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:

An α2-Na/K ATPase/α-adducin complex in astrocytes triggers non–cell autonomous neurodegeneration

Abstract

Perturbations of astrocytes trigger neurodegeneration in several diseases, but the glial cell–intrinsic mechanisms that induce neurodegeneration remain poorly understood. We found that a protein complex of α2-Na/K ATPase and α-adducin was enriched in astrocytes expressing mutant superoxide dismutase 1 (SOD1), which causes familial amyotrophic lateral sclerosis (ALS). Knockdown of α2-Na/K ATPase or α-adducin in mutant SOD1 astrocytes protected motor neurons from degeneration, including in mutant SOD1 mice in vivo. Heterozygous disruption of the α2-Na/K ATPase gene suppressed degeneration in vivo and increased the lifespan of mutant SOD1 mice. The pharmacological agent digoxin, which inhibits Na/K ATPase activity, protected motor neurons from mutant SOD1 astrocyte–induced degeneration. Notably, α2-Na/K ATPase and α-adducin were upregulated in spinal cord of sporadic and familial ALS patients. Collectively, our findings define chronic activation of the α2-Na/K ATPase/α-adducin complex as a critical glial cell–intrinsic mechanism of non–cell autonomous neurodegeneration, with implications for potential therapies for neurodegenerative diseases.

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

Figure 1: Upregulation of α-adducin in SOD1G93A astrocytes mediates non–cell autonomous degeneration of motor neurons.
Figure 2: Knockdown of α-adducin in SOD1G93A mice suppresses motor neuron degeneration in vivo.
Figure 3: Enrichment of the α2-Na/K ATPase/α-adducin complex in SOD1G93A astrocytes triggers motor neuron degeneration.
Figure 4: Heterozygous disruption of Atp1a2 in SOD1G93A mice suppresses motor neuron degeneration and enhances mouse lifespan.
Figure 5: Na/K ATPase stimulates mitochondrial respiration and expression of inflammatory genes in SOD1G93A astrocytes.
Figure 6: The α2-Na/K ATPase/α-adducin complex is upregulated in spinal cord in familial and sporadic ALS patients.

Similar content being viewed by others

References

  1. Allen, N.J. & Barres, B.A. Signaling between glia and neurons: focus on synaptic plasticity. Curr. Opin. Neurobiol. 15, 542–548 (2005).

    Article  CAS  PubMed  Google Scholar 

  2. Fields, R.D. & Stevens-Graham, B. New insights into neuron-glia communication. Science 298, 556–562 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Ilieva, H., Polymenidou, M. & Cleveland, D.W. Non–cell autonomous toxicity in neurodegenerative disorders: ALS and beyond. J. Cell Biol. 187, 761–772 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. McGann, J.C., Lioy, D.T. & Mandel, G. Astrocytes conspire with neurons during progression of neurological disease. Curr. Opin. Neurobiol. 22, 850–858 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Garden, G.A. et al. Polyglutamine-expanded ataxin-7 promotes non–cell-autonomous purkinje cell degeneration and displays proteolytic cleavage in ataxic transgenic mice. J. Neurosci. 22, 4897–4905 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Yoo, S.Y. et al. SCA7 knockin mice model human SCA7 and reveal gradual accumulation of mutant ataxin-7 in neurons and abnormalities in short-term plasticity. Neuron 37, 383–401 (2003).

    Article  CAS  PubMed  Google Scholar 

  7. Evert, B.O. et al. Inflammatory genes are upregulated in expanded ataxin-3–expressing cell lines and spinocerebellar ataxia type 3 brains. J. Neurosci. 21, 5389–5396 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Clement, A.M. et al. Wild-type nonneuronal cells extend survival of SOD1 mutant motor neurons in ALS mice. Science 302, 113–117 (2003).

    Article  CAS  PubMed  Google Scholar 

  9. Yamanaka, K. et al. Astrocytes as determinants of disease progression in inherited amyotrophic lateral sclerosis. Nat. Neurosci. 11, 251–253 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Faideau, M. et al. In vivo expression of polyglutamine-expanded huntingtin by mouse striatal astrocytes impairs glutamate transport: a correlation with Huntington's disease subjects. Hum. Mol. Genet. 19, 3053–3067 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Shin, J.Y. et al. Expression of mutant huntingtin in glial cells contributes to neuronal excitotoxicity. J. Cell Biol. 171, 1001–1012 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Rothstein, J.D., Van Kammen, M., Levey, A.I., Martin, L.J. & Kuncl, R.W. Selective loss of glial glutamate transporter GLT-1 in amyotrophic lateral sclerosis. Ann. Neurol. 38, 73–84 (1995).

    Article  CAS  PubMed  Google Scholar 

  13. Boillée, S. et al. Onset and progression in inherited ALS determined by motor neurons and microglia. Science 312, 1389–1392 (2006).

