Next Article in Journal
Targeted Therapy in Mesotheliomas: Uphill All the Way
Previous Article in Journal
Quantitative Bone SPECT/CT of Central Cartilaginous Bone Tumors: Relationship between SUVmax and Radiodensity in Hounsfield Unit
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Perspective

Cellular Transformation by Human Cytomegalovirus

by
Georges Herbein
1,2
1
Department Pathogens & Inflammation-EPILAB EA4266, University of Franche-Comté (UFC), 25000 Besançon, France
2
Department of Virology, CHU Besançon, 25000 Besançon, France
Cancers 2024, 16(11), 1970; https://doi.org/10.3390/cancers16111970
Submission received: 16 March 2024 / Revised: 13 May 2024 / Accepted: 17 May 2024 / Published: 22 May 2024
(This article belongs to the Section Infectious Agents and Cancer)

Abstract

:

Simple Summary

Discovering new oncoviruses is a main goal of virology research. In addition to its deleterious role in immunocompromised patients and during pregnancy leading to birth defects, the human cytomegalovirus’s (HCMV) potential role as an oncogenic agent has garnered significant attention recently. This perspective article focuses on the transforming potential of HCMV based on recently unveiled molecular and cellular characteristics of HCMV-infected cells.

Abstract

Epstein–Barr virus (EBV), Kaposi sarcoma human virus (KSHV), human papillomavirus (HPV), hepatitis B and C viruses (HBV, HCV), human T-lymphotropic virus-1 (HTLV-1), and Merkel cell polyomavirus (MCPyV) are the seven human oncoviruses reported so far. While traditionally viewed as a benign virus causing mild symptoms in healthy individuals, human cytomegalovirus (HCMV) has been recently implicated in the pathogenesis of various cancers, spanning a wide range of tissue types and malignancies. This perspective article defines the biological criteria that characterize the oncogenic role of HCMV and based on new findings underlines a critical role for HCMV in cellular transformation and modeling the tumor microenvironment as already reported for the other human oncoviruses.

1. Introduction

Human cytomegalovirus (HCMV) (also named HHV-5) belongs to the herpesvirus family, is an enveloped double-stranded DNA virus, and is ubiquitous with no seasonal variations [1]. Although usually asymptomatic in healthy individuals, HCMV infection can result in severe CMV disease in immunocompromised subjects and lead to congenital infections with severe neurological sequelae including sensorineural hearing loss in the pediatric population [2]. Recently the role of HCMV in cancer has been re-evaluated.
Several years ago a first paradigm named oncomodulation was forwarded to explain the role of HCMV in cancer where the virus will accelerate the transformation process in infected tumor cells. Although the high prevalence of HCMV in several tumors has been reported [3], it is difficult to know whether the presence of HCMV is incidental due to the viral infection on top of an already present tumor or if the HCMV by itself can start and favor the cellular transformation, and thereby could be defined as a genuine oncovirus [4]. Since stem cells are permissive to HCMV and the appearance of cancer stem cells (CSCs) is a main player in tumor initiation and tumor spread it’s worth questioning the genuine transformation potential of HCMV.
Epstein–Barr virus (EBV), Kaposi sarcoma human virus (KSHV), human papillomavirus (HPV), hepatitis B and C viruses (HBV, HCV), human T-lymphotropic virus-1 (HTLV-1), and the most recently discovered Merkel cell polyomavirus (MCPyV) belong to the group 1 carcinogens for humans, namely human oncoviruses [5,6]. Most oncoviruses transform cells through viral oncoproteins or activation of cellular oncoproteins. In addition, oncoviruses such as HCV favor neoplasm development mainly by chronic inflammation. The hallmarks of cancer were proposed as a set of functional capabilities acquired by human cells as they make their way from normalcy to neoplastic growth [7], and were twice updated in 2011 and 2022 to include additional biological factors [8]. Human oncoviruses fulfill the hallmarks of cancer [9], and recently HCMV has been recognized to display all the hallmarks of cancer as defined in 2011 [10].
Findings by our group and others indicated that HCMV displays oncogenic properties, in addition to the already reported oncomodulatory effect [3,4,10,11]. We will focus the present perspective article on the similarities between HCMV and the already described seven human oncoviruses, HCMV’s immunosuppressive impact on the tumor microenvironment, and the fulfillment of the recently reported hallmarks of cancer by HCMV-transformed cells.

2. HCMV Displays Oncogenic Traits Similar to Human Oncoviruses

In addition to the already recognized seven human oncoviruses, several points are in favor of a direct oncogenic role for HCMV [3,4,12,13,14,15] (Table 1). The stem cells are permissive to HCMV with Thy-1 and platelet-derived growth factor receptor alpha (PDGFRα) identified as stem cell markers that favor HCMV infection [16,17,18,19]. HCMV could favor oncogenesis through the infection of stem cells, the generation of cancer stem cells, and/or the dedifferentiation of mature cells toward stem cells or progenitor cells. HCMV can alter and impair DNA repair pathways parallel to enhanced cell survival [20,21]. Among the viral proteins, the immediate early protein 1 (IE1) favors stemness and EMT in glioblastoma cells [22,23,24]. We reported recently that the influence of oncoviruses and HCMV on Myc and EZH2 expression could be a key driver of cellular transformation. Oncoviruses and HCMV can modulate the levels of Myc and EZH2, two key oncogenic/stemness players, through various molecular mechanisms including among others cell cycle dysregulation, epigenetic modifications, apoptosis blockade, increased cell proliferation, and the generation of polyploid giant cancer cells (PGCCs) [25,26].
The alternance of lytic and latent phases occurs during the pathogenesis of oncoviruses of the Herpesviridae family, namely EBV and KSHV, with viral reactivation triggered by immune suppression and inflammation [70,71,72]. Similar to EBV and KSHV, in such a moving immune environment, HCMV variants emerge and HCMV fitness could be favored with the distribution of the virus between distinct anatomical compartments [73,74,75,76,77,78,79,80,81,82]. The diversity of HCMV strains could play a role in cellular transformation, although cancer-derived cell lines are not always fully permissive to HCMV replication [83,84,85,86,87]. In addition, very low levels of HCMV replication could be at play in the transformation process [88,89]. In fact, we isolated HCMV clinical strains in our laboratory, namely HCMV-DB and BL, which fully replicate in human mammary epithelial cells (HMECs), ovarian epithelial cells (OECs), prostate epithelial cells (PECs), and astrocytes followed by their transformation with the appearance in cultures of CMV-transformed HMECs (CTH cells), CMV-transformed OECs (CTO cells), CMV-transformed PECs (CTP cells), and CMV-elicited glioblastoma cells (CEGBCs) [60,61,62,63,66,67,68,69]. The HCMV-DB and BL strains were named high-risk oncogenic strains by the author (HR-HCMV) [90]. HR-HCMV strains were latent and/or replicated at low levels in chronically infected transformed cells and were reactivated following 12-O-tetradecanoyl-phorbol-13-acetate (TPA) treatment [62,67], similar to KSHV and EBV reactivation under TPA [91,92]. We believe that the latency phase is an important player in the transforming process of epithelial cells infected with HR-HCMV since in chronically infected transformed cells, we detect very low levels of HCMV with reactivation by MIEP activators such as TPA or HDAC inhibitors. In addition, in these transformed cultures, we observe time-by-time spontaneous transient “viral blips” indicating that transient lytic phases occur among the latently infected cells. Following the infection of epithelial cells and astrocytes by HR-HCMV, cellular dedifferentiation parallel to cancerous traits was present in CTH cells, CTO cells, CTP cells, and CEGBCs and could ultimately lead to the appearance of adenocarcinoma of poor prognosis such as triple-negative breast cancer, high-grade serous ovarian cancer (HGSOC), prostate cancer, and glioblastoma [62,63,64,65,67,69]. In addition, the direct role of HCMV in the appearance of glioblastoma is likely with the detection of the virus in all the glioblastoma biopsies tested in our group [66], similar to more than 99% of HPV DNA detection in cervical carcinoma [93]. Although many groups have confirmed the presence of HCMV in glioblastomas, others could not. We cannot exclude that optimized immunohistochemistry and PCR techniques are required to detect it (reviewed in [15]). Recently our group reported that human astrocytes chronically infected and transformed by oncogenic HR-HCMV strains (HCMV-DB strain and HCMV strains isolated from glioblastoma patients) generated spheroids that resulted in the appearance of glioblastoma-like tumors in xenografted mice [94]. The direct oncogenic role of HCMV following acute infection of primary epithelial cells (HMECs, OECs, and PECs) and human astrocytes is summarized in Table 2.

3. HCMV, like Human Oncoviruses, Favors an Immunosuppressive Tissue Microenvironment

Oncoviruses impact the TME with a chronic inflammatory environment and altered signaling of cell–cell and cell–extracellular matrix adhesion molecules that favor the spread of cancerous cells and metastasis [95]. TME is characterized by the presence of tumor-infiltrating myeloid cells, including tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), tumor-associated dendritic cells (TADCs), tumor-associated neutrophils (TANs), and cancer-associated fibroblasts (CAF). These cells display pro-tumorigenic activities with reactive oxygen species (ROS) production, modulation of inflammation, tumor progression, and angiogenesis; this could be fueled by oncoviruses [96]. Oncoviruses and HCMV along with EZH2 and Myc play a crucial role in shaping an immunosuppressive TME (reviewed in [25]). Similar to oncoviruses, HCMV infection leads to an immune-tolerant environment that will favor tumor appearance, growth, and spread parallel to increased viral fitness [10,97,98,99]. Critical viral players among others are the IE1/2 and pUS28 proteins known for favoring cell survival and sustained cell transformation [4]. HCMV favors tumor cell survival and promotes its own fitness, and HCMV blocks the apoptotic machinery within infected cells through IE2, pUL36, and pUL37 [10]. HCMV decreases viral-specific CD4+ and CD8+ T-cell response and NK activity through several viral proteins (pp65, vIL-10, gpUL40, pUL16, pUS18, and pUS20), which could favor tumor growth [10]. A bidirectional relationship between tumor cells and HCMV could be at play which will curtail viral replication and favor tumor escape with a limited deleterious accumulation of the inflammatory cells at the viral infection site [10,100,101,102]. In addition the production of immune-suppressive cytokines such as cellular IL-10, viral IL-10 and TGF-beta will favor the appearance of TAMs which will further accelerate the tumor spread [103]. HCMV infection profoundly modifies macrophage identity rewiring specific differentiation processes, favoring viral spread and curtailing innate tissue immunity [104]. Interestingly, the first high-risk HCMV strain identified in our group, namely HCMV-DB, is highly macrophage-tropic, favors an M1 to M2/TAM shift upon infection, and leads to the appearance of transformed cells (CTH, CTO, CTP, and CEGBCs) in culture [4,60,105].

4. HCMV Fulfills Previous and Current Hallmarks of Cancer

We previously reported that HCMV infection fulfills all the hallmarks of cancer as defined by Hanahan and Weinberg in 2011 [106] (Figure 1). Recently, Hanahan revisited the hallmarks of cancer and added the four following hallmarks: unlocking phenotypic plasticity, nonmutational epigenetic reprogramming, polymorphic microbiomes, and senescent cells [8].
Dedifferentiation depicts the phenotypic plasticity of our in vitro model with HCMV’s transformation of HMECs, OECs, PECs, and human astrocytes [60,62,66,67,69]. Luminal-to-basal transition upon oncogenic stress activation was described [107], highlighting the epithelial compartment plasticity during tumorigenesis [108], where differentiated luminal epithelial cells can revert into functional basal stem cells in vivo [109]. Further, HMECs’ dedifferentiation into the malignant progenitor-like phenotype can be triggered by the transcription factor special AT-rich binding protein-2 (SATB2) [110]. This conceptualizes that development is a bidirectional process [111] and that somatic cells can gradually dedifferentiate into primitive stages of the developmental hierarchy, where HCMV, in our model, could be a dedifferentiation vector. Similar to the dedifferentiation of mature HMEC infected with HCMV-DB and BL strains, we observed dedifferentiation of mature ovarian epithelial cells and prostate epithelial cells into immature progenitor cells with stemness traits [67,69]. Interestingly both high-risk HCMV-DB and BL strains dedifferentiated primary mature human astrocytes into neural-progenitor-like tumor cells similar to the cells detected in glioblastoma, especially the ones adopting the Lévy-like movement patterns [66,112].
Nonmutational epigenetic reprogramming is present in HCMV-transformed cells parallel to stemness, dedifferentiation, and polyploidy [4,10,113]. HCMV-transformed cells display deregulated p53 and Rb pathways parallel to increased Myc expression [12,114,115]. The enzymatic subunit of polycomb repressive complex 2 (PRC2), EZH2, a histone-lysine N-methyltransferase responsible for transcriptional silencing [116], is increased in CTH, CTO, CTP, and CEGBCs cells [64,66,67,69], as reported in several cancers of poor prognosis [117,118,119,120,121,122,123,124]. All in all, a nonmutational epigenetic reprogramming occurs in HCMV-transformed cells namely through EZH2 upregulation, a downstream target of the Myc oncogene [125,126].
The evidence is increasingly compelling that polymorphic variability in the microbiomes between individuals in a population can have a profound impact on cancer phenotypes [127,128]. The microbial constituents of microbiomes likely will influence the progression of the cancers caused by oncoviruses. In fact, most people infected with oncoviruses will never develop cancer, meaning that other factors including the microbiome could favor transformation. For example, in a study of women with high-risk HPV infections and high cervical cancer rates, significant bacterial and fungal profile changes were associated with cervical squamous intraepithelial lesions and HPV infections [129]. In addition, HCMV similar to most of the human oncoviruses, e.g., HPV, EBV, and KSHV, shows viral strain variability with distinct oncogenic potential that could be further enhanced by the tumor microbiome [130]. In agreement with this observation, our data indicate that the oncogenic potency of HCMV clinical strains varies between low- and high-risk strains [60,62,63,90]. Only the high-risk HCMV strains can trigger the appearance of PGCCs [90]. Recently, the essential role of intestinal microbiota in murine cytomegalovirus reactivation was reported [131], suggesting a potential role for the microbiota in the alternance of latent and lytic viral stages, thereby fueling CMV diversity. Therefore, it will be critical in the future to assess the microbia present in breast, prostate, and ovarian cancers in light of the HCMV strains (especially high-risk) detected in tumors and to decipher the exact role played by the microbiota in HCMV diversity parallel to cellular transformation.
Although the protective effects of senescence in limiting malignant progression have been reported [132,133], more recently, it has become clear that senescent cells stimulate tumor development and malignant progression [133,134]. Senescence favors transformation and tumor growth through the generation of a senescence-associated secretory phenotype (SASP) with the release of cytokines, chemokines and proteases [135,136,137], and transitory and reversible senescent cell states whereby senescent cancer cells can escape senescence and resume cell proliferation and acquire oncogenic traits [138,139]. Polyploid giant cancer cells (PGCCs) have been recently reported as a major player in cancer [140,141]. PGCCs exhibit features of senescent cells [142], questioning the role of senescence in the generation of PGCCs observed in infections with oncoviruses [26]. Although non-oncogenic viral infections (human respiratory syncytial virus, influenza A virus, HIV, measle virus, and dengue virus) favor senescence, most oncogenic viruses (EBV, KSHV, HBV, HCV, and MCPyV) and HCMV have been reported to trigger senescence but also to generate PGCCs [26,143]. Interestingly, HCMV is at the cross-road of senescence and oncogenesis [144], and as for other oncoviruses (e.g., HPV), not all individuals infected with HCMV will develop cancer and non-viral factors (lifestyle and personal immune system) could be also at play. Similar to HPV disease pathophysiology [145], although even healthy individuals might be at risk of HCMV-induced oncogenesis, immunocompromised individuals could develop persistent, treatment-refractory, and progressive HCMV-linked cancers. Following the acute infection of mammary epithelial cells, ovarian epithelial cells, and prostate epithelial cells with high-risk HCMV strains (DB and BL), our team has shown that transformed cells appear in culture including PGCCs [64,68,69] and are tumorigenic in xenografted NSG mice [60], indicating the direct involvement of HCMV in oncogenesis. As we already suggested previously [10], PGCCs should be included in the recently added hallmark of cancer, senescence [8].
All in all, HCMV fulfils the recently described four hallmarks of cancer in transformed cells [8], in addition to the previously reported ten hallmarks of cancer [106] (Figure 1).
Since HCMV fulfills all criteria of oncoviruses, this will ultimately pave the way to new therapeutic approaches including antiviral treatments targeting HCMV oncoprotein(s), immunotherapy, and prophylactic vaccination. To assess these new therapeutic strategies, innovative experimental animal models have to be developed such as the generation of glioblastoma in mice engrafted with HCMV-infected astrocytes [94] and the development of triple-negative breast tumors in NOD/SCID Gamma (NSG) mice engrafted with HCMV-infected mammary epithelial cells [60].

5. Conclusions

Although HCMV was considered as a herpesvirus with no transforming capacity, twenty years ago, the oncomodulation paradigm emerged that could explain the accelerated progression of cancers fueled by HCMV-infected tumor cells. Recently, a direct transforming role of high-risk oncogenic HCMV strains has been observed which could lead to the appearance of aggressive cancers, with poor prognosis. Based on similar oncogenic features compared to the already recognized seven human oncoviruses and the fulfillment of all the hallmarks of cancer, even the most recent ones described in 2022, the definitive classification of HCMV as the eighth human oncovirus has to be taken into account and will lead to new therapeutic approaches that are actively needed to curtail cancers, especially adenocarcinoma and glioblastoma of poor prognosis.

Funding

This work was supported by grants from the University of Franche-Comté (UFC) (CR3300), the Région Franche-Comté (2021-Y-08292 and 2021-Y-08290), and the Ligue contre le Cancer (CR3304) to Georges Herbein.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article.

