REFERENCES

1. López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging. Cell 2013;153:1194-217.

2. Ince S. Nikolaos frangogiannis: preventing dangerous remodeling after myocardial infarction. Circ Res 2016;119:25-8.

3. Dobaczewski M, Chen W, Frangogiannis NG. Transforming growth factor (TGF)-β signaling in cardiac remodeling. J Mol Cell Cardiol 2011;51:600-6.

4. Yue Y, Meng K, Pu Y, Zhang X. Transforming growth factor beta (TGF-β) mediates cardiac fibrosis and induces diabetic cardiomyopathy. Diabetes Res Clin Pract 2017;133:124-30.

5. Batlle E, Massagué J. Transforming growth factor-β signaling in immunity and cancer. Immunity 2019;50:924-40.

6. Ravenscroft TA, Phillips JB, Fieg E, et al. Heterozygous loss-of-function variants significantly expand the phenotypes associated with loss of GDF11. Genet Med 2021;23:1889-900.

7. Cui H, Kong Y, Zhang H. Oxidative stress, mitochondrial dysfunction, and aging. J Signal Transduct 2012;2012:646354.

8. Kikuchi K, Poss KD. Cardiac regenerative capacity and mechanisms. Annu Rev Cell Dev Biol 2012;28:719-41.

9. Vagnozzi RJ, Molkentin JD, Houser SR. New myocyte formation in the adult heart: endogenous sources and therapeutic implications. Circ Res 2018;123:159-76.

10. Conboy IM, Conboy MJ, Smythe GM, Rando TA. Notch-mediated restoration of regenerative potential to aged muscle. Science 2003;302:1575-7.

11. Carlson BM, Faulkner JA. Muscle transplantation between young and old rats: age of host determines recovery. Am J Physiol 1989;256:C1262-6.

12. Sinha M, Jang YC, Oh J, et al. Restoring systemic GDF11 levels reverses age-related dysfunction in mouse skeletal muscle. Science 2014;344:649-52.

13. Sinha I, Sinha-hikim AP, Wagers AJ, Sinha-hikim I. Testosterone is essential for skeletal muscle growth in aged mice in a heterochronic parabiosis model. Cell Tissue Res 2014;357:815-21.

14. Katsimpardi L, Litterman NK, Schein PA, et al. Vascular and neurogenic rejuvenation of the aging mouse brain by young systemic factors. Science 2014;344:630-4.

15. Loffredo FS, Steinhauser ML, Jay SM, et al. Growth differentiation factor 11 is a circulating factor that reverses age-related cardiac hypertrophy. Cell 2013;153:828-39.

16. Ruckh JM, Zhao JW, Shadrach JL, et al. Rejuvenation of regeneration in the aging central nervous system. Cell Stem Cell 2012;10:96-103.

17. Conboy IM, Conboy MJ, Wagers AJ, Girma ER, Weissman IL, Rando TA. Rejuvenation of aged progenitor cells by exposure to a young systemic environment. Nature 2005;433:760-4.

18. Miron VE, Boyd A, Zhao JW, et al. M2 microglia and macrophages drive oligodendrocyte differentiation during CNS remyelination. Nat Neurosci 2013;16:1211-8.

19. Brack AS, Conboy MJ, Roy S, et al. Increased Wnt signaling during aging alters muscle stem cell fate and increases fibrosis. Science 2007;317:807-10.

20. Villeda SA, Plambeck KE, Middeldorp J, et al. Young blood reverses age-related impairments in cognitive function and synaptic plasticity in mice. Nat Med 2014;20:659-63.

21. Salpeter SJ, Khalaileh A, Weinberg-Corem N, Ziv O, Glaser B, Dor Y. Systemic regulation of the age-related decline of pancreatic β-cell replication. Diabetes 2013;62:2843-8.

22. Baht GS, Silkstone D, Vi L, et al. Exposure to a youthful circulaton rejuvenates bone repair through modulation of β-catenin. Nat Commun 2015;6:7131.

