation[29],Citation[30] and reduced IL-2 Citation[27] and IFN-γ mRNA expression Citation[31] and their corresponding CD4+ and CD8+ T-cells Citation[32].

The Th1 response, especially TNF-α, IFN-γ and IL-2, has been postulated to be detrimental to the survival of the conceptus Citation[33-35]. When administered to pregnant mice, TNF-α, IFN-γ and IL-2 cause miscarriage Citation[36], which is then reversed by anti-TNF-α. In vitro, TNF-α and IL-2 inhibit outgrowth of human trophoblast cells Citation[37] and synergistically stimulate apoptosis of human primary villous trophoblast cells Citation[38]. One hypothesis states that the conceptus protects itself by secreting type 2 cytokines which downregulate harmful type 1 cytokines Citation[22].

What is the role of cytokines in recurrent miscarriage?

It is clear from the literature that cytokines do play an important role in recurrent miscarriage. In women with a history of recurrent miscarriage, embryotoxic activity was detected in supernatants from trophoblast-activated peripheral blood mononuclear cells, which were positive for TNF-α, TNF-β and IFN-γ. In contrast, normal women and men neither were embryotoxic nor contained Th1-type cytokines, but most contained the Th2-type cytokines IL-10 and IL-4 Citation[39]. Thus type-1 immunity to trophoblast antigens is associated with recurrent miscarriage. Mitogen-activated peripheral blood mononuclear cells derived from women with a history of recurrent miscarriage had higher levels of IL-2, IFN-γ and TNF-α, and lower levels of IL-4, IL-5, IL-6 and IL-10 than in normal pregnant women, indicating a greater type 1 bias in recurrent miscarriage and a greater type 2 bias in normal pregnancy Citation[40],Citation[41].

It has been shown that there is a pro type-2 shift in ratios of cytokine expression at the maternal–fetal interface in women with normal pregnancy as compared with women who suffer recurrent miscarriage Citation[27],Citation[42]. Th2-type cytokines are released spontaneously from placental tissue in culture Citation[43]. This has been shown in animal studies, together with the observation that pregnancy loss can be caused by Th1 cytokines Citation[36] and prevented by Th2 cytokines Citation[44].

An important factor in driving naive CD4+ T-cells toward Th1- or Th2-dominated populations is the predominance of a given cytokine in the microenvironment at the time of antigen presentation. Th1 differentiation is promoted by IFN-γ and IL-12, while IL-4 determines Th2 polarization Citation[20],Citation[21]. IL-12 is produced mainly by macrophages in response to certain microbial infections; therefore even subclinical infections can lead to increased IL-12 production resulting in an overall shift towards type 1 bias. Th1 development is also enhanced by IFN-γ. IL-12 and viral infections both lead to increased IFN-γ synthesis by NK cells, in turn enhancing Th1 response.

The conceptus may be considered an ‘innocent bystander’ while the mother fights infection with a vigorous Th1-type proinflammatory response Citation[34]. Women with recurrent miscarriage have significantly high levels of IL-12 compared with normal pregnant women Citation[45]. Hormones like relaxin (produced by the corpus luteum) may also contribute to differentiation of Th cells or in favoring the shift of already differentiated Th cells to a type 1 profile Citation[46]. On the other hand, progesterone has been shown to promote the development of a Th2 response, via an immunomodulatory protein known as progesterone-induced blocking factor, which inhibits several Th1-type responses in vitro Citation[47],Citation[48]. If there is a lack of suppression at the maternofetal interface, due to reduced levels of TGF-β2, a Th1 shift may be induced Citation[49].

It has been proposed that Th1 immune-mediated damage could be due to NK cells, similar to activated Th1 cells, releasing cytokines harmful to the trophoblast Citation[50]. Decidual NK cells produce IFN-γ, activating decidual macrophages to produce toxic levels of nitric oxide and TNF-αCitation[51]. Th1 cytokines may convert the NK cells to lymphokine-activated killer cells that have been shown to lyse trophoblasts Citation[19].

