Journal Search Engine
Search Advanced Search Adode Reader(link)
Download PDF Export Citaion korean bibliography PMC previewer
ISSN : 1226-7155(Print)
ISSN : 2287-6618(Online)
International Journal of Oral Biology Vol.49 No.3 pp.69-78
DOI : https://doi.org/10.11620/IJOB.2024.49.3.69

Decline in sestrin 2 expression during aging shifts mesenchymal stem cell differentiation from osteogenic to adipogenic lineage

Do Yeun Kim, Hyun-Jung Park, Jeong-Hwa Baek*
Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 08826,
Republic of Korea
*Correspondence to: Jeong-Hwa Baek, E-mail: baekjh@snu.ac.krhttps://orcid.org/0000-0002-2032-1683
May 23, 2024 August 29, 2024 September 9, 2024

Abstract


Sestrin 2 (SESN2) is a member of the sestrin family of stress-induced proteins that negatively regulate agingassociated biological processes. This study aims to investigate the role of SESN2 in regulating the differentiation potential and senescence of mesenchymal stem cells (MSCs) derived from young and elderly donors. Bulk RNA sequencing revealed a common decline in the SESN2 mRNA levels in MSCs from elderly individuals, which was confirmed via reverse transcription-polymerase chain reaction and western blot analyses. SESN2 knockdown in MSCs from young donors resulted in phenotypic changes similar to those in MSCs from elderly donors, including an enhanced expression of senescence and adipogenic markers and diminished expression of osteogenic markers. To confirm the effect of decreased SESN2 expression on osteogenic and adipogenic differentiation, we induced Sesn2 knockdown in mouse bone marrow-derived MSCs. Sesn2 knockdown suppressed the mRNA expression of osteogenic marker genes, alkaline phosphatase activity, and matrix mineralization. Furthermore, Sesn2 knockdown enhanced mRNA expression of the adipogenic marker genes and intracellular lipid accumulation. These results suggest that a decline in SESN2 expression during aging contributes to the shift of MSC differentiation from osteogenic to adipogenic lineage.



초록


    © The Korean Academy of Oral Biology. All rights reserved.

    This is an open-access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/bync/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

    Introduction

    The sestrin family comprises three highly conserved proteins: sestrin 1 (SESN1), sestrin 2 (SESN2), and sestrin 3 (SESN3). These proteins are stress-inducible, and play crucial roles in cellular responses to various stressors, such as oxidative stress, genotoxic stress, and nutrient deprivation [1]. Sestrins primarily function by regulating antioxidant defense mechanisms, maintaining metabolic homeostasis, and modulating cell survival pathways. Notably, they inhibit the mammalian target of rapamycin complex 1 (mTORC1) signaling pathway, a key regulator of cell growth and metabolism, thereby linking nutrient sensing to cellular stress responses [2,3]. Sestrin isoforms exhibit overlapping expression patterns and functions, but they can also display distinct expression profiles, regulatory mechanisms, and functions [4]. SESN1 is ubiquitously expressed in humans, including in skeletal muscle, heart, liver, and brain. SESN2 is highly expressed in various tissues, particularly in the kidney, lungs, liver, and brain. Similarly, SESN3 is prominently expressed in skeletal muscle, intestine, liver, and brain. Notably, in skeletal muscle, SESN1 expression levels are higher than those of SESN3 [5]. The p53 tumor suppressor protein induces the expression of both SESN1 and SESN2, whereas SESN3 expression is primarily regulated by FoxO transcription factors [4]. SESN1 and SESN3 are involved in the inhibition of oxidative stress, nutrient sensing, suppression of mTORC1, and induction of autophagy. Additionally, SESN3 plays a role in the regulation of mTORC2 and the maintenance of glucose homeostasis. Beyond the functions shared with SESN1, SESN2 is also involved in the inhibition of DNA damage and endoplasmic reticulum stress, as well as the maintenance of glucose and lipid homeostasis [4].

    Recent studies have highlighted the role of SESN2 as a regulator of aging and a potential therapeutic target for age-related diseases [6]. In response to oxidative stress, SESN2 is upregulated, activating antioxidant defenses and protecting cells from oxidative damage, a major contributor to aging and age-related diseases. Additionally, SESN2 may influence aging by inhibiting mTORC1, as hyperactivation of mTORC1 is linked to reduced autophagy, increased cellular senescence, and aging [7,8].

