Proteomic basis of mortality resilience mediated by FOXO3 longevity genotype

Timothy A. Donlon1, Brian J. Morris1, Randi Chen1, Eunjung Lim2, Eric K Morgen3, Kristen Fortney3, Naisha Shah3, Kamal Masaki4, Bradley J. Willcox4
1Department of Research, NIH Center of Biomedical Research Excellence for Clinical and Translational Research on Aging, Kuakini Medical Center, Honolulu, Hawaii, 96817, USA
2Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
3BioAge Labs Inc., 1445A S 50th St, Richmond, California, USA
4Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA

Tóm tắt

AbstractFOXO3 is a ubiquitous transcription factor expressed in response to cellular stress caused by nutrient deprivation, inflammatory cytokines, reactive oxygen species, radiation, hypoxia, and other factors. We showed previously that the association of inherited FOXO3 variants with longevity was the result of partial protection against mortality risk posed by aging-related life-long stressors, particularly cardiometabolic disease. We then referred to the longevity-associated genotypes as conferring “mortality resilience.” Serum proteins whose levels change with aging and are associated with mortality risk may be considered as “stress proteins.” They may serve as indirect measures of life-long stress. Our aims were to (1) identify stress proteins that increase with aging and are associated with an increased risk of mortality, and (2) to determine if FOXO3 longevity/resilience genotype dampens the expected increase in mortality risk they pose. A total of 4500 serum protein aptamers were quantified using the Somalogic SomaScan proteomics platform in the current study of 975 men aged 71–83 years. Stress proteins associated with mortality were identified. We then used age-adjusted multivariable Cox models to investigate the interaction of stress protein with FOXO3 longevity-associated rs12212067 genotypes. For all the analyses, the p values were corrected for multiple comparisons by false discovery rate. This led to the identification of 44 stress proteins influencing the association of FOXO3 genotype with reduced mortality. Biological pathways were identified for these proteins. Our results suggest that the FOXO3 resilience genotype functions by reducing mortality in pathways related to innate immunity, bone morphogenetic protein signaling, leukocyte migration, and growth factor response.

Từ khóa


Tài liệu tham khảo

Schachter F, Fauredelanef L, Guenot F, et al. Genetic associations with human longevity at the APOE and ACE loci. Nature Genet. 1994;6(1):29–32.

Willcox BJ, Donlon TA, He Q, et al. FOXO3A genotype is strongly associated with human longevity. Proc Natl Acad Sci USA. 2008;105:13987–92.

Morris BJ, Willcox BJ, Donlon TA. Genetic and epigenetic regulation of human aging and longevity. Biochim Biophys Acta Mol Basis Dis. 2019;1865(7):1718–44.

Donlon TA, Morris BJ, Chen R, et al. FOXO3 longevity interactome on chromosome 6. Aging Cell. 2017;16(5):1016-25.

Chen R, Morris BJ, Donlon TA, et al. Foxo3 longevity genotype mitigates the increased mortality risk in men with a cardiometabolic disease. Aging. 2020;12(23):23509–24.

Cheng Z. The FoxO-autophagy axis in health and disease. Trends Endocrinol Metab. 2019;30(9):658–71.

Morris BJ, Willcox DC, Donlon TA, Willcox BJ. FOXO3: a major gene for human longevity - a mini-review. Gerontology. 2015;61(6):515-25.

Xin Z, Ma Z, Jiang S, et al. FOXOs in the impaired heart: new therapeutic targets for cardiac diseases. Biochim Biophys Acta Mol Basis Dis. 2017;1863(2):486–98.

Hariharan N, Ikeda Y, Hong C, et al. Autophagy plays an essential role in mediating regression of hypertrophy during unloading of the heart. PLoS One. 2013;8(1):e51632.

Cao DJ, Jiang N, Blagg A, et al. Mechanical unloading activates FoxO3 to trigger Bnip3-dependent cardiomyocyte atrophy. J Am Heart Assoc. 2013;2(2):e000016.

