Open Access

A novel nutraceutical formulation increases telomere length and activates telomerase activity in middle‑aged rats

  • Authors:
    • Aristidis Tsatsakis
    • Elisavet Renieri
    • Dimitris Tsoukalas
    • Ana Maria Buga
    • Evangelia Sarandi
    • Elena Vakonaki
    • Persefoni Fragkiadaki
    • Athanasios Alegakis
    • Dragana Nikitovic
    • Daniela Calina
    • Demetrios A. Spandidos
    • Anca Oana Docea
  • View Affiliations

  • Published online on: October 27, 2023     https://doi.org/10.3892/mmr.2023.13119
  • Article Number: 232
  • Copyright: © Tsatsakis et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Telomeres are major contributors to cell fate and aging through their involvement in cell cycle arrest and senescence. The accelerated attrition of telomeres is associated with aging‑related diseases, and agents able to maintain telomere length (TL) through telomerase activation may serve as potential treatment strategies. The aim of the present study was to assess the potency of a novel telomerase activator on TL and telomerase activity in vivo. The administration of a nutraceutical formulation containing Centella asiatica extract, vitamin C, zinc and vitamin D3 in 18‑month‑old rats for a period of 3 months reduced the telomere shortening rate at the lower supplement dose and increased mean the TL at the higher dose, compared to pre‑treatment levels. TL was determined using the Q‑FISH method in peripheral blood mononuclear cells collected from the tail vein of the rats and cultured with RPMI‑1640 medium. In both cases, TLs were significantly longer compared to the untreated controls (P≤0.001). In addition, telomerase activity was increased in the peripheral blood mononuclear cells of both treatment groups. On the whole, the present study demonstrates that the nutraceutical formulation can maintain or even increase TL and telomerase activity in middle‑aged rats, indicating a potential role of this formula in the prevention and treatment of aging‑related diseases.

Introduction

Telomeres have been characterized as molecular clocks, playing a pivotal role in cell division arrest. These chromatin structures, composed of tandem DNA sequence repeats coated by the sheltering protein complex, are formed at chromosomal termini as protective caps against DNA degradation and recombination to ensure genomic stability and integrity (1). Each cell division leads to shorter telomeres due to the inability of DNA polymerase to fully replicate the end part of the lagging strand of DNA, described as the end replication problem. Telomere length (TL) loss can be compensated by telomerase, which functions as a reverse transcriptase, adding telomeric repeats at the end of chromosomes. The telomerase complex consists of a catalytic unit denominated as telomerase reverse transcriptase (TERT) and the telomerase RNA component (2). This ribonucleoprotein polymerase is abundantly expressed in highly proliferating cells, such as stem cells; however, its expression in somatic cells is reduced or absent (3). In cells with a low telomerase activity, telomeres continue to shorten, and when they reach a critically short length, cells undergo apoptosis or cell senescence. Beyond this critical length, excessive telomere shortening can lead to chromosomal fusions and genomic instability that increase the risk of cancer tumorigenesis through genomic alterations. Ultimately, cells need to reactivate telomerase to become immortal during tumor progression (4). The replicative past and potential of cells can be assessed through TL and telomerase activity estimation methods, focusing on short telomere load and the rate of telomere shortening that reflects cellular aging (5).

Telomere attrition and cellular senescence are identified as major contributors to the physiological process of aging (6). Furthermore, increased aging and age-related diseases have been shown to be associated with the disruption of telomere homeostasis, which has been mainly attributed to increased levels of oxidative stress and inflammation (7). To date, cumulative oxidative stress and inflammation have been strongly associated with age-related TL shortening (8). Research on the associations of telomere shortening with age has revealed that the inhibition of telomere attrition via telomere gene therapy in mice results in delayed physical aging (9). By contrast, it has been suggested that a short leukocyte TL is an independent risk factor that accounts for functional decline in elderly European populations (10). Furthermore, critically short telomeres have been shown to be associated with higher mortality rates and shorter lifespans (11).

The human lifespan has been considerably extended, leading to an increase in the elderly populations globally, stressing the need to develop ‘healthy aging’ strategies that will prevent and treat aging-related diseases (12). Research focusing on identifying effective natural or synthetic compounds with anti-aging properties has intensified in recent years (13). Specifically, there is increasing evidence to indicate that bioactive compounds, including nutrients and vitamins present in foods and supplements with antioxidant and anti-inflammatory properties can ameliorate age-related phenotypes, including telomere shortening (14,15). Of note, telomerase activity can be positively modified by natural compounds, such as Astragalus membranaceus plant extracts, cycloastragenol (CAG), curcumin C3 and vitamins, among others, which enable telomerase activation (13). Moreover, in a previous study, the authors demonstrated that a formulation containing Centella asiatica (C. asiatica) extract markedly enhanced telomerase activity in human peripheral blood mononuclear cells (PBMCs) in vitro (16). Notably, the demonstrated increase in telomerase activity was the highest reported in vitro, to date, at least to the best of our knowledge (16). Another study also demonstrated that the administration of the formulation containing C. asiatica extract, vitamin C, zinc and vitamin D3 for a period of 3 months restored TERT expression and increased telomerase activity in the brains of middle-aged rats. Furthermore, a structural reversibility effect was observed in the brains of middle-aged rats treated with the formulation, close to the differentiation of the grey matter from a young control group (17). Supporting evidence was provided from a complementary behavioral study of the same treated rats, where supplementation with the formulation improved the locomotor activity and decreased stress significantly in a dose-dependent manner in aged rats (12).

The present study aimed to examine the effects of the nutraceutical formulation containing C. asiatica extract, vitamin C, zinc and vitamin D3 on telomerase activity and TL using a murine model. The obtained results shed light onto the potential association of supplement administration with the abating of the aging process.

Materials and methods

Animal model

All animals were obtained from the University of Medicine and Pharmacy of Craiova Animal House, Craiova Romania, authorization no. 76/20.04.2016. The Ethical Committee of the University of Medicine and Pharmacy of Craiova, Craiova, Romania, approved the animal study (protocol no. 102/23.09.2019). All the procedures were according to the European directives for animal experiments (E.U. Directive 2010/63/E.U.as amended by Regulation E.U.2019/1010). A total of 24 Sprague-Dawley (CD-SD) male rats divided into four groups were used in the present study. Of the total 24 animals, 18 rats were 18 months old [body weight (bw) range, 500–580 g]. The middle-aged animals were randomly assigned to three groups as follows: Old group 1, old group 2 and old control group, with 6 rats in each group. The remaining 6 rats were 3 months old, and were denominated as the young control group. Male rats were selected over female animals to reduce the variability of responses due to potentially asynchronous estrous cycles (18). All animals were acclimatized to their new housing conditions for 2 weeks before the commencement of the study. The rats were kept in cages containing 2 or 3 animals during the study, under standard conditions with a constant temperature (22±1°C) and humidity (50±10%) and a 12-h dark/light cycle, receiving free access to standard animal feed and tap water. The study animals received the Reverse™ (Natural Doctor S.A.) supplement treatments presented in Table I for 3 months and were evaluated daily for signs of morbidity and mortality. At the end of the experiment (3 months), the animals received 5% sevoflurane anesthesia via nasal administration and were monitored until they were fully anesthetized. The rats were then sacrificed by exsanguination from the abdominal aorta for the analysis. As stated in current regulatory testing guidelines, when animals are evidently in pain, exhibiting signs of severe and enduring distress, or characterized as moribund, they were humanely euthanized rather than allowing them to survive to the end of the scheduled study period. However, in the present study, no animal death was observed (19,20). The animals were examined daily for any walking impairments that prevented access to food or water, excessive weight loss and emaciation, a lack of physical or mental alertness, difficulty breathing, and an inability to stand upright for long periods of time.

