Next Article in Journal
Development of Open-Field Behaviour in the Medaka, Oryzias latipes
Next Article in Special Issue
Prostaglandin E2 and Cancer: Insight into Tumor Progression and Immunity
Previous Article in Journal
Evaluation for the Genetic Association between Store-Operated Calcium Influx Pathway (STIM1 and ORAI1) and Human Hepatocellular Carcinoma in Patients with Chronic Hepatitis B Infection
Previous Article in Special Issue
IL-6, NLR, and SII Markers and Their Relation with Alterations in CD8+ T-Lymphocyte Subpopulations in Patients Treated for Lung Adenocarcinoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Lymphotropic Viruses: Chronic Inflammation and Induction of Cancers

1
Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, PA 17033, USA
2
Department of Microbiology and Immunology, Sylvester Comprehensive Cancer Center Miller School of Medicine, University of Miami, Miami, FL 33136, USA
*
Author to whom correspondence should be addressed.
Biology 2020, 9(11), 390; https://doi.org/10.3390/biology9110390
Submission received: 20 October 2020 / Revised: 4 November 2020 / Accepted: 6 November 2020 / Published: 10 November 2020
(This article belongs to the Special Issue Inflammation in Cancer: A Target for Prevention and Therapy)

Abstract

:

Simple Summary

Infection with viruses such as HTLV-1, EBV and KSHV has been linked to many cancers, including leukemias and lymphomas in humans worldwide. These viruses establish life-long latency after initial infections. Currently, there are no effective treatments for the prevention of cancers associated with these viruses. Studies have shown that chronic inflammation mediated by the transcription factors STAT3 and NF-κB in HTLV-1, EBV and KSHV-infected cells play critical roles in the development of viral-associated cancers. Inhibition of STAT3 and NF-κB activation in cells harboring these latent viruses leads to their destruction and the production of new virus particles. De novo infection by these viruses induces rapid NF-κB and STAT3 activation and creates a favorable environment for virus entry into host cells and viral latency. However, the host factors and the mechanisms required for rapid NF-κB and STAT3 activation during de novo infection and latency by these viruses are largely unknown. In this review, we will discuss the mechanisms that are specifically involved in NF-κB and STAT3 activation during de novo infection and latency by KSHV, EBV and HTLV-1.

Abstract

Inflammation induced by transcription factors, including Signal Transducers and Activators of Transcription (STATs) and NF-κB, in response to microbial pathogenic infections and ligand dependent receptors stimulation are critical for controlling infections. However, uncontrolled inflammation induced by these transcription factors could lead to immune dysfunction, persistent infection, inflammatory related diseases and the development of cancers. Although the induction of innate immunity and inflammation in response to viral infection is important to control virus replication, its effects can be modulated by lymphotropic viruses including human T-cell leukemia virus type 1 (HTLV-1), Κaposi’s sarcoma herpesvirus (KSHV), and Epstein Barr virus (EBV) during de novo infection as well as latent infection. These lymphotropic viruses persistently activate JAK-STAT and NF-κB pathways. Long-term STAT and NF-κB activation by these viruses leads to the induction of chronic inflammation, which can support the persistence of these viruses and promote virus-mediated cancers. Here, we review how HTLV-1, KSHV and EBV hijack the function of host cell surface molecules (CSMs), which are involved in the regulation of chronic inflammation, innate and adaptive immune responses, cell death and the restoration of tissue homeostasis. Thus, better understanding of CSMs-mediated chronic activation of STATs and NF-κB pathways in lymphotropic virus-infected cells may pave the way for therapeutic intervention in malignancies caused by lymphotropic viruses.

1. Introduction

Lymphotropic Viruses and Cancers

Infection with lymphotropic viruses, including Epstein Barr virus/human herpesvirus 4 (EBV/HH4), Κaposi’s sarcoma herpesvirus/human herpesvirus 8 (KSHV/HHV8), and human T-cell leukemia virus type 1 (HTLV-1), is associated with the development of cancers worldwide [1]. The rapid induction of inflammation by these viruses plays a critical role in the development of many cancers [2,3]. While both EBV and KSHV are linear double stranded DNA viruses [4,5], HTLV-1 is a retrovirus [6]. EBV was the first human lymphotropic virus associated with B-cell and T-cell lymphoproliferative disease, discovered in 1964 [7,8,9], and one of the most common viral infections in humans [10,11]. EBV has been associated with the etiology of multiple human cancers of both lymphoid and epithelial origin, including Burkitt’s Lymphoma, Hodgkin lymphoma, and Nasopharyngeal Carcinoma and EBV-associated gastric cancer [12]. KSHV, discovered in 1994, is associated with Kaposi’s sarcoma (KS), and two lymphoproliferative disorders called Primary effusion lymphoma (PEL) and multicentric Castleman’s Disease (MCD) [13,14,15]. HTLV-1 was discovered in 1980 and is linked to two main diseases: a malignant T-cell lymphoproliferation known as Adult T-cell Leukemia/Lymphoma (ATLL) and Tropical Spastic Paraparesis/HTLV-1-Associated Myelopathy (TSP/HAM) [16,17,18]. EBV, KSHV and HTLV-1 often establish a life-long asymptomatic latent infection to develop these cancers [19]. These tumor viruses exhibit a tropism for either B-, T-, epithelial or endothelial cells [19,20,21].

2. Inflammation and Oncogenesis

2.1. NF-κB

Nuclear factor-κB (NF-κB), a family of transcription factors, is ubiquitously expressed and is critical for the regulation of innate and adaptive immune functions [22,23]. NF-κB family members consist of NF-κB1 (p50), NF-κB2 (p52), RelA (p65), RelB and c-Rel, which can form various combinations of homo- and heterodimers [22,23]. All NF-κB subunits contain a conserved Rel homology domain that facilitates DNA binding, nuclear localization and dimerization of NF-κB proteins. NF-κB is activated by canonical and non-canonical signaling pathways. Canonical NF-κB signaling is responsible for the induction of pro-inflammatory cytokines, chemokines and inflammatory mediators in different types of innate immune cells [24]. The non-canonical NF-κB signaling pathway plays critical roles in lymphoid organogenesis, B-cell survival and maturation, dendritic cell activation and bone metabolism [25,26]. The signaling components and receptors involved in the activation of both canonical and non-canonical pathways are distinct.
The NF-κB proteins are normally held inactive in the cytoplasm by the inhibitor IκBα and other IκB family members, which contain ankyrin repeat domains. The NF-κB1 and NF-κB2 proteins are derived from precursor proteins, p105 and p100, respectively. In response to receptor stimulation or microbial infection, IκBα is rapidly phosphorylated by the IκB kinase (IKK) complex, consisting of two catalytically active kinases, IKKα and IKKβ, and a regulatory subunit IKKγ (NEMO), in the canonical NF-κB signaling pathway (Figure 1) [27]. Phosphorylation of IκBα leads to its polyubiquitination and proteasome-dependent degradation, which allows bound NF-κB dimers to translocate to the nucleus and activate gene expression (Figure 1). In contrast to the canonical NF-κB pathway, the non-canonical NF-κB pathway is activated by TNF receptor superfamily members, including BAFFR, CD40, lymphotoxin β receptor and RANK. The non-canonical NF-κB activation requires kinases NIK and IKKα that mediate p100 phosphorylation, ubiquitination, and processing to p52, which results in the generation of RelB and p52 dimers.
Chronic activation of NF-κB leads to the development of various autoimmune, inflammatory-related disorders and solid tumors, as well as leukemia and lymphoma. The transient transcriptional activity of NF-κB is normally maintained by one of the NF-κB target genes, IκBα, in a negative feedback loop, after stimulation with NF-κB activating receptors. Furthermore, a number of other NF-κB negative regulatory molecules, including A20 and Cyld, are involved in maintaining transient activation of NF-κB to prevent autoimmune, inflammatory related disorders and cancers [28,29,30].

2.2. STAT3

STAT3 is a member of the STAT (Signal Transducers and Activators of Transcription) transcription factor family discovered in 1994 [31,32,33]. The transcriptional activation of STAT3 by phosphorylation at tyrosine 705 (Y705) is regulated by a diverse array of cytokine receptors (e.g., Interleukin (IL)-6, IL-11, IL-23), growth factor receptors (e.g., epidermal growth factor (EGF)), G protein-coupled receptors (GPCRs) (e.g., sphingosine-1-phosphate receptor-1 (S1PR1), Angiotensin II type I receptor (AT1R)), ultraviolet (UV) radiation, oncogenes and stress. Although transient STAT3 phosphorylation and activation is essential for the development of immune cells such as T cells and B cells, cell growth and proliferation, cell migration, and cell death, chronic and hyper-activated STAT3 results in cancer-promoting inflammation, increased cell proliferation, cell survival, angiogenesis and metastases, and dysregulation of anti-tumor immunity [34,35]. Furthermore, activated STAT3 is a critical regulator of cell energy metabolism, which could have a significant impact on tumor transformation and growth [36]. Many of the STAT3-induced genes are cytokines and growth factors, which again bind to their receptors and activate STAT3 thereby maintaining chronic STAT3 activation in many cancers [37].
In an unstimulated cell, STAT3 is in an inactive state in the cytoplasm; however, it is activated by direct phosphorylation at tyrosine 705 and serine 727 residues by the receptor-associated Janus kinases (JAKs), tyrosine kinases, and select non-receptor tyrosine kinases such as EGFR, SRC and AB [34,35,38]. Phosphorylated STAT3 dimerizes or oligomerizes prior to nuclear translocation, DNA binding and induction of its target genes (Figure 1) [35,39]. Transient activation of STAT3 is a critical regulatory mechanism and is mediated by a variety of post-translational modifications of STAT3 and direct interactions with negative regulatory proteins [40]. Biologically active STAT3 dimers are destabilized by several tyrosine phosphatases, including PTPN6, PTPRD, PTPRT and PTPN11 [41]. Inactivation of these phosphatases by either genetic mutations or epigenetic regulation leads to chronic activation of STAT3 phosphorylation in tumor cells [42,43,44,45,46]. Most importantly, STAT3 activation is negatively regulated at the receptor level by STAT3 target genes, suppressors of cytokine signaling (SOCS) E3 ubiquitin ligases, which induce the degradation of cytokine receptor complexes [47]. STAT3 activation is also negatively regulated by protein inhibitor of activated STAT (PIAS), an E3 SUMO-protein ligase in the nucleolus (Figure 1) [48,49].

