Cellular senescence in skeletal muscle: age, sarcopenia, therapy
- Authors: Plekhova N.G.1, Novikova P.A.1, Voronova A.N.1, Korolev D.V.1, Shumatov V.B.1
-
Affiliations:
- Pacific State Medical University
- Issue: Vol 111, No 7 (2025)
- Pages: 1018-1041
- Section: REVIEW
- URL: https://kld-journal.fedlab.ru/0869-8139/article/view/691427
- DOI: https://doi.org/10.7868/S2658655X25070028
- EDN: https://elibrary.ru/mvjtdv
- ID: 691427
Cite item
Full Text
Abstract
Keywords
About the authors
N. G. Plekhova
Pacific State Medical University
Email: pl_nat@hotmail.com
Vladivostok, Russia
P. A. Novikova
Pacific State Medical UniversityVladivostok, Russia
A. N. Voronova
Pacific State Medical UniversityVladivostok, Russia
D. V. Korolev
Pacific State Medical UniversityVladivostok, Russia
V. B. Shumatov
Pacific State Medical UniversityVladivostok, Russia
References
- Arosio B, Calvani R, Ferri E, Coelho-Junior HJ, Carandina A, Campanelli F, Ghiglieri V, Marzetti E, Picca A (2023) Sarcopenia and cognitive decline in older adults: targeting the muscle-brain axis. Nutrients 15(8): 1853. https://doi.org/10.3390/nu15081853
- Dao T, Green AE, Kim YA, Bae SJ, Ha KT, Gariani K, Lee MR, Menzies KJ, Ryu D (2020) Sarcopenia and muscle aging: a brief overview. Endocrinol Metab (Seoul) 35(4): 716–732. https://doi.org/10.3803/EnM.2020.405
- Englund DA, Zhang X, Aversa Z, LeBrasseur NK (2021) Skeletal muscle aging, cellular senescence, and senotherapeutics: Current knowledge and future directions. Mech Ageing Dev 200: 111595. https://doi.org/10.1016/j.mad.2021.111595
- Marzetti E, Calvani R, Coelho-Júnior HJ, Landi F, Picca A (2024) Mitochondrial quantity and quality in age-related sarcopenia. Int J Mol Sci 25(4): 2052. https://doi.org/10.3390/ijms25042052
- Zhang L, Pitcher LE, Yousefzadeh MJ, Niedernhofer LJ, Robbins PD, Zhu Y (2022) Cellular senescence: a key therapeutic target in aging and diseases. J Clin Invest 132(15): e158450. https://doi.org/10.1172/JCI158450
- Rex N, Melk A, Schmitt R (2023) Cellular senescence and kidney aging. Clin Sci (Lond) 137(24): 1805–1821. https://doi.org/10.1042/CS20230140
- Melo Dos Santos LS, Trombetta-Lima M, Eggen B, Demaria M (2024) Cellular senescence in brain aging and neurodegeneration. Ageing Res Rev 93: 102141. https://doi.org/10.1016/j.arr.2023.102141
- De Magalhães JP (2024) Cellular senescence in normal physiology. Science 384(6702): 1300–1301. https://doi.org/10.1126/science.adj7050
- Aversa Z, Zhang X, Fielding RA, Lanza I, LeBrasseur NK (2019) The clinical impact and biological mechanisms of skeletal muscle aging. Bone 127: 26–36. https://doi.org/10.1016/j.bone.2019.05.021
- Игрункова АВ, Валиева ЯМ, Калиниченко АМ, Курков АВ, Попова КЮ, Шестаков ДЮ, Заборова ВА (2022) Клеточное старение: молекулярный и морфологический аспекты. Мол мед 20(4): 16–21. [Igrunkova AV, Valieva YaM, Kalinichenko AM, Kurkov AV, Popova KYu, Shestakov DYu, Zaborova VA (2022) Cellular senescence: molecular and morphological aspects. Mol Med 20(4): 16–21. (In Russ)]. https://doi.org/10.29296/24999490-2022-04-03
