Mesenchymal Stem Cell Senescence and Osteogenesis
Abstract
:1. Introduction
2. Cellular Senescence Process
2.1. Cell Cycle Arrest
2.2. Decreased Telomere Length and Response to DNA Injury
2.3. CDKN2A Locus Depression
2.4. Stress-Inducing Senescence and ROS
2.5. Aging Related Oncogene
2.6. Senescence-Associated Secretory Phenotype (SASP)
3. Mesenchymal Stem Cells (MSCs)
4. The Roles of Extracellular Vesicles
5. Characteristics of Senescent MSCs
6. Osteogenesis in Healthy and Senescent MSCs
7. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The Hallmarks of Aging. Cell 2013, 153, 1194–1217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Campisi, J.; D’Adda di Fagagna, F. Cellular senescence: When bad things happen to good cells. Nat. Rev. Mol. Cell Biol. 2007, 8, 729–740. [Google Scholar] [CrossRef] [PubMed]
- Collado, M.; Gil, J.; Efeyan, A.; Guerra, C.; Schuhmacher, A.J.; Barradas, M.; Benguría, A.; Zaballos, A.; Flores, J.M.; Barbacid, M.; et al. Tumour biology: Senescence in premalignant tumours. Nature 2005, 436, 642. [Google Scholar] [CrossRef]
- Kuilman, T.; Michaloglou, C.; Mooi, W.J.; Peeper, D.S. The essence of senescence. Genes Dev. 2010, 24, 2463–2479. [Google Scholar] [CrossRef] [Green Version]
- Muñoz-Espín, D.; Serrano, M. Cellular senescence: From physiology to pathology. Nat. Rev. Mol. Cell Biol. 2014, 15, 482–496. [Google Scholar] [CrossRef] [PubMed]
- Friedenstein, A.J.; Chailakhjan, R.K.; Lalykina, K. The development of fibroblast colonies in monolayer cultures of guinea-pig bone marrow and spleen cells. Cell Tissue Kinet. 1970, 3, 393–403. [Google Scholar] [CrossRef] [PubMed]
- Aboushady, I.M.; Salem, Z.A.; Sabry, D.; Mohamed, A. Comparative study of the osteogenic potential of mesenchymal stem cells derived from different sources. J. Clin. Exp. Dent. 2018, 10, e7–e13. [Google Scholar] [CrossRef] [PubMed]
- Macrin, D.; Joseph, J.P.; Pillai, A.A.; Devi, A. Eminent sources of adult mesenchymal stem cells and their therapeutic imminence. Stem Cell Rev. 2017, 13, 741–756. [Google Scholar] [CrossRef]
- Teng, S.W.; Lo, Y.S.; Liu, W.T.; Hsuan, Y.; Lin, W. Genome-wide comparison of mesenchymal stem cells derived from human placenta and umbilical cord. Taiwan. J. Obstet. Gynecol. 2017, 56, 664–671. [Google Scholar] [CrossRef]
- Horwitz, E.M.; Le Blanc, K.; Dominici, M.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.C.; Deans, R.J.; Krause, D.S.; Keating, A. Clarification of the nomenclature for MSC: The international society for cellular therapy position statement. Cytotherapy 2005, 7, 393–395. [Google Scholar] [CrossRef] [PubMed]
- Han, Z.C.; Du, W.J.; Han, Z.B.; Liang, L. New insights into the heterogeneity and functional diversity of human mesenchymal stem cells. Biomed. Mater. Eng. 2017, 28, S29–S45. [Google Scholar] [CrossRef]
- Liu, H.; Xia, X.; Li, B. Mesenchymal stem cell aging: Mechanisms and influences on skeletal and non-skeletal tissues. Exp. Biol. Med. 2015, 240, 1099–1106. [Google Scholar] [CrossRef] [Green Version]
- Coipeau, P.; Rosset, P.; Langonné, A.; Gaillard, J.; Delorme, B.; Rico, A.; Domenech, J.; Charbord, P.; Sensebé, L. Impaired differentiation potential of human trabecular bone mesenchymal stromal cells from elderly patients. Cytotherapy 2009, 11, 584–594. [Google Scholar] [CrossRef] [PubMed]
- 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, 919–926. [Google Scholar] [CrossRef] [PubMed]
- Moerman, E.J.; Teng, K.; Lipschitz, D.A.; Lecka-Czernik, B. Aging activates adipogenic and suppresses osteogenic programs in mesenchymal marrow stroma/stem cells: The role of PPAR-gamma2 transcription factor and TGFbeta/ BMP signaling pathways. Aging Cell 2004, 3, 379–389. [Google Scholar] [CrossRef] [Green Version]
- Chen, Q.; Shou, P.; Zheng, C.; Jiang, M.; Cao, G.; Yang, Q.; Cao, J.; Xie, N.; Velletri, T.; Zhang, X.; et al. Fate decision of mesenchymal stem cells: Adipocytes or osteoblasts? Cell Death Differ. 2016, 23, 1128–1139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lefterova, M.I.; Zhang, Y.; Steger, D.J.; Schupp, M.; Schug, J.; Cristancho, A.; Feng, D.; Zhuo, D.; Stoeckert, C.J.; Liu, X.S.; et al. PPAR gamma and C/EBP factors orchestrate adipocyte biology via adjacent binding on a genome-wide scale. Genes Dev. 2008, 22, 2941–2952. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, J.; Ko, J. A novel PPARγ2 modulator sLZIP controls the balance between adipogenesis and osteogenesis during mesenchymal stem cell differentiation. Cell Death Differ. 2014, 21, 1642–1655. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, H.N.; Chang, J.; Shao, L.; Han, L.I.; Iyer, S.; Manolagas, S.C.; O’Brien, C.A.; Jilka, R.L.; Zhou, D.; Almeida, M. DNA damage and senescence in osteoprogenitors expressing Osx1 may cause their decrease with age. Aging Cell 2017, 16, 693–703. [Google Scholar] [CrossRef]
