Diabetes Metab J.  2023 Jul;47(4):441-453. 10.4093/dmj.2022.0416.

Regulation of Cellular Senescence in Type 2 Diabetes Mellitus: From Mechanisms to Clinical Applications

Affiliations
  • 1Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
  • 2Tazuke Kofukai Medical Research Institute, Kitano Hospital, Osaka, Japan

Abstract

Cellular senescence is accelerated by hyperglycemia through multiple pathways. Therefore, senescence is an important cellular mechanism to consider in the pathophysiology of type 2 diabetes mellitus (T2DM) and an additional therapeutic target. The use of drugs that remove senescent cells has led to improvements in blood glucose levels and diabetic complications in animal studies. Although the removal of senescent cells is a promising approach for the treatment of T2DM, two main challenges limit its clinical application: the molecular basis of cellular senescence in each organ is yet to be understood, and the specific effect of removing senescent cells in each organ has to be determined. This review aims to discuss future applications of targeting senescence as a therapeutic option in T2DM and elucidate the characteristics of cellular senescence and senescence-associated secretory phenotype in the tissues important for regulating glucose levels: pancreas, liver, adipocytes, and skeletal muscle.

Keyword

Aging; Diabetes mellitus; type 2; Insulin-secreting cells; Senotherapeutics

Figure

  • Fig. 1 Role of cellular senescence in type 2 diabetes mellitus. The upper part of the figure depicts factors known to lead to senescence in cells: mitochondrial dysfunction, endoplasmic reticulum (ER) stress, and increased production of reactive oxygen species (ROS). Additional factors include telomere dysfunction or shortening, DNA damage, hyperglycemia, insulin resistance, and inflammatory signals. The lower part of the picture reflects some of the reported negative effects of cellular senescence: secretion of the senescence-associated secretory phenotype (SASP), which can promote the entry to senescence of neighboring cells and their dysfunction. The proliferation of cells is suppressed and they downregulate hallmark identity genes. Due to these characteristics, senescent cells can promote diabetes and metabolic diseases in multiple ways. Created in BioRender.

  • Fig. 2 Senotherapeutics used in diabetes and diabetes related diseases. Schematic representation of cellular senescence agents used and their target pathways. (1) Senescence molecules effectors, such as hyperglycemia and DNA damage, cause healthy cells to senesce; (2) Senescent cells secrete senescence-associated secretory phenotype (SASP) and lose their cell identity. (3) There are two types of senotherapeutics: senolytics, which eliminate senescent cells, and senomorphics, which inhibit SASP. The drugs in the figures have been used in mouse and human models of type 2 diabetes mellitus. Of these, dasatinib, quercetin, and fisetin have been used in clinical studies. Created in BioRender. HIF-1α, hypoxia-inducible factor 1α; PI3K, phosphoinositide 3-kinase; Bcl2, B-cell lymphoma 2; AMPK, AMP-activated protein kinase; mTOR, mammalian TORC1.