Wei X, Wu B, Zhao J, et al. Myocardial hypertrophic preconditioning attenuates cardiomyocyte hypertrophy and slows progression to heart failure through upregulation of S100A8/A9. Circulation. 2015;131(17):1506–17.

Sundaresan NR, Gupta M, Kim G, Rajamohan SB, Isbatan A, Gupta MP. Sirt3 blocks the cardiac hypertrophic response by augmenting Foxo3a-dependent antioxidant defense mechanisms in mice. J Clin Invest. 2009;119(9):2758–71.

Pajvani UB, Accili D. The new biology of diabetes. Diabetologia. 2015;58(11):2459–68.

Renault VM, Rafalski VA, Morgan AA, et al. FoxO3 regulates neural stem cell homeostasis. Cell Stem Cell. 2009;5:527–39.

Vidal RL, Figueroa A, Court FA, et al. Targeting the UPR transcription factor XBP1 protects against Huntington's disease through the regulation of FoxO1 and autophagy. Hum Mol Genet. 2012;21(10):2245–62.

Derisbourg MJ, Hartman MD, Denzel MS. Perspective: Modulating the integrated stress response to slow aging and ameliorate age-related pathology. Nat Aging. 2021;1(9):760–8.

Guerville F, De Souto BP, Ader I, et al. Revisiting the hallmarks of aging to identify markers of biological age. J Prev Alzheimers Dis. 2020;7(1):56–64.

Worth RM, Kagan A. Ascertainment of men of Japanese ancestry in Hawaii through World War II Selective Service registration. J Chronic Dis. 1970;23:389–97.

Kagan A, Harris BR, Winkelstein W Jr, et al. Epidemiologic studies of coronary heart disease and stroke in Japanese men living in Japan, Hawaii and California: demographic, physical, dietary and biochemical characteristics. J Chronic Dis. 1974;27:345–64.

Yano K, Reed DM, McGee DL. Ten-year incidence of coronary heart disease in the Honolulu Heart Program. Relationship to biologic and lifestyle characteristics. Am J Epidemiol. 1984;119:653–66.

Kagan A, editor. The Honolulu Heart Program: an epidemiological study of coronary heart disease and stroke. Amsterdam, The Netherlands: Harwood Academic Publishers; 1996.

Donlon TA, Curb JD, He Q, et al. FOXO3 gene variants and human aging: coding variants may not be key players. J Gerontol A Biol Sci Med Sci. 2012;67:1132–9.

Donlon TA, Willcox BJ, Morris BJ. FOXO3 cell resilience gene neighborhood. Aging. 2017; 9(12):2467-8.

Gold L, Ayers D, Bertino J, et al. Aptamer-based multiplexed proteomic technology for biomarker discovery. PLoS One. 2010;5(12):e15004.

Benjamini Y, Drai D, Elmer G, Kafkafi N, Golani I. Controlling the false discovery rate in behavior genetics research. Behav Brain Res. 2001;125:279–84.

Statistical Analysis System (SAS) version 9.4. SAS Institute, Cary, NC, USA. https://libguides.library.kent.edu/statconsulting/SAS. Accessed 22 Apr 2022

Stata 16. https://blog.stata.com/2019/06/26/stata-16-released/

Mostafavi S, Ray D, Warde-Farley D, Grouios C, Morris Q. GeneMANIA: a real-time multiple association network integration algorithm for predicting gene function. Genome Biol. 2008; 9(Suppl 1):S4.

Lee JC, Espéli M, Anderson CA, et al. Human SNP links differential outcomes in inflammatory and infectious disease to a FOXO3-regulated pathway. Cell. 2013;155:57–69.

Marlow G, Han DY, Triggs CM, Ferguson LR. Food intolerance: associations with the rs12212067 polymorphism of FOXO3 in Crohn's disease patients in New Zealand. J Nutrigenet Nutrigenomics. 2015;8(2):70–80.

Zhou YL, Wu WP, Cheng J, et al. CircFOXO3 rs12196996, a polymorphism at the gene flanking intron, is associated with circFOXO3 levels and the risk of coronary artery disease. Aging. 2020;12(13):13076-89.

