Skip to main content

Advertisement

Log in

A robust machine learning framework to identify signatures for frailty: a nested case-control study in four aging European cohorts

  • Original Article
  • Published:
GeroScience Aims and scope Submit manuscript

Abstract

Phenotype-specific omic expression patterns in people with frailty could provide invaluable insight into the underlying multi-systemic pathological processes and targets for intervention. Classical approaches to frailty have not considered the potential for different frailty phenotypes. We characterized associations between frailty (with/without disability) and sets of omic factors (genomic, proteomic, and metabolomic) plus markers measured in routine geriatric care. This study was a prevalent case control using stored biospecimens (urine, whole blood, cells, plasma, and serum) from 1522 individuals (identified as robust (R), pre-frail (P), or frail (F)] from the Toledo Study of Healthy Aging (R=178/P=184/F=109), 3 City Bordeaux (111/269/100), Aging Multidisciplinary Investigation (157/79/54) and InCHIANTI (106/98/77) cohorts. The analysis included over 35,000 omic and routine laboratory variables from robust and frail or pre-frail (with/without disability) individuals using a machine learning framework. We identified three protective biomarkers, vitamin D3 (OR: 0.81 [95% CI: 0.68–0.98]), lutein zeaxanthin (OR: 0.82 [95% CI: 0.70–0.97]), and miRNA125b-5p (OR: 0.73, [95% CI: 0.56–0.97]) and one risk biomarker, cardiac troponin T (OR: 1.25 [95% CI: 1.23–1.27]). Excluding individuals with a disability, one protective biomarker was identified, miR125b-5p (OR: 0.85, [95% CI: 0.81–0.88]). Three risks of frailty biomarkers were detected: pro-BNP (OR: 1.47 [95% CI: 1.27–1.7]), cardiac troponin T (OR: 1.29 [95% CI: 1.21–1.38]), and sRAGE (OR: 1.26 [95% CI: 1.01–1.57]). Three key frailty biomarkers demonstrated a statistical association with frailty (oxidative stress, vitamin D, and cardiovascular system) with relationship patterns differing depending on the presence or absence of a disability.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2

Similar content being viewed by others

Data Availability

Data will be made freely available

References

  1. Liu L-K, Guo C-Y, Lee W-J, Chen L-Y, Hwang A-C, Lin M-H, et al. Subtypes of physical frailty: Latent class analysis and associations with clinical characteristics and outcomes. Sci Rep. 2017;7(1):46417.

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Davies B, García F, Ara I, Artalejo FR, Rodriguez-Mañas L, Walter S. Relationship Between sarcopenia and frailty in the toledo study of healthy aging: a population based cross-sectional study. J Am Med Dir Assoc. 2018;19(4):282–6.

    CAS  PubMed  Google Scholar 

  3. Searle SD, Mitnitski A, Gahbauer EA, Gill TM, Rockwood K. A standard procedure for creating a frailty index. BMC Geriatr. 2008;8(1):24.

    PubMed  PubMed Central  Google Scholar 

  4. 3C Study Group. Vascular factors and risk of dementia: design of the Three-City Study and baseline characteristics of the study population. Neuroepidemiology. 2003;22(6):316–25.

    Google Scholar 

  5. Justice JN, Ferrucci L, Newman AB, Aroda VR, Bahnson JL, Divers J, et al. A framework for selection of blood-based biomarkers for geroscience-guided clinical trials: report from the TAME Biomarkers Workgroup. GeroScience. 2018;40(5–6):419–36.

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Viña J, Tarazona-Santabalbina FJ, Pérez-Ros P, Martínez-Arnau FM, Borras C, Olaso-Gonzalez G, et al. Biology of frailty: Modulation of ageing genes and its importance to prevent age-associated loss of function. Mol Aspects Med. 2016;50:88–108.

    PubMed  Google Scholar 

  7. Cutler RG, Mattson MP. The adversities of aging. Ageing Res Rev. 2006 Aug;5(3):221–38.

    CAS  PubMed  Google Scholar 

  8. Hayflick L. Biological aging is no longer an unsolved problem. Ann N Y Acad Sci. 2007;1100:1–13.

    CAS  PubMed  Google Scholar 

  9. Clegg A, Young J, Iliffe S, Rikkert MO, Rockwood K. Frailty in elderly people. Lancet. 2013;381(9868):752–62.

    PubMed  Google Scholar 

  10. Collard RM, Boter H, Schoevers RA, Oude Voshaar RC. Prevalence of frailty in community-dwelling older persons: a systematic review. J Am Geriatr Soc. 2012;60(8):1487–92.

    PubMed  Google Scholar 

  11. Siriwardhana DD, Hardoon S, Rait G, Weerasinghe MC, Walters KR. Prevalence of frailty and prefrailty among community-dwelling older adults in low-income and middle-income countries: a systematic review and meta-analysis. BMJ Open. 2018;8(3):e018195.

    PubMed  PubMed Central  Google Scholar 

  12. Liljas AEM, Carvalho LA, Papachristou E, De Oliveira C, Wannamethee SG, Ramsay SE, et al. Self-reported vision impairment and incident prefrailty and frailty in English community-dwelling older adults: findings from a 4-year follow-up study. J Epidemiol Community Health. 2017 Nov;71(11):1053–8.

    PubMed  PubMed Central  Google Scholar 

  13. Rodríguez-Mañas L, Féart C, Mann G, Viña J, Chatterji S, Chodzko-Zajko W, et al. Searching for an operational definition of frailty: a Delphi method based consensus statement. The Frailty Operative Definition-Consensus Conference Project. Journals Gerontol Ser A Biol Sci Med Sci. 2013;68(1):62–7.

    Google Scholar 

  14. Clegg A, Hassan-Smith Z. Frailty and the endocrine system. Lancet Diabetes Endocrinol. 2018;6(9):743–52.

    CAS  PubMed  Google Scholar 

  15. Alonso-Bouzón C, Carcaillon L, García-García FJ, Amor-Andrés MS, El Assar M, Rodríguez-Mañas L. Association between endothelial dysfunction and frailty: the Toledo Study for Healthy Aging. Age (Dordr). 2014;36(1):495–505.

    PubMed  Google Scholar 

  16. Pansarasa O, Pistono C, Davin A, Bordoni M, Mimmi MC, Guaita A, et al. Altered immune system in frailty: Genetics and diet may influence inflammation. Ageing Res Rev. 2019;54:100935.

    PubMed  Google Scholar 

  17. Arauna D, García F, Rodríguez-Mañas L, Marrugat J, Sáez C, Alarcón M, et al. Older adults with frailty syndrome present an altered platelet function and an increased level of circulating oxidative stress and mitochondrial dysfunction biomarker GDF-15. Free Radic Biol Med. 2020;149:64–71.

    CAS  PubMed  Google Scholar 

  18. Kochlik B, Stuetz W, Pérès K, Pilleron S, Féart C, García García FJ, et al. Associations of fat-soluble micronutrients and redox biomarkers with frailty status in the FRAILOMIC initiative. J Cachexia Sarcopenia Muscle. 2019;10(6):1339–46.

    PubMed  PubMed Central  Google Scholar 

  19. Butcher L, Carnicero JA, Gomez Cabrero D, Dartigues J-F, Pérès K, Garcia-Garcia FJ, et al. Increased levels of soluble receptor for advanced glycation end-products (RAGE) are associated with a higher risk of mortality in frail older adults. Age Ageing. 2019;48(5):696–702.

    PubMed  Google Scholar 

  20. El Assar M, Angulo J, Carnicero JA, Walter S, García-García FJ, López-Hernández E, et al. Frailty is associated with lower expression of genes involved in cellular response to stress: results from the Toledo Study for Healthy Aging. J Am Med Dir Assoc. 2017;18(8):734.e1–7.

    Google Scholar 

  21. Fried LP, Xue Q-L, Cappola AR, Ferrucci L, Chaves P, Varadhan R, et al. Nonlinear multisystem physiological dysregulation associated with frailty in older women: implications for etiology and treatment. Journals Gerontol Ser A Biol Sci Med Sci. 2009;64A(10):1049–57.

    Google Scholar 

  22. Trevisan C, Veronese N, Maggi S, Baggio G, Toffanello ED, Zambon S, et al. Factors influencing transitions between frailty states in elderly adults: The Progetto Veneto Anziani Longitudinal Study. J Am Geriatr Soc. 2017;65(1):179–84.

    PubMed  Google Scholar 

  23. Rodriguez-Mañas L, Laosa O, Vellas B, Paolisso G, Topinkova E, Oliva-Moreno J, et al. Effectiveness of a multimodal intervention in functionally impaired older people with type 2 diabetes mellitus. J Cachexia Sarcopenia Muscle. 2019;10(4):721–33.

    PubMed  PubMed Central  Google Scholar 

  24. Trombetti A, Hars M, Hsu F-C, Reid KF, Church TS, Gill TM, et al. Effect of physical activity on frailty. Ann Intern Med. 2018;168(5):309–16.

    PubMed  PubMed Central  Google Scholar 

  25. Nilsson R, Björkegren J, Tegnér J. On reliable discovery of molecular signatures. BMC Bioinformatics. 2009;10:38.

    PubMed  PubMed Central  Google Scholar 

  26. Nilsson R, Peña JM, Björkegren J, Tegnér J. Detecting multivariate differentially expressed genes. BMC Bioinformatics. 2007;8:150.

    PubMed  PubMed Central  Google Scholar 

  27. Angulo J, El Assar M, Álvarez-Bustos A, Rodríguez-Mañas L. Physical activity and exercise: strategies to manage frailty. Redox Biol. 2020;35:101513.

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Angulo J, El Assar M, Rodríguez-Mañas L. Frailty and sarcopenia as the basis for the phenotypic manifestation of chronic diseases in older adults. Mol Aspects Med. 2016;50:1–32.

    PubMed  Google Scholar 

  29. Huang S-T, Tange C, Otsuka R, Nishita Y, Peng L-N, Hsiao F-Y, et al. Subtypes of physical frailty and their long-term outcomes: a longitudinal cohort study. J Cachexia Sarcopenia Muscle. 2020;11(5):1223–31.

    PubMed  PubMed Central  Google Scholar 

  30. Pamoukdjian F, Laurent M, Martinez-Tapia C, Rolland Y, Paillaud E, Canoui-Poitrine F. Frailty parameters, morbidity and mortality in older adults with cancer: a structural equation modelling approach based on the fried phenotype. J Clin Med. 2020;9(6):1826.

    PubMed Central  Google Scholar 

  31. Garcia-Garcia FJ, Gutierrez Avila G, Alfaro-Acha A, Amor Andres MS, de la Torre Lanza MDLA, Escribano Aparicio MV, et al. The prevalence of frailty syndrome in an older population from Spain. The Toledo study for healthy aging. J Nutr Heal AGING. 2011;15(10):852–6.

    CAS  Google Scholar 

  32. Balzi D, Lauretani F, Barchielli A, Ferrucci L, Bandinelli S, Buiatti E, et al. Risk factors for disability in older persons over 3-year follow-up. Age Ageing. 2010;39(1):92–8.

    PubMed  Google Scholar 

  33. Ávila-Funes JA, Helmer C, Amieva H, Barberger-Gateau P, Le Goff M, Ritchie K, et al. Frailty among community-dwelling elderly people in France: the Three-City Study. Journals Gerontol Ser A Biol Sci Med Sci. 2008;63(10):1089–96.

    Google Scholar 

  34. Fried LP, Ferrucci L, Darer J, Williamson JD, Anderson G. Untangling the concepts of disability, frailty, and comorbidity: implications for improved targeting and care. Journals Gerontol Ser A Biol Sci Med Sci. 2004;59(3):M255–63.

    Google Scholar 

  35. Calvani R, Picca A, Marini F, Biancolillo A, Gervasoni J, Persichilli S, et al. Identification of biomarkers for physical frailty and sarcopenia through a new multi-marker approach: results from the BIOSPHERE study. GeroScience. 2020. https://doi.org/10.1007/s11357-020-00197-x.

  36. Pérès K, Matharan F, Allard M, Amieva H, Baldi I, Barberger-Gateau P, et al. Health and aging in elderly farmers: the AMI cohort. BMC Public Health. 2012;12:558.

    PubMed  PubMed Central  Google Scholar 

  37. Ferrucci L, Bandinelli S, Benvenuti E, Di Iorio A, Macchi C, Harris TB, et al. Subsystems contributing to the decline in ability to walk: bridging the gap between epidemiology and geriatric practice in the InCHIANTI study. J Am Geriatr Soc. 2000;48(12):1618–25.

    CAS  PubMed  Google Scholar 

  38. Fried LP, Tangen CM, Walston J, Newman AB, Hirsch C, Gottdiener J, et al. Frailty in older adults: evidence for a phenotype. Journals Gerontol A. 2001;56(3):146–56.

    Google Scholar 

  39. Erusalimsky JD, Grillari J, Grune T, Jansen-Duerr P, Lippi G, Sinclair AJ, et al. In search of ‘omics’-based biomarkers to predict risk of frailty and its consequences in older individuals: the FRAILOMIC Initiative. Gerontology. 2015;62(2):182–90.

    PubMed  Google Scholar 

  40. Lippi G, Jansen-Duerr P, Viña J, Durrance-Bagale A, Abugessaisa I, Gomez-Cabrero D, et al. Laboratory biomarkers and frailty: presentation of the FRAILOMIC initiative. Clin Chem Lab Med. 2015;53:e253–5. https://doi.org/10.1515/cclm-2015-0147.

    Article  CAS  PubMed  Google Scholar 

  41. Van Buuren S, Groothuis-Oudshoorn K. Multivariate imputation by chained equations. J Stat Softw. 2011;45(3):1–67.

    Google Scholar 

  42. Lagani V, Athineou G, Farcomeni A, Tsagris M, Tsamardinos I. Feature selection with the R package MXM: discovering statistically equivalent feature subsets. J Stat Software. 2017;1(7) SepAvailable from: https://www.jstatsoft.org/v080/i07.

  43. Breiman L. Random forests. Mach Learn. 2001;45(1):5–32.

    Google Scholar 

  44. Hand DJ, Till RJ. A simple generalisation of the area under the ROC curve for multiple class classification problems. Mach Learn. 2001;45(2):171–86.

    Google Scholar 

  45. Nagaraj S, Zoltowska KM, Laskowska-Kaszub K, Wojda U. microRNA diagnostic panel for Alzheimer’s disease and epigenetic trade-off between neurodegeneration and cancer. Ageing Res Rev. 2018. https://doi.org/10.1016/j.arr.2018.10.008.

  46. Mohri T, Nakajima M, Takagi S, Komagata S, Yokoi T. MicroRNA regulates human vitamin D receptor. Int J Cancer. 125(6):1328–33.

  47. Semba RD, Varadhan R, Bartali B, Ferrucci L, Ricks MO, Blaum C, et al. Low serum carotenoids and development of severe walking disability among older women living in the community: the Women’s Health and Aging Study I. Age Ageing. 2007;36(1):62–7.

    PubMed  Google Scholar 

  48. Bruyère O, Cavalier E, Buckinx F, Reginster J-Y. Relevance of vitamin D in the pathogenesis and therapy of frailty. Curr Opin Clin Nutr Metab Care. 2017;20(1) Available from: https://journals.lww.com/co-clinicalnutrition/Fulltext/2017/01000/Relevance_of_vitamin_D_in_the_pathogenesis_and.6.aspx.

  49. Costello LC, Franklin RB. Plasma citrate homeostasis: how it is regulated; and its physiological and clinical implications. an important, but neglected, relationship in medicine. HSOA J Hum Endocrinol. 2016 [cited 2019 Feb 10]. ;1(1). Available from: http://www.ncbi.nlm.nih.gov/pubmed/28286881

  50. Huang M, Que Y, Shen X. Correlation of the plasma levels of soluble RAGE and endogenous secretory RAGE with oxidative stress in pre-diabetic patients. J Diabetes Complications. 2015;29(3):422–6.

    PubMed  Google Scholar 

  51. Nakamura K, Yamagishi S, Adachi H, Kurita-Nakamura Y, Matsui T, Yoshida T, et al. Elevation of soluble form of receptor for advanced glycation end products (sRAGE) in diabetic subjects with coronary artery disease. Diabetes Metab Res Rev. 2007;23(5):368–71.

    CAS  PubMed  Google Scholar 

  52. Selvin E, Halushka MK, Rawlings AM, Hoogeveen RC, Ballantyne CM, Coresh J, et al. SRAGE and risk of diabetes, cardiovascular disease, and death. Diabetes. 2013;62(6):2116–21.

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Colhoun HM, Betteridge DJ, Durrington P, Hitman G, Neil A, Livingstone S, et al. Total soluble and endogenous secretory receptor for advanced glycation end products as predictive biomarkers of coronary heart disease risk in patients with type 2 diabetes. Diabetes. 2011;60(September):2379–85.

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Prasad K. Is there any evidence that AGE/sRAGE is a universal biomarker/risk marker for diseases? Mol Cell Biochem. 2019;451(1):139–44.

    CAS  PubMed  Google Scholar 

  55. Smedsrud MK, Gravning J, Omland T, Eek C, Mørkrid L, Skulstad H, et al. Sensitive cardiac troponins and N-terminal pro-B-type natriuretic peptide in stable coronary artery disease: correlation with left ventricular function as assessed by myocardial strain. Int J Cardiovasc Imaging. 2015;31(5):967–73.

    PubMed  Google Scholar 

  56. Daniels LB, Clopton P, DeFilippi CR, Sanchez OA, Bahrami H, Lima JAC, et al. Serial measurement of N-terminal pro-B-type natriuretic peptide and cardiac troponin T for cardiovascular disease risk assessment in the Multi-Ethnic Study of Atherosclerosis (MESA). Am Heart J. 2015;170(6):1170–83.

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Kajioka S, Takahashi-Yanaga F, Shahab N, Onimaru M, Matsuda M, Takahashi R, et al. Endogenous cardiac troponin T modulates Ca2+-mediated smooth muscle contraction. Sci Rep. 2012;2:1–7.

    Google Scholar 

  58. Seidelmann SB, Vardeny O, Claggett B, Yu B, Shah AM, Ballantyne CM, et al. An NPPB promoter polymorphism associated with elevated N-terminal pro-b-type natriuretic peptide and lower blood pressure, hypertension, and mortality. J Am Heart Assoc. 2017;6(4). https://doi.org/10.1161/JAHA.116.005257.

  59. Rattray NJW, Trivedi DK, Xu Y, Chandola T, Johnson CH, Marshall AD, et al. Metabolic dysregulation in vitamin E and carnitine shuttle energy mechanisms associate with human frailty. Nat Commun. 2019;10(1):5027.

    PubMed  PubMed Central  Google Scholar 

  60. Tegnér JN, Compte A, Auffray C, An G, Cedersund G, Clermont G, et al. Computational disease modeling - fact or fiction? BMC Syst Biol. 2009;3:56.

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgments

On behalf of the FRAILOMIC Initiative the authors would like to thank Perrine André MSc and Hermine Pellay MSc (Univ. Bordeaux, Inserm, Bordeaux Population Health Research Center, UMR 1219, F-33000 Bordeaux, France), Mariam El Assar MSc and Betty Davies MD (Fundación de Investigación Biomedica Hospital Unversitario de Getafe), Eleonora Talluri PhD(USL Centro Toscana, Firenze, Italy), Valeria Orrù MSc and Michele Marongiu MSc (Institute for Genetic and Biomedical Research, Caligari, Italy), Esther García-Esquinas PhD and Esther Lopez-Garcia PhD and Pilar Guallar MD PhD and Fernando Rodriguez Artalejo MD PhD (Faculty of Medicine, Universidad Autonoma de Madrid, Madrid, Spain), Ignacio Ara PhD (GENUD Toledo Research Group, Universidad Castilla-La Mancha, Toledo, Spain), José-María Sánchez-Puelles PhD (Molecular Pharmacology Lab, Centre of Biological Sciences, CSIC, Madrid, Spain), Paloma Moraga MSc (Sistemas Genomicos, Valencia, Spain).

