Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Role of maraviroc and/or rapamycin in the liver of IL10 KO mice with frailty syndrome

  • Laura Pérez-Martínez ,

    Roles Conceptualization, Data curation, Formal analysis, Investigation, Methodology, Resources, Supervision, Validation, Writing – original draft

    lperez@riojasalud.es

    Affiliation Centro de Investigación Biomédica de La Rioja (CIBIR), Logroño, Spain

  • Lourdes Romero,

    Roles Investigation, Methodology

    Affiliation Centro de Investigación Biomédica de La Rioja (CIBIR), Logroño, Spain

  • Eva M. Verdugo-Sivianes,

    Roles Conceptualization, Formal analysis, Investigation, Methodology, Resources

    Affiliations Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocío, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Sevilla, Spain, CIBERONC, Instituto de Salud Carlos III, Madrid, Spain

  • Sandra Muñoz-Galván,

    Roles Conceptualization, Formal analysis, Investigation, Methodology, Resources

    Affiliations Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocío, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Sevilla, Spain, CIBERONC, Instituto de Salud Carlos III, Madrid, Spain

  • Susana Rubio-Mediavilla,

    Roles Formal analysis, Resources

    Affiliation Servicio de Anatomía Patológica, Hospital San Pedro, Logroño, Spain

  • Ana Amiama-Roig,

    Roles Investigation, Methodology, Resources

    Affiliation Centro de Investigación Biomédica de La Rioja (CIBIR), Logroño, Spain

  • Amancio Carnero ,

    Roles Formal analysis, Funding acquisition, Investigation, Methodology, Resources, Supervision, Validation

    ‡ AC and JRB worked as seniors researchers in this work.

    Affiliations Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocío, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Sevilla, Spain, CIBERONC, Instituto de Salud Carlos III, Madrid, Spain

  • José-Ramón Blanco

    Roles Conceptualization, Formal analysis, Funding acquisition, Investigation, Resources, Software, Supervision, Validation

    ‡ AC and JRB worked as seniors researchers in this work.

    Affiliations Centro de Investigación Biomédica de La Rioja (CIBIR), Logroño, Spain, Servicio de Enfermedades Infecciosas, Hospital Universitario San Pedro, Logroño, Spain

Abstract

Cellular senescence and low-grade inflammation favor the acceleration of aging. The liver is an essential metabolic organ because changes related to its function are related to age-related diseases. The objective of this study was to evaluate the effects of maraviroc (MVC) and/or rapamycin (RAPA) on liver tissue in an experimental model of frailty syndrome in mice, since MVC and RAPA are two molecules able to decrease CCR5 expression, which is overexpressed in patients with frailty. Methods: Eighty male homozygous IL10KO mice were randomly assigned to one of 4 groups (n = 20): i) IL10KO group; ii) MVC group, iii) RAPA group, and iv) MVC-RAPA group. Liver samples were analyzed. Gene expression quantification and western blotting were also performed. The proinflammatory cytokines IL-6 and IL-18 were decreased in MVC and MVC/RAPA groups, IL-12 was decreased in RAPA and MVC/RAPA groups and TNF-α was decreased in all therapeutic groups. P21 was decreased in RAPA and MVC/RAPA groups, Galactosidase beta-1, was also significantly reduced in all therapeutic groups, as were NF-kB1, NF-kB2 and STAT3. In all groups, mTOR and CCL5 were significantly reduced. CCR5 expression was decreased in the MVC and MVC/RAPA groups. Conclusion: MVC and RAPA may protect against some factors involved in liver aging. More studies will be necessary to verify their clinical applications.

Introduction

Aging is directly related to the natural imbalance of the body. Therefore, it reduces survival and, at the same time, increases the risk of disease and death [1]. Aging causes not only a gradual loss of function or degeneration of the organism but also gain of function changes that allow cells to proliferate inappropriately [2]. In this context, aging is a major risk factor for the development of chronic diseases such as neurodegenerative and cardiovascular diseases, diabetes mellitus, osteoporosis, cancer and frailty. Indeed, the risk of developing frailty increases with age, suggesting an association between both processes [3]. Besides, the mechanisms of action of these diseases are closely related to aging [1].

The immunological changes associated with aging are characterized by a chronic low-grade inflammatory state (inflammaging) [4]. This inflammatory phenotype is associated with increases in inflammatory biomarkers such as C-reactive protein (CRP), interleukin-6 (IL-6), or tumor necrosis factor-alpha (TNF-α), which are associated with increased morbidity and mortality in older patients [3, 4].

The liver is a key organ that leads the energy metabolism of our body, connecting metabolic pathways between different tissues, including muscle and adipose tissue [5]. On the other hand, the imbalance of hepatic metabolism favors the development of diseases related to aging, such as insulin resistance, diabetes mellitus and non-alcoholic fatty liver [6, 7].

The development of aging in the liver is favored by genomic alterations and mitochondrial disorders that increase cellular senescence and the appearance of low-grade inflammation [8]. This metabolic damage in the aging liver can favor the cellular senescence of hepatocytes since they present altered genes involved in the glucose and protein synthesis pathway [9]. Therefore, senescent cells increase the activity of senescence-associated β-galactosidase (SA-β-gal), a biomarker related to increased lysosome levels [10]. Studying the levels of beta-galactosidase activity associated with cellular senescence can allow us to quantify the presence of senescent cells in the liver of ageing mice [10]. Moreover, cellular senescence is triggered by the activation of p16 ink4a and p21 Cip1 signaling [11]. In this context, senescent hepatocytes release proinflammatory cytokines such as IL-6 and TNF-α that are related to inflammation and age [12].

