Next Article in Journal
Histone H1.2 Inhibited EMCV Replication through Enhancing MDA5-Mediated IFN-β Signaling Pathway
Next Article in Special Issue
A Balancing Act: The Viral–Host Battle over RNA Binding Proteins
Previous Article in Journal
Detection and Genome Sequencing of Lumpy Skin Disease Viruses in Wildlife Game Species in South Africa
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Antiviral Defence Mechanisms during Early Mammalian Development

by
Felix Mueller
1,2,†,
Jeroen Witteveldt
1,† and
Sara Macias
1,*
1
Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, King’s Buildings, Charlotte Auerbach Road, Edinburgh EH9 3FL, UK
2
Centre for Virus Research, MRC-University of Glasgow, Garscube Campus, 464 Bearsden Road, Glasgow G61 1QH, UK
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Viruses 2024, 16(2), 173; https://doi.org/10.3390/v16020173
Submission received: 14 December 2023 / Revised: 11 January 2024 / Accepted: 20 January 2024 / Published: 24 January 2024
(This article belongs to the Special Issue Regulation of the Virus Lifecycle by Cellular RNA-Binding Proteins)

Abstract

:
The type-I interferon (IFN) response constitutes the major innate immune pathway against viruses in mammals. Despite its critical importance for antiviral defence, this pathway is inactive during early embryonic development. There seems to be an incompatibility between the IFN response and pluripotency, the ability of embryonic cells to develop into any cell type of an adult organism. Instead, pluripotent cells employ alternative ways to defend against viruses that are typically associated with safeguard mechanisms against transposable elements. The absence of an inducible IFN response in pluripotent cells and the constitutive activation of the alternative antiviral pathways have led to the hypothesis that embryonic cells are highly resistant to viruses. However, some findings challenge this interpretation. We have performed a meta-analysis that suggests that the susceptibility of pluripotent cells to viruses is directly correlated with the presence of receptors or co-receptors for viral adhesion and entry. These results challenge the current view of pluripotent cells as intrinsically resistant to infections and raise the fundamental question of why these cells have sacrificed the major antiviral defence pathway if this renders them susceptible to viruses.

1. Introduction

During the early stages of mammalian development, both the cells and embryonic structures critical for supporting embryogenesis and formation of all the tissues of the adult organism are established. After fertilisation, embryonic cells transition from a toti-potent to a pluripotent state during the establishment of the blastocyst [1]. The blastocyst consists of two distinct structures: the inner cell mass from which human and mouse embryonic stem cells (ESCs) are derived and the outer layer of cells, or trophoblast, that will form the foetal component of the placenta. Despite the importance of these early stages in embryonic development, early embryos are unable to utilise one of the most important pathways to defend from viruses, the type-I interferon (IFN) response. In this review, we will focus on the regulation of this innate immune pathway during embryonic development, the use of alternative antiviral strategies by pluripotent cells, and review the critical role of specific RNA-binding proteins in providing antiviral defence during this stage. Our meta-analysis suggests that despite the existence of alternative antiviral pathways, pluripotent cells are not generally resistant to viral infections. We have also found that the susceptibility of pluripotent cells to viruses seems to correlate with the expression of viral receptors and co-receptors. This challenges our current view of pluripotent cells being intrinsically immune to viruses.

2. The Type-I IFN Response and Early Development

The major innate immune response against viruses is the type-I IFN response. IFNs are secreted upon infection and are sensed by both the infected and neighbouring cells, resulting in the induction of a large group of interferon-stimulated genes (ISGs) [2]. Besides this immediate cellular response, type I IFNs are also responsible for priming the adaptive immune system through the activation of dendritic cells [3,4]. As triggering the IFN response has profound consequences at both the cellular and organism level, a tight regulation of its induction and consequent downregulation is crucial [5]. To ensure its correct activation, cells have developed specialised sensor-proteins, or pattern recognition receptors (PRRs), capable of recognising different virus-specific hallmarks or pathogen-associated molecular patterns (PAMPs). One of the most prominent viral PAMPs is the virus-derived nucleic acids. PRRs have evolved to recognise types of nucleic acids that are typically not present in mammalian cells or specific cellular compartments and that only accumulate upon viral infection. These include long double-stranded RNAs (dsRNAs), short dsRNAs with either 5′ di- or triphosphate ends and cytoplasmic DNA [6].
Several families of PRRs are involved in the recognition of viral RNA PAMPs. These include the cytoplasmic RIG-I-like receptor family (RLR), composed of the retinoic acid-inducible gene-I (RIG-I), the melanoma differentiation-associated protein 5 (MDA5) and the laboratory of genetics and physiology 2 (LGP2), in addition to the endosomal Toll-like receptor (TLR) family. Aside from these, individual proteins can act as PRRs for viral DNA, such as the cyclic GMP-AMP synthase (cGAS) for cytosolic DNA, the interferon-gamma inducible protein 16 (IFI16) for both nuclear and cytosolic DNA and the absent in melanoma 2 (AIM2) for cytosolic DNA. Upon recognition of the viral nucleic acids, these sensors activate downstream signalling through essential adaptor proteins such as the mitochondrial antiviral signalling (MAVS) protein for the RLR receptors, the TIR domain-containing adaptor molecule 1 (TICAM1) and the MYD88 innate immune signal transduction adaptor (MYD88) for the TLR pathways, and the stimulator of interferon response cGAMP interactor 1 (STING) for cGAS signalling. Although these signalling cascades are different, they all culminate in the expression of type I IFNs through the activation of the interferon regulatory factors 3 and 7 (IRF3/7) [7,8]. After expression and secretion, type-I IFNs signal in an auto- and paracrine manner by binding to the surface type-I IFN receptor (interferon alpha and beta receptor subunit 1 and 2, IFNAR1/2) in both the infected but also neighbouring cells. The binding of IFNs to IFNAR1/2 leads to the phosphorylation and dimerization of STAT1 and 2 (the signal transducer and activator of transcription 1 and 2), which translocate to the nucleus to initiate the transcription of hundreds of ISGs, which are responsible for establishing an antiviral cellular state and stimulating the adaptive immune response (Figure 1A) [9].
Despite the crucial importance of IFNs for antiviral defence, there seems to be an incompatibility between the ability to produce IFNs and pluripotency. For instance, cells derived from murine embryonic teratocarcinomas, which still retain certain pluripotent capacity, cannot produce IFNs in response to viral infections or stimulation with the viral mimic, dsRNA. These cells also appear to have a much-reduced response to stimulation with exogenous IFNs, as pre-treatment with IFNs does not improve protection from viral infection. Interestingly, the ability to produce IFNs and respond to exogenous IFNs is acquired after differentiation [10,11,12]. Similar results were obtained with both mouse and human embryonic stem cells (ESCs) as well as human induced pluripotent stem cells (hiPSCs) [13,14,15,16,17]. The absence of IFN production in hiPSCs is intriguing, as these cells originate from somatic IFN-competent cells but lose the ability to produce IFNs when reprogrammed to become pluripotent. Aside from ESCs, mouse oocytes are also incapable of producing IFNs when stimulated with dsRNAs. Instead, oocytes show an efficient RNA interference (RNAi) response, an antiviral system classically attributed to non-vertebrate organisms, which will be described in more detail in the next section [18,19]. Similar observations have been made in rat spermatogonia and human testis explants, which both fail to induce an IFN response upon viral infection or dsRNA (poly I:C) treatment [20,21,22]. These results suggest that the absence of a functional IFN response is characteristic of pluripotent cells, as well as gametes. Instead, adult stem cells and trophoblasts do appear to have a functional IFN response [23,24]. Aside from viruses and dsRNA, mouse ESCs are also not capable of activating an innate immune response against viral DNA mimics such as G3-YSD, lipopolysaccharide (LPS, which mimics bacterial infections) or bacteria [17,25,26].
The mechanisms responsible for silencing the IFN response in pluripotent cells have still not been completely elucidated. Thus far, low expression of critical PRRs and silencing of key innate immune genes by miRNAs have been associated with a poor ability to produce IFNs by ESCs. For instance, mouse and human ESCs and iPSCs are known to express low levels of MDA5 and TLR3 while maintaining similar levels of RIG-I compared to differentiated cells [13,14,16,27]. In addition, an ESC-specific miRNA was shown to silence MAVS expression in mouse ESCs, leading to inactivation of the whole RLR-sensing pathway. Both removal of the miRNA and ectopic expression of MAVS reconstituted a functional IFN response [17]. A possible mechanism explaining the poor response of hESCs to exogenous IFNs was suggested by Hong and Carmichael [15]. They reported high expression of the suppressor of cytokine signalling 1 (SOCS1) in hESCs, which decreased upon differentiation. Normally, this protein is expressed as part of a negative feedback loop of STAT1 signalling but was found to be responsible for dampening the response to IFNs in hESCs.

3. Alternative Antiviral Responses during Early Embryonic Development

The absence of a functional IFN response may pose a strong evolutionary force to develop alternative antiviral mechanisms. Thus far, three distinct alternative antiviral responses have been described in ESCs: antiviral RNAi, ERASE and the constitutive or intrinsic expression of a subset of ISGs (Figure 1B) [28,29,30,31,32]. Due to the high levels of activity of transposable elements (TEs) during early development and the viral origin of some TE families, some of the mechanisms acting to control the potentially deleterious effects of TEs during development also provide some level of antiviral defence [33].

3.1. Mammalian Antiviral RNAi

Since 2013, several reports have suggested a role for antiviral RNAi in mammalian somatic but also pluripotent cells [28,29,34,35,36]. RNAi is a post-transcriptional gene-silencing mechanism orchestrated by the RNA endonuclease activity of Dicer and the slicing activity of Argonaute (Ago2 in mammals) [37,38,39,40]. Antiviral RNAi starts by Dicer binding and cleaving long viral-derived dsRNAs into 21- to 23-nucleotide long small interfering RNAs (siRNAs). These siRNAs are next loaded onto the RNA-induced silencing complex (RISC) with Ago2, driving recognition by base-pairing of its cognate viral RNA. Upon binding, the viral target RNA is cleaved by Ago2, resulting in reduced viral RNA availability for replication and egress [41,42].
Typically, organisms where RNAi is crucial for antiviral defence express multiple Dicer genes, with forms designated for antiviral RNAi and others specialised in endogenous small RNA production, including microRNAs (miRNAs) [43]. In contrast, mammals express a single Dicer gene (DICER1) that is involved in both miRNA and siRNA biogenesis [44,45]. Despite its involvement in both biogenesis pathways, Dicer cleaves precursor miRNA substrates more efficiently than long dsRNAs, from which siRNAs can originate [36]. Interestingly, truncation of the N-terminal helicase domain of human Dicer leads to increased cleavage of perfectly complementary dsRNAs compared to full-length Dicer. Truncated Dicer generates siRNAs that are loaded onto the RISC complex and are able to inhibit the expression of target mRNAs, including viral RNAs [46]. A naturally occurring isoform of Dicer lacking the N-terminal helicase domain (DicerO) was found in mouse oocytes and also exhibited a more efficient cleavage of long dsRNAs compared to full-length Dicer [47]. Another natural, RNAi-proficient isoform of Dicer was detected in both a range of pluripotent and somatic mouse cells, but also in humans, including hESCs, hiPSCs and somatic cell lines [30]. Overexpression of this isoform (aviD) in DICER1−/− cells restored miRNA expression but also showed antiviral activity, with decreased Sindbis or Zika virus production. AviD is an alternatively spliced isoform of Dicer, lacking exons 7 and 8, which encode for one of the three helicase domains previously implicated in inhibiting siRNA processivity [46,47,48]. For the efficient processing of miRNA and siRNA substrates, the N-terminal helicase domain of Dicer needs to associate with its endonucleolytic RNase IIIb domain. While the N-terminal helicase domain of non-vertebrates allows for a certain structural flexibility and can change its conformation to accommodate different RNA substrates, the helicase domain of mammalian Dicer retains its conformation during RNA binding, limiting its potential substrates to shorter RNAs, including pre-miRNAs or shRNAs [49,50]. Thus, deletions within the N-terminal domain might help overcome these structural limitations and promote the processing of long dsRNA substrates, including those that are produced during viral replication [30,46,47]. However, whether the RNAi-based antiviral response plays a meaningful role in mammalian antiviral immunity remains unclear, as Dicer inactivation results in both siRNA and miRNA biogenesis inactivation, but also increased IFN responses [17,30,51].
Aside from its antiviral role, RNAi is also involved in the suppression of TEs. This functional overlap possibly stems from the shared evolutionary origin between some TE families and viruses. Knock-down of Dicer in early mouse embryos leads to increased levels of two types of autonomous long-terminal repeat (LTR) retrotransposons, the Intracisternal A-Particles (IAP) and the murine endogenous retrovirus-L (MERVL) [52]. This was later corroborated in mESC Dicer knockout cell lines and mouse oocytes, which both showed an increased expression of ERVs, but also of other types of TEs, including the long- and short-interspersed nuclear elements (LINEs and SINEs) [47,53,54,55,56,57,58,59].
While ESCs can produce IFNs upon differentiation, the relevance of RNAi-mediated antiviral responses is suggested to decrease after differentiation [28,29,30,32,60]. It is, therefore, hypothesised that the IFN and RNAi responses can inhibit each other. Despite this, antiviral RNAi has been detected in non-pluripotent cell lines, including A549, HEK293T, BHK-21 and primary murine lung fibroblasts (MLFs) [34,35]. Supporting the cross-inhibition of RNAi and IFNs, Witteveldt et al. [17] demonstrated that in the absence of the central factor for RNAi, Dicer, mESCs become capable of synthesising IFNs [17]. In turn, inactivation of the IFN response in somatic cells results in detectable RNAi activity [61] and some ISGs, such as LGP2, can inhibit antiviral Dicer activity in somatic cells [62]. The underlying mechanisms behind this apparent incompatibility between the RNAi and IFN response remain to be fully elucidated [7]. Furthermore, the functional role of antiviral RNAi compared to the IFN response remains to be determined.