    Mesenchymal stem/stromal cells (MSCs) are multipotent non-hematopoietic cells found in various tissues, including bone marrow, adipose tissue, skin, placenta, and thymus [9]. Bone marrow-derived MSCs (BMSCs) can differentiate into osteoblasts, chondrocytes, and adipocytes. However, aging impairs BMSCs differentiation into osteoblasts, the cells responsible for bone formation, while enhancing adipogenic differentiation, leading to fat cell accumulation in the bone marrow. This imbalance contributes to the reduced bone density and increased fracture risk observed in older individuals [10,11]. Various studies have identified the molecular mechanisms underlying these changes. For example, an increase in miR- 188 and a decrease in miR-130a expression in aging BMSCs are associated with bone loss and fat accumulation in bone marrow [12,13]. In addition, compared to cells from young mice, BMSCs obtained from aged mice exhibit significantly reduced lipolysis and fatty acid oxidative metabolic activity [14]. The predominant adipogenic differentiation of skeletal stem/ progenitor cells has been suggested to be associated with inflamm-aging, which in turn further triggers stem cell dysfunction [15]. Adipokines and other hormones released from mature adipocytes in aged bone marrow may create a vicious cycle that drives the differentiation fate of neighboring BMSCs toward adipocytes rather than osteoblasts [16]. Despite these findings, the mechanisms driving age-related changes in BMSCs differentiation require further investigation.

    Enhancement of AMP-activated protein kinase (AMPK) activity is known to promote osteogenesis and inhibit adipogenesis [17]. Given that SESN2 activates AMPK through mTORC1 inhibition [18], this study aims to investigate whether aging regulates SESN2 expression in BMSCs and determine SESN2’ s regulatory role in osteogenic and adipogenic differentiation of BMSCs.

    Materials and Methods

    1. Cell culture

    Human MSCs were purchased from Sigma-Aldrich, Promo- Cell, and STEMCELL Technologies. The age distribution of the human MSCs sources included both young (Y; 18 and 22 years old) and aged (O; 70 to 82 years old) donors. Mouse BMSCs were extracted from 8-week-old C57BL/6 mice obtained from Korea Research Institute of Bioscience and Biotechnology. The bone marrow was extracted from the mouse femur and tibia and cultured in MesenCultTM Basal Medium (mouse) (STEMCELL Technologies) until passage 3. Subsequently, CD31/ CD45-negative cells were sorted using FACSAria Fusion (BD Biosciences). Sorted mouse BMSCs and human MSCs were cultured in alpha-minimal essential medium high glucose (Hy- Clone) supplemented with 10% fetal bovine serum (HyClone), 100 U/mL penicillin, and 100 μg/mL streptomycin.

    Osteogenic differentiation was induced by supplementing the growth medium with 50 μg/mL ascorbic acid, 10 mM β-glycerophosphate, and 100 nM dexamethasone. The medium was changed every other day. At the end of the specified culture periods, cells were subjected to RNA isolation, alkaline phosphatase (ALP) activity assay, or staining for ALP or mineralized matrix (Alizarin Red S) as described previously [19].

    When cell confluency reached 100%, adipogenic differentiation was induced by adding supplements (1 μM dexamethasone, 0.5 mM IBMX, 50 μM indomethacin, and 10 μg/mL insulin) to the growth medium. The medium was changed every other day, and at the end of the culture periods, cells were subjected to RNA isolation or Oil Red O staining as described previously [20].

    2. Sestrin 2 knockdown using siRNA

    Non-targeting siRNAs and siRNAs for human and mouse sestrin 2 genes were purchased from Santa Cruz Biotechnology (human: sc-106544; mouse: sc-153380). For mouse BMSCs, control siRNA and Sesn2 siRNA were transfected using Lipofectamine 2000 (Invitrogen). For human MSCs, siRNA transfection was performed using an electroporator with a gold tip at 990 mV for 40 ms.

    3. Quantitative reverse transcription-polymerase chain reaction

    Total RNA was extracted from cells using QIAzol Lysis Reagent (QIAGEN), and cDNA was synthesized using AccuPower ® RT PreMix (Bioneer).

