Open access peer-reviewed chapter

Exosomes and Their Role in Viral Infections

Written By

Gulfaraz Khan, Waqar Ahmed and Pretty S. Philip

Submitted: 31 October 2016 Reviewed: 24 April 2017 Published: 12 July 2017

DOI: 10.5772/intechopen.69397

From the Edited Volume

Novel Implications of Exosomes in Diagnosis and Treatment of Cancer and Infectious Diseases

Edited by Jin Wang

Chapter metrics overview

2,653 Chapter Downloads

View Full Metrics

Abstract

Exosomes are excretory nano-vesicles that are formed by the cell’s endocytic system and shed from the surface of almost all types of cells. These tiny extracellular vesicles, once thought to be “garbage bags for cells,” carry a wide variety of molecules of cellular origin, including proteins, lipids, and RNAs, that are selectively incorporated during the formation of exosomes. Exosomes are now known to play a central role in several important biological processes such as cellular communication, intercellular transfer of bioactive molecules, and immune modulation. Recent advances in the field have shown that a number of animal viruses can exploit the exosomal pathway by incorporating specific cellular or viral factors within exosomes, in order to modulate the cellular microenvironment and influence downstream processes such as host immunity and virus spread. In this chapter, we provide an overview of our current understanding of exosome biogenesis and how this normal physiological process is hijacked by some pathogenic viruses. Viral components that appear to be selectively incorporated into exosomes and the potential role of these exosomes in viral pathogenesis are discussed. Identifying viral signatures in exosomes and their mode of action is fundamental for any future diagnostic and therapeutic strategies for viral infections.

Keywords

  • exosomes
  • viruses
  • immune modulation
  • pathogenesis
  • biomarkers

1. Introduction

Exosomes are nano-secretory vesicles ranging in size from 30 to 100 nm and having a density between 1.13 and 1.19 g/ml [1]. Exosomes are derived from the cell’s endosomal pathway, and their membranes are rich in lipids such as sphingolipids, ceramide, and cholesterol [2]. These tiny vesicles are released by virtually all cell types, but at varying degree, upon fusion of multivesicular bodies (MVBs) with the plasma membrane [35]. It is now well established that exosomes are not the cell’s trash bags, as initially thought, but rather they serve as important nano-vehicles for the transport of specific cargo in and out of the cells [6]. Depending on their cargo, exosomes can mediate certain intercellular communication processes [7, 8]. Although the mechanism of how this cargo is selected for packaging into these vesicles destined for excretion remains poorly understood, it is believed that the endosomal membranes play a central role in this process [9, 10].

A number of molecular mechanisms are involved in the formation of intraluminal vesicles (ILVs) and multivesicular bodies (MVBs) in a cell. One of the best studied and well-characterized group of proteins involved in this process belong to the ESCRT (endosomal sorting complex required for transport) family of proteins [1113]. These proteins are believed to play a role in the inward budding and scission of ILVs. One of the mechanism by which viruses hijack the exosome pathway is by directly interfering with the machinery involved in exosome biogenesis, such as the ESCRT proteins [14]. Others such as the oligomerization of the tetraspanin complexes [15], the sphingomyelinase pathways [16], phospholipase D2, and ADP ribosylation factor-6–mediated pathways have also been reported to be involved in the ILV budding process [17]. Another family of proteins that are essential for vesicular formation, trafficking, and fusion in eukaryotic cells belongs to a large family of highly conserved proteins known as Rab GTPases [18]. A number of Rab proteins such as Rab5 and Rab7 have been shown to be important in endosome maturation and sorting of material in the ILVs [19, 20]. Rab27 a/b are involved in the fusion of the ILVs with the plasma membrane and release of exosomes [50]. A number of other Rab GTPases are also found to play an instrumental role in exosome release. Depending on the cell type, Rab5, Rab7, Rab11, Rab27, and Rab35 have all been implicated in the release of vesicles. Altering the levels of any of these Rabs may lead to interference with progression of exosomal cargo at specific endocytic locations [20]. The fact that the exosomal pathway has some similarities with certain phases of viral life cycle has led to the observations that a number of viruses can indeed hijack the exosome pathway during their replication and pathogenesis [21, 22].

Advertisement

2. Viruses and the exosomal pathway

The endocytic pathway and the budding of viruses, especially enveloped viruses, share many common features. Both processes require generation of membrane curving, packaging of specific cargo, and membrane budding for release from the cell [22]. What is most surprising is that different viruses with very different evolutionary paths appear to converge in their use of the host endocytic pathway in the entry and exit from their host cells [23]. The receptor or clathrin-mediated endocytosis to enter the cell is found to be utilized by a number of viruses of the Flaviviridae family, which includes medically important pathogens such as hepatitis C (HCV), West Nile (WNV), Dengue, and Zika viruses [2427]. These viruses can enter the late endosomes and then fuse with the ILVs within the endosome compartments [28]. Recently, it was shown that HCV can incorporate its full-length RNA genome into the ILVs and be excreted out via exosomes, and retain infectivity [29, 30]. Since HCV is fairly small, it is possible that HCV infectious particles could be released directly within exosomes and account for infection. However, the observation that exosomes isolated from HCV sub-genomic replicon cell lines lacking HCV structural proteins remained infectious argues against the notion that mature viral particles are released in exosomes [30]. Currently, HCV and hepatitis A virus (HAV) are the only viruses that have been shown to incorporate their full-length genomic RNA within exosomes [31]. Another virus that can utilize the endosomal/exosomal system to deliver viral cargo to uninfected cells is the human immunodeficiency virus (HIV-1). Based on the similarities between HIV-1 assembly and egress, and exosome biogenesis, Gould et al. proposed the “Trojan exosome hypothesis,” in which they suggested that HIV has evolved to exploit the exosome system to infect cells in the absence of the receptor-mediated interaction [32, 33]. This hypothesis is supported by the observation that HIV virions are released together with exosomes, but the infectivity is reduced in the absence of exosomes, implying that the process of exosome release from HIV-infected cells probably also contributes to the release of HIV virions. This mechanism was demonstrated using HIV-infected dendritic cells, which were able to transfer the virus to closely associated uninfected T cells via exosomes [34, 35]. Unlike HCV, direct packaging of HIV genomic RNA into exosomes has not been observed, probably reflecting the findings that HIV predominately buds from the plasma membrane and not from the endosomal pathway [3638].

2.1. Viruses hijack the ESCRT and Rab GTPases involved in exosome biogenesis

Viruses are obligate intracellular parasites that hijack cellular pathways to complete their life cycle. In recent years, an accumulating body of data has emerged suggesting that some viruses can also manipulate with the vesicular trafficking machinery for their assembly, egress, and transmission [39, 40]. For example, HIV has been shown to exploit the ESCRT, lipid raft domains, and Rab GTPases components, all of which are involved in exosome biogenesis [23, 41, 42]. Specifically, HIV Gag has been shown to interact with exosomal tetraspanins, especially CD63 and CD81, to aid in virion egress [42]. Using electron microscopy, human herpesvirus 6 (HHV-6) virions have been shown to be present in MVBs and egress together with exosomes through the same pathway [43]. HHV-6 infection dramatically increases MVB formation, suggesting that the endosomal pathway is likely to be important for HHV-6 infection and assembly [43]. Furthermore, HHV-6 glycoprotein gB was found to co-localize with CD63 [43], but the importance of this association for virus egress remains to be demonstrated. Besides interfering with the ESCRT pathway, some viruses can also utilize the Rab GTPase complexes to assist in their replication and egress processes. Several negative strand RNA viruses, such as influenza A virus (IAV), hantavirus, and respiratory syncytial virus (RSV), have all been reported to utilize the Rab pathway for their transport to the plasma membrane for exit [4447]. It is known that interfering with Rab11 levels can inhibit or promote the release of exosome-containing contents such as transferrin, HSP-70, flotillin, and anthrax toxin [44, 48, 49]. In the case of hantavirus-infected cells, depletion of Rab11 results in a tenfold reduction in virion production [46]. Similarly, IAV and RSV also appear to hijack the Rab11 pathway to their benefit [45, 47]. Rab27a, another member of the Rab GTPase family, has also been shown to be essential for exosome biogenesis, particularly in the steps involving the fusion of MVBs with plasma membrane for the final release of exosomes [5051]. For example, in cytomegalovirus (CMV)-infected cells, the levels of Rab27a are increased and co-localized with the viral envelope components at assembly sites in the cytoplasm [52], but the molecular mechanisms and ultimate changes to exosome production remain to be elucidated. HIV proteins are also found to interact with Rab27a resulting in increased levels of exosome formation [41, 53]. Herpes simplex virus 1 (HSV-1) is another virus that appears to use Rab27a for its intracellular transport and exocytosis [54, 55]. Depletion or down-regulation of Rab27a leads to decrease in HSV-1 viral production [54, 55].

