Skip to main content
Erschienen in: Medical Microbiology and Immunology 3-4/2019

07.05.2019 | Original Investigation

Survey of cellular immune responses to human cytomegalovirus infection in the microenvironment of the uterine–placental interface

verfasst von: Takako Tabata, Matthew Petitt, June Fang-Hoover, Lenore Pereira

Erschienen in: Medical Microbiology and Immunology | Ausgabe 3-4/2019

Einloggen, um Zugang zu erhalten

Abstract

Congenital human cytomegalovirus (HCMV) infection is a leading cause of birth defects, yet there are no established treatments for preventing maternal–fetal transmission. During first trimester, HCMV replicates in basal decidua that functions as a reservoir for virus and source of transmission to the attached placenta and fetal hemiallograft but also contains immune cells, including natural killer cells, macrophages, and T cell subsets, that respond to pathogens, protecting the placenta and fetus. However, the specific cellular and cytokine responses to infection are unknown, nor are the immune correlates of protection that guide development of therapeutic strategies. Here we survey immune cell phenotypes in intact explants of basal decidua infected with a clinical pathogenic HCMV strain ex vivo and identify specific changes occurring in response to infection in the tissue environment. Using 4-color immunofluorescence microscopy, we found that at 3 days postinfection, virus replicates in decidual stromal cells and epithelial cells of endometrial glands. Infected cells and effector memory CD8+ T cells (TEM) in contact with them make IFN-γ. CD8+ TEM cells produce granulysin and cluster at sites of infection in decidua and the epithelium of endometrial glands. Quantification indicated expansion of two immune cell subtypes—CD8+ TEM cells and, to a lesser extent, iNKT cells. Approximately 20% of immune cells were found in pairs in both control and infected decidua, suggesting frequent cross-talk in the microenvironment of decidua. Our findings indicate a complex immune microenvironment in basal decidua and suggest CD8+ TEM cells play a role in early responses to decidual infection in seropositive women.
Literatur
1.
Zurück zum Zitat Pass RF, Fowler KB, Boppana SB, Britt WJ, Stagno S (2006) Congenital cytomegalovirus infection following first trimester maternal infection: symptoms at birth and outcome. J Clin Virol 35:216–220PubMed Pass RF, Fowler KB, Boppana SB, Britt WJ, Stagno S (2006) Congenital cytomegalovirus infection following first trimester maternal infection: symptoms at birth and outcome. J Clin Virol 35:216–220PubMed
2.
Zurück zum Zitat Enders G, Daiminger A, Bader U, Exler S, Enders M (2011) Intrauterine transmission and clinical outcome of 248 pregnancies with primary cytomegalovirus infection in relation to gestational age. J Clin Virol 52:244–246PubMed Enders G, Daiminger A, Bader U, Exler S, Enders M (2011) Intrauterine transmission and clinical outcome of 248 pregnancies with primary cytomegalovirus infection in relation to gestational age. J Clin Virol 52:244–246PubMed
3.
Zurück zum Zitat Picone O, Vauloup-Fellous C, Cordier AG, Guitton S, Senat MV, Fuchs F, Ayoubi JM, Grangeot Keros L, Benachi A (2013) A series of 238 cytomegalovirus primary infections during pregnancy: description and outcome. Prenat Diagn 33:751–758PubMed Picone O, Vauloup-Fellous C, Cordier AG, Guitton S, Senat MV, Fuchs F, Ayoubi JM, Grangeot Keros L, Benachi A (2013) A series of 238 cytomegalovirus primary infections during pregnancy: description and outcome. Prenat Diagn 33:751–758PubMed
4.
Zurück zum Zitat Faure-Bardon V, Magny JF, Parodi M, Couderc S, Garcia P, Maillotte AM, Benard M, Pinquier D, Astruc D, Patural H, Pladys P, Parat S, Guillois B, Garenne A, Bussieres L, Guilleminot T, Stirnemann J, Ghout I, Ville Y, Leruez-Ville M (2018) Sequelae of congenital cytomegalovirus (cCMV) following maternal primary infection are limited to those acquired in the first trimester of pregnancy. Clin Infect Dis. https://doi.org/10.1093/cid/ciy1128 CrossRef Faure-Bardon V, Magny JF, Parodi M, Couderc S, Garcia P, Maillotte AM, Benard M, Pinquier D, Astruc D, Patural H, Pladys P, Parat S, Guillois B, Garenne A, Bussieres L, Guilleminot T, Stirnemann J, Ghout I, Ville Y, Leruez-Ville M (2018) Sequelae of congenital cytomegalovirus (cCMV) following maternal primary infection are limited to those acquired in the first trimester of pregnancy. Clin Infect Dis. https://​doi.​org/​10.​1093/​cid/​ciy1128 CrossRef
5.