Conflicts of Interest

The author declares no conflicts of interest.

References

  1. Fulkerson, H.L.; Nogalski, M.T.; Collins-McMillen, D.; Yurochko, A.D. Overview of Human Cytomegalovirus Pathogenesis. Methods Mol. Biol. 2021, 2244, 1–18. [Google Scholar] [CrossRef]
  2. Aldè, M.; Binda, S.; Primache, V.; Pellegrinelli, L.; Pariani, E.; Pregliasco, F.; Di Berardino, F.; Cantarella, G.; Ambrosetti, U. Congenital Cytomegalovirus and Hearing Loss: The State of the Art. JCM 2023, 12, 4465. [Google Scholar] [CrossRef]
  3. Cobbs, C. Cytomegalovirus Is a Tumor-Associated Virus: Armed and Dangerous. Curr. Opin. Virol. 2019, 39, 49–59. [Google Scholar] [CrossRef] [PubMed]
  4. Herbein, G. The Human Cytomegalovirus, from Oncomodulation to Oncogenesis. Viruses 2018, 10, 408. [Google Scholar] [CrossRef]
  5. International Agency for Research on Cancer Weltgesundheitsorganisation (Ed.) IARC Monographs on the Evaluation of Carcinogenic Risks to Humans, Volume 100 B, Biological Agents: This Publication Represents the Views and Expert Opinions of an IARC Working Group on the Evaluation of Carcinogenic Risks to Humans, Which Met in Lyon. 2012; 24. [Google Scholar]
  6. Bouvard, V.; Baan, R.; Straif, K.; Grosse, Y.; Secretan, B.; Ghissassi, F.E.; Benbrahim-Tallaa, L.; Guha, N.; Freeman, C.; Galichet, L.; et al. A Review of Human Carcinogens—Part B: Biological Agents. Lancet Oncol. 2009, 10, 321–322. [Google Scholar] [CrossRef] [PubMed]
  7. Hanahan, D.; Weinberg, R.A. The Hallmarks of Cancer. Cell 2000, 100, 57–70. [Google Scholar] [CrossRef]
  8. Hanahan, D. Hallmarks of Cancer: New Dimensions. Cancer Discov. 2022, 12, 31–46. [Google Scholar] [CrossRef] [PubMed]
  9. Mesri, E.A.; Feitelson, M.A.; Munger, K. Human Viral Oncogenesis: A Cancer Hallmarks Analysis. Cell Host Microbe 2014, 15, 266–282. [Google Scholar] [CrossRef]
  10. Herbein, G. Tumors and Cytomegalovirus: An Intimate Interplay. Viruses 2022, 14, 812. [Google Scholar] [CrossRef]
  11. Söderberg-Nauclér, C. New Mechanistic Insights of the Pathogenicity of High-Risk Cytomegalovirus (CMV) Strains Derived from Breast Cancer: Hope for New Cancer Therapy Options. eBioMedicine 2022, 81, 104103. [Google Scholar] [CrossRef] [PubMed]
  12. Shen, Y.; Zhu, H.; Shenk, T. Human Cytomegalovirus IE1 and IE2 Proteins Are Mutagenic and Mediate “Hit-and-Run” Oncogenic Transformation in Cooperation with the Adenovirus E1A Proteins. Proc. Natl. Acad. Sci. USA 1997, 94, 3341–3345. [Google Scholar] [CrossRef] [PubMed]
  13. Soroceanu, L.; Cobbs, C.S. Is HCMV a Tumor Promoter? Virus Res. 2011, 157, 193–203. [Google Scholar] [CrossRef] [PubMed]
  14. Geisler, J.; Touma, J.; Rahbar, A.; Söderberg-Nauclér, C.; Vetvik, K. A Review of the Potential Role of Human Cytomegalovirus (HCMV) Infections in Breast Cancer Carcinogenesis and Abnormal Immunity. Cancers 2019, 11, 1842. [Google Scholar] [CrossRef] [PubMed]
  15. Peredo-Harvey, I.; Rahbar, A.; Söderberg-Nauclér, C. Presence of the Human Cytomegalovirus in Glioblastomas—A Systematic Review. Cancers 2021, 13, 5051. [Google Scholar] [CrossRef] [PubMed]
  16. Li, Q.; Wilkie, A.R.; Weller, M.; Liu, X.; Cohen, J.I. THY-1 Cell Surface Antigen (CD90) Has an Important Role in the Initial Stage of Human Cytomegalovirus Infection. PLoS Pathog. 2015, 11, e1004999. [Google Scholar] [CrossRef] [PubMed]
  17. Soroceanu, L.; Akhavan, A.; Cobbs, C.S. Platelet-Derived Growth Factor-α Receptor Activation Is Required for Human Cytomegalovirus Infection. Nature 2008, 455, 391–395. [Google Scholar] [CrossRef]
  18. Stegmann, C.; Hochdorfer, D.; Lieber, D.; Subramanian, N.; Stöhr, D.; Laib Sampaio, K.; Sinzger, C. A Derivative of Platelet-Derived Growth Factor Receptor Alpha Binds to the Trimer of Human Cytomegalovirus and Inhibits Entry into Fibroblasts and Endothelial Cells. PLoS Pathog. 2017, 13, e1006273. [Google Scholar] [CrossRef]
  19. Wu, Y.; Prager, A.; Boos, S.; Resch, M.; Brizic, I.; Mach, M.; Wildner, S.; Scrivano, L.; Adler, B. Human Cytomegalovirus Glycoprotein Complex gH/gL/gO Uses PDGFR-α as a Key for Entry. PLoS Pathog. 2017, 13, e1006281. [Google Scholar] [CrossRef]
  20. Li, J.-W.; Yang, D.; Yang, D.; Chen, Z.; Miao, J.; Liu, W.; Wang, X.; Qiu, Z.; Jin, M.; Shen, Z. Tumors Arise from the Excessive Repair of Damaged Stem Cells. Med. Hypotheses 2017, 102, 112–122. [Google Scholar] [CrossRef]
  21. Lilley, C.E.; Schwartz, R.A.; Weitzman, M.D. Using or Abusing: Viruses and the Cellular DNA Damage Response. Trends Microbiol. 2007, 15, 119–126. [Google Scholar] [CrossRef] [PubMed]
  22. Cobbs, C.S.; Soroceanu, L.; Denham, S.; Zhang, W.; Kraus, M.H. Modulation of Oncogenic Phenotype in Human Glioma Cells by Cytomegalovirus IE1–Mediated Mitogenicity. Cancer Res. 2008, 68, 724–730. [Google Scholar] [CrossRef] [PubMed]
  23. Soroceanu, L.; Matlaf, L.; Khan, S.; Akhavan, A.; Singer, E.; Bezrookove, V.; Decker, S.; Ghanny, S.; Hadaczek, P.; Bengtsson, H.; et al. Cytomegalovirus Immediate-Early Proteins Promote Stemness Properties in Glioblastoma. Cancer Res. 2015, 75, 3065–3076. [Google Scholar] [CrossRef] [PubMed]
  24. Fornara, O.; Bartek Jr, J.; Rahbar, A.; Odeberg, J.; Khan, Z.; Peredo, I.; Hamerlik, P.; Bartek, J.; Stragliotto, G.; Landázuri, N.; et al. Cytomegalovirus Infection Induces a Stem Cell Phenotype in Human Primary Glioblastoma Cells: Prognostic Significance and Biological Impact. Cell Death Differ. 2016, 23, 261–269. [Google Scholar] [CrossRef] [PubMed]
  25. El Baba, R.; Herbein, G. EZH2-Myc Hallmark in Oncovirus/Cytomegalovirus Infections and Cytomegalovirus’ Resemblance to Oncoviruses. Cells 2024, 13, 541. [Google Scholar] [CrossRef] [PubMed]
  26. Herbein, G.; Nehme, Z. Polyploid Giant Cancer Cells, a Hallmark of Oncoviruses and a New Therapeutic Challenge. Front. Oncol. 2020, 10, 567116. [Google Scholar] [CrossRef] [PubMed]
  27. Burd, E.M. Human Papillomavirus and Cervical Cancer. Clin. Microbiol. Rev. 2003, 16, 1–17. [Google Scholar] [CrossRef] [PubMed]
  28. Tomaić, V. Functional Roles of E6 and E7 Oncoproteins in HPV-Induced Malignancies at Diverse Anatomical Sites. Cancers 2016, 8, 95. [Google Scholar] [CrossRef] [PubMed]
  29. Zhang, W.; Liu, Y.; Zhao, N.; Chen, H.; Qiao, L.; Zhao, W.; Chen, J.J. Role of Cdk1 in the P53-Independent Abrogation of the Postmitotic Checkpoint by Human Papillomavirus E6. J. Virol. 2015, 89, 2553–2562. [Google Scholar] [CrossRef]
  30. Fan, X.; Liu, Y.; Heilman, S.A.; Chen, J.J. Human Papillomavirus E7 Induces Rereplication in Response to DNA Damage. J. Virol. 2013, 87, 1200–1210. [Google Scholar] [CrossRef]
  31. Pal, A.; Kundu, R. Human Papillomavirus E6 and E7: The Cervical Cancer Hallmarks and Targets for Therapy. Front. Microbiol. 2020, 10, 3116. [Google Scholar] [CrossRef] [PubMed]
  32. Kew, M.C. Hepatitis B Virus x Protein in the Pathogenesis of Hepatitis B Virus-induced Hepatocellular Carcinoma. J. Gastro Hepatol. 2011, 26, 144–152. [Google Scholar] [CrossRef] [PubMed]
  33. Li, T.; Wu, Y.; Tsai, H.; Sun, C.; Wu, Y.; Wu, H.; Pei, Y.; Lu, K.; Yen, T.T.; Chang, C.; et al. Intrahepatic Hepatitis B Virus Large Surface Antigen Induces Hepatocyte Hyperploidy via Failure of Cytokinesis. J. Pathol. 2018, 245, 502–513. [Google Scholar] [CrossRef] [PubMed]
  34. Musa, J.; Li, J.; Grünewald, T.G. Hepatitis B Virus Large Surface Protein Is Priming for Hepatocellular Carcinoma Development via Induction of Cytokinesis Failure. J. Pathol. 2019, 247, 6–8. [Google Scholar] [CrossRef] [PubMed]
  35. Moosavy, S.H.; Dvoodian, P.; Nazarnezhad, M.A.; Nejatizaheh, A.; Ephtekhar, E.; Mahboobi, H. Epidemiology, Transmission, Diagnosis, and Outcome of Hepatitis C Virus Infection. Electron. Physician 2017, 9, 5646–5656. [Google Scholar] [CrossRef] [PubMed]
  36. Machida, K.; Liu, J.-C.; McNamara, G.; Levine, A.; Duan, L.; Lai, M.M.C. Hepatitis C Virus Causes Uncoupling of Mitotic Checkpoint and Chromosomal Polyploidy through the Rb Pathway. J. Virol. 2009, 83, 12590–12600. [Google Scholar] [CrossRef] [PubMed]
  37. Moriya, K.; Fujie, H.; Shintani, Y.; Yotsuyanagi, H.; Tsutsumi, T.; Ishibashi, K.; Matsuura, Y.; Kimura, S.; Miyamura, T.; Koike, K. The Core Protein of Hepatitis C Virus Induces Hepatocellular Carcinoma in Transgenic Mice. Nat. Med. 1998, 4, 1065–1067. [Google Scholar] [CrossRef] [PubMed]
  38. Grassmann, R.; Aboud, M.; Jeang, K.-T. Molecular Mechanisms of Cellular Transformation by HTLV-1 Tax. Oncogene 2005, 24, 5976–5985. [Google Scholar] [CrossRef]
  39. Liang, M.-H.; Geisbert, T.; Yao, Y.; Hinrichs, S.H.; Giam, C.-Z. Human T-Lymphotropic Virus Type 1 Oncoprotein Tax Promotes S-Phase Entry but Blocks Mitosis. J. Virol. 2002, 76, 4022–4033. [Google Scholar] [CrossRef]
  40. Jin, D.-Y.; Spencer, F.; Jeang, K.-T. Human T Cell Leukemia Virus Type 1 Oncoprotein Tax Targets the Human Mitotic Checkpoint Protein MAD1. Cell 1998, 93, 81–91. [Google Scholar] [CrossRef]
  41. Malu, A.; Hutchison, T.; Yapindi, L.; Smith, K.; Nelson, K.; Bergeson, R.; Pope, J.; Romeo, M.; Harrod, C.; Ratner, L.; et al. The Human T-Cell Leukemia Virus Type-1 Tax Oncoprotein Dissociates NF-κB p65RelA-Stathmin Complexes and Causes Catastrophic Mitotic Spindle Damage and Genomic Instability. Virology 2019, 535, 83–101. [Google Scholar] [CrossRef]
  42. Mohanty, S.; Harhaj, E.W. Mechanisms of Oncogenesis by HTLV-1 Tax. Pathogens 2020, 9, 543. [Google Scholar] [CrossRef]
  43. Tsang, S.H.; Wang, R.; Nakamaru-Ogiso, E.; Knight, S.A.B.; Buck, C.B.; You, J. The Oncogenic Small Tumor Antigen of Merkel Cell Polyomavirus Is an Iron-Sulfur Cluster Protein That Enhances Viral DNA Replication. J. Virol. 2016, 90, 1544–1556. [Google Scholar] [CrossRef]
  44. Kwun, H.J.; Wendzicki, J.A.; Shuda, Y.; Moore, P.S.; Chang, Y. Merkel Cell Polyomavirus Small T Antigen Induces Genome Instability by E3 Ubiquitin Ligase Targeting. Oncogene 2017, 36, 6784–6792. [Google Scholar] [CrossRef] [PubMed]
  45. 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]
  46. Yang, J.F.; Liu, W.; You, J. Characterization of Molecular Mechanisms Driving Merkel Cell Polyomavirus Oncogene Transcription and Tumorigenic Potential. PLoS Pathog. 2023, 19, e1011598. [Google Scholar] [CrossRef]
  47. Lee, J.M.; Lee, K.-H.; Farrell, C.J.; Ling, P.D.; Kempkes, B.; Park, J.H.; Hayward, S.D. EBNA2 Is Required for Protection of Latently Epstein-Barr Virus-Infected B Cells against Specific Apoptotic Stimuli. J. Virol. 2004, 78, 12694–12697. [Google Scholar] [CrossRef]
  48. Pan, S.-H.; Tai, C.-C.; Lin, C.-S.; Hsu, W.-B.; Chou, S.-F.; Lai, C.-C.; Chen, J.-Y.; Tien, H.-F.; Lee, F.-Y.; Wang, W.-B. Epstein-Barr Virus Nuclear Antigen 2 Disrupts Mitotic Checkpoint and Causes Chromosomal Instability. Carcinogenesis 2009, 30, 366–375. [Google Scholar] [CrossRef]
  49. Parker, G.A.; Touitou, R.; Allday, M.J. Epstein-Barr Virus EBNA3C Can Disrupt Multiple Cell Cycle Checkpoints and Induce Nuclear Division Divorced from Cytokinesis. Oncogene 2000, 19, 700–709. [Google Scholar] [CrossRef] [PubMed]
  50. Parker, G.A.; Crook, T.; Bain, M.; Sara, E.A.; Farrell, P.J.; Allday, M.J. Epstein-Barr Virus Nuclear Antigen (EBNA)3C Is an Immortalizing Oncoprotein with Similar Properties to Adenovirus E1A and Papillomavirus E7. Oncogene 1996, 13, 2541–2549. [Google Scholar]
  51. Lajoie, V.; Lemieux, B.; Sawan, B.; Lichtensztejn, D.; Lichtensztejn, Z.; Wellinger, R.; Mai, S.; Knecht, H. LMP1 Mediates Multinuclearity through Downregulation of Shelterin Proteins and Formation of Telomeric Aggregates. Blood 2015, 125, 2101–2110. [Google Scholar] [CrossRef] [PubMed]
  52. Kieser, A.; Sterz, K.R. The Latent Membrane Protein 1 (LMP1). Curr. Top. Microbiol. Immunol. 2015, 391, 119–149. [Google Scholar] [CrossRef]
  53. Münz, C. Latency and Lytic Replication in Epstein–Barr Virus-Associated Oncogenesis. Nat. Rev. Microbiol. 2019, 17, 691–700. [Google Scholar] [CrossRef]
  54. Liu, J.; Martin, H.J.; Liao, G.; Hayward, S.D. The Kaposi’s Sarcoma-Associated Herpesvirus LANA Protein Stabilizes and Activates c-Myc. J. Virol. 2007, 81, 10451–10459. [Google Scholar] [CrossRef] [PubMed]
  55. Si, H.; Robertson, E.S. Kaposi’s Sarcoma-Associated Herpesvirus-Encoded Latency-Associated Nuclear Antigen Induces Chromosomal Instability through Inhibition of P53 Function. J. Virol. 2006, 80, 697–709. [Google Scholar] [CrossRef]
  56. Sun, Z.; Xiao, B.; Jha, H.C.; Lu, J.; Banerjee, S.; Robertson, E.S. Kaposi’s Sarcoma-Associated Herpesvirus-Encoded LANA Can Induce Chromosomal Instability through Targeted Degradation of the Mitotic Checkpoint Kinase Bub1. J. Virol. 2014, 88, 7367–7378. [Google Scholar] [CrossRef]
  57. Godden-Kent, D.; Talbot, S.J.; Boshoff, C.; Chang, Y.; Moore, P.; Weiss, R.A.; Mittnacht, S. The Cyclin Encoded by Kaposi’s Sarcoma-Associated Herpesvirus Stimulates Cdk6 to Phosphorylate the Retinoblastoma Protein and Histone H1. J. Virol. 1997, 71, 4193–4198. [Google Scholar] [CrossRef]
  58. Laman, H.; Coverley, D.; Krude, T.; Laskey, R.; Jones, N. Viral Cyclin–Cyclin-Dependent Kinase 6 Complexes Initiate Nuclear DNA Replication. Mol. Cell. Biol. 2001, 21, 624–635. [Google Scholar] [CrossRef]
  59. Li, T.; Gao, S.-J. Metabolic Reprogramming and Metabolic Sensors in KSHV-Induced Cancers and KSHV Infection. Cell Biosci. 2021, 11, 176. [Google Scholar] [CrossRef]
  60. Kumar, A.; Tripathy, M.K.; Pasquereau, S.; Al Moussawi, F.; Abbas, W.; Coquard, L.; Khan, K.A.; Russo, L.; Algros, M.-P.; Valmary-Degano, S.; et al. The Human Cytomegalovirus Strain DB Activates Oncogenic Pathways in Mammary Epithelial Cells. EBioMedicine 2018, 30, 167–183. [Google Scholar] [CrossRef]
  61. Moussawi, F.A.; Kumar, A.; Pasquereau, S.; Tripathy, M.K.; Karam, W.; Diab-Assaf, M.; Herbein, G. The Transcriptome of Human Mammary Epithelial Cells Infected with the HCMV-DB Strain Displays Oncogenic Traits. Sci. Rep. 2018, 8, 12574. [Google Scholar] [CrossRef] [PubMed]
  62. Nehme, Z.; Pasquereau, S.; Haidar Ahmad, S.; Coaquette, A.; Molimard, C.; Monnien, F.; Algros, M.-P.; Adotevi, O.; Diab Assaf, M.; Feugeas, J.-P.; et al. Polyploid Giant Cancer Cells, Stemness and Epithelial-Mesenchymal Plasticity Elicited by Human Cytomegalovirus. Oncogene 2021, 40, 3030–3046. [Google Scholar] [CrossRef] [PubMed]
  63. Haidar Ahmad, S.; Pasquereau, S.; El Baba, R.; Nehme, Z.; Lewandowski, C.; Herbein, G. Distinct Oncogenic Transcriptomes in Human Mammary Epithelial Cells Infected With Cytomegalovirus. Front. Immunol. 2021, 12, 772160. [Google Scholar] [CrossRef] [PubMed]
  64. Nehme, Z.; Pasquereau, S.; Haidar Ahmad, S.; El Baba, R.; Herbein, G. Polyploid Giant Cancer Cells, EZH2 and Myc Upregulation in Mammary Epithelial Cells Infected with High-Risk Human Cytomegalovirus. eBioMedicine 2022, 80, 104056. [Google Scholar] [CrossRef] [PubMed]
  65. Haidar Ahmad, S.; El Baba, R.; Herbein, G. Polyploid Giant Cancer Cells, Cytokines and Cytomegalovirus in Breast Cancer Progression. Cancer Cell Int. 2023, 23, 119. [Google Scholar] [CrossRef] [PubMed]
  66. El Baba, R.; Pasquereau, S.; Haidar Ahmad, S.; Monnien, F.; Abad, M.; Bibeau, F.; Herbein, G. EZH2-Myc Driven Glioblastoma Elicited by Cytomegalovirus Infection of Human Astrocytes. Oncogene 2023, 42, 2031–2045. [Google Scholar] [CrossRef] [PubMed]
  67. El Baba, R.; Haidar Ahmad, S.; Monnien, F.; Mansar, R.; Bibeau, F.; Herbein, G. Polyploidy, EZH2 Upregulation, and Transformation in Cytomegalovirus-Infected Human Ovarian Epithelial Cells. Oncogene 2023, 42, 3047–3061. [Google Scholar] [CrossRef] [PubMed]
  68. El Baba, R.; Pasquereau, S.; Haidar Ahmad, S.; Diab-Assaf, M.; Herbein, G. Oncogenic and Stemness Signatures of the High-Risk HCMV Strains in Breast Cancer Progression. Cancers 2022, 14, 4271. [Google Scholar] [CrossRef]
  69. Bouezzedine, F.; El Baba, R.; Haidar Ahmad, S.; Herbein, G. Polyploid Giant Cancer Cells Generated from Human Cytomegalovirus-Infected Prostate Epithelial Cells. Cancers 2023, 15, 4994. [Google Scholar] [CrossRef]
  70. Müller-Coan, B.G.; Caetano, B.F.R.; Pagano, J.S.; Elgui De Oliveira, D. Cancer Progression Goes Viral: The Role of Oncoviruses in Aggressiveness of Malignancies. Trends Cancer 2018, 4, 485–498. [Google Scholar] [CrossRef]
  71. Compton, T.; Kurt-Jones, E.A.; Boehme, K.W.; Belko, J.; Latz, E.; Golenbock, D.T.; Finberg, R.W. Human Cytomegalovirus Activates Inflammatory Cytokine Responses via CD14 and Toll-Like Receptor 2. J. Virol. 2003, 77, 4588–4596. [Google Scholar] [CrossRef] [PubMed]
  72. Niu, N.; Yao, J.; Bast, R.C.; Sood, A.K.; Liu, J. IL-6 Promotes Drug Resistance through Formation of Polyploid Giant Cancer Cells and Stromal Fibroblast Reprogramming. Oncogenesis 2021, 10, 65. [Google Scholar] [CrossRef] [PubMed]
  73. Jackson, S.E.; Mason, G.M.; Wills, M.R. Human Cytomegalovirus Immunity and Immune Evasion. Virus Res. 2011, 157, 151–160. [Google Scholar] [CrossRef] [PubMed]
  74. Kumar, A.; Herbein, G. Epigenetic Regulation of Human Cytomegalovirus Latency: An Update. Epigenomics 2014, 6, 533–546. [Google Scholar] [CrossRef] [PubMed]
  75. Renzette, N.; Gibson, L.; Bhattacharjee, B.; Fisher, D.; Schleiss, M.R.; Jensen, J.D.; Kowalik, T.F. Rapid Intrahost Evolution of Human Cytomegalovirus Is Shaped by Demography and Positive Selection. PLoS Genet. 2013, 9, e1003735. [Google Scholar] [CrossRef]
  76. Renzette, N.; Bhattacharjee, B.; Jensen, J.D.; Gibson, L.; Kowalik, T.F. Extensive Genome-Wide Variability of Human Cytomegalovirus in Congenitally Infected Infants. PLoS Pathog. 2011, 7, e1001344. [Google Scholar] [CrossRef] [PubMed]
  77. Dhingra, A.; Götting, J.; Varanasi, P.R.; Steinbrueck, L.; Camiolo, S.; Zischke, J.; Heim, A.; Schulz, T.F.; Weissinger, E.M.; Kay-Fedorov, P.C.; et al. Human Cytomegalovirus Multiple-Strain Infections and Viral Population Diversity in Haematopoietic Stem Cell Transplant Recipients Analysed by High-Throughput Sequencing. Med. Microbiol. Immunol. 2021, 210, 291–304. [Google Scholar] [CrossRef] [PubMed]
  78. Götting, J.; Lazar, K.; Suárez, N.M.; Steinbrück, L.; Rabe, T.; Goelz, R.; Schulz, T.F.; Davison, A.J.; Hamprecht, K.; Ganzenmueller, T. Human Cytomegalovirus Genome Diversity in Longitudinally Collected Breast Milk Samples. Front. Cell. Infect. Microbiol. 2021, 11, 664247. [Google Scholar] [CrossRef] [PubMed]
  79. Suárez, N.M.; Wilkie, G.S.; Hage, E.; Camiolo, S.; Holton, M.; Hughes, J.; Maabar, M.; Vattipally, S.B.; Dhingra, A.; Gompels, U.A.; et al. Human Cytomegalovirus Genomes Sequenced Directly From Clinical Material: Variation, Multiple-Strain Infection, Recombination, and Gene Loss. J. Infect. Dis. 2019, 220, 781–791. [Google Scholar] [CrossRef]
  80. Hage, E.; Wilkie, G.S.; Linnenweber-Held, S.; Dhingra, A.; Suárez, N.M.; Schmidt, J.J.; Kay-Fedorov, P.C.; Mischak-Weissinger, E.; Heim, A.; Schwarz, A.; et al. Characterization of Human Cytomegalovirus Genome Diversity in Immunocompromised Hosts by Whole-Genome Sequencing Directly From Clinical Specimens. J. Infect. Dis. 2017, 215, 1673–1683. [Google Scholar] [CrossRef]
  81. Suárez, N.M.; Blyth, E.; Li, K.; Ganzenmueller, T.; Camiolo, S.; Avdic, S.; Withers, B.; Linnenweber-Held, S.; Gwinner, W.; Dhingra, A.; et al. Whole-Genome Approach to Assessing Human Cytomegalovirus Dynamics in Transplant Patients Undergoing Antiviral Therapy. Front. Cell. Infect. Microbiol. 2020, 10, 267. [Google Scholar] [CrossRef] [PubMed]
  82. Görzer, I.; Guelly, C.; Trajanoski, S.; Puchhammer-Stöckl, E. Deep Sequencing Reveals Highly Complex Dynamics of Human Cytomegalovirus Genotypes in Transplant Patients over Time. J. Virol. 2010, 84, 7195–7203. [Google Scholar] [CrossRef] [PubMed]
  83. Gatherer, D.; Seirafian, S.; Cunningham, C.; Holton, M.; Dargan, D.J.; Baluchova, K.; Hector, R.D.; Galbraith, J.; Herzyk, P.; Wilkinson, G.W.G.; et al. High-Resolution Human Cytomegalovirus Transcriptome. Proc. Natl. Acad. Sci. USA 2011, 108, 19755–19760. [Google Scholar] [CrossRef] [PubMed]
