Hostname: page-component-8448b6f56d-jr42d Total loading time: 0 Render date: 2024-04-23T14:22:39.053Z Has data issue: false hasContentIssue false

Immunology of pregnancy: cellular mechanisms allowing fetal survival within the maternal uterus

Published online by Cambridge University Press:  27 April 2007

Ana Claudia Zenclussen*
Affiliation:
AG Reproduktionsimmunologie, Institut für Medizinische Immunologie, Charite, Medizinische Universität zu Berlin, Germany.
Anne Schumacher
Affiliation:
AG Reproduktionsimmunologie, Institut für Medizinische Immunologie, Charite, Medizinische Universität zu Berlin, Germany.
Maria Laura Zenclussen
Affiliation:
AG Reproduktionsimmunologie, Institut für Medizinische Immunologie, Charite, Medizinische Universität zu Berlin, Germany.
Paul Wafula
Affiliation:
AG Reproduktionsimmunologie, Institut für Medizinische Immunologie, Charite, Medizinische Universität zu Berlin, Germany.
Hans-Dieter Volk
Affiliation:
AG Reproduktionsimmunologie, Institut für Medizinische Immunologie, Charite, Medizinische Universität zu Berlin, Germany.
*
*Corresponding author: Ana Claudia Zenclussen, AG Reproduktionsimmunologie, Institut für Medizinische Immunologie, Charite, Medizinische Universität zu Berlin, CVK, Biomedizinisches Forschungszentrum, Raum 2.0534, Augustenburger Platz 1, 13353 Berlin, Germany. Tel: +49 30 450 559886; Fax: +49 30 450 559986; E-mail: ana.zenclussen@charite.de

Abstract

Pregnancy success remains a fascinating phenomenon to immunologists as it defies the immunological rules of rejection. Although it was previously thought that the maternal immune system does not see the fetus, it is now well documented that fetal cells reach the maternal body and encounter host immune cells. Natural tolerance mechanisms following this interaction remain to be fully elucidated. This article reviews the current literature on mechanisms of adaptive immunity, with emphasis on regulatory T cells and heme oxygenase 1 (HO-1). We propose a scenario in which regulatory T cells create a tolerant microenvironment at the fetal–maternal interface characterised by the presence of tolerance-associated molecules such as HO-1, which has been shown to be of vital importance for fetal survival.

Type
Research Article
Copyright
Copyright © Cambridge University Press 2007

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

References

1Medawar, P.B. (1953) Some immunological and endocrinological problems raised by the evolution of viviparity in vertebrates. Symp Soc Exper Biol 44, 320-338Google Scholar
2Billington, W.D. (1992) The normal fetomaternal immune relationship. Baillieres Clin Obstet Gynaecol 6, 417-438CrossRefGoogle ScholarPubMed
3Andrassy, J. et al. (2003) Tolerance to noninherited maternal MHC antigens in mice. J Immunol 171, 5554-5561CrossRefGoogle ScholarPubMed
4Yan, Z. et al. (2005) Male microchimerism in women without sons: quantitative assessment and correlation with pregnancy history. Am J Med 118, 899-906CrossRefGoogle ScholarPubMed
5Khoshrotehran, K. et al. (2005) Natural history of fetal cell microchimerism during and following murine pregnancy. J Reprod Immunol 66, 1-12CrossRefGoogle Scholar
6Nelson, J.L. (1998) Pregnancy, persistent microchimerism, and autoimmune disease. J Am Med Womens Assoc 53, 31-32Google ScholarPubMed