IL-10 homozygous knockout (IL-10tm/tm [IL10KO]) mice are an excellent tool for the study of frailty [13], a syndrome associated with aging [3], because they develop sarcopenia, muscular weakness and weight loss [14]. In this mouse model, the inflammatory signaling pathway has been altered by eliminating IL-10 (knockout of IL-10) [15]. This cytokine has anti-inflammatory activities because it suppresses the activation of macrophages and inhibits the production of inflammatory cytokines by Th1 cells [16]. Therefore, since these mice develop a chronic inflammation, they give rise to a mouse model with a disordered liver [17].

Increasing scientific evidence suggests that aging is a regulated process, and its course can be modified by modulating signal transduction pathways [18] including mammalian target of the rapamycin (mTOR), AMP-activated protein kinase (AMPK) [17] and a member of the signal transducer and activator of transcription 3 (STAT3) of signaling molecules [19].

Maraviroc (MVC), a specific C-C chemokine receptor type 5 (CCR5) antagonist, has shown some beneficial effects on certain factors involved in the development of frailty in mice [20]. CCR5 is a receptor that regulates the trafficking and effector functions of memory and effector T lymphocytes, macrophages, and immature dendritic cells [21]. One of CCR5 ligands is C-C motif chemokine ligand 5 (CCL5), which belongs to the CC family of inflammatory chemokine and plays an important role in the frailty syndrome [22] and inflammation [23]. In addition, rapamycin (RAPA), a specific inhibitor of mTOR pathway [23], not only extends the lifespan of aged mice [24], but can also decrease CCR5 mRNA expression [25], which is overexpressed in patients with frailty [26].

Studies by our group examined murine models of liver damage and observed that animals treated with MVC exhibited a better anti-inflammatory profile than control animals [5, 27]. Therefore, in the present study, our objective was to evaluate the effects of MVC and/or RAPA on a disordered liver by using an experimental mouse model of chronic inflammation and frailty, a pathology related to aging.

Results

In this study, IL-10 deficient mice were randomly assigned to one of the four groups (control, MVC, RAPA or MVC/RAPA) and received the corresponding treatment previously described [28]. All groups had similar survival rates [28]. The four groups had a similar baseline weight and none of the therapeutic interventions reduced body weight. In fact, there were no body weight changes both during and at the end of the experiment (week 24) [28]. We analyzed different parameters in the liver tissue.

The expression of proinflammatory cytokines is decreased after MVC and RAPA treatments

First, to evaluate the effect of the different treatments in the immune system, we measured the levels of different cytokines. At the mRNA level, compared to that in the control group, the IL-6 level was significantly lower in the MVC and MVC/RAPA groups (p<0.05 in both). The RAPA group also showed a clear tendency toward lower levels (p = 0.07) (Fig 1A). The expression of TNF-α was significantly lower in the MVC (p<0.0001), RAPA (p<0.05) and MVC/RAPA groups (p<0.01) (Fig 1B). In addition, the levels of IL-18, another proinflammatory cytokine, were also significantly lower in the MVC (p<0.01) and MVC/RAPA groups (p<0.05) (Fig 1C) and the IL-12 level was significantly lower in the MVC (p<0.001) and MVC/RAPA (p<0.0001) groups (Fig 1D). IL-1β, another proinflammatory cytokine was examined, and the MVC group showed a tendency toward lower levels (p = 0.07) (Fig 1E).

thumbnail
Fig 1. Liver expression of IL-6, TNF-α, IL-18, IL-12, IL-1β, CCL5 and CCR5 at the RNA level and STRING analysis separated by treatment groups.

https://doi.org/10.1371/journal.pone.0286201.g001

Therefore, the MVC and RAPA treatments and the combination of both (MVC/RAPA) induced a decrease in the mRNA expression of different cytokines. Direct interactions between cytokines IL-6, TNF-α, IL-18 and IL-12 were predicted by STRING analysis (Fig 1H–1J), separated by treatment groups, suggesting an extended phenotype beyond IL10, comprising all immune or inflammatory system.

The expression of CCR5 and CCL5 biomarkers is decreased after MVC and RAPA treatments

Then, we evaluate the expression of CCR5, target of both MVC and RAPA treatments, and its ligand CCL5. Compared to that in the control group, CCL5 mRNA expression was lower in the MVC (p<0.0001), RAPA (p<0.02) and MVC/RAPA groups (p<0.02) (Fig 1F). On the other hand, liver CCR5 expression was significantly reduced in the MVC (<0.0005) and MVC/RAPA groups (p<0.0001) (Fig 1G). Besides, direct interactions between CCL5 and CCR5 were predicted by STRING analysis (Fig 1H–1J).

Therefore, we corroborate that CCR5 and CCL5 mRNA expressions were reduced with MVC and RAPA treatments, as expected, since CCR5 is a target of MVC and it is also reduced after RAPA treatment.

