Next Article in Journal
Detection of Dengue Virus 1 and Mammalian Orthoreovirus 3, with Novel Reassortments, in a South African Family Returning from Thailand, 2017
Previous Article in Journal
The Altered Neonatal CD8+ T Cell Immunodominance Hierarchy during Influenza Virus Infection Impacts Peptide Vaccination
Previous Article in Special Issue
Parainfluenza Virus 5 V Protein Blocks Interferon Gamma-Mediated Upregulation of NK Cell Inhibitory Ligands and Improves NK Cell Killing of Neuroblastoma Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

No Time to Die: How Cytomegaloviruses Suppress Apoptosis, Necroptosis, and Pyroptosis

Leibniz Institute of Virology (LIV), 20251 Hamburg, Germany
*
Author to whom correspondence should be addressed.
Viruses 2024, 16(8), 1272; https://doi.org/10.3390/v16081272
Submission received: 5 July 2024 / Revised: 31 July 2024 / Accepted: 7 August 2024 / Published: 9 August 2024
(This article belongs to the Special Issue Viruses 2024 - A World of Viruses)

Abstract

:
Viruses are obligate intracellular pathogens as their replication depends on the metabolism of the host cell. The induction of cellular suicide, known as programmed cell death (PCD), has the potential to hinder viral replication and act as a first line of defense against viral pathogens. Apoptosis, necroptosis, and pyroptosis are three important PCD modalities. Different signaling pathways are involved in their execution, and they also differ in their ability to cause inflammation. Cytomegaloviruses (CMV), beta-herpesviruses with large double-stranded DNA genomes, encode a great variety of immune evasion genes, including several cell death suppressors. While CMV inhibitors of apoptosis and necroptosis have been known and studied for years, the first pyroptosis inhibitor has been identified and characterized only recently. Here, we describe how human and murine CMV interfere with apoptosis, necroptosis, and pyroptosis signaling pathways. We also discuss the importance of the different PCD forms and their viral inhibitors for the containment of viral replication and spread in vivo.

1. Cytomegalovirus Infection and Pathogenesis

Cytomegaloviruses (CMVs) are large, enveloped, double-stranded DNA viruses that infect mammals, including humans, in a species-specific manner. All CMVs are related members of the Betaherpesvirinae that have diverged as they coevolved with their hosts. They have adapted to subvert the immune system of their specific host, leading to widespread distribution and long-term coexistence [1,2,3].
Human cytomegalovirus (HCMV) is an opportunistic pathogen responsible for significant morbidity and mortality in immunocompromised patients such as hematopoietic stem cells and solid-organ transplant recipients [4]. HCMV infection can also be transmitted vertically from the mother to the unborn child, where it can cause long-term neurological sequelae. In fact, congenital HCMV infection is the most frequent infectious cause of birth defects worldwide. Due to the species-specificity of the CMVs, HCMV infection and pathogenesis cannot be studied in animals [5]. Therefore, CMVs of mice, rats, guinea pigs, and rhesus macaques are used to study CMV pathogenesis in its natural host organism. Although the rhesus macaque CMV and the guinea pig CMV models are especially well suited to study congenital HCMV infection, murine CMV (MCMV) has emerged as an excellent animal model where the interplay of viral modulators and host defense pathways can be investigated by using genetic mutants. Therefore, MCMV infection has been used to elucidate numerous mechanisms of infection, pathogenesis, and immune response to CMV in vivo.
The CMV genomes are 200–240 kbp linear double-stranded DNA molecules that encode more than 200 proteins. Most of them are involved in the modulation of virus–host interaction and the subversion of innate and adaptive immunity [6,7]. HCMV exhibits a broad tropism, infecting several cell types, with one of its pivotal features being its ability to induce different infection outcomes depending on the infected cell. Lytic infection occurs mainly in epithelial, endothelial, fibroblast, and smooth muscle cells and results in the release of infectious viral particles and spread to the neighboring cells [8]. For HCMV, hematopoietic cells of the myeloid lineage are a major site of latency. Reactivation from latent reservoirs can lead to recurrent disease, particularly in immunocompromised individuals [4]. After primary infection, HCMV induces innate immune responses involving the release of pro-inflammatory cytokines and interferons (IFNs) and activation of natural killer (NK) cells. CMV infection also triggers strong humoral and cell-mediated adaptive immune responses [9]. Like all herpesviruses, CMVs remain latent within their respective hosts for life, being controlled but not cleared by the antiviral immune response.
Innate immunity acts as the host’s first line of defense against CMV infection, activating different stress signals and defensive pathways. In particular, the activation of programmed cell death pathways provides a quick response against pathogens and results in the death and removal of infected cells. Because CMVs are highly dependent on the viability of infected cells to complete their viral life cycle, these viruses have evolved various mechanisms to suppress cell death starting from the early period after infection [10]. Here, we provide an overview of the molecular events triggered by CMV that lead to the induction of apoptosis, necroptosis, or pyroptosis and describe the evasion strategies used by these viruses to avoid or delay these responses. We also review the biological importance of viral cell death inhibition for infection and pathogenesis in the mouse model.

2. Overview of Programmed Cell Death Pathways Activated during CMV Infection

The evolution of CMVs with their natural hosts is a complex interplay that has occurred over millions of years, where the CMVs adapted to subvert the immune system and establish persistent infections without causing severe symptoms in immunocompetent hosts. This homeostasis has been shaped by host–pathogen interactions that include host recognition of the virus infection, while the virus evolved to evade the host’s immune responses.
Cell death can be traditionally classified into accidental cell death (ACD) or programmed cell death (PCD). Tightly regulated intracellular signaling pathways and their effector molecules orchestrate PCD, while ACD arises from unforeseen cellular injury. The best-studied forms of PCD are apoptosis, necroptosis, and pyroptosis, and these present very different features (Table 1) [11]. Apoptotic cell death has long been considered a critical cell death process for host defense by eliminating infected cells and preventing the spread of intracellular pathogens within the host [12]. Both physiological and pathogen-derived stimuli can induce apoptosis, and two main pathways result in its activation: the extrinsic and intrinsic pathways. In general, apoptosis leads to cell shrinkage, chromatin condensation, and cell fragmentation [12]. Apoptotic cells are rapidly cleared by phagocytic cells in tissues resulting in an inflammatory-silent form of cell death [13]. In the last two decades, other PCD forms such as necroptosis and pyroptosis have been identified as alternative death pathways that dominate under specific conditions [14,15]. Morphologically and mechanistically different from apoptosis, these PCDs are characterized by membrane rupture and initiation of a strong inflammatory response [11,16]. Although these pathways are often described as distinct signaling cascades, an increasing number of studies have revealed a substantial level of cross-talk between PCD forms leading to alternative outcomes, for instance, when caspase inhibition converts extrinsic apoptosis to necroptosis [17,18]. A complex form of PCD, termed PANoptosis, may be triggered via Z-nucleic acid binding protein 1 (ZBP1) when viruses fail to suppress cell death signaling (reviewed in [19,20]).

