Skip to main content
Top
Published in: Seminars in Immunopathology 2/2018

01-02-2018 | Review

Immunometabolism, pregnancy, and nutrition

Authors: Kristin Thiele, Lianghui Diao, Petra Clara Arck

Published in: Seminars in Immunopathology | Issue 2/2018

Login to get access

Abstract

The emerging field of immunometabolism has substantially progressed over the last years and provided pivotal insights into distinct metabolic regulators and reprogramming pathways of immune cell populations in various immunological settings. However, insights into immunometabolic reprogramming in the context of reproduction are still enigmatic. During pregnancy, the maternal immune system needs to actively adapt to the presence of the fetal antigens, i.e., by functional modifications of distinct innate immune cell subsets, the generation of regulatory T cells, and the suppression of an anti-fetal effector T cell response. Considering that metabolic pathways have been shown to affect the functional role of such immune cells in a number of settings, we here review the potential role of immunometabolism with regard to the molecular and cellular mechanisms necessary for successful reproduction. Since immunometabolism holds the potential for a therapeutic approach to alter the course of immune diseases, we further highlight how a targeted metabolic reprogramming of immune cells may be triggered by maternal anthropometric or nutritional aspects.
Literature
1.
go back to reference Cha J, Sun X, Dey SK (2012) Mechanisms of implantation: strategies for successful pregnancy. Nat Med 18:1754–1767PubMedCrossRef Cha J, Sun X, Dey SK (2012) Mechanisms of implantation: strategies for successful pregnancy. Nat Med 18:1754–1767PubMedCrossRef
2.
go back to reference Frolova AI, Moley KH (2011) Quantitative analysis of glucose transporter mRNAs in endometrial stromal cells reveals critical role of GLUT1 in uterine receptivity. Endocrinology 152:2123–2128PubMedPubMedCentralCrossRef Frolova AI, Moley KH (2011) Quantitative analysis of glucose transporter mRNAs in endometrial stromal cells reveals critical role of GLUT1 in uterine receptivity. Endocrinology 152:2123–2128PubMedPubMedCentralCrossRef
3.
go back to reference Kommagani R, Szwarc MM, Kovanci E et al (2013) Acceleration of the glycolytic flux by steroid receptor coactivator-2 is essential for endometrial decidualization. PLoS Genet 9:e1003900PubMedPubMedCentralCrossRef Kommagani R, Szwarc MM, Kovanci E et al (2013) Acceleration of the glycolytic flux by steroid receptor coactivator-2 is essential for endometrial decidualization. PLoS Genet 9:e1003900PubMedPubMedCentralCrossRef
4.
go back to reference Murdoch RN (1987) Glycolysis in the mouse uterus during the early post-implantation stages of pregnancy and the effects of acute doses of ethanol. Teratology 35:169–176PubMedCrossRef Murdoch RN (1987) Glycolysis in the mouse uterus during the early post-implantation stages of pregnancy and the effects of acute doses of ethanol. Teratology 35:169–176PubMedCrossRef
5.
go back to reference Krawczyk CM, Holowka T, Sun J et al (2010) Toll-like receptor-induced changes in glycolytic metabolism regulate dendritic cell activation. Blood 115:4742–4749PubMedPubMedCentralCrossRef Krawczyk CM, Holowka T, Sun J et al (2010) Toll-like receptor-induced changes in glycolytic metabolism regulate dendritic cell activation. Blood 115:4742–4749PubMedPubMedCentralCrossRef
6.
go back to reference Everts B, Amiel E, Huang SC et al (2014) TLR-driven early glycolytic reprogramming via the kinases TBK1-IKKvarepsilon supports the anabolic demands of dendritic cell activation. Nat Immunol 15:323–332PubMedPubMedCentralCrossRef Everts B, Amiel E, Huang SC et al (2014) TLR-driven early glycolytic reprogramming via the kinases TBK1-IKKvarepsilon supports the anabolic demands of dendritic cell activation. Nat Immunol 15:323–332PubMedPubMedCentralCrossRef
7.
go back to reference Rodriguez-Prados JC, Traves PG, Cuenca J et al (2010) Substrate fate in activated macrophages: a comparison between innate, classic, and alternative activation. J Immunol 185:605–614PubMedCrossRef Rodriguez-Prados JC, Traves PG, Cuenca J et al (2010) Substrate fate in activated macrophages: a comparison between innate, classic, and alternative activation. J Immunol 185:605–614PubMedCrossRef
8.
go back to reference Donnelly RP, Loftus RM, Keating SE et al (2014) mTORC1-dependent metabolic reprogramming is a prerequisite for NK cell effector function. J Immunol 193:4477–4484PubMedPubMedCentralCrossRef Donnelly RP, Loftus RM, Keating SE et al (2014) mTORC1-dependent metabolic reprogramming is a prerequisite for NK cell effector function. J Immunol 193:4477–4484PubMedPubMedCentralCrossRef
9.
go back to reference Michalek RD, Gerriets VA, Jacobs SR et al (2011) Cutting edge: distinct glycolytic and lipid oxidative metabolic programs are essential for effector and regulatory CD4+ T cell subsets. J Immunol 186:3299–3303PubMedPubMedCentralCrossRef Michalek RD, Gerriets VA, Jacobs SR et al (2011) Cutting edge: distinct glycolytic and lipid oxidative metabolic programs are essential for effector and regulatory CD4+ T cell subsets. J Immunol 186:3299–3303PubMedPubMedCentralCrossRef
10.
go back to reference Gubser PM, Bantug GR, Razik L et al (2013) Rapid effector function of memory CD8+ T cells requires an immediate-early glycolytic switch. Nat Immunol 14:1064–1072PubMedCrossRef Gubser PM, Bantug GR, Razik L et al (2013) Rapid effector function of memory CD8+ T cells requires an immediate-early glycolytic switch. Nat Immunol 14:1064–1072PubMedCrossRef
11.
go back to reference Shi LZ, Wang R, Huang G et al (2011) HIF1alpha-dependent glycolytic pathway orchestrates a metabolic checkpoint for the differentiation of TH17 and Treg cells. J Exp Med 208:1367–1376PubMedPubMedCentralCrossRef Shi LZ, Wang R, Huang G et al (2011) HIF1alpha-dependent glycolytic pathway orchestrates a metabolic checkpoint for the differentiation of TH17 and Treg cells. J Exp Med 208:1367–1376PubMedPubMedCentralCrossRef
12.
go back to reference Doughty CA, Bleiman BF, Wagner DJ et al (2006) Antigen receptor-mediated changes in glucose metabolism in B lymphocytes: role of phosphatidylinositol 3-kinase signaling in the glycolytic control of growth. Blood 107:4458–4465PubMedPubMedCentralCrossRef Doughty CA, Bleiman BF, Wagner DJ et al (2006) Antigen receptor-mediated changes in glucose metabolism in B lymphocytes: role of phosphatidylinositol 3-kinase signaling in the glycolytic control of growth. Blood 107:4458–4465PubMedPubMedCentralCrossRef
13.
go back to reference Huynh A, DuPage M, Priyadharshini B et al (2015) Control of PI(3) kinase in Treg cells maintains homeostasis and lineage stability. Nat Immunol 16:188–196PubMedPubMedCentralCrossRef Huynh A, DuPage M, Priyadharshini B et al (2015) Control of PI(3) kinase in Treg cells maintains homeostasis and lineage stability. Nat Immunol 16:188–196PubMedPubMedCentralCrossRef
14.
15.
go back to reference Wei J, Long L, Yang K et al (2016) Autophagy enforces functional integrity of regulatory T cells by coupling environmental cues and metabolic homeostasis. Nat Immunol 17:277–285PubMedPubMedCentralCrossRef Wei J, Long L, Yang K et al (2016) Autophagy enforces functional integrity of regulatory T cells by coupling environmental cues and metabolic homeostasis. Nat Immunol 17:277–285PubMedPubMedCentralCrossRef
16.
go back to reference O’Sullivan D, van der Windt GJ, Huang SC et al (2014) Memory CD8(+) T cells use cell-intrinsic lipolysis to support the metabolic programming necessary for development. Immunity 41:75–88PubMedPubMedCentralCrossRef O’Sullivan D, van der Windt GJ, Huang SC et al (2014) Memory CD8(+) T cells use cell-intrinsic lipolysis to support the metabolic programming necessary for development. Immunity 41:75–88PubMedPubMedCentralCrossRef
17.
go back to reference Infantino V, Convertini P, Cucci L et al (2011) The mitochondrial citrate carrier: a new player in inflammation. Biochem J 438:433–436PubMedCrossRef Infantino V, Convertini P, Cucci L et al (2011) The mitochondrial citrate carrier: a new player in inflammation. Biochem J 438:433–436PubMedCrossRef
18.
go back to reference Berod L, Friedrich C, Nandan A et al (2014) De novo fatty acid synthesis controls the fate between regulatory T and T helper 17 cells. Nat Med 20:1327–1333PubMedCrossRef Berod L, Friedrich C, Nandan A et al (2014) De novo fatty acid synthesis controls the fate between regulatory T and T helper 17 cells. Nat Med 20:1327–1333PubMedCrossRef
19.
go back to reference Wang R, Dillon CP, Shi LZ et al (2011) The transcription factor Myc controls metabolic reprogramming upon T lymphocyte activation. Immunity 35:871–882PubMedPubMedCentralCrossRef Wang R, Dillon CP, Shi LZ et al (2011) The transcription factor Myc controls metabolic reprogramming upon T lymphocyte activation. Immunity 35:871–882PubMedPubMedCentralCrossRef
20.
go back to reference Gerriets VA, Kishton RJ, Nichols AG et al (2015) Metabolic programming and PDHK1 control CD4+ T cell subsets and inflammation. J Clin Invest 125:194–207PubMedCrossRef Gerriets VA, Kishton RJ, Nichols AG et al (2015) Metabolic programming and PDHK1 control CD4+ T cell subsets and inflammation. J Clin Invest 125:194–207PubMedCrossRef
21.
go back to reference Posokhova EN, Khoshchenko OM, Chasovskikh MI et al (2008) Lipid synthesis in macrophages during inflammation in vivo: effect of agonists of peroxisome proliferator activated receptors alpha and gamma and of retinoid X receptors. Biochemistry (Mosc) 73:296–304CrossRef Posokhova EN, Khoshchenko OM, Chasovskikh MI et al (2008) Lipid synthesis in macrophages during inflammation in vivo: effect of agonists of peroxisome proliferator activated receptors alpha and gamma and of retinoid X receptors. Biochemistry (Mosc) 73:296–304CrossRef
22.
