Skip to main content

Advertisement

Log in

Disruption of leptin signalling in a mouse model of Alzheimer’s disease

  • Original Article
  • Published:
Metabolic Brain Disease Aims and scope Submit manuscript

Abstract

Disruption of leptin signalling has been implicated as playing a role in the development of Alzheimer’s disease (AD). Leptin has previously been shown to be affected by amyloid-beta (Aβ)-related signalling; however, pathways that link leptin to the disease pathogenesis have not been determined. To characterize the association between increasing age-dependent Aβ levels with leptin signalling and the vulnerable brain regions in AD, we assessed the mRNA and protein expression profile of leptin and leptin receptor (Ob-Rb) at 9 and 18-month-age in APP/PS1 mice. Immunohistochemical labelling demonstrated that leptin and Ob-Rb proteins were localised to neocortical and hippocampal neurons in APP/PS1 and wildtype (WT) mice. Neuronal leptin and Ob-Rb immunolabelling was more prominent in the neocortex of both groups at 9 month of age, while, at 18 months, labelling was reduced in the hippocampus of APP/PS1 mice relative to WT. Immunoblotting analysis demonstrated decreased hippocampal leptin levels, concomitantly with an increased Ob-Rb levels, in APP/PS1 mice compared with WT controls at 18 month of age. While no leptin mRNA was found in either of the groups analysed, Ob-Rb mRNA was significantly decreased in the hippocampus of APP/PS1 mice at both ages analysed. In addition, a significant decreased protein kinase B (Akt) activity concomitantly with an upregulation of suppressor of cytokine signaling-3 (SOCS3) and protein-tyrosine phosphatase 1B (PTP1B) transcripts was present. Thus, these results collectively indicate alterations of leptin signalling in the hippocampus of APP/PS1 mice, providing novel insights about the pathways that could link aberrant leptin signaling to the pathological changes of AD.

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

Access this article

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

Instant access to the full article PDF.

Fig. 1: Alterations in leptin levels in a pre-clinical state in APP/PS1 mice.
Fig. 2: Alterations in leptin levels in APP/PS1 mice.
Fig. 3: Alterations in leptin localization in APP/PS1 mice.
Fig. 4: Alterations in Ob-Rb levels in APP/PS1 mice.
Fig. 5: Alterations in Ob-Rb localization in APP/PS1 mice.
Fig. 6: Alterations in tyrosine phosphorylation of STAT3 in the neocortex and hippocampus of APP/PS1 mice.
Fig. 7: Alterations in SOCS3 and PTP1B in the neocortex and hippocampus of APP/PS1 mice.
Fig. 8: Leptin signaling pathways

Similar content being viewed by others

References

Download references

Acknowledgements

The authors would like to gratefully acknowledge Graeme McCormack for his excellent technical support. The authors declare that they have no conflict of interest. This work was supported by the funding from J.O. and J.R. Wicking Trust (Equity Trustees).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Carmen Fernandez-Martos.

Electronic supplementary material

Suppl. Fig.1

IHC negative controls. (A-H) Representative images processed without the primary antibodies, leptin and Ob-Rb, as controls, in both WT (A-G) and APP/PS1 (B-H) sections at 9 month of age. No non-specific staining was observed in any of the immunohistochemical controls. Images were collected on an Axio lab A.1 microscope and AxioCam ICc5 camera (both Zeiss, Germany), with ×5, ×20 and ×40 objectives. Scale bars = 20 μm. Black square indicates the hippocampal region of interest magnified with the ×40 objective. Abbreviations: CTX, neocortex; HP, hippocampus. (JPEG 2521 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

King, A., Brain, A., Hanson, K. et al. Disruption of leptin signalling in a mouse model of Alzheimer’s disease. Metab Brain Dis 33, 1097–1110 (2018). https://doi.org/10.1007/s11011-018-0203-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11011-018-0203-9

Keywords

Navigation