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.

  • Review Article
  • Published:

Inflammaging: a new immune–metabolic viewpoint for age-related diseases

Abstract

Ageing and age-related diseases share some basic mechanistic pillars that largely converge on inflammation. During ageing, chronic, sterile, low-grade inflammation — called inflammaging — develops, which contributes to the pathogenesis of age-related diseases. From an evolutionary perspective, a variety of stimuli sustain inflammaging, including pathogens (non-self), endogenous cell debris and misplaced molecules (self) and nutrients and gut microbiota (quasi-self). A limited number of receptors, whose degeneracy allows them to recognize many signals and to activate the innate immune responses, sense these stimuli. In this situation, metaflammation (the metabolic inflammation accompanying metabolic diseases) is thought to be the form of chronic inflammation that is driven by nutrient excess or overnutrition; metaflammation is characterized by the same mechanisms underpinning inflammaging. The gut microbiota has a central role in both metaflammation and inflammaging owing to its ability to release inflammatory products, contribute to circadian rhythms and crosstalk with other organs and systems. We argue that chronic diseases are not only the result of ageing and inflammaging; these diseases also accelerate the ageing process and can be considered a manifestation of accelerated ageing. Finally, we propose the use of new biomarkers (DNA methylation, glycomics, metabolomics and lipidomics) that are capable of assessing biological versus chronological age in metabolic diseases.

Key points

  • According to geroscience, inflammation is one of the seven evolutionarily conserved mechanistic pillars of ageing that are shared by age-related diseases, including metabolic diseases.

  • Inflammaging is the long-term result of the chronic physiological stimulation of the innate immune system, which can become damaging during ageing — a period of life largely unpredicted by evolution.

  • Inflammaging is the by-product of the degeneracy of a few receptors that can sense a variety of non-self, self and quasi-self damage signals (or ‘garbage’) and activate the innate immune system.

  • Inflammaging and metaflammation largely share the same molecular mechanisms, in which metaflammation can be conceptualized as a specific situation of chronic inflammation caused by nutrient excess.

  • The gut microbiota has a central role in metaflammation and inflammaging, as it can release inflammatory products and contribute to the circadian rhythms and crosstalk with other organs and systems.

  • Biomarkers of biological age, such as DNA methylation, glycomics, metabolomics and lipidomics, can be successfully applied to metabolic diseases.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: The seven pillars of ageing.
Fig. 2: The bow tie architecture of the inflammaging machinery.
Fig. 3: The gut microbiota as a key modulator of nutrition and inflammation.
Fig. 4: Inflammaging, age trajectories and age-related diseases.

Similar content being viewed by others

References

  1. Kennedy, B. K. et al. Geroscience: linking aging to chronic disease. Cell 159, 709–713 (2014). This paper identifies seven fields in ageing research to expand geroscience research and to extend healthspan; the link between ageing and chronic disease is the central point of this paper.

    CAS  PubMed  PubMed Central  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

  3. Franceschi, C. et al. Inflamm-aging. An evolutionary perspective on immunosenescence. Ann. NY Acad. Sci. 908, 244–254 (2000). This paper conceptualizes the inflammaging theory, starting from an evolutionary insight; the term inflammaging is used here.

    CAS  PubMed  Google Scholar 

  4. Tauber, A. I. Timeline: Metchnikoff and the phagocytosis theory. Nat. Rev. Mol. Cell Biol. 4, 897–901 (2003).

    CAS  PubMed  Google Scholar 

  5. Vitale, G., Salvioli, S. & Franceschi, C. Oxidative stress and the ageing endocrine system. Nat. Rev. Endocrinol. 9, 228–240 (2013).

    CAS  PubMed  Google Scholar 

  6. Gregor, M. F. & Hotamisligil, G. S. Inflammatory mechanisms in obesity. Annu. Rev. Immunol. 29, 415–445 (2011).

    CAS  PubMed  Google Scholar 

  7. Darwin, C. On the origin of species by means of natural selection, or, The preservation of favoured races in the struggle for life (John Murray, 1859).

  8. Fumagalli, M. et al. Signatures of environmental genetic adaptation pinpoint pathogens as the main selective pressure through human evolution. PLoS Genet. 7, e1002355 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Vasseur, E. & Quintana-Murci, L. The impact of natural selection on health and disease: uses of the population genetics approach in humans. Evol. Appl. 6, 596–607 (2013).

    PubMed  PubMed Central  Google Scholar 

  10. Ottaviani, E. & Franceschi, C. The invertebrate phagocytic immunocyte: clues to a common evolution of immune and neuroendocrine systems. Immunol. Today 18, 169–174 (1997).

    CAS  PubMed  Google Scholar 

  11. Chung, S. et al. Preadipocytes mediate lipopolysaccharide-induced inflammation and insulin resistance in primary cultures of newly differentiated human adipocytes. Endocrinology 147, 5340–5351 (2006).

    CAS  PubMed  Google Scholar 

  12. Charrière, G. et al. Preadipocyte conversion to macrophage: evidence of plasticity. J. Biol. Chem. 278, 9850–9855 (2003).

    PubMed  Google Scholar 

  13. Hotamisligil, G. S. & Erbay, E. Nutrient sensing and inflammation in metabolic diseases. Nat. Rev. Immunol. 8, 923–934 (2008). This paper describes metabolically triggered inflammation (termed metaflammation), starting from a detailed evolutionary description of the evolutionary history of metabolic and immune responses.

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Hotamisligil, G. S. Inflammation and metabolic disorders. Nature 444, 860–867 (2006).

    CAS  PubMed  Google Scholar 

  15. Hotamisligil, G. S. Inflammation, metaflammation and immunometabolic disorders. Nature 542, 177–185 (2017).

    CAS  PubMed  Google Scholar 

  16. Ye, J. & Keller, J. N. Regulation of energy metabolism by inflammation: a feedback response in obesity and calorie restriction. Aging 2, 361–368 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Lochmiller, R. L. & Deerenberg, C. Trade-offs in evolutionary immunology: just what is the cost of immunity? Oikos 88, 87–98 (2000).

    Google Scholar 

  18. Navarrete, A., van Schaik, C. P. & Isler, K. Energetics and the evolution of human brain size. Nature 480, 91–93 (2011).

    CAS  PubMed  Google Scholar 

  19. Potts, R. Evolution: big brains explained. Nature 480, 43–44 (2011).

    CAS  PubMed  Google Scholar 

  20. Ottaviani, E., Malagoli, D., Capri, M. & Franceschi, C. Ecoimmunology: is there any room for the neuroendocrine system? Bioessays 30, 868–874 (2008). The evolutionary considerations of this paper introduce the importance of the common origin of the immune and neuroendocrine system for the study of stress response and human health.

    PubMed  Google Scholar 

  21. Michael Muehlenbein, P. Human Evolutionary Biology (Cambridge Univ. Press, 2010).

  22. Sansoni, P. et al. The immune system in extreme longevity. Exp. Gerontol. 43, 61–65 (2008).

    CAS  PubMed  Google Scholar 

  23. Vescovini, R. et al. Naïve and memory CD8 T cell pool homeostasis in advanced aging: impact of age and of antigen-specific responses to cytomegalovirus. Age (Dordr.) 36, 625–640 (2014).

    CAS  Google Scholar 

  24. Sansoni, P. et al. New advances in CMV and immunosenescence. Exp. Gerontol. 55, 54–62 (2014).

    PubMed  Google Scholar 

  25. Low, H. et al. Cytomegalovirus restructures lipid rafts via a US28/CDC42-mediated pathway, enhancing cholesterol efflux from host cells. Cell Rep. 16, 186–200 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Yu, Y., Clippinger, A. J. & Alwine, J. C. Viral effects on metabolism: changes in glucose and glutamine utilization during human cytomegalovirus infection. Trends Microbiol. 19, 360–367 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Chen, S. et al. Cytomegalovirus seropositivity is associated with glucose regulation in the oldest old. Results from the Leiden 85-plus Study. Immun. Ageing 9, 18 (2012).

    PubMed  PubMed Central  Google Scholar 

  28. Almanzar, G. et al. Long-term cytomegalovirus infection leads to significant changes in the composition of the CD8+ T-cell repertoire, which may be the basis for an imbalance in the cytokine production profile in elderly persons. J. Virol. 79, 3675–3683 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Yurochko, A. D. & Huang, E. S. Human cytomegalovirus binding to human monocytes induces immunoregulatory gene expression. J. Immunol. 162, 4806–4816 (1999).

    CAS  PubMed  Google Scholar 

  30. Lohr, J. M. & Oldstone, M. B. A. Detection of cytomegalovirus nucleic acid sequences in pancreas in type 2 diabetes. Lancet 336, 644–648 (1990).

    CAS  PubMed  Google Scholar 

  31. Donath, M. Y. & Shoelson, S. E. Type 2 diabetes as an inflammatory disease. Nat. Rev. Immunol. 11, 98–107 (2011).

    CAS  PubMed  Google Scholar 

  32. Tieri, P. et al. Network, degeneracy and bow tie integrating paradigms and architectures to grasp the complexity of the immune system. Theor. Biol. Med. Model. 7, 32 (2010).

    PubMed  PubMed Central  Google Scholar 

  33. Edelman, G. M. & Gally, J. A. Degeneracy and complexity in biological systems. Proc. Natl Acad. Sci. USA 98, 13763–13768 (2001). In this paper, Gerald Edelman and Joseph Gally describe the concept of degeneracy; this paper discusses the degeneracy of many systems and the idea that degeneracy is also important for natural selection (Nobel Prize for Medicine in 1972).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Gay, N. J. & Gangloff, M. Structure and function of Toll receptors and their ligands. Annu. Rev. Biochem. 76, 141–165 (2007).

    CAS  PubMed  Google Scholar 

  35. Ottaviani, E., Malagoli, D. & Franceschi, C. Common evolutionary origin of the immune and neuroendocrine systems: from morphological and functional evidence to in silico approaches. Trends Immunol. 28, 497–502 (2007).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  37. Mathur, V. et al. Activation of the STING-dependent type I interferon response reduces microglial reactivity and neuroinflammation. Neuron 96, 1290–1302.e6 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Csete, M. & Doyle, J. Bow ties, metabolism and disease. Trends Biotechnol. 22, 446–450 (2004).

    CAS  PubMed  Google Scholar 

  39. Lee, J. Y. et al. Saturated fatty acid activates but polyunsaturated fatty acid inhibits Toll-like receptor 2 dimerized with Toll-like receptor 6 or 1. J. Biol. Chem. 279, 16971–16979 (2004).

    CAS  PubMed  Google Scholar 

  40. Huang, S. et al. Saturated fatty acids activate TLR-mediated proinflammatory signaling pathways. J. Lipid Res. 53, 2002–2013 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Zhao, L., Lee, J. Y. & Hwang, D. H. Inhibition of pattern recognition receptor-mediated inflammation by bioactive phytochemicals: a review of recent research. Nutr. Rev. 69, 310–320 (2011).

    PubMed  Google Scholar 

  42. Moossavi, S. Gliadin is an uncatalogued Toll-like receptor ligand. J. Med. Hypotheses Ideas 8, 44–47 (2014).

    CAS  Google Scholar 

  43. Wang, X. et al. Morphine activates neuroinflammation in a manner parallel to endotoxin. Proc. Natl Acad. Sci. USA 109, 6325–6330 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Lewis, S. S. et al. Glucuronic acid and the ethanol metabolite ethyl-glucuronide cause toll-like receptor 4 activation and enhanced pain. Brain Behav. Immun. 30, 24–32 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Byun, E.-B., Choi, H.-G., Sung, N.-Y. & Byun, E.-H. Green tea polyphenol epigallocatechin-3-gallate inhibits TLR4 signaling through the 67-kDa laminin receptor on lipopolysaccharide-stimulated dendritic cells. Biochem. Biophys. Res. Commun. 426, 480–485 (2012).

    CAS  PubMed  Google Scholar 

  46. Park, H.-J. et al. Phenethyl isothiocyanate regulates inflammation through suppression of the TRIF-dependent signaling pathway of Toll-like receptors. Life Sci. 92, 793–798 (2013).

    CAS  PubMed  Google Scholar 

  47. Pasparakis, M. & Vandenabeele, P. Necroptosis and its role in inflammation. Nature 517, 311–320 (2015).

    CAS  PubMed  Google Scholar 

  48. Revelo, X. S. et al. Nucleic acid-targeting pathways promote inflammation in obesity-related insulin resistance. Cell Rep. 16, 717–730 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Nishimoto, S. et al. Obesity-induced DNA released from adipocytes stimulates chronic adipose tissue inflammation and insulin resistance. Sci. Adv. 2, e1501332 (2016).

    PubMed  PubMed Central  Google Scholar 

  50. Ebersole, J. L. et al. Aging, inflammation, immunity and periodontal disease. Periodontol. 2000 72, 54–75 (2016).

    PubMed  Google Scholar 

  51. 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).

    PubMed  Google Scholar 

  52. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Biagi, E. et al. Through ageing, and beyond: gut microbiota and inflammatory status in seniors and centenarians. PLoS ONE 5, e10667 (2010).

    PubMed  PubMed Central  Google Scholar 

  54. Biagi, E., Candela, M., Franceschi, C. & Brigidi, P. The aging gut microbiota: new perspectives. Ageing Res. Rev. 10, 428–429 (2011).

    PubMed  Google Scholar 

  55. Cevenini, E., Monti, D. & Franceschi, C. Inflamm-ageing. Curr. Opin. Clin. Nutr. Metab. Care 16, 14–20 (2013).

    CAS  PubMed  Google Scholar 

  56. Biagi, E. et al. Gut microbiota and extreme longevity. Curr. Biol. 26, 1480–1485 (2016).

    CAS  PubMed  Google Scholar 

  57. Lee, Y. K. & Mazmanian, S. K. Has the microbiota played a critical role in the evolution of the adaptive immune system? Science 330, 1768–1773 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Groussin, M. et al. Unraveling the processes shaping mammalian gut microbiomes over evolutionary time. Nat. Commun. 8, 14319 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Moeller, A. H. et al. Cospeciation of gut microbiota with hominids. Science 353, 380–382 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Santoro, A. et al. Gut microbiota changes in the extreme decades of human life: a focus on centenarians. Cell. Mol. Life Sci. 75, 129–148 (2018).

    CAS  PubMed  Google Scholar 

  61. Franceschi, C. et al. Immunobiography and the heterogeneity of immune responses in the elderly: a focus on inflammaging and trained immunity. Front. Immunol. 8, 982 (2017).

    PubMed  PubMed Central  Google Scholar 

  62. Kundu, P., Blacher, E., Elinav, E. & Pettersson, S. Our gut microbiome: the evolving inner self. Cell 171, 1481–1493 (2017).

    CAS  PubMed  Google Scholar 

  63. Fransen, F. et al. Aged gut microbiota contributes to systemical inflammaging after transfer to germ-free mice. Front. Immunol. 8, 1385 (2017).

    PubMed  PubMed Central  Google Scholar 

  64. Thevaranjan, N. et al. Age-associated microbial dysbiosis promotes intestinal permeability, systemic inflammation, and macrophage dysfunction. Cell Host Microbe 21, 455–466.e4 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Everard, A. et al. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc. Natl Acad. Sci. USA 110, 9066–9071 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Kuehbacher, T. et al. Intestinal TM7 bacterial phylogenies in active inflammatory bowel disease. J. Med. Microbiol. 57, 1569–1576 (2008).

    CAS  PubMed  Google Scholar 

  67. Mukhopadhya, I., Hansen, R., El-Omar, E. M. & Hold, G. L. IBD—what role do Proteobacteria play? Nat. Rev. Gastroenterol. Hepatol. 9, 219–230 (2012).

    CAS  PubMed  Google Scholar 

  68. Rubio-Ruiz, M. E., Peredo-Escárcega, A. E., Cano-Martínez, A. & Guarner-Lans, V. An evolutionary perspective of nutrition and inflammation as mechanisms of cardiovascular disease. Int. J. Evol. Biol. 2015, 179791 (2015).