Liu J, Liu D, Zhang X, et al. NELL2 modulates cell proliferation and apoptosis via ERK pathway in the development of benign prostatic hyperplasia. Clin Sci (Lond). 2021;135(13):1591–608.

Anusuya GS, Kandasamy M, Jacob Raja SA, Sabarinathan S, Ravishankar P, Kandhasamy B. Bone morphogenetic proteins: signaling periodontal bone regeneration and repair. J Pharm Bioallied Sci. 2016;8(Suppl 1):S39–s41.

Miyazono K, Kamiya Y, Morikawa M. Bone morphogenetic protein receptors and signal transduction. J Biochem. 2010;147(1):35–51.

Rochette L, Zeller M, Cottin Y, Vergely C. GDF15: an emerging modulator of immunity and a strategy in COVID-19 in association with iron metabolism. Trends Endocrinol Metab. 2021;32(11):875–89.

Luan HH, Wang A, Hilliard BK, et al. GDF15 is an inflammation-induced central mediator of tissue tolerance. Cell. 2019;178(5):1231–44.e11.

Wang D, Day EA, Townsend LK, Djordjevic D, Jørgensen SB, Steinberg GR. GDF15: emerging biology and therapeutic applications for obesity and cardiometabolic disease. Nat Rev Endocrinol. 2021;17(10):592–607.

Patel S, Alvarez-Guaita A, Melvin A, et al. GDF15 provides an endocrine signal of nutritional stress in mice and humans. Cell Metab. 2019;29(3):707–18.e8.

Kim KH, Lee MS. GDF15 as a central mediator for integrated stress response and a promising therapeutic molecule for metabolic disorders and NASH. Biochim Biophys Acta Gen Subj. 2021;1865(3):129834.

Conte M, Martucci M, Chiariello A, Franceschi C, Salvioli S. Mitochondria, immunosenescence and inflammaging: a role for mitokines? Semin Immunopathol. 2020;42(5):607–17.

Baek SJ, Eling T. Growth differentiation factor 15 (GDF15): a survival protein with therapeutic potential in metabolic diseases. Pharmacol Ther. 2019;198:46–58.

Gonçalves R, Maciel ÁCC, Rolland Y, Vellas B, de Souto BP. Frailty biomarkers under the perspective of geroscience: a narrative review. Ageing Res Rev. 2022;81:101737.

Nourshargh S, Alon R. Leukocyte migration into inflamed tissues. Immunity. 2014;41(5):694–707.

Korpos É, Deák F, Kiss I. Matrilin-2, an extracellular adaptor protein, is needed for the regeneration of muscle, nerve and other tissues. Neural Regen Res. 2015;10(6):866–9.

Zhou YH, Hu Y, Yu L, et al. Weaponizing human EGF-containing fibulin-like extracellular matrix protein 1 (EFEMP1) for 21(st) century cancer therapeutics. Oncoscience. 2016;3(7-8):208–19.

GenCards: the human gene database. Version 5.10. 2022 https://www.genecards.org

Imler JL, Hoffmann JA. Toll receptors in innate immunity. Trends Cell Biol. 2001;11(7):304–11.

Taghavi M, Khosravi A, Mortaz E, Nikaein D, Athari SS. Role of pathogen-associated molecular patterns (PAMPS) in immune responses to fungal infections. Eur J Pharmacol. 2017;808:8–13.

Tang D, Kang R, Coyne CB, Zeh HJ, Lotze MT. PAMPs and DAMPs: signal 0s that spur autophagy and immunity. Immunol Rev. 2012;249(1):158–75.

Mahla RS, Reddy MC, Prasad DV, Kumar H. Sweeten PAMPs: role of sugar complexed PAMPs in innate immunity and vaccine biology. Front Immunol. 2013;4:248.

Chen X, Liu S, Goraya MU, Maarouf M, Huang S, Chen JL. Host immune response to influenza A virus infection. Front Immunol. 2018;9:320.

Seiler F, Hellberg J, Lepper PM, et al. FOXO transcription factors regulate innate immune mechanisms in respiratory epithelial cells. J Immunol (Baltimore). 2013;190(4):1603–13.