    The primer sequences for mouse genes were as follows (5′→3′): Gabph , TCAATGACAACTTTGTCAAGC and CCAGGGT T TCT TACTCCT TGG; Sesn2 , GGCGGTGGTGATGGGTCTAC and GACGACCCGGAAGTGGCCC; Alpl , CCAACTCTTTTGTGCCAG and GGCTACATTGGTGTTGAGCTTTT; Ibsp, CAGGGAGGCAGTGACTCTTC and AGTGTGGAAAGTGTGGCGTT; Bglap , CTGACAAAGCCTTCATGTCCAA and GCGCCGGAGTCTGTTCACTA; Cebpα , TGGTCCCCGTGTCCTCCTA and TCAGACCAGAAAGCTGAGTTGTG; Pparγ , CCGAAGAACCATCCGATTGAA and GCCCAAACCTGATGGCATT; Ap2 , AAAGAAGTGGGAGTGGGCTT and CTCTTGTGGAAGTCACGCCT; Plin1, TACCCTCCAGAAAAGATCGC and CTACCACCTTCTCGATGCTT; and Adipoq , TCTTGGTCCTAAGGGTGAGA and GTTGCAGTAGAACTTGCCAG.

    The primer sequences for the human genes were as follows (5′→3′): GAPDH , CCATCTTCCAGGAGCGAGATC and GCCTTCTCCATGGTGGTGAA; SESN2 , CCTGCTCAGGAGTCAGGTCA and CAACCTCCTCTGGAGGCACTT; P16 , GGGTCGGGTAGAGGAGGTG and CATCATGACCTGGATCGGC; P21 , ACCGAGACACCACTGGAGGG and CGAGGCACAAGGGTACAAGACA; ALPL , AACTTCCAGACCGGCTTGA and TTGCCGCGTGTCTT; OSX , ACCTACCCATCTGACTTTGCT and CCACTATTTCCCACTGCCTT; RUNX2 , CAGATGGGACTGTGGCTGT and GTGAAGACGGTTATGAAGG; BGLAP , CGCTACCTGTATCAATGGCTGG and CTCCTGAAAGCCGATGTGGTCA; LPL, GTGACCAAGGTAGACCAGCC and GAAGAGACTTCAGGCAGCGT; CEBPα , TCGGTGGACAAGAACAG and GCAGGCGGTCATT; CEBPβ, CGCTTACCTCGGCTACCA and ACGAGGAGGACGTGGAGAG; PPARγ , TTCTCCTATTGACCCAGAAAGC and CTCCACTTTGATTGCACTTTGG; AP2, CATCAGTGTGAATGGGGATG and GTGGAAGTGACGCCTTTCAT; and ADIPOQ, TTCACCGATGTCTCCCTTAGG and GGCATGACCAGGAAACCAC.

    4. Western blot analysis

    Protein samples for western blotting were extracted using PRO-PREPTM (iNtRON Biotechnology). Samples were subjected to sodium dodecyl sulfate-polyacrylamide gel electrophoresis and transferred onto a polyvinylidene difluoride membrane. Blocking was performed using Tris-buffered saline containing 0.1% Tween 20 and 5% skim milk. The membranes were incubated with primary antibodies, followed by incubation with horseradish peroxidase (HRP)-conjugated secondary antibodies. Pico ECL reagent was used for detection by a Bio-Image Analyzer (Vilber Lourmat).

    Antibodies for p16, p21, Lamin B, osteocalcin (OCN), osterix (OSX), RUNX2, SESN2, and β-actin were purchased from Santa Cruz Biotechnology. Antibodies for γH2AX, PPARγ, and adiponectin (ADIPOQ) were obtained from Cell Signaling Technology, and HRP-conjugated secondary antibodies were from Thermo Fisher Scientific.

    5. Senescence-associated β-galactosidase staining

    Cultured cells were stained using the Senescence β-Galactosidase Staining Kit (Cell Signaling Technology) according to the manufacturer’s instructions. Briefly, cells were washed with phosphate buffered saline (PBS) and fixed in β-gal staining fix solution for 15 minutes at room temperature. Cells were then washed with PBS and incubated with β-gal staining solution for 20–24 hours at 37℃. After washing with PBS, cells were observed under a bright-field microscope.

    6. Statistical analysis

    Quantitative data are represented as the mean ± standard deviation. Statistical significance was analyzed using Student’s t-test or one-way ANOVA with Tukey-Kramer post hoc test. A p-value less than 0.05 was considered significant.

    Results

    1. MSCs obtained from the elderly express higher levels of cellular senescence markers

    We first examined the differences in cellular senescence marker levels between human MSCs obtained from young and elderly donors. Compared to cells from young donors, MSCs from elderly donors exhibited strong senescence-associated β-galactosidase (SA β-gal) staining (Fig. 1A). Additionally, MSCs from elderly donors expressed higher levels of cellular senescence markers such as p16, p21, and γH2AX (Fig. 1B and 1C, Supplementary Table 1B). These results suggest that BMSCs obtained from elderly individuals have a higher population of cells exhibiting cellular senescence compared to those from younger individuals.