Zurück zum Zitat Pereira L, Maidji E, McDonagh S, Genbacev O, Fisher S (2003) Human cytomegalovirus transmission from the uterus to the placenta correlates with the presence of pathogenic bacteria and maternal immunity. J Virol 77:13301–13314PubMedPubMedCentral Pereira L, Maidji E, McDonagh S, Genbacev O, Fisher S (2003) Human cytomegalovirus transmission from the uterus to the placenta correlates with the presence of pathogenic bacteria and maternal immunity. J Virol 77:13301–13314PubMedPubMedCentral
6.
Zurück zum Zitat Weisblum Y, Panet A, Zakay-Rones Z, Haimov-Kochman R, Goldman-Wohl D, Ariel I, Falk H, Natanson-Yaron S, Goldberg MD, Gilad R, Lurain NS, Greenfield C, Yagel S, Wolf DG (2011) Modeling of human cytomegalovirus maternal–fetal transmission in a novel decidual organ culture. J Virol 85:13204–13213PubMedPubMedCentral Weisblum Y, Panet A, Zakay-Rones Z, Haimov-Kochman R, Goldman-Wohl D, Ariel I, Falk H, Natanson-Yaron S, Goldberg MD, Gilad R, Lurain NS, Greenfield C, Yagel S, Wolf DG (2011) Modeling of human cytomegalovirus maternal–fetal transmission in a novel decidual organ culture. J Virol 85:13204–13213PubMedPubMedCentral
7.
Zurück zum Zitat Pereira L, Petitt M, Fong A, Tsuge M, Tabata T, Fang-Hoover J, Maidji E, Zydek M, Zhou Y, Inoue N, Loghavi S, Pepkowitz S, Kauvar LM, Ogunyemi D (2014) Intrauterine growth restriction caused by underlying congenital cytomegalovirus infection. J Infect Dis 209:1573–1584PubMedPubMedCentral Pereira L, Petitt M, Fong A, Tsuge M, Tabata T, Fang-Hoover J, Maidji E, Zydek M, Zhou Y, Inoue N, Loghavi S, Pepkowitz S, Kauvar LM, Ogunyemi D (2014) Intrauterine growth restriction caused by underlying congenital cytomegalovirus infection. J Infect Dis 209:1573–1584PubMedPubMedCentral
8.
Zurück zum Zitat McDonagh S, Maidji E, Ma W, Chang HT, Fisher S, Pereira L (2004) Viral and bacterial pathogens at the maternal–fetal interface. J Infect Dis 190:826–834PubMed McDonagh S, Maidji E, Ma W, Chang HT, Fisher S, Pereira L (2004) Viral and bacterial pathogens at the maternal–fetal interface. J Infect Dis 190:826–834PubMed
9.
Zurück zum Zitat McDonagh S, Maidji E, Chang HT, Pereira L (2006) Patterns of human cytomegalovirus infection in term placentas: a preliminary analysis. J Clin Virol 35:210–215PubMed McDonagh S, Maidji E, Chang HT, Pereira L (2006) Patterns of human cytomegalovirus infection in term placentas: a preliminary analysis. J Clin Virol 35:210–215PubMed
10.
Zurück zum Zitat Nozawa N, Fang-Hoover J, Tabata T, Maidji E, Pereira L (2009) Cytomegalovirus-specific, high-avidity IgG with neutralizing activity in maternal circulation enriched in the fetal bloodstream. J Clin Virol 46(Suppl 4):S58–63PubMedPubMedCentral Nozawa N, Fang-Hoover J, Tabata T, Maidji E, Pereira L (2009) Cytomegalovirus-specific, high-avidity IgG with neutralizing activity in maternal circulation enriched in the fetal bloodstream. J Clin Virol 46(Suppl 4):S58–63PubMedPubMedCentral
11.
Zurück zum Zitat Tabata T, Petitt M, Fang-Hoover J, Zydek M, Pereira L (2016) Persistent cytomegalovirus infection in amniotic membranes of the human placenta. Am J Pathol 186:2970–2986PubMedPubMedCentral Tabata T, Petitt M, Fang-Hoover J, Zydek M, Pereira L (2016) Persistent cytomegalovirus infection in amniotic membranes of the human placenta. Am J Pathol 186:2970–2986PubMedPubMedCentral
12.
Zurück zum Zitat Tabata T, Petitt M, Puerta-Guardo H, Michlmayr D, Harris E, Pereira L (2018) Zika virus replicates in proliferating cells in explants from first-trimester human placentas, potential sites for dissemination of infection. J Infect Dis 217:1202–1213PubMed Tabata T, Petitt M, Puerta-Guardo H, Michlmayr D, Harris E, Pereira L (2018) Zika virus replicates in proliferating cells in explants from first-trimester human placentas, potential sites for dissemination of infection. J Infect Dis 217:1202–1213PubMed
13.
Zurück zum Zitat Weisblum Y, Oiknine-Djian E, Vorontsov OM, Haimov-Kochman R, Zakay-Rones Z, Meir K, Shveiky D, Elgavish S, Nevo Y, Roseman M, Bronstein M, Stockheim D, From I, Eisenberg I, Lewkowicz AA, Yagel S, Panet A, Wolf DG (2017) Zika virus infects early- and midgestation human maternal decidual tissues, inducing distinct innate tissue responses in the maternal-fetal interface. J Virol 91:e01905–01916PubMedPubMedCentral Weisblum Y, Oiknine-Djian E, Vorontsov OM, Haimov-Kochman R, Zakay-Rones Z, Meir K, Shveiky D, Elgavish S, Nevo Y, Roseman M, Bronstein M, Stockheim D, From I, Eisenberg I, Lewkowicz AA, Yagel S, Panet A, Wolf DG (2017) Zika virus infects early- and midgestation human maternal decidual tissues, inducing distinct innate tissue responses in the maternal-fetal interface. J Virol 91:e01905–01916PubMedPubMedCentral
14.
Zurück zum Zitat Fornara C, Furione M, Arossa A, Gerna G, Lilleri D (2016) Comparative magnitude and kinetics of human cytomegalovirus-specific CD4+(+) and CD8+(+) T-cell responses in pregnant women with primary versus remote infection and in transmitting versus non-transmitting mothers: its utility for dating primary infection in pregnancy. J Med Virol 88:1238–1246PubMed Fornara C, Furione M, Arossa A, Gerna G, Lilleri D (2016) Comparative magnitude and kinetics of human cytomegalovirus-specific CD4+(+) and CD8+(+) T-cell responses in pregnant women with primary versus remote infection and in transmitting versus non-transmitting mothers: its utility for dating primary infection in pregnancy. J Med Virol 88:1238–1246PubMed
15.
Zurück zum Zitat Lilleri D, Kabanova A, Revello MG, Percivalle E, Sarasini A, Genini E, Sallusto F, Lanzavecchia A, Corti D, Gerna G (2013) Fetal human cytomegalovirus transmission correlates with delayed maternal antibodies to gH/gL/pUL128-130-131 complex during primary infection. PLoS One 8:e59863PubMedPubMedCentral Lilleri D, Kabanova A, Revello MG, Percivalle E, Sarasini A, Genini E, Sallusto F, Lanzavecchia A, Corti D, Gerna G (2013) Fetal human cytomegalovirus transmission correlates with delayed maternal antibodies to gH/gL/pUL128-130-131 complex during primary infection. PLoS One 8:e59863PubMedPubMedCentral
17.
Zurück zum Zitat Tabata T, Petitt M, Puerta-Guardo H, Michlmayr D, Wang C, Fang-Hoover J, Harris E, Pereira L (2016) Zika virus targets different primary human placental cells, suggesting two routes for vertical transmission. Cell Host Microbe 20:155–166PubMedPubMedCentral Tabata T, Petitt M, Puerta-Guardo H, Michlmayr D, Wang C, Fang-Hoover J, Harris E, Pereira L (2016) Zika virus targets different primary human placental cells, suggesting two routes for vertical transmission. Cell Host Microbe 20:155–166PubMedPubMedCentral
19.
Zurück zum Zitat van den Heuvel M, Peralta C, Bashar S, Taylor S, Horrocks J, Croy BA (2005) Trafficking of peripheral blood CD56(bright) cells to the decidualizing uterus-new tricks for old dogmas? J Reprod Immunol 67:21–34PubMedPubMedCentral van den Heuvel M, Peralta C, Bashar S, Taylor S, Horrocks J, Croy BA (2005) Trafficking of peripheral blood CD56(bright) cells to the decidualizing uterus-new tricks for old dogmas? J Reprod Immunol 67:21–34PubMedPubMedCentral
20.
Zurück zum Zitat Chantakru S, Miller C, Roach LE, Kuziel WA, Maeda N, Wang WC, Evans SS, Croy BA (2002) Contributions from self-renewal and trafficking to the uterine NK cell population of early pregnancy. J Immunol 168:22–28PubMed Chantakru S, Miller C, Roach LE, Kuziel WA, Maeda N, Wang WC, Evans SS, Croy BA (2002) Contributions from self-renewal and trafficking to the uterine NK cell population of early pregnancy. J Immunol 168:22–28PubMed
21.
Zurück zum Zitat Lissauer D, Choudhary M, Pachnio A, Goodyear O, Moss PA, Kilby MD (2011) Cytomegalovirus sero positivity dramatically alters the maternal CD8++ T cell repertoire and leads to the accumulation of highly differentiated memory cells during human pregnancy. Hum Reprod 26:3355–3365PubMed Lissauer D, Choudhary M, Pachnio A, Goodyear O, Moss PA, Kilby MD (2011) Cytomegalovirus sero positivity dramatically alters the maternal CD8++ T cell repertoire and leads to the accumulation of highly differentiated memory cells during human pregnancy. Hum Reprod 26:3355–3365PubMed
22.
Zurück zum Zitat Mele F, Fornara C, Jarrossay D, Furione M, Arossa A, Spinillo A, Lanzavecchia A, Gerna G, Sallusto F, Lilleri D (2017) Phenotype and specificity of T cells in primary human cytomegalovirus infection during pregnancy: IL-7Rpos long-term memory phenotype is associated with protection from vertical transmission. PLoS One 12:e0187731PubMedPubMedCentral Mele F, Fornara C, Jarrossay D, Furione M, Arossa A, Spinillo A, Lanzavecchia A, Gerna G, Sallusto F, Lilleri D (2017) Phenotype and specificity of T cells in primary human cytomegalovirus infection during pregnancy: IL-7Rpos long-term memory phenotype is associated with protection from vertical transmission. PLoS One 12:e0187731PubMedPubMedCentral
23.
Zurück zum Zitat Tilburgs T, Schonkeren D, Eikmans M, Nagtzaam NM, Datema G, Swings GM, Prins F, van Lith JM, van der Mast BJ, Roelen DL, Scherjon SA, Claas FH (2010) Human decidual tissue contains differentiated CD8++ effector-memory T cells with unique properties. J Immunol 185:4470–4477PubMed Tilburgs T, Schonkeren D, Eikmans M, Nagtzaam NM, Datema G, Swings GM, Prins F, van Lith JM, van der Mast BJ, Roelen DL, Scherjon SA, Claas FH (2010) Human decidual tissue contains differentiated CD8++ effector-memory T cells with unique properties. J Immunol 185:4470–4477PubMed
24.
Zurück zum Zitat Tilburgs T, Strominger JL (2013) CD8++ effector T cells at the fetal–maternal interface, balancing fetal tolerance and antiviral immunity. Am J Reprod Immunol 69:395–407PubMed Tilburgs T, Strominger JL (2013) CD8++ effector T cells at the fetal–maternal interface, balancing fetal tolerance and antiviral immunity. Am J Reprod Immunol 69:395–407PubMed
25.
Zurück zum Zitat Lilleri D, Fornara C, Furione M, Zavattoni M, Revello MG, Gerna G (2007) Development of human cytomegalovirus-specific T cell immunity during primary infection of pregnant women and its correlation with virus transmission to the fetus. J Infect Dis 195:1062–1070PubMed Lilleri D, Fornara C, Furione M, Zavattoni M, Revello MG, Gerna G (2007) Development of human cytomegalovirus-specific T cell immunity during primary infection of pregnant women and its correlation with virus transmission to the fetus. J Infect Dis 195:1062–1070PubMed
26.
Zurück zum Zitat Dotiwala F, Mulik S, Polidoro RB, Ansara JA, Burleigh BA, Walch M, Gazzinelli RT, Lieberman J (2016) Killer lymphocytes use granulysin, perforin and granzymes to kill intracellular parasites. Nat Med 22:210–216PubMedPubMedCentral Dotiwala F, Mulik S, Polidoro RB, Ansara JA, Burleigh BA, Walch M, Gazzinelli RT, Lieberman J (2016) Killer lymphocytes use granulysin, perforin and granzymes to kill intracellular parasites. Nat Med 22:210–216PubMedPubMedCentral
27.
Zurück zum Zitat van der Zwan A, Bi K, Norwitz ER, Crespo AC, Claas FHJ, Strominger JL, Tilburgs T (2018) Mixed signature of activation and dysfunction allows human decidual CD8+(+) T cells to provide both tolerance and immunity. Proc Natl Acad Sci USA 115:385–390PubMed van der Zwan A, Bi K, Norwitz ER, Crespo AC, Claas FHJ, Strominger JL, Tilburgs T (2018) Mixed signature of activation and dysfunction allows human decidual CD8+(+) T cells to provide both tolerance and immunity. Proc Natl Acad Sci USA 115:385–390PubMed
28.
Zurück zum Zitat Deng A, Chen S, Li Q, Lyu SC, Clayberger C, Krensky AM (2005) Granulysin, a cytolytic molecule, is also a chemoattractant and proinflammatory activator. J Immunol 174:5243–5248PubMed Deng A, Chen S, Li Q, Lyu SC, Clayberger C, Krensky AM (2005) Granulysin, a cytolytic molecule, is also a chemoattractant and proinflammatory activator. J Immunol 174:5243–5248PubMed
29.
Zurück zum Zitat Whitmire JK, Tan JT, Whitton JL (2005) Interferon-gamma acts directly on CD8++ T cells to increase their abundance during virus infection. J Exp Med 201:1053–1059PubMedPubMedCentral Whitmire JK, Tan JT, Whitton JL (2005) Interferon-gamma acts directly on CD8++ T cells to increase their abundance during virus infection. J Exp Med 201:1053–1059PubMedPubMedCentral
30.