    PubMed  PubMed Central  Google Scholar 

  69. Ingram, D. K. & de Cabo, R. Calorie restriction in rodents: caveats to consider. Ageing Res. Rev. 39, 15–28 (2017). This review describes many crucial points that still need to be addressed in calorie restriction (such as the relation between calorie restriction and cognitive decline, the negative effect of calorie restriction and the role of genetics and gender in calorie restriction response) in future studies.

    PubMed  PubMed Central  Google Scholar 

  70. Mercken, E. M. et al. Conserved and species-specific molecular denominators in mammalian skeletal muscle aging. NPJ Aging Mech. Dis. 3, 8 (2017).

    PubMed  PubMed Central  Google Scholar 

  71. Lee, C. & Longo, V. Dietary restriction with and without caloric restriction for healthy aging. F1000Res. 5, 117 (2016).

    Google Scholar 

  72. Barzilai, N., Huffman, D. M., Muzumdar, R. H. & Bartke, A. The critical role of metabolic pathways in aging. Diabetes 61, 1315–1322 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Ristow, M. & Schmeisser, K. Mitohormesis: promoting health and lifespan by increased levels of reactive oxygen species (ROS). Dose Response 12, 288–341 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Rose, G., Santoro, A. & Salvioli, S. Mitochondria and mitochondria-induced signalling molecules as longevity determinants. Mech. Ageing Dev. 165, 115–128 (2017).

    CAS  PubMed  Google Scholar 

  75. Mirzaei, H., Suarez, J. A. & Longo, V. D. Protein and amino acid restriction, aging and disease: from yeast to humans. Trends Endocrinol. Metab. 25, 558–566 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Das, S. K., Balasubramanian, P. & Weerasekara, Y. K. Nutrition modulation of human aging: the calorie restriction paradigm. Mol. Cell. Endocrinol. 455, 148–157 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Keil, G., Cummings, E. & de Magalhães, J. P. Being cool: how body temperature influences ageing and longevity. Biogerontology 16, 383–397 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Walford, R. L. & Spindler, S. R. The response to calorie restriction in mammals shows features also common to hibernation: a cross-adaptation hypothesis. J. Gerontol. A Biol. Sci. Med. Sci. 52, B179–B183 (1997). This paper investigates commonalities among physiological events that occur during hibernation and under calorie restriction.

    CAS  PubMed  Google Scholar 

  79. Xu, R. et al. Hibernating squirrel muscle activates the endurance exercise pathway despite prolonged immobilization. Exp. Neurol. 247, 392–401 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Xu, Y. et al. Molecular signatures of mammalian hibernation: comparisons with alternative phenotypes. BMC Genomics 14, 567 (2013).

    PubMed  PubMed Central  Google Scholar 

  81. Tognini, P., Murakami, M. & Sassone-Corsi, P. Interplay between microbes and the circadian clock. Cold Spring Harb. Perspect. Biol. https://doi.org/10.1101/cshperspect.a028365 (2017).

    Article  Google Scholar 

  82. Ross, R. Atherosclerosis — an inflammatory disease. N. Engl. J. Med. 340, 115–126 (1999).

    CAS  PubMed  Google Scholar 

  83. Alipour, A. et al. Leukocyte activation by triglyceride-rich lipoproteins. Arterioscler. Thromb. Vasc. Biol. 28, 792–797 (2008).

    CAS  PubMed  Google Scholar 

  84. Sampson, M. J., Davies, I. R., Brown, J. C., Ivory, K. & Hughes, D. A. Monocyte and neutrophil adhesion molecule expression during acute hyperglycemia and after antioxidant treatment in type 2 diabetes and control patients. Arterioscler. Thromb. Vasc. Biol. 22, 1187–1193 (2002).

    CAS  PubMed  Google Scholar 

  85. Libby, P. Inflammation in atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 32, 2045–2051 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. van Oostrom, A. J. H. H. M. et al. Activation of leukocytes by postprandial lipemia in healthy volunteers. Atherosclerosis 177, 175–182 (2004).

    PubMed  Google Scholar 

  87. Wang, Y. I. et al. Triglyceride-rich lipoprotein modulates endothelial vascular cell adhesion molecule (VCAM)-1 expression via differential regulation of endoplasmic reticulum stress. PLoS ONE 8, e78322 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Gower, R. M. et al. CD11c/CD18 expression is upregulated on blood monocytes during hypertriglyceridemia and enhances adhesion to vascular cell adhesion molecule-1. Arterioscler. Thromb. Vasc. Biol. 31, 160–166 (2011).

    CAS  PubMed  Google Scholar 

  89. Higgins, L. J. & Rutledge, J. C. Inflammation associated with the postprandial lipolysis of triglyceride-rich lipoproteins by lipoprotein lipase. Curr. Atheroscler. Rep. 11, 199–205 (2009).

    CAS  PubMed  Google Scholar 

  90. van Oostrom, A. J. H. H. M. et al. Postprandial recruitment of neutrophils may contribute to endothelial dysfunction. J. Lipid Res. 44, 576–583 (2003).

    PubMed  Google Scholar 

  91. Erridge, C., Attina, T., Spickett, C. M. & Webb, D. J. A high-fat meal induces low-grade endotoxemia: evidence of a novel mechanism of postprandial inflammation. Am. J. Clin. Nutr. 86, 1286–1292 (2007).

    CAS  PubMed  Google Scholar 

  92. Manco, M., Putignani, L. & Bottazzo, G. F. Gut microbiota, lipopolysaccharides, and innate immunity in the pathogenesis of obesity and cardiovascular risk. Endocr. Rev. 31, 817–844 (2010).

    CAS  PubMed  Google Scholar 

  93. Musso, G., Gambino, R. & Cassader, M. Interactions between gut microbiota and host metabolism predisposing to obesity and diabetes. Annu. Rev. Med. 62, 361–380 (2011).

    CAS  PubMed  Google Scholar 

  94. Grosicki, G. J., Fielding, R. A. & Lustgarten, M. S. Gut microbiota contribute to age-related changes in skeletal muscle size, composition, and function: biological basis for a gut-muscle axis. Calcif. Tissue Int. 102, 433–442 (2017).

    PubMed  PubMed Central  Google Scholar 

  95. Giordano, A. et al. Obese adipocytes show ultrastructural features of stressed cells and die of pyroptosis. J. Lipid Res. 54, 2423–2436 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Li, H., Lee, J., He, C., Zou, M.-H. & Xie, Z. Suppression of the mTORC1/STAT3/Notch1 pathway by activated AMPK prevents hepatic insulin resistance induced by excess amino acids. Am. J. Physiol. Endocrinol. Metab. 306, E197–E209 (2014).

    CAS  PubMed  Google Scholar 

  97. Lee, P. L., Jung, S. M. & Guertin, D. A. The complex roles of mechanistic target of rapamycin in adipocytes and beyond. Trends Endocrinol. Metab. 28, 319–339 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Ozcan, U. et al. Loss of the tuberous sclerosis complex tumor suppressors triggers the unfolded protein response to regulate insulin signaling and apoptosis. Mol. Cell 29, 541–551 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Prattichizzo, F. et al. Inflammageing and metaflammation: the yin and yang of type 2 diabetes. Ageing Res. Rev. 41, 1–17 (2017).

    PubMed  Google Scholar 

  100. Franceschi, C. et al. Inflammaging and anti-inflammaging: a systemic perspective on aging and longevity emerged from studies in humans. Mech. Ageing Dev. 128, 92–105 (2007). This review describes not only the role of pro-inflammatory mechanisms in ageing but also the importance of the balance with anti-inflammatory factors (called anti-inflammaging) in healthy ageing.

    CAS  PubMed  Google Scholar 

  101. Franceschi, C. et al. The continuum of aging and age-related diseases: common mechanisms but different rates. Front. Med. 5, 61 (2018).

    Google Scholar 

  102. Olivieri, F. et al. MicroRNAs linking inflamm-aging, cellular senescence and cancer. Ageing Res. Rev. 12, 1056–1068 (2013).

    CAS  PubMed  Google Scholar 

  103. Al-Nedawi, K. et al. Gut commensal microvesicles reproduce parent bacterial signals to host immune and enteric nervous systems. FASEB J. 29, 684–695 (2015).

    CAS  PubMed  Google Scholar 

  104. Garagnani, P. et al. Methylation of ELOVL2 gene as a new epigenetic marker of age. Aging Cell 11, 1132–1134 (2012).

    CAS  PubMed  Google Scholar 

  105. Bocklandt, S. et al. Epigenetic predictor of age. PLoS ONE 6, e14821 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Horvath, S. DNA methylation age of human tissues and cell types. Genome Biol. 14, R115 (2013). This is one of the first papers to introduce the field of biological age, which is measured through DNA methylation profile and is described as methylation age in many human tissues.

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  108. Weidner, C. et al. Aging of blood can be tracked by DNA methylation changes at just three CpG sites. Genome Biol. 15, R24 (2014).

    PubMed  PubMed Central  Google Scholar 

  109. Bacalini, M. G. et al. Identification of a DNA methylation signature in blood cells from persons with Down Syndrome. Aging (Albany NY) 7, 82–96 (2015).

    CAS  Google Scholar 

  110. Guastafierro, T. et al. Genome-wide DNA methylation analysis in blood cells from patients with Werner syndrome. Clin. Epigenetics 9, 92 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Horvath, S. et al. Accelerated epigenetic aging in Down syndrome. Aging Cell 14, 491–495 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Horvath, S. et al. Decreased epigenetic age of PBMCs from Italian semi-supercentenarians and their offspring. Aging (Albany NY) 7, 1159–1170 (2015).

    CAS  Google Scholar 

  113. Horvath, S. & Ritz, B. R. Increased epigenetic age and granulocyte counts in the blood of Parkinson’s disease patients. Aging (Albany NY) 7, 1130–1142 (2015).

    CAS  Google Scholar 

  114. Horvath, S. et al. An epigenetic clock analysis of race/ethnicity, sex, and coronary heart disease. Genome Biol. 17, 171 (2016).

    PubMed  PubMed Central  Google Scholar 

  115. Petkovich, D. A. et al. Using DNA methylation profiling to evaluate biological age and longevity interventions. Cell Metab. 25, 954–960.e6 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Bacalini, M. G. et al. Systemic age-associated DNA hypermethylation of ELOVL2 gene: in vivo and in vitro evidences of a cell replication process. J. Gerontol. A Biol. Sci. Med. Sci. 72, 1015–1023 (2016).

    PubMed Central  Google Scholar 

  117. WHI-EMPC Investigators et al. DNA methylation signatures of chronic low-grade inflammation are associated with complex diseases. Genome Biol. 17, 255 (2016).

    PubMed Central  Google Scholar 

  118. Nevalainen, T. et al. Transcriptomic and epigenetic analyses reveal a gender difference in aging-associated inflammation: the Vitality 90+ study. Age (Dordr.) 37, 9814 (2015).