    Article  CAS  PubMed  Google Scholar 

  14. Nagai, M. et al. Astrocytes expressing ALS-linked mutated SOD1 release factors selectively toxic to motor neurons. Nat. Neurosci. 10, 615–622 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Di Giorgio, F.P., Carrasco, M.A., Siao, M.C., Maniatis, T. & Eggan, K. Non–cell autonomous effect of glia on motor neurons in an embryonic stem cell–based ALS model. Nat. Neurosci. 10, 608–614 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Dion, P.A., Daoud, H. & Rouleau, G.A. Genetics of motor neuron disorders: new insights into pathogenic mechanisms. Nat. Rev. Genet. 10, 769–782 (2009).

    Article  CAS  PubMed  Google Scholar 

  17. Rosen, D.R. et al. Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature 362, 59–62 (1993).

    Article  CAS  PubMed  Google Scholar 

  18. Gurney, M.E. et al. Motor neuron degeneration in mice that express a human Cu,Zn superoxide dismutase mutation. Science 264, 1772–1775 (1994).

    Article  CAS  PubMed  Google Scholar 

  19. Bruijn, L.I., Miller, T.M. & Cleveland, D.W. Unraveling the mechanisms involved in motor neuron degeneration in ALS. Annu. Rev. Neurosci. 27, 723–749 (2004).

    Article  CAS  PubMed  Google Scholar 

  20. Haidet-Phillips, A.M. et al. Astrocytes from familial and sporadic ALS patients are toxic to motor neurons. Nat. Biotechnol. 29, 824–828 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Goodman, L.S., Brunton, L.L., Chabner, B. & Knollmann, B.C. Goodman & Gilman's Pharmacological Basis of Therapeutics (McGraw-Hill, New York, 2011).

  22. Lehtinen, M.K. et al. A conserved MST-FOXO signaling pathway mediates oxidative-stress responses and extends life span. Cell 125, 987–1001 (2006).

    Article  CAS  PubMed  Google Scholar 

  23. Matsuoka, Y., Li, X. & Bennett, V. Adducin: structure, function and regulation. Cell. Mol. Life Sci. 57, 884–895 (2000).

    Article  CAS  PubMed  Google Scholar 

  24. Robledo, R.F. et al. Targeted deletion of alpha-adducin results in absent beta- and gamma-adducin, compensated hemolytic anemia, and lethal hydrocephalus in mice. Blood 112, 4298–4307 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Shan, X., Hu, J.H., Cayabyab, F.S. & Krieger, C. Increased phospho-adducin immunoreactivity in a murine model of amyotrophic lateral sclerosis. Neuroscience 134, 833–846 (2005).

    Article  CAS  PubMed  Google Scholar 

  26. Hu, J.H., Zhang, H., Wagey, R., Krieger, C. & Pelech, S.L. Protein kinase and protein phosphatase expression in amyotrophic lateral sclerosis spinal cord. J. Neurochem. 85, 432–442 (2003).

    Article  CAS  PubMed  Google Scholar 

  27. Kaplan, J.H. Biochemistry of Na,K-ATPase. Annu. Rev. Biochem. 71, 511–535 (2002).

    Article  CAS  PubMed  Google Scholar 

  28. Watts, A.G., Sanchez-Watts, G., Emanuel, J.R. & Levenson, R. Cell-specific expression of mRNAs encoding Na+, K(+)-ATPase alpha- and beta-subunit isoforms within the rat central nervous system. Proc. Natl. Acad. Sci. USA 88, 7425–7429 (1991).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Moseley, A.E. et al. Deficiency in Na,K-ATPase alpha isoform genes alters spatial learning, motor activity, and anxiety in mice. J. Neurosci. 27, 616–626 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Huang, G. et al. Death receptor 6 (DR6) antagonist antibody is neuroprotective in the mouse SOD1G93A model of amyotrophic lateral sclerosis. Cell Death Dis. 4, e841 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Hartford, A.K., Messer, M.L., Moseley, A.E., Lingrel, J.B. & Delamere, N.A. Na,K-ATPase alpha 2 inhibition alters calcium responses in optic nerve astrocytes. Glia 45, 229–237 (2004).

    Article  PubMed  Google Scholar 

  32. Nakahira, K. et al. Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome. Nat. Immunol. 12, 222–230 (2011).

    Article  CAS  PubMed  Google Scholar 

  33. Bulua, A.C. et al. Mitochondrial reactive oxygen species promote production of pro-inflammatory cytokines and are elevated in TNFR1-associated periodic syndrome (TRAPS). J. Exp. Med. 208, 519–533 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Zhou, R., Yazdi, A.S., Menu, P. & Tschopp, J. A role for mitochondria in NLRP3 inflammasome activation. Nature 469, 221–225 (2011).