The regulatory functions of the Rab GTPase mentioned above are still not fully understood. However, it is widely accepted that cells react to stimuli to adjust the distribution and levels of intracellular proteins as well as their degradation, secretion, and recycling [56]. Manipulation of specific steps in the endocytic pathway by viruses highlights the need for further research to unravel the complex interplay between regulators of the endocytic process and exosome release. Such studies may shed light to potential targets for anti-virals.

2.2. Viral signatures in exosomes

The discovery that certain features in the life cycle of viruses and the cellular endosomal/exosomal pathway are common, and that some viruses can exploit the exosomal pathway to their benefit, triggered a search to identify viral signatures in exosomes. This line of research has obvious downstream benefits, not only in terms of viral diagnosis, but also for understanding the mechanisms of viral-mediated pathogenesis. We now have a growing list of viral-specific components that have been identified in exosomes (Table 1). Moreover, functional analysis of excreted exosomes carrying viral components is beginning to shed light on how some viruses can modulate cellular processes as diverse as immune evasion, apoptosis, proliferation, and even viral infectivity (Table 1). In this context, one family of viruses that has been widely studied is the human herpesviruses. This family of viruses contains two members, namely Epstein-Barr virus (EBV) and Kaposi’s sarcoma virus (KSV), that are oncogenic and implicated in the pathogenesis of a number of human malignancies [57]. Both of these viruses have now been shown to exploit the exosome pathway to secrete various components ranging from proteins to various species of RNAs, including messenger RNAs (mRNA), microRNAs (miRNA), and small non-protein coding RNAs [5861]. In fact, viral miRNAs (vmiRNA) were first identified in EBV-infected cells [62] and subsequently shown to be excreted out of cells via exosomes [63]. It is now known that exosomes shed from EBV-infected cells contain a large number of viral miRNAs, most of which appear to be smaller products of larger BamH1 EBV transcripts [64, 65]. It is believed that these viral miRNAs, together with cellular miRNAs, play a role in modulating the expression of target genes in recipient cells (Figure 1) [59, 6567]. Recently, it was shown that the two non-protein coding EBV small RNAs, EBER-1 and EBER-2, are also consistently excreted from infected cells within exosomes [68]. EBERs are highly abundant EBV RNA polymerase II/III transcripts expressed in all EBV latently infected cells. The significance of their high abundance within infected cells, or the reasons for their release in exosomes, remains intriguing. One study showed that EBERs released from infected cells could induce innate immune responses via activation of Toll-like receptor 3-mediated signaling [69]. In addition to RNAs, a number of studies have shown that EBV-infected cells can also excrete viral-specific proteins, including the latent membrane protein 1 and 2A (LMP-1, LMP-2A) and the viral envelop glycoprotein 350 (gp350) [7073]. Export of these proteins via exosomes indicates another dimension to how EBV can modulate cellular processes, not only within the cells it infects, but also in the surrounding cells.

VirusMain cellular targetViral cargo reported in exosomesPotential effect of viral exosomesReferences
EBVLymphocytesLMP1, 2A, gp350, vmiRNA, EBERs, vRNAProliferation, apoptosis, immune evasion, viral reactivation[63, 68, 70, 72, 73, 92]
HSV-1Epithelial cellsVP16, HSV gB, ICP 127, vmiRNAIncrease infectivity, viral spread, and latency[116, 128]
CMVWBC, epithelial cellsCMV gBInfection of myeloid dendritic cells, increased viral infectivity[129]
HHV-8WBC, endothelial cellsvmiRNA, vRNAImmune modulation, cell metabolism[60, 61]
HIV-1LymphocytesvmiRTAR, vmiRNA, NefInhibition of apoptosis, stimulate proinflammatory cytokines, down-regulation of CD4 and MHC I, increased susceptibility of naïve T cells, antiviral activity[81, 82, 130, 131]
HTLV-1LymphocytesTax vmRNA, TAX, vmiRNAProinflammatory cytokines, damage to neurons[86, 132, 133]
HPVEpithelial cellsvmiRNAProliferation, apoptosis[134]
HAVHepatocytesHAV gRNA, HAV particlesImmune evasion, increased viral infectivity[31, 117, 135]
HBVHepatocytesvDNA, vRNA, HBsAgImmune evasion[118, 136]
HCVHepatocytesHCV gRNA, vmiRNA, vRNAImmune evasion[29, 124, 137]
RVFVWBCv-protein, vmRNAApoptosis, immune evasion[138]

Table 1.

Exosomes, their viral cargo, and their potential role in virus-mediated pathogenesis.

Viral-infected cells have been shown to shed exosomes containing cellular and viral-specific components. Table lists viral components that have been detected in exosomes. These include viral mRNAs, microRNAs (vmiRNA), non-protein coding RNAs (vRNA), full-length genomic RNA (gRNA), as well as virus-specific proteins. Depending on the exosomal cargo and type of recipient cells, different biological changes may be induced. Abbreviations: EBV, Epstein-Barr virus; HSV, herpes simplex virus; CMV, cytomegalovirus; HIV, human immunodeficiency virus; HTLV, human T-lymphotropic virus; HPV, human papillomavirus; HAV, hepatitis A virus; HBV, hepatitis B virus; HCV, hepatitis C virus; RVFV, Rift Valley fever virus.

Figure 1.

Viruses hijacking the exosomal pathway. Many different viruses have been shown to exploit the exosomal pathway to aid in their infection, spread, and pathogenesis. Three examples are illustrated here. EBV, a dsDNA virus of the herpes family, has been shown to export numerous viral microRNA (vmiRNA), viral mRNA, non-protein coding RNAs (EBERSs), latent membrane proteins (LMP-1 and 2A), and the envelop glycoprotein (gp350). Similarly, other viruses such as HIV, hepatitis A, B and C can also package their proteins and RNAs in exosomes. For HAV and HCV, full-length genomic RNA has been shown to be present in exosomes, which in the case of HCV has been demonstrated to be infectious and capable of producing virus particles.