MVC and RAPA treatments also reduced other biomarkers of the inflammatory response

Since RAPA is a specific inhibitor of mTOR pathway [23], we also measured mTOR expression levels in the different groups of treatment. mRNA levels of mTOR were notably reduced in the RAPA and MVC/RAPA groups (p<0.01 and p<0.05, respectively), and there was a clear trend in the MVC group (p = 0.07) (Fig 2).

thumbnail
Fig 2. Liver expression of mTOR, GLB-1, P21, P16, STAT3 and nuclear factor KB 1 and 2 at the mRNA level.

https://doi.org/10.1371/journal.pone.0286201.g002

To evaluate the presence of senescent cells in the liver tissue, we measured the levels of Galactosidase beta-1 (GLB-1), p21 Cip1 (P21) and p16 ink4a (P16). GLB-1 mRNA levels were also significantly reduced in the MVC (p<0.01), RAPA (p<0.001) and MVC/RAPA groups (p<0.001). All of the treatments seem to reduce the presence of this marker of senescent cells (Fig 2B). In addition, P21 levels was significantly reduced in the RAPA (p<0.01) and MVC/RAPA groups (p<0.0001) (Fig 2C) and P16 levels did not showed any significant difference (Fig 2D). In this context, this may indicate that cellular senescence is not promoted after MVC, RAPA or MVC/RAPA treatments.

Since previous reports demonstrated that IL10 repressed inflammation response via activation of STAT3 [19], we also measured STAT3 expression levels in the different groups of treatment. The expression of STAT3 was significantly lower in the MVC (p<0.05), RAPA (p<0.01) and MVC/RAPA groups (p<0.0001) (Fig 2E).

Furthermore, we measured the levels of NF-kB, a key transcription factor that regulates the inflammatory response [29]. NF-kB1 was examined, and the mRNA levels were significantly reduced in the MVC (p<0.05), RAPA (p<0.01) and MVC/RAPA groups (p<0.05). A similar outcome was observed regarding liver expression of NF-kB2 mRNA in the MVC (p<0.01), RAPA (p<0.05) and MVC/RAPA groups (p<0.05) (Fig 2F and 2G). Therefore, the expression of both NF-kB1 and NF-kB2 were reduced after all of the treatments. Direct interactions between others biomarkers: mTOR, GLB-1, P21, P16, STAT3, NF-kB1 and NF-kB2 were predicted by STRING analysis (Fig 1H–1J).

AKT/mTOR pathway is activated after MVC and RAPA treatments

Next, we measured the expression of some of these markers at the protein level, including their phosphorylated and activates forms.

Mice treated with MVC showed a significant increase in total NF-kB (p <0.05) and the RAPA group showed an increase in p-NF-kB (p<0.01). When NF-kB is activated, it is phosphorylated and translocate to the nucleus to induce the expression of some proinflammatory genes such as IL-1β and TNF-α. Therefore, it seems that NF-kB is activated after MVC and RAPA treatments.

Mice treated with MVC and MVC-RAPA showed significant increase in p-Akt (p <0.05). In the case of mTOR, we observed a decrease when mice were treated with the combination MVC-RAPA, but mice treated with MVC showed an increase in p-mTOR (p<0.01) and the MVC-RAPA group showed a significant increase in p-mTOR (p <0.001). Finally, we observed that mice treated with MVC, RAPA and MVC-RAPA reduced the levels of AMPK, which also inhibits mTOR. On the other hand, STAT3 (total and phosphorylated) was examined but didn´t show significant differences (Fig 3A and 3B and S1 Fig).

thumbnail
Fig 3. Analyses of the molecular pathways involved in the mechanism of action of MVC and RAPA.

https://doi.org/10.1371/journal.pone.0286201.g003

Therefore, it seems that AKT/mTOR pathway is activated after MVC and RAPA treatments. Given the specificity of RAPA for mTORC1 [37], and the differential activity to mTORC2, we can suggest that we predominantly detect mTORC2 phosphorylation and activity or some feed-back loop maintaining mTOR phosphorylated even on mTORC1 complex.

Discussion

Most chronic human diseases increase with age [28]. This makes it important to look for different strategies to slow down aging. It is known that the lifespan of mice can be modified by dietary, genetic, and pharmacological interventions [30]. Exciting findings have shown that rodent aging can be accelerated, stopped, or reversed simply by altering the systemic environment [31]. This growing line of research may offer strategies for treating aging. In this context, RAPA was the first drug to dramatically slow aging in mice [32]. RAPA also increases life expectancy in most studies and protects against many age-related diseases [32]. Other drugs, such as metformin and acarbose, also prolong the lifespan of mice [33, 34]. MVC also has some potential benefits on aging because it modifies certain factors involved in the development of frailty in mice, such as myostatin and certain inflammatory cytokines [28]. In relation to the liver safety of these therapeutic interventions, in a previous study [28] we observed that all of them reduced the transaminases levels.

mTOR has been implicated in many of the processes associated with aging, including cellular senescence, immune responses, stem cell regulation and mitochondrial function [35]. mTOR kinase functions within two multiprotein complexes called mTORC1 and mTORC2 with different subunits and specificities [35]. Inhibition of mTORC1 has prolonged lifespan in all species studied to date and has ameliorated multiple age-related pathologies, including decreased immune function [36].

In addition, some other reports also suggested that the coordination between mTOR and a STAT3 pathway in the modulation of innate immune response [19]. STAT3 was originally identified as a transcription factor that regulates gene expression in response to inflammatory stimulation evoked by ligands whose receptor complexes contain gp130, such as IL-6 [19]. IL-6, as a pro-inflammatory cytokine, it preferentially activates signal transducer and activator of transcription protein 3 (STAT3)-dependent gene expression [37]. Activation of STAT3 by IL-6 plays a crucial role in inflammation-induced disease pathogenesis [38]. On the other hand, it has been demonstrated that, important CCR5-activated signaling proteins such as janus kinase 2 (JAK2) and STAT3 were inhibited by maraviroc [39].