  84. Wilkinson, G.W.G.; Davison, A.J.; Tomasec, P.; Fielding, C.A.; Aicheler, R.; Murrell, I.; Seirafian, S.; Wang, E.C.Y.; Weekes, M.; Lehner, P.J.; et al. Human Cytomegalovirus: Taking the Strain. Med. Microbiol. Immunol. 2015, 204, 273–284. [Google Scholar] [CrossRef] [PubMed]
  85. Sijmons, S.; Thys, K.; Mbong Ngwese, M.; Van Damme, E.; Dvorak, J.; Van Loock, M.; Li, G.; Tachezy, R.; Busson, L.; Aerssens, J.; et al. High-Throughput Analysis of Human Cytomegalovirus Genome Diversity Highlights the Widespread Occurrence of Gene-Disrupting Mutations and Pervasive Recombination. J. Virol. 2015, 89, 7673–7695. [Google Scholar] [CrossRef] [PubMed]
  86. Xu, S.; Schafer, X.; Munger, J. Expression of Oncogenic Alleles Induces Multiple Blocks to Human Cytomegalovirus Infection. J. Virol. 2016, 90, 4346–4356. [Google Scholar] [CrossRef]
  87. Branch, K.M.; Garcia, E.C.; Chen, Y.M.; McGregor, M.; Min, M.; Prosser, R.; Whitney, N.; Spencer, J.V. Productive Infection of Human Breast Cancer Cell Lines with Human Cytomegalovirus (HCMV). Pathogens 2021, 10, 641. [Google Scholar] [CrossRef] [PubMed]
  88. Singh, P.; Neumann, D.M. Persistent HCMV Infection of a Glioblastoma Cell Line Contributes to the Development of Resistance to Temozolomide. Virus Res. 2020, 276, 197829. [Google Scholar] [CrossRef] [PubMed]
  89. Yang, Z.; Tang, X.; McMullen, T.P.W.; Brindley, D.N.; Hemmings, D.G. PDGFRα Enhanced Infection of Breast Cancer Cells with Human Cytomegalovirus but Infection of Fibroblasts Increased Prometastatic Inflammation Involving Lysophosphatidate Signaling. IJMS 2021, 22, 9817. [Google Scholar] [CrossRef]
  90. Herbein, G. High-Risk Oncogenic Human Cytomegalovirus. Viruses 2022, 14, 2462. [Google Scholar] [CrossRef]
  91. Cohen, A.; Brodie, C.; Sarid, R. An Essential Role of ERK Signalling in TPA-Induced Reactivation of Kaposi’s Sarcoma-Associated Herpesvirus. J. Gen. Virol. 2006, 87, 795–802. [Google Scholar] [CrossRef] [PubMed]
  92. Van Sciver, N.; Ohashi, M.; Pauly, N.P.; Bristol, J.A.; Nelson, S.E.; Johannsen, E.C.; Kenney, S.C. Hippo Signaling Effectors YAP and TAZ Induce Epstein-Barr Virus (EBV) Lytic Reactivation through TEADs in Epithelial Cells. PLoS Pathog. 2021, 17, e1009783. [Google Scholar] [CrossRef] [PubMed]
  93. Cerasuolo, A.; Annunziata, C.; Tortora, M.; Starita, N.; Stellato, G.; Greggi, S.; Maglione, M.G.; Ionna, F.; Losito, S.; Botti, G.; et al. Comparative Analysis of HPV16 Gene Expression Profiles in Cervical and in Oropharyngeal Squamous Cell Carcinoma. Oncotarget 2017, 8, 34070–34081. [Google Scholar] [CrossRef] [PubMed]
  94. Guyon, J.; Haidar Ahmad, S.; El Baba, R.; Le Quang, M.; Bikfalvi, A.; Daubon, T.; Herbein, G. Generation of Glioblastoma in Mice Engrafted with Human Cytomegalovirus-Infected Astrocytes. Cancer Gene Ther. 2024. [Google Scholar] [CrossRef] [PubMed]
  95. Guven-Maiorov, E.; Tsai, C.-J.; Nussinov, R. Oncoviruses Can Drive Cancer by Rewiring Signaling Pathways Through Interface Mimicry. Front. Oncol. 2019, 9, 1236. [Google Scholar] [CrossRef] [PubMed]
  96. Aghamajidi, A.; Farhangnia, P.; Pashangzadeh, S.; Damavandi, A.R.; Jafari, R. Tumor-Promoting Myeloid Cells in the Pathogenesis of Human Oncoviruses: Potential Targets for Immunotherapy. Cancer Cell Int. 2022, 22, 327. [Google Scholar] [CrossRef] [PubMed]
  97. Mocarski, E.S. Immunomodulation by Cytomegaloviruses: Manipulative Strategies beyond Evasion. Trends Microbiol. 2002, 10, 332–339. [Google Scholar] [CrossRef]
  98. Semmes, E.C.; Hurst, J.H.; Walsh, K.M.; Permar, S.R. Cytomegalovirus as an Immunomodulator across the Lifespan. Curr. Opin. Virol. 2020, 44, 112–120. [Google Scholar] [CrossRef]
  99. El Baba, R.; Herbein, G. Immune Landscape of CMV Infection in Cancer Patients: From “Canonical” Diseases Toward Virus-Elicited Oncomodulation. Front. Immunol. 2021, 12, 730765. [Google Scholar] [CrossRef]
  100. Cinatl, J.; Scholz, M.; Doerr, H.W. Role of Tumor Cell Immune Escape Mechanisms in Cytomegalovirus-mediated Oncomodulation. Med. Res. Rev. 2005, 25, 167–185. [Google Scholar] [CrossRef]
  101. Cinatl, J.; Scholz, M.; Kotchetkov, R.; Vogel, J.-U.; Wilhelm Doerr, H. Molecular Mechanisms of the Modulatory Effects of HCMV Infection in Tumor Cell Biology. Trends Mol. Med. 2004, 10, 19–23. [Google Scholar] [CrossRef]
  102. Margulies, B.J.; Browne, H.; Gibson, W. Identification of the Human Cytomegalovirus G Protein-Coupled Receptor Homologue Encoded by UL33 in Infected Cells and Enveloped Virus Particles. Virology 1996, 225, 111–125. [Google Scholar] [CrossRef]
  103. Chen, Y.; Song, Y.; Du, W.; Gong, L.; Chang, H.; Zou, Z. Tumor-Associated Macrophages: An Accomplice in Solid Tumor Progression. J. Biomed. Sci. 2019, 26, 78. [Google Scholar] [CrossRef] [PubMed]
  104. Baasch, S.; Giansanti, P.; Kolter, J.; Riedl, A.; Forde, A.J.; Runge, S.; Zenke, S.; Elling, R.; Halenius, A.; Brabletz, S.; et al. Cytomegalovirus Subverts Macrophage Identity. Cell 2021, 184, 3774–3793.e25. [Google Scholar] [CrossRef] [PubMed]
  105. Khan, K.A.; Coaquette, A.; Davrinche, C.; Herbein, G. Bcl-3-Regulated Transcription from Major Immediate-Early Promoter of Human Cytomegalovirus in Monocyte-Derived Macrophages. J. Immunol. 2009, 182, 7784–7794. [Google Scholar] [CrossRef]
  106. Hanahan, D.; Weinberg, R.A. Hallmarks of Cancer: The Next Generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed]
  107. Hein, S.M.; Haricharan, S.; Johnston, A.N.; Toneff, M.J.; Reddy, J.P.; Dong, J.; Bu, W.; Li, Y. Luminal Epithelial Cells within the Mammary Gland Can Produce Basal Cells upon Oncogenic Stress. Oncogene 2016, 35, 1461–1467. [Google Scholar] [CrossRef]
  108. Rodilla, V.; Fre, S. Cellular Plasticity of Mammary Epithelial Cells Underlies Heterogeneity of Breast Cancer. Biomedicines 2018, 6, 103. [Google Scholar] [CrossRef] [PubMed]
  109. Tata, P.R.; Mou, H.; Pardo-Saganta, A.; Zhao, R.; Prabhu, M.; Law, B.M.; Vinarsky, V.; Cho, J.L.; Breton, S.; Sahay, A.; et al. Dedifferentiation of Committed Epithelial Cells into Stem Cells in Vivo. Nature 2013, 503, 218–223. [Google Scholar] [CrossRef]
  110. Yu, W.; Ma, Y.; Ochoa, A.C.; Shankar, S.; Srivastava, R.K. Cellular Transformation of Human Mammary Epithelial Cells by SATB2. Stem Cell Res. 2017, 19, 139–147. [Google Scholar] [CrossRef]
  111. Takahashi, K.; Yamanaka, S. Induction of Pluripotent Stem Cells from Mouse Embryonic and Adult Fibroblast Cultures by Defined Factors. Cell 2006, 126, 663–676. [Google Scholar] [CrossRef] [PubMed]
  112. Venkataramani, V.; Yang, Y.; Schubert, M.C.; Reyhan, E.; Tetzlaff, S.K.; Wißmann, N.; Botz, M.; Soyka, S.J.; Beretta, C.A.; Pramatarov, R.L.; et al. Glioblastoma Hijacks Neuronal Mechanisms for Brain Invasion. Cell 2022, 185, 2899–2917.e31. [Google Scholar] [CrossRef]
  113. Merchut-Maya, J.M.; Bartek, J.; Bartkova, J.; Galanos, P.; Pantalone, M.R.; Lee, M.; Cui, H.L.; Shilling, P.J.; Brøchner, C.B.; Broholm, H.; et al. Human Cytomegalovirus Hijacks Host Stress Response Fueling Replication Stress and Genome Instability. Cell Death Differ. 2022, 29, 1639–1653. [Google Scholar] [CrossRef]
  114. Fortunato, E.A.; Spector, D.H. Viral Induction of Site-specific Chromosome Damage. Rev. Med. Virol. 2003, 13, 21–37. [Google Scholar] [CrossRef] [PubMed]
  115. Siew, V.-K.; Duh, C.-Y.; Wang, S.-K. Human Cytomegalovirus UL76 Induces Chromosome Aberrations. J. Biomed. Sci. 2009, 16, 107. [Google Scholar] [CrossRef]
  116. Cao, R.; Wang, L.; Wang, H.; Xia, L.; Erdjument-Bromage, H.; Tempst, P.; Jones, R.S.; Zhang, Y. Role of Histone H3 Lysine 27 Methylation in Polycomb-Group Silencing. Science 2002, 298, 1039–1043. [Google Scholar] [CrossRef] [PubMed]
  117. Kim, K.H.; Roberts, C.W.M. Targeting EZH2 in Cancer. Nat. Med. 2016, 22, 128–134. [Google Scholar] [CrossRef] [PubMed]
  118. Veneti, Z.; Gkouskou, K.; Eliopoulos, A. Polycomb Repressor Complex 2 in Genomic Instability and Cancer. IJMS 2017, 18, 1657. [Google Scholar] [CrossRef]
  119. Guo, S.; Li, X.; Rohr, J.; Wang, Y.; Ma, S.; Chen, P.; Wang, Z. EZH2 Overexpression in Different Immunophenotypes of Breast Carcinoma and Association with Clinicopathologic Features. Diagn. Pathol. 2016, 11, 41. [Google Scholar] [CrossRef]
  120. Kleer, C.G.; Cao, Q.; Varambally, S.; Shen, R.; Ota, I.; Tomlins, S.A.; Ghosh, D.; Sewalt, R.G.A.B.; Otte, A.P.; Hayes, D.F.; et al. EZH2 Is a Marker of Aggressive Breast Cancer and Promotes Neoplastic Transformation of Breast Epithelial Cells. Proc. Natl. Acad. Sci. USA 2003, 100, 11606–11611. [Google Scholar] [CrossRef]
  121. Reid, B.M.; Vyas, S.; Chen, Z.; Chen, A.; Kanetsky, P.A.; Permuth, J.B.; Sellers, T.A.; Saglam, O. Morphologic and Molecular Correlates of EZH2 as a Predictor of Platinum Resistance in High-Grade Ovarian Serous Carcinoma. BMC Cancer 2021, 21, 714. [Google Scholar] [CrossRef] [PubMed]
  122. Xu, K.; Wu, Z.J.; Groner, A.C.; He, H.H.; Cai, C.; Lis, R.T.; Wu, X.; Stack, E.C.; Loda, M.; Liu, T.; et al. EZH2 Oncogenic Activity in Castration-Resistant Prostate Cancer Cells Is Polycomb-Independent. Science 2012, 338, 1465–1469. [Google Scholar] [CrossRef] [PubMed]
  123. Gonzalez, M.E.; Moore, H.M.; Li, X.; Toy, K.A.; Huang, W.; Sabel, M.S.; Kidwell, K.M.; Kleer, C.G. EZH2 Expands Breast Stem Cells through Activation of NOTCH1 Signaling. Proc. Natl. Acad. Sci. USA 2014, 111, 3098–3103. [Google Scholar] [CrossRef]
  124. Wu, J.; Crowe, D.L. The Histone Methyltransferase EZH2 Promotes Mammary Stem and Luminal Progenitor Cell Expansion, Metastasis and Inhibits Estrogen Receptor-Positive Cellular Differentiation in a Model of Basal Breast Cancer. Oncol. Rep. 2015, 34, 455–460. [Google Scholar] [CrossRef] [PubMed]
  125. Koh, C.M.; Iwata, T.; Zheng, Q.; Bethel, C.; Yegnasubramanian, S.; De Marzo, A.M. Myc Enforces Overexpression of EZH2 in Early Prostatic Neoplasia via Transcriptional and Post-Transcriptional Mechanisms. Oncotarget 2011, 2, 669–683. [Google Scholar] [CrossRef]
  126. Kuser-Abali, G.; Alptekin, A.; Cinar, B. Overexpression of MYC and EZH2 Cooperates to Epigenetically Silence MST1 Expression. Epigenetics 2014, 9, 634–643. [Google Scholar] [CrossRef] [PubMed]
  127. Dzutsev, A.; Badger, J.H.; Perez-Chanona, E.; Roy, S.; Salcedo, R.; Smith, C.K.; Trinchieri, G. Microbes and Cancer. Annu. Rev. Immunol. 2017, 35, 199–228. [Google Scholar] [CrossRef]
  128. Helmink, B.A.; Khan, M.A.W.; Hermann, A.; Gopalakrishnan, V.; Wargo, J.A. The Microbiome, Cancer, and Cancer Therapy. Nat. Med. 2019, 25, 377–388. [Google Scholar] [CrossRef] [PubMed]
  129. Godoy-Vitorino, F.; Romaguera, J.; Zhao, C.; Vargas-Robles, D.; Ortiz-Morales, G.; Vázquez-Sánchez, F.; Sanchez-Vázquez, M.; De La Garza-Casillas, M.; Martinez-Ferrer, M.; White, J.R.; et al. Cervicovaginal Fungi and Bacteria Associated With Cervical Intraepithelial Neoplasia and High-Risk Human Papillomavirus Infections in a Hispanic Population. Front. Microbiol. 2018, 9, 2533. [Google Scholar] [CrossRef]
  130. Puchhammer-Stöckl, E.; Görzer, I. Cytomegalovirus and Epstein-Barr Virus Subtypes—The Search for Clinical Significance. J. Clin. Virol. 2006, 36, 239–248. [Google Scholar] [CrossRef]
  131. Kohda, C.; Ino, S.; Ishikawa, H.; Kuno, Y.; Nagashima, R.; Iyoda, M. The Essential Role of Intestinal Microbiota in Cytomegalovirus Reactivation. Microbiol. Spectr. 2023, 11, e02341-23. [Google Scholar] [CrossRef]
  132. He, S.; Sharpless, N.E. Senescence in Health and Disease. Cell 2017, 169, 1000–1011. [Google Scholar] [CrossRef]
  133. Kowald, A.; Passos, J.F.; Kirkwood, T.B.L. On the Evolution of Cellular Senescence. Aging Cell 2020, 19, e13270. [Google Scholar] [CrossRef] [PubMed]
  134. Wang, B.; Kohli, J.; Demaria, M. Senescent Cells in Cancer Therapy: Friends or Foes? Trends Cancer 2020, 6, 838–857. [Google Scholar] [CrossRef]
  135. Birch, J.; Gil, J. Senescence and the SASP: Many Therapeutic Avenues. Genes. Dev. 2020, 34, 1565–1576. [Google Scholar] [CrossRef]
  136. Faget, D.V.; Ren, Q.; Stewart, S.A. Unmasking Senescence: Context-Dependent Effects of SASP in Cancer. Nat. Rev. Cancer 2019, 19, 439–453. [Google Scholar] [CrossRef]
  137. Gorgoulis, V.; Adams, P.D.; Alimonti, A.; Bennett, D.C.; Bischof, O.; Bishop, C.; Campisi, J.; Collado, M.; Evangelou, K.; Ferbeyre, G.; et al. Cellular Senescence: Defining a Path Forward. Cell 2019, 179, 813–827. [Google Scholar] [CrossRef] [PubMed]
  138. De Blander, H.; Morel, A.-P.; Senaratne, A.P.; Ouzounova, M.; Puisieux, A. Cellular Plasticity: A Route to Senescence Exit and Tumorigenesis. Cancers 2021, 13, 4561. [Google Scholar] [CrossRef] [PubMed]
  139. Baker, D.J.; Childs, B.G.; Durik, M.; Wijers, M.E.; Sieben, C.J.; Zhong, J.; Saltness, R.A.; Jeganathan, K.B.; Verzosa, G.C.; Pezeshki, A.; et al. Naturally Occurring p16Ink4a-Positive Cells Shorten Healthy Lifespan. Nature 2016, 530, 184–189. [Google Scholar] [CrossRef]
  140. Liu, J.; Erenpreisa, J.; Sikora, E. Polyploid Giant Cancer Cells: An Emerging New Field of Cancer Biology. Semin. Cancer Biol. 2022, 81, 1–4. [Google Scholar] [CrossRef]
  141. Pienta, K.J.; Hammarlund, E.U.; Brown, J.S.; Amend, S.R.; Axelrod, R.M. Cancer Recurrence and Lethality Are Enabled by Enhanced Survival and Reversible Cell Cycle Arrest of Polyaneuploid Cells. Proc. Natl. Acad. Sci. USA 2021, 118, e2020838118. [Google Scholar] [CrossRef] [PubMed]
