Skip to main content

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

  • Article
  • Published:

MANF regulates metabolic and immune homeostasis in ageing and protects against liver damage

Abstract

Ageing is accompanied by altered intercellular communication, deregulated metabolic function, and inflammation. Interventions that restore a youthful state delay or reverse these processes, prompting the search for systemic regulators of metabolic and immune homeostasis. Here, we identified mesencephalic-astrocyte-derived neurotrophic factor (MANF), a secreted stress-response protein with immune modulatory properties, as an evolutionarily conserved regulator of systemic and, in particular, liver metabolic homeostasis. We show that MANF levels declined with age in flies, mice, and humans, and MANF overexpression extends lifespan in flies. MANF-deficient flies exhibit enhanced inflammation and shorter lifespans, and MANF heterozygous mice exhibit inflammatory phenotypes in various tissues, as well as progressive liver damage, fibrosis, and steatosis. We show that immune-cell-derived MANF protects against liver inflammation and fibrosis, whereas hepatocyte-derived MANF prevents hepatosteatosis. Liver rejuvenation by heterochronic parabiosis in mice further depends on MANF, whereas MANF supplementation ameliorates several hallmarks of liver ageing, prevents hepatosteatosis induced by diet, and improves age-related metabolic dysfunction. Our findings identify MANF as a systemic regulator of homeostasis in young animals, suggesting a therapeutic application for MANF in age-related 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: MANF protein levels decline with age in flies, mice, and humans.
Fig. 2: MANF overexpression extends lifespan in Drosophila.
Fig. 3: Mice with reduced MANF levels develop inflammation and liver damage.
Fig. 4: Immune-cell-derived MANF and hepatocyte-derived MANF contribute to liver homeostasis.
Fig. 5: MANF is required for full liver rejuvenation by heterochronic parabiosis.
Fig. 6: MANF improves liver damage, inflammation, and metabolic dysfunction in old mice.

Similar content being viewed by others

Data availability

All the data generated or analysed during this study are included in the published article and its Supplementary Information files, and are available from the corresponding author. Analysed RNA-sequencing data are available in Supplementary Table 1 and raw RNA-sequencing data are available under accession numbers GSE123115, GSE123116, and GSE123117 on the NCBI Gene Expression Omnibus database. Correspondence and requests for materials should be addressed to H.J., P.S.-V., and J.N.

References

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

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Franceschi, C. et al. Inflamm-aging. An evolutionary perspective on immunosenescence. Ann. N.Y. Acad. Sci. 908, 244–254 (2000).

    Article  CAS  PubMed  Google Scholar 

  3. Villeda, S. A. et al. The ageing systemic milieu negatively regulates neurogenesis and cognitive function. Nature 477, 90–94 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Smith, L. K. et al. beta2-microglobulin is a systemic pro-aging factor that impairs cognitive function and neurogenesis. Nat. Med. 21, 932–937 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Conboy, I. M. et al. Rejuvenation of aged progenitor cells by exposure to a young systemic environment. Nature 433, 760–764 (2005).

    Article  CAS  PubMed  Google Scholar 

  6. Rebo, J. et al. A single heterochronic blood exchange reveals rapid inhibition of multiple tissues by old blood. Nat. Commun. 7, 13363 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Ruckh, J. M. et al. Rejuvenation of regeneration in the aging central nervous system. Cell. Stem. Cell. 10, 96–103 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Murray, P. J. & Wynn, T. A. Protective and pathogenic functions of macrophage subsets. Nat. Rev. Immunol. 11, 723–737 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Miron, V. E. et al. M2 microglia and macrophages drive oligodendrocyte differentiation during CNS remyelination. Nat. Neurosci. 16, 1211–1218 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Li, H., Qi, Y. & Jasper, H. Preventing age-related decline of gut compartmentalization limits microbiota dysbiosis and extends lifespan. Cell. Host. Microbe 19, 240–253 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Clark, R. I. & Walker, D. W. Role of gut microbiota in aging-related health decline: insights from invertebrate models. Cell. Mol. Life Sci. 75, 93–101 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Clark, R. I., Walker, D. W. & Dionne, M. S. Metabolic and immune integration in aging and age-related disease. Aging 6, 3–4 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Gan, L., Chitturi, S. & Farrell, G. C. Mechanisms and implications of age-related changes in the liver: nonalcoholic fatty liver disease in the elderly. Curr. Gerontol. Geriatr. Res. 2011, 831536 (2011).

    PubMed  PubMed Central  Google Scholar 

  15. Sheedfar, F., Di Biase, S., Koonen, D. & Vinciguerra, M. Liver diseases and aging: friends or foes? Aging Cell. 12, 950–954 (2013).

