Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

An inflammatory aging clock (iAge) based on deep learning tracks multimorbidity, immunosenescence, frailty and cardiovascular aging

An Author Correction to this article was published on 26 July 2021

This article has been updated

Abstract

While many diseases of aging have been linked to the immunological system, immune metrics capable of identifying the most at-risk individuals are lacking. From the blood immunome of 1,001 individuals aged 8–96 years, we developed a deep-learning method based on patterns of systemic age-related inflammation. The resulting inflammatory clock of aging (iAge) tracked with multimorbidity, immunosenescence, frailty and cardiovascular aging, and is also associated with exceptional longevity in centenarians. The strongest contributor to iAge was the chemokine CXCL9, which was involved in cardiac aging, adverse cardiac remodeling and poor vascular function. Furthermore, aging endothelial cells in human and mice show loss of function, cellular senescence and hallmark phenotypes of arterial stiffness, all of which are reversed by silencing CXCL9. In conclusion, we identify a key role of CXCL9 in age-related chronic inflammation and derive a metric for multimorbidity that can be utilized for the early detection of age-related clinical phenotypes.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: The inflammatory clock of aging tracks with multimorbidity, frailty and exceptional longevity.
Fig. 2: The inflammatory clock of aging correlates with immunosenescence.
Fig. 3: CXCL9 is a major contributor to iAge.
Fig. 4: CXCL9 is an important regulator of endothelial cell aging.
Fig. 5: Early cellular senescence and loss of angiogenesis capacity in iPSC-derived aging endothelia is reversed by silencing CXCL9.
Fig. 6: CXCL9 promotes a vascular stiffness gene expression signature in the aging endothelium and impairs endothelial function.
Fig. 7: CXCL9 regulates endothelial cell senescence and capillary network formation in vivo.

Similar content being viewed by others

Data availability

The cell subpopulation, immune protein and cell signaling data for the Stanford Aging and Vaccination studies are publicly available on ImmPort Bioinformatics Repository (http://www.immport.org/immport-open/public/home/home) under the following study IDs SDY311 (cytokines, phosphoflow assays and CyTOF surface phenotyping), SDY312 (cytokines, phosphoflow assays and flow cytometry surface phenotyping), SDY314 (flow cytometry surface phenotyping), SDY315 (cytokines, phosphoflow assays and CyTOF surface phenotyping) and SDY478 (cytokines and CyTOF surface phenotyping). The gene expression data utilized in this study to compute gene expression-iAge has been uploaded to the Gene Expression Omnibus under accession number GSE168753. Our study complies in full with the STROBE statement, STARD guidelines and GATHER statement.

Code availability

The code used for the identification of immunotypes and construction of the inflammatory clock has been deposited on GitHub (https://github.com/) and is available under: https://github.com/clingsz/GAE. For the immunological characterization of immunotypes, we used R programming (https://www.r-project.org/). The LASSO and Elastic Net regularized generalized linear models package (glmnet) for R programming can be found at: https://cran.r-project.org/web/packages/glmnet/index.html. Maximum likelihood estimation is a function of the STATS4 R package found at: https://www.rdocumentation.org/packages/stats4/versions/3.4.1.

Change history

References

  1. Kotas, M. E. & Medzhitov, R. Homeostasis, inflammation, and disease susceptibility. Cell 160, 816–827 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Liu, F. et al. Mechanosignaling through YAP and TAZ drives fibroblast activation and fibrosis. Am. J. Physiol. Lung Cell Mol. Physiol. 308, L344–L357 (2015).

    Article  CAS  PubMed  Google Scholar 

  3. Crusz, S. M. & Balkwill, F. R. Inflammation and cancer: advances and new agents. Nat. Rev. Clin. Oncol. 12, 584–596 (2015).

    Article  CAS  PubMed  Google Scholar 

  4. Franceschi, C. & Campisi, J. Chronic inflammation (inflammaging) and its potential contribution to age-associated diseases. J. Gerontol. A Biol. Sci. Med. Sci. 69, S4–S9 (2014).

    Article  PubMed  Google Scholar 

  5. Furman, D. et al. Expression of specific inflammasome gene modules stratifies older individuals into two extreme clinical and immunological states. Nat. Med. 23, 174–184 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. López-Otín, C., Blasco, M. A., Partridge, L., Serrano, M. & Kroemer, G. The hallmarks of aging. Cell 153, 1194–1217 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Cavadas, C., Aveleira, C. A., Souza, G. F. P. & Velloso, L. A. The pathophysiology of defective proteostasis in the hypothalamus — from obesity to ageing. Nat. Rev. Endocrinol. 12, 723–733 (2016).

    Article  CAS  PubMed  Google Scholar 

  8. Efeyan, A., Comb, W. C. & Sabatini, D. M. Nutrient-sensing mechanisms and pathways. Nature 517, 302–310 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Grivennikov, S. I., Greten, F. R. & Karin, M. Immunity, inflammation, and cancer. Cell 140, 883–899 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Hunter, R. L. et al. Inflammation induces mitochondrial dysfunction and dopaminergic neurodegeneration in the nigrostriatal system. J. Neurochem. 100, 1375–1386 (2007).