7Chaouat, G., Kolb, J.P. and Wegmann, T.G. (1983) The murine placenta as an immunological barrier between the mother and the fetus. Immunol Rev 75, 31-60CrossRefGoogle ScholarPubMed
8Wegmann, T.G. (1987) Placental immunotrophism: maternal T cells enhance placental growth and function. Am J Reprod Immunol Microbiol 15, 67-69CrossRefGoogle Scholar
9Elbe-Burger, A. et al. (2000) Major histocompatibility complex class II- fetal skin dendritic cells are potent accessory cells of polyclonal T-cell responses. Immunology 101, 242-253CrossRefGoogle ScholarPubMed
10Tafuri, A. et al. (1995) T cell awareness of paternal alloantigens during pregnancy. Science 270, 630-633CrossRefGoogle ScholarPubMed
11Jiang, S.P. and Vacchio, M.S. (1998) Multiple mechanisms of peripheral T cell tolerance to the fetal “allograft”. J Immunol 160, 3086-309CrossRefGoogle Scholar
12Hunt, J.S. et al. (1988) Expression of class I HLA genes by trophoblast cells. Analysis by in situ hybridization. J Immunol 140, 1293-1299CrossRefGoogle Scholar
13Hunt, J.S. and Orr, H.T. (1992) HLA and maternal-fetal recognition. FASEB J 6, 2344-2348CrossRefGoogle ScholarPubMed
14Redline, R.W. and Lu, C.Y. (1989) Localization of fetal major histocompatibility complex antigens and maternal leukocytes in murine placenta. Implications for maternal-fetal immunological relationship. Lab Invest 61, 27-36Google ScholarPubMed
15Zuckermann, F.A. and Head, J.R. (1986) Expression of MHC antigens on murine trophoblast and their modulation by interferon. J Immunol 137, 846-853CrossRefGoogle ScholarPubMed
16Le Bouteiller, P. and Lenfant, F. (1996) Antigen-presenting function(s) of the nonclassical HLA-E, -F and -G class I molecules: the beginning of a story. Res Immunol 147, 301-313CrossRefGoogle Scholar
17Gardner, L. and Moffett, A. (2003) Dendritic cells in the human decidua. Biol Reprod 69, 1438-1446CrossRefGoogle ScholarPubMed
18Sasaki, Y. et al. (2004) Decidual and peripheral blood CD4+ CD25+  regulatory T cells in early pregnancy subjects and spontaneous abortion cases. Mol Hum Reprod 10, 347-353CrossRefGoogle ScholarPubMed
19Shiroishi, M. et al. (2006) Structural basis for recognition of the nonclassical MHC molecule HLA-G by the leukocyte Ig-like receptor B2 (LILRB2/LIR2/ILT4/CD85d). Proc Natl Acad Sci U S A 103, 16412-16417CrossRefGoogle ScholarPubMed
20Chang, C.C. et al. (2002) Tolerization of dendritic cells by T(S) cells: the crucial role of inhibitory receptors ILT3 and ILT4. Nat Immunol 3, 237-243CrossRefGoogle Scholar
21Velten, F.W. et al. (2004) A gene signature of inhibitory MHC receptors identifies a BDCA3(+) subset of IL-10-induced dendritic cells with reduced allostimulatory capacity in vitro. Eur J Immunol 34, 2800-2811CrossRefGoogle ScholarPubMed
22Zhang, J., Croy, B.A. and Tian, Z. (2005) Uterine natural killer cells: their choices, their missions. Cell Mol Immunol 2, 123-129Google ScholarPubMed
23Munn, D.H. et al. (1998) Prevention of allogeneic fetal rejection by tryptophan catabolism. Science 281, 1191-1193CrossRefGoogle ScholarPubMed
24Guleria, I. et al. (2005) A critical role for the programmed death ligand 1 in fetomaternal tolerance. J Exp Med 202, 231-237CrossRefGoogle ScholarPubMed
25Zhu, X.Y. et al. (2005) Blockade of CD86 signaling facilitates a Th2 bias at the maternalfetal interface and expands peripheral CD4+ CD25+  regulatory T cells to rescue abortion-prone fetuses. Biol Reprod 72, 338-345CrossRefGoogle ScholarPubMed
