Next Article in Journal
The Specificity of Epizootic and Epidemiological Processes in Natural Foci of Hemorrhagic Fever with Renal Syndrome and Tick-Borne Encephalitis in Russia, as the Basis for the Prospects of Creating a Combined Vaccine for the Prevention of These Infections
Next Article in Special Issue
EZH2 Inhibition by DS3201 Triggers the Kaposi’s Sarcoma-Associated Herpesvirus Lytic Cycle and Potentiates the Effects Induced by SAHA in Primary Effusion Lymphoma Cells
Previous Article in Journal
Molecular and Phylogenetic Evidence of Interfamilial Transmission of HTLV-1 in the Afro-Descendant Community of São José de Icatú in the Brazilian Amazon
Previous Article in Special Issue
Comparative Proteomic Analysis of Huh7 Cells Transfected with Sub-Saharan African Hepatitis B Virus (Sub)genotypes Reveals Potential Oncogenic Factors
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Oncoviral Infections and Small Extracellular Vesicles

by
Łukasz Ważny
1,
Theresa L. Whiteside
2,3,4 and
Monika Pietrowska
1,*
1
Maria Sklodowska-Curie National Research Institute of Oncology, 44-102 Gliwice, Poland
2
UPMC Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15232, USA
3
Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
4
Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
*
Author to whom correspondence should be addressed.
Viruses 2024, 16(8), 1291; https://doi.org/10.3390/v16081291
Submission received: 10 July 2024 / Revised: 5 August 2024 / Accepted: 12 August 2024 / Published: 13 August 2024
(This article belongs to the Special Issue Molecular and Cellular Biology of Human Oncogenic Viruses)

Abstract

:
Small extracellular vesicles (sEV) are small membrane-bound nanovesicles with a size range below 200 nm that are released by all types of cells. sEV carry a diverse cargo of proteins, lipids, glycans, and nucleic acids that mimic the content of producer cells. sEV mediate intercellular communication and play a key role in a broad variety of physiological and pathological conditions. Recently, numerous reports have emerged examining the role of sEV in viral infections. A significant number of similarities in the sEV biogenesis pathways and the replication cycles of viruses suggest that sEV might influence the course of viral infections in diverse ways. Besides directly modulating virus propagation by transporting the viral cargo (complete virions, proteins, RNA, and DNA), sEV can also modify the host antiviral response and increase the susceptibility of cells to infection. The network of mutual interactions is particularly complex in the case of oncogenic viruses, deserving special consideration because of its significance in cancer progression. This review summarizes the current knowledge of interactions between sEV and oncogenic viruses, focusing on sEV abilities to modulate the carcinogenic properties of oncoviruses.

1. Introduction

The rapid development of biological sciences that took place in the 20th century, including the development of electron microscopy (EM), has enabled the emergence of research on extracellular vesicles (EVs). Traditionally, the 1946 publication by Chargaff and West, which describes the procoagulant activity in a pellet from blood plasma obtained by ultracentrifugation, is considered as the first reported association of the thromboplastic factor with EVs [1,2]. It was not until 1967 that Peter Wolf described the detection by EM of small microparticles in plasma, referred to as “platelet dust” [3]. Research conducted in subsequent years has not only allowed for the identification of different types of EVs (among them, small extracellular vesicles; sEV aka exosomes) but has also led to the understanding that EVs are not a product of cell breakdown like apoptotic bodies. EVs are produced by live cells and represent a key element of intercellular communication, performing a variety of physiological and pathological functions [4]. The field of EV research has been characterized by dynamic developments in recent years, with an increasing number of publications focusing on the impact of EVs on viral infections.
The current definition of EVs identifies them as lipid bilayer membrane-enclosed nanovesicles released by all types of cells. Unlike cells, EVs cannot replicate independently and lack a nucleus [5]. EVs exhibit significant diversity in size, cellular origin, and biogenesis. Because of this heterogeneity, the nomenclature of EVs is not entirely clear, although the most commonly used terminology divides them into small EVs (sEV; with a diameter <200 nm) and large EVs (lEVs; with a diameter >200 nm) [5]. Further, based on their biogenesis, EVs can be divided into exosomes (EXOs) derived from the endocytic cell compartment; ectosomes (microvesicles, MVs) produced by pinching off from the surface of cell membrane; and apoptotic bodies [5,6]. The International Society for Extracellular Vesicles (ISEV) has recently issued recommendations that all vesicles smaller than 200 nm that originate from the endocytic cell compartment of producer cells should be referred to as “sEV”. [5]. EVs are present in all human biological fluids [7], including blood [8,9], urine [10], saliva [11,12], tears [13], ejaculate [14], bronchial fluid [15], cerebrospinal fluid [16], peritoneal fluid [17], amniotic fluid [18], milk [19], and bile [20]. In this review, we use the terms “sEV” or “exosomes” based on the vesicle size as well as cellular origin.
The biogenesis of sEV begins with the endocytosis of external materials and the formation of early endosomes in the cytosol of a producer cell. The endosomal membrane undergoes re-invagination, forming intraluminal vesicles (ILVs) within the multivesicular bodies (MVBs). Subsequently, the fusion of MVBs with the cellular membrane leads to the release of exosomes into the intercellular space [21]. The biogenesis of exosomes is tightly controlled by the endosomal sorting complex required for transport (ESCRT), which regulates the selection of exosomal cargo as well as ILV formation. Alternative ESCRT-independent pathways of exosome biosynthesis have also been described [22,23].
As mediators of intercellular communication, sEV are involved in a wide range of physiological functions. The role of sEV in stimulating both innate and acquired immune responses has been previously demonstrated [24,25]. sEV can fulfill these functions by transferring antigens, cytokines, and enzymes, as well as transcription factors and cytotoxic proteins, all of which can induce transcriptional and/or translational changes in recipient cells. An interesting example of such reprogramming is offered by sEV released by antigen-presenting cells (APCs) that can enhance antibacterial responses against Salmonella enterica. Emerson et al. demonstrated that sEV released by Salmonella-infected macrophages, when administered to BALB/c mice, protected the animals from fatal infection [26]. Luo et al. demonstrated anti-tumor effects of exosomes derived from NK cells (NK-EXOs) in ovarian cancer [27]. Others have reported that NK-EXOs carry numerous proteins with cytotoxic properties, including perforins, granzymes, and granulysin, as well as cytokines such as IFN-γ or TNF-α, promoting the elimination of viral infections or limiting tumorigenesis [28,29]. sEV are involved in various other physiological processes such as vessel growth, tissue regeneration, or neuronal protection against oxidative stress [30].
In addition to the many physiological functions sEV modulate, they are also involved in mediating pathological mechanisms [31]; for example, their role in the development of various cancers, e.g., lung cancer [32,33], breast cancer [31], melanoma [34], hepatocellular carcinoma [35], and cervical cancer [36], is well documented. Additionally, sEV involvement in the promotion of viral infections has been described [37]. The relationship between sEV and oncoviruses in the context of tumor development has been of special interest. Oncoviruses, a group of viruses capable of causing cancer, are associated with 12% of all human cancers [38]. Currently, this group includes seven viruses with different taxonomic affiliations, namely, human T-cell lymphotropic virus-1 (HTLV-1), hepatitis C virus (HCV), human herpesvirus-8 (HHV-8), Epstein–Barr virus (EBV), Merkel cell polyomavirus (MCPyV), human papillomavirus (HPV), and hepatitis B virus (HBV) [38]. The mere fact of oncovirus infection alone does not determine the occurrence of cancer, reflecting the complex relationships between oncoviruses and tumors. Research indicates that these processes appear to be strongly linked to sEV.

2. Types of Interactions between sEV and Viruses

Considering the remarkable ubiquity of sEV secretion by all types of cells, including immune cells, it should not be surprising that sEV influence the course of viral infections. sEV and viruses (especially enveloped viruses) exhibit many common features, both physical, chemical, and biological. Thus, the complete separation of sEV from virions during the isolation of the former is difficult or even impossible [39]. The mutual associations between sEV and viruses are also related to the interactions between the sEV biogenesis pathway and the viral replication cycle [40]. The effect of sEV on viruses can be twofold, acting as a “double-edged sword”. On the one hand, sEV can inhibit viral infection, e.g., by contributing to stimulation of antiviral immune mechanisms. On the other hand, they can promote viral infection e.g., by expanding the pool of cells susceptible to infection (transferring receptors for viruses or inducing receptor synthesis), increasing the efficiency of viral propagation and silencing the host immune response (see Figure 1) [41].

2.1. sEV and Inhibition of Viral Infections

2.1.1. Transfer of Host Restrictive Factors

The antiviral effect of EVs (including sEV) can take various forms, depending largely on the specific virus. It often includes mechanisms that affect cells even before viral infection, preventing its occurrence [41]. An example is the transmission by sEV of the host restriction factors, i.e., proteins involved in the processes of innate antiviral immunity. In the case of human immunodeficiency virus 1 (HIV-1), sEV have been shown to transport APOBEC3G (apolipoprotein B mRNA editing enzyme catalytic subunit 3G) and thereby increase the resistance of target cells to HIV-1 infection in vitro [42]. On the other hand, recent studies indicate the potential involvement of APOBEC3 molecules in limiting the progression of tumors induced by human papillomaviruses (HPV), which indicates a negative role of sEV in the transmission of these molecules during infection [43,44]. However, there are still no conclusive studies confirming the role of APOBEC3-carrying sEV in oncogenesis. Kwasnik et al. [45] showed that infection with Influenza A virus (IAV) alters the profile of RNA in sEV secreted by infected cells by increasing the level of miRNA encoding RSAD2 (radical S-adenosyl methionine domain containing 2; virus inhibitory protein; viperin)—a host protein that inhibits viral replication of IAV [46], HIV [47], or HCV [48]).

2.1.2. Binding and Neutralization of Virions by sEV Containing Entry Receptors

One of the ways for limiting viral infections is through increasing the presence of sEV that carry receptors recognized by viruses. By binding of virions to their surface, sEV can act as a “bait” and prevent the virus from entering cells [41]. This mechanism has been observed in different viruses, including the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and HIV-1 [49,50]. In the case of SARS-CoV-2, virions bind to sEV containing angiotensin-converting enzyme 2 (ACE-2). In HIV-1, the virus binds to CD4-coated sEV [51], although this effect might be negated by the viral protein Nef [52]. A similar mechanism has also been confirmed for the influenza virus. Suptawiwat et al. demonstrated that sEV secreted by bronchial epithelial cells carry sialic acid on the vesicle surface and can immobilize virions [53]. Previous research by Bedford et al. also suggested similar virion immobilizing effects of sEV isolated from mouse bronchial alveolar lavage fluid [54].

2.1.3. miRNA Transfer for Enhancing Immunity to Infection

Recent research has emphasized the crucial role of microRNAs (miRNAs) in regulating immune responses against viruses [55]. These molecules are commonly found in the cargo of various EVs, including sEV. The antiviral activity of sEV containing certain miRNAs has been demonstrated for HCV where the inhibition of HCV infection by sEV derived from umbilical cord mesenchymal stem cells (uMSCs) was linked to the presence of Let-7f, miR-145, miR-199a, and miR-221 [56]. Similarly, sEV secreted by uMSCs have also been shown to play a role in reducing the replication of influenza A/B virus and human coronaviruses [57]. The specific role of miRNA-125b in suppressing influenza virus replication by inducing of interferon-stimulated genes (ISGs) has also been reported [57].

2.1.4. Stimulation of Antiviral Immune Responses

sEV have a significant impact on both the innate and acquired immune responses to viruses. Several studies have demonstrated the ability of sEV to carry factors that modulate immune responses, particularly pro-inflammatory factors, including cytokines [58,59]. The study by Fitzgerald et al. reported that, of 33 plasma cytokines detected in soluble form, 11 were also secreted in the EV-bound form [60]. We have recently reported that in plasma of patients with HNSCC, 24/51 soluble cytokines were also carried in sEV, including IL6, TNFRII, IL-17a, IFNα, and IFNγ [61]. Additionally, Velandia-Romero et al. detected TNF-α, IL-6, complement protein C3, and metalloproteinase inhibitors in sEV released from dengue virus (DENV)-infected U937 macrophages and determined that DENV infection can increase the levels of certain inflammatory mediators in released sEV [62]. On the other hand, sEV can facilitate the recognition of viral pathogen-associated molecular patterns (PAMPs), such as viral proteins or nucleic acids, by transmitting them to appropriate pattern recognition receptors (PRRs), thereby promoting the initiation of antiviral responses [63,64]. The uptake of sEV containing viral antigens by antigen-presenting cells (APCs) can also contribute to a more efficient termination of infection by presenting viral antigens to effector cells [65,66].

2.2. sEV Effects on Viral Propagation

sEV play a critical role in the spread of viral infections by transporting bioactive viral particles, such as nucleic acids and proteins, or even whole virions. Furthermore, sEV can impair the host immune response or broaden viral tropism by delivering entry receptors to target cells and sensitizing them to infection. Here, we provide a comprehensive overview of the key mechanisms that contribute to the severity of viral infections mediated by sEV.