23.
go back to reference Carr EL, Kelman A, GS W et al (2010) Glutamine uptake and metabolism are coordinately regulated by ERK/MAPK during T lymphocyte activation. J Immunol 185:1037–1044PubMedPubMedCentralCrossRef Carr EL, Kelman A, GS W et al (2010) Glutamine uptake and metabolism are coordinately regulated by ERK/MAPK during T lymphocyte activation. J Immunol 185:1037–1044PubMedPubMedCentralCrossRef
24.
go back to reference Cobbold SP, Adams E, Farquhar CA et al (2009) Infectious tolerance via the consumption of essential amino acids and mTOR signaling. Proc Natl Acad Sci U S A 106:12055–12060PubMedPubMedCentralCrossRef Cobbold SP, Adams E, Farquhar CA et al (2009) Infectious tolerance via the consumption of essential amino acids and mTOR signaling. Proc Natl Acad Sci U S A 106:12055–12060PubMedPubMedCentralCrossRef
25.
26.
27.
go back to reference Frolova AI, Moley KH (2011) Glucose transporters in the uterus: an analysis of tissue distribution and proposed physiological roles. Reproduction 142:211–220PubMedPubMedCentralCrossRef Frolova AI, Moley KH (2011) Glucose transporters in the uterus: an analysis of tissue distribution and proposed physiological roles. Reproduction 142:211–220PubMedPubMedCentralCrossRef
28.
go back to reference Kim JY, Song H, Kim H et al (2009) Transcriptional profiling with a pathway-oriented analysis identifies dysregulated molecular phenotypes in the endometrium of patients with polycystic ovary syndrome. J Clin Endocrinol Metab 94:1416–1426PubMedPubMedCentralCrossRef Kim JY, Song H, Kim H et al (2009) Transcriptional profiling with a pathway-oriented analysis identifies dysregulated molecular phenotypes in the endometrium of patients with polycystic ovary syndrome. J Clin Endocrinol Metab 94:1416–1426PubMedPubMedCentralCrossRef
29.
go back to reference Frolova AI, O’Neill K, Moley KH (2011) Dehydroepiandrosterone inhibits glucose flux through the pentose phosphate pathway in human and mouse endometrial stromal cells, preventing decidualization and implantation. Mol Endocrinol 25:1444–1455PubMedPubMedCentralCrossRef Frolova AI, O’Neill K, Moley KH (2011) Dehydroepiandrosterone inhibits glucose flux through the pentose phosphate pathway in human and mouse endometrial stromal cells, preventing decidualization and implantation. Mol Endocrinol 25:1444–1455PubMedPubMedCentralCrossRef
30.
31.
go back to reference Arck PC, Hecher K (2013) Fetomaternal immune cross-talk and its consequences for maternal and offspring’s health. Nat Med 19:548–556PubMedCrossRef Arck PC, Hecher K (2013) Fetomaternal immune cross-talk and its consequences for maternal and offspring’s health. Nat Med 19:548–556PubMedCrossRef
32.
go back to reference Apps R, Murphy SP, Fernando R et al (2009) Human leucocyte antigen (HLA) expression of primary trophoblast cells and placental cell lines, determined using single antigen beads to characterize allotype specificities of anti-HLA antibodies. Immunology 127:26–39PubMedPubMedCentralCrossRef Apps R, Murphy SP, Fernando R et al (2009) Human leucocyte antigen (HLA) expression of primary trophoblast cells and placental cell lines, determined using single antigen beads to characterize allotype specificities of anti-HLA antibodies. Immunology 127:26–39PubMedPubMedCentralCrossRef
33.
go back to reference Madeja Z, Yadi H, Apps R et al (2011) Paternal MHC expression on mouse trophoblast affects uterine vascularization and fetal growth. Proc Natl Acad Sci U S A 108:4012–4017PubMedPubMedCentralCrossRef Madeja Z, Yadi H, Apps R et al (2011) Paternal MHC expression on mouse trophoblast affects uterine vascularization and fetal growth. Proc Natl Acad Sci U S A 108:4012–4017PubMedPubMedCentralCrossRef
34.
go back to reference Blois SM, Ilarregui JM, Tometten M et al (2007) A pivotal role for galectin-1 in fetomaternal tolerance. Nat Med 13:1450–1457PubMedCrossRef Blois SM, Ilarregui JM, Tometten M et al (2007) A pivotal role for galectin-1 in fetomaternal tolerance. Nat Med 13:1450–1457PubMedCrossRef
35.
go back to reference Nancy P, Tagliani E, Tay CS et al (2012) Chemokine gene silencing in decidual stromal cells limits T cell access to the maternal-fetal interface. Science 336:1317–1321PubMedPubMedCentralCrossRef Nancy P, Tagliani E, Tay CS et al (2012) Chemokine gene silencing in decidual stromal cells limits T cell access to the maternal-fetal interface. Science 336:1317–1321PubMedPubMedCentralCrossRef
36.
go back to reference Inada K, Shima T, Ito M et al (2015) Helios-positive functional regulatory T cells are decreased in decidua of miscarriage cases with normal fetal chromosomal content. J Reprod Immunol 107:10–19PubMedCrossRef Inada K, Shima T, Ito M et al (2015) Helios-positive functional regulatory T cells are decreased in decidua of miscarriage cases with normal fetal chromosomal content. J Reprod Immunol 107:10–19PubMedCrossRef
37.
go back to reference Clark DA (2016) The importance of being a regulatory T cell in pregnancy. J Reprod Immunol 116:60–69PubMedCrossRef Clark DA (2016) The importance of being a regulatory T cell in pregnancy. J Reprod Immunol 116:60–69PubMedCrossRef
38.
go back to reference Robertson SA, Guerin LR, Moldenhauer LM et al (2009) Activating T regulatory cells for tolerance in early pregnancy—the contribution of seminal fluid. J Reprod Immunol 83:109–116PubMedCrossRef Robertson SA, Guerin LR, Moldenhauer LM et al (2009) Activating T regulatory cells for tolerance in early pregnancy—the contribution of seminal fluid. J Reprod Immunol 83:109–116PubMedCrossRef
39.
go back to reference Aluvihare VR, Kallikourdis M, Betz AG, Regulatory T (2004) Cells mediate maternal tolerance to the fetus. Nat Immunol 5:266–271PubMedCrossRef Aluvihare VR, Kallikourdis M, Betz AG, Regulatory T (2004) Cells mediate maternal tolerance to the fetus. Nat Immunol 5:266–271PubMedCrossRef
40.
go back to reference Sasaki Y, Sakai M, Miyazaki S et al (2004) Decidual and peripheral blood CD4+CD25+ regulatory T cells in early pregnancy subjects and spontaneous abortion cases. Mol Hum Reprod 10:347–353PubMedCrossRef Sasaki Y, Sakai M, Miyazaki S et al (2004) Decidual and peripheral blood CD4+CD25+ regulatory T cells in early pregnancy subjects and spontaneous abortion cases. Mol Hum Reprod 10:347–353PubMedCrossRef
41.
go back to reference Tilburgs T, Roelen DL, van der Mast BJ et al (2008) Evidence for a selective migration of fetus-specific CD4+CD25 bright regulatory T cells from the peripheral blood to the decidua in human pregnancy. J Immunol 180:5737–5745PubMedCrossRef Tilburgs T, Roelen DL, van der Mast BJ et al (2008) Evidence for a selective migration of fetus-specific CD4+CD25 bright regulatory T cells from the peripheral blood to the decidua in human pregnancy. J Immunol 180:5737–5745PubMedCrossRef
42.
go back to reference Shima T, Sasaki Y, Itoh M et al (2010) Regulatory T cells are necessary for implantation and maintenance of early pregnancy but not late pregnancy in allogeneic mice. J Reprod Immunol 85:121–129PubMedCrossRef Shima T, Sasaki Y, Itoh M et al (2010) Regulatory T cells are necessary for implantation and maintenance of early pregnancy but not late pregnancy in allogeneic mice. J Reprod Immunol 85:121–129PubMedCrossRef
43.
go back to reference Arck P, Solano ME, Walecki M et al (2014) The immune privilege of testis and gravid uterus: same difference? Mol Cell Endocrinol 382:509–520PubMedCrossRef Arck P, Solano ME, Walecki M et al (2014) The immune privilege of testis and gravid uterus: same difference? Mol Cell Endocrinol 382:509–520PubMedCrossRef
45.
go back to reference Tilburgs T, Scherjon SA, van der Mast BJ et al (2009) Fetal-maternal HLA-C mismatch is associated with decidual T cell activation and induction of functional T regulatory cells. J Reprod Immunol 82:148–157PubMedCrossRef Tilburgs T, Scherjon SA, van der Mast BJ et al (2009) Fetal-maternal HLA-C mismatch is associated with decidual T cell activation and induction of functional T regulatory cells. J Reprod Immunol 82:148–157PubMedCrossRef
46.