Table I.

Treatments administered to the animal groups for 3 months daily and sampling time points (pre-and post-treatment).

Table I.

Treatments administered to the animal groups for 3 months daily and sampling time points (pre-and post-treatment).

Animal groupsOld group 1 (n=6)Old group 2 (n=6)Old control group (n=6)Young control group (n=6)
Treatments1 capsule per kg/body weight of Reverse™ supplement2 capsules per kg/body weight of Reverse™ supplement1.5 ml corn oil1.5 ml corn oil
Age (months) (pre-treatment)1818183
Age (months) (post-treatment)2121216
Treatment dose selection and administration

Reverse™ (Natural Doctor S.A.), notified as a food supplement (Notification no. 6704/21 1 2020) at the Greek National Organization for Medicines was administered at a dose of 1 or 2 capsules/kg bw/day. The supplement contains 9 mg C. asiatica extract (consisting of a >90% high purity single chemical entity, as assessed using high-performance liquid chromatography and gas chromatography) (21), vitamin C (200 mg as magnesium ascorbate), zinc (5 mg as zinc citrate) and vitamin D3 (50 µg as cholecalciferol) per capsule.

The doses administered to the rats were extrapolated from the human doses using the correction factor (fc) ratio for the species used and the safety factor value for humans. The fc is estimated as the ratio of the mean bw (kg) of the species used, to the species' body surface area (m2), according to the Food and Drug Administration guidelines being 6 for rats and 37 for humans, respectively. The fc ratio for drug dose conversion from rats to humans corresponds to a value of 6.2. The safety factor (fs) value for converting rat doses to humans is 1043. The reference bw for humans is 60 kg (22), which means the dose for humans (dH) is calculated to 1/60 capsules per kg bw for a single dose of 1 capsule and 2/60 capsules per kg bw for a single dose of 2 capsules. Based on the information provided above, the dose in rats (dR) was calculated as follows: dR=dH × fcratio × fs (Equation 1).

Based on Equation 1, the old group 1 receiving the equivalent of the 1 capsule/day human dose, was administered dR=1/60 capsules × 6.2×10=1.03 capsule per kg/bw. The Old Group 2, receiving the equivalent of the 2 capsules/day human dose, was issued with dR=2/60 capsule × 6.2×10=2.06 capsule per kg/bw.

Prior to treatment administration, the content of the capsules was suspended in corn oil, widely used as an inert suspension agent for non-water-soluble drugs in animal experiments (23), as a stock suspension with 1 capsule/1 ml corn oil concentration or 2 capsules/1 ml corn oil. Each rat received the equivalent of 1 capsule per kg/bw or 2 capsules per kg/bw, once per day at the same time for 3 months (Table I). The respective equivalent dose calculated according to the bw of each animal was diluted with corn oil until the final volume of 1.5 ml and administered by gavage.

The dose administered was within clinical recommendations relative to bw and similar to other animal studies evaluating the effects of vitamin C (24,25), vitamin D3 (25-hydroxyvitamin D) (26,27) and zinc (28). Specifically, as regards vitamin D3, the administered dose per capsule was below the upper limit and was safely administered without medical supervision. Concerning the safety levels of the ingested dose for vitamin D, the most common concern is the risk of hypercalcemia, which may be evoked when the serum 25-hydroxyvitamin D levels exceed 700 ng/ml, which is >7-fold higher than the levels of sufficiency (29). Cholecalciferol was selected over ergocalciferol since ergocalciferol is less stable and less potent than cholecalciferol, characterized as the only vitamin D form suitable for supplementation (21). For C. asiatica, there is no established clinical recommendation, at least to the best of our knowledge. The dose administered in the present study was markedly lower than the levels reported in the literature (30).

Sample collection and preparation for quantitative-fluorescent in situ hybridization (Q-FISH)

Peripheral blood from the tail vein (2 ml) was collected from each rat at the start of the experiment (pre-treatment) and after 3 months (post-treatment). The collected heparinized blood cultured in 50-ml falcon tubes with RPMI-1640 culture medium supplemented with 10% fetal bovine serum (FBS), 1% L-glutamine, 1% penicillin, phytohemagglutinin 100 µg/ml and streptomycin was stimulated for 72 h in a CO2 incubator with phytohemagglutinin. The cells were incubated at 37°C with 10 µg/ml colcemid for 2 h to obtain metaphases, followed by KCl hypotonic shock, and then were harvested and fixed in methanol/acetic acid (3:1). All reagents were obtained from MilliporeSigma.

Quantitative-fluorescent in situ hybridization (Q-FISH)

Several drops of a fixative solution containing cells from each culture were applied to three slides. Slides with the fixative solution containing cells were thereupon dried and incubated on a hot plate (55°C) overnight prior to hybridization. Fluorescence staining for telomeric DNA was performed using the peptide nucleic acid (PNA) fluorescent probe Cy3-(C3TA2)3 obtained from Panagene Inc. Each slide was hybridized with 20 µl of 60% formamide (MilliporeSigma) and 0.3 mg/ml Cy3 (C3TA2)3 PNA probe (HLB Panagene) diluted in 20 mM Tris pH 7.4 (MilliporeSigma) and covered using a coverslip (76×26 mm). Cell DNA was denatured by heat treatment for 10 min at 85°C. Following hybridization for 2 h at room temperature, the slides were washed at 55°C with a washing solution containing 0.05% Tween-20 (MilliporeSigma) (2×10 min). The chromosome preparations were counterstained with 4′,6-diamino-2-phenylindole (DAPI) (Thermo Fisher Scientific, Inc.) (0.5 µl/ml SSC) at room temperature for 20 min and then washed with SSC solution (MilliporeSigma) (2×2 min). The slides were air-dried and covered using mounting media and a coverslip before proceeding with image acquisition.