2.1. Inhibition of Apoptosis by CMVs

Apoptosis is the best-described form of programmed cell death, and its multifaceted functions contribute to the host’s ability to fight viral infections and maintain homeostasis. The extrinsic apoptosis pathway begins with the activation of cell-surface death receptors of the tumor necrosis factor (TNF) receptor family by their specific extracellular death ligands (Figure 1). These death receptors include TNF receptor 1 (TNFR1, activated by TNFα), FAS (activated by Fas ligand, FasL), and TNF-related apoptosis-inducing ligand (TRAIL) receptors 1 and 2 (activated by TRAIL) [21,22]. Activated FAS and TRAIL receptors recruit their adaptor protein Fas-associated death domain protein (FADD), which binds to apoptotic pro-caspase-8 or -10, and the cellular FADD-like IL-1β-converting enzyme (FLICE) inhibitor protein (cFLIP), forming the death-inducing signaling complex (DISC) [23,24,25,26]. Long and short cFLIP isoforms control the activation of the caspase cascade, promoting apoptotic cell death [27]. In contrast to Fas and TRAIL receptors, TNFR1 recruits the TNFR-associated protein with a death domain (TRADD) as an adaptor protein, which subsequently recruits FADD, TNF-associated factor-2 (TRAF2), the receptor-interacting protein kinase 1 (RIPK1) and the cellular inhibitor of apoptosis (cIAPs) [28,29,30]. After this complex is formed, FADD binds and activates caspase-8, which in turn activates the effector caspases-3/6/7 resulting in apoptotic cell death [31,32,33].
Intrinsic apoptosis (also known as the mitochondrial apoptosis pathway) is activated when intracellular stress interferes with the balance of the B-cell lymphoma 2 (BCL-2) family of pro- and anti-apoptotic proteins (Figure 1). The small “BH3-only” proteins are activated in response to stress or, in the case of BID (BCL-2 homology domain 3 (BH3)-interacting domain death agonist), connect the extrinsic with the intrinsic pathway [34,35]. They bind to anti-apoptotic proteins such as BCL-2, BCL-XL, and myeloid cell leukemia 1 (MCL-1), which control the activity of the pro-apoptotic BCL-2-associated X protein (BAX) and BCL-2 antagonist/killer (BAK) [36,37,38,39]. When cellular stress is sensed by cells, the anti-apoptotic proteins are inhibited, resulting in the release of BAX and BAK. These proteins then oligomerize and cause mitochondrial outer membrane permeabilization (MOMP) and the release of cytochrome c (cytC), SMAC/Diablo, and HtrA2/Omi [40,41,42,43]. Mitochondrial release of SMAC/Diablo and HtrA2/Omi overcomes the activity of inhibitor of apoptosis proteins (IAPs). In the cytosol, cytC binds to apoptotic protease-activating factor 1 (Apaf-1), forming an oligomeric complex [44,45]. This complex, also known as apoptosome, promotes autocatalytic activation of caspase-9, which subsequently activates the same effector caspases as caspase-8 in the extrinsic apoptotic pathway [46].
Over the years, the mechanisms of CMV inhibition of apoptosis have been studied in detail (Figure 1). In the course of CMV infection, multiple cellular processes can promote the release of pro-apoptotic stimuli. For instance, large amounts of viral glycoproteins are synthesized during viral infection. Unfolded or misfolded proteins in the secretory pathway can activate ER stress and initiate an unfolded protein response (UPR) [47]. If not resolved by counter-regulatory mechanisms, the UPR leads to the induction of apoptosis (in that case termed “terminal UPR”). During HCMV infection, the viral protein UL38 plays a key role in inhibiting apoptosis induced by ER stress (Figure 1). UL38 induces the accumulation of activating transcription factor 4 (ATF4) and suppresses the activation of c-Jun N-terminal kinase (JNK), a protein known to inhibit the action of anti-apoptotic proteins like BCL-2 and BIM [48,49,50]. Perturbation of mitochondrial metabolism by viral infection can also result in increased levels of oxidative stress and apoptosis. During the early stages of infection, the β2.7 non-coding RNA is produced by HCMV in large amounts, helping it counteract oxidative stress and maintain the mitochondrial membrane potential [51]. Because this RNA can be produced in high amounts with no need for translation, it has been proposed that this strategy allows HCMV to target mitochondria function quickly throughout the course of infection [51].
The CMVs are also known to encode proteins that directly interfere with cell surface expression of death receptors. The MCMV m166 viral gene product specifically inhibits TRAIL expression and is critical for replication in vivo [52]. Because HCMV glycoprotein UL141 is also capable of binding to TRAIL death receptors [53], restriction of the TRAIL/TRAIL-DR pathway seems to play a key role in the CMV life cycle. Direct control of death receptor signaling also contributes to effective viral pathogenesis. CMVs encode the viral inhibitor of caspase-8 activation (vICA), which directly binds to caspase-8, inhibiting its activity (Figure 1). The HCMV vICA is encoded by open reading frame (ORF) UL36, and its homolog in MCMV is encoded by ORF M36 (Figure 1). These proteins directly interact with the death domain of pro-caspase-8 to block caspase-8 activation, thereby reducing apoptotic cell death [54,55]. Caspase-8 suppression by M36 proteins was shown to be necessary for successful infection of monocyte-derived cells and control of viral dissemination [56]. Interestingly, a mutant MCMV expressing UL36 or overexpressing a dominant-negative FADD instead of M36 fully rescued the replication defect of the MCMV M36 deletion mutant in macrophages, showing that this form of apoptosis inhibition is conserved among the CMVs [57,58]. HCMV Immediate Early 2 protein (IE2) can also interfere with apoptosis signaling by inducing the expression of c-FLIP in human retinal pigment epithelial cells, an apoptotic inhibitor downstream of the FasL/FAS cascade [59,60].
MOMP is a central event in the intrinsic apoptotic pathway, and CMVs are known to encode viral inhibitors of pro-apoptotic BCL-2 family proteins. The viral mitochondria-localized inhibitor (vMIA), encoded by HCMV ORF UL37 exon 1 (UL37x1), binds and sequesters both BAX and BAK, thereby preventing MOMP and apoptosis [61,62,63,64] (Figure 1). In human fibroblasts, UL37x1 also inhibits mitochondrial serine protease HtrA2/Omi-dependent cell death, which is independent of apoptosis [65]. Although UL37x1 has an important anti-apoptotic role, it has also been shown to inhibit antiviral signaling in a step downstream of mitochondrial antiviral-signaling protein (MAVS) [66]. MCMV, by contrast, encodes two separate proteins to inhibit BAX and BAK. MCMV ORF m38.5, localized to the mitochondria, encodes a protein similar to HCMV vMIA that specifically inhibits BAX [64,67,68,69]. Later, a second inhibitor, encoded by ORF m41.1, was shown to associate with BAK at the mitochondrial membrane, acting as a viral inhibitor of BAK oligomerization (vIBO) [70] (Figure 1). Similarly, guinea pig CMV encodes specific inhibitors for BAX and BAK [71,72]. Different cells infected with MCMV mutants lacking m38.5 or m41.1 were shown to be sensitive to apoptosis induction [67,69,70], and MCMV replication in leucocytes was reduced when one of these gene products was absent [73,74]. In vivo, replication of an m41.1-deficient MCMV was attenuated in the liver and lungs of infected mice [73,74,75]. However, the same phenotype was not observed in mice infected with MCMV deleted of m38.5 [69]. Although single mutant studies disrupting m38.5 or m41.1 have shown that these are important for MCMV replication, the combined effect of both proteins for replication and pathogenesis was studied only recently. Combined suppression of BAX and BAK activation by m38.5 and m41.1 was shown to prevent intrinsic PCD, limit mitochondrial HtrA2/Omi signaling, and impair innate inflammation, which benefit sustained virus replication [76]. In vivo, MCMV dissemination to or replication to high titers in the salivary glands was compromised in mice infected with an MCMV mutant lacking both m38.5 and m41.1 [76].

2.2. Inhibition of Necroptosis by CMVs

Necroptosis can be induced by the activation of death receptors such as FAS and TNFR1 or by stimulation of the pattern recognition receptors Toll-like receptor 3 (TLR3) and TLR4 or the intracellular sensor ZBP1, also known as DAI or DLM1 [77]. In contrast to apoptosis, necroptosis leads to the release of intracellular contents and inflammation and occurs without caspase activation (Figure 2).
While TNFR signaling triggers the extrinsic apoptotic pathway, it can also induce necroptosis when the apoptotic initiator caspase-8 is inhibited [78,79]. When caspase-8 is inhibited, RIPK1 recruits RIPK3 through an RIP homotypic-interacting motif (RHIM)-dependent interaction, leading to the formation of a RIPK1–RIPK3 complex that initiates necroptosis [80,81]. Therefore, viral inhibition of caspase-8 by vICA blocks the extrinsic apoptosis pathway but sensitizes host cells to necroptosis. The activated RIPK3 subsequently recruits and phosphorylates the pore-forming protein mixed lineage kinase domain-like pseudokinase (MLKL) [82,83,84]. Phosphorylated MLKL oligomerizes and translocates to the plasma membrane, where it forms membrane-disrupting pores that lead to leakage of cytosolic content and inflammation [84,85]. Proteins that contain RHIMs play central roles in necroptotic cell death. Two other RHIM-containing proteins, ZBP1 and the TIR-domain-containing adaptor inducing interferon-β (TRIF), can activate RIPK3-dependent necroptosis. ZBP1 directly binds and activates RIPK3 through its RHIM, while the adaptor TRIF is recruited after stimulation of TLR3, leading to its interaction with RIPK3 through its RHIM domain [86,87,88] (Figure 2).
The M45 protein of MCMV is a potent cell death suppressor that contains a RHIM motif within its N-terminal domain and an induced protein aggregation motif (IPAM) located close to the C-terminus [89,90,91,92]. Although M45 strongly binds to RIPK1 with its C-terminal domain even when the RHIM-containing N-terminus is absent [93,94], the M45 RHIM domain is crucial for the inhibition of the necroptotic signaling. It inhibits RHIM-dependent activation of RIPK3, preventing necroptosis initiated by FAS or TNFR. Hence, it was named viral inhibitor of RIP activation (vIRA) [89,93] (Figure 2). As expected, MCMV expressing M45 with a mutated RHIM failed to inhibit necroptosis [95]. By interfering with RHIM–RHIM interactions, M45 can also inhibit ZBP1 and TRIF-mediated RIPK3 activation and subsequent necroptosis [87,88]. In MCMV-infected cells, Z-RNA resulting from newly transcribed viral RNA is sensed by ZBP1, which initiates necroptosis if not inhibited by M45 [96]. Besides RIPK1, M45 also binds strongly to the NF-κB essential modulator (NEMO), the regulatory subunit of the inhibitor of κB kinase (IKK) complex [93,94,97]. Both RIPK1 and NEMO are involved in the activation of the pro-inflammatory transcription factor NF-κB. The interaction of M45 with these proteins leads to the formation of insoluble protein aggregates. Aggregation of these proteins requires the IPAM located in the C-terminus of M45. It mediates self-interaction and is required for the formation of insoluble protein aggregates, which are targeted to autophagosomes for degradation in lysosomes [91]. Thus, M45 functions as a powerful inhibitor of both necroptosis and NF-κB activation. This ability to inhibit necroptosis is essential not only for MCMV infection of specific necroptosis-sensitive cell types but also for MCMV dissemination in vivo [95,98,99].
Similar to M45, the human alphaherpesviruses HSV-1 and HSV-2 R1 proteins (ICP6 and ICP10, respectively) also contain a RHIM domain and inhibit RIPK1/RIPK3-dependent necroptosis [100,101]. The HCMV protein homologous to M45, UL45, does not possess a RHIM domain, and an HCMV lacking UL45 does not exhibit the same phenotype as an M45-deficient MCMV. Although UL45 can inhibit RIPK1-mediated NF-κB activation by cooperating with the viral deubiquitinase UL48 [102], it does not impede necroptosis. In fact, the mechanism of necroptosis inhibition employed by HCMV is distinct from that of MCMV and targets a downstream step of RHIM protein signaling [103]. A recent study has shown that HCMV protein UL36 can function as a dual inhibitor of PCD: UL36 inhibits the extrinsic apoptosis pathway and blocks necroptosis by targeting MLKL and inducing its degradation [104] (Figure 2). Interestingly, UL36 was shown to bind to both human and murine MLKL. However, although UL36 was capable of reducing the levels of murine MLKL, it failed to inhibit necroptosis in murine cells [105].

2.3. Inhibition of Pyroptosis by CMVs

Pyroptosis is a lytic and highly inflammatory form of PCD, morphologically distinguishable from apoptosis. In pyroptosis, cells lose plasma membrane integrity, leading to cell swelling and rupture rather than the membrane blebbing in apoptosis. The detection of pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs) by cellular sensors such as NOD-like receptors (NLRs), AIM2-like receptors (ALRs) and RIG-I-like receptors (RLRs) initiate assembly of the inflammasome. While various inflammasomes serve to combat a wide array of pathogens, NLRP3 inflammasome, AIM2 inflammasome, and RIG-I inflammasome have been identified as crucial mediators of host responses to viral infection (reviewed in [106,107,108]). After sensing DAMPs or PAMPs, receptors recruit the adaptor protein ASC (apoptosis-associated speck-like protein containing a CARD), correlating with the oligomerization of ASC into macromolecular aggregates to form ASC specks [109,110]. The oligomeric inflammasome complex leads to the autoproteolytic cleavage of pro-caspase-1 into active caspase-1 consisting of the cleavage products p20 and p10 [111]. Activated caspase-1 processes the pro-inflammatory cytokine precursors pro-IL-1β and pro-IL-18 into their mature forms [112,113,114]. Moreover, caspase-1 also cleaves Gasdermin D (GSDMD), a key pyroptotic pore-forming protein, into an N-terminal (GSDMD-N) and C-terminal domain (GSDMD-C) [112,113]. GSDMD-N directly interacts with membrane phospholipids, leading to conformational changes that trigger oligomerization and the formation of a ring-shaped transmembrane pore [115,116,117] (Figure 3). GSDMD shares a common pore-forming mechanism with the entire GSDM protein family [115,118]. The formation of transmembrane pores by cleaved GSDMD and further plasma membrane rupture executed by NINJ1 protein [119,120,121] result in pyroptotic cell death and are essential for the release of the mature cytokines and other cellular components in the extracellular environment [114]. The release of mature cytokines IL-1β and IL-18 and proteins or “alarmins” derived from damaged cellular organelles such as the high-mobility group box 1 protein (HMGB1) contribute greatly to host responses against infected pathogens [122,123].
While it has been known for a long time that CMVs encode potent apoptosis and necroptosis inhibitors, a CMV inhibitor of pyroptosis has only recently been identified. NLRP3 detects a broad range of microbial motifs, endogenous danger signals and environmental irritants. The DNA sensors AIM2 and IFI16 are able to detect viral DNA upon viral infection. During infection with different herpesviruses, these receptors are known to be important sensors of viral infection and inducers of inflammation [107]. MCMV-infected AIM2-deficient mice were shown to have reduced IL-18 serum levels, and AIM2-deficient THP-1 cells were unable to efficiently control HCMV infection [124,125]. Interestingly, Horan et al. have shown that during CMV infection in myeloid cells, the viral DNA can be detected in the cytosol by IFI16 after ubiquitination and degradation of the viral capsid [126]. Only recently, the MCMV protein M84 was identified as an inhibitor of AIM2 inflammasome activation and signaling. The M84 protein interacts with both AIM2 and ASC through their pyrin domain to inhibit ASC speck formation (Figure 3). In the same way, M84 can also interact with other pyrin-domain-containing proteins, such as IFI203 and IFI204 [127]. The AIM2 inflammasome is mainly expressed in cells of the myeloid lineage such as macrophages and dendritic cells [128]. Compared with the WT virus, an M84stop mutant displayed a clear growth defect in macrophages. However, in fibroblasts, which lack AIM2 inflammasome expression, the M84stop mutant replicated to the same titers as the WT virus. The growth defect of the M84stop mutant was rescued when the AIM2 inflammasome was blocked (i.e., in AIM2 and ASC-deficient immortalized bone marrow-derived macrophages (iBMDM) or the presence of a caspase-1 inhibitor), suggesting that the AIM2 inflammasome restricts MCMV replication in macrophages and that M84 counteracts this restriction [127]. MCMV infection initially leads to transient caspase-1 activation and pro-inflammatory cytokine release [124]. Later, M84 inhibits caspase-1 activation and the release of IL-1β and IL-18 from MCMV-infected macrophages. Additionally, M84 also inhibits the cleavage of GSDMD, thereby reducing pyroptotic cell death. M84-mediated AIM2 inflammasome inhibition is important for MCMV replication and dissemination to the spleen and liver of infected mice and also inhibits the release of the inflammasome-activated cytokine IL-18 in the early stage of MCMV infection [127]. The stimulation of caspase-1-dependent canonical and caspase-11-dependent noncanonical pyroptotic pathways was shown in MCMV-induced retinitis of mice with murine acquired immunodeficiency syndrome (MAIDS) [129]. However, whether M84 inhibits caspase-11-dependent noncanonical pyroptosis remains unknown and requires further investigation.
Whether HCMV similarly interferes with inflammasome signaling remains unknown. The HCMV proteins UL83 and UL84 are most similar to MCMV M84 [130]. It was previously shown that HCMV protein UL83 interacts with the IFI16 pyrin domain to inhibit IFI16-mediated DNA sensing [131]. A UL83-deficient HCMV induced higher levels of caspase-1 activation compared to WT HCMV. A subsequent study showed that UL83 interacts with AIM2 and reduces the level of mature IL-1β in transfection assays [132]. Whether or not UL83 inhibits pyroptosis has not been investigated. The HCMV IE2 protein is also involved in the inhibition of the pro-IL-1β transcription and, independently, the induction of IL-1β protein degradation [133]. Further work will be required to fully understand the roles of HCMV proteins in modulating inflammasome signaling and pyroptosis.

3. Concluding Remarks and Perspectives

CMVs have evolved an array of mechanisms to inhibit PCD pathways in order to promote their replication. CMV inhibitors of the three major PCD pathways are described in this review. In some cases, HCMV and MCMV encode homologous viral proteins that function mechanistically in a very similar way. HCMV encodes multiple factors to subvert apoptotic response and mitochondrial cell death including UL36 (caspase-8 inhibitor); UL37x1 (binding and sequestering of BAX/BAK), UL38, which protects infected cells from endoplasmic reticulum stress; and β2.7 gene RNA, which is expressed much earlier during infection. Regarding the inhibition of necroptosis, HCMV is capable of inhibiting MLKL, an inhibitory mechanism that is substantially different from the one employed by MCMV.
The infection of mice with MCMV is a highly useful and practicable model to study CMV infection and pathogenesis in vivo [134]. Several studies have investigated the biological importance of MCMV cell death suppressors in this model. They revealed that viral inhibition of necroptosis is of great significance as M45-mutant MCMVs are highly attenuated and barely replicate and disseminate in vivo [92,95,98]. A strong attenuation was also observed in the absence of M36, the viral inhibitor of extrinsic apoptosis [58,135,136]. By contrast, the attenuation of MCMV mutants lacking inhibitors of intrinsic apoptosis (i.e., m38.5 or m41.1) or pyroptosis (M84) is more moderate [69,73,74,75,127]. However, this should not be interpreted to mean that these cell death pathways and their inhibition are of minor importance. MCMV and other viruses would not have acquired and maintained such inhibitors during their evolution if it were not to their benefit. A recent study has compared the impact of the different apoptosis and necroptosis inhibitors in MCMV-infected macrophages [76]. The results of the study suggested that MCMV employs sequential, non-redundant suppression strategies to specifically modulate PCD responses. Sequential activation of M45, M36 and m38.5/m41.1 initially suppresses RIPK3-dependent necroptosis, followed by inhibition of caspase-8-dependent extrinsic apoptosis and, lastly, intrinsic apoptosis regulated by BCL-2 family proteins and their viral inhibitors [76].
Until recently, very little was known about the importance of pyroptosis inhibition during CMV infection. Pyroptosis is usually mediated by inflammatory caspases and associated with the release of mature IL-1β and IL-18 [137]. MCMV M84 inhibits pyroptosis by inhibiting AIM2 inflammasome assembly and caspase-1-mediated cleavage of IL-1β, IL-18, and GSDMD [127]. It seems likely that HCMV encodes a similar inhibitor, but this remains to be demonstrated. As all of the N-terminal domains of GSDM family proteins can induce pyroptosis, sometimes without caspase activation [115], the term ‘pyroptosis’ has been re-defined as a GSDM-dependent type of cell death [138,139]. Considering the less-characterized functions of other Gasdermin proteins and their mechanisms of activation in different cell types during CMV infection, additional viral pyroptosis inhibitors might be identified in the future.
A recently described cytoplasmic multimeric protein complex named PANoptosome triggers a combined form of PCD dubbed PANoptosis [140]. The Kanneganti laboratory showed that ZBP1 interacts with RIPK3, caspase-6, and the NLRP3 inflammasome to form the ZBP1 PANoptosome complex that drives PANoptosis [141]. Later, the same laboratory defined another PANoptosome including AIM2, pyrin, and ZBP1 [142]. While the concept of PANoptosis is intriguing, many questions remain to be addressed. For example, it is unknown whether PANoptosomes are formed during CMV infection, whether the known PCD inhibitors are sufficient to inhibit all aspects of PANoptosis, or whether additional mechanisms of viral interference exist.
Overall, the interplay between PCD and CMV infection is complex and dynamic. Understanding these interactions is essential for developing strategies to combat CMV infections and designing novel antiviral therapies.

Author Contributions

Conceptualization and writing, Y.D., A.Á.-P. and W.B. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Landesforschungsförderung of the Free and Hanseatic City of Hamburg (LFF LV74 to W.B.). Y.D. was supported by a scholarship from the China Scholarship Council.

Conflicts of Interest

The authors declare no conflicts of interest. The funding sponsors had no role in the writing of the manuscript or in the decision to publish.

References

  1. Mocarski, E.S., Jr. Immunomodulation by cytomegaloviruses: Manipulative strategies beyond evasion. Trends Microbiol. 2002, 10, 332–339. [Google Scholar] [CrossRef] [PubMed]
  2. Hengel, H.; Brune, W.; Koszinowski, U.H. Immune evasion by cytomegalovirus--survival strategies of a highly adapted opportunist. Trends Microbiol. 1998, 6, 190–197. [Google Scholar] [CrossRef] [PubMed]
  3. Patro, A.R.K. Subversion of Immune Response by Human Cytomegalovirus. Front. Immunol. 2019, 10, 1155. [Google Scholar] [CrossRef] [PubMed]
  4. Griffiths, P.; Reeves, M. Pathogenesis of human cytomegalovirus in the immunocompromised host. Nat. Rev. Microbiol. 2021, 19, 759–773. [Google Scholar] [CrossRef] [PubMed]
  5. Brune, W. Molecular Basis of Cytomegalovirus Host Species Specificity. In Cytomegaloviruses: From Molecular Pathogenesis to Intervention; Reddehase, M., Ed.; Caister Academic Press: Wymondham, UK, 2013; Volume 1, pp. 322–329. [Google Scholar]
  6. Yu, D.; Silva, M.C.; Shenk, T. Functional map of human cytomegalovirus AD169 defined by global mutational analysis. Proc. Natl. Acad. Sci. USA 2003, 100, 12396–12401. [Google Scholar] [CrossRef] [PubMed]
  7. Dunn, W.; Chou, C.; Li, H.; Hai, R.; Patterson, D.; Stolc, V.; Zhu, H.; Liu, F. Functional profiling of a human cytomegalovirus genome. Proc. Natl. Acad. Sci. USA 2003, 100, 14223–14228. [Google Scholar] [CrossRef] [PubMed]
  8. Gerna, G.; Kabanova, A.; Lilleri, D. Human Cytomegalovirus Cell Tropism and Host Cell Receptors. Vaccines 2019, 7, 70. [Google Scholar] [CrossRef] [PubMed]
  9. Berry, R.; Watson, G.M.; Jonjic, S.; Degli-Esposti, M.A.; Rossjohn, J. Modulation of innate and adaptive immunity by cytomegaloviruses. Nat. Rev. Immunol. 2020, 20, 113–127. [Google Scholar] [CrossRef] [PubMed]
  10. Brune, W.; Andoniou, C.E. Die Another Day: Inhibition of Cell Death Pathways by Cytomegalovirus. Viruses 2017, 9, 249. [Google Scholar] [CrossRef]
  11. Ketelut-Carneiro, N.; Fitzgerald, K.A. Apoptosis, Pyroptosis, and Necroptosis-Oh My! The Many Ways a Cell Can Die. J. Mol. Biol. 2022, 434, 167378. [Google Scholar] [CrossRef]
  12. Elmore, S. Apoptosis: A review of programmed cell death. Toxicol. Pathol. 2007, 35, 495–516. [Google Scholar] [CrossRef] [PubMed]
  13. Nagata, S. Apoptosis and Clearance of Apoptotic Cells. Annu. Rev. Immunol. 2018, 36, 489–517. [Google Scholar] [CrossRef] [PubMed]
  14. Cai, Z.; Jitkaew, S.; Zhao, J.; Chiang, H.C.; Choksi, S.; Liu, J.; Ward, Y.; Wu, L.G.; Liu, Z.G. Plasma membrane translocation of trimerized MLKL protein is required for TNF-induced necroptosis. Nat. Cell Biol. 2014, 16, 55–65. [Google Scholar] [CrossRef] [PubMed]
  15. de Vasconcelos, N.M.; Lamkanfi, M. Recent Insights on Inflammasomes, Gasdermin Pores, and Pyroptosis. Cold Spring Harb. Perspect. Biol. 2020, 12, a036392. [Google Scholar] [CrossRef] [PubMed]
  16. Cookson, B.T.; Brennan, M.A. Pro-inflammatory programmed cell death. Trends Microbiol. 2001, 9, 113–114. [Google Scholar] [CrossRef] [PubMed]
  17. Upton, J.W.; Chan, F.K. Staying alive: Cell death in antiviral immunity. Mol. Cell 2014, 54, 273–280. [Google Scholar] [CrossRef] [PubMed]
  18. Wallach, D.; Kang, T.B.; Dillon, C.P.; Green, D.R. Programmed necrosis in inflammation: Toward identification of the effector molecules. Science 2016, 352, aaf2154. [Google Scholar] [CrossRef] [PubMed]
  19. Place, D.E.; Lee, S.; Kanneganti, T.D. PANoptosis in microbial infection. Curr. Opin. Microbiol. 2021, 59, 42–49. [Google Scholar] [CrossRef] [PubMed]
  20. Oh, S.; Lee, S. Recent advances in ZBP1-derived PANoptosis against viral infections. Front. Immunol. 2023, 14, 1148727. [Google Scholar] [CrossRef] [PubMed]
  21. Muzio, M. Signalling by proteolysis: Death receptors induce apoptosis. Int. J. Clin. Lab. Res. 1998, 28, 141–147. [Google Scholar] [CrossRef]
  22. Wajant, H. Death receptors. Essays Biochem. 2003, 39, 53–71. [Google Scholar] [CrossRef] [PubMed]
  23. Kim, J.W.; Choi, E.J.; Joe, C.O. Activation of death-inducing signaling complex (DISC) by pro-apoptotic C-terminal fragment of RIP. Oncogene 2000, 19, 4491–4499. [Google Scholar] [CrossRef] [PubMed]
  24. Wang, J.; Chun, H.J.; Wong, W.; Spencer, D.M.; Lenardo, M.J. Caspase-10 is an initiator caspase in death receptor signaling. Proc. Natl. Acad. Sci. USA 2001, 98, 13884–13888. [Google Scholar] [CrossRef] [PubMed]
  25. Schneider, P.; Thome, M.; Burns, K.; Bodmer, J.L.; Hofmann, K.; Kataoka, T.; Holler, N.; Tschopp, J. TRAIL receptors 1 (DR4) and 2 (DR5) signal FADD-dependent apoptosis and activate NF-kappaB. Immunity 1997, 7, 831–836. [Google Scholar] [CrossRef]
  26. Chinnaiyan, A.M.; O’Rourke, K.; Tewari, M.; Dixit, V.M. FADD, a novel death domain-containing protein, interacts with the death domain of Fas and initiates apoptosis. Cell 1995, 81, 505–512. [Google Scholar] [CrossRef] [PubMed]
  27. Irmler, M.; Thome, M.; Hahne, M.; Schneider, P.; Hofmann, K.; Steiner, V.; Bodmer, J.L.; Schroter, M.; Burns, K.; Mattmann, C.; et al. Inhibition of death receptor signals by cellular FLIP. Nature 1997, 388, 190–195. [Google Scholar] [CrossRef] [PubMed]
  28. Wong, G.H.; Goeddel, D.V. Fas antigen and p55 TNF receptor signal apoptosis through distinct pathways. J. Immunol. 1994, 152, 1751–1755. [Google Scholar] [CrossRef]
  29. Hsu, H.; Xiong, J.; Goeddel, D.V. The TNF receptor 1-associated protein TRADD signals cell death and NF-kappa B activation. Cell 1995, 81, 495–504. [Google Scholar] [CrossRef] [PubMed]
  30. Micheau, O.; Tschopp, J. Induction of TNF receptor I-mediated apoptosis via two sequential signaling complexes. Cell 2003, 114, 181–190. [Google Scholar] [CrossRef]
  31. Boldin, M.P.; Varfolomeev, E.E.; Pancer, Z.; Mett, I.L.; Camonis, J.H.; Wallach, D. A novel protein that interacts with the death domain of Fas/APO1 contains a sequence motif related to the death domain. J. Biol. Chem. 1995, 270, 7795–7798. [Google Scholar] [CrossRef]
  32. Scott, F.L.; Stec, B.; Pop, C.; Dobaczewska, M.K.; Lee, J.J.; Monosov, E.; Robinson, H.; Salvesen, G.S.; Schwarzenbacher, R.; Riedl, S.J. The Fas-FADD death domain complex structure unravels signalling by receptor clustering. Nature 2009, 457, 1019–1022. [Google Scholar] [CrossRef]
  33. Cohen, G.M. Caspases: The executioners of apoptosis. Biochem. J. 1997, 326 Pt 1, 1–16. [Google Scholar] [CrossRef]
  34. Tait, S.W.; Green, D.R. Mitochondria and cell death: Outer membrane permeabilization and beyond. Nat. Rev. Mol. Cell Biol. 2010, 11, 621–632. [Google Scholar] [CrossRef]
  35. Glover, H.L.; Schreiner, A.; Dewson, G.; Tait, S.W.G. Mitochondria and cell death. Nat. Cell Biol. 2024. [Google Scholar] [CrossRef]
  36. Willis, S.N.; Chen, L.; Dewson, G.; Wei, A.; Naik, E.; Fletcher, J.I.; Adams, J.M.; Huang, D.C. Proapoptotic Bak is sequestered by Mcl-1 and Bcl-xL, but not Bcl-2, until displaced by BH3-only proteins. Genes. Dev. 2005, 19, 1294–1305. [Google Scholar] [CrossRef]
  37. Fletcher, J.I.; Meusburger, S.; Hawkins, C.J.; Riglar, D.T.; Lee, E.F.; Fairlie, W.D.; Huang, D.C.; Adams, J.M. Apoptosis is triggered when prosurvival Bcl-2 proteins cannot restrain Bax. Proc. Natl. Acad. Sci. USA 2008, 105, 18081–18087. [Google Scholar] [CrossRef]
  38. Letai, A.; Bassik, M.C.; Walensky, L.D.; Sorcinelli, M.D.; Weiler, S.; Korsmeyer, S.J. Distinct BH3 domains either sensitize or activate mitochondrial apoptosis, serving as prototype cancer therapeutics. Cancer Cell 2002, 2, 183–192. [Google Scholar] [CrossRef]
  39. Czabotar, P.E.; Westphal, D.; Dewson, G.; Ma, S.; Hockings, C.; Fairlie, W.D.; Lee, E.F.; Yao, S.; Robin, A.Y.; Smith, B.J.; et al. Bax crystal structures reveal how BH3 domains activate Bax and nucleate its oligomerization to induce apoptosis. Cell 2013, 152, 519–531. [Google Scholar] [CrossRef]
  40. Liu, X.; Kim, C.N.; Yang, J.; Jemmerson, R.; Wang, X. Induction of apoptotic program in cell-free extracts: Requirement for dATP and cytochrome c. Cell 1996, 86, 147–157. [Google Scholar] [CrossRef]
  41. Du, C.; Fang, M.; Li, Y.; Li, L.; Wang, X. Smac, a mitochondrial protein that promotes cytochrome c-dependent caspase activation by eliminating IAP inhibition. Cell 2000, 102, 33–42. [Google Scholar] [CrossRef]
  42. Verhagen, A.M.; Ekert, P.G.; Pakusch, M.; Silke, J.; Connolly, L.M.; Reid, G.E.; Moritz, R.L.; Simpson, R.J.; Vaux, D.L. Identification of DIABLO, a mammalian protein that promotes apoptosis by binding to and antagonizing IAP proteins. Cell 2000, 102, 43–53. [Google Scholar] [CrossRef]
  43. Srinivasula, S.M.; Datta, P.; Fan, X.J.; Fernandes-Alnemri, T.; Huang, Z.; Alnemri, E.S. Molecular determinants of the caspase-promoting activity of Smac/DIABLO and its role in the death receptor pathway. J. Biol. Chem. 2000, 275, 36152–36157. [Google Scholar] [CrossRef]
  44. Li, P.; Nijhawan, D.; Budihardjo, I.; Srinivasula, S.M.; Ahmad, M.; Alnemri, E.S.; Wang, X. Cytochrome c and dATP-dependent formation of Apaf-1/caspase-9 complex initiates an apoptotic protease cascade. Cell 1997, 91, 479–489. [Google Scholar] [CrossRef]
  45. Zou, H.; Henzel, W.J.; Liu, X.; Lutschg, A.; Wang, X. Apaf-1, a human protein homologous to C. elegans CED-4, participates in cytochrome c-dependent activation of caspase-3. Cell 1997, 90, 405–413. [Google Scholar] [CrossRef]
  46. Bao, Q.; Shi, Y. Apoptosome: A platform for the activation of initiator caspases. Cell Death Differ. 2007, 14, 56–65. [Google Scholar] [CrossRef]
  47. Johnston, B.P.; McCormick, C. Herpesviruses and the Unfolded Protein Response. Viruses 2019, 12, 17. [Google Scholar] [CrossRef]
  48. Terhune, S.; Torigoi, E.; Moorman, N.; Silva, M.; Qian, Z.; Shenk, T.; Yu, D. Human cytomegalovirus UL38 protein blocks apoptosis. J. Virol. 2007, 81, 3109–3123. [Google Scholar] [CrossRef]
  49. Xuan, B.; Qian, Z.; Torigoi, E.; Yu, D. Human cytomegalovirus protein pUL38 induces ATF4 expression, inhibits persistent JNK phosphorylation, and suppresses endoplasmic reticulum stress-induced cell death. J. Virol. 2009, 83, 3463–3474. [Google Scholar] [CrossRef]
  50. Qian, Z.; Xuan, B.; Gualberto, N.; Yu, D. The human cytomegalovirus protein pUL38 suppresses endoplasmic reticulum stress-mediated cell death independently of its ability to induce mTORC1 activation. J. Virol. 2011, 85, 9103–9113. [Google Scholar] [CrossRef]
  51. Reeves, M.B.; Davies, A.A.; McSharry, B.P.; Wilkinson, G.W.; Sinclair, J.H. Complex I binding by a virally encoded RNA regulates mitochondria-induced cell death. Science 2007, 316, 1345–1348. [Google Scholar] [CrossRef]
  52. Verma, S.; Loewendorf, A.; Wang, Q.; McDonald, B.; Redwood, A.; Benedict, C.A. Inhibition of the TRAIL death receptor by CMV reveals its importance in NK cell-mediated antiviral defense. PLoS Pathog. 2014, 10, e1004268. [Google Scholar] [CrossRef]
  53. Smith, W.; Tomasec, P.; Aicheler, R.; Loewendorf, A.; Nemcovicova, I.; Wang, E.C.; Stanton, R.J.; Macauley, M.; Norris, P.; Willen, L.; et al. Human cytomegalovirus glycoprotein UL141 targets the TRAIL death receptors to thwart host innate antiviral defenses. Cell Host Microbe 2013, 13, 324–335. [Google Scholar] [CrossRef]
  54. Skaletskaya, A.; Bartle, L.M.; Chittenden, T.; McCormick, A.L.; Mocarski, E.S.; Goldmacher, V.S. A cytomegalovirus-encoded inhibitor of apoptosis that suppresses caspase-8 activation. Proc. Natl. Acad. Sci. USA 2001, 98, 7829–7834. [Google Scholar] [CrossRef]
  55. McCormick, A.L.; Skaletskaya, A.; Barry, P.A.; Mocarski, E.S.; Goldmacher, V.S. Differential function and expression of the viral inhibitor of caspase 8-induced apoptosis (vICA) and the viral mitochondria-localized inhibitor of apoptosis (vMIA) cell death suppressors conserved in primate and rodent cytomegaloviruses. Virology 2003, 316, 221–233. [Google Scholar] [CrossRef]
  56. Menard, C.; Wagner, M.; Ruzsics, Z.; Holak, K.; Brune, W.; Campbell, A.E.; Koszinowski, U.H. Role of murine cytomegalovirus US22 gene family members in replication in macrophages. J. Virol. 2003, 77, 5557–5570. [Google Scholar] [CrossRef]
  57. Chaudhry, M.Z.; Kasmapour, B.; Plaza-Sirvent, C.; Bajagic, M.; Casalegno Garduno, R.; Borkner, L.; Lenac Rovis, T.; Scrima, A.; Jonjic, S.; Schmitz, I.; et al. UL36 Rescues Apoptosis Inhibition and In vivo Replication of a Chimeric MCMV Lacking the M36 Gene. Front. Cell Infect. Microbiol. 2017, 7, 312. [Google Scholar] [CrossRef]
  58. Cicin-Sain, L.; Ruzsics, Z.; Podlech, J.; Bubic, I.; Menard, C.; Jonjic, S.; Reddehase, M.J.; Koszinowski, U.H. Dominant-negative FADD rescues the in vivo fitness of a cytomegalovirus lacking an antiapoptotic viral gene. J. Virol. 2008, 82, 2056–2064. [Google Scholar] [CrossRef]
  59. Chiou, S.H.; Liu, J.H.; Hsu, W.M.; Chen, S.S.; Chang, S.Y.; Juan, L.J.; Lin, J.C.; Yang, Y.T.; Wong, W.W.; Liu, C.Y.; et al. Up-regulation of Fas ligand expression by human cytomegalovirus immediate-early gene product 2: A novel mechanism in cytomegalovirus-induced apoptosis in human retina. J. Immunol. 2001, 167, 4098–4103. [Google Scholar] [CrossRef]
  60. Chiou, S.H.; Yang, Y.P.; Lin, J.C.; Hsu, C.H.; Jhang, H.C.; Yang, Y.T.; Lee, C.H.; Ho, L.L.; Hsu, W.M.; Ku, H.H.; et al. The immediate early 2 protein of human cytomegalovirus (HCMV) mediates the apoptotic control in HCMV retinitis through up-regulation of the cellular FLICE-inhibitory protein expression. J. Immunol. 2006, 177, 6199–6206. [Google Scholar] [CrossRef]
  61. Goldmacher, V.S.; Bartle, L.M.; Skaletskaya, A.; Dionne, C.A.; Kedersha, N.L.; Vater, C.A.; Han, J.W.; Lutz, R.J.; Watanabe, S.; Cahir McFarland, E.D.; et al. A cytomegalovirus-encoded mitochondria-localized inhibitor of apoptosis structurally unrelated to Bcl-2. Proc. Natl. Acad. Sci. USA 1999, 96, 12536–12541. [Google Scholar] [CrossRef]
  62. Arnoult, D.; Bartle, L.M.; Skaletskaya, A.; Poncet, D.; Zamzami, N.; Park, P.U.; Sharpe, J.; Youle, R.J.; Goldmacher, V.S. Cytomegalovirus cell death suppressor vMIA blocks Bax- but not Bak-mediated apoptosis by binding and sequestering Bax at mitochondria. Proc. Natl. Acad. Sci. USA 2004, 101, 7988–7993. [Google Scholar] [CrossRef]
  63. Poncet, D.; Larochette, N.; Pauleau, A.L.; Boya, P.; Jalil, A.A.; Cartron, P.F.; Vallette, F.; Schnebelen, C.; Bartle, L.M.; Skaletskaya, A.; et al. An anti-apoptotic viral protein that recruits Bax to mitochondria. J. Biol. Chem. 2004, 279, 22605–22614. [Google Scholar] [CrossRef]
  64. Norris, K.L.; Youle, R.J. Cytomegalovirus proteins vMIA and m38.5 link mitochondrial morphogenesis to Bcl-2 family proteins. J. Virol. 2008, 82, 6232–6243. [Google Scholar] [CrossRef]
  65. McCormick, A.L.; Roback, L.; Mocarski, E.S. HtrA2/Omi terminates cytomegalovirus infection and is controlled by the viral mitochondrial inhibitor of apoptosis (vMIA). PLoS Pathog. 2008, 4, e1000063. [Google Scholar] [CrossRef]
  66. Magalhaes, A.C.; Ferreira, A.R.; Gomes, S.; Vieira, M.; Gouveia, A.; Valenca, I.; Islinger, M.; Nascimento, R.; Schrader, M.; Kagan, J.C.; et al. Peroxisomes are platforms for cytomegalovirus’ evasion from the cellular immune response. Sci. Rep. 2016, 6, 26028. [Google Scholar] [CrossRef]
  67. Jurak, I.; Schumacher, U.; Simic, H.; Voigt, S.; Brune, W. Murine cytomegalovirus m38.5 protein inhibits Bax-mediated cell death. J. Virol. 2008, 82, 4812–4822. [Google Scholar] [CrossRef]
  68. Arnoult, D.; Skaletskaya, A.; Estaquier, J.; Dufour, C.; Goldmacher, V.S. The murine cytomegalovirus cell death suppressor m38.5 binds Bax and blocks Bax-mediated mitochondrial outer membrane permeabilization. Apoptosis 2008, 13, 1100–1110. [Google Scholar] [CrossRef]
  69. Manzur, M.; Fleming, P.; Huang, D.C.; Degli-Esposti, M.A.; Andoniou, C.E. Virally mediated inhibition of Bax in leukocytes promotes dissemination of murine cytomegalovirus. Cell Death Differ. 2009, 16, 312–320. [Google Scholar] [CrossRef]
  70. Cam, M.; Handke, W.; Picard-Maureau, M.; Brune, W. Cytomegaloviruses inhibit Bak- and Bax-mediated apoptosis with two separate viral proteins. Cell Death Differ. 2010, 17, 655–665. [Google Scholar] [CrossRef]
  71. Noguchi, K.; Majima, R.; Takahashi, K.; Iwase, Y.; Yamada, S.; Satoh, K.; Koshizuka, T.; Inoue, N. Identification and functional analyses of a cell-death inhibitor encoded by guinea pig cytomegalovirus gp38.1 in cell culture and in animals. J. Gen. Virol. 2020, 101, 1270–1279. [Google Scholar] [CrossRef]
  72. Satoh, K.; Takahashi, K.; Noguchi, K.; Kobayashi, Y.; Majima, R.; Iwase, Y.; Yamaguchi, K.; Masuda, Y.; Koshizuka, T.; Inoue, N. Characterization of the Second Apoptosis Inhibitor Encoded by Guinea Pig Cytomegalovirus. J. Virol. 2022, 96, e0162222. [Google Scholar] [CrossRef]
  73. Fleming, P.; Kvansakul, M.; Voigt, V.; Kile, B.T.; Kluck, R.M.; Huang, D.C.; Degli-Esposti, M.A.; Andoniou, C.E. MCMV-mediated inhibition of the pro-apoptotic Bak protein is required for optimal in vivo replication. PLoS Pathog. 2013, 9, e1003192. [Google Scholar] [CrossRef] [PubMed]
  74. Crosby, L.N.; McCormick, A.L.; Mocarski, E.S. Gene products of the embedded m41/m41.1 locus of murine cytomegalovirus differentially influence replication and pathogenesis. Virology 2013, 436, 274–283. [Google Scholar] [CrossRef] [PubMed]
  75. Handke, W.; Luig, C.; Popovic, B.; Krmpotic, A.; Jonjic, S.; Brune, W. Viral inhibition of BAK promotes murine cytomegalovirus dissemination to salivary glands. J. Virol. 2013, 87, 3592–3596. [Google Scholar] [CrossRef] [PubMed]
  76. Mandal, P.; Nagrani, L.N.; Hernandez, L.; McCormick, A.L.; Dillon, C.P.; Koehler, H.S.; Roback, L.; Alnemri, E.S.; Green, D.R.; Mocarski, E.S. Multiple Autonomous Cell Death Suppression Strategies Ensure Cytomegalovirus Fitness. Viruses 2021, 13, 1707. [Google Scholar] [CrossRef] [PubMed]
  77. Newton, K.; Strasser, A.; Kayagaki, N.; Dixit, V.M. Cell death. Cell 2024, 187, 235–256. [Google Scholar] [CrossRef] [PubMed]
  78. Weinlich, R.; Oberst, A.; Beere, H.M.; Green, D.R. Necroptosis in development, inflammation and disease. Nat. Rev. Mol. Cell Biol. 2017, 18, 127–136. [Google Scholar] [CrossRef] [PubMed]
  79. Samson, A.L.; Garnish, S.E.; Hildebrand, J.M.; Murphy, J.M. Location, location, location: A compartmentalized view of TNF-induced necroptotic signaling. Sci. Signal 2021, 14, eabc6178. [Google Scholar] [CrossRef] [PubMed]
  80. Cho, Y.S.; Challa, S.; Moquin, D.; Genga, R.; Ray, T.D.; Guildford, M.; Chan, F.K. Phosphorylation-driven assembly of the RIP1-RIP3 complex regulates programmed necrosis and virus-induced inflammation. Cell 2009, 137, 1112–1123. [Google Scholar] [CrossRef]
  81. Li, J.; McQuade, T.; Siemer, A.B.; Napetschnig, J.; Moriwaki, K.; Hsiao, Y.S.; Damko, E.; Moquin, D.; Walz, T.; McDermott, A.; et al. The RIP1/RIP3 necrosome forms a functional amyloid signaling complex required for programmed necrosis. Cell 2012, 150, 339–350. [Google Scholar] [CrossRef]
  82. Sun, L.; Wang, H.; Wang, Z.; He, S.; Chen, S.; Liao, D.; Wang, L.; Yan, J.; Liu, W.; Lei, X.; et al. Mixed lineage kinase domain-like protein mediates necrosis signaling downstream of RIP3 kinase. Cell 2012, 148, 213–227. [Google Scholar] [CrossRef] [PubMed]
  83. Zhao, J.; Jitkaew, S.; Cai, Z.; Choksi, S.; Li, Q.; Luo, J.; Liu, Z.G. Mixed lineage kinase domain-like is a key receptor interacting protein 3 downstream component of TNF-induced necrosis. Proc. Natl. Acad. Sci. USA 2012, 109, 5322–5327. [Google Scholar] [CrossRef] [PubMed]
  84. Murphy, J.M.; Czabotar, P.E.; Hildebrand, J.M.; Lucet, I.S.; Zhang, J.G.; Alvarez-Diaz, S.; Lewis, R.; Lalaoui, N.; Metcalf, D.; Webb, A.I.; et al. The pseudokinase MLKL mediates necroptosis via a molecular switch mechanism. Immunity 2013, 39, 443–453. [Google Scholar] [CrossRef] [PubMed]
  85. Wang, H.; Sun, L.; Su, L.; Rizo, J.; Liu, L.; Wang, L.F.; Wang, F.S.; Wang, X. Mixed lineage kinase domain-like protein MLKL causes necrotic membrane disruption upon phosphorylation by RIP3. Mol. Cell 2014, 54, 133–146. [Google Scholar] [CrossRef] [PubMed]
  86. Upton, J.W.; Kaiser, W.J. DAI Another Way: Necroptotic Control of Viral Infection. Cell Host Microbe 2017, 21, 290–293. [Google Scholar] [CrossRef]
  87. Upton, J.W.; Kaiser, W.J.; Mocarski, E.S. DAI/ZBP1/DLM-1 complexes with RIP3 to mediate virus-induced programmed necrosis that is targeted by murine cytomegalovirus vIRA. Cell Host Microbe 2012, 11, 290–297. [Google Scholar] [CrossRef]
  88. Kaiser, W.J.; Sridharan, H.; Huang, C.; Mandal, P.; Upton, J.W.; Gough, P.J.; Sehon, C.A.; Marquis, R.W.; Bertin, J.; Mocarski, E.S. Toll-like receptor 3-mediated necrosis via TRIF, RIP3, and MLKL. J. Biol. Chem. 2013, 288, 31268–31279. [Google Scholar] [CrossRef]
  89. Upton, J.W.; Kaiser, W.J.; Mocarski, E.S. Cytomegalovirus M45 cell death suppression requires receptor-interacting protein (RIP) homotypic interaction motif (RHIM)-dependent interaction with RIP1. J. Biol. Chem. 2008, 283, 16966–16970. [Google Scholar] [CrossRef]
  90. Lembo, D.; Brune, W. Tinkering with a viral ribonucleotide reductase. Trends Biochem. Sci. 2009, 34, 25–32. [Google Scholar] [CrossRef]
  91. Muscolino, E.; Schmitz, R.; Loroch, S.; Caragliano, E.; Schneider, C.; Rizzato, M.; Kim, Y.H.; Krause, E.; Juranic Lisnic, V.; Sickmann, A.; et al. Herpesviruses induce aggregation and selective autophagy of host signalling proteins NEMO and RIPK1 as an immune-evasion mechanism. Nat. Microbiol. 2020, 5, 331–342. [Google Scholar] [CrossRef]
  92. Muscolino, E.; Luoto, L.M.; Brune, W. Viral Induced Protein Aggregation: A Mechanism of Immune Evasion. Int. J. Mol. Sci. 2021, 22, 9624. [Google Scholar] [CrossRef]
  93. Mack, C.; Sickmann, A.; Lembo, D.; Brune, W. Inhibition of proinflammatory and innate immune signaling pathways by a cytomegalovirus RIP1-interacting protein. Proc. Natl. Acad. Sci. USA 2008, 105, 3094–3099. [Google Scholar] [CrossRef]
  94. Krause, E.; de Graaf, M.; Fliss, P.M.; Dölken, L.; Brune, W. Murine cytomegalovirus virion-associated protein M45 mediates rapid NF-kappaB activation after infection. J. Virol. 2014, 88, 9963–9975. [Google Scholar] [CrossRef]
  95. Upton, J.W.; Kaiser, W.J.; Mocarski, E.S. Virus inhibition of RIP3-dependent necrosis. Cell Host Microbe 2010, 7, 302–313. [Google Scholar] [CrossRef]
  96. Maelfait, J.; Liverpool, L.; Bridgeman, A.; Ragan, K.B.; Upton, J.W.; Rehwinkel, J. Sensing of viral and endogenous RNA by ZBP1/DAI induces necroptosis. EMBO J. 2017, 36, 2529–2543. [Google Scholar] [CrossRef]
  97. Fliss, P.M.; Jowers, T.P.; Brinkmann, M.M.; Holstermann, B.; Mack, C.; Dickinson, P.; Hohenberg, H.; Ghazal, P.; Brune, W. Viral mediated redirection of NEMO/IKKgamma to autophagosomes curtails the inflammatory cascade. PLoS Pathog. 2012, 8, e1002517. [Google Scholar] [CrossRef]
  98. Lembo, D.; Donalisio, M.; Hofer, A.; Cornaglia, M.; Brune, W.; Koszinowski, U.; Thelander, L.; Landolfo, S. The ribonucleotide reductase R1 homolog of murine cytomegalovirus is not a functional enzyme subunit but is required for pathogenesis. J. Virol. 2004, 78, 4278–4288. [Google Scholar] [CrossRef]
  99. Brune, W.; Menard, C.; Heesemann, J.; Koszinowski, U.H. A ribonucleotide reductase homolog of cytomegalovirus and endothelial cell tropism. Science 2001, 291, 303–305. [Google Scholar] [CrossRef]
  100. Guo, H.; Omoto, S.; Harris, P.A.; Finger, J.N.; Bertin, J.; Gough, P.J.; Kaiser, W.J.; Mocarski, E.S. Herpes simplex virus suppresses necroptosis in human cells. Cell Host Microbe 2015, 17, 243–251. [Google Scholar] [CrossRef]
  101. Huang, Z.; Wu, S.Q.; Liang, Y.; Zhou, X.; Chen, W.; Li, L.; Wu, J.; Zhuang, Q.; Chen, C.; Li, J.; et al. RIP1/RIP3 binding to HSV-1 ICP6 initiates necroptosis to restrict virus propagation in mice. Cell Host Microbe 2015, 17, 229–242. [Google Scholar] [CrossRef]
  102. Kwon, K.M.; Oh, S.E.; Kim, Y.E.; Han, T.H.; Ahn, J.H. Cooperative inhibition of RIP1-mediated NF-kappaB signaling by cytomegalovirus-encoded deubiquitinase and inactive homolog of cellular ribonucleotide reductase large subunit. PLoS Pathog. 2017, 13, e1006423. [Google Scholar] [CrossRef]
  103. Omoto, S.; Guo, H.; Talekar, G.R.; Roback, L.; Kaiser, W.J.; Mocarski, E.S. Suppression of RIP3-dependent necroptosis by human cytomegalovirus. J. Biol. Chem. 2015, 290, 11635–11648. [Google Scholar] [CrossRef]
  104. Fletcher-Etherington, A.; Nobre, L.; Nightingale, K.; Antrobus, R.; Nichols, J.; Davison, A.J.; Stanton, R.J.; Weekes, M.P. Human cytomegalovirus protein pUL36: A dual cell death pathway inhibitor. Proc. Natl. Acad. Sci. USA 2020, 117, 18771–18779. [Google Scholar] [CrossRef]
  105. Muscolino, E.; Castiglioni, C.; Brixel, R.; Frascaroli, G.; Brune, W. Species-Specific Inhibition of Necroptosis by HCMV UL36. Viruses 2021, 13, 2134. [Google Scholar] [CrossRef]
  106. Huerfano, S.; Sroller, V.; Brustikova, K.; Hornikova, L.; Forstova, J. The Interplay between Viruses and Host DNA Sensors. Viruses 2022, 14, 666. [Google Scholar] [CrossRef]
  107. Kumar, A.; Stavrakis, G.; Karaba, A.H. Herpesviruses and Inflammasomes: One Sensor Does Not Fit All. mBio 2022, 13, e0173721. [Google Scholar] [CrossRef]
  108. Yu, G.; Choi, Y.K.; Lee, S. Inflammasome diversity: Exploring novel frontiers in the innate immune response. Trends Immunol. 2024, 45, 248–258. [Google Scholar] [CrossRef]
  109. Lu, A.; Magupalli, V.G.; Ruan, J.; Yin, Q.; Atianand, M.K.; Vos, M.R.; Schroder, G.F.; Fitzgerald, K.A.; Wu, H.; Egelman, E.H. Unified polymerization mechanism for the assembly of ASC-dependent inflammasomes. Cell 2014, 156, 1193–1206. [Google Scholar] [CrossRef]
  110. Sborgi, L.; Ravotti, F.; Dandey, V.P.; Dick, M.S.; Mazur, A.; Reckel, S.; Chami, M.; Scherer, S.; Huber, M.; Bockmann, A.; et al. Structure and assembly of the mouse ASC inflammasome by combined NMR spectroscopy and cryo-electron microscopy. Proc. Natl. Acad. Sci. USA 2015, 112, 13237–13242. [Google Scholar] [CrossRef]
  111. Mariathasan, S.; Newton, K.; Monack, D.M.; Vucic, D.; French, D.M.; Lee, W.P.; Roose-Girma, M.; Erickson, S.; Dixit, V.M. Differential activation of the inflammasome by caspase-1 adaptors ASC and Ipaf. Nature 2004, 430, 213–218. [Google Scholar] [CrossRef]
  112. Kayagaki, N.; Stowe, I.B.; Lee, B.L.; O’Rourke, K.; Anderson, K.; Warming, S.; Cuellar, T.; Haley, B.; Roose-Girma, M.; Phung, Q.T.; et al. Caspase-11 cleaves gasdermin D for non-canonical inflammasome signalling. Nature 2015, 526, 666–671. [Google Scholar] [CrossRef] [PubMed]
  113. Shi, J.; Zhao, Y.; Wang, K.; Shi, X.; Wang, Y.; Huang, H.; Zhuang, Y.; Cai, T.; Wang, F.; Shao, F. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature 2015, 526, 660–665. [Google Scholar] [CrossRef] [PubMed]
  114. He, W.T.; Wan, H.; Hu, L.; Chen, P.; Wang, X.; Huang, Z.; Yang, Z.H.; Zhong, C.Q.; Han, J. Gasdermin D is an executor of pyroptosis and required for interleukin-1beta secretion. Cell Res. 2015, 25, 1285–1298. [Google Scholar] [CrossRef] [PubMed]
  115. Ding, J.; Wang, K.; Liu, W.; She, Y.; Sun, Q.; Shi, J.; Sun, H.; Wang, D.C.; Shao, F. Pore-forming activity and structural autoinhibition of the gasdermin family. Nature 2016, 535, 111–116. [Google Scholar] [CrossRef] [PubMed]
  116. Aglietti, R.A.; Estevez, A.; Gupta, A.; Ramirez, M.G.; Liu, P.S.; Kayagaki, N.; Ciferri, C.; Dixit, V.M.; Dueber, E.C. GsdmD p30 elicited by caspase-11 during pyroptosis forms pores in membranes. Proc. Natl. Acad. Sci. USA 2016, 113, 7858–7863. [Google Scholar] [CrossRef] [PubMed]
  117. Liu, X.; Zhang, Z.; Ruan, J.; Pan, Y.; Magupalli, V.G.; Wu, H.; Lieberman, J. Inflammasome-activated gasdermin D causes pyroptosis by forming membrane pores. Nature 2016, 535, 153–158. [Google Scholar] [CrossRef] [PubMed]
  118. Wang, Y.; Gao, W.; Shi, X.; Ding, J.; Liu, W.; He, H.; Wang, K.; Shao, F. Chemotherapy drugs induce pyroptosis through caspase-3 cleavage of a gasdermin. Nature 2017, 547, 99–103. [Google Scholar] [CrossRef] [PubMed]
  119. Kayagaki, N.; Kornfeld, O.S.; Lee, B.L.; Stowe, I.B.; O’Rourke, K.; Li, Q.; Sandoval, W.; Yan, D.; Kang, J.; Xu, M.; et al. NINJ1 mediates plasma membrane rupture during lytic cell death. Nature 2021, 591, 131–136. [Google Scholar] [CrossRef] [PubMed]
  120. Degen, M.; Santos, J.C.; Pluhackova, K.; Cebrero, G.; Ramos, S.; Jankevicius, G.; Hartenian, E.; Guillerm, U.; Mari, S.A.; Kohl, B.; et al. Structural basis of NINJ1-mediated plasma membrane rupture in cell death. Nature 2023, 618, 1065–1071. [Google Scholar] [CrossRef]
  121. David, L.; Borges, J.P.; Hollingsworth, L.R.; Volchuk, A.; Jansen, I.; Garlick, E.; Steinberg, B.E.; Wu, H. NINJ1 mediates plasma membrane rupture by cutting and releasing membrane disks. Cell 2024, 187, 2224–2235.e16. [Google Scholar] [CrossRef]
  122. Netea, M.G.; Simon, A.; van de Veerdonk, F.; Kullberg, B.J.; Van der Meer, J.W.; Joosten, L.A. IL-1beta processing in host defense: Beyond the inflammasomes. PLoS Pathog. 2010, 6, e1000661. [Google Scholar] [CrossRef]
  123. Lamkanfi, M.; Sarkar, A.; Vande Walle, L.; Vitari, A.C.; Amer, A.O.; Wewers, M.D.; Tracey, K.J.; Kanneganti, T.D.; Dixit, V.M. Inflammasome-dependent release of the alarmin HMGB1 in endotoxemia. J. Immunol. 2010, 185, 4385–4392. [Google Scholar] [CrossRef] [PubMed]
  124. Rathinam, V.A.; Jiang, Z.; Waggoner, S.N.; Sharma, S.; Cole, L.E.; Waggoner, L.; Vanaja, S.K.; Monks, B.G.; Ganesan, S.; Latz, E.; et al. The AIM2 inflammasome is essential for host defense against cytosolic bacteria and DNA viruses. Nat. Immunol. 2010, 11, 395–402. [Google Scholar] [CrossRef]
  125. Huang, Y.; Liu, L.; Ma, D.; Liao, Y.; Lu, Y.; Huang, H.; Qin, W.; Liu, X.; Fang, F. Human cytomegalovirus triggers the assembly of AIM2 inflammasome in THP-1-derived macrophages. J. Med. Virol. 2017, 89, 2188–2195. [Google Scholar] [CrossRef]
  126. Horan, K.A.; Hansen, K.; Jakobsen, M.R.; Holm, C.K.; Soby, S.; Unterholzner, L.; Thompson, M.; West, J.A.; Iversen, M.B.; Rasmussen, S.B.; et al. Proteasomal degradation of herpes simplex virus capsids in macrophages releases DNA to the cytosol for recognition by DNA sensors. J. Immunol. 2013, 190, 2311–2319. [Google Scholar] [CrossRef]
  127. Deng, Y.; Ostermann, E.; Brune, W. A cytomegalovirus inflammasome inhibitor reduces proinflammatory cytokine release and pyroptosis. Nat. Commun. 2024, 15, 786. [Google Scholar] [CrossRef] [PubMed]
  128. Jha, S.; Brickey, W.J.; Ting, J.P. Inflammasomes in Myeloid Cells: Warriors Within. Microbiol. Spectr. 2017, 5. [Google Scholar] [CrossRef]
  129. Carter, J.J.; Nemeno, J.G.E.; Oh, J.J.; Houghton, J.E.; Dix, R.D. Atypical cytomegalovirus retinal disease in pyroptosis-deficient mice with murine acquired immunodeficiency syndrome. Exp. Eye Res. 2021, 209, 108651. [Google Scholar] [CrossRef] [PubMed]
  130. Morello, C.S.; Cranmer, L.D.; Spector, D.H. In vivo replication, latency, and immunogenicity of murine cytomegalovirus mutants with deletions in the M83 and M84 genes, the putative homologs of human cytomegalovirus pp65 (UL83). J. Virol. 1999, 73, 7678–7693. [Google Scholar] [CrossRef]
  131. Li, T.; Chen, J.; Cristea, I.M. Human cytomegalovirus tegument protein pUL83 inhibits IFI16-mediated DNA sensing for immune evasion. Cell Host Microbe 2013, 14, 591–599. [Google Scholar] [CrossRef]
  132. Huang, Y.; Ma, D.; Huang, H.; Lu, Y.; Liao, Y.; Liu, L.; Liu, X.; Fang, F. Interaction between HCMV pUL83 and human AIM2 disrupts the activation of the AIM2 inflammasome. Virol. J. 2017, 14, 34. [Google Scholar] [CrossRef] [PubMed]
  133. Botto, S.; Abraham, J.; Mizuno, N.; Pryke, K.; Gall, B.; Landais, I.; Streblow, D.N.; Fruh, K.J.; DeFilippis, V.R. Human Cytomegalovirus Immediate Early 86-kDa Protein Blocks Transcription and Induces Degradation of the Immature Interleukin-1beta Protein during Virion-Mediated Activation of the AIM2 Inflammasome. mBio 2019, 10. [Google Scholar] [CrossRef] [PubMed]
  134. Brizic, I.; Lisnic, B.; Krstanovic, F.; Brune, W.; Hengel, H.; Jonjic, S. Mouse Models for Cytomegalovirus Infections in Newborns and Adults. Curr. Protoc. 2022, 2, e537. [Google Scholar] [CrossRef] [PubMed]
  135. Chaudhry, M.Z.; Casalegno-Garduno, R.; Sitnik, K.M.; Kasmapour, B.; Pulm, A.K.; Brizic, I.; Eiz-Vesper, B.; Moosmann, A.; Jonjic, S.; Mocarski, E.S.; et al. Cytomegalovirus inhibition of extrinsic apoptosis determines fitness and resistance to cytotoxic CD8 T cells. Proc. Natl. Acad. Sci. USA 2020, 117, 12961–12968. [Google Scholar] [CrossRef] [PubMed]
  136. Ebermann, L.; Ruzsics, Z.; Guzman, C.A.; van Rooijen, N.; Casalegno-Garduno, R.; Koszinowski, U.; Cicin-Sain, L. Block of death-receptor apoptosis protects mouse cytomegalovirus from macrophages and is a determinant of virulence in immunodeficient hosts. PLoS Pathog. 2012, 8, e1003062. [Google Scholar] [CrossRef] [PubMed]
  137. Broz, P.; Dixit, V.M. Inflammasomes: Mechanism of assembly, regulation and signalling. Nat. Rev. Immunol. 2016, 16, 407–420. [Google Scholar] [CrossRef] [PubMed]
  138. Shi, J.; Gao, W.; Shao, F. Pyroptosis: Gasdermin-Mediated Programmed Necrotic Cell Death. Trends Biochem. Sci. 2017, 42, 245–254. [Google Scholar] [CrossRef] [PubMed]
  139. Galluzzi, L.; Vitale, I.; Aaronson, S.A.; Abrams, J.M.; Adam, D.; Agostinis, P.; Alnemri, E.S.; Altucci, L.; Amelio, I.; Andrews, D.W.; et al. Molecular mechanisms of cell death: Recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ 2018, 25, 486–541. [Google Scholar] [CrossRef] [PubMed]
  140. Malireddi, R.K.S.; Kesavardhana, S.; Kanneganti, T.D. ZBP1 and TAK1: Master Regulators of NLRP3 Inflammasome/Pyroptosis, Apoptosis, and Necroptosis (PAN-optosis). Front. Cell Infect. Microbiol. 2019, 9, 406. [Google Scholar] [CrossRef]
  141. Zheng, M.; Karki, R.; Vogel, P.; Kanneganti, T.D. Caspase-6 Is a Key Regulator of Innate Immunity, Inflammasome Activation, and Host Defense. Cell 2020, 181, 674–687.e613. [Google Scholar] [CrossRef]
  142. Lee, S.; Karki, R.; Wang, Y.; Nguyen, L.N.; Kalathur, R.C.; Kanneganti, T.D. AIM2 forms a complex with pyrin and ZBP1 to drive PANoptosis and host defence. Nature 2021, 597, 415–419. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Inhibition of apoptosis by CMVs. The MCMV m166 protein inhibits TRAIL expression and HCMV UL141 prevents the expression of TRAIL death receptors on the cell surface. HCMV IE2 interferes with apoptosis signaling by inducing expression of cFLIP, an apoptotic inhibitor. HCMV protein UL36 and its homologous protein M36 in MCMV inhibit extrinsic apoptosis as viral inhibitors of Caspase-8 activation (vICA). HCMV protein UL37x1 and MCMV protein m38.5 are viral mitochondria-localized inhibitors of apoptosis (vMIA), which interfere with BAX. The m41.1 protein of MCMV inhibits apoptosis as a viral inhibitor of BAK oligomerization (vIBO). The HCMV β2.7 non-coding RNA counteracts oxidative stress and maintains the mitochondrial membrane potential. The HCMV protein UL38 inhibits apoptosis by inducing ATF4 accumulation to resolve ER stress. Additionally, UL38 curbs the activation of JNK, which suppresses anti-apoptotic BCL-2 and activates pro-apoptotic BH3 proteins. Created with BioRender.com.
Figure 1. Inhibition of apoptosis by CMVs. The MCMV m166 protein inhibits TRAIL expression and HCMV UL141 prevents the expression of TRAIL death receptors on the cell surface. HCMV IE2 interferes with apoptosis signaling by inducing expression of cFLIP, an apoptotic inhibitor. HCMV protein UL36 and its homologous protein M36 in MCMV inhibit extrinsic apoptosis as viral inhibitors of Caspase-8 activation (vICA). HCMV protein UL37x1 and MCMV protein m38.5 are viral mitochondria-localized inhibitors of apoptosis (vMIA), which interfere with BAX. The m41.1 protein of MCMV inhibits apoptosis as a viral inhibitor of BAK oligomerization (vIBO). The HCMV β2.7 non-coding RNA counteracts oxidative stress and maintains the mitochondrial membrane potential. The HCMV protein UL38 inhibits apoptosis by inducing ATF4 accumulation to resolve ER stress. Additionally, UL38 curbs the activation of JNK, which suppresses anti-apoptotic BCL-2 and activates pro-apoptotic BH3 proteins. Created with BioRender.com.
Viruses 16 01272 g001
Figure 2. Inhibition of necroptosis by CMVs. MCMV protein M45, the viral inhibitor of RIP activation (vIRA), binds and inhibits RIPK3 activation through its RIP homotypic interaction motif (RHIM). M45 thereby inhibits RIPK3-MLKL-mediated necroptosis. HCMV protein UL36 inhibits necroptosis by targeting MLKL and inducing its degradation. Created with BioRender.com.
Figure 2. Inhibition of necroptosis by CMVs. MCMV protein M45, the viral inhibitor of RIP activation (vIRA), binds and inhibits RIPK3 activation through its RIP homotypic interaction motif (RHIM). M45 thereby inhibits RIPK3-MLKL-mediated necroptosis. HCMV protein UL36 inhibits necroptosis by targeting MLKL and inducing its degradation. Created with BioRender.com.
Viruses 16 01272 g002
Figure 3. Inhibition of AIM2 inflammasome-mediated pyroptosis by MCMV. The M84 protein of MCMV interacts with AIM2 and ASC to inhibit inflammasome complex assembly. M84 thereby prevents downstream caspase-1 activation, the release of IL-1β and IL-18, GSDMD cleavage, and pyroptotic cell death. Created with BioRender.com.
Figure 3. Inhibition of AIM2 inflammasome-mediated pyroptosis by MCMV. The M84 protein of MCMV interacts with AIM2 and ASC to inhibit inflammasome complex assembly. M84 thereby prevents downstream caspase-1 activation, the release of IL-1β and IL-18, GSDMD cleavage, and pyroptotic cell death. Created with BioRender.com.
Viruses 16 01272 g003
Table 1. The main features of apoptosis, necroptosis, and pyroptosis. A comparison of the main features within the three programmed cell death (PCD) pathways.
Table 1. The main features of apoptosis, necroptosis, and pyroptosis. A comparison of the main features within the three programmed cell death (PCD) pathways.
ApoptosisNecroptosisPyroptosis
Lytic cell deathNoYesYes
Pore-forming proteinBAX/BAKMLKLGSDM proteins
Caspase activationYesNoYes
Inflammatory responseNoYesYes
Morphological featuresCell shrinkage
Nuclear condensation
Membrane blebbing
Cell swelling
Nuclear condensation
Membrane rupture
Cell swelling
Membrane rupture
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Deng, Y.; Águeda-Pinto, A.; Brune, W. No Time to Die: How Cytomegaloviruses Suppress Apoptosis, Necroptosis, and Pyroptosis. Viruses 2024, 16, 1272. https://doi.org/10.3390/v16081272

AMA Style

Deng Y, Águeda-Pinto A, Brune W. No Time to Die: How Cytomegaloviruses Suppress Apoptosis, Necroptosis, and Pyroptosis. Viruses. 2024; 16(8):1272. https://doi.org/10.3390/v16081272

Chicago/Turabian Style

Deng, Yingqi, Ana Águeda-Pinto, and Wolfram Brune. 2024. "No Time to Die: How Cytomegaloviruses Suppress Apoptosis, Necroptosis, and Pyroptosis" Viruses 16, no. 8: 1272. https://doi.org/10.3390/v16081272

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop