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.

  • Letter
  • Published:

Therapeutic inhibition of mTORC2 rescues the behavioral and neurophysiological abnormalities associated with Pten-deficiency

Abstract

Dysregulation of the mammalian target of rapamycin (mTOR) signaling, which is mediated by two structurally and functionally distinct complexes, mTORC1 and mTORC2, has been implicated in several neurological disorders1,2,3. Individuals carrying loss-of-function mutations in the phosphatase and tensin homolog (PTEN) gene, a negative regulator of mTOR signaling, are prone to developing macrocephaly, autism spectrum disorder (ASD), seizures and intellectual disability2,4,5. It is generally believed that the neurological symptoms associated with loss of PTEN and other mTORopathies (for example, mutations in the tuberous sclerosis genes TSC1 or TSC2) are due to hyperactivation of mTORC1-mediated protein synthesis1,2,4,6,7. Using molecular genetics, we unexpectedly found that genetic deletion of mTORC2 (but not mTORC1) activity prolonged lifespan, suppressed seizures, rescued ASD-like behaviors and long-term memory, and normalized metabolic changes in the brain of mice lacking Pten. In a more therapeutically oriented approach, we found that administration of an antisense oligonucleotide (ASO) targeting mTORC2’s defining component Rictor specifically inhibits mTORC2 activity and reverses the behavioral and neurophysiological abnormalities in adolescent Pten-deficient mice. Collectively, our findings indicate that mTORC2 is the major driver underlying the neuropathophysiology associated with Pten-deficiency, and its therapeutic reduction could represent a promising and broadly effective translational therapy for neurological disorders where mTOR signaling is dysregulated.

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: Genetic inhibition of mTORC1 (but not mTORC2) reverses increase in brain size, while genetic inhibition of mTORC2 (but not mTORC1) prolongs survival in Pten fb-KO mice.
Fig. 2: Genetic inhibition of mTORC2 prevents the development of seizures in Pten fb-KO mice.
Fig. 3: Genetic inhibition of mTORC2 prevents the behavioral abnormalities in Pten fb-KO mice.
Fig. 4: Injection of antisense oligonucleotide targeting mTORC2 component Rictor improved seizures, memory and ASD-like behaviors in Pten fb-KO mice.

Similar content being viewed by others

Data availability

The data supporting the findings of this study are available in this paper or the Supplementary Information. Full uncropped blots are available as Source data. Any other raw data that support the findings of this study are available from the corresponding author upon reasonable request. The request for Rictor-ASO will be promptly reviewed by Ionis Pharmaceuticals to verify that the request is subject to any intellectual property confidential obligations. If not, the ASO will be available upon completion of a standard Material Transfer Agreement with Ionis Pharmaceuticals.

References

  1. Ehninger, D. & Silva, A. J. Rapamycin for treating tuberous sclerosis and autism spectrum disorders. Trends Mol. Med. 17, 78–87 (2011).

    CAS  PubMed  Google Scholar 

  2. Winden, K. D., Ebrahimi-Fakhari, D. & Sahin, M. Abnormal mTOR activation in autism. Annu. Rev. Neurosci. 41, 1–23 (2018).

    CAS  PubMed  Google Scholar 

  3. Costa-Mattioli, M. & Monteggia, L. M. mTOR complexes in neurodevelopmental and neuropsychiatric disorders. Nat. Neurosci. 16, 1537–1543 (2013).

    CAS  PubMed  Google Scholar 

  4. Zhou, J. & Parada, L. F. PTEN signaling in autism spectrum disorders. Curr. Opin. Neurobiol. 22, 873–879 (2012).

    CAS  PubMed  Google Scholar 

  5. Knafo, S. & Esteban, J. A. PTEN: local and global modulation of neuronal function in health and disease. Trends Neurosci. 40, 83–91 (2017).

    CAS  PubMed  Google Scholar 

  6. Kelleher, R. J. 3rd & Bear, M. F. The autistic neuron: troubled translation? Cell 135, 401–406 (2008).

    CAS  PubMed  Google Scholar 

  7. Hoeffer, C. A. & Klann, E. mTOR signaling: at the crossroads of plasticity, memory and disease. Trends Neurosci. 33, 67–75 (2010).

