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.

  • Protocol
  • Published:

A toolbox for the longitudinal assessment of healthspan in aging mice

Abstract

The number of people aged over 65 is expected to double in the next 30 years. For many, living longer will mean spending more years with the burdens of chronic diseases such as Alzheimer’s disease, cardiovascular disease, and diabetes. Although researchers have made rapid progress in developing geroprotective interventions that target mechanisms of aging and delay or prevent the onset of multiple concurrent age-related diseases, a lack of standardized techniques to assess healthspan in preclinical murine studies has resulted in reduced reproducibility and slow progress. To overcome this, major centers in Europe and the United States skilled in healthspan analysis came together to agree on a toolbox of techniques that can be used to consistently assess the healthspan of mice. Here, we describe the agreed toolbox, which contains protocols for echocardiography, novel object recognition, grip strength, rotarod, glucose tolerance test (GTT) and insulin tolerance test (ITT), body composition, and energy expenditure. The protocols can be performed longitudinally in the same mouse over a period of 4–6 weeks to test how candidate geroprotectors affect cardiac, cognitive, neuromuscular, and metabolic health.

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: A graphic representation of the experimental design for healthspan assessment in response to a geroprotector of choice in C57BL6/J using the recommended toolbox.
Fig. 2: Representative images from high-resolution echocardiography systems.
Fig. 3: Novel object recognition (NOR) memory enhancement in exercised male mice.
Fig. 4: Assessment of muscle strength and neuromuscular function.
Fig. 5: Effects of dietary protein level on energy balance.
Fig. 6: Glucose and insulin tolerance tests.

Similar content being viewed by others

Data availability

All data shown in this paper are available from the authors upon reasonable request.

References

  1. Bellantuono, I. Find drugs that delay many diseases of old age. Nature 554, 293–295 (2018).

    CAS  PubMed  Google Scholar 

  2. Tchkonia, T. & Kirkland, J. L. Aging, cell senescence, and chronic disease: emerging therapeutic strategies. JAMA 320, 1319–1320 (2018).

    PubMed  Google Scholar 

  3. Martin-Montalvo, A. et al. Metformin improves healthspan and lifespan in mice. Nat. Commun. 4, 2192 (2013).

    PubMed  Google Scholar 

  4. Neff, F. et al. Rapamycin extends murine lifespan but has limited effects on aging. J. Clin. Invest. 123, 3272–3291 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Zhu, Y. et al. The Achilles’ heel of senescent cells: from transcriptome to senolytic drugs. Aging Cell 14, 644–658 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Arriola Apelo, S. I., Pumper, C. P., Baar, E. L., Cummings, N. E. & Lamming, D. W. Intermittent administration of rapamycin extends the life span of female C57BL/6J mice. J. Gerontol. A Biol. Sci. Med. Sci. 71, 876–881 (2016).

    PubMed  PubMed Central  Google Scholar 

  7. Richardson, A. et al. Measures of healthspan as indices of aging in mice-a recommendation. J. Gerontol. A Biol. Sci. Med. Sci. 71, 427–430 (2016).

    PubMed  Google Scholar 

  8. Kane, A. E., Keller, K. M., Heinze-Milne, S., Grandy, S. A. & Howlett, S. E. A murine frailty index based on clinical and laboratory measurements: links between frailty and pro-inflammatory cytokines differ in a sex-specific manner. J. Gerontol. A Biol. Sci. Med. Sci. 74, 275–282 (2019).

    CAS  PubMed  Google Scholar 

  9. Whitehead, J. C. et al. A clinical frailty index in aging mice: comparisons with frailty index data in humans. J. Gerontol. A Biol. Sci. Med. Sci. 69, 621–632 (2014).

    PubMed  Google Scholar 

  10. Hamm, R. J., Pike, B. R., O’Dell, D. M., Lyeth, B. G. & Jenkins, L. W. The rotarod test: an evaluation of its effectiveness in assessing motor deficits following traumatic brain injury. J. Neurotrauma 11, 187–196 (1994).

    CAS  PubMed  Google Scholar 

  11. Wolff, B. S., Raheem, S. A. & Saligan, L. N. Comparing passive measures of fatigue-like behavior in mice. Sci. Rep. 8, 14238 (2018).

    PubMed  PubMed Central  Google Scholar 

  12. Schafer, M. J. et al. Exercise prevents diet-induced cellular senescence in adipose tissue. Diabetes 65, 1606–1615 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Halldorsdottir, S., Carmody, J., Boozer, C. N., Leduc, C. A. & Leibel, R. L. Reproducibility and accuracy of body composition assessments in mice by dual energy x-ray absorptiometry and time domain nuclear magnetic resonance. Int. J. Body Compos. Res. 7, 147–154 (2009).

    PubMed  PubMed Central  Google Scholar 

  14. Potter, P. K. et al. Novel gene function revealed by mouse mutagenesis screens for models of age-related disease. Nat. Commun. 7, 12444 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Xie, K. et al. Epigenetic alterations in longevity regulators, reduced life span, and exacerbated aging-related pathology in old father offspring mice. Proc. Natl Acad. Sci. USA 115, E2348–E2357 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Lamming, D. W. et al. Young and old genetically heterogeneous HET3 mice on a rapamycin diet are glucose intolerant but insulin sensitive. Aging Cell 12, 712–718 (2013).

    CAS  PubMed  Google Scholar 

  17. Lamming, D. W. et al. Rapamycin-induced insulin resistance is mediated by mTORC2 loss and uncoupled from longevity. Science 335, 1638–1643 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Arriola Apelo, S. I. et al. Alternative rapamycin treatment regimens mitigate the impact of rapamycin on glucose homeostasis and the immune system. Aging Cell 15, 28–38 (2016).

    CAS  PubMed  Google Scholar 

  19. Stout, M. B. et al. 17alpha-Estradiol alleviates age-related metabolic and inflammatory dysfunction in male mice without inducing feminization. J. Gerontol. A Biol. Sci. Med. Sci. 72, 3–15 (2017).

    CAS  PubMed  Google Scholar 

  20. Alfaras, I. et al. Health benefits of late-onset metformin treatment every other week in mice. Npj. Aging Mech. Dis. 3, 16 (2017).

    PubMed  PubMed Central  Google Scholar 

  21. Diaz-Ruiz, A. et al.Overexpression of CYB5R3 and NQO1, two NAD(+) -producing enzymes, mimics aspects of caloric restriction. Aging Cell, e12767 (2018).

  22. Mitchell, S. J. et al. Nicotinamide improves aspects of healthspan, but not lifespan, in mice. Cell Metab. 27, 667–676 e664 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Allard, J. S. et al. Prolonged metformin treatment leads to reduced transcription of Nrf2 and neurotrophic factors without cognitive impairment in older C57BL/6J mice. Behav. Brain. Res. 301, 1–9 (2016).

    CAS  PubMed  Google Scholar 

  24. Lee, S. et al. Assessment of cognitive impairment in a mouse model of high-fat diet-induced metabolic stress with touchscreen-based automated battery system. Exp. Neurobiol. 27, 277–286 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Benice, T. S. & Raber, J. Object recognition analysis in mice using nose-point digital video tracking. J. Neurosci. Methods 168, 422–430 (2008).

    CAS  PubMed  Google Scholar 

  26. Bettis, T. & Jacobs, L. F. Sex differences in object recognition are modulated by object similarity. Behav. Brain Res. 233, 288–292 (2012).

    PubMed  Google Scholar 

  27. Prendergast, B. J., Onishi, K. G. & Zucker, I. Female mice liberated for inclusion in neuroscience and biomedical research. Neurosci. Biobehav. Rev. 40, 1–5 (2014).

    PubMed  Google Scholar 

  28. Clayton, J. A. & Collins, F. S. Policy: NIH to balance sex in cell and animal studies. Nature 509, 282–283 (2014).

    PubMed  PubMed Central  Google Scholar 

  29. McCullough, L. D., McCarthy, M. M. & de Vries, G. J. NIH policy: status quo is also costly. Nature 510, 340 (2014).

    CAS  PubMed  Google Scholar 

  30. Brooks, S. P., Pask, T., Jones, L. & Dunnett, S. B. Behavioural profiles of inbred mouse strains used as transgenic backgrounds. II: Cogn. tests. Genes Brain Behav. 4, 307–317 (2005).