go back to reference Wang WJ, Liu FJ, HM Q et al (2013) Regulation of the expression of Th17 cells and regulatory T cells by IL-27 in patients with unexplained early recurrent miscarriage. J Reprod Immunol 99:39–45PubMedCrossRef Wang WJ, Liu FJ, HM Q et al (2013) Regulation of the expression of Th17 cells and regulatory T cells by IL-27 in patients with unexplained early recurrent miscarriage. J Reprod Immunol 99:39–45PubMedCrossRef
47.
go back to reference Erlebacher A, Vencato D, Price KA et al (2007) Constraints in antigen presentation severely restrict T cell recognition of the allogeneic fetus. J Clin Invest 117:1399–1411PubMedPubMedCentralCrossRef Erlebacher A, Vencato D, Price KA et al (2007) Constraints in antigen presentation severely restrict T cell recognition of the allogeneic fetus. J Clin Invest 117:1399–1411PubMedPubMedCentralCrossRef
48.
go back to reference Moldenhauer LM, Diener KR, Thring DM et al (2009) Cross-presentation of male seminal fluid antigens elicits T cell activation to initiate the female immune response to pregnancy. J Immunol 182:8080–8093PubMedCrossRef Moldenhauer LM, Diener KR, Thring DM et al (2009) Cross-presentation of male seminal fluid antigens elicits T cell activation to initiate the female immune response to pregnancy. J Immunol 182:8080–8093PubMedCrossRef
49.
50.
go back to reference Blois SM, Alba Soto CD, Tometten M et al (2004) Lineage, maturity, and phenotype of uterine murine dendritic cells throughout gestation indicate a protective role in maintaining pregnancy. Biol Reprod 70:1018–1023PubMedCrossRef Blois SM, Alba Soto CD, Tometten M et al (2004) Lineage, maturity, and phenotype of uterine murine dendritic cells throughout gestation indicate a protective role in maintaining pregnancy. Biol Reprod 70:1018–1023PubMedCrossRef
51.
go back to reference Krey G, Frank P, Shaikly V et al (2008) In vivo dendritic cell depletion reduces breeding efficiency, affecting implantation and early placental development in mice. J Mol Med (Berl) 86:999–1011CrossRef Krey G, Frank P, Shaikly V et al (2008) In vivo dendritic cell depletion reduces breeding efficiency, affecting implantation and early placental development in mice. J Mol Med (Berl) 86:999–1011CrossRef
52.
go back to reference Plaks V, Birnberg T, Berkutzki T et al (2008) Uterine DCs are crucial for decidua formation during embryo implantation in mice. J Clin Invest 118:3954–3965PubMedPubMedCentral Plaks V, Birnberg T, Berkutzki T et al (2008) Uterine DCs are crucial for decidua formation during embryo implantation in mice. J Clin Invest 118:3954–3965PubMedPubMedCentral
53.
go back to reference Faas MM, de Vos P, Uterine NK (2017) Cells and macrophages in pregnancy. Placenta Faas MM, de Vos P, Uterine NK (2017) Cells and macrophages in pregnancy. Placenta
54.
go back to reference Cartwright JE, James-Allan L, Buckley RJ et al (2017) The role of decidual NK cells in pregnancies with impaired vascular remodelling. J Reprod Immunol 119:81–84PubMedCrossRef Cartwright JE, James-Allan L, Buckley RJ et al (2017) The role of decidual NK cells in pregnancies with impaired vascular remodelling. J Reprod Immunol 119:81–84PubMedCrossRef
55.
go back to reference Ratsep MT, Felker AM, Kay VR et al (2015) Uterine natural killer cells: supervisors of vasculature construction in early decidua basalis. Reproduction 149:R91–102PubMedCrossRef Ratsep MT, Felker AM, Kay VR et al (2015) Uterine natural killer cells: supervisors of vasculature construction in early decidua basalis. Reproduction 149:R91–102PubMedCrossRef
56.
go back to reference Evans J, Salamonsen LA, Winship A et al (2016) Fertile ground: human endometrial programming and lessons in health and disease. Nat Rev Endocrinol 12:654–667PubMedCrossRef Evans J, Salamonsen LA, Winship A et al (2016) Fertile ground: human endometrial programming and lessons in health and disease. Nat Rev Endocrinol 12:654–667PubMedCrossRef
57.
go back to reference Engler JB, Kursawe N, Solano ME et al (2017) Glucocorticoid receptor in T cells mediates protection from autoimmunity in pregnancy. Proc Natl Acad Sci U S A 114:E181–E190PubMedPubMedCentralCrossRef Engler JB, Kursawe N, Solano ME et al (2017) Glucocorticoid receptor in T cells mediates protection from autoimmunity in pregnancy. Proc Natl Acad Sci U S A 114:E181–E190PubMedPubMedCentralCrossRef
58.
go back to reference Thiele K, Lydon JP, DeMayo FJ et al (2016) Specific deletion of the progesterone receptor on CD11c dendritic cells reveals a progesterone-DC-crosstalk necessary to sustain fetal development. J Reprod Immunol 115:43–43CrossRef Thiele K, Lydon JP, DeMayo FJ et al (2016) Specific deletion of the progesterone receptor on CD11c dendritic cells reveals a progesterone-DC-crosstalk necessary to sustain fetal development. J Reprod Immunol 115:43–43CrossRef
59.
go back to reference Blois SM, Kammerer U, Alba Soto C et al (2007) Dendritic cells: key to fetal tolerance? Biol Reprod 77:590–598PubMedCrossRef Blois SM, Kammerer U, Alba Soto C et al (2007) Dendritic cells: key to fetal tolerance? Biol Reprod 77:590–598PubMedCrossRef
60.
go back to reference Blois SM, Joachim R, Kandil J et al (2004) Depletion of CD8+ cells abolishes the pregnancy protective effect of progesterone substitution with dydrogesterone in mice by altering the Th1/Th2 cytokine profile. J Immunol 172:5893–5899PubMedCrossRef Blois SM, Joachim R, Kandil J et al (2004) Depletion of CD8+ cells abolishes the pregnancy protective effect of progesterone substitution with dydrogesterone in mice by altering the Th1/Th2 cytokine profile. J Immunol 172:5893–5899PubMedCrossRef
61.
go back to reference Hao S, Zhao J, Zhou J et al (2007) Modulation of 17beta-estradiol on the number and cytotoxicity of NK cells in vivo related to MCM and activating receptors. Int Immunopharmacol 7:1765–1775PubMedCrossRef Hao S, Zhao J, Zhou J et al (2007) Modulation of 17beta-estradiol on the number and cytotoxicity of NK cells in vivo related to MCM and activating receptors. Int Immunopharmacol 7:1765–1775PubMedCrossRef
62.
go back to reference Laskarin G, Tokmadzic VS, Strbo N et al (2002) Progesterone induced blocking factor (PIBF) mediates progesterone induced suppression of decidual lymphocyte cytotoxicity. Am J Reprod Immunol 48:201–209PubMedCrossRef Laskarin G, Tokmadzic VS, Strbo N et al (2002) Progesterone induced blocking factor (PIBF) mediates progesterone induced suppression of decidual lymphocyte cytotoxicity. Am J Reprod Immunol 48:201–209PubMedCrossRef
63.
go back to reference Fournier T, Guibourdenche J, Evain-Brion D (2015) Review: hCGs: different sources of production, different glycoforms and functions. Placenta 36(Suppl 1):S60–S65PubMedCrossRef Fournier T, Guibourdenche J, Evain-Brion D (2015) Review: hCGs: different sources of production, different glycoforms and functions. Placenta 36(Suppl 1):S60–S65PubMedCrossRef
64.
go back to reference Khil LY, Jun HS, Kwon H et al (2007) Human chorionic gonadotropin is an immune modulator and can prevent autoimmune diabetes in NOD mice. Diabetologia 50:2147–2155PubMedCrossRef Khil LY, Jun HS, Kwon H et al (2007) Human chorionic gonadotropin is an immune modulator and can prevent autoimmune diabetes in NOD mice. Diabetologia 50:2147–2155PubMedCrossRef
65.
go back to reference Schumacher A, Heinze K, Witte J et al (2013) Human chorionic gonadotropin as a central regulator of pregnancy immune tolerance. J Immunol 190:2650–2658PubMedCrossRef Schumacher A, Heinze K, Witte J et al (2013) Human chorionic gonadotropin as a central regulator of pregnancy immune tolerance. J Immunol 190:2650–2658PubMedCrossRef
66.
go back to reference Schumacher A, Brachwitz N, Sohr S et al (2009) Human chorionic gonadotropin attracts regulatory T cells into the fetal-maternal Interface during early human pregnancy. J Immunol 182:5488–5497PubMedCrossRef Schumacher A, Brachwitz N, Sohr S et al (2009) Human chorionic gonadotropin attracts regulatory T cells into the fetal-maternal Interface during early human pregnancy. J Immunol 182:5488–5497PubMedCrossRef
67.
go back to reference Vacca P, Montaldo E, Croxatto D et al (2015) Identification of diverse innate lymphoid cells in human decidua. Mucosal Immunol 8:254–264PubMedCrossRef Vacca P, Montaldo E, Croxatto D et al (2015) Identification of diverse innate lymphoid cells in human decidua. Mucosal Immunol 8:254–264PubMedCrossRef
68.