Image analysis

The analysis of FISH Images from metaphase cells was performed using a Leica TCS Sp8 inverted laser scanning spectral confocal microscope (Leica Microsystems GmbH). For each slide, a total of 20–30 optical sections were captured. Metaphase spread images were captured using a 63X objective and a charge-coupled device camera at a 1024×1024 pixel resolution and 8-bit depth with a step of 250 nm. A 405-nm laser was used for the excitation and detection of DAPI for nuclear staining. In comparison, a 568-nm laser was utilized for the excitation and detection of Alexa Fluor 647 for telomere staining. Exposure and gain settings remained unaltered between captures to avoid differences between the replicates. For each slide, >10 different scanned images were obtained. The maximum projections and deconvolution of the images were performed using Leica Q-FISH software (Leica Application Suite-Advanced Fluorescence version 3.1.3 for Leica TCS SP8; Leica Microsystems, Inc.). The quantification of telomere fluorescence intensity was performed twice by two different researchers using ImageJ software 1.8.0 (National Institutes of Health). A total of two calibration steps were undertaken to ensure the accuracy of telomere fluorescence intensity quantification. First, images of fluorescent beads (orange beads, size 0.2 µm, Thermo Fisher Scientific, Inc.) were obtained before the samples. Fluorescence intensities of the beads (obtained using ImageJ software) were used to normalize and adjust the lamp intensity and alignment prior to sample analysis. Second, for the determination of the telomere fluorescence intensity the L5178Y-S cells (cat. no. 93050408; European Collection of Authenticated Cell Cultures) were included as a calibration standard in each experiment. The mean value of L5178Y-S cell telomere fluorescence intensities was used to normalize the respective values of samples between slides. Telomere fluorescence values were converted into kb according to the telomere fluorescence intensity of L5178Y-S cells that have an established TL of ~7 kb (31).

Telomerase activity assay

Blood samples for telomerase activity assay were collected from each animal at the beginning of the experiment (pre-treatment) and after 3 months (post-treatment). PBMCs were isolated from the blood samples using Ficoll-Hypaque (MilliporeSigma) gradient centrifugation at 277 × g for 5 min at room temperature in 15-ml falcon tubes. The extracted PBMCs were cultured in DMEM (F0455, Biochrom AG) supplemented with 10% FBS (10500-064, Invitrogen; Thermo Fisher Scientific, Inc.) with 4 mM glutamine (XCT1715, Biosera) and antibiotics (gentamycin; 15710-049, Gibco; Thermo Fisher Scientific, Inc.; and 100 U/ml penicillin/streptomycin; LMA4118, Biosera). Telomerase activity was quantified using a TeloTAGGG telomerase PCRELISA TRAP kit (MilliporeSigma), based on the telomeric repeat amplification protocol, as previously described (16). All measures for each condition were performed in triplicate.

Statistical analysis

The obtained data on TL were implemented into the specialized spreadsheet (BIOTEL 2.4) (32) to produce TL statistics, including percentiles, medians and telomere distribution. The TL data were further statistically analyzed using the IBM SPSS Statistics 24.0 package (IBM Corp.), while data for telomerase activity were processed using GraphPad Prism version 5.0 for Windows (GraphPad Software, Inc., www.graphpad.com). TL data are presented in two forms as follows: i) As the mean with 95% confidence intervals (CIs); and ii) as the median and interquartile range (IQR). Parametric (independent samples t-test) and non-parametric tests (Mann-Whitney test) for independent samples were applied to compare the means of two independent groups or, additionally/alternatively, the distribution of TL data. A similar approach using one-way ANOVA (parametric) and Kruskal-Wallis (non-parametric) for independent samples was applied to compare means for more than two separate groups or, additionally/alternatively, the distribution of TL data. These analyses (ANOVA and Kruskal-Wallis) was followed by post hoc analyses using the Bonferroni adjusted t-test, SNK, Dunnett's for ANOVA, and Dunn's test for Kruskal-Wallis.

Differences in paired design (pre-and post-treatment measures) were examined using a t-test for paired samples (parametric) or a Wilcoxon ranked sum test (non-parametric). Boxplots and scatterplots were applied for the graphical representation of the data. A value of P<0.05 was set as a significant statistical hypothesis.

Results

Effects of the nutraceutical formulation on the TL of the rats

Initially, the present study evaluated TL variations during the 3-month study period in the animals that did not receive treatment. These animals demonstrated a significant reduction in the mean and median TL during this period (Table II). Specifically, the pre-treatment mean TL of the young control group of 19.468 bp (19.146–19.789) was significantly longer compared to the TL mean value [14.535 bp (14.329–14.741)] at the study termination. Similarly, the pre-treatment mean TL values of the old control were decreased during the 3-month period (Table II).

Table II.

Descriptive statistics of TL and comparisons between pre-treatment (baseline).

Table II.

Descriptive statistics of TL and comparisons between pre-treatment (baseline).

Young controlOld controlOld group 1Old group 2




StatisticsPrePostPrePostPrePostPrePost
Min8.0616.5422.8051.9522.6714.0752.5432.905
95% LB19.14614.32910.2948.12710.24010.01010.20312.755
Mean19.46814.53510.5698.42010.47210.36910.46913.600
95% UB19.78914.74110.8438.71310.70310.72910.73414.446
1st16.61312.6137.6975.5938.7377.4078.0887.117
Median19.29714.43910.0687.10310.2219.14210.2929.064
3rd22.45816.36613.22010.68212.00912.63712.50219.385
Max31.37922.43822.17821.61617.83322.79219.30534.881
P-valueat(587)=82.42, P<0.001t(671)=76.35, P<0.001t(449)=1.45, P=0.148t(449)=−10.08, P<0.001
P-valuebz=−21.00, P<0,001z=−22.46, P<0.001z=−3.14, P=0.002z=−1.179, P=0.238
P-valuec Pre: F(2, 1581)=0.19, P=0.825
Post: F(2, 1581)=104.93, P<0.001
P-valued Pre: χ2(2)=0.463, P=0.793
Post: χ2(2)=136.40, P<0.001

a Paired samples t-test for pre-post comparisons for each group;

b Wilcoxon signed rank sum test for pre-post comparisons for each group;

c one way ANOVA comparing TL measures between groups of rats at pre-intervention (pre) and post-intervention (post) (young controls were not included in the analysis);

d non-parametric Kruskal-Wallis test comparing TL measures between groups of rats at pre-intervention (pre) and post-intervention (post) (young controls were not included in the analysis).

Of note, a mean TL reduction was not observed in the rats that received the treatment. Specifically, no significant differences were detected between the mean TL values at pre-treatment compared to post-treatment in the old group 1 [t (449)=1.45, P=0.148] (Table II). In the old group 2, the mean TL values at 13.600 bp (12.755–14.446) were statistically significantly higher at post-treatment compared to the baseline TL values of 10.469 bp (10.203–10.734), while the distribution was not significantly altered (z=−1.179, P=0.238) (Table II). The mean telomere length levels at baseline and at 3 months post-treatment are illustrated in Fig. 1 for the control groups and the groups that received treatments and indicative images from Q-FISH analysis referring to old group 2 pre-treatment and to old group 2 post-treatment are also presented.