Reference

1. Kirkland JL, Tchkonia T. Senolytic drugs: from discovery to translation. J Intern Med. 2020; 288:518–36.
Article
2. Wang L, Lankhorst L, Bernards R. Exploiting senescence for the treatment of cancer. Nat Rev Cancer. 2022; 22:340–55.
Article
3. Wang MJ, Chen F, Lau JT, Hu YP. Hepatocyte polyploidization and its association with pathophysiological processes. Cell Death Dis. 2017; 8:e2805.
Article
4. Celton-Morizur S, Merlen G, Couton D, Margall-Ducos G, Desdouets C. The insulin/Akt pathway controls a specific cell division program that leads to generation of binucleated tetraploid liver cells in rodents. J Clin Invest. 2009; 119:1880–7.
Article
5. Aguayo-Mazzucato C, Andle J, Lee TB, Midha A, Talemal L, Chipashvili V, et al. Acceleration of β cell aging determines diabetes and senolysis improves disease outcomes. Cell Metab. 2019; 30:129–42e4.
Article
6. Li Q, Hagberg CE, Silva Cascales H, Lang S, Hyvonen MT, Salehzadeh F, et al. Obesity and hyperinsulinemia drive adipocytes to activate a cell cycle program and senesce. Nat Med. 2021; 27:1941–53.
Article
7. Wan Y, Liu Z, Wu A, Khan AH, Zhu Y, Ding S, et al. Hyperglycemia promotes endothelial cell senescence through AQR/PLAU signaling axis. Int J Mol Sci. 2022; 23:2879.
Article
8. Kitada K, Nakano D, Ohsaki H, Hitomi H, Minamino T, Yatabe J, et al. Hyperglycemia causes cellular senescence via a SGLT2- and p21-dependent pathway in proximal tubules in the early stage of diabetic nephropathy. J Diabetes Complications. 2014; 28:604–11.
Article
9. Liu J, Huang K, Cai GY, Chen XM, Yang JR, Lin LR, et al. Receptor for advanced glycation end-products promotes premature senescence of proximal tubular epithelial cells via activation of endoplasmic reticulum stress-dependent p21 signaling. Cell Signal. 2014; 26:110–21.
Article
10. Liu J, Yang JR, Chen XM, Cai GY, Lin LR, He YN. Impact of ER stress-regulated ATF4/p16 signaling on the premature senescence of renal tubular epithelial cells in diabetic nephropathy. Am J Physiol Cell Physiol. 2015; 308:C621–30.
Article
11. Wiley CD, Campisi J. The metabolic roots of senescence: mechanisms and opportunities for intervention. Nat Metab. 2021; 3:1290–301.
Article
12. International Diabetes Federation. IDF Diabetes Atlas. 10th ed. Brussels: IDF;2021.
13. Hayflick L, Moorhead PS. The serial cultivation of human diploid cell strains. Exp Cell Res. 1961; 25:585–621.
Article
14. Hayflick L. The limited in vitro lifetime of human diploid cell strains. Exp Cell Res. 1965; 37:614–36.
Article
15. Burnet FM. Intrinsic mutagenesis: a genetic basis of ageing. Pathology. 1974; 6:1–11.
Article
16. Shay JW, Wright WE. Hayflick, his limit, and cellular ageing. Nat Rev Mol Cell Biol. 2000; 1:72–6.
Article
17. Harley CB, Futcher AB, Greider CW. Telomeres shorten during ageing of human fibroblasts. Nature. 1990; 345:458–60.
Article
18. Bodnar AG, Ouellette M, Frolkis M, Holt SE, Chiu CP, Morin GB, et al. Extension of life-span by introduction of telomerase into normal human cells. Science. 1998; 279:349–52.
Article
19. van Deursen JM. The role of senescent cells in ageing. Nature. 2014; 509:439–46.
Article
20. Palmer AK, Tchkonia T, LeBrasseur NK, Chini EN, Xu M, Kirkland JL. Cellular senescence in type 2 diabetes: a therapeutic opportunity. Diabetes. 2015; 64:2289–98.
Article
21. Campisi J, d’Adda di Fagagna F. Cellular senescence: when bad things happen to good cells. Nat Rev Mol Cell Biol. 2007; 8:729–40.
Article
22. Dimri GP, Lee X, Basile G, Acosta M, Scott G, Roskelley C, et al. A biomarker that identifies senescent human cells in culture and in aging skin in vivo. Proc Natl Acad Sci U S A. 1995; 92:9363–7.
Article
23. Gonzalez-Gualda E, Baker AG, Fruk L, Munoz-Espin D. A guide to assessing cellular senescence in vitro and in vivo. FEBS J. 2021; 288:56–80.
24. Leidal AM, Levine B, Debnath J. Autophagy and the cell biology of age-related disease. Nat Cell Biol. 2018; 20:1338–48.
Article
25. Matsunaga K, Saitoh T, Tabata K, Omori H, Satoh T, Kurotori N, et al. Two Beclin 1-binding proteins, Atg14L and Rubicon, reciprocally regulate autophagy at different stages. Nat Cell Biol. 2009; 11:385–96.
Article
26. Yamamoto-Imoto H, Minami S, Shioda T, Yamashita Y, Sakai S, Maeda S, et al. Age-associated decline of MondoA drives cellular senescence through impaired autophagy and mitochondrial homeostasis. Cell Rep. 2022; 38:110444.
Article
27. Palmer AK, Tchkonia T, Kirkland JL. Senolytics: potential for alleviating diabetes and its complications. Endocrinology. 2021; 162:bqab058.
Article
28. Coppe JP, Patil CK, Rodier F, Sun Y, Munoz DP, Goldstein J, et al. Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biol. 2008; 6:2853–68.
Article
29. Han X, Lei Q, Xie J, Liu H, Li J, Zhang X, et al. Potential regulators of the senescence-associated secretory phenotype during senescence and aging. J Gerontol A Biol Sci Med Sci. 2022; 77:2207–18.
Article
30. Rodier F, Coppe JP, Patil CK, Hoeijmakers WA, Munoz DP, Raza SR, et al. Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion. Nat Cell Biol. 2009; 11:973–9.
Article
31. Chien Y, Scuoppo C, Wang X, Fang X, Balgley B, Bolden JE, et al. Control of the senescence-associated secretory phenotype by NF-κB promotes senescence and enhances chemosensitivity. Genes Dev. 2011; 25:2125–36.
Article
32. Freund A, Patil CK, Campisi J. p38MAPK is a novel DNA damage response-independent regulator of the senescence-associated secretory phenotype. EMBO J. 2011; 30:1536–48.
Article
33. Xu M, Tchkonia T, Ding H, Ogrodnik M, Lubbers ER, Pirtskhalava T, et al. JAK inhibition alleviates the cellular senescence-associated secretory phenotype and frailty in old age. Proc Natl Acad Sci U S A. 2015; 112:E6301–10.
Article
34. Loo TM, Miyata K, Tanaka Y, Takahashi A. Cellular senescence and senescence-associated secretory phenotype via the cGAS-STING signaling pathway in cancer. Cancer Sci. 2020; 111:304–11.
Article
35. Mabrouk N, Ghione S, Laurens V, Plenchette S, Bettaieb A, Paul C. Senescence and cancer: role of nitric oxide (NO) in SASP. Cancers (Basel). 2020; 12:1145.
Article
36. Demaria M, Ohtani N, Youssef SA, Rodier F, Toussaint W, Mitchell JR, et al. An essential role for senescent cells in optimal wound healing through secretion of PDGF-AA. Dev Cell. 2014; 31:722–33.
Article
37. Yang G, Rosen DG, Zhang Z, Bast RC, Mills GB, Colacino JA, et al. The chemokine growth-regulated oncogene 1 (Gro-1) links RAS signaling to the senescence of stromal fibroblasts and ovarian tumorigenesis. Proc Natl Acad Sci U S A. 2006; 103:16472–7.
38. Laberge RM, Sun Y, Orjalo AV, Patil CK, Freund A, Zhou L, et al. MTOR regulates the pro-tumorigenic senescence-associated secretory phenotype by promoting IL1A translation. Nat Cell Biol. 2015; 17:1049–61.
39. Acosta JC, Banito A, Wuestefeld T, Georgilis A, Janich P, Morton JP, et al. A complex secretory program orchestrated by the inflammasome controls paracrine senescence. Nat Cell Biol. 2013; 15:978–90.
40. Munoz-Espin D, Serrano M. Cellular senescence: from physiology to pathology. Nat Rev Mol Cell Biol. 2014; 15:482–96.
41. Karin O, Agrawal A, Porat Z, Krizhanovsky V, Alon U. Senescent cell turnover slows with age providing an explanation for the Gompertz law. Nat Commun. 2019; 10:5495.
42. Gardner JP, Li S, Srinivasan SR, Chen W, Kimura M, Lu X, et al. Rise in insulin resistance is associated with escalated telomere attrition. Circulation. 2005; 111:2171–7.
43. Yu J, Nam D, Park KS. Substance P enhances cellular migration and inhibits senescence in human dermal fibroblasts under hyperglycemic conditions. Biochem Biophys Res Commun. 2020; 522:917–23.