Another virus which has attracted considerable attention is HIV. There are over 38 million people living with HIV, and there is still no cure [74]. Analysis of exosomes released from HIV-1-infected and non-infected cells shows that they differ in their densities [75]. This implies that the contents of the exosomes from infected and non-infected cells are clearly different [60, 61]. Although retroviruses are much smaller than herpesviruses, they are nevertheless still slightly larger than exosomes and as such it is unlikely that mature infectious HIV-1 particles could be packaged and excreted within exosomes. However, there is mounting evidence that HIV-1 egress is partly mediated by the endosomal pathway, and both exosomes and HIV-1 are released together in the same fraction [76]. HIV-1 Gag protein has been shown to interact with the exosomal membrane tetraspanins, CD63 and CD81, aiding in the assembly and exit of HIV-1 from infected cells [7780]. Moreover, several functionally active HIV-1 components have also been shown to be excreted out of infected cells using the exosomal “bus” (Figure 1). Once released, exosomes can bind to neighboring cells, travel passively through the blood stream to distant sites, and induce biological changes depending on the nature of the cargo they carry (Figure 1) [42]. Nef is one HIV-1 protein that has been shown to be released within exosomes [81]. Studies indicate that Nef plays an important role in activating resting bystander CD4+ T cells making them susceptible to HIV infection and viral replication [8183]. Reports have indicated that HIV-1 may also facilitate its spread to other cells by secreting viral co-receptors, CCR5 and CXCR4, in exosomes [84, 85]. In addition to functional proteins, exosomes from HIV-1-infected cells have been shown to carry several viral miRNAs, including vmiRTAR transcripts, vmiR88 and vmiR99 [23]. Similarly, another human retrovirus, the human T-lymphotropic virus 1 (HTLV-1), also appears to export viral components via the exosomal transport systems. Exosomes released from HTLV-1-infected cells contain not only viral mRNA transcripts, such as those for Tax, HBZ, and Env, but also the biologically active trans-activator protein, Tax [86, 87]. Moreover, HTLV-1 Tax protein has been demonstrated in exosomes isolated from cerebrospinal fluid of patients with HTLV-1-associated myelopathy/tropical spastic paraparesis, suggesting that HTLV-1 may modulate its microenvironment by selective secretion of specific viral cargo [88].

The list of components of cellular and microbial origin detected in exosomes is constantly expanding. This has led to the establishment of several online databases to catalogue the contents of exosomes. There is now substantial evidence indicating that many different types of pathogens, including bacteria, viruses, parasites, and even prions, can exploit the exosomal pathway [89, 90]. Of the viruses, members belonging to families as diverse as Bunyaviridae (enveloped RNA viruses) and Papillomaviridae (non-enveloped DNA viruses) have been shown to export their products in exosomes (Table 1). Moreover, studies are beginning to address the functional impact of exosomes carrying viral cargo in the pathogenesis of viral infections. One major challenge is to understand the mechanisms that regulate the selection of cargo to be packaged into exosomes and how we can use exosomes as biomarkers for viral infections and disease progression [89, 91].

Advertisement

3. Role of exosomes in viral pathogenesis

Exosomes released by viral-infected cells contain not only viral components, but also those of cellular origin [23, 67, 89]. It appears that viruses are able not only to export their own products in exosomes, but also to somehow influence which cellular products are packaged within the excretory vesicles. This is evident by the findings that exosomal cargo of cellular origin is clearly different from non-infected cells of the same type [60, 61, 92]. Thus, any pathophysiological impact of viral exosomes on recipient cells is by no means due to viral components only. An accumulating body of data indicates that exosomes from viral-infected cells can induce processes as diverse as immune evasion, apoptosis, proliferation, transcellular spread, and cytokine modulation (Table 1). The molecular details of how these processes are triggered are poorly understood and most probably dependent on multiple factors, including the type of cells releasing/receiving exosomes, nature of the exosomal cargo, mode of delivery, and stage of infection [1, 90]. This probably explains why apparently contradicting results have been reported in different studies [93, 94]. For EBV, it has been shown that uninfected epithelial cells exposed to exosomes derived from infected B cells are internalized via caveolar-dependent endocytosis and induce physiological changes in these cells [95]. Studies reported that exosomes derived from nasopharyngeal carcinoma (NPC) and from EBV-immortalized lymphoblastoid cell lines (LCLs) either inhibit proliferation of EBV-reactive CD4+ cells or induce apoptosis [70, 93, 96]. Similar results were observed with exosomes isolated from EBV-associated NPC patients and mice xenografted with NPC [97]. These pathophysiological changes were suggested to be due to viral and cellular components such as LMP-1, LMP-2A, viral miRNAs, and cellular galectin 9, excreted in exosomes from EBV-infected cells [72, 93, 97, 98]. The finding of EBV LMP-1 in exosomes is noteworthy [70, 93]. This is a well-known oncoprotein that plays a key role in the immortalization of EBV-infected cells [99]. Not surprisingly, LMP-1 has been extensively studied and shown to function as a constitutively activated receptor, signaling through the TRAF pathway leading to the activation of the master transcription factor, NFκB [100, 101]. LMP-2A is also an EBV latent protein expressed on the plasma membrane of latently infected cells [102]. Like LMP-1, LMP-2A also appears to be a constitutively activated receptor; while LMP-1 mimics CD40 receptor, LMP-2A mimics activated B-cell receptor (BCR), allowing infected cells to develop and survive, even in the absence of BCR signaling [103, 104]. Although it is not known how these membrane proteins are selected for export in exosomes, or what their functional impact is on recipient cells, it is tempting to postulate that the cell survival signals provided by LMP-2A and cell proliferation signals provided by LMP-1, if transferable to recipient cells, would be important in EBV pathogenesis.

Recently, we reported that exosomes from both EBV-infected and non-infected B cells are taken up by recipient cells, but only the exosomes from EBV-infected cells induced apoptosis in recipient cells in a dose-dependent manner [92]. We further showed that apoptosis was induced via the activation of the extrinsic pathway involving Fas-ligand (Fas-L) present in EBV exosomes. Moreover, the process could be blocked by using anti-Fas-L antibodies [92]. Another study reported that LCL-derived exosomes contain Fas-L and MHCII molecules and induce apoptosis in autologous CD4+ T cells [96]. Taken together, these studies indicate that one mechanism by which EBV could evade the body’s immune system may be by shedding exosomes containing signals that inhibit proliferation and/or promote apoptosis of anti–EBV-infiltrating lymphocytes. The fact that similar effects on bystander cells, albeit through different mechanisms, have also been reported for exosomes released from rotavirus and HIV-1–infected cells [105, 106] supports this hypothesis.

Another well-known mechanism by which some viruses can evade the immune responses is by down-regulating the expression of viral lytic genes and persisting in the infected cells in a latent state [107]. It’s a simple strategy; no viral antigens expressed in infected cells means no immune system can be triggered [108]. In this context, herpesviruses are among the most extensively studied [109, 110]. For example, herpes simplex type 1 (HSV-1) replicates in mucosal epithelial cells during primary infection and then enters sensory neurons where it establishes life-long latency [111]. During the latent state, although no viral proteins are expressed, numerous vmiRNAs have been detected, and some of these vmiRNAs appear to be central in suppressing viral gene expression and maintaining latency [112114]. The complexity of this process has been further exposed by recent findings indicating that HSV-1 can excrete vmiRNAs in exosomes, which on transfer to recipient cells, can suppress viral gene expression and viral spread to uninfected cells [115]. Furthermore, HSV-1 can also transfer antiviral factors, such as STING (stimulator of IFN genes), to suppress its cell-to-cell spread in circumstances that may be unfavorable [116]. Thus, inhibiting viral replication and spread in the face of a competent immune threat could be an important strategy for viruses to escape immune elimination and persist.

In some viral infections, such as with hepatitis B (HBV), non-infectious subviral particles are released into the serum, often at levels 1000s of fold higher compared to mature infectious particles [117, 118]. In evolutionary terms, it does not make sense why a virus would opt to shed enormous amounts of non-infectious subviral particles if it was not beneficial for the virus. One plausible hypothesis is that such subviral particles act as a decoy to divert the immune responses away from the bonafide infectious virions [118, 119]. HSV-1-infected cells can also release subviral particles, referred to as the L-particles. These particles have neither viral capsid nor viral DNA, and they are not infectious, but they do contain several HSV proteins [120, 121]. Recent studies suggest that the transfer of L particles to bystander cells can modulate the microenvironment to facilitate immune evasion and viral infection [122]. Similarly, there is evidence that some viruses can manipulate their microenvironment by secreting exosomes containing cargo that interferes with the host inflammatory and antiviral factors [119, 123].