TNF-α and IFN-γ can lead to apoptosis of trophoblast cells Citation[38], inhibition of secretion of granulocyte macrophage colony-stimulating factor from uterine epithelium Citation[22] and upregulation of the prothrombinase fg12 (procoagulant). The latter may be the most important effect as anti-fg12 antibodies significantly reduce the effect of TNF-α and IFN-γ and prevent miscarriage. It is proposed that miscarriage of normal karyotype human embryos could be due to a process of ‘cytokine-triggered vascular auto-amputation’Citation[52]. This involves activation of coagulation mechanisms leading to vasculitis, affecting the maternal blood supply to the implanted embryo and thrombus formation Citation[53].

Evidence shows that TNF-α, IFN-γ and NK cells cannot induce miscarriage separately, but a Th1–NK cell–macrophage triad is known to bring about miscarriage, which can in turn be suppressed by a Th2 cytokine response Citation[22],Citation[54]. One should note that, depending on their time of expression, stage of gestation, relative concentrations and receptor occupancy threshold, some type-1 cytokines may actually be beneficial rather than harmful to the pregnancy Citation[42].

Conclusion

The role of cytokines in recurrent miscarriage has become clearer over recent years. However, more research still needs to be done before one can devise treatment options for patients with recurrent miscarriage, based on the information available so far.

Declaration of interest: The authors report no conflicts of interest. The authors alone are responsible for the content and writing of the paper.