2.3. Crosstalk between NF-κB and STAT3 in Cancers

Recent studies have found that chronic infection or inflammation mediated by NF-κB and STAT3 plays a pivotal role in linking inflammation and cancer [50,51,52]. The genes induced by both NF-κB and STAT3 are critical regulators of cellular proliferation, angiogenesis, genomic instability, resistance to apoptosis, invasion and metastasis of tumor cells [53]. NF-κB and STAT3 activation are highly connected, and interact functionally with each other in a number of layers. Activation of NF-κB induces several cytokines and growth factors, such as IL-6, which is a major STAT3 activator. STAT3 activation also contributes to NF-κB activation by binding to RelA/p65 in the nucleus and enhancing RelA/p65 reversible acetylation, thereby prolonging its nuclear retention [54,55]. In addition, NF-κB and STAT3 bind together to a subset of gene promoters to cooperatively induce expression of their target genes [56].

3. Role of Cell Surface Molecules (CSMs) in EBV, KSHV or HTLV-1-Induced Inflammation during De Novo Infection

Most enveloped viruses, including EBV, KSHV and HTLV-1, rely on an optimal amount of certain cell surface molecules (CSMs) expressed on host cells for attachment, cell entry and spread. Inflammation induced by NF-κB and STAT3 upon attachment of KSHV or EBV envelope proteins to CSMs is necessary for their entry into target cells, for the establishment of latency and for the viral life cycle. However, it is unclear if HTLV-1 attachment to CSMs induces NF-κB and STAT3 activation and inflammation. In addition, the mechanisms and host CSMs involved in promoting the entry and inflammation of these viruses are not well understood.

3.1. CSMs in EBV-Induced Inflammation during De Novo Infection

EBV envelope glycoprotein 350 (gp350) binds to the cell surface receptor CD21 (type II complement receptor or CR2), which forms membrane complexes with CD35/CR1 and CD19, expressed on many human cell types such as B and T-lymphocytes, thymocytes, epithelial cells, follicular dendritic cells and endothelial cells. This membrane complex initiates EBV entry into the target cells [57,58,59]. At the same time, binding of EBV envelope gp350 to these receptor complexes rapidly induces activation of IKK and NF-κB (Figure 2) [60,61,62,63]. This activation of IKK and NF-κB is primarily mediated by phosphatidylinositol-3-kinase (PI3K) and protein kinase C (PKC) during EBV de novo infection. Similarly, STAT3 phosphorylation at Y705 and activation by EBV de novo infection has been reported [64,65]. Although STAT3 activation by EBV de novo infection is mediated through JAKs, the mechanism and CSMs involved in activating JAKs upon EBV de novo infection are not well understood. The activation of NF-κB and STAT3 is critical for the cell survival, proliferation, and adhesion of EBV infected cells, as well as immortalization [65,66,67].

3.2. CSMs in KSHV-Induced Inflammation during De Novo Infection

Like EBV, KSHV envelope glycoprotein gB, K8.1, and the complement control protein (KCP) promote attachment to cell surface associated heparan sulfate (HS), integrins (α3β1, αVβ3, and αVβ5) and EphA2 receptor tyrosine kinase (EphA2R), which initiates KSHV DNA entry into target cells [68]. Following attachment of KSHV glycoproteins to cell surface receptors, key signaling molecules including Focal Adhesion Kinase (FAK), non-receptor cytoplasmic tyrosine kinase Src, and PI3K are activated, which results in the activation of NF-κB [69]. Binding of KSHV to its receptors has been shown to activate NF-κB by a mechanism involving IKKβ and IKKε (Figure 2) [70]. Our recent findings suggest that cell surface expressed Cell adhesion molecule 1 (CADM1) is critical for KSHV de novo infection-induced NF-κB activation [71]. Although CADM1 and IKKβ and IKKε kinases are critical for NF-κB activation upon KSHV de novo infection, the mechanisms of CADM1 and IKKβ and IKKε activation are unclear. In addition, if CADM1 is required for IKKβ and IKKε early activation during KSHV de novo infection is unknown. It has been shown that KSHV de novo infection rapidly induces STAT3 phosphorylation at Y705 [65]. Activation of STAT3 is mediated by JAK2 upon KSHV de novo infection (Figure 2). In addition to activation of JAK2 and STAT3 by KSHV de novo infection, the JAK2 and STAT3 activating cell surface receptor gp130 is induced by KSHV infection. This upregulation of receptor gp130 maintains constitutive phosphorylation of JAK2/STAT3 in KSHV-infected cells (Figure 2). KSHV de novo infection-induced NF-κB and STAT3 activation ensures cell growth, cell survival and KSHV latency.

3.3. CSMs in HTLV-1-Induced Inflammation during De Novo Infection

Unlike EBV and KSHV, cell free HTLV-1 virus has extremely low infectivity [72]. HTLV-1 infection occurs through cell-to-cell contact between infected donor cells and uninfected target cells, in which viral particles are transmitted via viral biofilms, cellular conduits or viral synapse formation with essential contributions from cellular surface expressing lymphocyte function-associated antigen-1 (LFA1) and its ligand intracellular adhesion receptor 1 (ICAM-1) [73,74,75,76,77]. Upon binding of HTLV-1 to its cell surface receptors, glucose transporter 1 (Glut-1), neuropilin-1 (NRP-1, BDCA-4), and heparan sulfate proteoglycans (HSPGs), the viral RNA is delivered into the cytoplasm, reverse transcribed, and integrated into the host genome to form a provirus. Although the main target cells of HTLV-1 in vivo are CD4+ T-cells, HTLV-1 can infect a wide variety of cells in vitro, including CD8+ T-cells, endothelial cells, B-lymphocytes, myeloid cells and fibroblasts, possibly due to ubiquitous expression of the receptor Glut-1 [78,79]. Although it is not well understood if NF-κB and STAT3 are activated during HTLV-1 de novo infection, we speculate that NF-κB and STAT3 are activated, as NF-κB and STAT3-regulated inflammatory molecules, including IFN-γ, IL-1, TNF-α, IL-6 and CXCL10, are produced during HTLV-1 de novo infection [80]. These inflammatory molecules produced during HTLV-1 de novo infection play critical roles in the immortalization, survival and proliferation of infected cells. Furthermore, many NF-κB and STAT3 target genes induced during HTLV-1 de novo infection are critical for HTLV-1-mediated dysregulation of host type I Interferon antiviral signaling [81,82].

4. Role of CSMs in the Induction of Inflammation during EBV, KSHV, and HTLV-1 Latency

Humans that have been infected with EBV, KSHV or HTLV-1 remain infected for life due to viral latency in B cells and T cells. Upon entry into the target cells of these viruses, the viral latent proteins, such as homologue of FADD-like interleukin (IL)-1β-converting enzyme (FLICE/caspase-8) inhibitory protein (vFLIP), latency-associated nuclear antigen (LANA), Viral interleukin-6 (vIL-6) of KSHV, EBV-encoded nuclear antigens (EBNA), latent membrane proteins (LMPs) of EBV, and the trans-activator protein, Tax, and HTLV-1 basic leucine zipper factor (HBZ) of HTLV-1 are expressed. Expression of these viral oncogenes maintains chronic activation of both NF-κB and STAT3, and inflammation required for the immortalization and transformation of EBV, KSHV and HTLV-1-infected cells [65,83].

