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.

Hypothalamic stem cells control ageing speed partly through exosomal miRNAs

Subjects

An Author Correction to this article was published on 27 June 2018

This article has been updated

Abstract

It has been proposed that the hypothalamus helps to control ageing, but the mechanisms responsible remain unclear. Here we develop several mouse models in which hypothalamic stem/progenitor cells that co-express Sox2 and Bmi1 are ablated, as we observed that ageing in mice started with a substantial loss of these hypothalamic cells. Each mouse model consistently displayed acceleration of ageing-like physiological changes or a shortened lifespan. Conversely, ageing retardation and lifespan extension were achieved in mid-aged mice that were locally implanted with healthy hypothalamic stem/progenitor cells that had been genetically engineered to survive in the ageing-related hypothalamic inflammatory microenvironment. Mechanistically, hypothalamic stem/progenitor cells contributed greatly to exosomal microRNAs (miRNAs) in the cerebrospinal fluid, and these exosomal miRNAs declined during ageing, whereas central treatment with healthy hypothalamic stem/progenitor cell-secreted exosomes led to the slowing of ageing. In conclusion, ageing speed is substantially controlled by hypothalamic stem cells, partially through the release of exosomal miRNAs.

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

Figure 1: Ageing-associated htNSC loss and its impact on ageing speed.
Figure 2: Ageing acceleration and lifespan shortening due to htNSC loss.
Figure 3: Slowdown of ageing by hypothalamic implantation of IκBα-htNSCs.
Figure 4: Exosomes and exosomal miRNAs secreted by htNSCs.
Figure 5: Contribution of htNSCs to exosomal miRNAs in the CSF.
Figure 6: Slowdown of ageing by treatment of htNSC-derived exosomes.

Accession codes

Accessions

Gene Expression Omnibus

Change history

  • 27 June 2018

    The microarray data generated and analysed in this Article have been uploaded to the Gene Expression Omnibus (GEO) under accession number GSE113383. Accordingly, the statement in the 'Data availability' section of the Article has been rephrased online.

References

  1. Chang, H. C. & Guarente, L. SIRT1 mediates central circadian control in the SCN by a mechanism that decays with aging. Cell 153, 1448–1460 (2013)

    Article  CAS  Google Scholar 

  2. Fridell, Y. W., Sánchez-Blanco, A., Silvia, B. A. & Helfand, S. L. Targeted expression of the human uncoupling protein 2 (hUCP2) to adult neurons extends life span in the fly. Cell Metab. 1, 145–152 (2005)

    Article  CAS  Google Scholar 

  3. Alcedo, J. & Kenyon, C. Regulation of C. elegans longevity by specific gustatory and olfactory neurons. Neuron 41, 45–55 (2004)

    Article  CAS  Google Scholar 

  4. Riera, C. E. et al. TRPV1 pain receptors regulate longevity and metabolism by neuropeptide signaling. Cell 157, 1023–1036 (2014)

    Article  CAS  Google Scholar 

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

    Article  CAS  ADS  Google Scholar 

  6. Satoh, A. et al. Sirt1 extends life span and delays aging in mice through the regulation of Nk2 homeobox 1 in the DMH and LH. Cell Metab. 18, 416–430 (2013)

    Article  CAS  Google Scholar 

  7. Dacks, P. A., Moreno, C. L., Kim, E. S., Marcellino, B. K. & Mobbs, C. V. Role of the hypothalamus in mediating protective effects of dietary restriction during aging. Front. Neuroendocrinol. 34, 95–106 (2013)

    Article  CAS  Google Scholar 

  8. Sadagurski, M. et al. Transient early food restriction leads to hypothalamic changes in the long-lived crowded litter female mice. Physiol. Rep. 3, e12379 (2015)

    Article  Google Scholar 

  9. van Praag, H. et al. Functional neurogenesis in the adult hippocampus. Nature 415, 1030–1034 (2002)

    Article  CAS  ADS  Google Scholar 

  10. Encinas, J. M. et al. Division-coupled astrocytic differentiation and age-related depletion of neural stem cells in the adult hippocampus. Cell Stem Cell 8, 566–579 (2011)

    Article  CAS  Google Scholar 

  11. Kheirbek, M. A., Klemenhagen, K. C., Sahay, A. & Hen, R. Neurogenesis and generalization: a new approach to stratify and treat anxiety disorders. Nat. Neurosci. 15, 1613–1620 (2012)

    Article  CAS  Google Scholar 

  12. Merkle, F. T. et al. Adult neural stem cells in distinct microdomains generate previously unknown interneuron types. Nat. Neurosci. 17, 207–214 (2014)

    Article  CAS  Google Scholar 

  13. Sun, Y. et al. Neurogenin promotes neurogenesis and inhibits glial differentiation by independent mechanisms. Cell 104, 365–376 (2001)

    Article  CAS  Google Scholar 

  14. Molofsky, A. V. et al. Increasing p16INK4a expression decreases forebrain progenitors and neurogenesis during ageing. Nature 443, 448–452 (2006)

    Article  CAS  ADS  Google Scholar 

  15. Baruch, K. et al. Aging-induced type I interferon response at the choroid plexus negatively affects brain function. Science 346, 89–93 (2014)

    Article  CAS  ADS  Google Scholar 

  16. Greenberg, D. A. & Jin, K. Turning neurogenesis up a Notch. Nat. Med. 12, 884–885 (2006)

    Article  CAS  Google Scholar 

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

    Article  CAS  ADS  Google Scholar 

  18. Sun, F. et al. Notch1 signaling modulates neuronal progenitor activity in the subventricular zone in response to aging and focal ischemia. Aging Cell 12, 978–987 (2013)

    Article  CAS  Google Scholar 

  19. Li, J., Tang, Y. & Cai, D. IKKβ/NF-κB disrupts adult hypothalamic neural stem cells to mediate a neurodegenerative mechanism of dietary obesity and pre-diabetes. Nat. Cell Biol. 14, 999–1012 (2012)

    Article  CAS  Google Scholar 

  20. Lee, D. A. et al. Tanycytes of the hypothalamic median eminence form a diet-responsive neurogenic niche. Nat. Neurosci. 15, 700–702 (2012)

    Article  CAS  Google Scholar 

  21. McNay, D. E., Briançon, N., Kokoeva, M. V., Maratos-Flier, E. & Flier, J. S. Remodeling of the arcuate nucleus energy-balance circuit is inhibited in obese mice. J. Clin. Invest. 122, 142–152 (2012)

    Article  CAS  Google Scholar 

  22. Favaro, R. et al. Hippocampal development and neural stem cell maintenance require Sox2-dependent regulation of Shh. Nat. Neurosci. 12, 1248–1256 (2009)

    Article  CAS  Google Scholar 

  23. Molofsky, A. V. et al. Bmi-1 dependence distinguishes neural stem cell self-renewal from progenitor proliferation. Nature 425, 962–967 (2003)

    Article  CAS  ADS  Google Scholar 

  24. Strojnik, T., Røsland, G. V., Sakariassen, P. O., Kavalar, R. & Lah, T. Neural stem cell markers, nestin and musashi proteins, in the progression of human glioma: correlation of nestin with prognosis of patient survival. Surg. Neurol. 68, 133–143 (2007)

    Article  Google Scholar 

  25. Faiz, M. et al. Adult neural stem cells from the subventricular zone give rise to reactive astrocytes in the cortex after stroke. Cell Stem Cell 17, 624–634 (2015)

    Article  CAS  Google Scholar 

  26. Corti, S. et al. Neural stem cells LewisX+ CXCR4+ modify disease progression in an amyotrophic lateral sclerosis model. Brain 130, 1289–1305 (2007)

    Article  Google Scholar 

  27. Buch, T. et al. A Cre-inducible diphtheria toxin receptor mediates cell lineage ablation after toxin administration. Nat. Methods 2, 419–426 (2005)

    Article  CAS  Google Scholar 

  28. Shi, Y. et al. MicroRNA regulation of neural stem cells and neurogenesis. J. Neurosci. 30, 14931–14936 (2010)

    Article  CAS  Google Scholar 

  29. Li, Q. & Gregory, R. I. MicroRNA regulation of stem cell fate. Cell Stem Cell 2, 195–196 (2008)

    Article  CAS  Google Scholar 

  30. Boon, R. A. et al. MicroRNA-34a regulates cardiac ageing and function. Nature 495, 107–110 (2013)

    Article  CAS  ADS  Google Scholar 

  31. Fraczek, L. A., Martin, C. B. & Martin, B. K. c-Jun and c-Fos regulate the complement factor H promoter in murine astrocytes. Mol. Immunol. 49, 201–210 (2011)

    Article  CAS  Google Scholar 

  32. Zhang, X. et al. Hypothalamic IKKβ/NF-κB and ER stress link overnutrition to energy imbalance and obesity. Cell 135, 61–73 (2008)

    Article  CAS  Google Scholar 

  33. Soleimani, M. & Nadri, S. A protocol for isolation and culture of mesenchymal stem cells from mouse bone marrow. Nat. Protoc. 4, 102–106 (2009)

    Article  CAS  Google Scholar 

  34. Jin, K., Wang, X., Xie, L., Mao, X. O. & Greenberg, D. A. Transgenic ablation of doublecortin-expressing cells suppresses adult neurogenesis and worsens stroke outcome in mice. Proc. Natl Acad. Sci. USA 107, 7993–7998 (2010)

    Article  CAS  ADS  Google Scholar 

  35. Li, J. et al. Exosomes mediate the cell-to-cell transmission of IFN-α-induced antiviral activity. Nat. Immunol. 14, 793–803 (2013)

    Article  CAS  Google Scholar 

  36. Purkayastha, S., Zhang, G. & Cai, D. Uncoupling the mechanisms of obesity and hypertension by targeting hypothalamic IKK-β and NF-κB. Nat. Med. 17, 883–887 (2011)

    Article  CAS  Google Scholar 

  37. Fry, C. S. et al. Inducible depletion of satellite cells in adult, sedentary mice impairs muscle regenerative capacity without affecting sarcopenia. Nat. Med. 21, 76–80 (2015)

    Article  CAS  Google Scholar 

  38. Kaidanovich-Beilin, O., Lipina, T., Vukobradovic, I., Roder, J. & Woodgett, J. R. Assessment of social interaction behaviors. J. Vis. Exp. (48) 2473 (2011)

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

    Article  CAS  Google Scholar 

  40. Yan, J. et al. Obesity- and aging-induced excess of central transforming growth factor-β potentiates diabetic development via an RNA stress response. Nat. Med. 20, 1001–1008 (2014)

    Article  CAS  Google Scholar 

  41. Aponte, Y., Atasoy, D. & Sternson, S. M. AGRP neurons are sufficient to orchestrate feeding behavior rapidly and without training. Nat. Neurosci. 14, 351–355 (2011)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This study was supported by NIH R01 DK078750, AG031774, HL113180 and DK099136 (D.C.).

Author information

Authors and Affiliations

Authors

Contributions

Y.Z., M.S.K., J.Y. and C.H. performed hypothalamic injections and cell implantation; Y.Z. performed immunostaining, cloning, virus production and CSF sampling; M.S.K. performed behavioural experiments and exosome treatment; B.J. performed cell culture, exosome and miRNA characterization; J.Y. performed lifespan follow-up and initial behavioural and miRNA analysis; J.P.Z.-H. performed cell culture and imaging; D.C. conceived the hypothesis, designed and organized the study and wrote the paper.

Corresponding author

Correspondence to Dongsheng Cai.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

Reviewer Information Nature thanks K. Jin, T. Wyss-Coray 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.

Extended data figures and tables

Extended Data Figure 1 Ageing-related physiology and histology in C57BL/6 mice.

Male C57BL/6 mice at indicated ages (months, M) were maintained in standard housing conditions and under standard chow-feeding conditions without any experimental treatment, except for analysis of ageing-related physiological parameters and tissue histology as indicated. n = 18 mice per group (behaviours) and n = 5 mice per group (histology). Data are mean ± s.e.m.

Source data

Extended Data Figure 2 Viral injection and additional information on TK1/GCV model.

a, Lentiviruses of CMV-promoter-driven GFP were injected into the hypothalamic third ventricle (3V) of C57BL/6 mice via a pre-implanted cannula. One week after injection, brain sections were made and examined for GFP immunostaining. Scale bars, 50 μm. Images represent four independent experiments. b, c, AgRP-Cre mice and POMC-Cre mice received an injection in the hypothalamic third ventricle of Hsv-TK1 lentivirus followed by GCV treatment and were examined three months later for the number of AgRP and POMC neurons in the ARC through Cre immunostaining. d, AgRP-Cre mice and POMC-Cre mice were injected with rAAV2-FLEX-rev-ChR2:tdTomato virus or vehicle into the ARC, followed by injection of Sox2-promoter-driven Hsv-TK1 lentivirus (TK) or control lentivirus (Con) into the hypothalamic third ventricle. GCV was administrated into the third ventricle twice per week for three weeks. Subsequently these mice were subjected to an optogenetic stimulation-induced feeding response as described in the Methods. Food intake before and after optogenetic stimulation were also measured. e, MWM training information for Fig. 1f. **P < 0.01, ***P < 0.001; two-tailed Student’s t-test (b, c), one-way ANOVA with Tukey’s post hoc test (d, e); n = 4 mice per group (b, c), n = 5 mice per group (d) and n = 8 mice per group (e). Data are mean ± s.e.m.

Source data

Extended Data Figure 3 Ablation of htNSCs in hypothalamic third ventricular wall by DTR/diphtheria toxin.

Mid-aged male C57BL/6 mice (15 months old) were injected in the hypothalamic third ventricle with Sox2-promoter-directed DTR lentivirus (DTR) or control lentivirus, followed by four-week (twice per week) intraperitoneal injection of diphtheria toxin (DT) or vehicle. a, Diagram of lentiviral DTR. b, Evaluation of Sox2-promoter-driven DTR lentiviruses in cultured htNSCs by immunostaining of DTR and Sox2. Scale bars, 50 μm. Images represent 3 independent experiments. ce, Immunostaining of hypothalamic sections (c, d) and physiological analyses (e) of these mice at three months after viral injection. Control values in c and d represent similar observations in DTR/DT+ and DTR+/DT groups. *P < 0.05, **P < 0.01, ***P < 0.001; two-tailed Student’s t-test (c, d), one-way ANOVA with Tukey’s post hoc test (e); n = 5 mice per group (c, d) and n = 8 mice per group (e). Data are mean ± s.e.m.

Source data

Extended Data Figure 4 Assessing exosomes secreted by htNSCs.

a, Immunostaining of CD81 in cultured htNSCs and astrocytes. Scale bars, 10 μm. b, Electron microscopic images of htNSCs. Right, high magnification of the outlined area on the left. Black arrows indicate the presence of multivescular bodies. c, Flow cytometry analysis of CD81 of htNSC-derived secreted exosomes. Grey area indicates the appropriate isotype control. d, Purified exosomes secreted from cultured htNSCs were profiled using nanoparticle analysis. Data represent three independent experiments.

Extended Data Figure 5 Additional assessments on secreted exosomes from htNSCs.

a, Secreted exosomes isolated from htNSCs were subjected to fractioning by density-gradient ultracentrifugation, different fractions were analysed by immunoblotting with anti-TSG101 antibody. b, Secreted exosomes isolated from htNSCs were subjected to pull-down by anti-CD81 antibody or appropriate isotype control and then analysed for levels of candidate miRNAs. n = 4 independent biological samples per group (a) and n = 5 independent biological samples per group (b). Data are mean ± s.e.m.

Source data

Extended Data Figure 6 Small RNA bio-analyser assay of secreted exosomes.

a, Representative traces of exosomal small RNA and miRNA secreted by htNSCs, IκBα-htNSCs, MSCs and hypothalamic neuronal GT1-7 cells. b, Quantification of exosomal miRNAs secreted by indicated cells according to the results from small RNA/miRNA bio-analysis. n = 5 independent biological samples per group. Data are mean ± s.e.m.

Source data

Extended Data Figure 7 Growth factors and cytokines secreted by htNSCs.

Indicated cells were cultured in medium without EGF and bFGF for 2 days, and the medium was collected and analysed using the Mouse Growth Factor Array for indicated growth factors and cytokines. Array images contain three independent biological samples per cell model (blots on the top and the design of array on the bottom).

Extended Data Figure 8 Effects on htNSCs and animal physiology by Rab27a shRNA.

a, b, Cultured htNSCs were infected with Sox2-promoter-driven Cre lentivirus and Cre-dependent Rab27a shRNA or control scramble shRNA lentivirus (a) and examined for Ki67 by immunostaining (b) at 2–3 days after viral infection. Scale bars, 60 μm. c, C57BL/6 mice (12-month-old males) were injected in the hypothalamic third ventricle with Sox2-promoter-driven Cre lentivirus and Cre-dependent Rab27a shRNA or control scramble shRNA lentivirus or vehicle. Ageing-related physiology was analysed in mice at six weeks after viral injection. *P < 0.05, **P < 0.01; one-way ANOVA with Tukey’s post hoc test (c); n = 8 mice for vehicle, n = 9 mice for control shRNA, n = 10 mice for Rab27a shRNA (c). Data are mean ± s.e.m.

Source data

Extended Data Figure 9 Electron microscopic examination of htNSC-secreted exosomes.

Exosomes secreted from cultured htNSCs were obtained and purified using differential ultra-centrifugation and examined for the purity and size distribution by electron microscopy using the protocol detailed in the Methods. Scale bar, 250 nm.

Extended Data Figure 10 Additional information for the anti-ageing models used in this study.

a, C57BL/6 mice (16-month-old males) were treated via hypothalamic third-ventricle cannula with exosomes (100 ng protein, purified from htNSCs) or vehicle, three times per week for four months, and the hypothalamic tissues were dissected and examined for indicated mRNAs. Expression levels of mRNAs are presented in arbitrary units (au), and the value of each species in the control group was normalized to 1. b, The training session information for MWM in Fig. 6c. c, The training session information for MWM in Fig. 6d. *P < 0.05 (c) or as indicated (a); two-tailed Student’s t-test (a) or one-way ANOVA with Tukey’s post hoc test (c); n = 5 mice per group (a), n = 8 mice for Con/GCV with vehicle, n = 7 mice for TK1/GCV with vehicle and TK1/GCV with exosome (b), and n = 7 mice per group (c). Data are mean ± s.e.m.

Source data

Related audio

Supplementary information

Supplementary Figure 1

This file contains uncropped images of all western blots shown in the manuscript. (PDF 290 kb)

Supplementary Data 1

This file contains microarray data of Figure 4d. (XLSX 174 kb)

Supplementary Data 2

This file contains qPCR data of Figure 4e. (XLSX 12 kb)

PowerPoint slides

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, Y., Kim, M., Jia, B. et al. Hypothalamic stem cells control ageing speed partly through exosomal miRNAs. Nature 548, 52–57 (2017). https://doi.org/10.1038/nature23282

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature23282

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