2.2.1. Transport of Viral Cargo via sEV

En Bloc Transmission of Virions

For many years, the question of whether full virions are transported by sEV has been a topic of a debate and was considered controversial. The difficulties in separating virions and sEV, due to their similar physicochemical properties (such as size, density, structure, and content) have posed significant challenges and hindered the clear interpretation of test results [67]. When using the classical methods of sEV isolation, a possibility for co-isolation of virions should be considered. This requires the use of EM to confirm the presence of virions within sEV. Early research investigating the sEV potential for carrying full virions focused primarily on non-enveloped picornaviruses [68,69]. Feng et al. were the first to show that hepatitis A virus (HAV) can be released from cells inside sEV, taking on a form resembling an enveloped virus (quasi-enveloped HAV; eHAV), thereby confirming the existence of an alternative, non-lytic route of HAV exit from cells [68]. In the case of non-enveloped viruses, this mechanism may provide protection against the host’s immune response by concealing the virion within sEV [70]. The release of virions in sEV has been confirmed for viruses such as poliovirus [71], coxsackievirus B3 [72], hepatitis E Virus (HEV) [73], and rotavirus [74]. Given that enveloped viruses are already covered in a lipid bilayer, obtaining additional protection through sEV may seem unnecessary. However, it has been observed that even enveloped viruses use EVs (including sEV) to transport virions. Examples include the hepatitis B virus (HBV) [75] and HCV [76]. This strategy might be beneficial for the virus probably due to additional protection against the immune response, and it extends the range of potential target cells for the virus [77].

Transfer of Nucleic Acids

In viral infections, sEV can carry a wide array of different nucleic acids that are involved in the infection process. These nucleic acids can be categorized into three main types: full viral genome, mRNA, and non-coding RNAs [67]. The full viral genome, which consists of DNA or RNA (and comprises the entire genome of the virus), has been detected in EVs, including HIV-1 [78], HBV [79,80], and HCV [81]. Additionally, Mata-Rocha et al. detected part of the HPV-18 DNA genome in sEV secreted by HeLa cells with an integrated HPV-18 genome [82].
sEV can transport both mRNA that is a product of viral gene transcription and cellular mRNA encoding proteins that are important for the progression of viral infection [67]. This mechanism might also play a significant role in carcinogenesis caused by oncoviruses. If sEV carry mRNA transcripts that encode viral oncoproteins, the potential exists for the synthesis of these oncoproteins in target cells. Examples include mRNAs encoding the latent membrane protein (LMP-1) of the Epstein–Barr virus (EBV) [83], HBx of HBV [84], Tax of human T-lymphotropic virus (HTLV-1) [85], and E6/E7 of HPV [86]. A separate class of molecules that constitute the cargo of sEV are non-coding RNAs, among which miRNAs play a special role [67]. In many viral infections, sEV transport miRNAs encoded in the viral genome or specific host miRNAs that play a vital role in shaping the infection environment and whose expression level can be modified by the virus [87].

Transfer of Viral Proteins

The transmission of viral proteins is a fundamental mechanism by which sEV could influence the progression of an infection. This applies to both structural and non-structural proteins of the virus. In the case of structural proteins, numerous viral glycoproteins have been detected in sEV, such as Spike (SARS-CoV-2) [88], E1 and E2 (HCV) [89], Env (HIV) [90], and E (Zika) [91]. The presence of sEV coated with viral glycoproteins may challenge the humoral immune response, as they can capture neutralizing antibodies against viral glycoproteins and therefore prevent the targeting of true virions. This mechanism has been confirmed, for example, for the Zika virus (ZIKV) and SARS-CoV-2 [88,91].
In addition to structural proteins, sEV also transport non-structural and regulatory proteins of viruses, often with important functions. An example is the HIV Nef protein, which exhibits neurotoxic effects, and this effect is amplified by transport via sEV [92]. Of particular interest is also the transmission by sEV of viral oncoproteins such as LMP-1 (EBV) [93], HBx (HBV) [84], and Tax (HTLV-1) [94].
The role of sEV in infection may also be accomplished by the transport of certain host proteins. An example is the transmission of cellular receptors and coreceptors for the virus by sEV, which can lead to an increase in the pool of cells susceptible to infection. One of the first research papers to demonstrate the existence of such a mechanism is the publication by Mack et al., in which the authors showed the transfer of chemokine receptor type 5 (CCR5), i.e., a coreceptor for M-tropic strains of HIV, to cells without its endogenous expression [95].

2.3. Hijacking of sEV Biogenesis Pathways by Viruses

Viruses, as obligate parasites, rely on the host’s cellular machinery and metabolic pathways to complete their replication cycle [96]. The virus–host coevolution, which has lasted at least 3 billion years, has resulted in the development of mechanisms for controlling the host cell, ensuring the maximum propagation of new viruses and effective protection against the host’s immune system [97]. One such mechanism is the use of the EV biogenesis pathway, including sEV. Many viruses hijack the cellular machinery of EV biogenesis, such as the ESCRT and Rab-GTPase complex, which can be utilized to release virions and enclose them in a lipid envelope [37]. MVBs are employed by both enveloped viruses (e.g., HBV [98], DENV [99], SARS-CoV2 [100], HHV-6 [101,102], and HCMV [103]) and non-enveloped viruses (e.g., HAV [104], HEV [73], Enterovirus 71 [105], and Bluetongue virus [106,107]). In the case of some viruses, it is possible to utilize ESCRT to release virions from the cell without their entry into MVBs. An example of this is HIV-1, which acquires its lipid envelope in this process [108,109,110]. Other viruses, such as herpes simplex virus 1 (HSV-1), can use ESCRT to travel from the nucleus to the cytoplasm [111,112]. The use of sEV biogenesis pathways by viruses has been thoroughly summarized by Moulin et al. and is not further discussed here [37].

3. sEV Significance in Oncovirus Infections

Oncoviruses constitute a diverse group of viruses with differing taxonomic affiliations, structures, and replication cycles that have the potential to induce cancer. Although oncoviruses have the ability to induce cancer, this does not enhance their replication capacity, transmissibility, or viral load [113]. Carcinogenesis is not an integral part of the replication cycle of oncoviruses [114]. Thus, the development of cancer in response to induction by oncogenic virus appears to be more of a „biological accident” than an evolutionary adaptation [115]. In this context, sEV, as a key element of cell-to-cell communication, might play a crucial role in exacerbating the pathological effects of oncoviruses.
Efforts are ongoing to apply engineered sEV to new therapeutic strategies against oncoviruses. For example, a new form of therapy against HCV could include sEV loaded with anti-HCV miRNAs (let-7f, miR-145, miR-199a, and miR-221) [116]. In the case of HBV infection, sEV loaded with miR-574-5p may exhibit potential therapeutic effects [117]. In addition, the treatment of tumors induced by oncoviruses could be based on the use of sEV released by mesenchymal stromal cells (MSCs). The use of sEV derived from MSCs has many advantages over traditional cell therapies. By carrying cargo derived from MSCs, EVs provide paracrine effects similar to those mediated by MSCs, while also inducing low immunogenicity. Due to the smaller size of sEV compared to cells, there is also no risk of blood vessel blockage when administering sEV intravenously [118].
In this review, we summarize information on the role of sEV in the infections and pathogenesis of oncovirus-induced cancers. The discussed mechanisms of sEV-induced alterations in oncoviral infections are summarized in Table 1.

3.1. RNA Oncoviruses

3.1.1. Retroviruses

HTLV

In light of recent research, the significant role of sEV in HTLV-1 infection is becoming increasingly evident. Although intact HTLV-1 virions have not yet been detected in sEV, these vesicles are well-documented for carrying viral proteins (Tax and HBZ) and transcripts (tax, hbz, and env mRNA) [85,119], and recent reports also indicate the detection of genomic RNA [120]. HTLV-1 infection also alters the host protein expression profile carried by sEV [119]. Presumably, mechanisms related to the viral replication cycle promote the selective packaging of certain host proteins into sEV, as indicated by comparative proteomic analysis conducted by Jaworski et al. [119]. On the other hand, sEV released from infected cells promote cell-to-cell contact by carrying adhesion factors (ICAM-1 and LFA-1), which is crucial for HTLV-1 propagation [121]. The virus spreads 10,000 times more effectively this way compared to the release of free virions [122].
The carcinogenic effect of HTLV-1 infection appears to be amplified by sEV interactions. sEV inhibit apoptosis in target cells and transfer miRNA that promote oncogenic transformation in leukemias (miR-21 and miR-155) [123]. sEV from ATLL (adult T cell leukemia/lymphoma) cells not only carry the oncogenic Tax protein but also VEGF (vascular endothelial growth factor), a proangiogenic factor [123]. They induce morphological changes in MSCs, promote their proliferation, and activate the expression of genes associated with angiogenesis [123].

3.1.2. Flaviviruses

HCV

The relationship between sEV, HCV infection, and carcinogenesis is still not fully comprehended; however, recent research indicates that sEV have a significant impact on the effectiveness of HCV infection and are crucial in the transmission process between hepatocytes [81]. Released from infected cells, sEV carrying HCV virions can cause effective infections, similar to free virions, and are better protected against environmental influences [81,124]. An additional aim of the virus is to evade detection by the immune system by encapsulation within sEV, thus increasing the likelihood of infecting target cells [125]. Giannessi et al. proposed an interesting theoretical concept suggesting that sEV may facilitate the spread of infection by non-infectious, defective virions, for example, by allowing entry into cells by surface glycoprotein-defective virions that utilize the presence of cellular receptors on the surface of sEV [126,127].
Naïve liver cells can be infected by HCV RNA-sEV, secreted from HCV-infected cells, in a receptor-independent pathway, even in the presence of blocking antibodies directed against HCV receptors [125].
Table 1. The key mechanisms of sEV-mediated alterations that impact oncovirus infections.
Table 1. The key mechanisms of sEV-mediated alterations that impact oncovirus infections.
VirusActive Molecule in sEVMechanismEffectRefs.
Viral CargoCellular Cargo
RNA oncoviruses
HTLV-1Tax, mRNAs (tax, hbz, env), genomic RNAProinflammatory cytokines, miR-21, miR-155,VEGF, CD45,CD43, ICAM-1, LFA-1Provide protection for virions against neutralization and environmental factorsPromote infection[119]
Prevent Fas-mediated apoptosis by inducing the cFLIP and NFkB signaling pathwaysIncrease survival of target cells, inhibit apoptosis[119]
Transfer adhesion factors to the target cellIncrease cell-to-cell contact and agglutination potential[121,128]
Carry angiogenic factors and promote the proliferation of target cellsPromote tumor progression[123]
HCVintact virions, E2 protein, genomic RNA, viral RNAsmiR-122, miR-21, miR-34aTransfer of intact virions; enable the infection of target cells by defective virions (e.g., without surface glycoproteins)Increase infection efficiency[126]
Provide physical protection against the neutralization of virionsImpart resistance to neutralization by anti-HCV neutralizing antibodies[81]
Modify gene expression in target cells by transferring different miRNAsCreate favorable tumor environment[129]
Transport miR-122 to target cells, as well as HCV-RNA in a complex with miR-122-Ago2-HSP90Promotes HCV replication[125]
Transport miR-21 to target cellsHigh levels of miR-21 expression in plasma sEV are correlated with clinical-pathological features of HCC[130]
Transport of miR-34aInduction of apoptosis[131]
DNA oncoviruses
HBVintact virions, viral DNA, viral RNA, proteins (HBsAg, HBcAg, HBeAg, HBx, P pro-tein), HBV-miR-3miR-122, miR-21, miR-29, miR-135a-5pTransfer of intact virionsFacilitate virus propagation[75]
Transport of miR-135a-5p that alters gene expression in the target cellExhibit antiapoptotic activity towards cancer cells[132]
Decrease of RIG-I expression in NK cells and reduction of NFkB pathway activationInhibition of proliferation, cytotoxic activity and interferon ɣ production of NK cells[133]
Transport of HBV-miR-3Promotion of angiogenesis[134]
Transport viral HBx protein that lowers the level of expression of antitumorigenic miR-122Promote tumorigenesis [135]
HBV infection increases the expression of miR-21 and miR-29 in sEV, which alters gene expression in target cellsInhibition of IL-12 release by macrophages and dendritic cells[132]
HPVviral DNA, viral proteins (E6, E7), E6/E7 mRNAapoptosis inhibitors (surviving, XIAP, c-IAP1, c-IAP-2, ML-IAP), immunoregulatory molecules (CD276, CD47), calmodulin, MUC16, SIRPATransfer of viral oncoproteins E6/E7Promotion of tumorigenesis[86]
Transport of MUC16 and SIRPA proteinsInduce epithelial-mesenchymal transition[36]
Generate endoplasmic reticulum stress in target cells and decrease expression of proteins associated with tight junctions (ZO-1, claudin-5)Promotion of metastasis and tumor progression[36]
Transport of immunomodulatory factorsStimulate dendritic cell (DC) maturation and sustain their function[136]
Transport of apoptosis inhibitors (surviving, XIAP, c-IAP-1, c-IAP-2, ML-IAP)Inhibition of apoptosis of cancer cells[137]
EBVLMP-1, LMP-2A, gp350, viral RNA, viral miRNA, EBERsHIF1α, Galectin-9, EGFRTransfer oncogenic viral proteins LMP-1 and LMP-2AInduction of tumorigenesis[93,138]
Transport of HIF1α factorInduction of angiogenesis[139,140]
Increasing level of EGFR expressionPromotion of metastasis [141]
Viral protein LMP-1 modify content of cargo of sEV to increase expression proteins involved in EBV infectionIncrease infection efficiency[142]
miRNAs modulate viral and host gene expressionFacilitate infection and immune evasion[142,143]
KSHVviral RNA,
viral miRNA
IL-1β, IFI16Transported viral miRNAs affect cell metabolism and tumorigenesisCreation of microenvironment favorable to tumor development[144,145]
Removal of immune-inducing factors from the cell)Facilitate immune response evasion[146,147]
Complement system activationPromotion of long-term latency by activating NFκB pathway[148]
MCPyVviral oncoproteins, ALTOmiR-375Transport of viral oncoproteins and circular RNA (ALTO)Increase expression of host genes associated with pathogenesis of MCPyV infection[149,150]
VEGF: vascular endothelial growth factor; LFA-1: lymphocyte function-associated antigen 1; XIAP: X-linked inhibitor of apoptosis; c-IAP1-2: cellular inhibitor of apoptosis protein 1-2; ML-IAP: melanoma inhibitor of apoptosis; LMP-1: Epstein–Barr virus latent membrane protein 1; EBERs: Epstein–Barr virus-encoded small RNAs; EGFR: endothelial epidermal growth factor receptor; ALTO: alternative large T antigen open reading frame protein.
Bukong et al. detected HCV RNA in sEV that was found to be associated with Ago2 (Argonaute 2), HSP90 (heat shock protein 90), and miR-122, which may have a stabilizing function with regard to HCV RNA, as suggested by other studies [125]. This finding may explain the ineffectiveness of immunological therapies with anti-HCV-receptor antibodies and indicate the potential of alternative therapies such as miR-122 inhibitors or HSP90 inhibitors [151].
The pathogenesis of HCV-associated cancers is primarily mediated by the production of sEV, which creates a favorable environment for infection and tumor development [129]. Similar to other cancers, miRNAs are a particularly important class of bioactive molecules transported by sEV for the initiation and progression of hepatocellular cancer (HCC). An important example is miR-122, which is highly expressed in the liver and can be found in sEV [125,152]. This molecule participates in promoting HCV replication. Wang et al. discovered that high levels of miR-21 expression in plasma sEV are correlated with clinical-pathological features of HCC such as cirrhosis and tumor staging [130]. Badami et al. have reported an upregulation of miR34a expression in the HCC cell line Huh7.5 after HCV infection. The same study found that sEV secreted by infected cells also demonstrated increased miR34a expression, and when these sEV were applied to Huh7.5 cells, they induced apoptosis [131]. Other studies have demonstrated a correlation between the level of miR34a overexpression and the development of HCV-induced liver cirrhosis [153].

3.2. DNA Oncoviruses

3.2.1. Hepadnaviruses

HBV

Through various mechanisms, sEV promote HBV infection and the onset of HBV-related diseases (including HCC). The transmission of viral cargo by sEV secreted from HBV-infected cells is the main mechanism that promotes HBV propagation (see Figure 2) [133,154]. Recently, Wu et al. demonstrated the presence of full HBV virions in sEV for the first time [75]. There are several reports of the transfer of viral protein (HBx, HBsAg, HBeAg, HbcAg, and P protein), DNA (rcDNA and cccDNA), and RNA (XBx RNA, HBs/p RNA, and HBV-miR-3) in sEV released by infected cells [84,133,155]. In addition, studies using mass spectrometry and miRNA sequencing techniques have revealed differences in the expression profiles of proteins and host miRNAs in sEV derived from normal hepatocytes (THLE2 and THLE3) and HBV-infected hepatocytes (SNU-423 and SNU-182) [156]. According to functional analyses, many of these proteins and miRNAs are involved in the viral replication cycle, sEV biogenesis, and carcinogenesis, which may indicate an important role of sEV in the development of HCC induced by HBV infection [156].
Released from HBV-infected cells, sEV undergo endocytosis by monocytes, resulting in elevated PD-L1 levels in these cells. This observation is consistent with the high levels of PD-L1 expression in the monocytes of patients with chronic hepatitis B [157,158]. PD-L1 can inhibit the immune response against cancer by binding to PD-1 on T cells and promoting the exhaustion of these cells [159]. Additionally, sEV containing viral cargo can negatively affect other immune cells, such as NK cells [133]. In the study of Yang et al., the interaction of sEV containing HBV cargo with NK cells resulted in the disruption of functions such as proliferation, cytotoxic activity, and interferon ɣ production, and also caused a decrease in the expression of RIG-I receptors and inhibition of NF-κB and MAPK signaling pathways [133].
It has been shown that HBV proteins also affect the level of cellular miRNA expression in sEV, causing an increase in the expression of some miRNAs and a decrease in the expression of others, and thus can actively modulate the microenvironment, promoting the progression of CHB to HCC [154]. One example is the role of the viral HBx protein, which exhibits an oncogenic effect by lowering the expression level of miR-122, that is, miRNA involved in inhibiting the development of HCC by binding to genes responsible for proliferation [135]. HBx may also interact with the biogenesis pathways of sEV, leading to a change in their protein profile [84,160]. Moreover, the viral HBcAg protein increases the level of miR-135a-5p in sEV, which has a protective effect against apoptosis and also increases the proliferation of HCC cells and their resistance to chemotherapy [161]. Kouwaki et al. observed that HBV infection resulted in an increased expression of miR-21 and miR-29 in sEV, leading to the inhibition of IL-12 release by macrophages and dendritic cells [132].

3.2.2. Papillomaviruses

HPV

One of the first experiments confirming the transmission of cargo by sEV that could promote HPV-dependent carcinogenesis concerned the presence of survivin—a protein with anti-apoptotic activity [162,163]. Moreover, the presence of other apoptosis inhibitors, such as XIAP, c-IAP1, c-IAP2, and ML-IAP, has also been confirmed in sEV released from HPV-infected cells [164]. Honegger et al. found that sEV secreted by HeLa HPV-infected cells contain different miRNAs, and their composition depends on the endogenous expression of E6/E7 in infected cells [165]. It was observed that sEV secreted by oropharyngeal cancer (OPC) cells affected non-cancerous mammalian HPV (−) epithelial cells by inducing a epithelial-mesenchymal transition (EMT) and making them more invasive. HPV-16 E7 was also detected in previously uninfected HPV cells [166]. The main mechanism of oncogenesis conditioned by viral proteins E6/E7 was partially understood several decades ago and is based on the blocking of the most important cellular tumor suppressors by these proteins. The E6 protein blocks p53 by stimulating its degradation, while E7 binds to pRB, causing its inactivation and proteolysis, and releasing the E2F factor at the same time [167,168].
Ludwig et al. conducted a comparative proteomic analysis of sEV from HPV-infected and not-infected head and neck cancer (HNC) patient-derived cells. The results revealed that E6/E7, p16, and survivin proteins were detected only in HPV (+) cell-derived sEV, while multiple immunomodulatory proteins (TGF-β, FasL, OX40, OX40L, and HSP70) were detected in sEV from both HPV (+) and HPV (−) cells [169]. Interestingly, further studies revealed that HPV (+) cell sEV caused dendritic cell (DC) maturation and sustained their function, unlike HPV (−) cell sEV exhibiting the opposite effect [169]. This may explain previous observations indicating that HPV-infected HNC patients have a better prognosis than non-infected patients and respond better to some forms of therapy [136].

3.2.3. Herpesviruses

EBV

The presence of various viral elements in sEV, i.e., proteins, viral mRNA, miRNA, or Epstein–Barr virus-encoded small RNAs (EBERs) has been documented [170,171]. Among the EBV proteins found in sEV, LMP-1 and LMP-2A appear to be particularly important [93]. Latent membrane protein 1 (LMP-1) is a major EBV oncoprotein that is involved in B cell transformation by binding to TRAF6 and inducing signaling pathways typically triggered by CD40L binding to the CD40 receptor [172]. This leads to enhanced B cell viability, cell growth, and the inhibition of apoptosis [172]. Proteomic studies by Nkosi et al. showed that LMP-1 has the ability to modify the cargo of sEV secreted from infected B lymphocytes, increasing the content of proteins involved in EBV infection, endocytosis, apoptosis, and the MAPK and NF-Kb signaling pathways, as well as adhesion molecules [93]. Under the influence of LMP-1, the level of proteins involved in the cellular organization of organelles and catabolic processes in sEV was reduced [93]. In addition to modifying the protein profile of secreted sEV, LMP-1 may also enhance the process of sEV secretion [139]. Research in recent years has increasingly highlighted that LMP-1 is involved in the development and progression of various types of cancer, including gastric cancer [173], nasopharyngeal carcinoma (NPC) [138], and lymphoma [174]. The importance of LMP-1-carrying sEV in promoting different cancers is becoming evident [138].
In addition to viral factors, sEV released from EBV-infected cancer cells can carry cellular factors that promote tumor progression. This effect has been confirmed, for instance, in NPCs, where sEV from EBV (+) cells transport the promethastatic factor HIF1α [139]. Moreover, sEV from EBV (+) NPC cells may promote immune evasion, thereby ensuring uninterrupted tumor growth [142]. Galectin-9, which interacts with Tim3 and induces apoptosis in T cells, has also been identified in sEV secreted by EBV-infected NPCs [143]. NPC is characterized by a high level of EGFR expression, which in turn can be induced by LMP-1, and LMP-1 simultaneously promotes the release of EGFR into sEV [141].

KSHV

In the course of its replication cycle, KSHV exploits various metabolic pathways of the host cell, ultimately resulting in the formation of a microenvironment favorable for virus propagation and the evasion of the host’s immune response [146]. The manipulation of sEV biosynthesis pathways is one such mechanism that the virus utilizes [146]. In fact, KSHV-infected cells secrete a significantly higher number of sEV than uninfected cells [148]. Research has shown that sEV secreted by KSHV-infected cells contain IL-1β and IFI16 (interferon gamma inducible protein 16), two important factors involved in immune mechanisms [147]. This may indicate that KSHV uses sEV to remove immune-activating factors from the cell [146,147]. Surprisingly, according to results from Jeon et al., sEV from KSHV-infected epithelial cells can activate the complement system in both infected cells and neighboring cells [175]. Furthermore, this sEV-dependent activation of the complement system not only fails to block infection, but promotes persistent KSHV latency and increases the survival of infected cells, particularly by activating the NFkB pathway [175].
As in the case of EBV, KSHV also encodes viral miRNAs in its genome, which can affect host gene expression, immune processes or tumorigenesis [176]. It is not surprising that these viral miRNAs are also present in sEV secreted from KSHV-infected cells. Chugh et al. confirmed the presence of all KSHV miRNAs in sEV isolated from plasma of infected patients [144]. KSHV miRNAs can affect cell metabolism by adjusting it to maintain long-term KSHV latency. By decreasing mitochondrial biogenesis and inducing glycolytic pathways, KSHV miRNAs result in reduced oxygen consumption, increased glucose uptake, and increased lactate secretion, reminiscent of the KSHV-induced Warburg effect [177,178]. Other functions of individual KSHV miRNAs in the context of KSHV replication cycle have been described in detail in Dass et., therefore it will not be discussed here in more detail [179].

3.2.4. Polyomaviruses

MCPyV

The Merkel cell polyomavirus (MCPyV) is the latest discovered oncovirus, which was described by Feng et al. in 2008 [180]. Due to its relatively late discovery, this virus is the least understood oncogenic virus and there is limited research on the role of sEV in MCPyV infection. Konstantinell’s secretome studies revealed that sEV secreted by MCPyV-infected MCC cancer cells carry viral oncoproteins. An analysis of the sEV proteome secreted by MCPyV-infected and non-infected MCCs showed significant differences [181]. Yang et al. detected circular RNA encoding the viral protein ALTO (alternative large T antigen open reading frame) in sEV secreted by MCPyV-infected MCC cell lines. It has also been demonstrated that ALTO can promote the expression of host genes associated with the pathogenesis of MCPyV infection [149].
Research has shown that sEV released by MCCs also contain miRNAs. Fan et al. detected the presence of high levels of miR-375 [182]. Fan et al. demonstrated that the horizontal transfer of miR-375 to fibroblasts via sEV resulted in the polarization of these cells to cancer-associated fibroblasts (CAFs). This indicates the role of miR-375 in MCC to create a pro-cancer microenvironment [182].