3.2. Intrinsic Expression of ISGs

ISGs are the ultimate effector proteins that establish the antiviral state in mammalian cells. Most ISGs are generally lowly expressed in homeostatic conditions and are only induced upon activation of the JAK/STAT pathway [63,64]. However, pluripotent cells, including ESCs, adult tissue stem cells and iPSCs, constitutively express a subset of ISGs, suggesting that these cell types display a basal and intrinsic antiviral state [32,60]. This subset of ISGs corresponds to the top 20% of the highest expressed genes in both mouse and human ESCs, and its composition varies in a stage- and species-specific manner. Interestingly, ISGs that are known to have antiproliferative or pro-apoptotic properties, such as CH25H, TNFSF10 or IFI27, are not detected in ESCs, as these could interfere with embryogenesis [32,60,65,66,67]. Moreover, activation of the type-I IFN response results in defects with the differentiation of pluripotent cells [16,67]. Twenty-one ISGs have been identified as highly expressed in human ESCs, while 22 ISGs are considered highly expressed in mouse ESCs. Only 10 ISGs are shared between mouse and human ESCs, including members of the IFITM family, which are known to be potent antiviral factors [32,60,68,69,70]. Two of the highly expressed ISGs—Mov10 in mESCs and ADAR in hESCs—are also RNA-binding proteins (RBPs) and are especially interesting, as they are known to be involved in the regulation of both viral- and TE-derived RNAs. These represent additional examples of the functional overlap between TE control and antiviral defence in early development [71,72,73,74,75,76].

3.2.1. Adenosine Deaminase RNA Specific (ADAR)

Adar is a deaminase enzyme that acts on dsRNA, converting adenosines (A) to inosines (I) (A-I) [77,78,79,80]. This activity mutates nucleic acid sequences, as well as disrupts dsRNA structures. In human ESCs, ADAR (also known as ADAR1) is one of the highly expressed ISGs in the absence of infection [32]. ADAR can edit both virus- and TE-derived dsRNAs, and depending on the virus, deamination can have pro- or anti-viral consequences [81,82,83,84,85]. ADAR and ADARB1 have been reported to target all types of viruses (RNA and DNA viruses). For instance, Adar has been suggested to act as an antiviral factor against Encephalomyocarditis virus (EMCV) [86]. This virus forms circular RNA intermediates during replication, which are inaccessible for PKR binding and consequent immune responses. Adar can destabilise the stem structure of the circular RNAs by editing specific sequences, so-called reverse complementary matches (RCM), enabling the binding of PKR [87,88].
In addition to viruses, Adar can edit cellular RNAs. For instance, binding and editing of endogenous TE-derived dsRNAs prevents aberrant innate immune activation by MDA5 and PKR. This function is important for embryonic development, as Adar−/− mice are embryonically lethal (E11.5-12.5), and the lethality can be partially reverted by knocking out MDA5 and/or MAVS or fully reverted to birth by knocking out both PKR and MDA5. These findings suggest that activation of the IFN response and the mRNA translational shutoff are toxic for embryonic development [89,90,91,92,93,94,95,96,97,98,99,100,101,102,103]. In humans, inactivating mutations of ADAR are associated with type-I interferonopathy, Aicardi-Goutières syndrome. Patients show a constitutive activation of the IFN response, probably due to the accumulation of unedited dsRNAs [98,104].
Besides its role in immunity, both ADAR (ADAR1) and ADARB1 (ADAR2) have been shown to restrict human Alu and LINE-1 retrotransposons. Alu elements are primate-specific SINEs that make up to 10% of the human genome and are one of the major cellular targets for Adar [105,106]. Due to their repetitive nature, Alu elements are prone to form dsRNA secondary structures through complementary pairing between copies inserted in sense and antisense orientation. Adar can disrupt these structures through A-to-I conversion, which is reminiscent of the mechanism by which Adar restricts EMCV. LINE-1 is restricted by interactions of Adar(s) with the LINE-1 RNP complex, potentially interfering with the retrotransposition cycle. This function seems to be editing-independent, as both ADAR and ADARB1 mutants lacking deaminase activity remained capable of restricting LINE-1 mobilisation [75,105,106,107,108]. While ADAR most likely drives the LINE-1 RNP into stress granules, ADARB1 acts within the nucleus [107,109]. Although the mechanisms by which Adar(s) inhibit the LINE-1 retrotransposon are not fully clarified, it shows the versatile function of these proteins in viral and TE defence and its importance in restricting aberrant innate immunity in mice and humans [33].

3.2.2. Moloney Leukaemia Virus 10 (MOV10)

MOV10 is an ATP-dependent RNA helicase that was originally identified as an antiviral restriction factor against Molony murine leukaemia virus (MMLV) in mice. It is also one of the highly expressed ISGs in mESCs [32,110]. Since its discovery, it has been found to interact with other viruses, including Human Immunodeficiency Virus-1 (HIV-1), Influenza A virus (IAV) and Hepatitis B and C virus (HBV, HCV). The effect of MOV10 during infection appears variable and has been reported to be both pro- and anti-viral, depending on the virus or on the phase of the viral lifecycle that is affected [74]. Besides its role in viral infections, MOV10 has also been reported to strongly repress LINE-1 and IAP retrotransposition by various mechanisms [111,112]. First, MOV10 was shown to impair the retrotranscriptase activity of both IAP and LINE-1 elements [113]. Second, MOV10 in complex with RNase H2 was shown to inhibit LINE-1 retrotransposition by limiting the formation of DNA–RNA heteroduplexes during target-primed reverse transcription (TPRT) [114]. However, these findings contradict the role of RNAse H2 in other settings where it was found to facilitate LINE-1 retrotransposition [115,116]. More recently, MOV10 has been shown to facilitate the addition of non-templated uridines at the 3′-end of LINE-1 RNA by the terminal uridyltransferases (TUTases) 4/7. Introducing uridines at the 3′-end can trigger both LINE-1 RNA decay but also inhibit the TPRT process, as this requires complementarity of the poly-A tail with the T-rich insertion target sites [117]. Importantly, the absence of MOV10 is embryonic lethal, possibly because of its role in restricting potentially deleterious TE activity [112,118].

3.3. ERASE (Endogenous RTase/RNase H-Mediated Antiviral System)

Recently, an IFN and RNAi-independent antiviral mechanism in mouse ESCs, termed ERASE (Endogenous Reverse transcriptase (RTase)/RNase H-mediated antiviral system), has been identified. This mechanism relies on the ERV-derived reverse transcriptase activity and the endogenous RNase H1 enzyme [31]. RNase H1 belongs to the RNase H superfamily and degrades the RNA strand in RNA–DNA duplexes. Although it is unclear if RNase H1 can also regulate TE activity, it has been shown to partially rescue the LINE-1 retrotransposition defects of RNase H2 knockout cells [115,119]. In ERASE, viral RNAs are first reverse-transcribed to cDNA by the RTases from ERVs, resulting in DNA/RNA hybrids, which are next degraded by the host RNase H1 [31]. This process seems to require specific RTases. While overexpression of the full-length MusD ERV inhibited Encephalomyocarditis virus (EMCV) infection in mESCs, overexpression of the IAPs and LINE-1, which also encode their own RT enzyme, only had a minor effect. Inhibition of MusD-derived RTase by the RTase inhibitor azidothymidine (AZT) diminished the observed antiviral activity. Since mammals encode a wide variety of RTases, Wu et al. hypothesise that other TE-derived RTases might also be capable of acting in this manner [31].

4. Are Pluripotent Cells Insensitive to Viral Infections?

The presence of ESC-specific antiviral mechanisms and the initially limited number of viruses capable of establishing a successful infection in pluripotent cells led to the general assumption that ESCs are resistant to viruses. In more recent years, an increasing number of successful viral infection models, and even examples of viruses that infect ESCs better than somatic IFN-competent cells, have been reported [17]. An explanation for these observations could be due to one aspect of the viral lifecycle that is rarely addressed, which is the expression of essential (co-) receptors for viral binding and entry on the host cells. There is an enormous variation in the expression of receptors within the various cell types of an organism, and this is one of the main determinants of viral tropism. Here, we have performed a systematic review of the expression of viral receptors in pluripotent cells and compiled a table listing the different viruses used in infection studies in both mouse- and human-derived pluripotent cells (Table 1). For viral receptor expression, human and mouse pluripotent ESC RNA high-throughput sequencing datasets were analysed using DESeq2 [32,120,121]. Log2 normalised counts were used to classify viral receptor expression as follows: (-) not expressed for counts lower than 0.3; (+) lowly expressed for counts between 0.3 and 3; (++) expressed for counts between 3 and 8; (+++) highly expressed for counts over 8; (?) was used if the expression was unknown, or if the gene had no homolog in that particular species. For quantifying infectivity, several methods were considered, including flow cytometry, immunofluorescence and vRNA/vDNA expression data. Infectivity was classified as follows: (-) non-infectious; (+) poorly infectious; (++) infectious and (+++) highly susceptible; (ND) not determined.
Aside from the observation that there are more viruses able than unable to infect pluripotent cells, this meta-analysis also reveals an additional trend: in cases where the virus cannot or only poorly infects pluripotent cells, at least one of the main respective (co-) receptors is either lowly or not expressed. Conversely, in cases where viruses can establish robust infections in pluripotent cells, their (co-) receptors are all expressed. This latter observation is summarised in Figure 2, where we plotted the infectivity level for each virus on the y-axis against the average receptor expression levels on the x-axis. Despite a number of limitations with this approach, including combining essential and non-essential receptors and the subjective quantification of infectivity, it strongly suggests a correlation between infectivity and receptor expression. Our hypothesis has been experimentally confirmed in only a few reports where both infectivity and receptor expression were quantified in the same pluripotent cell line [126,130,143]. Interestingly, there does not seem to be a relationship between the levels of infectivity and the types of viruses, as dsDNA, ssRNA (+) and ssRNA (-) viruses all vary between infectious and non-infectious types. Viruses from the Flaviviridae or Herpesviridae family are known for their wide host range [183,184], but infectivity for both families in pluripotent cells is absent or very poor. This lack of infectivity seems to correlate with the absence of some of the major receptors (Figure 2). Based on these data, it seems reasonable to conclude that, for many viruses, the lack of the essential factors for binding and entry in pluripotent cells is a major but not only determining factor for their ability to establish successful infections. It, therefore, seems premature to conclude that pluripotent cells have a general resistance to most viruses solely based on the limited number of non-infectious viruses and the presence of alternative antiviral mechanisms. However, it is also inaccurate to conclude that alternative antiviral mechanisms do not provide a certain level of intrinsic antiviral immunity. Infections of mammalian cells, either somatic or pluripotent, with Nodamura virus (NoV), Encephalomyocarditis virus (EMCV) and Influenza A virus (IAV) revealed the presence of viral suppressors of RNAi, suggesting that viruses have also evolved ways to block antiviral RNAi to ensure a robust infection [28,29,35,41]. Knockout of the highly expressed ISGs IFITM1-3 in ESCs results in a considerable increase in viral susceptibility [32]. Interestingly, the IFITM proteins’ antiviral mode of action involves the inhibition of viral entry [68].
Taken together, viral susceptibility in early development is determined by a wide range of cellular characteristics, starting with the expression of (co-) receptors, an absence of the IFN response and the activity of alternative antiviral mechanisms.