Article
44. Sone H, Kagawa Y. Pancreatic beta cell senescence contributes to the pathogenesis of type 2 diabetes in high-fat diet-induced diabetic mice. Diabetologia. 2005; 48:58–67.
Article
45. Baker DJ, Wijshake T, Tchkonia T, LeBrasseur NK, Childs BG, van de Sluis B, et al. Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature. 2011; 479:232–6.
Article
46. Kantarjian H, Jabbour E, Grimley J, Kirkpatrick P. Dasatinib. Nat Rev Drug Discov. 2006; 5:717–8.
Article
47. Russo M, Milito A, Spagnuolo C, Carbone V, Rosen A, Minasi P, et al. CK2 and PI3K are direct molecular targets of quercetin in chronic lymphocytic leukaemia. Oncotarget. 2017; 8:42571–87.
Article
48. Zhu Y, Tchkonia T, Pirtskhalava T, Gower AC, Ding H, Giorgadze N, et al. The Achilles’ heel of senescent cells: from transcriptome to senolytic drugs. Aging Cell. 2015; 14:644–58.
Article
49. Hickson LJ, Langhi Prata LG, Bobart SA, Evans TK, Giorgadze N, Hashmi SK, et al. Senolytics decrease senescent cells in humans: preliminary report from a clinical trial of dasatinib plus quercetin in individuals with diabetic kidney disease. EBioMedicine. 2019; 47:446–56.
Article
50. Kirkland JL, Tchkonia T, Zhu Y, Niedernhofer LJ, Robbins PD. The clinical potential of senolytic drugs. J Am Geriatr Soc. 2017; 65:2297–301.
Article
51. Gastaldelli A, Cusi K. From NASH to diabetes and from diabetes to NASH: mechanisms and treatment options. JHEP Rep. 2019; 1:312–28.
Article
52. European Association for the Study of the Liver (EASL); European Association for the Study of Diabetes (EASD); European Association for the Study of Obesity (EASO). EASL-EASD-EASO clinical practice guidelines for the management of non-alcoholic fatty liver disease. J Hepatol. 2016; 64:1388–402.
53. Aravinthan A, Scarpini C, Tachtatzis P, Verma S, Penrhyn-Lowe S, Harvey R, et al. Hepatocyte senescence predicts progression in non-alcohol-related fatty liver disease. J Hepatol. 2013; 58:549–56.
Article
54. Baboota RK, Spinelli R, Erlandsson MC, Brandao BB, Lino M, Yang H, et al. Chronic hyperinsulinemia promotes human hepatocyte senescence. Mol Metab. 2022; 64:101558.
55. Ogrodnik M, Miwa S, Tchkonia T, Tiniakos D, Wilson CL, Lahat A, et al. Cellular senescence drives age-dependent hepatic steatosis. Nat Commun. 2017; 8:15691.
56. Baboota RK, Rawshani A, Bonnet L, Li X, Yang H, Mardinoglu A, et al. BMP4 and Gremlin 1 regulate hepatic cell senescence during clinical progression of NAFLD/NASH. Nat Metab. 2022; 4:1007–21.
57. Chen MS, Lee RT, Garbern JC. Senescence mechanisms and targets in the heart. Cardiovasc Res. 2022; 118:1173–87.
58. Cianflone E, Torella M, Biamonte F, De Angelis A, Urbanek K, Costanzo FS, et al. Targeting cardiac stem cell senescence to treat cardiac aging and disease. Cells. 2020; 9:1558.
59. Chi C, Li DJ, Jiang YJ, Tong J, Fu H, Wu YH, et al. Vascular smooth muscle cell senescence and age-related diseases: state of the art. Biochim Biophys Acta Mol Basis Dis. 2019; 1865:1810–21.
60. Zha Y, Zhuang W, Yang Y, Zhou Y, Li H, Liang J. Senescence in vascular smooth muscle cells and atherosclerosis. Front Cardiovasc Med. 2022; 9:910580.
Article
61. Saito Y, Chikenji TS. Diverse roles of cellular senescence in skeletal muscle inflammation, regeneration, and therapeutics. Front Pharmacol. 2021; 12:739510.
Article
62. Solovyeva EM, Ibebunjo C, Utzinger S, Eash JK, Dunbar A, Naumann U, et al. New insights into molecular changes in skeletal muscle aging and disease: differential alternative splicing and senescence. Mech Ageing Dev. 2021; 197:111510.
Article
63. Walaszczyk A, Dookun E, Redgrave R, Tual-Chalot S, Victorelli S, Spyridopoulos I, et al. Pharmacological clearance of senescent cells improves survival and recovery in aged mice following acute myocardial infarction. Aging Cell. 2019; 18:e12945.
64. Karnewar S, Neeli PK, Panuganti D, Kotagiri S, Mallappa S, Jain N, et al. Metformin regulates mitochondrial biogenesis and senescence through AMPK mediated H3K79 methylation: relevance in age-associated vascular dysfunction. Biochim Biophys Acta Mol Basis Dis. 2018; 1864(4 Pt A):1115–28.
Article
65. Kim SG, Sung JY, Kim JR, Choi HC. Quercetin-induced apoptosis ameliorates vascular smooth muscle cell senescence through AMP-activated protein kinase signaling pathway. Korean J Physiol Pharmacol. 2020; 24:69–79.
Article
66. Wan M, Gray-Gaillard EF, Elisseeff JH. Cellular senescence in musculoskeletal homeostasis, diseases, and regeneration. Bone Res. 2021; 9:41.
Article
67. Zhang H, Ryu D, Wu Y, Gariani K, Wang X, Luan P, et al. NAD+ repletion improves mitochondrial and stem cell function and enhances life span in mice. Science. 2016; 352:1436–43.
Article
68. Morley JE, Anker SD, von Haehling S. Prevalence, incidence, and clinical impact of sarcopenia: facts, numbers, and epidemiology-update 2014. J Cachexia Sarcopenia Muscle. 2014; 5:253–9.
Article
69. Sousa-Victor P, Gutarra S, Garcia-Prat L, Rodriguez-Ubreva J, Ortet L, Ruiz-Bonilla V, et al. Geriatric muscle stem cells switch reversible quiescence into senescence. Nature. 2014; 506:316–21.
Article
70. Li CW, Yu K, Shyh-Chang N, Jiang Z, Liu T, Ma S, et al. Pathogenesis of sarcopenia and the relationship with fat mass: descriptive review. J Cachexia Sarcopenia Muscle. 2022; 13:781–94.
Article
71. Choi KM. Sarcopenia and sarcopenic obesity. Korean J Intern Med. 2016; 31:1054–60.
Article
72. Minamino T, Orimo M, Shimizu I, Kunieda T, Yokoyama M, Ito T, et al. A crucial role for adipose tissue p53 in the regulation of insulin resistance. Nat Med. 2009; 15:1082–7.
Article
73. Chen YW, Harris RA, Hatahet Z, Chou KM. Ablation of XP-V gene causes adipose tissue senescence and metabolic abnormalities. Proc Natl Acad Sci U S A. 2015; 112:E4556–64.
Article
74. Liu Z, Jin L, Yang JK, Wang B, Wu KK, Hallenborg P, et al. The dysfunctional MDM2-p53 axis in adipocytes contributes to aging-related metabolic complications by induction of lipodystrophy. Diabetes. 2018; 67:2397–409.
Article
75. Xu M, Palmer AK, Ding H, Weivoda MM, Pirtskhalava T, White TA, et al. Targeting senescent cells enhances adipogenesis and metabolic function in old age. Elife. 2015; 4:e12997.
Article
76. Gustafson B, Nerstedt A, Smith U. Reduced subcutaneous adipogenesis in human hypertrophic obesity is linked to senescent precursor cells. Nat Commun. 2019; 10:2757.
Article
77. Justice JN, Nambiar AM, Tchkonia T, LeBrasseur NK, Pascual R, Hashmi SK, et al. Senolytics in idiopathic pulmonary fibrosis: results from a first-in-human, open-label, pilot study. EBioMedicine. 2019; 40:554–63.
Article
78. Rouault C, Marcelin G, Adriouch S, Rose C, Genser L, Ambrosini M, et al. Senescence-associated β-galactosidase in subcutaneous adipose tissue associates with altered glycaemic status and truncal fat in severe obesity. Diabetologia. 2021; 64:240–54.
Article
79. Xu M, Pirtskhalava T, Farr JN, Weigand BM, Palmer AK, Weivoda MM, et al. Senolytics improve physical function and increase lifespan in old age. Nat Med. 2018; 24:1246–56.
Article
80. Breccia M, Molica M, Alimena G. How tyrosine kinase inhibitors impair metabolism and endocrine system function: a systematic updated review. Leuk Res. 2014; 38:1392–8.
Article
81. Zhu Y, Tchkonia T, Fuhrmann-Stroissnigg H, Dai HM, Ling YY, Stout MB, et al. Identification of a novel senolytic agent, navitoclax, targeting the Bcl-2 family of anti-apoptotic factors. Aging Cell. 2016; 15:428–35.
Article
82. Lee S, Yu Y, Trimpert J, Benthani F, Mairhofer M, Richter-Pechanska P, et al. Virus-induced senescence is a driver and therapeutic target in COVID-19. Nature. 2021; 599:283–9.
Article
83. Pan R, Hogdal LJ, Benito JM, Bucci D, Han L, Borthakur G, et al. Selective BCL-2 inhibition by ABT-199 causes on-target cell death in acute myeloid leukemia. Cancer Discov. 2014; 4:362–75.
Article
84. Thompson PJ, Shah A, Ntranos V, Van Gool F, Atkinson M, Bhushan A. Targeted elimination of senescent beta cells prevents type 1 diabetes. Cell Metab. 2019; 29:1045–60e10.
Article
85. Kline MP, Rajkumar SV, Timm MM, Kimlinger TK, Haug JL, Lust JA, et al. ABT-737, an inhibitor of Bcl-2 family proteins, is a potent inducer of apoptosis in multiple myeloma cells. Leukemia. 2007; 21:1549–60.
Article
86. LaMoia TE, Shulman GI. Cellular and molecular mechanisms of metformin action. Endocr Rev. 2021; 42:77–96.
Article
87. Le Pelletier L, Mantecon M, Gorwood J, Auclair M, Foresti R, Motterlini R, et al. Metformin alleviates stress-induced cellular senescence of aging human adipose stromal cells and the ensuing adipocyte dysfunction. Elife. 2021; 10:e62635.
Article
88. Kim H, Yu MR, Lee H, Kwon SH, Jeon JS, Han DC, et al. Metformin inhibits chronic kidney disease-induced DNA damage and senescence of mesenchymal stem cells. Aging Cell. 2021; 20:e13317.
Article
89. Marycz K, Tomaszewski KA, Kornicka K, Henry BM, Wronski S, Tarasiuk J, et al. Metformin decreases reactive oxygen species, enhances osteogenic properties of adipose-derived multipotent mesenchymal stem cells in vitro, and increases bone density in vivo. Oxid Med Cell Longev. 2016; 2016:9785890.
90. Acar MB, Ayaz-Guner S, Gunaydin Z, Karakukcu M, Peluso G, Di Bernardo G, et al. Proteomic and biological analysis of the effects of metformin senomorphics on the mesenchymal stromal cells. Front Bioeng Biotechnol. 2021; 9:730813.
Article
91. Lee J, Kim YS, Kim E, Kim Y, Kim Y. Curcumin and hesperetin attenuate D-galactose-induced brain senescence in vitro and in vivo. Nutr Res Pract. 2020; 14:438–52.
Article
92. Yang L, Shi J, Wang X, Zhang R. Curcumin alleviates d-galactose-induced cardiomyocyte senescence by promoting autophagy via the SIRT1/AMPK/mTOR pathway. Evid Based Complement Alternat Med. 2022; 2022:2990843.
Article
93. Chuengsamarn S, Rattanamongkolgul S, Luechapudiporn R, Phisalaphong C, Jirawatnotai S. Curcumin extract for prevention of type 2 diabetes. Diabetes Care. 2012; 35:2121–7.
Article
94. Li C, Miao X, Wang S, Adhikari BK, Wang X, Sun J, et al. Novel curcumin C66 that protects diabetes-induced aortic damage was associated with suppressing JNK2 and upregulating Nrf2 expression and function. Oxid Med Cell Longev. 2018; 2018:5783239.
Article
95. Abdelsamia EM, Khaleel SA, Balah A, Abdel Baky NA. Curcumin augments the cardioprotective effect of metformin in an experimental model of type I diabetes mellitus; impact of Nrf2/HO-1 and JAK/STAT pathways. Biomed Pharmacother. 2019; 109:2136–44.
Article
96. Ahlqvist E, Storm P, Karajamaki A, Martinell M, Dorkhan M, Carlsson A, et al. Novel subgroups of adult-onset diabetes and their association with outcomes: a data-driven cluster analysis of six variables. Lancet Diabetes Endocrinol. 2018; 6:361–9.
Article
97. Carapeto PV, Aguayo-Mazzucato C. Effects of exercise on cellular and tissue aging. Aging (Albany NY). 2021; 13:14522–43.
Article
98. Shannon OM, Ashor AW, Scialo F, Saretzki G, Martin-Ruiz C, Lara J, et al. Mediterranean diet and the hallmarks of ageing. Eur J Clin Nutr. 2021; 75:1176–92.
Article
99. Nurk S, Koren S, Rhie A, Rautiainen M, Bzikadze AV, Mikheenko A, et al. The complete sequence of a human genome. Science. 2022; 376:44–53.
100. SenNet Consortium. NIH SenNet Consortium to map senescent cells throughout the human lifespan to understand physiological health. Nat Aging. 2022; 2:1090–100.
Full Text Links
  • DMJ
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr