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:

FSH blockade improves cognition in mice with Alzheimer’s disease

Abstract

Alzheimer’s disease has a higher incidence in older women, with a spike in cognitive decline that tracks with visceral adiposity, dysregulated energy homeostasis and bone loss during the menopausal transition1,2. Inhibiting the action of follicle-stimulating hormone (FSH) reduces body fat, enhances thermogenesis, increases bone mass and lowers serum cholesterol in mice3,4,5,6,7. Here we show that FSH acts directly on hippocampal and cortical neurons to accelerate amyloid-β and Tau deposition and impair cognition in mice displaying features of Alzheimer’s disease. Blocking FSH action in these mice abrogates the Alzheimer’s disease-like phenotype by inhibiting the neuronal C/EBPβ–δ-secretase pathway. These data not only suggest a causal role for rising serum FSH levels in the exaggerated Alzheimer’s disease pathophysiology during menopause, but also reveal an opportunity for treating Alzheimer’s disease, obesity, osteoporosis and dyslipidaemia with a single FSH-blocking agent.

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: FSH-blocking antibody reverses AD neuropathology and cognitive decline in Alzheimer’s mice.
Fig. 2: Neuronal FSH receptors in mouse and human brain.
Fig. 3: Targeted Fshr knockdown in the hippocampus ameliorates AD neuropathology and impaired spatial memory.
Fig. 4: Recombinant FSH induces AD pathologies and cognitive decline in 3xTg Mice.
Fig. 5: FSH-induced AD pathology is dampened in Cebpb+/− 3xTg mice.

Similar content being viewed by others

Data availability

All original western blots are provided in the Supplementary Information. The original unedited camera images for histology and immunohistochemistry are available online (https://osf.io/9hp8r/). There are no restrictions on data availability. Unique biological material will be made available to other investigators on request. Source data are provided with this paper.

References

  1. Sowers, M. et al. Changes in body composition in women over six years at midlife: ovarian and chronological aging. J. Clin. Endocrinol. Metab. 92, 895–901 (2007).

    Article  CAS  PubMed  Google Scholar 

  2. Sowers, M. R. et al. Hormone predictors of bone mineral density changes during the menopausal transition. J. Clin. Endocrinol. Metab. 91, 1261–1267 (2006).

    Article  CAS  PubMed  Google Scholar 

  3. Guo, Y. et al. Blocking FSH inhibits hepatic cholesterol biosynthesis and reduces serum cholesterol. Cell Res. 29, 151–166 (2019).

    Article  CAS  PubMed  Google Scholar 

  4. Han, X. et al. A novel follicle-stimulating hormone vaccine for controlling fat accumulation. Theriogenology 148, 103–111 (2020).

    Article  CAS  PubMed  Google Scholar 

  5. Ji, Y. et al. Epitope-specific monoclonal antibodies to FSHβ increase bone mass. Proc. Natl Acad. Sci. USA 115, 2192–2197 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Liu, P. et al. Blocking FSH induces thermogenic adipose tissue and reduces body fat. Nature 546, 107–112 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  7. Geng, W. et al. Immunization with FSHβ fusion protein antigen prevents bone loss in a rat ovariectomy-induced osteoporosis model. Biochem. Biophys. Res. Commun. 434, 280–286 (2013).

    Article  CAS  PubMed  Google Scholar 

  8. Fisher, D. W., Bennett, D. A. & Dong, H. Sexual dimorphism in predisposition to Alzheimer’s disease. Neurobiol. Aging 70, 308–324 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Andersen, K. et al. Gender differences in the incidence of AD and vascular dementia: the EURODEM studies. EURODEM Incidence Research Group. Neurology 53, 1992–1997 (1999).

    Article  CAS  PubMed  Google Scholar 

  10. Marongiu, R. Accelerated ovarian failure as a unique model to study peri-menopause influence on Alzheimer’s disease. Front. Aging Neurosci. 11, 242 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Matyi, J. M., Rattinger, G. B., Schwartz, S., Buhusi, M. & Tschanz, J. T. Lifetime estrogen exposure and cognition in late life: the Cache County study. Menopause 26, 1366–1374 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Zandi, P. P. et al. Hormone replacement therapy and incidence of Alzheimer disease in older women: the Cache County Study. JAMA 288, 2123–2129 (2002).

    Article  CAS  PubMed  Google Scholar 

  13. O’Brien, J., Jackson, J. W., Grodstein, F., Blacker, D. & Weuve, J. Postmenopausal hormone therapy is not associated with risk of all-cause dementia and Alzheimer’s disease. Epidemiol. Rev. 36, 83–103 (2014).

    Article  PubMed  Google Scholar 

  14. Shumaker, S. A. et al. Conjugated equine estrogens and incidence of probable dementia and mild cognitive impairment in postmenopausal women: Women’s Health Initiative Memory Study. JAMA 291, 2947–2958 (2004).

    Article  CAS  PubMed  Google Scholar 

  15. Short, R. A., Bowen, R. L., O’Brien, P. C. & Graff-Radford, N. R. Elevated gonadotropin levels in patients with Alzheimer disease. Mayo Clin. Proc. 76, 906–909 (2001).

    Article  CAS  PubMed  Google Scholar 

  16. Bowen, R. L., Isley, J. P. & Atkinson, R. L. An association of elevated serum gonadotropin concentrations and Alzheimer disease? J. Neuroendocrinol. 12, 351–354 (2000).

    Article  CAS  PubMed  Google Scholar 

  17. Randolph, J. F. Jr et al. Change in follicle-stimulating hormone and estradiol across the menopausal transition: effect of age at the final menstrual period. J. Clin. Endocrinol. Metab. 96, 746–754 (2011).

    Article  CAS  PubMed  Google Scholar 

  18. Epperson, C. N., Sammel, M. D. & Freeman, E. W. Menopause effects on verbal memory: findings from a longitudinal community cohort. J. Clin. Endocrinol. Metab. 98, 3829–3838 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Greendale, G. A. et al. Effects of the menopause transition and hormone use on cognitive performance in midlife women. Neurology 72, 1850–1857 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Meyer, P. M. et al. A population-based longitudinal study of cognitive functioning in the menopausal transition. Neurology 61, 801–806 (2003).

    Article  CAS  PubMed  Google Scholar 

  21. Zhu, L. L. et al. Blocking antibody to the β-subunit of FSH prevents bone loss by inhibiting bone resorption and stimulating bone synthesis. Proc. Natl Acad. Sci. USA 109, 14574–14579 (2012).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  22. Oddo, S. et al. Triple-transgenic model of Alzheimer’s disease with plaques and tangles: intracellular Aβ and synaptic dysfunction. Neuron 39, 409–421 (2003).

    Article  CAS  PubMed  Google Scholar 

  23. Webster, S. J., Bachstetter, A. D., Nelson, P. T., Schmitt, F. A. & Van Eldik, L. J. Using mice to model Alzheimer’s dementia: an overview of the clinical disease and the preclinical behavioral changes in 10 mouse models. Front. Genet. 5, 88 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Carroll, J. C. et al. Progesterone and estrogen regulate Alzheimer-like neuropathology in female 3xTg-AD mice. J. Neurosci. 27, 13357–13365 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Zhang, Z. et al. Cleavage of tau by asparagine endopeptidase mediates the neurofibrillary pathology in Alzheimer’s disease. Nat. Med. 20, 1254–1262 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Zhang, Z. et al. Delta-secretase cleaves amyloid precursor protein and regulates the pathogenesis in Alzheimer’s disease. Nat. Commun. 6, 8762 (2015).

    Article  ADS  CAS  PubMed  Google Scholar 

  27. Rocca, W. A. et al. Increased risk of cognitive impairment or dementia in women who underwent oophorectomy before menopause. Neurology 69, 1074–1083 (2007).

    Article  CAS  PubMed  Google Scholar 

  28. Tokuyama, N. et al. Individual and combining effects of anti-RANKL monoclonal antibody and teriparatide in ovariectomized mice. Bone Rep. 2, 1–7 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Rosen, C. J. & Zaidi, M. Contemporaneous reproduction of preclinical science: a case study of FSH and fat. Ann. N. Y. Acad. Sci. 1404, 17–19 (2017).

    Article  ADS  PubMed  Google Scholar 

  30. Minkeviciene, R. et al. Age-related decrease in stimulated glutamate release and vesicular glutamate transporters in APP/PS1 transgenic and wild-type mice. J. Neurochem. 105, 584–594 (2008).

    Article  CAS  PubMed  Google Scholar 

  31. Onos, K. D. et al. Enhancing face validity of mouse models of Alzheimer’s disease with natural genetic variation. PLoS Genet. 15, e1008155 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Volianskis, A., Kostner, R., Molgaard, M., Hass, S. & Jensen, M. S. Episodic memory deficits are not related to altered glutamatergic synaptic transmission and plasticity in the CA1 hippocampus of the APPswe/PS1deltaE9-deleted transgenic mice model of ss-amyloidosis. Neurobiol. Aging 31, 1173–1187 (2010).

    Article  CAS  PubMed  Google Scholar 

  33. Araujo, A. B. & Wittert, G. A. Endocrinology of the aging male. Best Pract. Res. Clin. Endocrinol. Metab. 25, 303–319 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Casadesus, G. et al. Increases in luteinizing hormone are associated with declines in cognitive performance. Mol. Cell. Endocrinol. 269, 107–111 (2007).

    Article  CAS  PubMed  Google Scholar 

  35. Berry, A., Tomidokoro, Y., Ghiso, J. & Thornton, J. Human chorionic gonadotropin (a luteinizing hormone homologue) decreases spatial memory and increases brain amyloid-β levels in female rats. Horm. Behav. 54, 143–152 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Liu, T., Wimalasena, J., Bowen, R. L. & Atwood, C. S. Luteinizing hormone receptor mediates neuronal pregnenolone production via up-regulation of steroidogenic acute regulatory protein expression. J. Neurochem. 100, 1329–1339 (2007).

    Article  CAS  PubMed  Google Scholar 

  37. Gera, S. et al. First-in-class humanized FSH blocking antibody targets bone and fat. Proc. Natl Acad. Sci. USA 117, 28971–28979 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Straccia, M. et al. Pro-inflammatory gene expression and neurotoxic effects of activated microglia are attenuated by absence of CCAAT/enhancer binding protein β. J. Neuroinflammation 8, 156 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Ramji, D. P. & Foka, P. CCAAT/enhancer-binding proteins: structure, function and regulation. Biochem. J. 365, 561–575 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Wang, Z. H. et al. Delta-secretase phosphorylation by SRPK2 enhances its enzymatic activity, provoking pathogenesis in Alzheimer’s disease. Mol. Cell 67, 812–825 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Sun, L. et al. FSH directly regulates bone mass. Cell 125, 247–260 (2006).

    Article  CAS  PubMed  Google Scholar 

  42. Hammond, S. L., Leek, A. N., Richman, E. H. & Tjalkens, R. B. Cellular selectivity of AAV serotypes for gene delivery in neurons and astrocytes by neonatal intracerebroventricular injection. PLoS ONE 12, e0188830 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  43. von Jonquieres, G. et al. Recombinant human myelin-associated glycoprotein promoter drives selective AAV-mediated transgene expression in oligodendrocytes. Front. Mol. Neurosci. 9, 13 (2016).

    Google Scholar 

  44. Randolph, J. F. Jr. et al. Reproductive hormones in the early menopausal transition: relationship to ethnicity, body size, and menopausal status. J. Clin. Endocrinol. Metab. 88, 1516–1522 (2003).

    Article  CAS  PubMed  Google Scholar 

  45. Ashe, K. H. & Zahs, K. R. Probing the biology of Alzheimer’s disease in mice. Neuron 66, 631–645 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Millward, C. A. et al. Mice with a deletion in the gene for CCAAT/enhancer-binding protein β are protected against diet-induced obesity. Diabetes 56, 161–167 (2007).

    Article  CAS  PubMed  Google Scholar 

  47. Zaidi, M. et al. Actions of pituitary hormones beyond traditional targets. J. Endocrinol. 237, R83–R98 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Silverman, E., Eimerl, S. & Orly, J. CCAAT enhancer-binding protein β and GATA-4 binding regions within the promoter of the steroidogenic acute regulatory protein (StAR) gene are required for transcription in rat ovarian cells. J. Biol. Chem. 274, 17987–17996 (1999).

    Article  CAS  PubMed  Google Scholar 

  49. Sirois, J. & Richards, J. S. Transcriptional regulation of the rat prostaglandin endoperoxide synthase 2 gene in granulosa cells. Evidence for the role of a cis-acting C/EBPβ promoter element. J. Biol. Chem. 268, 21931–21938 (1993).

    Article  CAS  PubMed  Google Scholar 

  50. Wang, H., Liu, X., Chen, S. & Ye, K. Spatiotemporal activation of the C/EBPβ/δ-secretase axis regulates the pathogenesis of Alzheimer’s disease. Proc. Natl Acad. Sci. USA 115, E12427–E12434 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  51. Sterneck, E., Tessarollo, L. & Johnson, P. F. An essential role for C/EBPβ in female reproduction. Genes Dev. 11, 2153–2162 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Minkeviciene, R. et al. Amyloid β-induced neuronal hyperexcitability triggers progressive epilepsy. J. Neurosci. 29, 3453–3462 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Zhang, Z. et al. 7,8-dihydroxyflavone prevents synaptic loss and memory deficits in a mouse model of Alzheimer’s disease. Neuropsychopharmacology 39, 638–650 (2014).

    Article  PubMed  Google Scholar 

  54. Xiang, J. et al. Delta-secretase-cleaved Tau antagonizes TrkB neurotrophic signalings, mediating Alzheimer’s disease pathologies. Proc. Natl Acad. Sci. USA 116, 9094–9102 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  56. Ioannidis, J. P. Why most published research findings are false. PLoS Med. 2, e124 (2005).

    Article  PubMed  PubMed Central  Google Scholar 

  57. Collins, F. S. & Tabak, L. A. Policy: NIH plans to enhance reproducibility. Nature 505, 612–613 (2014).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  58. McNutt, M. Reproducibility. Science 343, 229 (2014).

    Article  ADS  CAS  PubMed  Google Scholar 

  59. Mullard, A. Cancer reproducibility project yields first results. Nat. Rev. Drug Discov. 16, 77 (2017).

    PubMed  Google Scholar 

  60. Horrigan, S. K. et al. Replication study: melanoma genome sequencing reveals frequent PREX2 mutations. eLife 6, e21634 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  61. Horrigan, S. K., Reproducibility Project: Cancer Biology. Replication study: the CD47-signal regulatory protein alpha (SIRPa) interaction is a therapeutic target for human solid tumors. eLife 6, e18173 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work performed at Emory University School of Medicine was supported by NIH grants RF1 AG051538 and R01 AG065177 to K.Y. K.Y. thanks the staff at the Alzheimer’s Disease Research Center at Emory University for human samples; the Rodent Behavioral Core (RBC), one of the Emory Integrated Core Facilities; the Viral Vector Core of the Emory Neuroscience NINDS Core Facility (P30 NS055077); and the NIH’s Georgia Clinical and Translational Science Alliance (UL1 TR002378). Work at Icahn School of Medicine at Mount Sinai performed at the Center for Translational Medicine and Pharmacology (CeTMaP) was supported by U19 AG060917 to M.Z. and C.J.R.; R01 DK113627 to M.Z. and J.I.; and R01 AG071870, R01 AG074092 and U01 AG073148 to T.Y. and M.Z. M.Z. thanks the Harrington Discovery Institute for the Innovator–Scholar Award towards development of the FSH-Ab. C.J.R. acknowledges support from the NIH (P20 GM121301 to C.J.R.). We thank M. Ehrlich and S. Gandy for their intellectual contributions, and S. Babunovic for proofreading the final version of the paper.

Author information

Authors and Affiliations

Authors

Contributions

K.Y. conceived the idea that FSH may be a mediator of AD in post-menopausal women and, together with M.Z., jointly proposed that blocking FSH using M.Z.’s polyclonal FSH-Ab could prevent AD. Thereafter, V.R., M.Z. and T.Y. designed the experiments, analysed the data and jointly wrote the manuscript. J.X., S.S.K. and Z.W. designed and performed most of the experiments and analysed the data. X.L. prepared primary neurons and assisted with mouse experiments. P.C. performed RNAscope studies. V.R. and A.G. recorded and analysed RNAscope data. F.K., J.B. and S.M. conducted studies with APP/PS1 mice for contemporaneous replication6,29. K.I. and D.L. performed the Fshr mRNA expression studies. D.S., A.P. and S.G. generated FSH-Ab and Hu6. T.-C.K. and S.G. performed antibody distribution studies. P.K. and V.H. conceived and performed the ViewRNA studies with human brain. S.-P.Y. and Z.Z. assisted with data analysis and interpretation. In summary, K.Y.’s laboratory was primarily responsible for generating data in Figs. 1a–e, 2a–c, f, 3, 4 and 5 and Extended Data Figs. 1a–e, 2a–c, f and 310. M.Z.’s laboratory produced data in Figs. 1f and 2d–e, g and Extended Data Fig. 1i–p. Moreover, J.I., K.A.G. and C.J.R. assisted with the conception of experiments and manuscript preparation. T.Y., V.M. and V.R. checked image integrity with the help of J.X., S.S.K. and K.Y.; A.G. and T.F. edited and revised the manuscript. T.-C.K. rechecked the raw data files. T.Y., V.R., M.Z. and K.Y. oversaw overall data management and provenance.

Corresponding authors

Correspondence to Mone Zaidi or Keqiang Ye.

Ethics declarations

Competing interests

M.Z. is listed as an inventor on issued patents on inhibiting FSH for the prevention and treatment of osteoporosis and obesity: US patent numbers 5,436,285 (1995, awarded to Icahn School of Medicine at Mount Sinai (ISMMS)), 5,674,887 (1997, awarded to ISMMS and University of Pittsburgh), 8,435,948 (2013, awarded to ISMMS) and 11,034,761 (2021, awarded to ISMMS). M.Z. is also listed as an inventor on a pending patent application on composition and use of humanized monoclonal anti-FSH antibodies. These patents are owned by ISMMS, and M.Z. would be recipient of royalties according to institutional policy. M.Z. and K.Y. are listed as inventors of a pending patent application on the use of FSH as a target for preventing Alzheimer’s disease. The latter patent is jointly owned by ISMMS and Emory University, and M.Z. and K.Y. would be recipient of royalties according to institutional policy. The other authors declare no competing interests.

Peer review

Peer review information

Nature thanks Li Gan and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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 Fig. 1 Effects of Anti-FSHβ Antibody in Reversing Ovariectomy-Induced Neuropathology in 3xTg Mice.

a, Ovariectomized 3xTg mice displayed hypoplastic thread-like uteri and elevated serum FSH and LH levels. FSH-Ab (200 µg/mouse, every 2 days, i.p., 8 weeks) did not alter total serum FSH, LH or 17β-estradiol levels. Statistics: mean ± s.e.m., N = 8 mice per group, one-way ANOVA. b, Immunofluorescent micrographs showing enhanced labelling in the following pairs: amyloid β (Aβ, red) and thioflavin-S (Thio-S, green); Aβ (red) and cleaved APPC586 (green); and pTau (red) and Tau1–368 (green) in the hippocampus after OVX, and its amelioration with FSH-Ab (scale bar, 20 μm). c, Upregulation of Cebpb, Lgmn, App and Mapt in OVX mouse brains, with reversal to near-baseline with FSH-Ab. Statistics: mean ± s.e.m., N = 3 mice per group, one-way AVOVA. d, Immunofluorescence micrographs showing that OVX induces apoptosis (TUNEL, green) in hippocampal NeuN-positive neurons (red); this apoptosis is abolished by FSH-Ab (scale bar, 20 μm). e, Golgi staining on brain sections from CA1 region post-OVX shows a substantial reduction in spine numbers, which is corrected with the FSH-Ab (scale bar, 5 μm). Statistics: mean ± s.e.m., N = 3 mice per group (10 sections), one-way AVOVA. f, Transmission electron micrographic images and quantitative analysis of synapses in hippocampal sections post-OVX treated with IgG or FSH-Ab (scale bar, 1 μm). Statistics: mean ± s.e.m., N = 3 mice per group (8 sections), one-way AVOVA. g, Morris Water Maze testing shows no differences in swim speed. Statistics: mean ± s.e.m., N = 9 mice per group, one-way ANOVA. h, Cognitive testing using the Novel Object Recognition test revealed the absence of significant difference between APP/PS1 and non-transgenic mice in Discrimination Index [(Novel Object Head Entry – Familiar Object Head Entry)/Total Head Entry]; the result is expected at 9 months of age in APP/PS1 mice. Thus, no effect of FSH-Ab was noted at this age, despite the reduction in Aβ40 and Aβ42 accumulation shown in Fig. 1f. Statistics: mean ± s.e.m., mice per group 9, 10, 4 and 4 from left to right; Whisker plot, upper and lower ends of the whiskers show maxima and minima, line in box shows median, and upper and lower box boundaries show 75th and 25th percentile, respectively; unpaired two-tailed Student’s t-test; i, ELISA showing no cross-reactivity of FSH-Ab with LH. j, Western immunoblot showing no change in expression of the LHCGR in whole brain lysates upon OVX or FSH-Ab treatment (N = 2 mice per group). k, IVIS imaging of isolated tissues from mice injected with AlexaFluor750–FSH, i.v., showing localization of FSH in the brain (N = 3 mice per group). l, Western immunoblots of whole brain lysates showing that i.p. injection of human FSH (5 IU) causes an elevation of brain FSH (N = 3 mice per group). m, Immunofluorescence micrographs showing the detection of peripherally injected (i.p.) biotinylated FSH-Ab (red) and biotinylated goat IgG (red) in brain sections (scale bar, 20 μm). Note the absence of cellular or nuclear co-localization with MAP2 or DAPI, respectively. n, Representative PET image shows that 89Zr-labelled humanized monoclonal FSH-Ab (89Zr-Hu6), injected i.v., is localized to live brain (arrows). γ-counting in perfused tissue shows presence of 89Zr-Hu6 in dissected brain tissue at 24 and 48 h post-injection (N = 4 mice). o, IVIS imaging and quantitation with AlexaFluor750-labelled Hu6, given i.v. shows localization in perfused whole brain tissue; N = 3 mice per group. Control (Ctrl): phosphate-buffered saline (PBS). p, Confirmatory immunofluorescence on the same mice (o) using anti-human IgG showing Hu6 localization (red) in proximity to CD31+ endothelial cells (green) (scale bar, 100 µm). For gel source data, see Supplementary Fig. 1.

Source data

Extended Data Fig. 2 FSHR Activation Triggers Amyloidogenic Protein Accumulation.

a, Western immunoblots showing the effect of activating neuronal FSHRs by FSH (30 ng/mL) in human SH-SY5Y and primary rat neuronal cells on the expression of C/EBPβ, AEP, as well as the cleavage of amyloid precursor protein (APP) and Tau using antibodies noted in ‘Methods’. FSH (30 ng/mL) likewise stimulated the expression of CEBPB, LGMN, APP and MAPT (qPCR) (b); AEP activity (c); and certain inflammatory cytokines (ELISA), namely IL-6 and IL-1β (d). Statistics: mean ± s.e.m.; Mice per group, (b) 3, (c) 6, and (d) 6; one-way ANOVA. e, Western immunoblotting showing C/EBPβ, AEP, APP, cleaved APP1–585, total Tau, and cleaved Tau1–368 in response to FSH or PBS following transfection with human FSHR siRNA (si-FSHR) for SH-SY5Y cells or rat Fshr siRNA (si-Fshr) for primary rat neurons, or appropriate scrambled siRNAs. f, mRNA levels of CEBPB, LGMN, APP and MAPT in SH-SY5Y cells incubated with FSH after control or si-FSHR transfection. g, AEP activity after incubation with FSH in control or si-FSHR-transfected SH-SY5Y cells. h, IL-6 and IL-1β levels (ELISAs) in SH-SY5Y cells incubated with FSH following control or si-FSHR infection. Statistics: mean ± s.e.m.; (f) 3 biological replicates; (g, h) 6 mice per group; one-way ANOVA.

Source data

Extended Data Fig. 3 FSH Induces APP and Tau Cleavage Through C/EBPβ and AEP/δ-Secretase Activation in Human SH-SY5Y Cells and Rat Cortical Neurons.

a, Western immunoblots showing the effect of FSH (30 ng/mL) on Tau, APP, AEP and FSHR of knocking down C/EBPβ expression by lentiviral infection with shRNA-Cebpb (sh-Cebpb) or reducing δ-secretase activity by adeno-associated virus infection of AEPC189S in both human SH-SY5Y cells and rat cortical neurons. The stimulatory action of FSH was reversed at 48 h. b, c, Effect of FSH (30 ng/mL) on APP, APPC586, pTau and Tau1–368 accumulation (immunofluorescence, scale bar, 40 μm, b) and AEP activity (c) in rat cortical neurons infected with sh-Cebpb or AAV-AEPC189S. Statistics: (c) mean ± s.e.m.; N = 6 mice per group; one-way ANOVA. d, Western immunoblots showing the time course of FSH effects on C/EBPβ, phosphorylated C/EBPβ (pC/EBPβ), AEP, pAEPS226, total AKT, pAKTS473, total ERK1/2, pERK1/2, total SRPK2, pSRPK2T492 and pNFκB-p65. e, Western immunoblots showing the effect of a 30-minute incubation with FSH (30 ng/mL) on levels of C/EBPβ, AEP, pAEPS226, total AKT and pAKTS473, total ERK1/2 and pERK1/2, total SRPK2 and pSRPK2T492 in the presence or absence of the cAMP inhibitor SQ22536 (100 µM), Gαi inhibitor pertussis toxin (PTX, 50 ng/ml), AKTi-1/2 inhibitor (10 μM) and ERK1/2 inhibitor PD98059 (10 μM).

Source data

Extended Data Fig. 4 Targeted Knockdown of Fshr in the Hippocampus Diminishes AD Pathologies.

a, Quantitative PCR shows significantly reduced expression of Fshr, Cebpb, Lgmn, App and Mapt. b, Immunohistochemistry of the hippocampus shows reduced accumulation of Aβ and pTau, as well as of proteinaceous deposits (silver staining) in si-Fshr-injected OVX mice (scale bar, 50 μm). c, The two isoforms of Aβ, namely Aβ40 and Aβ42, were also reduced. d, Notable is the marked increase in dendritic spines (Golgi staining) (scale bar, 5 μm). Statistics: mean ± s.e.m., (a) 3 biological replicates; (c) 5 mice per group; (d) 10 sections from 3 mice per group; unpaired two-tailed Student’s t-test.

Source data

Extended Data Fig. 5 Recombinant FSH Triggers AD Pathology in 3xTg Mice.

a, Serum FSH levels—both mouse (endogenous) and human (exogenous)—24 h after i.p. injection of 2, 5 or 10 IU human recombinant FSH. b, Serum LH levels also shown. Female 3xTg mice were injected with recombinant FSH (5 IU per mouse, daily, i.p., 3 months). c, Immunohistochemistry for Aβ or pTau in hippocampus post-FSH injection (scale bar, 50 μm). d, Silver staining of the prefrontal cortex, and hippocampal CA1 and dentate gyrus (DG) regions showing enhanced proteinaceous deposits in FSH-injected mice (scale bar, 50 μm). e, Brain mRNA levels of Cebpb, Lgmn, App and Mapt. f, Golgi staining of brain sections from the CA1 region shows reduced spine numbers in FSH-injected mice (scale bar, 5 μm). g, Transmission electron micrographs of hippocampal sections showing reduced synapse numbers post-FSH (scale bar, 1 μm). Immunofluorescence micrographs showing the following image pairs in the hippocampus and/or cortex post-FSH: (h) Aβ (red) and cleaved APPC586 (green); (i) pTau (red) and cleaved Tau1–368 (green); (j) Aβ (red) and thioflavin-S (green); and (k) NeuN (red) and TUNEL (green) (scale bar, 20 μm). l, Immunofluorescence showing co-localization of C/EBPβ, AEP, Aβ and pTau to NeuN-positive neurons upon FSH stimulation [10x (scale bar, 300 μm) and 40x (scale bar, 50 μm) magnifications]. Statistics: mean ± s.e.m., (a, b) 3 mice per group; (e) 3 biological replicates, (f, g) 10 sections from 3 mice per group; unpaired two-tailed Student’s t-test.

Source data

Extended Data Fig. 6 Effect of Recombinant FSH in Triggering AD Pathology in Ovariectomized 3xTg Mice With Oestrogen Replacement.

3xTg mice were ovariectomized at 3 months and supplemented with 17β-estradiol using 90-day-release pellets (E2, 0.36 mg) to render them biochemically eugonadal. The mice were randomly divided to be injected with PBS or recombinant human FSH (5 IU per mouse, daily, i.p., 3 months). a, Serum level of FSH and 17β-estradiol. b, Western immunoblotting showing increased C/EBPβ, AEP, cleaved APP1–373 and APP1–585, total Tau, cleaved Tau1–368 and pTau in the brain after FSH injection. c, Brain AEP enzymatic activity also shown. d, Immunohistochemistry of the hippocampus shows increased expression of Aβ and pTau in the FSH group. Silver staining showed increased proteinaceous deposits in FSH-treated mice (scale bar, 50 μm). Statistics: mean ± s.e.m., mice per group; (a) 4 and (c) 5; unpaired two-tailed Student’s t-test.

Source data

Extended Data Fig. 7 Effect of Recombinant FSH in Triggering AD Pathology and Cognitive Decline in Male Mice.

Male 3xTg mice were injected with recombinant FSH at 5 IU per mouse daily, i.p. for 3 months. a, Western immunoblots showing increased C/EBPβ, AEP, cleaved APP1–373 and APP1–585, total Tau, cleaved Tau1–368 and pTau in the brain (3 mice per group). b, c, Brain AEP activity (b) and Aβ isoforms, Aβ40 and Aβ42 (c) were also increased with FSH. d, Morris Water Maze test shows enhanced escape latency to mount the platform (seconds). Also shown are integrated escape latency (area under the curve, AUC) and percentage of time spent in the target quadrant (Probe Trial Test). e, Silver staining of the prefrontal cortex, and hippocampus CA1 and dentate gyrus (DG) regions showing enhanced proteinaceous deposits in FSH-injected mice (scale bar, 50 μm). f, Immunohistochemistry for Aβ or pTau in the hippocampus post-FSH injection (scale bar, 50 μm). g, Brain mRNA levels of Cebpb, Lgmn, App and Mapt. h, Golgi staining of brain sections from the CA1 region of the hippocampus showing reduced spine numbers in FSH-injected mice (scale bar, 5 μm). i, Transmission electron micrographs of hippocampal sections showing reduced synapse numbers post-FSH (scale bar, 1 μm). Immunofluorescence micrographs showing the following image pairs: (j) Aβ (red) and cleaved APPC586 (green); (k) pTau (red) and cleaved Tau1–368 (green); (l) Aβ (red) and thioflavin-S (green); and (m) NeuN (red) and TUNEL (green) in the hippocampus and/or cortex of male 3xTg mice after FSH (scale bar, 20 μm). Statistics: mean ± s.e.m., mice per group, (b, c) 5, (d) 7, (g) 3, (h, i), 3 (10 sections); unpaired two-tailed Student’s t-test.

Source data

Extended Data Fig. 8 Effect of FSH in Triggering AD Pathology and Cognitive Decline in Female Wild Type and APP-KI Mice.

In APP-KI mice, three amino acid substitutions (G601R;F606Y;R609H) are knocked into Aβ-coding exon 14 of the APP gene—this results in the non-transgenic expression at basal levels of oligomerizable human Aβ. Female wild type and APP-KI mice were injected with recombinant FSH (5 IU, daily, i.p. 3 months). a–d, In wild type mice, Western immunoblotting showing increased C/EBPβ, AEP, cleaved APP1–373 and APP1–585, total Tau, and cleaved Tau1–368 in whole brain (3 mice per group) (a), as well as increased silver staining (b), elevated AEP activity (c) and increases Aβ isoforms, Aβ40 and Aβ42 (d) upon FSH treatment. e, Morris Water Maze test, however, showed no difference in escape latency to mount the platform (seconds). Also shown are no differences in integrated escape latency (area under the curve, AUC) and percentage of time spent in the target quadrant (Probe Trial Test). f–I, Western immunoblotting showing elevations in C/EBPβ, AEP, cleaved APP1–373 and APP1–585, and cleaved Tau1–368 in whole brain (f), along with enhancements in silver staining (g), AEP activity (h), and Aβ isoforms (i) in APP-KI mice in response to FSH injection. j, There was also a significant spatial memory deficit on the Morris Water Maze test. k, Immunofluorescence micrographs showed increases in the hippocampus and/or cortex of female APP-KI mice post-FSH injected in the following pairs: Aβ (red) and cleaved APPC586 (green); Aβ (red) and thioflavin-S (green); and NeuN (red) and TUNEL (green). Scale bar: (b, g) 50 μm, (k) 100 μm (magnified view, 10 μm). Statistics: mean ± s.e.m.; mice per group, (c, d, h, i) 5, (e, j) 8; unpaired two-tailed Student’s t-test.

Source data

Extended Data Fig. 9 C/EBPβ Mediates FSH-Induced AD Neuropathology and Cognitive Decline in 3xTg Mice.

a, Cebpb, Lgmn, App and Mapt mRNA expression following FSH injection to 3xTg or Cebpb+/− 3xTg mice. Statistics: mean\(\,\pm \) s.e.m., N = 3 biological replicates, one-way ANOVA. b, Immunohistochemistry for Aβ and pTau and silver staining for proteinaceous deposits (scale bar, 50 μm). c–e, Immunofluorescence staining for Aβ (red) and C/EBPβ (green) (c) and for pTau (red) and C/EBPβ (green) (d) (scale bar, 20 μm), and Golgi staining for dendritic spines (e) in the hippocampus in female 3xTg or Cebpb+/− 3xTg mice, post-FSH (scale bar, 5 μm). Statistics: mean ± s.e.m., 10 sections from 3 mice per group, one-way ANOVA. f, Morris Water Maze testing showed no difference in swim speed. Statistics: 9 mice per group, one-way ANOVA.

Source data

Extended Data Fig. 10 C/EBPβ Mediates Ovariectomy-Induced AD Neuropathology and Cognitive Decline in 3xTg Mice.

a, Cebpb, Lgmn, App and Mapt mRNA expression following ovariectomy of 3xTg or Cebpb+/− 3xTg mice. Statistics: mean ± s.e.m., 3 biological replicates, one-way ANOVA. b, Immunohistochemistry for Aβ and pTau and silver staining for proteinaceous deposits (scale bar, 50 μm). c–e, Immunofluorescence staining for Aβ (red) and C/EBPβ (green) (c) and for pTau (red) and C/EBPβ (green) (d) (scale bar, 20 μm), and Golgi staining for dendritic spines (e) in the hippocampus in female 3xTg or Cebpb+/− 3xTg mice (scale bar, 5 μm), post-OVX. Statistics: mean ± s.e.m., 10 sections from 3 mice per group, one-way ANOVA. f, Morris Water Maze test showed no difference in the swim speed between 3xTg and Cebpb+/− 3xTg mice. Statistics: left to right: 7, 8, 8 mice per group, one-way ANOVA.

Source data

Supplementary information

Supplementary Information

Supplementary Fig. 1: uncropped agarose gels and western blots. Supplementary Table 1: information about the key antibodies used in this study.

Reporting Summary

Supplementary Video 1

PET scan of a mouse injected with 89Zr-labelled FSH antibody. Four C57BL/6 mice were injected through the tail vein with 100 µCi of the 89Zr-radiolabelled monoclonal FSH antibody Hu6. After 24 h, the mice were anaesthetized and then imaged using a Mediso NanoScan PET/CT scanner. In this representative video, 89Zr-labelled Hu6 was noted in the brain. These data provide compelling evidence that the FSH antibody can cross the blood–brain barrier.

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xiong, J., Kang, S.S., Wang, Z. et al. FSH blockade improves cognition in mice with Alzheimer’s disease. Nature 603, 470–476 (2022). https://doi.org/10.1038/s41586-022-04463-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-022-04463-0

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: 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