Author Contributions

F.M., J.W. and S.M. carried out the primary literature research and co-wrote the manuscript. F.M. and J.W. designed the figures. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by a Leverhulme Trust Research Grant (RPG-2020-355) and a Senior Research Fellowship from the Wellcome Trust (221737/Z/20/Z) to S.M. F.M. was funded by a Darwin Trust PhD fellowship.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Figures have been designed using ‘Smart Servier Medical Art’ (https://smart.servier.com) under a CC BY 3.0 (https://creativecommons.org/licenses/by/3.0/), accessed on 10 October 2023.

Acknowledgments

We thank Alasdair Ivens (University of Edinburgh) for his bioinformatic analyses of the transcriptomic datasets of human and mouse ESCs.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study, in the collection, analyses, or interpretation of data, in the writing of the manuscript, or in the decision to publish the results.

References

  1. Wilkinson, A.L.; Zorzan, I.; Rugg-Gunn, P.J. Epigenetic Regulation of Early Human Embryo Development. Cell Stem Cell 2023, 12, 1569–1584. [Google Scholar] [CrossRef] [PubMed]
  2. McNab, F.; Mayer-Barber, K.; Sher, A.; Wack, A.; O’Garra, A. Type I Interferons in Infectious Disease. Nat. Rev. Immunol. 2015, 15, 87–103. [Google Scholar] [CrossRef] [PubMed]
  3. Le Bon, A.; Tough, D.F. Links between Innate and Adaptive Immunity via Type I Interferon. Curr. Opin. Immunol. 2002, 14, 432–436. [Google Scholar] [CrossRef]
  4. Hertzog, P.J. Overview. Type I Interferons as Primers, Activators and Inhibitors of Innate and Adaptive Immune Responses. Immunol. Cell Biol. 2012, 90, 471–473. [Google Scholar] [CrossRef]
  5. Chen, K.; Liu, J.; Cao, X. Regulation of Type I Interferon Signaling in Immunity and Inflammation: A Comprehensive Review. J. Autoimmun. 2017, 83, 1–11. [Google Scholar] [CrossRef]
  6. Roers, A.; Hiller, B.; Hornung, V. Recognition of Endogenous Nucleic Acids by the Innate Immune System. Immunity 2016, 44, 739–754. [Google Scholar] [CrossRef] [PubMed]
  7. Watson, S.F.; Knol, L.I.; Witteveldt, J.; Macias, S. Crosstalk Between Mammalian Antiviral Pathways. Noncoding RNA 2019, 5, 29. [Google Scholar] [CrossRef] [PubMed]
  8. Dalskov, L.; Gad, H.H.; Hartmann, R. Viral Recognition and the Antiviral Interferon Response. EMBO J. 2023, 42, e112907. [Google Scholar] [CrossRef]
  9. Philips, R.L.; Wang, Y.; Cheon, H.; Kanno, Y.; Gadina, M.; Sartorelli, V.; Horvath, C.M.; Darnell, J.E.; Stark, G.R.; O’Shea, J.J. The JAK-STAT Pathway at 30: Much Learned, Much More to Do. Cell 2022, 185, 3857–3876. [Google Scholar] [CrossRef]
  10. Burke, D.C.; Graham, C.F.; Lehman, J.M. Appearance of Interferon Inducibility and Sensitivity during Differentiation of Murine Teratocarcinoma Cells in Vitro. Cell 1978, 13, 243–248. [Google Scholar] [CrossRef]
  11. Francis, M.K.; Lehman, J.M. Control of Beta-Interferon Expression in Murine Embryonal Carcinoma F9 Cells. Mol. Cell. Biol. 1989, 9, 3553–3556. [Google Scholar] [CrossRef] [PubMed]
  12. Ellis, M.J.; Goodbourn, S. NF-Kappa B-Independent Activation of Beta-Interferon Expression in Mouse F9 Embryonal Carcinoma Cells. Nucleic Acids Res. 1994, 22, 4489–4496. [Google Scholar] [CrossRef]
  13. Wang, R.; Wang, J.; Paul, A.M.; Acharya, D.; Bai, F.; Huang, F.; Guo, Y.-L. Mouse Embryonic Stem Cells Are Deficient in Type I Interferon Expression in Response to Viral Infections and Double-Stranded RNA. J. Biol. Chem. 2013, 288, 15926–15936. [Google Scholar] [CrossRef] [PubMed]
  14. Chen, L.L.; Yang, L.; Carmichael, G.G. Molecular Basis for an Attenuated Cytoplasmic DsRNA Response in Human Embryonic Stem Cells. Cell Cycle 2010, 9, 3552–3564. [Google Scholar] [CrossRef] [PubMed]
  15. Hong, X.X.; Carmichael, G.G. Innate Immunity in Pluripotent Human Cells: Attenuated Response to Interferon-Beta. J. Biol. Chem. 2013, 288, 16196–16205. [Google Scholar] [CrossRef]
  16. Eggenberger, J.; Blanco-Melo, D.; Panis, M.; Brennand, K.J.; tenOever, B.R. Type I Interferon Response Impairs Differentiation Potential of Pluripotent Stem Cells. Proc. Natl. Acad. Sci. USA 2019, 116, 1384–1393. [Google Scholar] [CrossRef]
  17. Witteveldt, J.; Knol, L.I.; Macias, S. MicroRNA-Deficient Mouse Embryonic Stem Cells Acquire a Functional Interferon Response. eLife 2019, 8, e44171. [Google Scholar] [CrossRef]
  18. Svoboda, P.; Stein, P.; Hayashi, H.; Schultz, R.M. Selective Reduction of Dormant Maternal MRNAs in Mouse Oocytes by RNA Interference. Dev. Camb. Engl. 2000, 127, 4147–4156. [Google Scholar] [CrossRef]
  19. Wianny, F.; Zernicka-Goetz, M. Specific Interference with Gene Function by Double-Stranded RNA in Early Mouse Development. Nat. Cell Biol. 2000, 2, 70–75. [Google Scholar] [CrossRef]
  20. Dejucq, N.; Lienard, M.-O.; Guillaume, E.; Dorval, I.; Jégou, B. Expression of Interferons-α and -γ in Testicular Interstitial Tissue and Spermatogonia of the Rat. Endocrinology 1998, 139, 3081–3087. [Google Scholar] [CrossRef]
  21. Matusali, G.; Houzet, L.; Satie, A.-P.; Mahé, D.; Aubry, F.; Couderc, T.; Frouard, J.; Bourgeau, S.; Bensalah, K.; Lavoué, S.; et al. Zika Virus Infects Human Testicular Tissue and Germ Cells. J. Clin. Investig. 2018, 128, 4697–4710. [Google Scholar] [CrossRef] [PubMed]
  22. Kuassivi, O.N.; Abiven, H.; Satie, A.-P.; Cartron, M.; Mahé, D.; Aubry, F.; Mathieu, R.; Rebours, V.; Le Tortorec, A.; Dejucq-Rainsford, N. Human Testicular Germ Cells, a Reservoir for Zika Virus, Lack Antiviral Response Upon Zika or Poly(I:C) Exposure. Front. Immunol. 2022, 13, 909341. [Google Scholar] [CrossRef] [PubMed]
  23. Yu, L.; Xu, Y.; Wang, F.; Yang, C.; Liu, G.; Song, X. Functional Roles of Pattern Recognition Receptors That Recognize Virus Nucleic Acids in Human Adipose-Derived Mesenchymal Stem Cells. BioMed Res. Int. 2016, 2016, 9872138. [Google Scholar] [CrossRef] [PubMed]
  24. Fendereski, M.; Neupane, B.; Nazneen, F.; Bai, F.; Guo, Y.-L. Mouse Trophoblast Cells Can Provide IFN-Based Antiviral Protection to Embryonic Stem Cells via Paracrine Signaling. J. Immunol. 2022, 208, 2761–2770. [Google Scholar] [CrossRef] [PubMed]
  25. D’Angelo, W.; Chen, B.; Gurung, C.; Guo, Y.-L. Characterization of Embryonic Stem Cell-Differentiated Fibroblasts as Mesenchymal Stem Cells with Robust Expansion Capacity and Attenuated Innate Immunity. Stem Cell Res. Ther. 2018, 9, 278. [Google Scholar] [CrossRef] [PubMed]
  26. Yu, J.; Rossi, R.; Hale, C.; Goulding, D.; Dougan, G. Interaction of Enteric Bacterial Pathogens with Murine Embryonic Stem Cells. Infect. Immun. 2009, 77, 585–597. [Google Scholar] [CrossRef]
  27. Chen, G.-Y.; Hwang, S.-M.; Su, H.-J.; Kuo, C.-Y.; Luo, W.-Y.; Lo, K.-W.; Huang, C.-C.; Chen, C.-L.; Yu, S.-H.; Hu, Y.-C. Defective Antiviral Responses of Induced Pluripotent Stem Cells to Baculoviral Vector Transduction. J. Virol. 2012, 86, 8041–8049. [Google Scholar] [CrossRef]
  28. Li, Y.; Lu, J.; Han, Y.; Fan, X.; Ding, S.W. RNA Interference Functions as an Antiviral Immunity Mechanism in Mammals. Science 2013, 342, 231–234. [Google Scholar] [CrossRef]
  29. Maillard, P.V.; Ciaudo, C.; Marchais, A.; Li, Y.; Jay, F.; Ding, S.W.; Voinnet, O. Antiviral RNA Interference in Mammalian Cells. Science 2013, 342, 235–238. [Google Scholar] [CrossRef]
  30. Poirier, E.Z.; Buck, M.D.; Chakravarty, P.; Carvalho, J.; Frederico, B.; Cardoso, A.; Healy, L.; Ulferts, R.; Beale, R.; Reis e Sousa, C. An Isoform of Dicer Protects Mammalian Stem Cells against Multiple RNA Viruses. Science 2021, 373, 231–236. [Google Scholar] [CrossRef]
  31. Wu, J.; Wu, C.; Xing, F.; Cao, L.; Zeng, W.; Guo, L.; Li, P.; Zhong, Y.; Jiang, H.; Luo, M.; et al. Endogenous Reverse Transcriptase and RNase H-Mediated Antiviral Mechanism in Embryonic Stem Cells. Cell Res. 2021, 31, 998–1010. [Google Scholar] [CrossRef] [PubMed]
  32. Wu, X.; Dao Thi, V.L.; Huang, Y.; Billerbeck, E.; Saha, D.; Hoffmann, H.H.; Wang, Y.; Silva, L.A.V.; Sarbanes, S.; Sun, T.; et al. Intrinsic Immunity Shapes Viral Resistance of Stem Cells. Cell 2018, 172, 423–438.e25. [Google Scholar] [CrossRef] [PubMed]
  33. Gazquez-Gutierrez, A.; Witteveldt, J.; Heras, S.R.; Macias, S. Sensing of Transposable Elements by the Antiviral Innate Immune System. RNA 2021, 27, 735–752. [Google Scholar] [CrossRef] [PubMed]
  34. Kong, J.; Bie, Y.; Ji, W.; Xu, J.; Lyu, B.; Xiong, X.; Qiu, Y.; Zhou, X. Alphavirus Infection Triggers Antiviral RNAi Immunity in Mammals. Cell Rep. 2023, 42, 112441. [Google Scholar] [CrossRef] [PubMed]
  35. Li, Y.; Basavappa, M.; Lu, J.; Dong, S.; Cronkite, D.A.; Prior, J.T.; Reinecker, H.-C.; Hertzog, P.; Han, Y.; Li, W.-X.; et al. Induction and Suppression of Antiviral RNA Interference by Influenza A Virus in Mammalian Cells. Nat. Microbiol. 2016, 2, 16250. [Google Scholar] [CrossRef]
  36. Wang, J.; Li, Y. Current Advances in Antiviral RNA Interference in Mammals. FEBS J. 2023, 291, 208–216. [Google Scholar] [CrossRef] [PubMed]
  37. Fire, A.; Xu, S.; Montgomery, M.K.; Kostas, S.A.; Driver, S.E.; Mello, C.C. Potent and Specific Genetic Interference by Double-Stranded RNA in Caenorhabditis Elegans. Nature 1998, 391, 806–811. [Google Scholar] [CrossRef] [PubMed]
  38. Gregory, R.I.; Chendrimada, T.P.; Cooch, N.; Shiekhattar, R. Human RISC Couples MicroRNA Biogenesis and Posttranscriptional Gene Silencing. Cell 2005, 123, 631–640. [Google Scholar] [CrossRef]
  39. Wilson, R.C.; Doudna, J.A. Molecular Mechanisms of RNA Interference. Annu. Rev. Biophys. 2013, 42, 217–239. [Google Scholar] [CrossRef]
  40. Rivas, F.V.; Tolia, N.H.; Song, J.-J.; Aragon, J.P.; Liu, J.; Hannon, G.J.; Joshua-Tor, L. Purified Argonaute2 and an SiRNA Form Recombinant Human RISC. Nat. Struct. Mol. Biol. 2005, 12, 340–349. [Google Scholar] [CrossRef]
  41. Maillard, P.V.; van der Veen, A.G.; Poirier, E.Z.; Reis e Sousa, C. Slicing and Dicing Viruses: Antiviral RNA Interference in Mammals. EMBO J. 2019, 38, e100941. [Google Scholar] [CrossRef] [PubMed]
  42. Schuster, S.; Miesen, P.; van Rij, R.P. Antiviral RNAi in Insects and Mammals: Parallels and Differences. Viruses 2019, 11, 448. [Google Scholar] [CrossRef] [PubMed]
  43. de Jong, D.; Eitel, M.; Jakob, W.; Osigus, H.-J.; Hadrys, H.; DeSalle, R.; Schierwater, B. Multiple Dicer Genes in the Early-Diverging Metazoa. Mol. Biol. Evol. 2009, 26, 1333–1340. [Google Scholar] [CrossRef] [PubMed]