Zurück zum Zitat Krummel MF, Mahale JN, Uhl LFK, Hardison EA, Mujal AM, Mazet JM, Weber RJ, Gartner ZJ, Gerard A (2018) Paracrine costimulation of IFN-gamma signaling by integrins modulates CD8+ T cell differentiation. Proc Natl Acad Sci U S A 115:11585–11590PubMedPubMedCentral Krummel MF, Mahale JN, Uhl LFK, Hardison EA, Mujal AM, Mazet JM, Weber RJ, Gartner ZJ, Gerard A (2018) Paracrine costimulation of IFN-gamma signaling by integrins modulates CD8+ T cell differentiation. Proc Natl Acad Sci U S A 115:11585–11590PubMedPubMedCentral
31.
Zurück zum Zitat Albayati Z, Alyami A, Alomar S, Middleton D, Bonnett L, Aleem S, Flanagan BF, Christmas SE (2017) The influence of cytomegalovirus on expression of HLA-G and its ligand KIR2DL4 by human peripheral blood leucocyte subsets. Scand J Immunol 86:396–407PubMed Albayati Z, Alyami A, Alomar S, Middleton D, Bonnett L, Aleem S, Flanagan BF, Christmas SE (2017) The influence of cytomegalovirus on expression of HLA-G and its ligand KIR2DL4 by human peripheral blood leucocyte subsets. Scand J Immunol 86:396–407PubMed
32.
Zurück zum Zitat Kos FJ, Engleman EG (1995) Requirement for natural killer cells in the induction of cytotoxic T cells. J Immunol 155:578–584PubMed Kos FJ, Engleman EG (1995) Requirement for natural killer cells in the induction of cytotoxic T cells. J Immunol 155:578–584PubMed
33.
Zurück zum Zitat Kos FJ, Chin CS (2002) Costimulation of T cell receptor-triggered IL-2 production by Jurkat T cells via fibroblast growth factor receptor 1 upon its engagement by CD56. Immunol Cell Biol 80:364–369PubMed Kos FJ, Chin CS (2002) Costimulation of T cell receptor-triggered IL-2 production by Jurkat T cells via fibroblast growth factor receptor 1 upon its engagement by CD56. Immunol Cell Biol 80:364–369PubMed
34.
Zurück zum Zitat Sorensen V, Zhen Y, Zakrzewska M, Haugsten EM, Walchli S, Nilsen T, Olsnes S, Wiedlocha A (2008) Phosphorylation of fibroblast growth factor (FGF) receptor 1 at Ser777 by p38 mitogen-activated protein kinase regulates translocation of exogenous FGF1 to the cytosol and nucleus. Mol Cell Biol 28:4129–4141PubMedPubMedCentral Sorensen V, Zhen Y, Zakrzewska M, Haugsten EM, Walchli S, Nilsen T, Olsnes S, Wiedlocha A (2008) Phosphorylation of fibroblast growth factor (FGF) receptor 1 at Ser777 by p38 mitogen-activated protein kinase regulates translocation of exogenous FGF1 to the cytosol and nucleus. Mol Cell Biol 28:4129–4141PubMedPubMedCentral
35.
Zurück zum Zitat Ahmed Z, Schuller AC, Suhling K, Tregidgo C, Ladbury JE (2008) Extracellular point mutations in FGFR2 elicit unexpected changes in intracellular signalling. Biochem J 413:37–49PubMed Ahmed Z, Schuller AC, Suhling K, Tregidgo C, Ladbury JE (2008) Extracellular point mutations in FGFR2 elicit unexpected changes in intracellular signalling. Biochem J 413:37–49PubMed
37.
Zurück zum Zitat O'Reilly V, Zeng SG, Bricard G, Atzberger A, Hogan AE, Jackson J, Feighery C, Porcelli SA, Doherty DG (2011) Distinct and overlapping effector functions of expanded human CD4++, CD8+ alpha+ and CD4+-CD8+alpha-invariant natural killer T cells. PLoS One 6:e28648PubMedPubMedCentral O'Reilly V, Zeng SG, Bricard G, Atzberger A, Hogan AE, Jackson J, Feighery C, Porcelli SA, Doherty DG (2011) Distinct and overlapping effector functions of expanded human CD4++, CD8+ alpha+ and CD4+-CD8+alpha-invariant natural killer T cells. PLoS One 6:e28648PubMedPubMedCentral
38.