4.1. CSMs and Chronic Inflammation during EBV Latency

EBV infection-induced CSMs such as CD40 and CD137 expression on EBV-infected T or NK cells play critical roles in the constitutive activation of NF-κB and survival of infected cells [84,85,86]. Studies have shown that chronic activation of STAT3 is thought to be triggered by IL-6, which is induced by persistent activation of NF-κB [87,88]. Activation of NF-κB and STAT3 following B lymphocyte infection with EBV in vitro leads to the induction of chronic inflammation, which is critical for the continuous proliferation of infected B cells and the establishment of lymphoblastoid cell lines (LCLs) [89,90]. During this latency, known as latency type III, a total of nine genes are expressed, including EBV-encoded RNAs (EBER 1 and 2), EBV nuclear antigens (EBNAs 1, 2, 3A, 3B, 3C, and LP) and EBV latent membrane proteins (LMPs 1, 2A, and 2B). These genes play a key role in cell proliferation, migration and survival [12,91]. During latency II, EBERs, EBNA-1, LMP1 and LMP2 are expressed and this is typically observed in EBV-associated Hodgkin’s lymphoma, NPC and T/NK cell lymphomas [92,93,94]. During latency I, only EBERs and EBNA-1 are expressed and this occurs in Burkitt’s lymphoma [95,96,97]. In addition to the latent infection, the lytic form of EBV infection also occurs occasionally to produce new virus particles. During EBV lytic infection BILF1, a viral G protein–coupled receptor (vGPCR) is expressed, which downregulates cell surface HLA class I expression and inhibits T cell recognition of EBV-infected cells [98,99,100].
Both LMP1 and BILF1 viral proteins are expressed on the cell surface and are persistently activated [66]. In addition to STAT3 activation by LMP1 via NF-κB-IL-6, LMP1 can directly induce activation of STAT3 via JAK3 in an EBV-negative NPC line (Figure 2) [65,101]. LMP1 also causes STAT3 phosphorylation at S727 through Protein kinase C-δ (PKCδ) and Extracellular signal-regulated kinase (ERK) in epithelial cells [102,103].

4.2. CSMs and Chronic Inflammation during KSHV Latency

Several KSHV oncogenes, including vFLIP, vIL6, vGPCR, K15 and K1, expressed after successful KSHV de novo infection are critical for KSHV oncogenesis. While vGPCR, K15 and K1 are induced during KSHV lytic replication, they are constantly expressed at a low level during KSHV latency [104,105,106]. vGPCR, K15 and K1 are membrane proteins which are partly exposed to the cell surface and chronically activate NF-κB [106,107,108]. While vFLIP is mostly localized in the cytosolic compartment of PEL cells, it initiates NF-κB activation in membrane lipid rafts of PEL cells. Our recent findings suggest that Cell Adhesion Molecule 1 (CADM1), which is highly induced during KSHV de novo infection, is essential for vFLIP to maintain chronic NF-κB activation in PEL cells [71]. We also found that vGPCR, which is a close homolog to mammalian chemokine receptors CXCR1 and CXCR2, required CADM1 to activate NF-κB (Figure 2) [71]. A recent report suggests the possible role of CADM1 in the regulation of STAT3 activation in squamous cell carcinoma progression [109]. vGPCR also chronically activates STAT3 phosphorylation at Y705 [110]. However, the mechanism of STAT3 activation by vGPCR is poorly understood. In addition, whether CADM1 is required for vGPCR to activate STAT3 is not known. Studies have shown that chronic activation of STAT3 by KSHV vIL6 is critical for the survival of PEL cells. The survival and proliferation of PEL cells were significantly diminished after knocking down CADM1 [111,112]. It is possible that CADM1 may also be required for vIL6-mediated STAT3 activation in PEL cells.

4.3. CSMs and Chronic Inflammation during HTLV-1 Latency

The two HTLV-1 oncogenes, Tax and HBZ play important roles in maintaining chronic inflammation to promote HTLV-1 pathogenesis [113,114]. Tax expression during the early stages of HTLV-1 latent infection is crucial for chronic NF-κB activation and induction of inflammation that is necessary for the development of HTLV-1-associated diseases. When Tax is conjugated by K63-linked polyubiquitination in membrane lipid rafts, it mediates the persistent activation of NF-κB by triggering chronic phosphorylation and activation of the IKK kinase complex [115]. This activation of IKK leads to the phosphorylation and proteasomal degradation of IκBα. Recent findings suggest that Tax interacts with CADM1 in membrane lipid rafts and initiates IKK kinase complex activation. Tax also interacts with the CSM Scribble, a prominent regulator of T-cell polarity, to mediate activation of NF-κB in T-cells [116]. Tax also mediates STAT3 activation by inducing interleukin-6 receptor (IL-6R) expression in HTLV-1-infected cells [117]. When Tax expression is lost in about 60% of ATLL patients HBZ protein and RNA promote the growth and survival of the leukemic cells [118,119]. While HBZ inhibits the classical NF-κB pathway probably to facilitate escape from host immune responses and avoid senescence [120], it maintains chronic inflammation in mice similar to those in HTLV-1-infected individuals [121]. HBZ interacts with STAT3 and modulates the IL-10/JAK/STAT signaling pathway to promote T cell proliferation [122]. Although CADM1 is one of the most highly expressed molecules in ATLL cells, it is not clear whether it is essential for HBZ-induced inflammation in ATLL cells.

5. Role of CSMs in the Negative Regulation of NF-κB and STAT3 Inhibition

Mounting evidence has revealed that transient induction of inflammation by NF-κB and STAT3 in response to injury or infection is essential to activate immune responses. Chronic inflammation mediated via NF-κB and STAT3 is inhibited by NF-κB negative regulatory proteins (A20 and Cyld) and STAT3 negative regulatory (SOCSs) proteins to prevent chronic inflammation-induced autoimmune diseases and cancers and to restore homeostasis and tissue repair. Most of these NF-κB and STAT3 negative regulators are induced by NF-κB and STAT3 themselves and function in negative feedback loops. A20 and Cyld are deubiquitinating enzymes that inhibit the ubiquitination and activation of key IKK kinase complex activators, including TRAFs and RIP1, in receptor-mediated NF-κB activation [123,124,125]. The DUB activity of A20 is dispensable for the inhibition of IKK and NF-κB activation as it can inhibit NF-κB activation in a non-catalytic manner via zinc-finger 4 (ZF4) and ZF7 [126,127]. In addition, A20 disrupts E2:E3 ubiquitin enzyme complexes independently of DUB activity to inhibit NF-κB signaling [128]. Interestingly, it is not well understood how these molecules are activated in response to NF-κB and STAT3 activation. In addition, it is unclear if the CSMs that are involved in the activation of NF-κB and STAT3 initiate the activation of NF-κB and STAT3 negative regulators. Recently, we and others have shown that the HTLV-1 Tax protein targets NF-κB negative regulatory complexes to maintain chronic activation of NF-κB [29,125,128,129,130]. Interestingly, the CSM CADM1 is critical for Tax to target NF-κB negative regulatory complexes to maintain chronic activation of NF-κB in HTLV-1-infected cells [115] (Figure 2). CADM1 is also critical for the KSHV oncogenes vGPCR and vFLIP to maintain chronic activation of NF-κB in KSHV-infected PEL cells. Thus, it is possible that CADM1 may also inhibit NF-κB negative regulatory complexes. Because CADM1 is highly upregulated in both HTLV-1 and KSHV-infected cells, where STAT3 is chronically activated, it is likely that CADM1 may also be involved in directly or indirectly inhibiting STAT3 negative regulatory mechanisms. It would not be surprising if CADM1 is also induced by EBV infection and involved in chronic activation of both NF-κB and STAT3 by either targeting NF-κB and STAT3 inhibitory mechanisms or directly activating NF-κB and STAT3 signaling.

6. Conclusions

A large number of CSMs are involved in maintaining cell–cell communication and conveying various signals from the extracellular environment to inside the cell nucleus in multi-cellular organisms. While conveying extracellular signals, they also recognize various pathogens present in the extracellular environment and transmit danger signals. However, certain pathogenic viruses such as EBV, KSHV and HTLV-1 have evolved multiple strategies to dysregulate the normal function of CSMs to benefit viral replication or persistence. In this review, we have highlighted select CSMs that are involved in facilitating the entry of EBV, KSHV and HTLV-1 into their target cells, NF-κB and STAT3 activation, and inflammation essential for EBV, KSHV and HTLV-1 pathogenesis. Chronic inflammation mediated by CSMs during de novo infection by these pathogens creates a favorable environment for the initiation of latency and subsequent diseases. Although studies have shown that acute induction of inflammation by NF-κB and STAT3 in response to infection eliminates or limits pathogen spread, chronic inflammation driven by NF-κB and STAT3 activation create favorable environments for EBV, KSHV and HTLV-1. Oncogenes expressed during EBV, KSHV or HTLV-1 latency further dysregulate the function of NF-κB and STAT3 negative regulators to maintain chronic activation of NF-κB and STAT3. Most of these viral oncogenes hijack host CSMs to dysregulate NF-κB and STAT3 negative regulators in lymphotropic virus-infected cells. Therefore, research into the precise mechanisms of CSMs-mediated chronic inflammation in EBV, KSHV and HTLV-1 infections may result in new therapies to target the cancers caused by these viruses.

Author Contributions

Conceptualization, N.S. and E.W.H.; writing—original draft preparation, N.S.; writing—review and editing, N.S. and E.W.H.; funding acquisition, N.S. All authors have read and agreed to the published version of the manuscript.

Funding

N.S. was funded by the American Cancer Society Grant RSG-16-254-01-MPC.

Acknowledgments

The authors apologize for not citing all relevant papers due to space considerations.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Parkin, D.M. The global health burden of infection-associated cancers in the year 2002. Int. J. Cancer 2006, 118, 3030–3044. [Google Scholar] [CrossRef] [Green Version]
  2. Coussens, L. Session 2: Inflammation and Cancer. Toxicol. Pathol. 2004, 32, 732. [Google Scholar] [CrossRef]
  3. Jha, H.C.; Banerjee, S.; Robertson, E.S. The Role of Gammaherpesviruses in Cancer Pathogenesis. Pathogens 2016, 5, 18. [Google Scholar] [CrossRef] [Green Version]
  4. Baer, R.; Bankier, A.T.; Biggin, M.D.; Deininger, P.L.; Farrell, P.J.; Gibson, T.J.; Hatfull, G.; Hudson, G.S.; Satchwell, S.C.; Séguin, C.; et al. DNA sequence and expression of the B95-8 Epstein—Barr virus genome. Nat. Cell Biol. 1984, 310, 207–211. [Google Scholar] [CrossRef]
  5. Russo, J.J.; Bohenzky, R.A.; Chien, M.-C.; Chen, J.; Yan, M.; Maddalena, D.; Parry, J.P.; Peruzzi, D.; Edelman, I.S.; Chang, Y.; et al. Nucleotide sequence of the Kaposi sarcoma-associated herpesvirus (HHV8]. Proc. Natl. Acad. Sci. USA 1996, 93, 14862–14867. [Google Scholar] [CrossRef] [Green Version]
  6. Vandamme, A.-M. The simian origins of the pathogenic human T-cell lymphotropic virus type I. Trends Microbiol. 1998, 6, 477–483. [Google Scholar] [CrossRef]
  7. Carbone, A.; Gloghini, A.; Dotti, G. EBV-Associated Lymphoproliferative Disorders: Classification and Treatment. Oncology 2008, 13, 577–585. [Google Scholar] [CrossRef]
  8. Thorley-Lawson, D.A. Epstein-Barr virus: Exploiting the immune system. Nat. Rev. Immunol. 2001, 1, 75–82. [Google Scholar] [CrossRef]
  9. Hoeger, B.; Serwas, N.; Boztug, K. Human NF-κB1 Haploinsufficiency and Epstein–Barr Virus-Induced Disease—Molecular Mechanisms and Consequences. Front. Immunol. 2018, 8, 1978. [Google Scholar] [CrossRef] [Green Version]
  10. Epstein, M.A.; Achong, B.G.; Barr, Y.M. Virus Particles in Cultured Lymphoblasts from Burkitt’s Lymphoma. Lancet 1964, 1, 702–703. [Google Scholar] [CrossRef]
  11. Velapasamy, S.; Dawson, C.W.; Young, L.S.; Paterson, I.C.; Yap, L.F. The Dynamic Roles of TGF-beta Signalling in EBV-Associated Cancers. Cancers 2018, 10, 247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Young, L.S.; Yap, L.F.; Murray, P.G. Epstein–Barr virus: More than 50 years old and still providing surprises. Nat. Rev. Cancer 2016, 16, 789–802. [Google Scholar] [CrossRef] [PubMed]
  13. Chang, Y.; Cesarman, E.; Pessin, M.S.; Lee, F.; Culpepper, J.; Knowles, D.M.A.; Moore, P.S. Identification of herpesvirus-like DNA sequences in AIDS-associated Kaposi’s sarcoma. Science 1994, 266, 1865–1869. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Cesarman, E.; Chang, Y.; Moore, P.S.; Said, J.W.; Knowles, D.M. Kaposi’s Sarcoma–Associated Herpesvirus-Like DNA Sequences in AIDS-Related Body-Cavity–Based Lymphomas. N. Engl. J. Med. 1995, 332, 1186–1191. [Google Scholar] [CrossRef]
  15. Soulier, J.; Grollet, L.; Oksenhendler, E.; Cacoub, P.; Cazals-Hatem, D.; Babinet, P.; d’Agay, M.F.; Clauvel, J.P.; Raphael, M.; Degos, L.; et al. Kaposi’s sarcoma-associated herpesvirus-like DNA sequences in multicentric Castleman’s disease. Blood 1995, 86, 1276–1280. [Google Scholar] [CrossRef] [Green Version]
  16. Yoshida, M.; Miyoshi, I.; Hinuma, Y. Isolation and characterization of retrovirus from cell lines of human adult T-cell leukemia and its implication in the disease. Proc. Natl. Acad. Sci. USA 1982, 79, 2031–2035. [Google Scholar] [CrossRef] [Green Version]
  17. Uchiyama, T.; Yodoi, J.; Sagawa, K.; Takatsuki, K.; Uchino, H. Adult T-cell leukemia: Clinical and hematologic features of 16 cases. Blood 1997, 50, 481–492. [Google Scholar] [CrossRef] [Green Version]
  18. Gessain, A.; Vernant, J.; Maurs, L.; Barin, F.; Gout, O.; Calender, A.; De Thé, G. Antibodies to human T-lymphotropic virus type-I in patients with tropical spastic paraparesis. Lancet 1985, 326, 407–410. [Google Scholar] [CrossRef]
  19. Morrison, B.J.; Labo, N.; Miley, W.J.; Whitby, D. Serodiagnosis for Tumor Viruses. Semin. Oncol. 2015, 42, 191–206. [Google Scholar] [CrossRef] [Green Version]
  20. Myoung, J.; Ganem, D. Active lytic infection of human primary tonsillar B cells by KSHV and its noncytolytic control by activated CD4+ T cells. J. Clin. Investig. 2011, 121, 1130–1140. [Google Scholar] [CrossRef] [Green Version]
  21. Pique, C.; Jones, K.S. Pathways of cell-cell transmission of HTLV-1. Front. Microbiol. 2012, 3, 378. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Keller, S.A.; Hernandez-Hopkins, D.; Vider, J.; Ponomarev, V.; Hyjek, E.; Schattner, E.J.; Cesarman, E. NF-κB is essential for the progression of KSHV- and EBV-infected lymphomas in vivo. Blood 2006, 107, 3295–3302. [Google Scholar] [CrossRef] [PubMed]
  23. Read, S.A.; Douglas, M.W. Virus induced inflammation and cancer development. Cancer Lett. 2014, 345, 174–181. [Google Scholar] [CrossRef] [PubMed]
  24. Allen, I.C.; Wilson, J.E.; Schneider, M.; Lich, J.D.; Roberts, R.A.; Arthur, J.C.; Woodford, R.M.T.; Davis, B.K.; Uronis, J.M.; Herfarth, H.H.; et al. NLRP12 suppresses colon inflammation and tumorigenesis through the negative regulation of noncanonical NF-kappaB signaling. Immunity 2012, 36, 742–754. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Dejardin, E. The alternative NF-κB pathway from biochemistry to biology: Pitfalls and promises for future drug development. Biochem. Pharmacol. 2006, 72, 1161–1179. [Google Scholar] [CrossRef]
  26. Sun, S.C. Non-canonical NF-kappaB signaling pathway. Cell Res. 2011, 21, 71–85. [Google Scholar] [CrossRef] [Green Version]
  27. Hayden, M.S.; Ghosh, S. NF- B, the first quarter-century: Remarkable progress and outstanding questions. Genes Dev. 2012, 26, 203–234. [Google Scholar] [CrossRef] [Green Version]
  28. Lork, M.; Verhelst, K.; Beyaert, R. CYLD, A20 and OTULIN deubiquitinases in NF-kappaB signaling and cell death: So similar, yet so different. Cell Death Differ. 2017, 24, 1172–1183. [Google Scholar] [CrossRef]
  29. Pujari, R.; Hunte, R.; Khan, W.N.; Shembade, N. A20-mediated negative regulation of canonical NF-kappaB signaling pathway. Immunol. Res. 2013, 57, 166–171. [Google Scholar] [CrossRef]
  30. Shembade, N.; Harhaj, E.W. Regulation of NF-kappaB signaling by the A20 deubiquitinase. Cell. Mol. Immunol. 2012, 9, 123–130. [Google Scholar] [CrossRef] [Green Version]
  31. Lütticken, C.; Wegenka, U.; Yuan, J.; Buschmann, J.; Schindler, C.; Ziemiecki, A.; Harpur, A.G.; Wilks, A.F.; Yasukawa, K.; Taga, T.; et al. Association of transcription factor APRF and protein kinase Jak1 with the interleukin-6 signal transducer gp130. Science 1994, 263, 89–92. [Google Scholar] [CrossRef] [PubMed]
  32. Standke, G.J.; Meier, V.S.; Groner, B. Mammary gland factor activated by prolactin on mammary epithelial cells and acute-phase response factor activated by interleukin-6 in liver cells share DNA binding and transactivation potential. Mol Endocrinol. 1994, 8, 469–477. [Google Scholar] [PubMed] [Green Version]
  33. Zhong, Z.; Wen, Z.; Darnell, J.E. Stat3: A STAT family member activated by tyrosine phosphorylation in response to epidermal growth factor and interleukin-6. Science 1994, 264, 95–98. [Google Scholar] [CrossRef] [PubMed]
  34. Yu, H.; Pardoll, D.M.; Jove, R. STATs in cancer inflammation and immunity: A leading role for STAT3. Nat. Rev. Cancer 2009, 9, 798–809. [Google Scholar] [CrossRef] [PubMed]
  35. Zou, S.; Tong, Q.; Liu, B.; Huang, W.; Tian, Y.; Fu, X. Targeting STAT3 in Cancer Immunotherapy. Mol. Cancer 2020, 19, 1–19. [Google Scholar] [CrossRef] [PubMed]
  36. Poli, V.; Camporeale, A. STAT3-Mediated Metabolic Reprograming in Cellular Transformation and Implications for Drug Resistance. Front. Oncol. 2015, 5, 121. [Google Scholar] [CrossRef] [Green Version]
  37. Zhang, H.-F.; Lai, R. STAT3 in Cancer—Friend or Foe? Cancers 2014, 6, 1408–1440. [Google Scholar] [CrossRef] [Green Version]
  38. Prasad, S.; Pandey, M.K.; Yadav, V.R.; Aggarwal, B.B. Gambogic acid inhibits STAT3 phosphorylation through activation of protein tyrosine phosphatase SHP-1: Potential role in proliferation and apoptosis. Cancer Prev. Res. 2011, 4, 1084–1094. [Google Scholar] [CrossRef] [Green Version]
  39. Sgrignani, J.; Garofalo, M.; Matkovic, M.; Merulla, J.; Catapano, C.V.; Cavalli, A. Structural Biology of STAT3 and Its Implications for Anticancer Therapies Development. Int. J. Mol. Sci. 2018, 19, 1591. [Google Scholar] [CrossRef] [Green Version]
  40. Yu, H.; Kortylewski, M.; Pardoll, D.M. Crosstalk between cancer and immune cells: Role of STAT3 in the tumour microenvironment. Nat. Rev. Immunol. 2007, 7, 41–51. [Google Scholar] [CrossRef]
  41. Kim, M.; Morales, L.D.; Jang, I.; Cho, Y.; Kim, D.J. Protein Tyrosine Phosphatases as Potential Regulators of STAT3 Signaling. Int. J. Mol. Sci. 2018, 19, 2708. [Google Scholar] [CrossRef] [Green Version]
  42. Kim, D.J.; Tremblay, M.L.; DiGiovanni, J. Protein Tyrosine Phosphatases, TC-PTP, SHP1, and SHP2, Cooperate in Rapid Dephosphorylation of Stat3 in Keratinocytes Following UVB Irradiation. PLoS ONE 2010, 5, e10290. [Google Scholar] [CrossRef] [Green Version]
  43. Peyser, N.D.; Du, Y.; Li, H.; Lui, V.W.Y.; Xiao, X.; Chan, T.A.; Grandis, J.R. Loss-of-Function PTPRD Mutations Lead to Increased STAT3 Activation and Sensitivity to STAT3 Inhibition in Head and Neck Cancer. PLoS ONE 2015, 10, e0135750. [Google Scholar] [CrossRef] [Green Version]
  44. Tartaglia, M.; Niemeyer, C.M.; Fragale, A.; Song, X.; Buechner, J.; Jung, A.; Hählen, K.; Hasle, H.; Licht, J.D.; Gelb, B.D. Somatic mutations in PTPN11 in juvenile myelomonocytic leukemia, myelodysplastic syndromes and acute myeloid leukemia. Nat. Genet. 2003, 34, 148–150. [Google Scholar] [CrossRef]
  45. Peyser, N.D.; Freilino, M.; Wang, L.; Zeng, Y.; Li, H.; Johnson, D.E.; Grandis, J.R. Frequent promoter hypermethylation of PTPRT increases STAT3 activation and sensitivity to STAT3 inhibition in head and neck cancer. Oncogene 2015, 35, 1163–1169. [Google Scholar] [CrossRef] [Green Version]
  46. Lui, V.W.Y.; Peyser, N.D.; Ng, P.K.-S.; Hritz, J.; Zeng, Y.; Lu, Y.; Li, H.; Wang, L.; Gilbert, B.R.; General, I.J.; et al. Frequent mutation of receptor protein tyrosine phosphatases provides a mechanism for STAT3 hyperactivation in head and neck cancer. Proc. Natl. Acad. Sci. USA 2014, 111, 1114–1119. [Google Scholar] [CrossRef] [Green Version]
  47. Babon, J.J.; Varghese, L.N.; Nicola, N.A. Inhibition of IL-6 family cytokines by SOCS3. Semin. Immunol. 2014, 26, 13–19. [Google Scholar] [CrossRef] [Green Version]
  48. Saydmohammed, M.; Joseph, D.; Syed, V. Curcumin suppresses constitutive activation of STAT-3 by up-regulating protein inhibitor of activated STAT-3 (PIAS-3] in ovarian and endometrial cancer cells. J. Cell. Biochem. 2010, 110, 447–456. [Google Scholar] [CrossRef]
  49. Chung, C.D.; Liao, J.; Liu, B.; Rao, X.; Jay, P.; Berta, P.; Shuai, K. Specific Inhibition of Stat3 Signal Transduction by PIAS3. Science 1997, 278, 1803–1805. [Google Scholar] [CrossRef]
  50. Mittal, D.; Gubin, M.M.; Schreiber, R.D.; Smyth, M.J. New insights into cancer immunoediting and its three component phases—Elimination, equilibrium and escape. Curr. Opin. Immunol. 2014, 27, 16–25. [Google Scholar] [CrossRef] [Green Version]
  51. Ji, Z.; He, L.; Regev, A.; Struhl, K. Inflammatory regulatory network mediated by the joint action of NF-kB, STAT3, and AP-1 factors is involved in many human cancers. Proc. Natl. Acad. Sci. USA 2019, 116, 9453–9462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. De Martel, C.; Franceschi, S. Infections and cancer: Established associations and new hypotheses. Crit. Rev. Oncol. 2009, 70, 183–194. [Google Scholar] [CrossRef] [PubMed]
  53. Fan, Y.; Mao, R.; Yang, J. NF-kappaB and STAT3 signaling pathways collaboratively link inflammation to cancer. Protein Cell 2013, 4, 176–185. [Google Scholar] [CrossRef] [Green Version]
  54. Lee, H.; Herrmann, A.; Deng, J.H.; Kujawski, M.; Niu, G.; Li, Z.; Forman, S.; Jove, R.; Pardoll, D.M.; Yu, H. Persistently activated Stat3 maintains constitutive NF-kappaB activity in tumors. Cancer Cell 2009, 15, 283–293. [Google Scholar] [CrossRef] [Green Version]
  55. Chen, L.-F.; Fischle, W.; Verdin, E.; Greene, W.C. Duration of Nuclear NF-kappa B Action Regulated by Reversible Acetylation. Science 2001, 293, 1653–1657. [Google Scholar] [CrossRef] [Green Version]
  56. Yang, J.; Liao, X.; Agarwal, M.K.; Barnes, L.; Auron, P.E.; Stark, G.R. Unphosphorylated STAT3 accumulates in response to IL-6 and activates transcription by binding to NFkappaB. Genes Dev. 2007, 21, 1396–1408. [Google Scholar] [CrossRef] [Green Version]
  57. D’Addario, M.; Libermann, T.A.; Xu, J.; Ahmad, A.; Menezes, J. Epstein-Barr virus and its glycoprotein-350 upregulate IL-6 in human B-lymphocytes via CD21, involving activation of NF-κB and different signaling pathways. J. Mol. Biol. 2001, 308, 501–514. [Google Scholar] [CrossRef]
  58. Fingeroth, J.D.; Weis, J.J.; Tedder, T.F.; Strominger, J.L.; Biro, P.A.; Fearon, D.T. Epstein-Barr virus receptor of human B lymphocytes is the C3d receptor CR2. Proc. Natl. Acad. Sci. USA 1984, 81, 4510–4514. [Google Scholar] [CrossRef] [Green Version]
  59. D’Addario, M.; Ahmad, A.; Morgan, A.; Menezes, J. Binding of the Epstein-Barr virus major envelope glycoprotein gp350 results in the upregulation of the TNF-alpha gene expression in monocytic cells via NF-kappaB involving PKC, PI3-K and tyrosine kinases. J. Mol. Biol. 2000, 298, 765–778. [Google Scholar] [CrossRef]
  60. Bouillie, S.; Barel, M.; Frade, R. Signaling through the EBV/C3d receptor (CR2, CD21] in human B lymphocytes: Activation of phosphatidylinositol 3-kinase via a CD19-independent pathway. J. Immunol. 1999, 162, 136–143. [Google Scholar]
  61. Arredouani, M.S.; Bhasin, M.K.; Sage, D.R.; Dunn, L.K.; Gill, M.B.; Agnani, D.; Libermann, T.A.; Fingeroth, J.D. Analysis of Host Gene Expression Changes Reveals Distinct Roles for the Cytoplasmic Domain of the Epstein-Barr Virus Receptor/CD21 in B-Cell Maturation, Activation, and Initiation of Virus Infection. J. Virol. 2014, 88, 5559–5577. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. D’Addario, M.; Ahmad, A.; Xu, J.W.; Menezes, J. Epstein-Barr virus envelope glycoprotein gp350 induces NF-kappaB activation and IL-1beta synthesis in human monocytes-macrophages involving PKC and PI3-K. FASEB J. 1999, 13, 2203–2213. [Google Scholar]
  63. Sun, S.C.; Cesarman, E. NF-kappaB as a target for oncogenic viruses. Curr. Top. Microbial. Immunol. 2011, 349, 197–244. [Google Scholar]
  64. Koganti, S.; De La Paz, A.; Freeman, A.F.; Bhaduri-McIntosh, S. B Lymphocytes from Patients with a Hypomorphic Mutation in STAT3 Resist Epstein-Barr Virus-Driven Cell Proliferation. J. Virol. 2013, 88, 516–524. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Li, X.; Bhaduri-McIntosh, S. A Central Role for STAT3 in Gammaherpesvirus-Life Cycle and -Diseases. Front. Microbiol. 2016, 7, 1052. [Google Scholar] [CrossRef]
  66. De Oliveira, D.E.; Ballon, G.; Cesarman, E. NF-κB signaling modulation by EBV and KSHV. Trends Microbiol. 2010, 18, 248–257. [Google Scholar] [CrossRef]
  67. Takada, H.; Imadome, K.I.; Shibayama, H.; Yoshimori, M.; Wang, L.; Saitoh, Y.; Uota, S.; Yamaoka, S.; Koyama, T.; Shimizu, N.; et al. EBV induces persistent NF-kappaB activation and contributes to survival of EBV-positive neoplastic T- or NK-cells. PLoS ONE 2017, 12, e0174136. [Google Scholar]
  68. Kumar, B.; Chandran, B. KSHV Entry and Trafficking in Target Cells—Hijacking of Cell Signal Pathways, Actin and Membrane Dynamics. Viruses 2016, 8, 305. [Google Scholar] [CrossRef]
  69. Sadagopan, S.; Sharma-Walia, N.; Veettil, M.V.; Raghu, H.; Sivakumar, R.; Bottero, V.; Chandran, B. Kaposi’s sarcoma-associated herpesvirus induces sustained NF-kappaB activation during de novo infection of primary human dermal microvascular endothelial cells that is essential for viral gene expression. J. Virol. 2007, 81, 3949–3968. [Google Scholar] [CrossRef] [Green Version]
  70. He, Z.; Zhao, J.; Zhang, J.; Jung, J.U.; Feng, P. NF-kappaB activation coordinated by IKKbeta and IKKepsilon enables latent infection of Kaposi’s sarcoma-associated herpesvirus. J. Virol. 2014, 88, 444–455. [Google Scholar] [CrossRef] [Green Version]
  71. Hunte, R.; Alonso, P.; Thomas, R.; Bazile, C.A.; Ramos, J.C.; van der Weyden, L.; Dominguez-Bendala, J.; Khan, W.N.; Shembade, N. CADM1 is essential for KSHV-encoded vGPCR-and vFLIP-mediated chronic NF-kappaB activation. PLoS Pathog. 2018, 14, e1006968. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Dutartre, H.; Clavière, M.; Journo, C.; Mahieux, R. Cell-Free versus Cell-to-Cell Infection by Human Immunodeficiency Virus Type 1 and Human T-Lymphotropic Virus Type 1: Exploring the Link among Viral Source, Viral Trafficking, and Viral Replication. J. Virol. 2016, 90, 7607–7617. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Pasquier, A.; Alais, S.; Roux, L.; Thoulouze, M.-I.; Alvarez, K.; Journo, C.; Dutartre, H.; Mahieux, R. How to Control HTLV-1-Associated Diseases: Preventing de Novo Cellular Infection Using Antiviral Therapy. Front. Microbiol. 2018, 9, 278. [Google Scholar] [CrossRef] [PubMed]
  74. Igakura, T.; Stinchcombe, J.C.; Goon, P.K.C.; Taylor, G.P.; Weber, J.N.; Griffiths, G.M.; Tanaka, Y.; Osame, M.; Bangham, C.R.M. Spread of HTLV-I Between Lymphocytes by Virus-Induced Polarization of the Cytoskeleton. Science 2003, 299, 1713–1716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Thoulouze, M.-I.; Alcover, A. Can viruses form biofilms? Trends Microbiol. 2011, 19, 257–262. [Google Scholar] [CrossRef] [PubMed]
  76. Van Prooyen, N.; Gold, H.; Andresen, V.; Schwartz, O.; Jones, K.; Ruscetti, F.; Lockett, S.; Gudla, P.; Venzon, D.; Franchini, G. Human T-cell leukemia virus type 1 p8 protein increases cellular conduits and virus transmission. Proc. Natl. Acad. Sci. USA 2010, 107, 20738–20743. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Pais-Correia, A.-M.; Sachse, M.; Guadagnini, S.; Robbiati, V.; Lasserre, R.; Gessain, A.; Gout, O.; Alcover, A.; Thoulouze, M.-I. Biofilm-like extracellular viral assemblies mediate HTLV-1 cell-to-cell transmission at virological synapses. Nat. Med. 2009, 16, 83–89. [Google Scholar] [CrossRef] [PubMed]
  78. Longo, D.L.; Gelmann, E.P.; Cossman, J.; Young, R.A.; Gallo, R.C.; O’Brien, S.; Matis, L.A. Isolation of HTLV-transformed B-lymphocyte clone from a patient with HTLV-associated adult T-cell leukaemia. Nat. Cell Biol. 1984, 310, 505–506. [Google Scholar] [CrossRef]
  79. Koyanagi, Y.; Itoyama, Y.; Nakamura, N.; Takamatsu, K.; Kira, J.-I.; Iwamasa, T.; Goto, I.; Yamamoto, N. In Vivo Infection of Human T-Cell Leukemia Virus Type I in Non-T Cells. Virology 1993, 196, 25–33. [Google Scholar] [CrossRef]
  80. Futsch, N.; Prates, G.; Mahieux, R.; Casseb, J.; Dutartre, H. Cytokine Networks Dysregulation during HTLV-1 Infection and Associated Diseases. Viruses 2018, 10, 691. [Google Scholar] [CrossRef] [Green Version]
  81. Olière, S.; Hernandez, E.; Lézin, A.; Arguello, M.; Douville, R.; Nguyên, T.L.-A.; Olindo, S.; Panelatti, G.; Kazanji, M.; Wilkinson, P.; et al. HTLV-1 Evades Type I Interferon Antiviral Signaling by Inducing the Suppressor of Cytokine Signaling 1 (SOCS1]. PLoS Pathog. 2010, 6, e1001177. [Google Scholar] [CrossRef] [PubMed]
  82. Charoenthongtrakul, S.; Zhou, Q.; Shembade, N.; Harhaj, N.S.; Harhaj, E.W. Human T cell leukemia virus type 1 Tax inhibits innate antiviral signaling via NF-kappaB-dependent induction of SOCS1. J. Virol. 2011, 85, 6955–6962. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Chen, L.; Liu, D.; Zhang, Y.; Zhang, H.; Cheng, H. The autophagy molecule Beclin 1 maintains persistent activity of NF-kappaB and Stat3 in HTLV-1-transformed T lymphocytes. Biochem. Biophys. Res. Commun. 2015, 465, 739–745. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Arai, A. Chronic active Epstein–Barr virus infection: A bi-faceted disease with inflammatory and neoplastic elements. Immunol. Med. 2018, 41, 162–169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Imadome, K.-I.; Shimizu, N.; Arai, A.; Miura, O.; Watanabe, K.; Nakamura, H.; Nonoyama, S.; Yamamoto, K.; Fujiwara, S. Coexpression of CD40 and CD40 Ligand in Epstein-Barr Virus–Infected T and NK Cells and Their Role in Cell Survival. J. Infect. Dis. 2005, 192, 1340–1348. [Google Scholar] [CrossRef] [PubMed]
  86. Yoshimori, M.; Imadome, K.-I.; Komatsu, H.; Wang, L.; Saitoh, Y.; Yamaoka, S.; Fukuda, T.; Kurata, M.; Koyama, T.; Shimizu, N.; et al. CD137 Expression Is Induced by Epstein-Barr Virus Infection through LMP1 in T or NK Cells and Mediates Survival Promoting Signals. PLoS ONE 2014, 9, e112564. [Google Scholar] [CrossRef]
  87. Chen, H.; Hutt-Fletcher, L.; Cao, L.; Hayward, S.D. A Positive Autoregulatory Loop of LMP1 Expression and STAT Activation in Epithelial Cells Latently Infected with Epstein-Barr Virus. J. Virol. 2003, 77, 4139–4148. [Google Scholar] [CrossRef] [Green Version]
  88. Onozawa, E.; Shibayama, H.; Takada, H.; Imadome, K.-I.; Aoki, S.; Yoshimori, M.; Shimizu, N.; Fujiwara, S.; Koyama, T.; Miura, O.; et al. STAT3 is constitutively activated in chronic active Epstein-Barr virus infection and can be a therapeutic target. Oncotarget 2018, 9, 31077–31089. [Google Scholar] [CrossRef]
  89. Omi, N.; Tokuda, Y.; Ikeda, Y.; Ueno, M.; Mori, K.; Sotozono, C.; Kinoshita, S.; Nakano, M.; Tashiro, K. Efficient and reliable establishment of lymphoblastoid cell lines by Epstein-Barr virus transformation from a limited amount of peripheral blood. Sci. Rep. 2017, 7, 43833. [Google Scholar] [CrossRef] [Green Version]
  90. Babcock, G.J.; Decker, L.L.; Volk, M.; Thorley-Lawson, D.A. EBV Persistence in Memory B Cells In Vivo. Immunity 1998, 9, 395–404. [Google Scholar] [CrossRef] [Green Version]
  91. Brink, A.A.; Dukers, D.F.; Brule, A.J.V.D.; Oudejans, J.J.; Middeldorp, J.M.; Meijer, C.J.; Jiwa, M. Presence of Epstein-Barr virus latency type III at the single cell level in post-transplantation lymphoproliferative disorders and AIDS related lymphomas. J. Clin. Pathol. 1997, 50, 911–918. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Roughan, J.E.; Thorley-Lawson, D.A. The Intersection of Epstein-Barr Virus with the Germinal Center. J. Virol. 2009, 83, 3968–3976. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Pallesen, G.; Hamilton-Dutoit, S.J.; Rowe, M.; Young, L. Expression of Epstein-Barr virus latent gene products in tumour cells of Hodgkin’s disease. Lancet 1991, 337, 320–322. [Google Scholar] [CrossRef]
  94. Brooks, L.; Yao, Q.Y.; Rickinson, A.B.; Young, L.S. Epstein-Barr virus latent gene transcription in nasopharyngeal carcinoma cells: Coexpression of EBNA1, LMP1, and LMP2 transcripts. J. Virol. 1992, 66, 2689–2697. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Rowe, M.; Rowe, D.T.; Gregory, C.D.; Young, L.S.; Farrell, P.J.; Rupani, H.; Rickinson, A.B. Differences in B cell growth phenotype reflect novel patterns of Epstein-Barr virus latent gene expression in Burkitt’s lymphoma cells. EMBO J. 1987, 6, 2743–2751. [Google Scholar] [CrossRef]
  96. Thorley-Lawson, D.A. EBV Persistence—Introducing the Virus. Curr. Top. Microbiol. Immunol. 2015, 390, 151–209. [Google Scholar] [CrossRef] [Green Version]
  97. Sugiura, M.; Imai, S.; Tokunaga, M.; Koizumi, S.; Uchizawa, M.; Okamoto, K.; Osato, T. Transcriptional analysis of Epstein-Barr virus gene expression in EBV-positive gastric carcinoma: Unique viral latency in the tumour cells. Br. J. Cancer 1996, 74, 625–631. [Google Scholar] [CrossRef]
  98. Zuo, J.; Currin, A.; Griffin, B.D.; Shannon-Lowe, C.; Thomas, W.A.; Ressing, M.E.; Wiertz, E.J.H.J.; Rowe, M. The Epstein-Barr Virus G-Protein-Coupled Receptor Contributes to Immune Evasion by Targeting MHC Class I Molecules for Degradation. PLoS Pathog. 2009, 5, e1000255. [Google Scholar] [CrossRef] [Green Version]
  99. Zuo, J.; Quinn, L.L.; Tamblyn, J.; Thomas, W.A.; Feederle, R.; Delecluse, H.-J.; Hislop, A.D.; Rowe, M. The Epstein-Barr Virus-Encoded BILF1 Protein Modulates Immune Recognition of Endogenously Processed Antigen by Targeting Major Histocompatibility Complex Class I Molecules Trafficking on both the Exocytic and Endocytic Pathways. J. Virol. 2010, 85, 1604–1614. [Google Scholar] [CrossRef] [Green Version]
  100. Griffin, B.D.; Gram, A.M.; Mulder, A.; Van Leeuwen, D.; Claas, F.H.J.; Wang, F.; Ressing, M.E.; Wiertz, E. EBV BILF1 Evolved To Downregulate Cell Surface Display of a Wide Range of HLA Class I Molecules through Their Cytoplasmic Tail. J. Immunol. 2013, 190, 1672–1684. [Google Scholar] [CrossRef] [Green Version]
  101. Liu, Y.-P.; Tan, Y.-N.; Wang, Z.-L.; Zeng, L.; Lu, Z.-X.; Li, L.-L.; Luo, W.; Tang, M.; Cao, Y. Phosphorylation and nuclear translocation of STAT3 regulated by the Epstein-Barr virus latent membrane protein 1 in nasopharyngeal carcinoma. Int. J. Mol. Med. 2008, 21, 153–162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Kung, C.-P.; Meckes, D.G.; Raab-Traub, N. Epstein-Barr Virus LMP1 Activates EGFR, STAT3, and ERK through Effects on PKC. J. Virol. 2011, 85, 4399–4408. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Suarez, A.A.R.; Van Renne, N.; Baumert, T.F.; Lupberger, J. Viral manipulation of STAT3: Evade, exploit, and injure. PLoS Pathog. 2018, 14, e1006839. [Google Scholar] [CrossRef] [Green Version]
  104. Sharp, T.V.; Wang, H.-W.; Koumi, A.; Hollyman, D.; Endo, Y.; Ye, H.; Du, M.-Q.; Boshoff, C. K15 Protein of Kaposi’s Sarcoma-Associated Herpesvirus Is Latently Expressed and Binds to HAX-1, a Protein with Antiapoptotic Function. J. Virol. 2002, 76, 802–816. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Chandriani, S.; Ganem, D. Array-based transcript profiling and limiting-dilution reverse transcription-PCR analysis identify additional latent genes in Kaposi’s sarcoma-associated herpesvirus. J. Virol. 2010, 84, 5565–5573. [Google Scholar] [CrossRef] [Green Version]
  106. Azzi, S.; Smith, S.S.; Dwyer, J.; Leclair, H.M.; Alexia, C.; Hebda, J.K.; Dupin, N.; Bidere, N.; Gavard, J. YGLF motif in the Kaposi sarcoma herpes virus G-protein-coupled receptor adjusts NF-kappaB activation and paracrine actions. Oncogene 2014, 33, 5609–5618. [Google Scholar] [CrossRef] [Green Version]
  107. Prakash, O.; Tang, Z.-Y.; Peng, X.; Coleman, R.; Gill, J.; Farr, G.; Samaniego, F. Tumorigenesis and Aberrant Signaling in Transgenic Mice Expressing the Human Herpesvirus-8 K1 Gene. J. Natl. Cancer Inst. 2002, 94, 926–935. [Google Scholar] [CrossRef] [Green Version]
  108. Brinkmann, M.M.; Glenn, M.; Rainbow, L.; Kieser, A.; Henke-Gendo, C.; Chulz, T.F. Activation of mitogen-activated protein kinase and NF-kappaB pathways by a Kaposi’s sarcoma-associated herpesvirus K15 membrane protein. J. Virol. 2003, 77, 9346–9358. [Google Scholar] [CrossRef] [Green Version]
  109. Vallath, S.; Sage, E.K.; Kolluri, K.K.; Lourenco, S.N.; Teixeira, V.S.; Chimalapati, S.; George, P.J.; Janes, S.M.; Giangreco, A. CADM1 inhibits squamous cell carcinoma progression by reducing STAT3 activity. Sci. Rep. 2016, 6, 24006. [Google Scholar] [CrossRef] [Green Version]
  110. Burger, M.; Hartmann, T.; Burger, J.A.; Schraufstatter, I.U. KSHV-GPCR and CXCR2 transforming capacity and angiogenic responses are mediated through a JAK2-STAT3-dependent pathway. Oncogene 2005, 24, 2067–2075. [Google Scholar] [CrossRef] [Green Version]
  111. Wu, J.; Xu, Y.; Mo, D.; Huang, P.; Sun, R.; Huang, L.; Pan, S.; Xu, J. Kaposi’s sarcoma-associated herpesvirus (KSHV) vIL-6 promotes cell proliferation and migration by upregulating DNMT1 via STAT3 activation. PLoS ONE 2014, 9, e93478. [Google Scholar] [CrossRef] [PubMed]
  112. Chen, D.; Gao, Y.; Nicholas, J. Human Herpesvirus 8 Interleukin-6 Contributes to Primary Effusion Lymphoma Cell Viability via Suppression of Proapoptotic Cathepsin D, a Cointeraction Partner of Vitamin K Epoxide Reductase Complex Subunit 1 Variant 2. J. Virol. 2013, 88, 1025–1038. [Google Scholar] [CrossRef] [Green Version]
  113. Mohanty, S.; Harhaj, E.W. Mechanisms of Oncogenesis by HTLV-1 Tax. Pathogens 2020, 9, 543. [Google Scholar] [CrossRef]
  114. Shembade, N.; Harhaj, E.W. Role of post-translational modifications of HTLV-1 Tax in NF-kappaB activation. World J. Boil. Chem. 2010, 1, 13–20. [Google Scholar] [CrossRef]
  115. Pujari, R.; Hunte, R.; Thomas, R.; van der Weyden, L.; Rauch, D.; Ratner, L.; Nyborg, J.K.; Ramos, J.C.; Takai, Y.; Shembade, N. Human T-cell leukemia virus type 1 (HTLV-1] tax requires CADM1/TSLC1 for inactivation of the NF-kappaB inhibitor A20 and constitutive NF-kappaB signaling. PLoS Pathog. 2015, 11, e1004721. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Arpin-André, C.; Mesnard, J.-M. The PDZ Domain-binding Motif of the Human T Cell Leukemia Virus Type 1 Tax Protein Induces Mislocalization of the Tumor Suppressor hScrib in T cells. J. Biol. Chem. 2007, 282, 33132–33141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Horiuchi, S.; Yamamoto, N.; Dewan, Z.; Takahashi, Y.; Yamashita, A.; Yoshida, T.; Nowell, M.A.; Richards, P.; Jones, S.A.; Yamamoto, N. Human T-cell leukemia virus type-I Tax induces expression of interleukin-6 receptor (IL-6R): Shedding of soluble IL-6R and activation of STAT3 signaling. Int. J. Cancer 2006, 119, 823–830. [Google Scholar] [CrossRef]
  118. Satou, Y.; Yasunaga, J.-I.; Yoshida, M.; Matsuoka, M. HTLV-I basic leucine zipper factor gene mRNA supports proliferation of adult T cell leukemia cells. Proc. Natl. Acad. Sci. USA 2006, 103, 720–725. [Google Scholar] [CrossRef] [Green Version]
  119. Usui, T.; Yanagihara, K.; Tsukasaki, K.; Murata, K.; Hasegawa, H.; Yamada, Y.; Kamihira, S. Characteristic expression of HTLV-1 basic zipper factor (HBZ) transcripts in HTLV-1 provirus-positive cells. Retrovirology 2008, 5, 34. [Google Scholar] [CrossRef] [Green Version]
  120. Zhao, T.; Matsuoka, M. HBZ and its roles in HTLV-1 oncogenesis. Front. Microbiol. 2012, 3, 247. [Google Scholar] [CrossRef] [Green Version]
  121. Satou, Y.; Yasunaga, J.-I.; Zhao, T.; Yoshida, M.; Miyazato, P.; Takai, K.; Shimizu, K.; Ohshima, K.; Green, P.L.; Ohkura, N.; et al. HTLV-1 bZIP Factor Induces T-Cell Lymphoma and Systemic Inflammation In Vivo. PLoS Pathog. 2011, 7, e1001274. [Google Scholar] [CrossRef] [PubMed]
  122. Higuchi, Y.; Yasunaga, J.-I.; Mitagami, Y.; Tsukamoto, H.; Nakashima, K.; Ohshima, K.; Matsuoka, M. HTLV-1 induces T cell malignancy and inflammation by viral antisense factor-mediated modulation of the cytokine signaling. Proc. Natl. Acad. Sci. USA 2020, 117, 13740–13749. [Google Scholar] [CrossRef] [PubMed]
  123. Sun, S.C. CYLD: A tumor suppressor deubiquitinase regulating NF-kappaB activation and diverse biological processes. Cell Death Differ. 2010, 17, 25–34. [Google Scholar] [CrossRef] [PubMed]
  124. Shembade, N.; Harhaj, N.S.; Liebl, D.J.; Harhaj, E.W. Essential role for TAX1BP1 in the termination of TNF-alpha-, IL-1- and LPS-mediated NF-kappaB and JNK signaling. EMBO J. 2007, 26, 3910–3922. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Shembade, N.; Harhaj, N.S.; Parvatiyar, K.; Copeland, N.G.; Jenkins, N.A.; Matesic, L.E.; Harhaj, E.W. The E3 ligase Itch negatively regulates inflammatory signaling pathways by controlling the function of the ubiquitin-editing enzyme A20. Nat. Immunol. 2008, 9, 254–262. [Google Scholar] [CrossRef] [PubMed]
  126. Razani, B.; Whang, M.I.; Kim, F.S.; Nakamura, M.C.; Sun, X.; Advincula, R.; Turnbaugh, J.A.; Pendse, M.; Tanbun, P.; Achacoso, P.; et al. Non-catalytic ubiquitin binding by A20 prevents psoriatic arthritis–like disease and inflammation. Nat. Immunol. 2020, 21, 422–433. [Google Scholar] [CrossRef]
  127. Skaug, B.; Chen, J.; Du, F.; He, J.; Ma, A.; Chen, Z.J. Direct, Noncatalytic Mechanism of IKK Inhibition by A20. Mol. Cell 2011, 44, 559–571. [Google Scholar] [CrossRef] [Green Version]
  128. Shembade, N.; Ma, A.; Harhaj, E.W. Inhibition of NF-kappaB signaling by A20 through disruption of ubiquitin enzyme complexes. Science 2010, 327, 1135–1139. [Google Scholar] [CrossRef] [Green Version]
  129. Wu, X.-F.; Zhang, M.; Sun, S.-C. Mutual regulation between deubiquitinase CYLD and retroviral oncoprotein Tax. Cell Biosci. 2011, 1, 27. [Google Scholar] [CrossRef] [Green Version]
  130. Shembade, N.; Harhaj, E.W. A20 inhibition of NFκB and inflammation: Targeting E2:E3 ubiquitin enzyme complexes. Cell Cycle 2010, 9, 2481–2482. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Activation and negative regulation of NF-κB and STAT3. (A) Activation of NF-κB in response to the stimulation of TNFR1, IL-1R, or TLRs. A20 is induced by NF-κB in the TNFR1, IL-1R, or TLRs pathways, functioning as a negative feedback loop. (B) Activation and negative regulation of the STAT3 signaling pathway. The activation of STAT3 is mediated in response to cell surface receptor (growth factor or cytokine receptors) stimulation that leads to the recruitment and activation of the JAK family of kinases, which, in turn, phosphorylate STAT3. Phosphorylated STAT3 dimerize and translocate to the nucleus, where they directly regulate gene expression. This activation loop can be turned off by PIAS and SOCS proteins.
Figure 1. Activation and negative regulation of NF-κB and STAT3. (A) Activation of NF-κB in response to the stimulation of TNFR1, IL-1R, or TLRs. A20 is induced by NF-κB in the TNFR1, IL-1R, or TLRs pathways, functioning as a negative feedback loop. (B) Activation and negative regulation of the STAT3 signaling pathway. The activation of STAT3 is mediated in response to cell surface receptor (growth factor or cytokine receptors) stimulation that leads to the recruitment and activation of the JAK family of kinases, which, in turn, phosphorylate STAT3. Phosphorylated STAT3 dimerize and translocate to the nucleus, where they directly regulate gene expression. This activation loop can be turned off by PIAS and SOCS proteins.
Biology 09 00390 g001
Figure 2. Cell surface molecules-induced STAT3 and NF-κB activation during the early and late phases of infection by lymphotropic viruses. (A) Upon viral (Epstein Barr virus (EBV) or Κaposi’s sarcoma herpesvirus (KSHV)) attachment to cell surface molecules, STAT3 and NF-κB activation, viral entry into target cells, and viral latency establishment occurs. Rapid inflammation induced by STAT3 and NF-κB activation helps to establish viral latency. (B) During viral latency (viral late phase) the viral latent proteins and/or oncogenes, vIL6 and vFLIP of KSHV, LMP1 and BLIF1 of EBV, and Tax and HBZ of human T-cell leukemia virus type 1 (HTLV-1), are expressed. Viral proteins interact with cell surface molecules and maintain chronic activation of STAT3 and NF-κB, essential for EBV, KSHV and HTLV-1 oncogenesis. Viral oncogenes may hijack cell CSMs to inhibit the function of A20 and CylD proteins in NF-κB, and SOCS proteins in STAT3 pathways to maintain chronic activation of NF-κB and STAT3 in lymphotropic virus-infected cells.
Figure 2. Cell surface molecules-induced STAT3 and NF-κB activation during the early and late phases of infection by lymphotropic viruses. (A) Upon viral (Epstein Barr virus (EBV) or Κaposi’s sarcoma herpesvirus (KSHV)) attachment to cell surface molecules, STAT3 and NF-κB activation, viral entry into target cells, and viral latency establishment occurs. Rapid inflammation induced by STAT3 and NF-κB activation helps to establish viral latency. (B) During viral latency (viral late phase) the viral latent proteins and/or oncogenes, vIL6 and vFLIP of KSHV, LMP1 and BLIF1 of EBV, and Tax and HBZ of human T-cell leukemia virus type 1 (HTLV-1), are expressed. Viral proteins interact with cell surface molecules and maintain chronic activation of STAT3 and NF-κB, essential for EBV, KSHV and HTLV-1 oncogenesis. Viral oncogenes may hijack cell CSMs to inhibit the function of A20 and CylD proteins in NF-κB, and SOCS proteins in STAT3 pathways to maintain chronic activation of NF-κB and STAT3 in lymphotropic virus-infected cells.
Biology 09 00390 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Harhaj, E.W.; Shembade, N. Lymphotropic Viruses: Chronic Inflammation and Induction of Cancers. Biology 2020, 9, 390. https://doi.org/10.3390/biology9110390

AMA Style

Harhaj EW, Shembade N. Lymphotropic Viruses: Chronic Inflammation and Induction of Cancers. Biology. 2020; 9(11):390. https://doi.org/10.3390/biology9110390

Chicago/Turabian Style

Harhaj, Edward W., and Noula Shembade. 2020. "Lymphotropic Viruses: Chronic Inflammation and Induction of Cancers" Biology 9, no. 11: 390. https://doi.org/10.3390/biology9110390

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