One-way ANOVA and the Kruskal-Wallis test were applied to examine the differences between the baseline TL of the old group 1, old group 2 and the old control group. No statistically significant differences were determined for the baseline mean TL among the old control groups. Similarly, no significant differences were detected in TL distributions [median of the overall old control group, 10.068; median old group 1, 10.221; and old group 2, 10.292; χ2(2)=0.463, P=0.793] (Table II).

On the other hand, significant differences in the mean and median TLs among these groups were identified post-treatment (Table II). The old control group displayed lower mean TL values (8.420, 8.127–8.713 bp) compared to the intervention groups [old group 1, 10.369 bp (10.010–10.729 bp); old group 2, 13.600 bp (12.755–14.446 bp); F (2.1581)=104.93, P<0.001]. The median TL values post-treatment exhibited similar differences [χ2(2)=136.40, P<0.001]. The TL distribution per animal at the two set time points (pre-and post-treatment) is presented in Fig. 2. The results of the Kolmogorov-Smirnov test for normality are presented in Table SI.

Independent statistical analysis using the adjusted t-test (LSD, Bonferroni or Dunnett's) likewise did not detect any differences in the TL pre-treatment values between the old controls and the old treatment groups (old group 1 and old group 2). On the other hand, significant differences in TL values were detected between the old controls and the rats in old group 1, and old group 1 vs. the rats in old group 2. A detailed post hoc analysis of pairwise differences is presented in Tables SII and SIII).

Effects of treatment with the nutraceutical formulation on the TL reduction rate

To evaluate the effects of the nutraceutical formulation treatment on the rate of TL change, the mean and median difference in TL per month (TL pre- and TL post-treatment)/month was calculated and represented for each group (Table SIV). A decrease in the median difference in TL per month was detected in all the study groups. In younger rats (untreated young control), the magnitude of the TL reduction rate of TL values was shown to be relatively high, whereas the reduction rate of TL values for the old untreated control group was decreased to ~50% as compared to the reduction rate of the young controls. Treated with supplements, rats showed a complex pattern in TL changes. The median TL reduction rate of the old group 1 and old group 2 was <50% compared to the reduction rate of the old control group (Fig. 3). Nevertheless, the mean TL reduction rate was low for old group 1, whereas the TL of old group 2 exhibited a considerable increase with a mean of −1044 bases/month (Fig. 3).

Effects of aging on rat TL

Furthermore, the effects of aging on TL were evaluated in the old and young rat groups. The median TL and quartile range values at 3 months (young control pre-), 6 months (young control post), 18 months (old control pre) and 21 months (old control post) are presented (Fig. 4). A TL decrement is apparent, and TL vs. time dependence can be characterized by a linear regression line TL=−1743 * time + 19.700 bp with an R2=0.932.

Telomerase activity

The baseline telomerase activity of the PBMCs collected from all rat groups measured as fluorescence at 450 nm OD is presented in Fig. 5. The mean telomerase activity of the PBMCs was 0.355±0.033 for the young control group, while the old control group exhibited a mean activity of 0.163±0.03. Old group 1 and old group 2 exhibited similar baseline telomerase activities with a mean of 0.168±0.03 and 0.163±0.03, respectively. One-way ANOVA revealed a statistically significant difference between the young group and all old groups [(F(3, 12)=37,94, P<0.001)]. Similar results were obtained with the post-hoc Dunnett's test (t=8.784, P<0.001).

The telomerase activity of the PBMCs form the control and treated groups at 3 months post-treatment is presented in Fig. 6. One-way ANOVA revealed a statistically significant difference between the young control group (0.343±0.03), old control group (0.118±0.02), treated old group1 (0.258±0.017) and treated old group2 (0.298±0.033) with an F(3, 12)=52.17 and a P-value of <0.001. In continuation, the analysis of telomerase activity values with the Dunnett's test revealed a significant difference between the old and young controls (t=11.82, P<0,05), and between old group 1 and the young controls (t=4.464, P<0.05). Notably, no marked differences in telomerase activity were detected between old group 2 and the young controls (t=2.364, P>0.05). Statistically significant differences at the P<0.001 level were found for old group 1 and old group 2 vs. the old control CD-SD rats (P>0.001).

A two-way ANOVA was applied to compare the telomerase activity of PBMCs expressed at OD450 nm fluorescence between four groups (two controls and two treated groups) at two time points (baseline and following 3 months of treatment). The analysis revealed that treatment × time interaction presented a significant effect [F(3, 12)=16.72, P=0.007]. The administration of the C. asiatica-containing supplements induced significant effects on PBMC telomerase activity with F(3, 12)=69.95, P<0.001, and F(1, 12)=5.46, P=0.005, respectively (Fig. 7). Furthermore, the Bonferroni post hoc test revealed differences in telomerase activity in old group 1 CD-SD rats (t=3.657, P<0.05) and old group 2 CD-SD rats (t=5.485, P<0.001) when comparing the baseline and the levels at 3 months after treatment.

Discussion

Telomere shortening is strongly associated with DNA damage, oxidative stress and inflammation (1), without omitting various other factors, including genetic and epigenetic mediators, which affect telomere length regulation (33).

Natural molecules with demonstrated antioxidant and anti-inflammatory capacities are being increasingly recognized as potent agents which can be used to restore telomere attrition for the improvement of aging-related diseases (3436).

The effects of natural compounds on TL regulation have been ascribed to the central role of oxidative stress and inflammation to telomere shortening and telomerase, suggesting that nutrients with anti-inflammatory and antioxidant capacity can reduce cell sensitivity to telomere loss (37). Th authors have previously demonstrated that the nutraceutical formulation containing C. asiatica extract, vitamin C (as magnesium ascorbate), zinc (as zinc citrate) and vitamin D3 (as cholecalciferol) has a beneficial effect on aging-associated telomerase decline, behavior and brain morphology change in vivo (17). The present study examined the potency of the formulation in middle-aged rats and revealed that it results in telomere shortening attenuation and the activation of telomerase of PBMCs in a dose-dependent manner. Specifically, the treatment maintained the rat PBMC TL at the lower dose, whereas the higher dose increased the median TLs, compared to the pre-treatment levels. In terms of translating these findings in humans, it is supported that two capsules of the nutraceutical formulation per day were more potent in activating telomerase and increasing the telomere length of PBMCs than 1 capsule. Clinical trials on the efficacy of the nutraceutical formulation to telomere shortening and other aging biomarkers are required to further evaluate these effects.

Data from epidemiological studies and clinical trials have demonstrated that the increased intake of micronutrients, such as vitamins D, C, E and A, and dietary fiber in the context of a balanced diet, e.g., the Mediterranean diet, is associated with longer telomeres, as reviewed by Galiè et al (38). In addition, the consumption of nuts and seeds, a rich source of anti-inflammatory polyunsaturated fatty acids, has been showon to be associated with longer telomeres, and improved metabolic and blood lipid profiles, possibly mediated by an antioxidant effect (39).

