Pregnancy-Induced Alterations in NK Cell Phenotype and Function.
Journal: 2019/November - Frontiers in Immunology
ISSN: 1664-3224
Abstract:
Pregnant women are particularly susceptible to complications of influenza A virus infection, which may result from pregnancy-induced changes in the function of immune cells, including natural killer (NK) cells. To better understand NK cell function during pregnancy, we assessed the ability of the two main subsets of NK cells, CD56dim, and CD56bright NK cells, to respond to influenza-virus infected cells and tumor cells. During pregnancy, CD56dim and CD56bright NK cells displayed enhanced functional responses to both infected and tumor cells, with increased expression of degranulation markers and elevated frequency of NK cells producing IFN-γ. To better understand the mechanisms driving this enhanced function, we profiled CD56dim and CD56bright NK cells from pregnant and non-pregnant women using mass cytometry. NK cells from pregnant women displayed significantly increased expression of several functional and activation markers such as CD38 on both subsets and NKp46 on CD56dim NK cells. NK cells also displayed diminished expression of the chemokine receptor CXCR3 during pregnancy. Overall, these data demonstrate that functional and phenotypic shifts occur in NK cells during pregnancy that can influence the magnitude of the immune response to both infections and tumors.
Relations:
Content
Citations
(8)
References
(57)
Diseases
(1)
Genes
(1)
Organisms
(1)
Processes
(2)
Anatomy
(1)
Affiliates
(2)
Similar articles
Articles by the same authors
Discussion board
Front Immunol 10: 2469

Pregnancy-Induced Alterations in NK Cell Phenotype and Function

+5 authors
Click here for additional data file.(18K, DOCX)
Click here for additional data file.(5.8M, PDF)
Department of Medicine, Stanford University, Palo Alto, CA, United States
Department of Stanford Immunology Program, Stanford University, Palo Alto, CA, United States
Department of Statistics, Stanford University, Palo Alto, CA, United States
Department of Pediatrics, Stanford University, Palo Alto, CA, United States
Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
Department of Obstetrics and Gynecology, Stanford Prevention Research Center, Stanford University School of Medicine, Stanford University, Palo Alto, CA, United States
Department of Microbiology and Immunology, Stanford University, Palo Alto, CA, United States
Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, United States
Chan Zuckerberg Biohub, San Francisco, CA, United States
Edited by: Henrik Mei, Deutsches Rheuma-Forschungszentrum (DRFZ), Germany
Reviewed by: Kanutte Huse, Oslo University Hospital, Norway; Quirin Hammer, Karolinska Institute (KI), Sweden; Bertram Bengsch, Freiburg University Medical Center, Germany
*Correspondence: Mathieu Le Gars ude.drofnats@hsilbc
Catherine A. Blish moc.liamg@1sragel.ueihtam
This article was submitted to NK and Innate Lymphoid Cell Biology, a section of the journal Frontiers in Immunology
Edited by: Henrik Mei, Deutsches Rheuma-Forschungszentrum (DRFZ), Germany
Reviewed by: Kanutte Huse, Oslo University Hospital, Norway; Quirin Hammer, Karolinska Institute (KI), Sweden; Bertram Bengsch, Freiburg University Medical Center, Germany
Received 2019 May 23; Accepted 2019 Oct 3.
This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

Abstract

Pregnant women are particularly susceptible to complications of influenza A virus infection, which may result from pregnancy-induced changes in the function of immune cells, including natural killer (NK) cells. To better understand NK cell function during pregnancy, we assessed the ability of the two main subsets of NK cells, CD56, and CD56 NK cells, to respond to influenza-virus infected cells and tumor cells. During pregnancy, CD56 and CD56 NK cells displayed enhanced functional responses to both infected and tumor cells, with increased expression of degranulation markers and elevated frequency of NK cells producing IFN-γ. To better understand the mechanisms driving this enhanced function, we profiled CD56 and CD56 NK cells from pregnant and non-pregnant women using mass cytometry. NK cells from pregnant women displayed significantly increased expression of several functional and activation markers such as CD38 on both subsets and NKp46 on CD56 NK cells. NK cells also displayed diminished expression of the chemokine receptor CXCR3 during pregnancy. Overall, these data demonstrate that functional and phenotypic shifts occur in NK cells during pregnancy that can influence the magnitude of the immune response to both infections and tumors.

Keywords: NK cells, pregnancy, influenza virus, cancer cells, NK repertoire
Abstract

Acknowledgments

We thank our study volunteers for their participation, Sally Mackey for regulatory and data management, Sue Swope for consenting and conducting study visits, and the staff of the Stanford Vaccine Program for overall study coordination. This manuscript has been released as a Pre-Print at Biorxiv (62).

Acknowledgments

Footnotes

Funding. This was supported by an Elizabeth and Russell Siegelman Fellowship in Infectious Diseases from the Stanford Child Health Research Institute (CHRI) to AK, a Stanford CHRI post-doctoral fellowship to ML, the CHRI – Stanford Clinical and Translational Science Award grant number UL1 TR000093 (AK), a National Institutes of Health (NIH) Training Grant: Viral Infections in Children T32 AI78896-05 (AK), a Smith Family Stanford Graduate Fellowship (NB), Ruth L. Kirschstein NRSA 1F31HD089675 (NB), a Clinical Scientist Development Award #2013099 from the Doris Duke Charitable Foundation (CB), the McCormick Faculty Award (CB), Tasha and John Morgridge Endowed Faculty Scholar in Pediatric Translational Medicine from Stanford CHRI and Stanford University School of Medicine (CB), a NIH Director's New Innovator Award DP2AI112193 (CB), an Infrastructure and Opportunity Fund (CB) as part of the Stanford Human Immunology Project Consortium (HIPC) Grant U19AI090019 (MD), and an investigator award from the Chan Zuckerberg Biohub (CB). Clinical cohorts were supported by NIH U19AI057229 (MD) and an NIH/NCRR CTSA award UL1 RR025744 (H. Greenberg).

Footnotes
Click here for additional data file.(18K, DOCX)Click here for additional data file.(5.8M, PDF)

References

  • 1. Erlebacher A. Immunology of the maternal-fetal interface. Ann Rev Immunol. (2013) 31:387–411. 10.1146/annurev-immunol-032712-100003 [] [[PubMed]
  • 2. Kourtis AP, Read JS, Jamieson DJ. Pregnancy and infection. N Engl J Med. (2014) 370:2211–8. 10.1056/NEJMra1213566 ] [
  • 3. Periolo N, Avaro M, Czech A, Russo M, Benedetti E, Pontoriero A, et al. . Pregnant women infected with pandemic influenza A(H1N1)pdm09 virus showed differential immune response correlated with disease severity. J Clin Virol. (2015) 64:52–8. 10.1016/j.jcv.2015.01.009 [] [[PubMed]
  • 4. PrabhuDas M, Bonney E, Caron K, Dey S, Erlebacher A, Fazleabas A, et al. . Immune mechanisms at the maternal-fetal interface: perspectives and challenges. Nat Immunol. (2015) 16:328–34. 10.1038/ni.3131 ] [
  • 5. Littauer EQ, Esser ES, Antao OQ, Vassilieva EV, Compans RW, Skountzou I. H1N1 influenza virus infection results in adverse pregnancy outcomes by disrupting tissue-specific hormonal regulation. PLoS Pathogens. (2017) 13:e1006757. 10.1371/journal.ppat.1006757 ] [
  • 6. Pazos M, Sperling RS, Moran TM, Kraus TA. The influence of pregnancy on systemic immunity. Immunol Res. (2012) 54:254–61. 10.1007/s12026-012-8303-9 [] [[PubMed]
  • 7. Omer SB, Bednarczyk R, Madhi SA, Klugman KP. Benefits to mother and child of influenza vaccination during pregnancy. Hum Vacc Immunother. (2012) 8:130–7. 10.4161/hv.8.1.18601 [] [[PubMed]
  • 8. Raj RS, Bonney EA, Phillippe M. Influenza, immune system, and pregnancy. Reprod Sci. (2014) 21:1434–51. 10.1177/1933719114537720 ] [
  • 9. Siston AM, Rasmussen SA, Honein MA, Fry AM, Seib K, Callaghan WM, et al. . Pandemic 2009 influenza A(H1N1) virus illness among pregnant women in the United States. JAMA. (2010) 303:1517–25. 10.1001/jama.2010.479 ] [
  • 10. Aghaeepour N, Ganio EA, Mcilwain D, Tsai AS, Tingle M, Van Gassen S, et al. . An immune clock of human pregnancy. Sci Immunol. (2017) 2:eaan2946. 10.1126/sciimmunol.aan2946 ] [
  • 11. Gars ML, Le Gars M, Kay AW, Bayless NL, Aziz N, Dekker CL, et al. . Increased proinflammatory responses of monocytes and plasmacytoid dendritic cells to influenza A virus infection during pregnancy. J Infect Dis. (2016) 214:1666–71. 10.1093/infdis/jiw448 ] [
  • 12. Kay AW, Fukuyama J, Aziz N, Dekker CL, Mackey S, Swan GE, et al. . Enhanced natural killer-cell and T-cell responses to influenza A virus during pregnancy. Proc Natl Acad Sci USA. (2014) 111:14506–11. 10.1073/pnas.1416569111 ] [
  • 13. Kraus TA, Engel SM, Sperling RS, Kellerman L, Lo Y, Wallenstein S, et al. . Characterizing the pregnancy immune phenotype: results of the viral immunity and pregnancy (VIP) study. J Clin Immunol. (2012) 32:300–11. 10.1007/s10875-011-9627-2 [] [[PubMed]
  • 14. Kraus TA, Sperling RS, Engel SM, Lo Y, Kellerman L, Singh T, et al. . Peripheral blood cytokine profiling during pregnancy and post-partum periods. Am J Reprod Immunol. (2010) 64:411–26. 10.1111/j.1600-0897.2010.00889.x [] [[PubMed]
  • 15. Abdul-Careem MF, Mian MF, Yue G, Gillgrass A, Chenoweth MJ, Barra NG, et al. . Critical role of natural killer cells in lung immunopathology during influenza infection in mice. J Infect Dis. (2012) 206:167–77. 10.1093/infdis/jis340 [] [[PubMed]
  • 16. Zhou G, Juang SWW, Kane KP. NK cells exacerbate the pathology of influenza virus infection in mice. Eur J Immunol. (2013) 43:929–38. 10.1002/eji.201242620 [] [[PubMed]
  • 17. Gazit R, Gruda R, Elboim M, Arnon TI, Katz G, Achdout H, et al. . Lethal influenza infection in the absence of the natural killer cell receptor gene Ncr1. Nat Immunol. (2006) 7:517–23. 10.1038/ni1322 [] [[PubMed]
