Abstract
Telomeres shorten with each cell division, serving as biomarkers of aging, with human tissues exhibiting short telomeres and restricted telomerase expression. In contrast, mice have longer telomeres and widespread telomerase activity, limiting their relevance as models for human telomere biology. To address this, we engineer a mouse strain with a humanized mTert gene (hmTert), replacing specific non-coding sequences with human counterparts. The hmTert gene, which is repressed in adult tissues except the gonads and thymus, closely mimics human TERT regulation. This modification rescues telomere dysfunction in mTert-knockout mice. Successive intercrosses of Terth/- mice stabilized telomere length below 10 kb, while Terth/h mice achieve a human-like average length of 10–12 kb, compared to 50 kb in wildtype mice. Despite shortened telomeres, Terth/h mice maintain normal body weight and cell homeostasis. These mice, with humanized telomere regulation, represent a valuable model to study human aging and cancer.
Similar content being viewed by others

Introduction
Telomeres play key roles in the development of many human diseases, especially age-related disorders and cancer. Telomeres are replenished by telomerase, a ribonucleoprotein complex containing TERT, TERC, and accessory proteins1. Humans are born with 10–15 kb telomeres and adults have 5–15 kb of telomeres2,3. Telomerase activity is very low or undetectable in most adult human tissues. As a result, telomeres progressively shorten upon successive cell divisions and become exhausted in aged somatic tissues, triggering replicative senescence or cell death, and thereby functioning as an aging clock. Mutations in human telomerase genes lead to dyskeratosis congenita, a prototypical telomere biology disorder that presents as a multi-system syndrome with a broad spectrum of clinical manifestations, including aplastic anemia and cancer4. Additionally, short telomere-induced replicative senescence/cell death in human cells also functions as a tumor-suppressing mechanism5.
There exist important differences in telomerase regulation and telomere length among mammal species. In humans, telomerase expression is restricted to a small number of organs, such as testis, ovary, and thymus, and telomeres are exhausted in many aged tissues. However, a comparative study of over 60 mammals showed that many organisms, including mice, did not have telomere-mediated replicative aging6. Telomerase expression in mice is less restricted, with most tissues expressing significant levels of Tert mRNA and telomerase activity7,8. Laboratory inbred strains, such as C57BL/6, have long telomeres. Telomerase-null mice (Tert or Terc-KO) survive up to 6 generations with no discernible phenotypes in early generations, indicating that mice have adequate telomere reserves that are not exhausted for multiple generations without telomerase9.
Mouse models of human diseases have become a central part of biomedical research. Laboratory mice provide the most experimentally accessible mammalian models that share genes, organs, and systemic physiology with humans. However, many mouse models fail to mimic human disease progression, posing translational challenges and limiting their use in human disease research. This may have contributed to the high failure rates of human clinical trials, particularly in oncology, predicating the need for improved preclinical data from animal models10.
Beyond differences in telomerase expression and telomere length, the telomere biology in humans and mice exhibits several other notable distinctions. For example, mice have two Pot1 proteins, Pot1a and Pot1b, whereas humans have only one POT1 protein11. Additionally, a key amino acid difference (M492K) in the RTEL1 proteins of humans and mice has been associated with shorter telomeres12. While these differences likely influence telomere maintenance, we have focused on the regulation of the TERT gene, the limiting catalytic subunit of telomerase.
To develop a mouse model with human-like telomere homeostasis, we previously engineered a humanized mTert allele, hmTert, by replacing the 5′ intergenic region (5’IR), introns 2 and 6 of the mTert gene with their human counterparts in mouse embryonic stem cells (ESCs)13. In the present study, we successfully obtained C57BL/6 J mice with a germline hmTert gene. Tert mRNA expression and telomerase activity in these mice were distinct from those in wildtype C57BL/6 J mice, but remarkably similar to those in humans. Importantly, the hmTert gene was able to rescue telomere deficiency when it was crossed into the 5th generation of mTert knockout (KO) mice. Crucially, the introduction of hmTert allele led to a recalibration of telomere equilibrium length in these mice. Following successive intercrosses of Terth/- mice and subsequent incrosses of Terth/h mice, this equilibrium settled at a range of 8–10 kb and 10–12 kb, respectively. The equilibriums were notably shorter than that in wildtype C57BL/6 J mice, but resembled human telomere lengths. This adjustment was also associated with a marked decrease in colonocyte proliferation following treatment with dextran sodium sulfate (DSS), suggesting that the shorten telomeres had a restrictive impact on the proliferative potential of somatic cells in these mice. In summary, the humanization of the mTert gene in C57BL/6 J mice culminated in the development of a murine population that closely replicated human-like telomere homeostasis.
Results
Telomerase regulation in mice with the hmTert allele
The hmTert gene, containing the human 5’IR (23 kb), introns 2 (11 kb) and 6 (5.5 kb) (Fig. 1a) was highly expressed in embryonic stem cells and stringently repressed upon differentiation13. Terth/+ mice were obtained and mated with Tert+/- mice14, generating mice of Terth/-, Tert+/-, and Terth/+ genotypes. As shown in Fig. 1b, telomerase activity was readily detected in the majority of adult tissues in Tert+/- mice, yet it was present in a limited number of tissues in their Terth/- littermates. A direct comparison of mTert and hmTert mRNAs in Terthm/+ mice showed that mTert mRNA was expressed in most organs, whereas high hmTert mRNA expression was found only in thymus (Fig. 1c). Relatively low levels of hmTert mRNA were detected in testis and ovary, and very low levels in intestine and spleen. The hmTert mRNA expression pattern was similar to that of the hTERT mRNA in human tissues (Fig. 1d), suggesting that the postnatal regulation of the hmTert gene recapitulated that of hTERT in humans.
a Genomic maps of hTERT, mTert, and hmTert loci. Arrows indicate the directions of transcription. Vertical lines are exons; black and dark grey regions represent repetitive sequences, TEs, and VNTRs, respectively. Human and mouse 5’IR and introns 2 & 6 are labeled in blue and red, respectively. b Telomerase expression in tissues from Tert+/- and Terth/- littermates. Telomerase activities were determined by TRAP assay. 0.5 µg protein extracts from 4-month-old mice were used except for thymus (0.12 µg). +, h, and - refer to mTert, hmTert, and mTert-KO alleles, respectively. Organs from at least three different mice were examined and data from one representative mouse are shown here. c The expression of Tert mRNAs in adult mice. Tissues were collected from 4-month-old Terth/+ mice. hmTert and mTert mRNAs were distinguished by using primers overlapping with the silent mutations in exon 2 of the hmTert allele. P values, two tailed student’s t test, are shown. d hTERT mRNA expression in human tissues. Tert/TERT mRNA data were determined by qRT-PCR assay, normalized to 18S rRNA, and compared to those in Terth/+ ESCs or human ESC H1 cells (1.0). For c, d means and standard deviations (±SDs) of three technical repeats are shown. Repeated experiments showed similar results. Source data for panels b–d are provided as Source Data files.
During postnatal development, hmTert expression in testis, ovary, thymus, and intestine was more pronounced within the first two weeks of newborns and decreased with age in young mice (Supplementary Fig. 1a). Only thymus maintained significant hmTert expression. By comparison, mTert expression was high in most organs of newborns and decreased upon postnatal development, except for ovary, liver, and spleen, where high and low mTert mRNA levels were maintained in adults, respectively.