69.
go back to reference Muzzio DO, Soldati R, Ehrhardt J et al (2014) B cell development undergoes profound modifications and adaptations during pregnancy in mice. Biol Reprod 91:115PubMedCrossRef Muzzio DO, Soldati R, Ehrhardt J et al (2014) B cell development undergoes profound modifications and adaptations during pregnancy in mice. Biol Reprod 91:115PubMedCrossRef
70.
go back to reference Huang B, Faucette AN, Pawlitz MD et al (2017) Interleukin-33-induced expression of PIBF1 by decidual B cells protects against preterm labor. Nat Med 23:128–135PubMedCrossRef Huang B, Faucette AN, Pawlitz MD et al (2017) Interleukin-33-induced expression of PIBF1 by decidual B cells protects against preterm labor. Nat Med 23:128–135PubMedCrossRef
71.
go back to reference Mor G, Aldo P, Alvero AB (2017) The unique immunological and microbial aspects of pregnancy. Nat Rev Immunol 17:469–482PubMedCrossRef Mor G, Aldo P, Alvero AB (2017) The unique immunological and microbial aspects of pregnancy. Nat Rev Immunol 17:469–482PubMedCrossRef
72.
go back to reference Frauwirth KA, Riley JL, Harris MH et al (2002) The CD28 signaling pathway regulates glucose metabolism. Immunity 16:769–777PubMedCrossRef Frauwirth KA, Riley JL, Harris MH et al (2002) The CD28 signaling pathway regulates glucose metabolism. Immunity 16:769–777PubMedCrossRef
74.
go back to reference Papathanassoglou E, El-Haschimi K, Li XC et al (2006) Leptin receptor expression and signaling in lymphocytes: kinetics during lymphocyte activation, role in lymphocyte survival, and response to high fat diet in mice. J Immunol 176:7745–7752PubMedCrossRef Papathanassoglou E, El-Haschimi K, Li XC et al (2006) Leptin receptor expression and signaling in lymphocytes: kinetics during lymphocyte activation, role in lymphocyte survival, and response to high fat diet in mice. J Immunol 176:7745–7752PubMedCrossRef
75.
go back to reference van der Windt GJ, Everts B, Chang CH et al (2012) Mitochondrial respiratory capacity is a critical regulator of CD8+ T cell memory development. Immunity 36:68–78PubMedCrossRef van der Windt GJ, Everts B, Chang CH et al (2012) Mitochondrial respiratory capacity is a critical regulator of CD8+ T cell memory development. Immunity 36:68–78PubMedCrossRef
77.
go back to reference Rath M, Muller I, Kropf P et al (2014) Metabolism via arginase or nitric oxide synthase: two competing arginine pathways in macrophages. Front Immunol 5:532PubMedPubMedCentralCrossRef Rath M, Muller I, Kropf P et al (2014) Metabolism via arginase or nitric oxide synthase: two competing arginine pathways in macrophages. Front Immunol 5:532PubMedPubMedCentralCrossRef
79.
go back to reference Monticelli LA, Buck MD, Flamar AL et al (2016) Arginase 1 is an innate lymphoid-cell-intrinsic metabolic checkpoint controlling type 2 inflammation. Nat Immunol 17:656–665PubMedPubMedCentralCrossRef Monticelli LA, Buck MD, Flamar AL et al (2016) Arginase 1 is an innate lymphoid-cell-intrinsic metabolic checkpoint controlling type 2 inflammation. Nat Immunol 17:656–665PubMedPubMedCentralCrossRef
81.
82.
go back to reference Lee GK, Park HJ, Macleod M et al (2002) Tryptophan deprivation sensitizes activated T cells to apoptosis prior to cell division. Immunology 107:452–460PubMedPubMedCentralCrossRef Lee GK, Park HJ, Macleod M et al (2002) Tryptophan deprivation sensitizes activated T cells to apoptosis prior to cell division. Immunology 107:452–460PubMedPubMedCentralCrossRef
83.
go back to reference Palsson-McDermott EM, Curtis AM, Goel G et al (2015) Pyruvate kinase M2 regulates Hif-1alpha activity and IL-1beta induction and is a critical determinant of the warburg effect in LPS-activated macrophages. Cell Metab 21:65–80PubMedPubMedCentralCrossRef Palsson-McDermott EM, Curtis AM, Goel G et al (2015) Pyruvate kinase M2 regulates Hif-1alpha activity and IL-1beta induction and is a critical determinant of the warburg effect in LPS-activated macrophages. Cell Metab 21:65–80PubMedPubMedCentralCrossRef
86.
go back to reference Delgoffe GM, Kole TP, Zheng Y et al (2009) The mTOR kinase differentially regulates effector and regulatory T cell lineage commitment. Immunity 30:832–844PubMedPubMedCentralCrossRef Delgoffe GM, Kole TP, Zheng Y et al (2009) The mTOR kinase differentially regulates effector and regulatory T cell lineage commitment. Immunity 30:832–844PubMedPubMedCentralCrossRef
87.
go back to reference Brown EJ, Albers MW, Shin TB et al (1994) A mammalian protein targeted by G1-arresting rapamycin-receptor complex. Nature 369:756–758PubMedCrossRef Brown EJ, Albers MW, Shin TB et al (1994) A mammalian protein targeted by G1-arresting rapamycin-receptor complex. Nature 369:756–758PubMedCrossRef
88.
go back to reference Gabardi S, Baroletti SA (2010) Everolimus: a proliferation signal inhibitor with clinical applications in organ transplantation, oncology, and cardiology. Pharmacotherapy 30:1044–1056PubMedCrossRef Gabardi S, Baroletti SA (2010) Everolimus: a proliferation signal inhibitor with clinical applications in organ transplantation, oncology, and cardiology. Pharmacotherapy 30:1044–1056PubMedCrossRef
89.
go back to reference Mayer CT, Berod L, Sparwasser T (2012) Layers of dendritic cell-mediated T cell tolerance, their regulation and the prevention of autoimmunity. Front Immunol 3:183PubMedPubMedCentralCrossRef Mayer CT, Berod L, Sparwasser T (2012) Layers of dendritic cell-mediated T cell tolerance, their regulation and the prevention of autoimmunity. Front Immunol 3:183PubMedPubMedCentralCrossRef
90.
91.
go back to reference Procaccini C, Carbone F, Di Silvestre D et al (2016) The proteomic landscape of human ex vivo regulatory and conventional T cells reveals specific metabolic requirements. Immunity 44:406–421PubMedPubMedCentralCrossRef Procaccini C, Carbone F, Di Silvestre D et al (2016) The proteomic landscape of human ex vivo regulatory and conventional T cells reveals specific metabolic requirements. Immunity 44:406–421PubMedPubMedCentralCrossRef
92.
go back to reference Beier UH, Angelin A, Akimova T et al (2015) Essential role of mitochondrial energy metabolism in Foxp3(+) T-regulatory cell function and allograft survival. FASEB J 29:2315–2326PubMedPubMedCentralCrossRef Beier UH, Angelin A, Akimova T et al (2015) Essential role of mitochondrial energy metabolism in Foxp3(+) T-regulatory cell function and allograft survival. FASEB J 29:2315–2326PubMedPubMedCentralCrossRef
93.
go back to reference Almeida L, Lochner M, Berod L et al (2016) Metabolic pathways in T cell activation and lineage differentiation. Semin Immunol 28:514–524PubMedCrossRef Almeida L, Lochner M, Berod L et al (2016) Metabolic pathways in T cell activation and lineage differentiation. Semin Immunol 28:514–524PubMedCrossRef
96.
go back to reference Procaccini C, De Rosa V, Galgani M et al (2010) An oscillatory switch in mTOR kinase activity sets regulatory T cell responsiveness. Immunity 33:929–941PubMedPubMedCentralCrossRef Procaccini C, De Rosa V, Galgani M et al (2010) An oscillatory switch in mTOR kinase activity sets regulatory T cell responsiveness. Immunity 33:929–941PubMedPubMedCentralCrossRef
97.
go back to reference Yin Y, Choi SC, Xu Z et al (2015) Normalization of CD4+ T cell metabolism reverses lupus. Sci Transl Med 7:274ra218CrossRef Yin Y, Choi SC, Xu Z et al (2015) Normalization of CD4+ T cell metabolism reverses lupus. Sci Transl Med 7:274ra218CrossRef
98.
go back to reference Yang Z, Shen Y, Oishi H et al (2016) Restoring oxidant signaling suppresses proarthritogenic T cell effector functions in rheumatoid arthritis. Sci Transl Med 8:331ra338 Yang Z, Shen Y, Oishi H et al (2016) Restoring oxidant signaling suppresses proarthritogenic T cell effector functions in rheumatoid arthritis. Sci Transl Med 8:331ra338
99.
go back to reference Bednarski JJ, Warner RE, Rao T et al (2003) Attenuation of autoimmune disease in Fas-deficient mice by treatment with a cytotoxic benzodiazepine. Arthritis Rheum 48:757–766PubMedCrossRef Bednarski JJ, Warner RE, Rao T et al (2003) Attenuation of autoimmune disease in Fas-deficient mice by treatment with a cytotoxic benzodiazepine. Arthritis Rheum 48:757–766PubMedCrossRef
100.
go back to reference Sun Y, Tian T, Gao J et al (2016) Metformin ameliorates the development of experimental autoimmune encephalomyelitis by regulating T helper 17 and regulatory T cells in mice. J Neuroimmunol 292:58–67PubMedCrossRef Sun Y, Tian T, Gao J et al (2016) Metformin ameliorates the development of experimental autoimmune encephalomyelitis by regulating T helper 17 and regulatory T cells in mice. J Neuroimmunol 292:58–67PubMedCrossRef
101.