23. Elabd C, Cousin W, Upadhyayula P, et al. Oxytocin is an age-specific circulating hormone that is necessary for muscle maintenance and regeneration. Nat Commun 2014;5:4082.

24. Smith LK, He Y, Park JS, et al. β2-microglobulin is a systemic pro-aging factor that impairs cognitive function and neurogenesis. Nat Med 2015;21:932-7.

25. Bieri G, Schroer AB, Villeda SA. Blood-to-brain communication in aging and rejuvenation. Nat Neurosci 2023;26:379-93.

26. Hsiao YT, Shimizu I, Yoshida Y, Minamino T. Role of circulating molecules in age-related cardiovascular and metabolic disorders. Inflamm Regen 2022;42:2.

27. Pluvinage JV, Wyss-Coray T. Systemic factors as mediators of brain homeostasis, ageing and neurodegeneration. Nat Rev Neurosci 2020;21:93-102.

28. Rando TA, Jones DL. Regeneration, rejuvenation, and replacement: turning back the clock on tissue aging. Cold Spring Harb Perspect Biol 2021;13:a040907.

29. Egerman MA, Cadena SM, Gilbert JA, et al. GDF11 increases with age and inhibits skeletal muscle regeneration. Cell Metab 2015;22:164-74.

30. Peng L, Gagliano-Jucá T, Pencina KM, et al. Age trends in growth and differentiation factor-11 and myostatin levels in healthy men, and differential response to testosterone, measured using liquid chromatography-tandem mass spectrometry. J Gerontol A Biol Sci Med Sci 2022;77:763-9.

31. Smith SC, Zhang X, Zhang X, et al. GDF11 does not rescue aging-related pathological hypertrophy. Circ Res 2015;117:926-32.

32. Harper SC, Johnson J, Borghetti G, et al. GDF11 decreases pressure overload-induced hypertrophy, but can cause severe cachexia and premature death. Circ Res 2018;123:1220-31.

33. Hammers DW, Merscham-Banda M, Hsiao JY, Engst S, Hartman JJ, Sweeney HL. Supraphysiological levels of GDF11 induce striated muscle atrophy. EMBO Mol Med 2017;9:531-44.

34. Jones JE, Cadena SM, Gong C, et al. Supraphysiologic administration of GDF11 induces cachexia in part by upregulating GDF15. Cell Rep 2018;22:1522-30.

35. McPherron AC, Lawler AM, Lee SJ. Regulation of skeletal muscle mass in mice by a new TGF-beta superfamily member. Nature 1997;387:83-90.

36. McPherron AC, Lee SJ. Double muscling in cattle due to mutations in the myostatin gene. Proc Natl Acad Sci USA 1997;94:12457-61.

37. Mosher DS, Quignon P, Bustamante CD, et al. A mutation in the myostatin gene increases muscle mass and enhances racing performance in heterozygote dogs. PLoS Genet 2007;3:e79.

38. Medeiros EF, Phelps MP, Fuentes FD, Bradley TM. Overexpression of follistatin in trout stimulates increased muscling. Am J Physiol Regul Integr Comp Physiol 2009;297:R235-42.

39. McPherron AC, Lawler AM, Lee SJ. Regulation of anterior/posterior patterning of the axial skeleton by growth/differentiation factor 11. Nat Genet 1999;22:260-4.

40. McPherron AC, Huynh TV, Lee SJ. Redundancy of myostatin and growth/differentiation factor 11 function. BMC Dev Biol 2009;9:24.

41. Harmon EB, Apelqvist AA, Smart NG, Gu X, Osborne DH, Kim SK. GDF11 modulates NGN3+ islet progenitor cell number and promotes beta-cell differentiation in pancreas development. Development 2004;131:6163-74.

42. Liu JP. The function of growth/differentiation factor 11 (Gdf11) in rostrocaudal patterning of the developing spinal cord. Development 2006;133:2865-74.

43. Wu HH, Ivkovic S, Murray RC, et al. Autoregulation of neurogenesis by GDF11. Neuron 2003;37:197-207.

44. Kim J, Wu HH, Lander AD, Lyons KM, Matzuk MM, Calof AL. GDF11 controls the timing of progenitor cell competence in developing retina. Science 2005;308:1927-30.

45. Li Y, Choi WJ, Wei W, et al. Aging-associated changes in cerebral vasculature and blood flow as determined by quantitative optical coherence tomography angiography. Neurobiol Aging 2018;70:148-59.

46. Bullitt E, Zeng D, Mortamet B, et al. The effects of healthy aging on intracerebral blood vessels visualized by magnetic resonance angiography. Neurobiol Aging 2010;31:290-300.

47. Graff BJ, Payne SJ, El-Bouri WK. The ageing brain: investigating the role of age in changes to the human cerebral microvasculature with an in silico model. Front Aging Neurosci 2021;13:632521.

48. Knox EG, Aburto MR, Clarke G, Cryan JF, O'Driscoll CM. The blood-brain barrier in aging and neurodegeneration. Mol Psychiatry 2022;27:2659-73.

49. Chen MB, Yang AC, Yousef H, et al. Brain endothelial cells are exquisite sensors of age-related circulatory cues. Cell Rep 2020;30:4418-32.e4.

50. Ximerakis M, Lipnick SL, Innes BT, et al. Single-cell transcriptomic profiling of the aging mouse brain. Nat Neurosci 2019;22:1696-708.

51. Middeldorp J, Lehallier B, Villeda SA, et al. Preclinical assessment of young blood plasma for alzheimer disease. JAMA Neurol 2016;73:1325-33.

52. Villeda SA, Luo J, Mosher KI, et al. The ageing systemic milieu negatively regulates neurogenesis and cognitive function. Nature 2011;477:90-4.

53. Ximerakis M, Holton KM, Giadone RM, et al. Heterochronic parabiosis reprograms the mouse brain transcriptome by shifting aging signatures in multiple cell types. Nat Aging 2023;3:327-45.

54. He Y, Zhang H, Yung A, et al. ALK5-dependent TGF-β signaling is a major determinant of late-stage adult neurogenesis. Nat Neurosci 2014;17:943-52.

55. Shen Q, Goderie SK, Jin L, et al. Endothelial cells stimulate self-renewal and expand neurogenesis of neural stem cells. Science 2004;304:1338-40.

56. Kokovay E, Goderie S, Wang Y, et al. Adult SVZ lineage cells home to and leave the vascular niche via differential responses to SDF1/CXCR4 signaling. Cell Stem Cell 2010;7:163-73.

57. Kawauchi S, Kim J, Santos R, Wu HH, Lander AD, Calof AL. Foxg1 promotes olfactory neurogenesis by antagonizing Gdf11. Development 2009;136:1453-64.

58. Mayweather BA, Buchanan SM, Rubin LL. GDF11 expressed in the adult brain negatively regulates hippocampal neurogenesis. Mol Brain 2021;14:134.

59. Zhang W, Guo Y, Li B, et al. GDF11 rejuvenates cerebrovascular structure and function in an animal model of alzheimer's disease. J Alzheimers Dis 2018;62:807-19.

60. Ma J, Zhang L, Niu T, et al. Growth differentiation factor 11 improves neurobehavioral recovery and stimulates angiogenesis in rats subjected to cerebral ischemia/reperfusion. Brain Res Bull 2018;139:38-47.

61. Hudobenko J, Ganesh BP, Jiang J, et al. Growth differentiation factor-11 supplementation improves survival and promotes recovery after ischemic stroke in aged mice. Aging 2020;12:8049-66.

62. Walker RG, Czepnik M, Goebel EJ, et al. Structural basis for potency differences between GDF8 and GDF11. BMC Biol 2017;15:19.

63. Lian J, Walker RG, D'Amico A, et al. Functional substitutions of amino acids that differ between GDF11 and GDF8 impact skeletal development and skeletal muscle. Life Sci Alliance 2023;6:e202201662.

64. Lee SJ, Lehar A, Rydzik R, et al. Functional replacement of myostatin with GDF-11 in the germline of mice. Skelet Muscle 2022;12:7.

65. Rebbapragada A, Benchabane H, Wrana JL, Celeste AJ, Attisano L. Myostatin signals through a transforming growth factor β-like signaling pathway to block adipogenesis. Mol Cell Biol 2003;23:7230-42.

66. Walker RG, McCoy JC, Czepnik M, et al. Molecular characterization of latent GDF8 reveals mechanisms of activation. Proc Natl Acad Sci USA 2018;115:E866-75.

67. Le VQ, Iacob RE, Tian Y, et al. Tolloid cleavage activates latent GDF8 by priming the pro-complex for dissociation. EMBO J 2018;37:384-97.

68. McCoy JC, Goebel EJ, Thompson TB. Characterization of tolloid-mediated cleavage of the GDF8 procomplex. Biochem J 2021;478:1733-47.

69. Roh JD, Hobson R, Chaudhari V, et al. Activin type II receptor signaling in cardiac aging and heart failure. Sci Transl Med 2019;11:eaau8680.

70. Goff S, Hulmes DJS, Moali C. BMP-1/tolloid-like proteinases synchronize matrix assembly with growth factor activation to promote morphogenesis and tissue remodeling. Matrix Biol 2015;44-6:14-23.

71. Troilo H, Bayley CP, Barrett AL, Lockhart-Cairns MP, Jowitt TA, Baldock C. Mammalian tolloid proteinases: role in growth factor signalling. FEBS Lett 2016;590:2398-407.

72. Lee SJ, McPherron AC. Regulation of myostatin activity and muscle growth. Proc Natl Acad Sci USA 2001;98:9306-11.

73. Wolfman NM, McPherron AC, Pappano WN, et al. Activation of latent myostatin by the BMP-1/tolloid family of metalloproteinases. Proc Natl Acad Sci USA 2003;100:15842-6.

74. Walker RG, Poggioli T, Katsimpardi L, et al. Biochemistry and biology of GDF11 and myostatin similarities, differences, and questions for future investigation. Circ Res 2016;118:1125-41;discussion 1142.

75. Ge G, Hopkins DR, Ho WB, Greenspan DS. GDF11 forms a bone morphogenetic protein 1-activated latent complex that can modulate nerve growth factor-induced differentiation of PC12 cells. Mol Cell Biol 2005;25:5846-58.

76. Lee SJ. Genetic analysis of the role of proteolysis in the activation of latent myostatin. PLoS One 2008;3:e1628.

77. Cox TC, Lidral AC, McCoy JC, et al. Mutations in GDF11 and the extracellular antagonist, Follistatin, as a likely cause of Mendelian forms of orofacial clefting in humans. Hum Mutat 2019;40:1813-25.

78. Poggioli T, Vujic A, Yang P, et al. Circulating growth differentiation factor 11/8 levels decline with age. Circ Res 2016;118:29-37.

79. Ozek C, Krolewski RC, Buchanan SM, Rubin LL. Growth differentiation factor 11 treatment leads to neuronal and vascular improvements in the hippocampus of aged mice. Sci Rep 2018;8:17293.

80. Glass DJ. Elevated GDF11 is a risk factor for age-related frailty and disease in humans. Cell Metab 2016;24:7-8.

81. Rodgers BD. The immateriality of circulating GDF11. Circ Res 2016;118:1472-4.

82. Zhou Y, Sharma N, Dukes D, et al. GDF11 treatment attenuates the recovery of skeletal muscle function after injury in older rats. AAPS J 2017;19:431-7.

83. Olson KA, Beatty AL, Heidecker B, et al. Association of growth differentiation factor 11/8, putative anti-ageing factor, with cardiovascular outcomes and overall mortality in humans: analysis of the heart and soul and HUNT3 cohorts. Eur Heart J 2015;36:3426-34.

84. Hathout Y, Brody E, Clemens PR, et al. Large-scale serum protein biomarker discovery in Duchenne muscular dystrophy. Proc Natl Acad Sci USA 2015;112:7153-8.

85. Schafer MJ, Atkinson EJ, Vanderboom PM, et al. Quantification of GDF11 and myostatin in human aging and cardiovascular disease. Cell Metab 2016;23:1207-15.

86. Xu J, Gontier G, Chaker Z, Lacube P, Dupont J, Holzenberger M. Longevity effect of IGF-1R+/- mutation depends on genetic background-specific receptor activation. Aging Cell 2014;13:19-28.

87. Conover CA. PAPP-A: a new anti-aging target? Aging Cell 2010;9:942-6.

88. Mohammed I, Hollenberg MD, Ding H, Triggle CR. A critical review of the evidence that metformin is a putative anti-aging drug that enhances healthspan and extends lifespan. Front Endocrinol 2021;12:718942.

89. Blagosklonny MV. Cell senescence, rapamycin and hyperfunction theory of aging. Cell Cycle 2022;21:1456-67.

90. Rodgers BD, Eldridge JA. Reduced circulating GDF11 is unlikely responsible for age-dependent changes in mouse heart, muscle, and brain. Endocrinology 2015;156:3885-8.

91. Demontis F, Patel VK, Swindell WR, Perrimon N. Intertissue control of the nucleolus via a myokine-dependent longevity pathway. Cell Rep 2014;7:1481-94.

92. Du GQ, Shao ZB, Wu J, et al. Targeted myocardial delivery of GDF11 gene rejuvenates the aged mouse heart and enhances myocardial regeneration after ischemia-reperfusion injury. Basic Res Cardiol 2017;112:7.

93. Ma S, Upneja A, Galecki A, et al. Cell culture-based profiling across mammals reveals DNA repair and metabolism as determinants of species longevity. eLife 2016;5:e19130.

94. Duran J, Troncoso MF, Lagos D, Ramos S, Marin G, Estrada M. GDF11 modulates Ca2+-dependent Smad2/3 signaling to prevent cardiomyocyte hypertrophy. Int J Mol Sci 2018;19:1508.

95. Zhu HZ, Zhang LY, Zhai ME, et al. GDF11 Alleviates pathological myocardial remodeling in diabetic cardiomyopathy through SIRT1-dependent regulation of oxidative stress and apoptosis. Front Cell Dev Biol 2021;9:686848.

96. Zhang C, Wang Y, Ge Z, et al. GDF11 attenuated ANG II-induced hypertrophic cardiomyopathy and expression of ANP, BNP and Beta-MHC through down- regulating CCL11 in mice. Curr Mol Med 2018;18:661-71.

97. Garrido-Moreno V, Díaz-Vegas A, López-Crisosto C, et al. GDF-11 prevents cardiomyocyte hypertrophy by maintaining the sarcoplasmic reticulum-mitochondria communication. Pharmacol Res 2019;146:104273.

98. Amato AA, Sivakumar K, Goyal N, et al. Treatment of sporadic inclusion body myositis with bimagrumab. Neurology 2014;83:2239-46.

99. Lach-Trifilieff E, Minetti GC, Sheppard K, et al. An antibody blocking activin type II receptors induces strong skeletal muscle hypertrophy and protects from atrophy. Mol Cell Biol 2014;34:606-18.

100. Morvan F, Rondeau JM, Zou C, et al. Blockade of activin type II receptors with a dual anti-ActRIIA/IIB antibody is critical to promote maximal skeletal muscle hypertrophy. Proc Natl Acad Sci USA 2017;114:12448-53.

101. Polesskaya A, Seale P, Rudnicki MA. Wnt signaling induces the myogenic specification of resident CD45+ adult stem cells during muscle regeneration. Cell 2003;113:841-52.

102. Gayraud-Morel B, Chrétien F, Tajbakhsh S. Skeletal muscle as a paradigm for regenerative biology and medicine. Regen Med 2009;4:293-319.

The Journal of Cardiovascular Aging

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/