    CAS  PubMed  Google Scholar 

  8. Lipton, J. O. & Sahin, M. The neurology of mTOR. Neuron 84, 275–291 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Wullschleger, S., Loewith, R. & Hall, M. N. TOR signaling in growth and metabolism. Cell 124, 471–484 (2006).

    CAS  PubMed  Google Scholar 

  10. Saxton, R. A. & Sabatini, D. M. mTOR signaling in growth, metabolism and disease. Cell 169, 361–371 (2017).

    CAS  PubMed  Google Scholar 

  11. Sarbassov, D. D. et al. Rictor, a novel binding partner of mTOR, defines a rapamycin-insensitive and raptor-independent pathway that regulates the cytoskeleton. Curr. Biol. 14, 1296–1302 (2004).

    CAS  PubMed  Google Scholar 

  12. Hay, N. & Sonenberg, N. Upstream and downstream of mTOR.Genes Dev. 18, 1926–1945 (2004).

    CAS  PubMed  Google Scholar 

  13. Buffington, S. A., Huang, W. & Costa-Mattioli, M. Translational control in synaptic plasticity and cognitive dysfunction. Annu. Rev. Neurosci. 37, 17–38 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Crino, P. B. The mTOR signalling cascade: paving new roads to cure neurological diseaseNat. Rev. Neurol. 12, 379–392 (2016).

    CAS  PubMed  Google Scholar 

  15. Zhou, J. et al. Pharmacological inhibition of mTORC1 suppresses anatomical, cellular and behavioral abnormalities in neural-specific Pten knock-out mice. J. Neurosci. 29, 1773–1783 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Ljungberg, M. C., Sunnen, C. N., Lugo, J. N., Anderson, A. E. & D’Arcangelo, G. Rapamycin suppresses seizures and neuronal hypertrophy in a mouse model of cortical dysplasia. Dis. Models Mech. 2, 389–398 (2009).

    CAS  Google Scholar 

  17. Nguyen, L. H. et al. mTOR inhibition suppresses established epilepsy in a mouse model of cortical dysplasia. Epilepsia 56, 636–646 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Hobert, J. A., Embacher, R., Mester, J. L., Frazier, T. W.II & Eng, C. Biochemical screening and PTEN mutation analysis in individuals with autism spectrum disorders and macrocephaly. Eur. J. Hum. Genet 22, 273–276 (2014).

    CAS  PubMed  Google Scholar 

  19. Tilot, A. K. et al. Neural transcriptome of constitutional Pten dysfunction in mice and its relevance to human idiopathic autism spectrum disorder. Mol. Psychiatry 21, 118–125 (2016).

    CAS  PubMed  Google Scholar 

  20. McBride, K. L. et al. Confirmation study of PTEN mutations among individuals with autism or developmental delays/mental retardation and macrocephaly.Autism Res. 3, 137–141 (2010).

    PubMed  Google Scholar 

  21. Hu, W. F., Chahrour, M. H. & Walsh, C. A. The diverse genetic landscape of neurodevelopmental disorders. Annu. Rev. Genomics Hum. Genet. 15, 195–213 (2014).

    PubMed  Google Scholar 

  22. Mirzaa, G. M. & Poduri, A. Megalencephaly and hemimegalencephaly: breakthroughs in molecular etiology. Am. J. Med. Genet. C Semin. Med. Genet. 166C, 156–172 (2014).

    PubMed  Google Scholar 

  23. Kwon, C. H. et al. Pten regulates neuronal arborization and social interaction in mice. Neuron 50, 377–388 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Backman, S. A. et al. Deletion of Pten in mouse brain causes seizures, ataxia and defects in soma size resembling Lhermitte–Duclos disease. Nat. Genet. 29, 396–403 (2001).

    CAS  PubMed  Google Scholar 

  25. Kwon, C. H. et al. Pten regulates neuronal soma size: a mouse model of Lhermitte–Duclos disease. Nat. Genet. 29, 404–411 (2001).

    CAS  PubMed  Google Scholar 

  26. Ostendorf, A. P. & Wong, M. mTOR inhibition in epilepsy: rationale and clinical perspectives. CNS Drugs 29, 91–99 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Noebels, J. L Single-gene determinants of epilepsy comorbidity. Cold Spring Harb. Perspect. Med. 5, a022756 (2015).

    PubMed  PubMed Central  Google Scholar 

  28. Fombonne, E. The epidemiology of autism: a review. Psychol. Med. 29, 769–786 (1999).

    CAS  PubMed  Google Scholar 

  29. Kazdoba, T. M. et al. Translational mouse models of autism: advancing toward pharmacological therapeutics. Curr. Top. Behav. Neurosci. 28, 1–52 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Jiang, Y. H. & Ehlers, M. D. Modeling autism by SHANK gene mutations in mice. Neuron 78, 8–27 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. American Psychiatric Association & American Psychiatric Association DSM-5 Task Force Diagnostic and Statistical Manual of Mental Disorders: DSM-5 (American Psychiatric Association, 2013).

  32. Lopez, B. R., Lincoln, A. J., Ozonoff, S. & Lai, Z. Examining the relationship between executive functions and restricted, repetitive symptoms of autistic disorder. J. Autism Dev. Disord. 35, 445–460 (2005).

    PubMed  Google Scholar 

  33. Stoica, L. et al. Selective pharmacogenetic inhibition of mammalian target of rapamycin complex I (mTORC1) blocks long-term synaptic plasticity and memory storage. Proc. Natl Acad. Sci. USA 108, 3791–3796 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Huang, W. et al. mTORC2 controls actin polymerization required for consolidation of long-term memory. Nat. Neurosci. 16, 441–448 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Garcia-Cao, I. et al. Systemic elevation of PTEN induces a tumor-suppressive metabolic state. Cell 149, 49–62 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Hagiwara, A. et al. Hepatic mTORC2 activates glycolysis and lipogenesis through Akt, glucokinase and SREBP1c. Cell Metab. 15, 725–738 (2012).

    CAS  PubMed  Google Scholar 

  37. Masui, K. et al. mTOR complex 2 controls glycolytic metabolism in glioblastoma through FoxO acetylation and upregulation of c-Myc. Cell Metab. 18, 726–739 (2013).

    CAS  PubMed  Google Scholar 

  38. Panasyuk, G. et al. PPARγ contributes to PKM2 and HK2 expression in fatty liver. Nat. Commun. 3, 672 (2012).

    PubMed  Google Scholar 

  39. Robey, R. B. & Hay, N. Is Akt the ‘Warburg kinase’? Akt-energy metabolism interactions and oncogenesis. Semin. Cancer Biol. 19, 25–31 (2009).

    CAS  PubMed  Google Scholar 

  40. Rinaldi, C. & Wood, M. J. A. Antisense oligonucleotides: the next frontier for treatment of neurological disorders. Nat. Rev. Neurol. 14, 9–21 (2018).

    CAS  PubMed  Google Scholar 

  41. Vickers, T. A. et al. Efficient reduction of target RNAs by small interfering RNA and RNase H-dependent antisense agents. A comparative analysis. J. Biol. Chem. 278, 7108–7118 (2003).

    CAS  PubMed  Google Scholar 

  42. Cioffi, C. L. et al. Selective inhibition of A-Raf and C-Raf mRNA expression by antisense oligodeoxynucleotides in rat vascular smooth muscle cells: role of A-Raf and C-Raf in serum-induced proliferation. Mol. Pharmacol. 51, 383–389 (1997).

    CAS  PubMed  Google Scholar 

  43. Zhang, H. et al. Reduction of liver Fas expression by an antisense oligonucleotide protects mice from fulminant hepatitis. Nat. Biotechnol. 18, 862–867 (2000).

    CAS  PubMed  Google Scholar 

  44. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S. L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009).

