Skip to main content

The expression level of chicken telomerase reverse transcriptase in tumors induced by ALV-J is positively correlated with methylation and mutation of its promoter region

Abstract

Avian leukosis virus subgroup J (ALV-J) can cause neoplastic diseases in poultry and is still widely prevalent in China. Chicken telomerase reverse transcriptase (chTERT) is the core component of telomerase, which is closely related to the occurrence and development of tumors. Our previous studies showed that chTERT is overexpressed in ALV-J tumors, but the mechanism is still not completely clear. Therefore, this study aims to analyze the possible molecular mechanism of chTERT overexpression in ALV-J tumors from the perspective of DNA methylation and promoter mutation. Methylation sequencing of the chTERT amplicon showed that ALV-J replication promoted the methylation level of the chTERT promoter. And the methylation level of the chTERT promoter in ALV-J tumors was significantly higher than that in tumor-adjacent and normal tissues. Compared with the tumor-adjacent and normal tissues, the chTERT promoter in each ALV-J tumors tested had a mutation of −183 bp C > T, and 36.0% (9/25) of the tumors also had mutations of −184 bp T > C, −73 bp::GGCCC and −56 bp A > T in the chTERT promoter, which formed the binding sites for the transcription factors NFAT5, TFAP2A and ZEB1, respectively. The results of RT–qPCR and Western blotting showed that the occurrence of these mutations significantly increased the expression level of chTERT. In conclusion, this study demonstrated that the high expression of chTERT in ALV-J tumors is positively correlated with the level of hypermethylation and mutation in its promoter, which provides a new perspective for further research on the molecular mechanism of chTERT in ALV-J tumorigenesis.

Introduction

Avian leukosis (AL) is caused by avian leukosis virus (ALV), which can induce a variety of neoplastic diseases in poultry. The disease has various clinical manifestations, including myeloma, lymphoma, hemangioma, and fibrosarcoma [1]. It is a kind of provenance disease that seriously harms the poultry industry worldwide [2]. Since the 1990s, AL has caused significant economic losses to China’s chicken industry, especially avian leukosis virus subgroup J (ALV-J) [3]. It seriously harms the healthy development of white-feathered broilers, egg-type broilers, yellow-feathered broilers and local broilers and is still widely prevalent in China [4].

Telomerase reverse transcriptase (TERT), as the core component of telomerase regulation, is closely related to the development of cancer and cell growth [5]. Significantly elevated expression levels of human TERT (hTERT) have been found in most human malignancies, such as sarcomas [6], brain tumors [7], colorectal tumors [8], and breast cancer [9], increasing the risk of tumor recurrence. Studies have shown that mutations and methylation in the hTERT promoter region play a crucial role in the regulation of hTERT transcription levels and telomerase activity [10,11,12,13]. Preliminary studies by our group have shown that the chicken TERT (chTERT) gene is one of the main integration sites for ALV-J provirus to insert into the host genome, and it is significantly overexpressed in ALV-J tumors [14]. However, the reasons for its high expression are still not completely clear.

5'-Methylcytosine (5mC) is the main form of DNA methylation and one of the earliest and most thoroughly studied epigenetic regulatory mechanisms, playing an important role in cancer, gene expression, aging, atherosclerosis, Alzheimer’s disease and other diseases [15,16,17]. Studies have shown that the increased expression of hTERT in colorectal cancer and gastric cancer was associated with the degree of hypermethylation of the hTERT gene, which seriously affected recurrence after treatment [18, 19]. In patients with liver cancer, the methylation level of the hTERT promoter in cancer tissues was significantly higher than that in tumor-adjacent tissues, and the expression level of the hTERT gene was also increased by tens of times [20]. However, it has been reported that when CpG islands with low methylation overlie the promoter region of the gene, telomerase activity is inhibited, which is inconsistent with a previous study showing that methylation in the promoter region of the hTERT gene promoted its gene expression level [11, 21]. In conclusion, the mechanisms by which methylation in the TERT promoter region regulates gene expression in different tumor diseases are not entirely the same and may be methylation-dependent or methylation-independent, requiring precise and targeted studies. The 5' region of the chTERT gene and its promoter region are part of a large CpG island of approximately 4 kb, suggesting that chTERT expression may be regulated by methylation [22]. Therefore, we were inspired to actively explore whether there are significant differences in the methylation levels of the chTERT gene and its promoter region in tumors induced by ALV-J compared with those in tumor-adjacent tissues or normal tissues, as well as the methylation effects on chTERT gene expression and telomerase activity.

The formation of many tumors is inseparable from the occurrence of genetic mutations, but previous studies have shown that genetic mutations mostly occur in coding regions. However, a study found that there was a high frequency of mutations in the hTERT promoter region in melanoma, reaching approximately 70.0%, and the mutation sites were concentrated in −57 bp T > G, −124 bp G > A (C228T) and −146 bp G > A (C250T). After the mutation, the sequence of GGAA/T or CCGGAA/T was obtained, forming a new transcriptional binding site for the Ets ternary complex factor, thereby actively participating in the regulation of hTERT expression and promoting the occurrence and development of tumors [23, 24]. Subsequently, two popular mutation sites, C228T and C250T, were also found in adrenal tumors, which can promote the expression of hTERT and increase the activity of telomerase, providing a suitable environment for tumor growth [25]. In addition, scientists have successively found mutations in the hTERT promoter region in thyroid cancer [26], liposarcoma [27], hepatocellular carcinoma [28], urothelial cancer and other tumors [29, 30], but the mutation sites and mutation frequencies were not the same. Since the first discovery of TERT promoter mutations in melanoma, an increasing number of studies have shown that TERT promoter mutations play an important role in tumorigenesis [31, 32]. However, most of the current studies on TERT promoter mutations focus on human tumor diseases, and there are few studies on chicken-related tumor diseases. Therefore, it is of great significance to identify the mutation of the chTERT promoter region and its regulatory effect on gene expression in ALV-J-induced tumors in a timely manner.