Becker T, Loch G, Beyer M, et al. FOXO-dependent regulation of innate immune homeostasis. Nature. 2010;463:369–73.

McHugh DR, Koumis E, Jacob P, et al. DAF-16 and SMK-1 contribute to innate immunity during adulthood in Caenorhabditis elegans. G3 (Bethesda). 2020;10(5):1521–39.

Boehm AM, Khalturin K, Erxleben FA, et al. FoxO is a critical regulator of stem cell maintenance in immortal Hydra: Proc Natl Acad Sci USA. 2013;109:19697-19602, 2012.

Mah LJ, El-Osta A, Karagiannis TC. gammaH2AX: a sensitive molecular marker of DNA damage and repair. Leukemia. 2010;24(4):679–86.

Barretto EC, Polan DM, Beevor-Potts AN, Lee B, Grewal SS. Tolerance to hypoxia is promoted by FOXO regulation of the innate immunity transcription factor NF-κB/relish in Drosophila. Genetics. 2020;215(4):1013–25.

Alasiri G, Fan LY, Zona S, et al. ER stress and cancer: the FOXO forkhead transcription factor link. Mol Cell Endocrinol. 2018;462(Pt B):67–81.

Hughes CE, Nibbs RJB. A guide to chemokines and their receptors. FEBS J. 2018;285(16):2944–71.

Johansen KH, Golec DP, Thomsen JH, Schwartzberg PL, Okkenhaug K. PI3K in T cell adhesion and trafficking. Front Immunol. 2021;12:708908.

Kagan BL, Ganz T, Lehrer RI. Defensins: a family of antimicrobial and cytotoxic peptides. Toxicology. 1994;87(1-3):131–49.

Rice WG, Ganz T, Kinkade JM Jr, Selsted ME, Lehrer RI, Parmley RT. Defensin-rich dense granules of human neutrophils. Blood. 1987;70(3):757–65.

Lehrer RI, Lichtenstein AK, Ganz T. Defensins: antimicrobial and cytotoxic peptides of mammalian cells. Annu Rev Immunol. 1993;11:105–28.

Lee HJ, Kim JM, Kim KH, Heo JI, Kwak SJ, Han JA. Genotoxic stress/p53-induced DNAJB9 inhibits the pro-apoptotic function of p53. Cell Death Differ. 2015;22(1):86–95.

Kelley N, Jeltema D, Duan Y, He Y. The NLRP3 inflammasome: an overview of mechanisms of activation and regulation. Int J Mol Sci. 2019;20(13):3328.

Liu Y, Zhang W, Wu X, Gong J. Foxo3a-dependent Bim transcription protects mice from a high fat diet via inhibition of activation of the NLRP3 inflammasome by facilitating autophagy flux in Kupffer cells. Oncotarget. 2017;8(21):34258–67.

Lin L, Hron JD, Peng SL. Regulation of NF-κB, Th activation, and autoinflammation by the forkhead transcription factor Foxo3a. Immunity. 2004;21:203–13.

Zhou W, Cao Q, Peng Y, et al. FoxO4 inhibits NF-κB and protects mice against colonic injury and inflammation. Gastroenterology. 2009;137(4):1403–14.

Tominaga K. The emerging role of senescent cells in tissue homeostasis and pathophysiology. Pathobiol Aging Age Relat Dis. 2015;5:27743.

Qi XF, Chen ZY, Xia JB, et al. FoxO3a suppresses the senescence of cardiac microvascular endothelial cells by regulating the ROS-mediated cell cycle. J Mol Cell Cardiol. 2015;81:114–26.

Tokunaga R, Zhang W, Naseem M, et al. CXCL9, CXCL10, CXCL11/CXCR3 axis for immune activation – a target for novel cancer therapy. Cancer Treat Rev. 2018;63:40–7.

Menier C, Rouas-Freiss N, Favier B, LeMaoult J, Moreau P, Carosella ED. Recent advances on the non-classical major histocompatibility complex class I HLA-G molecule. Tissue Antigens. 2010;75(3):201–6.

Zhong L, Chen XF, Wang T, et al. Soluble TREM2 induces inflammatory responses and enhances microglial survival. J Exp Med. 2017;214(3):597–607.

Zhu Z, Zhang X, Dong W, et al. TREM2 suppresses the proinflammatory response to facilitate PRRSV infection via PI3K/NF-κB signaling. PLoS Pathog. 2020;16(5):e1008543.

Hay N. Interplay between FOXO, TOR, and Akt. Biochim Biophys Acta. 2011;1813(11):1965–70.

Roy SK, Srivastava RK, Shankar S. Inhibition of PI3K/AKT and MAPK/ERK pathways causes activation of FOXO transcription factor, leading to cell cycle arrest and apoptosis in pancreatic cancer. J Mol Signal. 2010;5:10.

Sun Y, Liu WZ, Liu T, Feng X, Yang N, Zhou HF. Signaling pathway of MAPK/ERK in cell proliferation, differentiation, migration, senescence and apoptosis. J Recept Signal Transduct Res. 2015;35(6):600–4.

Breit SN, Brown DA, Tsai VW. The GDF15-GFRAL pathway in health and metabolic disease: friend or foe? Annu Rev Physiol. 2021;83:127–51.

Namwanje M, Brown CW. Activins and inhibins: roles in development, physiology, and disease. Cold Spring Harb Perspect Biol. 2016;8(7).

Hung WT, Wu FJ, Wang CJ, Luo CW. DAN (NBL1) specifically antagonizes BMP2 and BMP4 and modulates the actions of GDF9, BMP2, and BMP4 in the rat ovary. Biol Reprod. 2012;86(5):1–9.

Yang X, Cao N, Chen L, Liu L, Zhang M, Cao Y. Suppression of cell tumorigenicity by non-neural pro-differentiation factors via inhibition of neural property in tumorigenic cells. Front Cell Dev Biol. 2021;9:714383.

Maquart FX, Monboisse JC. Extracellular matrix and wound healing. Pathol Biol (Paris). 2014;62(2):91–5.

Haines BP, Wheldon LM, Summerbell D, Heath JK, Rigby PW. Regulated expression of FLRT genes implies a functional role in the regulation of FGF signalling during mouse development. Dev Biol. 2006;297(1):14–25.

Kim KA, Kakitani M, Zhao J, et al. Mitogenic influence of human R-spondin1 on the intestinal epithelium. Science. 2005;309(5738):1256–9.

Wong VS, Yeung A, Schultz W, Brubaker PL. R-spondin-1 is a novel beta-cell growth factor and insulin secretagogue. J Biol Chem. 2010;285(28):21292–302.

Hoshino A, Kim HS, Bojmar L, et al. Extracellular vesicle and particle biomarkers define multiple human cancers. Cell. 2020;182(4):1044–61.e18.

Brazil DP, Church RH, Surae S, Godson C, Martin F. BMP signalling: agony and antagony in the family. Trends Cell Biol. 2015;25(5):249–64.

Koike N, Kassai Y, Kouta Y, Miwa H, Konishi M, Itoh N. Brorin, a novel secreted bone morphogenetic protein antagonist, promotes neurogenesis in mouse neural precursor cells. J Biol Chem. 2007;282(21):15843–50.

Sun M, Forsman C, Sergi C, Gopalakrishnan R, O'Connor MB, Petryk A. The expression of twisted gastrulation in postnatal mouse brain and functional implications. Neuroscience. 2010;169(2):920–31.

Murrow L, Debnath J. Autophagy as a stress-response and quality-control mechanism: implications for cell injury and human disease. Annu Rev Pathol. 2013;8:105–37.

Milan G, Romanello V, Pescatore F, et al. Regulation of autophagy and the ubiquitin-proteasome system by the FoxO transcriptional network during muscle atrophy. Nature Commun. 2015;6:6670.

Ortolan TG, Tongaonkar P, Lambertson D, Chen L, Schauber C, Madura K. The DNA repair protein rad23 is a negative regulator of multi-ubiquitin chain assembly. Nat Cell Biol. 2000;2(9):601–8.

Meusser B, Hirsch C, Jarosch E, Sommer T. ERAD: the long road to destruction. Nat Cell Biol. 2005;7(8):766–72.

Can ND, Basturk E, Kizilboga T, et al. Interactome analysis of Bag-1 isoforms reveals novel interaction partners in endoplasmic reticulum-associated degradation. PLoS One. 2021;16(8):e0256640.

D'Arcy MS. Cell death: a review of the major forms of apoptosis, necrosis and autophagy. Cell Biol Int. 2019;43(6):582–92.

Grodzicky T, Elkon KB. Apoptosis: a case where too much or too little can lead to autoimmunity. Mt Sinai J Med. 2002;69(4):208–19.

Mortezaee K, Salehi E, Mirtavoos-Mahyari H, et al. Mechanisms of apoptosis modulation by curcumin: implications for cancer therapy. J Cell Physiol. 2019;234(8):12537–50.

Kurowska M, Rudnicka W, Kontny E, et al. Fibroblast-like synoviocytes from rheumatoid arthritis patients express functional IL-15 receptor complex: endogenous IL-15 in autocrine fashion enhances cell proliferation and expression of Bcl-x(L) and Bcl-2. J Immunol (Baltimore). 2002;169(4):1760–7.

Liang R, Ghaffari S. Stem cells seen through the FOXO lens: an evolving paradigm. Curr Top Dev Biol. 2018;127:23–47.

Kops GJ, Dansen TB, Polderman PE, et al. Forkhead transcription factor FOXO3a protects quiescent cells from oxidative stress. Nature. 2002;419:316–21.

Sengupta A, Molkentin JD, Paik JH, DePinho RA, Yutzey KE. FoxO transcription factors promote cardiomyocyte survival upon induction of oxidative stress. J Biol Chem. 2011;286:7468–78.

Webb AE, Brunet A. FOXO transcription factors: key regulators of cellular quality control. Trends Biochem Sci. 2014;39:159–69.

Fasano C, Disciglio V, Bertora S, Lepore Signorile M, Simone C. FOXO3a from the nucleus to the mitochondria: a round trip in cellular stress response. Cells. 2019;8(9):1110.

Gurkar AU, Robinson AR, Cui Y, et al. Dysregulation of DAF-16/FOXO3A-mediated stress responses accelerates oxidative DNA damage induced aging. Redox Biol. 2018;18:191–9.

Andrade J, Shi C, Costa ASH, et al. Control of endothelial quiescence by FOXO-regulated metabolites. Nat Cell Biol. 2021;23(4):413–23.

Warr MR, Binnewies M, Flach J, et al. FOXO3A directs a protective autophagy program in haematopoietic stem cells. Nature. 2013;494:323–7.

Santo EE, Paik J. FOXO in neural cells and diseases of the nervous system. Curr Top Dev Biol. 2018;127:105–18.

Gopinath SD, Webb AE, Brunet A, Rando TA. FOXO3 promotes quiescence in adult muscle stem cells during the process of self-renewal. Stem Cell Reports. 2014;2(4):414–26.

Sathyan S, Ayers E, Gao T, et al. Plasma proteomic profile of age, health span, and all-cause mortality in older adults. Aging Cell. 2020;19(11):e13250.

Tanaka T, Basisty N, Fantoni G, et al. Plasma proteomic biomarker signature of age predicts health and life span. eLife. 2020:9.

Eiriksdottir T, Ardal S, Jonsson BA, et al. Predicting the probability of death using proteomics. Commun Biol. 2021;4(1):758.

Frankum R, Jameson TSO, Knight BA, et al. Extreme longevity variants at the FOXO3 locus may moderate FOXO3 isoform levels. GeroScience. 2021.

Nguetse CN, Kremsner PG, Velavan TP. FOXO3A regulatory polymorphism and susceptibility to severe malaria in Gabonese children. Immunogenetics. 2015;67(2):67–71.

Alonso A, Domènech E, Julià A, et al. Identification of risk loci for Crohn's disease phenotypes using a genome-wide association study. Gastroenterology. 2015;148(4):794–805.