- Pabla P, Jones EJ, Piasecki M, Phillips BE (2024) Skeletal muscle dysfunction with advancing age. Clin Sci (Lond) 138(14): 863–882. https://doi.org/10.1042/CS20231197
- Brooks SV, Guzman SD, Ruiz LP (2023) Skeletal muscle structure, physiology, and function. Handb Clin Neurol 195: 3–16. https://doi.org/10.1016/B978-0-323-98818-6.00013-3
- Granic A, Martin-Ruiz C, Dodds RM, Robinson L, Spyridopoulos I, Kirkwood TB, von Zglinicki T, Sayer AA (2020) Immuno profiles are not associated with muscle strength, physical performance and sarcopenia risk in very old adults: the Newcastle 85+ study. Mech Ageing Dev 190: 111321. https://doi.org/10.1016/j.mad.2020.111321
- Park J, Shin DW (2022) Senotherapeutics and their molecular mechanism for improving aging. Biomol Ther (Seoul) 30(6): 490–500. https://doi.org/10.4062/biomolther.2022.114
- Falvino A, Gasperini B, Cariati I, Bonanni R, Chiavoghilefu A, Gasbarra E, Botta A, Tancredi V, Tarantino U (2024) Cellular senescence: the driving force of musculoskeletal diseases. Biomedicines 12(9): 1948. https://doi.org/10.3390/biomedicines12091948
- Moiseeva V, Cisneros A, Sica V, Deryagin O, Lai Y, Jung S, Andres E, An J, Segales J, Ortet L, Lukesova V, Volpe G, Benguria A, Dopazo A, Benitah SA, Urano Y, Del Sol A, Esteban MA, Ohkawa Y, Serrano AL, Perdiguero E, Munoz-Canoves P (2023) Senescence atlas reveals an aged-like inflamed niche that blunts muscle regeneration. Nature 613: 169–178. https://doi.org/10.1038/s41586-022-05535-x
- Burton DGA, Stolzing A (2018) Cellular senescence: Immunosurveillance and future immunotherapy. Ageing Res Rev 43: 17–25. https://doi.org/10.1016/j.arr.2018.02.001
- Xu M, Pirtskhalava T, Farr JN, Weigand BM, Palmer A, Weivoda MM, Inman CL, Ogrodnik M, Hachfeld CM, Fraser DG, Onken JL, Johnson KO, Verzosa GC, Langhi LGP, Weigl M, Giorgadze N, LeBrasseur NK, Miller JD, Jurk D, Singh RJ, Allison DB, Ejima K, Hubbard GB, Saito Y, Chikenji TS (2021) Diverse Roles of Cellular Senescence in skeletal muscle inflammation, regeneration, and therapeutics. Front Pharmacol 12: 739510. https://doi.org/10.3389/fphar.2021.739510
- Cai Y, Han Z, Cheng H, Li H, Wang K, Chen J, Liu ZX, Xie Y, Lin Y, Zhou S, Wang S, Zhou X, Jin S (2024) The impact of ageing mechanisms on musculoskeletal system diseases in the elderly. Front Immunol 15: 1405621. https://doi.org/10.3389/fimmu.2024.1405621
- Dungan CM, Peck BD, Walton RG, Huang Z, Bamman MM, Kern PA, Peterson CA (2020) In vivo analysis of γH2AX+ cells in skeletal muscle from aged and obese humans. Faseb J 34: 7018–7035. https://doi.org/10.1096/fj.202000111rr
- Larijani B, Foroughi-Heravani N, Alaei S, Rezaei-Tavirani M, Alavi-Moghadam S, Payab M, Goodarzi P, Tayanloo-Beik A, Aghayan HR, Arjmand B (2021) Opportunities and challenges in stem cell aging. Adv Exp Med Biol 1341: 143–175. https://doi.org/10.1007/5584_2021_624
- Sousa-Victor P, Gutarra S, Garcia-Prat L, Rodriguez-Ubreva J, Ortet L, Ruiz-Bonilla V, Jardi M, Ballestar E, Gonzalez S, Serrano AL, Perdiguero E, Munoz-Canoves P (2014) Geriatric muscle stem cells switch reversible quiescence into senescence. Nature 506: 316–321. https://doi.org/10.1038/nature13013