  142. Zhang, X.; Yao, J.; Li, X.; Niu, N.; Liu, Y.; Hajek, R.A.; Peng, G.; Westin, S.; Sood, A.K.; Liu, J. Targeting Polyploid Giant Cancer Cells Potentiates a Therapeutic Response and Overcomes Resistance to PARP Inhibitors in Ovarian Cancer. Sci. Adv. 2023, 9, eadf7195. [Google Scholar] [CrossRef] [PubMed]
  143. Seoane, R.; Vidal, S.; Bouzaher, Y.H.; El Motiam, A.; Rivas, C. The Interaction of Viruses with the Cellular Senescence Response. Biology 2020, 9, 455. [Google Scholar] [CrossRef] [PubMed]
  144. Bouezzedine, F.; El Baba, R.; Morot-Bizot, S.; Diab-Assaf, M.; Herbein, G. Cytomegalovirus at the Crossroads of Immunosenescence and Oncogenesis. Explor. Immunol. 2023, 3, 17–27. [Google Scholar] [CrossRef]
  145. Hewavisenti, R.V.; Arena, J.; Ahlenstiel, C.L.; Sasson, S.C. Human Papillomavirus in the Setting of Immunodeficiency: Pathogenesis and the Emergence of next-Generation Therapies to Reduce the High Associated Cancer Risk. Front. Immunol. 2023, 14, 1112513. [Google Scholar] [CrossRef]
Figure 1. HCMV (high-risk strains) fulfills all the hallmarks of cancer described by Hanahan in 2022 [8].
Figure 1. HCMV (high-risk strains) fulfills all the hallmarks of cancer described by Hanahan in 2022 [8].
Cancers 16 01970 g001
Table 1. Transforming potential of human oncoviruses and HCMV. * HCV core, NS3, NS5A, and NS5B proteins potentiate oncogenic transformation; ** potential transforming HCMV proteins, still to be confirmed.
Table 1. Transforming potential of human oncoviruses and HCMV. * HCV core, NS3, NS5A, and NS5B proteins potentiate oncogenic transformation; ** potential transforming HCMV proteins, still to be confirmed.
Viral AgentOncoproteinsCancer Cell Lines/Tumor TypesAssociated/Described OutcomesReferences
HPVE6, E7, E5
  • HPV-positive cervical cancer cells Ca Ski, SiHa
  • HPV16 E6 and E7 expressing esophageal squamous cell carcinoma
  • Continued proliferation to confluency
  • Lack of apoptosis
  • Reduction in G0/G1 cell cycle arrest
  • PGCCs
  • Establishment of a cancer stem-like phenotype
  • Enhancement of migration, invasion, and spherogenesis
  • Elevated levels of proteins involved in EMT
[27,28,29,30,31]
HBVHBx, LHBs
  • HepG2 cells
  • pX-transfected hepatocellular carcinoma cells
  • Increased tumorigenicity, self-renewal, stemness
  • PGCCs
[32,33,34]
HCVNone (HCV core, NS3, NS5A, NS5B *) PGCCs[35,36,37]
HTLV-1Tax
  • MT2 and MT4 cells
  • Increased anti-apoptotic proteins
  • Rb depletion
  • PGCCs
[38,39,40,41,42]
MCPyVLT-Ag PGCCs[43,44,45,46]
EBVLMP1, EBNA2, EBNA3C, BNRF1
  • Burkitt’s lymphoma-derived cells
  • LMP1-expressing nasopharyngeal carcinoma cells
  • BHRF1-expressing nasopharyngeal carcinoma cells
  • Increased cell proliferation
  • Apoptosis suppression
  • Invasion
  • Colony formation ability
  • PGCCs
  • Tumor formation in nude mice
[47,48,49,50,51,52,53]
KSHVLANA
Cyclin K
  • LANA-expressing human breast cancer cell line MCF7
  • Inhibition of G2 arrest
  • PGCCs
[54,55,56,57,58,59]
HCMVIE1, IE2, US28 **
  • CMV-transformed human mammary epithelial cells (CTH cells), ovarian epithelial cells (CTO cells), prostate epithelial cells (CTP cells), and astrocytes (CEGBCs or CMV-elicited glioblastoma Cells)
  • Sustained cell proliferation
  • Increased telomerase activity
  • Colony formation ability
  • Stemness
  • EMT
  • Tumor formation in immunodeficient mice
[60,61,62,63,64,65,66,67,68,69]
Table 2. Direct oncogenic effect in epithelial cells and astrocytes infected with high-risk HCMV strains results in the generation of transformed CTH, CTO, CTP, and CEGBC cells.
Table 2. Direct oncogenic effect in epithelial cells and astrocytes infected with high-risk HCMV strains results in the generation of transformed CTH, CTO, CTP, and CEGBC cells.
HCMV-Transformed Primary Human CellOncogenic High-Risk HCMV StrainsPhenotypic Features of HCMV-Transformed CellsMolecular Characteristics of
HCMV-Transformed Cells
References
CTH cell
(CMV-transformed human mammary epithelial cells)
DB, BL
HCMV strains isolated from TNBC tumors
  • Cellular proliferation of heterogeneous cells
  • PGCCs, giant cell cycling
  • Colony formation in soft agar
  • Dedifferentiation of mature cells during the transformation process
  • Stemness
  • EMT, pEMT
  • Tumor formation in NSG mice
  • Inactivation of pRb and p53
  • Activation of telomerase activity
  • Activation of c-Myc, Akt, STAT3
  • Enhancement of EZH2
  • Detection of HCMV genes and proteins in transformed cells
  • Reaction of latent virus from transformed cells
[60,61,62,63,64,65,68]
CTO cells
(CMV-transformed ovarian epithelial cells)
DB, BL
HCMV strains isolated from HGSOC tumors
  • Cellular proliferation of heterogeneous cells
  • PGCCs, giant cell cycling
  • Colony formation in soft agar
  • Dedifferentiation of mature cells during the transformation process
  • Stemness
  • EMT and pEMT
  • Inactivation of pRb and p53
  • Decreased telomerase activity
  • Activation of c-Myc and Akt
  • Enhancement of EZH2
  • Detection of HCMV genes and proteins in transformed cells
[67]
CTP cell
(CMV-transformed prostate epithelial cells)
DB, BL
  • Cellular proliferation of heterogeneous cells
  • PGCCs, giant cell cycling
  • Colony formation in soft agar
  • Dedifferentiation of mature cells during the transformation process
  • Stemness
  • EMT
  • Inactivation of pRb and p53
  • Increased telomerase activity
  • Activation of c-Myc
  • Enhancement of EZH2
  • Detection of HCMV genes and proteins in transformed cells
[69]
CEGBC
(CMV-elicited glioblastoma Cells)
DB, BL
HCMV strains isolated from glioblastoma tumors
  • Cellular proliferation of heterogeneous cells
  • PGCCs
  • Colony formation in soft agar
  • Dedifferentiation of mature cells during the transformation process
  • Stemness
  • EMT
  • Spheroid formation
  • Invasiveness in vitro
  • Inactivation of pRb and p53
  • Increased telomerase activity
  • Activation of c-Myc and Akt
  • Enhancement of EZH2
  • Detection of HCMV genes and proteins in transformed cells
[66]
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

Herbein, G. Cellular Transformation by Human Cytomegalovirus. Cancers 2024, 16, 1970. https://doi.org/10.3390/cancers16111970

AMA Style

Herbein G. Cellular Transformation by Human Cytomegalovirus. Cancers. 2024; 16(11):1970. https://doi.org/10.3390/cancers16111970

Chicago/Turabian Style

Herbein, Georges. 2024. "Cellular Transformation by Human Cytomegalovirus" Cancers 16, no. 11: 1970. https://doi.org/10.3390/cancers16111970

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