    PubMed  PubMed Central  Google Scholar 

  45. Zhu, P. J. et al. Suppression of PKR promotes network excitability and enhanced cognition by interferon-γ-mediated disinhibition. Cell 147, 1384–1396 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Buffington, S. A. et al. Microbial reconstitution reverses maternal diet-induced social and synaptic deficits in offspring. Cell 165, 1762–1775 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Silverman, J. L., Yang, M., Lord, C. & Crawley, J. N. Behavioural phenotyping assays for mouse models of autism. Nat. Rev. Neurosci. 11, 490–502 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Deacon, R. M. & Rawlins, J. N. T-maze alternation in the rodent. Nat. Protoc. 1, 7–12 (2006).

    PubMed  Google Scholar 

  49. Johnson, J. L., Huang, W., Roman, G. & Costa-Mattioli, M. TORC2: a novel target for treating age-associated memory impairment. Sci. Rep. 5, 15193 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Zhu, P. J., Chen, C. J., Mays, J., Stoica, L. & Costa-Mattioli, M. mTORC2, but not mTORC1, is required for hippocampal mGluR-LTD and associated behaviors. Nat. Neurosci. 21, 799–802 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Huang, W et al. Translational control by eIF2α phosphorylation regulates vulnerability to the synaptic and behavioral effects of cocaine. eLife 5, e12052 (2016).

    PubMed  PubMed Central  Google Scholar 

  52. Ma, Y., Bai, R. K., Trieu, R. & Wong, L. J. Mitochondrial dysfunction in human breast cancer cells and their transmitochondrial cybrids. Biochim. Biophys. Acta 1797, 29–37 (2010).

    CAS  PubMed  Google Scholar 

  53. Frazier, A. E. & Thorburn, D. R. Biochemical analyses of the electron transport chain complexes by spectrophotometry. Methods Mol. Biol. 837, 49–62 (2012).

    CAS  PubMed  Google Scholar 

  54. Venegas, V. & Halberg, M. C. Measurement of mitochondrial DNA copy number. Methods Mol. Biol. 837, 327–335 (2012).

    CAS  PubMed  Google Scholar 

  55. Swayze, E. E. et al. Antisense oligonucleotides containing locked nucleic acid improve potency but cause significant hepatotoxicity in animals. Nucleic Acids Res. 35, 687–700 (2007).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank A. Placzek and members of the Costa-Mattioli laboratory for comments on the manuscript. This work was supported by funds to M.C.-M. (NIMH 096816, Department of Defense AR120254 and Sammons Enterprises), J.N. (NINDS NS29709) and J.C. (NS085171).

Author information

Authors and Affiliations

Authors

Contributions

C.-J.C. and M.C.-M. conceived and planned experiments. C.-J.C., M.S., J.M., H.Z., R.L., J.P., I.-C.W., J.H.P. and L.S. performed the experiments and analyzed the data. J.C. and J.L.N. contributed to the seizures study design and analysis, and B.A.K. contributed to the mitochondria function analysis. P.J.-N. and F.R. designed the ASOs, performed experiments to test Rictor knockdown and immune response activation. C.-J.C. and M.C.M. wrote the manuscript, with contributions from M.S., J.M., P.J.-N., J.C. and J.L.N.

Corresponding author

Correspondence to Mauro Costa-Mattioli.

Ethics declarations

Competing interests

P.J.-N. and F.R. are employed by Ionis Pharmaceutical, a company that develops ASO therapies. The authors declare no other competing interests.

Additional information

Peer review information Brett Benedetti was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 Selective inhibition of mTORC1 or mTORC2 activity in Pten fb-KO mice.