- Imai, S.; Guarente, L. NAD+ and sirtuins in aging and disease. Trends Cell Biol. 2014, 24, 464–471. [Google Scholar] [CrossRef]
- McReynolds, M.R.; Chellappa, K.; Baur, J.A. Age-related NAD(+) decline. Exp. Gerontol. 2020, 134, 110888. [Google Scholar] [CrossRef]
- Yoshino, J.; Baur, J.A.; Imai, S.I. NAD(+) intermediates: The biology and therapeutic potential of NMN and NR. Cell Metab. 2018, 27, 513–528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rajman, L.; Chwalek, K.; Sinclair, D.A. Therapeutic potential of NAD-boosting molecules: The in vivo evidence. Cell Metab. 2018, 27, 529–547. [Google Scholar] [CrossRef] [Green Version]
- Ucer, S.; Iyer, S.; Kim, H.N.; Han, L.; Rutlen, C.; Allison, K.; Thostenson, J.D.; De Cabo, R.; Jilka, R.L.; O’Brien, C.; et al. The effects of aging and sex steroid deficiency on the murine skeleton are independent and mechanistically distinct. J. Bone Min. Res. 2017, 32, 560–574. [Google Scholar] [CrossRef] [PubMed]
- Almeida, M.; Han, L.; Martin-Millan, M.; O’Brien, C.A.; Manolagas, S.C. Oxidative stress antagonizes Wnt signaling in osteoblast precursors by diverting betacatenin from T cell factor- to forkhead box O-mediated transcription. J. Biol. Chem. 2007, 282, 27298–27305. [Google Scholar] [CrossRef] [Green Version]
- Hoogeboom, D.; Essers, M.A.; Polderman, P.E.; Voets, E.; Smits, L.M.; Boudewijn, M.T. Interaction of FOXO with beta-catenin inhibits beta-catenin/ T cell factor activity. J. Biol. Chem. 2008, 283, 9224–9230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Essers, M.A.; De Vries-Smits, L.M.; Barker, N.; Polderman, P.E.; Burgering, B.M.; Korswagen, H.C. Functional interaction between beta-catenin and FOXO in oxidative stress signaling. Science 2005, 308, 1181–1184. [Google Scholar] [CrossRef] [PubMed]
- Bruder, S.P.; Jaiswal, N.; Haynesworth, S.E. Growth kinetics, self-renewal, and the osteogenic potential of purified human mesenchymal stem cells during extensive subcultivation and following cryopreservation. J. Cell. Biochem. 1997, 64, 278–294. [Google Scholar] [CrossRef]
- Watanabe, S. Impact of senescence-associated secretory phenotype and its potential as a therapeutic target for senescence-associated diseases. Cancer Sci. 2017, 108, 563–569. [Google Scholar] [CrossRef] [Green Version]
- Noren, H.N. Techniques to induce and quantify cellular senescence. J. Vis. Exp. 2017, 123, e55533. [Google Scholar] [CrossRef]
- Jones, E. Identification of senescent cells in multipotent mesenchymal stromal cell cultures: Current methods and future directions. Cytotherapy 2019, 21, 803–819. [Google Scholar] [CrossRef]
- Chicas, A.; Wang, X.; Zhang, C.; McCurrach, M.; Zhao, Z.; Mert, O.; Dickins, R.A.; Narita, M.; Zhang, M.; Lowe, S.W. Dissecting the unique role of the retinoblastoma tumor suppressor during cellular senescence. Cancer Cell 2010, 17, 376–387. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Beauséjour, C.M.; Krtolica, A.; Galimi, F.; Narita, M.; Lowe, S.W.; Yaswen, P.; Campisi, J. Reversal of human cellular senescence: Roles of the p53 and p16 pathways. EMBO J. 2003, 22, 4212–4222. [Google Scholar] [CrossRef]
- Salama, R.; Sadaie, M.; Hoare, M.; Narita, M. Cellular senescence and its effector programs. Genes Dev. 2014, 28, 99–114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rodier, F.; Campisi, J. Four faces of cellular senescence. J. Cell Biol. 2011, 192, 547–556. [Google Scholar] [CrossRef] [PubMed]
- Takahashi, A.; Ohtani, N.; Yamakoshi, K.; Iida, S.I.; Tahara, H.; Nakayama, K.; Nakayama, K.I.; Ide, T.; Saya, H.; Hara, E. Mitogenic signalling and the p16INK4a-Rb pathway cooperate to enforce irreversible cellular senescence. Nat. Cell Biol. 2006, 8, 1291–1297. [Google Scholar] [CrossRef]
- Parrinello, S.; Samper, E.; Krtolica, A.; Goldstein, J.; Melov, S.; Campisi, J. Oxygen sensitivity severely limits the replicative lifespan of murine fibroblasts. Nat. Cell Biol. 2003, 5, 741–747. [Google Scholar] [CrossRef] [PubMed]
- Passos, J.F.; Saretzki, G.; Ahmed, S.; Nelson, G.; Richter, T.; Peters, H.; Wappler, I.; Birket, M.J.; Harold, G.; Schaeuble, K.; et al. Mitochondrial dysfunction accounts for the stochastic heterogeneity in telomere-dependent senescence. PLoS Biol. 2007, 5, e110. [Google Scholar] [CrossRef] [PubMed]
- Fumagalli, M.; Rossiello, F.; Clerici, M.; Barozzi, S.; Cittaro, D.; Kaplunov, J.M.; Bucci, G.; Dobreva, M.; Matti, V.; Beausejour, C.M.; et al. Telomeric DNA damage is irreparable and causes persistent DNA-damage-response activation. Nat. Cell Biol. 2012, 14, 355–365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, W.Y.; Sharpless, N.E. The regulation of INK4/ ARF in cancer and aging. Cell 2006, 127, 265–275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gil, J.; Peters, G. Regulation of the INK4b-ARF-INK4a tumour suppressor locus: All for one or one for all. Nat. Rev. Mol. Cell Biol. 2006, 7, 667–677. [Google Scholar] [CrossRef]
- Krishnamurthy, J.; Torrice, C.; Ramsey, M.R.; Kovalev, G.I.; Al-Regaiey, K.; Su, L.; Sharpless, N.E. Ink4a/Arf expression is a biomarker of aging. J. Clin. Investig. 2004, 114, 1299–1307. [Google Scholar] [CrossRef] [PubMed]
- Jacobs, J.J.; Kieboom, K.; Marino, S.; DePinho, R.A.; Van Lohuizen, M. The oncogene and Polycomb- group gene bmi-1 regulates cell proliferation and senescence through the ink4a locus. Nature 1999, 397, 164–168. [Google Scholar] [CrossRef]
- Bracken, A.P.; Kleine-Kohlbrecher, D.; Dietrich, N.; Pasini, D.; Gargiulo, G.; Beekman, C.; Theilgaard-Mönch, K.; Minucci, S.; Porse, B.T.; Marine, J.C.; et al. The Polycomb group proteins bind throughout the INK4A-ARF locus and are disassociated in senescent cells. Genes Dev. 2007, 21, 525–530. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Velimezi, G.; Liontos, M.; Vougas, K.; Roumeliotis, T.; Bartkova, J.; Sideridou, M.; Dereli-Oz, A.; Kocylowski, M.; Pateras, I.S.; Evangelou, K.; et al. Functional interplay between the DNA-damage-response kinase ATM and ARF tumour suppressor protein in human cancer. Nat. Cell Biol. 2013, 15, 967–977. [Google Scholar] [CrossRef]
- Evangelou, K.; Bartkova, J.; Kotsinas, A.; Pateras, I.S.; Liontos, M.; Velimezi, G.; Kosar, M.; Liloglou, T.; Trougakos, I.P.; Dyrskjot, L.; et al. The DNA damage checkpoint precedes activation of ARF in response to escalating oncogenic stress during tumorigenesis. Cell Death Differ. 2013, 20, 1485–1497. [Google Scholar] [CrossRef] [Green Version]
- Chen, Q.; Fischer, A.; Reagan, J.D.; Yan, L.J.; Ames, B.N. Oxidative DNA damage and senescence of human diploid fibroblast cells. Proc. Natl. Acad. Sci. USA 1995, 92, 4337–4341. [Google Scholar] [CrossRef] [Green Version]
- Lee, A.C.; Fenster, B.E.; Ito, H.; Takeda, K.; Bae, N.S.; Hirai, T.; Yu, Z.X.; Ferrans, V.J.; Howard, B.H.; Finkel, T. Ras proteins induce senescence by altering the intracellular levels of reactive oxygen species. J. Biol. Chem. 1999, 274, 7936–7940. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Macip, S.; Igarashi, M.; Fang, L.; Chen, A.; Pan, Z.Q.; Lee, S.W.; Aaronson, S.A. Inhibition of p21-mediated ROS accumulation can rescue p21-induced senescence. EMBO J. 2002, 21, 2180–2188. [Google Scholar] [CrossRef]
- Bartkova, J.; Rezaei, N.; Liontos, M.; Karakaidos, P.; Kletsas, D.; Issaeva, N.; Vassiliou, L.V.F.; Kolettas, E.; Niforou, K.; Zoumpourlis, V.C.; et al. Oncogene-induced senescence is part of the tumorigenesis barrier imposed by DNA damage checkpoints. Nature 2006, 444, 633–637. [Google Scholar] [CrossRef] [PubMed]
- Di Micco, R.; Fumagalli, M.; Cicalese, A.; Piccinin, S.; Gasparini, P.; Luise, C.; Schurra, C.; Nuciforo, P.G.; Bensimon, A.; Maestro, R.; et al. Oncogene-induced senescence is a DNA damage response triggered by DNA hyper-replication. Nature 2006, 444, 638–642. [Google Scholar] [CrossRef]
- Efeyan, A.; Serrano, M. p53: Guardian of the genome and policeman of the oncogenes. Cell Cycle 2007, 6, 1006–1010. [Google Scholar] [CrossRef] [PubMed]
- Halazonetis, T.D.; Gorgoulis, V.G.; Bartek, J. An oncogene-induced DNA damage model for cancer development. Science 2008, 319, 1352–1355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Evan, G.I.; D’Adda di Fagagna, F. Cellular senescence: Hot or what? Curr. Opin. Genet. Dev. 2009, 19, 25–31. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Wu, Q.; Wang, Y.; Li, L.; Bu, H.; Bao, J. Senescence of mesenchymal stem cells (Review). Int. J. Mol. Med. 2017, 39, 775–782. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Campisi, J. Aging, cellular senescence, and cancer. Annu. Rev. Physiol. 2013, 75, 685–705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Coppe, J.P.; Desprez, P.Y.; Krtolica, A.; Campisi, J. The senescence-associated secretory phenotype: The dark side of tumor suppression. Annu. Rev. Pathol. 2010, 5, 99–118. [Google Scholar] [CrossRef] [Green Version]
- Acosta, J.C.; O’Loghlen, A.; Banito, A.; Guijarro, M.V.; Augert, A.; Raguz, S.; Fumagalli, M.; Da Costa, M.; Brown, C.; Popov, N.; et al. Chemokine signaling via the CXCR2 receptor reinforces senescence. Cell 2008, 133, 1006–1018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Coppé, J.P.; Patil, C.K.; Rodier, F.; Sun, Y.U.; Muñoz, D.P.; Goldstein, J.; Nelson, P.S.; Desprez, P.Y.; Campisi, J. Senescence-associated secretory phenotypes reveal cell-non autonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biol. 2008, 6, 2853–2868. [Google Scholar] [CrossRef]
- Kuilman, T.; Michaloglou, C.; Vredeveld, L.C.; Douma, S.; Van Doorn, R.; Desmet, C.J.; Aarden, L.A.; Mooi, W.J.; Peeper, D.S. Oncogene-induced senescence relayed by an interleukin-dependent inflammatory network. Cell 2008, 133, 1019–1031. [Google Scholar] [CrossRef] [Green Version]
- Xue, W.; Zender, L.; Miething, C.; Dickins, R.A.; Hernando, E.; Krizhanovsky, V.; Cordon-Cardo, C.; Lowe, S.W. Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas. Nature 2007, 445, 656–660. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hoenicke, L.; Zender, L. Immune surveillance of senescent cells—Biological significance in cancer- and non-cancer pathologies. Carcinogenesis 2012, 33, 1123–1126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Acosta, J.C.; Banito, A.; Wuestefeld, T.; Georgilis, A.; Janich, P.; Morton, J.P.; Athineos, D.; Kang, T.W.; Lasitschka, F.; Andrulis, M.; et al. A complex secretory program orchestrated by the inflammasome controls paracrine senescence. Nat. Cell Biol. 2013, 15, 978–990. [Google Scholar] [CrossRef] [PubMed]
- Li, C.Y.; Wu, X.Y.; Tong, J.B.; Yang, X.X.; Zhao, J.L.; Zheng, Q.F.; Zhao, G.B.; Ma, Z.J. Comparative analysis of human mesenchymal stem cells from bone marrow and adipose tissue under xeno-free conditions for cell therapy. Stem Cell Res. Ther. 2015, 6, 55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Neri, S. Genetic stability of mesenchymal stromal cells for regenerative medicine applications: A fundamental biosafety aspect. Int. J. Mol. Sci. 2019, 20, 2406. [Google Scholar] [CrossRef] [Green Version]
- Lv, F.J.; Tuan, R.S.; Cheung, K.M.; Leung, V.Y. Concise review: The surface markers and identity of human mesenchymal stem cells. Stem Cells 2014, 32, 1408–1419. [Google Scholar] [CrossRef]
- Dominici, M.L.B.K.; Le Blanc, K.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.C.; Krause, D.S.; Deans, R.J.; Keating, A.; Prockop, D.J.; Horwitz, E.M. Minimal criteria for defining multipotent mesenchymal stromal cells. The international society for cellular therapy position statement. Cytotherapy 2006, 8, 315–317. [Google Scholar] [CrossRef] [PubMed]
- Graf, T. Covering the stem cell explosion at the 2017 ISSCR conference in Boston. Stem Cell Rep. 2017, 9, 1017–1023. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bianco, P.; Cao, X.; Frenette, P.S.; Mao, J.J.; Robey, P.G.; Simmons, P.J.; Wang, C.Y. The meaning, the sense and the significance: Translating the science of mesenchymal stem cells into medicine. Nat. Med. 2013, 19, 35–42. [Google Scholar] [CrossRef] [Green Version]
- Mastrolia, I.; Foppiani, E.M.; Murgia, A.; Candini, O.; Samarelli, A.V.; Grisendi, G.; Veronesi, E.; Horwitz, E.M.; Dominici, M. Challenges in clinical development of mesenchymal stromal/stem cells: Concise review. Stem Cells Transl. Med. 2019, 8, 1135–1148. [Google Scholar] [CrossRef] [Green Version]
- Wagner, D.R.; Karnik, S.; Gunderson, Z.J.; Nielsen, J.J.; Fennimore, A.; Promer, H.J.; Lowery, J.W.; Loghmani, M.T.; Low, P.S.; McKinley, T.O.; et al. Dysfunctional stem and progenitor cells impair fracture healing with age. World J. Stem Cells 2019, 11, 281–296. [Google Scholar] [CrossRef] [PubMed]
- Moretta, L.; Uccelli, A.; Pistoia, V. Mesenchymal stromal cells and immunity: Introductory overview. Immunol. Lett. 2015, 168, 127–128. [Google Scholar] [CrossRef] [PubMed]
- English, K. Mechanisms of mesenchymal stromal cell immunomodulation. Immunol. Cell Biol. 2013, 91, 19–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhao, K.; Lou, R.; Huang, F.; Peng, Y.; Jiang, Z.; Huang, K.; Wu, X.; Zhang, Y.; Fan, Z.; Zhou, H.; et al. Immunomodulation effects of mesenchymal stromal cells on acute graft-versus-host disease after hematopoietic stem cell transplantation. Biol. Blood Marrow Transplant. 2015, 21, 97–104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Uccelli, A.; Moretta, L.; Pistoia, V. Mesenchymal stem cells in health and disease. Nat. Rev. Immunol. 2008, 8, 726–736. [Google Scholar] [CrossRef] [PubMed]
- Mun, C.H.; Kang, M.I.; Shin, Y.D.; Kim, Y.; Park, Y.B. The expression of immunomodulation-related cytokines and genes of adipose- and bone marrow-derived human mesenchymal stromal cells from early to late passages. Tissue Eng. Regen. Med. 2018, 15, 771–779. [Google Scholar] [CrossRef] [PubMed]
- Harrell, C.R.; Fellabaum, C.; Jovicic, N.; Djonov, V.; Arsenijevic, N.; Volarevic, V. Molecular mechanisms responsible for therapeutic potential of mesenchymal stem cell-derived secretome. Cells 2019, 8, 467. [Google Scholar] [CrossRef] [Green Version]
- Van Niel, G.; D’Angelo, G.; Raposo, G. Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell Biol. 2018, 19, 213–228. [Google Scholar] [CrossRef] [PubMed]
- Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the International society for extracellular vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 2018, 7, 1535750. [Google Scholar] [CrossRef] [Green Version]
- Shimoda, M.; Khokha, R. Metalloproteinases in extracellular vesicles. Biochim. Biophys. Acta Mol. Cell Res. 2017, 1864, 1989–2000. [Google Scholar] [CrossRef]
- Zhang, Y.; Jin, X.; Liang, J.; Guo, Y.; Sun, G.; Zeng, X.; Yin, H. Extracellular vesicles derived from ODNstimulated macrophages transfer and activate Cdc42 in recipient cells and thereby increase cellular permissiveness to EV uptake. Sci. Adv. 2019, 5, eaav1564. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhou, S.; Greenberger, J.S.; Epperly, M.W.; Goff, J.P.; Adler, C.; Leboff, M.S.; Glowacki, J. Age-related intrinsic changes in human bone-marrow-derived mesenchymal stem cells and their differentiation to osteoblasts. Aging Cell 2008, 7, 335–343. [Google Scholar] [CrossRef] [Green Version]
- Singh, M.; Piekorz, R.P. Senescence-associated lysosomal α-L-fucosidase (SA-α-Fuc): A sensitive and more robust biomarker for cellular senescence beyond SA-β-Gal. Cell Cycle 2013, 12, 1996. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yu, Y.; Park, Y.S.; Kim, H.S.; Kim, H.Y.; Jin, Y.M.; Jung, S.C.; Ryu, K.H.; Jo, I. Characterization of long-term in vitro culture-related alterations of human tonsil-derived mesenchymal stem cells: Role for CCN1 in replicative senescence-associated increase in osteogenic differentiation. J. Anat. 2014, 225, 510–518. [Google Scholar] [CrossRef] [PubMed]
- Simmons, P.J.; Torok-Storb, B. Identification of stromal cell precursors in human bone marrow by a novel monoclonal antibody, STRO-1. Blood 1991, 78, 55–62. [Google Scholar] [CrossRef] [Green Version]
- Jung, E.M.; Kwon, O.; Kwon, K.S.; Cho, Y.S.; Rhee, S.K.; Min, J.K.; Oh, D.B. Evidences for correlation between the reduced VCAM-1 expression and hyaluronan synthesis during cellular senescence of human mesenchymal stem cells. Biochem. Biophys Res. Commun. 2011, 404, 463–469. [Google Scholar] [CrossRef] [PubMed]
- Laschober, G.T.; Brunauer, R.; Jamnig, A.; Fehrer, C.; Greiderer, B.; Lepperdinger, G. Leptin receptor/CD295 is upregulated on primary human mesenchymal stem cells of advancing biological age and distinctly marks the subpopulation of dying cells. Exp. Gerontol. 2009, 44, 57–62. [Google Scholar] [CrossRef]
- Astudillo, P.; Ríos, S.; Pastenes, L.; Pino, A.M.; Rodríguez, J.P. Increased adipogenesis of osteoporotic humanmesenchymal stem cells (MSCs) characterizes by impaired leptin action. J. Cell Biochem. 2008, 103, 1054–1065. [Google Scholar] [CrossRef]
- Abuna, R.P.; Stringhetta-Garcia, C.T.; Fiori, L.P.; Dornelles, R.C.M.; Rosa, A.L.; Beloti, M.M. Aging impairs osteoblast differentiation of mesenchymal stem cells grown on titanium by favoring adipogenesis. J. Appl. Oral Sci. 2016, 24, 376–382. [Google Scholar] [CrossRef]
- Kosar, M. Senescence-associated heterochromatin foci are dispensable for cellular senescence, occur in a cell type- and insult-dependent manner and follow expression of p16(ink4a). Cell Cycle 2011, 10, 457–468. [Google Scholar] [CrossRef] [Green Version]
- Narita, M. Rb-Mediated heterochromatin formation and silencing of E2F target genes during cellular senescence. Cell 2003, 113, 703–716. [Google Scholar] [CrossRef] [Green Version]
- Koch, C.M. Pluripotent stem cells escape from senescence associated DNA methylation changes. Genome Res. 2013, 23, 248–259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wagner, W. The Link between epigenetic clocks for aging and senescence. Front. Genet. 2019, 10, 303. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wagner, W. Senescence-associated DNA methylation is stochastically acquired in subpopulations of mesenchymal stem cells. Aging Cell 2017, 16, 183–191. [Google Scholar] [CrossRef] [Green Version]
- Easwaran, H. DNA methylation in senescence, aging and cancer. Oncoscience 2019, 6, 291–293. [Google Scholar]
- Gronthos, S.; Cakouros, D. Epigenetic regulation of bone marrow stem cell aging: Revealing epigenetic signatures associated with hematopoietic and mesenchymal stem cell aging. Aging Dis. 2019, 10, 174–189. [Google Scholar] [CrossRef] [Green Version]
- Childs, B.G. Senescent cells: A therapeutic target for cardiovascular disease. J. Clin. Investig. 2018, 128, 1217–1228. [Google Scholar] [CrossRef]
- Debacq-Chainiaux, F. Protocols to detect senescence associated beta-galactosidase (SA-betagal) activity, a biomarker of senescent cells in culture and in vivo. Nat. Protoc. 2009, 4, 1798–1806. [Google Scholar] [CrossRef]
- Sikora, E. Is senescence-associated b-galactosidase a marker of neuronal senescence? Oncotarget 2016, 7, 81099–81109. [Google Scholar]
- Akyurekli, C. A systematic review of preclinical studies on the therapeutic potential of mesenchymal stromal cell-derived microvesicles. Stem Cell Rev. Rep. 2015, 11, 150–160. [Google Scholar] [CrossRef]
- Xie, H. Development of an angiogenesis-promoting microvesicle-alginatepolycaprolactone composite graft for bone tissue engineering applications. PeerJ 2016, 4, e2040. [Google Scholar] [CrossRef]
- Patel, P.L. Derepression of hTERT gene expression promotes escape from oncogene-induced cellular senescence. Proc. Natl. Acad. Sci. USA 2016, 113, E5024–E5033. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fujii, M.; Kawai, Y.; Endoh, M.; Hossain, M.N.; Nakabayashi, K.; Ayusawa, D. Expression of RAB27B is up-regulated in senescent human cells. Mech. Ageing Dev. 2006, 127, 639–642. [Google Scholar] [CrossRef]
- Takahashi, A.; Okada, R.; Nagao, K.; Kawamata, Y.; Hanyu, A.; Yoshimoto, S.; Takasugi, M.; Watanabe, S.; Kanemaki, M.T.; Obuse, C.; et al. Exosomes maintain cellular homeostasis by excreting harmful DNA from cells. Nat. Commun. 2017, 8, 15287. [Google Scholar] [CrossRef] [Green Version]
- Prattichizzo, F.; Micolucci, L.; Cricca, M.; De Carolis, S.; Mensà, E.; Ceriello, A.; Procopio, A.D.; Bonafè, M.; Olivieri, F. Exosome-based immunomodulation during aging: A nanoperspective on inflamm-aging. Mech. Ageing Dev. 2017, 168, 44–53. [Google Scholar] [CrossRef]
- Wang, Y.; Fu, B.; Sun, X.; Li, D.; Huang, Q.; Zhao, W.; Chen, X. Differentially expressed microRNAs in bone marrow mesenchymal stem cell-derived microvesicles in young and older rats and their effect on tumor growth factor-beta1-mediated epithelial-mesenchymal transition in HK2 cells. Stem Cell Res. Ther. 2015, 6, 185. [Google Scholar] [CrossRef] [Green Version]
- Yu, K.R.; Kang, K.S. Aging-related genes in mesenchymal stem cells: A mini-review. Gerontology 2013, 59, 557–563. [Google Scholar] [CrossRef]
- Karsenty, G. Transcriptional control of skeletogenesis. Annu Rev. Genom. Hum. Genet. 2008, 9, 183–196. [Google Scholar] [CrossRef] [Green Version]
- Alt, E.U.; Senst, C.; Murthy, S.N.; Slakey, D.P.; Dupin, C.L.; Chaffin, A.E.; Kadowitz, P.J.; Izadpanah, R. Aging alters tissue resident mesenchymal stem cell properties. Stem Cell Res. 2012, 8, 215–225. [Google Scholar] [CrossRef] [Green Version]
- Edwards, J.R.; Mundy, G.R. Advances in osteoclast biology: Old findings and new insights from mouse models. Nat. Rev. Rheumatol. 2011, 7, 235–243. [Google Scholar] [CrossRef]
- Crane, J.L.; Cao, X. Function of matrix IGF-1 in coupling bone resorption and formation. J. Mol. Med. 2014, 92, 107–115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rachner, T.D.; Khosla, S.; Hofbauer, L.C. Osteoporosis: Now and the future. Lancet 2011, 377, 1276–1287. [Google Scholar] [CrossRef] [Green Version]
- Pittenger, M.F.; Mackay, A.F.; Beck, S.C.; Jaiswal, S.K.; Douglas, R.; Mosca, J.D. Multilineage potential of adult human mesenchymal stem cells. Science 1999, 284, 143–147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stolzing, A.; Coleman, N.; Scutt, A. Glucose-induced replicative senescence in mesenchymal stem cells. Rejuvenation Res. 2006, 9, 31–35. [Google Scholar] [CrossRef]
- Campisi, J.; Rodier, F.; Bhaumik, D. Two faces of p53: Aging and tumor suppression. Nucleic Acid Res. 2007, 35, 7475–7484. [Google Scholar] [CrossRef]
- Zindy, F.; Quelle, D.E.; Roussel, M.F.; Sherr, C.J. Expression of the p16INK4a tumor suppressor versus other INK4 family members during mouse development and aging. Oncogene 1997, 15, 203–211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sedelnikova, O.A.; Horikawa, I.; Zimonjic, D.B.; Popescu, N.C.; Bonner, W.M.; Barrett, J.C. Senescing human cells and ageing mice accumulate DNA lesions with unrepairable double-strand breaks. Nat. Cell Biol. 2004, 6, 168–170. [Google Scholar] [CrossRef]
- Thomas, D.M.; Carty, S.A.; Piscopo, D.M.; Lee, J.S.; Wang, W.F.; Forrester, W.C.; Hinds, P.W. The retinoblastoma protein acts as a transcriptional coactivator required for osteogenic differentiation. Mol. Cell 2001, 8, 303–316. [Google Scholar] [CrossRef]
- Calo, E.; Quintero-Estades, J.A.; Danielian, P.S.; Nedelcu, S.; Berman, S.D.; Lees, J.A. Rb regulates fate choice and lineage commitment in vivo. Nature 2010, 466, 1110–1114. [Google Scholar] [CrossRef] [Green Version]
- Wang, X.; Kua, H.Y.; Hu, Y.; Guo, K.; Zeng, Q.; Wu, Q.; Ng, H.H.; Karsenty, G.; de Crombrugghe, B.; Yeh, J.; et al. p53 functions as a negative regulator of osteoblastogenesis, osteoblast-dependent osteoclastogenesis, and bone remodeling. J. Cell Biol. 2006, 172, 115–125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Molchadsky, A.; Shats, I.; Goldfinger, N.; Pevsner-Fischer, M.; Olson, M.; Rinon, A.; Tzahor, E.; Lozano, G.; Zipori, D.; Sarig, R.; et al. p53 plays a role in mesenchymal differentiation programs, in a cell fate dependent manner. PLoS ONE 2008, 3, e3707. [Google Scholar] [CrossRef] [Green Version]
- Atashi, F.; Modaresi, A.; Pepper, M.S. The role of reactive oxygen species in mesenchymal stem cell adipogenic and osteogenic differentiation: A review. Stem Cells Dev. 2015, 24, 1150–1163. [Google Scholar] [CrossRef] [Green Version]
- Xu, J.; Qian, J.; Xie, X.; Lin, L.; Zou, Y.; Fu, M.; Huang, Z.; Zhang, G.; Su, Y.; Ge, J. High density lipoprotein protect mesenchymal stem cells from oxidative stress induced apoptosis via activation of the P13K/Akt pathway and suppression of reactive oxygen species. Int. J. Mol. Sci. 2012, 13, 7104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jiang, Y.; Mishima, H.; Sakai, S.; Liu, Y.K.; Ohyabu, Y.; Uemura, T. Gene expression analysis of major liniage defining factors in human bone marrow cells: Effect of aging, gender, and age related disorders. J. Orthop. Res. 2008, 26, 910–917. [Google Scholar] [CrossRef]
- Komori, T. Signaling networks in RUNX2 dependent bone development. J. Cell Biochem. 2011, 112, 750–755. [Google Scholar] [CrossRef] [PubMed]
- Tu, B.; Peny, Z.X.; Fan, Q.M.; Du, L.; Yan, W.; Tang, T. Osteosarcoma cells promote the production of pro-tumor cytokins in MSC by inhibiting their osteogenic differentiation through the TGF-ß/smad2/3 pathway. Exp. Cell Res. 2014, 320, 164–173. [Google Scholar] [CrossRef] [PubMed]
- Xu, C.; Wang, J.; Zhu, T.; Shen, Y.; Tang, X.; Fang, L.; Xu, Y. Cross-talking between PPAR and WNT signaling and its regulation in mesenchymal stem cell differentiation. Curr. Stem Cell Res. Ther. 2016, 11, 247–254. [Google Scholar] [CrossRef] [PubMed]
- Gorgoulis, V.; Adams, P.D.; Alimonti, A.; Bennett, D.C.; Bischof, O.; Bishop, C.; Campisi, J.; Collado, M.; Evangelou, K.; Ferbeyre, G.; et al. Cellular senescence: Defining a path forward. Cell 2019, 179, 813–827. [Google Scholar] [CrossRef]
- Giatti, S.; Mastrangelo, R.; D’Antonio, M.; Pesaresi, M.; Romano, S.; Diviccaro, S.; Caruso, D.; Mitro, N.; Melcangi, R.C. Neuroactive steroids and diabetic complications in the nervous system. Front. Neuroendocrinol. 2018, 48, 58–69. [Google Scholar] [CrossRef] [PubMed]
- Haigis, M.C.; Sinclair, D.A. Mammalian sirtuins: Biological insights and disease relevance. Annu. Rev. Pathol. 2010, 5, 253–295. [Google Scholar] [CrossRef] [Green Version]
- Baur, J.A.; Ungvari, Z.; Minor, R.K.; Le Couteur, D.G.; De Cabo, R. Are sirtuins viable targets for improving healthspan and lifespan? Nat. Rev. Drug Discov. 2012, 11, 443–461. [Google Scholar] [CrossRef] [Green Version]
- Michan, S.; Sinclair, D. Sirtuins in mammals: Insights into their biological function. Biochem. J. 2007, 404, 1–13. [Google Scholar] [CrossRef] [Green Version]
- Manolagas, S.C. Birth and death of bone cells: Basic regulatory mechanisms and implications for the pathogenesis and treatment of osteoporosis. Endocr. Rev. 2000, 21, 115–137. [Google Scholar] [PubMed] [Green Version]
- Long, F. Building strong bones: Molecular regulation of the osteoblast lineage. Nat. Rev. Mol. Cell Biol. 2012, 13, 27–38. [Google Scholar] [CrossRef] [PubMed]
- Iyer, S.; Ambrogini, E.; Bartell, S.M.; Han, L.; Roberson, P.K.; de Cabo, R.; Jilka, R.L.; Weinstein, R.S.; O’Brien, C.A.; Manolagas, S.C.; et al. FOXOs attenuate bone formation by suppressing Wnt signaling. J. Clin. Investig. 2013, 123, 3409–3419. [Google Scholar] [CrossRef] [PubMed]
- Chini, E.N.; Chini, C.C.S.; Espindola Netto, J.M.; De Oliveira, G.C.; Van Schooten, W. The pharmacology of CD38/NADase: An emerging target in cancer and diseases of aging. Trends Pharmacol. Sci. 2018, 39, 424–436. [Google Scholar] [CrossRef]
- Camacho-Pereira, J.; Tarragó, M.G.; Chini, C.C.; Nin, V.; Escande, C.; Warner, G.M.; Puranik, A.S.; Schoon, R.A.; Reid, J.M.; Galina, A.; et al. CD38 dictates age-related NAD decline and mitochondrial dysfunction through an SIRT3-dependent mechanism. Cell Metab. 2016, 23, 1127–1139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tarragó, M.G.; Chini, C.C.; Kanamori, K.S.; Warner, G.M.; Caride, A.; de Oliveira, G.C.; Rud, M.; Samani, A.; Hein, K.Z.; Huang, R.; et al. A potent and specific CD38 inhibitor ameliorates age-related metabolic dysfunction by reversing tissue NAD(+) decline. Cell Metab. 2018, 27, 1081–1095. [Google Scholar] [CrossRef] [Green Version]
- Aksoy, P.; White, T.A.; Thompson, M.; Chini, E.N. Regulation of intracellular levels of NAD: A novel role for CD38. Biochem. Biophys. Res. Commun. 2006, 345, 1386–1392. [Google Scholar] [CrossRef] [PubMed]
- Young, G.S.; Choleris, E.; Lund, F.E.; Kirkland, J.B. Decreased cADPR and increased NAD+ in the Cd38−/− mouse. Biochem. Biophys. Res. Commun. 2006, 346, 188–192. [Google Scholar] [CrossRef] [PubMed]
- Iyer, S.; Han, L.; Bartell, S.M.; Kim, H.N.; Gubrij, I.; De Cabo, R.; O’Brien, C.A.; Manolagas, S.C.; Almeida, M. Sirtuin1 (Sirt1) promotes cortical bone formation by preventing beta (β)-catenin sequestration by FoxO transcription factors in osteoblast progenitors. J. Biol. Chem. 2014, 289, 24069–24078. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Simic, P.; Zainabadi, K.; Bell, E.; Sykes, D.B.; Saez, B.; Lotinun, S.; Baron, R.; Scadden, D.; Schipani, E.; Guarente, L. SIRT1 regulates differentiation of mesenchymal stem cells by deacetylating beta-catenin. EMBO Mol. Med. 2013, 5, 430–440. [Google Scholar] [CrossRef] [PubMed]
- Mercken, E.M.; Mitchell, S.J.; Martin-Montalvo, A.; Minor, R.K.; Almeida, M.; Gomes, A.P.; Scheibye-Knudsen, M.; Palacios, H.H.; Licata, J.J.; Zhang, Y.; et al. SRT2104 extends survival of male mice on a standard diet and preserves bone and muscle mass. Aging Cell 2014, 13, 787–796. [Google Scholar] [CrossRef] [PubMed]
- Zainabadi, K.; Liu, C.J.; Caldwell, A.L.M.; Guarente, L. SIRT1 is a positive regulator of in vivo bone mass and a therapeutic target for osteoporosis. PLoS ONE 2017, 12, e0185236. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Tjempakasari, A.; Suroto, H.; Santoso, D. Mesenchymal Stem Cell Senescence and Osteogenesis. Medicina 2022, 58, 61. https://doi.org/10.3390/medicina58010061
Tjempakasari A, Suroto H, Santoso D. Mesenchymal Stem Cell Senescence and Osteogenesis. Medicina. 2022; 58(1):61. https://doi.org/10.3390/medicina58010061
Chicago/Turabian StyleTjempakasari, Artaria, Heri Suroto, and Djoko Santoso. 2022. "Mesenchymal Stem Cell Senescence and Osteogenesis" Medicina 58, no. 1: 61. https://doi.org/10.3390/medicina58010061