    CAS  Google Scholar 

  119. Xu, C.-J. et al. DNA methylation in childhood asthma: an epigenome-wide meta-analysis. Lancet Respir. Med. 6, 379–388 (2018).

    CAS  PubMed  Google Scholar 

  120. Nojima, M. et al. Correlation between global methylation level of peripheral blood leukocytes and serum C reactive protein level modified by MTHFR polymorphism: a cross-sectional study. BMC Cancer 18, 184 (2018).

    PubMed  PubMed Central  Google Scholar 

  121. Dall’Olio, F. et al. N-Glycomic biomarkers of biological aging and longevity: a link with inflammaging. Ageing Res. Rev. 12, 685–698 (2013).

    PubMed  Google Scholar 

  122. Vanhooren, V. et al. N-Glycomic changes in serum proteins during human aging. Rejuvenation Res. 10, 521–531a (2007). This paper introduces the use of N-glycan level changes as a measurement of general health or for age-related disease progression.

    PubMed  Google Scholar 

  123. Vanhooren, V. et al. Serum N-glycan profile shift during human ageing. Exp. Gerontol. 45, 738–743 (2010).

    CAS  PubMed  Google Scholar 

  124. Borelli, V. et al. Plasma N-glycome signature of Down Syndrome. J. Proteome Res. 14, 4232–4245 (2015).

    CAS  PubMed  Google Scholar 

  125. Holst, S. et al. in High-Throughput Glycomics and Glycoproteomics (eds Lauc, G. & Wuhrer, M.) 185–196 (Springer New York, 2017).

  126. Krištic´, J. et al. Glycans are a novel biomarker of chronological and biological ages. J. Gerontol. A Biol. Sci. Med. Sci. 69, 779–789 (2014).

    PubMed  Google Scholar 

  127. Menni, C. et al. Glycosylation profile of immunoglobulin G is cross-sectionally associated with cardiovascular disease risk score and subclinical atherosclerosis in two independent cohorts. Circ. Res. 122, 1555–1564 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  128. Testa, R. et al. N-glycomic changes in serum proteins in Type 2 diabetes mellitus correlate with complications and with metabolic syndrome parameters. PLoS ONE 10, e0119983 (2015).

    PubMed  PubMed Central  Google Scholar 

  129. de Kreutzenberg, S. V. et al. Metformin improves putative longevity effectors in peripheral mononuclear cells from subjects with prediabetes. A randomized controlled trial. Nutr. Metab. Cardiovasc. Dis. 25, 686–693 (2015).

    PubMed  Google Scholar 

  130. Keser, T. et al. Increased plasma N-glycome complexity is associated with higher risk of type 2 diabetes. Diabetologia 60, 2352–2360 (2017).

    CAS  PubMed  Google Scholar 

  131. Plomp, R. et al. Subclass-specific IgG glycosylation is associated with markers of inflammation and metabolic health. Sci. Rep. 7, 12325 (2017).

    PubMed  PubMed Central  Google Scholar 

  132. Wahl, A. et al. Genome-wide association study on immunoglobulin G glycosylation patterns. Front. Immunol. 9, 277 (2018).

    PubMed  PubMed Central  Google Scholar 

  133. Montoliu, I. et al. Serum profiling of healthy aging identifies phospho- and sphingolipid species as markers of human longevity. Aging (Albany NY) 6, 9–25 (2014).

    CAS  Google Scholar 

  134. Gonzalez-Covarrubias, V. et al. Lipidomics of familial longevity. Aging Cell 12, 426–434 (2013).

    CAS  PubMed  Google Scholar 

  135. Li-Gao, R. et al. Postprandial metabolite profiles associated with type 2 diabetes clearly stratify individuals with impaired fasting glucose. Metabolomics 14, 13 (2018).

    PubMed  Google Scholar 

  136. Liu, J. et al. Metabolomics based markers predict type 2 diabetes in a 14-year follow-up study. Metabolomics 13, 104 (2017).

    PubMed  PubMed Central  Google Scholar 

  137. Okekunle, A. P. et al. Abnormal circulating amino acid profiles in multiple metabolic disorders. Diabetes Res. Clin. Pract. 132, 45–58 (2017).

    CAS  PubMed  Google Scholar 

  138. Wang, S. M. et al. Identification of serum metabolites associated with obesity and traditional risk factors for metabolic disease in Chinese adults. Nutr. Metab. Cardiovasc. Dis. 28, 112–118 (2017).

    PubMed  Google Scholar 

  139. Leal-Witt, M. J. et al. Untargeted metabolomics identifies a plasma sphingolipid-related signature associated with lifestyle intervention in prepubertal children with obesity. Int. J. Obes. 42, 72–78 (2017).

    Google Scholar 

  140. BIOS Consortium et al. Blood lipids influence DNA methylation in circulating cells. Genome Biol. 17, 138 (2016).

    PubMed Central  Google Scholar 

  141. Martucci, M. et al. Mediterranean diet and inflammaging within the hormesis paradigm. Nutr. Rev. 75, 442–455 (2017).

    PubMed  PubMed Central  Google Scholar 

  142. Lee, R. J. et al. Bacterial d-amino acids suppress sinonasal innate immunity through sweet taste receptors in solitary chemosensory cells. Sci. Signal. 10, eaam7703 (2017).

    PubMed  PubMed Central  Google Scholar 

  143. Lee, R. J. et al. T2R38 taste receptor polymorphisms underlie susceptibility to upper respiratory infection. J. Clin. Invest. 122, 4145–4159 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  144. Prince, A. The bitter taste of infection. J. Clin. Invest. 122, 3847–3849 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  145. Harris, N. The enigmatic tuft cell in immunity. Science 351, 1264–1265 (2016).