Inhibiting mTOR with RAPA delays aging and increases lifespan in mice [23]. In our study, MVC, RAPA or MVC-RAPA reduced the mRNA levels of mTOR. However, there is evidence of increased mTOR activity, at least in the liver, in mice in which the aging process has been delayed, such as due to gene mutations [24] (Fig 4). Given the specificity of RAPA for mTORC1 [40], and the differential activity to mTORC2, we can suggest that we predominantly detect mTORC2 phosphorylation and activity, or some feed-back loop maintaining mTOR phosphorylated even on mTORC1 complex. mTORC2 might maintain the phosphorylation of AKT by direct phosphorylation maintaining some feedback loop on mTORC1 phosphorylation, even in the presence of RAPA, which will inhibit further mTORC1, but not mTORC2, activity [40].

thumbnail
Fig 4. Schematic graph of the postulated mechanism by which MVC and RAPA interfere in an aged mouse liver model.

https://doi.org/10.1371/journal.pone.0286201.g004

In our study, all mice that were treated with MVC also showed higher protein levels of p-mTOR. The phosphorylation of mTOR in mTORC1 by Akt occurs directly via Rheb (Ras family of GTPase) [41]. However, since the mechanism seems the same in both cases, it might be more related to AKT activity than mTOR. There are proteins and transcription factors that can be activated or inhibited after the phosphorylation of the Akt pathway [4145] without involving mTORC1. The frailty in this model might partly relay in these pathways, like oxidative stress, which has also been related to aging [46, 47]. Nrf2 is a key factor in downstream of PI3K/Akt and is involved in the regulation of oxidative stress and inflammatory response [48]. Therefore, PI3K/Akt pathway may play an important role in the activation of Nrf2. In our study, both MVC and MVC/RAPA activated AKT, and further research with AKT inhibitors [48] or antioxidants, could help to discriminate the effects on the relation between inflammation and frailty syndrome.

In addition to activated Akt, inflammatory and oxidative stress stimulate the NF-kB family of transcription factors [40] (Fig 4). In mouse models, the inhibition of NF-kB has been shown to lead to late onset of age-related symptoms because most of the genes under the transcriptional control of NF-kB are involved in biologic pathways associated with aging, including immune responses, cell senescence, apoptosis and metabolism [40]. All of these factors contribute to age-related tissue degeneration [6]. These changes have been described in liver tissue from mice and rats [8]. Our results showed that at the mRNA level, there was a decrease in NF-kB expression in the liver (1&2) in all therapeutic groups, while at the protein level; there was an increase in total NF-kB expression in the MVC group. This increase was the same as that observed in the muscles of aged IL-10 mice [28]. NF-kB activation is related to many of the known lifespan regulators, including mTOR [49]. Therefore, NF-kB represents a potential antiaging therapeutic target. Consistent with our results, mTOR and NF-kB signaling are coregulated [50] (Fig 1). Another component that contributes to the increase in NF-kB activity that is associated with aging is control of the expression of inflammatory cytokines [51]. Increasingly, inflammation is being linked to aging and chronic diseases [52]. The basal inflammatory response increases with age, leading to low-level chronic inflammation that is likely maladaptive and promotes aging. In this animal model of frailty, we observed a decrease in the mRNA levels of proinflammatory cytokines (IL-6, TNF-α, IL-18 and IL-12) (Fig 1). Therefore, the main protective effect of MVC and RAPA depends on a proinflammatory pathway. This inflammatory phenotype accelerates aging (Fig 4).

Since rapamycin was reported in 2009 to increase the lifespan of mice, there are more than twenty studies published showing that cellular senescence is attenuated by rapamycin [53]. Selvarani R et al. shows a list of different studies in which rapamycin reduces or blocks senescence in a variety of cells from humans, mice, and rats [53]. In addition to suppressing markers of senescence, such as P21 or SA-β-gal-positive cells, rapamycin reduced/prevented the senescent associated secretory phenotype (SASP) phenotype (e.g., the expression and secretion of proinflammatory cytokines by senescent cells such as Il-6 and TNF-α) [51]. This SASP causes DNA damage of sufficient magnitude to induce senescence. Furthermore, SASPs produced by senescent cells could play a role in the age-related increase in chronic inflammation [54, 55]. Therefore, cellular senescence could be an important mechanism underlying aging, because it affects tissue regeneration and drives chronic low-grade inflammation, which exacerbates the aging process [54, 55]. In the same way, in our previous study, we detected at the mRNA muscle level, the expression of IL-6 were significantly lower in the MVC-RAPA group and showed a tendency in the MVC group [28]. Likewise, in muscle samples, IL-18 mRNA levels, another proinflammatory cytokine, were significantly lower in the MVC group [28].

To better understand the mechanism underlying these observations, in this animal model, we did not observe a synergistic, additive or antagonistic effect on the levels of CCR5 or CCL5 mRNA in the liver in the MVC-RAPA group. CCL5 plays an active role in the recruitment of a variety of leukocytes to inflammatory sites [25] (Fig 1). In collaboration with certain cytokines that are released by T cells (e.g., IL-2), CCL5 induces the activation and proliferation of certain natural killer cells to generate chemokine-activated C-C killer cells [56]. CCL5/CCR5 interactions can act as growth factors, inducing the recruitment of additional inflammatory cells and participating in immune evasion [26].