The formulation tested herein consisted of nutrients that have been previously reported to improve aging-related and/or oxidative stress markers. C. asiatica is a herb utilized in traditional Chinese medicine for the treatment of various pathologies (40). The extract of this herb contains some pentacyclic triterpenoids, mainly asiaticoside, madecasosside, asiatic acid, madecassic acid and other components, including centellose and centelloside (41). Notably, the extracts of C. asiatica have been shown to exert potent antioxidant and anti-inflammatory effects. Specifically, the oral administration of C. asiatica has been shown to attenuate inflammatory markers and enhance the antioxidative function of diabetic rats (42). Moreover, carbon tetrachloride (CCl4)-induced liver fibrosis has been shown to be ameliorated by oral treatment with C. asiatica. Specifically, C. asiatica was shown to decrease CCl4-induced inflammation, oxidative stress and hepatocyte apoptosis, and to modify Bcl-2/Bax signaling in the livers of rats (43).

Furthermore, vitamin D is a fat-soluble micronutrient that participates in critical cellular functions beyond the absorption of calcium and its deficiency has been related to numerous inflammatory and aging-associated pathologies, including hypertension, cognitive decline and cardiometabolic complications (44). One of its extraskeletal effects is the regulation of the pace of aging as a regulator of mitochondrial function, oxidative stress and senescence (45). The results of an epidemiological study suggest that vitamin D can reduce the production of inflammatory mediators, such as C-reactive protein and interleukin-6, known to mitigate telomere shortening (46). Moreover, it was previously demonstrated that supplementation with vitamin D for 12 months improved cognitive function by reducing oxidative stress in older adults with mild cognitive impairment. This was associated with an increased TL (47). A separate, recent epidemiological study suggested that genetic variations in the gene encoding vitamin D binding protein (GC-rs2282679) were associated with a low TL, suggesting that the vitamin D status may affect TL early in life (48).

A cross-sectional study performed on a cohort of 2,483 males demonstrated that 25-hydroxyvitamin D (25(OH)D) and 1,25-dihydroxy vitamin D plasma levels were not associated with a low TL (49). On the other hand, higher plasma 25(OH)D levels appear to be associated with longer telomeres in women, and this association appears to be modulated by calcium intake (50). There is additional epidemiological evidence linking high serum levels of vitamin D with a longer TL. As previously demonstrated, the treatment of patients undergoing hemodialysis with vitamin D resulted in a longer peripheral PBMC TL compared to that of untreated patients (51). Additionally, epidemiological studies have positively associated vitamin C intake with a longer TL, and the vitamin's antioxidative properties have further been associated with abated telomere shortening of peripheral blood cells (5254). There is also in vitro evidence to support the potency of zinc to decrease the aging of mesenchymal stem cells via an increase in TL, telomerase expression and activity, with an additional decrease in the percentage of senescent cells and the epigenetic modification of the hTERT gene promoter (55).

The present study further demonstrated that the administration of the nutraceutical formulation containing C. asiatica extract, vitamin C, zinc and vitamin D3 increased the telomerase activity of PBMCs in a dose-dependent manner in a rat model. Specifically, the telomerase activity of medium-aged rats was reversed to the levels of the young controls at the higher dose. These findings are in line with those of previous studies demonstrating the potency of formulation and its naturally occurring constituents on telomerase activation (17). Moreover, the authors also recently demonstrated that C. asiatica extract increased telomerase activity in human PBMCs (16). The potency of the C. asiatica-containing formulation to activate telomerase was significantly more pronounced compared to other multi-nutrient formulations (16). Furthermore, vitamin C treatment has been found to be capable of mesenchymal stem cell sheet formation and tissue regeneration by inducing telomerase activity in vitro (56). In addition, vitamin C has been shown to activate telomerase in periodontal ligament stem cells, resulting in their enhanced expression of extracellular matrix components and stem cell markers (57). Moreover, the treatment of human pluripotent stem cell-derived cardiomyocytes with vitamin C has been found to result in the upregulation of human telomerase RNA (57). Likewise, supplementation with vitamin D has been demonstrated to significantly enhance the PBMC telomerase activity in a cohort of overweight African Americans (58). In a separate cohort of obese African Americans, vitamin D supplementation was associated with reduced epigenetic aging (59). Notwithstanding, it has been highlighted that the combination of nutrients and natural compounds exerts more significant effects than single compounds (13,16,60). Thus, it can be hypothesized that the beneficial effects of treatment on TL and telomerase may be due to the synergy of the constituents of the supplement. However, further studies are required to validate this.

The present study has several limitations which should be mentioned. First, the relatively low number of animals in the control and treatment groups can be mitigated by increasing animal numbers in future studies. In addition, the present study did not assess the putative mechanisms of action. In future studies, this point can be addressed by examining antioxidant, anti-inflammatory and various epigenetic markers. Finally, there is a general limitation in extrapolating results obtained in animal models to clinical efficacy that can be attributed to internal and external validity (61). In the present study, strict measures were taken to ensure internal validity and control bias. However, the issue of external validity, including the incapacity of animal models to emulate the complexity of human conditions and species-related differences, remain. One approach would be to perform the study with a larger number of animals exposed to some level of stress, e.g., sleep interruption, to better mimic the everyday pressures facing the majority of the human population. Furthermore, there are several differences between rats, as well as mice and humans regarding telomeres, telomerase and lifespan. Small animals such as mice and rats are characterized by longer telomeres and a higher telomerase expression in various tissues compared to humans, albeit with a lower lifespan (62). Still, they are the gold standard in aging research, with numerous reports on the impact of telomerase activation and underlying anti-aging mechanisms (63). Collectively, it was observed that the administration of the nutraceutical supplement resulted in telomerase activation in both PBMCs and the brains of rats in accordance with the amelioration of brain structure and function and the increase in TL. However, additional studies are required to define the association of these effects, given that rats have long telomeres, and other non-canonical pathways of TERT may be involved (64). Finally, epidemiological studies would also provide a better overview of the putative role of multi-nutrient supplements on TL and telomerase activity in humans.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

The present study was supported by the European Union's ‘Horizon Europe’ framework programme project name: ‘MAgnetically steerable wireless Nanodevices for the tarGeted delivery of therapeutIc agents in any vascular rEgion of the body’ (ANGIE H2020-EIC-FETPROACT-2019). The study was also partially funded by the Spin-Off Company of the University of Crete, Toxplus S.A., by the start-up company LifePlus and by the Special Research Account of University of Crete (ELKE nos. 3464).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author upon reasonable request.