It was reported that hTERT expression progressively declined during T cell differentiation15 and T cells rapidly upregulated hTERT mRNA to support robust cell division and differentiation16. Consistently, resting mouse T cells expressed little hmTert mRNA and its level increase dramatically in CD4+ and CD8+ T cells following stimulation by CD3/CD28 antibodies for 48 and 72 h (Supplementary Fig. 1b). mTert RNA was readily detected in resting T cells and its level also increased during T cell activation. The increase of hmTert and mTert RNAs correlated with EdU incorporation, and thus cell proliferation, in T cells (Supplementary Fig. 1c).
Telomere lengths in Tert+/-, Terth/-, and Tert-KO mice
C57BL/6 J mice have an average telomere length of approximately 50 kb. To reduce telomere length in mice with hmTert genes, Tert+/h F1 mice were crossbred to Tert+/- mice to produce Terth/- and Tert+/- offspring, which were then bred with successive generations of Tert-/- mice (Fig. 2a). Telomere length of these mice were examined by flow cytometry following fluorescence in situ hybridization (Flow-FISH) and telomere restriction fragment (TRF) analysis by Southern blotting (Fig. 2b, c). As the breeding of Tert-/- mice resulted in shortened telomeres, similar decreases in telomere length were observed in both Tert+/- and Terth/- mice over successive generations. In each generation, Terth/- mice had shorter telomeres on average compared to their Tert+/- counterparts. In G6 Terth/- mice, the average telomere length dropped to roughly 50% of the length in wildtype mice, or ~25 kb. The litter sizes of Tert+/-xTert-/- and Terth/-xTert-/- mattings declined over generations, while the litter sizes of homozygous Tert-/- incrosses decreased even more dramatically (Fig. 2d). In several attempts, G6 Tert-/- mice produced only one offspring which died prematurely, ultimately ending this breeding strategy. However, the reduction in telomere length in Tert+/- and Terth/- mice did not negatively impact their overall health and well-being, as evidence by their normal body weight in G6 mice (Fig. 2e).
a Breeding strategy. Telomere length of splenocytes from 2-month-old Tert+/-, Terth/-, and Tert-/- mice were determined by Flow-FISH (b) and telomere restriction fragment (TRF) analysis (c). b Telomere Flow-FISH. Telomere signals were detected by hybridization to FAM-(CCCTAA)3 oligonucleotide. Fluorescence signals were compared to that of wildtype C57BL/6 J mice (1.0). c TRF analysis. Splenocyte genomic DNAs were digested with HinfI and RsaI, followed by pulsed-field gel electrophoresis and Southern blotting. Positions of size markers are shown on the left (kb). d Litter sizes of breeding between Tert+/- and Tert-/- (red triangles), Terth/- and Tert-/- (blue circles), Tert-/- and Tert-/- (white squares) mice. e Body weight of male (upper) and female (lower) mice at 8-week of age. f Testis weight of mice at 10–15-week age. g H&E staining of seminiferous tubules in testes from Tert+/-, Terth/-, and Tert-/- mice. Yellow arrowheads indicate aberrant tubules. h Average percentages of aberrant seminiferous tubules in testes. +/+, n = 3; +/-: G3, n = 3; G4, n = 2, G5, n = 3; h/-: G3, n = 3, G4, n = 3, G5, n = 4; -/-: G3, n = 3, G4, n = 3, G5, n = 3. i, j, Survival curves of mice with mTert, hmTert, and mTert-KO alleles. Mice were bred as shown in panel a. Kaplan-Meier survival curves of G4 (i) and G5 (j) mice are shown. P-values of survival curve comparisons were calculated using logrank test. Each datapoint in panels b, d, e, f, & h represents one biological repeat (one animal). Means ± SDs are indicated. Source data for panels b–f and h–j are provided as Source Data files.
Maintaining testis homeostasis by the hmTert gene
Due to the impact of shorter telomeres on mouse fertility, we analyzed the testes of these mice. As illustrated in Fig. 2f–h, Tert-/- mice showed testicular atrophy as well as a progressive loss of germ cells in seminiferous tubules starting from G3 mice and worsening in G4 and G5 mice, as previously reported in telomerase deficient mice17. Tert+/- mice also exhibited a low level of testicular defects in G4 and G5, but such defects were absent in Terth/- mice. These data indicate that both mTert and hmTert genes help preserve germ cells in the testis, with hmTert showing a slight advantage in preventing germ cell loss.
Sustaining mouse lifespan by the hmTert gene
Previous studies have shown that telomere deficiency can impact mouse survival and lifespan18,19. Our results align with those findings, as shown in Fig. 2i, j, G4 and G5 Tert-/- mice had a significantly shortened lifespan, with median survival of approximately 440 and 320 days, respectively. However, the majority of wildtype Tert+/+, G4 and G5 Tert+/- and Terth/- mice lived past 500 days, indicating that the presence of a mTert or hmTert gene could sustain longevity even when telomeres were relatively short.
Rescuing telomere dysfunction in Tert-/- mice by the hmTert gene
Offspring inherit not only parents’ genotypes but also the lengths of their telomeres. G5 Tert-/- mice displayed severe telomere dysfunction, evidenced by tissue dystrophy, reduced body weight, and a shortened lifespan. The next generation, denoted as G6 Tert-/-m and G6 Tert-/-h, was generated by crossing G5 Tert-/- mice with G5 Tert+/- and Terth/- mice, respectively (Fig. 3a). Examination of telomere length through TRF and Flow-FISH analyses indicated that the average telomere lengths in G6 Terth/- and Tert+/- mice were comparable to, or slightly longer than, those of their G5 Tert-/- parents and G6 Tert-/- siblings (Fig. 3b and Supplementary Fig. 2a, b). However, despite these similarities, G6 Terth/- and Tert+/- mice exhibited significantly extended lifespans compared to their G5 Tert-/- mice (Fig. 3c). Notably, while all G5 Tert-/- mice died within 383 days, three out of 14 G6 Tert-/-h mice survived beyond the entire 460-day experimental period. This resilience could be attributed to the inheritance of short yet functional telomeres from their G5 Terth/- parents. Taken together, these findings suggest that the hmTert allele in Terth/- mice has the capacity to restore the shortest and likely most impaired telomeres, even in the context of their overall short average telomere length.
a Mouse breeding scheme. b TRF analysis of representative animals. Splenocyte genomic DNAs were digested with HinfI and RsaI, followed by pulsed-field gel electrophoresis and Southern blotting. c Kaplan-Meier survival curves of mice. P-values comparing indicated paired curves were determined using logrank tests. d Body weight of male (upper) and female (lower) mice at 8-week of age. e Testis weight of mice at 10−15-week age. Each datapoint in panels d & e represents a biological repeat. Means ± SDs are shown. P values shown are calculated using one-way Anova. Source data for panels b–e are provided as Source Data files.