In addition to immune modulation, exosomes released from some viral-infected cells can promote infection and enhance viral spread. A good example of this is HIV-1. Exosome-mediated transfer of HIV-1 co-receptors CCR5 and CXCR4 to recipient cells that do not normally express these receptors can facilitate HIV-1 infection in these cells [84, 85]. In the case of hepatitis C virus, it has been reported that infected cells release exosomes containing full-length viral genomic RNA as well as viral-specific proteins [29, 30, 124]. Importantly, HCV RNA carrying exosomes could transmit the infection to non-infected cells and establish a productive infection [29, 30, 124]. This receptor-independent mechanism of HCV transmission would prevent the virus from being exposed to antibodies that would normally be effective in neutralizing cell-free virus [125, 126]. Some viruses can also manipulate with the endocytic pathway, not for export of their cargo, but for virion assembly and egress from the infected cells during replication. For example, HSV-1 can interact with Rab27a via its tegument protein and its glycoproteins gH and gD [55]. Depletion of Rab27a results in significant reduction in both viral production and viral egress, highlighting the importance of the Rab27a in the egress of HSV-1 [55]. A similar phenomenon has been reported for several other members of the herpesviridae family, including cytomegalovirus (CMV) and human herpesvirus 6 (HHV-6) [23].

Although we have focused on how viruses exploit the exosome system to aid their infection and pathogenesis, it should be borne in mind that the shedding of exosomes is a normal physiological process and it also plays a role in defending against infections [90, 127]. For example, a recent study reported that exosomes isolated from semen, but not from blood of healthy individuals, were able to inhibit the replication of HIV-1 in in vitro culture [42]. Remarkably, this anti-viral activity of semen exosomes appeared to be restricted to retroviruses and had no effect on HSV-1 or HSV-2 replication [5]. Ironically, some viruses are able to not only overcome these defense mechanisms but also exploit them to their benefit.

Advertisement

4. Conclusions

Our current understanding of microvesicle biology and function, especially in regard to virus infections, is still in its early stages. The study of viral exosomes has shown that the transfer of viral and cellular factors in exosomes enables the manipulation of the neighboring unaffected cells. Microvesicle-mediated communication allows the virus to respond and control the cellular microenvironment. A number of reports suggest that viruses utilize the cellular vesiculation pathway for virus budding/assembly, immune evasion, and intercellular communication. Understanding the role of exosomes in the host-viral interactions can open new avenues of understanding the disease mechanisms and future diagnostic and therapeutic interventions.

Advertisement

Acknowledgments

The work in Khan Laboratory is supported by grants 31R90, 31016, and 31M263 from UAEU.

References

  1. 1. van der Pol E, Böing AN, Harrison P, Sturk A, Nieuwland R. Classification, functions, and clinical relevance of extracellular vesicles. Pharmacological Reviews. 2012;64:676-705
  2. 2. De Toro J, Herschlik L, Waldner C, Mongini C. Emerging roles of exosomes in normal and pathological conditions: New insights for diagnosis and therapeutic applications. Frontiers in Immunology. 2015;6:203
  3. 3. Lässer C, Seyed Alikhani V, Ekström K, Eldh M, Torregrosa Paredes P, Bossios A, Sjöstrand M, Gabrielsson S, Lötvall J, Valadi H. Human saliva, plasma and breast milk exosomes contain RNA: Uptake by macrophages. Journal of Translational Medicine. 2011;9:9
  4. 4. Street JM, Barran PE, Mackay CL, Weidt S, Balmforth C, Walsh TS, Chalmers RT, Webb DJ, Dear JW. Identification and proteomic profiling of exosomes in human cerebrospinal fluid. Journal of Translational Medicine. 2012;10:5
  5. 5. Madison MN, Roller RJ, Okeoma CM. Human semen contains exosomes with potent anti-HIV-1 activity. Retrovirology. 2014;11:102
  6. 6. Villanueva MT. Microenvironment: Small containers, important cargo. Nature Reviews Cancer. 2014;14:764
  7. 7. Pant S, Hilton H, Burczynski ME. The multifaceted exosome: Biogenesis, role in normal and aberrant cellular function, and frontiers for pharmacological and biomarker opportunities. Biochemical Pharmacology. 2012;83:1484-1494
  8. 8. Robbins PD, Morelli AE. Regulation of immune responses by extracellular vesicles. Nature Reviews Immunology. 2014;14:195-208
  9. 9. Hurley JH, Odorizzi G. Get on the exosome bus with ALIX. Nature Cell Biology. 2012;14: 654-655
  10. 10. Stoorvogel W. Resolving sorting mechanisms into exosomes. Cell Research. 2015;25(5): 531-532
  11. 11. Raiborg C, Stenmark H. The ESCRT machinery in endosomal sorting of ubiquitylated membrane proteins. Nature. 2009;458:445-452
  12. 12. Hurley JH. The ESCRT complexes. Critical Reviews in Biochemistry and Molecular Biology. 2010;45:463-487
  13. 13. Colombo M, Raposo G, Théry C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annual Review of Cell and Developmental Biology. 2014;30:255-289
  14. 14. Votteler J, Sundquist WI. Virus budding and the ESCRT pathway. Cell Host & Microbe. 2013;14:232-241
  15. 15. van Niel G, Charrin S, Simoes S, Romao M, Rochin L, Saftig P, Marks MS, Rubinstein E, Raposo G. The tetraspanin CD63 regulates ESCRT-independent and -dependent endosomal sorting during melanogenesis. Developmental Cell. 2011;21:708-721
  16. 16. Trajkovic K, Hsu C, Chiantia S, Rajendran L, Wenzel D, Wieland F, Schwille P, Brügger B, Simons M. Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science. 2008;319:1244-1247
  17. 17. Ghossoub R, Lembo F, Rubio A, Gaillard CB, Bouchet J, Vitale N, Slavík J, Machala M, Zimmermann P. Syntenin-ALIX exosome biogenesis and budding into multivesicular bodies are controlled by ARF6 and PLD2. Nature Communications. 2014;5:3477
  18. 18. Chavrier P, Parton RG, Hauri HP, Simons K, Zerial M. Localization of low molecular weight GTP binding proteins to exocytic and endocytic compartments. Cell. 1990;62: 317-329
  19. 19. Vitelli R, Santillo M, Lattero D, Chiariello M, Bifulco M, Bruni CB, Bucci C. Role of the small GTPase Rab7 in the late endocytic pathway. Journal of Biological Chemistry. 1997;272:4391-4397
  20. 20. Zerial M, McBride H. Rab proteins as membrane organizers. Nature Reviews Molecular Cell Biology. 2001;2:107-117
  21. 21. Gruenberg J. Viruses and endosome membrane dynamics. Current Opinion in Cell Biology. 2009;21:582-588
  22. 22. de Armas-Rillo L, Valera M-S, Marrero-Hernández S, Valenzuela-Fernández A. Membrane dynamics associated with viral infection. Reviews in Medical Virology. 2016;26:146-160
  23. 23. Alenquer M, Amorim MJ. Exosome biogenesis, regulation, and function in viral infection. Viruses. 2015;7:5066-5083
  24. 24. Nour AM, Modis Y. Endosomal vesicles as vehicles for viral genomes. Trends in Cell Biology. 2014;24:449-454
  25. 25. Piccini LE, Castilla V, Damonte EB. Dengue-3 virus entry into vero cells: Role of clathrin-mediated endocytosis in the outcome of infection. PloS One. 2015;10:e0140824
  26. 26. Smit JM, Moesker B, Rodenhuis-Zybert I, Wilschut J. Flavivirus cell Entry and membrane fusion. Viruses. 2011;3:160-171
  27. 27. Hamel R, Dejarnac O, Wichit S, Ekchariyawat P, Neyret A, Luplertlop N, Perera-Lecoin M, Surasombatpattana P, Talignani L, Thomas F, Cao-Lormeau V-M, Choumet V, Briant L, Desprès P, Amara A, Yssel H, Missé D. Biology of zika virus infection in human skin cells. Journal of Virology. 2015;89:8880-8896
  28. 28. Cohen FS. How viruses invade cells. Biophysical Journal. 2016;110:1028-1032
  29. 29. Ramakrishnaiah V, Thumann C, Fofana I, Habersetzer F, Pan Q, Ruiter PE de, Willemsen R, Demmers JAA, Raj VS, Jenster G, Kwekkeboom J, Tilanus HW, Haagmans BL, Baumert TF, Laan LJW van der. Exosome-mediated transmission of hepatitis C virus between human hepatoma Huh7.5 cells. Proceedings of the National Academy of Sciences 2013;110:13109-13113
  30. 30. Longatti A, Boyd B, Chisari FV. Virion-independent transfer of replication-competent hepatitis C virus RNA between permissive cells. Journal of Virology. 2015;89:2956-2961
  31. 31. Longatti A. The dual role of exosomes in hepatitis A and C virus transmission and viral immune activation. Viruses. 2015;7:6707-6715
  32. 32. Izquierdo-Useros N, Naranjo-Gomez M, Erkizia I, Puertas MC, Borras FE, Blanco J, Martinez-Picado J. HIV and mature dendritic cells: Trojan exosomes riding the trojan horse? PLOS Pathogens 2010;6(3):e1000740
  33. 33. Gould SJ, Booth AM, Hildreth JEK. The Trojan exosome hypothesis. Proceedings of the National Academy of Sciences. 2003;100:10592-10597
  34. 34. Wiley RD, Gummuluru S. Immature dendritic cell-derived exosomes can mediate HIV-1 trans infection. Proceedings of the National Academy of Sciences of the United States of America. 2006;103:738-743
  35. 35. Piguet V, Steinman RM. The interaction of HIV with dendritic cells: Outcomes and pathways. Trends in Immunology. 2007;28:503-510
  36. 36. Welsch S, Keppler OT, Habermann A, Allespach I, Krijnse-Locker J, Kräusslich H-G. HIV-1 buds predominantly at the plasma membrane of primary human macrophages. PLOS Pathogens. 2007;3:e36
  37. 37. Woodward CL, Cheng SN, Jensen GJ. Electron cryo-tomography studies of maturing HIV-1 particles reveal the assembly pathway of the viral core. Journal of Virology. 2014:JVI.02997-14
  38. 38. Yandrapalli N, Muriaux D, Favard C. Lipid domains in HIV-1 assembly. Frontiers in Microbiology. 2014;5:220
  39. 39. Chahar HS, Bao X, Casola A. Exosomes and their role in the life cycle and pathogenesis of RNA viruses. Viruses. 2015;7:3204-3225
  40. 40. Anderson MR, Kashanchi F, Jacobson S. Exosomes in viral disease. Neurotherapeutics. 2016;13:535-546
  41. 41. Meng B, Ip NCY, Prestwood LJ, Abbink TEM, Lever AML. Evidence that the endosomal sorting complex required for transport-II (ESCRT-II) is required for efficient human immunodeficiency virus-1 (HIV-1) production. Retrovirology. 2015;12:72
  42. 42. Madison MN, Okeoma CM. Exosomes: Implications in HIV-1 pathogenesis. Viruses. 2015;7:4093-4118
  43. 43. Mori Y, Koike M, Moriishi E, Kawabata A, Tang H, Oyaizu H, Uchiyama Y, Yamanishi K. Human herpesvirus-6 induces MVB formation, and virus egress occurs by an exosomal release pathway. Traffic 2008;9:1728-1742
  44. 44. Savina A, Vidal M, Colombo MI. The exosome pathway in K562 cells is regulated by Rab11. Journal of Cell Science. 2002;115(Pt 12):2505-2515
  45. 45. Bruce EA, Digard P, Stuart AD. The Rab11 pathway is required for influenza A virus budding and filament formation. Journal of Virology. 2010;84:5848-5859
  46. 46. Rowe RK, Suszko JW, Pekosz A. Roles for the recycling endosome, Rab8, and Rab11 in hantavirus release from epithelial cells. Virology. 2008;382:239-249
  47. 47. Utley TJ, Ducharme NA, Varthakavi V, Shepherd BE, Santangelo PJ, Lindquist ME, Goldenring JR, Crowe JE. Respiratory syncytial virus uses a Vps4-independent budding mechanism controlled by Rab11-FIP2. Proceedings of the National Academy of Sciences of the United States of America. 2008;105:10209-10214
  48. 48. Abrami L, Brandi L, Moayeri M, Brown MJ, Krantz BA, Leppla SH, Goot FG. Hijacking multivesicular bodies enables long-term and exosome-mediated long-distance action of anthrax toxin. Cell Reports. 2013;5:986-996
  49. 49. Bhuin T, Roy JK. Rab11 in disease progression. International Journal of Molecular and Cellular Medicine. 2015;4:1-8
  50. 50. Ostrowski M, Carmo NB, Krumeich S, Fanget I, Raposo G, Savina A, Moita CF, Schauer K, Hume AN, Freitas RP, Goud B, Benaroch P, Hacohen N, Fukuda M, Desnos C, Seabra MC, Darchen F, Amigorena S, Moita LF, Thery C. Rab27a and Rab27b control different steps of the exosome secretion pathway. Nature Cell Biology. 2010;12:19-30
  51. 51. Bobrie A, Krumeich S, Reyal F, Recchi C, Moita LF, Seabra MC, Ostrowski M, Théry C. Rab27a supports exosome-dependent and -independent mechanisms that modify the tumor microenvironment and can promote tumor progression. Cancer Research. 2012;72:4920-4930
  52. 52. Fraile-Ramos A, Cepeda V, Elstak E, Sluijs P. Rab27a is required for human cytomegalovirus assembly. PloS One. 2010;5:e15318
  53. 53. Gerber PP, Cabrini M, Jancic C, Paoletti L, Banchio C, Bilderling C, Sigaut L, Pietrasanta LI, Duette G, Freed EO, Basile G de S, Moita CF, Moita LF, Amigorena S, Benaroch P, Geffner J, Ostrowski M. Rab27a controls HIV-1 assembly by regulating plasma membrane levels of phosphatidylinositol 4,5-bisphosphate. Journal of Cell Biology. 2015;209:435-452
  54. 54. Miranda-Saksena M, Boadle RA, Aggarwal A, Tijono B, Rixon FJ, Diefenbach RJ, Cunningham AL. Herpes simplex virus utilizes the large secretory vesicle pathway for anterograde transport of tegument and envelope proteins and for viral exocytosis from growth cones of human fetal axons. Journal of Virology. 2009;83:3187-3199
  55. 55. Bello-Morales R, Crespillo AJ, Fraile-Ramos A, Tabarés E, Alcina A, López-Guerrero JA. Role of the small GTPase Rab27a during herpes simplex virus infection of oligodendrocytic cells. BMC Microbiology 2012;12:265
  56. 56. White IJ, Bailey LM, Aghakhani MR, Moss SE, Futter CE. EGF stimulates annexin 1-dependent inward vesiculation in a multivesicular endosome subpopulation. The EMBO Journal. 2006;25:1-12
  57. 57. Morales-Sánchez A, Fuentes-Pananá EM. Human viruses and cancer. Viruses. 2014;6: 4047-4079
  58. 58. Chugh PE, Sin S-H, Ozgur S, Henry DH, Menezes P, Griffith J, Eron JJ, Damania B, Dittmer DP. Systemically circulating viral and tumor-derived microRNAs in KSHV-associated malignancies. PLOS Pathogens. 2013;9:e1003484
  59. 59. Canitano A, Venturi G, Borghi M, Ammendolia MG, Fais S. Exosomes released in vitro from Epstein–Barr virus (EBV)-infected cells contain EBV-encoded latent phase mRNAs. Cancer Letters. 2013;337:193-199
  60. 60. Meckes Jr DG, Gunawardena HP, Dekroon RM, Heaton PR, Edwards RH, Ozgur S, Griffith JD, Damania B, Raab-Traub N. Modulation of B-cell exosome proteins by gamma herpesvirus infection. Proceedings of the National Academy of Sciences of the United States of America. 2013;110:E2925-2933
  61. 61. Hoshina S, Sekizuka T, Kataoka M, Hasegawa H, Hamada H, Kuroda M, Katano H. Profile of exosomal and intracellular microRNA in gamma-herpesvirus-infected lymphoma cell lines. PLOS ONE 2016;11:e0162574
  62. 62. Pfeffer S, Zavolan M, Grässer FA, Chien M, Russo JJ, Ju J, John B, Enright AJ, Marks D, Sander C, Tuschl T. Identification of virus-encoded microRNAs. Science 2004;304:734-736
  63. 63. Pegtel DM, Cosmopoulos K, Thorley-Lawson DA, Eijndhoven MAJ van, Hopmans ES, Lindenberg JL, Gruijl TD de, Würdinger T, Middeldorp JM. Functional delivery of viral miRNAs via exosomes. Proceedings of the National Academy of Sciences. 2010;107:6328-6333
  64. 64. Edwards RH, Marquitz AR, Raab-Traub N. Epstein-Barr virus BART microRNAs are produced from a large intron prior to splicing. Journal of Virology. 2008;82:9094-9106
  65. 65. Hooykaas MJG, Kruse E, Wiertz EJHJ, Lebbink RJ. Comprehensive profiling of functional Epstein-Barr virus miRNA expression in human cell lines. BMC Genomics. 2016;17:644
  66. 66. Shinozaki-Ushiku A, Kunita A, Isogai M, Hibiya T, Ushiku T, Takada K, Fukayama M. Profiling of virus-encoded microRNAs in Epstein-Barr virus-associated gastric carcinoma and their roles in gastric carcinogenesis. Journal of Virology. 2015;89:5581-5591
  67. 67. Gallo A, Vella S, Miele M, Timoneri F, Di Bella M, Bosi S, Sciveres M, Conaldi PG. Global profiling of viral and cellular non-coding RNAs in Epstein-Barr virus-induced lymphoblastoid cell lines and released exosome cargos. Cancer Letters. 2017;388:334-343
  68. 68. Ahmed W, Philip PS, Tariq S, Khan G. Epstein-Barr virus-encoded small RNAs (EBERs) are present in fractions related to exosomes released by EBV-transformed cells. PloS One. 2014;9:e99163
  69. 69. Iwakiri D, Zhou L, Samanta M, Matsumoto M, Ebihara T, Seya T, Imai S, Fujieda M, Kawa K, Takada K. Epstein-Barr virus (EBV)-encoded small RNA is released from EBV-infected cells and activates signaling from toll-like receptor 3. Journal of Experimental Medicine. 2009;206(10):2091-2099
  70. 70. Flanagan J, Middeldorp J, Sculley T. Localization of the Epstein-Barr virus protein LMP 1 to exosomes. Journal of General Virology. 2003;84(Pt 7):1871-1879
  71. 71. Verweij FJ, van Eijndhoven MAJ, Hopmans ES, Vendrig T, Wurdinger T, Cahir-McFarland E, Kieff E, Geerts D, van der Kant R, Neefjes J, Middeldorp JM, Pegtel DM. LMP1 association with CD63 in endosomes and secretion via exosomes limits constitutive NF-κB activation. The EMBO Journal. 2011;30:2115-2129
  72. 72. Ikeda M, Longnecker R. Cholesterol is critical for Epstein-Barr virus latent membrane protein 2A trafficking and protein stability. Virology. 2007;360:461-468
  73. 73. Vallhov H, Gutzeit C, Johansson SM, Nagy N, Paul M, Li Q, Friend S, George TC, Klein E, Scheynius A, Gabrielsson S. Exosomes containing glycoprotein 350 released by EBV-transformed B cells selectively target B cells through CD21 and block EBV infection in vitro. Journal of Immunology. 2011;186:73-82
  74. 74. GBD 2015 HIV Collaborators, Wang H, Wolock TM, Carter A, Nguyen G, Kyu HH, Gakidou E, Hay SI, Mills EJ, Trickey A, Msemburi W, Coates MM, Mooney MD, Fraser MS, Sligar A, Salomon J, Larson HJ, Friedman J, Abajobir AA, Abate KH, Abbas KM, Razek MMAE, Abd-Allah F, Abdulle AM, Abera SF, Abubakar I, Abu-Raddad LJ, Abu-Rmeileh NME, Abyu GY, Adebiyi AO, et al. Estimates of global, regional, and national incidence, prevalence, and mortality of HIV, 1980-2015: the Global Burden of Disease Study 2015. The Lancet HIV. 2016;3:e361-387
  75. 75. Cantin R, Diou J, Bélanger D, Tremblay AM, Gilbert C. Discrimination between exosomes and HIV-1: Purification of both vesicles from cell-free supernatants. Journal of Immunological Methods 2008;338:21-30
  76. 76. Wu L, KewalRamani VN. Dendritic-cell interactions with HIV: Infection and viral dissemination. Nature Reviews Immunology. 2006;6:859-868
  77. 77. Grigorov B, Attuil-Audenis V, Perugi F, Nedelec M, Watson S, Pique C, Darlix J-L, Conjeaud H, Muriaux D. A role for CD81 on the late steps of HIV-1 replication in a chronically infected T cell line. Retrovirology. 2009;6:28
  78. 78. Izquierdo-Useros N, Naranjo-Gómez M, Archer J, Hatch SC, Erkizia I, Blanco J, Borràs FE, Puertas MC, Connor JH, Fernández-Figueras MT, Moore L, Clotet B, Gummuluru S, Martinez-Picado J. Capture and transfer of HIV-1 particles by mature dendritic cells converges with the exosome-dissemination pathway. Blood. 2009;113:2732-2741
  79. 79. Jolly C, Sattentau QJ. Human immunodeficiency virus type 1 assembly, budding, and cell-cell spread in T cells take place in tetraspanin-enriched plasma membrane domains. Journal of Virology. 2007;81:7873-7884
  80. 80. Sato K, Aoki J, Misawa N, Daikoku E, Sano K, Tanaka Y, Koyanagi Y. Modulation of human immunodeficiency virus type 1 infectivity through incorporation of tetraspanin proteins. Journal of Virology. 2008;82:1021-1033
  81. 81. Campbell TD, Khan M, Huang M-B, Bond VC, Powell MD. HIV-1 Nef protein is secreted into vesicles that can fuse with target cells and virions. Ethnicity & Disease. 2008;18(2 Suppl 2):S2-14-19
  82. 82. Carvalho JV, Castro RO, Silva EZM, Silveira PP, Silva-Januário ME, Arruda E, Jamur MC, Oliver C, Aguiar RS, daSilva LLP. Nef neutralizes the ability of exosomes from CD4+ T cells to act as decoys during HIV-1 infection. PloS One. 2014;9:e113691
  83. 83. Arenaccio C, Chiozzini C, Columba-Cabezas S, Manfredi F, Affabris E, Baur A, Federico M. Exosomes from human immunodeficiency virus type 1 (HIV-1)-infected cells license quiescent CD4+ T lymphocytes To replicate HIV-1 through a Nef- and ADAM17-dependent mechanism. Journal of Virology. 2014;88:11529-11539
  84. 84. Mack M, Kleinschmidt A, Brühl H, Klier C, Nelson PJ, Cihak J, Plachý J, Stangassinger M, Erfle V, Schlöndorff D. Transfer of the chemokine receptor CCR5 between cells by membrane-derived microparticles: A mechanism for cellular human immunodeficiency virus 1 infection. Nature Medicine. 2000;6:769-775
  85. 85. Rozmyslowicz T, Majka M, Kijowski J, Murphy SL, Conover DO, Poncz M, Ratajczak J, Gaulton GN, Ratajczak MZ. Platelet- and megakaryocyte-derived microparticles transfer CXCR4 receptor to CXCR4-null cells and make them susceptible to infection by X4-HIV. AIDS (London, England). 2003;17:33-42
  86. 86. Jaworski E, Narayanan A, Van Duyne R, Shabbeer-Meyering S, Iordanskiy S, Saifuddin M, Das R, Afonso PV, Sampey GC, Chung M, Popratiloff A, Shrestha B, Sehgal M, Jain P, Vertes A, Mahieux R, Kashanchi F. Human T-lymphotropic virus type 1-infected cells secrete exosomes that contain Tax protein. Journal of Biological Chemistry. 2014;289: 22284-22305
  87. 87. Narayanan A, Jaworski E, Van Duyne R, Iordanskiy S, Guendel I, Das R, Currer R, Sampey G, Chung M, Kehn-Hall K, Bailey C, Popratiloff A, Kashanchi F. Exosomes derived from HTLV-1 infected cells contain the viral protein Tax. Retrovirology. 2014;11(Suppl 1):O46
  88. 88. Anderson M, Lepene B, Kashanchi F, Jacobson S. Detection of human T-cell lymphotropic virus type I proteins in exosomes from HAM/TSP patient CSF by novel nanotrap technology (I7-5E). Neurology 2015;84(14 Supplement):I7-5E
  89. 89. Fleming A, Sampey G, Chung M-C, Bailey C, van Hoek ML, Kashanchi F, Hakami RM. The carrying pigeons of the cell: Exosomes and their role in infectious diseases caused by human pathogens. Pathogens and Disease. 2014;71:109-120
  90. 90. Schorey JS, Cheng Y, Singh PP, Smith VL. Exosomes and other extracellular vesicles in host–pathogen interactions. EMBO Reports. 2015;16:24-43
  91. 91. Soung YH, Ford S, Zhang V, Chung J. Exosomes in cancer diagnostics. Cancers. 2017;9(1): pii:E8
  92. 92. Ahmed W, Philip PS, Attoub S, Khan G. Epstein-Barr virus infected cells release Fas-ligand in exosomal fractions and induce apoptosis in recipient cells via the extrinsic pathway. Journal of General Virology. 2015;96:3646-3659
  93. 93. Keryer-Bibens C, Pioche-Durieu C, Villemant C, Souquere S, Nishi N, Hirashima M, Middeldorp J, Busson P. Exosomes released by EBV-infected nasopharyngeal carcinoma cells convey the viral Latent Membrane Protein 1 and the immunomodulatory protein galectin 9. BMC Cancer. 2006;6:283
  94. 94. Gutzeit C, Nagy N, Gentile M, Lyberg K, Gumz J, Vallhov H, Puga I, Klein E, Gabrielsson S, Cerutti A, Scheynius A. Exosomes derived from Burkitt’s lymphoma cell lines induce proliferation, differentiation, and class-switch recombination in B Cells. Journal of Immunology. 2014;192:5852-5862
  95. 95. Nanbo A, Kawanishi E, Yoshida R, Yoshiyama H. Exosomes derived from Epstein-Barr virus-infected cells are internalized via caveola-dependent endocytosis and promote phenotypic modulation in target cells. Journal of Virology. 2013;87:10334-10347
  96. 96. Klinker MW, Lizzio V, Reed TJ, Fox DA, Lundy SK. Human B cell-derived lymphoblastoid cell lines constitutively produce fas ligand and secrete MHCII+FasL+ Killer exosomes. Frontiers in Immunology. 2014;5:1-10
  97. 97. Klibi J, Niki T, Riedel A, Pioche-Durieu C, Souquere S, Rubinstein E, Le Moulec S, Moulec SLE, Guigay J, Hirashima M, Guemira F, Adhikary D, Mautner J, Busson P. Blood diffusion and Th1-suppressive effects of galectin-9-containing exosomes released by Epstein-Barr virus-infected nasopharyngeal carcinoma cells. Blood. 2009;113:1957-1966
  98. 98. Meckes DG. Exosomal communication goes viral. Journal of Virology. 2015;89:5200-5203
  99. 99. Wang D, Liebowitz D, Kieff E. An EBV membrane protein expressed in immortalized lymphocytes transforms established rodent cells. Cell. 1985;43(3 Pt 2):831-840
  100. 100. Mosialos G, Birkenbach M, Yalamanchili R, VanArsdale T, Ware C, Kieff E. The Epstein-Barr virus transforming protein LMP1 engages signaling proteins for the tumor necrosis factor receptor family. Cell. 1995;80:389-399
  101. 101. Lavorgna A, Harhaj EW. EBV LMP1: New and shared pathways to NF-κB activation. Proceedings of the National Academy of Sciences of the United States of America. 2012;109:2188-2189
  102. 102. Longnecker R, Kieff E. A second Epstein-Barr virus membrane protein (LMP2) is expressed in latent infection and colocalizes with LMP1. Journal of Virology. 1990;64: 2319-2326
  103. 103. Caldwell RG, Wilson JB, Anderson SJ, Longnecker R. Epstein-Barr virus LMP2A drives B cell development and survival in the absence of normal B cell receptor signals. Immunity. 1998;9:405-411
  104. 104. Casola S, Otipoby KL, Alimzhanov M, Humme S, Uyttersprot N, Kutok JL, Carroll MC, Rajewsky K. B cell receptor signal strength determines B cell fate. Nature Immunology. 2004;5:317-327
  105. 105. Barreto A, Rodríguez L-S, Rojas OL, Wolf M, Greenberg HB, Franco MA, Angel J. Membrane vesicles released by intestinal epithelial cells infected with rotavirus inhibit T-cell function. Viral Immunology. 2010;23:595-608
  106. 106. Lenassi M, Cagney G, Liao M, Vaupotič T, Bartholomeeusen K, Cheng Y, Krogan NJ, Plemenitaš A, Peterlin BM. HIV Nef is secreted in exosomes and triggers apoptosis in bystander CD4+ T cells. Traffic. 2010;11:110-122
  107. 107. Oldstone MBA. Viral persistence: Parameters, mechanisms and future predictions. Virology. 2006;344:111-118
  108. 108. Cox JE, Sullivan CS. Balance and stealth: The role of noncoding RNAs in the regulation of virus gene expression. Annual Review of Virology. 2014;1:89-109
  109. 109. Stern-Ginossar N, Elefant N, Zimmermann A, Wolf DG, Saleh N, Biton M, Horwitz E, Prokocimer Z, Prichard M, Hahn G, Goldman-Wohl D, Greenfield C, Yagel S, Hengel H, Altuvia Y, Margalit H, Mandelboim O. Host immune system gene targeting by a viral miRNA. Science. 2007;317:376-381
  110. 110. Cullen BR. MicroRNAs as mediators of viral evasion of the immune system. Nature Immunology. 2013;14:205-210
  111. 111. Nicoll MP, Proença JT, Efstathiou S. The molecular basis of herpes simplex virus latency. FEMS Microbiology Reviews. 2012;36:684-705
  112. 112. Umbach JL, Kramer MF, Jurak I, Karnowski HW, Coen DM, Cullen BR. MicroRNAs expressed by herpes simplex virus 1 during latent infection regulate viral mRNAs. Nature. 2008;454:780-783
  113. 113. Du T, Han Z, Zhou G, Roizman B. Patterns of accumulation of miRNAs encoded by herpes simplex virus during productive infection, latency, and on reactivation. Proceedings of the National Academy of Sciences of the United States of America. 2015;112:E49–E55
  114. 114. Piedade D, Azevedo-Pereira JM. The role of microRNAs in the pathogenesis of herpesvirus infection. Viruses. 2016;8(6):pii: E156
  115. 115. Han Z, Liu X, Chen X, Zhou X, Du T, Roizman B, Zhou G. miR-H28 and miR-H29 expressed late in productive infection are exported and restrict HSV-1 replication and spread in recipient cells. Proceedings of the National Academy of Sciences of the United States of America. 2016;113:E894–E901
  116. 116. Kalamvoki M, Du T, Roizman B. Cells infected with herpes simplex virus 1 export to uninfected cells exosomes containing STING, viral mRNAs, and microRNAs. Proceedings of the National Academy of Sciences of the United States of America. 2014;111:E4991-4996
  117. 117. Chai N, Chang HE, Nicolas E, Han Z, Jarnik M, Taylor J. Properties of subviral particles of hepatitis B virus. Journal of Virology. 2008;82:7812-7817
  118. 118. Jiang B, Himmelsbach K, Ren H, Boller K, Hildt E. Subviral hepatitis B virus filaments, like infectious viral particles, are released via multivesicular bodies. Journal of Virology. 2016;90:3330-3341
  119. 119. Kouwaki T, Fukushima Y, Daito T, Sanada T, Yamamoto N, Mifsud EJ, Leong CR, Tsukiyama-Kohara K, Kohara M, Matsumoto M, Seya T, Oshiumi H. Extracellular vesicles including exosomes regulate innate immune responses to hepatitis B virus infection. Frontiers in Immunology. 2016;7:335
  120. 120. McLauchlan J, Addison C, Craigie MC, Rixon FJ. Noninfectious L-particles supply functions which can facilitate infection by HSV-1. Virology. 1992;190:682-688
  121. 121. Rixon FJ, Addison C, McLauchlan J. Assembly of enveloped tegument structures (L particles) can occur independently of virion maturation in herpes simplex virus type 1-infected cells. Journal of General Virology. 1992;73:277-284
  122. 122. Heilingloh CS, Kummer M, Mühl-Zürbes P, Drassner C, Daniel C, Klewer M, Steinkasserer A. L particles transmit viral proteins from herpes simplex virus 1-infected mature dendritic cells to uninfected bystander cells, inducing CD83 downmodulation. Journal of Virology. 2015;89:11046-11055
  123. 123. Chen R, Zhao X, Wang Y, Xie Y, Liu J. Hepatitis B virus X protein is capable of down-regulating protein level of host antiviral protein APOBEC3G. Scientific Reports. 2017;7: 40783
  124. 124. Bukong TN, Momen-Heravi F, Kodys K, Bala S, Szabo G. Exosomes from hepatitis C infected patients transmit HCV infection and contain replication competent viral RNA in complex with Ago2-miR122-HSP90. PLOS Pathogens. 2014;10:e1004424
  125. 125. Sattentau Q. Avoiding the void: cell-to-cell spread of human viruses. Nature Reviews Microbiology. 2008;6:815-826
  126. 126. Brimacombe CL, Grove J, Meredith LW, Hu K, Syder AJ, Flores MV, Timpe JM, Krieger SE, Baumert TF, Tellinghuisen TL, Wong-Staal F, Balfe P, McKeating JA. Neutralizing antibody-resistant hepatitis C virus cell-to-cell transmission. Journal of Virology. 2011;85:596-605
  127. 127. Li J, Liu K, Liu Y, Xu Y, Zhang F, Yang H, Liu J, Pan T, Chen J, Wu M, Zhou X, Yuan Z. Exosomes mediate the cell-to-cell transmission of IFN-α-induced antiviral activity. Nature Immunology. 2013;14:793-803
  128. 128. Temme S, Eis-Hübinger AM, McLellan AD, Koch N. The herpes simplex virus-1 encoded glycoprotein B diverts HLA-DR into the exosome pathway. Journal of Immunology. 2010;184:236-243
  129. 129. Plazolles N, Humbert J-M, Vachot L, Verrier B, Hocke C, Halary F. Pivotal advance: The promotion of soluble DC-SIGN release by inflammatory signals and its enhancement of cytomegalovirus-mediated cis-infection of myeloid dendritic cells. Journal of Leukocyte Biology. 2011;89:329-342
  130. 130. Arenaccio C, Anticoli S, Manfredi F, Chiozzini C, Olivetta E, Federico M. Latent HIV-1 is activated by exosomes from cells infected with either replication-competent or defective HIV-1. Retrovirology. 2015;12:87
  131. 131. Sampey GC, Saifuddin M, Schwab A, Barclay R, Punya S, Chung M-C, Hakami RM, Zadeh MA, Lepene B, Klase ZA, El-Hage N, Young M, Iordanskiy S, Kashanchi F. Exosomes from HIV-1-infected cells stimulate production of Pro-inflammatory cytokines through Trans-activating response (TAR) RNA. Journal of Biological Chemistry. 2016;291:1251-1266
  132. 132. Dhib-Jalbut S, Hoffman PM, Yamabe T, Sun D, Xia J, Eisenberg H, Bergey G, Ruscetti FW. Extracellular human T-cell lymphotropic virus type I Tax protein induces cytokine production in adult human microglial cells. Annals of Neurology. 1994;36:787-790
  133. 133. Shembade N, Harhaj EW. Role of post-translational modifications of HTLV-1 Tax in NF-κB activation. World Journal of Biological Chemistry. 2010;1:13-20
  134. 134. Honegger A, Schilling D, Bastian S, Sponagel J, Kuryshev V, Sültmann H, Scheffner M, Hoppe-Seyler K, Hoppe-Seyler F. Dependence of intracellular and exosomal microRNAs on viral E6/E7 oncogene expression in HPV-positive tumor cells. PLOS Pathogens. 2015;11:e1004712
  135. 135. Feng Z, Hensley L, McKnight KL, Hu F, Madden V, Ping L, Jeong S-H, Walker C, Lanford RE, Lemon SM. A pathogenic picornavirus acquires an envelope by hijacking cellular membranes. Nature. 2013;496:367-371
  136. 136. Yang Y, Han Q, Hou Z, Zhang C, Tian Z, Zhang J. Exosomes mediate hepatitis B virus (HBV) transmission and NK-cell dysfunction. Cellular & Molecular Immunology. 2017;14(5):465-475
  137. 137. Dreux M, Garaigorta U, Boyd B, Décembre E, Chung J, Whitten-Bauer C, Wieland S, Chisari FV. Short-Range exosomal transfer of viral RNA from infected cells to plasmacytoid dendritic cells triggers innate immunity. Cell Host & Microbe. 2012;12:558-570
  138. 138. Ahsan NA, Sampey GC, Lepene B, Akpamagbo Y, Barclay RA, Iordanskiy S, Hakami RM, Kashanchi F. Presence of viral RNA and proteins in exosomes from cellular clones resistant to rift valley fever virus infection. Frontiers in Microbiology. 2016;7:139

Written By

Gulfaraz Khan, Waqar Ahmed and Pretty S. Philip

Submitted: 31 October 2016 Reviewed: 24 April 2017 Published: 12 July 2017