Funding

This work was supported by the European Union’s Seventh Framework Programme (FP7/2007-2013) FRAILOMIC Project (grant number 305483). The Three-City Study was conducted under a partnership agreement between the Institut National de la Santé et de la Recherche Médicale, Victor Segalen – Bordeaux2 University and the Sanofi-Synthélabo company. The Fondation pour la Recherche Médicale funded the preparation and beginning of the study. The 3C-Study was also sponsored by the Caisse Nationale Maladie des Travailleurs Salariés, Direction Générale de la Santé, Conseils Régionaux of Aquitaine and Bourgogne, Fondation de France, Ministry of Research-INSERM Program Cohortes et collections de données biologiques, the Fondation Plan Alzheimer (FCS 2009-2012), and the Caisse Nationale pour la Solidarité et l’Autonomie. The InCHIANTI study baseline (1998–2000) was supported as a ‘targeted project’ (ICS110.1/RF97.71) by the Italian Ministry of Health and in part by the U.S. National Institute on Aging (Contracts: 263 MD 9164 and 263 MD 821336) and by the Intramural Research Program of the National Institute on Aging, National Institutes of Health, Baltimore, Maryland.

Author information

Authors and Affiliations

Authors

Consortia

Contributions

The authors of this manuscript certify that they comply with the ethical guidelines for authorship and publishing in the Journal.

Corresponding author

Correspondence to Leocadio Rodriguez-Mañas.

Ethics declarations

Ethics approval

The original study protocols were approved by ethical committee according to the principles of the Declaration of Helsinki and all participants signed a written consent with participants agree to sample retention/analysis and data publication.

Conflicts of interest

Stefan Walter, Rebeca Miñambres, Lucía Bernard, Lee Butcher, Jorge Erusalimsky, Francisco José García-García, José Antonio Carnicero, Tim Hardman, Mattias Hacki, Johannes Grillari, Edoardo Fiorillo, Francesco Cucca, Matteo Cesari, Isabelle Carrie, Marco Colpo, Stefania Bandinelli, Karine Peres, Jean Francois Dartigues, Catherine Helmer,José Viña, Gloria Olaso, Irene Garcia, Jorge Garcia, Pidder Janssen-Dürr, Tilman Grune, Daniela Weber, Giuseppe Lippi, Chiara Bonaguri, and Alan Sinclair declare no conflicts of interest. David Gomez-Cabrero, Imad Abugesaissa and Jesper Tegner have been paid as consultants by YouHealth SB. David Gomez-Cabrero has received a speaker honorarium from Sanofi Aventis. Harald Mischak is the co-founder and co-owner of Mosaiques Diagnostics. Petra Zürbig is employed by Mosaiques Diagnostics. Catherine Féart received fees for conferences from Danone Institute and Nutricia, and served as consultant for Laboratoire Lescuyer and Cholé'Doc. Leocadio Rodriguez-Mañas has received fees for conferences from Abbott Laboratories and Novartis.

Additional information

Publisher’s note

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

David Gómez-Cabrero and Stefan Walter contributed equally to this work

Supplementary Information

ESM 1

(DOCX 8326 kb)

ESM 2

(XLSX 36 kb)

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gomez-Cabrero, D., Walter, S., Abugessaisa, I. et al. A robust machine learning framework to identify signatures for frailty: a nested case-control study in four aging European cohorts. GeroScience 43, 1317–1329 (2021). https://doi.org/10.1007/s11357-021-00334-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11357-021-00334-0

Keywords

Navigation