4. Conclusions

Research conducted in recent years has recognized sEV as key factors in the pathogenesis of cancer induced by oncoviruses. sEV carry a wide spectrum of bioactive macromolecules involved in both the development of viral infections and in carcinogenesis. However, there still exists a lack of detailed information about the mechanisms that govern the interactions between viruses, sEV, and cancer. Research in this area is especially important for the design of future anticancer therapies, which should take into account the pathologic mechanisms mediated by sEV. Future anti-viral therapies might also be able to take advantage of targeting sEV, which promote viral infections

Author Contributions

Conceptualization, M.P. and Ł.W.; writing—original draft preparation, Ł.W.; writing—review and editing, Ł.W., T.L.W. and M.P.; visualization, Ł.W.; supervision, T.L.W. and M.P.; project administration, T.L.W. and M.P.; funding acquisition, T.L.W. and M.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded in part by NIDCR grant R01-DE031299 from the National Institutes of Health to TLW and MP.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

This work is dedicated to the memory of Łukasz Skoczylas. The authors of this paper would like to express their gratitude for the inspiration to write this review article and for the collected materials, as well as to convey their highest appreciation for Łukasz’s dedication to research in this topic. We would like to thank Marta Gawin for her assistance with editorial revisions.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Couch, Y.; Buzàs, E.I.; Vizio, D.D.; Gho, Y.S.; Harrison, P.; Hill, A.F.; Lötvall, J.; Raposo, G.; Stahl, P.D.; Théry, C.; et al. A Brief History of Nearly EV-erything—The Rise and Rise of Extracellular Vesicles. J. Extracell. Vesicles 2021, 10, e12144. [Google Scholar] [CrossRef]
  2. Chargaff, E.; West, R. The Biological Significance of the Thromboplastic Protein of Blood. J. Biol. Chem. 1946, 166, 189–197. [Google Scholar] [CrossRef]
  3. Wolf, P. The Nature and Significance of Platelet Products in Human Plasma. Br. J. Haematol. 1967, 13, 269–288. [Google Scholar] [CrossRef] [PubMed]
  4. Yuana, Y.; Sturk, A.; Nieuwland, R. Extracellular Vesicles in Physiological and Pathological Conditions. Blood Rev. 2013, 27, 31–39. [Google Scholar] [CrossRef]
  5. Welsh, J.A.; Goberdhan, D.C.I.; O’Driscoll, L.; Buzas, E.I.; Blenkiron, C.; Bussolati, B.; Cai, H.; Di Vizio, D.; Driedonks, T.A.P.; Erdbrügger, U.; et al. Minimal Information for Studies of Extracellular Vesicles (MISEV2023): From Basic to Advanced Approaches. J. Extracell. Vesicles 2024, 13, e12404. [Google Scholar] [CrossRef] [PubMed]
  6. Jia, Y.; Yu, L.; Ma, T.; Xu, W.; Qian, H.; Sun, Y.; Shi, H. Small Extracellular Vesicles Isolation and Separation: Current Techniques, Pending Questions and Clinical Applications. Theranostics 2022, 12, 6548–6575. [Google Scholar] [CrossRef] [PubMed]
  7. Lim, Y.-J.; Jung, G.N.; Park, W.-T.; Seo, M.-S.; Lee, G.W. Therapeutic Potential of Small Extracellular Vesicles Derived from Mesenchymal Stem Cells for Spinal Cord and Nerve Injury. Front. Cell Dev. Biol. 2023, 11, 1151357. [Google Scholar] [CrossRef] [PubMed]
  8. Nieuwland, R.; Siljander, P.R. A Beginner’s Guide to Study Extracellular Vesicles in Human Blood Plasma and Serum. J. Extracell. Vesicles 2024, 13, e12400. [Google Scholar] [CrossRef] [PubMed]
  9. Małys, M.S.; Köller, M.C.; Papp, K.; Aigner, C.; Dioso, D.; Mucher, P.; Schachner, H.; Bonelli, M.; Haslacher, H.; Rees, A.J.; et al. Small Extracellular Vesicles Are Released Ex Vivo from Platelets into Serum and from Residual Blood Cells into Stored Plasma. J. Extracell. Biol. 2023, 2, e88. [Google Scholar] [CrossRef] [PubMed]
  10. Hadpech, S.; Chaiyarit, S.; Phuangkham, S.; Sukphan, S.; Thongboonkerd, V. The Modulatory Effects of Large and Small Extracellular Vesicles from Normal Human Urine on Calcium Oxalate Crystallization, Growth, Aggregation, Adhesion on Renal Cells, and Invasion through Extracellular Matrix: An in Vitro Study. Biomed. Pharmacother. 2024, 173, 116393. [Google Scholar] [CrossRef] [PubMed]
  11. Tengler, L.; Tiedtke, M.; Schütz, J.; Bieback, K.; Uhlig, S.; Theodoraki, M.-N.; Nitschke, K.; Worst, T.S.; Seiz, E.; Scherl, C.; et al. Optimization of Extracellular Vesicles Preparation from Saliva of Head and Neck Cancer Patients. Sci. Rep. 2024, 14, 946. [Google Scholar] [CrossRef] [PubMed]
  12. Han, P.; Bartold, P.M.; Ivanovski, S. The Emerging Role of Small Extracellular Vesicles in Saliva and Gingival Crevicular Fluid as Diagnostics for Periodontitis. J. Periodontal. Res. 2022, 57, 219–231. [Google Scholar] [CrossRef] [PubMed]
  13. Cross, T.; Øvstebø, R.; Brusletto, B.S.; Trøseid, A.-M.S.; Olstad, O.K.; Aspelin, T.; Jackson, C.J.; Chen, X.; Utheim, T.P.; Haug, K.B.F. RNA Profiles of Tear Fluid Extracellular Vesicles in Patients with Dry Eye-Related Symptoms. Int. J. Mol. Sci. 2023, 24, 15390. [Google Scholar] [CrossRef] [PubMed]
  14. Höög, J.L.; Lötvall, J. Diversity of Extracellular Vesicles in Human Ejaculates Revealed by Cryo-Electron Microscopy. J. Extracell. Vesicles 2015, 4, 28680. [Google Scholar] [CrossRef] [PubMed]
  15. Jonak, S.T.; Liu, Z.; Liu, J.; Li, T.; D’Souza, B.V.; Schiaffino, J.A.; Oh, S.; Xie, Y.-H. Analyzing Bronchoalveolar Fluid Derived Small Extracellular Vesicles Using Single-Vesicle SERS for Non-Small Cell Lung Cancer Detection. Sens. Diagn. 2023, 2, 90–99. [Google Scholar] [CrossRef] [PubMed]
  16. Krušić Alić, V.; Malenica, M.; Biberić, M.; Zrna, S.; Valenčić, L.; Šuput, A.; Kalagac Fabris, L.; Wechtersbach, K.; Kojc, N.; Kurtjak, M.; et al. Extracellular Vesicles from Human Cerebrospinal Fluid Are Effectively Separated by Sepharose CL-6B—Comparison of Four Gravity-Flow Size Exclusion Chromatography Methods. Biomedicines 2022, 10, 785. [Google Scholar] [CrossRef]
  17. Quiralte, M.; Barquín, A.; Yagüe Fernández, M.; Navarro, P.; Grazioso, T.P.; Sevillano, E.; Rodriguez Moreno, J.F.; Balarezo-Saldivar, A.; Peinado, H.; Izquierdo, E.; et al. Proteomic Profiles of Peritoneal-Derived Small Extracellular Vesicles Correlate with Outcome in Ovarian Cancer Patients. J. Clin. Investig. 2024, 134, e176161. [Google Scholar] [CrossRef] [PubMed]
  18. Costa, A.; Quarto, R.; Bollini, S. Small Extracellular Vesicles from Human Amniotic Fluid Samples as Promising Theranostics. Int. J. Mol. Sci. 2022, 23, 590. [Google Scholar] [CrossRef] [PubMed]
  19. Vahkal, B.; Kraft, J.; Ferretti, E.; Chung, M.; Beaulieu, J.-F.; Altosaar, I. Review of Methodological Approaches to Human Milk Small Extracellular Vesicle Proteomics. Biomolecules 2021, 11, 833. [Google Scholar] [CrossRef] [PubMed]
  20. Muraki, R.; Morita, Y.; Ida, S.; Kitajima, R.; Furuhashi, S.; Takeda, M.; Kikuchi, H.; Hiramatsu, Y.; Takanashi, Y.; Hamaya, Y.; et al. Phosphatidylcholine in Bile-derived Small Extracellular Vesicles as a Novel Biomarker of Cholangiocarcinoma. Cancer Med. 2023, 12, 13007–13018. [Google Scholar] [CrossRef] [PubMed]
  21. Zhang, Y.; Liu, Y.; Liu, H.; Tang, W.H. Exosomes: Biogenesis, Biologic Function and Clinical Potential. Cell Biosci. 2019, 9, 19. [Google Scholar] [CrossRef] [PubMed]
  22. Kenific, C.M.; Zhang, H.; Lyden, D. An Exosome Pathway without an ESCRT. Cell Res. 2021, 31, 105–106. [Google Scholar] [CrossRef] [PubMed]
  23. Rezaie, J.; Ajezi, S.; Avci, Ç.B.; Karimipour, M.; Geranmayeh, M.H.; Nourazarian, A.; Sokullu, E.; Rezabakhsh, A.; Rahbarghazi, R. Exosomes and Their Application in Biomedical Field: Difficulties and Advantages. Mol. Neurobiol. 2018, 55, 3372–3393. [Google Scholar] [CrossRef] [PubMed]
  24. Arteaga-Blanco, L.A.; Bou-Habib, D.C. The Role of Extracellular Vesicles from Human Macrophages on Host-Pathogen Interaction. Int. J. Mol. Sci. 2021, 22, 10262. [Google Scholar] [CrossRef] [PubMed]
  25. Hazrati, A.; Soudi, S.; Malekpour, K.; Mahmoudi, M.; Rahimi, A.; Hashemi, S.M.; Varma, R.S. Immune Cells-Derived Exosomes Function as a Double-Edged Sword: Role in Disease Progression and Their Therapeutic Applications. Biomark. Res. 2022, 10, 30. [Google Scholar] [CrossRef]
  26. Emerson, L.E.; Barker, H.; Tran, T.; Barker, S.; Enslow, S.; Ou, M.; Hoffman, C.; Jones, M.; Pascual, D.W.; Edelmann, M.J. Extracellular Vesicles Elicit Protective Immune Responses against Salmonella Infection. J. Extracell. Vesicles 2022, 11, e12267. [Google Scholar] [CrossRef] [PubMed]
  27. Luo, H.; Zhou, Y.; Zhang, J.; Zhang, Y.; Long, S.; Lin, X.; Yang, A.; Duan, J.; Yang, N.; Yang, Z.; et al. NK Cell-Derived Exosomes Enhance the Anti-Tumor Effects against Ovarian Cancer by Delivering Cisplatin and Reactivating NK Cell Functions. Front. Immunol. 2023, 13, 1087689. [Google Scholar] [CrossRef] [PubMed]
  28. Razizadeh, M.H.; Zafarani, A.; Taghavi-Farahabadi, M.; Khorramdelazad, H.; Minaeian, S.; Mahmoudi, M. Natural Killer Cells and Their Exosomes in Viral Infections and Related Therapeutic Approaches: Where Are We? Cell Commun. Signal. 2023, 21, 261. [Google Scholar] [CrossRef] [PubMed]
  29. Wu, C.-H.; Li, J.; Li, L.; Sun, J.; Fabbri, M.; Wayne, A.S.; Seeger, R.C.; Jong, A.Y. Extracellular Vesicles Derived from Natural Killer Cells Use Multiple Cytotoxic Proteins and Killing Mechanisms to Target Cancer Cells. J. Extracell. Vesicles 2019, 8, 1588538. [Google Scholar] [CrossRef] [PubMed]
  30. Iraci, N.; Leonardi, T.; Gessler, F.; Vega, B.; Pluchino, S. Focus on Extracellular Vesicles: Physiological Role and Signalling Properties of Extracellular Membrane Vesicles. Int. J. Mol. Sci. 2016, 17, 171. [Google Scholar] [CrossRef] [PubMed]
  31. Yi, X.; Huang, D.; Li, Z.; Wang, X.; Yang, T.; Zhao, M.; Wu, J.; Zhong, T. The Role and Application of Small Extracellular Vesicles in Breast Cancer. Front. Oncol. 2022, 12, 980404. [Google Scholar] [CrossRef] [PubMed]
  32. Liu, Y.; Xia, Y.; Smollar, J.; Mao, W.; Wan, Y. The Roles of Small Extracellular Vesicles in Lung Cancer: Molecular Pathology, Mechanisms, Diagnostics, and Therapeutics. Biochim. Biophys. Acta Rev. Cancer 2021, 1876, 188539. [Google Scholar] [CrossRef] [PubMed]
  33. Zhang, X.; Sai, B.; Wang, F.; Wang, L.; Wang, Y.; Zheng, L.; Li, G.; Tang, J.; Xiang, J. Hypoxic BMSC-Derived Exosomal miRNAs Promote Metastasis of Lung Cancer Cells via STAT3-Induced EMT. Mol. Cancer 2019, 18, 40. [Google Scholar] [CrossRef] [PubMed]
  34. Benito-Martín, A.; Jasiulionis, M.G.; García-Silva, S. Extracellular Vesicles and Melanoma: New Perspectives on Tumor Microenvironment and Metastasis. Front. Cell Dev. Biol. 2022, 10, 1061982. [Google Scholar] [CrossRef]
  35. Schneider, N.; Hermann, P.C.; Eiseler, T.; Seufferlein, T. Emerging Roles of Small Extracellular Vesicles in Gastrointestinal Cancer Research and Therapy. Cancers 2024, 16, 567. [Google Scholar] [CrossRef] [PubMed]
  36. Acevedo-Sánchez, V.; Rodríguez-Hernández, R.M.; Aguilar-Ruíz, S.R.; Torres-Aguilar, H.; Romero-Tlalolini, M.d.l.A. Extracellular Vesicles in Cervical Cancer and HPV Infection. Membranes 2021, 11, 453. [Google Scholar] [CrossRef] [PubMed]
  37. Moulin, C.; Crupi, M.J.F.; Ilkow, C.S.; Bell, J.C.; Boulton, S. Extracellular Vesicles and Viruses: Two Intertwined Entities. Int. J. Mol. Sci. 2023, 24, 1036. [Google Scholar] [CrossRef] [PubMed]
  38. Mui, U.N.; Haley, C.T.; Tyring, S.K. Viral Oncology: Molecular Biology and Pathogenesis. J. Clin. Med. 2017, 6, 111. [Google Scholar] [CrossRef] [PubMed]
  39. Nolte-‘t Hoen, E.; Cremer, T.; Gallo, R.C.; Margolis, L.B. Extracellular Vesicles and Viruses: Are They Close Relatives? Proc. Natl. Acad. Sci. USA 2016, 113, 9155–9161. [Google Scholar] [CrossRef] [PubMed]
  40. Alenquer, M.; Amorim, M.J. Exosome Biogenesis, Regulation, and Function in Viral Infection. Viruses 2015, 17, 5066–5083. [Google Scholar] [CrossRef]
  41. Bou, J.-V.; Taguwa, S.; Matsuura, Y. Trick-or-Trap: Extracellular Vesicles and Viral Transmission. Vaccines 2023, 11, 1532. [Google Scholar] [CrossRef] [PubMed]
  42. Khatua, A.K.; Taylor, H.E.; Hildreth, J.E.K.; Popik, W. Exosomes Packaging APOBEC3G Confer Human Immunodeficiency Virus Resistance to Recipient Cells. J. Virol. 2009, 83, 512–521. [Google Scholar] [CrossRef]
  43. Warren, C.J.; Santiago, M.L.; Pyeon, D. APOBEC3: Friend or Foe in Human Papillomavirus Infection and Oncogenesis? Annu. Rev. Virol. 2022, 9, 375–395. [Google Scholar] [CrossRef] [PubMed]
  44. Lovšin, N.; Gangupam, B.; Bergant Marušič, M. The Intricate Interplay between APOBEC3 Proteins and DNA Tumour Viruses. Pathogens 2024, 13, 187. [Google Scholar] [CrossRef] [PubMed]
  45. Kwasnik, M.; Socha, W.; Czech, B.; Wasiak, M.; Rola, J.; Rozek, W. Protein-Coding Region Derived Small RNA in Exosomes from Influenza A Virus–Infected Cells. Int. J. Mol. Sci. 2023, 24, 867. [Google Scholar] [CrossRef] [PubMed]
  46. Wang, X.; Hinson, E.R.; Cresswell, P. The Interferon-Inducible Protein Viperin Inhibits Influenza Virus Release by Perturbing Lipid Rafts. Cell Host Microbe 2007, 2, 96–105. [Google Scholar] [CrossRef] [PubMed]
  47. Nasr, N.; Maddocks, S.; Turville, S.G.; Harman, A.N.; Woolger, N.; Helbig, K.J.; Wilkinson, J.; Bye, C.R.; Wright, T.K.; Rambukwelle, D.; et al. HIV-1 Infection of Human Macrophages Directly Induces Viperin Which Inhibits Viral Production. Blood 2012, 120, 778–788. [Google Scholar] [CrossRef] [PubMed]
  48. Jiang, D.; Guo, H.; Xu, C.; Chang, J.; Gu, B.; Wang, L.; Block, T.M.; Guo, J.-T. Identification of Three Interferon-Inducible Cellular Enzymes That Inhibit the Replication of Hepatitis C Virus. J. Virol. 2008, 82, 1665–1678. [Google Scholar] [CrossRef]
  49. Fernbach, S.; Hale, B.G. SARS-CoV-2 Takes the Bait: Exosomes as Endogenous Decoys. PLoS Biol. 2022, 20, e3001787. [Google Scholar] [CrossRef] [PubMed]
  50. El-Shennawy, L.; Hoffmann, A.D.; Dashzeveg, N.K.; McAndrews, K.M.; Mehl, P.J.; Cornish, D.; Yu, Z.; Tokars, V.L.; Nicolaescu, V.; Tomatsidou, A.; et al. Circulating ACE2-Expressing Extracellular Vesicles Block Broad Strains of SARS-CoV-2. Nat. Commun. 2022, 13, 405. [Google Scholar] [CrossRef] [PubMed]
  51. Lee, J.-H. The Complex Role of Extracellular Vesicles in HIV Infection. BMB Rep. 2023, 56, 335–340. [Google Scholar] [CrossRef] [PubMed]
  52. Carvalho, J.V.d.; Castro, R.O.d.; Silva, E.Z.M.d.; Silveira, P.P.; Silva-Januário, M.E.d.; Arruda, E.; Jamur, M.C.; Oliver, C.; Aguiar, R.S.; daSilva, L.L.P. Nef Neutralizes the Ability of Exosomes from CD4+ T Cells to Act as Decoys during HIV-1 Infection. PLoS ONE 2014, 9, e113691. [Google Scholar] [CrossRef] [PubMed]
  53. Suptawiwat, O.; Ruangrung, K.; Boonarkart, C.; Puthavathana, P.; Maneechotesuwan, K.; Charngkaew, K.; Chomanee, N.; Auewarakul, P. Microparticle and Anti-Influenza Activity in Human Respiratory Secretion. PLoS ONE 2017, 12, e0183717. [Google Scholar] [CrossRef] [PubMed]
  54. Bedford, J.G.; Infusini, G.; Dagley, L.F.; Villalon-Letelier, F.; Zheng, M.Z.M.; Bennett-Wood, V.; Reading, P.C.; Wakim, L.M. Airway Exosomes Released During Influenza Virus Infection Serve as a Key Component of the Antiviral Innate Immune Response. Front. Immunol. 2020, 11, 887. [Google Scholar] [CrossRef] [PubMed]
  55. Bauer, A.N.; Majumdar, N.; Williams, F.; Rajput, S.; Pokhrel, L.R.; Cook, P.P.; Akula, S.M. MicroRNAs: Small but Key Players in Viral Infections and Immune Responses to Viral Pathogens. Biology 2023, 12, 1334. [Google Scholar] [CrossRef] [PubMed]
  56. Qian, X.; Xu, C.; Fang, S.; Zhao, P.; Wang, Y.; Liu, H.; Yuan, W.; Qi, Z. Exosomal MicroRNAs Derived From Umbilical Mesenchymal Stem Cells Inhibit Hepatitis C Virus Infection. Stem Cells Transl. Med. 2016, 5, 1190–1203. [Google Scholar] [CrossRef] [PubMed]
  57. Oh, S.-J.; Lee, E.-N.; Park, J.-H.; Lee, J.K.; Cho, G.J.; Park, I.-H.; Shin, O.S. Anti-Viral Activities of Umbilical Cord Mesenchymal Stem Cell-Derived Small Extracellular Vesicles Against Human Respiratory Viruses. Front. Cell. Infect. Microbiol. 2022, 12. [Google Scholar] [CrossRef] [PubMed]
  58. Heidari, F.; Seyedebrahimi, R.; Yang, P.; Farsani, M.; Ababzadeh, S.; Kalhor, N.; Manoochehri, H.; Sheykhhasan, M.; Azimzadeh, M. Exosomes in Viral Infection: Effects for Pathogenesis and Treatment Strategies. Biocell 2023, 47, 2597–2608. [Google Scholar] [CrossRef]
  59. Souza, A.G.; Colli, L.M. Extracellular Vesicles and Interleukins: Novel Frontiers in Diagnostic and Therapeutic for Cancer. Front. Immunol. 2022, 13, 836922. [Google Scholar] [CrossRef] [PubMed]
  60. Fitzgerald, W.; Freeman, M.L.; Lederman, M.M.; Vasilieva, E.; Romero, R.; Margolis, L. A System of Cytokines Encapsulated in ExtraCellular Vesicles. Sci. Rep. 2018, 8, 8973. [Google Scholar] [CrossRef] [PubMed]
  61. Hong, C.S.; Diergaarde, B.; Whiteside, T.L. Small Extracellular Vesicles in Plasma Carry Luminal Cytokines That Remain Undetectable by Antibody-Based Assays in Cancer Patients and Healthy Donors. BJC Rep. 2024, 2, 16. [Google Scholar] [CrossRef] [PubMed]
  62. Velandia-Romero, M.L.; Calderón-Peláez, M.A.; Balbás-Tepedino, A.; Márquez-Ortiz, R.A.; Madroñero, L.J.; Barreto Prieto, A.; Castellanos, J.E. Extracellular Vesicles of U937 Macrophage Cell Line Infected with DENV-2 Induce Activation in Endothelial Cells EA.Hy926. PLoS ONE 2020, 15, e0227030. [Google Scholar] [CrossRef] [PubMed]
  63. Bernard, M.A.; Zhao, H.; Yue, S.C.; Anandaiah, A.; Koziel, H.; Tachado, S.D. Novel HIV-1 MiRNAs Stimulate TNFα Release in Human Macrophages via TLR8 Signaling Pathway. PLoS ONE 2014, 9, e106006. [Google Scholar] [CrossRef] [PubMed]
  64. Assil, S.; Webster, B.; Dreux, M. Regulation of the Host Antiviral State by Intercellular Communications. Viruses 2015, 7, 4707–4733. [Google Scholar] [CrossRef] [PubMed]
  65. El Safadi, D.; Mokhtari, A.; Krejbich, M.; Lagrave, A.; Hirigoyen, U.; Lebeau, G.; Viranaicken, W.; Krejbich-Trotot, P. Exosome-Mediated Antigen Delivery: Unveiling Novel Strategies in Viral Infection Control and Vaccine Design. Vaccines 2024, 12, 280. [Google Scholar] [CrossRef] [PubMed]
  66. Lindenbergh, M.F.S.; Stoorvogel, W. Antigen Presentation by Extracellular Vesicles from Professional Antigen-Presenting Cells. Annu. Rev. Immunol. 2018, 36, 435–459. [Google Scholar] [CrossRef] [PubMed]
  67. Rey-Cadilhac, F.; Rachenne, F.; Missé, D.; Pompon, J. Viral Components Trafficking with(in) Extracellular Vesicles. Viruses 2023, 15, 2333. [Google Scholar] [CrossRef] [PubMed]
  68. Feng, Z.; Hensley, L.; McKnight, K.L.; Hu, F.; Madden, V.; Ping, L.; Jeong, S.-H.; Walker, C.; Lanford, R.E.; Lemon, S.M. A Pathogenic Picornavirus Acquires an Envelope by Hijacking Cellular Membranes. Nature 2013, 496, 367–371. [Google Scholar] [CrossRef] [PubMed]
  69. Robinson, S.M.; Tsueng, G.; Sin, J.; Mangale, V.; Rahawi, S.; McIntyre, L.L.; Williams, W.; Kha, N.; Cruz, C.; Hancock, B.M.; et al. Coxsackievirus B Exits the Host Cell in Shed Microvesicles Displaying Autophagosomal Markers. PLoS Pathog. 2014, 10, e1004045. [Google Scholar] [CrossRef] [PubMed]
  70. Altan-Bonnet, N. Extracellular Vesicles Are the Trojan Horses of Viral Infection. Curr. Opin. Microbiol. 2016, 32, 77–81. [Google Scholar] [CrossRef] [PubMed]
  71. Yang, J.E.; Rossignol, E.D.; Chang, D.; Zaia, J.; Forrester, I.; Raja, K.; Winbigler, H.; Nicastro, D.; Jackson, W.T.; Bullitt, E. Complexity and Ultrastructure of Infectious Extracellular Vesicles from Cells Infected by Non-Enveloped Virus. Sci. Rep. 2020, 10, 7939. [Google Scholar] [CrossRef] [PubMed]
  72. Fu, Y.; Xiong, S. Exosomes Mediate Coxsackievirus B3 Transmission and Expand the Viral Tropism. PLoS Pathog. 2023, 19, e1011090. [Google Scholar] [CrossRef] [PubMed]
  73. Nagashima, S.; Jirintai, S.; Takahashi, M.; Kobayashi, T.; Tanggis; Nishizawa, T.; Kouki, T.; Yashiro, T.; Okamoto, H. Hepatitis E Virus Egress Depends on the Exosomal Pathway, with Secretory Exosomes Derived from Multivesicular Bodies. J. Gen. Virol. 2014, 95 Pt 10, 2166–2175. [Google Scholar] [CrossRef] [PubMed]
  74. Iša, P.; Pérez-Delgado, A.; Quevedo, I.R.; López, S.; Arias, C.F. Rotaviruses Associate with Distinct Types of Extracellular Vesicles. Viruses 2020, 12, 763. [Google Scholar] [CrossRef] [PubMed]
  75. Wu, Q.; Glitscher, M.; Tonnemacher, S.; Schollmeier, A.; Raupach, J.; Zahn, T.; Eberle, R.; Krijnse-Locker, J.; Basic, M.; Hildt, E. Presence of Intact Hepatitis B Virions in Exosomes. Cell. Mol. Gastroenterol. Hepatol. 2023, 15, 237–259. [Google Scholar] [CrossRef] [PubMed]
  76. Liu, Z.; Zhang, X.; Yu, Q.; He, J.J. Exosome-Associated Hepatitis C Virus in Cell Cultures and Patient Plasma. Biochem. Biophys. Res. Commun. 2014, 455, 218–222. [Google Scholar] [CrossRef] [PubMed]
  77. Chatterjee, S.; Kordbacheh, R.; Sin, J. Extracellular Vesicles: A Novel Mode of Viral Propagation Exploited by Enveloped and Non-Enveloped Viruses. Microorganisms 2024, 12, 274. [Google Scholar] [CrossRef] [PubMed]
  78. Columba Cabezas, S.; Federico, M. Sequences within RNA Coding for HIV-1 Gag P17 Are Efficiently Targeted to Exosomes. Cell. Microbiol. 2013, 15, 412–429. [Google Scholar] [CrossRef]
  79. Sukriti, S.; Choudhary, M.C.; Maras, J.S.; Sharma, S.; Thangariyal, S.; Singh, A.; Das, S.; Islam, M.; Sharma, S.; Trehanpati, N.; et al. Extracellular Vesicles from Hepatitis B Patients Serve as Reservoir of Hepatitis B Virus DNA. J. Viral. Hepat. 2019, 26, 211–214. [Google Scholar] [CrossRef] [PubMed]
  80. Sanada, T.; Hirata, Y.; Naito, Y.; Yamamoto, N.; Kikkawa, Y.; Ishida, Y.; Yamasaki, C.; Tateno, C.; Ochiya, T.; Kohara, M. Transmission of HBV DNA Mediated by Ceramide-Triggered Extracellular Vesicles. Cell. Mol. Gastroenterol. Hepatol. 2017, 3, 272–283. [Google Scholar] [CrossRef] [PubMed]
  81. Ramakrishnaiah, V.; Thumann, C.; Fofana, I.; Habersetzer, F.; Pan, Q.; de Ruiter, P.E.; Willemsen, R.; Demmers, J.A.A.; Stalin Raj, V.; Jenster, G.; et al. Exosome-Mediated Transmission of Hepatitis C Virus between Human Hepatoma Huh7.5 Cells. Proc. Natl. Acad. Sci. USA 2013, 110, 13109–13113. [Google Scholar] [CrossRef] [PubMed]
  82. Mata-Rocha, M.; Rodríguez-Hernández, R.M.; Chávez-Olmos, P.; Garrido, E.; Robles-Vázquez, C.; Aguilar-Ruiz, S.; Torres-Aguilar, H.; González-Torres, C.; Gaytan-Cervantes, J.; Mejía-Aranguré, J.M.; et al. Presence of HPV DNA in Extracellular Vesicles from HeLa Cells and Cervical Samples. Enferm. Infecc. Microbiol. Clin. (Engl. Ed.) 2020, 38, 159–165. [Google Scholar] [CrossRef] [PubMed]
  83. Canitano, A.; Venturi, G.; Borghi, M.; Ammendolia, M.G.; Fais, S. Exosomes Released in Vitro from Epstein-Barr Virus (EBV)-Infected Cells Contain EBV-Encoded Latent Phase mRNAs. Cancer Lett. 2013, 337, 193–199. [Google Scholar] [CrossRef] [PubMed]
  84. Kapoor, N.R.; Chadha, R.; Kumar, S.; Choedon, T.; Reddy, V.S.; Kumar, V. The HBx Gene of Hepatitis B Virus Can Influence Hepatic Microenvironment via Exosomes by Transferring Its mRNA and Protein. Virus Res. 2017, 240, 166–174. [Google Scholar] [CrossRef] [PubMed]
  85. Otaguiri, K.K.; Dos Santos, D.F.; Slavov, S.N.; Depieri, L.V.; Palma, P.V.B.; Meirelles, F.V.; Covas, D.T.; da Silveira, J.C.; Kashima, S. TAX-mRNA-Carrying Exosomes from Human T Cell Lymphotropic Virus Type 1-Infected Cells Can Induce Interferon-Gamma Production In Vitro. AIDS Res. Hum. Retroviruses 2018, 34, 1075–1082. [Google Scholar] [CrossRef] [PubMed]
  86. Chiantore, M.V.; Mangino, G.; Iuliano, M.; Zangrillo, M.S.; De Lillis, I.; Vaccari, G.; Accardi, R.; Tommasino, M.; Columba Cabezas, S.; Federico, M.; et al. Human Papillomavirus E6 and E7 Oncoproteins Affect the Expression of Cancer-Related microRNAs: Additional Evidence in HPV-Induced Tumorigenesis. J. Cancer Res. Clin. Oncol. 2016, 142, 1751–1763. [Google Scholar] [CrossRef] [PubMed]
  87. Wower, I.K.; Brandebourg, T.D.; Wower, J. New Insights on the Mobility of Viral and Host Non-Coding RNAs Reveal Extracellular Vesicles as Intriguing Candidate Antiviral Targets. Pathogens 2020, 9, 876. [Google Scholar] [CrossRef] [PubMed]
  88. Troyer, Z.; Alhusaini, N.; Tabler, C.O.; Sweet, T.; de Carvalho, K.I.L.; Schlatzer, D.M.; Carias, L.; King, C.L.; Matreyek, K.; Tilton, J.C. Extracellular Vesicles Carry SARS-CoV-2 Spike Protein and Serve as Decoys for Neutralizing Antibodies. J. Extracell. Vesicles 2021, 10, e12112. [Google Scholar] [CrossRef] [PubMed]
  89. Masciopinto, F.; Giovani, C.; Campagnoli, S.; Galli-Stampino, L.; Colombatto, P.; Brunetto, M.; Yen, T.S.B.; Houghton, M.; Pileri, P.; Abrignani, S. Association of Hepatitis C Virus Envelope Proteins with Exosomes. Eur. J. Immunol. 2004, 34, 2834–2842. [Google Scholar] [CrossRef] [PubMed]
  90. Arakelyan, A.; Fitzgerald, W.; Zicari, S.; Vanpouille, C.; Margolis, L. Extracellular Vesicles Carry HIV Env and Facilitate Hiv Infection of Human Lymphoid Tissue. Sci. Rep. 2017, 7, 1695. [Google Scholar] [CrossRef] [PubMed]
  91. Zhao, F.; Xu, Y.; Liu, N.; Lv, D.; Chen, Y.; Liu, Z.; Jin, X.; Xiao, M.; Lavillette, D.; Zhong, J.; et al. Extracellular Vesicles from Zika Virus-Infected Cells Display Viral E Protein That Binds ZIKV-Neutralizing Antibodies to Prevent Infection Enhancement. EMBO J. 2023, 42, e112096. [Google Scholar] [CrossRef] [PubMed]
  92. Sami Saribas, A.; Cicalese, S.; Ahooyi, T.M.; Khalili, K.; Amini, S.; Sariyer, I.K. HIV-1 Nef Is Released in Extracellular Vesicles Derived from Astrocytes: Evidence for Nef-Mediated Neurotoxicity. Cell Death Dis. 2018, 8, e2542. [Google Scholar] [CrossRef] [PubMed]
  93. Nkosi, D.; Sun, L.; Duke, L.C.; Meckes, D.G., Jr. Epstein-Barr Virus LMP1 Manipulates the Content and Functions of Extracellular Vesicles to Enhance Metastatic Potential of Recipient Cells. PLOS Pathog. 2020, 16, e1009023. [Google Scholar] [CrossRef]
  94. Narayanan, A.; Jaworski, E.; Van Duyne, R.; Iordanskiy, S.; Guendel, I.; Das, R.; Currer, R.; Sampey, G.; Chung, M.; Kehn-Hall, K.; et al. Exosomes Derived from HTLV-1 Infected Cells Contain the Viral Protein Tax. Retrovirology 2014, 11 (Suppl. 1), O46. [Google Scholar] [CrossRef]
  95. Mack, M.; Kleinschmidt, A.; Brühl, H.; Klier, C.; Nelson, P.J.; 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. Nat. Med. 2000, 6, 769–775. [Google Scholar] [CrossRef] [PubMed]
  96. Thaker, S.K.; Ch’ng, J.; Christofk, H.R. Viral Hijacking of Cellular Metabolism. BMC Biol. 2019, 17, 59. [Google Scholar] [CrossRef] [PubMed]
  97. Koonin, E.V.; Dolja, V.V.; Krupovic, M. The Logic of Virus Evolution. Cell Host Microbe 2022, 30, 917–929. [Google Scholar] [CrossRef] [PubMed]
  98. Zheng, Y.; Wang, M.; Li, S.; Bu, Y.; Xu, Z.; Zhu, G.; Wu, C.; Zhao, K.; Li, A.; Chen, Q.; et al. Hepatitis B Virus Hijacks TSG101 to Facilitate Egress via Multiple Vesicle Bodies. PLoS Pathog. 2023, 19, e1011382. [Google Scholar] [CrossRef] [PubMed]
  99. Mishra, R.; Lata, S.; Ali, A.; Banerjea, A.C. Dengue Haemorrhagic Fever: A Job Done via Exosomes? Emerg. Microbes Infect. 2019, 8, 1626–1635. [Google Scholar] [CrossRef]
  100. Gurunathan, S.; Kang, M.H.; Kim, J.-H. Diverse Effects of Exosomes on COVID-19: A Perspective of Progress From Transmission to Therapeutic Developments. Front. Immunol. 2021, 12, 716407. [Google Scholar] [CrossRef] [PubMed]
  101. 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. [Google Scholar] [CrossRef] [PubMed]
  102. Eliassen, E.; Lum, E.; Pritchett, J.; Ongradi, J.; Krueger, G.; Crawford, J.R.; Phan, T.L.; Ablashi, D.; Hudnall, S.D. Human Herpesvirus 6 and Malignancy: A Review. Front. Oncol. 2018, 8, 512. [Google Scholar] [CrossRef] [PubMed]
  103. Flomm, F.J.; Soh, T.K.; Schneider, C.; Wedemann, L.; Britt, H.M.; Thalassinos, K.; Pfitzner, S.; Reimer, R.; Grünewald, K.; Bosse, J.B. Intermittent Bulk Release of Human Cytomegalovirus. PLoS Pathog. 2022, 18, e1010575. [Google Scholar] [CrossRef] [PubMed]
  104. Gholizadeh, O.; Akbarzadeh, S.; Ghazanfari Hashemi, M.; Gholami, M.; Amini, P.; Yekanipour, Z.; Tabatabaie, R.; Yasamineh, S.; Hosseini, P.; Poortahmasebi, V. Hepatitis A: Viral Structure, Classification, Life Cycle, Clinical Symptoms, Diagnosis Error, and Vaccination. Can. J. Infect. Dis. Med. Microbiol. 2023, 2023, 4263309. [Google Scholar] [CrossRef] [PubMed]
  105. Zhang, R.; Chen, J.; Zi, R.; Ji, L.; Hu, J.; Wu, Z.; Fu, Y. Enterovirus 71-Induced Autophagosome Fusion with Multivesicular Bodies Facilitates Viral RNA Packaging into Exosomes. Microb. Pathog. 2022, 173 Pt A, 105875. [Google Scholar] [CrossRef]
  106. Bhattacharya, B.; Celma, C.C.; Roy, P. Influence of Cellular Trafficking Pathway on Bluetongue Virus Infection in Ovine Cells. Viruses 2015, 7, 2378–2403. [Google Scholar] [CrossRef] [PubMed]
  107. Roy, P. Bluetongue Virus Assembly and Exit Pathways. Adv. Virus. Res. 2020, 108, 249–273. [Google Scholar] [CrossRef] [PubMed]
  108. Hurley, J.H.; Cada, A.K. Inside Job: How the ESCRTs release HIV-1 from infected cells. Biochem. Soc. Trans. 2018, 46, 1029–1036. [Google Scholar] [CrossRef] [PubMed]
  109. Meng, B.; Ip, N.C.Y.; Abbink, T.E.M.; Kenyon, J.C.; Lever, A.M.L. ESCRT-II Functions by Linking to ESCRT-I in Human Immunodeficiency Virus-1 Budding. Cell. Microbiol. 2020, 22, e13161. [Google Scholar] [CrossRef] [PubMed]
  110. Hudait, A.; Hurley, J.H.; Voth, G.A. Dynamics of Upstream ESCRT Organization at the HIV-1 Budding Site. Biophys. J. 2023, 122, 2655–2674. [Google Scholar] [CrossRef] [PubMed]
  111. Arii, J.; Watanabe, M.; Maeda, F.; Tokai-Nishizumi, N.; Chihara, T.; Miura, M.; Maruzuru, Y.; Koyanagi, N.; Kato, A.; Kawaguchi, Y. ESCRT-III Mediates Budding across the Inner Nuclear Membrane and Regulates Its Integrity. Nat. Commun. 2018, 9, 3379. [Google Scholar] [CrossRef] [PubMed]
  112. Barnes, J.; Wilson, D.W. Seeking Closure: How Do Herpesviruses Recruit the Cellular ESCRT Apparatus? J. Virol. 2019, 93, e00392-19. [Google Scholar] [CrossRef] [PubMed]
  113. Moore, P.S.; Chang, Y. Why Do Viruses Cause Cancer? Highlights of the First Century of Human Tumour Virology. Nat. Rev. Cancer 2010, 10, 878–889. [Google Scholar] [CrossRef] [PubMed]
  114. Chang, Y.; Moore, P.S.; Weiss, R.A. Human Oncogenic Viruses: Nature and Discovery. Philosophical. Trans. R. Soc. B Biol. Sci. 2017, 372, 20160264. [Google Scholar] [CrossRef] [PubMed]
  115. Green, V.A.; Munshi, S.U.; Marakalala, M.J.; Mourão, M.M. Molecular Mechanisms of Viral Infection and Propagation: An Overview of the Second Advanced Summer School in Africa. IUBMB Life 2010, 62, 573–583. [Google Scholar] [CrossRef] [PubMed]
  116. Owliaee, I.; Khaledian, M.; Boroujeni, A.K.; Shojaeian, A. Engineered Small Extracellular Vesicles as a Novel Platform to Suppress Human Oncovirus-Associated Cancers. Infect. Agents Cancer 2023, 18, 69. [Google Scholar] [CrossRef] [PubMed]
  117. Wu, W.; Wu, D.; Yan, W.; Wang, Y.; You, J.; Wan, X.; Xi, D.; Luo, X.; Han, M.; Ning, Q. Interferon-Induced Macrophage-Derived Exosomes Mediate Antiviral Activity Against Hepatitis B Virus Through miR-574-5p. J. Infect Dis. 2021, 223, 686–698. [Google Scholar] [CrossRef] [PubMed]
  118. Chinnici, C.M.; Russelli, G.; Bulati, M.; Miceli, V.; Gallo, A.; Busà, R.; Tinnirello, R.; Conaldi, P.G.; Iannolo, G. Mesenchymal Stromal Cell Secretome in Liver Failure: Perspectives on COVID-19 Infection Treatment. World J. Gastroenterol. 2021, 27, 1905–1919. [Google Scholar] [CrossRef] [PubMed]
  119. Jaworski, E.; Narayanan, A.; Van Duyne, R.; Shabbeer-Meyering, S.; Iordanskiy, S.; Saifuddin, M.; Das, R.; Afonso, P.V.; Sampey, G.C.; Chung, M.; et al. Human T-Lymphotropic Virus Type 1-Infected Cells Secrete Exosomes That Contain Tax Protein. J. Biol. Chem. 2014, 289, 22284–22305. [Google Scholar] [CrossRef] [PubMed]
  120. Kawano, K.; Hashikura, Y.; Umekita, K. Purification Method of Extracellular Vesicles Derived from Human T-Cell Leukemia Virus Type 1-Infected Cells without Virions. Viruses 2024, 16, 249. [Google Scholar] [CrossRef]
  121. Pinto, D.O.; Al Sharif, S.; Mensah, G.; Cowen, M.; Khatkar, P.; Erickson, J.; Branscome, H.; Lattanze, T.; DeMarino, C.; Alem, F.; et al. Extracellular Vesicles from HTLV-1 Infected Cells Modulate Target Cells and Viral Spread. Retrovirology 2021, 18, 6. [Google Scholar] [CrossRef] [PubMed]
  122. Kalinichenko, S.; Komkov, D.; Mazurov, D. HIV-1 and HTLV-1 Transmission Modes: Mechanisms and Importance for Virus Spread. Viruses 2022, 14, 152. [Google Scholar] [CrossRef] [PubMed]
  123. El-Saghir, J.; Nassar, F.; Tawil, N.; El-Sabban, M. ATL-Derived Exosomes Modulate Mesenchymal Stem Cells: Potential Role in Leukemia Progression. Retrovirology 2016, 13, 73. [Google Scholar] [CrossRef] [PubMed]
  124. Dreux, M.; Garaigorta, U.; Boyd, B.; Décembre, E.; Chung, J.; Whitten-Bauer, C.; Wieland, S.; Chisari, F.V. Short-Range Exosomal Transfer of Viral RNA from Infected Cells to Plasmacytoid Dendritic Cells Triggers Innate Immunity. Cell Host Microbe 2012, 12, 558–570. [Google Scholar] [CrossRef]
  125. Bukong, T.; 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 Pathog. 2014, 10, e1004424. [Google Scholar] [CrossRef] [PubMed]
  126. Karamichali, E.; Chihab, H.; Kakkanas, A.; Marchio, A.; Karamitros, T.; Pogka, V.; Varaklioti, A.; Kalliaropoulos, A.; Martinez-Gonzales, B.; Foka, P.; et al. HCV Defective Genomes Promote Persistent Infection by Modulating the Viral Life Cycle. Front. Microbiol. 2018, 9, 2942. [Google Scholar] [CrossRef] [PubMed]
  127. Giannessi, F.; Aiello, A.; Franchi, F.; Percario, Z.A.; Affabris, E. The Role of Extracellular Vesicles as Allies of HIV, HCV and SARS Viruses. Viruses 2020, 12, 571. [Google Scholar] [CrossRef] [PubMed]
  128. Pinto, D.O.; DeMarino, C.; Pleet, M.L.; Cowen, M.; Branscome, H.; Al Sharif, S.; Jones, J.; Dutartre, H.; Lepene, B.; Liotta, L.A.; et al. HTLV-1 Extracellular Vesicles Promote Cell-to-Cell Contact. Front. Microbiol. 2019, 10, 2147. [Google Scholar] [CrossRef] [PubMed]
  129. Sasaki, R.; Kanda, T.; Yokosuka, O.; Kato, N.; Matsuoka, S.; Moriyama, M. Exosomes and Hepatocellular Carcinoma: From Bench to Bedside. Int. J. Mol. Sci. 2019, 20, 1406. [Google Scholar] [CrossRef]
  130. Wang, H.; Hou, L.; Li, A.; Duan, Y.; Gao, H.; Song, X. Expression of Serum Exosomal MicroRNA-21 in Human Hepatocellular Carcinoma. Biomed Res. Int. 2014, 2014, 864894. [Google Scholar] [CrossRef] [PubMed]
  131. Badami, E.; Carcione, C.; Chinnici, C.M.; Tinnirello, R.; Conaldi, P.G.; Iannolo, G. HCV Interplay With Mir34a: Implications in Hepatocellular Carcinoma. Front. Oncol. 2021, 11, 803278. [Google Scholar] [CrossRef] [PubMed]
  132. Kouwaki, T.; Fukushima, Y.; Daito, T.; Sanada, T.; Yamamoto, N.; Mifsud, E.J.; Leong, C.R.; Tsukiyama-Kohara, K.; Kohara, M.; Matsumoto, M.; et al. Extracellular Vesicles Including Exosomes Regulate Innate Immune Responses to Hepatitis B Virus Infection. Front. Immunol. 2016, 7, 335. [Google Scholar] [CrossRef] [PubMed]
  133. Yang, Y.; Han, Q.; Hou, Z.; Zhang, C.; Tian, Z.; Zhang, J. Exosomes Mediate Hepatitis B Virus (HBV) Transmission and NK-Cell Dysfunction. Cell. Mol. Immunol. 2017, 14, 465–475. [Google Scholar] [CrossRef] [PubMed]
  134. Chen, H.; Cao, D.; Han, N.; Zhang, M.; Jiang, W.; Wang, X.; Zeng, Q.; Tang, H. Hepatitis B Virus-Encoded MicroRNA (HBV-miR-3) Inhibits FIH-1 Expression to Promote Tumor Angiogenesis in HBV-Related Hepatocellular Carcinoma. J. Hepatocell. Carcinoma 2023, 10, 2337–2353. [Google Scholar] [CrossRef] [PubMed]
  135. Fu, S.; Zhou, R.; Li, N.; Huang, Y.; Fan, X.-G. Hepatitis B Virus X Protein in Liver Tumor Microenvironment. Tumor. Biol. 2016, 37, 15371–15381. [Google Scholar] [CrossRef] [PubMed]
  136. Nagel, R.; Martens-de Kemp, S.R.; Buijze, M.; Jacobs, G.; Braakhuis, B.J.M.; Brakenhoff, R.H. Treatment Response of HPV-Positive and HPV-Negative Head and Neck Squamous Cell Carcinoma Cell Lines. Oral Oncol. 2013, 49, 560–566. [Google Scholar] [CrossRef]
  137. Honegger, A.; Leitz, J.; Bulkescher, J.; Hoppe-Seyler, K.; Hoppe-Seyler, F. Silencing of Human Papillomavirus (HPV) E6/E7 Oncogene Expression Affects Both the Contents and the Amounts of Extracellular Microvesicles Released from HPV-Positive Cancer Cells. Int. J. Cancer 2013, 133, 1631–1642. [Google Scholar] [CrossRef] [PubMed]
  138. Lo, A.K.-F.; Dawson, C.W.; Lung, H.L.; Wong, K.-L.; Young, L.S. The Role of EBV-Encoded LMP1 in the NPC Tumor Microenvironment: From Function to Therapy. Front. Oncol. 2021, 11, 640207. [Google Scholar] [CrossRef] [PubMed]
  139. Aga, M.; Bentz, G.L.; Raffa, S.; Torrisi, M.R.; Kondo, S.; Wakisaka, N.; Yoshizaki, T.; Pagano, J.S.; Shackelford, J. Exosomal HIF1α Supports Invasive Potential of Nasopharyngeal Carcinoma-Associated LMP1-Positive Exosomes. Oncogene 2014, 33, 4613–4622. [Google Scholar] [CrossRef] [PubMed]
  140. Gallo, A.; Jang, S.-I.; Ong, H.L.; Perez, P.; Tandon, M.; Ambudkar, I.; Illei, G.; Alevizos, I. Targeting the Ca2+ Sensor STIM1 by Exosomal Transfer of Ebv-miR-BART13-3p Is Associated with Sjögren’s Syndrome. EBioMedicine 2016, 10, 216–226. [Google Scholar] [CrossRef] [PubMed]
  141. Meckes, D.G.; Shair, K.H.Y.; Marquitz, A.R.; Kung, C.-P.; Edwards, R.H.; Raab-Traub, N. Human Tumor Virus Utilizes Exosomes for Intercellular Communication. Proc. Natl. Acad. Sci. USA 2010, 107, 20370–20375. [Google Scholar] [CrossRef] [PubMed]
  142. Mrizak, D.; Martin, N.; Barjon, C.; Jimenez-Pailhes, A.-S.; Mustapha, R.; Niki, T.; Guigay, J.; Pancré, V.; de Launoit, Y.; Busson, P.; et al. Effect of Nasopharyngeal Carcinoma-Derived Exosomes on Human Regulatory T Cells. J. Natl. Cancer Inst. 2015, 107, 363. [Google Scholar] [CrossRef] [PubMed]
  143. Klibi, J.; Niki, T.; Riedel, A.; Pioche-Durieu, C.; Souquere, S.; Rubinstein, E.; Le Moulec, S.; Guigay, J.; Hirashima, M.; Guemira, F.; et al. 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. [Google Scholar] [CrossRef] [PubMed]
  144. Chugh, P.E.; Sin, S.-H.; Ozgur, S.; Henry, D.H.; Menezes, P.; Griffith, J.; Eron, J.J.; Damania, B.; Dittmer, D.P. Systemically Circulating Viral and Tumor-Derived microRNAs in KSHV-Associated Malignancies. PLoS Pathog. 2013, 9, e1003484. [Google Scholar] [CrossRef] [PubMed]
  145. 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. [Google Scholar] [CrossRef] [PubMed]
  146. Barrett, L.; Dai, L.; Wang, S.; Qin, Z. Kaposi’s Sarcoma-Associated Herpesvirus and Extracellular Vesicles. J. Med. Virol. 2021, 93, 3294–3299. [Google Scholar] [CrossRef] [PubMed]
  147. Singh, V.V.; Kerur, N.; Bottero, V.; Dutta, S.; Chakraborty, S.; Ansari, M.A.; Paudel, N.; Chikoti, L.; Chandran, B. Kaposi’s Sarcoma-Associated Herpesvirus Latency in Endothelial and B Cells Activates Gamma Interferon-Inducible Protein 16-Mediated Inflammasomes. J. Virol. 2013, 87, 4417–4431. [Google Scholar] [CrossRef] [PubMed]
  148. Jeon, H.; Lee, J.; Lee, S.; Kang, S.-K.; Park, S.J.; Yoo, S.-M.; Lee, M.-S. Extracellular Vesicles From KSHV-Infected Cells Stimulate Antiviral Immune Response Through Mitochondrial DNA. Front. Immunol. 2019, 10, 876. [Google Scholar] [CrossRef]
  149. Yang, R.; Lee, E.E.; Kim, J.; Choi, J.H.; Kolitz, E.; Chen, Y.; Crewe, C.; Salisbury, N.J.H.; Scherer, P.E.; Cockerell, C.; et al. Characterization of ALTO-Encoding Circular RNAs Expressed by Merkel Cell Polyomavirus and Trichodysplasia Spinulosa Polyomavirus. PLoS Pathog. 2021, 17, e1009582. [Google Scholar] [CrossRef]
  150. Renwick, N.; Cekan, P.; Masry, P.A.; McGeary, S.E.; Miller, J.B.; Hafner, M.; Li, Z.; Mihailovic, A.; Morozov, P.; Brown, M.; et al. Multicolor microRNA FISH Effectively Differentiates Tumor Types. J. Clin. Investig. 2013, 123, 2694–2702. [Google Scholar] [CrossRef] [PubMed]
  151. Emanuelson, C.; Ankenbruck, N.; Kumbhare, R.; Thomas, M.; Connelly, C.; Baktash, Y.; Randall, G.; Deiters, A. Transcriptional Inhibition of MicroRNA miR-122 by Small Molecules Reduces Hepatitis C Virus Replication in Liver Cells. J. Med. Chem. 2022, 65, 16338–16352. [Google Scholar] [CrossRef] [PubMed]
  152. Panigrahi, M.; Palmer, M.A.; Wilson, J.A. MicroRNA-122 Regulation of HCV Infections: Insights from Studies of miR-122-Independent Replication. Pathogens 2022, 11, 1005. [Google Scholar] [CrossRef] [PubMed]
  153. Cabral, B.C.A.; Hoffmann, L.; Bottaro, T.; Costa, P.F.; Ramos, A.L.A.; Coelho, H.S.M.; Villela-Nogueira, C.A.; Ürményi, T.P.; Faffe, D.S.; Silva, R. Circulating microRNAs Associated with Liver Fibrosis in Chronic Hepatitis C Patients. Biochem. Biophys. Rep. 2020, 24, 100814. [Google Scholar] [CrossRef] [PubMed]
  154. Liu, Z.; Li, Y.; Wang, Y.; Bai, X.; Zhang, Y. Exosomes in HBV Infection. Clin. Chim. Acta 2023, 538, 65–69. [Google Scholar] [CrossRef] [PubMed]
  155. Jia, X.; Chen, J.; Megger, D.A.; Zhang, X.; Kozlowski, M.; Zhang, L.; Fang, Z.; Li, J.; Chu, Q.; Wu, M.; et al. Label-Free Proteomic Analysis of Exosomes Derived from Inducible Hepatitis B Virus-Replicating HepAD38 Cell Line. Mol. Cell. Proteom. 2017, 16 (Suppl. 1), S144–S160. [Google Scholar] [CrossRef] [PubMed]
  156. Todorova, V.K.; Byrum, S.D.; Mackintosh, S.G.; Jamshidi-Parsian, A.; Gies, A.J.; Washam, C.L.; Jenkins, S.V.; Spiva, T.; Bowman, E.; Reyna, N.S.; et al. Exosomal MicroRNA and Protein Profiles of Hepatitis B Virus-Related Hepatocellular Carcinoma Cells. Int. J. Mol. Sci. 2023, 24, 13098. [Google Scholar] [CrossRef] [PubMed]
  157. Kakizaki, M.; Yamamoto, Y.; Otsuka, M.; Kitamura, K.; Ito, M.; Kawai, H.D.; Muramatsu, M.; Kagawa, T.; Kotani, A. Extracellular Vesicles Secreted by HBV-Infected Cells Modulate HBV Persistence in Hydrodynamic HBV Transfection Mouse Model. J. Biol. Chem. 2020, 295, 12449–12460. [Google Scholar] [CrossRef] [PubMed]
  158. Huang, Z.-Y.; Xu, P.; Li, J.-H.; Zeng, C.-H.; Song, H.-F.; Chen, H.; Zhu, Y.-B.; Song, Y.-Y.; Lu, H.-L.; Shen, C.-P.; et al. Clinical Significance of Dynamics of Programmed Death Ligand-1 Expression on Circulating CD14+ Monocytes and CD19+ B Cells with the Progression of Hepatitis B Virus Infection. Viral. Immunol. 2017, 30, 224–231. [Google Scholar] [CrossRef] [PubMed]
  159. Blank, C.; Mackensen, A. Contribution of the PD-L1/PD-1 Pathway to T-Cell Exhaustion: An Update on Implications for Chronic Infections and Tumor Evasion. Cancer Immunol. Immunother. 2007, 56, 739–745. [Google Scholar] [CrossRef] [PubMed]
  160. Zhao, X.; Wu, Y.; Duan, J.; Ma, Y.; Shen, Z.; Wei, L.; Cui, X.; Zhang, J.; Xie, Y.; Liu, J. Quantitative Proteomic Analysis of Exosome Protein Content Changes Induced by Hepatitis B Virus in Huh-7 Cells Using SILAC Labeling and LC-MS/MS. J. Proteome Res. 2014, 13, 5391–5402. [Google Scholar] [CrossRef] [PubMed]
  161. Wei, X.-C.; Xia, Y.-R.; Zhou, P.; Xue, X.; Ding, S.; Liu, L.-J.; Zhu, F. Hepatitis B Core Antigen Modulates Exosomal miR-135a to Target Vesicle-Associated Membrane Protein 2 Promoting Chemoresistance in Hepatocellular Carcinoma. World J. Gastroenterol. 2021, 27, 8302–8322. [Google Scholar] [CrossRef] [PubMed]
  162. Guenat, D.; Hermetet, F.; Prétet, J.-L.; Mougin, C. Exosomes and Other Extracellular Vesicles in HPV Transmission and Carcinogenesis. Viruses 2017, 9, 211. [Google Scholar] [CrossRef] [PubMed]
  163. Khan, S.; Jutzy, J.M.S.; Aspe, J.R.; McGregor, D.W.; Neidigh, J.W.; Wall, N.R. Survivin Is Released from Cancer Cells via Exosomes. Apoptosis 2011, 16, 1–12. [Google Scholar] [CrossRef] [PubMed]
  164. Valenzuela, M.M.A.; Ferguson Bennit, H.R.; Gonda, A.; Diaz Osterman, C.J.; Hibma, A.; Khan, S.; Wall, N.R. Exosomes Secreted from Human Cancer Cell Lines Contain Inhibitors of Apoptosis (IAP). Cancer Microenviron. 2015, 8, 65–73. [Google Scholar] [CrossRef] [PubMed]
  165. 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 Pathog. 2015, 11, e1004712. [Google Scholar] [CrossRef] [PubMed]
  166. Kannan, A.; Hertweck, K.L.; Philley, J.V.; Wells, R.B.; Dasgupta, S. Genetic Mutation and Exosome Signature of Human Papilloma Virus Associated Oropharyngeal Cancer. Sci. Rep. 2017, 7, 46102. [Google Scholar] [CrossRef] [PubMed]
  167. Yim, E.-K.; Park, J.-S. The Role of HPV E6 and E7 Oncoproteins in HPV-Associated Cervical Carcinogenesis. Cancer Res. Treat. 2005, 37, 319–324. [Google Scholar] [CrossRef] [PubMed]
  168. Dyson, N.; Howley, P.M.; Münger, K.; Harlow, E. The Human Papilloma Virus-16 E7 Oncoprotein Is Able to Bind to the Retinoblastoma Gene Product. Science 1989, 243, 934–937. [Google Scholar] [CrossRef] [PubMed]
  169. Ludwig, S.; Sharma, P.; Theodoraki, M.-N.; Pietrowska, M.; Yerneni, S.S.; Lang, S.; Ferrone, S.; Whiteside, T.L. Molecular and Functional Profiles of Exosomes From HPV(+) and HPV(−) Head and Neck Cancer Cell Lines. Front. Oncol. 2018, 8, 445. [Google Scholar] [CrossRef] [PubMed]
  170. Pennisi, R.; Trischitta, P.; Costa, M.; Venuti, A.; Tamburello, M.P.; Sciortino, M.T. Update of Natural Products and Their Derivatives Targeting Epstein–Barr Infection. Viruses 2024, 16, 124. [Google Scholar] [CrossRef]
  171. Pegtel, D.M.; Cosmopoulos, K.; Thorley-Lawson, D.A.; van Eijndhoven, M.A.J.; Hopmans, E.S.; Lindenberg, J.L.; de Gruijl, T.D.; Würdinger, T.; Middeldorp, J.M. Functional Delivery of Viral miRNAs via Exosomes. Proc. Natl. Acad. Sci. USA 2010, 107, 6328–6333. [Google Scholar] [CrossRef] [PubMed]
  172. Giehler, F.; Ostertag, M.S.; Sommermann, T.; Weidl, D.; Sterz, K.R.; Kutz, H.; Moosmann, A.; Feller, S.M.; Geerlof, A.; Biesinger, B.; et al. Epstein-Barr Virus-Driven B Cell Lymphoma Mediated by a Direct LMP1-TRAF6 Complex. Nat. Commun. 2024, 15, 414. [Google Scholar] [CrossRef] [PubMed]
  173. Huang, X.; Zhang, M.; Zhang, Z. The Role of LMP1 in Epstein-Barr Virus-Associated Gastric Cancer. Curr. Cancer Drug Targets 2024, 24, 127–141. [Google Scholar] [CrossRef] [PubMed]
  174. Zeng, M.; Chen, Y.; Jia, X.; Liu, Y. The Anti-Apoptotic Role of EBV-LMP1 in Lymphoma Cells. Cancer Manag. Res. 2020, 12, 8801–8811. [Google Scholar] [CrossRef] [PubMed]
  175. Jeon, H.; Yoo, S.-M.; Choi, H.S.; Mun, J.Y.; Kang, H.-G.; Lee, J.; Park, J.; Gao, S.-J.; Lee, M.-S. Extracellular Vesicles from KSHV-Infected Endothelial Cells Activate the Complement System. Oncotarget 2017, 8, 99841–99860. [Google Scholar] [CrossRef] [PubMed]
  176. Hussein, H.A.M.; Alfhili, M.A.; Pakala, P.; Simon, S.; Hussain, J.; McCubrey, J.A.; Akula, S.M. miRNAs and Their Roles in KSHV Pathogenesis. Virus Res. 2019, 266, 15–24. [Google Scholar] [CrossRef]
  177. Yogev, O.; Lagos, D.; Enver, T.; Boshoff, C. Kaposi’s Sarcoma Herpesvirus microRNAs Induce Metabolic Transformation of Infected Cells. PLoS Pathog. 2014, 10, e1004400. [Google Scholar] [CrossRef] [PubMed]
  178. Yogev, O.; Henderson, S.; Hayes, M.J.; Marelli, S.S.; Ofir-Birin, Y.; Regev-Rudzki, N.; Herrero, J.; Enver, T. Herpesviruses Shape Tumour Microenvironment through Exosomal Transfer of Viral microRNAs. PLoS Pathog. 2017, 13, e1006524. [Google Scholar] [CrossRef] [PubMed]
  179. Dass, D.; Dhotre, K.; Chakraborty, M.; Nath, A.; Banerjee, A.; Bagchi, P.; Mukherjee, A. miRNAs in Herpesvirus Infection: Powerful Regulators in Small Packages. Viruses 2023, 15, 429. [Google Scholar] [CrossRef] [PubMed]
  180. Feng, H.; Shuda, M.; Chang, Y.; Moore, P.S. Clonal Integration of a Polyomavirus in Human Merkel Cell Carcinoma. Science 2008, 319, 1096–1100. [Google Scholar] [CrossRef] [PubMed]
  181. Konstantinell, A.; Bruun, J.-A.; Olsen, R.; Aspar, A.; Škalko-Basnet, N.; Sveinbjørnsson, B.; Moens, U. Secretomic Analysis of Extracellular Vesicles Originating from Polyomavirus-Negative and Polyomavirus-Positive Merkel Cell Carcinoma Cell Lines. Proteomics 2016, 16, 2587–2591. [Google Scholar] [CrossRef] [PubMed]
  182. Fan, K.; Spassova, I.; Gravemeyer, J.; Ritter, C.; Horny, K.; Lange, A.; Gambichler, T.; Ødum, N.; Schrama, D.; Schadendorf, D.; et al. Merkel Cell Carcinoma-Derived Exosome-Shuttle miR-375 Induces Fibroblast Polarization by Inhibition of RBPJ and P53. Oncogene 2021, 40, 980–996. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The influence of small extracellular vesicles (sEV) in viral infections. sEV can promote (A) or inhibit viral infections (B). The mechanisms underlying the sEV ability to facilitate viral infections are multifaceted, including the transmission of viral cargo, the delivery of entry receptors for the virus to new cells, and the binding of neutralizing antibodies. On the other hand, sEV antiviral effects are associated with the delivery of host restriction factors, such as APOBEC3G (apolipoprotein B mRNA editing enzyme catalytic subunit 3G) and IFTIM3 (interferon-induced transmembrane protein 3), as well as miRNAs that condition immunity to infection. sEV can also bind to virions (the „bait” mechanism), induce antiviral immune responses by delivering viral pathogen-associated molecular patterns (PAMPs) to antigen presenting cells (APCs), and promote the activation of natural killer cells (NK cells) and proliferation of CD4+ and CD8+ T cells. Created with BioRender.com.
Figure 1. The influence of small extracellular vesicles (sEV) in viral infections. sEV can promote (A) or inhibit viral infections (B). The mechanisms underlying the sEV ability to facilitate viral infections are multifaceted, including the transmission of viral cargo, the delivery of entry receptors for the virus to new cells, and the binding of neutralizing antibodies. On the other hand, sEV antiviral effects are associated with the delivery of host restriction factors, such as APOBEC3G (apolipoprotein B mRNA editing enzyme catalytic subunit 3G) and IFTIM3 (interferon-induced transmembrane protein 3), as well as miRNAs that condition immunity to infection. sEV can also bind to virions (the „bait” mechanism), induce antiviral immune responses by delivering viral pathogen-associated molecular patterns (PAMPs) to antigen presenting cells (APCs), and promote the activation of natural killer cells (NK cells) and proliferation of CD4+ and CD8+ T cells. Created with BioRender.com.
Viruses 16 01291 g001
Figure 2. Role of sEV in HBV infection and the development of HBV-induced liver disease. sEV secreted from HBV-infected cells transport bioactive cargo and may promote progression of HBV-dependent HCC. Created with BioRender.com.
Figure 2. Role of sEV in HBV infection and the development of HBV-induced liver disease. sEV secreted from HBV-infected cells transport bioactive cargo and may promote progression of HBV-dependent HCC. Created with BioRender.com.
Viruses 16 01291 g002
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

Ważny, Ł.; Whiteside, T.L.; Pietrowska, M. Oncoviral Infections and Small Extracellular Vesicles. Viruses 2024, 16, 1291. https://doi.org/10.3390/v16081291

AMA Style

Ważny Ł, Whiteside TL, Pietrowska M. Oncoviral Infections and Small Extracellular Vesicles. Viruses. 2024; 16(8):1291. https://doi.org/10.3390/v16081291

Chicago/Turabian Style

Ważny, Łukasz, Theresa L. Whiteside, and Monika Pietrowska. 2024. "Oncoviral Infections and Small Extracellular Vesicles" Viruses 16, no. 8: 1291. https://doi.org/10.3390/v16081291

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