- Sousa-Victor P, Garcia-Prat L, Munoz-Canoves P (2022) Control of satellite cell function in muscle regeneration and its disruption in ageing. Nat Rev Mol Cell Biol 23: 204–226. https://doi.org/10.1038/s41580-021-00421-2
- Chikenji TS, Saito Y, Konari N, Nakano M, Mizue Y, Otani M, Fujimiya M (2019) p16INK4A-expressing mesenchymal stromal cells restore the senescence-clearance-regeneration sequence that is impaired in chronic muscle inflammation. Ebiomedicine 44: 86–97. https://doi.org/10.1016/j.ebiom.2019.05.012
- Mu X, Tang Y, Lu A, Takayama K, Usas A, Wang B, Weiss K, Huard J (2015) The role of notch signaling in muscle progenitor cell depletion and the rapid onset of histopathology in muscular dystrophy. Hum Mol Genet 24: 2923–2937. https://doi.org/10.1093/hmg/ddv055
- Sousa-Victor P, Perdiguero E, Munoz-Canoves P (2014) Geroconversion of aged muscle stem cells under regenerative pressure. Cell Cycle 13: 3183–3190. https://doi.org/10.4161/15384101.2014.965072
- Saito Y, Chikenji TS (2021) Diverse Roles of Cellular Senescence in Skeletal Muscle Inflammation, Regeneration, and Therapeutics. Front Pharmacol 12: 739510. https://doi.org/10.3389/fphar.2021.739510
- Zhu P, Zhang C, Gao Y, Wu F, Zhou Y, Wu WS (2019) The transcription factor Slug represses p16Ink4a and regulates murine muscle stem cell aging. Nat Commun 10: 2568. https://doi.org/10.1038/s41467-019-10479-4
- Kudryashova E, Kramerova I, Spencer MJ (2012) Satellite cell senescence underlies myopathy in a mouse model of limb-girdle muscular dystrophy 2H. J Clin Invest 122: 1764–1776. https://doi.org/10.1172/jci59581
- Sugihara H, Teramoto N, Nakamura K, Shiga T, Shirakawa T, Matsuo M, Ogasawara M, Nishino I, Matsuwaki T, Nishihara M, Yamanouchi K (2020) Cellular senescence-mediated exacerbation of duchenne muscular dystrophy. Sci Rep 10: 16385. https://doi.org/10.1038/s41598-020-73315-6
- Alcalde-Estévez E, Asenjo-Bueno A, Sosa P, Olmos G, Plaza P, Caballero-Mora MА, Rodríguez-Puyol D, Ruíz-Torres MP, López-Ongil S (2020) Endothelin-1 induces cellular senescence and fibrosis in cultured myoblasts. A potential mechanism of aging-related sarcopenia. Aging 12: 11200–11223. https://doi.org/10.18632/aging.103450
- Yu S, Ren B, Chen H, Goltzman D, Yan J, Miao D (2021) 1,25-Dihydroxyvitamin D deficiency induces sarcopenia by inducing skeletal muscle cell senescence. Am J Transl Res 13: 12638–12649. https://doi.org/10.18632/aging.103450
- Moustogiannis A, Philippou A, Taso O, Zevolis E, Pappa M, Chatzigeorgiou A, Koutsilieris M (2021) The effects of muscle cell aging on myogenesis. Int J Mol Sci 22: 3721. https://doi.org/10.3390/ijms22073721
- Scott RW, Arostegui M, Schweitzer R, Rossi FMV, Underhill TM (2019) Hic1 Defines Quiescent Mesenchymal Progenitor Subpopulations with Distinct Functions and Fates in Skeletal Muscle Regeneration. Cell Stem Cell 25(6): 797–813e9. https://doi.org/10.1016/j.stem.2019.11.004
- Theret M, Rossi FMV, Contreras O (2021) Evolving roles of muscle-resident fibro-adipogenic progenitors in health, regeneration, neuromuscular disorders, and aging. Front Physiol 12: 673404. https://doi.org/10.3389/fphys.2021.673404