    CAS  Google Scholar 

  31. Feridooni, H. A., Sun, M. H., Rockwood, K. & Howlett, S. E. Reliability of a frailty index based on the clinical assessment of health deficits in male C57BL/6J mice. J. Gerontol. A Biol. Sci. Med. Sci. 70, 686–693 (2015).

    PubMed  Google Scholar 

  32. Sukoff Rizzo, S. J. et al. Assessing healthspan and lifespan measures in aging mice: optimization of testing protocols, replicability, and rater reliability. Curr. Protoc. Mouse Biol. 8, e45 (2018).

    PubMed  Google Scholar 

  33. Palmer, A. K. et al. Targeting senescent cells alleviates obesity-induced metabolic dysfunction. Aging Cell, e12950 (2019).

  34. Xu, M. et al. Senolytics improve physical function and increase lifespan in old age. Nat. Med. 24, 1246–1256 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Mitchell, S. J. et al. Effects of sex, strain, and energy intake on hallmarks of aging in mice. Cell Metab. 23, 1093–1112 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Cummings, N. E. et al. Restoration of metabolic health by decreased consumption of branched-chain amino acids. J. Physiol. 596, 623–645 (2018).

    CAS  PubMed  Google Scholar 

  37. Xie, K. et al. Every-other-day feeding extends lifespan but fails to delay many symptoms of aging in mice. Nat. Commun. 8, 155 (2017).

    PubMed  PubMed Central  Google Scholar 

  38. McGreevy, K. R. et al. Intergenerational transmission of the positive effects of physical exercise on brain and cognition. Proc. Natl Acad. Sci. USA (2019).

  39. Lamming, D. W. et al. Depletion of Rictor, an essential protein component of mTORC2, decreases male lifespan. Aging Cell 13, 911–917 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Zhu, Y. et al. New agents that target senescent cells: the flavone, fisetin, and the BCL-XL inhibitors, A1331852 and A1155463. Aging (Albany NY) 9, 955–963 (2017).

    Google Scholar 

  41. Yousefzadeh, M. J. et al. Fisetin is a senotherapeutic that extends health and lifespan. EbioMedicine 36, 18–28 (2018).

    PubMed  PubMed Central  Google Scholar 

  42. Xu, M. et al. Targeting senescent cells enhances adipogenesis and metabolic function in old age. eLife 4, e12997 (2015).

    PubMed  PubMed Central  Google Scholar 

  43. Mercken, E. M. et al. SRT2104 extends survival of male mice on a standard diet and preserves bone and muscle mass. Aging Cell 13, 787–796 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Mitchell, S. J. et al. The SIRT1 activator SRT1720 extends lifespan and improves health of mice fed a standard diet. Cell Rep. 6, 836–843 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Minor, R. K. et al. SRT1720 improves survival and healthspan of obese mice. Sci. Rep. 1, 70 (2011).

    PubMed  PubMed Central  Google Scholar 

  46. Kane, A. E. et al. Impact of longevity interventions on a validated mouse clinical frailty index. J. Gerontol. A Biol. Sci. Med. Sci. 71, 333–339 (2016).

    CAS  PubMed  Google Scholar 

  47. Kane, A. E. et al. A comparison of two mouse frailty assessment tools. J. Gerontol. A Biol. Sci. Med. Sci. 72, 904–909 (2017).

    PubMed  PubMed Central  Google Scholar 

  48. Yu, D. et al. Short-term methionine deprivation improves metabolic health via sexually dimorphic, mTORC1-independent mechanisms. FASEB J. 32, 3471–3482 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Ramos, F. J. et al. Rapamycin reverses elevated mTORC1 signaling in lamin A/C-deficient mice, rescues cardiac and skeletal muscle function, and extends survival. Sci. Transl. Med. 4, 144ra103 (2012).

    PubMed  PubMed Central  Google Scholar 

  50. Darcy, J. et al. Increased environmental temperature normalizes energy metabolism outputs between normal and Ames dwarf mice. Aging 10, 2709–2722 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Cohen, D. E., Supinski, A. M., Bonkowski, M. S., Donmez, G. & Guarente, L. P. Neuronal SIRT1 regulates endocrine and behavioral responses to calorie restriction. Genes Dev. 23, 2812–2817 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Bitto, A. et al. Transient rapamycin treatment can increase lifespan and healthspan in middle-aged mice. eLife 5, https://doi.org/10.7554/eLife.16351 (2016).

  53. Chen, C., Liu, Y., Liu, Y. & Zheng, P. mTOR regulation and therapeutic rejuvenation of aging hematopoietic stem cells. Sci. Signal 2, ra75 (2009).

    PubMed  PubMed Central  Google Scholar 

  54. Harrison, D. E. et al. Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature 460, 392–395 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Dai, D. F. et al. Altered proteome turnover and remodeling by short-term caloric restriction or rapamycin rejuvenate the aging heart. Aging Cell 13, 529–539 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Flynn, J. M. et al. Late-life rapamycin treatment reverses age-related heart dysfunction. Aging Cell 12, 851–862 (2013).

    CAS  PubMed  Google Scholar 

  57. Hou, Y. et al. NAD+ supplementation normalizes key Alzheimer’s features and DNA damage responses in a new AD mouse model with introduced DNA repair deficiency. Proc. Natl Acad. Sci. USA 115, E1876–E1885 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Spilman, P. et al. Inhibition of mTOR by rapamycin abolishes cognitive deficits and reduces amyloid-beta levels in a mouse model of Alzheimer’s disease. PLoS ONE 5, e9979 (2010).

    PubMed  PubMed Central  Google Scholar 

  59. Baur, J. A. et al. Resveratrol improves health and survival of mice on a high-calorie diet. Nature 444, 337–342 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Fontana, L., Kennedy, B. K., Longo, V. D., Seals, D. & Melov, S. Medical research: treat ageing. Nature 511, 405–407 (2014).

    CAS  PubMed  Google Scholar 

  61. Mitchell, S. J. et al. Daily fasting improves health and survival in male mice independent of diet composition and calories. Cell Metab. 29, 221–228 e223 (2019).

    CAS  PubMed  Google Scholar 

  62. Wilkinson, J. E. et al. Rapamycin slows aging in mice. Aging Cell 11, 675–682 (2012).

    CAS  PubMed  Google Scholar 

  63. Turturro, A. et al. Growth curves and survival characteristics of the animals used in the Biomarkers of Aging Program. J. Gerontol. A Biol. Sci. Med. Sci. 54, B492–501 (1999).

    CAS  PubMed  Google Scholar 

  64. Dumas, S. N. & Lamming, D. W. Next generation strategies for geroprotection via mTORC1 inhibition. J. Gerontol. A Biol. Sci. Med. Sci., https://doi.org/10.1093/gerona/glz056 (2019).

    Google Scholar 

  65. Harrison, D. E. et al. Acarbose, 17-alpha-estradiol, and nordihydroguaiaretic acid extend mouse lifespan preferentially in males. Aging Cell 13, 273–282 (2014).

    CAS  PubMed  Google Scholar 

  66. Lamming, D. W. Diminished mTOR signaling: a common mode of action for endocrine longevity factors. SpringerPlus 3, 735 (2014).

    PubMed  PubMed Central  Google Scholar 

  67. Strong, R. et al. Longer lifespan in male mice treated with a weakly estrogenic agonist, an antioxidant, an alpha-glucosidase inhibitor or a Nrf2-inducer. Aging Cell 15, 872–884 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Miller, R. A. et al. Rapamycin-mediated lifespan increase in mice is dose and sex dependent and metabolically distinct from dietary restriction. Aging Cell 13, 468–477 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Fontana, L. et al. Decreased consumption of branched-chain amino acids improves metabolic health. Cell Rep. 16, 520–530 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. De Leon, E. R. et al. Age-dependent protection of insulin secretion in diet induced obese mice. Sci. Rep. 8, 17814 (2018).

    PubMed  PubMed Central  Google Scholar 

  71. Gill, J. F., Santos, G., Schnyder, S. & Handschin, C. PGC-1alpha affects aging-related changes in muscle and motor function by modulating specific exercise-mediated changes in old mice. Aging Cell 17, e12697 (2018).

    Google Scholar 

  72. Graber, T. G., Ferguson-Stegall, L., Liu, H. & Thompson, L. V. Voluntary aerobic exercise reverses frailty in old mice. J. Gerontol. A Biol. Sci. Med. Sci. 70, 1045–1058 (2015).

    PubMed  Google Scholar 

  73. Chellappa, K. et al. Hypothalamic mTORC2 is essential for metabolic health and longevity. Aging Cell 18, e13014 (2019).

    PubMed  PubMed Central  Google Scholar 

  74. Yu, D. et al. Calorie-restriction-induced insulin sensitivity is mediated by adipose mTORC2 and not required for lifespan extension. Cell Rep. 29, 236–248.e3 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Nadon, N. L., Strong, R., Miller, R. A. & Harrison, D. E. NIA interventions testing program: investigating putative aging intervention agents in a genetically heterogeneous mouse model. EbioMedicine 21, 3–4 (2017).

    PubMed  Google Scholar 

  76. Yuan, R. et al. Genetic coregulation of age of female sexual maturation and lifespan through circulating IGF1 among inbred mouse strains. Proc. Natl Acad. Sci. USA 109, 8224–8229 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Ryan, D. P. et al. A paternal methyl donor-rich diet altered cognitive and neural functions in offspring mice. Mol. Psychiatry 23, 1345–1355 (2018).

    CAS  PubMed  Google Scholar 

  78. Fernandez-Twinn, D. S., Constancia, M. & Ozanne, S. E. Intergenerational epigenetic inheritance in models of developmental programming of adult disease. Semin. Cell Dev. Biol. 43, 85–95 (2015).

    PubMed  PubMed Central  Google Scholar 

  79. Martin-Gronert, M. S. & Ozanne, S. E. Early life programming of obesity. Med. Wieku Rozwoj. 17, 7–12 (2013).

    PubMed  Google Scholar 

  80. Menting, M. D. et al. Maternal obesity in pregnancy impacts offspring cardiometabolic health: Systematic review and meta-analysis of animal studies. Obes. Rev. 20, 675–685 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Dearden, L., Bouret, S. G. & Ozanne, S. E. Sex and gender differences in developmental programming of metabolism. Mol. Metab. 15, 8–19 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Evered, L., Scott, D. A. & Silbert, B. Cognitive decline associated with anesthesia and surgery in the elderly: does this contribute to dementia prevalence? Curr. Opin. Psychiatry 30, 220–226 (2017).

    PubMed  Google Scholar 

  83. Schulte, P. J. et al. Association between exposure to anaesthesia and surgery and long-term cognitive trajectories in older adults: report from the Mayo Clinic Study of Aging. Br. J. Anaesth. 121, 398–405 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Steinmetz, J. & Rasmussen, L. S. Anesthesia and the risk of dementia in the elderly. Presse Med. 47, e45–e51 (2018).

    PubMed  Google Scholar 

  85. Butterfield, N. N., Graf, P., Ries, C. R. & MacLeod, B. A. The effect of repeated isoflurane anesthesia on spatial and psychomotor performance in young and aged mice. Anesth. Analg. 98, 1305–1311 (2004).

    CAS  PubMed  Google Scholar 

  86. Xu, H. et al. Smaller sized inhaled anesthetics have more potency on senescence-accelerated prone-8 mice compared with senescence-resistant-1 mice. J. Alzheimers Dis. 39, 29–34 (2014).

    PubMed  Google Scholar 

  87. Li, X. M. et al. Disruption of hippocampal neuregulin 1-ErbB4 signaling contributes to the hippocampus-dependent cognitive impairment induced by isoflurane in aged mice. Anesthesiology 121, 79–88 (2014).

    CAS  PubMed  Google Scholar 

  88. Li, R. L. et al. Postoperative impairment of cognitive function in old mice: a possible role for neuroinflammation mediated by HMGB1, S100B, and RAGE. J. Surg. Res. 185, 815–824 (2013).

    CAS  PubMed  Google Scholar 

  89. Campbell, J. E. & Drucker, D. J. Pharmacology, physiology, and mechanisms of incretin hormone action. Cell Metab. 17, 819–837 (2013).

    CAS  PubMed  Google Scholar 

  90. Linnemann, A. K. et al. Glucagon-like peptide-1 regulates cholecystokinin production in beta-cells to protect from apoptosis. Mol. Endocrinol. 29, 978–987 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Renner, S., Blutke, A., Streckel, E., Wanke, R. & Wolf, E. Incretin actions and consequences of incretin-based therapies: lessons from complementary animal models. J. Pathol. 238, 345–358 (2016).

    CAS  PubMed  Google Scholar 

  92. Leiter, E. H., Premdas, F., Harrison, D. E. & Lipson, L. G. Aging and glucose homeostasis in C57BL/6J male mice. FASEB J. 2, 2807–2811 (1988).

    CAS  PubMed  Google Scholar 

  93. Ennaceur, A. & Delacour, J. A new one-trial test for neurobiological studies of memory in rats. 1: behavioral data. Behav. Brain Res. 31, 47–59 (1988).

    CAS  PubMed  Google Scholar 

  94. Leger, M. et al. Object recognition test in mice. Nat. Prot. 8, 2531–2537 (2013).

    CAS  Google Scholar 

  95. Servick, K. Mouse microbes may make scientific studies harder to replicate. Science, https://doi.org/10.1126/science.aah7199 (2016).

  96. Cheng, C. J., Gelfond, J. A. L., Strong, R. & Nelson, J. F. Genetically heterogeneous mice exhibit a female survival advantage that is age- and site-specific: results from a large multi-site study. Aging Cell, e12905, https://doi.org/10.1111/acel.12905 (2019).

    PubMed  PubMed Central  Google Scholar 

  97. Kalueff, A. V., Keisala, T., Minasyan, A., Kuuslahti, M. & Tuohimaa, P. Temporal stability of novelty exploration in mice exposed to different open field tests. Behav. Process. 72, 104–112 (2006).

    Google Scholar 

  98. Heyser, C. J. & Chemero, A. Novel object exploration in mice: not all objects are created equal. Behav. Process. 89, 232–238 (2012).

    Google Scholar 

  99. Ennaceur, A. One-trial object recognition in rats and mice: methodological and theoretical issues. Behav. Brain Res. 215, 244–254 (2010).

    CAS  PubMed  Google Scholar 

  100. Raber, J. Novel images and novel locations of familiar images as sensitive translational cognitive tests in humans. Behav. Brain Res. 285, 53–59 (2015).

    PubMed  Google Scholar 

  101. Frick, K. M. & Gresack, J. E. Sex differences in the behavioral response to spatial and object novelty in adult C57BL/6 mice. Behav. Neurosci. 117, 1283–1291 (2003).

    PubMed  Google Scholar 

  102. Prut, L. & Belzung, C. The open field as a paradigm to measure the effects of drugs on anxiety-like behaviors: a review. Eur. J. Pharmacol. 463, 3–33 (2003).

    CAS  PubMed  Google Scholar 

  103. Klein, S. M. et al. Noninvasive in vivo assessment of muscle impairment in the mdx mouse model—a comparison of two common wire hanging methods with two different results. J. Neurosci. Methods 203, 292–297 (2012).

    CAS  PubMed  Google Scholar 

  104. Karp, N. A., Segonds-Pichon, A., Gerdin, A. K., Ramirez-Solis, R. & White, J. K. The fallacy of ratio correction to address confounding factors. Lab. Anim. 46, 245–252 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Gregg, T. et al. Pancreatic beta-cells from mice offset ageaAssociated mitochondrial deficiency with reduced KATP channel activity. Diabetes 65, 2700–2710 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Schnelle, M. et al. Echocardiographic evaluation of diastolic function in mouse models of heart disease. J. Mol. Cell. Cardiol. 114, 20–28 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Majumder, S. et al. Lifelong rapamycin administration ameliorates age-dependent cognitive deficits by reducing IL-1beta and enhancing NMDA signaling. Aging Cell 11, 326–335 (2012).