    Article  CAS  PubMed  Google Scholar 

  11. Jurk, D. et al. Chronic inflammation induces telomere dysfunction and accelerates ageing in mice. Nat. Commun. 5, 4172 (2014).

    Article  CAS  Google Scholar 

  12. Lasry, A. & Ben-Neriah, Y. Senescence-associated inflammatory responses: aging and cancer perspectives. Trends Immunol. 36, 217–228 (2015).

    Article  CAS  PubMed  Google Scholar 

  13. Nathan, C. & Cunningham-Bussel, A. Beyond oxidative stress: an immunologist’s guide to reactive oxygen species. Nat. Rev. Immunol. 13, 349–361 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Oh, J., Lee, Y. D. & Wagers, A. J. Stem cell aging: mechanisms, regulators and therapeutic opportunities. Nat. Med. 20, 870–880 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Thevaranjan, N. et al. Age-associated microbial dysbiosis promotes intestinal permeability, systemic inflammation, and macrophage dysfunction. Cell Host Microbe 21, 455–466 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Alpert, A. et al. A clinically meaningful metric of immune age derived from high-dimensional longitudinal monitoring. Nat. Med. 25, 487–495 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Goldberg, E. L. & Dixit, V. D. Drivers of age-related inflammation and strategies for healthspan extension. Immunol. Rev. 265, 63–74 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Franceschi, C., Garagnani, P., Vitale, G., Capri, M. & Salvioli, S. Inflammaging and garb-aging. Trends Endocrinol. Metab. 28, 199–212 (2017).

    Article  CAS  PubMed  Google Scholar 

  19. Morrisette-Thomas, V. et al. Inflamm-aging does not simply reflect increases in pro-inflammatory markers. Mechanisms Ageing Dev. 139, 49–57 (2014).

    Article  CAS  Google Scholar 

  20. Furman, D. et al. Chronic inflammation in the etiology of disease across the life span. Nat. Med. 25, 1822–1832 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Blazkova, J. et al. Multicenter systems analysis of human blood reveals immature neutrophils in males and during pregnancy. J. Immunol. 198, 2479–2488 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Brodin, P. et al. Variation in the human immune system is largely driven by non-heritable influences. Cell 160, 37–47 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Furman, D. et al. Systems analysis of sex differences reveals an immunosuppressive role for testosterone in the response to influenza vaccination. Proc. Natl Acad. Sci. USA 111, 869–874 (2014).

    Article  CAS  PubMed  Google Scholar 

  24. Furman, D. et al. Apoptosis and other immune biomarkers predict influenza vaccine responsiveness. Mol. Syst. Biol. 9, 659 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Furman, D. et al. Cytomegalovirus infection enhances the immune response to influenza. Sci. Transl. Med. 7, 281ra43 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Price, J. V. et al. Characterization of influenza vaccine immunogenicity using influenza antigen microarrays. PLoS ONE 8, e64555 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Roskin, K. M. et al. IgH sequences in common variable immune deficiency reveal altered B cell development and selection. Sci. Transl. Med. 7, 302ra135 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Shen-Orr Shai, S. et al. Defective signaling in the JAK–STAT pathway tracks with chronic inflammation and cardiovascular risk in aging humans. Cell Syst. 3, 374–384 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Wang, C. et al. Effects of aging, cytomegalovirus infection, and EBV infection on human B cell repertoires. J. Immunol. 192, 603–611 (2014).

    Article  CAS  PubMed  Google Scholar 

  30. Montoya, J. G. et al. Cytokine signature associated with disease severity in chronic fatigue syndrome patients. Proc. Natl Acad. Sci. USA 114, E7150–E7158 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Maecker, H. T. et al. New tools for classification and monitoring of autoimmune diseases. Nat. Rev. Rheumatol. 8, 317–328 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Ridker, P. M. et al. Antiinflammatory therapy with canakinumab for atherosclerotic disease. N. Engl. J. Med. 377, 1119–1131 (2017).

    Article  CAS  PubMed  Google Scholar 

  33. Barnett, K. et al. Epidemiology of multimorbidity and implications for health care, research, and medical education: a cross-sectional study. Lancet 380, 37–43 (2012).

    Article  PubMed  Google Scholar 

  34. Zou, H. & Hastie, T. Regularization and variable selection via the Elastic Net. J. R. Stat. Soc. B Stat. Methodol. 67, 301–320 (2005).

    Article  Google Scholar 

  35. Rockwood, K., Rockwood, M. R. & Mitnitski, A. Physiological redundancy in older adults in relation to the change with age in the slope of a frailty index. J. Am. Geriatr. Soc. 58, 318–323 (2010).

    Article  PubMed  Google Scholar 

  36. Mahmood, S. S., Levy, D., Vasan, R. S. & Wang, T. J. The Framingham Heart Study and the epidemiology of cardiovascular disease: a historical perspective. Lancet 383, 999–1008 (2014).

    Article  PubMed  Google Scholar 

  37. Bertsch, T. et al. C-reactive protein and the acute phase reaction in geriatric patients. Z. Gerontologie Geriatr. 48, 595–600 (2015).

    Article  Google Scholar 

  38. Scheller, J., Chalaris, A., Schmidt-Arras, D. & Rose-John, S. The pro- and anti-inflammatory properties of the cytokine interleukin-6. Biochim. Biophys. Acta 1813, 878–888 (2011).

    Article  CAS  PubMed  Google Scholar 

  39. Krutzik, P. O., Irish, J. M., Nolan, G. P. & Perez, O. D. Analysis of protein phosphorylation and cellular signaling events by flow cytometry: techniques and clinical applications. Clin. Immunol. 110, 206–221 (2004).

    Article  CAS  PubMed  Google Scholar 

  40. Kisseleva, T., Bhattacharya, S., Braunstein, J. & Schindler, C. W. Signaling through the JAK/STAT pathway, recent advances and future challenges. Gene 285, 1–24 (2002).

    Article  CAS  PubMed  Google Scholar 

  41. Altara, R. et al. Left ventricular dysfunction and CXCR3 ligands in hypertension: from animal experiments to a population-based pilot study. PLoS ONE 10, e0141394 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Altara, R. et al. CXCL10 is a circulating inflammatory marker in patients with advanced heart failure: a pilot study. J. Cardiovasc. Transl. Res. 9, 302–314 (2016).

    Article  PubMed  Google Scholar 

  43. Altara, R., Mallat, Z., Booz, G. W. & Zouein, F. A. The CXCL10/CXCR3 axis and cardiac inflammation: implications for immunotherapy to treat infectious and noninfectious diseases of the heart. J. Immunol. Res. 2016, 4396368 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Hardison, J. L., Wrightsman, R. A., Carpenter, P. M., Lane, T. E. & Manning, J. E. The chemokines CXCL9 and CXCL10 promote a protective immune response but do not contribute to cardiac inflammation following infection with Trypanosoma cruzi. Infect. Immun. 74, 125–134 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Vlachopoulos, C., Aznaouridis, K. & Stefanadis, C. Prediction of cardiovascular events and all-cause mortality with arterial stiffness: a systematic review and meta-analysis. J. Am. Coll. Cardiol. 55, 1318–1327 (2010).

    Article  PubMed  Google Scholar 

  46. Ben-Shlomo, Y. et al. Aortic pulse wave velocity improves cardiovascular event prediction: an individual participant meta-analysis of prospective observational data from 17,635 subjects. J. Am. Coll. Cardiol. 63, 636–646 (2014).

    Article  PubMed  Google Scholar 

  47. Chirinos, J. A. Large artery stiffness, microvascular function, and cardiovascular risk. Circ. Cardiovasc Imaging 9, e005903 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Figueiredo, V. N. et al. Vascular stiffness and endothelial dysfunction: correlations at different levels of blood pressure. Blood Press. 21, 31–38 (2012).

    Article  CAS  PubMed  Google Scholar 

  49. Vanhoutte, P. M., Feletou, M. & Taddei, S. Endothelium-dependent contractions in hypertension. Br. J. Pharmacol. 144, 449–458 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Castellon, X. & Bogdanova, V. Chronic inflammatory diseases and endothelial dysfunction. Aging Dis. 7, 81–89 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Harvey, A., Montezano, A. C. & Touyz, R. M. Vascular biology of ageing: implications in hypertension. J. Mol. Cell Cardiol. 83, 112–121 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Kamo, T., Akazawa, H. & Komuro, I. Cardiac nonmyocytes in the hub of cardiac hypertrophy. Circ. Res. 117, 89–98 (2015).

    Article  CAS  PubMed  Google Scholar 

  53. Arnaoutova, I. & Kleinman, H. K. In vitro angiogenesis: endothelial cell tube formation on gelled basement membrane extract. Nat. Protoc. 5, 628–635 (2010).

    Article  CAS  PubMed  Google Scholar 

  54. Mukai, N. et al. A comparison of the tube forming potentials of early and late endothelial progenitor cells Exp. Cell. Res. 314, 430–440 (2008).

    Article  CAS  PubMed  Google Scholar 

  55. Tousoulis, D., Kampoli, A. M., Tentolouris, C., Papageorgiou, N. & Stefanadis, C. The role of nitric oxide on endothelial function. Curr. Vasc. Pharmacol. 10, 4–18 (2012).

    Article  CAS  PubMed  Google Scholar 

  56. Voyta, J. C., Via, D. P., Butterfield, C. E. & Zetter, B. R. Identification and isolation of endothelial cells based on their increased uptake of acetylated low-density lipoprotein. J. Cell Biol. 99, 2034–2040 (1984).