  44. Margis, R.; Fusaro, A.F.; Smith, N.A.; Curtin, S.J.; Watson, J.M.; Finnegan, E.J.; Waterhouse, P.M. The Evolution and Diversification of Dicers in Plants. FEBS Lett. 2006, 580, 2442–2450. [Google Scholar] [CrossRef] [PubMed]
  45. Rossi, J.J. Mammalian Dicer Finds a Partner. EMBO Rep. 2005, 6, 927–929. [Google Scholar] [CrossRef] [PubMed]
  46. Kennedy, E.M.; Whisnant, A.W.; Kornepati, A.V.R.; Marshall, J.B.; Bogerd, H.P.; Cullen, B.R. Production of Functional Small Interfering RNAs by an Amino-Terminal Deletion Mutant of Human Dicer. Proc. Natl. Acad. Sci. USA 2015, 112, E6945–E6954. [Google Scholar] [CrossRef] [PubMed]
  47. Flemr, M.; Malik, R.; Franke, V.; Nejepinska, J.; Sedlacek, R.; Vlahovicek, K.; Svoboda, P. A Retrotransposon-Driven Dicer Isoform Directs Endogenous Small Interfering RNA Production in Mouse Oocytes. Cell 2013, 155, 807–816. [Google Scholar] [CrossRef]
  48. Ma, E.; MacRae, I.J.; Kirsch, J.F.; Doudna, J.A. Autoinhibition of Human Dicer by Its Internal Helicase Domain. J. Mol. Biol. 2008, 380, 237–243. [Google Scholar] [CrossRef]
  49. Gu, S.; Jin, L.; Zhang, Y.; Huang, Y.; Zhang, F.; Valdmanis, P.N.; Kay, M.A. The Loop Position of ShRNAs and Pre-MiRNAs Is Critical for the Accuracy of Dicer Processing in Vivo. Cell 2012, 151, 900–911. [Google Scholar] [CrossRef]
  50. Zapletal, D.; Kubicek, K.; Svoboda, P.; Stefl, R. Dicer Structure and Function: Conserved and Evolving Features. EMBO Rep. 2023, 24, e57215. [Google Scholar] [CrossRef]
  51. Gurung, C.; Fendereski, M.; Sapkota, K.; Guo, J.; Huang, F.; Guo, Y.L. Dicer Represses the Interferon Response and the Double-Stranded RNA-Activated Protein Kinase Pathway in Mouse Embryonic Stem Cells. J. Biol. Chem. 2021, 296, 100264. [Google Scholar] [CrossRef] [PubMed]
  52. Svoboda, P.; Stein, P.; Anger, M.; Bernstein, E.; Hannon, G.J.; Schultz, R.M. RNAi and Expression of Retrotransposons MuERV-L and IAP in Preimplantation Mouse Embryos. Dev. Biol. 2004, 269, 276–285. [Google Scholar] [CrossRef]
  53. Bodak, M.; Cirera-Salinas, D.; Yu, J.; Ngondo, R.P.; Ciaudo, C. Dicer, a New Regulator of Pluripotency Exit and LINE-1 Elements in Mouse Embryonic Stem Cells. FEBS Open Bio 2017, 7, 204–220. [Google Scholar] [CrossRef] [PubMed]
  54. Calabrese, J.M.; Seila, A.C.; Yeo, G.W.; Sharp, P.A. RNA Sequence Analysis Defines Dicer’s Role in Mouse Embryonic Stem Cells. Proc. Natl. Acad. Sci. USA 2007, 104, 18097–18102. [Google Scholar] [CrossRef] [PubMed]
  55. Tam, O.H.; Aravin, A.A.; Stein, P.; Girard, A.; Murchison, E.P.; Cheloufi, S.; Hodges, E.; Anger, M.; Sachidanandam, R.; Schultz, R.M.; et al. Pseudogene-Derived Small Interfering RNAs Regulate Gene Expression in Mouse Oocytes. Nature 2008, 453, 534–538. [Google Scholar] [CrossRef]
  56. Watanabe, T.; Takeda, A.; Tsukiyama, T.; Mise, K.; Okuno, T.; Sasaki, H.; Minami, N.; Imai, H. Identification and Characterization of Two Novel Classes of Small RNAs in the Mouse Germline: Retrotransposon-Derived SiRNAs in Oocytes and Germline Small RNAs in Testes. Genes Dev. 2006, 20, 1732–1743. [Google Scholar] [CrossRef] [PubMed]
  57. Watanabe, T.; Totoki, Y.; Toyoda, A.; Kaneda, M.; Kuramochi-Miyagawa, S.; Obata, Y.; Chiba, H.; Kohara, Y.; Kono, T.; Nakano, T.; et al. Endogenous SiRNAs from Naturally Formed DsRNAs Regulate Transcripts in Mouse Oocytes. Nature 2008, 453, 539–543. [Google Scholar] [CrossRef]
  58. Babiarz, J.E.; Ruby, J.G.; Wang, Y.; Bartel, D.P.; Blelloch, R. Mouse ES Cells Express Endogenous ShRNAs, SiRNAs, and Other Microprocessor-Independent, Dicer-Dependent Small RNAs. Genes Dev. 2008, 22, 2773–2785. [Google Scholar] [CrossRef]
  59. Berrens, R.V.; Andrews, S.; Spensberger, D.; Santos, F.; Dean, W.; Gould, P.; Sharif, J.; Olova, N.; Chandra, T.; Koseki, H.; et al. An EndosiRNA-Based Repression Mechanism Counteracts Transposon Activation during Global DNA Demethylation in Embryonic Stem Cells. Cell Stem Cell 2017, 21, 694–703.e7. [Google Scholar] [CrossRef]
  60. Wu, X.; Kwong, A.C.; Rice, C.M. Antiviral Resistance of Stem Cells. Curr. Opin. Immunol. 2019, 56, 50–59. [Google Scholar] [CrossRef]
  61. Maillard, P.V.; Van der Veen, A.G.; Deddouche-Grass, S.; Rogers, N.C.; Merits, A.; Reis e Sousa, C. Inactivation of the Type I Interferon Pathway Reveals Long Double-Stranded RNA-Mediated RNA Interference in Mammalian Cells. EMBO J. 2016, 35, 2505–2518. [Google Scholar] [CrossRef] [PubMed]
  62. van der Veen, A.G.; Maillard, P.V.; Schmidt, J.M.; Lee, S.A.; Deddouche-Grass, S.; Borg, A.; Kjaer, S.; Snijders, A.P.; Reis e Sousa, C. The RIG-I-like Receptor LGP2 Inhibits Dicer-Dependent Processing of Long Double-Stranded RNA and Blocks RNA Interference in Mammalian Cells. EMBO J. 2018, 37, e97479. [Google Scholar] [CrossRef] [PubMed]
  63. Schoggins, J.W.; Wilson, S.J.; Panis, M.; Murphy, M.Y.; Jones, C.T.; Bieniasz, P.; Rice, C.M. A Diverse Range of Gene Products Are Effectors of the Type I Interferon Antiviral Response. Nature 2011, 472, 481–485. [Google Scholar] [CrossRef] [PubMed]
  64. Schoggins, J.W.; Rice, C.M. Interferon-Stimulated Genes and Their Antiviral Effector Functions. Curr. Opin. Virol. 2011, 1, 519–525. [Google Scholar] [CrossRef]
  65. Borden, E.C.; Hogan, T.F.; Voelkel, J.G. Comparative Antiproliferative Activity in Vitro of Natural Interferons Alpha and Beta for Diploid and Transformed Human Cells. Cancer Res. 1982, 42, 4948–4953. [Google Scholar] [PubMed]
  66. Chawla-Sarkar, M.; Lindner, D.J.; Liu, Y.-F.; Williams, B.R.; Sen, G.C.; Silverman, R.H.; Borden, E.C. Apoptosis and Interferons: Role of Interferon-Stimulated Genes as Mediators of Apoptosis. Apoptosis 2003, 8, 237–249. [Google Scholar] [CrossRef] [PubMed]
  67. Hertzog, P.J.; Hwang, S.Y.; Kola, I. Role of Interferons in the Regulation of Cell Proliferation, Differentiation, and Development. Mol. Reprod. Dev. 1994, 39, 226–232. [Google Scholar] [CrossRef]
  68. Bailey, C.C.; Zhong, G.; Huang, I.-C.; Farzan, M. IFITM-Family Proteins: The Cell’s First Line of Antiviral Defense. Annu. Rev. Virol. 2014, 1, 261–283. [Google Scholar] [CrossRef]
  69. Grow, E.J.; Flynn, R.A.; Chavez, S.L.; Bayless, N.L.; Wossidlo, M.; Wesche, D.J.; Martin, L.; Ware, C.B.; Blish, C.A.; Chang, H.Y.; et al. Intrinsic Retroviral Reactivation in Human Preimplantation Embryos and Pluripotent Cells. Nature 2015, 522, 221–225. [Google Scholar] [CrossRef]
  70. Wu, X.; Robotham, J.M.; Lee, E.; Dalton, S.; Kneteman, N.M.; Gilbert, D.M.; Tang, H. Productive Hepatitis C Virus Infection of Stem Cell-Derived Hepatocytes Reveals a Critical Transition to Viral Permissiveness during Differentiation. PLoS Pathog. 2012, 8, e1002617. [Google Scholar] [CrossRef]
  71. Gallois-Montbrun, S.; Kramer, B.; Swanson, C.M.; Byers, H.; Lynham, S.; Ward, M.; Malim, M.H. Antiviral Protein APOBEC3G Localizes to Ribonucleoprotein Complexes Found in P Bodies and Stress Granules. J. Virol. 2007, 81, 2165–2178. [Google Scholar] [CrossRef]
  72. Goodier, J.L. Restricting Retrotransposons: A Review. Mob. DNA 2016, 7, 16. [Google Scholar] [CrossRef] [PubMed]
  73. Goodier, J.L.; Cheung, L.E.; Kazazian, H.H., Jr. MOV10 RNA Helicase Is a Potent Inhibitor of Retrotransposition in Cells. PLoS Genet. 2012, 8, e1002941. [Google Scholar] [CrossRef] [PubMed]
  74. Nawaz, A.; Shilikbay, T.; Skariah, G.; Ceman, S. Unwinding the Roles of RNA Helicase MOV10. WIREs RNA 2022, 13, e1682. [Google Scholar] [CrossRef] [PubMed]
  75. Orecchini, E.; Doria, M.; Antonioni, A.; Galardi, S.; Ciafrè, S.A.; Frassinelli, L.; Mancone, C.; Montaldo, C.; Tripodi, M.; Michienzi, A. ADAR1 Restricts LINE-1 Retrotransposition. Nucleic Acids Res. 2017, 45, 155–168. [Google Scholar] [CrossRef]
  76. Wang, X.; Han, Y.; Dang, Y.; Fu, W.; Zhou, T.; Ptak, R.G.; Zheng, Y.-H. Moloney Leukemia Virus 10 (MOV10) Protein Inhibits Retrovirus Replication. J. Biol. Chem. 2010, 285, 14346–14355. [Google Scholar] [CrossRef]
  77. Gerber, A.; O’Connell, M.A.; Keller, W. Two Forms of Human Double-Stranded RNA-Specific Editase 1 (HRED1) Generated by the Insertion of an Alu Cassette. RNA 1997, 3, 453–463. [Google Scholar]
  78. Gerber, A.P.; Keller, W. RNA Editing by Base Deamination: More Enzymes, More Targets, New Mysteries. Trends Biochem. Sci. 2001, 26, 376–384. [Google Scholar] [CrossRef]
  79. O’Connell, M.A. RNA Editing: Rewriting Receptors. Curr. Biol. CB 1997, 7, R437–R439. [Google Scholar] [CrossRef]
  80. O’Connell, M.A.; Gerber, A.; Keller, W. Purification of Human Double-Stranded RNA-Specific Editase 1 (HRED1) Involved in Editing of Brain Glutamate Receptor B Pre-MRNA. J. Biol. Chem. 1997, 272, 473–478. [Google Scholar] [CrossRef]
  81. Bass, B.L.; Weintraub, H.; Cattaneo, R.; Billeter, M.A. Biased Hypermutation of Viral RNA Genomes Could Be Due to Unwinding/Modification of Double-Stranded RNA. Cell 1989, 56, 331. [Google Scholar] [CrossRef] [PubMed]
  82. Pfaller, C.K.; George, C.X.; Samuel, C.E. Adenosine Deaminases Acting on RNA (ADARs) and Viral Infections. Annu. Rev. Virol. 2021, 8, 239–264. [Google Scholar] [CrossRef] [PubMed]
  83. Samuel, C.E. Adenosine Deaminases Acting on RNA (ADARs) Are Both Antiviral and Proviral. Virology 2011, 411, 180–193. [Google Scholar] [CrossRef] [PubMed]
  84. Taylor, D.R.; Puig, M.; Darnell, M.E.R.; Mihalik, K.; Feinstone, S.M. New Antiviral Pathway That Mediates Hepatitis C Virus Replicon Interferon Sensitivity through ADAR1. J. Virol. 2005, 79, 6291–6298. [Google Scholar] [CrossRef] [PubMed]
  85. Ward, S.V.; George, C.X.; Welch, M.J.; Liou, L.-Y.; Hahm, B.; Lewicki, H.; de la Torre, J.C.; Samuel, C.E.; Oldstone, M.B. RNA Editing Enzyme Adenosine Deaminase Is a Restriction Factor for Controlling Measles Virus Replication That Also Is Required for Embryogenesis. Proc. Natl. Acad. Sci. USA 2011, 108, 331–336. [Google Scholar] [CrossRef]
  86. Piontkivska, H.; Wales-McGrath, B.; Miyamoto, M.; Wayne, M.L. ADAR Editing in Viruses: An Evolutionary Force to Reckon With. Genome Biol. Evol. 2021, 13, evab240. [Google Scholar] [CrossRef]
  87. Liu, C.-X.; Li, X.; Nan, F.; Jiang, S.; Gao, X.; Guo, S.-K.; Xue, W.; Cui, Y.; Dong, K.; Ding, H.; et al. Structure and Degradation of Circular RNAs Regulate PKR Activation in Innate Immunity. Cell 2019, 177, 865–880.e21. [Google Scholar] [CrossRef]
  88. Ivanov, A.; Memczak, S.; Wyler, E.; Torti, F.; Porath, H.T.; Orejuela, M.R.; Piechotta, M.; Levanon, E.Y.; Landthaler, M.; Dieterich, C.; et al. Analysis of Intron Sequences Reveals Hallmarks of Circular RNA Biogenesis in Animals. Cell Rep. 2015, 10, 170–177. [Google Scholar] [CrossRef]
  89. Chung, H.; Calis, J.J.A.; Wu, X.; Sun, T.; Yu, Y.; Sarbanes, S.L.; Dao Thi, V.L.; Shilvock, A.R.; Hoffmann, H.-H.; Rosenberg, B.R.; et al. Human ADAR1 Prevents Endogenous RNA from Triggering Translational Shutdown. Cell 2018, 172, 811–824.e14. [Google Scholar] [CrossRef]
  90. Gannon, H.S.; Zou, T.; Kiessling, M.K.; Gao, G.F.; Cai, D.; Choi, P.S.; Ivan, A.P.; Buchumenski, I.; Berger, A.C.; Goldstein, J.T.; et al. Identification of ADAR1 Adenosine Deaminase Dependency in a Subset of Cancer Cells. Nat. Commun. 2018, 9, 5450. [Google Scholar] [CrossRef]
  91. Hartner, J.C.; Schmittwolf, C.; Kispert, A.; Müller, A.M.; Higuchi, M.; Seeburg, P.H. Liver Disintegration in the Mouse Embryo Caused by Deficiency in the RNA-Editing Enzyme ADAR1. J. Biol. Chem. 2004, 279, 4894–4902. [Google Scholar] [CrossRef] [PubMed]
  92. Hartner, J.C.; Walkley, C.R.; Lu, J.; Orkin, S.H. ADAR1 Is Essential for the Maintenance of Hematopoiesis and Suppression of Interferon Signaling. Nat. Immunol. 2009, 10, 109–115. [Google Scholar] [CrossRef] [PubMed]
  93. Liddicoat, B.J.; Piskol, R.; Chalk, A.M.; Ramaswami, G.; Higuchi, M.; Hartner, J.C.; Li, J.B.; Seeburg, P.H.; Walkley, C.R. RNA Editing by ADAR1 Prevents MDA5 Sensing of Endogenous DsRNA as Nonself. Science 2015, 349, 1115–1120. [Google Scholar] [CrossRef] [PubMed]
  94. Mannion, N.M.; Greenwood, S.M.; Young, R.; Cox, S.; Brindle, J.; Read, D.; Nellåker, C.; Vesely, C.; Ponting, C.P.; McLaughlin, P.J.; et al. The RNA-Editing Enzyme ADAR1 Controls Innate Immune Responses to RNA. Cell Rep. 2014, 9, 1482–1494. [Google Scholar] [CrossRef] [PubMed]
  95. Pestal, K.; Funk, C.C.; Snyder, J.M.; Price, N.D.; Treuting, P.M.; Stetson, D.B. Isoforms of RNA-Editing Enzyme ADAR1 Independently Control Nucleic Acid Sensor MDA5-Driven Autoimmunity and Multi-Organ Development. Immunity 2015, 43, 933–944. [Google Scholar] [CrossRef]
  96. Pujantell, M.; Riveira-Muñoz, E.; Badia, R.; Castellví, M.; Garcia-Vidal, E.; Sirera, G.; Puig, T.; Ramirez, C.; Clotet, B.; Esté, J.A.; et al. RNA Editing by ADAR1 Regulates Innate and Antiviral Immune Functions in Primary Macrophages. Sci. Rep. 2017, 7, 13339. [Google Scholar] [CrossRef]
  97. Quin, J.; Sedmík, J.; Vukić, D.; Khan, A.; Keegan, L.P.; O’Connell, M.A. ADAR RNA Modifications, the Epitranscriptome and Innate Immunity. Trends Biochem. Sci. 2021, 46, 758–771. [Google Scholar] [CrossRef]
  98. Rice, G.I.; Kasher, P.R.; Forte, G.M.A.; Mannion, N.M.; Greenwood, S.M.; Szynkiewicz, M.; Dickerson, J.E.; Bhaskar, S.S.; Zampini, M.; Briggs, T.A.; et al. Mutations in ADAR1 Cause Aicardi-Goutières Syndrome Associated with a Type I Interferon Signature. Nat. Genet. 2012, 44, 1243–1248. [Google Scholar] [CrossRef]
  99. Sinigaglia, K.; Cherian, A.; Vukic, D.; Melicherova, J.; Linhartova, P.; Du, Q.; Zerad, L.; Stejskal, S.; Malik, R.; Prochazka, J.; et al. Aberrant Activation of the Innate Immune Sensor PKR by Self DsRNA Is Prevented by Direct Interaction with ADAR1. bioRxiv 2023. [Google Scholar] [CrossRef]
  100. Vitali, P.; Scadden, A.D.J. Double-Stranded RNAs Containing Multiple IU Pairs Are Sufficient to Suppress Interferon Induction and Apoptosis. Nat. Struct. Mol. Biol. 2010, 17, 1043–1050. [Google Scholar] [CrossRef]
  101. Wang, Q.; Miyakoda, M.; Yang, W.; Khillan, J.; Stachura, D.L.; Weiss, M.J.; Nishikura, K. Stress-Induced Apoptosis Associated with Null Mutation of ADAR1 RNA Editing Deaminase Gene. J. Biol. Chem. 2004, 279, 4952–4961. [Google Scholar] [CrossRef] [PubMed]
  102. Yang, J.-H.; Luo, X.; Nie, Y.; Su, Y.; Zhao, Q.; Kabir, K.; Zhang, D.; Rabinovici, R. Widespread Inosine-Containing MRNA in Lymphocytes Regulated by ADAR1 in Response to Inflammation. Immunology 2003, 109, 15–23. [Google Scholar] [CrossRef] [PubMed]
  103. Hu, S.-B.; Heraud-Farlow, J.; Sun, T.; Liang, Z.; Goradia, A.; Taylor, S.; Walkley, C.R.; Li, J.B. ADAR1p150 Prevents MDA5 and PKR Activation via Distinct Mechanisms to Avert Fatal Autoinflammation. Mol. Cell 2023, 83, 3869–3884.e7. [Google Scholar] [CrossRef]
  104. Crow, Y.J. Aicardi-Goutières Syndrome. Handb. Clin. Neurol. 2013, 113, 1629–1635. [Google Scholar] [CrossRef] [PubMed]
  105. Athanasiadis, A.; Rich, A.; Maas, S. Widespread A-to-I RNA Editing of Alu-Containing MRNAs in the Human Transcriptome. PLoS Biol. 2004, 2, e391. [Google Scholar] [CrossRef]
  106. Levanon, E.Y.; Eisenberg, E.; Yelin, R.; Nemzer, S.; Hallegger, M.; Shemesh, R.; Fligelman, Z.Y.; Shoshan, A.; Pollock, S.R.; Sztybel, D.; et al. Systematic Identification of Abundant A-to-I Editing Sites in the Human Transcriptome. Nat. Biotechnol. 2004, 22, 1001–1005. [Google Scholar] [CrossRef]
  107. Frassinelli, L.; Orecchini, E.; Al-Wardat, S.; Tripodi, M.; Mancone, C.; Doria, M.; Galardi, S.; Ciafrè, S.A.; Michienzi, A. The RNA Editing Enzyme ADAR2 Restricts L1 Mobility. RNA Biol. 2021, 18, 75–87. [Google Scholar] [CrossRef] [PubMed]
  108. Zhao, K.; Du, J.; Peng, Y.; Li, P.; Wang, S.; Wang, Y.; Hou, J.; Kang, J.; Zheng, W.; Hua, S.; et al. LINE1 Contributes to Autoimmunity through Both RIG-I- and MDA5-Mediated RNA Sensing Pathways. J. Autoimmun. 2018, 90, 105–115. [Google Scholar] [CrossRef]
  109. Orecchini, E.; Frassinelli, L.; Michienzi, A. Restricting Retrotransposons: ADAR1 Is Another Guardian of the Human Genome. RNA Biol. 2017, 14, 1485–1491. [Google Scholar] [CrossRef]
  110. Jaenisch, R.; Jähner, D.; Nobis, P.; Simon, I.; Löhler, J.; Harbers, K.; Grotkopp, D. Chromosomal Position and Activation of Retroviral Genomes Inserted into the Germ Line of Mice. Cell 1981, 24, 519–529. [Google Scholar] [CrossRef]
  111. Li, X.; Zhang, J.; Jia, R.; Cheng, V.; Xu, X.; Qiao, W.; Guo, F.; Liang, C.; Cen, S. The MOV10 Helicase Inhibits LINE-1 Mobility. J. Biol. Chem. 2013, 288, 21148–21160. [Google Scholar] [CrossRef] [PubMed]
  112. Goodier, J.L.; Cheung, L.E.; Kazazian, H.H., Jr. Mapping the LINE1 ORF1 Protein Interactome Reveals Associated Inhibitors of Human Retrotransposition. Nucleic Acids Res. 2013, 41, 7401–7419. [Google Scholar] [CrossRef] [PubMed]
  113. Skariah, G.; Seimetz, J.; Norsworthy, M.; Lannom, M.C.; Kenny, P.J.; Elrakhawy, M.; Forsthoefel, C.; Drnevich, J.; Kalsotra, A.; Ceman, S. Mov10 Suppresses Retroelements and Regulates Neuronal Development and Function in the Developing Brain. BMC Biol. 2017, 15, 54. [Google Scholar] [CrossRef] [PubMed]
  114. Choi, J.; Hwang, S.-Y.; Ahn, K. Interplay between RNASEH2 and MOV10 Controls LINE-1 Retrotransposition. Nucleic Acids Res. 2018, 46, 1912–1926. [Google Scholar] [CrossRef] [PubMed]
  115. Benitez-Guijarro, M.; Lopez-Ruiz, C.; Tarnauskaitė, Ž.; Murina, O.; Mian Mohammad, M.; Williams, T.C.; Fluteau, A.; Sanchez, L.; Vilar-Astasio, R.; Garcia-Canadas, M.; et al. RNase H2, Mutated in Aicardi-Goutières Syndrome, Promotes LINE-1 Retrotransposition. EMBO J. 2018, 37, e98506. [Google Scholar] [CrossRef]
  116. Bartsch, K.; Knittler, K.; Borowski, C.; Rudnik, S.; Damme, M.; Aden, K.; Spehlmann, M.E.; Frey, N.; Saftig, P.; Chalaris, A.; et al. Absence of RNase H2 Triggers Generation of Immunogenic Micronuclei Removed by Autophagy. Hum. Mol. Genet. 2017, 26, 3960–3972. [Google Scholar] [CrossRef]
  117. Warkocki, Z.; Krawczyk, P.S.; Adamska, D.; Bijata, K.; Garcia-Perez, J.L.; Dziembowski, A. Uridylation by TUT4/7 Restricts Retrotransposition of Human LINE-1s. Cell 2018, 174, 1537–1548.e29. [Google Scholar] [CrossRef]
  118. Lu, C.; Luo, Z.; Jäger, S.; Krogan, N.J.; Peterlin, B.M. Moloney Leukemia Virus Type 10 Inhibits Reverse Transcription and Retrotransposition of Intracisternal A Particles. J. Virol. 2012, 86, 10517–10523. [Google Scholar] [CrossRef]
  119. Lockhart, A.; Pires, V.B.; Bento, F.; Kellner, V.; Luke-Glaser, S.; Yakoub, G.; Ulrich, H.D.; Luke, B. RNase H1 and H2 Are Differentially Regulated to Process RNA-DNA Hybrids. Cell Rep. 2019, 29, 2890–2900.e5. [Google Scholar] [CrossRef]
  120. Sladitschek, H.L.; Neveu, P.A. A Gene Regulatory Network Controls the Balance between Mesendoderm and Ectoderm at Pluripotency Exit. Mol. Syst. Biol. 2019, 15, e9043. [Google Scholar] [CrossRef]
  121. Love, M.I.; Huber, W.; Anders, S. Moderated Estimation of Fold Change and Dispersion for RNA-Seq Data with DESeq2. Genome Biol. 2014, 15, 550. [Google Scholar] [CrossRef] [PubMed]
  122. Smith-Arica, J.R.; Thomson, A.J.; Ansell, R.; Chiorini, J.; Davidson, B.; McWhir, J. Infection Efficiency of Human and Mouse Embryonic Stem Cells Using Adenoviral and Adeno-Associated Viral Vectors. Cloning Stem Cells 2003, 5, 51–62. [Google Scholar] [CrossRef] [PubMed]
  123. O’Donnell, J.; Taylor, K.A.; Chapman, M.S. Adeno-Associated Virus-2 and Its Primary Cellular Receptor—Cryo-EM Structure of a Heparin Complex. Virology 2009, 385, 434–443. [Google Scholar] [CrossRef] [PubMed]
  124. Pillay, S.; Meyer, N.L.; Puschnik, A.S.; Davulcu, O.; Diep, J.; Ishikawa, Y.; Jae, L.T.; Wosen, J.E.; Nagamine, C.M.; Chapman, M.S.; et al. An Essential Receptor for Adeno-Associated Virus Infection. Nature 2016, 530, 108–112. [Google Scholar] [CrossRef] [PubMed]
  125. Summerford, C.; Samulski, R.J. Membrane-Associated Heparan Sulfate Proteoglycan Is a Receptor for Adeno-Associated Virus Type 2 Virions. J. Virol. 1998, 72, 1438–1445. [Google Scholar] [CrossRef]
  126. Brokhman, I.; Pomp, O.; Fishman, L.; Tennenbaum, T.; Amit, M.; Itzkovitz-Eldor, J.; Goldstein, R.S. Genetic Modification of Human Embryonic Stem Cells with Adenoviral Vectors: Differences of Infectability between Lines and Correlation of Infectability with Expression of the Coxsackie and Adenovirus Receptor. Stem Cells Dev. 2009, 18, 447–457. [Google Scholar] [CrossRef]
  127. Arnberg, N. Adenovirus Receptors: Implications for Targeting of Viral Vectors. Trends Pharmacol. Sci. 2012, 33, 442–448. [Google Scholar] [CrossRef]
  128. Haisma, H.J.; Boesjes, M.; Beerens, A.M.; van der Strate, B.W.A.; Curiel, D.T.; Plüddemann, A.; Gordon, S.; Bellu, A.R. Scavenger Receptor A: A New Route for Adenovirus 5. Mol. Pharm. 2009, 6, 366–374. [Google Scholar] [CrossRef]