Authors' contributions

AT, DT, DAS, DN and AOD conceptualized the study. AT, DT, AOD, DN and DC were involved in the supervision of the study. AMB, AOD, DC, ER, ES, EV and PF performed the experiments, sample treatment and analyses. ER, DN, ES, DT, AMB, EV, PF, AA, AOD, AT and DAS were involved in the writing and editing of the manuscript. AA performed the statistical analyses. AT and AA confirm the authenticity of the raw data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

All animals were obtained from the University of Medicine and Pharmacy of Craiova Animal House, Craiova Romania, authorization no. 76/20.04.2016. The Ethical Committee of the University of Medicine and Pharmacy of Craiova, Craiova, Romania, approved the animal study (protocol no. 102/23.09.2019). All the procedures were according to the European directives for animal experiments (E.U. Directive 2010/63/E.U.as amended by Regulation E.U.2019/1010).

Patient consent for publication

Not applicable.

Competing interests

DAS is the Editor-in-Chief for the journal, but had no personal involvement in the reviewing process, or any influence in terms of adjudicating on the final decision, for this article. DT is a scientific advisor for Natural Doctor S.A. The other authors declare that they have no competing interests.

References

1 

Chakravarti D, LaBella KA and De Pinho RA: Telomeres: History, health, and hallmarks of aging. Cell. 184:306–322. 2021. View Article : Google Scholar : PubMed/NCBI

2 

Poole JC, Andrews LG and Tollefsbol TO: Activity, function, and gene regulation of the catalytic subunit of telomerase (hTERT). Gene. 269:1–12. 2001. View Article : Google Scholar : PubMed/NCBI

3 

Razgonova MP, Zakharenko AM, Golokhvast KS, Thanasoula M, Sarandi E, Nikolouzakis K, Fragkiadaki P, Tsoukalas D, Spandidos DA and Tsatsakis A: Telomerase and telomeres in aging theory and chronographic aging theory (Review). Mol Med Rep. 22:1679–1694. 2020. View Article : Google Scholar : PubMed/NCBI

4 

Maciejowski J and de Lange T: Telomeres in cancer: Tumour suppression and genome instability. Nat Rev Mol Cell Biol. 18:175–186. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Hao LY, Armanios M, Strong MA, Karim B, Feldser DM, Huso D and Greider CW: Short telomeres, even in the presence of telomerase, limit tissue renewal capacity. Cell. 123:1121–1131. 2005. View Article : Google Scholar : PubMed/NCBI

6 

López-Otín C, Blasco MA, Partridge L, Serrano M and Kroemer G: The hallmarks of aging. Cell. 153:1194–1217. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Fernandes SG, Dsouza R and Khattar E: External environmental agents influence telomere length and telomerase activity by modulating internal cellular processes: Implications in human aging. Environ Toxicol Pharmacol. 85:1036332021. View Article : Google Scholar : PubMed/NCBI

8 

Li H, Lin L, Ye S, Li H and Fan J: Baseline Assessment of nutrient and heavy metal contamination in the seawater and sediment of Yalujiang Estuary. Mar Pollut Bull. 117:499–506. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Derevyanko A, Whittemore K, Schneider RP, Jiménez V, Bosch F and Blasco MA: Gene therapy with the TRF1 telomere gene rescues decreased TRF1 levels with aging and prolongs mouse health span. Aging Cell. 16:1353–1368. 2017. View Article : Google Scholar : PubMed/NCBI

10 

Rojas DM, Nilsson A, Ponsot E, Brummer RJ, Fairweather-Tait S, Jennings A, de Groot LCPGM, Berendsen A, Pietruszka B, Madej D, et al: Short telomere length is related to limitations in physical function in elderly European adults. Front Physiol. 9:11102018. View Article : Google Scholar : PubMed/NCBI

11 

Steenstrup T, Kark JD, Verhulst S, Thinggaard M, Hjelmborg JVB, Dalgård C, Kyvik KO, Christiansen L, Mangino M, Spector TD, et al: Telomeres and the natural lifespan limit in humans. Aging (Albany NY). 9:1130–1142. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Tsoukalas D, Zlatian O, Mitroi M, Renieri E, Tsatsakis A, Izotov BN, Burada F, Sosoi S, Burada E, Buga AM, et al: A novel nutraceutical formulation can improve motor activity and decrease the stress level in a murine model of middle-age animals. J Clin Med. 10:6242021. View Article : Google Scholar : PubMed/NCBI

13 

Selim AM, Nooh MM, El-Sawalhi MM and Ismail NA: Amelioration of age-related alterations in rat liver: Effects of curcumin C3 complex, Astragalus membranaceus and blueberry. Exp Gerontol. 137:1109822020. View Article : Google Scholar : PubMed/NCBI

14 

Tsoukalas D, Fragkiadaki P, Docea AO, Alegakis AK, Sarandi E, Vakonaki E, Salataj E, Kouvidi E, Nikitovic D, Kovatsi L, et al: Association of nutraceutical supplements with longer telomere length. Int J Mol Med. 44:218–226. 2019.PubMed/NCBI

15 

Renieri E, Vakonaki E, Karzi V, Fragkiadaki P and Tsatsakis A: Telomere length: Associations with nutrients and xenobiotics. Toxicological Risk Assessment and Multi-System Health Impacts from Exposure. Tsatsakis A: 1st edition. Academic Press; 2021, View Article : Google Scholar

16 

Tsoukalas D, Fragkiadaki P, Docea AO, Alegakis AK, Sarandi E, Thanasoula M, Spandidos DA, Tsatsakis A, Razgonova MP and Calina D: Discovery of potent telomerase activators: Unfolding new therapeutic and anti-aging perspectives. Mol Med Rep. 20:3701–3708. 2019.PubMed/NCBI

17 

Tsoukalas D, Buga AM, Docea AO, Sarandi E, Mitrut R, Renieri E, Spandidos DA, Rogoveanu I, Cercelaru L, Niculescu M, et al: Reversal of brain aging by targeting telomerase: A nutraceutical approach. Int J Mol Med. 48:1992021. View Article : Google Scholar : PubMed/NCBI

18 

Zucker I and Beery AK: Males still dominate animal studies. Nature. 465:6902010. View Article : Google Scholar : PubMed/NCBI

19 

Environmental Protection Agency (EPA), . Health Effects Test Guidelines OPPTS 870.3700 Prenatal Developmental Toxicity Study. EPA; Washington, DC: 1998

20 

Organization for Economic Co-operation and Development (OECD), . Guidance Document on the Recognition, Assessment, and Use of Clinical Signs as Humane Endpoints for Experimental Animals Used in Safety Evaluation. OECD Series on Testing and Assessment. No. 19. OECD Publishing; Paris, France: 2002

21 

Gad SC, Cassidy CD, Aubert N, Spainhour B and Robbe H: Nonclinical vehicle use in studies by multiple routes in multiple species. Int J Toxicol. 25:499–521. 2006. View Article : Google Scholar : PubMed/NCBI

22 

Nair A and Jacob S: A simple practice guide for dose conversion between animals and human. J Basic Clin Pharm. 7:27–31. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Vieth R: Vitamin D supplementation: Cholecalciferol, calcifediol, and calcitriol. Eur J Clin Nutr. 74:1493–1497. 2020. View Article : Google Scholar : PubMed/NCBI

24 

Dietary Reference Intakes for Vitamin A, Vitamin K, Arsenic, Boron, Chromium, Copper, Iodine, IronManganese, Molybdenum, Nickel, Silicon, Vanadium, Zinc: Dietary Reference Intakes for Vitamin A, Vitamin K, Arsenic, Boron, Chromium, Copper, Iodine, Iron, Manganese, Molybdenum, Nickel, Silicon, Vanadium, and Zinc. National Academies Press (US); 2000

25 

Sil S, Ghosh T, Gupta P, Ghosh R, Kabir SN and Roy A: Dual role of vitamin C on the neuroinflammation mediated neurodegeneration and memory impairments in colchicine induced rat model of Alzheimer disease. J Mol Neurosci. 60:421–435. 2016. View Article : Google Scholar : PubMed/NCBI

26 

Holick MF, Binkley NC, Bischoff-Ferrari HA, Gordon CM, Hanley DA, Heaney RP, Murad MH and Weaver CM: Guidelines for preventing and treating vitamin D deficiency and insufficiency revisited. J Clin Endocrinol Metabol. 97:1153–1158. 2012. View Article : Google Scholar

27 

Williamson L, Hayes A, Hanson ED, Pivonka P, Sims NA and Gooi JH: High dose dietary vitamin D3 increases bone mass and strength in mice. Bone Rep. 6:44–50. 2017. View Article : Google Scholar : PubMed/NCBI

28 

Dietary Reference Intakes for Vitamin C, Vitamin E, Selenium, Carotenoids, Dietary Reference Intakes for Vitamin C, Vitamin E, Selenium, and Carotenoids. National Academies Press; 2000

29 

Hathcock JN, Shao A, Vieth R and Heaney R: Risk assessment for vitamin D. Am J Clin Nut. 85:6–18. 2007. View Article : Google Scholar

30 

Rao SB, Chetana M and Devi PU: Centella asiatica treatment during postnatal period enhances learning and memory in mice. Physiol Behav. 86:449–457. 2005. View Article : Google Scholar : PubMed/NCBI

31 

McIlrath J, Bouffler SD, Samper E, Cuthbert A, Wojcik A, Szumiel I, Bryant PE, Riches AC, Thompson A, Blasco MA, et al: Telomere length abnormalities in mammalian radiosensitive cells. Cancer Res. 61:912–915. 2001.PubMed/NCBI

32 

Tsatsakis A, Tsoukalas D, Fragkiadaki P, Vakonaki E, Tzatzarakis M, Sarandi E, Nikitovic D, Tsilimidos G and Alegakis AK: Developing BioTel: A semi-automated spreadsheet for estimating telomere length and biological age. Front Genet. 10:842019. View Article : Google Scholar : PubMed/NCBI

33 

Kansara K and Gupta SS: Chapter 14. DNA damage, repair, and maintenance of telomere length: Role of nutritional supplements. Mutagenicity: Assays and Applications. Academic Press; pp. 287–307. 2017

34 

Guo J, Huang X, Dou L, Yan M, Shen T, Tang W and Li J: Aging and aging-related diseases: from molecular mechanisms to interventions and treatments. Signal Transduct Target Ther. 7:3912022. View Article : Google Scholar : PubMed/NCBI

35 

Jacczak B, Rubiś B and Totoń E: Potential of naturally derived compounds in telomerase and telomere modulation in skin senescence and aging. Int J Mol Sci. 22:63812021. View Article : Google Scholar : PubMed/NCBI

36 

Grosso G, Godos J, Currenti W, Micek A, Falzone L, Libra M, Giampieri F, Forbes-Hernández TY, Quiles JL, Battino M, et al: The effect of dietary polyphenols on vascular health and hypertension: Current evidence and mechanisms of action. Nutrients. 14:5452022. View Article : Google Scholar : PubMed/NCBI

37 

Davinelli S, Trichopoulou A, Corbi G, De Vivo I and Scapagnini G: The potential nutrigeroprotective role of Mediterranean diet and its functional components on telomere length dynamics. Ageing Res Rev. 49:1–10. 2019. View Article : Google Scholar : PubMed/NCBI

38 

Galiè S, Canudas S, Muralidharan J, García-Gavilán J, Bulló M and Salas-Salvadó J: Impact of nutrition on telomere health: Systematic review of observational cohort studies and randomized clinical trials. Adv Nutr. 11:576–601. 2020. View Article : Google Scholar : PubMed/NCBI

39 

Godos J, Giampieri F, Micek A, Battino M, Forbes-Hernández TY, Quiles JL, Paladino N, Falzone L and Grosso G: Effect of Brazil nuts on selenium status, blood lipids, and biomarkers of oxidative stress and inflammation: A systematic review and meta-analysis of randomized clinical trials. Antioxidants (Basel). 11:4032022. View Article : Google Scholar : PubMed/NCBI

40 

Sun B, Wu L, Wu Y, Zhang C, Qin L, Hayashi M, Kudo M, Gao M and Liu T: Therapeutic potential of centella asiatica and its triterpenes: A review. Front Pharmacol. 11:5680322020. View Article : Google Scholar : PubMed/NCBI

41 

Pizzorno M and Murray J: Textbook of natural medicine. 4th edition. Elsevier Health Sciences; 2012

42 

Giribabu N, Karim K, Kilari EK, Nelli SR and Salleh N: Oral administration of Centella asiatica (L.) Urb leave aqueous extract ameliorates cerebral oxidative stress, inflammation, and apoptosis in male rats with type-2 diabetes. Inflammopharmacology. 28:1599–1622. 2020. View Article : Google Scholar : PubMed/NCBI

43 

Wei L, Chen Q, Guo A, Fan J, Wang R and Zhang H: Asiatic acid attenuates CCl4-induced liver fibrosis in rats by regulating the PI3K/AKT/mTOR and Bcl-2/Bax signaling pathways. Int Immunopharmacol. 60:1–8. 2018. View Article : Google Scholar : PubMed/NCBI

44 

Meehan M and Penckofer S: The role of vitamin D in the aging adult. J Aging Gerontol. 2:60–71. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Sosa-Díaz E, Hernández-Cruz EY and Pedraza-Chaverri J: The role of vitamin D on redox regulation and cellular senescence. Free Radic Biol Med. 193:253–273. 2022. View Article : Google Scholar : PubMed/NCBI

46 

Mazidi M, Kengne AP and Banach M: Mineral and vitamin consumption and telomere length among adults in the United States. Pol Arch Intern Med. 127:87–90. 2017.PubMed/NCBI

47 

Yang T, Wang H, Xiong Y, Chen C, Duan K, Jia J and Ma F: Vitamin D supplementation improves cognitive function through reducing oxidative stress regulated by telomere length in older adults with mild cognitive impairment: A 12-month randomized controlled trial. J Alzheimers Dis. 78:1509–1518. 2020. View Article : Google Scholar : PubMed/NCBI

48 

Normando P, Santos-Rebouças C, Leung C, Epel E, da Fonseca AC, Zembrzuski V, Faerstein E and Bezerra FF: Variants in gene encoding for vitamin D binding protein were associated with leukocyte telomere length: The Pró-Saúde study. Nutrition. 71:1106182020. View Article : Google Scholar : PubMed/NCBI

49 

Julin B, Shui IM, Prescott J, Giovannucci EL and De Vivo I: Plasma vitamin D biomarkers and leukocyte telomere length in men. Eur J Nutr. 56:501–508. 2017. View Article : Google Scholar : PubMed/NCBI

50 

Liu JJ, Prescott J, Giovannucci E, Hankinson SE, Rosner B, Han J and De Vivo I: Plasma vitamin D biomarkers and leukocyte telomere length. Am J Epidemiol. 177:1411–1417. 2013. View Article : Google Scholar : PubMed/NCBI

51 

Borras M, Panizo S, Sarró F, Valdivielso JM and Fernandez E: Assessment of the potential role of active vitamin D treatment in telomere length: A case-control study in Hemodialysis patients. Clin Ther. 34:849–856. 2012. View Article : Google Scholar : PubMed/NCBI

52 

Lee JY, Shin C and Baik I: Longitudinal associations between micronutrient consumption and leukocyte telomere length. J Hum Nutr Diet. 30:236–243. 2017. View Article : Google Scholar : PubMed/NCBI

53 

Sen A, Marsche G, Freudenberger P, Schallert M, Toeglhofer AM, Nagl C, Schmidt R, Launer LJ and Schmidt H: Association between higher plasma lutein, zeaxanthin, and vitamin C concentrations and longer telomere length: Results of the Austrian stroke prevention study. J Am Geriatr Soc. 62:222–229. 2014. View Article : Google Scholar : PubMed/NCBI

54 

Marcon F, Siniscalchi E, Crebelli R, Saieva C, Sera F, Fortini P, Simonelli V and Palli D: Diet-related telomere shortening and chromosome stability. Mutagenesis. 27:49–57. 2012. View Article : Google Scholar : PubMed/NCBI

55 

Farahzadi R, Fathi E, Mesbah-Namin SA and Zarghami N: Zinc sulfate contributes to promote telomere length extension via increasing telomerase gene expression, telomerase activity and change in the TERT gene promoter CpG island methylation status of human adipose-derived mesenchymal stem cells. PLoS One. 12:e01880522017. View Article : Google Scholar : PubMed/NCBI

56 

Wei F, Qu C, Song T, Ding G, Fan Z, Liu D, Liu Y, Zhang C, Shi S and Wang S: Vitamin C treatment promotes mesenchymal stem cell sheet formation and tissue regeneration by elevating telomerase activity. J Cell Physiol. 227:3216–3224. 2012. View Article : Google Scholar : PubMed/NCBI

57 

Kim YY, Ku SY, Huh Y, Liu HC, Kim SH, Choi YM and Moon SY: Anti-aging effects of vitamin C on human pluripotent stem cell-derived cardiomyocytes. Age (Dordr). 35:1545–1557. 2013. View Article : Google Scholar : PubMed/NCBI

58 

Zhu H, Guo D, Li K, Pedersen-White J, Stallmann-Jorgensen IS, Huang Y, Parikh S, Liu K and Dong Y: Increased telomerase activity and vitamin D supplementation in overweight African Americans. Int J Obes (Lond). 36:805–809. 2012. View Article : Google Scholar : PubMed/NCBI

59 

Chen L, Dong Y, Bhagatwala J, Raed A, Huang Y and Zhu H: Effects of Vitamin D 3 supplementation on epigenetic aging in overweight and obese African Americans with suboptimal Vitamin D status: A randomized clinical trial. J Gerontol A Biol Sci Med Sci. 74:91–98. 2019. View Article : Google Scholar : PubMed/NCBI

60 

Athanasopoulou S, Kapetanou M, Magouritsas MG, Mougkolia N, Taouxidou P, Papacharalambous M, Sakellaridis F and Gonos E: Anti-oxidant and antiaging properties of a novel synergistic nutraceutical complex: Readouts from an in cellulo study and an in vivo prospective, randomized trial. Antioxidants (Basel). 11:4682022. View Article : Google Scholar : PubMed/NCBI

61 

Ferreira GS, Veening-Griffioen DH, Boon WPC, Moors EHM and van Meer PJK: Levelling the translational gap for animal to human efficacy data. Animals (Basel). 10:11992020. View Article : Google Scholar : PubMed/NCBI

62 

Calado RT and Dumitriu B: Telomere dynamics in mice and humans. Semin Hematol. 50:165–174. 2013. View Article : Google Scholar : PubMed/NCBI

63 

Folgueras AR, Freitas-Rodríguez S, Velasco G and López-Otín C: Mouse models to disentangle the hallmarks of human aging. Circ Res. 123:905–924. 2018. View Article : Google Scholar : PubMed/NCBI

64 

Apostolova N and Victor VM: Molecular strategies for targeting anti-oxidants to mitochondria: Therapeutic implications. Antioxid Redox Signal. 22:686–729. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2023
Volume 28 Issue 6

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Tsatsakis A, Renieri E, Tsoukalas D, Buga AM, Sarandi E, Vakonaki E, Fragkiadaki P, Alegakis A, Nikitovic D, Calina D, Calina D, et al: A novel nutraceutical formulation increases telomere length and activates telomerase activity in middle‑aged rats. Mol Med Rep 28: 232, 2023
APA
Tsatsakis, A., Renieri, E., Tsoukalas, D., Buga, A.M., Sarandi, E., Vakonaki, E. ... Docea, A.O. (2023). A novel nutraceutical formulation increases telomere length and activates telomerase activity in middle‑aged rats. Molecular Medicine Reports, 28, 232. https://doi.org/10.3892/mmr.2023.13119
MLA
Tsatsakis, A., Renieri, E., Tsoukalas, D., Buga, A. M., Sarandi, E., Vakonaki, E., Fragkiadaki, P., Alegakis, A., Nikitovic, D., Calina, D., Spandidos, D. A., Docea, A. O."A novel nutraceutical formulation increases telomere length and activates telomerase activity in middle‑aged rats". Molecular Medicine Reports 28.6 (2023): 232.
Chicago
Tsatsakis, A., Renieri, E., Tsoukalas, D., Buga, A. M., Sarandi, E., Vakonaki, E., Fragkiadaki, P., Alegakis, A., Nikitovic, D., Calina, D., Spandidos, D. A., Docea, A. O."A novel nutraceutical formulation increases telomere length and activates telomerase activity in middle‑aged rats". Molecular Medicine Reports 28, no. 6 (2023): 232. https://doi.org/10.3892/mmr.2023.13119