  • 18. Narni-Mancinelli E, Jaeger BN, Bernat C, Fenis A, Kung S, De Gassart A, et al. . Tuning of natural killer cell reactivity by NKp46 and Helios calibrates T cell responses. Science. (2012) 335:344–8. 10.1126/science.1215621 [] [[PubMed]
  • 19. Mauad T, Hajjar LA, Callegari GD, da Silva LFF, Schout D, Galas FRBG, et al. . Lung pathology in fatal novel human influenza A (H1N1) infection. Am J Respirat Critic Care Med. (2010) 181:72–9. 10.1164/rccm.200909-1420OC [] [[PubMed]
  • 20. Strauss-Albee DM, Horowitz A, Parham P, Blish CA. Coordinated regulation of NK receptor expression in the maturing human immune system. J Immunol. (2014) 193:4871–9. 10.4049/jimmunol.1401821 ] [
  • 21. Vivier E, Tomasello E, Baratin M, Walzer T, Ugolini S. Functions of natural killer cells. Nat Immunol. (2008) 9:503–10. 10.1038/ni1582 [] [[PubMed]
  • 22. Cooper MA, Fehniger TA, Turner SC, Chen KS, Ghaheri BA, Ghayur T, et al Human natural killer cells: a unique innate immunoregulatory role for the CD56bright subset. Blood. (2001) 97:3146–51. 10.1182/blood.V97.10.3146 [] [[PubMed][Google Scholar]
  • 23. Nagler A, Lanier LL, Cwirla S, Phillips JH. Comparative studies of human FcRIII-positive and negative natural killer cells. J Immunol. (1989) 143:3183–91. [[PubMed]
  • 24. Kay AW, Strauss-Albee DM, Blish CA. Application of mass cytometry (CyTOF) for functional and phenotypic analysis of natural killer cells. Methods Mol Biol. (2016) 1441:13–26. 10.1007/978-1-4939-3684-7_2 ] [
  • 25. Strauss-Albee DM, Blish CA. CyTOF: single cell mass cytometry for evaluation of complex innate cellular phenotypes. In: Montgomery R, Bucala R, editors. , editors. Experimental Approaches for the Investigation of Innate Immunity. World Scientific; Yale University School of Medicine (2015). p. 27–39. 10.1142/9789814678735_0003 [[PubMed]
  • 26. Horowitz A, Strauss-Albee DM, Leipold M, Kubo J, Nemat-Gorgani N, Dogan OC, et al. . Genetic and environmental determinants of human NK cell diversity revealed by mass cytometry. Sci Transl Med. (2013) 5:208ra145. 10.1126/scitranslmed.3006702 ] [
  • 27. Seiler C, Kronstad LM, Simpson LJ, Le Gars M, Vendrame E, Blish CA, et al Uncertainty Quantification in Multivariate Mixed Models for Mass Cytometry Data. (2019). Retrieved from: (accessed October 15, 2019).[PubMed][Google Scholar]
  • 28. Gentleman R, Carey V, Huber W, Irizarry R, Dudoit S. Bioinformatics and Computational Biology Solutions Using R and Bioconductor. Springer Science & Business Media (2006). 10.1007/0-387-29362-0 [[PubMed]
  • 29. Ripley B, Venables B, Bates DM, Hornik K, Gebhardt A, Firth D. MASS: Support Functions and Datasets for Venables and Ripley's MASS. R Package Version 7–3. (2011). [PubMed]
  • 30. Kronstad LM, Seiler C, Vergara R, Holmes SP, Blish CA. Differential induction of IFN-α and modulation of CD112 and CD54 expression govern the magnitude of NK cell IFN-γ response to influenza A viruses. J Immunol. (2018) 201:2117–31. 10.4049/jimmunol.1800161 ] [
  • 31. Hilton HG, Parham P. Missing or altered self: human NK cell receptors that recognize HLA-C. Immunogenetics. (2017) 69:567–79. 10.1007/s00251-017-1001-y ] [
  • 32. Veenstra van Nieuwenhoven AL, Bouman A, Moes H, Heineman MJ, de Leij LFMH, Santema J, et al Cytokine production in natural killer cells and lymphocytes in pregnant women compared with women in the follicular phase of the ovarian cycle. Fertil Steril. (2002) 77:1032–7. 10.1016/S0015-0282(02)02976-X [] [[PubMed][Google Scholar]