    Article  CAS  PubMed  Google Scholar 

  57. Boisen, L., Drasbek, K. R., Pedersen, A. S. & Kristensen, P. Evaluation of endothelial cell culture as a model system of vascular ageing. Exp. Gerontol. 45, 779–787 (2010).

    Article  CAS  PubMed  Google Scholar 

  58. Brandes, R. P., Fleming, I. & Busse, R. Endothelial aging. Cardiovascular Res. 66, 286–294 (2005).

    Article  CAS  Google Scholar 

  59. Takahashi, K. & Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663–676 (2006).

    Article  CAS  PubMed  Google Scholar 

  60. Sayed, N. et al. Clinical trial in a dish using iPSCs shows lovastatin improves endothelial dysfunction and cellular cross-talk in LMNA cardiomyopathy. Sci. Transl. Med. 12, eaax9276 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Rivard, A. et al. Age-Dependent Impairment of Angiogenesis. Circulation 99, 111–120 (1999).

    Article  CAS  PubMed  Google Scholar 

  62. Minamino, T. & Komuro, I. Vascular cell senescence: contribution to atherosclerosis. Circ. Res. 100, 15–26 (2007).

    Article  CAS  PubMed  Google Scholar 

  63. Yin, H. & Pickering, J. G. Cellular senescence and vascular disease: novel routes to better understanding and therapy. Can. J. Cardiol. 32, 612–623 (2016).

    Article  PubMed  Google Scholar 

  64. Uryga, A. K. & Bennett, M. R. Ageing induced vascular smooth muscle cell senescence in atherosclerosis. J. Physiol. 594, 2115–2124 (2016).

    Article  CAS  PubMed  Google Scholar 

  65. Jia, G., Aroor, A. R., Jia, C. & Sowers, J. R. Endothelial cell senescence in aging-related vascular dysfunction. Biochim. Biophys. Acta, Mol. Basis Dis. 1865, 1802–1809 (2019).

    Article  CAS  Google Scholar 

  66. Stojanovic, S. D., Fiedler, J., Bauersachs, J., Thum, T. & Sedding, D. G. Senescence-induced inflammation: an important player and key therapeutic target in atherosclerosis. Eur. Heart J. 41, 2983–2996 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Zieman, S. J., Melenovsky, V. & Kass, D. A. Mechanisms, pathophysiology, and therapy of arterial stiffness. Arter. Thromb. Vasc. Biol. 25, 932–943 (2005).

    Article  CAS  Google Scholar 

  68. Kohn, J. C., Lampi, M. C. & Reinhart-King, C. A. Age-related vascular stiffening: causes and consequences. Front. Genet. 6, 112 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  69. Lu, P., Takai, K., Weaver, V. M. & Werb, Z. Extracellular matrix degradation and remodeling in development and disease. Cold Spring Harb. Perspect. Biol. 3, a005082 (2011).

    Article  Google Scholar 

  70. Kuzuya, M. et al. Glycation cross-links inhibit matrix metalloproteinase-2 activation in vascular smooth muscle cells cultured on collagen lattice. Diabetologia 44, 433–436 (2001).

    Article  CAS  PubMed  Google Scholar 

  71. González-Amaro, R., Diaz-González, F. & Sánchez-Madrid, F. Adhesion molecules in inflammatory diseases. Drugs 56, 977–988 (1998).

    Article  PubMed  Google Scholar 

  72. Khanamiri, S. et al. Marked vascular dysfunction in a case of peripartum cardiomyopathy. J. Vasc. Res. 56, 11–15 (2019).

    Article  CAS  PubMed  Google Scholar 

  73. Sayed, N. et al. Transdifferentiation of human fibroblasts to endothelial cells: role of innate immunity. Circulation 131, 300–309 (2015).

    Article  CAS  PubMed  Google Scholar 

  74. Hannum, G. et al. Genome-wide methylation profiles reveal quantitative views of human aging rates. Mol. Cell. 49, 359–367 (2013).

    Article  CAS  PubMed  Google Scholar 

  75. Horvath, S. DNA methylation age of human tissues and cell types. Genome Biol. 14, R115 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  76. Holly, A. C. et al. Towards a gene expression biomarker set for human biological age. Aging Cell 12, 324–326 (2013).

    Article  CAS  PubMed  Google Scholar 

  77. Ishikawa, M. et al. Plasma and serum lipidomics of healthy white adults shows characteristic profiles by subjects’ gender and age. PLoS ONE 9, e91806 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  78. Yu, Z. et al. Human serum metabolic profiles are age dependent. Aging Cell. 11, 960–967 (2012).

    Article  CAS  PubMed  Google Scholar 

  79. Menni, C. et al. Metabolomic markers reveal novel pathways of ageing and early development in human populations. Int J. Epidemiol. 42, 1111–1119 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  80. Hertel, J. et al. Measuring biological age via metabonomics: the metabolic age score. J. Proteome Res. 15, 400–410 (2016).

    Article  CAS  PubMed  Google Scholar 

  81. Collino, S. et al. Metabolic signatures of extreme longevity in northern Italian centenarians reveal a complex remodeling of lipids, amino acids, and gut microbiota metabolism. PLoS ONE 8, e56564 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Lawton, K. A. et al. Analysis of the adult human plasma metabolome. Pharmacogenomics 9, 383–397 (2008).

    Article  CAS  PubMed  Google Scholar 

  83. Rahman, S. A. & Adjeroh, D. A. Deep learning using convolutional LSTM estimates biological age from physical activity. Sci. Rep. 9, 11425 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  84. Tang X., Wang Z., Luo W. and Gao S. Face aging with identity-preserved conditional generative adversarial networks. Proc. IEEE/CVF Conf. Computer Vision Pattern Recognition https://doi.org/10.1109/CVPR.2018.00828 (2018).

  85. Zhang K. et al. Fine-grained age estimation in the wild with attention LSTM networks. IEEE Trans. Circuits Systems Video Technol. https://doi.org/10.1109/TCSVT.2019.2936410 (2019).

  86. Putin, E. et al. Deep biomarkers of human aging: application of deep neural networks to biomarker development. Aging 8, 1021–1033 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Mamoshina, P. et al. Population specific biomarkers of human aging: a big data study using South Korean, Canadian, and Eastern European patient populations. J. Gerontol. A Biol. Sci. Med Sci. 73, 1482–1490 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  88. Subramanian, N., Torabi-Parizi, P., Gottschalk, R. A., Germain, R. N. & Dutta, B. Network representations of immune system complexity. WIREs Syst. Biol. Med. 7, 13–38 (2015).

    Article  CAS  Google Scholar 

  89. Bengio, Y. Learning deep architectures for AI. Found. Trends Mach. Learn. 2, 1–127 (2009).

    Article  Google Scholar 

  90. Tanaka, T. et al. Plasma proteomic signature of age in healthy humans. Aging Cell. 17, e12799 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  91. Fourati, S. et al. Pre-vaccination inflammation and B-cell signalling predict age-related hyporesponse to hepatitis B vaccination. Nat. Commun. 7, 10369 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Lehallier, B. et al. Undulating changes in human plasma proteome profiles across the lifespan. Nat. Med. 25, 1843–1850 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Bonfante, H. L. et al. CCL2, CXCL8, CXCL9 and CXCL10 serum levels increase with age but are not altered by treatment with hydroxychloroquine in patients with osteoarthritis of the knees. Int J. Rheum. Dis. 20, 1958–1964 (2017).

    Article  CAS  PubMed  Google Scholar 

  94. Roubenoff, R. et al. Monocyte cytokine production in an elderly population: effect of age and inflammation. J. Gerontol. A Biol. Sci. Med Sci. 53, M20–M26 (1998).

    Article  CAS  PubMed  Google Scholar 

  95. Torres, K. C. L. et al. Immune senescence and biomarkers profile of Bambui aged population-based cohort. Exp. Gerontol. 103, 47–56 (2018).

    Article  CAS  PubMed  Google Scholar 

  96. Shurin, G. V. et al. Dynamic alteration of soluble serum biomarkers in healthy aging. Cytokine 39, 123–129 (2007).

    Article  CAS  PubMed  Google Scholar 

  97. Zheng, Y. et al. Age-related pro-inflammatory and pro-angiogenic changes in human aqueous humor. Int J. Ophthalmol. 11, 196–200 (2018).

    PubMed  PubMed Central  Google Scholar 

  98. Sandovici, I. et al. Ageing is associated with molecular signatures of inflammation and type 2 diabetes in rat pancreatic islets. Diabetologia 59, 502–511 (2016).

    Article  CAS  PubMed  Google Scholar 

  99. de Araujo, F. F. et al. CXCL9 and CXCL10 display an age-dependent profile in Chagas patients: a cohort study of aging in Bambui, Brazil. Infect. Dis. Poverty 9, 51 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  100. Zhou, L. et al. Age-specific changes in the molecular phenotype of patients with moderate-to-severe atopic dermatitis. J. Allergy Clin. Immunol. 144, 144–156 (2019).

    Article  CAS  PubMed  Google Scholar 

  101. Amorim, J. S. C. et al. Inflammatory markers and occurrence of falls: Bambui cohort study of aging. Rev. Saude Publica 53, 35 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  102. Chiu, B. C. et al. Differential effects of ageing on cytokine and chemokine responses during type-1 (mycobacterial) and type-2 (schistosomal) pulmonary granulomatous inflammation in mice. Mech. Ageing Dev. 123, 313–326 (2002).

    Article  CAS  PubMed  Google Scholar 

  103. Janowski, A. M. et al. NLRC4 suppresses melanoma tumor progression independently of inflammasome activation. J. Clin. Investig. 126, 3917–3928 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  104. Versari, D., Daghini, E., Virdis, A., Ghiadoni, L. & Taddei, S. Endothelial dysfunction as a target for prevention of cardiovascular disease. Diabetes Care 32, S314–S321 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Mezzaroma, E. et al. The inflammasome promotes adverse cardiac remodeling following acute myocardial infarction in the mouse. Proc. Natl Acad. Sci. USA 108, 19725–19730 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Nevers, T. et al. Left ventricular T-cell recruitment contributes to the pathogenesis of heart failure. Circulation Heart Fail. 8, 776–787 (2015).

    Article  CAS  Google Scholar 

  107. Rawlings, J. S., Rosler, K. M. & Harrison, D. A. The JAK/STAT signaling pathway. J. Cell Sci. 117, 1281–1283 (2004).

    Article  CAS  PubMed  Google Scholar 

  108. Shuai, K. & Liu, B. Regulation of gene-activation pathways by PIAS proteins in the immune system. Nat. Rev. Immunol. 5, 593–605 (2005).

    Article  CAS  PubMed  Google Scholar 

  109. van Boxel-Dezaire, A. H. H., Rani, M. R. S. & Stark, G. R. Complex modulation of cell type-specific signaling in response to type I interferons. Immunity 25, 361–372 (2006).

    Article  PubMed  Google Scholar 

  110. Matsa, E. et al. Transcriptome profiling of patient-specific human iPSC-cardiomyocytes predicts individual drug safety and efficacy responses in vitro. Cell Stem Cell 19, 311–325 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Gu, M. et al. Patient-specific iPSC-derived endothelial cells uncover pathways that protect against pulmonary hypertension in BMPR2 mutation carriers. Cell Stem Cell 20, 490–504 (2017).

    Article  CAS  PubMed  Google Scholar 

  112. Lian, X. et al. Robust cardiomyocyte differentiation from human pluripotent stem cells via temporal modulation of canonical Wnt signaling. Proc. Natl Acad. Sci. USA 109, E1848–E1857 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Nagueh, S. F. et al. Recommendations for the evaluation of left ventricular diastolic function by echocardiography. Eur. J. Echocardiogr. 10, 165–193 (2009).

    Article  PubMed  Google Scholar 

  114. Redfield, M. M., Jacobsen, S. J., Borlaug, B. A., Rodeheffer, R. J. & Kass, D. A. Age- and gender-related ventricular-vascular stiffening: a community-based study. Circulation 112, 2254–2262 (2005).

    Article  PubMed  Google Scholar 

  115. Lang, R. M. et al. Chamber Quantification Writing G, American Society of Echocardiography’s G, Standards C and European Association of E. Recommendations for chamber quantification: a report from the American Society of Echocardiography’s Guidelines and Standards Committee and the Chamber Quantification Writing Group, developed in conjunction with the European Association of Echocardiography, a branch of the European Society of Cardiology. J. Am. Soc. Echocardiogr. 18, 1440–1463 (2005).

    Article  PubMed  Google Scholar 

  116. Godec, J. et al. Compendium of immune signatures identifies conserved and species-specific biology in response to inflammation. Immunity 44, 194–206 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Friedman, J., Hastie, T. & Tibshirani, R. Regularization paths for generalized linear models via coordinate descent. J. Stat. Softw. 33, 1–22 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  118. Gold, L. et al. Aptamer-based multiplexed proteomic technology for biomarker discovery. PLoS ONE 5, e15004 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Behzadi-Khormouji, H. et al. Deep learning, reusable and problem-based architectures for detection of consolidation on chest X-ray images. Comput. Methods Prog. Biomed. 185, 105162 (2020).

    Article  Google Scholar 

  120. Kingma, D. & Ba, J. A. A method for stochastic optimization. In 3rd International Conference for Learning Representations (2014).

  121. Dai, H., Leeder, J. S. & Cui, Y. A modified generalized Fisher method for combining probabilities from dependent tests. Front. Genet. 5, 32 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  122. Foroutan, M. et al. Single sample scoring of molecular phenotypes. BMC Bioinf. 19, 404 (2018).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank the study participants for their time and dedication and the staff of the Stanford-LPCH Vaccine Program for recruiting participants and conducting the studies. Support for the conduct of these studies was from The Buck Institute for Research on Aging (to D.F.), the Ellison Foundation, National Institutes of Health (NIH) U19 AI057229, U19 AI090019 (to M.M.D.) and NIH/NCRR CTSA award number UL1 RR025744. This work was also supported by grants to C.F. from the EU Horizon 2020 Project PROPAG-AGEING (grant 634821), the EU JPND ADAGE project, the Ministry of Education and Science of the Russian Federation Agreement (agreement no. 075-15-2020-808). We gratefully acknowledge additional funding support from the NIH K01 HL135455, Stanford TRAM scholar award (N.S.), the Paul F. Glenn Foundation and the NIH Stanford Alzheimer’s Disease Research Center P50AG047366.

