Mutual Interference between Cytomegalovirus and Reconstitution of Protective Immunity after Hematopoietic Cell Transplantation

Matthias J Reddehase, Matthias J Reddehase

Abstract

Hematopoietic cell transplantation (HCT) is a therapy option for aggressive forms of hematopoietic malignancies that are resistant to standard antitumoral therapies. Hematoablative treatment preceding HCT, however, opens a "window of opportunity" for latent Cytomegalovirus (CMV) by releasing it from immune control with the consequence of reactivation of productive viral gene expression and recurrence of infectious virus. A "window of opportunity" for the virus represents a "window of risk" for the patient. In the interim between HCT and reconstitution of antiviral immunity, primarily mediated by CD8(+) T cells, initially low amounts of reactivated virus can expand exponentially, disseminate to essentially all organs, and cause multiple organ CMV disease, with interstitial pneumonia (CMV-IP) representing the most severe clinical manifestation. Here, I will review predictions originally made in the mouse model of experimental HCT and murine CMV infection, some of which have already paved the way to translational preclinical research and promising clinical trials of a preemptive cytoimmunotherapy of human CMV disease. Specifically, the mouse model has been pivotal in providing "proof of concept" for preventing CMV disease after HCT by adoptive transfer of preselected, virus epitope-specific effector and memory CD8(+) T cells bridging the critical interim. However, CMV is not a "passive antigen" but is a pathogen that actively interferes with the reconstitution of protective immunity by infecting bone marrow (BM) stromal cells that otherwise form niches for hematopoiesis by providing the structural microenvironment and by producing hematopoietically active cytokines, the hemopoietins. Depending on the precise conditions of HCT, reduced homing of transplanted hematopoietic stem- and progenitor cells to infected BM stroma and impaired colony growth and lineage differentiation can lead to "graft failure." In consequence, uncontrolled virus spread causes morbidity and mortality. In the race between viral BM pathology and reconstitution of antiviral immunity following HCT, exogenous reconstitution of virus-specific CD8(+) T cells by adoptive cell transfer as an interventional strategy can turn the balance toward control of CMV.

Keywords: CD8 T cells; Cytomegalovirus; adoptive cell transfer; bone marrow stroma; hematopoietic cell transplantation; hemopoietins; immunotherapy; reconstitution.

Figures

Figure 1
Figure 1
Negative impact of CMV infection on survival rates after HCT demonstrated in the murine model. (Top) Sketch of the experimental model, transplanting increasing numbers of hematopoietic cells (HC) to BALB/c recipients immunocompromised by hematoablative total-body γ-irradiation with single doses increasing from 5 to 7 Gy. (Bottom) Kaplan–Meier survival curves depending on the two variables HC dose (increasing reconstitution) and dose of γ-irradiation (increasing ablation). (Green lines) HCT, no infection. (Red lines), HCT and intraplantar infection with a constant dose of purified mCMV. Green- and red-shaded panels correspond to the bone marrow histology shown in Figures 3A,B, respectively. Reproduced from Ref. (59) with permission from Caister Academic Press, Norfolk, UK.
Figure 2
Figure 2
Infection of BM stromal cells in primary cell culture. (A) Images of the typically flattened, uninfected myofibroblastic BM stroma cells co-expressing smooth muscle cell marker α-SM-actin (a1, red fluorescence) and endothelial cell marker von Willebrand factor concentrated in Weibel–Palade bodies (a2, green fluorescence). (a3) merge. (B) Cytopathic effect of mCMV infection in the BM stroma cells. Detection of intranuclear E1 protein (spotty green fluorescence) indicates that infection has proceeded to the early (E) phase of the viral replicative cycle. Bar markers: 10 μm. Images illustrate information reported in Ref. (59).
Figure 3
Figure 3
Bone marrow aplasia caused by infection. (A) Overview image of BM histology performed on day 14 after HCT, revealing a beginning repopulation of the BM of the HCT recipients with myeloid lineage colonies homing to the BM stroma in a femoral diaphysis in the absence of infection. The image corresponds to survival shown in Figure 1, green-shaded panel. (B) Images document absence of hematopoiesis in infected HCT recipients under conditions otherwise identical to those in (A), corresponding to high mortality shown in Figure 1, red-shaded panel. (b1) Overview, showing empty stromal network in a femoral epiphysis. (b2) Detail, with immunohistological staining of viral intranuclear protein IE1 (brown staining), detecting in situ infected, network-forming stromal cells. Bar markers: 25 μm, throughout. Note that the size and shape of the stromal cells in situ matches their size and shape in cell culture (compare Figure 3B, b2 with Figure 2A). Images reproduced in rearranged form from Ref. (59, 84, 86) with permissions from Caister Academic Press, Norfolk, UK and from the Journal of Virology, American Society for Microbiology.
Figure 4
Figure 4
Infection inhibits HC engraftment. (A) Sketch of the experimental design of transplanting male (XY, gene sry+) HC to female (XX, gene sry−) BALB/c recipients, resulting in transplantation chimeras with sry+ HC and sry− stromal cells. Flash symbol: total-body γ-irradiation (B) PCR quantitation of sry+ HC engraftment on day 14 after HCT without infection (Ø) or with infection (CMV). Modified from Ref. (86) with permission from the Journal of Virology, American Society for Microbiology.
Figure 5
Figure 5
Sketch of serial transplantation and re-transplantation protocols. (A) Serial transplantation of HC for quantitating BM-repopulating hematopoietic stem cells (HSC) depending on a past productive infection of primary HCT recipients. Infected primary HCT recipients become lastingly deficient in hematopoiesis reflected by reduced numbers of HSC. (B) Localization of the cause of enduring hematopoietic deficiency in primary HCT recipients with a past productive infection. Failure to cure by re-transplantation with normal HC localizes the hematopoietic deficiency to the BM stroma. This Figure illustrates the experimental regimens used to demonstrate enduring bone marrow stroma deficiency reported in Ref. (82, 83).
Figure 6
Figure 6
Model of “closed” and “open” hematopoietic niches. (Left) Low-dose HCT: in the model example, a single transplanted HSC has a 20% chance to occupy a single “open niche” (green) out of a total of five niches of which four are closed (red) due to CMV infection of the BM stromal cells that form the niche. (Right) High-dose HCT: high probability of occupying the single “open niche” out of five niches when five HSC are transferred. Note that the probability for a successful lodging event is <100% since more than one HSC may try to lodge to a “closed niche,” but the overall probability for the event that at least one out of five transplanted HSC lodges to the single “open niche” is increased. Importantly, the rate of stroma cell infection is not notably influenced by the dose of transplanted HC over the range tested. Stop symbol: hematopoiesis is not supported. Bent arrow: successful HSC lodging and self-renewal. This Figure provides a graphical new explanation for data published in Ref. (86), corresponding to Figure 4B.
Figure 7
Figure 7
Infection of HCT recipients impedes the reconstitution of CD8+ T cells. (Top) Sketch of the experimental protocol, transplanting HC from BALB/c mice to immunocompromised (flash symbol: sublethal γ-irradiation with a dose of 6 Gy) mutant mice not expressing the MHC-I molecule Ld. (Bottom) Cytofluorometric analysis of Ld expression in the recipients revealed chimerism within the spleen-derived CD8+ T-cell population that shifted toward donor-type reconstitution with increasing doses of transplanted HC (left panel, HCT). This shift is impeded by infection (right panel, HCT + infection). *100% mortality. Percentages of donor-derived CD8+ T cells are indicated. Modified from Ref. (96) with permission from the Journal of Virology, American Society for Microbiology.
Figure 8
Figure 8
CD8+ T-cell infiltration of infected lungs after HCT coincides with control of the infection in nodular inflammatory foci (NIF). HCT was performed with the high dose of 107 HC, a condition under which infected recipients survive (recall Figure 1). (A) Coincidence of the peak of CD8+ T-cell infiltration (top panel) and the onset of decline in titers of infectious virus (bottom panel). Symbols represent data from individual mice with the median values marked. Reproduced from Ref. (59, 109) with permissions from Caister Academic Press, Norfolk, UK and from the Journal of Virology, American Society for Microbiology. (B) Confinement of pulmonary infection in NIF; shown here is a NIF with peribronchiolar localization. Two-color IHC with red-staining of IE1 protein in infected lung cells and black staining of T cells. The bar marker represents 25 μm. This image has been the cover photograph of Journal of Virology, volume 74, issue no. 16 (August 2000), accompanying the publication cited here as Ref. (61). Reproduced with permission from the American Society for Microbiology.
Figure 9
Figure 9
Survival of infected HCT recipients depends on the reconstitution of CD8+ T cells. In the process of an ongoing reconstitution after HCT, T-cell subsets were depleted on days 7 and 14. Shown are Kaplan–Meier survival curves that reveal 100% mortality only when CD8+ T cells were depleted. Reproduced from Ref. (59, 61) with permissions from Caister Academic Press, Norfolk, UK and from the Journal of Virology, American Society for Microbiology.
Figure 10
Figure 10
CD8+ T cells are essential for confining the infection to nodular inflammatory foci (NIF) during reconstitution after HCT. Two-color IHC images of viral pathology in the liver correspond to the survival/mortality data shown in Figure 9. (A) Uncontrolled virus spread in liver tissue after HCT and depletion of CD8+ T cells. The red arrow representatively points to an infected hepatocyte identified by red-staining of intranuclear IE1 protein. The black arrow representatively points to an infiltrating CD4+ T cell identified by black staining of cell surface CD3ε. Note that the CD4+ T cells are randomly distributed, do not form NIF, and apparently fail to limit the virus spread. (B) Lack of liver tissue infiltration, absence of NIF, and uncontrolled spread of the infection after depletion of both T-cell subsets, which excludes participation of other CD3ε-expressing immune cells, such as γ/δ T cells and NKT cells, in the control of infection. As the infection is not controlled, cells of innate immunity (unstained) apparently are also not functional in antiviral protection after HCT. (C) Confinement of infection to NIF in the absence of T-cell depletion. (D) Confinement of infection to NIF is maintained after depletion of CD4+ T cells, indicating that formation of NIF and control of the infection by CD8+ T cells do not critically require CD4+ T cell help. Bar markers represent 25 μm. Reproduced in rearranged form from Ref. (59) with permission from Caister Academic Press, Norfolk, UK.
Figure 11
Figure 11
Synopsis of the mutual interference between Cytomegalovirus and reconstitution of protective immunity after HCT. See the body of the text for explanation. HC, hematopoietic cells; MC, mast cells; SCF, stem cell factor; G-CSF, granulocyte colony-stimulating factor; IL-6, interleukin-6; NIF, nodular inflammatory focus/foci. Red TCR symbol: CMV-specific. Blue TCR symbol: unrelated TCR specificity. Red-colored nuclei: infected cells.

References

    1. Davison AJ, Holton M, Dolan A, Dargan DJ, Gatherer D, Hayward GS. Comparative genomics of primate cytomegaloviruses. In: Reddehase MJ, editor. Cytomegaloviruses: From Molecular Pathogenesis to Intervention. (Vol. 1), Norfolk, UK: Caister Academic Press; (2013). p. 1–22.
    1. Ho M. The history of Cytomegalovirus and its diseases. Med Microbiol Immunol (2008) 197:65–73.10.1007/s00430-007-0066-x
    1. Roizman B, Sears AE. An inquiry into the mechanisms of herpes simplex virus latency. Annu Rev Microbiol (1987) 41:543–71.10.1146/annurev.mi.41.100187.002551
    1. Poole E, Sinclair J. Sleepless latency of human Cytomegalovirus. Med Microbiol Immunol (2015) 204:421–9.10.1007/s00430-015-0401-6
    1. Reeves M, Sinclair J. Epigenetic regulation of human Cytomegalovirus gene expression: impact on latency and reactivation. In: Reddehase MJ, editor. Cytomegaloviruses: From Molecular Pathogenesis to Intervention. (Vol. 1), Norfolk, UK: Caister Academic Press; (2013). p. 330–46.
    1. Slobedman B, Avdic S, Abendroth A. Transcription associated with human Cytomegalovirus latency. In: Reddehase MJ, editor. Cytomegaloviruses: From Molecular Pathogenesis to Intervention. (Vol. 1), Norfolk, UK: Caister Academic Press; (2013). p. 347–62.
    1. Cannon MJ, Grosse SD, Fowler KB. The epidemiology and public health impact of congenital Cytomegalovirus infection. In: Reddehase MJ, editor. Cytomegaloviruses: From Molecular Pathogenesis to Intervention. (Vol. 2), Norfolk, UK: Caister Academic Press; (2013). p. 26–48.
    1. Adler SP, Nigro G. Clinical Cytomegalovirus research: congenital infection. In: Reddehase MJ, editor. Cytomegaloviruses: From Molecular Pathogenesis to Intervention. (Vol. 2), Norfolk, UK: Caister Academic Press; (2013). p. 55–72.
    1. Hummel M, Abecassis MM. A model for reactivation of CMV from latency. J Clin Virol (2002) 25:S123–36.10.1016/S1386-6532(02)00088-4
    1. Kim SJ, Varghese TK, Zhang Z, Zhao LC, Thomas G, Hummel M, et al. Renal ischemia/reperfusion injury activates the enhancer domain of the human Cytomegalovirus major immediate early promoter. Am J Transplant (2005) 5:1606–13.10.1111/j.1600-6143.2005.00912.x
    1. Boppana SB, Britt WJ. Synopsis of clinical aspects of human Cytomegalovirus disease. In: Reddehase MJ, editor. Cytomegaloviruses: From Molecular Pathogenesis to Intervention. (Vol. 2), Norfolk, UK: Caister Academic Press; (2013). p. 1–25.
    1. Sissons JG, Wills MR. How understanding immunology contributes to managing CMV disease in immunosuppressed patients: now and in future. Med Microbiol Immunol (2015) 204:307–16.10.1007/s00430-015-0415-0
    1. Smith SM, Streblow DN, Caposio P, Nelson JA. Humanized mouse models of Cytomegalovirus pathogenesis and latency. In: Reddehase MJ, editor. Cytomegaloviruses: From Molecular Pathogenesis to Intervention. (Vol. 1), Norfolk, UK: Caister Academic Press; (2013). p. 417–36.
    1. Smith MG. Propagation in tissue cultures of a cytopathogenic virus from human salivary gland virus (SGV) disease. Proc Soc Exp Biol Med (1956) 92:424–30.10.3181/00379727-92-22498
    1. Reddehase MJ. Margaret Gladys Smith, mother of Cytomegalovirus: 60th anniversary of Cytomegalovirus isolation. Med Microbiol Immunol (2015) 204:239–41.10.1007/s00430-015-0416-z
    1. Ostermann E, Pawletko K, Indenbirken D, Schumacher U, Brune W. Stepwise adaptation of murine Cytomegalovirus to cells of a foreign host for identification of host range determinants. Med Microbiol Immunol (2015) 204:461–9.10.1007/s00430-015-0400-7
    1. Redwood AJ, Shellam GR, Smith LM. Molecular evolution of murine Cytomegalovirus genomes. In: Reddehase MJ, editor. Cytomegaloviruses: From Molecular Pathogenesis to Intervention. (Vol. 1), Norfolk, UK: Caister Academic Press; (2013). p. 23–37.
    1. Reddehase MJ. Antigens and immunoevasins: opponents in Cytomegalovirus immune surveillance. Nat Rev Immunol (2002) 2:831–44.10.1038/nri932
    1. Powers C, DeFilippis V, Malouli D, Früh K. Cytomegalovirus immune evasion. Curr Top Microbiol Immunol (2008) 325:333–59.10.1007/978-3-540-77349-8_19
    1. Vidal S, Krmpotic A, Pyzik M, Jonjic S. Innate immunity to Cytomegalovirus in the murine model. In: Reddehase MJ, editor. Cytomegaloviruses: From Molecular Pathogenesis to Intervention. (Vol. 2), Norfolk, UK: Caister Academic Press; (2013). p. 191–213.
    1. Mc Gregor A, McVoy MA, Schleiss MR. The guinea pig model of congenital Cytomegalovirus infection. In: Reddehase MJ, editor. Cytomegaloviruses: From Molecular Pathogenesis to Intervention. (Vol. 2), Norfolk, UK: Caister Academic Press; (2013). p. 87–117.
    1. Voigt S, Ettinger J, Streblow DN. The rat model of Cytomegalovirus infection and vascular disease. In: Reddehase MJ, editor. Cytomegaloviruses: From Molecular Pathogenesis to Intervention. (Vol. 2), Norfolk, UK: Caister Academic Press; (2013). p. 310–34.
    1. Früh K, Malouli D, Oxford KL, Barry PA. Non-human-primate models of Cytomegalovirus infection, prevention, and therapy. In: Reddehase MJ, editor. Cytomegaloviruses: From Molecular Pathogenesis to Intervention. (Vol. 2), Norfolk, UK: Caister Academic Press; (2013). p. 461–94.
    1. Benedict CA, Crozat K, Degli-Esposti M, Dalod M. Host genetic models in Cytomegalovirus immunology. In: Reddehase MJ, editor. Cytomegaloviruses: From Molecular Pathogenesis to Intervention. (Vol. 2), Norfolk, UK: Caister Academic Press; (2013). p. 310–34.
    1. Wilkinson GW, Davison AJ, Tomasec P, Fielding CA, Aicheler R, Murrell I, et al. Human Cytomegalovirus: taking the strain. Med Microbiol Immunol (2015) 204:273–84.10.1007/s00430-015-0411-4
    1. Reddehase MJ, Weiland F, Münch K, Jonjic S, Lüske A, Koszinowski UH. Interstitial murine Cytomegalovirus pneumonia after irradiation: characterization of cells that limit viral replication during established infection of the lungs. J Virol (1985) 55:264–73.
    1. Reddehase MJ, Mutter W, Koszinowski UH. In vivo application of recombinant interleukin 2 in the immunotherapy of established Cytomegalovirus infection. J Exp Med (1987) 165:650–6.10.1084/jem.165.3.650
    1. Reddehase MJ, Jonjić S, Weiland F, Mutter W, Koszinowski UH. Adoptive immunotherapy of murine Cytomegalovirus adrenalitis in the immunocompromised host: CD4-helper-independent antiviral function of CD8-positive memory T lymphocytes derived from latently infected donors. J Virol (1988) 62:1061–5.
    1. Holtappels R, Böhm V, Podlech J, Reddehase MJ. CD8 T-cell-based immunotherapy of Cytomegalovirus infection: “proof of concept” provided by the murine model. Med Microbiol Immunol (2008) 197:125–34.10.1007/s00430-008-0093-2
    1. Ebert S, Podlech J, Gillert-Marien D, Gergely KM, Büttner JK, Fink A, et al. Parameters determining the efficacy of adoptive CD8 T-cell therapy of Cytomegalovirus infection. Med Microbiol Immunol (2012) 201:527–39.10.1007/s00430-012-0258-x
    1. Riddell SR, Watanabe KS, Goodrich JM, Li CR, Agha ME, Greenberg PD. Restoration of viral immunity in immunodeficient humans by the adoptive transfer of T cell clones. Science (1992) 257:238–41.10.1126/science.1352912
    1. Walter EA, Greenberg PD, Gilbert MJ, Finch RJ, Watanabe KS, Thomas ED, et al. Reconstitution of cellular immunity against Cytomegalovirus in recipients of allogeneic bone marrow by transfer of T-cell clones from the donor. N Engl J Med (1995) 333:1038–44.10.1056/NEJM199510193331603
    1. Einsele H, Roosnek E, Rufer N, Sinzger C, Riegler S, Löffler J, et al. Infusion of Cytomegalovirus (CMV)-specific T cells for the treatment of CMV infection not responding to antiviral chemotherapy. Blood (2002) 99:3916–22.10.1182/blood.V99.11.3916
    1. Peggs KS, Verfuerth S, Pizzey A, Khan N, Guiver M, Moss PA, et al. Adoptive cellular therapy for early Cytomegalovirus infection after allogeneic stem-cell transplantation with virus-specific T-cell lines. Lancet (2003) 362:1375–7.10.1016/S0140-6736(03)14634-X
    1. Cobbold M, Khan N, Pourgheysari B, Tauro S, McDonald D, Osman H, et al. Adoptive transfer of Cytomegalovirus-specific CTL to stem cell transplant patients after selection by HLA-peptide tetramers. J Exp Med (2005) 202:379–86.10.1084/jem.20040613
    1. Odendahl M, Grigoleit GU, Bönig H, Neuenhahn M, Albrecht J, Anderl F, et al. Clinical-scale isolation of ‘minimally manipulated’ Cytomegalovirus-specific donor lymphocytes for the treatment of refractory Cytomegalovirus disease. Cytotherapy (2014) 16:1245–56.10.1016/j.jcyt.2014.05.023
    1. Reddehase MJ, Koszinowski UH. Significance of herpesvirus immediate early gene expression in cellular immunity to Cytomegalovirus infection. Nature (1984) 312:369–71.10.1038/312369a0
    1. Reddehase MJ, Mutter W, Münch K, Bühring HJ, Koszinowski UH. CD8-positive T lymphocytes specific for murine Cytomegalovirus immediate-early antigens mediate protective immunity. J Virol (1987) 61:3102–8.
    1. Reddehase MJ, Rothbard JB, Koszinowski UH. A pentapeptide as minimal antigenic determinant for MHC class I-restricted T lymphocytes. Nature (1989) 337:651–3.10.1038/337651a0
    1. Del Val M, Schlicht HJ, Volkmer H, Messerle M, Reddehase MJ, Koszinowski UH. Protection against lethal Cytomegalovirus infection by a recombinant vaccine containing a single nonameric T-cell epitope. J Virol (1991) 65:3641–6.
    1. Stinski MF, Isomura H. Role of the Cytomegalovirus major immediate early enhancer in acute infection and reactivation from latency. Med Microbiol Immunol (2008) 197:223–31.10.1007/s00430-007-0069-7
    1. Kurz SK, Rapp M, Steffens HP, Grzimek NK, Schmalz S, Reddehase MJ. Focal transcriptional activity of murine Cytomegalovirus during latency in the lungs. J Virol (1999) 73:482–94.
    1. Grzimek NK, Dreis D, Schmalz S, Reddehase MJ. Random, asynchronous, and asymmetric transcriptional activity of enhancer-flanking major immediate-early genes ie1/3 and ie2 during murine Cytomegalovirus latency in the lungs. J Virol (2001) 75:2692–705.10.1128/JVI.75.6.2692-2705.2001
    1. Simon CO, Holtappels R, Tervo HM, Böhm V, Däubner T, Oehrlein-Karpi SA, et al. CD8 T cells control Cytomegalovirus latency by epitope-specific sensing of transcriptional reactivation. J Virol (2006) 80:10436–56.10.1128/JVI.01248-06
    1. Reddehase MJ, Simon CO, Seckert CK, Lemmermann N, Grzimek NK. Murine model of Cytomegalovirus latency and reactivation. Curr Top Microbiol Immunol (2008) 325:315–31.10.1007/978-3-540-77349-8_18
    1. Holtappels R, Pahl-Seibert MF, Thomas D, Reddehase MJ. Enrichment of immediate-early 1 (m123/pp89) peptide-specific CD8 T cells in a pulmonary CD62L(lo) memory-effector cell pool during latent murine Cytomegalovirus infection of the lungs. J Virol (2000) 74:11495–503.10.1128/JVI.74.24.11495-11503.2000
    1. Karrer U, Sierro S, Wagner M, Oxenius A, Hengel H, Koszinowski UH, et al. Memory inflation: continuous accumulation of antiviral CD8+ T cells over time. J Immunol (2003) 170:2022–9; Correction in J Immunol (2003) 17:3895.10.4049/jimmunol.170.4.2022
    1. Seckert CK, Griessl M, Büttner JK, Scheller S, Simon CO, Kropp KA, et al. Viral latency drives ‘memory inflation’: a unifying hypothesis linking two hallmarks of Cytomegalovirus infection. Med Microbiol Immunol (2012) 201:551–66.10.1007/s00430-012-0273-y
    1. Seckert CK, Grießl M, Büttner JK, Freitag K, Lemmermann NA, Hummel MA, et al. Immune surveillance of Cytomegalovirus latency and reactivation in murine models: link to ‘memory inflation’. In: Reddehase MJ, editor. Cytomegaloviruses: From Molecular Pathogenesis to Intervention. (Vol. 1), Norfolk, UK: Caister Academic Press; (2013). p. 374–416.
    1. Borysiewicz LK, Hickling JK, Graham S, Sinclair J, Cranage MP, Smith GL, et al. Human Cytomegalovirus-specific cytotoxic T cells. Relative frequency of stage-specific CTL recognizing the 72-kD immediate early protein and glycoprotein B expressed by recombinant vaccinia viruses. J Exp Med (1988) 168:919–31.10.1084/jem.168.3.919
    1. McLaughlin-Taylor E, Pande H, Forman SJ, Tanamachi B, Li CR, Zaia JA, et al. Identification of the major late human Cytomegalovirus matrix protein pp65 as a target antigen for CD8+ virus-specific cytotoxic T lymphocytes. J Med Virol (1994) 43:103–10.10.1002/jmv.1890430119
    1. Wills MR, Carmichael AJ, Mynard K, Jin X, Weekes MP, Plachter B, et al. 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) 70:7569–79.
    1. Büscher N, Paulus C, Nevels M, Tenzer S, Plachter B. The proteome of human Cytomegalovirus virions and dense bodies is conserved across different strains. Med Microbiol Immunol (2015) 204:285–93.10.1007/s00430-015-0397-y
    1. Kern F, Surel IP, Faulhaber N, Frömmel C, Schneider-Mergener J, Schönemann C, et al. Target structures of the CD8(+)-T-cell response to human Cytomegalovirus: the 72-kilodalton major immediate-early protein revisited. J Virol (1999) 73:8179–84.
    1. Bunde T, Kirchner A, Hoffmeister B, Habedank D, Hetzer R, Cherepnev G, et al. Protection from Cytomegalovirus after transplantation is correlated with immediate early 1-specific CD8 T cells. J Exp Med (2005) 201:1031–6.10.1084/jem.20042384
    1. Sylwester AW, Mitchell BL, Edgar JB, Taormina C, Pelte C, Ruchti F, et al. Broadly targeted human Cytomegalovirus-specific CD4+ and CD8+ T cells dominate the memory compartments of exposed subjects. J Exp Med (2005) 202:673–85.10.1084/jem.20050882
    1. Thomas S, Klobuch S, Podlech J, Plachter B, Hoffmann P, Renzaho A, et al. Evaluating human T-cell therapy of Cytomegalovirus organ disease in HLA-transgenic mice. PLoS Pathog (2015) 11:e1005049.10.1371/journal.ppat.1005049
    1. Revello MG, Gerna G. State of the art and trends in Cytomegalovirus diagnostics. In: Reddehase MJ, editor. Cytomegaloviruses: From Molecular Pathogenesis to Intervention. (Vol. 2), Norfolk, UK: Caister Academic Press; (2013). p. 380–99.
    1. Holtappels R, Ebert S, Podlech J, Fink A, Böhm V, Lemmermann NA, et al. Murine model for cytoimmunotherapy of CMV disease after haematopoietic cell transplantation. In: Reddehase MJ, editor. Cytomegaloviruses: From Molecular Pathogenesis to Intervention. (Vol. 2), Norfolk, UK: Caister Academic Press; (2013). p. 352–79.
    1. Podlech J, Holtappels R, Wirtz N, Steffens HP, Reddehase MJ. Reconstitution of CD8 T cells is essential for the prevention of multiple-organ Cytomegalovirus histopathology after bone marrow transplantation. J Gen Virol (1998) 79:2099–104.10.1099/0022-1317-79-9-2099
    1. Podlech J, Holtappels R, Pahl-Seibert MF, Steffens HP, Reddehase MJ. Murine model of interstitial Cytomegalovirus pneumonia in syngeneic bone marrow transplantation: persistence of protective pulmonary CD8-T-cell infiltrates after clearance of acute infection. J Virol (2000) 74:7496–507.10.1128/JVI.74.16.7496-7507.2000
    1. Bianco P, Riminucci M, Gronthos S, Robey PG. Bone marrow stromal stem cells: nature, biology, and potential applications. Stem Cells (2001) 19:180–92.10.1634/stemcells.19-3-180
    1. Busch FW, Mutter W, Koszinowski UH, Reddehase MJ. Rescue of myeloid lineage-committed preprogenitor cells from Cytomegalovirus-infected bone marrow stroma. J Virol (1991) 65:981–4.
    1. Apperley JF, Dowding C, Hibbin J, Buiter J, Matutes E, Sissons PJ, et al. The effect of Cytomegalovirus on hemopoiesis: in vitro evidence for selective infection of marrow stromal cells. Exp Hematol (1989) 17:38–45.
    1. Simmons P, Kaushansky K, Torok-Storb B. Mechanisms of Cytomegalovirus-mediated myelosuppression: perturbation of stromal cell function versus direct infection of myeloid cells. Proc Natl Acad Sci U S A (1990) 87:1386–90.10.1073/pnas.87.4.1386
    1. Taylor-Wiedeman J, Sissons JG, Borysiewicz LK, Sinclair JH. Monocytes are a major site of persistence of human Cytomegalovirus in peripheral blood mononuclear cells. J Gen Virol (1991) 72:2059–64.10.1099/0022-1317-72-9-2059
    1. Maciejewski JP, Bruening EE, Donahue RE, Mocarski ES, Young NS, St Jeor SC. Infection of hematopoietic progenitor cells by human Cytomegalovirus. Blood (1992) 80:170–8.
    1. Söderberg-Nauclér C, Fish KN, Nelson JA. Reactivation of latent human Cytomegalovirus by allogeneic stimulation of blood cells from healthy donors. Cell (1997) 91:119–26.10.1016/S0092-8674(01)80014-3
    1. Hahn G, Jores R, Mocarski ES. Cytomegalovirus remains latent in a common precursor of dendritic and myeloid cells. Proc Natl Acad Sci U S A (1998) 95:3937–42.10.1073/pnas.95.7.3937
    1. Goodrum FD, Jordan CT, High K, Shenk T. Human Cytomegalovirus gene expression during infection of primary hematopoietic progenitor cells: a model for latency. Proc Natl Acad Sci U S A (2002) 99:16255–60.10.1073/pnas.252630899
    1. Emery VC. Relative importance of Cytomegalovirus load as a risk factor for Cytomegalovirus disease in the immunocompromised host. In: Scholz M, Rabenau HF, Doerr HW, Cinatl J, editors. CMV-Related Immunopathology. Basel, Swiss: Karger; (1998). p. 288–301.
    1. Crapnell KB, Almeida-Porada G, Khaiboullina S, St Jeor SC, Zanjani ED. Human haematopoietic stem cells that mediate long-term in vivo engraftment are not susceptible to infection by human Cytomegalovirus. Br J Haematol (2004) 124:676–84.10.1111/j.1365-2141.2004.04827.x
    1. Seckert CK, Renzaho A, Reddehase MJ, Grzimek NK. Hematopoietic stem cell transplantation with latently infected donors does not transmit virus to immunocompromised recipients in the murine model of Cytomegalovirus infection. Med Microbiol Immunol (2008) 197:251–9.10.1007/s00430-008-0094-1
    1. Adler B, Sinzger C. Cytomegalovirus interstrain variance in cell type tropism. In: Reddehase MJ, editor. Cytomegaloviruses: From Molecular Pathogenesis to Intervention. (Vol. 1), Norfolk, UK: Caister Academic Press; (2013). p. 297–321.
    1. Wagner FM, Brizic I, Prager A, Trsan T, Arapovic M, Lemmermann NA, et al. The viral chemokine MCK-2 of murine Cytomegalovirus promotes infection as part of a gH/gL/MCK-2 complex. PLoS Pathog (2013) 9:e1003493.10.1371/journal.ppat.1003493
    1. Lemmermann NA, Krmpotic A, Podlech J, Brizic I, Prager A, Adler H, et al. Non-redundant and redundant roles of Cytomegalovirus gH/gL complexes in host organ entry and intra-tissue spread. PLoS Pathog (2015) 11:e1004640.10.1371/journal.ppat.1004640
    1. Messerle M, Crnkovic I, Hammerschmidt W, Ziegler H, Koszinowski UH. Cloning and mutagenesis of a herpesvirus genome as an infectious bacterial artificial chromosome. Proc Natl Acad Sci U S A (1997) 94:14759–63.10.1073/pnas.94.26.14759
    1. Wagner M, Jonjic S, Koszinowski UH, Messerle M. Systematic excision of vector sequences from the BAC-cloned herpesvirus genome during virus reconstitution. J Virol (1999) 73:7056–60.
    1. Lemmermann NA, Kropp KA, Seckert CK, Grzimek NK, Reddehase MJ. Reverse genetics modification of Cytomegalovirus antigenicity and immunogenicity by CD8 T-cell epitope deletion and insertion. J Biomed Biotechnol (2011) 2011:812742.10.1155/2011/812742
    1. Jordan S, Krause J, Prager A, Mitrovic M, Jonjic S, Koszinowski UH, et al. Virus progeny of murine Cytomegalovirus bacterial artificial chromosome pSM3fr show reduced growth in salivary glands due to a fixed mutation of MCK-2. J Virol (2011) 85:10346–53.10.1128/JVI.00545-11
    1. Mutter W, Reddehase MJ, Busch FW, Bühring HJ, Koszinowski UH. Failure in generating hemopoietic stem cells is the primary cause of death from Cytomegalovirus disease in the immunocompromised host. J Exp Med (1988) 167:1645–58.10.1084/jem.167.5.1645
    1. Reddehase MJ. Bone marrow dysfunction in irradiated, Cytomegalovirus-infected mice. Transplant Proc (1991) 23:10–1.
    1. Reddehase MJ, Dreher-Stumpp L, Angele P, Balthesen M, Susa M. Hematopoietic stem cell deficiency resulting from Cytomegalovirus infection of bone marrow stroma. Ann Hematol (1992) 64:A125–7.10.1007/BF01715364
    1. Mayer A, Podlech J, Kurz S, Steffens HP, Maiberger S, Thalmeier K, et al. Bone marrow failure by Cytomegalovirus is associated with an in vivo deficiency in the expression of essential stromal hemopoietin genes. J Virol (1997) 71:4589–98.
    1. Dobonici M, Podlech J, Steffens HP, Maiberger S, Reddehase MJ. Evidence against a key role for transforming growth factor-beta1 in Cytomegalovirus-induced bone marrow aplasia. J Gen Virol (1998) 79:867–76.10.1099/0022-1317-79-4-867
    1. Steffens HP, Podlech J, Kurz S, Angele P, Dreis D, Reddehase MJ. Cytomegalovirus inhibits the engraftment of donor bone marrow cells by downregulation of hemopoietin gene expression in recipient stroma. J Virol (1998) 72:5006–15.
    1. Chabannon C, Torok-Storb B. Stem cell-stromal cell interactions. Curr Top Microbiol Immunol (1992) 177:123–36.
    1. Schofield R. The pluripotent stem cell. Clin Haematol (1979) 8:221–37.
    1. Heazlewood SY, Oteiza A, Cao H, Nilsson SK. Analyzing hematopoietic stem cell homing, lodgment, and engraftment to better understand the bone marrow niche. Ann N Y Acad Sci (2014) 1310:119–28.10.1111/nyas.12329
    1. Birbrair A, Frenette PS. Niche heterogeneity in the bone marrow. Ann N Y Acad Sci (2016) 1370:82–96.10.1111/nyas.13016
    1. Hoggatt J, Kfoury Y, Scadden DT. Hematopoietic stem cell niche in health and disease. Annu Rev Pathol (2016) 11:555–81.10.1146/annurev-pathol-012615-044414
    1. Yu VW, Scadden DT. Heterogeneity of the bone marrow niche. Curr Opin Hematol (2016) 23:331–8.10.1097/MOH.0000000000000265
    1. Driessen RL, Johnston HM, Nilsson SK. Membrane-bound stem cell factor is a key regulator in the initial lodgment of stem cells within the endosteal marrow region. Exp Hematol (2003) 31:1284–91.10.1016/j.exphem.2003.08.015
    1. Quinnan GV, Kirmani N, Rook AH, Manischewitz JF, Jackson L, Moreschi G, et al. 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) 307:7–13.10.1056/NEJM198207013070102
    1. Reusser P, Riddell SR, Meyers JD, Greenberg PD. Cytotoxic T-lymphocyte response to Cytomegalovirus after human allogeneic bone marrow transplantation: pattern of recovery and correlation with Cytomegalovirus infection and disease. Blood (1991) 78:1373–80.
    1. Alterio de Goss M, Holtappels R, Steffens HP, Podlech J, Angele P, Dreher L, et al. Control of Cytomegalovirus in bone marrow transplantation chimeras lacking the prevailing antigen-presenting molecule in recipient tissues rests primarily on recipient-derived CD8 T cells. J Virol (1998) 72:7733–44.
    1. Jonjić S, Pavić I, Lucin P, Rukavina D, Koszinowski UH. Efficacious control of Cytomegalovirus infection after long-term depletion of CD8+ T lymphocytes. J Virol (1990) 64:5457–64.
    1. Polić B, Jonjić S, Pavić I, Crnković I, Zorica I, Hengel H, et al. Lack of MHC class I complex expression has no effect on spread and control of Cytomegalovirus infection in vivo. J Gen Virol (1996) 77:217–25.10.1099/0022-1317-77-2-217
    1. Khairallah C, Netzer S, Villacreces A, Juzan M, Rousseau B, Dulanto S, et al. γδ T cells confer protection against murine Cytomegalovirus (MCMV). PLoS Pathog (2015) 11:e1004702.10.1371/journal.ppat.1004702
    1. Bukowski JF, Warner JF, Dennert G, Welsh RM. Adoptive transfer studies demonstrating the antiviral effect of natural killer cells in vivo. J Exp Med (1985) 161:40–52.10.1084/jem.161.1.40
    1. Klenovsek K, Weisel F, Schneider A, Appelt U, Jonjic S, Messerle M, et al. Protection from CMV infection in immunodeficient hosts by adoptive transfer of memory B cells. Blood (2007) 110:3472–9.10.1182/blood-2007-06-095414
    1. Jeitziner SM, Walton SM, Torti N, Oxenius A. Adoptive transfer of Cytomegalovirus-specific effector CD4+ T cells provides antiviral protection from murine CMV infection. Eur J Immunol (2013) 43:2886–95.10.1002/eji.201343690
    1. Sell S, Dietz M, Schneider A, Holtappels R, Mach M, Winkler TH. Control of murine Cytomegalovirus infection by γδ T cells. PLoS Pathog (2015) 11:e1004481.10.1371/journal.ppat.1004481
    1. Riddell SR. Pathogenesis of Cytomegalovirus pneumonia in immunocompromised hosts. Semin Respir Infect (1995) 10:199–208.
    1. Seo S, Boeckh M. Clinical Cytomegalovirus research: haematopoietic cell transplantation. In: Reddehase MJ, editor. Cytomegaloviruses: From Molecular Pathogenesis to Intervention. (Vol. 2), Norfolk, UK: Caister Academic Press; (2013). p. 335–51.
    1. Brody AR, Craighead JE. Pathogenesis of pulmonary Cytomegalovirus infection in immunosuppressed mice. J Infect Dis (1974) 129:677–89.10.1093/infdis/129.6.677
    1. Jordan MC. Interstitial pneumonia and subclinical infection after intranasal inoculation of murine Cytomegalovirus. Infect Immun (1978) 21:275–80.
    1. Shanley JD, Pesanti EL, Nugent KM. The pathogenesis of pneumonitis due to murine Cytomegalovirus. J Infect Dis (1982) 146:388–96.10.1093/infdis/146.3.388
    1. Holtappels R, Podlech J, Geginat G, Steffens HP, Thomas D, Reddehase MJ. Control of murine Cytomegalovirus in the lungs: relative but not absolute immunodominance of the immediate-early 1 nonapeptide during the antiviral cytolytic T-lymphocyte response in pulmonary infiltrates. J Virol (1998) 72:7201–12.
    1. Balthesen M, Messerle M, Reddehase MJ. Lungs are a major organ site of Cytomegalovirus latency and recurrence. J Virol (1993) 67:5360–6.
    1. Kataoka N, Shinohara H, Takayama K, Takaku K, Kondo S, Yonehara S, et al. Concanamycin A, a powerful tool for characterization and estimation of contribution of perforin- and Fas-based lytic pathways in cell-mediated cytotoxicity. J Immunol (1996) 156:3678–86.
    1. Seckert CK, Schader SI, Ebert S, Thomas D, Freitag K, Renzaho A, et al. Antigen-presenting cells of haematopoietic origin prime Cytomegalovirus-specific CD8 T-cells but are not sufficient for driving memory inflation during viral latency. J Gen Virol (2011) 92:1994–2005.10.1099/vir.0.031815-0
    1. Lefrançois L, Obar JJ. Once a killer, always a killer: from cytotoxic T cell to memory cell. Immunol Rev (2010) 235:206–18.10.1111/j.0105-2896.2010.00895.x
    1. Böhm V, Podlech J, Thomas D, Deegen P, Pahl-Seibert MF, Lemmermann NA, et al. Epitope-specific in vivo protection against Cytomegalovirus disease by CD8 T cells in the murine model of preemptive immunotherapy. Med Microbiol Immunol (2008) 197:135–44.10.1007/s00430-008-0092-3
    1. Sacher T, Podlech J, Mohr CA, Jordan S, Ruzsics Z, Reddehase MJ, et al. The major virus-producing cell type during murine Cytomegalovirus infection, the hepatocyte, is not the source of virus dissemination in the host. Cell Host Microbe (2008) 3:263–72.10.1016/j.chom.2008.02.014
    1. Ebert S, Becker M, Lemmermann NA, Büttner JK, Michel A, Taube C, et al. Mast cells expedite control of pulmonary murine Cytomegalovirus infection by enhancing the recruitment of protective CD8 T cells to the lungs. PLoS Pathog (2014) 10:e1004100.10.1371/journal.ppat.1004100
    1. Holtappels R, Lemmermann NA, Podlech J, Ebert S, Reddehase MJ. Reconstitution of CD8 T cells protective against Cytomegalovirus in a mouse model of hematopoietic cell transplantation: dynamics and inessentiality of epitope immunodominance. Front Immunol (2016) 7:232.10.3389/fimmu.2016.00232
    1. Walton SM, Torti N, Mandaric S, Oxenius A. T-cell help permits memory CD8(+) T-cell inflation during Cytomegalovirus latency. Eur J Immunol (2011) 41:2248–59.10.1002/eji.201141575
    1. Becker M, Lemmermann NA, Ebert S, Baars P, Renzaho A, Podlech J, et al. Mast cells as rapid innate sensors of Cytomegalovirus by TLR3/TRIF signaling-dependent and -independent mechanisms. Cell Mol Immunol (2015) 12:192–201.10.1038/cmi.2014.73
    1. Podlech J, Ebert S, Becker M, Reddehase MJ, Stassen M, Lemmermann NA. Mast cells: innate attractors recruiting protective CD8 T cells to sites of Cytomegalovirus infection. Med Microbiol Immunol (2015) 204:327–34.10.1007/s00430-015-0386-1
    1. Munks MW, Gold MC, Zajac AL, Doom CM, Morello CS, Spector DH, et al. Genome-wide analysis reveals a highly diverse CD8 T cell response to murine Cytomegalovirus. J Immunol (2006) 176:3760–6.10.4049/jimmunol.176.6.3760
    1. Nikolich-Zugich J, Slifka MK, Messaoudi I. The many important facets of T-cell repertoire diversity. Nat Rev Immunol (2004) 4:123–32.10.1038/nri1292
    1. Jenkins MK, Chu HH, McLachlan JB, Moon JJ. On the composition of the preimmune repertoire of T cells specific for peptide-major histocompatibility complex ligands. Annu Rev Immunol (2010) 28:275–94.10.1146/annurev-immunol-030409-101253
    1. Jenkins MK, Moon JJ. The role of naive T cell precursor frequency and recruitment in dictating immune response magnitude. J Immunol (2012) 188:4135–40.10.4049/jimmunol.1102661
    1. Jaeger S, Fernandez B, Ferrier P. Epigenetic aspects of lymphocyte antigen receptor gene rearrangement or ‘when stochasticity completes randomness’. Immunology (2013) 139:141–50.10.1111/imm.12057
    1. Holtappels R, Simon CO, Munks MW, Thomas D, Deegen P, Kühnapfel B, et al. Subdominant CD8 T-cell epitopes account for protection against Cytomegalovirus independent of immunodomination. J Virol (2008) 82:5781–96.10.1128/JVI.00155-08
    1. Nauerth M, Weißbrich B, Knall R, Franz T, Dössinger G, Bet J, et al. TCR-ligand koff rate correlates with the protective capacity of antigen-specific CD8+ T cells for adoptive transfer. Sci Transl Med (2013) 5:192ra87.10.1126/scitranslmed.3005958
    1. Lemmermann NA, Gergely K, Böhm V, Deegen P, Däubner T, Reddehase MJ. Immune evasion proteins of murine Cytomegalovirus preferentially affect cell surface display of recently generated peptide presentation complexes. J Virol (2010) 84:1221–36.10.1128/JVI.02087-09
    1. Lemmermann NA, Fink A, Podlech J, Ebert S, Wilhelmi V, Böhm V, et al. Murine Cytomegalovirus immune evasion proteins operative in the MHC class I pathway of antigen processing and presentation: state of knowledge, revisions, and questions. Med Microbiol Immunol (2012) 201:497–512.10.1007/s00430-012-0257-y
    1. Holtappels R, Thomas D, Podlech J, Reddehase MJ. Two antigenic peptides from genes m123 and m164 of murine Cytomegalovirus quantitatively dominate CD8 T-cell memory in the H-2d haplotype. J Virol (2002) 76:151–64.10.1128/JVI.76.1.151-164.2002
    1. Däubner T, Fink A, Seitz A, Tenzer S, Müller J, Strand D, et al. A novel transmembrane domain mediating retention of a highly motile herpesvirus glycoprotein in the endoplasmic reticulum. J Gen Virol (2010) 91:1524–34.10.1099/vir.0.018580-0
    1. Fink A, Büttner JK, Thomas D, Holtappels R, Reddehase MJ, Lemmermann NA. Noncanonical expression of a murine Cytomegalovirus early protein CD8 T-cell epitope as an immediate early epitope based on transcription from an upstream gene. Viruses (2014) 6:808–31.10.3390/v6020808
    1. Ebert S, Lemmermann NA, Thomas D, Renzaho A, Reddehase MJ, Holtappels R. Immune control in the absence of immunodominant epitopes: implications for immunotherapy of Cytomegalovirus infection with antiviral CD8 T cells. Med Microbiol Immunol (2012) 201:541–50.10.1007/s00430-012-0268-8
    1. Steffens HP, Kurz S, Holtappels R, Reddehase MJ. Preemptive CD8 T-cell immunotherapy of acute Cytomegalovirus infection prevents lethal disease, limits the burden of latent viral genomes, and reduces the risk of virus recurrence. J Virol (1998) 72:1797–804.
    1. Pahl-Seibert MF, Juelch M, Podlech J, Thomas D, Deegen P, Reddehase MJ, et al. Highly protective in vivo function of Cytomegalovirus IE1 epitope-specific memory CD8 T cells purified by T-cell receptor-based cell sorting. J Virol (2005) 79:5400–13.10.1128/JVI.79.9.5400-5413.2005
    1. Reddehase MJ, Balthesen M, Rapp M, Jonjić S, Pavić I, Koszinowski UH. The conditions of primary infection define the load of latent viral genome in organs and the risk of recurrent Cytomegalovirus disease. J Exp Med (1994) 179:185–93.10.1084/jem.179.1.185
    1. Wills MR, Mason GM, Sissons JG. Adaptive cellular immunity to human Cytomegalovirus. In: Reddehase MJ, editor. Cytomegaloviruses: From Molecular Pathogenesis to Intervention. (Vol. 2), Norfolk, UK: Caister Academic Press; (2013). p. 141–71.

Source: PubMed

3
Abonnere