Human endogenous retrovirus regulates the initiation and progression of cancers (Review)

  • Authors:
    • Srishti Sahu
    • Bharat Singh
    • Ambak Kumar Rai
  • View Affiliations

  • Published online on: August 3, 2022     https://doi.org/10.3892/mco.2022.2576
  • Article Number: 143
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The expression of genes is altered in various diseases and is responsible for the disease's initiation, progression and pathology. Several other genes, predominantly inactivated, may become activated in a given condition and contribute to the initiation and progression of the disease. Similarly, human endogenous viruses (HERVs) are an incomplete, non‑productive and inactive viral sequence present in the heterochromatin of the human genome, and are often referred to as junk DNA. HERVs were inserted into the host genome millions of years ago. However, they were silenced due to multiple mutations and recombination that occurred over time. However, their expression is increased in cancers due to either epigenetic or transcriptional dysregulation. Some of the HERVs having intact open reading frames have been reported to express virus‑like particles, functional peptides and proteins involved in tumorigenesis. To summarize, there is involvement of different HERVs in the initiation and progression of several cancers. The present review aims to provide concise information on HERV and its involvement in the initiation and progression of multiple types of cancer.

1. Introduction

Human endogenous retroviruses (HERVs) are molecular remnants of exogenous retroviruses that infected the human germline millions of years ago (horizontal transmission). These genetic fragments are stably integrated into the human genome; they are called endogenous retroviruses (1). They are often considered ‘non-functional DNA’ and account for approximately 8.29% of the human genome (2). They are inherited from parents to the offspring like any other gene (vertical transmission) (Fig. 1). The retroviruses are known for their transforming potential in their animal host via reverse transcription, but the HERVs are typically silenced or non-productive due to the accumulation of mutations and therefore rendered inadequate to produce virions (3). However, due to epigenetic dysregulation, some HERVs can retain their potency, produce virus-like particles, and express some immunogenic protein products, such as Syncytins (4), Np9, and Rec (5). The production of these proteins by the retroelements affects the biological functions and cancer immunoregulation (2).

HERVs are not usually expressed in normal cells. Nevertheless, some of their gene products and viral components can be expressed in human cells as antigens in some instances. Their expression has been reported to have a dual impact on human physiology (2). They aid in human physiological functions like regulating pluripotency of embryonic stem cells (6), involvement in placental morphogenesis (7), modulating the innate immune response (8), and regulating gene expression (9). On the contrary, they are also involved in the pathogenesis of multiple sclerosis (10), rheumatoid arthritis (11), schizophrenia (12), AIDS (13), cellular senescence (14), and diabetes (15). HERVs have gained a significant attraction due to their association with various cancers and their progression (3). Due to their abnormal expression in multiple malignancies and their pleiotropic role in oncogenesis, extensive research has targeted HERV antigens for immunotherapy by triggering both innate and adaptive immune response (16,17). Blocking the expression and function of HERVs in tumor cells via small interfering RNA (siRNA) or CRISPR (18), and anti-viral drugs (19) have also been studied by researchers (20). Since HERVs are extensively present in the human genome, their gene products can also be used as biomarkers to detect cancer progression (21).

2. Structure and classification of HERVs

HERVs have diverse structures ranging from solo LTR (long terminal repeats) to partially or fully intact open reading frames (ORFs) (22). The most active HERV group reported having a relatively intact ORF is the HERV-K HML-2 subtype (23). Classical HERVs structure contains the general components of the retroviruses, including protein-encoding sequences, GAG (gene-specific antigen), PRO (protease gene), POL (polymerase gene), and ENV (envelope gene) flanked by non-coding LTRs (Fig. 2) which are the regulatory region and can act as a promoter or an enhancer. Until now, 31 discrete groups of HERVs have been discovered. They can be categorized into three classes of retroviruses based on their similar phylogenetic origin to exogenous viruses (24) (Fig. 3).

3. Activation of HERVs

Generally, HERVs, having intact ORF, remain inactive due to CpG hypermethylation of their sequence which is catalyzed by DNA methylase-1(25). However, they can be activated by multiple factors like exogenous viruses such as human immunodeficiency virus (HIV) (26), Kaposi sarcoma-associated herpesvirus (KSHV), also known as human herpesvirus 8 (HHV8) (27), Epstein-Barr virus (28), and human T-cell leukemia virus-1(29). Also, epigenetic modifications (DNA demethylation, histone modification) (30), chemical substances (hydroquinone (31), phorbol-12-myristate-13-acetate (PMA) (32), phytohemagglutinin (PHA) (33), cupric salt (34)), physical factors (X-rays, UV-B) (35,36), and cytokines (37) (Fig. 4).

4. Role of HERV in cancer

Upon their activation, HERVs can be transcribed into full-length mRNA (38), spliced mRNA (39), and non-coding mRNA (40), resulting in either intact protein (41) or truncated protein (42). These HERV elements are found to be widely expressed in a variety of cancers like germ cell cancers (43), colorectal cancer (40), breast cancer (41), prostate cancer (44), ovarian cancer (45), lung cancer (46), melanoma (47), bladder cancer (48), lymphoma (39), hepatobiliary cancer (49), soft tissue sarcoma (50), Kaposi's sarcoma (27), seminomas (51), choriocarcinoma (52), and leukemia (53). Thus, these are potential biomarkers for cancers (49) and may have therapeutic potential if pursued to understand the causal relationship with individual disease types. However, according to the literature, HERVs have a dual opposing role in some cancer where they can either promote carcinogenesis or suppress it (2). Both the aspects of HERV mediated regulation will be discussed in the upcoming sections.

Cancer promoting effects

The Env proteins (Eg. syncytins, Np9, Rec) of different HERV groups HK2, HERV-W, HERV-V, HERV-H, and HERV-P (54) have been reported to suppress the immune system (55), affect the cell signaling pathways (56), and trigger cell-to-cell fusion (57) (Fig. 5). Thus, having an oncogenic effect. They may promote cell proliferation, growth, migration, invasion, metastasis, and stemness in different types of cancers, such as breast cancer (41), melanoma (58), leukemia (59), Kaposi's sarcoma (27), pancreatic cancer (60), etc. Therefore, HERV Env proteins are an exciting target for anti-cancer therapy. Few studies have shown that the expression of several HERV groups like HERV-W and HERV-K has contributed to cancer stemness or pluripotency in colorectal cancer (19) and melanoma cells (58), respectively. However, the underlying mechanisms are yet to be studied. Further, non-allelic recombination of HERV sequences causes their translocation to different regions in the genome. Their new proximity may activate their expression resulting in the activation of specific oncogenes (61) or disruption of a tumor suppressor gene (62). Besides, HERV LTR can act as an alternative promoter that can regulate cellular gene expression, leading to abnormal gene expression, such as switching on of proto-oncogenes, and finally contributing to tumorigenesis (16) (Fig. 6). Also, HERV-E derivative exon E1B has been observed to downregulate the surface expression of CD5 on T-cell in the case of T-cell acute lymphoblastic leukemia (T-ALL), thereby, inhibiting its functions and causing uncontrolled proliferation of leukemic T-cells (63) (refer to section 5.5). Therefore, it can be concluded that HERVs have a major role in oncogenesis, especially the Env protein of different HERV subgroups (3,27,41,56-59,64-67) (Table I).

Table I

Putative pro-oncogenic activity of Env molecules of important HERV groups.

Table I

Putative pro-oncogenic activity of Env molecules of important HERV groups.

First author/s, yearHERV groupTumor typeMoleculesOncogenic effect(Refs.)
Zhou et al, 2016; Lemaître et al, 2017HERV- K/HK2Breast cancerEnvIncrease proliferation, migration and invasiveness of the tumor, and cell-to-cell fusion(41,56)
Argaw-Denboba et al, 2017 MelanomaEnvInvolved in EMT, stemness and maintenance of tumor cells(58)
Chen et al, 2013 LeukemiaEnv (Np9)Increase in the growth of the tumor(59)
Grandi and Tramontano, 2018; Denne et al, 2007 Germline cancerEnv (Np9, Rec)Increase in cell proliferation and growth of the tumor, and inhibition of apoptosis of tumor cells(3,64)
Li et al, 2017 Pancreatic cancerEnvIncrease in the tumor growth, cell proliferation and metastasis of the tumor(60)
Dai et al, 2018 Kaposi's sarcomaEnv (Np9)Increase in cell proliferation and invasiveness of the tumor(27)
Bjerregaard et al, 2006HERV-WBreast cancerEnv (syncytin)Involved in cell-to-cell fusion(57)
Strissel et al, 2012 Endometrial cancerEnv (syncytin)Involved in cell-to-cell fusion(65)
Yu et al, 2014 Bladder urothelial cells carcinomaEnv (syncytin)Increase in cell proliferation and stemness of tumor(66)
Li et al, 2013 NeuroblastomaEnv (syncytin)Neuron excitotoxicity and neurological diseases(67)

[i] HERV, human endogenous retrovirus; Env, envelope protein.

Cancer suppressing effects

On the contrary, HERVs can exert suppressive effects on cancer instead of promoting it. The HERV protein products have been reported to stimulate innate, humoral, and cellular immune responses against malignant tumors by acting as an antigen like PAMP (pathogen-associated molecular patterns) recognized by pattern recognition receptors (PRRs) of immune cells (68). This triggers an immune response and causes the pro-inflammatory signals to exert an anti-viral effect against the HERV antigens by treating them as exogenous infections (2,69). This phenomenon has been reported in the case of clear cell renal cell carcinoma (ccRCC), where the infiltration of CD8+ cytotoxic T-cells triggered by HERV-E antigen was increased in ccRCC patients with hematopoietic stem cell transfer, which negatively affected the cancer progression (70). It might happen due to viral mimicry, which causes the activation of the interferon signaling pathway to upregulate the antitumor immune responses (3,16,71) like HERV-W interacts explicitly with the TLR4 and CD14 receptors, inducing the production of IL-1β, IL-6, and TNF-α pro-inflammatory cytokines. These cytokines further activate the dendritic cells, resulting in a Th-1 response (72). Different HERV Env peptides activate specific cytotoxic T-cells and dendritic cells (DCs) in cancers like ovarian, breast, and colorectal cancer (2). Activation of B-cells and production of antibodies have been seen in the case of breast cancer (73). Hence, it can be concluded that triggering the viral mimicry pathway and targeting the HERV proteins/transcripts can be a potential anti-cancer therapy.

To summarize, HERVs, especially the HK-2 group (Env protein) (3,27,41,56-60,64-67), play an accessory role in promoting carcinogenesis by suppressing the immune response by inhibiting tumor suppressor genes and activating multiple oncogenic signaling pathways. However, the viral mimicry effect of the HERV antigens induces an anti-cancer response. Therefore, HERVs remain an attractive target for immunotherapy.

5. Expression of HERV in various cancer types

Germ cell cancers

Most germ cell tumors (GCTs) like teratocarcinoma, multiple GCTs, and testicular cancers are known to express HERV-K for a long time. Mueller et al (74) performed a study on different stages of GCTs and suggested that the expression of HERV-K is regulated by the epigenetic mechanisms occurring during different stages of cellular development, which also affects the neighboring cells. Its expression is linked with oncogenesis, migration, and resistance to chemotherapy and correlates with poor prognosis (42). A variety of HERV-K viral particles promote tumor development in multiple GCTs (75). Teratocarcinoma is known to be a classical model for the study of HERV-K. An increase in the Np9 accessory protein in teratocarcinoma has the oncogenic potential (42,64). An increased expression of Gag protein in teratocarcinoma was also seen due to hypomethylation of the HERV-K sequence (76). Likewise, an increase in syncytin-1 transcribed by HERV-W Env was seen in seminoma patients, which may be involved in oncogenesis (2).

Breast cancer

HERV-K is the most reported and studied ERV in breast cancer. It is associated with tumor metastasis and invasion. It is also involved in cancer cell stemness and endothelial to mesenchymal transition (EMT) (41). HERV-K Env is involved in the carcinogenesis of breast cancer. The Env proteins downregulate the p53 cancer suppressor gene, causing the induction of cancer (77). They are also involved in the activation and upregulation of the RAS/ERK pathway, thus, causing the growth and proliferation of tumor cells (78). Anti-HERV-K Env antibody was able to inhibit tumor growth and induced breast cancer cells apoptosis, thus, showing an anti-tumor response (73). Increased HERV ENV, GAG mRNA, and RT (reverse transcriptase) expression in breast cancer are associated with poor prognosis (79,80). Detection of Env proteins in the early stages suggests that they may be involved in initiating oncogenesis in breast cancer (81). Thus, understanding the downstream function of Env may offer a new therapeutic target besides improving our knowledge. Besides, using vaccines against HERV-K Env may prevent breast cancer (82). Also, HERV-K RT can be used as an early prognostic biomarker for breast cancer as its expression was found in patients who develop cancer (79).

Both HERV-FRD Env and HERV-W Env (syncytin-1 and syncytin-2) proteins are expressed in breast cancer cells, promoting cell-to-cell fusion between endothelial cells and breast cancer cells (65). Non-coding RNA encoded by HERVs also promotes cancer progression in breast cancer. ZMYND8 protein, which is involved in suppressing metastatic cancer genes (VEGFR, TROJAN, CD44, and Slug), is degraded by ubiquitination by long non-coding RNA derived from HERV (83,84).

Skin cancers

HERV-K protein expression and HERV-K specific antibodies are found in different melanoma cell lines, assisting in cell-to-cell fusion. HERV-K proteins are immunogenic; therefore, antibodies are generated against them, resulting in increased antibody titer, which is correlated with poor prognosis. The Env protein maintains the tumor cell stemness and promotes phenotypic switching of tumorigenic cells, making them non-adherent and malignant (3,20). The overall expression of Env, Rec, Np9 and Gag has been reported in melanoma patients (85,86). Similarly, HERV-H is also found in a cell line Hs294T of melanoma which promotes dedifferentiation of tumor cells and helps them escape the immune cells (87). Further, HERV-W Env is expressed in cutaneous T cell lymphoma (CTCL), which promotes cell fusion (88), similar to its function in mediating trophoblast fusion during placental development (89).