  • 33. Glasner A, Zurunic A, Meningher T, Lenac Rovis T, Tsukerman P, Bar-On Y, et al. . Elucidating the mechanisms of influenza virus recognition by Ncr1. PLoS ONE. (2012) 7:e36837. 10.1371/journal.pone.0036837 ] [
  • 34. Mandelboim O, Lieberman N, Lev M, Paul L, Arnon TI, Bushkin Y, et al. . Recognition of haemagglutinins on virus-infected cells by NKp46 activates lysis by human NK cells. Nature. (2001) 409:1055–60. 10.1038/35059110 [] [[PubMed]
  • 35. Rasmussen SA, Jamieson DJ, Bresee JS. Pandemic influenza and pregnant women. Emerg Infect Dis. (2008) 14:95–100. 10.3201/eid1401.070667 ] [
  • 36. Centers for Disease Control and Prevention (CDC) Estimates of deaths associated with seasonal influenza — United States, 1976-2007. MMWR. MMWR Morb Mortal Wkly Rep. (2010) 59:1057–62. [[PubMed]
  • 37. Carlin LE, Hemann EA, Zacharias ZR, Heusel JW, Legge KL. Natural killer cell recruitment to the lung during influenza A virus infection is dependent on CXCR3, CCR5, and virus exposure dose. Front Immunol. (2018) 9:781. 10.3389/fimmu.2018.00781 ] [
  • 38. Kim HM, Kang YM, Song BM, Kim HS, Seo SH. The 2009 pandemic H1N1 influenza virus is more pathogenic in pregnant mice than seasonal H1N1 influenza virus. Viral Immunol. (2012) 25:402–10. 10.1089/vim.2012.0007 [] [[PubMed]
  • 39. Cheung CY, Poon LLM, Lau AS, Luk W, Lau YL, Shortridge KF, et al. . Induction of proinflammatory cytokines in human macrophages by influenza A (H5N1) viruses: a mechanism for the unusual severity of human disease?Lancet. (2002) 360:1831–7. 10.1016/S0140-6736(02)11772-7 [] [[PubMed]
  • 40. de Jong MD, Simmons CP, Thanh TT, Hien VM, Smith GJD, Chau TNB, et al. . Fatal outcome of human influenza A (H5N1) is associated with high viral load and hypercytokinemia. Nat Med. (2006) 12:1203–7. 10.1038/nm1477 ] [
  • 41. Kobasa D, Jones SM, Shinya K, Kash JC, Copps J, Ebihara H, et al. . Aberrant innate immune response in lethal infection of macaques with the 1918 influenza virus. Nature. (2007) 445:319–23. 10.1038/nature05495 [] [[PubMed]
  • 42. Deaglio S, Mallone R, Baj G, Arnulfo A, Surico N, Dianzani U, et al. . CD38/CD31, a receptor/ligand system ruling adhesion and signaling in human leukocytes. Chem Immunol. (2000) 75:99–120. 10.1159/000058765 [] [[PubMed]
  • 43. Lee HC. Structure and enzymatic functions of human CD38. Mol Med. (2006) 12:317–23. 10.2119/2006-00086.Lee ] [
  • 44. Deaglio S, Zubiaur M, Gregorini A, Bottarel F, Ausiello CM, Dianzani U, et al. . Human CD38 and CD16 are functionally dependent and physically associated in natural killer cells. Blood. (2002) 99:2490–8. 10.1182/blood.V99.7.2490 [] [[PubMed]
  • 45. Mallone R, Funaro A, Zubiaur M, Baj G, Ausiello CM, Tacchetti C, et al. . Signaling through CD38 induces NK cell activation. Int Immunol. (2001) 13:397–409. 10.1093/intimm/13.4.397 [] [[PubMed]
  • 46. Sconocchia G, Titus JA, Mazzoni A, Visintin A, Pericle F, Hicks SW, et al. . CD38 triggers cytotoxic responses in activated human natural killer cells. Blood. (1999) 94:3864–71. [[PubMed]
  • 47. Muñoz P, Mittelbrunn M, de la Fuente H, Pérez-Martínez M, García-Pérez A, Ariza-Veguillas A, et al. . Antigen-induced clustering of surface CD38 and recruitment of intracellular CD38 to the immunologic synapse. Blood. (2008) 111:3653–64. 10.1182/blood-2007-07-101600 [] [[PubMed]
  • 48. Koopman LA, Kopcow HD, Rybalov B, Boyson JE, Orange JS, Schatz F, et al. . Human decidual natural killer cells are a unique NK cell subset with immunomodulatory potential. J Exp Med. (2003) 198:1201–12. 10.1084/jem.20030305 ] [