    CAS  PubMed  Google Scholar 

  108. Lin, A. L. et al. Chronic rapamycin restores brain vascular integrity and function through NO synthase activation and improves memory in symptomatic mice modeling Alzheimer’s disease. J. Cereb. Blood Flow. Metab. 33, 1412–1421 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Ozcelik, S. et al. Rapamycin attenuates the progression of tau pathology in P301S tau transgenic mice. PLoS ONE 8, e62459 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Caccamo, A., Majumder, S., Richardson, A., Strong, R. & Oddo, S. Molecular interplay between mammalian target of rapamycin (mTOR), amyloid-beta, and Tau: effects on cognitive impairments. J. Biol. Chem. 285, 13107–13120 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Lee, A. S. et al. Aged skeletal muscle retains the ability to fully regenerate functional architecture. Bioarchitecture 3, 25–37 (2013).

    PubMed  PubMed Central  Google Scholar 

  112. van Norren, K. et al. Behavioural changes are a major contributing factor in the reduction of sarcopenia in caloric-restricted ageing mice. J. Cachexia Sarcopenia Muscle 6, 253–268 (2015).

    PubMed  PubMed Central  Google Scholar 

  113. Ge, X. et al. Grip strength is potentially an early indicator of age-related decline in mice. Pathobiol. Aging Age Relat. Dis. 6, 32981 (2016).

    PubMed  Google Scholar 

  114. Hirsch, C. H., Buzkova, P., Robbins, J. A., Patel, K. V. & Newman, A. B. Predicting late-life disability and death by the rate of decline in physical performance measures. Age Ageing 41, 155–161 (2012).

    PubMed  Google Scholar 

  115. Bogue, M. A. et al. Accessing data resources in the mouse phenome database for genetic analysis of murine life span and health span. J. Gerontol. A Biol. Sci. Med. Sci. 71, 170–177 (2016).

    CAS  PubMed  Google Scholar 

  116. Xu, M. et al. JAK inhibition alleviates the cellular senescence-associated secretory phenotype and frailty in old age. Proc. Natl Acad. Sci. USA 112, E6301–6310 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Peterson, R. L., Parkinson, K. C. & Mason, J. B. Manipulation of ovarian function significantly influenced sarcopenia in postreproductive-age mice. J. Transplant. 2016, 4570842 (2016).

    PubMed  PubMed Central  Google Scholar 

  118. Graber, T. G., Ferguson-Stegall, L., Kim, J. H. & Thompson, L. V. C57BL/6 neuromuscular healthspan scoring system. J. Gerontol. A Biol. Sci. Med. Sci. 68, 1326–1336 (2013).

    PubMed  PubMed Central  Google Scholar 

  119. Simmons, D. A. et al. A small molecule TrkB ligand reduces motor impairment and neuropathology in R6/2 and BACHD mouse models of Huntington’s disease. J. Neurosci. 33, 18712–18727 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Jeong, H. et al. Sirt1 mediates neuroprotection from mutant huntingtin by activation of the TORC1 and CREB transcriptional pathway. Nat. Med. 18, 159–165 (2011).

    PubMed  Google Scholar 

  121. Zhang, Y. et al. Rapamycin extends life and health in C57BL/6 mice. J. Gerontol. A Biol. Sci. Med. Sci. 69, 119–130 (2014).

    CAS  PubMed  Google Scholar 

  122. Cummings, N. E. & Lamming, D. W. Regulation of metabolic health and aging by nutrient-sensitive signaling pathways. Mol. Cell. Endocrinol. 455, 13–22 (2017).

    CAS  PubMed  Google Scholar 

  123. Lamming, D. W. & Anderson, R. M. Metabolic effects of caloric restriction in eLS. (John Wiley & Sons, Ltd, 2014).

  124. Malloy, V. L. et al. Methionine restriction decreases visceral fat mass and preserves insulin action in aging male Fischer 344 rats independent of energy restriction. Aging Cell 5, 305–314 (2006).

    CAS  PubMed  Google Scholar 

  125. Green, C. L. & Lamming, D. W. Regulation of metabolic health by essential dietary amino acids. Mech. Ageing Dev. 177, 186–200 (2018).