    Article  CAS  PubMed  Google Scholar 

  16. Park, J. H. et al. Daumone fed late in life improves survival and reduces hepatic inflammation and fibrosis in mice. Aging Cell. 13, 709–718 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Larsen, C. M. et al. Interleukin-1-receptor antagonist in type 2 diabetes mellitus. N. Eng. J. Med. 356, 1517–1526 (2007).

    Article  CAS  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  19. Neves, J. et al. Immune modulation by MANF promotes tissue repair and regenerative success in the retina. Science 353, aaf3646 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Lindholm, P. & Saarma, M. Novel CDNF/MANF family of neurotrophic factors. Dev. Neurobiol. 70, 360–371 (2010).

    CAS  PubMed  Google Scholar 

  21. Galli, E. et al. Increased circulating concentrations of mesencephalic astrocyte-derived neurotrophic factor in children with type 1 diabetes. Sci. Rep. 6, 29058 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Wu, T. et al. Circulating mesencephalic astrocyte-derived neurotrophic factor is increased in newly diagnosed prediabetic and diabetic patients, and is associated with insulin resistance. Endocr. J. 64, 403–410 (2017).

    Article  CAS  PubMed  Google Scholar 

  23. Lindholm, P. et al. MANF is widely expressed in mammalian tissues and differently regulated after ischemic and epileptic insults in rodent brain. Mol. Cell. Neurosci. 39, 356–371 (2008).

    Article  CAS  PubMed  Google Scholar 

  24. Lindahl, M., Saarma, M. & Lindholm, P. Unconventional neurotrophic factors CDNF and MANF: structure, physiological functions and therapeutic potential. Neurobiol. Dis. 97, 90–102 (2017).

    Article  CAS  PubMed  Google Scholar 

  25. Voutilainen, M. H. et al. Mesencephalic astrocyte-derived neurotrophic factor is neurorestorative in rat model of Parkinson’s disease. J. Neurosci. 29, 9651–9659 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Chen, L. et al. Mesencephalic astrocyte-derived neurotrophic factor is involved in inflammation by negatively regulating the NF-kappaB pathway. Sci. Rep. 5, 8133 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Yang, S., Huang, S., Gaertig, M. A., Li, X. J. & Li, S. Age-dependent decrease in chaperone activity impairs MANF expression, leading to Purkinje cell degeneration in inducible SCA17 mice. Neuron 81, 349–365 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Glass, D. et al. Gene expression changes with age in skin, adipose tissue, blood and brain. Genome Biol. 14, R75 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Ayyaz, A. & Jasper, H. Intestinal inflammation and stem cell homeostasis in aging Drosophila melanogaster. Front. Cell. Infect. Microbiol. 3, 98 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Yang, S. et al. MANF regulates hypothalamic control of food intake and body weight. Nat. Commun. 8, 579 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Lindahl, M. et al. MANF is indispensable for the proliferation and survival of pancreatic beta cells. Cell Rep. 7, 366–375 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Schmucker, D. L. Age-related changes in liver structure and function: implications for disease? Exp. Gerontol. 40, 650–659 (2005).

    Article  CAS  PubMed  Google Scholar 

  33. Rabinowitz, S. S. & Gordon, S. Macrosialin, a macrophage-restricted membrane sialoprotein differentially glycosylated in response to inflammatory stimuli. J. Exp. Med. 174, 827–836 (1991).

    Article  CAS  PubMed  Google Scholar 

  34. Kinoshita, M. et al. Characterization of two F4/80-positive Kupffer cell subsets by their function and phenotype in mice. J. Hepatol. 53, 903–910 (2010).

    Article  CAS  PubMed  Google Scholar 

  35. Walker, D. G. & Lue, L. F. Immune phenotypes of microglia in human neurodegenerative disease: challenges to detecting microglial polarization in human brains. Alzheimers Res. Ther. 7, 56 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Oishi, Y. & Manabe, I. Macrophages in age-related chronic inflammatory diseases. N.P.J. Aging Mech. Dis. 2, 16018 (2016).

    Article  Google Scholar 

  37. Baker, D. J. et al. Naturally occurringp16(Ink4a)-positive cells shorten healthy lifespan. Nature 530, 184–189 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Higami, Y. et al. Aging accelerates but life-long dietary restriction suppresses apoptosis-related Fas expression on hepatocytes. Am. J. Pathol. 151, 659–663 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Gregg, S. Q. et al. A mouse model of accelerated liver aging caused by a defect in DNA repair. Hepatology 55, 609–621 (2012).

    Article  CAS  PubMed  Google Scholar 

  40. Le Couteur, D. G., et al. in Calorie Restriction, Aging and Longevity (eds Everitt, A., Rattan, S., le Couteur, D. & de Cabo, R.) Ch. 11 (Springer, Dordrecht, 2010).

  41. Yang, L., Yang, L., Dong, C. & Li, L. The class D scavenger receptor CD68 contributes to mouse chronic liver injury. Immunol. Res. 66, 414–424 (2018).

    Article  CAS  PubMed  Google Scholar 

  42. Stahl, E. C. & Brown, B. N. Kupffer cell subsets differ between young and aged murine livers. J. Immunol. 196(1 Supplement), 126.131 (2016).

    Google Scholar 

  43. Gordon, S. Alternative activation of macrophages. Nat. Rev. Immunol. 3, 23–35 (2003).

    Article  CAS  PubMed  Google Scholar 

  44. Singh, P. et al. Lymphoid neogenesis and immune infiltration in aged liver. Hepatology 47, 1680–1690 (2008).

    Article  PubMed  Google Scholar 

  45. White, R. R. et al. Comprehensive transcriptional landscape of aging mouse liver. BMC Genom. 16, 899 (2015).

    Article  Google Scholar 

  46. Bataller, R. & Brenner, D. A. Liver fibrosis. J. Clin. Invest. 115, 209–218 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Warren, A. et al. The effects of old age on hepatic stellate cells. Curr. Gerontol. Geriatr. Res. 2011, 439835 (2011).

    PubMed  PubMed Central  Google Scholar 

  48. Palgi, M., Greco, D., Lindstrom, R., Auvinen, P. & Heino, T. I. Gene expression analysis of Drosophila Manf mutants reveals perturbations in membrane traffic and major metabolic changes. BMC Genom. 13, 134 (2012).

    Article  CAS  Google Scholar 

  49. Heckmann, B. L., Zhang, X., Xie, X. & Liu, J. The G0/G1 switch gene 2 (G0S2): regulating metabolism and beyond. Biochim. Biophys. Acta 1831, 276–281 (2013).

    Article  CAS  PubMed  Google Scholar 

  50. Yang, X. et al. The G(0)/G(1) switch gene 2 regulates adipose lipolysis through association with adipose triglyceride lipase. Cell. Metab. 11, 194–205 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Jung, S. et al. Analysis of fractalkine receptor CX(3)CR1 function by targeted deletion and green fluorescent protein reporter gene insertion. Mol. Cell. Biol. 20, 4106–4114 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Auffray, C. et al. Monitoring of blood vessels and tissues by a population of monocytes with patrolling behavior. Science 317, 666–670 (2007).

    Article  CAS  PubMed  Google Scholar 

  53. Castellano, J. M. et al. Human umbilical cord plasma proteins revitalize hippocampal function in aged mice. Nature 544, 488–492 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Sato, S. et al. Circadian reprogramming in the liver identifies metabolic pathways of aging. Cell 170, 664–677 e611 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Kanfi, Y. et al. The sirtuin SIRT6 regulates lifespan in male mice. Nature 483, 218–221 (2012).

    Article  CAS  PubMed  Google Scholar 

  56. Swindell, W. R. Genes and gene expression modules associated with caloric restriction and aging in the laboratory mouse. BMC Genom. 10, 585 (2009).

    Article  Google Scholar 

  57. Houtkooper, R. H. et al. The metabolic footprint of aging in mice. Sci. Rep. 1, 134 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Elabd, C. et al. Oxytocin is an age-specific circulating hormone that is necessary for muscle maintenance and regeneration. Nat. Commun. 5, 4082 (2014).

    Article  CAS  PubMed  Google Scholar 

  59. Loffredo, F. S. et al. Growth differentiation factor 11 is a circulating factor that reverses age-related cardiac hypertrophy. Cell 153, 828–839 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Zhang, G. et al. Hypothalamic programming of systemic ageing involving IKK-beta, NF-kappaB and GnRH. Nature 497, 211–216 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Neves, J., Sousa-Victor, P. & Jasper, H. Rejuvenating strategies for stem cell-based therapies in aging. Cell. Stem. Cell. 20, 161–175 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Malhi, H. & Kaufman, R. J. Endoplasmic reticulum stress in liver disease. J. Hepatol. 54, 795–809 (2011).

    Article  CAS  PubMed  Google Scholar 

  63. Wang, Y. et al. The g0/g1 switch gene 2 is an important regulator of hepatic triglyceride metabolism. PLoS ONE 8, e72315 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Sugaya, Y. & Satoh, H. Liver-specific G0 /G1 switch gene 2 (G0s2) expression promotes hepatic insulin resistance by exacerbating hepatic steatosis in male Wistar rats. J. Diabetes 9, 754–763 (2017).

    Article  CAS  PubMed  Google Scholar 

  65. Heckmann, B. L. et al. Liver X receptor alpha mediates hepatic triglyceride accumulation through upregulation of G0/G1 Switch Gene 2 expression. JCI Insight 2, e88735 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  66. Huang, S. C. et al. Cell-intrinsic lysosomal lipolysis is essential for alternative activation of macrophages. Nat. Immunol. 15, 846–855 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Day, C. P. Pathogenesis of steatohepatitis. Best practice & research. Clin. Gastroenterol. 16, 663–678 (2002).

    CAS  Google Scholar 

  68. Fontana, L. et al. Aging promotes the development of diet-induced murine steatohepatitis but not steatosis. Hepatology 57, 995–1004 (2013).

    Article  CAS  PubMed  Google Scholar 

  69. Yavarna, T. et al. High diagnostic yield of clinical exome sequencing in Middle Eastern patients with Mendelian disorders. Hum. Genet. 134, 967–980 (2015).

    Article  CAS  PubMed  Google Scholar 

  70. Sousa-Victor, P. et al. Geriatric muscle stem cells switch reversible quiescence into senescence. Nature 506, 316–321 (2014).

    Article  CAS  PubMed  Google Scholar 

  71. Parkhurst, C. N. et al. Microglia promote learning-dependent synapse formation through brain-derived neurotrophic factor. Cell 155, 1596–1609 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Villeda, S. A. et al. Young blood reverses age-related impairments in cognitive function and synaptic plasticity in mice. Nat. Med. 20, 659–663 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Sun, Z. et al. Hepatic Hdac3 promotes gluconeogenesis by repressing lipid synthesis and sequestration. Nat. Med. 18, 934–942 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Liu, F., Song, Y. & Liu, D. Hydrodynamics-based transfection in animals by systemic administration of plasmid DNA. Gene Ther. 6, 1258–1266 (1999).

    Article  CAS  PubMed  Google Scholar 

  75. Tiscornia, G., Singer, O. & Verma, I. M. Production and purification of lentiviral vectors. Nat. Protoc. 1, 241–245 (2006).

    Article  CAS  PubMed  Google Scholar 

  76. Huang da, W., Sherman, B. T. & Lempicki, R. A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 4, 44–57 (2009).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We thank M. Konjikusic for technical help with the mouse colony, A. Ireland and M. Ray from Calico Labs for help with library preparation and RNA sequencing, and H. Liu for providing an expert pathologist evaluation of liver sections. Work in H.J.’s laboratory is supported by NIH grant nos. AG052989, AG050104, and AG047497, and some work was supported by Calico Labs. Work in D.A.L.’s laboratory is supported by NIH grant no. EY025779. Work in S.A.V.’s lab is supported by NIH grant no. AG055797. P.S.-V. and J.N. are supported by the Glenn Foundation for Medical Research.

Author information

Authors and Affiliations

Authors

Contributions

P.S.-V., J.N., D.A.L., and H.J. conceived the study. P.S.-V. and J.N. designed and analysed all of the experiments. P.S.-V., J.N., and W.C.-C. performed experiments and collected data. P.B.V. and S.A.V. performed and supervised heterochronic parabiosis experiment. R.R.R. and C.Y.L. assisted with in vivo mouse experiments. I.S. performed transcriptomic analysis. G.A.K. and N.V.B. provided human samples, plasmids for HTV experiments and other reagents, provided expertise for experimental design and analysis, and supervised transcriptomic analysis experiments. P.S.-V. and J.N. interpreted the results, prepared the figures, and wrote the manuscript, with input from D.A.L. and H.J. D.A.L. and H.J. supervised the study. All of the authors revised the manuscript.

Corresponding author

Correspondence to Heinrich Jasper.

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.

Supplementary information

Supplementary Information

Supplementary Figures 1–13 and Supplementary Tables 2–5

Reporting Summary

Supplementary Table 1

Reads per kilobase of transcript per million values for the RNA sequencing analysis

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sousa-Victor, P., Neves, J., Cedron-Craft, W. et al. MANF regulates metabolic and immune homeostasis in ageing and protects against liver damage. Nat Metab 1, 276–290 (2019). https://doi.org/10.1038/s42255-018-0023-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-018-0023-6

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research