Author information

Authors and Affiliations

Authors

Contributions

D.F. conceived, conceptualized and designed the study; coordinated the biological analysis of samples and contributed to the analysis of experimental data and interpretation of the results. D.F., M.M.D., C.L.D. and J.G.M. conceived the study, provided guidance and funding. J.C.W. and F.H. provided guidance for the experimental work. N.S. and L.C. conducted in vitro and in vivo mice and EC experiments. D.F., B.L., Y.H., K.N., A.A. and T.G. conducted deep-learning and statistical analyses. S.S.O., V.J., R.T. and T.H. provided guidance for the in silico analysis of experimental data. N.S., Y.R.-H., F.H. and H.T.M. carried out or supervised the human data measurements; T.K., A.G. and Z.K.-R. helped to edit the manuscript. C.F., T.W.-C., B.L., R.O., D.M. collaborated with the study in centenarians. N.S., Y.H. and D.F. wrote the manuscript. All authors approved the final version of the manuscript.

Corresponding author

Correspondence to David Furman.

Ethics declarations

Competing interests

D.F. and M.M.D. are co-founders of Edifice Health, a company that utilizes iAge. The remaining authors declare no competing interests.

Additional information

Peer review information Nature Aging thanks M. Luisa Iruela-Arispe and the other, anonymous, reviewers for their contribution to the peer review of this work.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 1000 Immunomes Study design: systematic analysis of immune systems via ‘OMICS’ approaches.

The Stanford 1000 Immunomes Project consist of 1001 ambulatory subjects age 8 to 96 (34% males, 66% females) recruited during the years 2007 to 2016 for a longitudinal study of aging and vaccination, and for an independent study of chronic fatigue syndrome from which only healthy controls were included. For all samples of the Stanford 1KIP, deep immune phenotyping was conducted at the Stanford Human Immune Monitoring Center, where peripheral blood specimens were isolated and analyzed using standard procedures. Peripheral blood samples were obtained by venipuncture and peripheral blood mononuclear cells or whole blood samples were used for determination of cellular phenotypes and frequencies (N = 935) and for investigation of in vitro cellular responses to a variety of cytokine stimulations (N = 818); serum samples were obtained and used for protein content determination (including a total of 50 cytokines, chemokines and growth factors) (N = 1001). Clinical characterization was assessed via clinical questionnaire in a total of 902 subjects who completed the full set of 53 clinical items. From a total of 97 healthy young and older adults, comprehensive cardiovascular phenotyping was also conducted

Extended Data Fig. 2

Age distribution of the Stanford 1KIP cohort.

Source data

Extended Data Fig. 3 Estimation of the GAE code length and accuracy of age prediction.

We used 5-fold cross-validation to identify the best code length, among lengths from 1 to 10. We selected the length of code k, whose performance was not statistically significantly worse than that of longer codes (paired t-test p-value > 0.05). Within each fold we performed nested 3-fold cross-validation to select hyper-parameters (depth, weight decay and guidance-ratio). In our experiment, the best code length is 5 (a) as adding one more code (6) does not significantly improve the total loss (p = 0.18). After obtaining the best code length as 5, we used the 5-fold-cross-validation to select the best hyper-parameter setting (depth = 2, guidance-ratio = 0.2, L2 = 0.001) on all GAE with code length 5. Finally, we trained the GAE on the whole dataset with the selected best hyper-parameter setting and obtained the predictive function as the inflammatory clock predictor. GAE was compared to other machine learning methods such as autoencoder, neural networks, PCA, and RAW in (b). For the neural network, 2 fully connected layers with 5 nodes in each layer and tanh activation function were used. For PCA and RAW, we used elastic net to predict age. The GAE method outperforms linear methods for protein data reconstruction and prediction of chronological age (b). In (c), we found that the predictive performance of gradient boosting decision tree (GBDT) has similar performance as PCA. We conclude that GAE is superior to traditional machine learning methods.

Source data

Extended Data Fig. 4 Elimination of batch effect for serum immune protein data.

. Immune protein data from serum samples were subjected to normalization and batch correction procedures (See Methods) to ensure data from different sources can be combined and used as a whole. a, Spearman correlation between immune protein features and batch ID shows a strong dependency of data source on top 4 components (raw data, green line), which reaches a steady state after component 5. Data normalization and batch correction removes batch effect as indicated by lower mean absolute Spearman correlation between all features and batch id (blue line), which indicates impossibility to distinguish sample source from corrected data. b, Upper panel: immune protein expression heatmap of uncorrected data, Lower panel: immune protein expression heatmap of corrected data. The two batches come from two study cohorts, the Chronic Fatigue Syndrome Study (CFS) and Aging and vaccination study cohort (Flu).

Source data

Extended Data Fig. 5 iAge predictive of multi-mordity.

To select for predictors of comorbidity without bias, based on available data for all 902 subjects while controlling for the age effect, age-adjusted cross-validation was performed (a). By applying differential penalty values for each regressor, age variable is ‘forced in’, while imposing a stringent penalty (the lasso penalty) to all other features, so that selected variables do not correlate with age. A Mean Absolute Error (MAE) for the prediction of comorbidity of 0.41 is observed (b). Eighteen features are selected including inflammatory clock, high cholesterol and BMI (c) and immune parameters such as total CD8 (+) T cells, plasmablasts and transitional B cells (negative predictors) and IgD+CD27- and IgD-CD27- B cells, effector CD8 (+) T cells, total lymphocytes and monocytes, and central memory T cells (positive predictors) (d)

Source data

Extended Data Fig. 6 Univariate Regression between Age and CXCL9.

Significant correlation between age and CXCL9 using univariate regression analysis. We used linear regression where CXCL9 were regressed onto age. Correlation coefficient (R2) and p-value of F-test of overall significance are reported.

Source data

Extended Data Fig. 7 Luminex data for cardiovascular validation cohort.

In a validation study, 97 healthy adults (aged 25–90) well matched for cardiovascular risk factors, were selected from a total of 151 recruited subjects. Immune protein analysis was conducted in samples from these subjects. CXCL9, HGF, CXCL1, and LIF were found to change in the same direction in both the Stanford 1KIP and the validation cohort.

Source data

Extended Data Fig. 8 Human blood endothelial progenitor cells and mice endothelial cells.

a, Representative images of human blood progenitor endothelial cells from young (left) and old (right) individuals. b, Representative images of capillary-like networks show impaired tube formation by human BECs of old individuals compared to young. To further confirm the potential contribution of CXCL9 in cardiovascular aging, we assessed its expression in young (3–4 month) and old mice (2 yr.) endothelial cells (c). ECs isolated from old mice showed higher levels of CXCL9 (P value = 0.023) (d), while at the same time showed impaired EC function as evident by decreased tube formation (P value = 0.042) (a, f). Figure S8: All data represented as mean ± SEM, n = 3, *P < 0.05. Statistical analyses were performed using Student’s t-test (paired). Scale bar: 50 μm.

Extended Data Fig. 9 Expression of CXCR3 RNA in different tissue types.

CXCR3 was not expressed in iPSC induced cardiomyocytes (iPSC-CM), Fibroblast, or iPSC. However, it is highly expressed in iPSC induced endothelial cells and Human Umbilical Vein Endothelial Cells (HUVEC). All data represented as mean ± SEM.

Source data

Extended Data Fig. 10 Validation of the effects of CXCL9 on endothelial function.

Representative images of capillary-like networks from scramble- and CXCL9-KD hiPSC-ECs show that CXCL9-KD hiPSC-ECs retain their capacity to form tubes even at later passages when compared to scramble that showed impaired tube formation towards later passages of hiPSC-ECs. Scale bar: 50 μm. Experiment was repeated 3 times.

Supplementary information

Supplementary Information

Supplementary Figs. 1 and 2, and Tables 1–3.

Reporting Summary

Source data

Source Data Fig. 1

Age, number of comorbidities, frailty source data.

Source Data Fig. 2

CyTOF, cytokine source data.

Source Data Fig. 3

Cytokine data,CXCL9, arterial stiffness, PWV raw data.

Source Data Fig. 4

CXCL9 expression in blood EC (relative fold change). Tube formation in blood EC (number of tubes). NO production (μmol l−1). LDL uptake (mg protein h−1). Tube formation in iPSC-EC (number of tubes). LDL uptake (mg protein h−1). NO production (μmol l−1).

Source Data Fig. 5

iPSC-KO and Scramble gene expression data. Count data of specific pathways. Enrichment scores of pathways.

Source Data Fig. 6

iPSC-KO and Scramble gene expression data. Percentage relaxation of aortas to acetylcholine.

Source Data Fig. 7

Cell count for proliferation assay. SA-b-Gal activity. count for CD31+ capillaries.

Source Data Extended Data Fig. 2

Demographics source data.

Source Data Extended Data Fig. 3

Raw data used in predictive models.

Source Data Extended Data Fig. 4

Batch corrected before and after cytokine expression.

Source Data Extended Data Fig. 5

CyTOF source data.

Source Data Extended Data Fig. 6

Age and CXCL9 source data.

Source Data Extended Data Fig. 7

Raw data of cytokine expression in validation dataset.

Source Data CXCL9 expression in mouse EC (relative fold change).

Tube formation in mouse ECs (number of tubes).

Source Data Extended Data Fig. 9

Expression of CXCR3 RNA source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sayed, N., Huang, Y., Nguyen, K. et al. An inflammatory aging clock (iAge) based on deep learning tracks multimorbidity, immunosenescence, frailty and cardiovascular aging. Nat Aging 1, 598–615 (2021). https://doi.org/10.1038/s43587-021-00082-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43587-021-00082-y

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing