Systems-based based approaches have begun to shed light on extrinsic factors that contribute to immune system variation. Among these, CMV (HHV-5, a β-herpesvirus) imposes a surprisingly profound impact. Most of the world’s population is CMV+, and the virus goes through three distinct infection phases en route to establishing lifelong détente with its host. Immune control of CMV in each phase recruits unique arms of host defense, and in turn the virus employs multiple immune-modulatory strategies that help facilitate the establishment of lifelong persistence. In this review, we explain how CMV shapes immunity and discuss the impact it may have on overall health.

In recent years, systems approaches have been used to begin elucidating the extent that the immune system differs between individuals. These have focused largely on measuring the composition, phenotype, and gene-expression patterns of circulating immune cell populations (13). The hope is that this will begin to clarify how variation in these parameters relates to disease, how they might impact vaccination responses, and, ultimately, how they help develop a blueprint for better overall health. Concurrent technological advancements have allowed the measurement of multiple, individual immune cell frequencies and immune-regulating cytokines from patient blood samples (e.g., mass cytometry and multiplex ELISA), and in combination with unbiased genomics approaches will ultimately help to spawn personalized medicine (4, 5). Notably, nonheritable factors are major drivers of immune system variation, with one prime example being the microbiome (6). However, quite surprisingly, another major factor is human CMV (HCMV) infection (HHV-5, a β-herpesvirus) (7). Identical twins discordant for HCMV infection vary in >50% of ∼200 measured immune parameters (7), a remarkable impact for a single, common viral infection. In this review, we discuss unique aspects of the broad immune response to HCMV infection that arise during its distinct phases of infection, specific strategies used by CMV to target them, and how this may ultimately impact health and disease.

HCMV is the prototypic member of the Betaherpesvirinae and is a dsDNA virus with an ∼236-kb genome expressing up to ∼750 protein-encoding open reading frames emanating from complex control of transcription and splicing (8, 9). CMVs, similar to all Herpesviridae, have coevolved with their individual hosts for millions of years to establish a persistent/latent infection that is never cleared (10). Consequently, CMV replication is species specific, in large part because the virus has evolved many fine-tuned strategies to inhibit various immune defenses unique to those hosts (11, 12). In the United States, infection rates of prepubescent children range from 30 to 60%, varying by sex, ethnicity, and socioeconomic status, increasing steadily to 50–90% by the age of 50 y. In general, infection is lowest in non-Hispanic white males, persons of higher education, and those living in less crowded conditions (13). The overall incidence of CMV infection is significantly higher in South America, Asia, and Africa (>90%) as compared with the United States and Western Europe (14). Consequently, the fact that many people living today in the Western developed world remain uninfected with CMV into adulthood has facilitated assessing how it impacts immunity.

CMV infection has three distinct phases: 1) a systemic replication phase in many peripheral tissues that strongly activates the innate immune system and specific NK cell populations and primes a diverse Ab and T effector memory (Tem) cell response; 2) a tissue-localized persistent phase that continues for months to years and continues to shape innate and adaptive immunity; and 3) multisite latency with restricted viral gene expression that promotes immune inflation during a subsequent lifetime (1518) (Fig. 1, Table I). Despite this prolonged and multifaceted interaction with its host, CMV normally only causes acute disease when immunity is naive or compromised, exemplifying how coevolution over millennia has resulted in a largely nonpathogenic détente. However, the extremely broad impact of CMV on immune system homeostasis in healthy adults likely impacts aspects of health and disease over time.

FIGURE 1.

CMV impacts the immune system during the course of a lifetime. Top panel, Depicted are the three phases of CMV infection: I) the acute/systemic phase, which is controlled after a few weeks; II) the persistent replication phase that can continue for months to years in people (weeks to months in mice); and III) the percolating latency phase where an ongoing détente between the virus and its host continues to shape and inflate host defenses. Bottom panel, CMV impacts both innate and adaptive cell populations and immune responses during its three-phase infection, and shown are several that are directly impacted.

FIGURE 1.

CMV impacts the immune system during the course of a lifetime. Top panel, Depicted are the three phases of CMV infection: I) the acute/systemic phase, which is controlled after a few weeks; II) the persistent replication phase that can continue for months to years in people (weeks to months in mice); and III) the percolating latency phase where an ongoing détente between the virus and its host continues to shape and inflate host defenses. Bottom panel, CMV impacts both innate and adaptive cell populations and immune responses during its three-phase infection, and shown are several that are directly impacted.

Close modal
Table I.
CMV infection phases in healthy individuals
PhaseI. Acute InfectionII. PersistenceIII. Latency/Reactivation
CMV genome expression Yes Yes Minimal/suppression of the major immediate-early promoter 
Virion production Yes Yes No/sporadic during reactivation 
Organs implicated Visceral organs (spleen, liver, lung, gastrointestinal tract) Mucosal organs (salivary glands)/liver CD14+ monocytes/CD34 progenitors/endothelial progenitors? 
Shedding in body fluids Yes Yes No 
Serology IgM IgM/IgG (low to high avidity) IgG (high avidity)/occasional IgM 
PhaseI. Acute InfectionII. PersistenceIII. Latency/Reactivation
CMV genome expression Yes Yes Minimal/suppression of the major immediate-early promoter 
Virion production Yes Yes No/sporadic during reactivation 
Organs implicated Visceral organs (spleen, liver, lung, gastrointestinal tract) Mucosal organs (salivary glands)/liver CD14+ monocytes/CD34 progenitors/endothelial progenitors? 
Shedding in body fluids Yes Yes No 
Serology IgM IgM/IgG (low to high avidity) IgG (high avidity)/occasional IgM 

Phase I: the innate immune response to initial CMV infection.

Primary CMV infection in healthy people is essentially asymptomatic, and therefore studies assessing the activation and contribution of innate defenses during the phase I infection in people are sparse. Additionally, the species-specific replication of CMV necessitates using animal models to help define key innate control mechanisms. In this regard, mouse CMV (MCMV) has been the most instructive (19), but rhesus (20, 21) and guinea pig (22) CMV models have also been very instructional. Most mouse and guinea pig infections are done systemically (i.p. or i.v.), with s.c. infection often performed in monkeys. How the initial activation/priming of innate and adaptive immune responses with these infection routes and doses might differ from that of natural mucosal infection is an important question and is discussed later.

NK cells and type I IFN (IFN-I) are key regulators of early CMV control (23, 24). MCMV induces two major systemic phases of IFN-I. The initial phase is produced by splenic stromal cells and is proportional to the infecting dose, peaks in sera at 10–12 h, and is dependent on B cell–derived lymphotoxin αβ signaling (25). The second phase occurs at 36–48 h in response to the first burst of MCMV production and is produced by both plasmacytoid dendritic cells (DC) and conventional DC. In addition to IFN-I, these DC subsets produce high levels of tissue-localized and systemic IL-12 and IL-18 (26, 27), which further NK cell activation (2830) and help to prime adaptive responses. Notably, second-phase IFN-I does not directly restrict MCMV replication levels (31), whereas the first phase does (32).

Work with MCMV during the last three decades has revealed many mechanisms by which NK cells contribute to antiviral defense in various inbred mouse strains (33), and more recent advances in genome sequencing has furthered this (34). It is clear that altered NK cell licensing and expression of disparate activating and inhibitory receptors underlie many of these differences (35, 36). In C57BL/6 mice ∼50% of splenic NK cells express the Ly49H activating receptor, which binds the MCMV m157 protein expressed on the surface of infected cells, promoting their resistance to viral replication in the spleen (3742). m157 induces the activation and proliferation of Ly49H+ NK cells during the first week of infection (43, 44), which then restrict MCMV replication mainly by cytolysis (45). Notably, in the absence of Ly49H–m157 interactions, the role of NK cells in restricting MCMV replication can be minimal (e.g., in BALB/c mice) (33, 46), in large part due to CMV neutralizing several NK cell effector pathways (35).

Several other innate effector cell types also make important contributions in both the direct and indirect control of early MCMV infection. Lymphoid tissue sinus-localized macrophages can capture MCMV during initial infection and restrict it from productively infecting reticular stromal cells (47). Mast cells are activated within hours of infection and produce chemokines that recruit CD8 T cells to infection sites within the first week (48, 49). Neutrophils are rapidly recruited by NK-produced IL-22 and contribute to viral control via TRAIL (50). Invariant NKT cells are activated by the first and second waves of innate cytokines, independently of CD1d, and help to control early MCMV replication (51, 52). The importance of invariant NKT cells in responding to HCMV infection is also suggested by viral restriction of CD1d expression (53, 54). Various myeloid cell types also help to shape the earliest response to CMV infection, but they also play a key role in disseminating CMV to sites of persistence and harboring latent viral genome in mice and humans, highlighting their complex role in CMV defenses (5560).

CMV shaping of the NK cell compartment.

A key role for NK cells in controlling HCMV infection is supported from a patient case study who lacked them (24). Historically, NK cells were thought to respond generally to viral infection, functioning en masse as front-line innate defenders. However, more recent data, many in mice, indicate that populations of memory-like NK cells that display properties of adaptive lymphocytes can be primed during phase I (61) and may be preferentially maintained during later phases of infection. One of these is the m157-driven expansion of Ly49H+ NK cells in C57BL/6 mice, which remain at higher levels for months postinfection and provide better MCMV protection when adoptively transferred into naive mice (62). Interestingly, healthy CMV-infected people >60 y of age show a significant increase in the numbers of CD56dim NK cells in blood (63). These cells also display a unique CD57+NKG2Chi cell surface phenotype (64), a distinct transcription factor expression profile, and epigenetic remodeling of the Ifnγ gene locus (65). They also preferentially expand in CMV-infected transplant patients, correlating with control of systemic viremia (64). Some studies indicate that NKG2C/HLA-E interactions might be impacting this NK cell subset (66, 67), and a currently unidentified HCMV-encoded NKG2C ligand has also been postulated (similar to Ly49H-m157 in mice), but the precise mechanisms underlying why NKG2Chi NK cells expand in CMV+ patients are yet to be fully defined (68). However, as NKG2Chi NK cells can comprise as much as 50% of the entire repertoire in some infected individuals (64), which results in a substantial reduction in overall NK pool diversity, this may impact the ability to respond to heterologous infections or tumor immune surveillance. Interestingly, a population of CD56+CD8+ NKT-like cells also expands in peripheral blood of middle-aged persons infected with CMV, but may be more a result of aging than being virus imposed per se (69).

Phase II: the adaptive immune response during CMV persistence.

The CMV-specific T cell response shows several unique characteristics (18) and is differentially impacted by the three infection phases. MCMV replication is ultimately controlled during the first week in most peripheral organs, correlating with the initial expansion of effector T cells, but replicates for many more weeks at high levels in selected mucosal sites such as the salivary gland. A robust and diverse epitope-specific CD8 and CD4 T cell response is primed in mice during this first week, most of which contract and go on to establish a stable memory pool in the next 1–2 wk (70, 71). In contrast, selected populations of T cells do not significantly contract, expand only during this second phase, and/or continue to increase during viral persistence, and these have been termed inflationary memory cells (15, 7173). These inflationary T cells display a largely Tem phenotype, are maintained by a relatively small population of CD27hi CD8 T cells that are more central memory–like (74), depend less on the immunoproteasome than do stable memory cells, are dependent on the initial dose of viral inoculum (75), and require the continuous transcriptional activity of FOXO1 (76). Notably, despite the phenotype of these inflationary T cells being consistent with their continued exposure to Ag, they do not show typical signs of exhausted T cells induced by several other chronic viral infections (77, 78).

As in mice, CMV induces a broad CD8 and CD4 T cell response that typically comprises 10–20% of all circulating T cells in CMV+ persons (79). HCMV also induces the expansion of selected, epitope-specific Tem cell subsets over time in healthy individuals, reaching staggeringly high numbers in some people (>20% for a single, epitope-specific response) (80, 81), and again typically show no signs of exhaustion. The expansion of these Tem cells scores as one of the most prominent CMV-imposed impacts in discordant monozygotic twins (7). Interestingly, these inflationary HCMV-specific T cells can use unique cosignaling pathways for their activation (82). Many lack expression of CD27 and/or CD28 (83), and the emergence of high numbers of HCMV-specific CD4 T cells displaying a cytolytic phenotype is also seen (84). Differential cosignaling requirements for stable and inflationary memory pools (8589) and induction of CD4 CTLs (90) is also observed in CMV-infected mice.

CMV-specific Ab responses are strongly induced. Polyclonal human Ig containing high HCMV Ab levels has been reported to provide some protection against congenital CMV infection and/or disease in at-risk pregnant women (91), but not all trials could reproduce this (92). This may be due to differing HCMV IgG avidity (93), or its specificity for particular envelope protein complexes that mediate viral entry into fibroblasts (gB/gH/gL, trimer) and other cell types (gH/gL/UL128–131, pentamer) (94, 95). In mice, transfer of MCMV Ig can reduce viral spread within tissues and restrict dissemination from sites of initial infection (96). The levels of MCMV IgG increase steadily throughout persistence/latency, although avidity appears to plateau several weeks before phase II is ultimately controlled, whereas IgM levels drop significantly after phase I (17). Inflation of HCMV IgG has also been observed in healthy infected persons (97). Mice deficient in B7-CD28 cosignaling are severely compromised in MCMV IgG responses (17), and in combination with their crippled T cell response never control phase II infection (R. Arens and C. Benedict, unpublished observations). Adoptive transfer of memory B cells into immune-deficient RAG−/− mice also reduces MCMV replication in both prophylactic and therapeutic settings, and it provides longer protection than serum Ig transfer (98). As seen for MCMV T cell responses (75), the magnitude of the Ig and memory B cell response is dose-dependent, although Ig inflation and affinity maturation does still occur at low doses (17). As natural infection by nursing or saliva transfer in both mice and people is likely to be relatively inefficient (99, 100), the relative initial infection efficiency may partly explain why CMV+ individuals often show significant variation in immune inflation decades later. Whether selected memory B cell subsets inflate in CMV-infected mice or humans is currently unknown, but B cells in CMV+ twins do show altered IL-10 signaling responses (7). Interestingly, some HCMV Ags appear to induce poor memory B cell responses during primary infection (101), suggesting that well-designed vaccine approaches may be able to outperform natural immunity. Despite all evidence that CMV induces a robust B cell/Ig response that is protective in several therapeutic settings, it is noteworthy that immune control of phase I and II MCMV infection is unchanged in mice lacking B cells (102), although control of viral reactivation in phase III is compromised to some extent (103), indicating that significant redundancy exists for immune control of primary infection.

Phase III: percolating CMV latency and its impact on the immune system.

After high-level persistent CMV replication during phase II is ultimately brought under control, viral latency is established and a host–virus détente ensues for life in phase III. Latency as defined originally for HSV is generally appreciated to be 1) stable, nuclear, and extrachromosomal maintenance of the viral genome in selected cells and tissue sites for the life of the host, 2) a state of restricted gene expression with no viral production, and 3) associated with the ability to reactivate from this state to full-replicative potential under the right conditions (104). CMV encodes the largest genome of the Herpesviridae (105), displays a broad cellular tropism, and is capable of establishing latency in several cell types (106). This differs from HSV, which establishes latency only in neurons. For HCMV, latency has been studied almost exclusively in hematopoietic precursor and myeloid lineage cells, in part because these cells are relatively easy to access from patients, but primarily because these cells clearly harbor latent viral genome in CMV+ people and good experimental models have been established (59). In mice, MCMV can establish latency in both endothelial and lymphoid-tissue reticular cell types (107, 108), and can be experimentally reactivated from various tissues (109).

Although less is known about the molecular and cell-intrinsic regulators of latency for MCMV than HCMV, mouse studies have revealed key aspects of how phase III infection impacts immune inflation (16). Inflationary T cells are maintained at higher levels when adoptively transferred into latently infected mice than into naive uninfected mice, indicating that sporadic/abortive CMV gene expression during phase III latency facilitates continued exposure of immune cells to viral Ag and accentuates immune inflation (73). The relevant cellular source driving this inflation is radioresistant stromal cells (110, 111), suggesting that CMV latency in nonmyeloid cells is a major driver. Interestingly, a spread-deficient MCMV mutant can also induce memory inflation, albeit at lower absolute levels (112). This suggests that a latency program can be triggered shortly after initial CMV entry in some cells, perhaps due to their specific differentiation/activation state, with other cellular states favoring the initiation of lytic replication and phase I infection. Evidence exists for a key role of lymphotoxin and IFN-I signaling in regulating this choice (113, 114). Importantly, these two paths do not have to be mutually exclusive, and they are likely to be conserved among the Herpesviridae (115). However, recent studies show that significantly more viral gene expression likely occurs during HCMV latency than dogma might predict (116118), many of which perform immune-modulatory functions (119, 120), and this may help to explain why T cells of particular Ag specificity undergo immune inflation and others do not.

Intriguingly, healthy CMV+ people with strong pre-existing immunity can be reinfected (121), and in mice reinfection enhances T cell inflation (122). The significant variability in the magnitude of immune inflation between individuals may depend in part on reinfection frequency. As many genetically distinct strains of HCMV exist (123), reinfection may just simply represent an insufficient ability of prior immunity to protect against a heterologous virus. However, a genetically identical rhesus CMV isolate can reinfect monkeys multiple times and induce strong primary Tem responses, a strategy that is being developed as a vaccination approach (124). Interestingly, the ability of rhesus CMV to superinfect in the presence of strong pre-existing immunity is dependent on viral immune evasion strategies that dampen CD8 T cell responses (125).

Finally, γδ T cells also contribute to controlling MCMV replication and limiting virus-induced organ disease (126), and specific clonotypes expand in mice and show inflation within selected organs, similar to that observed for αβ T cells. Support for a role of γδ T cells in controlling, or at a minimum strongly responding to, HCMV infection also exists in both healthy persons and transplant patients (7, 127, 128). However, as it remains unclear what self and/or CMV Ags γδ T cells recognize, it is difficult to know what phase of infection may be most critical in promoting their inflation.

Despite inducing a robust and diverse innate and adaptive immune response, CMV successfully progresses through its three-phase infection and establishes lifelong détente with its host. To aid in this, CMV has developed many sophisticated mechanisms targeting host immunity. It is likely that only about a third of the >750 CMV open reading frames are required for the nuts-and-bolts of entry and lytic replication (129), suggesting that the rest have evolved to combat the host immune response. Our laboratory has studied several of these immune-modulatory genes during the last two decades, primarily focusing on those that target and/or intersect with CD28 family and TNF family immune signaling pathways. These and many other CMV immune evasion mechanisms have been reviewed in recent years (12, 35, 55, 130135), so we only briefly discuss those that we deem to be the most likely to impact immune inflation.

CMV targeting of NK cells.

As already discussed, CMV induces the expansion of specific NK cell subsets. In addition to encoding proteins that specifically bind and activate NK cells (e.g., m157-Ly49H), CMV commensurately uses proteins to restrict NK cell–activating ligands and their receptors on the surface of infected cells. HCMV blocks expression of NKG2D ligands such as MICA/B and ULBP family proteins (35). HCMV UL141 restricts the surface expression of CD155 and CD122, which activate NK via DNAM-1 and CD94 (136). Additionally, UL141 also binds and inhibits expression of the TRAIL death receptors to block NK-mediated killing by this TNF family cytokine (137), highlighting the polyfunctionality that a single CMV immunomodulatory protein can have. MCMV utilizes a similar strategy via m166 to inhibit TRAIL apoptosis (46). HCMV also encodes proteins that bind NK inhibitory receptors, such as the MHC class I homolog UL18 that binds to LIR-1 (138), and UL40 that stabilizes HLA-E expression to promote NKG2A engagement (66). In fact, m157 can bind the inhibitory receptor Ly49i encoded in Ly49H 129/J mice, which likely attenuates NK cell activation (39). Collectively, this multilayered approach will almost certainly contribute to the CMV-induced expansion, function, and/or memory of specific NK subsets

CMV targeting of T cells.

CMV also utilizes multiple strategies to regulate detection by both CD8 and CD4 T cells. First, multiple HCMV and MCMV proteins operate to reduce cell surface MHC expression during lytic replication, thereby limiting T cell activation during phase I and II of infection (133, 139). In infected cells, the HCMV US2, 3, 6, 10, and 11 and MCMV m152/gp140, m04/gp34, and m05/gp48 proteins downregulate MHC membrane expression and limit peptide presentation to T cells. MCMV restricts expression of costimulatory molecules (e.g., B7 and CD40) and preferentially enhances expression of inhibitory ligands (e.g., PD-L1) as another T cell blockade strategy (140143), and viral mutants unable to block B7-CD28 signaling induce higher CD4 T cell response and show reduced replication in phase II (144). HCMV employs similar strategies to inhibit B7-CD28 signaling and promote death of T cells that encounter infected DC (145), although the specific viral proteins responsible have not been identified as for MCMV. HCMV UL144 is an ortholog of HVEM that selectively binds the coinhibitory molecule BTLA and potently blocks T cell proliferation (146148). UL144 can also activate NF-κB and is expressed during latency (120, 149), suggesting that it may impact T cell responses in phase III of infection. Additionally, all primate CMVs encode orthologs of IL-10 (150), and HCMV viral IL-10 is expressed during both lytic and latent infection, perhaps contributing to the creation of a latency niche where effector T cells are suppressed (117). Notably, HCMV can also promote production of host IL-10 by virus-specific CD4 T cells that encounter MHC class II–presented latency Ags (119, 151). This strategy of hijacking host IL-10 immunosuppression to promote viral persistence is similarly employed by MCMV (152, 153). Finally, CMV encodes both chemokines and chemokine receptors, which impact T cells and operate to fine-tune the inflammatory environment during all phases of infection (154).

Does the broad impact of CMV on the immune system ultimately contribute to human disease? CMV can be a serious human pathogen when immunity is compromised or naive, as in the case of transplant patients and fetal infection, and this is why vaccine development is a top priority (19, 155). However, there is a paucity of direct evidence for whether the lifelong détente in healthy people is deleterious, and the field remains largely agnostic on whether CMV is a driver or a passenger in associated diseases. CMV+ persons have been reported to exhibit a higher incidence of all-cause mortality, mainly from increased cardiovascular disease (CVD) (156158), and recent meta-analysis indicates CMV+ individuals have a 22% higher risk of developing CVD (159). As CMV infects vascular endothelial cells, a direct role for the virus is possible. Intriguingly, CMV CD4 T cells can express CX3CR1, which binds fractalkine expressed by activated/damaged endothelium, suggesting a mechanism by which CMV may kick-start or amplify CVD during multiple phases of infection (81, 160, 161). CVD is an autoinflammatory disease, and consequently whether CMV infection may be linked with other common autoimmune disorders such as rheumatoid arthritis, systemic sclerosis, systemic lupus erythematosus, diabetes, Sjögren’s syndrome, and several intestinal disorders has also been examined (162). However, because most data are from small patient cohorts and/or case studies, causation remains unclear.

Evidence for CMV negatively impacting immune function over time is suggested by its association with inflammaging and immune senescence in the elderly (163), with recent results indicating that it likely accelerates these processes (164). Very old Swedish cohorts exhibit an immune risk phenotype that includes CD4T/CD8T ratios <1 in blood (165, 166). However, the apparent ability of CMV to dial up the homeostatic rheostat of the immune system may also provide some benefit, as younger CMV+ people have been reported to mount better vaccination responses to influenza (167).

Although HCMV is not generally considered an oncogenic virus, viral nucleic acid has been reported to be present in various tumor types at high frequency (168), with the most notable example in recent years being that of glioblastoma (169). However, the absence of replicating virus, the low percentage of glioma cells harboring viral genomes, and the inability of some groups to detect HCMV at all has again made the contribution of CMV controversial. Nevertheless, some recent clinical results suggest that including CMV-targeted approaches in the treatment of glioblastoma may be helpful (170, 171). Future studies assessing whether the high proportions of inflationary CMV Tem might be preferentially recruited to tumor sites will be of interest, as their unique phenotype may impact the tumor microenvironment regardless of whether viral Ag-specific recognition occurs.

It is becoming more clear that immune system diversity among healthy individuals does not segregate into distinct groups, but instead varies across a continuum to generate multiple immunotypes (164). These immunotypes vary less in younger people, and even less in twins, and they highlight the importance of nonheritable factors such as CMV infection in shaping an individuals immune system (7, 164). Interestingly, if someone varies highly in the levels of one particular immune cell type, this does not correlate with them being an outlier for overall immune cell composition (164). CMV activates many arms of the immune system, and together with its modulatory strategies results in a major impact on its homeostasis. Given the multitude of acute and persistent infections that we encounter during a lifetime, it is quite extraordinary that CMV can shape immunity to this degree, and we would argue that this stems from unique aspects of its three-phase infection. It seems reasonable to hypothesize that this may ultimately impact health and/or disease at various levels, and the rapid advancements being made in systems-based immunity are likely to bring this into clearer focus in the coming years.

This work was supported by National Institutes of Health/National Institute for Allergy and Infectious Diseases Grants AI101423 and AI113349 (to C.A.B.).

Abbreviations used in this article:

CVD

cardiovascular disease

DC

dendritic cell

HCMV

human CMV

IFN-I

type I IFN

MCMV

mouse CMV

Tem

T effector memory.

1
Orrù
,
V.
,
M.
Steri
,
G.
Sole
,
C.
Sidore
,
F.
Virdis
,
M.
Dei
,
S.
Lai
,
M.
Zoledziewska
,
F.
Busonero
,
A.
Mulas
, et al
.
2013
.
Genetic variants regulating immune cell levels in health and disease.
Cell
155
:
242
256
.
2
Tsang
,
J. S.
,
P. L.
Schwartzberg
,
Y.
Kotliarov
,
A.
Biancotto
,
Z.
Xie
,
R. N.
Germain
,
E.
Wang
,
M. J.
Olnes
,
M.
Narayanan
,
H.
Golding
, et al
CHI Consortium
.
2014
.
Global analyses of human immune variation reveal baseline predictors of postvaccination responses. [Published erratum appears in 2014 Cell 158: 226.]
Cell
157
:
499
513
.
3
Pulendran
,
B.
2014
.
Systems vaccinology: probing humanity’s diverse immune systems with vaccines.
Proc. Natl. Acad. Sci. USA
111
:
12300
12306
.
4
Spitzer
,
M. H.
,
P. F.
Gherardini
,
G. K.
Fragiadakis
,
N.
Bhattacharya
,
R. T.
Yuan
,
A. N.
Hotson
,
R.
Finck
,
Y.
Carmi
,
E. R.
Zunder
,
W. J.
Fantl
, et al
.
2015
.
An interactive reference framework for modeling a dynamic immune system.
Science
349
:
1259425
.
5
Tian
,
Q.
,
N. D.
Price
,
L.
Hood
.
2012
.
Systems cancer medicine: towards realization of predictive, preventive, personalized and participatory (P4) medicine.
J. Intern. Med.
271
:
111
121
.
6
Lee
,
Y. K.
,
S. K.
Mazmanian
.
2010
.
Has the microbiota played a critical role in the evolution of the adaptive immune system?
Science
330
:
1768
1773
.
7
Brodin
,
P.
,
V.
Jojic
,
T.
Gao
,
S.
Bhattacharya
,
C. J. L.
Angel
,
D.
Furman
,
S.
Shen-Orr
,
C. L.
Dekker
,
G. E.
Swan
,
A. J.
Butte
, et al
.
2015
.
Variation in the human immune system is largely driven by non-heritable influences.
Cell
160
:
37
47
.
8
Stern-Ginossar
,
N.
,
B.
Weisburd
,
A.
Michalski
,
V. T. K.
Le
,
M. Y.
Hein
,
S. X.
Huang
,
M.
Ma
,
B.
Shen
,
S. B.
Qian
,
H.
Hengel
, et al
.
2012
.
Decoding human cytomegalovirus.
Science
338
:
1088
1093
.
9
Balázs
,
Z.
,
D.
Tombácz
,
A.
Szűcs
,
Z.
Csabai
,
K.
Megyeri
,
A. N.
Petrov
,
M.
Snyder
,
Z.
Boldogkői
.
2017
.
Long-read sequencing of human cytomegalovirus transcriptome reveals RNA isoforms carrying distinct coding potentials.
Sci. Rep.
7
:
15989
.
10
McGeoch
,
D. J.
,
F. J.
Rixon
,
A. J.
Davison
.
2006
.
Topics in herpesvirus genomics and evolution.
Virus Res.
117
:
90
104
.
11
Mocarski
,
E. S.
,
J. W.
Upton
,
W. J.
Kaiser
.
2011
.
Viral infection and the evolution of caspase 8-regulated apoptotic and necrotic death pathways.
Nat. Rev. Immunol.
12
:
79
88
.
12
Loewendorf
,
A.
,
C. A.
Benedict
.
2010
.
Modulation of host innate and adaptive immune defenses by cytomegalovirus: timing is everything.
J. Intern. Med.
267
:
483
501
.
13
Bate
,
S. L.
,
S. C.
Dollard
,
M. J.
Cannon
.
2010
.
Cytomegalovirus seroprevalence in the United States: the national health and nutrition examination surveys, 1988–2004.
Clin. Infect. Dis.
50
:
1439
1447
.
14
Cannon
,
M. J.
,
D. S.
Schmid
,
T. B.
Hyde
.
2010
.
Review of cytomegalovirus seroprevalence and demographic characteristics associated with infection.
Rev. Med. Virol.
20
:
202
213
.
15
Karrer
,
U.
,
S.
Sierro
,
M.
Wagner
,
A.
Oxenius
,
H.
Hengel
,
U. H.
Koszinowski
,
R. E.
Phillips
,
P.
Klenerman
.
2003
.
Memory inflation: continuous accumulation of antiviral CD8+ T cells over time.
J. Immunol.
170
:
2022
2029
.
16
Seckert
,
C. K.
,
M.
Griessl
,
J. K.
Büttner
,
S.
Scheller
,
C. O.
Simon
,
K. A.
Kropp
,
A.
Renzaho
,
B.
Kühnapfel
,
N. K. A.
Grzimek
,
M. J.
Reddehase
.
2012
.
Viral latency drives “memory inflation”: a unifying hypothesis linking two hallmarks of cytomegalovirus infection.
Med. Microbiol. Immunol.
201
:
551
566
.
17
Welten
,
S. P. M.
,
A.
Redeker
,
R. E. M.
Toes
,
R.
Arens
.
2016
.
Viral persistence induces antibody inflation without altering antibody avidity.
J. Virol.
90
:
4402
4411
.
18
Klenerman
,
P.
,
A.
Oxenius
.
2016
.
T cell responses to cytomegalovirus.
Nat. Rev. Immunol.
16
:
367
377
.
19
Benedict
,
C. A.
2013
.
A CMV vaccine: TREATing despite the TRICKs.
Expert Rev. Vaccines
12
:
1235
1237
.
20
Powers
,
C.
,
K.
Früh
.
2008
.
Rhesus CMV: an emerging animal model for human CMV.
Med. Microbiol. Immunol.
197
:
109
115
.
21
Itell
,
H. L.
,
A.
Kaur
,
J. D.
Deere
,
P. A.
Barry
,
S. R.
Permar
.
2017
.
Rhesus monkeys for a nonhuman primate model of cytomegalovirus infections.
Curr. Opin. Virol.
25
:
126
133
.
22
Schleiss
,
M. R.
2008
.
Comparison of vaccine strategies against congenital CMV infection in the guinea pig model.
J. Clin. Virol.
41
:
224
230
.
23
Verma
,
S.
,
C. A.
Benedict
.
2011
.
Sources and signals regulating type I interferon production: lessons learned from cytomegalovirus.
J. Interferon Cytokine Res.
31
:
211
218
.
24
Biron
,
C. A.
,
K. S.
Byron
,
J. L.
Sullivan
.
1989
.
Severe herpesvirus infections in an adolescent without natural killer cells.
N. Engl. J. Med.
320
:
1731
1735
.
25
Schneider
,
K.
,
A.
Loewendorf
,
C.
De Trez
,
J.
Fulton
,
A.
Rhode
,
H.
Shumway
,
S.
Ha
,
G.
Patterson
,
K.
Pfeffer
,
S. A.
Nedospasov
, et al
.
2008
.
Lymphotoxin-mediated crosstalk between B cells and splenic stroma promotes the initial type I interferon response to cytomegalovirus.
Cell Host Microbe
3
:
67
76
.
26
Andrews
,
D. M.
,
A. A.
Scalzo
,
W. M.
Yokoyama
,
M. J.
Smyth
,
M. A.
Degli-Esposti
.
2003
.
Functional interactions between dendritic cells and NK cells during viral infection.
Nat. Immunol.
4
:
175
181
.
27
Dalod
,
M.
,
T. P.
Salazar-Mather
,
L.
Malmgaard
,
C.
Lewis
,
C.
Asselin-Paturel
,
F.
Brière
,
G.
Trinchieri
,
C. A.
Biron
.
2002
.
Interferon α/β and interleukin 12 responses to viral infections: pathways regulating dendritic cell cytokine expression in vivo.
J. Exp. Med.
195
:
517
528
.
28
Bukowski
,
J. F.
,
J. F.
Warner
,
G.
Dennert
,
R. M.
Welsh
.
1985
.
Adoptive transfer studies demonstrating the antiviral effect of natural killer cells in vivo.
J. Exp. Med.
161
:
40
52
.
29
Orange
,
J. S.
,
C. A.
Biron
.
1996
.
Characterization of early IL-12, IFN-alphabeta, and TNF effects on antiviral state and NK cell responses during murine cytomegalovirus infection.
J. Immunol.
156
:
4746
4756
.
30
Pien
,
G. C.
,
A. R.
Satoskar
,
K.
Takeda
,
S.
Akira
,
C. A.
Biron
.
2000
.
Cutting edge: selective IL-18 requirements for induction of compartmental IFN-γ responses during viral infection.
J. Immunol.
165
:
4787
4791
.
31
Swiecki
,
M.
,
S.
Gilfillan
,
W.
Vermi
,
Y.
Wang
,
M.
Colonna
.
2010
.
Plasmacytoid dendritic cell ablation impacts early interferon responses and antiviral NK and CD8+ T cell accrual.
Immunity
33
:
955
966
.
32
Verma
,
S.
,
Q.
Wang
,
G.
Chodaczek
,
C. A.
Benedict
.
2013
.
Lymphoid-tissue stromal cells coordinate innate defense to cytomegalovirus.
J. Virol.
87
:
6201
6210
.
33
Pyzik
,
M.
,
B.
Charbonneau
,
E. M.
Gendron-Pontbriand
,
M.
Babić
,
A.
Krmpotić
,
S.
Jonjić
,
S. M.
Vidal
.
2011
.
Distinct MHC class I–dependent NK cell–activating receptors control cytomegalovirus infection in different mouse strains.
J. Exp. Med.
208
:
1105
1117
.
34
Gillespie
,
A.
,
H.
Lee
,
C.
Robertson
,
M.
Cabot
,
M. G.
Brown
.
2017
.
Genome-wide exome analysis of Cmv5-disparate mouse strains that differ in host resistance to murine cytomegalovirus infection.
G3 (Bethesda).
7
:
1979
1984
.
35
Lisnić
,
B.
,
V. J.
Lisnić
,
S.
Jonjić
.
2015
.
NK cell interplay with cytomegaloviruses.
Curr. Opin. Virol.
15
:
9
18
.
36
Nash
,
W. T.
,
J.
Teoh
,
H.
Wei
,
A.
Gamache
,
M. G.
Brown
.
2014
.
Know thyself: NK-cell inhibitory receptors prompt self-tolerance, education, and viral control.
Front. Immunol.
5
:
175
.
37
Daniels
,
K. A.
,
G.
Devora
,
W. C.
Lai
,
C. L.
O’Donnell
,
M.
Bennett
,
R. M.
Welsh
.
2001
.
Murine cytomegalovirus is regulated by a discrete subset of natural killer cells reactive with monoclonal antibody to Ly49H.
J. Exp. Med.
194
:
29
44
.
38
Smith
,
H. R. C.
,
J. W.
Heusel
,
I. K.
Mehta
,
S.
Kim
,
B. G.
Dorner
,
O. V.
Naidenko
,
K.
Iizuka
,
H.
Furukawa
,
D. L.
Beckman
,
J. T.
Pingel
, et al
.
2002
.
Recognition of a virus-encoded ligand by a natural killer cell activation receptor.
Proc. Natl. Acad. Sci. USA
99
:
8826
8831
.
39
Arase
,
H.
,
E. S.
Mocarski
,
A. E.
Campbell
,
A. B.
Hill
,
L. L.
Lanier
.
2002
.
Direct recognition of cytomegalovirus by activating and inhibitory NK cell receptors.
Science
296
:
1323
1326
.
40
Brown
,
M. G.
,
A. O.
Dokun
,
J. W.
Heusel
,
H. R.
Smith
,
D. L.
Beckman
,
E. A.
Blattenberger
,
C. E.
Dubbelde
,
L. R.
Stone
,
A. A.
Scalzo
,
W. M.
Yokoyama
.
2001
.
Vital involvement of a natural killer cell activation receptor in resistance to viral infection.
Science
292
:
934
937
.
41
Lee
,
S. H.
,
S.
Girard
,
D.
Macina
,
M.
Busà
,
A.
Zafer
,
A.
Belouchi
,
P.
Gros
,
S. M.
Vidal
.
2001
.
Susceptibility to mouse cytomegalovirus is associated with deletion of an activating natural killer cell receptor of the C-type lectin superfamily.
Nat. Genet.
28
:
42
45
.
42
Scalzo
,
A. A.
,
N. A.
Fitzgerald
,
A.
Simmons
,
A. B.
La Vista
,
G. R.
Shellam
.
1990
.
Cmv-1, a genetic locus that controls murine cytomegalovirus replication in the spleen.
J. Exp. Med.
171
:
1469
1483
.
43
Dokun
,
A. O.
,
S.
Kim
,
H. R.
Smith
,
H. S.
Kang
,
D. T.
Chu
,
W. M.
Yokoyama
.
2001
.
Specific and nonspecific NK cell activation during virus infection.
Nat. Immunol.
2
:
951
956
.
44
Geurs
,
T. L.
,
Y. M.
Zhao
,
E. B.
Hill
,
A. R.
French
.
2009
.
Ly49H engagement compensates for the absence of type I interferon signaling in stimulating NK cell proliferation during murine cytomegalovirus infection.
J. Immunol.
183
:
5830
5836
.
45
Parikh
,
B. A.
,
S. J.
Piersma
,
M. A.
Pak-Wittel
,
L.
Yang
,
R. D.
Schreiber
,
W. M.
Yokoyama
.
2015
.
Dual requirement of cytokine and activation receptor triggering for cytotoxic control of murine cytomegalovirus by NK cells.
PLoS Pathog.
11
:
e1005323
.
46
Verma
,
S.
,
A.
Loewendorf
,
Q.
Wang
,
B.
McDonald
,
A.
Redwood
,
C. A.
Benedict
.
2014
.
Inhibition of the TRAIL death receptor by CMV reveals its importance in NK cell-mediated antiviral defense.
PLoS Pathog.
10
:
e1004268
.
47
Farrell
,
H. E.
,
K.
Bruce
,
C.
Lawler
,
R. D.
Cardin
,
N. J.
Davis-Poynter
,
P. G.
Stevenson
.
2016
.
Type 1 interferons and NK cells limit murine cytomegalovirus escape from the lymph node subcapsular sinus.
PLoS Pathog.
12
:
e1006069
.
48
Podlech
,
J.
,
S.
Ebert
,
M.
Becker
,
M. J.
Reddehase
,
M.
Stassen
,
N. A. W.
Lemmermann
.
2015
.
Mast cells: innate attractors recruiting protective CD8 T cells to sites of cytomegalovirus infection.
Med Microbiol Immunol.
204
:
327
334
.
49
Becker
,
M.
,
N. A.
Lemmermann
,
S.
Ebert
,
P.
Baars
,
A.
Renzaho
,
J.
Podlech
,
M.
Stassen
,
M. J.
Reddehase
.
2015
.
Mast cells as rapid innate sensors of cytomegalovirus by TLR3/TRIF signaling-dependent and -independent mechanisms.
Cell. Mol. Immunol.
12
:
192
201
.
50
Stacey
,
M. A.
,
M.
Marsden
,
T. A.
Pham N
,
S.
Clare
,
G.
Dolton
,
G.
Stack
,
E.
Jones
,
P.
Klenerman
,
A. M.
Gallimore
,
P. R.
Taylor
, et al
.
2014
.
Neutrophils recruited by IL-22 in peripheral tissues function as TRAIL-dependent antiviral effectors against MCMV.
Cell Host Microbe
15
:
471
483
.
51
Tyznik
,
A. J.
,
S.
Verma
,
Q.
Wang
,
M.
Kronenberg
,
C. A.
Benedict
.
2014
.
Distinct requirements for activation of NKT and NK cells during viral infection.
J. Immunol.
192
:
3676
3685
.
52
Tyznik
,
A. J.
,
E.
Tupin
,
N. A.
Nagarajan
,
M. J.
Her
,
C. A.
Benedict
,
M.
Kronenberg
.
2008
.
Cutting edge: the mechanism of invariant NKT cell responses to viral danger signals.
J. Immunol.
181
:
4452
4456
.
53
Raftery
,
M. J.
,
M.
Hitzler
,
F.
Winau
,
T.
Giese
,
B.
Plachter
,
S. H. E.
Kaufmann
,
G.
Schönrich
.
2008
.
Inhibition of CD1 antigen presentation by human cytomegalovirus.
J. Virol.
82
:
4308
4319
.
54
Yuan
,
W.
,
A.
Dasgupta
,
P.
Cresswell
.
2006
.
Herpes simplex virus evades natural killer T cell recognition by suppressing CD1d recycling.
Nat. Immunol.
7
:
835
842
.
55
Brinkmann
,
M. M.
,
F.
Dağ
,
H.
Hengel
,
M.
Messerle
,
U.
Kalinke
,
L.
Čičin-Šain
.
2015
.
Cytomegalovirus immune evasion of myeloid lineage cells.
Med. Microbiol. Immunol.
204
:
367
382
.
56
Chan
,
G.
,
M. T.
Nogalski
,
E. V.
Stevenson
,
A. D.
Yurochko
.
2012
.
Human cytomegalovirus induction of a unique signalsome during viral entry into monocytes mediates distinct functional changes: a strategy for viral dissemination.
J. Leukoc. Biol.
92
:
743
752
.
57
Salazar-Mather
,
T. P.
,
K. L.
Hokeness
.
2003
.
Calling in the troops: regulation of inflammatory cell trafficking through innate cytokine/chemokine networks.
Viral Immunol.
16
:
291
306
.
58
Sinclair
,
J.
,
M.
Reeves
.
2014
.
The intimate relationship between human cytomegalovirus and the dendritic cell lineage.
Front. Microbiol.
5
:
389
.
59
Goodrum
,
F.
2015
.
Human cytomegalovirus latency: approaching the gordian knot.
Annu. Rev. Virol.
3
:
333
357
.
60
Daley-Bauer
,
L. P.
,
L. J.
Roback
,
G. M.
Wynn
,
E. S.
Mocarski
.
2014
.
Cytomegalovirus hijacks CX3CR1hi patrolling monocytes as immune-privileged vehicles for dissemination in mice.
Cell Host Microbe
15
:
351
362
.
61
O’Sullivan
,
T. E.
,
J. C.
Sun
,
L. L.
Lanier
.
2015
.
Natural killer cell memory.
Immunity
43
:
634
645
.
62
Sun
,
J. C.
,
J. N.
Beilke
,
L. L.
Lanier
.
2009
.
Adaptive immune features of natural killer cells. [Published erratum appears in 2009 Nature 457: 1168.]
Nature
457
:
557
561
.
63
Chidrawar
,
S.
,
N.
Khan
,
W.
Wei
,
A.
McLarnon
,
N.
Smith
,
L.
Nayak
,
P.
Moss
.
2009
.
Cytomegalovirus-seropositivity has a profound influence on the magnitude of major lymphoid subsets within healthy individuals.
Clin. Exp. Immunol.
155
:
423
432
.
64
Lopez-Vergès
,
S.
,
J. M.
Milush
,
B. S.
Schwartz
,
M. J.
Pando
,
J.
Jarjoura
,
V. A.
York
,
J. P.
Houchins
,
S.
Miller
,
S.-M.
Kang
,
P. J.
Norris
, et al
.
2011
.
Expansion of a unique CD57+NKG2Chi natural killer cell subset during acute human cytomegalovirus infection.
Proc. Natl. Acad. Sci. USA
108
:
14725
14732
.
65
Schlums
,
H.
,
F.
Cichocki
,
B.
Tesi
,
J.
Theorell
,
V.
Beziat
,
T. D.
Holmes
,
H.
Han
,
S. C. C.
Chiang
,
B.
Foley
,
K.
Mattsson
, et al
.
2015
.
Cytomegalovirus infection drives adaptive epigenetic diversification of NK cells with altered signaling and effector function.
Immunity
42
:
443
456
.
66
Tomasec
,
P.
,
V. M.
Braud
,
C.
Rickards
,
M. B.
Powell
,
B. P.
McSharry
,
S.
Gadola
,
V.
Cerundolo
,
L. K.
Borysiewicz
,
A. J.
McMichael
,
G. W.
Wilkinson
.
2000
.
Surface expression of HLA-E, an inhibitor of natural killer cells, enhanced by human cytomegalovirus gpUL40.
Science
287
:
1031
1033
.
67
Llano
,
M.
,
M.
Gumá
,
M.
Ortega
,
A.
Angulo
,
M.
López-Botet
.
2003
.
Differential effects of US2, US6 and US11 human cytomegalovirus proteins on HLA class Ia and HLA-E expression: impact on target susceptibility to NK cell subsets.
Eur. J. Immunol.
33
:
2744
2754
.
68
Pupuleku
,
A.
,
M.
Costa-García
,
D.
Farré
,
H.
Hengel
,
A.
Angulo
,
A.
Muntasell
,
M.
López-Botet
.
2017
.
Elusive role of the CD94/NKG2C NK cell receptor in the response to cytomegalovirus: novel experimental observations in a reporter cell system.
Front. Immunol.
8
:
1317
.
69
Hassouneh
,
F.
,
C.
Campos
,
N.
López-Sejas
,
C.
Alonso
,
R.
Tarazona
,
R.
Solana
,
A.
Pera
.
2016
.
Effect of age and latent CMV infection on CD8+ CD56+ T cells (NKT-like) frequency and functionality.
Mech. Ageing Dev.
158
:
38
45
.
70
Munks
,
M. W.
,
K. S.
Cho
,
A. K.
Pinto
,
S.
Sierro
,
P.
Klenerman
,
A. B.
Hill
.
2006
.
Four distinct patterns of memory CD8 T cell responses to chronic murine cytomegalovirus infection.
J. Immunol.
177
:
450
458
.
71
Arens
,
R.
,
P.
Wang
,
J.
Sidney
,
A.
Loewendorf
,
A.
Sette
,
S. P.
Schoenberger
,
B.
Peters
,
C. A.
Benedict
.
2008
.
Cutting edge: murine cytomegalovirus induces a polyfunctional CD4 T cell response.
J. Immunol.
180
:
6472
6476
.
72
Holtappels
,
R.
,
M. F.
Pahl-Seibert
,
D.
Thomas
,
M. J.
Reddehase
.
2000
.
Enrichment of immediate-early 1 (m123/pp89) peptide-specific CD8 T cells in a pulmonary CD62Llo memory-effector cell pool during latent murine cytomegalovirus infection of the lungs.
J. Virol.
74
:
11495
11503
.
73
Snyder
,
C. M.
,
K. S.
Cho
,
E. L.
Bonnett
,
S.
van Dommelen
,
G. R.
Shellam
,
A. B.
Hill
.
2008
.
Memory inflation during chronic viral infection is maintained by continuous production of short-lived, functional T cells.
Immunity
29
:
650
659
.
74
Quinn
,
M.
,
H.
Turula
,
M.
Tandon
,
B.
Deslouches
,
T.
Moghbeli
,
C. M.
Snyder
.
2015
.
Memory T cells specific for murine cytomegalovirus re-emerge after multiple challenges and recapitulate immunity in various adoptive transfer scenarios.
J. Immunol.
194
:
1726
1736
.
75
Redeker
,
A.
,
S. P. M.
Welten
,
R.
Arens
.
2014
.
Viral inoculum dose impacts memory T-cell inflation.
Eur. J. Immunol.
44
:
1046
1057
.
76
Delpoux
,
A.
,
R. H.
Michelini
,
S.
Verma
,
C.-Y.
Lai
,
K. D.
Omilusik
,
D. T.
Utzschneider
,
A. J.
Redwood
,
A. W.
Goldrath
,
C. A.
Benedict
,
S. M.
Hedrick
.
2018
.
Continuous activity of Foxo1 is required to prevent anergy and maintain the memory state of CD8+ T cells.
J. Exp. Med.
215
:
575
594
.
77
Cicin-Sain
,
L.
,
R.
Arens
.
2017
.
Exhaustion and inflation at antipodes of T cell responses to chronic virus infection.
Trends Microbiol
. DOI: 10.1016/j.tim.2017.11.012.
78
Wherry
,
E. J.
,
M.
Kurachi
.
2015
.
Molecular and cellular insights into T cell exhaustion.
Nat. Rev. Immunol.
15
:
486
499
.
79
Sylwester
,
A. W.
,
B. L.
Mitchell
,
J. B.
Edgar
,
C.
Taormina
,
C.
Pelte
,
F.
Ruchti
,
P. R.
Sleath
,
K. H.
Grabstein
,
N. A.
Hosken
,
F.
Kern
, et al
.
2005
.
Broadly targeted human cytomegalovirus-specific CD4+ and CD8+ T cells dominate the memory compartments of exposed subjects.
J. Exp. Med.
202
:
673
685
.
80
Khan
,
N.
,
N.
Shariff
,
M.
Cobbold
,
R.
Bruton
,
J. A.
Ainsworth
,
A. J.
Sinclair
,
L.
Nayak
,
P. A. H.
Moss
.
2002
.
Cytomegalovirus seropositivity drives the CD8 T cell repertoire toward greater clonality in healthy elderly individuals.
J. Immunol.
169
:
1984
1992
.
81
Pachnio
,
A.
,
M.
Ciaurriz
,
J.
Begum
,
N.
Lal
,
J.
Zuo
,
A.
Beggs
,
P.
Moss
.
2016
.
Cytomegalovirus infection leads to development of high frequencies of cytotoxic virus-specific CD4+ T cells targeted to vascular endothelium.
PLoS Pathog.
12
:
e1005832
.
82
Waller
,
E. C. P.
,
N.
McKinney
,
R.
Hicks
,
A. J.
Carmichael
,
J. G. P.
Sissons
,
M. R.
Wills
.
2007
.
Differential costimulation through CD137 (4-1BB) restores proliferation of human virus-specific “effector memory” (CD28 CD45RAHI) CD8+ T cells.
Blood
110
:
4360
4366
.
83
van de Berg
,
P. J. E. J.
,
A.
van Stijn
,
I. J. M.
Ten Berge
,
R. A. W.
van Lier
.
2008
.
A fingerprint left by cytomegalovirus infection in the human T cell compartment.
J. Clin. Virol.
41
:
213
217
.
84
van Leeuwen
,
E. M. M.
,
E. B. M.
Remmerswaal
,
M. T. M.
Vossen
,
A. T.
Rowshani
,
P. M. E.
Wertheim-van Dillen
,
R. A. W.
van Lier
,
I. J. M.
ten Berge
.
2004
.
Emergence of a CD4+CD28 granzyme B+, cytomegalovirus-specific T cell subset after recovery of primary cytomegalovirus infection.
J. Immunol.
173
:
1834
1841
.
85
Arens
,
R.
,
A.
Loewendorf
,
A.
Redeker
,
S.
Sierro
,
L.
Boon
,
P.
Klenerman
,
C. A.
Benedict
,
S. P.
Schoenberger
.
2011
.
Differential B7–CD28 costimulatory requirements for stable and inflationary mouse cytomegalovirus-specific memory CD8 T cell populations.
J. Immunol.
186
:
3874
3881
.
86
Humphreys
,
I. R.
,
S.-W.
Lee
,
M.
Jones
,
A.
Loewendorf
,
E.
Gostick
,
D. A.
Price
,
C. A.
Benedict
,
C. F.
Ware
,
M.
Croft
.
2010
.
Biphasic role of 4-1BB in the regulation of mouse cytomegalovirus-specific CD8+ T cells.
Eur. J. Immunol.
40
:
2762
2768
.
87
Snyder
,
C. M.
,
A.
Loewendorf
,
E. L.
Bonnett
,
M.
Croft
,
C. A.
Benedict
,
A. B.
Hill
.
2009
.
CD4+ T cell help has an epitope-dependent impact on CD8+ T cell memory inflation during murine cytomegalovirus infection.
J. Immunol.
183
:
3932
3941
.
88
Humphreys
,
I. R.
,
A.
Loewendorf
,
C.
de Trez
,
K.
Schneider
,
C. A.
Benedict
,
M. W.
Munks
,
C. F.
Ware
,
M.
Croft
.
2007
.
OX40 costimulation promotes persistence of cytomegalovirus-specific CD8 T cells: a CD4-dependent mechanism.
J. Immunol.
179
:
2195
2202
.
89
Welten
,
S. P. M.
,
A.
Redeker
,
K. L.
Franken
,
C. A.
Benedict
,
H.
Yagita
,
F. M.
Wensveen
,
J.
Borst
,
C. J. M.
Melief
,
R. A. W.
van Lier
,
K. P. J. M.
van Gisbergen
,
R.
Arens
.
2013
.
CD27-CD70 costimulation controls T cell immunity during acute and persistent cytomegalovirus infection.
J. Virol.
87
:
6851
6865
.
90
Verma
,
S.
,
D.
Weiskopf
,
A.
Gupta
,
B.
McDonald
,
B.
Peters
,
A.
Sette
,
C. A.
Benedict
.
2015
.
Cytomegalovirus-specific CD4 T cells are cytolytic and mediate vaccine protection. [Published erratum appears in 2017 J. Virol. 91: e00959-17]
J. Virol.
90
:
650
658
.
91
Nigro
,
G.
,
S. P.
Adler
.
2013
.
Hyperimmunoglobulin for prevention of congenital cytomegalovirus disease.
Clin. Infect. Dis.
57
(
Suppl. 4
):
S193
S195
.
92
Revello
,
M. G.
,
T.
Lazzarotto
,
B.
Guerra
,
A.
Spinillo
,
E.
Ferrazzi
,
A.
Kustermann
,
S.
Guaschino
,
P.
Vergani
,
T.
Todros
,
T.
Frusca
, et al
CHIP Study Group
.
2014
.
A randomized trial of hyperimmune globulin to prevent congenital cytomegalovirus.
N. Engl. J. Med.
370
:
1316
1326
.
93
Furione
,
M.
,
V.
Rognoni
,
A.
Sarasini
,
M.
Zavattoni
,
D.
Lilleri
,
G.
Gerna
,
M. G.
Revello
.
2013
.
Slow increase in IgG avidity correlates with prevention of human cytomegalovirus transmission to the fetus.
J. Med. Virol.
85
:
1960
1967
.
94
Wussow
,
F.
,
F.
Chiuppesi
,
H.
Contreras
,
D. J.
Diamond
.
2017
.
Neutralization of human cytomegalovirus entry into fibroblasts and epithelial cells.
Vaccines (Basel)
5
:
E39
.
95
Vanarsdall
,
A. L.
,
D. C.
Johnson
.
2012
.
Human cytomegalovirus entry into cells.
Curr. Opin. Virol.
2
:
37
42
.
96
Wirtz
,
N.
,
S. I.
Schader
,
R.
Holtappels
,
C. O.
Simon
,
N. A. W.
Lemmermann
,
M. J.
Reddehase
,
J.
Podlech
.
2008
.
Polyclonal cytomegalovirus-specific antibodies not only prevent virus dissemination from the portal of entry but also inhibit focal virus spread within target tissues.
Med. Microbiol. Immunol.
197
:
151
158
.
97
Vescovini
,
R.
,
A. R.
Telera
,
M.
Pedrazzoni
,
B.
Abbate
,
P.
Rossetti
,
I.
Verzicco
,
M.-C.
Arcangeletti
,
M. C.
Medici
,
A.
Calderaro
,
R.
Volpi
, et al
.
2016
.
Impact of persistent cytomegalovirus infection on dynamic changes in human immune system profile.
PLoS One
11
:
e0151965
.
98
Klenovsek
,
K.
,
F.
Weisel
,
A.
Schneider
,
U.
Appelt
,
S.
Jonjic
,
M.
Messerle
,
B.
Bradel-Tretheway
,
T. H.
Winkler
,
M.
Mach
.
2007
.
Protection from CMV infection in immunodeficient hosts by adoptive transfer of memory B cells.
Blood
110
:
3472
3479
.
99
Farrell
,
H. E.
,
C.
Lawler
,
C. S. E.
Tan
,
K.
MacDonald
,
K.
Bruce
,
M.
Mach
,
N.
Davis-Poynter
,
P. G.
Stevenson
.
2016
.
Murine cytomegalovirus exploits olfaction to enter new hosts.
MBio
7
:
e00251
e16
.
100
Schleiss
,
M. R.
2006
.
Acquisition of human cytomegalovirus infection in infants via breast milk: natural immunization or cause for concern?
Rev. Med. Virol.
16
:
73
82
.
101
Dauby
,
N.
,
D.
Sartori
,
C.
Kummert
,
S.
Lecomte
,
E.
Haelterman
,
M.-L.
Delforge
,
C.
Donner
,
M.
Mach
,
A.
Marchant
.
2016
.
Limited effector memory B-cell response to envelope glycoprotein B during primary human cytomegalovirus infection.
J. Infect. Dis.
213
:
1642
1650
.
102
Jonjić
,
S.
,
I.
Pavić
,
B.
Polić
,
I.
Crnković
,
P.
Lucin
,
U. H.
Koszinowski
.
1994
.
Antibodies are not essential for the resolution of primary cytomegalovirus infection but limit dissemination of recurrent virus.
J. Exp. Med.
179
:
1713
1717
.
103
Polić
,
B.
,
H.
Hengel
,
A.
Krmpotić
,
J.
Trgovcich
,
I.
Pavić
,
P.
Luccaronin
,
S.
Jonjić
,
U. H.
Koszinowski
.
1998
.
Hierarchical and redundant lymphocyte subset control precludes cytomegalovirus replication during latent infection.
J. Exp. Med.
188
:
1047
1054
.
104
Roizman
,
B.
,
A. E.
Sears
.
1987
.
An inquiry into the mechanisms of herpes simplex virus latency.
Annu. Rev. Microbiol.
41
:
543
571
.
105
Davison
,
A. J.
,
R.
Eberle
,
B.
Ehlers
,
G. S.
Hayward
,
D. J.
McGeoch
,
A. C.
Minson
,
P. E.
Pellett
,
B.
Roizman
,
M. J.
Studdert
,
E.
Thiry
.
2009
.
The order Herpesvirales.
Arch. Virol.
154
:
171
177
.
106
Dupont
,
L.
,
M. B.
Reeves
.
2016
.
Cytomegalovirus latency and reactivation: recent insights into an age old problem.
Rev. Med. Virol.
26
:
75
89
.
107
Mercer
,
J. A.
,
C. A.
Wiley
,
D. H.
Spector
.
1988
.
Pathogenesis of murine cytomegalovirus infection: identification of infected cells in the spleen during acute and latent infections.
J. Virol.
62
:
987
997
.
108
Seckert
,
C. K.
,
A.
Renzaho
,
H. M.
Tervo
,
C.
Krause
,
P.
Deegen
,
B.
Kühnapfel
,
M. J.
Reddehase
,
N. K. A.
Grzimek
.
2009
.
Liver sinusoidal endothelial cells are a site of murine cytomegalovirus latency and reactivation.
J. Virol.
83
:
8869
8884
.
109
Cardin
,
R. D.
,
G. C.
Schaefer
,
J. R.
Allen
,
N. J.
Davis-Poynter
,
H. E.
Farrell
.
2009
.
The M33 chemokine receptor homolog of murine cytomegalovirus exhibits a differential tissue-specific role during in vivo replication and latency.
J. Virol.
83
:
7590
7601
.
110
Torti
,
N.
,
S. M.
Walton
,
T.
Brocker
,
T.
Rülicke
,
A.
Oxenius
.
2011
.
Non-hematopoietic cells in lymph nodes drive memory CD8 T cell inflation during murine cytomegalovirus infection.
PLoS Pathog.
7
:
e1002313
.
111
Seckert
,
C. K.
,
S. I.
Schader
,
S.
Ebert
,
D.
Thomas
,
K.
Freitag
,
A.
Renzaho
,
J.
Podlech
,
M. J.
Reddehase
,
R.
Holtappels
.
2011
.
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.
92
:
1994
2005
.
112
Snyder
,
C. M.
,
K. S.
Cho
,
E. L.
Bonnett
,
J. E.
Allan
,
A. B.
Hill
.
2011
.
Sustained CD8+ T cell memory inflation after infection with a single-cycle cytomegalovirus.
PLoS Pathog.
7
:
e1002295
.
113
Benedict
,
C. A.
,
T. A.
Banks
,
L.
Senderowicz
,
M.
Ko
,
W. J.
Britt
,
A.
Angulo
,
P.
Ghazal
,
C. F.
Ware
.
2001
.
Lymphotoxins and cytomegalovirus cooperatively induce interferon-beta, establishing host-virus détente.
Immunity
15
:
617
626
.
114
Dağ
,
F.
,
L.
Dölken
,
J.
Holzki
,
A.
Drabig
,
A.
Weingärtner
,
J.
Schwerk
,
S.
Lienenklaus
,
I.
Conte
,
R.
Geffers
,
C.
Davenport
, et al
.
2014
.
Reversible silencing of cytomegalovirus genomes by type I interferon governs virus latency.
PLoS Pathog.
10
:
e1003962
.
115
Penkert
,
R. R.
,
R. F.
Kalejta
.
2011
.
Tegument protein control of latent herpesvirus establishment and animation.
Herpesviridae
2
:
3
.
116
Cheng
,
S.
,
K.
Caviness
,
J.
Buehler
,
M.
Smithey
,
J.
Nikolich-Žugich
,
F.
Goodrum
.
2017
.
Transcriptome-wide characterization of human cytomegalovirus in natural infection and experimental latency.
Proc. Natl. Acad. Sci. USA
114
:
E10586
E10595
.
117
Poole
,
E.
,
J.
Sinclair
.
2015
.
Sleepless latency of human cytomegalovirus.
Med. Microbiol. Immunol.
204
:
421
429
.
118
Rossetto
,
C. C.
,
M.
Tarrant-Elorza
,
G. S.
Pari
.
2013
.
Cis and trans acting factors involved in human cytomegalovirus experimental and natural latent infection of CD14+ monocytes and CD34+ cells.
PLoS Pathog.
9
:
e1003366
.
119
Mason
,
G. M.
,
S.
Jackson
,
G.
Okecha
,
E.
Poole
,
J. G. P.
Sissons
,
J.
Sinclair
,
M. R.
Wills
.
2013
.
Human cytomegalovirus latency-associated proteins elicit immune-suppressive IL-10 producing CD4+ T cells.
PLoS Pathog.
9
:
e1003635
.
120
Poole
,
E.
,
A.
Walther
,
K.
Raven
,
C. A.
Benedict
,
G. M.
Mason
,
J.
Sinclair
.
2013
.
The myeloid transcription factor GATA-2 regulates the viral UL144 gene during human cytomegalovirus latency in an isolate-specific manner.
J. Virol.
87
:
4261
4271
.
121
Görzer
,
I.
,
H.
Kerschner
,
M.
Redlberger-Fritz
,
E.
Puchhammer-Stöckl
.
2010
.
Human cytomegalovirus (HCMV) genotype populations in immunocompetent individuals during primary HCMV infection.
J. Clin. Virol.
48
:
100
103
.
122
Trgovcich
,
J.
,
M.
Kincaid
,
A.
Thomas
,
M.
Griessl
,
P.
Zimmerman
,
V.
Dwivedi
,
V.
Bergdall
,
P.
Klenerman
,
C. H.
Cook
.
2016
.
Cytomegalovirus reinfections stimulate CD8 T-memory inflation.
PLoS One
11
:
e0167097
.
123
Wilkinson
,
G. W. G.
,
A. J.
Davison
,
P.
Tomasec
,
C. A.
Fielding
,
R.
Aicheler
,
I.
Murrell
,
S.
Seirafian
,
E. C. Y.
Wang
,
M.
Weekes
,
P. J.
Lehner
, et al
.
2015
.
Human cytomegalovirus: taking the strain.
Med. Microbiol. Immunol.
204
:
273
284
.
124
Früh
,
K.
,
L.
Picker
.
2017
.
CD8+ T cell programming by cytomegalovirus vectors: applications in prophylactic and therapeutic vaccination.
Curr. Opin. Immunol.
47
:
52
56
.
125
Hansen
,
S. G.
,
C. J.
Powers
,
R.
Richards
,
A. B.
Ventura
,
J. C.
Ford
,
D.
Siess
,
M. K.
Axthelm
,
J. A.
Nelson
,
M. A.
Jarvis
,
L. J.
Picker
,
K.
Früh
.
2010
.
Evasion of CD8+ T cells is critical for superinfection by cytomegalovirus.
Science
328
:
102
106
.
126
Khairallah
,
C.
,
S.
Netzer
,
A.
Villacreces
,
M.
Juzan
,
B.
Rousseau
,
S.
Dulanto
,
A.
Giese
,
P.
Costet
,
V.
Praloran
,
J.-F.
Moreau
, et al
.
2015
.
γδ T cells confer protection against murine cytomegalovirus (MCMV).
PLoS Pathog.
11
:
e1004702
.
127
Lafarge
,
X.
,
P.
Merville
,
M. C.
Cazin
,
F.
Bergé
,
L.
Potaux
,
J. F.
Moreau
,
J.
Déchanet-Merville
.
2001
.
Cytomegalovirus infection in transplant recipients resolves when circulating γδ T lymphocytes expand, suggesting a protective antiviral role.
J. Infect. Dis.
184
:
533
541
.
128
Pitard
,
V.
,
D.
Roumanes
,
X.
Lafarge
,
L.
Couzi
,
I.
Garrigue
,
M. E.
Lafon
,
P.
Merville
,
J. F.
Moreau
,
J.
Déchanet-Merville
.
2008
.
Long-term expansion of effector/memory Vδ2-γδ T cells is a specific blood signature of CMV infection.
Blood
112
:
1317
1324
.
129
Yu
,
D.
,
M. C.
Silva
,
T.
Shenk
.
2003
.
Functional map of human cytomegalovirus AD169 defined by global mutational analysis.
Proc. Natl. Acad. Sci. USA
100
:
12396
12401
.
130
Jackson
,
S. E.
,
A.
Redeker
,
R.
Arens
,
D.
van Baarle
,
S. P. H.
Berg
,
C. A.
Benedict
,
L.
Cicin-Sain
,
A. B.
Hill
,
M. R.
Wills
.
2017
.
CMV immune evasion and manipulation of the immune system with aging.
Geroscience
39
:
273
291
.
131
Hook
,
L.
,
M.
Hancock
,
I.
Landais
,
R.
Grabski
,
W.
Britt
,
J. A.
Nelson
.
2014
.
Cytomegalovirus microRNAs.
Curr. Opin. Virol.
7
:
40
46
.
132
Kaiser
,
W. J.
,
J. W.
Upton
,
E. S.
Mocarski
.
2013
.
Viral modulation of programmed necrosis.
Curr. Opin. Virol.
3
:
296
306
.
133
Lemmermann
,
N. A. W.
,
A.
Fink
,
J.
Podlech
,
S.
Ebert
,
V.
Wilhelmi
,
V.
Böhm
,
R.
Holtappels
,
M. J.
Reddehase
.
2012
.
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.
201
:
497
512
.
134
Alexandre
,
Y. O.
,
C. D.
Cocita
,
S.
Ghilas
,
M.
Dalod
.
2014
.
Deciphering the role of DC subsets in MCMV infection to better understand immune protection against viral infections.
Front. Microbiol.
5
:
378
.
135
Marshall
,
E. E.
,
A. P.
Geballe
.
2009
.
Multifaceted evasion of the interferon response by cytomegalovirus.
J. Interferon Cytokine Res.
29
:
609
619
.
136
Tomasec
,
P.
,
E. C. Y.
Wang
,
A. J.
Davison
,
B.
Vojtesek
,
M.
Armstrong
,
C.
Griffin
,
B. P.
McSharry
,
R. J.
Morris
,
S.
Llewellyn-Lacey
,
C.
Rickards
, et al
.
2005
.
Downregulation of natural killer cell-activating ligand CD155 by human cytomegalovirus UL141.
Nat. Immunol.
6
:
181
188
.
137
Smith
,
W.
,
P.
Tomasec
,
R.
Aicheler
,
A.
Loewendorf
,
I.
Nemčovičová
,
E. C. Y.
Wang
,
R. J.
Stanton
,
M.
Macauley
,
P.
Norris
,
L.
Willen
, et al
.
2013
.
Human cytomegalovirus glycoprotein UL141 targets the TRAIL death receptors to thwart host innate antiviral defenses.
Cell Host Microbe
13
:
324
335
.
138
Cosman
,
D.
,
N.
Fanger
,
L.
Borges
,
M.
Kubin
,
W.
Chin
,
L.
Peterson
,
M. L.
Hsu
.
1997
.
A novel immunoglobulin superfamily receptor for cellular and viral MHC class I molecules.
Immunity
7
:
273
282
.
139
Halenius
,
A.
,
C.
Gerke
,
H.
Hengel
.
2015
.
Classical and non-classical MHC I molecule manipulation by human cytomegalovirus: so many targets—but how many arrows in the quiver?
Cell. Mol. Immunol.
12
:
139
153
.
140
Loewendorf
,
A.
,
C.
Krüger
,
E. M.
Borst
,
M.
Wagner
,
U.
Just
,
M.
Messerle
.
2004
.
Identification of a mouse cytomegalovirus gene selectively targeting CD86 expression on antigen-presenting cells.
J. Virol.
78
:
13062
13071
.
141
Mintern
,
J. D.
,
E. J.
Klemm
,
M.
Wagner
,
M. E.
Paquet
,
M. D.
Napier
,
Y. M.
Kim
,
U. H.
Koszinowski
,
H. L.
Ploegh
.
2006
.
Viral interference with B7-1 costimulation: a new role for murine cytomegalovirus fc receptor-1.
J. Immunol.
177
:
8422
8431
.
142
Benedict
,
C. A.
,
A.
Loewendorf
,
Z.
Garcia
,
B. R.
Blazar
,
E. M.
Janssen
.
2008
.
Dendritic cell programming by cytomegalovirus stunts naive T cell responses via the PD-L1/PD-1 pathway.
J. Immunol.
180
:
4836
4847
.
143
Loewendorf
,
A. I.
,
L.
Steinbrueck
,
C.
Peter
,
A.
Busche
,
C. A.
Benedict
,
P. C.
Kay-Jackson
.
2011
.
The mouse cytomegalovirus glycoprotein m155 inhibits CD40 expression and restricts CD4 T cell responses.
J. Virol.
85
:
5208
5212
.
144
Arens
,
R.
,
A.
Loewendorf
,
M. J.
Her
,
K.
Schneider-Ohrum
,
G. R.
Shellam
,
E.
Janssen
,
C. F.
Ware
,
S. P.
Schoenberger
,
C. A.
Benedict
.
2011
.
B7-mediated costimulation of CD4 T cells constrains cytomegalovirus persistence.
J. Virol.
85
:
390
396
.
145
Raftery
,
M. J.
,
M.
Schwab
,
S. M.
Eibert
,
Y.
Samstag
,
H.
Walczak
,
G.
Schönrich
.
2001
.
Targeting the function of mature dendritic cells by human cytomegalovirus: a multilayered viral defense strategy.
Immunity
15
:
997
1009
.
146
Benedict
,
C. A.
,
K. D.
Butrovich
,
N. S.
Lurain
,
J.
Corbeil
,
I.
Rooney
,
P.
Schneider
,
J.
Tschopp
,
C. F.
Ware
.
1999
.
Cutting edge: a novel viral TNF receptor superfamily member in virulent strains of human cytomegalovirus.
J. Immunol.
162
:
6967
6970
.
147
Cheung
,
T. C.
,
I. R.
Humphreys
,
K. G.
Potter
,
P. S.
Norris
,
H. M.
Shumway
,
B. R.
Tran
,
G.
Patterson
,
R.
Jean-Jacques
,
M.
Yoon
,
P. G.
Spear
, et al
.
2005
.
Evolutionarily divergent herpesviruses modulate T cell activation by targeting the herpesvirus entry mediator cosignaling pathway.
Proc. Natl. Acad. Sci. USA
102
:
13218
13223
.
148
Šedý
,
J. R.
,
M. O.
Balmert
,
B. C.
Ware
,
W.
Smith
,
I.
Nemčovičova
,
P. S.
Norris
,
B. R.
Miller
,
D.
Aivazian
,
C. F.
Ware
.
2017
.
A herpesvirus entry mediator mutein with selective agonist action for the inhibitory receptor B and T lymphocyte attenuator.
J. Biol. Chem.
292
:
21060
21070
.
149
Poole
,
E.
,
C. A.
King
,
J. H.
Sinclair
,
A.
Alcami
.
2006
.
The UL144 gene product of human cytomegalovirus activates NFκB via a TRAF6-dependent mechanism.
EMBO J.
25
:
4390
4399
.
150
Slobedman
,
B.
,
P. A.
Barry
,
J. V.
Spencer
,
S.
Avdic
,
A.
Abendroth
.
2009
.
Virus-encoded homologs of cellular interleukin-10 and their control of host immune function.
J. Virol.
83
:
9618
9629
.
151
Jackson
,
S. E.
,
G. X.
Sedikides
,
G. M.
Mason
,
G.
Okecha
,
M. R.
Wills
.
2017
.
HCMV specific CD4+ T cells are poly-functional and can respond to HCMV infected dendritic cells in vitro.
J. Virol
.
152
Humphreys
,
I. R.
,
C.
de Trez
,
A.
Kinkade
,
C. A.
Benedict
,
M.
Croft
,
C. F.
Ware
.
2007
.
Cytomegalovirus exploits IL-10–mediated immune regulation in the salivary glands.
J. Exp. Med.
204
:
1217
1225
.
153
Clement
,
M.
,
M.
Marsden
,
M. A.
Stacey
,
J.
Abdul-Karim
,
S.
Gimeno Brias
,
D.
Costa Bento
,
M. J.
Scurr
,
P.
Ghazal
,
C. T.
Weaver
,
G.
Carlesso
, et al
.
2016
.
Cytomegalovirus-specific IL-10-producing CD4+ T cells are governed by type-I IFN-induced IL-27 and promote virus persistence.
PLoS Pathog.
12
:
e1006050
.
154
McSharry
,
B. P.
,
S.
Avdic
,
B.
Slobedman
.
2012
.
Human cytomegalovirus encoded homologs of cytokines, chemokines and their receptors: roles in immunomodulation.
Viruses
4
:
2448
2470
.
155
Plotkin
,
S.
2015
.
The history of vaccination against cytomegalovirus.
Med. Microbiol. Immunol.
204
:
247
254
.
156
Savva
,
G. M.
,
A.
Pachnio
,
B.
Kaul
,
K.
Morgan
,
F. A.
Huppert
,
C.
Brayne
,
P. A. H.
Moss
;
Medical Research Council Cognitive Function and Ageing Study
.
2013
.
Cytomegalovirus infection is associated with increased mortality in the older population.
Aging Cell
12
:
381
387
.
157
Simanek
,
A. M.
,
J. B.
Dowd
,
G.
Pawelec
,
D.
Melzer
,
A.
Dutta
,
A. E.
Aiello
.
2011
.
Seropositivity to cytomegalovirus, inflammation, all-cause and cardiovascular disease-related mortality in the United States.
PLoS One
6
:
e16103
.
158
Caposio
,
P.
,
S. L.
Orloff
,
D. N.
Streblow
.
2011
.
The role of cytomegalovirus in angiogenesis.
Virus Res.
157
:
204
211
.
159
Wang
,
H.
,
G.
Peng
,
J.
Bai
,
B.
He
,
K.
Huang
,
X.
Hu
,
D.
Liu
.
2017
.
Cytomegalovirus infection and relative risk of cardiovascular disease (ischemic heart disease, stroke, and cardiovascular death): a meta-analysis of prospective studies up to 2016.
J. Am. Heart Assoc.
6
:
e005025
.
160
Bolovan-Fritts
,
C. A.
,
S. A.
Spector
.
2008
.
Endothelial damage from cytomegalovirus-specific host immune response can be prevented by targeted disruption of fractalkine-CX3CR1 interaction.
Blood
111
:
175
182
.
161
van de Berg
,
P. J. E. J.
,
S. L.
Yong
,
E. B. M.
Remmerswaal
,
R. A. W.
van Lier
,
I. J. M.
ten Berge
.
2012
.
Cytomegalovirus-induced effector T cells cause endothelial cell damage.
Clin. Vaccine Immunol.
19
:
772
779
.
162
Halenius
,
A.
,
H.
Hengel
.
2014
.
Human cytomegalovirus and autoimmune disease.
BioMed Res. Int.
2014
:
472978
.
163
Nikolich-Zugich
,
J.
,
F.
Goodrum
,
K.
Knox
,
M. J.
Smithey
.
2017
.
Known unknowns: how might the persistent herpesvirome shape immunity and aging?
Curr. Opin. Immunol.
48
:
23
30
.
164
Kaczorowski
,
K. J.
,
K.
Shekhar
,
D.
Nkulikiyimfura
,
C. L.
Dekker
,
H.
Maecker
,
M. M.
Davis
,
A. K.
Chakraborty
,
P.
Brodin
.
2017
.
Continuous immunotypes describe human immune variation and predict diverse responses.
Proc. Natl. Acad. Sci. USA
114
:
E6097
E6106
.
165
Pawelec
,
G.
,
A.
Akbar
,
C.
Caruso
,
R.
Effros
,
B.
Grubeck-Loebenstein
,
A.
Wikby
.
2004
.
Is immunosenescence infectious?
Trends Immunol.
25
:
406
410
.
166
Hadrup
,
S. R.
,
J.
Strindhall
,
T.
Køllgaard
,
T.
Seremet
,
B.
Johansson
,
G.
Pawelec
,
P.
thor Straten
,
A.
Wikby
.
2006
.
Longitudinal studies of clonally expanded CD8 T cells reveal a repertoire shrinkage predicting mortality and an increased number of dysfunctional cytomegalovirus-specific T cells in the very elderly.
J. Immunol.
176
:
2645
2653
.
167
Furman
,
D.
,
V.
Jojic
,
S.
Sharma
,
S. S.
Shen-Orr
,
C. J. L.
Angel
,
S.
Onengut-Gumuscu
,
B. A.
Kidd
,
H. T.
Maecker
,
P.
Concannon
,
C. L.
Dekker
, et al
.
2015
.
Cytomegalovirus infection enhances the immune response to influenza.
Sci. Transl. Med.
7
:
281ra43
.
168
Söderberg-Nauclér
,
C.
2006
.
Does cytomegalovirus play a causative role in the development of various inflammatory diseases and cancer?
J. Intern. Med.
259
:
219
246
.
169
Dziurzynski
,
K.
,
S. M.
Chang
,
A. B.
Heimberger
,
R. F.
Kalejta
,
S. R.
McGregor Dallas
,
M.
Smit
,
L.
Soroceanu
,
C. S.
Cobbs
;
HCMV and Gliomas Symposium
.
2012
.
Consensus on the role of human cytomegalovirus in glioblastoma.
Neuro Oncol.
14
:
246
255
.
170
Foster
,
H.
,
I. V.
Ulasov
,
C. S.
Cobbs
.
2017
.
Human cytomegalovirus-mediated immunomodulation: effects on glioblastoma progression.
Biochim Biophys Acta.
1868
:
273
276
.
171
Crough
,
T.
,
L.
Beagley
,
C.
Smith
,
L.
Jones
,
D. G.
Walker
,
R.
Khanna
.
2012
.
Ex vivo functional analysis, expansion and adoptive transfer of cytomegalovirus-specific T-cells in patients with glioblastoma multiforme.
Immunol. Cell Biol.
90
:
872
880
.

The authors have no financial conflicts of interest.