Prostate cancer

Until now, only HERV-K expression has been reported in the case of prostate cancer. HERV-K Env protein was upregulated in prostate cancer patients (90). Targeting the Env protein via CRISPR/Cas9 downregulated the proto-oncogene SF2/ASF and RAS pathway expression in prostate cancer cell lines (44). Likewise, HERV-K Gag protein expression was also upregulated in prostate cancer due to demethylation and androgen stimulation (91). Gag protein expressions are also associated with smoking, old age, and disease status, leading to more aggressive prostate cancer (90). Anti-HERV-K Gag antibody titer was increased in stage III and stage IV of cancer compared to stage I and II, promoting carcinogenesis and depicting worse survival (91). Both HERV mRNA and anti-HERV antibodies have been reported to be used as potential biomarkers in prostate cancer (90).

Blood cancers

The expression of HERVs has been reported in acute myeloid leukemia (AML) (12,92-94), acute lymphoblastic leukemia (ALL) (12,93,94), acute mixed lineage leukemia (AMLL) (94), chronic myeloid leukemia (CML) (12), chronic lymphoblastic leukemia (CLL) (94), chronic mixed myeloid leukemia (CMML) (92), non-Hodgkin's lymphoma (NHL) and other lymphomas (94-96), essential thrombopenia (ET) (97), and myelodysplastic syndrome (MDS) (53) (Table II). The Np9 protein expressed by HERV-K activates a cascade of cellular signaling pathways (β-catenin, ERK, AKT, and Notch1), which helps in the proliferation of leukemia cells by increasing le1ukemia stem and progenitor cells (2,20). Expressions of HERV-K Env and Gag proteins are also reported in lymphoma patients. Likewise, HERV-W Env (syncytin-1) expression is found in leukemic patients and is a potential diagnostic marker (94). How these viral antigens influence oncogenesis is not clearly understood.

Table II

Expression of reported HERVs in different blood cancers.

Table II

Expression of reported HERVs in different blood cancers.

First author/s, yearHERV groupExpression in blood cancer type(Refs.)
Chen et al, 2013; Saini et al, 2020; Contreras-Galindo et al, 2008; Tatkiewicz et al, 2020; Morgan and Brodsky, 2004HERV-KAML, ALL, CML, ET, multiple myeloma, B cell lymphoma, large cell lymphoma, mantle cell lymphoma(59,92,95-97)
Saini et al, 2020; Alqahtani et al, 2016; Sun et al, 2010HERV-WAML, ALL, AMLL, CML, CLL, CMML, MDS, NHL(92-94)
Saini et al, 2020HERV-EAML, CMML, MDS(92)

[i] HERV, human endogenous retrovirus; AML, acute myeloid leukemia; ALL, acute lymphoblastic leukemia; CML, chronic myeloid leukemia; ET, essential thrombopenia; AMLL, acute mixed lineage leukemia; CLL, chronic lymphoblastic leukemia; CMML, chronic mixed myeloid leukemia; MDS, myelodysplastic syndrome; NHL, non-Hodgkin's lymphoma.

In a study performed by Rai et al (63) on T-ALL, exon E1B was observed to be regulating the surface expression of the CD5 gene on the T-cells. Exon E1B is a non-conventional exon of CD5 and a derivative of the HERV-E sequence. Exon E1B expression is seen to be upregulated in the case of T-ALL, while there is downregulation of conventional exon E1A. Due to the lack of leader peptide in the case of exon E1B, surface expression of CD5 is decreased and it is accumulated inside the cytoplasm. Consequently, the negative regulation function of CD5 is compromised, causing the uncontrolled proliferation of leukemic T-cells, thus, promoting carcinogenesis (63).

Kidney cancer

Cytotoxic T-cells were involved in the regression of kidney cancer in clear cell renal cell carcinoma (ccRCC) patients undergoing hematopoietic stem cell transfer. After the investigation, it was found that CT-RCC, a highly immunogenic antigen encoded by HERV-E, induces the activation of CD8+ T-cells and, therefore, triggers an immune response against the RCC cells. This led to tumor regression in-vitro and in-vivo (70). Further, it was found that an increase in the HERV-E expression was strongly correlated with the non-functional von Hippel Lindau (VHL) tumor suppressor gene. Absence of VHL protein induced the expression of HIF-2α, which regulated the expression of HERV-E due to the presence of hypoxia regulatory element (HRE) on the 5' LTR of HERV-E (98-100). A full-length protein of HERV-E, Env expression, was also selectively expressed in ccRCC patients, which can serve as a biomarker for ccRCC (101).

Kaposi's sarcoma

Kaposi's sarcoma is caused by the infection of human herpesvirus 8 (HHV8), also known as Kaposi's sarcoma-associated herpesvirus (KSHV), and is the leading cause of mortality in HIV infection (102). It is characterized by the most common AIDS-related malignancies, which still require effective treatment options. Kaposi's sarcoma is a classic example of activation of HERV through exogenous viral infection. KSHV infection was found to upregulate the production of HERV-K Np9 protein, which advanced the invasiveness of primary endothelial cells by the action of disintegrins and metalloproteinases, contributing to carcinogenesis increasing the morbidity among Kaposi's sarcoma patients (27).

Ovarian cancer

Various HERVs have been reported to be expressed in ovarian cancer. Both HERV-K Env and RT expressions were increased in ovarian cancer compared to adjacent healthy and benign tissues. HERV-K Env antigens triggered the proliferation and activation of specific cytotoxic T-cells and IFNγ production. This led to the lysis of autologous tumor cells (45). Also, demethylation of ovarian cancer cells by DNA methyltransferase inhibitors (DNMTis) induces the production of double-stranded RNA (dsRNA) of HERV, which activates the viral defense pathway, enhancing the innate immune response and apoptosis (103).

Colorectal cancer

Various HERV expression in colorectal cancer (CRC) has been reported, including HERV-K, HERV-W, HERV-H, HERV-FRD, and HERV-3. HERV-K is involved in cell growth, proliferation, migration, and colonization (20). Expression of HERV-W is correlated with poor prognosis in syncytintal cancer (104). HERV-H Env exerts an immune-modulatory effect (40). HERVs are also suggested to be used as a biomarker and clinical examination for better predicting CRC patient survival (105).

Pancreatic cancer

HERV-K Env plays a significant role in pancreatic cancer, whose high expression is associated with a poor prognosis. It promotes tumor cell proliferation, growth, and metastasis. In particular, HERV-K Env interferes with the signal transduction pathway RAS/ERK/RSK pathway and thus promotes carcinogenesis. HERV-K RT activity was also observed in pancreatic cancer tissues (60).

6. Conclusion

HERVs have been associated with cancer for a long time. Their abnormal level of expression has been found in a variety of cancers. Different groups of HERV are found to be overexpressed in different cancers. Multiple factors are responsible for their activation like epigenetic dysregulation (30), exogenous infections (26-29), radiations (35,36), cytokines (37), chemical induction (31-34), etc. They encode highly immunogenic antigens whose expression can promote or inhibit cancer advancement by modulating the immune system. HERVs are correlated with tumor cell proliferation, migration, decreased apoptosis, endothelial to mesenchymal transition (EMT), and immune suppression, thus initiating and promoting oncogenesis (20). Since the expression of HERV is a natural phenomenon, each HERV protein must be characterized separately to elucidate its role in the pathogenesis of different cancer and other diseases. Future studies may shed light on the effect of vaccination against a specific epitope of HERV elements and monoclonal antibody (MAB) on the control and prevention of certain cancers. It suggests the need to develop an onco-immunotherapy approach for rapidly evolving cancer types.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

SS was involved in conceptualization, wrote the original draft and was involved in visualization. BS reviewed and edited the manuscript. AKR was involved in conceptualization and provided supervision. Data authentication is not applicable. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Wang X, Huang J and Zhu F: Human endogenous retroviral envelope protein syncytin-1 and inflammatory abnormalities in neuropsychological diseases. Front Psychiatry. 9(422)2018.PubMed/NCBI View Article : Google Scholar

2 

Gao Y, Yu XF and Chen T: Human endogenous retroviruses in cancer: Expression, regulation and function. Oncol Lett. 21(121)2021.PubMed/NCBI View Article : Google Scholar

3 

Grandi N and Tramontano E: HERV envelope proteins: physiological role and pathogenic potential in cancer and autoimmunity. Front Microbiol. 9(462)2018.PubMed/NCBI View Article : Google Scholar

4 

Durnaoglu S, Lee SK and Ahnn J: Syncytin, envelope protein of human endogenous retrovirus (HERV): No longer ‘fossil’in human genome. Anim Cells Syst (Seoul). 25:358–368. 2022.PubMed/NCBI View Article : Google Scholar

5 

Salavatiha Z, Soleimani-Jelodar R and Jalilvand S: The role of endogenous retroviruses-K in human cancer. Rev Med Virol. 30:1–13. 2020.PubMed/NCBI View Article : Google Scholar

6 

Santoni FA, Guerra J and Luban J: HERV-H RNA is abundant in human embryonic stem cells and a precise marker for pluripotency. Retrovirology. 9(111)2012.PubMed/NCBI View Article : Google Scholar

7 

Mi S, Lee X, Li X, Veldman GM, Finnerty H, Racie L, LaVallie E, Tang XY, Edouard P, Howes S, et al: Syncytin is a captive retroviral envelope protein involved in human placental morphogenesis. Nature. 403:785–789. 2000.PubMed/NCBI View Article : Google Scholar

8 

Alcazer V, Bonaventura P and Depil S: Human endogenous retroviruses (HERVs): Shaping the innate immune response in cancers. Cancers (Basel). 12(610)2020.PubMed/NCBI View Article : Google Scholar

9 

Kim HS: Genomic impact, chromosomal distribution and transcriptional regulation of HERV elements. Mol Cells. 33:539–544. 2012.PubMed/NCBI View Article : Google Scholar

10 

Kremer D, Gruchot J, Weyers V, Oldemeier L, Göttle P, Healy L, Ho Jang J, Kang T, Xu Y, Volsko C, et al: pHERV-W envelope protein fuels microglial cell-dependent damage of myelinated axons in multiple sclerosis. Proc Natl Acad Sci USA. 116:15216–15225. 2019.PubMed/NCBI View Article : Google Scholar

11 

Dervan E, Bhattacharyya DD, McAuliffe JD, Khan FH and Glynn SA: Ancient adversary-HERV-K (HML-2) in cancer. Front Oncol. 11(658489)2021.PubMed/NCBI View Article : Google Scholar

12 

Huang WJ, Liu ZC, Wei W, Wang GH, Wu JG and Zhu F: Human endogenous retroviral pol RNA and protein detected and identified in the blood of individuals with schizophrenia. Schizophr Res. 83:193–199. 2006.PubMed/NCBI View Article : Google Scholar

13 

Monde K, Terasawa H, Nakano Y, Soheilian F, Nagashima K, Maeda Y and Ono A: Molecular mechanisms by which HERV-K Gag interferes with HIV-1 Gag assembly and particle infectivity. Retrovirology. 14(27)2017.PubMed/NCBI View Article : Google Scholar

14 

Liu X, Liu Z, Sun L, Ren J, Wu Z, Jiang X, Ji Q, Wang Q, Fan Y, Cai Y, et al: Resurrection of human endogenous retroviruses during aging reinforces senescence. bioRxiv: 2021.02.22.432260, 2021.

15 

Levet S, Charvet B, Bertin A, Deschaumes A, Perron H and Hober D: Human endogenous retroviruses and type 1 diabetes. Curr Diab Rep. 19(141)2019.PubMed/NCBI View Article : Google Scholar

16 

Hurst TP and Magiorkinis G: Activation of the innate immune response by endogenous retroviruses. J Gen Virol. 96:1207–1218. 2015.PubMed/NCBI View Article : Google Scholar

17 

Smith CC, Beckermann KE, Bortone DS, De Cubas AA, Bixby LM, Lee SJ, Panda A, Ganesan S, Bhanot G, Wallen EM, et al: Endogenous retroviral signatures predict immunotherapy response in clear cell renal cell carcinoma. J Clin Invest. 128:4804–4820. 2018.PubMed/NCBI View Article : Google Scholar

18 

Mangeney M, Pothlichet J, Renard M, Ducos B and Heidmann T: Endogenous retrovirus expression is required for murine melanoma tumor growth in vivo. Cancer Res. 65:2588–2591. 2005.PubMed/NCBI View Article : Google Scholar

19 

Díaz-Carballo D, Acikelli AH, Klein J, Jastrow H, Dammann P, Wyganowski T, Guemues C, Gustmann S, Bardenheuer W, Malak S, et al: Therapeutic potential of antiviral drugs targeting chemorefractory colorectal adenocarcinoma cells overexpressing endogenous retroviral elements. J Exp Clin Cancer Res. 34(81)2015.PubMed/NCBI View Article : Google Scholar

20 

Müller MD, Holst PJ and Nielsen KN: A systematic review of expression and immunogenicity of human endogenous retroviral proteins in cancer and discussion of therapeutic approaches. Int J Mol Sci. 23(1330)2022.PubMed/NCBI View Article : Google Scholar

21 

Liu CH, Grandi N, Palanivelu L, Tramontano E and Lin LT: Contribution of human retroviruses to disease development-A focus on the HIV- and HERV-cancer relationships and treatment strategies. Viruses. 12(852)2020.PubMed/NCBI View Article : Google Scholar

22 

Kassiotis G: Endogenous retroviruses and the development of cancer. J Immunol. 192:1343–1349. 2014.PubMed/NCBI View Article : Google Scholar

23 

Subramanian RP, Wildschutte JH, Russo C and Coffin JM: Identification, characterization, and comparative genomic distribution of the HERV-K (HML-2) group of human endogenous retroviruses. Retrovirology. 8(90)2011.PubMed/NCBI View Article : Google Scholar

24 

Curty G, Marston JL, de Mulder Rougvie M, Leal FE, Nixon DF and Soares MA: Human endogenous retrovirus K in cancer: A potential biomarker and immunotherapeutic target. Viruses. 12(726)2020.PubMed/NCBI View Article : Google Scholar

25 

Lavie L, Kitova M, Maldener E, Meese E and Mayer J: CpG methylation directly regulates transcriptional activity of the human endogenous retrovirus family HERV-K(HML-2). J Virol. 79:876–883. 2005.PubMed/NCBI View Article : Google Scholar

26 

Contreras-Galindo R, López P, Vélez R and Yamamura Y: HIV-1 infection increases the expression of human endogenous retroviruses type K (HERV-K) in vitro. AIDS Res Hum Retroviruses. 23:116–122. 2007.PubMed/NCBI View Article : Google Scholar

27 

Dai L, Del Valle L, Miley W, Whitby D, Ochoa AC, Flemington EK and Qin Z: Transactivation of human endogenous retrovirus K (HERV-K) by KSHV promotes Kaposi's sarcoma development. Oncogene. 37:4534–4545. 2018.PubMed/NCBI View Article : Google Scholar

28 

Sutkowski N, Conrad B, Thorley-Lawson DA and Huber BT: Epstein-Barr virus transactivates the human endogenous retrovirus HERV-K18 that encodes a superantigen. Immunity. 15:579–589. 2001.PubMed/NCBI View Article : Google Scholar

29 

Toufaily C, Landry S, Leib-Mosch C, Rassart E and Barbeau B: Activation of LTRs from different human endogenous retrovirus (HERV) families by the HTLV-1 tax protein and T-cell activators. Viruses. 3:2146–2159. 2011.PubMed/NCBI View Article : Google Scholar

30 

Romanish MT, Cohen CJ and Mager DL: Potential mechanisms of endogenous retroviral-mediated genomic instability in human cancer. In: Seminars in cancer biology. Elsevier, pp246-253, 2010.

31 

Conti A, Rota F, Ragni E, Favero C, Motta V, Lazzari L, Bollati V, Fustinoni S and Dieci G: Hydroquinone induces DNA hypomethylation-independent overexpression of retroelements in human leukemia and hematopoietic stem cells. Biochem Biophys Res Commun. 474:691–695. 2016.PubMed/NCBI View Article : Google Scholar

32 

Johnston JB, Silva C, Holden J, Warren KG, Clark AW and Power C: Monocyte activation and differentiation augment human endogenous retrovirus expression: Implications for inflammatory brain diseases. Ann Neurol. 50:434–442. 2001.PubMed/NCBI View Article : Google Scholar

33 

Kelleher CA, Wilkinson DA, Freeman JD, Mager DL and Gelfand EW: Expression of novel transposon-containing mRNAs in human T cells. J Gen Virol. 77:1101–1110. 1996.PubMed/NCBI View Article : Google Scholar

34 

Karimi A, Sheervalilou R and Kahroba H: A new insight on activation of human endogenous retroviruses (HERVs) in malignant melanoma upon exposure to CuSO4. Biol Trace Elem Res. 191:70–74. 2019.PubMed/NCBI View Article : Google Scholar

35 

Lee JR, Ahn K, Kim YJ, Jung YD and Kim HS: Radiation-induced human endogenous retrovirus (HERV)-R env gene expression by epigenetic control. Radiat Res. 178:379–384. 2012.PubMed/NCBI View Article : Google Scholar

36 

Reiche J, Pauli G and Ellerbrok H: Differential expression of human endogenous retrovirus K transcripts in primary human melanocytes and melanoma cell lines after UV irradiation. Melanoma Res. 20:435–440. 2010.PubMed/NCBI View Article : Google Scholar

37 

Katsumata K, Ikeda H, Sato M, Ishizu A, Kawarada Y, Kato H, Wakisaka A, Koike T and Yoshiki T: Cytokine regulation of env gene expression of human endogenous retrovirus-R in human vascular endothelial cells. Clin Immunol. 93:75–80. 1999.PubMed/NCBI View Article : Google Scholar

38 

Montesion M, Bhardwaj N, Williams ZH, Kuperwasser C and Coffin JM: Mechanisms of HERV-K (HML-2) transcription during human mammary epithelial cell transformation. J Virol. 92:e01258–17. 2017.PubMed/NCBI View Article : Google Scholar

39 

Barth M, Gröger V, Cynis H and Staege MS: Identification of human endogenous retrovirus transcripts in Hodgkin lymphoma cells. Mol Biol Rep. 46:1885–1893. 2019.PubMed/NCBI View Article : Google Scholar

40 

Liang Q, Xu Z, Xu R, Wu L and Zheng S: Expression patterns of non-coding spliced transcripts from human endogenous retrovirus HERV-H elements in colon cancer. PLoS One. 7(e29950)2012.PubMed/NCBI View Article : Google Scholar

41 

Zhou F, Li M, Wei Y, Lin K, Lu Y, Shen J, Johanning GL and Wang-Johanning F: Activation of HERV-K Env protein is essential for tumorigenesis and metastasis of breast cancer cells. Oncotarget. 7:84093–84117. 2016.PubMed/NCBI View Article : Google Scholar

42 

Chan SM, Sapir T, Park SS, Rual JF, Contreras-Galindo R, Reiner O and Markovitz DM: The HERV-K accessory protein Np9 controls viability and migration of teratocarcinoma cells. PLoS One. 14(e0212970)2019.PubMed/NCBI View Article : Google Scholar

43 

Sauter M, Roemer K, Best B, Afting M, Schommer S, Seitz G, Hartmann M and Mueller-Lantzsch N: Specificity of antibodies directed against Env protein of human endogenous retroviruses in patients with germ cell tumors. Cancer Res. 56:4362–4365. 1996.PubMed/NCBI

44 

Ibba G, Piu C, Uleri E, Serra C and Dolei A: Disruption by SaCas9 endonuclease of HERV-Kenv, a retroviral gene with oncogenic and neuropathogenic potential, inhibits molecules involved in cancer and amyotrophic lateral sclerosis. Viruses. 10(412)2018.PubMed/NCBI View Article : Google Scholar

45 

Rycaj K, Plummer JB, Yin B, Li M, Garza J, Radvanyi L, Ramondetta LM, Lin K, Johanning GL, Tang DG and Wang-Johanning F: Cytotoxicity of human endogenous retrovirus K-specific T cells toward autologous ovarian cancer cells. Clin Cancer Res. 21:471–483. 2015.PubMed/NCBI View Article : Google Scholar

46 

Zare M, Mostafaei S, Ahmadi A, Azimzadeh Jamalkandi S, Abedini A, Esfahani-Monfared Z, Dorostkar R and Saadati M: Human endogenous retrovirus env genes: Potential blood biomarkers in lung cancer. Microb Pathog. 115:189–193. 2018.PubMed/NCBI View Article : Google Scholar

47 

Krishnamurthy J, Rabinovich BA, Mi T, Switzer KC, Olivares S, Maiti SN, Plummer JB, Singh H, Kumaresan PR, Huls HM, et al: Genetic engineering of T cells to target HERV-K, an ancient retrovirus on melanoma. Clin Cancer Res. 21:3241–3251. 2015.PubMed/NCBI View Article : Google Scholar

48 

Kreimer U, Schulz WA, Koch A, Niegisch G and Goering W: HERV-K and LINE-1 DNA methylation and reexpression in urothelial carcinoma. Front Oncol. 3(255)2013.PubMed/NCBI View Article : Google Scholar

49 

Grabski DF, Hu Y, Sharma M and Rasmussen SK: Close to the bedside: A systematic review of endogenous retroviruses and their impact in oncology. J Surg Res. 240:145–155. 2019.PubMed/NCBI View Article : Google Scholar

50 

Giebler M, Staege MS, Blauschmidt S, Ohm LI, Kraus M, Würl P, Taubert H and Greither T: Elevated HERV-K expression in soft tissue sarcoma is associated with worsened relapse-free survival. Front Microbiol. 9(211)2018.PubMed/NCBI View Article : Google Scholar

51 

Galli UM, Sauter M, Lecher B, Maurer S, Herbst H, Roemer K and Mueller-Lantzsch N: Human endogenous retrovirus rec interferes with germ cell development in mice and may cause carcinoma in situ, the predecessor lesion of germ cell tumors. Oncogene. 24:3223–3228. 2005.PubMed/NCBI View Article : Google Scholar

52 

Aagaard L, Bjerregaard B, Kjeldbjerg AL, Pedersen FS, Larsson LI and Rossi JJ: Silencing of endogenous envelope genes in human choriocarcinoma cells shows that envPb1 is involved in heterotypic cell fusions. J Gen Virol. 93(1696)2012.PubMed/NCBI View Article : Google Scholar

53 

Bergallo M, Montanari P, Mareschi K, Merlino C, Berger M, Bini I, Daprà V, Galliano I and Fagioli F: Expression of the pol gene of human endogenous retroviruses HERV-K and -W in leukemia patients. Arch Virol. 162:3639–3644. 2017.PubMed/NCBI View Article : Google Scholar

54 

Mangeney M, Renard M, Schlecht-Louf G, Bouallaga I, Heidmann O, Letzelter C, Richaud A, Ducos B and Heidmann T: Placental syncytins: Genetic disjunction between the fusogenic and immunosuppressive activity of retroviral envelope proteins. Proc Natl Acad Sci USA. 104:20534–20539. 2007.PubMed/NCBI View Article : Google Scholar

55 

Cianciolo GJ, Copeland TD, Oroszlan S and Snyderman R: Inhibition of lymphocyte proliferation by a synthetic peptide homologous to retroviral envelope proteins. Science. 230:453–455. 1985.PubMed/NCBI View Article : Google Scholar

56 

Lemaître C, Tsang J, Bireau C, Heidmann T and Dewannieux M: A human endogenous retrovirus-derived gene that can contribute to oncogenesis by activating the ERK pathway and inducing migration and invasion. PLoS Pathog. 13(e1006451)2017.PubMed/NCBI View Article : Google Scholar

57 

Bjerregaard B, Holck S, Christensen I and Larsson LI: Syncytin is involved in breast cancer-endothelial cell fusions. Cell Mol Life Sci. 63:1906–1911. 2006.PubMed/NCBI View Article : Google Scholar

58 

Argaw-Denboba A, Balestrieri E, Serafino A, Cipriani C, Bucci I, Sorrentino R, Sciamanna I, Gambacurta A, Sinibaldi-Vallebona P and Matteucci C: HERV-K activation is strictly required to sustain CD133+ melanoma cells with stemness features. J Exp Clin Cancer Res. 36(20)2017.PubMed/NCBI View Article : Google Scholar

59 

Chen T, Meng Z, Gan Y, Wang X, Xu F, Gu Y, Xu X, Tang J, Zhou H, Zhang X, et al: The viral oncogene Np9 acts as a critical molecular switch for co-activating β-catenin, ERK, Akt and Notch1 and promoting the growth of human leukemia stem/progenitor cells. Leukemia. 27:1469–1478. 2013.PubMed/NCBI View Article : Google Scholar

60 

Li M, Radvanyi L, Yin B, Rycaj K, Li J, Chivukula R, Lin K, Lu Y, Shen J, Chang DZ, et al: Downregulation of human endogenous retrovirus type K (HERV-K) viral env RNA in pancreatic cancer cells decreases cell proliferation and tumor growth. Clin Cancer Res. 23:5892–5911. 2017.PubMed/NCBI View Article : Google Scholar

61 

Zhang M, Liang JQ and Zheng S: Expressional activation and functional roles of human endogenous retroviruses in cancers. Rev Med Virol. 29(e2025)2019.PubMed/NCBI View Article : Google Scholar

62 

Cherkasova E, Malinzak E, Rao S, Takahashi Y, Senchenko VN, Kudryavtseva AV, Nickerson ML, Merino M, Hong JA, Schrump DS, et al: Inactivation of the von Hippel-Lindau tumor suppressor leads to selective expression of a human endogenous retrovirus in kidney cancer. Oncogene. 30:4697–4706. 2011.PubMed/NCBI View Article : Google Scholar

63 

Rai AK, Singh A, Saxena A, Seth T, Raina V and Mitra DK: Exonal switch down-regulates the expression of CD5 on blasts of acute T cell leukaemia. Clin Exp Immunol. 190:340–350. 2017.PubMed/NCBI View Article : Google Scholar

64 

Denne M, Sauter M, Armbruester V, Licht JD, Roemer K and Mueller-Lantzsch N: Physical and functional interactions of human endogenous retrovirus proteins Np9 and rec with the promyelocytic leukemia zinc finger protein. J Virol. 81:5607–5616. 2007.PubMed/NCBI View Article : Google Scholar

65 

Strissel PL, Ruebner M, Thiel F, Wachter D, Ekici AB, Wolf F, Thieme F, Ruprecht K, Beckmann MW and Strick R: Reactivation of codogenic endogenous retroviral (ERV) envelope genes in human endometrial carcinoma and prestages: Emergence of new molecular targets. Oncotarget. 3:1204–1219. 2012.PubMed/NCBI View Article : Google Scholar

66 

Yu H, Liu T, Zhao Z, Chen Y, Zeng J, Liu S and Zhu F: Mutations in 3'-long terminal repeat of HERV-W family in chromosome 7 upregulate syncytin-1 expression in urothelial cell carcinoma of the bladder through interacting with c-Myb. Oncogene. 33:3947–3958. 2014.PubMed/NCBI View Article : Google Scholar

67 

Li S, Liu ZC, Yin SJ, Chen YT, Yu HL, Zeng J, Zhang Q and Zhu F: Human endogenous retrovirus W family envelope gene activates the small conductance Ca2+-activated K+ channel in human neuroblastoma cells through CREB. Neuroscience. 247:164–174. 2013.PubMed/NCBI View Article : Google Scholar

68 

Barbalat R, Ewald SE, Mouchess ML and Barton GM: Nucleic acid recognition by the innate immune system. Annu Rev Immunol. 29:185–214. 2011.PubMed/NCBI View Article : Google Scholar

69 

Bannert N, Hofmann H, Block A and Hohn O: HERVs new role in cancer: From accused perpetrators to cheerful protectors. Front Microbiol. 9(178)2018.PubMed/NCBI View Article : Google Scholar

70 

Panda A, de Cubas AA, Stein M, Riedlinger G, Kra J, Mayer T, Smith CC, Vincent BG, Serody JS, Beckermann KE, et al: Endogenous retrovirus expression is associated with response to immune checkpoint blockade in clear cell renal cell carcinoma. JCI Insight. 3(e121522)2018.PubMed/NCBI View Article : Google Scholar

71 

Cao W, Kang R, Xiang Y and Hong J: Human endogenous retroviruses in clear cell renal cell carcinoma: Biological functions and clinical values. Onco Targets Ther. 13:7877–7885. 2020.PubMed/NCBI View Article : Google Scholar

72 

Rolland A, Jouvin-Marche E, Viret C, Faure M, Perron H and Marche PN: The envelope protein of a human endogenous retrovirus-W family activates innate immunity through CD14/TLR4 and promotes Th1-like responses. J Immunol. 176:7636–7644. 2006.PubMed/NCBI View Article : Google Scholar

73 

Wang-Johanning F, Li M, Esteva FJ, Hess KR, Yin B, Rycaj K, Plummer JB, Garza JG, Ambs S and Johanning GL: Human endogenous retrovirus type K antibodies and mRNA as serum biomarkers of early-stage breast cancer. Int J Cancer. 134:587–595. 2014.PubMed/NCBI View Article : Google Scholar

74 

Mueller T, Hantsch C, Volkmer I and Staege MS: Differentiation-dependent regulation of human endogenous retrovirus K sequences and neighboring genes in germ cell tumor cells. Front Microbiol. 9(1253)2018.PubMed/NCBI View Article : Google Scholar

75 

Kleiman A, Senyuta N, Tryakin A, Sauter M, Karseladze A, Tjulandin S, Gurtsevitch V and Mueller-Lantzsch N: HERV-K(HML-2) GAG/ENV antibodies as indicator for therapy effect in patients with germ cell tumors. Int J Cancer. 110:459–461. 2004.PubMed/NCBI View Article : Google Scholar

76 

Götzinger N, Sauter M, Roemer K and Mueller-Lantzsch N: Regulation of human endogenous retrovirus-K Gag expression in teratocarcinoma cell lines and human tumours. J Gen Virol. 77:2983–2990. 1996.PubMed/NCBI View Article : Google Scholar

77 

Wang Z, Zheng Y, Park HJ, Li J, Carr JR, Chen YJ, Kiefer MM, Kopanja D, Bagchi S, Tyner AL and Raychaudhuri P: Targeting FoxM1 effectively retards p53-null lymphoma and sarcoma. Mol Cancer Ther. 12:759–767. 2013.PubMed/NCBI View Article : Google Scholar

78 

von Lintig FC, Dreilinger AD, Varki NM, Wallace AM, Casteel DE and Boss GR: Ras activation in human breast cancer. Breast Cancer Res Treat. 62:51–62. 2000.PubMed/NCBI View Article : Google Scholar

79 

Golan M, Hizi A, Resau JH, Yaal-Hahoshen N, Reichman H, Keydar I and Tsarfaty I: Human endogenous retrovirus (HERV-K) reverse transcriptase as a breast cancer prognostic marker. Neoplasia. 10:521–533. 2008.PubMed/NCBI View Article : Google Scholar

80 

Johanning GL, Malouf GG, Zheng X, Esteva FJ, Weinstein JN, Wang-Johanning F and Su X: Expression of human endogenous retrovirus-K is strongly associated with the basal-like breast cancer phenotype. Sci Rep. 7(41960)2017.PubMed/NCBI View Article : Google Scholar

81 

Kim HJ, Moon BI, Lee JW, Kim SC and Kim HJ: Age-related reduction of antibody response against the human endogenous retrovirus K envelope in women. Oncotarget. 7:17327–17337. 2016.PubMed/NCBI View Article : Google Scholar

82 

Zhou F, Krishnamurthy J, Wei Y, Li M, Hunt K, Johanning GL, Cooper LJ and Wang-Johanning F: Chimeric antigen receptor T cells targeting HERV-K inhibit breast cancer and its metastasis through downregulation of Ras. Oncoimmunology. 4(e1047582)2015.PubMed/NCBI View Article : Google Scholar

83 

Jin X, Xu XE, Jiang YZ, Liu YR, Sun W, Guo YJ, Ren YX, Zuo WJ, Hu X, Huang SL, et al: The endogenous retrovirus-derived long noncoding RNA TROJAN promotes triple-negative breast cancer progression via ZMYND8 degradation. Sci Adv. 5(eaat9820)2019.PubMed/NCBI View Article : Google Scholar

84 

Li N, Li Y, Lv J, Zheng X, Wen H, Shen H, Zhu G, Chen TY, Dhar SS, Kan PY, et al: ZMYND8 reads the dual histone mark H3K4me1-H3K14ac to antagonize the expression of metastasis-linked genes. Mol Cell. 63:470–484. 2016.PubMed/NCBI View Article : Google Scholar

85 

Büscher K, Hahn S, Hofmann M, Trefzer U, Ozel M, Sterry W, Löwer J, Löwer R, Kurth R and Denner J: Expression of the human endogenous retrovirus-K transmembrane envelope, Rec and Np9 proteins in melanomas and melanoma cell lines. Melanoma Res. 16:223–234. 2006.PubMed/NCBI View Article : Google Scholar

86 

Hahn S, Ugurel S, Hanschmann KM, Strobel H, Tondera C, Schadendorf D, Löwer J and Löwer R: Serological response to human endogenous retrovirus K in melanoma patients correlates with survival probability. AIDS Res Hum Retroviruses. 24:717–723. 2008.PubMed/NCBI View Article : Google Scholar

87 

Kudo-Saito C, Yura M, Yamamoto R and Kawakami Y: Induction of immunoregulatory CD271+ cells by metastatic tumor cells that express human endogenous retrovirus H. Cancer Res. 74:1361–1370. 2014.PubMed/NCBI View Article : Google Scholar

88 

Maliniemi P, Vincendeau M, Mayer J, Frank O, Hahtola S, Karenko L, Carlsson E, Mallet F, Seifarth W, Leib-Mösch C and Ranki A: Expression of human endogenous retrovirus-w including syncytin-1 in cutaneous T-cell lymphoma. PLoS One. 8(e76281)2013.PubMed/NCBI View Article : Google Scholar

89 

Blond JL, Lavillette D, Cheynet V, Bouton O, Oriol G, Chapel-Fernandes S, Mandrand B, Mallet F and Cosset FL: An envelope glycoprotein of the human endogenous retrovirus HERV-W is expressed in the human placenta and fuses cells expressing the type D mammalian retrovirus receptor. J Virol. 74:3321–3329. 2000.PubMed/NCBI View Article : Google Scholar

90 

Wallace TA, Downey RF, Seufert CJ, Schetter A, Dorsey TH, Johnson CA, Goldman R, Loffredo CA, Yan P, Sullivan FJ, et al: Elevated HERV-K mRNA expression in PBMC is associated with a prostate cancer diagnosis particularly in older men and smokers. Carcinogenesis. 35:2074–2083. 2014.PubMed/NCBI View Article : Google Scholar

91 

Reis BS, Jungbluth AA, Frosina D, Holz M, Ritter E, Nakayama E, Ishida T, Obata Y, Carver B, Scher H, et al: Prostate cancer progression correlates with increased humoral immune response to a human endogenous retrovirus GAG protein. Clin Cancer Res. 19:6112–6125. 2013.PubMed/NCBI View Article : Google Scholar

92 

Saini SK, Ørskov AD, Bjerregaard AM, Unnikrishnan A, Holmberg-Thydén S, Borch A, Jensen KV, Anande G, Bentzen AK, Marquard AM, et al: Human endogenous retroviruses form a reservoir of T cell targets in hematological cancers. Nat Commun. 11(5660)2020.PubMed/NCBI View Article : Google Scholar

93 

Alqahtani S, Promtong P, Oliver AW, He XT, Walker TD, Povey A, Hampson L and Hampson IN: Silver nanoparticles exhibit size-dependent differential toxicity and induce expression of syncytin-1 in FA-AML1 and MOLT-4 leukaemia cell lines. Mutagenesis. 31:695–702. 2016.PubMed/NCBI View Article : Google Scholar

94 

Sun Y, Ouyang DY, Pang W, Tu YQ, Li YY, Shen XM, Tam SC, Yang HY and Zheng YT: Expression of syncytin in leukemia and lymphoma cells. Leuk Res. 34:1195–1202. 2010.PubMed/NCBI View Article : Google Scholar

95 

Contreras-Galindo R, Kaplan MH, Leissner P, Verjat T, Ferlenghi I, Bagnoli F, Giusti F, Dosik MH, Hayes DF, Gitlin SD and Markovitz DM: Human endogenous retrovirus K (HML-2) elements in the plasma of people with lymphoma and breast cancer. J Virol. 82:9329–9336. 2008.PubMed/NCBI View Article : Google Scholar

96 

Tatkiewicz W, Dickie J, Bedford F, Jones A, Atkin M, Kiernan M, Maze EA, Agit B, Farnham G, Kanapin A and Belshaw R: Characterising a human endogenous retrovirus (HERV)-derived tumour-associated antigen: Enriched RNA-Seq analysis of HERV-K(HML-2) in mantle cell lymphoma cell lines. Mob DNA. 11(9)2020.PubMed/NCBI View Article : Google Scholar

97 

Morgan D and Brodsky I: Human endogenous retrovirus (HERV-K) particles in megakaryocytes cultured from essential thrombocythemia peripheral blood stem cells. Exp Hematol. 32:520–525. 2004.PubMed/NCBI View Article : Google Scholar

98 

Takahashi Y, Harashima N, Kajigaya S, Yokoyama H, Cherkasova E, McCoy JP, Hanada K, Mena O, Kurlander R, Tawab A, et al: Regression of human kidney cancer following allogeneic stem cell transplantation is associated with recognition of an HERV-E antigen by T cells. J Clin Invest. 118:1099–1109. 2008.PubMed/NCBI View Article : Google Scholar

99 

Rao S, Abdul T, Kurlander R, Harashima N, Lundqvist A, Hong J, Malinzak E, Smith A, Cherkasova E, McCoy P, et al: The human endogenous retrovirus (HERV) derived kidney cancer antigen CT-RCC1 induces proliferation of CD8+ antigen-specific T-cells in vitro that kill renal cell carcinoma (RCC) and is up-regulated by inhibiting histone deacetylase. Cancer Res. 68 (9 Suppl)(S1033)2008.

100 

Haruta M, Gray WM and Sussman MR: Regulation of the plasma membrane proton pump (H(+)-ATPase) by phosphorylation. Curr Opin Plant Biol. 28:68–75. 2015.PubMed/NCBI View Article : Google Scholar

101 

Weyerer V, Strissel PL, Stöhr C, Eckstein M, Wach S, Taubert H, Brandl L, Geppert CI, Wullich B, Cynis H, et al: Endogenous retroviral-K envelope is a novel tumor antigen and prognostic indicator of renal cell carcinoma. Front Oncol. 11(657187)2021.PubMed/NCBI View Article : Google Scholar

102 

Gabaev I, Williamson JC, Crozier TWM, Schulz TF and Lehner PJ: Quantitative proteomics analysis of lytic KSHV infection in human endothelial cells reveals targets of viral immune modulation. Cell Rep. 33(108249)2020.PubMed/NCBI View Article : Google Scholar

103 

Chiappinelli KB, Strissel PL, Desrichard A, Li H, Henke C, Akman B, Hein A, Rote NS, Cope LM, Snyder A, et al: Inhibiting DNA methylation causes an interferon response in cancer via dsRNA including endogenous retroviruses. Cell. 162:974–986. 2015.PubMed/NCBI View Article : Google Scholar

104 

Larsen JM, Christensen IJ, Nielsen HJ, Hansen U, Bjerregaard B, Talts JF and Larsson LI: Syncytin immunoreactivity in colorectal cancer: Potential prognostic impact. Cancer Lett. 280:44–49. 2009.PubMed/NCBI View Article : Google Scholar

105 

Golkaram M, Salmans ML, Kaplan S, Vijayaraghavan R, Martins M, Khan N, Garbutt C, Wise A, Yao J, Casimiro S, et al: HERVs establish a distinct molecular subtype in stage II/III colorectal cancer with poor outcome. NPJ Genom Med. 6(13)2021.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

October-2022
Volume 17 Issue 4

Print ISSN: 2049-9450
Online ISSN:2049-9469

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Sahu S, Singh B and Rai AK: Human endogenous retrovirus regulates the initiation and progression of cancers (Review). Mol Clin Oncol 17: 143, 2022
APA
Sahu, S., Singh, B., & Rai, A.K. (2022). Human endogenous retrovirus regulates the initiation and progression of cancers (Review). Molecular and Clinical Oncology, 17, 143. https://doi.org/10.3892/mco.2022.2576
MLA
Sahu, S., Singh, B., Rai, A. K."Human endogenous retrovirus regulates the initiation and progression of cancers (Review)". Molecular and Clinical Oncology 17.4 (2022): 143.
Chicago
Sahu, S., Singh, B., Rai, A. K."Human endogenous retrovirus regulates the initiation and progression of cancers (Review)". Molecular and Clinical Oncology 17, no. 4 (2022): 143. https://doi.org/10.3892/mco.2022.2576