go back to reference Gatza E, Wahl DR, Opipari AW et al (2011) Manipulating the bioenergetics of alloreactive T cells causes their selective apoptosis and arrests graft-versus-host disease. Sci Transl Med 3:67ra68CrossRef Gatza E, Wahl DR, Opipari AW et al (2011) Manipulating the bioenergetics of alloreactive T cells causes their selective apoptosis and arrests graft-versus-host disease. Sci Transl Med 3:67ra68CrossRef
103.
go back to reference Nguyen HD, Chatterjee S, Haarberg KM et al (2016) Metabolic reprogramming of alloantigen-activated T cells after hematopoietic cell transplantation. J Clin Invest 126:1337–1352PubMedPubMedCentralCrossRef Nguyen HD, Chatterjee S, Haarberg KM et al (2016) Metabolic reprogramming of alloantigen-activated T cells after hematopoietic cell transplantation. J Clin Invest 126:1337–1352PubMedPubMedCentralCrossRef
104.
go back to reference Glick GD, Rossignol R, Lyssiotis CA et al (2014) Anaplerotic metabolism of alloreactive T cells provides a metabolic approach to treat graft-versus-host disease. J Pharmacol Exp Ther 351:298–307PubMedPubMedCentralCrossRef Glick GD, Rossignol R, Lyssiotis CA et al (2014) Anaplerotic metabolism of alloreactive T cells provides a metabolic approach to treat graft-versus-host disease. J Pharmacol Exp Ther 351:298–307PubMedPubMedCentralCrossRef
106.
go back to reference Procaccini C, De Rosa V, Galgani M et al (2012) Leptin-induced mTOR activation defines a specific molecular and transcriptional signature controlling CD4+ effector T cell responses. J Immunol 189:2941–2953PubMedCrossRef Procaccini C, De Rosa V, Galgani M et al (2012) Leptin-induced mTOR activation defines a specific molecular and transcriptional signature controlling CD4+ effector T cell responses. J Immunol 189:2941–2953PubMedCrossRef
107.
go back to reference Wilk S, Scheibenbogen C, Bauer S, Jenke A, Rother M, Guerreiro M, Kudernatsch R, Goerner N, Poller W, Elligsen-Merkel D, Utku N, Magrane J, Volk HD, Skurk C (2011) Adiponectin is a negative regulator of antigen-activated T cells. Eur J Immunol 41:2323–2332 Wilk S, Scheibenbogen C, Bauer S, Jenke A, Rother M, Guerreiro M, Kudernatsch R, Goerner N, Poller W, Elligsen-Merkel D, Utku N, Magrane J, Volk HD, Skurk C (2011) Adiponectin is a negative regulator of antigen-activated T cells. Eur J Immunol 41:2323–2332
108.
go back to reference Piccio L, Cantoni C, Henderson JG et al (2013) Lack of adiponectin leads to increased lymphocyte activation and increased disease severity in a mouse model of multiple sclerosis. Eur J Immunol 43:2089–2100PubMedPubMedCentralCrossRef Piccio L, Cantoni C, Henderson JG et al (2013) Lack of adiponectin leads to increased lymphocyte activation and increased disease severity in a mouse model of multiple sclerosis. Eur J Immunol 43:2089–2100PubMedPubMedCentralCrossRef
110.
go back to reference Ahima RS, Prabakaran D, Mantzoros C et al (1996) Role of leptin in the neuroendocrine response to fasting. Nature 382:250–252PubMedCrossRef Ahima RS, Prabakaran D, Mantzoros C et al (1996) Role of leptin in the neuroendocrine response to fasting. Nature 382:250–252PubMedCrossRef
111.
go back to reference Balsells M, Garcia-Patterson A, Corcoy R (2016) Systematic review and meta-analysis on the association of prepregnancy underweight and miscarriage. Eur J Obstet Gynecol Reprod Biol 207:73–79PubMedCrossRef Balsells M, Garcia-Patterson A, Corcoy R (2016) Systematic review and meta-analysis on the association of prepregnancy underweight and miscarriage. Eur J Obstet Gynecol Reprod Biol 207:73–79PubMedCrossRef
112.
go back to reference Huh JY, Park YJ, Ham M et al (2014) Crosstalk between adipocytes and immune cells in adipose tissue inflammation and metabolic dysregulation in obesity. Molecules and Cells 37:365–371PubMedPubMedCentralCrossRef Huh JY, Park YJ, Ham M et al (2014) Crosstalk between adipocytes and immune cells in adipose tissue inflammation and metabolic dysregulation in obesity. Molecules and Cells 37:365–371PubMedPubMedCentralCrossRef
113.
go back to reference Yang H, Youm YH, Vandanmagsar B et al (2010) Obesity increases the production of proinflammatory mediators from adipose tissue T cells and compromises TCR repertoire diversity: implications for systemic inflammation and insulin resistance. J Immunol 185:1836–1845PubMedPubMedCentralCrossRef Yang H, Youm YH, Vandanmagsar B et al (2010) Obesity increases the production of proinflammatory mediators from adipose tissue T cells and compromises TCR repertoire diversity: implications for systemic inflammation and insulin resistance. J Immunol 185:1836–1845PubMedPubMedCentralCrossRef
114.
go back to reference Nishimura S, Manabe I, Nagasaki M et al (2009) CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nat Med 15:914–920PubMedCrossRef Nishimura S, Manabe I, Nagasaki M et al (2009) CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nat Med 15:914–920PubMedCrossRef
115.
go back to reference Han JM, Patterson SJ, Speck M et al (2014) Insulin inhibits IL-10-mediated regulatory T cell function: implications for obesity. J Immunol 192:623 LP–623629CrossRef Han JM, Patterson SJ, Speck M et al (2014) Insulin inhibits IL-10-mediated regulatory T cell function: implications for obesity. J Immunol 192:623 LP–623629CrossRef
116.
go back to reference Mattioli B, Straface E, Quaranta MG et al (2005) Leptin promotes differentiation and survival of human dendritic cells and licenses them for Th1 priming. J Immunol 174:6820–6828PubMedCrossRef Mattioli B, Straface E, Quaranta MG et al (2005) Leptin promotes differentiation and survival of human dendritic cells and licenses them for Th1 priming. J Immunol 174:6820–6828PubMedCrossRef
117.
118.
go back to reference Rocha VZ, Folco EJ, Sukhova G et al (2008) Interferon-gamma, a Th1 cytokine, regulates fat inflammation: a role for adaptive immunity in obesity. Circ Res 103:467–476PubMedPubMedCentralCrossRef Rocha VZ, Folco EJ, Sukhova G et al (2008) Interferon-gamma, a Th1 cytokine, regulates fat inflammation: a role for adaptive immunity in obesity. Circ Res 103:467–476PubMedPubMedCentralCrossRef
119.
go back to reference Keane KN, Calton EK, Carlessi R et al. (2017) The bioenergetics of inflammation: insights into obesity and type 2 diabetes. Eur J Clin Nutr Keane KN, Calton EK, Carlessi R et al. (2017) The bioenergetics of inflammation: insights into obesity and type 2 diabetes. Eur J Clin Nutr
120.
go back to reference Matjila MJ, Hoffman A, van der Spuy ZM (2017) Medical conditions associated with recurrent miscarriage—is BMI the tip of the iceberg? Eur J Obstet Gynecol Reprod Biol 214:91–96PubMedCrossRef Matjila MJ, Hoffman A, van der Spuy ZM (2017) Medical conditions associated with recurrent miscarriage—is BMI the tip of the iceberg? Eur J Obstet Gynecol Reprod Biol 214:91–96PubMedCrossRef
121.
go back to reference Bhandari HM, Tan BK, Quenby S (2016) Superfertility is more prevalent in obese women with recurrent early pregnancy miscarriage. BJOG 123:217–222PubMedCrossRef Bhandari HM, Tan BK, Quenby S (2016) Superfertility is more prevalent in obese women with recurrent early pregnancy miscarriage. BJOG 123:217–222PubMedCrossRef
122.
go back to reference Boonstra A, Barrat FJ, Crain C et al (2001) 1α,25-dihydroxyvitamin D3 has a direct effect on naive CD4+ T Cells to enhance the development of Th2 cells. J Immunol 167:4974 LP–4974980CrossRef Boonstra A, Barrat FJ, Crain C et al (2001) 1α,25-dihydroxyvitamin D3 has a direct effect on naive CD4+ T Cells to enhance the development of Th2 cells. J Immunol 167:4974 LP–4974980CrossRef
123.
go back to reference Gorman S, Kuritzky LA, Judge MA et al (2007) Topically applied 1,25-dihydroxyvitamin D3 enhances the suppressive activity of CD4+ CD25+ cells in the draining lymph nodes. J Immunol 179:6273–6283PubMedCrossRef Gorman S, Kuritzky LA, Judge MA et al (2007) Topically applied 1,25-dihydroxyvitamin D3 enhances the suppressive activity of CD4+ CD25+ cells in the draining lymph nodes. J Immunol 179:6273–6283PubMedCrossRef
124.
go back to reference Kinoshita M, Kayama H, Kusu T et al (2012) Dietary folic acid promotes survival of Foxp3+ regulatory T cells in the colon. J Immunol 189:2869–2878PubMedCrossRef Kinoshita M, Kayama H, Kusu T et al (2012) Dietary folic acid promotes survival of Foxp3+ regulatory T cells in the colon. J Immunol 189:2869–2878PubMedCrossRef
125.
go back to reference Griffin MD, Lutz WH, Phan VA et al (2000) Potent inhibition of dendritic cell differentiation and maturation by vitamin D analogs. Biochem Biophys Res Commun 270:701–708PubMedCrossRef Griffin MD, Lutz WH, Phan VA et al (2000) Potent inhibition of dendritic cell differentiation and maturation by vitamin D analogs. Biochem Biophys Res Commun 270:701–708PubMedCrossRef
126.