  • 49. Carlino C, Stabile H, Morrone S, Bulla R, Soriani A, Agostinis C, et al. . Recruitment of circulating NK cells through decidual tissues: a possible mechanism controlling NK cell accumulation in the uterus during early pregnancy. Blood. (2008) 111:3108–15. 10.1182/blood-2007-08-105965 [] [[PubMed]
  • 50. Keskin DB, Allan DSJ, Rybalov B, Andzelm MM, Stern JNH, Kopcow HD, et al. . TGFβ promotes conversion of CD16+ peripheral blood NK cells into CD16– NK cells with similarities to decidual NK cells. Proc Natl Acad Sci USA. (2007) 104:3378–83. 10.1073/pnas.0611098104 ] [
  • 51. Goodridge JP, Lathbury LJ, John E, Charles AK, Christiansen FT, Witt CS. The genotype of the NK cell receptor, KIR2DL4, influences INF secretion by decidual natural killer cells. Mol Hum Reproduct. (2009) 15:489–97. 10.1093/molehr/gap039 [] [[PubMed]
  • 52. Li C, Houser BL, Nicotra ML, Strominger JL. HLA-G homodimer-induced cytokine secretion through HLA-G receptors on human decidual macrophages and natural killer cells. Proc Natl Acad Sci USA. (2009) 106:5767–72. 10.1073/pnas.0901173106 ] [
  • 53. Rajagopalan S, Bryceson YT, Kuppusamy SP, Geraghty DE, van der Meer A, Joosten I, et al. . Activation of NK cells by an endocytosed receptor for soluble HLA-G. PLoS Biol. (2005) 4:e9. 10.1371/journal.pbio.0040009 ] [
  • 54. Gamliel M, Goldman-Wohl D, Isaacson B, Gur C, Stein N, Yamin R, et al. . Trained memory of human uterine NK cells enhances their function in subsequent pregnancies. Immunity. (2018) 48:951–62.e5. 10.1016/j.immuni.2018.03.030 [] [[PubMed]
  • 55. Glasner A, Isaacson B, Viukov S, Neuman T, Friedman N, Mandelboim M, et al. . Increased NK cell immunity in a transgenic mouse model of NKp46 overexpression. Sci Rep. (2017) 7:13090. 10.1038/s41598-017-12998-w ] [
  • 56. Shi L, Li K, Guo Y, Banerjee A, Wang Q, Lorenz UM, et al. . Modulation of NKG2D, NKp46, and Ly49C/I facilitates natural killer cell-mediated control of lung cancer. Proc Natl Acad Sci USA. (2018) 115:11808–13. 10.1073/pnas.1804931115 ] [
  • 57. Han B, Mao F-Y, Zhao Y-L, Lv Y-P, Teng Y-S, Duan M, et al. . Altered NKp30, NKp46, NKG2D, and DNAM-1 expression on circulating NK cells is associated with tumor progression in human gastric cancer. J Immunol Res. (2018) 2018:6248590. 10.1155/2018/6248590 ] [
  • 58. Ichikawa A, Kuba K, Morita M, Chida S, Tezuka H, Hara H, et al. . CXCL10-CXCR3 enhances the development of neutrophil-mediated fulminant lung injury of viral and nonviral origin. Am J Respirat Critic Care Med. (2013) 187:65–77. 10.1164/rccm.201203-0508OC ] [
  • 59. Soudja SM, Ruiz AL, Marie JC, Lauvau G. Inflammatory monocytes activate memory CD8 T and innate NK lymphocytes independent of cognate antigen during microbial pathogen invasion. Immunity. (2012) 37:549–62. 10.1016/j.immuni.2012.05.029 ] [
  • 60. Arruvito L, Giulianelli S, Flores AC, Paladino N, Barboza M, Lanari C, et al. . NK cells expressing a progesterone receptor are susceptible to progesterone-induced apoptosis. J Immunol. (2008) 180:5746–53. 10.4049/jimmunol.180.8.5746 [] [[PubMed]
  • 61. Nilsson N, Carlsten H. Estrogen induces suppression of natural killer cell cytotoxicity and augmentation of polyclonal B cell activation. Cell Immunol. (1994) 158:131–9. 10.1006/cimm.1994.1262 [] [[PubMed]
  • 62. Le Gars M, Seiler C, Kay AW, Bayless NL, Starosvetsky E, Moore L, et al CD38 contributes to human natural killer cell responses through a role in immune synapse formation. bioRxiv [Preprint]. 10.1101/349084 [[PubMed][Google Scholar]
Collaboration tool especially designed for Life Science professionals.Drag-and-drop any entity to your messages.