    CAS  PubMed  Google Scholar 

  146. Howitt, M. R. et al. Tuft cells, taste-chemosensory cells, orchestrate parasite type 2 immunity in the gut. Science 351, 1329–1333 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  147. Libert, S. & Pletcher, S. D. Modulation of longevity by environmental sensing. Cell 131, 1231–1234 (2007).

    CAS  PubMed  Google Scholar 

  148. Waterson, M. J. et al. Water sensor ppk28 modulates Drosophila lifespan and physiology through AKH signaling. Proc. Natl Acad. Sci. USA 111, 8137–8142 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  149. Zhang, Q. et al. Circulating mitochondrial DAMPs cause inflammatory responses to injury. Nature 464, 104–107 (2010). This paper describes in detail the consequences of the evolutionarily conserved similarities among DAMPs and bacterial PAMPs and their role in inflammation.

    CAS  PubMed  PubMed Central  Google Scholar 

  150. Pinti, M. et al. Circulating mitochondrial DNA increases with age and is a familiar trait: implications for ‘inflamm-aging’. Eur. J. Immunol. 44, 1552–1562 (2014).

    CAS  PubMed  Google Scholar 

  151. Weinberg, S. E., Sena, L. A. & Chandel, N. S. Mitochondria in the regulation of innate and adaptive immunity. Immunity 42, 406–417 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  152. Lund, J., Sato, A., Akira, S., Medzhitov, R. & Iwasaki, A. Toll-like receptor 9–mediated recognition of herpes simplex virus-2 by plasmacytoid dendritic cells. J. Exp. Med. 198, 513–520 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  153. Silver, A. C., Arjona, A., Walker, W. E. & Fikrig, E. The circadian clock controls toll-like receptor 9-mediated innate and adaptive immunity. Immunity 36, 251–261 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  154. Fetissov, S. O. Role of the gut microbiota in host appetite control: bacterial growth to animal feeding behaviour. Nat. Rev. Endocrinol. 13, 11–25 (2016). This Review describes the data relevant to the possible involvement of gut bacteria in the regulation of host appetite, integrating of the energy status of both the host and its gut microbiota.

    PubMed  Google Scholar 

  155. Liang, X. & FitzGerald, G. A. Timing the microbes: the circadian rhythm of the gut microbiome. J. Biol. Rhythms 32, 505–515 (2017).

    PubMed  Google Scholar 

  156. Thaiss, C. A. et al. Transkingdom control of microbiota diurnal oscillations promotes metabolic homeostasis. Cell 159, 514–529 (2014). This paper presents an example of how a symbiotic community may synchronize its interdependent physiological activities to the geophysical clock to promote homeostasis; moreover, it reveals that feeding rhythms direct microbiota diurnal oscillations and how external intervention (such as chronic jet lag) may lead to dysbiosis.

    CAS  PubMed  Google Scholar 

  157. Leone, V. et al. Effects of diurnal variation of gut microbes and high-fat feeding on host circadian clock function and metabolism. Cell Host Microbe 17, 681–689 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  158. Thaiss, C. A. et al. Microbiota diurnal rhythmicity programs host transcriptome oscillations. Cell 167, 1495–1510.e12 (2016).

    CAS  PubMed  Google Scholar 

  159. Froy, O. Circadian rhythms, nutrition and implications for longevity in urban environments. Proc. Nutr. Soc. https://doi.org/10.1017/S0029665117003962 (2017).

    Article  PubMed  Google Scholar 

  160. Thaiss, C. A., Levy, M. & Elinav, E. Chronobiomics: the biological clock as a new principle in host–microbial interactions. PLOS Pathog. 11, e1005113 (2015).

    PubMed  PubMed Central  Google Scholar 

  161. Patterson, R. E. & Sears, D. D. Metabolic effects of intermittent fasting. Annu. Rev. Nutr. 37, 371–393 (2017).

    CAS  PubMed  Google Scholar 

  162. Baker, P. W., Rye, K. A., Gamble, J. R., Vadas, M. A. & Barter, P. J. Ability of reconstituted high density lipoproteins to inhibit cytokine-induced expression of vascular cell adhesion molecule-1 in human umbilical vein endothelial cells. J. Lipid Res. 40, 345–353 (1999).

    CAS  PubMed  Google Scholar 

  163. Xia, P., Vadas, M. A., Rye, K. A., Barter, P. J. & Gamble, J. R. High density lipoproteins (HDL) interrupt the sphingosine kinase signaling pathway. A possible mechanism protection against atherosclerosis HDL. J. Biol. Chem. 274, 33143–33147 (1999).

    CAS  PubMed  Google Scholar 

  164. Perrin-Cocon, L. et al. High-density lipoprotein phospholipids interfere with dendritic cell Th1 functional maturation. Immunobiology 217, 91–99 (2012).

    CAS  PubMed  Google Scholar 

  165. Baker, P. W., Rye, K. A., Gamble, J. R., Vadas, M. A. & Barter, P. J. Phospholipid composition of reconstituted high density lipoproteins influences their ability to inhibit endothelial cell adhesion molecule expression. J. Lipid Res. 41, 1261–1267 (2000).

    CAS  PubMed  Google Scholar 

  166. Litman, B. J., Lewis, E. N. & Levin, I. W. Packing characteristics of highly unsaturated bilayer lipids: Raman spectroscopic studies of multilamellar phosphatidylcholine dispersions. Biochemistry 30, 313–319 (1991).

    CAS  PubMed  Google Scholar 

  167. Ulevitch, R. J., Johnston, A. R. & Weinstein, D. B. New function for high density lipoproteins. Their participation in intravascular reactions of bacterial lipopolysaccharides. J. Clin. Invest. 64, 1516–1524 (1979).

    CAS  PubMed  PubMed Central  Google Scholar 

  168. Vishnyakova, T. G. et al. Binding and internalization of lipopolysaccharide by Cla-1, a human orthologue of rodent scavenger receptor B1. J. Biol. Chem. 278, 22771–22780 (2003).

    CAS  PubMed  Google Scholar 

  169. Murphy, A. J. et al. High-density lipoprotein reduces the human monocyte inflammatory response. Arterioscler. Thromb. Vasc. Biol. 28, 2071–2077 (2008).

    CAS  PubMed  Google Scholar 

  170. Peshavariya, H. et al. Reconstituted high-density lipoprotein suppresses leukocyte NADPH oxidase activation by disrupting lipid rafts. Free Radic. Res. 43, 772–782 (2009).

    CAS  PubMed  Google Scholar 

  171. Kabouridis, P. S. & Jury, E. C. Lipid rafts and T-lymphocyte function: Implications for autoimmunity. FEBS Lett. 582, 3711–3718 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  172. Gupta, N. & DeFranco, A. L. Lipid rafts and B cell signaling. Semin. Cell Dev. Biol. 18, 616–626 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  173. Fulop, T. et al. Immunosenescence and inflamm-aging as two sides of the same coin: friends or foes? Front. Immunol. 8, 1960 (2017).

    PubMed  Google Scholar 

  174. Doyle, S. & Menaker, M. Circadian photoreception in vertebrates. Cold Spring Harb. Symp. Quant. Biol. 72, 499–508 (2007).

    CAS  PubMed  Google Scholar 

  175. Schibler, U. et al. Clock-talk: interactions between central and peripheral circadian oscillators in mammals. Cold Spring Harb. Symp. Quant. Biol. 80, 223–232 (2015).

    PubMed  Google Scholar 

  176. Olivieri, F., Rippo, M. R., Procopio, A. D. & Fazioli, F. Circulating inflamma-miRs in aging and age-related diseases. Front. Genet. 4, 121 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  177. Zhao, J. et al. Metabolic profiles of biological aging in American Indians: the strong heart family study. Aging 6, 176–186 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  178. Takiyama, N. & Matsumoto, K. Age-and sex-related differences of serum carnitine in a Japanese population. J. Am. Coll. Nutr. 17, 71–74 (1998).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was partly supported by Fondazione Cassa di Risparmio delle Province Lombarde (CARIPLO) (Rif. 2015–0564 to C.F. and Rif. 2016–0835); European Union (EU) FP7 Project HUMAN (Health and the Understanding of Metabolism, Aging and Nutrition) (grant agreement 602757) and EU Joint Programme – Neurodegenerative Disease Research (JPND) Adage to C.F.; EU H2020 Project PROPAG-AGEING (grant agreement 634821) to C.F. and P.G.; the Italian Ministry of Health Ricerca Finalizzata Young Researchers (under 40)–Giovani Ricercatori (GR-2013-02358026) to A.S.; Basic Research Projects of the Alma Mater Studiorum - University of Bologna (ALMA-IDEA-2017) to C.G.; and a grant of the Ministry of Education and Science of the Russian Federation (agreement 074-02-2018-330) "Digitalized and Personalized Medicine of Healthy Aging (DPM-AGEING)" at Lobachevsky State University of Nizhny Novgorod to C.F.

Author information

Authors and Affiliations

Authors

Contributions

All authors researched the data for the article, provided substantial contribution to the discussion of the content, wrote the article and reviewed and/or edited the manuscript before submission.

Corresponding author

Correspondence to Cristina Giuliani.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

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

Glossary terms

Geroscience

A research field that tries to understand the molecular relationship and link between ageing and age-related chronic diseases; the basic assumption is that the mechanisms driving ageing and those driving age-related diseases largely overlap.

Inflammasome

A multiprotein intracellular complex that detects pathogenic microorganisms and sterile stressors.

Immunosenescence

A process that refers to all of the most marked changes that occur with ageing in the adaptive immune system; this process is responsible for the increased susceptibility of elderly individuals to new infectious diseases and it is also linked to inflammatory age-related diseases.

Mitokines

Signalling proteins and peptides produced in response to mitochondrial stress (such as oxidative stress and unfolded proteins); they can either be encoded by nuclear DNA or mitochondrial DNA.

Poikilotherms

Cold-blooded animals that have biological strategies that allow them to elude or endure exposures to environmental temperatures that are below the freezing point of their body fluid.

Homeotherms

Organisms with a constant body temperature that is largely independent of the temperature of its surroundings.

Metabolic endotoxaemia

A low-grade, chronic elevation in plasma lipopolysaccharide (10–50 times lower than septic conditions).

Metabolomics

The systematic identification and quantification of the small molecule metabolic products (the metabolome) of a biological system.

Lipidomics

The study of the structure and function of the complete set of lipids (the lipidome).

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Franceschi, C., Garagnani, P., Parini, P. et al. Inflammaging: a new immune–metabolic viewpoint for age-related diseases. Nat Rev Endocrinol 14, 576–590 (2018). https://doi.org/10.1038/s41574-018-0059-4

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41574-018-0059-4

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