go back to reference Griffin MD, Lutz W, Phan VA et al (2001) Dendritic cell modulation by 1alpha,25 dihydroxyvitamin D3 and its analogs: a vitamin D receptor-dependent pathway that promotes a persistent state of immaturity in vitro and in vivo. Proc Natl Acad Sci U S A 98:6800–6805PubMedPubMedCentralCrossRef Griffin MD, Lutz W, Phan VA et al (2001) Dendritic cell modulation by 1alpha,25 dihydroxyvitamin D3 and its analogs: a vitamin D receptor-dependent pathway that promotes a persistent state of immaturity in vitro and in vivo. Proc Natl Acad Sci U S A 98:6800–6805PubMedPubMedCentralCrossRef
127.
go back to reference Troen AM, Mitchell B, Sorensen B et al (2006) Unmetabolized folic acid in plasma is associated with reduced natural killer cell cytotoxicity among postmenopausal women. J Nutr 136:189–194PubMedCrossRef Troen AM, Mitchell B, Sorensen B et al (2006) Unmetabolized folic acid in plasma is associated with reduced natural killer cell cytotoxicity among postmenopausal women. J Nutr 136:189–194PubMedCrossRef
128.
go back to reference Shaat N, Ignell C, Katsarou A et al. (2017) Glucose homeostasis, beta cell function, and insulin resistance in relation to vitamin D status after gestational diabetes mellitus. Acta Obstet Gynecol Scand. n/a-n/a Shaat N, Ignell C, Katsarou A et al. (2017) Glucose homeostasis, beta cell function, and insulin resistance in relation to vitamin D status after gestational diabetes mellitus. Acta Obstet Gynecol Scand. n/a-n/a
129.
go back to reference Lu M, Xu Y, Lv L et al (2016) Association between vitamin D status and the risk of gestational diabetes mellitus: a meta-analysis. Arch Gynecol Obstet 293:959–966PubMedCrossRef Lu M, Xu Y, Lv L et al (2016) Association between vitamin D status and the risk of gestational diabetes mellitus: a meta-analysis. Arch Gynecol Obstet 293:959–966PubMedCrossRef
130.
go back to reference Smith TA, Kirkpatrick DR, Kovilam O et al (2015) Immunomodulatory role of vitamin D in the pathogenesis of preeclampsia. Expert Rev Clin Immunol 11:1055–1063PubMedPubMedCentralCrossRef Smith TA, Kirkpatrick DR, Kovilam O et al (2015) Immunomodulatory role of vitamin D in the pathogenesis of preeclampsia. Expert Rev Clin Immunol 11:1055–1063PubMedPubMedCentralCrossRef
131.
go back to reference Murthi P, Yong HEJ, Ngyuen TPH et al. (2016) Role of the placental vitamin D receptor in modulating feto-placental growth in fetal growth restriction and preeclampsia-affected pregnancies. In 43–43 Murthi P, Yong HEJ, Ngyuen TPH et al. (2016) Role of the placental vitamin D receptor in modulating feto-placental growth in fetal growth restriction and preeclampsia-affected pregnancies. In 43–43
132.
go back to reference McKay JA, Xie L, Adriaens M et al. (2016) Organ-specific gene expression changes in the fetal liver and placenta in response to maternal folate depletion. Nutrients 8. McKay JA, Xie L, Adriaens M et al. (2016) Organ-specific gene expression changes in the fetal liver and placenta in response to maternal folate depletion. Nutrients 8.
133.
go back to reference Lehr S, Hartwig S, Sell H (2012) Adipokines: a treasure trove for the discovery of biomarkers for metabolic disorders. PROTEOMICS – Clinical Applications 6:91–101PubMedCrossRef Lehr S, Hartwig S, Sell H (2012) Adipokines: a treasure trove for the discovery of biomarkers for metabolic disorders. PROTEOMICS – Clinical Applications 6:91–101PubMedCrossRef
134.
go back to reference Blüher M, Mantzoros CS (2015) From leptin to other adipokines in health and disease: facts and expectations at the beginning of the 21st century. Metabolism 64:131–145PubMedCrossRef Blüher M, Mantzoros CS (2015) From leptin to other adipokines in health and disease: facts and expectations at the beginning of the 21st century. Metabolism 64:131–145PubMedCrossRef
135.
go back to reference Perez-Perez A, Vilarino-Garcia T, Fernandez-Riejos P et al. Role of leptin as a link between metabolism and the immune system. Cytokine Growth Factor Rev 2017: Perez-Perez A, Vilarino-Garcia T, Fernandez-Riejos P et al. Role of leptin as a link between metabolism and the immune system. Cytokine Growth Factor Rev 2017:
136.
go back to reference Macdonald SPJ, Bosio E, Neil C et al (2017) Resistin and NGAL are associated with inflammatory response, endothelial activation and clinical outcomes in sepsis. Inflamm Res 66:611–619PubMedCrossRef Macdonald SPJ, Bosio E, Neil C et al (2017) Resistin and NGAL are associated with inflammatory response, endothelial activation and clinical outcomes in sepsis. Inflamm Res 66:611–619PubMedCrossRef
137.
go back to reference White RT, Damm D, Hancock N et al (1992) Human adipsin is identical to complement factor D and is expressed at high levels in adipose tissue. J Biol Chem 267:9210–9213PubMed White RT, Damm D, Hancock N et al (1992) Human adipsin is identical to complement factor D and is expressed at high levels in adipose tissue. J Biol Chem 267:9210–9213PubMed
138.
go back to reference Procaccini C, Galgani M, De Rosa V et al (2012) Intracellular metabolic pathways control immune tolerance. Trends Immunol 33:1–7PubMedCrossRef Procaccini C, Galgani M, De Rosa V et al (2012) Intracellular metabolic pathways control immune tolerance. Trends Immunol 33:1–7PubMedCrossRef
139.
go back to reference Mor G, Cardenas I, Abrahams V, Guller S (2011) Inflammation and pregnancy: the role of the immune system at the implantation site. Ann N Y Acad Sci 1221:80–87 Mor G, Cardenas I, Abrahams V, Guller S (2011) Inflammation and pregnancy: the role of the immune system at the implantation site. Ann N Y Acad Sci 1221:80–87
141.
go back to reference Malik NM, Carter ND, Murray JF et al (2001) Leptin requirement for conception, implantation, and gestation in the mouse. Endocrinology 142:5198–5202PubMedCrossRef Malik NM, Carter ND, Murray JF et al (2001) Leptin requirement for conception, implantation, and gestation in the mouse. Endocrinology 142:5198–5202PubMedCrossRef
142.
go back to reference Io M (2009) Weight gain during pregnancy: reexamining the guidelines. National Academies Press, Washington, DC Io M (2009) Weight gain during pregnancy: reexamining the guidelines. National Academies Press, Washington, DC
143.
144.
go back to reference Engels G, Hierweger AM, Hoffmann J et al (2017) Pregnancy-related immune adaptation promotes the emergence of highly virulent H1N1 influenza virus strains in allogenically pregnant mice. Cell Host Microbe 21:321–333PubMedCrossRef Engels G, Hierweger AM, Hoffmann J et al (2017) Pregnancy-related immune adaptation promotes the emergence of highly virulent H1N1 influenza virus strains in allogenically pregnant mice. Cell Host Microbe 21:321–333PubMedCrossRef
145.
go back to reference van Riel D, Mittrucker HW, Engels G et al (2016) Influenza pathogenicity during pregnancy in women and animal models. Semin Immunopathol 38:719–726PubMedCrossRef van Riel D, Mittrucker HW, Engels G et al (2016) Influenza pathogenicity during pregnancy in women and animal models. Semin Immunopathol 38:719–726PubMedCrossRef
146.
147.
go back to reference Feuerer M, Herrero L, Cipolletta D et al (2009) Lean, but not obese, fat is enriched for a unique population of regulatory T cells that affect metabolic parameters. Nat Med 15:930–939PubMedPubMedCentralCrossRef Feuerer M, Herrero L, Cipolletta D et al (2009) Lean, but not obese, fat is enriched for a unique population of regulatory T cells that affect metabolic parameters. Nat Med 15:930–939PubMedPubMedCentralCrossRef
148.
go back to reference Winer DA, Winer S, Shen L et al (2011) B cells promote insulin resistance through modulation of T cells and production of pathogenic IgG antibodies. Nat Med 17:610–U134PubMedPubMedCentralCrossRef Winer DA, Winer S, Shen L et al (2011) B cells promote insulin resistance through modulation of T cells and production of pathogenic IgG antibodies. Nat Med 17:610–U134PubMedPubMedCentralCrossRef
149.
150.
go back to reference Silha JV, Krsek M, Skrha JV et al (2003) Plasma resistin, adiponectin and leptin levels in lean and obese subjects: correlations with insulin resistance. Eur J Endocrinol 149:331–335PubMedCrossRef Silha JV, Krsek M, Skrha JV et al (2003) Plasma resistin, adiponectin and leptin levels in lean and obese subjects: correlations with insulin resistance. Eur J Endocrinol 149:331–335PubMedCrossRef
151.
go back to reference Steppan CM, Bailey ST, Bhat S et al (2001) The hormone resistin links obesity to diabetes. Nature 409:307–312PubMedCrossRef Steppan CM, Bailey ST, Bhat S et al (2001) The hormone resistin links obesity to diabetes. Nature 409:307–312PubMedCrossRef
152.
go back to reference Degawa-Yamauchi M, Bovenkerk JE, Juliar BE et al (2003) Serum resistin (FIZZ3) protein is increased in obese humans. J Clin Endocrinol Metab 88:5452–5455PubMedCrossRef Degawa-Yamauchi M, Bovenkerk JE, Juliar BE et al (2003) Serum resistin (FIZZ3) protein is increased in obese humans. J Clin Endocrinol Metab 88:5452–5455PubMedCrossRef
153.
go back to reference Kuzmicki M, Telejko B, Szamatowicz J et al (2009) High resistin and interleukin-6 levels are associated with gestational diabetes mellitus. Gynecol Endocrinol 25:258–263PubMedCrossRef Kuzmicki M, Telejko B, Szamatowicz J et al (2009) High resistin and interleukin-6 levels are associated with gestational diabetes mellitus. Gynecol Endocrinol 25:258–263PubMedCrossRef
154.
go back to reference Siddiqui K, P George T. Resistin (2017) Role in development of gestational diabetes mellitus. Biomark Med. 11: Siddiqui K, P George T. Resistin (2017) Role in development of gestational diabetes mellitus. Biomark Med. 11:
155.
go back to reference Lobo TF, Torloni MR, Gueuvoghlanian-Silva BY et al (2013) Resistin concentration and gestational diabetes: a systematic review of the literature. J Reprod Immunol 97:120–127PubMedCrossRef Lobo TF, Torloni MR, Gueuvoghlanian-Silva BY et al (2013) Resistin concentration and gestational diabetes: a systematic review of the literature. J Reprod Immunol 97:120–127PubMedCrossRef
156.
go back to reference Sivakumar K, Bari MF, Adaikalakoteswari A et al (2013) Elevated fetal adipsin/acylation-stimulating protein (ASP) in obese pregnancy: novel placental secretion via Hofbauer cells. J Clin Endocrinol Metab 98:4113–4122PubMedPubMedCentralCrossRef Sivakumar K, Bari MF, Adaikalakoteswari A et al (2013) Elevated fetal adipsin/acylation-stimulating protein (ASP) in obese pregnancy: novel placental secretion via Hofbauer cells. J Clin Endocrinol Metab 98:4113–4122PubMedPubMedCentralCrossRef
158.
go back to reference Henson MC, Swan KF, O'Neil JS (1998) Expression of placental leptin and leptin receptor transcripts in early pregnancy and at term. Obstet Gynecol 92:1020–1028PubMed Henson MC, Swan KF, O'Neil JS (1998) Expression of placental leptin and leptin receptor transcripts in early pregnancy and at term. Obstet Gynecol 92:1020–1028PubMed
159.
go back to reference Hassink SG, de Lancey E, Sheslow DV et al (1997) Placental leptin: an important new growth factor in intrauterine and neonatal development? Pediatrics 100:E1PubMedCrossRef Hassink SG, de Lancey E, Sheslow DV et al (1997) Placental leptin: an important new growth factor in intrauterine and neonatal development? Pediatrics 100:E1PubMedCrossRef
160.
go back to reference Cameo P, Bischof P, Calvo JC (2003) Effect of leptin on progesterone, human chorionic gonadotropin, and interleukin-6 secretion by human term trophoblast cells in culture. Biol Reprod 68:472–477PubMedCrossRef Cameo P, Bischof P, Calvo JC (2003) Effect of leptin on progesterone, human chorionic gonadotropin, and interleukin-6 secretion by human term trophoblast cells in culture. Biol Reprod 68:472–477PubMedCrossRef
161.
162.
go back to reference Maymo JL, Perez AP, Sanchez-Margalet V et al (2009) Up-regulation of placental leptin by human chorionic gonadotropin. Endocrinology 150:304–313PubMedCrossRef Maymo JL, Perez AP, Sanchez-Margalet V et al (2009) Up-regulation of placental leptin by human chorionic gonadotropin. Endocrinology 150:304–313PubMedCrossRef
163.
go back to reference Cella F, Giordano G, Cordera R (2000) Serum leptin concentrations during the menstrual cycle in normal-weight women: effects of an oral triphasic estrogen-progestin medication. Eur J Endocrinol 142:174–178PubMedCrossRef Cella F, Giordano G, Cordera R (2000) Serum leptin concentrations during the menstrual cycle in normal-weight women: effects of an oral triphasic estrogen-progestin medication. Eur J Endocrinol 142:174–178PubMedCrossRef
164.
go back to reference Abelenda M, Puerta M (2004) Leptin release is decreased in white adipocytes isolated from progesterone-treated rats. Endocr Res 30:335–342PubMedCrossRef Abelenda M, Puerta M (2004) Leptin release is decreased in white adipocytes isolated from progesterone-treated rats. Endocr Res 30:335–342PubMedCrossRef
165.
go back to reference Zhao M, Chen YH, Chen X, Dong XT, Zhou J, Wang H, Wu SX, Zhang C, Xu DX (2014) Folic acid supplementation during pregnancy protects against lipopolysaccharide-induced neural tube defects in mice. Toxicol Lett 224:201–208 Zhao M, Chen YH, Chen X, Dong XT, Zhou J, Wang H, Wu SX, Zhang C, Xu DX (2014) Folic acid supplementation during pregnancy protects against lipopolysaccharide-induced neural tube defects in mice. Toxicol Lett 224:201–208
166.
go back to reference Antony AC (2007) In utero physiology: role of folic acid in nutrient delivery and fetal development. Am J Clin Nutr 85:598S–603SPubMed Antony AC (2007) In utero physiology: role of folic acid in nutrient delivery and fetal development. Am J Clin Nutr 85:598S–603SPubMed
167.
go back to reference Maggini S, Wintergerst ES, Beveridge S, Hornig DH (2007) Selected vitamins and trace elements support immune function by strengthening epithelial barriers and cellular and humoral immune responses. Br J Nutr 98(Suppl 1):S29–S35 Maggini S, Wintergerst ES, Beveridge S, Hornig DH (2007) Selected vitamins and trace elements support immune function by strengthening epithelial barriers and cellular and humoral immune responses. Br J Nutr 98(Suppl 1):S29–S35
168.
go back to reference Håberg SE, London SJ, Stigum H, Nafstad P, Nystad W (2009) Folic acid supplements in pregnancy and early childhood respiratory health. Arch Dis Child 94:180–184 Håberg SE, London SJ, Stigum H, Nafstad P, Nystad W (2009) Folic acid supplements in pregnancy and early childhood respiratory health. Arch Dis Child 94:180–184
169.
go back to reference Courtemanche C, Elson-Schwab I, Mashiyama ST et al (2004) Folate deficiency inhibits the proliferation of primary human CD8+ T lymphocytes in vitro. J Immunol 173:3186 LP–3183192CrossRef Courtemanche C, Elson-Schwab I, Mashiyama ST et al (2004) Folate deficiency inhibits the proliferation of primary human CD8+ T lymphocytes in vitro. J Immunol 173:3186 LP–3183192CrossRef
170.
go back to reference Yamaguchi T, Hirota K, Nagahama K et al (2007) Control of immune responses by antigen-specific regulatory T cells expressing the folate receptor. Immunity 27:145–159PubMedCrossRef Yamaguchi T, Hirota K, Nagahama K et al (2007) Control of immune responses by antigen-specific regulatory T cells expressing the folate receptor. Immunity 27:145–159PubMedCrossRef
171.
go back to reference Who F (2004) Vitamin and mineral requirements in human nutrition, 2nd edn. World Health Organization, Geneva Who F (2004) Vitamin and mineral requirements in human nutrition, 2nd edn. World Health Organization, Geneva
172.
go back to reference Caprio M, Infante M, Calanchini M et al (2017) Vitamin D: not just the bone. Evidence for beneficial pleiotropic extraskeletal effects. Eat Weight Disord 22:27–41PubMedCrossRef Caprio M, Infante M, Calanchini M et al (2017) Vitamin D: not just the bone. Evidence for beneficial pleiotropic extraskeletal effects. Eat Weight Disord 22:27–41PubMedCrossRef
173.
go back to reference Tamblyn JA, Hewison M, Wagner CL et al (2015) Immunological role of vitamin D at the maternal-fetal interface. J Endocrinol 224:R107–R121PubMedCrossRef Tamblyn JA, Hewison M, Wagner CL et al (2015) Immunological role of vitamin D at the maternal-fetal interface. J Endocrinol 224:R107–R121PubMedCrossRef
174.
go back to reference Nunn JD, Katz DR, Barker S et al (1986) Regulation of human tonsillar T-cell proliferation by the active metabolite of vitamin D3. Immunology 59:479–484PubMedPubMedCentral Nunn JD, Katz DR, Barker S et al (1986) Regulation of human tonsillar T-cell proliferation by the active metabolite of vitamin D3. Immunology 59:479–484PubMedPubMedCentral
175.
go back to reference Vanherwegen AS, Gysemans C, Mathieu C (2017) Vitamin D endocrinology on the cross-road between immunity and metabolism. Mol Cell Endocrinol 453:52–67 Vanherwegen AS, Gysemans C, Mathieu C (2017) Vitamin D endocrinology on the cross-road between immunity and metabolism. Mol Cell Endocrinol 453:52–67
176.
go back to reference Alemzadeh R, Kichler J, Babar G et al (2008) Hypovitaminosis D in obese children and adolescents: relationship with adiposity, insulin sensitivity, ethnicity, and season. Metabolism 57:183–191PubMedCrossRef Alemzadeh R, Kichler J, Babar G et al (2008) Hypovitaminosis D in obese children and adolescents: relationship with adiposity, insulin sensitivity, ethnicity, and season. Metabolism 57:183–191PubMedCrossRef
178.
go back to reference Diemert A, Lezius S, Pagenkemper M et al (2016) Maternal nutrition, inadequate gestational weight gain and birth weight: results from a prospective birth cohort. BMC Pregnancy Childbirth 16:224PubMedPubMedCentralCrossRef Diemert A, Lezius S, Pagenkemper M et al (2016) Maternal nutrition, inadequate gestational weight gain and birth weight: results from a prospective birth cohort. BMC Pregnancy Childbirth 16:224PubMedPubMedCentralCrossRef
179.
go back to reference Wu G, Bazer FW, Davis TA et al (2009) Arginine metabolism and nutrition in growth, health and disease. Amino Acids 37:153–168PubMedCrossRef Wu G, Bazer FW, Davis TA et al (2009) Arginine metabolism and nutrition in growth, health and disease. Amino Acids 37:153–168PubMedCrossRef
180.
181.
182.
go back to reference Bronte V, Zanovello P (2005) Regulation of immune responses by L-arginine metabolism. Nat Rev Immunol 5:641–654PubMedCrossRef Bronte V, Zanovello P (2005) Regulation of immune responses by L-arginine metabolism. Nat Rev Immunol 5:641–654PubMedCrossRef
183.
go back to reference Munder M, Schneider H, Luckner C et al (2006) Suppression of T-cell functions by human granulocyte arginase. Blood 108:1627 LP–1621634CrossRef Munder M, Schneider H, Luckner C et al (2006) Suppression of T-cell functions by human granulocyte arginase. Blood 108:1627 LP–1621634CrossRef
185.
go back to reference Kim JY, Kang JS, Kim HM et al (2009) Inhibition of phenotypic and functional maturation of dendritic cells by manassantin a. J Pharmacol Sci 109:583–592PubMedCrossRef Kim JY, Kang JS, Kim HM et al (2009) Inhibition of phenotypic and functional maturation of dendritic cells by manassantin a. J Pharmacol Sci 109:583–592PubMedCrossRef
186.
go back to reference Simioni PU, Fernandes LGR, Tamashiro WMSC (2016) Downregulation of L-arginine metabolism in dendritic cells induces tolerance to exogenous antigen. Int J Immunopathol Pharmacol 30:44–57CrossRef Simioni PU, Fernandes LGR, Tamashiro WMSC (2016) Downregulation of L-arginine metabolism in dendritic cells induces tolerance to exogenous antigen. Int J Immunopathol Pharmacol 30:44–57CrossRef
187.
go back to reference Greenberg SS, Lancaster JR, Xie J et al (1997) Effects of NO synthase inhibitors, arginine-deficient diet, and amiloride in pregnant rats. Am J Physiol—Regul Integr Comp Physiol 273:R1031 LP–R10R1045CrossRef Greenberg SS, Lancaster JR, Xie J et al (1997) Effects of NO synthase inhibitors, arginine-deficient diet, and amiloride in pregnant rats. Am J Physiol—Regul Integr Comp Physiol 273:R1031 LP–R10R1045CrossRef
188.
go back to reference Buhimschi IR, Saade G, Chwalisz K et al (1998) The nitric oxide pathway in pre-eclampsia: pathophysiological implications. Hum Reprod Update 4:25–42PubMedCrossRef Buhimschi IR, Saade G, Chwalisz K et al (1998) The nitric oxide pathway in pre-eclampsia: pathophysiological implications. Hum Reprod Update 4:25–42PubMedCrossRef
189.
go back to reference Kim YJ, Park HS, Lee HY et al (2006) Reduced l-arginine level and decreased placental eNOS activity in preeclampsia. Placenta 27:438–444PubMedCrossRef Kim YJ, Park HS, Lee HY et al (2006) Reduced l-arginine level and decreased placental eNOS activity in preeclampsia. Placenta 27:438–444PubMedCrossRef
190.
go back to reference Rytlewski K, Olszanecki R, Lauterbach R et al (2006) Effects of oral L-arginine on the foetal condition and neonatal outcome in preeclampsia: a preliminary report. Basic Clin Pharmacol Toxicol 99:146–152PubMedCrossRef Rytlewski K, Olszanecki R, Lauterbach R et al (2006) Effects of oral L-arginine on the foetal condition and neonatal outcome in preeclampsia: a preliminary report. Basic Clin Pharmacol Toxicol 99:146–152PubMedCrossRef
191.
go back to reference Rytlewski K, Olszanecki R, Lauterbach R et al (2008) Effects of oral l-arginine on the pulsatility indices of umbilical artery and middle cerebral artery in preterm labor. Eur J Obstet Gynecol Reprod Biol 138:23–28PubMedCrossRef Rytlewski K, Olszanecki R, Lauterbach R et al (2008) Effects of oral l-arginine on the pulsatility indices of umbilical artery and middle cerebral artery in preterm labor. Eur J Obstet Gynecol Reprod Biol 138:23–28PubMedCrossRef
192.
go back to reference Brown LD, Green AS, Limesand SW et al (2011) Maternal amino acid supplementation for intrauterine growth restriction. Front biosci (Scholar edition) 3:428–444 Brown LD, Green AS, Limesand SW et al (2011) Maternal amino acid supplementation for intrauterine growth restriction. Front biosci (Scholar edition) 3:428–444
193.
go back to reference Gui S, Jia J, Niu X et al (2013) Arginine supplementation for improving maternal and neonatal outcomes in hypertensive disorder of pregnancy: a systematic review. J Renin-Angiotensin-Aldosterone Syst 15:88–96PubMedCrossRef Gui S, Jia J, Niu X et al (2013) Arginine supplementation for improving maternal and neonatal outcomes in hypertensive disorder of pregnancy: a systematic review. J Renin-Angiotensin-Aldosterone Syst 15:88–96PubMedCrossRef
194.
go back to reference Lukaszewski MA, Delahaye F, Vieau D et al (2012) Is the adipose tissue a key target of developmental programming of adult adiposity by maternal undernutrition? Adipocyte 1:64–67PubMedPubMedCentralCrossRef Lukaszewski MA, Delahaye F, Vieau D et al (2012) Is the adipose tissue a key target of developmental programming of adult adiposity by maternal undernutrition? Adipocyte 1:64–67PubMedPubMedCentralCrossRef
195.
go back to reference Jaquet D, Gaboriau A, Czernichow P et al (2001) Relatively low serum leptin levels in adults born with intra-uterine growth retardation. Int J Obes Relat Metab Disord 25:491–495PubMedCrossRef Jaquet D, Gaboriau A, Czernichow P et al (2001) Relatively low serum leptin levels in adults born with intra-uterine growth retardation. Int J Obes Relat Metab Disord 25:491–495PubMedCrossRef
196.
go back to reference Palou M, Konieczna J, Torrens JM et al (2012) Impaired insulin and leptin sensitivity in the offspring of moderate caloric-restricted dams during gestation is early programmed. J Nutr Biochem 23:1627–1639PubMedCrossRef Palou M, Konieczna J, Torrens JM et al (2012) Impaired insulin and leptin sensitivity in the offspring of moderate caloric-restricted dams during gestation is early programmed. J Nutr Biochem 23:1627–1639PubMedCrossRef
197.
go back to reference Palou M, Priego T, Sanchez J et al (2010) Sexual dimorphism in the lasting effects of moderate caloric restriction during gestation on energy homeostasis in rats is related with fetal programming of insulin and leptin resistance. Nutr Metab (Lond) 7:69CrossRef Palou M, Priego T, Sanchez J et al (2010) Sexual dimorphism in the lasting effects of moderate caloric restriction during gestation on energy homeostasis in rats is related with fetal programming of insulin and leptin resistance. Nutr Metab (Lond) 7:69CrossRef
198.
go back to reference Vickers MH1, Breier BH, Cutfield WS, Hofman PL, Gluckman PD (2000) Fetal origins of hyperphagia, obesity, and hypertension and postnatal amplification by hypercaloric nutrition. Am J Physiol Endocrinol Metab 279:E83–87 Vickers MH1, Breier BH, Cutfield WS, Hofman PL, Gluckman PD (2000) Fetal origins of hyperphagia, obesity, and hypertension and postnatal amplification by hypercaloric nutrition. Am J Physiol Endocrinol Metab 279:E83–87
199.
go back to reference Grasemann C, Herrmann R, Starschinova J et al (2017) Effects of fetal exposure to high-fat diet or maternal hyperglycemia on L-arginine and nitric oxide metabolism in lung. Nutr Diabetes e244:7 Grasemann C, Herrmann R, Starschinova J et al (2017) Effects of fetal exposure to high-fat diet or maternal hyperglycemia on L-arginine and nitric oxide metabolism in lung. Nutr Diabetes e244:7
200.
go back to reference Mayor RS, Finch KE, Zehr J, Morselli E, Neinast MD, Frank AP, Hahner LD, Wang J, Rakheja D, Palmer BF, Rosenfeld CR, Savani RC, Clegg DJ (2015) Maternal high-fat diet is associated with impaired fetal lung development. Am J Physiol Lung Cell Mol Physiol 309:L360–368 Mayor RS, Finch KE, Zehr J, Morselli E, Neinast MD, Frank AP, Hahner LD, Wang J, Rakheja D, Palmer BF, Rosenfeld CR, Savani RC, Clegg DJ (2015) Maternal high-fat diet is associated with impaired fetal lung development. Am J Physiol Lung Cell Mol Physiol 309:L360–368
Metadata
Title
Immunometabolism, pregnancy, and nutrition
Authors
Kristin Thiele
Lianghui Diao
Petra Clara Arck
Publication date
01-02-2018
Publisher
Springer Berlin Heidelberg
Published in
Seminars in Immunopathology / Issue 2/2018
Print ISSN: 1863-2297
Electronic ISSN: 1863-2300
DOI
https://doi.org/10.1007/s00281-017-0660-y

Other articles of this Issue 2/2018

Seminars in Immunopathology 2/2018 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.