    PubMed  PubMed Central  Google Scholar 

  126. Fang, Y. et al. Duration of rapamycin treatment has differential effects on metabolism in mice. Cell Metab. 17, 456–462 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Brewer, R. A., Gibbs, V. K. & Smith, D. L. Jr. Targeting glucose metabolism for healthy aging. Nutr. Healthy Aging 4, 31–46 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  128. Tschop, M. H. et al. A guide to analysis of mouse energy metabolism. Nat. Methods 9, 57–63 (2011).

    PubMed  PubMed Central  Google Scholar 

  129. Lusk, G. ANIMAL CALORIMETRY: Twenty-Fourth Paper. ANALYSIS OF THE OXIDATION OF MIXTURES OF CARBOHYDRATE AND FAT. J. Biol. Chem. 59, 41–42 (1924).

    CAS  Google Scholar 

  130. Goodpaster, B. H. & Sparks, L. M. Metabolic flexibility in health and disease. Cell Metab. 25, 1027–1036 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Solon-Biet, S. M. et al. Dietary protein to carbohydrate ratio and caloric restriction: comparing metabolic outcomes in mice. Cell Rep. 11, 1529–1534 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  132. U.S. Department of Health and Human Services. Health, United States, 2016 With Chartbook on Long-term Trends in Health (National Center for Heath Statistics, 2016).

  133. Taffet, G. E., Pham, T. T. & Hartley, C. J. The age-associated alterations in late diastolic function in mice are improved by caloric restriction. J. Gerontol. A Biol. Sci. Med. Sci. 52, B285–290 (1997).

    CAS  PubMed  Google Scholar 

  134. Casaclang-Verzosa, G., Enriquez-Sarano, M., Villaraga, H. R. & Miller, J. D. Echocardiographic approaches and protocols for comprehensive phenotypic characterization of valvular heart disease in mice. J. Vis. Exp. https://doi.org/10.3791/54110 (2017).

Download references

Acknowledgements

We thank K. R. McGreevy from the Cajal Institute for her assistance with Fig. 3, and D. E. Cohen for enabling D.W.L. to attend the 2017 MouseAGE annual meeting. This article is based on work from COST Action (BM1402: MouseAGE), supported by COST (European Cooperation in Science and Technology; I.B., P.K.P., D.E., L.J.T.). Part of this work has been funded by the European Union Research and Innovation Program Horizon 2020 (grant agreement number 730879; I.B.). The Lamming laboratory is supported in part by the NIH (AG051974, AG056771 and AG062328) and by the US Department of Veterans Affairs (I01-BX004031), and this work was supported using facilities and resources from the William S. Middleton Memorial Veterans Hospital. D.E. was supported by a grant from the Helmholtz-Future Topic ‘Aging and Metabolic Programming’ (AMPro), the Mayo clinic is funded by Robert and Arlene Kogod, the Connor Group, and partially by NIA grant AG13925. R.dC., C.D.G., and I.N. are supported by the Intramural Research Program of the NIA/NIH. This work does not necessarily represent the official views of the NIH, the Department of Veterans Affairs or the United States Government.

Author information

Authors and Affiliations

Authors

Contributions

I.B., R.d.C., D.E., C.D.G., I.N.-E., S.M., P.K.P., T.T., J.L.T., A.L. and D.W.L. contributed equally to the development of the protocols. I.B. and D.L. wrote the manuscript. All other authors edited and approved the final manuscript.

Corresponding authors

Correspondence to I. Bellantuono or D. W. Lamming.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Protocols thanks Erin R. Hascup, Susan Howlett and the other, anonymous, reviewer(s) 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.

Related links

Key references using this protocol

Martin-Montalvo, A. et al. Nat. Commun. 4, 2192 (2013): https://doi.org/10.1038/ncomms3192.

Neff, F. et al. J. Clin. Invest. 123, 3272–3291 (2013): https://doi.org/10.1172/JCI67674.

Zhu, Y. et al. Aging Cell 14, 644–658 (2015): https://doi.org/10.1111/acel.12344.

Supplemantary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bellantuono, I., de Cabo, R., Ehninger, D. et al. A toolbox for the longitudinal assessment of healthspan in aging mice. Nat Protoc 15, 540–574 (2020). https://doi.org/10.1038/s41596-019-0256-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-019-0256-1

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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