  129. Böhnke, J.; Pinkert, S.; Schmidt, M.; Binder, H.; Bilz, N.C.; Jung, M.; Reibetanz, U.; Beling, A.; Rujescu, D.; Claus, C. Coxsackievirus B3 Infection of Human IPSC Lines and Derived Primary Germ-Layer Cells Regarding Receptor Expression. Int. J. Mol. Sci. 2021, 22, 1220. [Google Scholar] [CrossRef]
  130. Scassa, M.E.; Jaquenod de Giusti, C.; Questa, M.; Pretre, G.; Richardson, G.A.V.; Bluguermann, C.; Romorini, L.; Ferrer, M.F.; Sevlever, G.E.; Miriuka, S.G.; et al. Human Embryonic Stem Cells and Derived Contractile Embryoid Bodies Are Susceptible to Coxsakievirus B Infection and Respond to Interferon Iβ Treatment. Stem Cell Res. 2011, 6, 13–22. [Google Scholar] [CrossRef]
  131. Hübner, D.; Jahn, K.; Pinkert, S.; Böhnke, J.; Jung, M.; Fechner, H.; Rujescu, D.; Liebert, U.G.; Claus, C. Infection of IPSC Lines with Miscarriage-Associated Coxsackievirus and Measles Virus and Teratogenic Rubella Virus as a Model for Viral Impairment of Early Human Embryogenesis. ACS Infect. Dis. 2017, 3, 886–897. [Google Scholar] [CrossRef] [PubMed]
  132. Shafren, D.R.; Bates, R.C.; Agrez, M.V.; Herd, R.L.; Burns, G.F.; Barry, R.D. Coxsackieviruses B1, B3, and B5 Use Decay Accelerating Factor as a Receptor for Cell Attachment. J. Virol. 1995, 69, 3873–3877. [Google Scholar] [CrossRef] [PubMed]
  133. Bazzone, L.E.; King, M.; MacKay, C.R.; Kyawe, P.P.; Meraner, P.; Lindstrom, D.; Rojas-Quintero, J.; Owen, C.A.; Wang, J.P.; Brass, A.L.; et al. A Disintegrin and Metalloproteinase 9 Domain (ADAM9) Is a Major Susceptibility Factor in the Early Stages of Encephalomyocarditis Virus Infection. mBio 2019, 10, e02734-18. [Google Scholar] [CrossRef] [PubMed]
  134. Hammoumi, S.; Guy, M.; Eloit, M.; Bakkali-Kassimi, L. Encephalomyocarditis Virus May Use Different Pathways to Initiate Infection of Primary Human Cardiomyocytes. Arch. Virol. 2012, 157, 43–52. [Google Scholar] [CrossRef] [PubMed]
  135. Weidemann, W.; Klukas, C.; Klein, A.; Simm, A.; Schreiber, F.; Horstkorte, R. Lessons from GNE-Deficient Embryonic Stem Cells: Sialic Acid Biosynthesis Is Involved in Proliferation and Gene Expression. Glycobiology 2010, 20, 107–117. [Google Scholar] [CrossRef]
  136. Martin, M.J.; Muotri, A.; Gage, F.; Varki, A. Human Embryonic Stem Cells Express an Immunogenic Nonhuman Sialic Acid. Nat. Med. 2005, 11, 228–232. [Google Scholar] [CrossRef] [PubMed]
  137. Huber, S.A. VCAM-1 Is a Receptor for Encephalomyocarditis Virus on Murine Vascular Endothelial Cells. J. Virol. 1994, 68, 3453–3458. [Google Scholar] [CrossRef] [PubMed]
  138. Wash, R.; Calabressi, S.; Franz, S.; Griffiths, S.J.; Goulding, D.; Tan, E.-P.; Wise, H.; Digard, P.; Haas, J.; Efstathiou, S.; et al. Permissive and Restricted Virus Infection of Murine Embryonic Stem Cells. J. Gen. Virol. 2012, 93, 2118–2130. [Google Scholar] [CrossRef]
  139. Dukhovny, A.; Sloutskin, A.; Markus, A.; Yee, M.B.; Kinchington, P.R.; Goldstein, R.S. Varicella-Zoster Virus Infects Human Embryonic Stem Cell-Derived Neurons and Neurospheres but Not Pluripotent Embryonic Stem Cells or Early Progenitors. J. Virol. 2012, 86, 3211–3218. [Google Scholar] [CrossRef]
  140. Connolly, S.A.; Jardetzky, T.S.; Longnecker, R. The Structural Basis of Herpesvirus Entry. Nat. Rev. Microbiol. 2021, 19, 110–121. [Google Scholar] [CrossRef]
  141. Lafemina, R.L.; Hayward, G.S. Differences in Cell Type-Specific Blocks to Immediate Early Gene Expression and DNA Replication of Human, Simian and Murine Cytomegalovirus. J. Gen. Virol. 1988, 69, 355–374. [Google Scholar] [CrossRef]
  142. Gonczol, E.; Andrews, P.W.; Plotkin, S.A. Cytomegalovirus Replicates in Differentiated but Not in Undifferentiated Human Embryonal Carcinoma Cells. Science 1984, 224, 159–161. [Google Scholar] [CrossRef] [PubMed]
  143. Berger, A.A.; Gil, Y.; Panet, A.; Weisblum, Y.; Oiknine-Djian, E.; Gropp, M.; Steiner, D.; Reubinoff, B.E.; Wolf, D.G. Transition toward Human Cytomegalovirus Susceptibility in Early Human Embryonic Stem Cell-Derived Neural Precursors. J. Virol. 2015, 89, 11159–11164. [Google Scholar] [CrossRef] [PubMed]
  144. Soroceanu, L.; Akhavan, A.; Cobbs, C.S. Platelet-Derived Growth Factor-α Receptor Activation Is Required for Human Cytomegalovirus Infection. Nature 2008, 455, 391–395. [Google Scholar] [CrossRef] [PubMed]
  145. Nguyen, C.C.; Kamil, J.P. Pathogen at the Gates: Human Cytomegalovirus Entry and Cell Tropism. Viruses 2018, 10, 704. [Google Scholar] [CrossRef] [PubMed]
  146. Shen, H.; Cheng, T.; Preffer, F.I.; Dombkowski, D.; Tomasson, M.H.; Golan, D.E.; Yang, O.; Hofmann, W.; Sodroski, J.G.; Luster, A.D.; et al. Intrinsic Human Immunodeficiency Virus Type 1 Resistance of Hematopoietic Stem Cells Despite Coreceptor Expression. J. Virol. 1999, 73, 728–737. [Google Scholar] [CrossRef]
  147. Weichold, F.F.; Zella, D.; Barabitskaja, O.; Maciejewski, J.P.; Dunn, D.E.; Sloand, E.M.; Young, N.S. Neither Human Immunodeficiency Virus-1 (HIV-1) nor HIV-2 Infects Most-Primitive Human Hematopoietic Stem Cells as Assessed in Long-Term Bone Marrow Cultures. Blood 1998, 91, 907–915. [Google Scholar] [CrossRef]
  148. Dragic, T.; Litwin, V.; Allaway, G.P.; Martin, S.R.; Huang, Y.; Nagashima, K.A.; Cayanan, C.; Maddon, P.J.; Koup, R.A.; Moore, J.P.; et al. HIV-1 Entry into CD4+ Cells Is Mediated by the Chemokine Receptor CC-CKR-5. Nature 1996, 381, 667–673. [Google Scholar] [CrossRef]
  149. Feng, Y.; Broder, C.C.; Kennedy, P.E.; Berger, E.A. HIV-1 Entry Cofactor: Functional CDNA Cloning of a Seven-Transmembrane, G Protein-Coupled Receptor. Science 1996, 272, 872–877. [Google Scholar] [CrossRef]
  150. Hastie, E.; Cataldi, M.; Marriott, I.; Grdzelishvili, V.Z. Understanding and Altering Cell Tropism of Vesicular Stomatitis Virus. Virus Res. 2013, 176, 16–32. [Google Scholar] [CrossRef]
  151. Zahedi-Amiri, A.; Sequiera, G.L.; Dhingra, S.; Coombs, K.M. Influenza a Virus-Triggered Autophagy Decreases the Pluripotency of Human-Induced Pluripotent Stem Cells. Cell Death Dis. 2019, 10, 337. [Google Scholar] [CrossRef] [PubMed]
  152. Sempere Borau, M.; Stertz, S. Entry of Influenza A Virus into Host Cells—Recent Progress and Remaining Challenges. Curr. Opin. Virol. 2021, 48, 23–29. [Google Scholar] [CrossRef] [PubMed]
  153. Windhaber, S.; Xin, Q.; Lozach, P.-Y. Orthobunyaviruses: From Virus Binding to Penetration into Mammalian Host Cells. Viruses 2021, 13, 872. [Google Scholar] [CrossRef] [PubMed]
  154. Naaman, H.; Rabinski, T.; Yizhak, A.; Mizrahi, S.; Avni, Y.S.; Taube, R.; Rager, B.; Weinstein, Y.; Rall, G.; Gopas, J.; et al. Measles Virus Persistent Infection of Human Induced Pluripotent Stem Cells. Cell. Reprogram. 2018, 20, 17–26. [Google Scholar] [CrossRef] [PubMed]
  155. Schneider-Schaulies, J.; Dunster, L.M.; Schwartz-Albiez, R.; Krohne, G.; ter Meulen, V. Physical Association of Moesin and CD46 as a Receptor Complex for Measles Virus. J. Virol. 1995, 69, 2248–2256. [Google Scholar] [CrossRef] [PubMed]
  156. Dunster, L.M.; Schneider-Schaulies, J.; Löffler, S.; Lankes, W.; Schwartz-Albiez, R.; Lottspeich, F.; ter Meulen, V. Moesin: A Cell Membrane Protein Linked with Susceptibility to Measles Virus Infection. Virology 1994, 198, 265–274. [Google Scholar] [CrossRef] [PubMed]
  157. Kawasaki, H.; Kosugi, I.; Arai, Y.; Iwashita, T.; Tsutsui, Y. Mouse Embryonic Stem Cells Inhibit Murine Cytomegalovirus Infection through a Multi-Step Process. PLoS ONE 2011, 6, e17492. [Google Scholar] [CrossRef]
  158. Lane, R.K.; Guo, H.; Fisher, A.D.; Diep, J.; Lai, Z.; Chen, Y.; Upton, J.W.; Carette, J.; Mocarski, E.S.; Kaiser, W.J. Necroptosis-Based CRISPR Knockout Screen Reveals Neuropilin-1 as a Critical Host Factor for Early Stages of Murine Cytomegalovirus Infection. Proc. Natl. Acad. Sci. USA 2020, 117, 20109–20116. [Google Scholar] [CrossRef]
  159. Swartzendruber, D.E.; Lehman, J.M. Neoplastic Differentiation: Interaction of Simian Virus 40 and Polyoma Virus with Murine Teratocarcinoma Cells in Vitro. J. Cell. Physiol. 1975, 85, 179–187. [Google Scholar] [CrossRef]
  160. Cremisi, C.; Babinet, C. Negative Regulation of Early Polyomavirus Expression in Mouse Embryonal Carcinoma Cells. J. Virol. 1986, 59, 761–763. [Google Scholar] [CrossRef]
  161. Mayberry, C.L.; Bond, A.C.; Wilczek, M.P.; Mehmood, K.; Maginnis, M.S. Sending Mixed Signals: Polyomavirus Entry and Trafficking. Curr. Opin. Virol. 2021, 47, 95–105. [Google Scholar] [CrossRef] [PubMed]
  162. Kwak, D.H.; Seo, B.B.; Chang, K.T.; Choo, Y.K. Roles of Gangliosides in Mouse Embryogenesis and Embryonic Stem Cell Differentiation. Exp. Mol. Med. 2011, 43, 379–388. [Google Scholar] [CrossRef] [PubMed]
  163. Graziano, V.R.; Walker, F.C.; Kennedy, E.A.; Wei, J.; Ettayebi, K.; Strine, M.S.; Filler, R.B.; Hassan, E.; Hsieh, L.L.; Kim, A.S.; et al. CD300lf Is the Primary Physiologic Receptor of Murine Norovirus but Not Human Norovirus. PLoS Pathog. 2020, 16, e1008242. [Google Scholar] [CrossRef] [PubMed]
  164. Villa, N.Y.; Bais, S.; Chan, W.M.; Meacham, A.M.; Wise, E.; Rahman, M.M.; Moreb, J.S.; Rosenau, E.H.; Wingard, J.R.; McFadden, G.; et al. Ex Vivo Virotherapy with Myxoma Virus Does Not Impair Hematopoietic Stem and Progenitor Cells. Cytotherapy 2016, 18, 465–480. [Google Scholar] [CrossRef]
  165. Zhang, L.; Bukreyev, A.; Thompson, C.I.; Watson, B.; Peeples, M.E.; Collins, P.L.; Pickles, R.J. Infection of Ciliated Cells by Human Parainfluenza Virus Type 3 in an In Vitro Model of Human Airway Epithelium. J. Virol. 2005, 79, 1113–1124. [Google Scholar] [CrossRef] [PubMed]
  166. Battles, M.B.; McLellan, J.S. Respiratory Syncytial Virus Entry and How to Block It. Nat. Rev. Microbiol. 2019, 17, 233–245. [Google Scholar] [CrossRef] [PubMed]
  167. Bilz, N.C.; Willscher, E.; Binder, H.; Böhnke, J.; Stanifer, M.L.; Hübner, D.; Boulant, S.; Liebert, U.G.; Claus, C. Teratogenic Rubella Virus Alters the Endodermal Differentiation Capacity of Human Induced Pluripotent Stem Cells. Cells 2019, 8, 870. [Google Scholar] [CrossRef]
  168. Mori, Y.; Sakata, M.; Sakai, S.; Okamoto, T.; Nakatsu, Y.; Taguwa, S.; Otsuki, N.; Maeda, Y.; Hanada, K.; Matsuura, Y.; et al. Membrane Sphingomyelin in Host Cells Is Essential for Nucleocapsid Penetration into the Cytoplasm after Hemifusion during Rubella Virus Entry. mBio 2022, 13, e01698-22. [Google Scholar] [CrossRef]
  169. Chen, J.; Neil, J.A.; Tan, J.P.; Rudraraju, R.; Mohenska, M.; Sun, Y.B.Y.; Walters, E.; Bediaga, N.G.; Sun, G.; Zhou, Y.; et al. A Placental Model of SARS-CoV-2 Infection Reveals ACE2-Dependent Susceptibility and Differentiation Impairment in Syncytiotrophoblasts. Nat. Cell Biol. 2023, 25, 1223–1234. [Google Scholar] [CrossRef]
  170. Li, W.; Moore, M.J.; Vasilieva, N.; Sui, J.; Wong, S.K.; Berne, M.A.; Somasundaran, M.; Sullivan, J.L.; Luzuriaga, K.; Greenough, T.C.; et al. Angiotensin-Converting Enzyme 2 Is a Functional Receptor for the SARS Coronavirus. Nature 2003, 426, 450–454. [Google Scholar] [CrossRef]
  171. Glowacka, I.; Bertram, S.; Müller, M.A.; Allen, P.; Soilleux, E.; Pfefferle, S.; Steffen, I.; Tsegaye, T.S.; He, Y.; Gnirss, K.; et al. Evidence That TMPRSS2 Activates the Severe Acute Respiratory Syndrome Coronavirus Spike Protein for Membrane Fusion and Reduces Viral Control by the Humoral Immune Response. J. Virol. 2011, 85, 4122–4134. [Google Scholar] [CrossRef] [PubMed]
  172. Wybenga, L.E.; Epand, R.F.; Nir, S.; Chu, J.W.; Sharom, F.J.; Flanagan, T.D.; Epand, R.M. Glycophorin as a Receptor for Sendai Virus. Biochemistry 1996, 35, 9513–9518. [Google Scholar] [CrossRef] [PubMed]
  173. Bitzer, M.; Lauer, U.; Baumann, C.; Spiegel, M.; Gregor, M.; Neubert, W.J. Sendai Virus Efficiently Infects Cells via the Asialoglycoprotein Receptor and Requires the Presence of Cleaved F0 Precursor Proteins for This Alternative Route of Cell Entry. J. Virol. 1997, 71, 5481–5486. [Google Scholar] [CrossRef] [PubMed]
  174. Tsai, B.; Gilbert, J.M.; Stehle, T.; Lencer, W.; Benjamin, T.L.; Rapoport, T.A. Gangliosides Are Receptors for Murine Polyoma Virus and SV40. EMBO J. 2003, 22, 4346–4355. [Google Scholar] [CrossRef] [PubMed]
  175. Caruso, M.; Cavaldesi, M.; Gentile, M.; Sthandier, O.; Amati, P.; Garcia, M.-I. Role of Sialic Acid-Containing Molecules and the Alpha4beta1 Integrin Receptor in the Early Steps of Polyomavirus Infection. J. Gen. Virol. 2003, 84, 2927–2936. [Google Scholar] [CrossRef] [PubMed]
  176. Fried, H.; Cahan, L.D.; Paulson, J.C. Polyoma Virus Recognizes Specific Sialyligosaccharide Receptors on Host Cells. Virology 1981, 109, 188–192. [Google Scholar] [CrossRef] [PubMed]
  177. Rose, P.P.; Hanna, S.L.; Spiridigliozzi, A.; Wannissorn, N.; Beiting, D.P.; Ross, S.R.; Hardy, R.W.; Bambina, S.A.; Heise, M.T.; Cherry, S. Natural Resistance-Associated Macrophage Protein Is a Cellular Receptor for Sindbis Virus in Both Insect and Mammalian Hosts. Cell Host Microbe 2011, 10, 97–104. [Google Scholar] [CrossRef] [PubMed]
  178. Clark, L.E.; Clark, S.A.; Lin, C.; Liu, J.; Coscia, A.; Nabel, K.G.; Yang, P.; Neel, D.V.; Lee, H.; Brusic, V.; et al. VLDLR and ApoER2 Are Receptors for Multiple Alphaviruses. Nature 2022, 602, 475–480. [Google Scholar] [CrossRef]
  179. Lipton, H.L.; Kumar, A.S.M.; Hertzler, S.; Reddi, H.V. Differential Usage of Carbohydrate Co-Receptors Influences Cellular Tropism of Theiler’s Murine Encephalomyelitis Virus Infection of the Central Nervous System. Glycoconj. J. 2006, 23, 39–49. [Google Scholar] [CrossRef]
  180. Fan, W.; Qian, P.; Wang, D.; Zhi, X.; Wei, Y.; Chen, H.; Li, X. Integrin Avβ3 Promotes Infection by Japanese Encephalitis Virus. Res. Vet. Sci. 2017, 111, 67–74. [Google Scholar] [CrossRef]
  181. Oliveira, L.G.; Peron, J.P.S. Viral Receptors for Flaviviruses: Not Only Gatekeepers. J. Leukoc. Biol. 2019, 106, 695–701. [Google Scholar] [CrossRef] [PubMed]
  182. Laureti, M.; Narayanan, D.; Rodriguez-Andres, J.; Fazakerley, J.K.; Kedzierski, L. Flavivirus Receptors: Diversity, Identity, and Cell Entry. Front. Immunol. 2018, 9, 2180. [Google Scholar] [CrossRef] [PubMed]
  183. Zhang, Y.; Yan, Y.; Li, S.; Yuan, F.; Wen, D.; Jia, N.; Xiong, T.; Zhang, X.; Zheng, A. Broad Host Tropism of Flaviviruses during the Entry Stage. Microbiol. Spectr. 2023, 11, e0528122. [Google Scholar] [CrossRef] [PubMed]
  184. Sinzger, C.; Digel, M.; Jahn, G. Cytomegalovirus Cell Tropism. Curr. Top. Microbiol. Immunol. 2008, 325, 63–83. [Google Scholar] [CrossRef]
Figure 1. Overview of the antiviral responses of somatic vs. pluripotent cells. (A) Somatic cells have specialised cytoplasmic receptors that sense viral double-stranded RNAs (dsRNAs). These sensors include RIG-I and MDA5, which signal through MAVS to activate the transcription factors IRF3/7, leading to the expression of type I IFNs. Secreted IFNs bind the IFNAR1 and 2 receptors, and through JAK/STAT signalling, lead to the expression of hundreds of ISGs and establishment of an antiviral state. (B) Contrary to somatic cells, pluripotent cells lack a functional IFN response and fail to produce type I IFNs upon challenge with viruses or viral mimics. Instead, pluripotent cells rely on alternative strategies, including (1) RNA interference, where viral dsRNAs are cleaved by Dicer to generate antiviral siRNAs, which are loaded onto the RNA-induced silencing complex (RISC), (2) RNase H1-mediated degradation of viral RNA (ERASE) after generation of viral DNA/RNA hybrids and, (3) constitutive or intrinsic expression of a subset of ISGs.
Figure 1. Overview of the antiviral responses of somatic vs. pluripotent cells. (A) Somatic cells have specialised cytoplasmic receptors that sense viral double-stranded RNAs (dsRNAs). These sensors include RIG-I and MDA5, which signal through MAVS to activate the transcription factors IRF3/7, leading to the expression of type I IFNs. Secreted IFNs bind the IFNAR1 and 2 receptors, and through JAK/STAT signalling, lead to the expression of hundreds of ISGs and establishment of an antiviral state. (B) Contrary to somatic cells, pluripotent cells lack a functional IFN response and fail to produce type I IFNs upon challenge with viruses or viral mimics. Instead, pluripotent cells rely on alternative strategies, including (1) RNA interference, where viral dsRNAs are cleaved by Dicer to generate antiviral siRNAs, which are loaded onto the RNA-induced silencing complex (RISC), (2) RNase H1-mediated degradation of viral RNA (ERASE) after generation of viral DNA/RNA hybrids and, (3) constitutive or intrinsic expression of a subset of ISGs.
Viruses 16 00173 g001
Figure 2. Summary of virus infectivity and (co-) receptor expression in pluripotent human and mouse cells. The figure shows a correlation between susceptibility to viral infections and expression of the viral (co-) receptors. This correlation suggests that the ability of a virus to establish a successful infection is also determined by the ability to enter (grey triangles depict a level of receptor expression or infectivity).
Figure 2. Summary of virus infectivity and (co-) receptor expression in pluripotent human and mouse cells. The figure shows a correlation between susceptibility to viral infections and expression of the viral (co-) receptors. This correlation suggests that the ability of a virus to establish a successful infection is also determined by the ability to enter (grey triangles depict a level of receptor expression or infectivity).
Viruses 16 00173 g002
Table 1. List of viruses used in pluripotent human and mouse cells, including ESCs, iPSCs and teratocarcinoma-derived cells. The table shows the level of infectivity in the pluripotent human and mouse cells, the (co-) receptors known for each virus and the expression levels (mRNA) of each receptor in human and mouse ESCs.
Table 1. List of viruses used in pluripotent human and mouse cells, including ESCs, iPSCs and teratocarcinoma-derived cells. The table shows the level of infectivity in the pluripotent human and mouse cells, the (co-) receptors known for each virus and the expression levels (mRNA) of each receptor in human and mouse ESCs.
VirusInfectivity in Pluripotent CellsReferences InfectivityReceptorsExpression in Pluripotent Cells References
Receptors/Expression
Human 1Mouse 1Human 2Mouse 2
Adeno-associated virus 2
(AAV2)
++++++[122]Heparan sulfate proteoglycans++++++[123,124,125]
AAVR++++++
Adenovirus
(Ad5)
+++
+++
+++
ND
[122,126]CAR++++++[127,128]
VCAM-1++
Heparan sulfate proteoglycans++++++
Scavenger receptor A++++++
Coxsackie B virus
(CVB1&3)
++
++
ND
ND
[129,130,131]CAR++++++[129,132]
DAF++++++
Encephalomyocarditis virus
(EMCV)
ND+++[29]Adam9++++++[133,134,135,136,137]
Sialic acid (not essential for all strains)++
VCAM-1 (only relevant for specific cell types)++
Herpex simplex virus
(HSV-1)
ND
++
+++
ND
[138,139]Nectin-1++++++[140]
Human Cytomegalo
virus
(hCMV)
-
-
-
-
ND
ND
[141,142,143]PDGFRα++++[144,145]
EGFR++++
Neurophilin2 (Nrp2)++++++
CD46++++
BSG (CD147)++++++
Thy1 (CD90)+++++
Heparan sulfate proteoglycans (HSPG)++++++
αvβ3 Integrin ++++
CD13+++++
CD151++++++
Human immunodeficiency virus
(HIV1/2)
-ND[146,147]CD4+++[148,149]
CCR5-+
CXCR4+++
Indiana vesiculovirus (VSV)+++ND[32]LDLR++++++[150]
HSP90B1 (Gp96)++++++
TLR4 ++
TLR13?+
Influenza A virus
(IAV)
ND
+++
+++
+++
ND
ND
[17,32,151]Sialic Acid α2,3++[135,136,152]
La Crosse encephalitis virus
(LACV)
ND+++[13]C-type lectins??[153]
DC-SIGN (cd209)++
Mincle (Clec4E)++
Dectin-1 (Clec7a)+-
Dectin-2 (Clec6a)+?
Measles virus
(MV)
++ND[131,154] MSN++++++[155,156]
CD46+++++
Mouse Cytomegalo
virus
(mCMV)
ND
ND
+
+
[141,157]Neurophilin-1 (Nrp1)++++[158]
Mouse Polyoma virus
(mPyV)
ND
ND
-
-
[159,160]GT1a?-[135,136,161,162]
GD1a?-
GT1b?+
Glycosaminoglycans++
ITGA4 (α4β1 integrin)+++
Murine Norovirus
(MNV1/3)
ND-Personal observation CD300lf-+[163]
Myxomavirus
(MyxV)
-ND[164] Unknown
Nodamura virus
(NoV)
++ND[29]Unknown
Parainfluenza virus type 3
(PIV3)
+++ND[32]Sialic acid α2-6++[135,165]
Nucleolin++++++
Respiratory syncytial virus
(RSV)
+ND[32]Glycosaminoglycans++[166]
Nucleolin++++++
CX3CR1++
Rubella virus
(RV)
++
++
ND
ND
[131,167]Sphingomyelin (SGMS1)++++++[168]
SARS-CoV-2++ND[169]ACE2+++[170,171]
TMPRSS2++++
Sendai virus
(SeV)
ND+++[13]GYPA++[172,173]
ASGR1++++
Fucosylated glycans++
Sialic acid α2-3++
Simian vacuolating
virus 40
(SV40)
ND-[159]Ganglioside GM1?-[135,136,161,162,174,175,176]
ITGA4 (α4β1 integrin)+++
Sialic acid++++
glycosaminoglycans++
Sindbis virus
(SINV)
ND+++Personal observation NRAMP2 (SLC11A2)++++++[177,178]
VLDLR+++++
LRP8 (ApoER2)++++++
Theiler’s encephalo
myelitis virus
(TMEV(GDVII)
ND+++[17]Heparan sulfate (essential co-receptor)++++++[179]
West Nile virus (WNV)
Yellow fever virus
(YFV)
Zika virus
(ZIKV)
Dengue virus (DENV)
-
-

+

-
+
ND

ND

ND
[13,32]αvβ3 Integrin/ITGAV++++++[180,181,182]
DC-Sign (CD209)++
HAVCR1 (TIM-1)-+
TIMD4 (TIM-4)-
AXL+++++
TYRO3++++++
Mertk++
Varicella zoster virus (VZV)-ND[139]αV integrin/ITGAV++++++[140]
1 Infectivity score for human and mouse pluripotent cells was obtained using data generated by several methods, including flow cytometry, immunofluorescence and vRNA/vDNA expression data. Infectivity is classified as follows: (-) non-infectious; (+) poorly infectious; (++) infectious; (+++) highly susceptible; (ND) not determined. 2 Expression levels of the known (co-) receptors in human and mouse pluripotent cells were extracted from RNA-seq data [32,120]. To this end, log2 normalised counts after DESeq2 analysis were used. Receptor level expression is classified as follows: (-) not expressed (log2 norm counts < 0.3); (+) lowly expressed (log2 norm counts 0.3–3); (++) expressed (log2 norm counts 3–8); (+++) highly expressed (log2 norm counts >8); (?) if the expression is unknown, or if the gene has no homologous in that particular species.
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

Mueller, F.; Witteveldt, J.; Macias, S. Antiviral Defence Mechanisms during Early Mammalian Development. Viruses 2024, 16, 173. https://doi.org/10.3390/v16020173

AMA Style

Mueller F, Witteveldt J, Macias S. Antiviral Defence Mechanisms during Early Mammalian Development. Viruses. 2024; 16(2):173. https://doi.org/10.3390/v16020173

Chicago/Turabian Style

Mueller, Felix, Jeroen Witteveldt, and Sara Macias. 2024. "Antiviral Defence Mechanisms during Early Mammalian Development" Viruses 16, no. 2: 173. https://doi.org/10.3390/v16020173

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