- Giuliani G, Rosina M, Reggio A (2022) Signaling pathways regulating the fate of fibro/adipogenic progenitors (FAPs) in skeletal muscle regeneration and disease. FEBS J 289(21): 6484–6517. https://doi.org/10.1111/febs.16080
- Farup J, Madaro L, Puri PL, Mikkelsen UR (2015) Interactions between muscle stem cells, mesenchymal-derived cells and immune cells in muscle homeostasis, regeneration and disease. Cell Death Dis 6(7): e1830. https://doi.org/10.1038/cddis.2015.198
- Francis TG, Jaka O, Harridge SDR, Ellison-hughes GM, Lazarus NR (2022) Human primary skeletal muscle-derived myoblasts and fibroblasts reveal different senescent phenotypes. JCSM Rapid Commun 5: 226–238. https://doi.org/10.1002/rco2.67
- Molina T, Fabre P, Dumont NA (2021) Fibro-adipogenic progenitors in skeletal muscle homeostasis, regeneration and diseases. Open Biol 11(12): 210110. https://doi.org/10.1098/rsob.210110
- Vumbaca S, Giuliani G, Fiorentini V, Tortolici F, Cerquone Perpetuini A, Riccio F, Sennato S, Gargioli C, Fuoco C, Castagnoli L, Cesareni G (2021) Characterization of the skeletal muscle secretome reveals a role for extracellular vesicles and IL1α/IL1β in restricting fibro/adipogenic progenitor adipogenesis. Biomolecules 11(8): 1171. https://doi.org/10.3390/biom11081171
- Riparini G, Simone JM, Sartorelli V (2022) FACS-isolation and culture of fibro-adipogenic progenitors and muscle stem cells from unperturbed and injured mouse skeletal muscle. J Vis Exp 184: 10.3791/63983. https://doi.org/10.3791/63983
- Ratnayake D, Nguyen PD, Rossello FJ, Wimmer VC, Tan JL, Galvis LA, Julier Z, Wood AJ, Boudier T, Isiaku AI, Berger S, Oorschot V, Sonntag C, Rogers KL, Marcelle C, Lieschke GJ, Martino MM, Bakkers J, Currie PD (2021) Macrophages provide a transient muscle stem cell niche via NAMPT secretion. Nature 591: 281–287. https://doi.org/10.1038/s41586-021-03199-7
- Kuswanto W, Burzyn D, Panduro M, Wang KK, Jang YC, Wagers AJ, Benoist C, Mathis D (2016) Poor repair of skeletal muscle in aging mice reflects a defect in local, interleukin-33-dependent accumulation of regulatory T cells. Immunity 44: 355–367. https://doi.org/10.1016/j.immuni.2016.01.009
- Baker DJ, Weaver RL, van Deursen JM (2013) p21 both attenuates and drives senescence and aging in BubR1 progeroid mice. Cell Rep 3: 1164–1174. https://doi.org/10.1016/j.celrep.2013.03.028
- Chiche A, Le Roux I, von Joest M, Sakai H, Aguín SB, Cazin C, Salam R, Fiette L, Alegria O, Flamant P, Tajbakhsh S, Li H (2017) Injury-induced senescence enables in vivo reprogramming in skeletal muscle. Cell Stem Cell 20: 407–414e4. https://doi.org/10.1016/j.stem.2016.11.020
- Murach KA, Dimet-Wiley AL, Wen Y, Brightwell CR, Latham CM, Dungan CM, Fry CS, Watowich SJ (2022) Late-life exercise mitigates skeletal muscle epigenetic aging. Aging Cell J 21(1): e13527. https://doi.org/ 10.1111/acel.13527
- Liang W, Xu F, Li L, Peng C, Sun H, Qiu J, Sun J (2024) Epigenetic control of skeletal muscle atrophy. Cell Mol Biol Lett 29(1): 99. https://doi.org/ 10.1186/s11658-024-00618-1
- Liu JC, Dong SS, Shen H, Yang DY, Chen BB, Ma XY, Peng YR, Xiao HM, Deng HW (2022) Multi-omics research in sarcopenia: Current progress and future prospects. Ageing Res Rev 76: 101576. https://doi.org/ 10.1016/j.arr.2022.101576
- Hannum G, Guinney J, Zhao L, Zhang L, Hughes G, Sadda S, Klotzle B, Bibikova M, Fan JB, Gao Y, Deconde R, Chen M, Rajapakse I, Friend S, Ideker T, Zhang K (2013) Genome-wide methylation profiles reveal quantitative views of human aging rates. Mol Cell 49(2): 359–367. https://doi.org/10.1016/j.molcel.2012.10.016
- Horvath S (2015) DNA methylation age of human tissues and cell types. Genome Biol 14(10): R115. https://doi.org/ 10.1186/gb-2013-14-10-r115 015-0649-6
- Moresi V, Marroncelli N, Coletti D, Adamo S (2015) Regulation of skeletal muscle development and homeostasis by gene imprinting, histone acetylation and microRNA. Biochim Biophys Acta 1849(3): 309–316. https://doi.org/10.1016/j.bbagrm.2015.01.002
- Buonaiuto G, Desideri F, Taliani V, Ballarino M (2021) Muscle Regeneration and RNA: New Perspectives for Ancient Molecules. Cells Sep 10(10): 2512. https://doi.org/10.3390/cells10102512
- Liu Z, Liang Q, Ren Y, Guo C, Ge X, Wang L, Cheng Q, Luo P, Zhang Y, Han X (2023) Immunosenescence: molecular mechanisms and diseases. Signal Transduct Target Ther 8(1): 200. https://doi.org/10.1038/s41392-023-01451-2
- Barbé-Tuana F, Funchal G, Schmitz CRR, Maurmann RM, Bauer ME (2020) The interplay between immunosenescence and age-related diseases. Semin Immunopathol 42(5): 545–557. https://doi.org/ 10.1007/s00281-020-00806-z
- Yousefzadeh MJ, Flores RR, Zhu Y, Schmiechen ZC, Brooks RW, Trussoni CE, Cui Y, Angelini L, Lee KA, McGowan SJ, Burrack AL, Wang D, Dong Q, Lu A, Sano T, O'Kelly RD, McGuckian CA, Kato JI, Bank MP, Wade EA, Pillai SPS, Klug J, Ladiges WC, Burd CE, Lewis SE, LaRusso NF, Vo NV, Wang Y, Kelley EE, Huard J, Stromnes IM, Robbins PD, Niedernhofer LJ (2021) An aged immune system drives senescence and ageing of solid organs. Nature 594: 100–105. https://doi.org/10.1038/s41586-021-03547-7
- Ming GF, Wu K, Hu K, Chen Y, Xiao J (2016) NAMPT regulates senescence, proliferation, and migration of endothelial progenitor cells through the SIRT1 AS lncRNA/miR-22/SIRT1 pathway. Biochem Biophys Res Commun 478: 1382–1388. https://doi.org/10.1016/j.bbrc.2016.08.133
- Strasser B, Wolters M, Weyh C, Krüger K, Ticinesi A (2021) The effects of lifestyle and diet on gut microbiota composition, inflammation and muscle performance in our aging society. Nutrients 13(6): 2045. https://doi.org/10.3390/nu13062045
- Ticinesi A, Nouvenne A, Cerundolo N, Catania P, Prati B, Tana C, Meschi T (2019) Gut microbiota, muscle mass and function in aging: a focus on physical frailty and sarcopenia. Nutrients 11(7): 1633. https://doi.org/10.3390/nu11071633
- Liu C, Cheung WH, Li J, Chow SK, Yu J, Wong SH, Ip M, Sung JJY, Wong RMY (2021) Understanding the gut microbiota and sarcopenia: a systematic review. J Cachexia Sarcopenia Muscle 12(6): 1393–1407. https://doi.org/10.1002/jcsm.12784
- Grahnemo L, Nethander M, Coward E, Gabrielsen ME, Sree S, Billod JM, Sjögren K, Engstrand L, Dekkers KF, Fall T, Langhammer A, Hveem K, Ohlsson C (2023) Identification of three bacterial species associated with increased appendicular lean mass: the HUNT study. Nat Commun 14(1): 2250. https://doi.org/10.1038/s41467-023-37978-9
- Granic A, Sayer AA, Jurk D, Lanza IR, Khosla S, Fielding RA, Nair KS, Schafer MJ, Passos JF, LeBrasseur NK (2022) Characterization of cellular senescence in aging skeletal muscle. Nat Aging 2: 601–615. https://doi.org/10.1038/s43587-022-00250-8
- Fielding RA, Atkinson EJ, Aversa Z, White TA, Heeren AA, Achenbach SJ, Mielke MM, Cummings SR, Pahor M, Leeuwenburgh C, LeBrasseur NK (2022) Associations between biomarkers of cellular senescence and physical function in humans: Observations from the lifestyle interventions for elders (LIFE) study. Geroscience 44: 2757–2770. https://doi.org/10.1007/s11357-022-00685-2
- Gerosa L, Malvandi AM, Malavolta M, Provinciali M, Lombardi G (2023) Exploring cellular senescence in the musculoskeletal system: Any insights for biomarkers discovery? Ageing Res Rev 88: 101943. https://doi.org/10.1016/j.arr.2023.101943
- Da Silva PFL, Ogrodnik M, Kucheryavenko O, Glibert J, Miwa S, Cameron K, Ishaq A, Saretzki G, Nagaraja-Grellscheid S, Nelson G, von Zglinicki T (2019) The bystander effect contributes to the accumulation of senescent cells in vivo. Aging Cell 18: e12848. https://doi.org/10.1111/acel.12848
- Wan M, Gray-Gaillard EF, Elisseeff JH (2021) Cellular senescence in musculoskeletal homeostasis, diseases, and regeneration. Bone Res 9(1): 41. https://doi.org/10.1038/s41413-021-00164-y
- Mijares A, Allen PD, Lopez JR (2021) Senescence is associated with elevated intracellular resting [Ca2 +] in mice skeletal muscle fibers. An in vivo study. Front Physiol 11: 601189. https://doi.org/10.3389/fphys.2020.601189
- Kumari R, Jat P (2021) Mechanisms of cellular senescence: cell cycle arrest and senescence associated secretory phenotype. Front Cell Dev Biol 29(9): 645593. https://doi.org/10.3389/fcell.2021.645593
- Hall BM, Balan V, Gleiberman AS (2016) Aging of mice is associated with p16(Ink4a)- and beta-galactosidase-positive macrophage accumulation that can be induced in young mice by senescent cells. Aging (Albany NY) 8(7): 1294–1315. https://doi.org/10.18632/aging.100991
- Alsharidah M, Lazarus NR, George TE, Agley CC, Velloso CP, Harridge SD (2013) Primary human muscle precursor cells obtained from young and old donors produce similar proliferative, differentiation and senescent profiles in culture. Aging Сell 12: 333–344. https://doi.org/10.1111/acel.12051
- Sugihara H, Teramoto N, Yamanouchi K, Matsuwaki T, Nishihara M (2018) Oxidative stress-mediated senescence in mesenchymal progenitor cells causes the loss of their fibro/adipogenic potential and abrogates myoblast fusion. Aging 10: 747–763. https://doi.org/10.18632/aging.101425
- Zhu S, Tian Z, Torigoe D, Zhao J, Xie P, Sugizaki T, Sato M, Horiguchi H, Terada K, Kadomatsu T, Miyata K, Oike Y (2019) Aging- and obesity-related peri-muscular adipose tissue accelerates muscle atrophy. PLoS One 14(8): e0221366. https://doi.org/10.1371/journal.pone.0221366
- Tezze C, Romanello V, Desbats MA, Fadini GP, Albiero M, Favaro G, Ciciliot S, Soriano ME, Morbidoni V, Cerqua C, Loefler S, Kern H, Franceschi C, Salvioli S, Conte M, Blaauw B, Zampieri S, Salviati L, Scorrano L, Sandri M (2017) Age-Associated Loss of OPA1 in Muscle Impacts Muscle Mass, Metabolic Homeostasis, Systemic Inflammation, and Epithelial Senescence. Cell Metab 5(6): 1374–1389.e6. https://doi.org/10.1016/j.cmet.2017.04.021
- Chen XK, Yi ZN, Wong GTC, Hasan KMM, Kwan JSK, Ma ACH, Chang RC (2021) Is exercise a senolytic medicine? A Systematic Review. Aging Cell 20: e13294. https://doi.org/10.1111/acel.13294
- Tieland M, Trouwborst I, Clark BC (2018) Skeletal muscle performance and ageing. J Cachexia Sarcopenia Muscle 9(1): 3–19. https://doi.org/10.1002/jcsm.12238
- Zhang L, Pitcher LE, Prahalad V, Niedernhofer LJ, Robbins PD (2023) Targeting cellular senescence with senotherapeutics: senolytics and senomorphics. FEBS J 290(5): 1362–1383. https://doi.org/10.1111/febs.16350
- Cai Y, Zhou H, Zhu Y, Sun Q, Ji Y, Xue A, Wang Y, Chen W, Yu X, Wang L, Chen H, Li C, Luo T, Deng H (2020) Elimination of senescent cells by beta-galactosidase-targeted prodrug attenuates inflammation and restores physical function in aged mice. Cell Res 30: 574–589. https://doi.org/10.1038/s41422-020-0314-9
- Chang J, Wang Y, Shao L, Laberge RM, Demaria M, Campisi J, Janakiraman K, Sharpless NE, Ding S, Feng W, Luo Y, Wang X, Aykin-Burns N, Krager K, Ponnappan U, Hauer-Jensen M, Meng A, Zhou D (2016) Clearance of senescent cells by ABT263 rejuvenates aged hematopoietic stem cells in mice. Nat Med 22: 78–83. https://doi.org/10.1038/s41422-020-0314-9
- Baar MP, Brandt RMC, Putavet DA, Klein JDD, Derks KWJ, Bourgeois BRM, Stryeck S, Rijksen Y, van Willigenburg H, Feijtel DA, van der Pluijm I, Essers J, van Cappellen WA, van IJcken WF, Houtsmuller AB, Pothof J, de Bruin RWF, Madl T, Hoeijmakers JHJ, Campisi J, de Keizer PLJ (2017) Targeted apoptosis of senescent cells restores tissue homeostasis in response to chemotoxicity and aging. Cell 169(1): 132–147.e16. https://doi.org/10.1016/j.cell.02.031
- Chung L, Maestas DR Jr, Lebid A, Mageau A, Rosson GD, Wu X, Wolf MT, Tam AJ, Vanderzee I, Wang X, Andorko JI, Zhang H, Narain R, Sadtler K, Fan H, Čiháková D, Le Saux CJ, Housseau F, Pardoll DM, Elisseeff JH (2020) Interleukin 17 and senescent cells regulate the foreign body response to synthetic material implants in mice and humans. Sci Transl Med 2(539): eaax3799. https://doi.org/10.1126/scitranslmed.aax3799
- Khor SC, Wan Ngah WZ, Mohd Yusof YA, Abdul Karim N, Makpol S (2017) Tocotrienol-rich fraction ameliorates antioxidant defense mechanisms and improves replicative senescence-associated oxidative stress in human myoblasts. Oxid Med Cell Longev 2017: 3868305. https://doi.org/10.1155/2017/3868305
- Lim JJ, Wan Zurinah WN, Mouly V, Norwahidah AK (2019) Tocotrienol-rich fraction (TRF) treatment promotes proliferation capacity of stress-induced premature senescence myoblasts and modulates the renewal of satellite cells: microarray analysis. Oxid Med Cell Longev 2019: 9141343. https://doi.org/10.1155/2019/9141343
- Yanai K, Kaneko S, Ishii H, Aomatsu A, Ito K, Hirai K, Ookawara S, Ishibashi K, Morishita Y (2020) MicroRNAs in sarcopenia: a systematic review. Front Med (Lausanne) 7: 180.
Supplementary files