References

  • Szekeres-Bartho J, Balasch J. Progestagen therapy for recurrent miscarriage. Hum Reprod Update 2008; 14: 27–35
  • Nybo Andersen A M, Wohlfahrt J, Christens P, Olsen J, Melbye M. Maternal age and fetal loss: population based register linkage study. BMJ 2000; 320: 1708–1712
  • Stirrat G M. Recurrent miscarriage. Lancet 1990; 336: 673–675
  • Rai R, Regan L. Recurrent miscarriage. Lancet 2006; 368: 601–611
  • Alberman E. The epidemiology of repeated abortion. Early pregnancy loss: mechanisms and treatment, R W Beard, F Sharp. RCOG Press, London 2008; 9–17
  • Royal College of Obstetricians and Gynaecologists. The Investigation and Treatment of Couples with Recurrent Miscarriage [Internet]. RCOG, London May, 2003, [cited]. Available from: http://www.rcog.org.uk/resources/Public/pdf/Recurrent_Miscarriage_No17.pdf
  • Regan L, Rai R. Thrombophilia and pregnancy loss. J Reprod Immunol 2002; 55: 163–180
  • King A. Uterine leukocytes and decidualization. Hum Reprod Update 2000; 6: 28–36
  • Fitzgerald K A, O'Neill L AJ, Gearing A JH, Callard R E. The cytokine facts book. Academic Press, London 2001
  • Taniguchi T. Cytokine signaling through nonreceptor protein tyrosine kinases. Science 1995; 268: 251–255
  • Sacks G P, Redman C, Sargent I. The immunology of human pregnancy. Progress in obstetrics and gynaecology15th ed, J Studd. Elsevier Science Limited, Amsterdam 2003; 17–32
  • Medawar P. Some immunological and endocrinological problemsraised by the evolution of viviparity in vertebrates. Symp Soc Exp Biol 1953; 7: 320–338
  • Shwartzman G. A new phenomenon of local skin reactivity to B. typhosus culture filtrate. Proc Soc Exp Biol Med 1928; 151: 560–561
  • Beller F K, Schmidt E H, Holzgreve W, Hauss J. Septicemia during pregnancy: a study in different species of experimental animals. Am J Obstet Gynecol 1985; 151: 967–975
  • Mori W. The Shwartzman reaction: a review including clinical manifestations and proposal for a univisceral or single organ third type. Histopathology 1981; 5: 113–126
  • Ozmen L, Pericin M, Hakimi J, Chizzonite R A, Wysocka M, Trinchieri G, Gately M, Garotta G. Interleukin 12, interferon γ, and tumor necrosis factor α are the key cytokines of the generalized Shwartzman reaction. J Exp Med 1994; 180: 907–915
  • Fearon D T. Seeking wisdom in innate immunity. Nature 1997; 388: 323–324
  • Matzinger P. Tolerance, danger, and the extended family. Annu Rev Immunol 1994; 12: 991–1045
  • Raghupathy R. Pregnancy: success and failure within the Th1/Th2/Th3 paradigm. Semin Immunol 2001; 13: 219–227
  • Mosmann T R, Sad S. The expanding universe of T-cell subsets: Th1, Th2 and more. Immunol Today 1996; 17: 138–146
  • Coffman R L, Romagnani S. Redirection of Th1 and Th2 responses. Springer-Verlag, Berlin 1999
  • Wegmann T G, Lin H, Guilbert L, Mosmann T R. Bidirectional cytokine interactions in the maternal–fetal relationship: is successful pregnancy a TH2 phenomenon. Immunol Today 1993; 14: 353–356
  • Piccinni M P, Giudizi M G, Biagiotti R, Beloni L, Giannarini L, Sampognaro S, Parronchi P, Manetti R, Annunziato F, Livi C, et al. Progesterone favors the development of human T helper cells producing Th2-type cytokines and promotes both IL-4 production and membrane CD30 expression in established Th1 cell clones. J Immunol 1995; 155: 128–133
  • Kelly R W, Carr G G, Elliott C L, Tulppala M, Critchley H O. Prostaglandin and cytokine release by trophoblastic villi. Hum Reprod 1995; 10: 3289–3292
  • Kelly R W, Critchley H O. A T-helper-2 bias in decidua: the prostaglandin contribution of the macrophage and trophoblast. J Reprod Immunol 1997; 33: 181–187
  • Robertson S A, Seamark R F, Guilbert L J, Wegmann T G. The role of cytokines in gestation. Crit Rev Immunol 1994; 14: 239–292
  • Vince G S, Johnson P M. Is there a Th2 bias in human pregnancy?. J Reprod Immunol 1996; 32: 101–104
  • Sacks G P, Clover L M, Bainbridge D R, Redman C W, Sargent I L. Flow cytometric measurement of intracellular Th1 and Th2 cytokine production by human villous and extravillous cytotrophoblast. Placenta 2001; 22: 550–559
  • Hanna N, Hanna I, Hleb M, Wagner E, Dougherty J, Balkundi D, Padbury J, Sharma S. Gestational age-dependent expression of IL-10 and its receptor in human placental tissues and isolated cytotrophoblasts. J Immunol 2000; 164: 5721–5728
  • Marzi M, Vigano A, Trabattoni D, Villa M L, Salvaggio A, Clerici E, Clerici M. Characterization of type 1 and type 2 cytokine production profile in physiologic and pathologic human pregnancy. Clin Exp Immunol 1996; 106: 127–133
  • Kruse N, Greif M, Moriabadi N F, Marx L, Toyka K V, Rieckmann P. Variations in cytokine mRNA expression during normal human pregnancy. Clin Exp Immunol 2000; 119: 317–322
  • Reinhard G, Noll A, Schlebusch H, Mallmann P, Ruecker A V. Shifts in the TH1/TH2 balance during human pregnancy correlate with apoptotic changes. Biochem Biophys Res Commun 1998; 245: 933–938
  • Raghupathy R. Th1-type immunity is incompatible with successful pregnancy. Immunol Today 1997; 18: 478–482
  • Clark D A, Arck P C, Chaouat G. Why did your mother reject you? Immunogenetic determinants of the response to environmental selective pressure expressed at the uterine level. Am J Reprod Immunol 1999; 41: 5–22
  • Hill J A, Polgar K, Harlow B L, Anderson D J. Evidence of embryo- and trophoblast-toxic cellular immune response(s) in women with recurrent spontaneous abortion. Am J Obstet Gynecol 1992; 166: 1044–1052
  • Chaouat G, Menu E, Clark D A, Dy M, Minkowski M, Wegmann T G. Control of fetal survival in CBA×DBA/2 mice by lymphokine therapy. J Reprod Fertil 1990; 89: 447–458
  • Haimovici F, Hill J A, Anderson D J. The effects of soluble products of activated lymphocytes and macrophages on blastocyst implantation events in vitro. Biol Reprod 1991; 44: 69–75
  • Yui J, Garcia-Lloret M, Wegmann T G, Guilbert L J. Cytotoxicity of tumour necrosis factor-α and γ-interferon against primary human placental trophoblasts. Placenta 1994; 15: 819–835
  • Hill J A, Polgar K, Anderson D J. T-helper 1-type immunity to trophoblast in women with recurrent spontaneous abortion. JAMA 1995; 273: 1933–1936
  • Makhseed M, Raghupathy R, Azizieh F, Al Azemi M M, Hassan N A, Bandar A. Mitogen-induced cytokine responses of maternal peripheral blood lymphocytes indicate a differential Th-type bias in normal pregnancy and pregnancy failure. Am J Reprod Immunol 1999; 42: 273–281
  • Raghupathy R, Makhseed M, Azizieh F, Omu A, Gupta M, Farhat R. Cytokine production by maternal lymphocytes during normal human pregnancy and in unexplained recurrent spontaneous abortion. Hum Reprod 2000; 15: 713–718
  • Guilbert L J. There is a bias against type 1 (inflammatory) cytokine expression and function in pregnancy. J Reprod Immunol 1996; 32: 105–110
  • Lin H, Mosmann T R, Guilbert L, Tuntipopipat S, Wegmann T G. Synthesis of T helper 2-type cytokines at the maternal–fetal interface. J Immunol 1993; 151: 4562–4573
  • Chaouat G, Assal M A, Martal J, Raghupathy R, Elliott J F, Mosmann T, Wegmann T G. IL-10 prevents naturally occurring fetal loss in the CBA×DBA/2 mating combination, and local defect in IL-10 production in this abortion-prone combination is corrected by in vivo injection of IFN-τ. J Immunol 1995; 154: 4261–4268
  • Wilson R, McInnes I, Leung B, McKillop J H, Walker J J. Altered interleukin 12 and nitric oxide levels in recurrent miscarriage. Eur J Obstet Gynecol Reprod Biol 1997; 75: 211–214
  • Piccinni M P, Romagnani S. Regulation of fetal allograft survival by a hormone-controlled Th1- and Th2-type cytokines. Immunol Res 1996; 15: 141–150
  • Szekeres-Bartho J, Chaouat G. Lymphocyte-derived progesterone-induced blocking factor corrects resorption in a murine abortion system. Am J Reprod Immunol 1990; 23: 26–28
  • Szekeres-Bartho J, Wegmann T G. A progesterone-dependent immunomodulatory protein alters the Th1/Th2 balance. J Reprod Immunol 1996; 31: 81–95
  • Lea R G, Underwood J, Flanders K C, Hirte H, Banwatt D, Finotto S, Ohno I, Daya S, Harley C, Michel M, et al. A subset of patients with recurrent spontaneous abortion is deficient in transforming growth factor β-2-producing ‘suppressor cells’ in uterine tissue near the placental attachment site. Am J Reprod Immunol 1995; 34: 52–64
  • Baines M G, Duclos A J, Antecka E, Haddad E K. Decidual infiltration and activation of macrophages leads to early embryo loss. Am J Reprod Immunol 1997; 37: 471–477
  • Haddad E K, Duclos A J, Lapp W S, Baines M G. Early embryo loss is associated with the prior expression of macrophage activation markers in the decidua. J Immunol 1997; 158: 4886–4892
  • Clark D A, Chaouat G, Arck P C, Mittruecker H W, Levy G A. Cytokine-dependent abortion in CBA×DBA/2 mice is mediated by the procoagulant fgl2 prothrombinase [correction of prothombinase]. J Immunol 1998; 160: 545–549
  • Clark D A, Ding J W, Chaouat G, Coulam C B, August C, Levy G A. The emerging role of immunoregulation of fibrinogen-related procoagulant Fgl2 in the success or spontaneous abortion of early pregnancy in mice and humans. Am J Reprod Immunol 1999; 42: 37–43
  • Arck P, Dietl J, Clark D. From the decidual cell internet: trophoblast-recognizing T cells. Biol Reprod 1999; 60: 227–233

Reprints and Corporate Permissions

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

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

Academic Permissions

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

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

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