Based on our study, mTOR and NF-kB signaling are coregulated in the liver of IL-10tm/tm [IL10KO] mice, demonstrating that the main protective effect in this animal model of MVC and RAPA could depends on a proinflammatory pathway (Fig 4). This inflammatory phenotype accelerates aging. In summary, our data suggest that the use of MVC and/or RAPA could have protective effects on some factors involved in liver aging. Additional studies will be necessary before justifying a randomized, controlled trial to determine their beneficial effects.

Material and methods

Ethics information

All procedures were carried out in accordance with the European Communities Council Directive (86/609/ CEE) on animal experiments and with approval from the ethical committee on animal welfare of our institution (Comité Ético de Experimentación Animal del Centro de Investigación Biomédica de La Rioja, CEEA-CIBIR). In addition, all the study was carried out in compliance with the ARRIVE guidelines (https://arriveguidelines.org). Consent to participate not applicable.

The Materials and Methods section has been published elsewhere [28]. Briefly, a total of 80 male homozygous IL-10-deficient mice (B6.129P2-IL10tm1Cgn/J) were purchased from Jackson Laboratory (Bar Harbor, ME, USA). When the animals were approximately 6 weeks old, they were randomly assigned (n = 20) to one of 4 groups and fed for 24 weeks: i) the IL-10KO group (IL-10KO) received a standard rodent diet and tap water; ii) the preventive MVC group received the same diet as the IL-10KO group and were administered MVC (Pfizer, New York, N York) in their drinking water (300 mg/L) [6, 26, 28]; iii) the preventive RAPA group received the same diet as the IL-10KO group and were administered RAPA in their drinking water (1.5 mg/kg/day) [28]; and iv) the preventive MVC plus RAPA group (MVC-RAPA) received the same diet as the IL-10KO group and were administered MVC plus RAPA in their drinking water at the same concentration as in the MVC and RAPA groups.

The mice were observed daily, and all observations were recorded. All animals were sacrificed on week 24 by CO2 exposure. Blood samples were collected under anesthesia after a 4-hour fasting period. Internal organs were examined macroscopically and weighed.

Gene expression quantification

Total RNA was extracted and purified from liver samples using an RNA RNeasy Mini Kit (Qiagen, Valencia, CA) and treated with DNase I (Qiagen) according to the manufacturer’s instructions [28]. cDNA was synthesized by reverse transcription of 1 μg of total RNA using the SuperScript III First-Strand Synthesis kit (Invitrogen, Carlsbad) in a total volume of 20 μl according to the manufacturer’s instructions, followed by amplification using SYBR Green (Takara Bio Inc, Shiga, Japan) The PCR primer sequences are listed in S1 Table. The amplification and detection of specific products were performed using an ABI PRISM 7300 system (Applied Biosystems, Foster City, CA, USA). All reactions were run in duplicate for each sample. The expression of respective genes was normalized to glyceraldehyde 3-phosphate dehydrogenase (GAPDH) as an internal control [28]. All gene interactions were investigated using the STRING database [57].

Western blot analysis

Liver samples were lysed using a homogenizer in RIPA lysis buffer (Sigma Aldrich, St. Louis, MO). The cell lysate was centrifuged at 10,000 g for 10 minutes at 4°C [28]. The concentration of total protein in each sample was determined by the Bradford method. 5´adenosine monophosphate-activated protein kinase (AMPK) [58], phosphorylated AMPK (pAMPK) [59], protein kinase-B (Akt) [60], phosphorylated Akt (pAkt) [61], nuclear factor-kB (NF-kB) [62], phosphorylated NF-kB (pNF-kB) [63], mammalian target of (mTOR) [64], phosphorylated mTOR (pmTOR) [65], signal transducer and activator of transcription 3 (STAT3) [66] and phosphorylated STAT3 [66] were evaluated by Western blotting (WB). GAPDH was used as an internal control [67] S2 Table.

Proteins were analyzed by colorimetry using a secondary antibody bound to peroxidase (anti-rabbit or mouse IgG, Cell Signaling, Danvers, MA) and incubated with the corresponding substrate [28]. After high-resolution scanning, the concentration of each protein was evaluated by densitometry using Image J software. The density values for each of the test samples were normalized to the values of GAPDH, which was used as a loading control [18].

Statistical analysis

The data are presented in the figures as the mean ± SE (standard error of the mean). For all data, the Kruskal–Wallis test was used followed by the Mann–Whitney U test. Correlations between variables were determined using the Spearman rank-sum test. We used two-way ANOVA for independent sample comparison. All data were analyzed with GraphPad Prism 6 software and were considered statistically significant when p<0.05.

Supporting information

S1 Table. List of primers employed by SybrGreen for real-time PCR.

https://doi.org/10.1371/journal.pone.0286201.s001

(PDF)

S1 Fig. Original blots of the western blot analysis of Fig 3.

https://doi.org/10.1371/journal.pone.0286201.s003

(PDF)

Acknowledgments

Maraviroc and Rapamycin, were a generous gift from Pfizer.

References

  1. 1. Niccoli T, Partridge L. Ageing as a risk factor for disease. Curr Biol. 2012; 22: 741–752. pmid:22975005
  2. 2. Campisi J. Aging, cellular senescence, and cancer. Annu Rev Physiol. 2013; 75:685–705. pmid:23140366
  3. 3. Shephard RJ. Frailty: Pathophysiology, Phenotype and Patient Care. Nestlé Nutr Inst Workshop Ser. 2015; 83:29–39.
  4. 4. Franceschi C, Bonafe M, Valensin S, Olivieri F, De Luca M, Ottaviani E, et al. Inflamm-aging. An evolutionary perspective on immunosenescence. Ann N Y Acad Sci. 2000; 908:244–54. pmid:10911963
  5. 5. Trefts E, Gannon M, Wasserman DH. The liver. Curr Biol. 2017; 27: R1147–R1151. pmid:29112863
  6. 6. Perez-Martinez L, Perez-Matute P, Aguilera-Lizarraga J, Rubio-Mediavilla S, Narro J, Recio E, et al. Maraviroc, a CCR5 antagonist, ameliorates the development of hepatic steatosis in a mousemodel of non-alcoholic fatty liver disease (NAFLD). J Antimicrob Chemother. 2014; 69:1903–1910. pmid:24651825
  7. 7. Papatheodoridi AM, Chrysavgis L, Koutsilieris M, Chatzigeorgiou A. The role of senescence in the development of nonalcoholic fatty liver disease and progression to nonalcoholic steatohepatitis. Hepatology. 2000; 71:363–374.
  8. 8. Hunt NJ, Kang SWS, Lockwood GP, Le Couteur DG, Cogger VC. Hallmarks of Aging in the Liver. Comput Struct Biotechnol J. 2019; 7: 1151–1161. pmid:31462971
  9. 9. Aravinthan A, Challis B, Shannon N, Hoare M, Heaney J, Alexander GJM. Selective insulin resistant in hepatocyte senescence. Exp Cell Res. 2015; 331: 38–45.
  10. 10. Debacq-Chainiaux F, Erusalimsky JD, Campisi J, Toussaint O. Protocols to detect senescence-associated beta-galactosidase activity (SA-betagal), a biomarker of senescent cells in culture and in vivo. Nat Protocol. 2009; 4: 1798–1806.
  11. 11. Sharpless NE, Sherr CJ. Forging a signature of in vivo senescence. Nat Rev Cancer. 2015; 15 (7):397–408. pmid:26105537
  12. 12. Lasry A, Ben-Neriah Y. Senescence-associated inflammatory responses: aging and cancer perspectives. Trends Immunol. 2015; 36: 217–228. pmid:25801910
  13. 13. Kõks S, Dogan S, Tuna BG, Gonzalez-Navarro H, Potter P, E Vandernbrouke R. Mouse models of ageing and their relevance to disease. Mech Ageing Dev. 2016; 160: 41–53. pmid:27717883
  14. 14. Zglinicki T, Varela Nieto I, Brites D, Karagianni N, Ortolano S, Georgopoulos S, et al. Frailty in mouse ageing: A conceptual approach. Mech Ageing Dev. 2016; 160:34–40. pmid:27443148
  15. 15. Walston J, Fedarko N, Yang H, Leng S, Beamer B, Espinoza S, et al. The physical and biological characterization of a frail mouse model. J Gerontol A Biol Sci Med Sci. 2008; 63: 391–398. pmid:18426963
  16. 16. Fiorentino DF, Zlotnik A, Mosmann TR, Howard M, O´Garra A. IL-10 inhibits cytokineproduction by activated macrophages. J Immunol 147; 1991: 3815–22. pmid:1940369
  17. 17. Kühn R, Löhler J, Rennick D, Rajewsky K, Müller W. Interleukin-10-deficient mice develop chronic enterocolitis. Cell. 1993; 75: 263–274. pmid:8402911
  18. 18. Mannick JB, Morris M, Hockey HU, Roma G, Beibel M, Kulmatycki K, et al. TORC1 inhibition enhances immune function and reduces infections in the elderly. Sci Transl Med. 2018; 10: eaaq1564.18. pmid:29997249
  19. 19. Itoh Y, Saitoh M, Miyazawa K. Smad3-STAT3 crosstalk in pathophysiological contexts. Acta Biochim Biophys Sin. 2018; 1; 50 (1):82–90. pmid:29140406
  20. 20. Laplana M, Fibla J. Distribution of functional polymorphic variants of inflammation-related genes RANTES and CCR5 in long-lived individuals. Cytokine. 2012; 58:10–13. pmid:22265023
  21. 21. Oppermann M. Chemokine receptor CCR5: insights into structure, function, and regulation. Cell Signal. 2004; 16(11):1201–10. pmid:15337520
  22. 22. Lu Y, Tan CT, Shwe Zin Nyunt, Wing Hei E, Camous X, Kared H, et al. Inflammatory and immune markers associated with physical frailty syndrome: findings from Singapore longitudinal aging studies. Oncotarget. 2016; 7:28783–28795. pmid:27119508
  23. 23. Harrison DE, Strong R, Sharp Z, Nelson JF, Astle CA, Flurkey K, et al. Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature. 2019; 460: 392–395.
  24. 24. Wilkinson JE, Burmeister L, Brooks SV, Chan CC, Friedline S, Harrison DE, et al. Rapamycin slows aging in mice. Aging. Cell. 2012; 11:675–682. pmid:22587563
