NK Cell Memory to Cytomegalovirus: Implications for Vaccine Development
Abstract
:1. Introduction
2. NK Cells
3. NK Cell Memory
3.1. Antigen-Specific Memory in Liver-Resident NK Cells
3.2. Cytokine-Induced NK Memory
3.3. CMV-Driven NK Memory
4. NK Cells and HCMV
5. HCMV Vaccination: Current Strategies
5.1. Challenges of HCMV Vaccine Development
5.2. Current Candidates
6. NK Cells in HCMV Vaccination
6.1. NK Cell Support of Adaptive Responses
6.2. NK Cells as Effectors of the Antibody Response
6.3. Induction of NK Cell Memory
7. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Dupont, L.; Reeves, M.B. Cytomegalovirus latency and reactivation: Recent insights into an age old problem. Rev. Med. Virol. 2016, 26, 75–89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Staras, S.A.; Dollard, S.C.; Radford, K.W.; Flanders, W.D.; Pass, R.F.; Cannon, M.J. Seroprevalence of cytomegalovirus infection in the United States, 1988–1994. Clin. Infect. Dis. 2006, 43, 1143–1151. [Google Scholar] [CrossRef] [PubMed]
- Zuhair, M.; Smit, G.; Wallis, G.; Jabbar, F.; Smith, C.; Devleesschauwer, B.; Griffiths, P. Estimation of the worldwide seroprevalence of cytomegalovirus: A systematic review and meta-analysis. Rev. Med. Virol. 2019, 29, e2034. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- De Vries, J.J.; Vossen, A.C.; Kroes, A.C.; van der Zeijst, B.A. Implementing neonatal screening for congenital cytomegalovirus: Addressing the deafness of policy makers. Rev. Med. Virol. 2011, 21, 54–61. [Google Scholar] [CrossRef] [PubMed]
- Cannon, M.J.; Griffiths, P.D.; Aston, V.; Rawlinson, W.D. Universal newborn screening for congenital CMV infection: What is the evidence of potential benefit? Rev. Med. Virol. 2014, 24, 291–307. [Google Scholar] [CrossRef] [Green Version]
- Legendre, C.; Pascual, M. Improving outcomes for solid-organ transplant recipients at risk from cytomegalovirus infection: Late-onset disease and indirect consequences. Clin. Infect. Dis. 2008, 46, 732–740. [Google Scholar] [CrossRef] [Green Version]
- Ljungman, P.; Hakki, M.; Boeckh, M. Cytomegalovirus in hematopoietic stem cell transplant recipients. Hematol. Oncol. Clin. N. Am. 2011, 25, 151–169. [Google Scholar] [CrossRef]
- Griffiths, P.; Baraniak, I.; Reeves, M. The pathogenesis of human cytomegalovirus. J. Pathol. 2015, 235, 288–297. [Google Scholar] [CrossRef]
- Arvin, A.M.; Fast, P.; Myers, M.; Plotkin, S.; Rabinovich, R.; National Vaccine Advisory Committee. Vaccine development to prevent cytomegalovirus disease: Report from the National Vaccine Advisory Committee. Clin. Infect. Dis. 2004, 39, 233–239. [Google Scholar] [CrossRef] [PubMed]
- Plotkin, S.A.; Wang, D.; Oualim, A.; Diamond, D.J.; Kotton, C.N.; Mossman, S.; Carfi, A.; Anderson, D.; Dormitzer, P.R. The status of vaccine development against the human cytomegalovirus. J. Infect. Dis. 2020, 221, S113–S122. [Google Scholar] [CrossRef] [Green Version]
- Herberman, R.B.; Nunn, M.E.; Lavrin, D.H. Natural cytotoxic reactivity of mouse lymphoid cells against syngeneic acid allogeneic tumors. I. Distribution of reactivity and specificity. Int. J. Cancer. 1975, 16, 216–229. [Google Scholar] [CrossRef] [PubMed]
- Mace, E.M.; Orange, J.S. Emerging insights into human health and NK cell biology from the study of NK cell deficiencies. Immunol. Rev. 2019, 287, 202–225. [Google Scholar] [CrossRef] [PubMed]
- Kärre, K. Natural killer cell recognition of missing self. Nat. Immunol. 2008, 9, 477–480. [Google Scholar] [CrossRef] [PubMed]
- Lanier, L.L. Up on the tightrope: Natural killer cell activation and inhibition. Nat. Immunol. 2008, 9, 495–502. [Google Scholar] [CrossRef]
- Arase, H.; Mocarski, E.S.; Campbell, A.E.; Hill, A.B.; Lanier, L.L. Direct recognition of cytomegalovirus by activating and inhibitory NK cell receptors. Science 2002, 296, 1323–1326. [Google Scholar] [CrossRef]
- Doherty, D.G.; O’Farrelly, C. Innate and adaptive lymphoid cells in the human liver. Immunol. Rev. 2000, 174, 5–20. [Google Scholar] [CrossRef]
- Male, V. Liver-resident NK cells: The human factor. Trends Immunol. 2017, 38, 307–309. [Google Scholar] [CrossRef]
- Peng, H.; Tian, Z. Re-examining the origin and function of liver-resident NK cells. Trends Immunol. 2015, 36, 293–299. [Google Scholar] [CrossRef]
- Monin, L.; Whettlock, E.M.; Male, V. Immune responses in the human female reproductive tract. Immunology 2020, 160, 106–115. [Google Scholar] [CrossRef] [Green Version]
- O’Leary, J.G.; Goodarzi, M.; Drayton, D.L.; von Andrian, U.H. T cell-and B cell-independent adaptive immunity mediated by natural killer cells. Nat. Immunol. 2006, 7, 507–516. [Google Scholar] [CrossRef]
- Paust, S.; Gill, H.S.; Wang, B.Z.; Flynn, M.P.; Moseman, E.A.; Senman, B.; Szczepanik, M.; Telenti, A.; Askenase, P.W.; Compans, R.W.; et al. Critical role for the chemokine receptor CXCR6 in NK cell-mediated antigen-specific memory of haptens and viruses. Nat. Immunol. 2010, 11, 1127–1135. [Google Scholar] [CrossRef] [PubMed]
- Peng, H.; Jiang, X.; Chen, Y.; Sojka, D.K.; Wei, H.; Gao, X.; Sun, R.; Yokoyama, W.M.; Tian, Z. Liver-resident NK cells confer adaptive immunity in skin-contact inflammation. J. Clin. Investig. 2013, 123, 1444–1456. [Google Scholar] [CrossRef] [Green Version]
- Wight, A.; Mahmoud, A.B.; Scur, M.; Tu, M.M.; Rahim, M.M.A.; Sad, S.; Makrigiannis, A.P. Critical role for the Ly49 family of class I MHC receptors in adaptive natural killer cell responses. Proc. Natl. Acad. Sci. USA 2018, 115, 11579–11584. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Reeves, R.K.; Li, H.; Jost, S.; Blass, E.; Li, H.; Schafer, J.L.; Varner, V.; Manickam, C.; Eslamizar, L.; Altfeld, M.; et al. Antigen-specific NK cell memory in rhesus macaques. Nat. Immunol. 2015, 16, 927–932. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nikzad, R.; Angelo, L.S.; Aviles-Padilla, K.; Le, D.T.; Singh, V.K.; Bimler, L.; Vukmanovic-Stejic, M.; Vendrame, E.; Ranganath, T.; Simpson, L.; et al. Human natural killer cells mediate adaptive immunity to viral antigens. Sci. Immunol. 2019, 10, 4. [Google Scholar] [CrossRef] [PubMed]
- Cooper, M.A.; Elliott, J.M.; Keyel, P.A.; Yang, L.; Carrero, J.A.; Yokoyama, W.M. Cytokine-induced memory-like natural killer cells. Proc. Natl. Acad. Sci. USA 2009, 106, 1915–1919. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ni, J.; Miller, M.; Stojanovic, A.; Garbi, N.; Cerwenka, A. Sustained effector function of IL-12/15/18-preactivated NK cells against established tumors. J. Exp. Med. 2012, 209, 2351–2365. [Google Scholar] [CrossRef]
- Romee, R.; Rosario, M.; Berrien-Elliott, M.M.; Wagner, J.A.; Jewell, B.A.; Schappe, T.; Leong, J.W.; Abdel-Latif, S.; Schneider, S.E.; Willey, S.; et al. Cytokine-induced memory-like natural killer cells exhibit enhanced responses against myeloid leukemia. Sci. Transl. Med. 2016, 8, 357ra123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Romee, R.; Schneider, S.E.; Leong, J.W.; Chase, J.M.; Keppel, C.R.; Sullivan, R.P.; Cooper, M.A.; Fehniger, T.A. Cytokine activation induces human memory-like NK cells. Blood 2012, 120, 4751–4760. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ni, J.; Hölsken, O.; Miller, M.; Hammer, Q.; Luetke-Eversloh, M.; Romagnani, C.; Cerwenka, A. Adoptively transferred natural killer cells maintain long-term antitumor activity by epigenetic imprinting and CD4+ T cell help. Oncoimmunology 2016, 5, e1219009. [Google Scholar] [CrossRef] [Green Version]
- Hammer, Q.; Romagnani, C. About training and memory: NK-cell adaptation to viral infections. Adv. Immunol. 2017, 133, 171–207. [Google Scholar] [CrossRef] [PubMed]
- Dokun, A.O.; Kim, S.; Smith, H.R.; Kang, H.S.; Chu, D.T.; Yokoyama, W.M. Specific and nonspecific NK cell activation during virus infection. Nat. Immunol. 2001, 2, 951–956. [Google Scholar] [CrossRef] [PubMed]
- Sun, J.C.; Beilke, J.N.; Lanier, L.L. Adaptive immune features of natural killer cells. Nature 2009, 457, 557–561. [Google Scholar] [CrossRef] [PubMed]
- Min-Oo, G.; Lanier, L.L. Cytomegalovirus generates long-lived antigen-specific NK cells with diminished bystander activation to heterologous infection. J. Exp. Med. 2014, 211, 2669–2680. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sun, J.C.; Madera, S.; Bezman, N.A.; Beilke, J.N.; Kaplan, M.H.; Lanier, L.L. Proinflammatory cytokine signaling required for the generation of natural killer cell memory. J. Exp. Med. 2012, 209, 947–954. [Google Scholar] [CrossRef] [Green Version]
- Nabekura, T.; Kanaya, M.; Shibuya, A.; Fu, G.; Gascoigne, N.R.; Lanier, L.L. Costimulatory molecule DNAM-1 is essential for optimal differentiation of memory natural killer cells during mouse cytomegalovirus infection. Immunity 2014, 40, 225–234. [Google Scholar] [CrossRef] [Green Version]
- Weizman, O.E.; Adams, N.M.; Schuster, I.S.; Krishna, C.; Pritykin, Y.; Lau, C.; Degli-Esposti, M.A.; Leslie, C.S.; Sun, J.C.; O’Sullivan, T.E. ILC1 confer early host protection at initial sites of viral infection. Cell 2017, 171, 795–808.e12. [Google Scholar] [CrossRef]
- Weizman, O.E.; Song, E.; Adams, N.M.; Hildreth, A.D.; Riggan, L.; Krishna, C.; Aguilar, O.A.; Leslie, C.S.; Carlyle, J.R.; Sun, J.C.; et al. Mouse cytomegalovirus-experienced ILC1s acquire a memory response dependent on the viral glycoprotein m12. Nat. Immunol. 2019, 20, 1004–1011. [Google Scholar] [CrossRef]
- Aguilar, O.A.; Berry, R.; Rahim, M.M.A.; Reichel, J.J.; Popović, B.; Tanaka, M.; Fu, Z.; Balaji, G.R.; Lau, T.N.H.; Tu, M.M.; et al. A viral immunoevasin controls innate immunity by targeting the prototypical natural killer cell receptor family. Cell 2017, 169, 58–71. [Google Scholar] [CrossRef] [Green Version]
- Gumá, M.; Angulo, A.; Vilches, C.; Gómez-Lozano, N.; Malats, N.; López-Botet, M. Imprint of human cytomegalovirus infection on the NK cell receptor repertoire. Blood 2004, 104, 3664–3671. [Google Scholar] [CrossRef] [Green Version]
- Lopez-Vergès, S.; Milush, J.M.; Schwartz, B.S.; Pando, M.J.; Jarjoura, J.; York, V.A.; Houchins, J.P.; Miller, S.; Kang, S.M.; Norris, P.J.; et al. Expansion of a unique CD57⁺NKG2Chi natural killer cell subset during acute human cytomegalovirus infection. Proc. Natl Acad. Sci. USA 2011, 108, 14725–14732. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Foley, B.; Cooley, S.; Verneris, M.R.; Pitt, M.; Curtsinger, J.; Luo, X.; Lopez-Vergès, S.; Lanier, L.L.; Weisdorf, D.; Miller, J.S. Cytomegalovirus reactivation after allogeneic transplantation promotes a lasting increase in educated NKG2C+ natural killer cells with potent function. Blood 2012, 119, 2665–2674. [Google Scholar] [CrossRef]
- Béziat, V.; Liu, L.L.; Malmberg, J.A.; Ivarsson, M.A.; Sohlberg, E.; Björklund, A.T.; Retière, C.; Sverremark-Ekström, E.; Traherne, J.; Ljungman, P.; et al. NK cell responses to cytomegalovirus infection lead to stable imprints in the human KIR repertoire and involve activating KIRs. Blood 2013, 121, 2678–2688. [Google Scholar] [CrossRef]
- Schlums, H.; Cichocki, F.; Tesi, B.; Theorell, J.; Beziat, V.; Holmes, T.D.; Han, H.; Chiang, S.C.; Foley, B.; Mattsson, K.; et al. Cytomegalovirus infection drives adaptive epigenetic diversification of NK cells with altered signaling and effector function. Immunity 2015, 42, 443–456. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Smith, S.L.; Kennedy, P.R.; Stacey, K.B.; Worboys, J.D.; Yarwood, A.; Seo, S.; Solloa, E.H.; Mistretta, B.; Chatterjee, S.S.; Gunaratne, P.; et al. Diversity of peripheral blood human NK cells identified by single-cell RNA sequencing. Blood Adv. 2020, 4, 1388–1406. [Google Scholar] [CrossRef] [PubMed]
- Liu, L.L.; Landskron, J.; Ask, E.H.; Enqvist, M.; Sohlberg, E.; Traherne, J.A.; Hammer, Q.; Goodridge, J.P.; Larsson, S.; Jayaraman, J.; et al. Critical role of CD2 Co-stimulation in adaptive natural killer cell responses revealed in NKG2C-deficient humans. Cell Rep. 2016, 15, 1088–1099. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hammer, Q.; Rückert, T.; Borst, E.M.; Dunst, J.; Haubner, A.; Durek, P.; Heinrich, F.; Gasparoni, G.; Babic, M.; Tomic, A.; et al. Peptide-specific recognition of human cytomegalovirus strains controls adaptive natural killer cells. Nat. Immunol. 2018, 19, 453–463. [Google Scholar] [CrossRef] [PubMed]
- Foley, B.; Cooley, S.; Verneris, M.R.; Curtsinger, J.; Luo, X.; Waller, E.K.; Anasetti, C.; Weisdorf, D.; Miller, J.S. Human cytomegalovirus (CMV)-induced memory-like NKG2C(+) NK cells are transplantable and expand in vivo in response to recipient CMV antigen. J. Immunol. 2012, 189, 5082–5088. [Google Scholar] [CrossRef] [Green Version]
- Kuijpers, T.W.; Baars, P.A.; Dantin, C.; van den Burg, M.; van Lier, R.A.; Roosnek, E. Human NK cells can control CMV infection in the absence of T cells. Blood 2008, 112, 914–915. [Google Scholar] [CrossRef] [Green Version]
- Gumá, M.; Budt, M.; Sáez, A.; Brckalo, T.; Hengel, H.; Angulo, A.; López-Botet, M. Expansion of CD94/NKG2C+ NK cells in response to human cytomegalovirus-infected fibroblasts. Blood 2006, 107, 3624–3631. [Google Scholar] [CrossRef] [Green Version]
- Rölle, A.; Pollmann, J.; Ewen, E.M.; Le, V.T.; Halenius, A.; Hengel, H.; Cerwenka, A. IL-12-producing monocytes and HLA-E control HCMV-driven NKG2C+ NK cell expansion. J. Clin. Investig. 2014, 124, 5305–5316. [Google Scholar] [CrossRef] [Green Version]
- Della Chiesa, M.; Falco, M.; Bertaina, A.; Muccio, L.; Alicata, C.; Frassoni, F.; Locatelli, F.; Moretta, L.; Moretta, A. Human cytomegalovirus infection promotes rapid maturation of NK cells expressing activating killer Ig-like receptor in patients transplanted with NKG2C-/-umbilical cord blood. J. Immunol. 2014, 192, 1471–1479. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jackson, S.E.; Mason, G.M.; Wills, M.R. Human cytomegalovirus immunity and immune evasion. Virus Res. 2011, 157, 151–160. [Google Scholar] [CrossRef]
- Quinnan, G.V., Jr.; Kirmani, N.; Rook, A.H.; Manischewitz, J.F.; Jackson, L.; Moreschi, G.; Santos, G.W.; Saral, R.; Burns, W.H. Cytotoxic t cells in cytomegalovirus infection: HLA-restricted T-lymphocyte and non-T-lymphocyte cytotoxic responses correlate with recovery from cytomegalovirus infection in bone-marrow-transplant recipients. N. Engl. J. Med. 1982, 307, 7–13. [Google Scholar] [CrossRef]
- Biron, C.A.; Byron, K.S.; Sullivan, J.L. Severe herpesvirus infections in an adolescent without natural killer cells. N. Engl. J. Med. 1989, 320, 1731–1735. [Google Scholar] [CrossRef]
- Cottineau, J.; Kottemann, M.C.; Lach, F.P.; Kang, Y.H.; Vély, F.; Deenick, E.K.; Lazarov, T.; Gineau, L.; Wang, Y.; Farina, A.; et al. Inherited GINS1 deficiency underlies growth retardation along with neutropenia and NK cell deficiency. J. Clin. Investig. 2017, 127, 1991–2006. [Google Scholar] [CrossRef] [PubMed]
- Chen, K.C.; Stanton, R.J.; Banat, J.J.; Wills, M.R. Leukocyte immunoglobulin-like receptor 1-expressing human natural killer cell subsets differentially recognize isolates of human cytomegalovirus through the viral major histocompatibility complex class I homolog ul18. J. Virol. 2016, 90, 3123–3137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Berry, R.; Watson, G.M.; Jonjic, S.; Degli-Esposti, M.A.; Rossjohn, J. Modulation of innate and adaptive immunity by cytomegaloviruses. Nat. Rev. Immunol. 2020, 20, 113–127. [Google Scholar] [CrossRef]
- Kim, Y.; Park, B.; Cho, S.; Shin, J.; Cho, K.; Jun, Y.; Ahn, K. Human cytomegalovirus UL18 utilizes US6 for evading the NK and T-cell responses. PLoS Pathog. 2008, 4, e1000123. [Google Scholar] [CrossRef] [Green Version]
- Wang, E.C.; McSharry, B.; Retiere, C.; Tomasec, P.; Williams, S.; Borysiewicz, L.K.; Braud, V.M.; Wilkinson, G.W. UL40-mediated NK evasion during productive infection with human cytomegalovirus. Proc. Natl. Acad. Sci. USA 2002, 99, 7570–7575. [Google Scholar] [CrossRef] [Green Version]
- Cosman, D.; Müllberg, J.; Sutherland, C.L.; Chin, W.; Armitage, R.; Fanslow, W.; Kubin, M.; Chalupny, N.J. ULBPs, novel MHC class I-related molecules, bind to CMV glycoprotein UL16 and stimulate NK cytotoxicity through the NKG2D receptor. Immunity 2001, 14, 123–133. [Google Scholar] [CrossRef]
- De Pelsmaeker, S.; Romero, N.; Vitale, M.; Favoreel, H.W. Herpesvirus evasion of natural killer cells. J. Virol. 2018, 92, e02105–e02117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rölle, A.; Mousavi-Jazi, M.; Eriksson, M.; Odeberg, J.; Soderberg-Naucler, C.; Cosman, D.; Karre, K.; Cerboni, C. Effects of human cytomegalovirus infection on ligands for the activating NKG2D receptor of NK cells: Up-regulation of UL16-binding protein (ULBP)1 and ULBP2 is counter- acted by the viral UL16 protein. J. Immunol. 2003, 171, 902–908. [Google Scholar] [CrossRef] [PubMed]
- Fielding, C.A.; Aicheler, R.; Stanton, R.J.; Wang, E.C.; Han, S.; Seirafian, S.; Davies, J.; McSharry, B.P.; Weekes, M.P.; Antrobus, P.R.; et al. Two novel human cytomegalovirus NK cell evasion functions target MICA for lysosomal degradation. PLoS Pathog. 2014, 10, e1004058. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rölle, A.; Halenius, A.; Ewen, E.M.; Cerwenka, A.; Hengel, H.; Momburg, F. CD2-CD58 interactions are pivotal for the activation and function of adaptive natural killer cells in human cytomegalovirus infection. Eur. J. Immunol. 2016, 46, 2420–2425. [Google Scholar] [CrossRef] [PubMed]
- Bryceson, Y.T.; March, M.E.; Ljunggren, H.G.; Long, E.O. Synergy among receptors on resting NK cells for the activation of natural cytotoxicity and cytokine secretion. Blood 2006, 107, 159–166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Long, E.O.; Kim, H.S.; Liu, D.; Peterson, M.E.; Rajagopalan, S. Controlling natural killer cell responses: Integration of signals for activation and inhibition. Annu. Rev. Immunol. 2013, 31, 227–258. [Google Scholar] [CrossRef] [Green Version]
- Arnon, T.I.; Achdout, H.; Levi, O.; Markel, G.; Saleh, N.; Katz, G.; Gazit, R.; Gonen-Gross, T.; Hanna, J.; Nahari, E.; et al. Inhibition of the NKp30 activating receptor by pp65 of human cytomegalovirus. Nat. Immunol. 2005, 6, 515–523. [Google Scholar] [CrossRef]
- Seidel, E.; Le, V.; Bar-On, Y.; Tsukerman, P.; Enk, J.; Yamin, R.; Stein, N.; Schmiedel, D.; Oiknine Djian, E.; Weisblum, Y.; et al. Dynamic Co-evolution of host and pathogen: HCMV downregulates the prevalent allele MICA∗008 to escape elimination by NK cells. Cell Rep. 2015, 10, 968–982. [Google Scholar] [CrossRef] [Green Version]
- Elek, S.D.; Stern, H. Development of a vaccine against mental retardation caused by cytomegalovirus infection in utero. Lancet 1974, 1, 1–5. [Google Scholar] [CrossRef]
- Plotkin, S.A.; Smiley, M.L.; Friedman, H.M.; Starr, S.E.; Fleisher, G.R.; Wlodaver, C.; Dafoe, D.C.; Friedman, A.D.; Grossman, R.A.; Barker, C.F. Towne-vaccine-induced prevention of cytomegalovirus disease after renal transplants. Lancet 1984, 1, 528–530. [Google Scholar] [CrossRef]
- Plotkin, S.A.; Starr, S.E.; Friedman, H.M.; Gönczöl, E.; Weibel, R.E. Protective effects of Towne cytomegalovirus vaccine against low-passage cytomegalovirus administered as a challenge. J. Infect. Dis. 1989, 159, 860–865. [Google Scholar] [CrossRef] [PubMed]
- Isaacson, M.K.; Compton, T. Human cytomegalovirus glycoprotein B is required for virus entry and cell-to-cell spread but not for virion attachment, assembly, or egress. J. Virol. 2009, 83, 3891–3903. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bernstein, D.I.; Munoz, F.M.; Callahan, S.T.; Rupp, R.; Wootton, S.H.; Edwards, K.M.; Turley, C.B.; Stanberry, L.R.; Patel, S.M.; Mcneal, M.M.; et al. Safety and efficacy of a cytomegalovirus glycoprotein B (gB) vaccine in adolescent girls: A randomized clinical trial. Vaccine 2016, 34, 313–319. [Google Scholar] [CrossRef] [Green Version]
- Pass, R.F.; Duliegè, A.M.; Boppana, S.; Sekulovich, R.; Percell, S.; Britt, W.; Burke, R.L. A subunit cytomegalovirus vaccine based on recombinant envelope glycoprotein B and a new adjuvant. J. Infect. Dis. 1999, 180, 970–975. [Google Scholar] [CrossRef] [Green Version]
- Griffiths, P.D.; Stanton, A.; McCarrell, E.; Smith, C.; Osman, M.; Harber, M.; Davenport, A.; Jones, G.; Wheeler, D.C.; O’Beirne, J.; et al. Cytomegalovirus glycoprotein-B vaccine with MF59 adjuvant in transplant recipients: A phase 2 randomised placebo-controlled trial. Lancet 2011, 377, 1256–1263. [Google Scholar] [CrossRef] [Green Version]
- Schleiss, M.R.; Choi, K.Y.; Anderson, J.; Mash, J.G.; Wettendorff, M.; Mossman, S.; Van Damme, M. Glycoprotein B (gB) vaccines adjuvanted with AS01 or AS02 protect female guinea pigs against cytomegalovirus (CMV) viremia and offspring mortality in a CMV-challenge model. Vaccine 2014, 32, 2756–2762. [Google Scholar] [CrossRef] [Green Version]
- GSK. A Follow-Up Study to Evaluate the Long-Term Persistence of GSK Biologicals’ Candidate CMV Vaccine Administered to Male Adults. 2012. Available online: https://www.gsk-studyregister.com/en/trial-details/?id=115429 (accessed on 14 June 2020).
- Barel, M.T.; Hassink, G.C.; van Voorden, S.; Wiertz, E.J. Human cytomegalovirus-encoded US2 and US11 target unassembled MHC class I heavy chains for degradation. Mol. Immunol. 2006, 43, 1258–1266. [Google Scholar] [CrossRef]
- Wiertz, E.J.; Tortorella, D.; Bogyo, M.; Yu, J.; Mothes, W.; Jones, T.R.; Rapoport, T.A.; Ploegh, H.L. Sec61-mediated transfer of a membrane protein from the endoplasmic reticulum to the proteasome for destruction. Nature 1996, 384, 432–438. [Google Scholar] [CrossRef]
- Jones, T.R.; Hanson, L.K.; Sun, L.; Slater, J.S.; Stenberg, R.M.; Campbell, A.E. Multiple independent loci within the human cytomegalovirus unique short region down-regulate expression of major histocompatibility complex class I heavy chains. J. Virol. 1995, 69, 4830–4841. [Google Scholar] [CrossRef] [Green Version]
- Jones, T.R.; Wiertz, E.J.; Sun, L.; Fish, K.N.; Nelson, J.A.; Ploegh, H.L. Human cytomegalovirus US3 impairs transport and maturation of major histocompatibility complex class I heavy chains. Proc. Natl. Acad. Sci. USA 1996, 93, 11327–11333. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lehner, P.J.; Karttunen, J.T.; Wilkinson, G.W.; Cresswell, P. The human cytomegalovirus US6 glycoprotein inhibits transporter associated with antigen processing-dependent peptide translocation. Proc. Natl. Acad. Sci. USA 1997, 94, 6904–6909. [Google Scholar] [CrossRef] [Green Version]
- Park, B.; Spooner, E.; Houser, B.L.; Strominger, J.L.; Ploegh, H.L. The HCMV membrane glycoprotein US10 selectively targets HLA-G for degradation. J. Exp. Med. 2010, 207, 2033–2041. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ulbrecht, M.; Martinozzi, S.; Grzeschik, M.; Hengel, H.; Ellwart, J.W.; Pla, M.; Weiss, E.H. Cutting edge: The human cytomegalovirus UL40 gene product contains a ligand for HLA-E and prevents NK cell-mediated lysis. J. Immunol. 2000, 164, 5019–5022. [Google Scholar] [CrossRef] [Green Version]
- Nachmani, D.; Zimmermann, A.; Oiknine Djian, E.; Weisblum, Y.; Livneh, Y.; Khanh Le, V.T.; Galun, E.; Horejsi, V.; Isakov, O.; Shomron, N.; et al. MicroRNA editing facilitates immune elimination of HCMV infected cells. PLoS Pathog. 2014, 10, e1003963. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Charpak-Amikam, Y.; Kubsch, T.; Seidel, E.; Oiknine-Djian, E.; Cavaletto, N.; Yamin, R.; Schmiedel, D.; Wolf, D.; Gribaudo, G.; Messerle, M.; et al. Human cytomegalovirus escapes immune recognition by NK cells through the downregulation of B7-H6 by the viral genes US18 and US20. Sci. Rep. 2017, 7, 8661. [Google Scholar] [CrossRef] [Green Version]
- Welte, S.A.; Sinzger, C.; Lutz, S.Z.; Singh-Jasuja, H.; Sampaio, K.L.; Eknigk, U.; Rammensee, H.G.; Steinle, A. Selective intracellular retention of virally induced NKG2D ligands by the human cytomegalovirus UL16 glycoprotein. Eur. J. Immunol. 2003, 33, 194–203. [Google Scholar] [CrossRef]
- Ashiru, O.; Bennett, N.J.; Boyle, L.H.; Thomas, M.; Trowsdale, J.; Wills, M.R. NKG2D ligand MICA is retained in the cis-Golgi apparatus by human cytomegalovirus protein UL142. J. Virol. 2009, 83, 12345–12354. [Google Scholar] [CrossRef] [Green Version]
- Dassa, L.; Seidel, E.; Oiknine-Djian, E.; Yamin, R.; Wolf, D.G.; Le-Trilling, V.; Mandelboim, O. The human cytomegalovirus protein UL148A downregulates the NK cell-activating ligand MICA to avoid NK cell attack. J. Virol. 2018, 92, e00162–e00218. [Google Scholar] [CrossRef] [Green Version]
- Stern-Ginossar, N.; Elefant, N.; Zimmermann, A.; Wolf, D.G.; Saleh, N.; Biton, M.; Horwitz, E.; Prokocimer, Z.; Prichard, M.; Hahn, G.; et al. Host immune system gene targeting by a viral miRNA. Science 2007, 317, 376–381. [Google Scholar] [CrossRef] [Green Version]
- Eagle, R.A.; Traherne, J.A.; Hair, J.R.; Jafferji, I.; Trowsdale, J. ULBP6/RAET1L is an additional human NKG2D ligand. Eur. J. Immunol. 2009, 39, 3207–3216. [Google Scholar] [CrossRef] [PubMed]
- Bennett, N.J.; Ashiru, O.; Morgan, F.J.; Pang, Y.; Okecha, G.; Eagle, R.A.; Trowsdale, J.; Sissons, J.G.; Wills, M.R. Intracellular sequestration of the NKG2D ligand ULBP3 by human cytomegalovirus. J. Immunol. 2010, 185, 1093–1102. [Google Scholar] [CrossRef] [PubMed]
- Fielding, C.A.; Weekes, M.P.; Nobre, L.V.; Ruckova, E.; Wilkie, G.S.; Paulo, J.A.; Chang, C.; Suárez, N.M.; Davies, J.A.; Antrobus, R.; et al. Control of immune ligands by members of a cytomegalovirus gene expansion suppresses natural killer cell activation. eLife 2017, 6, e22206. [Google Scholar] [CrossRef] [PubMed]
- Prod’homme, V.; Sugrue, D.M.; Stanton, R.J.; Nomoto, A.; Davies, J.; Rickards, C.R.; Cochrane, D.; Moore, M.; Wilkinson, G.; Tomasec, P. Human cytomegalovirus UL141 promotes efficient downregulation of the natural killer cell activating ligand CD112. J. Gen. Virol. 2010, 91, 2034–2039. [Google Scholar] [CrossRef]
- Tomasec, P.; Wang, E.C.; Davison, A.J.; Vojtesek, B.; Armstrong, M.; Griffin, C.; McSharry, B.P.; Morris, R.J.; Llewellyn-Lacey, S.; Rickards, C.; et al. Downregulation of natural killer cell-activating ligand CD155 by human cytomegalovirus UL141. Nat. Immunol. 2005, 6, 181–188. [Google Scholar] [CrossRef] [Green Version]
- Wang, E.; Pjechova, M.; Nightingale, K.; Vlahava, V.M.; Patel, M.; Ruckova, E.; Forbes, S.K.; Nobre, L.; Antrobus, R.; Roberts, D.; et al. Suppression of costimulation by human cytomegalovirus promotes evasion of cellular immune defenses. Proc. Natl. Acad. Sci. USA 2018, 115, 4998–5003. [Google Scholar] [CrossRef] [Green Version]
- Corrales-Aguilar, E.; Trilling, M.; Hunold, K.; Fiedler, M.; Le, V.T.; Reinhard, H.; Ehrhardt, K.; Mercé-Maldonado, E.; Aliyev, E.; Zimmermann, A.; et al. Human cytomegalovirus Fcγ binding proteins gp34 and gp68 antagonize Fcγ receptors I, II and III. PLoS Pathog. 2014, 10, e1004131. [Google Scholar] [CrossRef] [Green Version]
- Smith, W.; Tomasec, P.; Aicheler, R.; Loewendorf, A.; Nemčovičová, I.; Wang, E.C.; Stanton, R.J.; Macauley, M.; Norris, P.; Willen, L.; et al. Human cytomegalovirus glycoprotein UL141 targets the TRAIL death receptors to thwart host innate antiviral defenses. Cell Host Microbe 2013, 13, 324–335. [Google Scholar] [CrossRef] [Green Version]
- Zischke, J.; Mamareli, P.; Pokoyski, C.; Gabaev, I.; Buyny, S.; Jacobs, R.; Falk, C.S.; Lochner, M.; Sparwasser, T.; Schulz, T.F.; et al. The human cytomegalovirus glycoprotein pUL11 acts via CD45 to induce T cell IL-10 secretion. PLoS Pathog. 2017, 13, e1006454. [Google Scholar] [CrossRef] [Green Version]
- Prod’homme, V.; Griffin, C.; Aicheler, R.J.; Wang, E.C.; McSharry, B.P.; Rickards, C.R.; Stanton, R.J.; Borysiewicz, L.K.; López-Botet, M.; Wilkinson, G.W.; et al. The human cytomegalovirus MHC class I homolog UL18 inhibits LIR-1+ but activates LIR-1-NK cells. J. Immunol. 2007, 178, 4473–4481. [Google Scholar] [CrossRef] [Green Version]
- Stanton, R.J.; Prod’homme, V.; Purbhoo, M.A.; Moore, M.; Aicheler, R.J.; Heinzmann, M.; Bailer, S.M.; Haas, J.; Antrobus, R.; Weekes, M.P.; et al. HCMV pUL135 remodels the actin cytoskeleton to impair immune recognition of infected cells. Cell Host Microbe 2014, 16, 201–214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hahn, G.; Revello, M.G.; Patrone, M.; Percivalle, E.; Campanini, G.; Sarasini, A.; Wagner, M.; Gallina, A.; Milanesi, G.; Koszinowski, U.; et al. Human cytomegalovirus UL131-128 genes are indispensable for virus growth in endothelial cells and virus transfer to leukocytes. J. Virol. 2004, 78, 10023–10033. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kabanova, A.; Perez, L.; Lilleri, D.; Marcandalli, J.; Agatic, G.; Becattini, S.; Preite, S.; Fuschillo, D.; Percivalle, E.; Sallusto, F.; et al. Antibody-driven design of a human cytomegalovirus gHgLpUL128L subunit vaccine that selectively elicits potent neutralizing antibodies. Proc. Natl. Acad. Sci. USA 2014, 111, 17965–17970. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Murrell, I.; Bedford, C.; Ladell, K.; Miners, K.L.; Price, D.A.; Tomasec, P.; Wilkinson, G.W.G.; Stanton, R.J. The pentameric complex drives immunologically covert cell-cell transmission of wild-type human cytomegalovirus. Proc. Natl. Acad. Sci. USA 2017, 114, 6104–6109. [Google Scholar] [CrossRef] [Green Version]
- Lilleri, D.; Kabanova, A.; Revello, M.G.; Percivalle, E.; Sarasini, A.; Genini, E.; Sallusto, F.; Lanzavecchia, A.; Corti, D.; Gerna, G. Fetal human cytomegalovirus transmission correlates with delayed maternal antibodies to gH/gL/pUL128-130-131 complex during primary infection. PLoS ONE 2013, 8, e5986. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ishida, J.H.; Patel, A.; Mehta, A.K.; Gatault, P.; McBride, J.M.; Burgess, T.; Derby, M.A.; Snydman, D.R.; Emu, B.; Feierbach, B.; et al. Phase 2 randomized, double-blind, placebo-controlled trial of RG7667, a combination monoclonal antibody, for prevention of cytomegalovirus infection in high-risk kidney transplant recipients. Antimicrob. Agents Chemother. 2017, 61, e01794–e01816. [Google Scholar] [CrossRef] [Green Version]
- Vanarsdall, A.L.; Chin, A.L.; Liu, J.; Jardetzky, T.S.; Mudd, J.O.; Orloff, S.L.; Streblow, D.; Mussi-Pinhata, M.M.; Yamamoto, A.Y.; Duarte, G.; et al. HCMV trimer-and pentamer-specific antibodies synergize for virus neutralization but do not correlate with congenital transmission. Proc. Natl. Acad. Sci. USA 2019, 116, 3728–3733. [Google Scholar] [CrossRef] [Green Version]
- Plotkin, S.; Robinson, J.M.; Cunningham, G.; Iqbal, R.; Larsen, S. The complexity and cost of vaccine manufacturing—An overview. Vaccine 2017, 35, 4064–4071. [Google Scholar] [CrossRef]
- Manicklal, S.; Emery, V.C.; Lazzarotto, T.; Boppana, S.B.; Gupta, R.K. The “silent” global burden of congenital cytomegalovirus. Clin. Microbiol. Rev. 2013, 26, 86–102. [Google Scholar] [CrossRef] [Green Version]
- Azevedo, L.S.; Pierrotti, L.C.; Abdala, E.; Costa, S.F.; Strabelli, T.M.; Campos, S.V.; Ramos, J.F.; Latif, A.Z.; Litvinov, N.; Maluf, N.Z.; et al. Cytomegalovirus infection in transplant recipients. clinics (Sao Paulo) 2015, 70, 515–523. [Google Scholar] [CrossRef]
- Wills, M.R.; Carmichael, A.J.; Mynard, K.; Jin, X.; Weekes, M.P.; Plachter, B.; Sissons, J.G. The human cytotoxic T-lymphocyte (CTL) response to cytomegalovirus is dominated by structural protein pp65: Frequency, specificity, and T-cell receptor usage of pp65-specific CTL. J. Virol. 1996, 70, 7569–7579. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sylwester, A.W.; Mitchell, B.L.; Edgar, J.B.; Taormina, C.; Pelte, C.; Ruchti, F.; Sleath, P.R.; Grabstein, K.H.; Hosken, N.A.; Kern, F.; et al. Broadly targeted human cytomegalovirus-specific CD4+ and CD8+ T cells dominate the memory compartments of exposed subjects. J. Exp. Med. 2005, 202, 673–685. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sacre, K.; Carcelain, G.; Cassoux, N.; Fillet, A.M.; Costagliola, D.; Vittecoq, D.; Salmon, D.; Amoura, Z.; Katlama, C.; Autran, B. Repertoire, diversity, and differentiation of specific CD8 T cells are associated with immune protection against human cytomegalovirus disease. J. Exp. Med. 2005, 201, 1999–2010. [Google Scholar] [CrossRef] [Green Version]
- Pötzsch, S.; Spindler, N.; Wiegers, A.K.; Fisch, T.; Rücker, P.; Sticht, H.; Grieb, N.; Baroti, T.; Weisel, F.; Stamminger, T.; et al. B cell repertoire analysis identifies new antigenic domains on glycoprotein b of human cytomegalovirus which are target of neutralizing antibodies. PLoS Pathog. 2011, 7, e1002172. [Google Scholar]
- Schrader, J.W.; McLean, G.R. Location, location, timing: Analysis of cytomegalovirus epitopes for neutralizing antibodies. Immunol. Lett. 2007, 112, 58–60. [Google Scholar] [CrossRef]
- Bialas, K.M.; Westreich, D.; Cisneros de la Rosa, E.; Nelson, C.S.; Kauvar, L.M.; Fu, T.M.; Permar, S.R. Maternal antibody responses and nonprimary congenital cytomegalovirus infection of HIV-1-exposed infants. J. Infect. Dis. 2016, 214, 1916–1923. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Baraniak, I.; Kropff, B.; McLean, G.R.; Pichon, S.; Piras-Douce, F.; Milne, R.S.B.; Smith, C.; Mach, M.; Griffiths, P.D.; Reeves, M.B. Epitope-specific humoral responses to human cytomegalovirus glycoprotein-B vaccine with MF59: Anti-AD2 levels correlate with protection from viremia. J. Infect. Dis. 2018, 217, 1907–1917. [Google Scholar] [CrossRef]
- Liu, Y.; Freed, D.C.; Li, L.; Tang, A.; Li, F.; Murray, E.M.; Adler, S.P.; McVoy, M.A.; Rupp, R.E.; Barrett, D.; et al. A replication-defective human cytomegalovirus vaccine elicits humoral immune responses analogous to those with natural infection. J. Virol. 2019, 93, e00747–e00819. [Google Scholar] [CrossRef]
- John, S.; Yuzhakov, O.; Woods, A.; Deterling, J.; Hassett, K.; Shaw, C.A.; Ciaramella, G. Multi-antigenic human cytomegalovirus mRNA vaccines that elicit potent humoral and cell-mediated immunity. Vaccine 2018, 36, 1689–1699. [Google Scholar] [CrossRef]
- Horowitz, A.; Behrens, R.H.; Okell, L.; Fooks, A.R.; Riley, E.M. NK cells as effectors of acquired immune responses: Effector CD4+ T cell-dependent activation of NK cells following vaccination. J. Immunol. 2010, 185, 2808–2818. [Google Scholar] [CrossRef] [Green Version]
- Darboe, A.; Danso, E.; Clarke, E.; Umesi, A.; Touray, E.; Wegmuller, R.; Moore, S.E.; Riley, E.M.; Goodier, M.R. Enhancement of cytokine-driven NK cell IFN-γ production after vaccination of HCMV infected Africans. Eur. J. Immunol. 2017, 47, 1040–1050. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wagstaffe, H.R.; Pickering, H.; Houghton, J.; Mooney, J.P.; Wolf, A.S.; Prevatt, N.; Behrens, R.H.; Holland, M.J.; Riley, E.M.; Goodier, M.R. Influenza vaccination primes human myeloid cell cytokine secretion and NK cell function. J. Immunol. 2019, 203, 1609–1618. [Google Scholar] [CrossRef] [PubMed]
- Farsakoglu, Y.; Palomino-Segura, M.; Latino, I.; Zanaga, S.; Chatziandreou, N.; Pizzagalli, D.U.; Rinaldi, A.; Bolis, M.; Sallusto, F.; Stein, J.V.; et al. Influenza vaccination induces NK-cell-mediated type-II IFN response that regulates humoral immunity in an IL-6-dependent manner. Cell Rep. 2019, 26, 2307–2315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pejoski, D.; de Rham, C.; Martinez-Murillo, P.; Santoro, F.; Auderset, F.; Medaglini, D.; Pozzi, G.; Vono, M.; Lambert, P.H.; Huttner, A.; et al. Rapid dose-dependent Natural Killer (NK) cell modulation and cytokine responses following human rVSV-ZEBOV Ebolavirus vaccination. NPJ Vaccines 2020, 5, 32. [Google Scholar] [CrossRef] [Green Version]
- Rechtien, A.; Richert, L.; Lorenzo, H.; Martrus, G.; Hejblum, B.; Dahlke, C.; Kasonta, R.; Zinser, M.; Stubbe, H.; Matschl, U.; et al. Systems vaccinology identifies an early innate immune signature as a correlate of antibody responses to the Ebola vaccine rVSV-ZEBOV. Version 2. Cell Rep. 2017, 20, 2251–2261. [Google Scholar] [CrossRef] [Green Version]
- Michaelsen, T.E.; Aase, A.; Norderhaug, L.; Sandlie, I. Antibody dependent cell-mediated cytotoxicity induced by chimeric mouse-human IgG subclasses and IgG3 antibodies with altered hinge region. Mol. Immunol. 1992, 29, 319–326. [Google Scholar] [CrossRef]
- Yates, N.L.; Liao, H.X.; Fong, Y.; DeCamp, A.; Vandergrift, N.A.; Williams, W.T. Vaccine-induced Env V1-V2 IgG3 correlates with lower HIV-1 infection risk and declines soon after vaccination. Sci. Transl. Med. 2014, 6, 228ra39. [Google Scholar] [CrossRef] [Green Version]
- Wagstaffe, H.R.; Clutterbuck, E.A.; Bockstal, V.; Stoop, J.N.; Luhn, K.; Douoguih, M.; Shukarev, G.; Snape, M.D.; Pollard, A.J.; Riley, E.M.; et al. Antibody-dependent natural killer cell activation after Ebola vaccination. J. Infect. Dis. 2019, 10, jiz657. [Google Scholar] [CrossRef] [Green Version]
- Heuser, C.; Ganser, M.; Hombach, A.; Brand, H.; Denton, G.; Hanisch, F.G.; Abken, H. An anti-MUC1-antibody-interleukin-2 fusion protein that activates resting NK cells to lysis of MUC1-positive tumour cells. Br. J. Cancer 2003, 89, 1130–1139. [Google Scholar] [CrossRef] [Green Version]
- Dou, Y.; Fu, B.; Sun, R.; Li, W.; Hu, W.; Tian, Z.; Wei, H. Influenza vaccine induces intracellular immune memory of human NK cells. PLoS ONE 2015, 10, e0121258. [Google Scholar] [CrossRef] [Green Version]
- Junqueira-Kipnis, A.P.; Trentini, M.M.; Marques Neto, L.M.; Kipnis, A. Live vaccines have different NK cells and neutrophils requirements for the development of a protective immune response against tuberculosis. Front. Immunol. 2020, 11, 741. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Martins, K.A.; Steffens, J.T.; van Tongeren, S.A.; Wells, J.B.; Bergeron, A.A.; Dickson, S.P.; Dye, J.M.; Salazar, A.M.; Bavari, S. Toll-like receptor agonist augments virus-like particle-mediated protection from Ebola virus with transient immune activation. PLoS ONE 2014, 9, e89735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dérian, N.; Bellier, B.; Pham, H.P.; Tsitoura, E.; Kazazi, D.; Huret, C.; Mavromara, P.; Klatzmann, D.; Six, A. Early transcriptome signatures from immunized mouse dendritic cells predict late vaccine-induced T-cell responses. PLoS Comput. Biol. 2016, 12, e1004801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gomes, A.C.; Mohsen, M.O.; Mueller, J.E.; Leoratti, F.M.S.; Cabral-Miranda, G.; Bachmann, M.F. Early transcriptional signature in dendritic cells and the induction of protective T cell responses upon immunization with VLPs containing TLR ligands—A role for CCL2. Front. Immunol. 2019, 10, 1679. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Leigh, N.D.; Bian, G.; Ding, X.; Liu, H.; Aygun-Sunar, S.; Burdelya, L.G.; Gudkov, A.V.; Cao, X. A flagellin-derived toll-like receptor 5 agonist stimulates cytotoxic lymphocyte-mediated tumor immunity. PLoS ONE 2014, 9, e85587. [Google Scholar] [CrossRef]
- Ma, F.; Zhang, J.; Zhang, J.; Zhang, C. The TLR7 agonists imiquimod and gardiquimod improve DC-based immunotherapy for melanoma in mice. Cell. Mol. Immunol. 2010, 7, 381–388. [Google Scholar] [CrossRef] [Green Version]
- Goodier, M.R.; Jonjić, S.; Riley, E.M.; Juranić Lisnić, V. CMV and natural killer cells: Shaping the response to vaccination. Eur. J. Immunol. 2018, 48, 50–65. [Google Scholar] [CrossRef]
Receptor Type | Receptor | Ligand | HCMV Immune Evasin | Evasion Mechanism |
---|---|---|---|---|
Activating | Activating KIR | MHC-Ia | US2, US3, US6, US11 | Reduction in MHC-I surface expression; US2, US11- proteasomal degradation [79,80,81]; US3- ER retention of MHC-I [82]; US6- blocks TAP mediated peptide translocation [83]. |
- | HLA-G | US2, US10 | Reduction in HLA-G surface expression; US2- proteasomal degradation [79,80,81]; US10- degradation of HLA-G [84]. | |
CD94-NKG2C/E/H | HLA-E | US6, UL40, miR376a | UL40 encodes a TAP-independent signal peptide that stabilises HLA-E surface expression [85]. miR376a blocks HLA-E surface expression [86]. US6 blocks TAP dependent peptide translocation [83]. | |
NKp30 | B7-H6 | US18, US20, pp65 (UL83) | US18, US20, Lysosomal degradation of activation receptor ligands [87]. | |
- | BAT-3 | pp65 (UL83) | pp65-binds NKp30 causing CD3ζ dissociation [68]. | |
NKG2D | MICA/B | UL16, UL142, UL148A, US9, US18, US20, miR-UL112 | UL16, UL142- intracellular retention [88,89]; UL148A, US18, US20- lysosomal degradation [87,90]; US9- proteasomal degradation (MICA*008) [69], miR-UL112- downregulation of MICB expression [91]. | |
- | ULBPs | UL16, UL142, US12, US13, US20 | UL16, UL142- intracellular retention [88,92,93]; US20- lysosomal degradation [87]; US12, US13- downregulation of ULBPs [94]. | |
DNAM-1 | CD112 | UL141 (requires US2) | ER retention [95]. | |
- | CD155 | UL141 | ER retention [96]. | |
TACTILE | CD111 | - | - | |
- | CD155 | UL141 | ER retention [96]. | |
2B4 | CD48 | - | - | |
CD2 | LFA-3 | UL148 | Lysosomal degradation [97]. | |
CD16 | Fc of IgG | RL11-13, UL119-UL118 | Fcγ binding and inhibition of Fc receptor signalling, inhibition of ADCC [98]. | |
TRAIL | TRAIL-R1/-R2 | UL141 | ER retention [99]. | |
CD45 | pUL11 | pUL11 | Inhibition of CD45 mediated signalling through direct binding of pUL11 [100]. | |
Inhibitory | Inhibitory KIR | MHC-I polymorphisms | - | - |
LIR-1 | MHC-Ia | UL18 | MHC-I homolog [101]. | |
CD94-NKG2A/B | HLA-E | UL40 | UL40 encodes a TAP-independent signal peptide that stabilises HLA-E surface expression [85]. | |
TACTILE | CD111 | - | - | |
- | CD155 | UL141 | ER retention [96]. | |
TIGIT/ PVRIG | CD112 | UL141 (requires US2) | ER retention [95]. | |
- | CD155 | UL141 | ER retention [96]. | |
Cadherins | KLRG1 | - | - | |
LLT1 | CD161 (NKR-P1A | - | - | |
- | WAVE2/F-actin | UL135 | Suppression of immune synapse formation [102]. |
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Forrest, C.; Gomes, A.; Reeves, M.; Male, V. NK Cell Memory to Cytomegalovirus: Implications for Vaccine Development. Vaccines 2020, 8, 394. https://doi.org/10.3390/vaccines8030394
Forrest C, Gomes A, Reeves M, Male V. NK Cell Memory to Cytomegalovirus: Implications for Vaccine Development. Vaccines. 2020; 8(3):394. https://doi.org/10.3390/vaccines8030394
Chicago/Turabian StyleForrest, Calum, Ariane Gomes, Matthew Reeves, and Victoria Male. 2020. "NK Cell Memory to Cytomegalovirus: Implications for Vaccine Development" Vaccines 8, no. 3: 394. https://doi.org/10.3390/vaccines8030394
APA StyleForrest, C., Gomes, A., Reeves, M., & Male, V. (2020). NK Cell Memory to Cytomegalovirus: Implications for Vaccine Development. Vaccines, 8(3), 394. https://doi.org/10.3390/vaccines8030394