In this study, DF-1 cells and LMH cells were used as in vitro models. The methylation and mutation characteristics of the chTERT promoter region in chicken tumor cells and normal cells were determined by methylation sequencing and Sanger sequencing, and the effect of ALV-J replication on the chTERT promoter methylation level was analyzed. ALV-J tumor tissues and tumor-adjacent and normal tissues were used as research objects to investigate the methylation and mutation characteristics of the chTERT promoter region in ALV-J tumors. Moreover, the influence of these differences on chTERT expression was analyzed statistically. It is expected that this study can preliminarily answer the scientific question of how the high expression of chTERT was formed in ALV-J tumors and provide a useful reference and theoretical support for the research of other avian tumor diseases.

Materials and methods

Cells, virus, ALV-J tumors and antibodies

The avian leghorn male hepatoma cell line LMH and chicken fibroblast cell line DF-1 were maintained in our laboratory and cultured in DMEM/F12 or DMEM (Gibco, Thermo Fisher Scientific, Inc., Grand Island, NY, USA) supplemented with 10% fetal bovine serum (FBS) and maintained at 37 °C with 5% CO2. The ALV-J Hc1 strain was maintained in our laboratory. Twenty-five tumor tissue samples induced by chronic transformed ALV-J and their corresponding tumor-adjacent and normal tissues were confirmed and collected from our previous ALV-J artificial tumorigenicity experiment and clinical tumor cases in large-scale breeding poultry farms (Additional file 1). The chicken single factor serum of chTERT was prepared and preserved by our laboratory in the early stage. The anti-GAPDH primary monoclonal antibody was purchased from Abcam, Inc. (Cambridge, UK). IRDye® 800CW goat anti-rabbit IgG and donkey anti-chicken secondary antibodies were purchased from LI-COR Biosciences, Ltd. (Nebraska, USA).

Prediction of chTERT promoter methylation

We referred to the related sequences of the chTERT promoter region and coding region on NCBI (GenBank: EU650197.1, NM.001031007) by using the online software MethPrimer to analyze the possible methylation CpG sites in the chTERT promoter region. The results (Figure 1) showed that there were 2 CpG islands from 600 bp upstream to 800 bp downstream of ATG of the initiation codon, which contained more than 110 CpG sites. Therefore, in this study, this sequence was used as a target gene for DNA methylation analysis in the chTERT promoter region, and the position of the amplicon relative to the ATG initiation codon in the chTERT coding region was −627 bp to + 873 bp.

Figure 1
figure 1

Prediction of DNA methylation in the chTERT promoter region.

Target gene bisulfite sequencing

DNA was extracted from cells or tissue samples according to the instructions of the DNA extraction kit (Omega, Norcross, GA, USA). Target gene bisulfite sequencing was conducted by E-Gene Co., Ltd. (Shenzhen, China). Briefly, primers (Table 1) were designed to recognize regions without CpG sites to avoid amplification bias of methylated versus unmethylated sequences. Bisulfite sequencing PCR (BSP) validation experiments were conducted as follows: 500 ng of genomic DNA was converted using a ZYMO EZ DNA Methylation-Gold Kit™ (Zymo Research, California, USA) according to the manufacturer’s instructions. After purification of the converted products, PCR amplification was carried out in a final reaction volume of 50 µL consisting of 3 µL purified conversion fractions, 4 µL 2.5 mM dNTP, 5 µL 10× buffer, 1 µL BSP primers, 0.5 µL JumpStart™ Taq DNA Polymerase (Sigma-Aldrich, St. Louis, Missouri, USA) and 36.5 µL water and the following thermal cycling program was 94 °C 1 min, 30 cycles of 94 °C 10 s, 58 °C 30 s, 72 °C 30 s then extension of 5 min at 72 °C and products were held at 12 °C. After amplification, the PCR products were further used for library construction, and the final libraries were quantified by an Agilent 2100 Bioanalyzer (Agilent Technologies, California, USA) and real-time PCR assay and then sequenced by Illumina HiSeq.

Table 1 BSP Primer sequences for methylation analysis of the chTERT amplicon

PCR amplification and Sanger sequencing of the chTERT promoter region

PCR was used to amplify an approximately 1000 bp fragment at the end of the chTERT promoter region (approximately 1000 bp upstream of ATG) to analyze its mutation characteristics. The primer sequences (5’-GTTGGTGGTATGGCAGTA-3′, 5′-TCCTCCCGCGCTACATTG-3′) were used for amplification, and the annealing temperature was 57 °C. Phanta® Max DNA Polymerase (Vazyme, Nanjing, China) was used for amplification. The products were detected by 1% agarose gel electrophoresis in 1 × TAE with EB staining, and the PCR product was recovered by a Gel Extraction Kit (Omega) and cloned into the pMD-18 T vector according to the manufacturer’s instructions (Takara, Tokyo, Japan). After that, recombinants were transformed into E. coli DH5α competent cells (Takara), and monoclonal clones were selected on LB plates containing ampicillin. DNA was extracted from positive clones using a Plasmid Mini Kit (Omega) and sent to Sangon Biotech (Shanghai, China) for Sanger sequencing.

Real-time fluorescence quantitative PCR (RT–qPCR)

The tissue samples were homogenized by a cryogenic grinder, and then the total RNA was extracted from tissues with TRIzol reagent (Fastagen Biotech, Shanghai, China) according to the manufacturer’s recommendations. cDNA was synthesized from the total RNA template with random primers using a PrimeScript RT Reagent kit (TaKaRa). The RT–qPCR was performed using Hieff® qPCR SYBR Green Master Mix (YEASEN, Shanghai, China) on a CFX96TM Real-time PCR System (Bio–Rad, California, USA). Expression levels were quantified using the 2−ΔΔCt method and normalized to GAPDH expression. The sequences of the primers used were the same as those used in our previous study [33].

Western blotting

The tissue samples were homogenized at low temperature, after which NP40 lysis buffer (Beyotime, Shanghai, China) was used to extract protein. The concentration of extracted protein was determined by the BCA Protein Assay Kit (Beyotime). Subsequently, equal amounts of total protein were separated by SDS–polyacrylamide gel electrophoresis (SDS–PAGE) (Beyotime) and then transferred onto a nitrocellulose membrane. The membranes were immunoblotted with primary antibodies at 4 °C overnight followed by a corresponding secondary antibody at 37 °C for 1 h. Finally, the blots were scanned using an Odyssey Infrared Imaging System (LI-COR, Nebraska, USA).