  25. 25. Gilliam BL, Riedel DJ, Redfield RR. Clinical use of CCR5 inhibitors in HIV and beyond. J Transl Med. 2011; 9: Suppl 1:S9. pmid:21284908
  26. 26. De Fanis UC, Wang GC, Fedarko NS, Walston JD, Casolaro V, Leng SX. T-lymphocytes expressing CC chemokine receptor-5 are increased in frail older adults. J Am Geriatr. 2008; 56: 904–908. pmid:18384587
  27. 27. Ochoa-Callejero L, Pérez-Martinez L, Rubio-Mediavilla S, Oteo JA, Martinez A, Blanco JR. Maraviroc, a CCR5 antagonist, prevents the development of hepatocellular carcinoma in a model of mouse. PLoS One. 2013; 8: e53992.
  28. 28. Pérez-Martínez L, Romero L, Muñoz-Galvan S, Verdugo-Sivianes E, Rubio-Mediavilla S, Oteo JA, et al. Implications of maraviroc and/or rapamycin in a mouse model of fragility. Aging (Albany NY). 2020; 12: 8565–8582. pmid:32353830
  29. 29. Salminen A, Kaarniranta K. NF-kappaB signaling in the aging process. J Clin Immunol. 2009; 29: 397–405. pmid:19408108
  30. 30. Kennedy BK, Pennypacker JK. Drugs that modulate aging: the promising yet difficult path ahead. Transl Res. 2014; 163: 456–465. pmid:24316383
  31. 31. Rando TA, Wyss-Coray T. Stem cells as vehicles for youthful regeneration of aged tissues. J Gerontol A Biol Sci Med Sci. 2014; 69: Suppl 1:39–42. pmid:24833585
  32. 32. Jonhson SC, Rabinovitch PS, Kaeberlein M. mTOR is a key modulator of ageing and age-related disease. Nature. 2013; 493: 338–45. pmid:23325216
  33. 33. Harrison DE, Strong R, Allison DB, Ames BN, Astle CM, Atamma H, et al. Acarbose, 17-alpha-estradiol, and nordihydroguaiaretic acid extend mouse lifespan preferentially in males. Aging Cell. 2014; 13: 273–282.
  34. 34. Martin-Montalvo A, Mercken EM, Mitchell SJ, Palacios HH, Mote PL, Scheibye-Knudsen M, et al. Metformin improves healthspan and lifespan in mice. Nat Commun. 2013; 4: 2192. pmid:23900241
  35. 35. Papadopoli D, Boulay K, Kasak L, Pollak M, Mallette F, Topisirovic I, et al. mTOR as a central regulator of lifespan and aging. F1000Res 8: F1000 Faculty Rev. 2019; 998. pmid:31316753
  36. 36. Mannick JB, Morris M, Hockey HU, Roma G, Beibel M, Kulmatycki K, et al. TORC1 inhibition enhances immune function and reduces infections in the elderly. Sci Transl Med. 2018; 11; 10:eaaq1564. pmid:29997249
  37. 37. Chen SC, Chang YL, Wang DL, Cheng JJ. Herbal remedy magnolol suppresses IL‐6‐induced STAT3 activation and gene expression in endothelial cells. Br J Pharmacol. 2006; 148 (2): 226–232. pmid:16520748
  38. 38. Matsumoto S, Hara T, Mitsuyama K, Yamamoto M, Tsuruta O, Sata M et al. Essential roles of IL-6 trans-signaling in colonic epithelial cells, induced by the IL-6/soluble–IL-6 receptor derived from lamina propria macrophages, on the development of colitis-associated premalignant cancer in a murine model. J Immunol. 2010; 184 (3):1543–1551. pmid:20042582
  39. 39. Zi J, Yuan S, Qiao J, Zhao K, Xu L, Qi K, et al. Treatment with the C-C chemokine receptor type 5 (CCR5)-inhibitor maraviroc suppresses growth and induces apoptosis of acute lymphoblastic leukemia cells. Am J Cancer Res. 2017; 7(4):869–880. pmid:28469959
  40. 40. Lamming DW. Inhibition of the mechanistic target of rapamycin (mTOR)-rapamycin and beyond. Cold Spring Harb Perspect Med. 2016; 2; 6:a025924. pmid:27048303
  41. 41. Yang H, Jiang X, Li B, Yang HJ, Miller M, Yang A, et al. Mechanisms of mTORC1 activation by RHEB and inhibition by PRAS40. Nature. 2017; 552: 368–373. pmid:29236692
  42. 42. Schreiber KH, Arriola Apelo SI, Yu D, Brinkman JA, Velarde MC, Syed FA, et al. A novel rapamycin analog is highly selective for mTORC1 in vivo. Nat Commun. 2019; 19: 10:3194. pmid:31324799
  43. 43. Li J, Kim, SG, Blenis J. Rapamycin: one drug, many effects. Cell Metab. 2014; 4; 19:373–379.
  44. 44. Carnero A, Paramio JM. The PTEN/PI3K/AKT Pathway in vivo. Cancer Mouse Models. Front Oncol. 2014; 23; 4:252.
  45. 45. Revathidevi S, Munirajan AK. Akt in cancer: Mediator and more. Semin Cancer Biol. 2019; 59:80–91. pmid:31173856
  46. 46. El Assar M, Angulo J, Rodriguez-Mañas L. Oxidative stress and vascular inflammation in aging. Free Radic Biol Med. 2013; 65:380–401. pmid:23851032
  47. 47. Zhang H, Davies KJA, Forman HJ. Oxidative stress response and Nrf2 signaling in aging. Free Radic Biol Med. 2015; 88(Pt B):314–336. pmid:26066302
  48. 48. Sun X, Chen L, He Z. PI3K/Akt-Nrf2 and Anti-Inflammation Effect of Macrolides in Chronic Obstructive Pulmonary Disease. Curr Drug Metab. 2019; 20:301–304. pmid:30827233
  49. 49. Adler A, Sinha S, Kawahara T, Zhang JY, Segal E, Chang HY. Motif module map reveals enforcement of aging by continual NF-kappaB activity. Genes Dev. 2007; 21: 3244–3257. pmid:18055696
  50. 50. Dan HC, Baldwin AS. Differential involvement of IkappaB kinases alpha and beta in cytokine- and insulin-induced mammalian target of rapamycin activation determined by Akt. J Immunol. 2008; 180: 7582–7589. pmid:18490760
  51. 51. Coppe J P, Patil CK, Rodier F, Krtolica A, Beausejour CM, Parrinello S, et al. A human-like senescence-associated secretory phenotype is conserved in mouse cells dependent on physiological oxygen. PLoS One. 2010; 5: e9188. pmid:20169192
  52. 52. Salvioli S, Monti D, Lanzarini C, Conte M, Pirazzini C, Bacalini MG, et al. Immune system, cell senescence, aging and longevity—inflamm-aging reappraised. Curr Pharm Des. 2013; 19: 1675–1679. pmid:23589904
  53. 53. Selvarani R, Mohammed S. Effect of rapamycin on aging and age-related diseases-past and future. Geroscience. 2021; 43:1135–1158. pmid:33037985
  54. 54. Childs BG, Gluscevic M. Senescent cells: an emerging target for diseases of ageing. Nat Rev Drug Discov. 2017; 16: 718–735. pmid:28729727
  55. 55. Tchkonia T, Zhu Y, Deursen J, Campisi J, Kirkland JL. Cellular senescence and the senescent secretory phenotype: therapeutic opportunities. J Clin Invest. 2013; 123:966–72. pmid:23454759
  56. 56. Soria G, Ben-Baruch A. The inflammatory chemokines CCL2 and CCL5 in breast cancer. Cancer Lett. 2008; 267: 271–285. pmid:18439751
  57. 57. Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, et al. STRING v11: proteinprotein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 2019; 47:D607–13. pmid:30476243
  58. 58. Li Q, Xu Q, Tan J, Hu L, Ge C, Xu M, et al. Carminic acid supplementation protects against fructose-induced kidney injury mainly through suppressing inflammation and oxidative stress via improving Nrf-2 signaling. Aging (Albany NY). 2021; 13: 10326–10353. pmid:33819919
  59. 59. Niu Y, Zhang Y, Zhang W, Lu J, Chen Y, Hao W, et al. Canagliflozin ameliorates NLRP3 inflammasome-mediated inflammation through inhibiting NF-κB signaling and upregulating Bif-1. Front Pharmacol. 2022; 28: 820541.
  60. 60. Zhang ZM, Liu ZH, Nie Q, Zhang XM, Yang LQ, Wang C, et al. Metformin improves high-fat diet-induced insulin resistance in mice by downregulating the expression of long noncoding RNA NONMMUT031874.2. Exp Ther Med. 2022; 23: 332. pmid:35401798
  61. 61. Fu J, Zhao J, Zhang H, Fan X, Geng W, Qio S. MicroRNA-451a prevents cutaneous squamous cell carcinoma progression via the 3-phosphoinositide-dependent protein kinase-1-mediated PI3K/AKT signaling pathway. Exp Ther Med. 2021; 21: 116. pmid:33335579
  62. 62. Chen Q, Ren D, Wu J, Yu H, Chen X, Wang J, et al. Shenyan Kangfu tablet alleviates diabetic kidney disease through attenuating inflammation and modulating the gut microbiota. J Nat Med. 2021; 75: 84–98. pmid:32997272
  63. 63. Wang J, Chen J, Qiu D, Zeng Z. Regulatory role of DEPTOR-mediated cellular autophagy and mitochondrial reactive oxygen species in angiogenesis in multiple myeloma. Int J Mol Med. 2021; 47: 643–658.
  64. 64. Wang B, Qin Y, Wu Q, Li X, Xie D, Zhao Z, et al. mTOR signaling pathway regulates the release of proinflammatory molecule CCL5 implicated in the pathogenesis of autism spectrum disorder. Front Immunol. 2022; 13: 818518. pmid:35422816
  65. 65. Xie B, Zhao M, Song D, Wu K, Yi L, Li W, et al. Induction of autophagy and suppression of type I IFN secretion by CSFV. Autophagy. 2021; 17: 925–947. pmid:32160078
  66. 66. Liu SP, Harn HJ, Chien YJ, Chang CH, Hsu CY, Fu RH, et al. n-Butylidenephthalide (BP) maintains stem cell pluripotency by activating Jak2/Stat3 pathway and increases the efficiency of iPS cells generation. PLoS One. 2012;7:e44024. pmid:22970157
  67. 67. Nakasuka F, Tabata S, Sakamoto T, Hirayama A, Ebi H, Yamada T, et al. TGF-β-dependent reprogramming of amino acid metabolism induces epithelial-mesenchymal transition in non-small cell lung cancers. Communications biology. 2021; 4: 782.