Further examination of genotypes among G6 progeny indicated that fewer G6 Tert-/- offspring were born compared to their Terth/- siblings (Supplementary Fig. 2c), indicating that some Tert-/- offspring died during prenatal development. Although G6 Tert-/-m and G6 Tert-/-h mice had reduced body and testis weights compared to their Tert+/- and Terth/- littermates, respectively, their testes weighed significantly more than their G5 Tert-/- parents (Fig. 3d, e). In addition, G6 Tert-/-h mice had a slightly increased testis weight compared to G6 Tert-/-m mice, suggesting that the hmTert gene was functionally similar to, or somewhat better than, the mTert gene for rescuing testicular defects. Overall, G6 offspring showed better general health compared to their G5 Tert-/- parents, demonstrating the telomere function-restoring capacities of both the hmTert and mTert genes within a single generation.
hmTERT function in immune system
Previous studies have shown that hematopoietic cells’ proliferative capacity was compromised in telomerase-deficient mice, and human short telomere syndromes cause anemia, decreased erythropoiesis, and T cell immunodeficiency20,21,22. Consistent with earlier reports, peripheral blood from G5 Tert-/- mice exhibited a slight decrease in white blood cell (WBC) counts, a statistically significant reduction in red blood cell (RBC) counts, and normal platelet numbers (Fig. 4a). Among WBCs, there was a marked decrease of lymphocytes observed in G5 Tert-/- mice, accompanied by corresponding increases in neutrophils and monocytes. Both G6 Tert+/- and Terth/- mice had blood cell counts similar to wildtype mice. The lymphocyte and neutrophil cell percentages within WBCs of G6 Tert-/-h mice were between those of wildtype mice and their G5 Tert-/- parents. G5 Tert-/- mice had a dramatically reduced number of CD8+ T cells and a significant increase of the CD4/CD8 T cell ratio (Fig. 4b). Additionally, CD19+ B cells decreased in G5 Tert-/- mice. All these cell counts in G6 Tert+/- and Terth/- mice were restored to the levels found in wildtype mice. Further analyses of T and B cell counts in spleen and bone marrow revealed similar changes in G5 Tert-/- mice and Tert+/- and Terth/- offspring (Supplementary Fig. 3). In short, our data indicate that the hmTert gene effectively rescued blood cell defects in G5 Tert-/- mice within one generation.
a Whole blood cell counts in adult mice of 3–6 months by hematology analyses. b Lymphocyte counts in peripheral blood. Cells were stained using antibodies and analyzed by flow cytometry. Each datapoint represents one animal. Means ± SDs are indicated. P values shown are calculated using one-way Anova. Source data are provided as Source Data files.
hmTERT function in small intestine
The gastrointestinal tract is another high proliferation tissue that is affected by telomere dysfunction23. Depletion of the intestinal epithelial crypts and severe villus atrophy were observed in small intestines of older G5 Tert-/- (≥ 8 months), but not in G6 Tert+/- and Terth/- mice (Fig. 5a). The intestinal lesions probably contributed to the loss of body weight and overall poor health of Tert-/- animals due to decreased nutritional absorption.
a Histopathology of small intestines of adult mice of 8–10 months. The bar indicates 100 µm. b The expression of genes regulating cellular senescence and proliferation in small intestine. Each column represents an individual mouse. Relative mRNA levels were determined by qRT-PCR and normalized to 18S rRNA. Means ± SDs are shown. Source data are provided as a Source Data file.
The intestinal defects are likely a consequence of cell cycle inhibition and cellular senescence induced by telomere dysfunction. Therefore, the expression of genes involved in cell proliferation was examined. Tert mRNA was readily detected in the intestines of Tert+/+ and Tert+/- mice, but not in those of Terth/- or Tert-/- mice, confirming that the hmTert gene is strictly regulated in adult tissues (Fig. 5b). Markers of cell proliferation, Ki-67 and PCNA, and the cell cycle inhibitor p21 were found in the intestine of all genotypes. Senescence-associated genes, p16Ink4a and IL-6, were upregulated in G5 Tert-/- and G6 Tert-/-h mice, but not in any mice with mTert or hmTert genes. TNF-α, another pro-inflammatory cytokine secreted by senescent cells, appeared to be expressed in mouse intestines of all genotypes. Therefore, our data suggest that cellular senescence occurred in Tert-/- intestines with significant telomere dysfunction but was suppressed by the presence of the hmTert gene in this tissue.
Telomere shortening during intercrosses of Terth/- mice
Despite its restricted expression in adult tissues, the hmTert gene rescued telomere dysfunction in G5 Tert-/- mice with relatively short average telomeres of ~25 kb. Our next objective was to determine the telomere length setpoint influenced by the hmTert gene. To this end, G4 Terth/- mice were continuously intercrossed for 16 generations, from G4.1 to G4.16 (Fig. 6a). Using Flow-FISH, we monitored telomere length of Terth/- mice in splenocytes at each generation. As depicted in Fig. 6b, the average telomere length of Terth/- mice decreased from 60% to 18% of that observed in wildtype mice from G4 to G4.14, eventually stabilizing at 18–19% in the last three generations (G4.14 to G4.16). Throughout the breeding generations, both male and female Terth/- mice maintained body weights similar to those of wildtype mice (Fig. 6c). During this process, litter sizes varied, but were largely maintained (Fig. 6d). Male mice also maintained their testis weight (Fig. 6e). Figure 6f compares the average telomere length of all three genotypes in each generation, from G4.10 to G4.16. It demonstrates that Terth/h mice in general had longer telomere than their Terth/- siblings, and that Tert-/- mice consistently exhibited the shortest telomeres across generations. Overall, our data indicated that average mouse telomeres could be shorten to below 10 kb without affecting their overall health, at least at a young age, as long as they have the hmTert gene.
a Breeding strategy. Terth/- progeny from G4 Terth/- parents were intercrossed. b Telomere length as determined by Flow-FISH. Splenocytes from 2-month-old mice were used for the analyses. P value within the groups of G4.14, G4.15, and G4.16 was calculated using one-way ANOVA. c Body weight of 8-week-old male and female mice. d Litter sizes. e Testis weight of mice at 10–15-week age. f Flow-FISH comparing telomere lengths of Terth/h, Terth/-, and Tert-/- littermates. Each datapoint in panels b–f represents one animal. Means ± SDs are shown. n ≥ 3. g TRF analysis. Splenocyte genomic DNAs were digested with HinfI and RsaI, followed by 0.6% Agarose gel electrophoresis and Southern blotting. Sizes are indicated on the left (kb). MW, molecular weight marker. NHF (P11), passage 11 normal human foreskin fibroblasts. A longer exposure is shown in Supplementary Fig. 8a. h Genotype ratios of progeny in intercrosses at 7, 21, and 56 postnatal days. n = 21 ~ 93. Source data for panels b–h are provided as Source Data files.
Telomere length in later generations of mice was also verified using TRF analysis (Fig. 6g). Two types of telomeres were found in these mice: discrete bands of variable sizes and intensities between 15 and 20 kb, and shorter human-like telomere smears. The upper bands were sensitive to Bal-31 exonuclease digestion, indicating that they were indeed telomeres (Supplementary Fig. 4). The mean telomere lengths were 7–9 kb in G4.14–G4.16 Terth/h mice and 6–8 kb in the Terth/- mice (Supplementary Table 1). In a G4.14 Tert-/- mouse, the telomere smear was much less apparent. Figure 6h shows that, from G4.2 to G4.12, approximately 50% of total born mice were of heterozygous Terth/- genotype, while homozygous Terth/h and Tert-/- mice each accounted for about 25% of total progeny, following Mendelian genetics. However, there was a sharp decline in the numbers of Tert-/- mice born from G4.13 to G4.16. The few Tert-/- mice that were born were small and die at young ages. These data indicated that short telomeres in late-generation Tert-/- embryos could no longer sustain mouse development.
Maintaining stable human-like telomeres in homozygous Terth/h mice
To assess the stability of short telomeres in mice with the hmTert genes, homozygous Terth/h offspring from G4, G4.8, and G4.14 were incrossed for 13, 9, and 2 generations, respectively (Fig. 7a). Average telomere lengths in their progeny were measured by Flow-FISH. The results, depicted in Fig. 7b, revealed a gradual decrease in telomere length across successive generations. In G4 Terth/h mice, telomeres decreased from 60% to 30%, while in G4.8 Terth/h mice, the decline went from 34% to 24%. G4.14 Terth/h mice exhibited telomere lengths approximately 24% of the wildtype, and their offspring maintained telomeres at 22–23% of wildtype length during two successive generations of incrossing. These findings indicate that telomere length in Terth/h mice stabilized at a shortened but consistent ranges of 21–24% of wildtype mice, equivalent to an average telomere length of 10–12 kb, similar to reported leukocyte telomere lengths of 9.5 ± 0.7 kb and 10-11 kb in newborn humans2,24. Additionally, TRF analysis confirmed the presence of variable discrete telomere bands between 15–20 kb and a human-like telomere smear (Fig. 7c). For G4.8(g-i) and G4.14(a–b) mice, the average lengths of telomere smears were approximately 10 kb. Regardless of having shortened telomeres, these mice exhibited good health, as demonstrated by stable body weight, litter sizes, and testis weight (Fig. 7d–f).
a Breeding schemes. Terth/h progeny from G4, G4.8, and G4.14 Terth/- parents were successively incrossed. b Relative telomere signals. Telomere signals were determined by Flow-FISH and normalized to that of wildtype C57BL/6 J mice (50 kb). n ≥ 3. c TRF analysis. A longer exposure is shown in Supplementary Fig. 8b. d Body weight. e Litter sizes. f Testis weight. Each data point represents one animal. Means ± SDs are shown in panels b, & d–f. Source data for panels b–f are provided as Source Data files.
To further determine the health status of Terth/h mice with human-like telomeres, we performed hematology analysis on peripheral blood samples from G4.8 g, h, and i mice aged 2–3 months. As shown in Supplementary Fig. 5, the result indicated normal red blood cell counts, hemoglobin, and hematocrit in all groups of mice. Although the average WBC count in G4.8 h was lower than those of G4.8 g, G4.8i, and wildtype mice, it still fell within the normal WBC range of physiological data for C57BL/6 J mice published by the Jackson Laboratory25. Within WBC populations, all groups of mice displayed normal percentages of lymphocytes, monocytes, and neutrophils, further supporting the notion that Terth/h mice with human-like telomeres maintain good health at early stages of their life.
Comparing hmTert and mTert gene functions
In order to directly compare the functions of the hmTert and mTert genes and establish a another line of Terth/h mice, we conducted intercrosses using G6 Terth/- and Tert+/- mice (Fig. 8a). Consistent with our previous findings, telomere length in G6 Terth/- mice decreased from 55% to 18–19% of wildtype telomere length in C57BL/6 J mice across 12 successive intercrosses (Fig. 8b). In contrast, the average telomere lengths in G6 Tert+/- mice remained stable at 55% of wildtype telomere length over eight intercrosses. This result aligned with a previous study in which Tert+/- mice were intercrossed for 17 generations, stabilizing their average telomere length at approximately 50% of wildtype telomeres26. Importantly, G6 Terth/- mice exhibited consistent litter sizes and development with appropriate body and testis weights across the entire breeding process (Fig. 8c–e). Therefore, our data indicate that the Tert loci play a crucial role in regulating telomere length homeostasis, and the hmTert gene genetically determines short telomeres in mice.
a Breeding strategy. G6 Tert+/- and Terth/- mice from Fig. 2a were independently intercrossed. b Relative telomere signals as determined by Flow-FISH and normalized to that of wildtype C57BL/6 J mice. c, Body weight. d Litter sizes. e Testis weight. Each data point represents one animal. Means ± SDs are shown. Source data for panels b–e are provided as Source Data files.
Hematopoiesis following bone marrow ablation
A major problem in patients with telomere biology disorders are hematopoietic defects, manifested as decreased marrow reserves and increased sensitivities to cytotoxic agents27. Here, we examined the responses of Terth/h mice with short telomeres (G4.8i) to the chemotherapeutic agent 5-fluorouracil, or 5-FU. The numbers of white blood cells, red blood cells, and lymphocytes in the peripheral bood of both C57BL/6 J (Tert+/+) and Terth/h mice decreased significantly on days 6 and 11 post 5-FU injection (Supplementary Fig. 6). The numbers of platelets in both experimental groups also declined on day 6 but increased on day 11. The recovery of platelet numbers in Terth/h mice appeared to be slightly slower compared to those in wildtype mice. Therefore, the data showed that the ability to sustain and recover from 5-FU-induced damage in the hematopoietic system was not significantly impacted by short telomeres in Terth/h mice.
Decreased in vivo cell proliferation capacity in Terth/h mice
Ulcerative colitis is an inflammatory disease associated with telomere shortening and accelerated colon aging in human patients28. G4.8 h Terth/h mice remained in good health, and their gastrointestinal tracts appeared normal. As depicted in Fig. 9d,e, cellular proliferation, assessed by EdU incorporation, in the colons of G4.8 h Terth/h mice were similar to that wildtype mice. To evaluate tissue renewal capacity under pathological conditions, mice were subjected to a 6-day dextran sodium sulfate (DSS) treatment to induce conditions akin to ulcerative colitis29,30 (Fig. 9a). DSS treatment led to similar colon shortening and spleen enlargement in both Tert+/+ and Terth/h mice, indicating DSS-induced comparable inflammatory responses in both groups (Fig. 9b, c). The toxicity of DSS to colonic epithelial cells triggered a regenerative response upon toxin removal29. In Terth/h mice, an average of about 2 EdU-positive cells per crypt cross-section were observed, significantly fewer than the average of 7 EdU-labeled cells per crypt cross-section in wildtype mice (Fig. 9d, e). These results suggested that, while Terth/h mice with human-like short telomeres maintained tissue homeostasis during postnatal development and adulthood under normal physiological conditions, tissue renewal was more limited under pathological conditions due to their short telomeres and absence of telomerase activity.
a Experimental strategy. 7–8-month-old Tert+/+ (wildtype C57BL/6 J) and Terth/h (G4.8 h) mice were given drinking water with or without 3% DSS for 6 days, followed by 1 day of pure drinking water. Intraperitoneal EdU injection was performed 2 h before tissue collection. b Representative images of colons and spleens following DSS treatment. c Spleen weight. Spleen weight was normalized to the body weight of each mouse. Each data point represents one animal. n = 4. d EdU staining of colon crypt sections. Colon tissues were labeled with anti-EdU (white) and E-cadherin (green) antibodies, as well as Hoechst dye for nuclear staining (blue). Scale bar represents 100 µm. Representative images are shown. e Quantification of EdU-positive cells. Each datapoint in panel e represents the average number of EdU-positive cells per colon crypt in 30 crypts from one animal. n = 6. Means ± SDs are shown in panels c, e. P values, calculated using two-way Anova, are shown. Source data for panels c and e are provided as Source Data files.
Telomere integrity in mouse cells with human-like telomeres
To assess telomere integrity in somatic cells from mice with human-like short telomeres, mouse embryo fibroblasts (MEFs) were prepared from individual embryos of G4.16 Terth/- intercrosses. To minimize telomere damages, MEFs were culture in 3% oxygen31. Telomere length in passage 4 MEFs was measured using telomere fluorescence in situ hybridization (FISH) analysis. As shown in Supplementary Fig. 7a, both Terth/h and Terth/- MEFs exhibited lower telomere fluorescence compared to wildtype (Tert+/+) MEFs. Quantitative measurements revealed that the average telomere fluorescence signals of Terth/h and Terth/- MEFs were approximately 18% and 15% of those in wildtype MEFs, respectively (Supplementary Fig. 7b). These MEFs were stained with antibodies against TRF2 and γH2A.X. Damaged telomeres were identified as telomere dysfunction-induced foci (TIFs) through the colocalization of TRF2 and γH2A.X staining. As shown in Supplementary Fig. 7, panels c and d, Terth/h and wildtype MEFs had similar numbers of TIFs. However, TIF numbers were elevated in Terth/- MEFs and further increased in Tert-/- MEFs. These results indicate that while short telomeres in Terth/h mice are functional, telomere dysfunction is present in cells from Terth/- and Tert-/- mice.
Discussion
While significant progress has been made in unraveling the mechanisms governing telomere maintenance and regulation, the genetic factors contributing to the variation in telomere length among species and individuals are still not fully understood. Moreover, the discrepancies in telomerase expression and telomere length between mice and humans pose significant challenges for studying human cancer and age-related diseases using mouse models. To overcome this obstacle, we engineered a humanized version of the mTert gene, known as hmTert, by replacing the 5’IR and introns 2 and 6 with their human counterparts in mouse ESCs13,32. In this report, we generated and characterized mouse lines with germline hmTert alleles. Our findings demonstrate that the regulation of the hmTert gene resemble that of the hTERT gene during postnatal development and in adult tissues and the hmTert gene resets mouse telomere to human length. Given the inability of hTERT protein to function with mTerc and other telomerase accessory proteins33,34, the generation of a similar mouse model using alternative approaches, such as complementing a mTert knockout allele with a transgenic hTERT gene, is not feasible7.
Telomere homeostasis is regulated by telomerase, the shelterin protein complex, and other telomere-protecting proteins, such as Rtel1 helicase35,36. The shelterin proteins exhibit structural and functional conservation between mice and humans, with the exception of POT1. Mice possess two paralogs, mPot1a and mPot1b, and they both bind TPP1 and perform the same overall functions as the single human POT1 protein11. Our data indicate that humanization of the mTert gene resets equilibrium telomere length. During both intercrosses of Terth/- mice and incrosses of Terth/h mice, average telomere lengths progressively shortened until reaching 9–10 kb and 10–12 kb, respectively. The new equilibrium point of telomere length for Terth/h mice is similar to that observed in human telomeres, resulting a mouse strain with humanized telomerase expression and telomere length. These Terth/h mice, named HuT mice, possess a C57BL/6 J genetic background.
Our data unequivocally establish the Tert gene as a primary genetic determinant of telomere length in mice. A fundamental difference between the hTERT and mTert genes lies in the rich context of repetitive sequences that encompassing the hTERT promoter, including transposable elements (TEs) and variable number tandem repeats (VNTRs), primarily within the 5’ IR and intron 2 (Fig. 1a). These elements play a crucial role in maintaining stringent hTERT repression in most adult tissues while allowing its expression in key organs, such as the thymus and gonads13,37,38,39. Although the precise molecular basis for the shorter telomeres in Terth/h and Terth/- mice compared to Tert+/+ or Tert+/- mice warrants further investigation, we have observed the hmTert gene more effectively rescues telomere dysfunction-induced seminiferous tubule atrophy compared to the mTert allele. This is particularly noteworthy, even with the lower hmTert mRNA expression in the testis compared to mTert mRNA (Fig. 1c). It is conceivable that the higher expression of mTert throughout germ cell development results in longer telomeres in progeny. Additionally, our previous data have indicated that the hTERT promoter activity is especially high in elongating spermatids8. These cells are undergoing dramatic epigenetic reprogramming, involving the replacement of canonical histones with protamines and extensive DNA methylation40, and exhibit elevated telomerase activity41. The robust expression of hmTert/hTERT in these cells likely contributes to maintaining telomere integrity and germline cell survival during this crucial stage, allowing haploid germ cells with short telomeres to develop into sperm. This also provides a plausible explanation for the efficient rescue of telomere dysfunction by hmTert in the testis. Further investigation into hmTert/hTERT expression in the testis cells may yield deeper insights into this phenomenon at the cellular level.
Mice with genotypically wildtype characteristics but short telomeres have been reported previously23,26,42. These mice were obtained through homozygosity induction following extensive intercrosses of heterozygous Tert+/- or Terc+/- mice23,26. Another approach involved a single amino acid change (M492K) in the Rtel1 gene (Known as Telomouse), leading to much shorter human-like telomere length12. Although telomeres in these mice were shorter than those in wildtype C57BL/6 mice, most of them – except for those in Telomice – were still substantially longer than human telomeres. Furthermore, all previously reported mice with shorter telomeres carried the mTert gene and their telomerase expression remained widespread in somatic tissues. In contrast, HuT mice display human-like short telomeres and show minimal telomerase activity in most adult tissues. Additionally, the rate of telomere shortening per generation was higher during Terth/- intercrosses than in Terth/h incrosses. Terth/- offspring also exhibited shorter telomeres than their Terth/h siblings during intercrosses. These data indicate that the hmTert gene is haploinsufficient, rendering Terth/- mice an ideal model for dyskeratosis congenita (DC), a human genetic syndrome resulting from mutations in telomerase genes.
CAST/EiJ Terc+/- mice exhibited a DC-like phenotype after six generations of intercrosses23, whereas Terth/- mice were intercrossed for 16 generations and only displayed mild phenotypes associated with telomere dysfunction. Terth/- mice achieved significantly shorter telomeres compared to either Tert+/- or Terc+/- intercrosses23,26. This difference may also be related to the observation that the hmTert gene may function more effectively than the mTert gene in repairing critically shortened telomeres in germ cells (Fig. 2g, h). The protection of germ cells with short telomeres may enable mice with the hmTert gene to maintain human-like short telomeres.
The average telomere lengths in HuT mice are comparable to those observed in humans. TRF analyses revealed that telomere signals in Terth/- and Terth/h mice consist of discrete bands measuring 15–20 kb, along with a lower-molecular-weight smear reminiscent of human telomeres. Notably, these discrete telomere bands were distinct from interstitial telomere bands, as they varied among individual mice and were highly susceptible to Bal-31 exonuclease digestion. This suggests that these bands likely represent ultra-long telomeres present in C57BL/6 J mice that have not reached the length of human telomeres. Since the shortest telomeres are prone to dysfunction and can limit cell proliferation, the presence of a few longer telomeres may have minimal impact on aging and tumorigenesis. Moreover, it is anticipated that these long telomeres will continue to shorten over successive generations during the breeding of these mice.
It is widely recognized that the shortest telomeres, rather than the average telomere length, are crucial in maintaining chromosomal stability and triggering replicative senescence43. Therefore, it is not surprising that Terth/h mice with very short telomeres (HuT mice) remain in good health during the early stages of life. Previous research has documented variations in the rates of telomere shortening among different mammalian species3. For example, human peripheral blood cell telomeres shortened at a chronological rate of 31–72 bp/year, while C57BL/6 mice experienced a much more rapid rate of 7000 bp/year44. Although the rate of telomere attrition in HuT mice has yet to be determined, the significantly reduced telomere reserve in HuT mice may impose limitations on cell proliferation and tissue renewal. Indeed, our experiments revealed that colonocyte proliferation was constrained in HuT mice under a DSS-induced ultracerative colitis condition. Telomere attrition has been implicated as a counting mechanism for cellular replicative senescence in vitro and organismal aging in vivo. Our objective is to further investigate the aging process in HuT mice and determine whether their cells undergo replicative senescence.
The development of HuT mice has the potential to catalyze a paradigm shift in cancer and aging research using mouse models. First, the presence of short telomeres in HuT mice is likely to impede tumor cell proliferation during cancer development, as dysfunctional telomeres can induce chromosomal instability and significantly impact the course of cancer progression. Second, the limited telomere reserve in HuT mice and humans can also exert a profound influence on the tumor microenvironment and the host immune system. Recent research suggests that host T cell immune deficiency, rather than tumor cell chromosome instability, predisposes patients with short telomere syndromes to squamous cancers45. Thus, telomere-driven immune senescence may contribute to the high tumor incidences in aged humans and HuT mice. Third, chemotherapy drugs can induce DNA damage in both normal cells and tumor cells. Considering that tissues and cells with short telomeres are inherently more susceptible to such damage, this vulnerability could synergize with the cytotoxic effects of cancer drugs, particularly in older humans and HuT mice. As a result, HuT mice may provide a more accurate preclinical model for evaluating drug toxicity. Due to the vast difference in telomere length between HuT mice and other laboratory mice, introducing a second transgene while maintaining short telomeres can be challenging. However, it is feasible to edit genes in HuT mice by injecting CRISPR/Cas9 components into zygotes. Taken together, HuT mice are expected to serve as a mouse model for studying tumorigenesis and age-related diseases, offering valuable insights into cancer development and potential therapies.
In conclusion, the process of telomere attrition plays a pivotal role in the development of various human diseases. To gain a deeper understanding of the fundamental disparities in telomerase regulation and telomere homeostasis between mice and humans, we have developed genetically engineered HuT mice. Our data demonstrates that the hmTert allele is a functional gene and contributes to establishing a telomere length setpoint in mice that closely resembles that of humans. These mice exhibit humanized telomerase expression and telomere length homeostasis, and ongoing investigations are focused on studying their aging process and susceptibility to age-related diseases. There may be additional differences between mouse and human cells beyond the dissimilarities in telomere homeostasis. For example, the p16Ink4a/Rb tumor suppressor pathway is more robust in human cells46 and mouse cells are more sensitive to oxidative stress31. The molecular mechanisms underlying these distinctions may be probed by determining whether and how somatic cells from HuT mice acquire the Hayflick limit. Therefore, the development of HuT mice represents an opportunity to address previously unattainable inquiries concerning human aging and cancer, opening avenues for scientific exploration.
Methods
Generation of mice with hmTert alleles
The engineering of the hmTert gene (Fig. 1a) in mouse ESCs (Terth/+) was previously reported13. The ESCs (G4, 129xC57BL/6)47 were injected into blastocysts of C57BL/6 J albino hosts. The resulting male chimera mice were bred with C57BL/6 J female mice. Mice with a germline hmTert allele (F0) were obtained and crossed again with C57BL/6 J mice to generate F1 Tert+/h mice with 88% C57BL/6 J background. The F1 Tert+/h mice were backcrossed with C57BL/6 J mice for three generations before Terth/+ mice were used for mRNA expression analyses. Tert+/h mice were also crossed with Tert-knockout (Tert-KO) mice48. Mice are maintained at 23 ± 2°C, 40–60% humidity, and 12:12 h light/dark cycle conditions and with ad libitum food, water and enriched environment available. All animal experiments were conducted in accordance with the NIH Guide for the Care and Use of Animals and were approved under protocols ASAF#6659 and ASAF#6693 by the Institutional Animal Care and Use Committee of Washington State University.
Gene expression and telomerase activity
Total RNAs were extracted from mouse and human tissues (Penn State Cancer Institute Tissue Bank)8. mRNA expression analyses were performed using quantitative PCR and data were normalized to 18S ribosomal RNA. Primer sequences are provided in Supplementary Table 3. Telomerase activities were determined using a modified telomeric repeat amplification protocol (TRAP) assay13. Tissues and cell extracts were adjusted to same concentration and 0.5 µg samples were used in each reaction. ESCs (Terth/h, Tert+/+, and Tert+/h) served as positive controls.
Telomere length measurements
Telomere lengths were measured using two independent methods. For TRF analysis13, genomic DNAs were digested with HinfI and RsaI, and subjected to pulsed-field gel electrophoresis using CHEF-DR III Pulsed field Electrophoresis Systems (for telomeres over 20 kb) or regular 0.6% Agarose gel (for telomeres less than 20 kb), followed by Southern blotting using a (TTAGGG)3-Biotin probe. Hybridization signal was developed with Chemiluminescent Nucleic Acid Detection Module Kit (Thermo Fisher). For exonuclease digestion, splenocyte genomic DNAs (5 µg) were digested with 1 unit Bal-31 nuclease (Takara) at 30 °C for 5, 10, and 15 min, and reactions were stopped with 33 mM EGTA. Purified DNA samples were analyzed by TRF analysis. Telomere lengths were also measured by Flow-FISH. Telomere signals were detected by hybridization to FAM-(CCCTAA)3 oligonucleotide and converted to arbitrary units of molecular equivalents of soluble fluorescence.
T cell culture and stimulation
CD4+ and CD8+ T cells were isolated from splenocytes using MojoSort™ T cells isolation kits (Biolegend, USA). Briefly, spleens were collected in RPMI-1640 medium (Gibco, USA) as soon as mice were sacrificed. The splenocytes were isolated through spleen crushing and filtration using a 70 μm strainer. Red blood cells were lysed with 1x RBC lysis buffer, splenocytes were incubated with biotin-antibody cocktails on ice for 20 min, and streptavidin nanobeads beads were added for 5 min, followed by pulldown of non-T cells. 6-well plates were coated by 10 μg/ml anti-mouse CD3e antibody in PBS for 2 h at 37 °C. The isolated CD4 or CD8 T cells were stimulated in CD3e antibody coated plates, together with added 2 μg/ml anti-mouse CD28 antibody for 24 h. 10 ng/ml IL-2 was added at 48, 72 and 96 h and activated T cells were examined for cell proliferation using Click-iT® Plus EdU Flow Cytometry Assay Kit (ThermoFisher, USA) or harvested for gene expression analyses.
Histological analyses
Fresh intestines were fixed in 10% formalin solution for 48 h, embedded in paraffin, sectioned at 5 μm, and stained with hematoxylin and eosin (H&E). Mouse testes were fixed in Bouin’s fixative for 3 days and similarly processed. Slides were evaluated for aberrant seminiferous tubules in a blinded fashion and at least three mice of each experimental group were analyzed.
Hematological analysis
Whole blood samples were collected from submandibular veins of 3–6 months mice. Numbers of WBCs, RBCs, platelets, lymphocytes, neutrophils, and monocytes were determined on a Hemogram Analyzer (Abaxis HM5). For Bone marrow ablation, age-matched animals (11 months) were injected intraperitoneally with 150 mg/kg 5-Fluorouracil (5-FU, Selleckchem, Cat. S1209) was given through23. Blood samples (60 µl) were collected via the cheek vein before the 5-FU injection, and at 6- and 11-days post-injection.
FACS analyses
Peripheral blood, splenocytes, and femur bone marrow (BM) were collected from mice of 3–6 months. Peripheral blood was washed twice with PBS by centrifugation in the presence of heparin, followed by removing RBCs with 1x RBC lysis buffer (Thermo Fisher, USA). WBCs were counted using hemocytometers, stained with CD4, CD8, and CD19 antibodies, and analyzed by flow cytometry. Splenocytes and bone marrow cells, flushed from femurs by syringes with 20 ml PBS buffer, were similarly processed for cell counting, FACS staining, and analyses.
Dextran sulfate socium treatment
Female mice of 7–8 months of age were subjected to a 6-day period of 3% Dextran sulfate sodium (DSS) (Cat. 160110, MP biomedical) administration in drinking water, followed by a day of regular drinking water before tissue collection. Body weight of treated mice were monitored. For EdU labeling, 150 µl of 10 mg/ml EdU solution was injected intraperitoneally into mice two hours before sacrifice. Paraformaldehyde-fixed tissues were embedded in OCT matrix. EdU was detected using the Click-iT Plus EdU Cell Proliferation Kit (Alexa Fluor™ 594, C10639, ThermoFisher). Tissue sections were stained overnight with an anti-E-Cadherin antibody (1:250, Cat. 610181, BD Biosciences), followed by incubation with FITC-conjugated secondary antibody and Hoechst 33342 (1:2000). Images were captured using a Zeiss AXIO Image M2 microscope and processed using FIJI and Adobe Photoshop 2023.
Mouse embryo fibroblasts (MEFs) and telomere analyses
MEFs were isolated from E13.5 embryos of G4.16 Terth/- intercrosses as described previously49 and cultured with DMEM with 10% FBS and incubated in 5% CO2 and 3% oxygen. Passage 4 MEFs were seeded into 8-well chamber slides (LabTek, 154534) and stained with antibodies against TRF2 and γH2AX as described50. Cell images were captured with a ZEISS M2 microscope and analyzed using ImageJ software. Co-localized foci were analyzed with ComDet V0.5.5 plugins in ImageJ.
Telomere length was quantified by qFISH using peptide nucleic acid (PNA) probes as described previously13. Metaphase spreads were prepared, and qFISH was conducted per the manufacturer’s instructions (PNA Bio Inc.), using telomere-specific TelC-Cy3 (CCCTAA repeats) and centromere-specific CENPB-Alxa488 (ATTCGTTGGAAACGGGA) PNA probes. Fluorescence images were captured on an Axioscope Z2 (Zeiss, Germany), and telomere signal integration was conducted from a minimum of 15 metaphases. Quantitative analysis was done as described by Perner et al.51 using Isis software (MetaSystems, Germany).
Statistical analysis
GraphPad Prism 10 was used for statistical analyses, including two-tailed student’s t-tests for comparisons of gene expression, one or two-way Anova for comparisons of body and organ weights and cell numbers among different experimental groups, and logrank tests for mouse survival curves.
Reporting summary
Further information on research design is available in the Nature Portfolio Reporting Summary linked to this article.
Data availability
All data supporting the findings of this study are available within the paper and its Supplementary Information. Source data are provided with this paper.
References
Venteicher, A. S. et al. A human telomerase holoenzyme protein required for Cajal body localization and telomere synthesis. Science 323, 644–648 (2009).
Okuda, K. et al. Telomere length in the newborn. Pediatr. Res. 52, 377–381 (2002).
Whittemore, K., Vera, E., Martinez-Nevado, E., Sanpera, C. & Blasco, M. A. Telomere shortening rate predicts species life span. Proc. Natl Acad. Sci. USA 116, 15122–15127 (2019).
Kirwan, M. & Dokal, I. Dyskeratosis congenita, stem cells and telomeres. Biochim Biophys. Acta 1792, 371–379 (2009).
Greenberg, R. A. et al. Short dysfunctional telomeres impair tumorigenesis in the INK4a(delta2/3) cancer-prone mouse. Cell 97, 515–525 (1999).
Gomes, N. M. et al. Comparative biology of mammalian telomeres: hypotheses on ancestral states and the roles of telomeres in longevity determination. Aging Cell 10, 761–768 (2011).
Horikawa, I. et al. Differential cis-regulation of human versus mouse TERT gene expression in vivo: identification of a human-specific repressive element. Proc. Natl Acad. Sci. USA 102, 18437–18442 (2005).
Jia, W. et al. A BAC transgenic reporter recapitulates in vivo regulation of human telomerase reverse transcriptase in development and tumorigenesis. FASEB J. 25, 979–989 (2011).
Blasco, M. A. et al. Telomere shortening and tumor formation by mouse cells lacking telomerase RNA. Cell 91, 25–34 (1997).
Singh, M., Murriel, C. L. & Johnson, L. Genetically engineered mouse models: closing the gap between preclinical data and trial outcomes. Cancer Res. 72, 2695–2700 (2012).
Palm, W., Hockemeyer, D., Kibe, T. & de Lange, T. Functional dissection of human and mouse POT1 proteins. Mol. Cell Biol. 29, 471–482 (2009).
Smoom, R. et al. Telomouse-a mouse model with human-length telomeres generated by a single amino acid change in RTEL1. Nat. Commun. 14, 6708 (2023).
Cheng et al. Engineering a humanized telomerase reverse transcriptase gene in mouse embryonic stem cells. Sci. Rep. 9, 9683 (2019).
Erdmann, N., Liu, Y. & Harrington, L. Distinct dosage requirements for the maintenance of long and short telomeres in mTert heterozygous mice. Proc. Natl Acad. Sci. USA 101, 6080–6085 (2004).
Patrick, M. S. et al. Human T Cell Differentiation Negatively Regulates Telomerase Expression Resulting in Reduced Activation-Induced Proliferation and Survival. Front Immunol. 10, 1993 (2019).
Weng, N. P., Levine, B. L., June, C. H. & Hodes, R. J. Regulated expression of telomerase activity in human T lymphocyte development and activation. J. Exp. Med. 183, 2471–2479 (1996).
Herrera, E. et al. Disease states associated with telomerase deficiency appear earlier in mice with short telomeres. EMBO J. 18, 2950–2960 (1999).
Rudolph, K. L. et al. Longevity, stress response, and cancer in aging telomerase-deficient mice. Cell 96, 701–712 (1999).
Choudhury, A. R. et al. Cdkn1a deletion improves stem cell function and lifespan of mice with dysfunctional telomeres without accelerating cancer formation. Nat. Genet 39, 99–105 (2007).
Lee, H. W. et al. Essential role of mouse telomerase in highly proliferative organs. Nature 392, 569–574 (1998).
Raval, A. et al. Reversibility of Defective Hematopoiesis Caused by Telomere Shortening in Telomerase Knockout Mice. PLoS One 10, e0131722 (2015).
Wagner, C. L. et al. Short telomere syndromes cause a primary T cell immunodeficiency. J. Clin. Invest 128, 5222–5234 (2018).
Hao, L. Y. et al. Short telomeres, even in the presence of telomerase, limit tissue renewal capacity. Cell 123, 1121–1131 (2005).
Factor-Litvak, P. et al. Leukocyte Telomere Length in Newborns: Implications for the Role of Telomeres in Human Disease. Pediatrics 137 https://doi.org/10.1542/peds.2015-3927 (2016).
(The Jackson Laboratory, 2007).
Chiang, Y. J. et al. Telomere length is inherited with resetting of the telomere set-point. Proc. Natl Acad. Sci. USA 107, 10148–10153 (2010).
Thongon, N. et al. Hematopoiesis under telomere attrition at the single-cell resolution. Nat. Commun. 12, 6850 (2021).
Chakravarti, D. et al. Telomere dysfunction instigates inflammation in inflammatory bowel disease. Proc Natl Acad Sci USA 118 https://doi.org/10.1073/pnas.2024853118 (2021).
Chassaing, B., Aitken, J. D., Malleshappa, M. & Vijay-Kumar, M. Dextran sulfate sodium (DSS)-induced colitis in mice. Curr. Protoc. Immunol. 104, 15 25 11–15 25 14 (2014).
Wang, J. et al. Resveratrol Attenuates Inflammatory Bowel Disease in Mice by Regulating SUMO1. Biol. Pharm. Bull. 43, 450–457 (2020).
Parrinello, S. et al. Oxygen sensitivity severely limits the replicative lifespan of murine fibroblasts. Nat. Cell Biol. 5, 741–747 (2003).
Zhang, F., Cheng, Wang, S. & Zhu, J. Crispr/Cas9-mediated cleavages facilitate homologous recombination during genetic engineering of a large chromosomal region. Biotechnol. Bioeng. 117, 2816–2826 (2020).
Beattie, T. L., Zhou, W., Robinson, M. O. & Harrington, L. Reconstitution of human telomerase activity in vitro. Curr. Biol. 8, 177–180 (1998).
Chen, J. L. & Greider, C. W. Determinants in mammalian telomerase RNA that mediate enzyme processivity and cross-species incompatibility. Embo J. 22, 304–314 (2003).
de Lange, T. Shelterin: the protein complex that shapes and safeguards human telomeres. Genes Dev. 19, 2100–2110 (2005).
Ding, H. et al. Regulation of murine telomere length by Rtel: an essential gene encoding a helicase-like protein. Cell 117, 873–886 (2004).
Cheng, D. et al. Regulation of human and mouse telomerase genes by genomic contexts and transcription factors during embryonic stem cell differentiation. Sci. Rep. 7, 16444 (2017).
Chiba, K. et al. Cancer-associated TERT promoter mutations abrogate telomerase silencing. Elife 4 https://doi.org/10.7554/eLife.07918 (2015).
Wang, S., Zhao, Y., Hu, C. & Zhu, J. Differential repression of human and mouse TERT genes during cell differentiation. Nucleic Acids Res. 37, 2618–2629 (2009).
Hammoud, S. S. et al. Distinctive chromatin in human sperm packages genes for embryo development. Nature 460, 473–478 (2009).
Tanemura, K. et al. Dynamic rearrangement of telomeres during spermatogenesis in mice. Dev. Biol. 281, 196–207 (2005).
Meznikova, M., Erdmann, N., Allsopp, R. & Harrington, L. A. Telomerase reverse transcriptase-dependent telomere equilibration mitigates tissue dysfunction in mTert heterozygotes. Dis. models mechanisms 2, 620–626 (2009).
Hemann, M. T., Strong, M. A., Hao, L. Y. & Greider, C. W. The shortest telomere, not average telomere length, is critical for cell viability and chromosome stability. Cell 107, 67–77 (2001).
Vera, E., Bernardes de Jesus, B., Foronda, M., Flores, J. M. & Blasco, M. A. The rate of increase of short telomeres predicts longevity in mammals. Cell Rep. 2, 732–737 (2012).
Schratz, K. E. et al. T cell immune deficiency rather than chromosome instability predisposes patients with short telomere syndromes to squamous cancers. Cancer Cell 41, 807–817 e806 (2023).
Smogorzewska, A. & de Lange, T. Different telomere damage signaling pathways in human and mouse cells. EMBO J. 21, 4338–4348 (2002).
George, S. H. et al. Developmental and adult phenotyping directly from mutant embryonic stem cells. Proc. Natl Acad. Sci. USA 104, 4455–4460 (2007).
Liu, Y. et al. The telomerase reverse transcriptase is limiting and necessary for telomerase function in vivo. Curr. Biol. 10, 1459–1462 (2000).
Zhu, J. Y., Abate, M., Rice, P. W. & Cole, C. N. The ability of simian virus 40 large T antigen to immortalize primary mouse embryo fibroblasts cosegregates with its ability to bind to p53. J. Virol. 65, 6872–6880 (1991).
Zhang, J. et al. Telomere dysfunction in Tert knockout mice delays Braf(V600E) -induced melanoma development. Int J. Cancer 154, 548–560 (2024).
Perner, S. et al. Quantifying telomere lengths of human individual chromosome arms by centromere-calibrated fluorescence in situ hybridization and digital imaging. Am. J. Pathol. 163, 1751–1756 (2003).
Acknowledgements
This work was supported in part by NIH Grants R21OD021432, R01AG073423, and R35GM149529 to JZ, a Team Science Award (ME220261) from Department of Defense to JZ and GPR, and Health Sciences & Services Authority (HSSA) of Spokane County. We would like to express our gratitude to Drs. Yie Liu and Lea Harrington for their generous gift of mTert knockout mice. We thank the Program of Laboratory Animal Resources (PLAR) and Histology Core of Washington State University Spokane. We also thank Dr. Gang Chen for his support on statistical data analyses.
Author information
Authors and Affiliations
Contributions
Conception and design: J Zhu; Data acquisition: F Zhang, D Cheng, KI Porter, EA Heck; Data analysis and interpretation: F Zhang, D Cheng, S.Wang, J Zhu; Manuscript writing and revision: J Zhu, F Zhang, D Cheng, S Wang, CJ Davis, GP Robertson; Study supervision: J Zhu.
Corresponding author
Ethics declarations
Competing interests
The authors declare no competing interest.
Peer review
Peer review information
Nature Communications thanks the anonymous reviewer(s) for their contribution to the peer review of this work. A peer review file is available.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary information
Source data
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, 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 you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. 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-nc-nd/4.0/.
About this article
Cite this article
Zhang, F., Cheng, D., Porter, K.I. et al. Modification of the telomerase gene with human regulatory sequences resets mouse telomeres to human length. Nat Commun 16, 1211 (2025). https://doi.org/10.1038/s41467-025-56559-6
Received:
Accepted:
Published:
DOI: https://doi.org/10.1038/s41467-025-56559-6
This article is cited by
-
A promising new TERT activator: unraveling the molecular mechanisms behind its natural aging mitigation
Frontiers of Medicine (2025)