Zurück zum Zitat Ishikawa H, Tanaka K, Kutsukake E, Fukui T, Sasaki H, Hata A, Noda S, Matsumoto T (2010) IFN-gamma production downstream of NKT cell activation in mice infected with influenza virus enhances the cytolytic activities of both NK cells and viral antigen-specific CD8++ T cells. Virology 407:325–332PubMed Ishikawa H, Tanaka K, Kutsukake E, Fukui T, Sasaki H, Hata A, Noda S, Matsumoto T (2010) IFN-gamma production downstream of NKT cell activation in mice infected with influenza virus enhances the cytolytic activities of both NK cells and viral antigen-specific CD8++ T cells. Virology 407:325–332PubMed
39.
Zurück zum Zitat Tyznik AJ, Verma S, Wang Q, Kronenberg M, Benedict CA (2014) Distinct requirements for activation of NKT and NK cells during viral infection. J Immunol 192:3676–3685PubMed Tyznik AJ, Verma S, Wang Q, Kronenberg M, Benedict CA (2014) Distinct requirements for activation of NKT and NK cells during viral infection. J Immunol 192:3676–3685PubMed
40.
Zurück zum Zitat Pass RF, Zhang C, Evans A, Simpson T, Andrews W, Huang ML, Corey L, Hill J, Davis E, Flanigan C, Cloud G (2009) Vaccine prevention of maternal cytomegalovirus infection. N Engl J Med 360:1191–1199PubMedPubMedCentral Pass RF, Zhang C, Evans A, Simpson T, Andrews W, Huang ML, Corey L, Hill J, Davis E, Flanigan C, Cloud G (2009) Vaccine prevention of maternal cytomegalovirus infection. N Engl J Med 360:1191–1199PubMedPubMedCentral
41.
Zurück zum Zitat Baraniak I, Kropff B, Ambrose L, McIntosh M, McLean GR, Pichon S, Atkinson C, Milne RSB, Mach M, Griffiths PD, Reeves MB (2018) Protection from cytomegalovirus viremia following glycoprotein B vaccination is not dependent on neutralizing antibodies. Proc Natl Acad Sci USA 115:6273–6278PubMedPubMedCentral Baraniak I, Kropff B, Ambrose L, McIntosh M, McLean GR, Pichon S, Atkinson C, Milne RSB, Mach M, Griffiths PD, Reeves MB (2018) Protection from cytomegalovirus viremia following glycoprotein B vaccination is not dependent on neutralizing antibodies. Proc Natl Acad Sci USA 115:6273–6278PubMedPubMedCentral
42.
Zurück zum Zitat Baraniak I, Kropff B, McLean GR, Pichon S, Piras-Douce F, Milne RSB, Smith C, Mach M, Griffiths PD, Reeves MB (2018) Epitope-specific humoral responses to human cytomegalovirus glycoprotein-B vaccine with MF59: anti-AD2 levels correlate with protection from viremia. J Infect Dis 217:1907–1917PubMedPubMedCentral Baraniak I, Kropff B, McLean GR, Pichon S, Piras-Douce F, Milne RSB, Smith C, Mach M, Griffiths PD, Reeves MB (2018) Epitope-specific humoral responses to human cytomegalovirus glycoprotein-B vaccine with MF59: anti-AD2 levels correlate with protection from viremia. J Infect Dis 217:1907–1917PubMedPubMedCentral
43.
Zurück zum Zitat Baker K, Rath T, Lencer WI, Fiebiger E, Blumberg RS (2013) Cross-presentation of IgG-containing immune complexes. Cell Mol Life Sci 70:1319–1334PubMed Baker K, Rath T, Lencer WI, Fiebiger E, Blumberg RS (2013) Cross-presentation of IgG-containing immune complexes. Cell Mol Life Sci 70:1319–1334PubMed
44.
Zurück zum Zitat Rath T, Kuo TT, Baker K, Qiao SW, Kobayashi K, Yoshida M, Roopenian D, Fiebiger E, Lencer WI, Blumberg RS (2013) The immunologic functions of the neonatal Fc receptor for IgG. J Clin Immunol 33(Suppl 1):S9–17PubMed Rath T, Kuo TT, Baker K, Qiao SW, Kobayashi K, Yoshida M, Roopenian D, Fiebiger E, Lencer WI, Blumberg RS (2013) The immunologic functions of the neonatal Fc receptor for IgG. J Clin Immunol 33(Suppl 1):S9–17PubMed
45.
Zurück zum Zitat Baker K, Qiao SW, Kuo TT, Aveson VG, Platzer B, Andersen JT, Sandlie I, Chen Z, de Haar C, Lencer WI, Fiebiger E, Blumberg RS (2011) Neonatal Fc receptor for IgG (FcRn) regulates cross-presentation of IgG immune complexes by CD8+-CD11b+ dendritic cells. Proc Natl Acad Sci USA 108:9927–9932PubMedPubMedCentral Baker K, Qiao SW, Kuo TT, Aveson VG, Platzer B, Andersen JT, Sandlie I, Chen Z, de Haar C, Lencer WI, Fiebiger E, Blumberg RS (2011) Neonatal Fc receptor for IgG (FcRn) regulates cross-presentation of IgG immune complexes by CD8+-CD11b+ dendritic cells. Proc Natl Acad Sci USA 108:9927–9932PubMedPubMedCentral
46.
Zurück zum Zitat Kagan KO, Enders M, Schampera MS, Baeumel E, Hoopmann M, Geipel A, Berg C, Goelz R, De Catte L, Wallwiener D, Brucker S, Adler SP, Jahn G, Hamprecht K (2018) Prevention of maternal–fetal transmission of CMV by hyperimmunoglobulin (HIG) administered after a primary maternal CMV infectionin early gestation. Ultrasound Obstet Gynecol. https://doi.org/10.1002/uog.19164 CrossRefPubMed Kagan KO, Enders M, Schampera MS, Baeumel E, Hoopmann M, Geipel A, Berg C, Goelz R, De Catte L, Wallwiener D, Brucker S, Adler SP, Jahn G, Hamprecht K (2018) Prevention of maternal–fetal transmission of CMV by hyperimmunoglobulin (HIG) administered after a primary maternal CMV infectionin early gestation. Ultrasound Obstet Gynecol. https://​doi.​org/​10.​1002/​uog.​19164 CrossRefPubMed
47.
Zurück zum Zitat Buxmann H, Stackelberg OM, Schlosser RL, Enders G, Gonser M, Meyer-Wittkopf M, Hamprecht K, Enders M (2012) Use of cytomegalovirus hyperimmunoglobulin for prevention of congenital cytomegalovirus disease: a retrospective analysis. J Perinat Med 40:439–446PubMed Buxmann H, Stackelberg OM, Schlosser RL, Enders G, Gonser M, Meyer-Wittkopf M, Hamprecht K, Enders M (2012) Use of cytomegalovirus hyperimmunoglobulin for prevention of congenital cytomegalovirus disease: a retrospective analysis. J Perinat Med 40:439–446PubMed
48.
Zurück zum Zitat Nigro G, Adler SP, La Torre R, Best AM (2005) Passive immunization during pregnancy for congenital cytomegalovirus infection. N Engl J Med 353:1350–1362PubMed Nigro G, Adler SP, La Torre R, Best AM (2005) Passive immunization during pregnancy for congenital cytomegalovirus infection. N Engl J Med 353:1350–1362PubMed
49.
Zurück zum Zitat Revello MG, Lazzarotto T, Guerra B, Spinillo A, Ferrazzi E, Kustermann A, Guaschino S, Vergani P, Todros T, Frusca T, Arossa A, Furione M, Rognoni V, Rizzo N, Gabrielli L, Klersy C, Gerna G, Group CS (2014) A randomized trial of hyperimmune globulin to prevent congenital cytomegalovirus. N Engl J Med 370:1316–1326 Revello MG, Lazzarotto T, Guerra B, Spinillo A, Ferrazzi E, Kustermann A, Guaschino S, Vergani P, Todros T, Frusca T, Arossa A, Furione M, Rognoni V, Rizzo N, Gabrielli L, Klersy C, Gerna G, Group CS (2014) A randomized trial of hyperimmune globulin to prevent congenital cytomegalovirus. N Engl J Med 370:1316–1326
50.
Zurück zum Zitat Qiao SW, Kobayashi K, Johansen FE, Sollid LM, Andersen JT, Milford E, Roopenian DC, Lencer WI, Blumberg RS (2008) Dependence of antibody-mediated presentation of antigen on FcRn. Proc Natl Acad Sci USA 105:9337–9342PubMedPubMedCentral Qiao SW, Kobayashi K, Johansen FE, Sollid LM, Andersen JT, Milford E, Roopenian DC, Lencer WI, Blumberg RS (2008) Dependence of antibody-mediated presentation of antigen on FcRn. Proc Natl Acad Sci USA 105:9337–9342PubMedPubMedCentral
51.
Zurück zum Zitat Lee J, Zhang T, Hwang I, Kim A, Nitschke L, Kim M, Scott JM, Kamimura Y, Lanier LL, Kim S (2015) Epigenetic modification and antibody-dependent expansion of memory-like NK cells in human cytomegalovirus-infected individuals. Immunity 42:431–442PubMedPubMedCentral Lee J, Zhang T, Hwang I, Kim A, Nitschke L, Kim M, Scott JM, Kamimura Y, Lanier LL, Kim S (2015) Epigenetic modification and antibody-dependent expansion of memory-like NK cells in human cytomegalovirus-infected individuals. Immunity 42:431–442PubMedPubMedCentral
52.
Zurück zum Zitat Sun JC, Beilke JN, Lanier LL (2010) Immune memory redefined: characterizing the longevity of natural killer cells. Immunol Rev 236:83–94PubMedPubMedCentral Sun JC, Beilke JN, Lanier LL (2010) Immune memory redefined: characterizing the longevity of natural killer cells. Immunol Rev 236:83–94PubMedPubMedCentral
53.
Zurück zum Zitat Liu ST, Sharon-Friling R, Ivanova P, Milne SB, Myers DS, Rabinowitz JD, Brown HA, Shenk T (2011) Synaptic vesicle-like lipidome of human cytomegalovirus virions reveals a role for SNARE machinery in virion egress. Proc Natl Acad Sci USA 108:12869–12874PubMedPubMedCentral Liu ST, Sharon-Friling R, Ivanova P, Milne SB, Myers DS, Rabinowitz JD, Brown HA, Shenk T (2011) Synaptic vesicle-like lipidome of human cytomegalovirus virions reveals a role for SNARE machinery in virion egress. Proc Natl Acad Sci USA 108:12869–12874PubMedPubMedCentral
54.
Zurück zum Zitat McEwen-Smith RM, Salio M, Cerundolo V (2015) CD1d-dependent endogenous and exogenous lipid antigen presentation. Curr Opin Immunol 34:116–125PubMed McEwen-Smith RM, Salio M, Cerundolo V (2015) CD1d-dependent endogenous and exogenous lipid antigen presentation. Curr Opin Immunol 34:116–125PubMed
55.
Zurück zum Zitat Gordon CL, Miron M, Thome JJ, Matsuoka N, Weiner J, Rak MA, Igarashi S, Granot T, Lerner H, Goodrum F, Farber DL (2017) Tissue reservoirs of antiviral T cell immunity in persistent human CMV infection. J Exp Med 214:651–667PubMedPubMedCentral Gordon CL, Miron M, Thome JJ, Matsuoka N, Weiner J, Rak MA, Igarashi S, Granot T, Lerner H, Goodrum F, Farber DL (2017) Tissue reservoirs of antiviral T cell immunity in persistent human CMV infection. J Exp Med 214:651–667PubMedPubMedCentral
56.
Zurück zum Zitat Vento-Tormo R, Efremova M, Botting RA, Turco MY, Vento-Tormo M, Meyer KB, Park JE, Stephenson E, Polanski K, Goncalves A, Gardner L, Holmqvist S, Henriksson J, Zou A, Sharkey AM, Millar B, Innes B, Wood L, Wilbrey-Clark A, Payne RP, Ivarsson MA, Lisgo S, Filby A, Rowitch DH, Bulmer JN, Wright GJ, Stubbington MJT, Haniffa M, Moffett A, Teichmann SA (2018) Single-cell reconstruction of the early maternal-fetal interface in humans. Nature 563:347–353PubMed Vento-Tormo R, Efremova M, Botting RA, Turco MY, Vento-Tormo M, Meyer KB, Park JE, Stephenson E, Polanski K, Goncalves A, Gardner L, Holmqvist S, Henriksson J, Zou A, Sharkey AM, Millar B, Innes B, Wood L, Wilbrey-Clark A, Payne RP, Ivarsson MA, Lisgo S, Filby A, Rowitch DH, Bulmer JN, Wright GJ, Stubbington MJT, Haniffa M, Moffett A, Teichmann SA (2018) Single-cell reconstruction of the early maternal-fetal interface in humans. Nature 563:347–353PubMed
58.
Zurück zum Zitat Gao GF, Willcox BE, Wyer JR, Boulter JM, O'Callaghan CA, Maenaka K, Stuart DI, Jones EY, Van Der Merwe PA, Bell JI, Jakobsen BK (2000) Classical and nonclassical class I major histocompatibility complex molecules exhibit subtle conformational differences that affect binding to CD8+alphaalpha. J Biol Chem 275:15232–15238PubMed Gao GF, Willcox BE, Wyer JR, Boulter JM, O'Callaghan CA, Maenaka K, Stuart DI, Jones EY, Van Der Merwe PA, Bell JI, Jakobsen BK (2000) Classical and nonclassical class I major histocompatibility complex molecules exhibit subtle conformational differences that affect binding to CD8+alphaalpha. J Biol Chem 275:15232–15238PubMed
Metadaten
Titel
Survey of cellular immune responses to human cytomegalovirus infection in the microenvironment of the uterine–placental interface
verfasst von
Takako Tabata
Matthew Petitt
June Fang-Hoover
Lenore Pereira
Publikationsdatum
07.05.2019
Verlag
Springer Berlin Heidelberg
Erschienen in
Medical Microbiology and Immunology / Ausgabe 3-4/2019
Print ISSN: 0300-8584
Elektronische ISSN: 1432-1831
DOI
https://doi.org/10.1007/s00430-019-00613-w

Weitere Artikel der Ausgabe 3-4/2019

Medical Microbiology and Immunology 3-4/2019 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.