Statistical analysis

All the results are presented as the means ± standard deviations. Statistical analysis was performed by Student’s t test using GraphPad Prism software, and a P value of < 0.05 was considered significant.

Results

The methylation level of the chTERT promoter region in LMH cells is higher than that in DF-1 cells

LMH and DF-1 cells were used as in vitro models of chicken tumor cells and healthy cells, respectively, to analyze the difference between them in methylation levels of the chTERT promoter region. The results showed that the methylation level of the chTERT promoter region in LMH cells was higher than that in DF-1 cells (Figure 2). Our previous study showed that telomerase activity was positive in LMH cells and negative in DF-1 cells [33]; that is, chTERT was not expressed in DF-1 cells. In conclusion, the expression of chTERT in LMH cells is related to the hypermethylation level of the promoter region.

Figure 2
figure 2

Comparison of chTERT promoter methylation levels in LMH and DF-1 cells.

Replication of ALV-J promotes methylation of the chTERT promoter region

LMH cells and DF-1 cells were infected with ALV-J at 1.0 multiplicity of infection (MOI) and maintained for 7 days, and the cell DNA was extracted for chTERT amplicon methylation sequencing. The results showed that ALV-J replication promoted the methylation of the chTERT promoter region in both DF-1 cells (Figure 3) and LMH cells (Figure 4), and the thermal map of the methylation level is shown in Figure. 5, the corresponding methylation matrix data are shown in Additional file 2. Our previous study showed that the replication of ALV-J upregulated the expression level of chTERT [33]. In conclusion, the expression level of chTERT is positively correlated with the methylation level of its promoter region.

Figure 3
figure 3

Replication of ALV-J promotes methylation of the chTERT promoter region in DF-1 cells.

Figure 4
figure 4

Replication of ALV-J promotes methylation of the chTERT promoter region in LMH cells.

Figure 5
figure 5

Heatmap of the DNA methylation level of the chTERT amplicon in LMH cells and DF-1 cells.

The high expression of chTERT in ALV-J tumors correlates with the high methylation level of its promoter region

Some of the ALV-J tumor tissues were selected for methylation sequencing of the chTERT promoter region. The results showed that the methylation level of the chTERT promoter region in ALV-J tumor tissue was higher than that in tumor-adjacent tissues and normal tissues (Figure 6), suggesting that the high expression of chTERT in ALV-J tumor tissues (Figure 7) was related to the high methylation level of its promoter region; the heatmap of its methylation level is shown in Figure 8, the corresponding methylation matrix data are shown in Additional file 3.

Figure 6
figure 6

The DNA methylation level change curve of the chTERT amplicon in ALV-J tumors and tumor-adjacent and normal tissues. T: tumor tissues; TA: tumor-adjacent tissues; N: normal tissues.

Figure 7
figure 7

chTERT was significantly highly expressed in ALV-J tumor tissues. Analysis of telomerase activity (A) and chTERT protein expression levels (B) in tumor tissue, tumor-adjacent and normal tissues.

Figure 8
figure 8

Heatmap of the DNA methylation level of the chTERT amplicon in ALV-J tumors and tumor-adjacent and normal tissues. T: tumor tissues; TA: tumor-adjacent tissues; N: normal tissues.

Mutation of the chTERT promoter region promotes its methylation and forms binding sites for the transcription factors ZEB1, TFAP2A and NFAT5

The sequence of approximately 1000 bp at the end of the chTERT promoter region was amplified by PCR (Additional file 4), and Sanger sequencing was performed. Then, the sequences were compared to find a unified rule. The results showed (Additional file 5) that there were no uniform mutations in the chTERT promoter region in tumor-adjacent tissues compared with normal tissues. However, compared with tumor-adjacent or normal tissues, the −183 bp of the chTERT promoter region in all ALV-J tumor tissues was mutated from T to C (−183 bp C > T). In addition, nine tumor tissues (n = 25) were further mutated; that is, the chTERT promoter at the −184 bp site was mutated from C to T (−184 bp T > C), and the −56 bp was mutated from T to A (−56 bp A > T). Moreover, after −73 bp in the chTERT promoter region of this part of the tumor tissue, there was the insertion of 5 bases GGCCC (−73 bp::GGCCC). In this study, tumor tissues with further mutations in this region were called Mutant, and those without such mutations were called Wild type.

Further analysis showed that the mutation of −183 bp C > T in ALV-J tumor tissue makes this site a CpG site that may be methylated. As seen from Figure 9, the methylation level of −183 bp in the chTERT promoter region of some tumor tissue samples (such as Kidney_T1, Liver_T2, Liver_T3, and Mesentery_T) is indeed higher than that of the tumor-adjacent and normal tissues. This finding indirectly suggests that mutations at this site may promote chTERT expression through methylation. On this basis, the sequences before and after the mutation were imported into the JASPAR database for analysis, and it was found that the mutation or insertion of −184 bp T > C, −73 bp::GGCCC and −56 bp A > T forms the binding sites of transcription factors of nuclear factor of activated T cells 5 (NFAT5), transcription factor AP-2a (TFAP2A) and E box zinc finger E-box binding homeobox 1, ZEB1), respectively.

Figure 9
figure 9

Schematic diagram of mutations in the chTERT promoter region in ALV-J tumors relative to its tumor-adjacent and normal tissues.

The high expression of chTERT in ALV-J tumors correlates with its promoter mutation

To understand how the mutation of the chTERT promoter region in ALV-J tumor tissues affects the expression of chTERT, the expression of chTERT in wild-type and mutant tumors was analyzed by Western blot and RT–qPCR. The results showed that the mRNA and protein expression levels of chTERT in mutant tumor tissues were significantly higher than those in wild-type tissues (Figure 10, p < 0.01), suggesting that the high expression of chTERT in ALV-J tumors was also significantly related to the mutation of its promoter region.

Figure 10
figure 10

The expression levels of chTERT mRNA (A) and protein (B) in tumor tissues with the wild-type or mutant chTERT promoter.

Discussion

Studies have shown that transcriptional reactivation of TERT is associated with methylation and mutation of its promoter region in a variety of human tumor diseases [34,35,36,37]. However, there is no relevant research demonstrating the difference in the methylation level of chTERT in the promoter region of ALV-J-induced tumors, nor whether there are mutations in the promoter region or the characteristics of the mutations. Therefore, this study focused on the chTERT promoter region to analyze the methylation and expression of this promoter region in the LMH tumor cell line and DF-1 normal cell line, as well as in ALV-J tumor tissues, tumor-adjacent tissues and normal tissues, and to explore the effects of these differences and characteristics on chTERT expression.

In this study, the differences in chTERT promoter methylation levels between LMH cells and DF-1 cells were analyzed in vitro, and the results showed that the methylation levels of the chTERT promoter in telomerase-positive LMH cells were significantly higher than those in telomerase-negative DF-1 cells, but the exact reason for this is still unknown. After reviewing the literature, we speculated that this might be related to the different immortalization mechanism of the two cells. DF-1 cells are telomerase-negative immortalized cell lines, which are not cancer cells in the strict sense, and their immortal proliferation does not depend on the expression of chTERT, but on another mechanism, namely the alternative lengthening of telomeres [38]. LMH cells are telomerase-positive chicken tumor cell lines, and their immortal proliferation depends on the expression of telomerase or chTERT. In order to obtain continuous and stable expression of telomerase activity, the chTERT promoter has a high level of methylation. These results suggested that the reactivation of telomerase in LMH cells was related to the high methylation level of the chTERT promoter region.

To understand the effect of ALV-J replication on chTERT promoter methylation, ALV-J was inoculated with 1.0 MOI on DF-1 and LMH cells. The results showed that after ALV-J inoculation, the methylation level of the chTERT promoter region consistently showed an increasing trend. Our previous research showed that replication of ALV-J upregulated the expression of chTERT and telomerase activity, indicating that methylation in the chTERT promoter region could promote its expression. This was further confirmed by comparing the methylation levels of the chTERT promoter region in ALV-J tumor tissues, tumor-adjacent tissues and normal tissues. That is, the methylation level of the chTERT promoter region in tumor tissues was higher than that in tumor-adjacent and normal tissues, which was positively correlated with the expression of chTERT. This is consistent with the conclusions of studies on human small-cell lung cancer, thyroid cancer, hepatocellular carcinoma and other cancer diseases [20, 39, 40]. Imperfectly, allele specific methylation of chTERT promoter was not analyzed in this study. Indeed, this is because this research was not designed in a more comprehensive way. Secondly, due to the large number of chickens, which are economic animals, the sources of tumor samples are complicated, it is less feasible to trace their genetic information and lack of database support. Therefore, if based on this research to supplement the experimental analysis of the allele specific methylation, its feasibility is worthy to be considered, but it can be further explored and analyzed in detail in our subsequent new research. Allele specific methylation analysis is interesting, but we believe that the lack of this data may not affect the main thrust of this study.

Previous studies have shown that when the insertion of chronically transformed ALV-J is integrated near the proto-oncogene of the host gene, its insertion integration site is usually located ± 2500 bp away from the transcription start site [14]. Therefore, to avoid the influence of ALV-J insertion and integration as much as possible and to analyze the mutation of the methylated region in the chTERT promoter region and for the convenience of the experiment, PCR amplification was only performed on the gene sequence 1000 bp upstream of ATG at the end of the promoter region to analyze the mutation characteristics. The results showed that there was no difference in the promoter sequence between tumor-adjacent tissues and normal tissues. Compared with tumor-adjacent and normal tissues, ALV-J tumors have more significant mutations in the promoter sequence of this segment, and all have the mutation of −183 bp C > T. On this basis, some tumor tissues also have the mutation of −183 bp C > T, 184 bp T > C, −73 bp::GGCCC and −56 bp A > T; these mutation sites are not identical to the mutation hotspots of C228T and C250T in the hTERT promoter region in human tumor diseases [25, 41, 42] because in different tumor diseases, the mutation frequency and mutation sites of TERT are not completely consistent. On the other hand, this study also attempted to compare the mutation of the chTERT promoter region in LMH cells and DF-1 cells, but no uniform mutation pattern was found after comparison, which was not consistent with the mutation characteristics of chTERT in ALV-J tumors or tumor-adjacent and normal tissues. However, this was not difficult to understand, because even the corresponding cells in vitro cannot completely simulate the pathological model at the tissue level in vivo, there must be differences.

Combined with the results of methylation sequencing, it was found that the mutation of −183 bp C > T makes this site a CpG site that may be methylated. It has also been proven that methylation at this site in the chTERT promoter region has indeed occurred in some tumor tissues. On this basis, the JASPAR database was used to analyze the differences caused by the sequence differences before and after the mutation. The results showed that the mutations of −184 bp T > C, −73 bp::GGCCC and −56 bp A > T formed transcriptional binding sites that could be bound by the transcription factors NFAT5, TFAP2A and ZEB1, respectively. Western blot and RT–qPCR analyses showed that the expression of chTERT in mutant tumor tissues was significantly higher than that in wild-type tumor tissues. This is because the mutation of −183 bp C > T promotes an increase in the methylation level, thereby upregulating the expression of chTERT. On the other hand, it is presumed to be regulated by the transcription factors NFAT5, TFAP2A and ZEB1.

After reviewing the literature, it was determined that NFAT5 has similar functions to TERT and plays an important role in various activities, such as immune regulation, metabolic regulation, DNA damage repair and tumorigenesis [43,44,45], such as non-small-cell lung cancer [46], pancreatic cancer [47], chronic lymphocytic leukemia [48] and melanoma [49]. It plays an important role in the occurrence and development of various human tumor diseases, and studies have shown that NFAT5 can regulate the transcription of the mouse TERT gene and promote the expression of TERT [50]. TFAP2A is closely related to the process of regulating the cell cycle, epithelial-mesenchymal transition and apoptosis and can participate in regulating the proliferation and migration of cervical cancer [51], ovarian cancer [52] and other tumor cells and promote the occurrence and development of tumors. As a transcriptional regulator containing multiple functional domains, ZEB1 can participate in the regulation of the expression of various oncogenes, including hTERT; for example, ZEB1 can promote the occurrence and development of breast cancer by upregulating the expression of hTERT [53]. In colorectal cancer, the hTERT/ZEB1 complex directly regulates E-cadherin to promote EMT [54]. This study speculates that one of the main reasons why the expression of chTERT in mutant tumor tissues is significantly higher than that in wild-type tumor tissues is that NFAT5, TFAP2A and ZEB1 are actively involved in regulating the expression of chTERT, but their interaction with chTERT and the regulatory mechanism remain to be further elucidated, which also provides new scientific questions and research directions for future generations to further study the mechanism of chTERT in ALV-J-induced tumorigenesis.

In summary, this study demonstrated that the high expression of chTERT in ALV-J tumors was positively correlated with the hypermethylation and mutation level of its promoter region, and we analyzed the molecular mechanism by which chTERT promotes the tumorigenicity of ALV-J from the perspective of DNA methylation and promoter mutation. This study provides a new perspective for further research on the role and molecular mechanism of chTERT in tumorigenicity by ALV.

Abbreviations

chTERT:

chicken telomerase reverse transcriptase

hTERT:

human telomerase reverse transcriptase

AL:

avian leucosis

ALV:

avian leukosis viruses

ALV-J:

avian leukosis virus subgroup J

TRAP:

telomeric repeat amplification protocol

MOI:

multiplicity of infection

RT–qPCR:

real-time quantitative PCR

5mC:

5'-Methylcytosine

FBS:

fetal bovine serum

BSP:

bisulfite sequencing PCR

NFAT5:

nuclear factor of activated T cells 5

TFAP2A:

transcription factor AP-2a

ZEB1:

E box zinc finger E-box binding homeobox 1

References

  1. Xu M, Hang F, Qian K, Shao H, Ye J, Qin A (2022) Chicken hepatomegaly and splenomegaly associated with novel subgroup J avian leukosis virus infection. BMC Vet Res 18:32. https://doi.org/10.1186/s12917-022-03139-1

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Freick M, Schreiter R, Weber J, Vahlenkamp TW, Heenemann K (2022) Avian leukosis virus (ALV) is highly prevalent in fancy-chicken flocks in Saxony. Arch Virol 167:1169–1174. https://doi.org/10.1007/s00705-022-05404-y

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Deng Q, Li M, He C, Lu Q, Gao Y, Li Q, Shi M, Wang P, Wei P (2021) Genetic diversity of avian leukosis virus subgroup J (ALV-J): toward a unified phylogenetic classification and nomenclature system. Virus Evol 7:veab037. https://doi.org/10.1093/ve/veab037

    Article  PubMed  PubMed Central  Google Scholar 

  4. Liu P, Li L, Jiang Z, Yu Y, Chen X, Xiang Y, Chen J, Li Y, Cao W (2021) Molecular characteristics of subgroup J avian leukosis virus isolated from yellow breeder chickens in Guangdong, China, during 2016–2019. Infect Genet Evol 89:104721. https://doi.org/10.1016/j.meegid.2021.104721

    Article  CAS  PubMed  Google Scholar 

  5. Kwon JH, Yi JW (2021) Correlation between telomerase reverse transcriptase messenger RNA expression and survival of patients with papillary thyroid carcinoma. Surgery 169:43–49. https://doi.org/10.1016/j.surg.2020.04.054

    Article  PubMed  Google Scholar 

  6. Koelsche C, Renner M, Hartmann W, Brandt R, Lehner B, Waldburger N, Alldinger I, Schmitt T, Egerer G, Penzel R, Wardelmann E, Schirmacher P, von Deimling A, Mechtersheimer G (2014) TERT promoter hotspot mutations are recurrent in myxoid liposarcomas but rare in other soft tissue sarcoma entities. J Exp Clin Cancer Res 33:33. https://doi.org/10.1186/1756-9966-33-33

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Li X, Qian X, Wang B, Xia Y, Zheng Y, Du L, Xu D, Xing D, DePinho RA, Lu Z (2020) Programmable base editing of mutated TERT promoter inhibits brain tumour growth. Nat Cell Biol 22:282–288. https://doi.org/10.1038/s41556-020-0471-6

    Article  CAS  PubMed  Google Scholar 

  8. Jung SJ, Park JH, Hwang I, Lee JH (2020) Different TERT expression between colorectal adenoma and serrated polyp. Medicina (Kaunas) 56:463. https://doi.org/10.3390/medicina56090463

    Article  PubMed Central  Google Scholar 

  9. Wazir U, Orakzai M, Martin TA, Jiang WG, Mokbel K (2019) Correlation of TERT and stem cell markers in the context of human breast cancer. Cancer Genomics Proteomics 16:121–127. https://doi.org/10.21873/cgp.20117

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Salgado C, Roelse C, Nell R, Gruis N, van Doorn R, van der Velden P (2020) Interplay between TERT promoter mutations and methylation culminates in chromatin accessibility and TERT expression. PLoS One 15:e0231418. https://doi.org/10.1371/journal.pone.0231418

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Fotouhi O, Ghaderi M, Wang N, Zedenius J, Kjellman M, Xu D, Juhlin CC, Larsson C (2019) Telomerase activation in small intestinal neuroendocrine tumours is associated with aberrant TERT promoter methylation, but not hot-spot mutations. Epigenetics 14:1224–1233. https://doi.org/10.1080/15592294.2019.1634987

    Article  PubMed  PubMed Central  Google Scholar 

  12. Lee DD, Komosa M, Sudhaman S, Leao R, Zhang CH, Apolonio JD, Hermanns T, Wild PJ, Klocker H, Nassiri F, Zadeh G, Diplas BH, Yan H, Gallinger S, Pugh TJ, Ramaswamy V, Taylor MD, Castelo-Branco P, Nunes NM, Tabori U (2021) Dual role of allele-specific DNA hypermethylation within the TERT promoter in cancer. J Clin Invest 131:e146915. https://doi.org/10.1172/JCI146915

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Rowland TJ, Bonham AJ, Cech TR (2020) Allele-specific proximal promoter hypomethylation of the telomerase reverse transcriptase gene (TERT) associates with TERT expression in multiple cancers. Mol Oncol 14:2358–2374. https://doi.org/10.1002/1878-0261.12786

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Li Y, Liu X, Yang Z, Xu C, Liu D, Qin J, Dai M, Hao J, Feng M, Huang X, Tan L, Cao W, Liao M (2014) The MYC, TERT, and ZIC1 genes are common targets of viral integration and transcriptional deregulation in avian leukosis virus subgroup J-induced myeloid leukosis. J Virol 88:3182–3191. https://doi.org/10.1128/JVI.02995-13

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Rygiel CA, Goodrich JM, Solano-Gonzalez M, Mercado-Garcia A, Hu H, Tellez-Rojo MM, Peterson KE, Dolinoy DC (2021) Prenatal Lead (Pb) exposure and peripheral blood DNA methylation (5mC) and hydroxymethylation (5hmC) in Mexican adolescents from the ELEMENT birth cohort. Environ Health Perspect 129:67002. https://doi.org/10.1289/EHP8507

    Article  CAS  PubMed  Google Scholar 

  16. Mo Z, Cao Z, Luo S, Chen Y, Zhang S (2020) Novel molecular subtypes associated with 5mC methylation and their role in hepatocellular carcinoma immunotherapy. Front Mol Biosci 7:562441. https://doi.org/10.3389/fmolb.2020.562441

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Esopi D, Graham MK, Brosnan-Cashman JA, Meyers J, Vaghasia A, Gupta A, Kumar B, Haffner MC, Heaphy CM, De Marzo AM, Meeker AK, Nelson WG, Wheelan SJ, Yegnasubramanian S (2020) Pervasive promoter hypermethylation of silenced TERT alleles in human cancers. Cell Oncol (Dordr) 43:847–861. https://doi.org/10.1007/s13402-020-00531-7

    Article  CAS  PubMed Central  Google Scholar 

  18. Nomoto K, Maekawa M, Sugano K, Ushiama M, Fukayama N, Fujita S, Kakizoe T (2002) Methylation status and expression of human telomerase reverse transcriptase mRNA in relation to hypermethylation of the p16 gene in colorectal cancers as analyzed by bisulfite PCR-SSCP. Jpn J Clin Oncol 32:3–8. https://doi.org/10.1093/jjco/hyf001

    Article  PubMed  Google Scholar 

  19. Liu L, Liu C, Fotouhi O, Fan Y, Wang K, Xia C, Shi B, Zhang G, Wang K, Kong F, Larsson C, Hu S, Xu D (2017) TERT promoter hypermethylation in gastrointestinal cancer: a potential stool biomarker. Oncologist 22:1178–1188. https://doi.org/10.1634/theoncologist.2017-0064

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Yu J, Yuan X, Sjoholm L, Liu T, Kong F, Ekstrom TJ, Bjorkholm M, Xu D (2018) Telomerase reverse transcriptase regulates DNMT3B expression/aberrant DNA methylation phenotype and AKT activation in hepatocellular carcinoma. Cancer Lett 434:33–41. https://doi.org/10.1016/j.canlet.2018.07.013

    Article  CAS  PubMed  Google Scholar 

  21. Yuan T, Zhao W, Niu Y, Fu Y, Lu L, Niu D (2019) Exploration of the temporal-spatial expression pattern and DNA methylation-related regulation of the duck telomerase reverse transcriptase gene. Poult Sci 98:3257–3267. https://doi.org/10.3382/ps/pez240

    Article  CAS  PubMed  Google Scholar 

  22. Lam G, Beemon K (2018) ALV integration-associated hypomethylation at the TERT promoter locus. Viruses 10:74. https://doi.org/10.3390/v10020074

    Article  CAS  PubMed Central  Google Scholar 

  23. Horn S, Figl A, Rachakonda PS, Fischer C, Sucker A, Gast A, Kadel S, Moll I, Nagore E, Hemminki K, Schadendorf D, Kumar R (2013) TERT promoter mutations in familial and sporadic melanoma. Science 339:959–961. https://doi.org/10.1126/science.1230062

    Article  CAS  PubMed  Google Scholar 

  24. Huang FW, Hodis E, Xu MJ, Kryukov GV, Chin L, Garraway LA (2013) Highly recurrent TERT promoter mutations in human melanoma. Science 339:957–959. https://doi.org/10.1126/science.1229259

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Liu T, Brown TC, Juhlin CC, Andreasson A, Wang N, Backdahl M, Healy JM, Prasad ML, Korah R, Carling T, Xu D, Larsson C (2014) The activating TERT promoter mutation C228T is recurrent in subsets of adrenal tumors. Endocr Relat Cancer 21:427–434. https://doi.org/10.1530/ERC-14-0016

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Stenman A, Hysek M, Jatta K, Branstrom R, Darai-Ramqvist E, Paulsson JO, Wang N, Larsson C, Zedenius J, Juhlin CC (2019) TERT promoter mutation spatial heterogeneity in a metastatic follicular thyroid carcinoma: implications for clinical work-up. Endocr Pathol 30:246–248. https://doi.org/10.1007/s12022-019-09580-7

    Article  CAS  PubMed  Google Scholar 

  27. Kunieda J, Yamashita K, Togashi Y, Baba S, Sakata S, Inamura K, Ae K, Matsumoto S, Machinami R, Kitagawa M, Takeuchi K (2022) High prevalence of TERT aberrations in myxoid liposarcoma: TERT reactivation may play a crucial role in tumorigenesis. Cancer Sci 113:1078–1089. https://doi.org/10.1111/cas.15256

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Kwa WT, Effendi K, Yamazaki K, Kubota N, Hatano M, Ueno A, Masugi Y, Sakamoto M (2020) Telomerase reverse transcriptase (TERT) promoter mutation correlated with intratumoral heterogeneity in hepatocellular carcinoma. Pathol Int 70:624–632. https://doi.org/10.1111/pin.12974

    Article  CAS  PubMed  Google Scholar 

  29. Wang CC, Huang CY, Jhuang YL, Chen CC, Jeng YM (2018) Biological significance of TERT promoter mutation in papillary urothelial neoplasm of low malignant potential. Histopathology 72:795–803. https://doi.org/10.1111/his.13441

    Article  PubMed  Google Scholar 

  30. Wang X, Lopez-Beltran A, Osunkoya AO, Wang M, Zhang S, Davidson DD, Emerson RE, Williamson SR, Tan PH, Kaimakliotis HZ, Baldridge LA, MacLennan GT, Montironi R, Cheng L (2017) TERT promoter mutation status in sarcomatoid urothelial carcinomas of the upper urinary tract. Future Oncol 13:705–714. https://doi.org/10.2217/fon-2016-0414

    Article  CAS  PubMed  Google Scholar 

  31. Thomas C, Thierfelder F, Trager M, Soschinski P, Muther M, Edelmann D, Forster A, Geiler C, Kim HY, Filipski K, Harter PN, Schittenhelm J, Eckert F, Ntoulias G, May SA, Stummer W, Onken J, Vajkoczy P, Schuller U, Heppner FL, Capper D, Koch A, Kaul D, Paulus W, Hasselblatt M, Schweizer L (2021) TERT promoter mutation and chromosome 6 loss define a high-risk subtype of ependymoma evolving from posterior fossa subependymoma. Acta Neuropathol 141:959–970. https://doi.org/10.1007/s00401-021-02300-8

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Munoz-Jimenez MT, Blanco L, Ruano Y, Carrillo R, Santos-Briz A, Riveiro-Falkenbach E, Requena L, Kutzner H, Garrido MC, Rodriguez-Peralto JL (2021) TERT promoter mutation in sebaceous neoplasms. Virchows Arch 479:551–558. https://doi.org/10.1007/s00428-021-03083-9

    Article  CAS  PubMed  Google Scholar 

  33. Xiang Y, Yu Y, Li Q, Jiang Z, Li J, Liang C, Chen J, Li Y, Chen X, Cao W (2021) Mutual regulation between chicken telomerase reverse transcriptase and the Wnt/beta-catenin signalling pathway inhibits apoptosis and promotes the replication of ALV-J in LMH cells. Vet Res 52:110. https://doi.org/10.1186/s13567-021-00979-x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Teske N, Karschnia P, Weller J, Siller S, Dorostkar MM, Herms J, von Baumgarten L, Tonn JC, Thon N (2022) Extent, pattern, and prognostic value of MGMT promotor methylation: does it differ between glioblastoma and IDH-wildtype/TERT-mutated astrocytoma? J Neurooncol 156:317–327. https://doi.org/10.1007/s11060-021-03912-6

    Article  CAS  PubMed  Google Scholar 

  35. Zhao J, Li L, Guo L, Wang R, Zhao Y, Li W, Liu Y, Ma Y, Jia J (2021) Nano-gold PCR in detection of TERT methylation and its correlation with hepatitis B-related hepatocellular carcinoma. J Biomed Nanotechnol 17:1284–1292. https://doi.org/10.1166/jbn.2021.3103

    Article  CAS  PubMed  Google Scholar 

  36. Wang Y, Chen Y, Li C, Xiao Z, Yuan H, Zhang Y, Pang D, Tang X, Li M, Ouyang H (2022) TERT promoter revertant mutation inhibits melanoma growth through intrinsic apoptosis. Biology (Basel) 11:141. https://doi.org/10.3390/biology11010141

    Article  CAS  Google Scholar 

  37. Terzi NK, Yilmaz I, Oz AB (2021) The place and prognostic value of TERT promoter mutation in molecular classification in grade II-III glial tumors and primary glioblastomas. Turk Patoloji Derg 38:90–98. https://doi.org/10.5146/tjpath.2021.01555

    Article  CAS  Google Scholar 

  38. O’Hare TH, Delany ME (2011) Molecular and cellular evidence for the alternative lengthening of telomeres (ALT) mechanism in chicken. Cytogenet Genome Res 135:65–78. https://doi.org/10.1159/000330125

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. McKelvey BA, Umbricht CB, Zeiger MA (2020) Telomerase reverse transcriptase (TERT) regulation in thyroid cancer: a review. Front Endocrinol (Lausanne) 11:485. https://doi.org/10.3389/fendo.2020.00485

    Article  Google Scholar 

  40. Zhai G, Li J, Zheng J, An P, Chen X, Wang X, Li C (2020) hTERT promoter methylation promotes small cell lung cancer progression and radiotherapy resistance. J Radiat Res 61:674–683. https://doi.org/10.1093/jrr/rraa052

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Hayashi Y, Fujita K, Nojima S, Tomiyama E, Matsushita M, Koh Y, Nakano K, Wang C, Ishizuya Y, Kato T, Hatano K, Kawashima A, Ujike T, Uemura M, Imamura R, Morii E, Nonomura N (2020) TERT C228T mutation in non-malignant bladder urothelium is associated with intravesical recurrence for patients with non-muscle invasive bladder cancer. Mol Oncol 14:2375–2383. https://doi.org/10.1002/1878-0261.12746

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Hirata M, Fujita K, Fujihara S, Mizuo T, Nakabayashi R, Kono T, Namima D, Fujita N, Yamana H, Kamada H, Tani J, Kobara H, Tsutsui K, Matsuda Y, Ono M, Masaki T (2022) Telomerase reverse transcriptase promoter mutations in human hepatobiliary, pancreatic and gastrointestinal cancer cell lines. In Vivo 36:94–102. https://doi.org/10.21873/invivo.12680

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Lu F, Song Y, Cui S, Zhao H, Chen Y, Du H (2021) LncRNA MIAT promotes the proliferation, migration, and invasion of melanoma cells through recruiting TCF12 and activating NFAT5. Am J Transl Res 13:12588–12600

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Laban H, Siegmund S, Zappe M, Trogisch FA, Heineke J, Torre C, Fisslthaler B, Arnold C, Lauryn J, Buttner M, Mogler C, Kato K, Adams RH, Kuk H, Fischer A, Hecker M, Kuebler WM, Korff T (2021) NFAT5/TonEBP limits pulmonary vascular resistance in the hypoxic lung by controlling mitochondrial reactive oxygen species generation in arterial smooth muscle cells. Cells 10:3293. https://doi.org/10.3390/cells10123293

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Gwon DH, Kim SI, Lee SH, Noh C, Kim Y, Yun S, Lee WH, Oh JY, Kim DW, Hong J, Lee SY (2021) NFAT5 deficiency alleviates formalin-induced inflammatory pain through mTOR. Int J Mol Sci 22:2587. https://doi.org/10.3390/ijms22052587

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Dou Y, Tian W, Wang H, Lv S (2021) Circ_0001944 contributes to glycolysis and tumor growth by upregulating NFAT5 through acting as a decoy for miR-142-5p in non-small cell lung cancer. Cancer Manag Res 13:3775–3787. https://doi.org/10.2147/CMAR.S302814

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Jiang Y, He R, Jiang Y, Liu D, Tao L, Yang M, Lin C, Shen Y, Fu X, Yang J, Li J, Huo Y, Hua R, Liu W, Zhang J, Shen B, Zhang Z, Sun Y (2019) Transcription factor NFAT5 contributes to the glycolytic phenotype rewiring and pancreatic cancer progression via transcription of PGK1. Cell Death Dis 10:948. https://doi.org/10.1038/s41419-019-2072-5

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Chen BL, Li Y, Xu S, Nie Y, Zhang J (2021) NFAT5 Regulated by STUB1, Facilitates malignant cell survival and p38 MAPK Activation by upregulating AQP5 in chronic lymphocytic leukemia. Biochem Genet 59:870–883. https://doi.org/10.1007/s10528-021-10040-3

    Article  CAS  PubMed  Google Scholar 

  49. Kim DH, Kim KS, Ramakrishna S (2018) NFAT5 promotes in vivo development of murine melanoma metastasis. Biochem Biophys Res Commun 505:748–754. https://doi.org/10.1016/j.bbrc.2018.09.171

    Article  CAS  PubMed  Google Scholar 

  50. Fujiki T, Udono M, Kotake Y, Yamashita M, Shirahata S, Katakura Y (2010) NFAT5 regulates transcription of the mouse telomerase reverse transcriptase gene. Exp Cell Res 316:3342–3350. https://doi.org/10.1016/j.yexcr.2010.10.001

    Article  CAS  PubMed  Google Scholar 

  51. Zhang P, Hou Q, Yue Q (2020) MiR-204-5p/TFAP2A feedback loop positively regulates the proliferation, migration, invasion and EMT process in cervical cancer. Cancer Biomark 28:381–390. https://doi.org/10.3233/CBM-191064

    Article  CAS  PubMed  Google Scholar 

  52. Xu H, Wang L, Jiang X (2021) Silencing of lncRNA DLEU1 inhibits tumorigenesis of ovarian cancer via regulating miR-429/TFAP2A axis. Mol Cell Biochem 476:1051–1061. https://doi.org/10.1007/s11010-020-03971-9

    Article  CAS  PubMed  Google Scholar 

  53. Yu P, Shen X, Yang W, Zhang Y, Liu C, Huang T (2018) ZEB1 stimulates breast cancer growth by up-regulating hTERT expression. Biochem Biophys Res Commun 495:2505–2511. https://doi.org/10.1016/j.bbrc.2017.12.139

    Article  CAS  PubMed  Google Scholar 

  54. Qin Y, Tang B, Hu CJ, Xiao YF, Xie R, Yong X, Wu YY, Dong H, Yang SM (2016) An hTERT/ZEB1 complex directly regulates E-cadherin to promote epithelial-to-mesenchymal transition (EMT) in colorectal cancer. Oncotarget 7:351–361. https://doi.org/10.18632/oncotarget.5968

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We would like to thank the Springer Nature Author Services for editing the English language, grammar and spelling.

Funding

This study was supported by the Key-Area Research and Development Program of Guangdong Province (2020B020222001), the Poultry Industry Technology System of Guangdong (2021KJ128), and the China Agriculture Research System of MOF and MARA (CARS-41).

Author information

Authors and Affiliations

Authors

Contributions

YX and WC conceived and designed the study. YX, QC, QL and CL performed the experiments. YX and WC analyzed the data and wrote the paper. YX, WC and CL contributed to critical revision of the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Weisheng Cao.

Ethics declarations

Ethics approval and consent to participate

All experiments involving chickens were performed in qualified facilities, and experimental protocols (2021c059) were approved by the biosafety committee of South China Agriculture University. Housing animals were conducted in accordance with the guidelines of the experimental animal administration and ethics committee of South China Agriculture University.

Availability of data and materials

The data on which the conclusions of the manuscript rely are presented in the main paper and supplementary information.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1. ALV-J Tumor tissues and their sources and quantities.

Additional file 2. Matrix of methylation levels of chTERT amplicon CG sites in LMH cells and DF-1 cells.

13567_2022_1069_MOESM3_ESM.doc

Additional file 3. Matrix of methylation levels of chTERT amplicon CG sites in ALV-J tumor, tumor-adjacent and normal tissues.

Additional file 4. Agarose gel electrophoresis of PCR amplification of the chTERT promoter region.

Lanes 1-8: different tissue samples; Lane 9: negative control.

Additional file 5. Analysis of DNA sequence mutations in the promoter region of chTERT.

Green box: normal tissues; blue box: tumor-adjacent tissues; red box: tumor tissues; orange box: mutant tumors. T: tumor tissues; TA: tumor-adjacent tissues; N: normal tissues.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xiang, Y., Chen, Q., Li, Q. et al. The expression level of chicken telomerase reverse transcriptase in tumors induced by ALV-J is positively correlated with methylation and mutation of its promoter region. Vet Res 53, 49 (2022). https://doi.org/10.1186/s13567-022-01069-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13567-022-01069-2

Keywords