(a) Schematics of generation of forebrain neuron specific Pten-deficient (Pten fb-KO) and double knockout mice (Pten-Rptor fb-DKO and Pten-Rictor fb-DKO). (b-e) Representative western blot (b) and quantification show reduced PTEN levels in Pten fb-KO mice (c, n = 5, t = 5.22, P < 0.0001), Pten-Rptor fb-DKO (c, n = 5, t = 5.34, P< 0.0001) and Pten-Rictor fb-DKO (c, n = 5, t= 4.50, P= 0.0004) and reduced Raptor (d, n = 5 per group, vs. control t = 3.52, P= 0.0028) or Rictor (e, n = 5, t = 3.83, P= 0.0015) levels in the hippocampus of Pten-Rptor fb-DKO mice or Pten-Rictor fb-DKO mice, respectively. Representative western blot (f) and quantification (g-h) show increased mTORC1 (g, p-S6 at S240/244) and mTORC2 activity (h, p-Akt at S473) in cortex of Pten fb-KO mice (n = 6 per group, p-S6: t = 2.32, P = 0.03, p-Akt: t = 2.81, P = 0.0111). The increased mTORC1 activity in the cortex of Pten fb-KO mice was reduced in Pten-Rptor fb-DKO mice (n = 6, vs. Pten fb-KO, t = 2.87, P = 0.0091), but not in Pten-Rictor fb-DKO mice (n = 6, vs. Pten fb-KO, t = 0.13, P= 0.8958). By contrast, the increased mTORC2 activity in the cortex of Pten fb-KO mice was reduced in Pten-Rictor fb-DKO mice (t = 3.02, P= 0.0066), but not in Pten-Rptor fb-DKO mice (t = 3.92, P= 0.0009). Data are mean ± SEM. Statistics were based on one-way ANOVA followed by uncorrected two-sided Fisher’s LSD method for pairwise comparisons. n.s., not significant.

Source data

Extended Data Fig. 2 Genetic inhibition of mTORC1 (but not mTORC2) reverses the increased brain/body weight ratio in Pten fb-KO mice.

(a) Crystal violet staining of mid-sagittal brain sections from control, Pten fb-KO mice, Pten-Rptor fb-DKO and Pten-Rictor fb-DKO mice (3 mice per group were used for the experiment). (b) Body weight of control (n = 11), Pten fb-KO (n = 9), Pten-Rictor fb-DKO mice (n = 13) and Pten-Rptor fb-DKO (n = 9) was similar (F3,38 = 0.93; P= 0.443). (c) Brain weight relative to body weight in control (n = 11, 0.94 ± 0.04%), Pten fb-KO (n = 9, 2.45 ± 0.34%, vs. control, t= 5.13, P< 0.0001), Pten-Rptor fb-DKO (n = 8, 2.12 ± 0.08%, vs. Pten fb-KO, t= 3.145, P= 0.0034) and Pten-Rictor fb-DKO mice (n = 11, 2.52 ± 0.07%, vs. Pten fb-KO, t= 0.21, P= 0.7810). Box graphs are presented as median, 25th percentile, 75th percentile, minimum, and maximum of the group. Statistics were based on one-way ANOVA followed by uncorrected two-sided Fisher’s LSD method for pairwise comparisons or paired t-test. n.s., not significant.

Extended Data Fig. 3 Genetic inhibition of mTORC2 (but not mTORC1) reverses the increased excitatory synaptic transmission in Pten fb-KO mice.

(a-c) Summary data (a-b) and sample traces (c) show increased frequency (a, Control n = 7, Pten fb-KO n = 6, t = 4.62, P < 0.0001), but not amplitude (b, Control n = 7, Pten fb-KO n = 6, t = 0.47, P= 0.641) of spontaneous excitatory postsynaptic currents (sEPSCs) in CA1 neurons from Pten fb-KO mice compared to control littermates. Unlike in Pten-Rptor fb-DKO mice (n = 9; a, c, Control vs. Pten-Rptor fb-DKO: t = 6.26, P < 0.0001), in Pten-Rictor fb-DKO mice (n = 5), sEPSC frequency is restored to control values (a, c, Control vs. Pten-Rictor fb-DKO: t= 0.86, P = 0.99). Data are mean ± SEM. Statistics are based on one-way ANOVA followed by uncorrected two-sided Fisher’s LSD method for pairwise comparisons. (d-f) Summary data (d-e) and sample traces (f) show similar frequency (d, Control n = 8, Pten fb-KO n = 14, t = 0.47, P = 0.636) and amplitude (e, Control n = 8, Pten fb-KO n = 14, t = 0.09, P = 0.925) of spontaneous inhibitory postsynaptic currents (sIPSCs) in CA1 neurons from control and Pten fb-KO mice. Statistics were based two-sided Student’s t-test Data are mean ± SEM. (g) Input-output curves in CA1 pyramidal neurons show that Pten fb-KO mice and Pten-Rptor fb-DKO mice fired more action potentials (APs) than control littermates at 350pA or higher current injection (n = 7 per group, control vs. Pten fb-KO, 350pA: t = 2.76, P = 0.020; 400 pA: t = 3.23, P = 0.0041; 450 pA: t = 4.13, P = 0.0002). By contrast, the increased excitability in Pten fb-KO mice is reduced in Pten-Rictor fb-DKO mice (n = 7 per group, Pten fb-KO vs. Pten-Rictor fb-DKO, 350 pA: t = 2.76, P = 0.020; 400 pA: t = 3.05, P = 0.010; 450 pA: t = 3.42, P = 0.002. Data are mean ± SEM. Statistics were based on two-way ANOVA followed by Bonferroni’s two-sided multiple comparison test. (h) Representative response elicited in CA1 pyramidal neurons (at -70 mV) upon injection of 400 pA for 500 ms.

Extended Data Fig. 4 Genetic inhibition of mTORC2 rescues the deficits in reciprocal social interaction and long-term object recognition memory in Pten fb-KO mice.

(a) Schematic of the reciprocal social interaction task. (b) Compared to controls (n = 11), Pten fb-KO mice (n= 9) show decreased social interaction (t = 3.42, P = 0.0023). The deficits in reciprocal social interaction are restored in Pten-Rictor fb-DKO mice (n = 7, vs. Pten fb-KO mice, t = 4.69, P < 0.0001). (c) Normal freezing responses in Pten fb-KO mice during training. All groups show similar freezing responses either before (naïve: control, n = 15 vs. Pten fb-KO, n = 12, t = 0.13, P = 0.8948; control vs. Pten-Rptor fb-DKO, n = 12, t = 0.32, P = 0.7941; control vs. Pten-Rictor fb-DKO, n = 12, t = 0.04, P = 0.9668) or immediately after training (post-shock: control vs. Pten fb-KO, t = 0.66, P = 0.5904; control vs. Pten-Rptor fb-DKO, t = 1.46, P = 0.1492; control vs. Pten-Rictor fb-DKO, t = 1.23, P = 0.2238). (d) Schematic of the object recognition task. (e) Pten fb-KO mice showed impaired long-term object recognition memory [control (n = 9), Pten fb-KO (n = 6), t = 4.89, P < 0.0001), which was rescued in Pten-Rictor fb DKO mice [Pten-Rictor fb DKO mice (n= 8) vs. Pten fb-KO mice, t = 3.71, P = 0.0014; Pten-Rictor fb DKO mice vs. control, t = 1.18, P = 0.2518]. Data are mean ± SEM. Statistics were based on two-way ANOVA followed by uncorrected two-sided Fisher’s LSD method for pairwise comparisons. n.s., not significant.

Extended Data Fig. 5 Selective increase in p-Akt in Pten fb-KO mice is reduced by genetic inhibition of mTORC2.

Representative western blots (a) and quantifications (b-d) show that compared to control mice, p-Akt at Ser473 (but not p-PKC or p-NDRG1) is the only mTORC2 downstream target whose activity is upregulated in the brain of Pten fb-KO mice [control vs. Pten fb-KO (n = 5-6 per group), p-Akt (b): t = 6.11, P< 0.0001, p-PKC (c): t = 0.64, P = 0.5291, p-NDRG1 (d): t = 0.69, P = 0.4983). As expected, genetic deletion of mTORC2 reduced the activity of all three mTORC2 targets (p-Akt, p-PKC and p-NDRG1) in the brain of Pten-Rictor fb-DKO (n = 6, vs. control p-Akt (b): t = 0.89, P= 0.3903, p-PKC (c): t = 4.56, P = 0.0004, p-NDRG1 (d): t = 2.31, P = 0.0359; vs. Pten fb-KO, p-Akt: t = 7.33, P< 0.0001, p-PKC: t = 3.91, P = 0.0014, p-NDRG1: t = 2.99, P = 0.0090). Compared to Pten fb-KO mice, genetic deletion of mTORC1 in Pten-Rptor fb-DKO did not decrease p-Akt (e-f:,Pten fb-KO (n= 4) vs. Pten-Rptor fb-DKO (n= 6), t = 4.59, P = 0.0005; control (n= 5) vs. Pten-Rptor fb-DKO, t = 9.31, P < 0.0001), p-PKC (e,g: Pten fb-KO (n= 4) vs. Pten-Rptor fb-DKO (n= 5), t = 0.65, P = 0.5332; control (n= 4) vs. Pten-Rptor fb-DKO, t = 0.73, P = 0.4820) or p-NDRG1 (e,h: Pten fb-KO (n= 4) vs. Pten-Rptor fb-DKO (n= 5), t = 1.19, P = 0.2581; control (n= 4) vs. Pten-Rptor fb-DKO, t = 0.46, P = 0.6527). Data are mean ± SEM. Statistics were based on one-way ANOVA followed by uncorrected two-sided Fisher’s LSD method for pairwise comparisons. n.s., not significant.

Source data

Extended Data Fig. 6 Genetic inhibition of mTORC2 rescues the changes in glucose metabolism in the brain of Pten fb-KO mice.

(a) Schematic of mTORC2 regulation of glucose metabolism through the phosphorylation of Akt. (b-d) Metabolomic study revealed an increase in glycolysis metabolites in Pten fb-KO mice, which is normalized when mTORC2, but not mTORC1, is inhibited. The level of glucose (b), glucose-6-phosphate (c) and lactate (d) were increased in the cortex of Pten fb-KO mice (control (n= 8) vs. Pten fb-KO (n= 7): glucose, t = 3.51, P = 0.0017; glucose-6-phosphate, t = 2.77, P = 0.0103; lactate, t= 2.41, P = 0.024). The changes in glucose metabolism are reversed in Pten-Rictor fb-DKO mice (n= 7, vs. control: glucose, t = 0.067, P = 0.9469; glucose-6-phosphate, t = 0.39, P = 0.7009; lactate t = 1.39, P = 0.1771), but not in Pten-Rptor fb-DKO mice [Pten-Rictor fb-KO mice (n = 7) vs. Pten-Rptor fb-DKO (n = 7): glucose, t = 1.07, P = 0.2962; glucose-6-phosphate, t = 1.55, P = 0.1355; lactate t = 0.39, P = 0.6993]. Data are mean ± SEM. Statistics were based on one-way ANOVA followed by uncorrected two-sided Fisher’s LSD method for pairwise comparisons. n.s., not significant.

Extended Data Fig. 7 No major changes in mitochondria number, electron transport chain function or the abundance of TCA cycle metabolites in the brain of Pten fb-KO mice.

(a-b) No differences in mitochondria DNA copy number (a, n = 5 per group, F3,16 = 0.21, P = 0.1229) or ETC enzyme activity [b, control (n = 4) Pten fb-KO (n = 3), Pten-Rptor fb-KO (n = 3), Pten-Rictor fb-KO (n = 3), F3,9 = 0.16, P = 0.3782) were observed between the cortexes of control and Pten-deficient mice. (c) The abundance of TCA cycle metabolites did not dramatically change in the brain of Pten fb-KO mice [control (n = 8) vs. Pten fb-KO (n = 7): citrate, t = 0.60, P = 0.5512; alpha-ketoglutarate, t = 1.45, P = 0.1545; malate, t= 1.79, P = 0.0858, fumarate, t = 0.3983, P = 0.69. Control vs. Pten-Rptor fb-DKO (n = 7): citrate, t= 0.95, P = 0.3522; alpha-ketoglutarate, t = 0.62, P = 0.5404; malate, t= 0.41, P = 0.6833; fumarate, t = 1.37, P = 0.1816. control vs. Pten-Rictor fb-DKO n = 7, citrate, t = 0.5512, P = 0.5864; alpha-ketoglutarate, t = 0.2, P = 0.8455; malate, t= 0.43, P = 0.6687, fumarate, t = 0.39, P= 0.6973]. Data are mean ± SEM. Statistics were based on one-way ANOVA followed by uncorrected two-sided Fisher’s LSD method for pairwise comparisons. n.s., not significant.

Extended Data Fig. 8 ASO-A (Rictor-ASO) reduces mTORC2 (but not mTORC1) activity in cortical neurons in culture.

(a-b) Cortical neurons were treated for 72 hours with different ASOs (10 μM) designed to target Rictor mRNA. Representative western blots (a) and quantification (b) show that ASO-A strongly reduces both Rictor protein level (n= 4 per group, t6 = 10.94, P = 0.0001) and mTORC2 activity (t6 = 7.25, P = 0.0010), but had no effect on mTORC1 activity (t6 = 0.33, P = 0.9846). Data are mean ± SEM. Values were compared relative to the vehicle treated group and statistics were based on two-sided t-test comparisons. n.s., not significant.

Source data

Extended Data Fig. 9 Rictor-ASO selectively reduces Rictor mRNA levels in vivo and is safe.

Control mice were injected with Rictor-ASO and monitored for 5 weeks after injection. (a) Rictor-ASO reduced Rictor mRNA levels in spinal cord (n= 4 per group, t6 = 40.76, P < 0.0001), cortex (n= 4 per group, t6 = 16.77, P < 0.0001) and cerebellum (n= 4 per group, t6 = 11.44, P < 0.001). (b) Rictor-ASO-injected mice showed normal weight gain several weeks post-injection (F5,30 = 2,10, P = 0.0875). (c) mRNA expression of gliosis marker (Gfap) and microglia inflammatory markers (Aif1, CD68) were normal in the brain of Rictor-ASO injected mice (Aif1: n= 4 per group, t6 = 0.60, P = 0.544; CD68: n= 4 per group, t6 = 2.12, P = 0.783; Gfap: n= 4 per group, t6 = 0.17, P = 0.8673). Circles represent number of animals per condition. (d) Control mice were injected with Rictor-ASO and the hippocampus was isolated 2 weeks post-injection. Rictor-ASO selectively reduces Rictor mRNA levels, but did not change the expression of Nisch, Plus7, GM6943, GM9265, GM9295 and GM5954 (n = 4 per group, PBS vs. Rictor-ASO: Rictor: t6 = 3.06, P = 0.0223; Nisch: t6 = 1.11, P = 0.3094; Pus7l: t6 = 1.40, P = 0.2106; GM6943: t6 = 0.54, P = 0.6061, GM9265: t6 = 0.48, P = 0.6513; GM9295: t6 = 0.39, P = 0.71; GM5954: t6 = 1.19, P = 0.2763). Data are mean ± SEM. n represents one biological independent mouse. Statistics were based on two-sided t-test comparisons. n.s., not significant.

Extended Data Fig. 10 A control ASO (Control-ASO) failed to reduce Rictor mRNA levels and inhibit mTORC2 activity.

(a) Treatment with Control-ASO did not reduce Rictor mRNA [PBS (n = 6) vs. Control-ASO (n = 4), t8 = 1.88, P= 0.0968]. Representative western blot image (b) and quantification of (c-d) show that Control-ASO did not decrease Rictor protein levels (n = 4 per group, PBS vs. control-ASO t6 = 0.57, P= 0.5864) or mTORC2 activity (n = 4 per group, PBS vs. control-ASO, t6 = 0.71, P= 0.5018). Data are mean ± SEM. Statistics were based on two-sided t-test comparisons. n.s., not significant.

Source Data

Supplementary information

Supplementary Information

Supplementary Table 1, Supplementary Methods and Supplementary References

Reporting Summary

Source data

Source Data Fig. 1

Unprocessed western blots

Source Data Fig. 4

Unprocessed western blots

Source Data Extended Data Fig. 1

Unprocessed western blots

Source Data Extended Data Fig. 5

Unprocessed western blots

Source Data Extended Data Fig. 8

Unprocessed western blots

Source Data Extended Data Fig. 10

Unprocessed western blots

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, CJ., Sgritta, M., Mays, J. et al. Therapeutic inhibition of mTORC2 rescues the behavioral and neurophysiological abnormalities associated with Pten-deficiency. Nat Med 25, 1684–1690 (2019). https://doi.org/10.1038/s41591-019-0608-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-019-0608-y

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