    2. MSCs from the elderly express lower levels of osteogenic marker genes and higher levels of adipogenic marker genes compared to those from the young

    We next examined the basal expression levels of osteogenic and adipogenic differentiation marker genes in MSCs from young and elderly donors. As shown in Fig. 2A, MSCs cultured in growth medium expressed some of these marker proteins. Western blot results indicated that MSCs from elderly donors expressed lower levels of osteogenic marker proteins (RUNX2, OSX, and OCN) while expressing higher levels of adipogenic markers (PPARγ and ADIPOQ) compared to MSCs from young donors. Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) also demonstrated similar results, showing lower levels of osteogenic marker mRNAs (ALPL, RUNX2, and OSX) and higher levels of adipogenic marker mRNAs (PPARγ and AP2) in MSCs from elderly donors (Fig. 2B, Supplementary Table 2B). These results suggest that MSCs from elderly individuals have already acquired more adipogenic characteristics with a corresponding decrease in osteogenic traits, even in the absence of exogenous differentiation signals.

    To further confirm that MSCs from elderly donors have higher potential for adipogenic differentiation but lower potential for osteogenic differentiation, the cells were cultured for 14 days under osteogenic (Fig. 2C, Supplementary Table 2C) or adipogenic conditions (Fig. 2D, Supplementary Table 2D). qRTPCR results demonstrated that under differentiation-inducing conditions, MSCs from elderly donors exhibited significantly enhanced expression levels of adipogenic marker genes and suppressed levels of osteogenic marker genes compared to MSCs from young donors. These results confirm that during aging, the differentiation potential of MSCs shifts from the osteogenic to the adipogenic lineage.

    3. The expression levels of SESN2 are decreased in MSCs from the elderly

    Bulk RNA sequencing (RNA-seq) was performed using MSCs from young and elderly donors. RNA-seq results showed that among the sestrin family members, the average FPKM levels of SESN2 were significantly lower in MSCs from elderly donors compared to those from young donors (Fig. 3A). To confirm the RNA-seq results, mRNA and protein samples were prepared from six batches of human MSCs obtained from 18-, 22-, 70-, 77-, and 82-year-old donors. Both qRT-PCR and western blot analyses (Fig. 3B and 3C, Supplementary Table 3B) demonstrated that MSCs from elderly donors expressed lower levels of SESN2 compared to MSCs from young donors. These results suggest that SESN2 expression in MSCs declines with aging.

    4. Knockdown of SESN2 in young human MSCs suppresses osteogenic marker gene expression while enhancing the expression of adipogenic and senescence marker genes

    To examine whether the decrease in SESN2 expression contributes to the phenotypic changes observed in MSCs from elderly donors, SESN2 knockdown was performed in young MSCs using siRNAs. Knockdown of SESN2 resulted in more than a three-fold induction of senescence marker gene (P16 and P21) expression (Fig. 4A, Supplementary Table 4A). Furthermore, SESN2 knockdown in young MSCs altered the basal expression levels of osteogenic and adipogenic marker genes in the absence of exogenous differentiation-inducing stimuli (Fig. 4B and 4C, Supplementary Table 4B and 4C). Similar to the phenotype of MSCs from elderly donors, SESN2 - knockdown young MSCs expressed lower levels of osteogenic marker gene mRNAs but higher levels of adipogenic marker gene mRNAs. These results suggest that the decline in SESN2 expression levels in MSCs during aging plays a role in shifting the differentiation fate of BMSCs from the osteogenic to the adipogenic lineage.

    5. S esn2 knockdown inhibits osteogenic differentiation but enhances adipogenic differentiation of mouse BMSCs

    To further confirm the regulatory effect of SESN2 loss on the osteogenic and adipogenic differentiation potential of BMSCs, similar experiments were performed using mouse BMSCs prepared from 8-week-old mice. Knockdown of Sesn2 in mouse BMSCs significantly decreased the mRNA levels of osteogenic marker genes, including Alpl, Ibsp, and Bglap (Fig. 5A, Supplementary Table 5A). Additionally, ALP activity and matrix mineral deposition were reduced in Sesn2-knockdown BMSCs (Fig. 5B and 5C, Supplementary Table 5B and 5C).

    When mouse BMSCs were incubated under adipogenic conditions, Sesn2 knockdown significantly enhanced the expression of adipogenic marker mRNAs, including Cebpα, PPARγ, Adipoq, and Plin1 (Fig. 6A, Supplementary Table 6A). Furthermore, Oil Red O staining showed that intracellular lipid accumulation was increased by Sesn2 knockdown (Fig. 6B). These results suggest that SESN2 in BMSCs plays a role in regulating differentiation fate in favor of the osteogenic lineage.

    Discussion

    In this study, we demonstrated that SESN2 expression levels are decreased in MSCs from elderly donors compared to those from young donors. Furthermore, SESN2 knockdown in MSCs from young donors altered their cellular phenotypes to mimic those of MSCs from elderly donors, resulting in enhanced adipogenic potential and cellular senescence marker expression, but diminished osteogenic potential.

    Recent studies indicate that senescent cells accumulate in the bone microenvironment with aging and that targeting these cells with senolytic and/or senomorphic drugs prevents aging-related bone loss, at least in mice [21]. Various cell types in the bone microenvironment, including osteoblast progenitors, osteoblasts, osteocytes, myeloid cells, and B and T cells, increase the expression of senescence-associated secretory phenotype factors with aging. Consistent with previous reports, we found that MSCs from elderly donors exhibited an increased number of SA β-gal-positive senescent cells and higher levels of p16, p21, and γH2AX expression [22]. Additionally, MSCs from elderly individuals showed higher basal levels of adipogenic genes and increased adipogenic marker gene expression under adipogenic conditions, while exhibiting lower levels of osteogenic marker gene expression both in the absence and presence of osteogenic stimuli. These results align with previous findings and the bone phenotypes observed in the elderly individuals [10,11].

    In this study, bulk RNA-seq analysis of MSCs from young and elderly donors revealed a common decline in SESN2 mRNA levels in MSCs from elderly individuals. Sestrins are induced by various stressors and negatively regulate agingassociated biological processes. While no prior reports specifically noted a decline in SESN2 expression in MSCs from aged donors, some studies have linked reduced SESN2 levels to aging-associated musculoskeletal diseases. For instance, the number of SESN2-positive chondrocytes in articular cartilage declines with aging and osteoarthritis in both humans and mice [23]. Additionally, serum SESN2 levels are significantly reduced in sarcopenic and frail older subjects compared to non-sarcopenic and non-frail ones, respectively [24,25]. These findings, along with our results, suggest that a decline in SESN2 is associated with aging-induced loss of bone and muscle.

    To explore the functional role of SESN2 in MSC phenotypic maintenance, we performed knockdown experiments. SESN2 knockdown in MSCs from young donors increased the expression levels of p16 and p21 mRNAs. Previous studies reported that mouse embryonic fibroblasts from SESN2-deficient mice exhibit cellular senescence phenotypes, including enhanced SA β-gal staining and decreased cell proliferation [26]. Additionally, SESN2 overexpression reduces SA β-gal activity and levels of p16 and p21 proteins in macrophages [27]. These findings, together with our results, suggest that maintaining SESN2 expression plays a role in preventing senescence of MSCs.

    SESN2 knockdown also altered the differentiation potential of young MSCs, favoring the adipogenic lineage. Similarly, SESN2 knockdown significantly enhanced intracellular lipid accumulation and adipogenic marker gene expression in mouse BMSCs in this study. These results are consistent with reports showing that a loss of Sesn in Drosophila leads to triglyceride accumulation, reversible by AMPK activation or TOR inhibition [2].

    Consistent with enhanced adipogenic differentiation, SESN2 knockdown inhibited osteogenic differentiation and matrix mineralization in mouse BMSCs. These results suggest that SESN2 plays a role in maintaining the osteogenic differentiation potential of MSCs in bone marrow. Although direct regulatory effects of SESN2 on MSC osteogenic differentiation have not been reported, SESN2 in aortic valve macrophages inhibits oxidized low-density lipoprotein-induced M1 polarization, indirectly suppressing the osteogenic differentiation of co-cultured valvular interstitial cells [28]. The inhibitory effect of SESN2 on osteogenic differentiation of valvular interstitial cells is mediated through its antioxidant and anti-inflammatory effects in macrophages, suggesting a potential indirect regulatory mechanism. Therefore, direct regulatory effect of SESN2 on osteogenic differentiation of valvular interstitial cells remains elusive.

    In this study, we did not investigate the molecular mechanisms underlying SESN2 deficiency-induced phenotypic changes in MSCs. However, previous studies have shown that oxidative stress and mTORC1 activation favor adipogenic lineage, while AMPK activation favors osteogenic lineage in MSC differentiation [29-31]. Thus, it is speculated that SESN2 regulates MSC differentiation potential by inhibiting oxidative stress and mTORC1 activity and activating AMPK. Further studies are needed to elucidate these regulatory mechanisms in MSCs.

    In conclusion, we demonstrated for the first time that a decline in SESN2 expression during aging contributes to the shift in MSC differentiation fate from osteogenic to adipogenic lineage. These results enhance our understanding of the pathophysiological mechanisms underlying aging-associated bone loss.

    Funding

    This study was supported by the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT) (No. 2019R1A2C1003709).

    Conflicts of Interest

    No potential conflict of interest relevant to this article was reported.

    Supplementary Data

    Supplementary data is available at http://www.kijob.or.kr only.

    Figure

    IJOB-49-3-69_F1.gif

    Human mesenchymal stem cells (MSCs) obtained from the elderly exhibit higher expression levels of cellular senescence markers compared to MSCs from the young. (A) MSCs from old individuals (O) showed a higher level of senescence-associated β-galactosidase staining compared to MSCs from young individuals (Y). (B, C) The expression levels of cellular senescence markers were examined by western blot analysis (B) and qRT-PCR (C). mRNA levels of each gene were normalized to GAPDH and presented as a ratio to relative to values obtained from young MSCs. Quantitative data are presented as the mean ± SD.

    qRT-PCR, quantitative reverse transcription-polymerase chain reaction; SD, standard deviation.

    *p < 0.05, compared to Y.

    IJOB-49-3-69_F2.gif

    Mesenchymal stem cells (MSCs) from elderly individuals exhibit higher adipogenic but lower osteogenic potential than those from young individuals. (A, B) Young and old human MSCs were cultured in growth medium, followed by western blot analysis (A) and qRT-PCR (B) to compare the expression levels of osteogenic and adipogenic markers. (C, D) MSCs were incubated for 14 days in osteogenic medium (C) or adipogenic medium (D), followed by qRT-PCR. Quantitative data are presented as the mean ± SD.

    OCN, osteocalcin; OSX, osterix; ADIPOQ, adiponectin; qRT-PCR, quantitative reverse transcription-polymerase chain reaction; SD, standard deviation.

    *p < 0.05, compared to Y.

    IJOB-49-3-69_F3.gif

    Mesenchymal stem cells (MSCs) from elderly individuals express lower levels of sestrin 2 (SESN2) compared to those from young individuals. (A) Bulk RNA sequencing (RNA-seq) of young and old human MSCs showed SESN2 mRNA expression declined in MSCs from elderly individuals. Average FPKM of each group was used to calculate fold change and p-value. (B, C) RNA-seq results were verified using RNA and protein samples obtained from MSCs of two young adults (Y: 18 and 22 years old) and four elderly individuals (O: 70 to 82 years old). mRNA expression levels of SESN2 were examined by qRT-PCR (B) and SESN2 protein levels were confirmed by western blot analysis (C). Relative mRNA levels are presented as a ratio relative to values obtained from young MSCs (from 18-year-old donor). Quantitative data are presented as the mean ± SD and statistical significance was tested by one-way ANOVA and Tukey-Kramer post hoc analysis.

    qRT-PCR, quantitative reverse transcription-polymerase chain reaction; SD, standard deviation.

    *p < 0.05.

    IJOB-49-3-69_F4.gif

    Sestrin 2 (SESN2) knockdown in young mesenchymal stem cells (MSCs) suppresses osteogenic marker gene expression while enhancing adipogenic gene expression. Young MSCs were transiently transfected with non-targeting control siRNA (si CON) or SESN2-targeting siRNA (si SESN2), followed by incubation in growth medium for 72 hours. mRNAs were then prepared for qRT-PCR analyses of cellular senescence markers (A), osteogenic markers (B), and adipogenic marker genes (C). Quantitative data are presented as the mean ± SD.

    qRT-PCR, quantitative reverse transcription-polymerase chain reaction; SD, standard deviation.

    *p < 0.05, compared to si CON.

    IJOB-49-3-69_F5.gif

    Sestrin 2 (Sesn2) knockdown inhibits osteogenic differentiation of mouse bone marrow-derived MSCs (BMSCs). BMSCs prepared from 8-week-old mice were transfected with non-targeting control siRNA (si Con) or Sesn2 siRNA (si Sesn2). These cells were cultured in osteogenic medium for the indicated periods, and osteogenic differentiation was evaluated. (A) 10 days after osteogenic induction, mRNA expression levels of osteogenic markers were examined by qRT-PCR. (B) Alkaline phosphatase (ALP) activity assay (upper panel) and ALP staining (lower panel) were performed after 5 days of osteogenic differentiation. (C) Alizarin Red S staining (lower panel) was performed after 10 days of osteogenic differentiation. The stain was then dissolved out, and optical density was measured. Quantitative data are presented as the mean ± SD.

    MSCs, mesenchymal stem cells; qRT-PCR, quantitative reverse transcription-polymerase chain reaction; SD, standard deviation.

    *p < 0.05, compared to si Con.

    IJOB-49-3-69_F6.gif

    Loss of Sestrin 2 (Sesn2) expression promotes adipogenic differentiation of mouse bone marrow-derived MSCs (BMSCs). Young mouse BMSCs were transfected with nontargeting control siRNA or SESN2 siRNA and incubated in growth medium for 24 hours. (A) Cells were then incubated in adipogenic medium for 3 days, followed by qRT-PCR analyses and (B) Oil Red O staining (×40 magnification). Quantitative data are presented as the mean ± SD.

    MSCs, mesenchymal stem cells; qRT-PCR, quantitative reverse transcription-polymerase chain reaction; SD, standard deviation.

    *p < 0.05, compared to si Con.

    Table

    Reference

    1. Lee JH, Budanov AV, Karin M. Sestrins orchestrate cellular metabolism to attenuate aging. Cell Metab 2013;18:792-801.
    2. Lee JH, Budanov AV, Park EJ, Birse R, Kim TE, Perkins GA, Ocorr K, Ellisman MH, Bodmer R, Bier E, Karin M. Sestrin as a feedback inhibitor of TOR that prevents age-related pathologies. Science 2010;327:1223-8.
    3. Budanov AV, Lee JH, Karin M. Stressin’ Sestrins take an aging fight. EMBO Mol Med 2010;2:388-400.
    4. Chen Y, Huang T, Yu Z, Yu Q, Wang Y, Hu J, Shi J, Yang G. The functions and roles of sestrins in regulating human diseases. Cell Mol Biol Lett 2022;27:2.
    5. Xu D, Shimkus KL, Lacko HA, Kutzler L, Jefferson LS, Kimball SR. Evidence for a role for Sestrin1 in mediating leucine-induced activation of mTORC1 in skeletal muscle. Am J Physiol Endocrinol Metab 2019;316:E817-28.
    6. Fang H, Shi X, Wan J, Zhong X. Role of sestrins in metabolic and aging-related diseases. Biogerontology 2024;25:9-22.
    7. Guillén C, Benito M. mTORC1 overactivation as a key aging factor in the progression to type 2 diabetes mellitus. Front Endocrinol (Lausanne) 2018;9:621.
    8. Johnson SC, Rabinovitch PS, Kaeberlein M. mTOR is a key modulator of ageing and age-related disease. Nature 2013; 493:338-45.
    9. Musina RA, Bekchanova ES, Sukhikh GT. Comparison of mesenchymal stem cells obtained from different human tissues. Bull Exp Biol Med 2005;139:504-9.
    10. Bergman RJ, Gazit D, Kahn AJ, Gruber H, McDougall S, Hahn TJ. Age-related changes in osteogenic stem cells in mice. J Bone Miner Res 1996;11:568-77.
    11. Justesen J, Stenderup K, Ebbesen EN, Mosekilde L, Steiniche T, Kassem M. Adipocyte tissue volume in bone marrow is increased with aging and in patients with osteoporosis. Biogerontology 2001;2:165-71.
    12. Lin Z, He H, Wang M, Liang J. MicroRNA-130a controls bone marrow mesenchymal stem cell differentiation towards the osteoblastic and adipogenic fate. Cell Prolif 2019;52:e12688.
    13. Li CJ, Cheng P, Liang MK, Chen YS, Lu Q, Wang JY, Xia ZY, Zhou HD, Cao X, Xie H, Liao EY, Luo XH. MicroRNA-188 regulates age-related switch between osteoblast and adipocyte differentiation. J Clin Invest 2015;125:1509-22.
    14. Li X, Wang X, Zhang C, Wang J, Wang S, Hu L. Dysfunction of metabolic activity of bone marrow mesenchymal stem cells in aged mice. Cell Prolif 2022;55:e13191.
    15. Josephson AM, Bradaschia-Correa V, Lee S, Leclerc K, Patel KS, Muinos Lopez E, Litwa HP, Neibart SS, Kadiyala M, Wong MZ, Mizrahi MM, Yim NL, Ramme AJ, Egol KA, Leucht P. Age-related inflammation triggers skeletal stem/progenitor cell dysfunction. Proc Natl Acad Sci U S A 2019;116:6995- 7004.
    16. Ganguly P, El-Jawhari JJ, Giannoudis PV, Burska AN, Ponchel F, Jones EA. Age-related changes in bone marrow mesenchymal stromal cells: a potential impact on osteoporosis and osteoarthritis development. Cell Transplant 2017;26:1520-9.
    17. Wang YG, Qu XH, Yang Y, Han XG, Wang L, Qiao H, Fan QM, Tang TT, Dai KR. AMPK promotes osteogenesis and inhibits adipogenesis through AMPK-Gfi1-OPN axis. Cell Signal 2016; 28:1270-82.
    18. Ling NXY, Kaczmarek A, Hoque A, Davie E, Ngoei KRW, Morrison KR, Smiles WJ, Forte GM, Wang T, Lie S, Dite TA, Langendorf CG, Scott JW, Oakhill JS, Petersen J. mTORC1 directly inhibits AMPK to promote cell proliferation under nutrient stress. Nat Metab 2020;2:41-9.
    19. Qadir AS, Um S, Lee H, Baek K, Seo BM, Lee G, Kim GS, Woo KM, Ryoo HM, Baek JH. miR-124 negatively regulates osteogenic differentiation and in vivo bone formation of mesenchymal stem cells. J Cell Biochem 2015;116:730-42.
    20. Lee HL, Woo KM, Ryoo HM, Baek JH. Distal-less homeobox 5 inhibits adipogenic differentiation through the down-regulation of peroxisome proliferator-activated receptor γ expression. J Cell Physiol 2013;228:87-98.
    21. Khosla S, Farr JN, Monroe DG. Cellular senescence and the skeleton: pathophysiology and therapeutic implications. J Clin Invest 2022;132:e154888.
    22. Stenderup K, Justesen J, Clausen C, Kassem M. Aging is associated with decreased maximal life span and accelerated senescence of bone marrow stromal cells. Bone 2003;33: 919-26.
    23. Shen T, Alvarez-Garcia O, Li Y, Olmer M, Lotz MK. Suppression of Sestrins in aging and osteoarthritic cartilage: dysfunction of an important stress defense mechanism. Osteoarthritis Cartilage 2017;25:287-96.
    24. Rajan SP, Anwar M, Jain B, Khan MA, Dey S, Dey AB. Serum sestrins: potential predictive molecule in human sarcopenia. Aging Clin Exp Res 2021;33:1315-24.
    25. Rai N, Venugopalan G, Pradhan R, Ambastha A, Upadhyay AD, Dwivedi S, Dey AB, Dey S. Exploration of novel antioxidant protein Sestrin in frailty syndrome in elderly. Aging Dis 2018;9:220-7.
    26. Hwang CY, Han YH, Lee SM, Cho SM, Yu DY, Kwon KS. Sestrin2 attenuates cellular senescence by inhibiting NADPH oxidase 4 expression. Ann Geriatr Med Res 2020;24:297-304.
    27. Kong S, Li J, Pan X, Zhao C, Li Y. Allicin regulates Sestrin2 ubiquitination to affect macrophage autophagy and senescence, thus inhibiting the growth of hepatoma cells. Tissue Cell 2024;88:102398.
    28. Wang H, Xi J, Zhang Z, Li J, Guo L, Li N, Sun Y, Li X, Han X. Sestrin2 is increased in calcific aortic disease and inhibits osteoblastic differentiation in valvular interstitial cells via the nuclear factor E2-related factor 2 pathway. J Cardiovasc Pharmacol 2022;80:609-15.
    29. Atashi F, Modarressi A, Pepper MS. The role of reactive oxygen species in mesenchymal stem cell adipogenic and osteogenic differentiation: a review. Stem Cells Dev 2015;24:1150- 63.
    30. Martin SK, Fitter S, Dutta AK, Matthews MP, Walkley CR, Hall MN, Ruegg MA, Gronthos S, Zannettino AC. Brief report: the differential roles of mTORC1 and mTORC2 in mesenchymal stem cell differentiation. Stem Cells 2015;33:1359-65.
    31. Kim EK, Lim S, Park JM, Seo JK, Kim JH, Kim KT, Ryu SH, Suh PG. Human mesenchymal stem cell differentiation to the osteogenic or adipogenic lineage is regulated by AMPactivated protein kinase. J Cell Physiol 2012;227:1680-7.