26Malan Borel, I. et al. (1991) IgG asymmetric molecules with anti-paternal activity isolated from sera and placental of pregnant human. J Reprod Immunol 20, 129-140CrossRefGoogle Scholar
27Lin, H. et al. (1993) Synthesis of T helper 2-type cytokines at the feto-maternal interfase. J Immunol 151, 4562-4573CrossRefGoogle Scholar
28Tangri, S. and Raghupathy, R. (1993) Expression of cytokines in placentas of mice undergoing immunologically mediated spontaneous fetal resorption. Biol Reprod 49, 850-856CrossRefGoogle Scholar
29Aluvihare, V., Kallikourdis, M. and Betz, A. (2004) Regulatory T cells mediate maternal tolerance to the fetus. Nat Immunol 3, 266-271CrossRefGoogle Scholar
30Zenclussen, A.C. et al. (2005) Abnormal T cell reactivity against paternal antigens in spontaneous abortion: Adoptive transfer of pregnancy-induced CD4+ CD25+  T regulatory cells prevents fetal rejection in a murine abortion model. Am J Pathol 166, 811-822CrossRefGoogle Scholar
31Stewart, C.L. et al. (1992) Blastocyst implantation depends on maternal expression of leukaemia inhibitory factor. Nature 359, 76-79CrossRefGoogle ScholarPubMed
32Zenclussen, M.L. et al. (2006) Overexpression of heme oxygenase-1 by adenoviral gene transfer improves pregnancy outcome in a murine model of abortion. J Reprod Immunol 69, 35-52CrossRefGoogle Scholar
33Abrahams, V.M. et al. (2005) A role for TLRs in the regulation of immune cell migration by first trimester trophoblast cells. J Immunol 175, 8096-8104CrossRefGoogle ScholarPubMed
34Fest, S. et al. (2007) Trophoblast–macrophage interactions: a regulatory network for the protection of pregnancy. Am J Reprod Immunol 57, 55-66CrossRefGoogle Scholar
35Baban, B. et al. (2004) Indoleamine 2,3-dioxygenase expression is restricted to fetal trophoblast giant cells during murine gestation and is maternal genome specific. J Reprod Immunol 61, 67-77CrossRefGoogle ScholarPubMed
36Svensson, L. et al. (2001) The Th2 cytokines IL-4 and IL-10 are not crucial for the completion of allogeneic pregnancy in mice. J Reprod Immunol 51, 3-7CrossRefGoogle Scholar
37Fallon, P.G. et al. (2002) IL-4 induces characteristic Th2 even in the combined absence of IL-5, IL-9 and IL-13. Immunity 17, 7-17CrossRefGoogle ScholarPubMed
38Raghupathy, R. (1997) Th1-type immunity is incompatible with successful pregnancy. Immunol Today 18, 478-482CrossRefGoogle ScholarPubMed
39Saito, S. (2001) Cytokine network at the feto-maternal interface. J Reprod Immunol 47, 87-103CrossRefGoogle Scholar
40Piccinni, M.P. et al. (1998) Defective production of both, leukemia inhibitor factor and type 2 T-helper cytokines by decidual T cells in unexplained recurrent abortions. Nat Med 4, 1020-1024CrossRefGoogle ScholarPubMed
41Zenclussen, A.C. et al. (2003) Murine abortion is associated with enhanced interleukin-6 levels at the feto-maternal interface. Cytokine 24, 150-160CrossRefGoogle ScholarPubMed
42Gorivodsky, M. et al. (1999) TGF beta 2 mRNA expression and pregnancy failure in mice. Am J Reprod Immunol 42, 124-133CrossRefGoogle ScholarPubMed
43Dünker, N. and Krieglstein, K. (2002) Tgfbeta2 -/-  Tgfbeta3 -/- double knockout mice display severe midline fusion defects and early embryonic lethality. Anat Embryol 206, 73-83Google ScholarPubMed
44Dealtry, G., O'Farrell, M. and Fernandez, N. (2000) The Th2 cytokine environment of the placenta. Int Arch Allergy Immunol 123, 107-119CrossRefGoogle ScholarPubMed
45Chaouat, G. et al. (1995) IL-10 prevents naturally occurring fetal loss in the CBA x DBA/2 mating combination, and local defect in IL-10 production in this abortion-prone combination is corrected by in vivo injection of IFN-tau. J Immunol 154, 4261-4268CrossRefGoogle ScholarPubMed
46White, C.A. et al. (2004) Effect of interleukin-10 null mutation on maternal immune response and reproductive outcome in mice. Biol Reprod 70, 123-131CrossRefGoogle ScholarPubMed
47Kruse, A. et al. (1999) Evidence of specialized leukocyte-vascular homing interactions at the maternal/foetal interface. Eur J Immunol 29, 1116-11263.0.CO;2-4>CrossRefGoogle Scholar
48Austrup, D. et al. (1997) P- and E-selectin mediate recruitment of T-helper-1 but not T-helper-2 cells into inflamed tissue. Nature 385, 81-83CrossRefGoogle ScholarPubMed
49Bertoja, A.Z. et al. (2005) Anti-P and E-selectin therapy prevents abortion in the CBA/J x DBA/2J combination by blocking the migration of Th1 lymphocytes into the foetal-maternal interface. Cell Immunol 238, 97-102CrossRefGoogle ScholarPubMed
50Zenclussen, A.C. et al. (2001) Upregulation of decidual P-selectin expression is associated with an increased number of Th1 cell populations in patients suffering from spontaneous abortion. Cell Immunol 213, 94-103CrossRefGoogle Scholar
51Zenclussen, A.C. et al. (2004) Introducing a mouse model for pre-eclampsia: adoptive transfer of activated Th1 cells leads to pre-eclampsia-like symptoms exclusively in pregnant mice. Eur J Immunol 34, 377-387CrossRefGoogle ScholarPubMed
52Pijnenborg, R. et al. (1996) Attachment and differentiation in vitro of trophoblast from normal and preeclamptic human placentas. Am J Obstet Gynecol 175, 30-36CrossRefGoogle ScholarPubMed
53Faas, M.M. et al. (1994) A new animal model for human preeclampsia: ultralow dose endotoxin infusion in pregnant rats. Am J Obstet Gynecol 171, 158-164CrossRefGoogle Scholar
54Akira, S., Taga, T. and Kishimoto, T. (1993) Interleukin-6 in biology and medicine. Adv Immunol 54, 1-78CrossRefGoogle ScholarPubMed
55Nishino, E. et al. (1990) Trophoblast-derived interleukin-6 (IL-6) regulates human chorionic gonadotropin release through IL-6 receptor on human trophoblasts. J Clin Endocrinol Metab 71, 436-441CrossRefGoogle ScholarPubMed
56Wu, M.Y. et al. (2001) Mouse embryo toxicity of IL-6 in peritoneal fluids from women with or without endometriosis. Acta Obstet Gynecol Scand 80, 7-11Google ScholarPubMed
57Evans, R. et al. (1992) Synergistic interaction of bacterial lipopolysaccharide and the monocyte-macrophage colony-stimulating factor, potential quantitative and qualitative changes in macrophage-produced cytokine bioactivity. J Leukoc Biol 51, 93-96CrossRefGoogle ScholarPubMed
58Wang, Y.Y., Tawfik, O. and Wood, G.W. (1998) Endotoxin-induced abortion in mice is mediated by activated fetal macrophages. J Leukoc Biol 63, 40-50CrossRefGoogle ScholarPubMed
59Meisser, A. et al. (1999) Effects of interleukin-6 (IL-6) on cytotrophoblastic cells. Mol Hum Reprod 5, 1055-1058CrossRefGoogle ScholarPubMed
60Margni, R.A. and Zenclussen, A.C. (2001) During pregnancy, in the context of a Th2 type cytokine prolife, serum IL-6 levels might condition the quality of the synthesized antibodies. Am J Reprod Immunol 46, 181-187CrossRefGoogle Scholar
61Margni, R.A. and Malan Borel, I. (1998) Paradoxical behavior of asymmetric IgG antibodies. Immunol Rev 163, 77-87CrossRefGoogle ScholarPubMed
62Zenclussen, A.C. et al. (2001) Asymmetric antibodies and pregnancy. Am J Reprod Immunol 45, 289-294CrossRefGoogle ScholarPubMed
63Zenclussen, A.C. et al. (2000) Interleukin-6 and soluble interleukin-6 receptor serum levels in recurrent spontaneous abortion women immunized with paternal white cells. Am J Reprod Immunol 44, 22-29CrossRefGoogle ScholarPubMed
64Bettelli, E. et al. (2006) Reciprocal developmental pathways for the generation of pathogenic effector Th17 and regulatory T cells. Nature 441, 235-238CrossRefGoogle ScholarPubMed
65Sakaguchi, S. (2004) Naturally arising CD4+ regulatory T cells for immunologic self-tolerance and negative control of immune responses. Annu Rev Immunol 22, 531-562CrossRefGoogle ScholarPubMed
66Waldmann, H. et al. (2004) Regulatory T cells and organ transplantation. Semin Immunol 16, 119-126CrossRefGoogle ScholarPubMed
67Sakaguchi, S. et al. (1995) Immunological self-tolerance maintained by activated T-cells expressing IL-2 receptor-chains (CD25). Breakdown of a single mechanism of self tolerance causes various auto-immune diseases. J Immunol 155, 1151-1164CrossRefGoogle Scholar
68Chaouat, G. and Voisin, G.A. (1981) Regulatory T cells in pregnancy. V. Allopregnancy-induced T-cell-suppressor factor is H-2 restricted and bears Ia determinants. Cell Immunol 62, 186-95CrossRefGoogle ScholarPubMed
69Somerset, D.A. et al. (2004) Normal human pregnancy is associated with an elevation in the immune suppressive CD25+  CD4+  regulatory T-cell subset. Immunology 112, 38-43CrossRefGoogle ScholarPubMed
70Sasaki, Y. et al. (2004) Decidual and peripheral blood CD4+ CD25+  regulatory T cells in early pregnancy subjects and spontaneous abortion cases. Mol Hum Reprod 10, 347-353CrossRefGoogle ScholarPubMed
71Karim, M. et al. (2004) Alloantigen-induced CD25+ CD4+  regulatory T cells can develop in vivo from CD25-CD4+  precursors in a thymus-independent process. J Immunol 172, 923-928CrossRefGoogle Scholar
72Zenclussen, A.C. (2006) Regulatory T cells induce a privileged tolerant microenvironment at the fetal-maternal interface. Eur J Immunol 36, 82-94CrossRefGoogle ScholarPubMed
73Sollwedel, A. et al. (2005) Protection from abortion by HO-1 up-regulation is associated with increased levels of Bag-1 and neuropilin-1 at the fetal-maternal interface. J Immunol 175, 4875-4885CrossRefGoogle ScholarPubMed
74Metcalfe, S.M. and De, SMuthukumarana, P.A. (2005) Transplantation tolerance: gene expression profiles comparing allotolerance vs. allorejection. Int Immunopharmacol 5, 33-39CrossRefGoogle ScholarPubMed
75Abraham, N.G., Friedland, M.L. and Levere, R.D. (1983) Heme metabolism in hepatic and erythroid cells. In Progress in Hematology (Brown, E., ed.), pp. 75-130, Grune & Stratton, Inc. (G&S), New YorkGoogle Scholar
76Abraham, N.G. et al. (1988) The physiological significance of heme oxygenase. Int J Biochem 20, 543-558CrossRefGoogle ScholarPubMed
77Beri, R. and Chandra, R. (1993) Chemistry and biology of heme. Effect of metal salts, organometals and metalloporphyrins on heme synthesis and catabolism, with special reference to clinical implications and interactions with cytochrome P-450. Drug Metab Rev 25, 49-152CrossRefGoogle ScholarPubMed
78Sassa, S. and Nagai, T. (1996) The role of heme in gene expression. Int J Hematol 63, 167-178CrossRefGoogle ScholarPubMed
79Ponka, P. (1999) Cell biology of heme. Am J Med Sci 318, 241-256CrossRefGoogle ScholarPubMed
80Vercellotti, G.M. et al. (1994) Heme and the vasculature: an oxidative hazard that induces antioxidant defenses in the endothelium. Artif Cells Blood Substit Immobil Biotechnol 22, 207-213CrossRefGoogle ScholarPubMed
81Jeney, V. et al. (2002) Pro-oxidant and cytotoxic effects of circulating heme. Blood 100, 879-887CrossRefGoogle ScholarPubMed
82Balla, G. et al. (1991) Hemin: a possible physiological mediator of low density lipoprotein oxidation and endothelial injury. Arterioscler Thromb 11, 1700-1711CrossRefGoogle ScholarPubMed
83Ryter, S.W. and Tyrrell, R.M. (2000) The heme synthesis and degradation pathways: role in oxidant sensitivity. Heme oxygenase has both pro- and antioxidant properties. Free Radic Biol Med 28, 289-309CrossRefGoogle ScholarPubMed
84Otterbein, L.E. and Choi, A.M. (2000) Heme oxygenase: colors of defense against cellular stress. Am J Physiol Lung Cell Mol Physiol 279, L1029-L1037CrossRefGoogle ScholarPubMed
85Yesilkaya, A., Altinayak, R. and Korgun, D.K. (2000) The antioxidant effect of free bilirubin on cumene-hydroperoxide treated human leukocytes. Gen Pharmacol 35, 17-20CrossRefGoogle ScholarPubMed
86Vedernikov, Y.P., Graser, T. and Vanin, A.F. (1989) Similar endothelium-independent arterial relaxation by carbon monoxide and nitric oxide. Biomed Biochim Acta 48, 601-603Google ScholarPubMed
87Ryter, S.W. and Choi, A.M. (2002) Heme oxygenase-1: molecular mechanisms of gene expression in oxygen-related stress. Antioxid Redox Signal 4, 625-632CrossRefGoogle ScholarPubMed
88Maines, M.D. (1997) The heme oxygenase system: a regulator of second messenger gases. Annu Rev Pharmacol Toxicol 37, 517-554CrossRefGoogle ScholarPubMed
89Montellano, P.R. (2000) The mechanism of heme oxygenase. Curr Opin Chem Biol 4, 221-227CrossRefGoogle ScholarPubMed
90Morse, D. and Choi, A.M. (2002) Heme oxygenase-1. The “emerging molecule” has arrived. Am J Respir Cell Mol Biol 27, 8-16CrossRefGoogle ScholarPubMed
91Li, X. and Clark, J.D. (2000) Chronic morphine exposure and the expression of heme oxygenase type 2. Brain Res Mol Brain Res 75, 179-184CrossRefGoogle ScholarPubMed
92Liu, N. et al. (2000) Developmentally regulated expression of two transcripts for heme oxygenase-2 with a first exon unique to rat testis: control by corticosterone of the oxygenase protein expression. Gene 241, 175-183CrossRefGoogle ScholarPubMed
93Maines, M.D. and Panahian, N. (2001) The heme oxygenase system and cellular defense mechanisms. Do HO-1 and HO-2 have different functions? Adv Exp Med Biol 502, 249-272CrossRefGoogle ScholarPubMed
94McCoubrey, W.K. Jr., Huang, T.J. and Maines, M.D. (1997) Isolation and characterization of a cDNA from the rat brain that encodes hemoprotein heme oxygenase-3. Eur J Biochem 247, 725-732CrossRefGoogle ScholarPubMed
95Woo, J. et al. (2000) Alleviation of graft-versus-host disease after conditioning with cobalt-protoporphyrin, an inducer of heme oxygenase-1. Transplantation 69, 623-633CrossRefGoogle ScholarPubMed
96Chauveau, C. et al. (2002) Gene transfer of heme oxygenase-1 and carbon monoxide delivery inhibit chronic rejection. Am J Transplant 2, 581-592CrossRefGoogle ScholarPubMed
97Soares, M.P. et al. (1998) Expression of heme oxygenase-1 can determine cardiac xenograft survival. Nat Med 4, 1073-1077CrossRefGoogle ScholarPubMed
98Zenclussen, A.C. et al. (2005) Heme oxygenase as a therapeutic target in immunological pregnancy complications. Int Immunopharmacol 5, 41-51CrossRefGoogle ScholarPubMed
99Ihara, N. et al. (1998) Developmental changes of gene expression in heme metabolic enzymes in rat placenta. FEBS Lett 439, 163-167CrossRefGoogle ScholarPubMed
100Zenclussen, A.C. et al. (2002) Heme oxygenase is downregulated in stress-triggered and IL-12 mediated murine abortion. Scand J Immunol 55, 560-569CrossRefGoogle ScholarPubMed
101Barber, A. et al. (2001) Heme oxygenase expression in human placenta and placental bed: reduced expression of placenta endothelial HO-2 in preeclampsia and fetal growth restriction. FASEB J 15, 1158-1168CrossRefGoogle ScholarPubMed
102Zenclussen, A.C. et al. (2003) Heme oxygenases in pregnancy II: HO-2 is downregulated in human pathological pregnancies. Am J Reprod Immunol 50, 66-76CrossRefGoogle Scholar
103Denschlag, D. et al. (2004) The size of a microsatellite polymorphism of the haem oxygenase 1 gene is associated with idiopathic recurrent miscarriage. Mol Hum Reprod 10, 211-214CrossRefGoogle ScholarPubMed
104Poss, K. and Tonegawa, S. (1997) Reduced stress defense in heme oxygenase-1 deficient cells. Proc Natl Acad Sci U S A 94, 10925-10930CrossRefGoogle ScholarPubMed
105Townsend, P.A. et al. (2003) BAG-1: a multifunctional regulator of cell growth and survival. Biochim Biophys Acta 1603, 83-98Google ScholarPubMed
106Choi, B.M. et al. (2005) Critical role of heme oxygenase-1 in Foxp3-mediated immune suppression. Biochem Biophys Res Commun 327, 1066-1071CrossRefGoogle ScholarPubMed
107Jaffe, R., Dorgan, A. and Abramowicz, J.S. (1995) Color Doppler imaging of the uteroplacental circulation in the first trimester: value in predicting pregnancy failure or complication. Am J Roentgenol 164, 1255-1258CrossRefGoogle ScholarPubMed
108Trudinger, B.J., Giles, W.B. and Cook, C.M. (1985) Uteroplacental blood flow velocitytime waveforms in normal and complicated pregnancy. Br J Obstet Gynaecol 92, 39-45CrossRefGoogle ScholarPubMed
109Appleton, S.D. et al. (2002) Heme oxygenase activity in placenta: direct dependence on oxygen availability. Am J Physiol Heart Circ Physiol 282, 2055-2059CrossRefGoogle ScholarPubMed
110Zhou, Y., Damsky, C.H. and Fisher, S.J. (1997) Pre-eclampsia is associated with failure of human cytotrophoblasts to mimic a vascular adhesion phenotype. One cause of defective endovascular invasion in this syndrome? J Clin Invest 99, 2152-2164CrossRefGoogle Scholar
111Marks, G.S. et al. (1991) Does carbon monoxide have a physiological function? Trends Pharmacol Sci 12, 185-188CrossRefGoogle ScholarPubMed
112Verma, A. et al. (1993) Carbon monoxide: a putative neural messenger. Science 259, 381-384CrossRefGoogle ScholarPubMed
113Lyall, F. et al. (2000) Hemeoxygenase expression in human placenta and placental bed implies a role in regulation of trophoblast invasion and placental function. FASEB J 14, 208-219CrossRefGoogle ScholarPubMed
114Khoury, J.C. et al. (2004) Consequences of smoking and caffeine consumption during pregnancy in women with type 1 diabetes. J Matern Fetal Neonatal Med 15, 44-50CrossRefGoogle ScholarPubMed
115Taylor, C. and Faulk, M.P. (1981) Preventing recent abortion with leukocyte transfusions. Lancet 2, 68-70CrossRefGoogle Scholar
116Takeshita, T. (2004) Diagnosis and treatment of recurrent miscarriage associated with immunological disorders: is paternal lymphocyte immunization a relic of the past? J Nippon Med Sch 71, 308-313CrossRefGoogle ScholarPubMed
117Hill, J.A. (1997) Immunotherapy for recurrent pregnancy loss: “standard of care or buyer beware”. J Soc Gynecol Invest 4, 267-273Google Scholar
118Christiansen, O.B., Nielsen, H.S. and Kolte, A.M. (2006) Future directions of failed implantation and recurrent miscarriage research. Reprod Biomed Online 13, 71-83CrossRefGoogle ScholarPubMed
119Mowbray, J.F. et al. (1985) Controlled trials of treatment of recurrent spontaneous abortion by immunostimulation with paternal cells. Lancet 1(8435), 941-943CrossRefGoogle ScholarPubMed
120Marget, M. et al. (2005) Analysis of sIL-2R, CD4, CD8, CD25, CD45RO and FOXP3 before and after lymphocyte immune therapy of women with recurrent miscarriages. Reproductive Immunology Workshop (September 22, 2005, Kiel, Germany); Meeting of the Germany Society of Immunology (Deutsche Gesellschaft für Immunologie). Abstract no. M10Google Scholar

Further reading, resources and contacts

The following websites provide patient support for miscarriage:

Guleria, I. and Sayegh, M. (2007) Maternal acceptance of the fetus: true human tolerance. J Immunol 178, 3345-3351CrossRefGoogle ScholarPubMed
Hunt, J. (2006) Stranger in a strange land. Immunol Rev 213, 36-47CrossRefGoogle Scholar
Moffett, A. and Loke, C. (2006) Immunology of placentation in eutherian mammals. Nat Rev Immunol 6, 584-594CrossRefGoogle ScholarPubMed
Kling, C. et al. (2006) Transfusion related risks of intradermal allogeneic lymphocyte immunotherapy: single cases in a large cohort and review of the literature. Am J Reprod Immunol 56, 157-171CrossRefGoogle Scholar
Abrahams, V.M. and Mor, G. (2005) Toll-like receptors and their role in the trophoblast. Placenta 26, 540-547CrossRefGoogle ScholarPubMed
http://fertilityusa.com/ (Advanced Reproductive Care Inc)Google Scholar
http://smfm.org/ (The Society for Maternal-Fetal Medicine)Google Scholar
http://royaninstitute.org/ (The Royan Institute, Iran)Google Scholar
Guleria, I. and Sayegh, M. (2007) Maternal acceptance of the fetus: true human tolerance. J Immunol 178, 3345-3351CrossRefGoogle ScholarPubMed
Hunt, J. (2006) Stranger in a strange land. Immunol Rev 213, 36-47CrossRefGoogle Scholar
Moffett, A. and Loke, C. (2006) Immunology of placentation in eutherian mammals. Nat Rev Immunol 6, 584-594CrossRefGoogle ScholarPubMed
Kling, C. et al. (2006) Transfusion related risks of intradermal allogeneic lymphocyte immunotherapy: single cases in a large cohort and review of the literature. Am J Reprod Immunol 56, 157-171CrossRefGoogle Scholar
Abrahams, V.M. and Mor, G. (2005) Toll-like receptors and their role in the trophoblast. Placenta 26, 540-547CrossRefGoogle ScholarPubMed
http://fertilityusa.com/ (Advanced Reproductive Care Inc)Google Scholar
http://smfm.org/ (The Society for Maternal-Fetal Medicine)Google Scholar
http://royaninstitute.org/ (The Royan Institute, Iran)Google Scholar