    Article  CAS  PubMed  Google Scholar 

  35. Wu, D.C., Re, D.B., Nagai, M., Ischiropoulos, H. & Przedborski, S. The inflammatory NADPH oxidase enzyme modulates motor neuron degeneration in amyotrophic lateral sclerosis mice. Proc. Natl. Acad. Sci. USA 103, 12132–12137 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Phatnani, H.P. et al. Intricate interplay between astrocytes and motor neurons in ALS. Proc. Natl. Acad. Sci. USA 110, E756–E765 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Brooks, B.R., Miller, R.G., Swash, M. & Munsat, T.L. El Escorial revisited: revised criteria for the diagnosis of amyotrophic lateral sclerosis. Amyotroph. Lateral Scler. Other Motor Neuron Disord. 1, 293–299 (2000).

    Article  CAS  PubMed  Google Scholar 

  38. Ellis, D.Z., Rabe, J. & Sweadner, K.J. Global loss of Na,K-ATPase and its nitric oxide–mediated regulation in a transgenic mouse model of amyotrophic lateral sclerosis. J. Neurosci. 23, 43–51 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Martin, L.J. et al. Motor neuron degeneration in amyotrophic lateral sclerosis mutant superoxide dismutase-1 transgenic mice: mechanisms of mitochondriopathy and cell death. J. Comp. Neurol. 500, 20–46 (2007).

    Article  CAS  PubMed  Google Scholar 

  40. Kaphzan, H., Buffington, S.A., Jung, J.I., Rasband, M.N. & Klann, E. Alterations in intrinsic membrane properties and the axon initial segment in a mouse model of Angelman syndrome. J. Neurosci. 31, 17637–17648 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Kaphzan, H. et al. Genetic reduction of the alpha1 subunit of Na/K-ATPase corrects multiple hippocampal phenotypes in Angelman syndrome. Cell Reports 4, 405–412 (2013).

    Article  CAS  PubMed  Google Scholar 

  42. Efendiev, R. et al. Hypertension-linked mutation in the adducin alpha-subunit leads to higher AP2-mu2 phosphorylation and impaired Na+,K+-ATPase trafficking in response to GPCR signals and intracellular sodium. Circ. Res. 95, 1100–1108 (2004).

    Article  CAS  PubMed  Google Scholar 

  43. Torielli, L. et al. alpha-adducin mutations increase Na/K pump activity in renal cells by affecting constitutive endocytosis: implications for tubular Na reabsorption. Am. J. Physiol. Renal Physiol. 295, F478–F487 (2008).

    Article  CAS  PubMed  Google Scholar 

  44. Cusi, D. et al. Polymorphisms of alpha-adducin and salt sensitivity in patients with essential hypertension. Lancet 349, 1353–1357 (1997).

    Article  CAS  PubMed  Google Scholar 

  45. Kennedy, D.J. et al. CD36 and Na/K-ATPase-alpha1 form a pro-inflammatory signaling loop in kidney. Hypertension 61, 216–224 (2013).

    Article  CAS  PubMed  Google Scholar 

  46. Liu, J., Kennedy, D.J., Yan, Y. & Shapiro, J.I. Reactive oxygen species modulation of Na/K-ATPase regulates fibrosis and renal proximal tubular sodium handling. Int. J. Nephrol. 2012, 381320 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Wang, J.K. et al. Cardiac glycosides provide neuroprotection against ischemic stroke: discovery by a brain slice-based compound screening platform. Proc. Natl. Acad. Sci. USA 103, 10461–10466 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Piccioni, F., Roman, B.R., Fischbeck, K.H. & Taylor, J.P. A screen for drugs that protect against the cytotoxicity of polyglutamine-expanded androgen receptor. Hum. Mol. Genet. 13, 437–446 (2004).

    Article  CAS  PubMed  Google Scholar 

  49. Corcoran, L.J., Mitchison, T.J. & Liu, Q. A novel action of histone deacetylase inhibitors in a protein aggresome disease model. Curr. Biol. 14, 488–492 (2004).

    Article  CAS  PubMed  Google Scholar 

  50. Burkhardt, M.F. et al. A cellular model for sporadic ALS using patient-derived induced pluripotent stem cells. Mol. Cell. Neurosci. 56, 355–364 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Chandra, S., Gallardo, G., Fernandez-Chacon, R., Schluter, O.M. & Sudhof, T.C. Alpha-synuclein cooperates with CSPalpha in preventing neurodegeneration. Cell 123, 383–396 (2005).

    Article  CAS  PubMed  Google Scholar 

  52. Gaudilliere, B., Shi, Y. & Bonni, A. RNA interference reveals a requirement for myocyte enhancer factor 2A in activity-dependent neuronal survival. J. Biol. Chem. 277, 46442–46446 (2002).

    Article  CAS  PubMed  Google Scholar 

  53. Gingras, M., Gagnon, V., Minotti, S., Durham, H.D. & Berthod, F. Optimized protocols for isolation of primary motor neurons, astrocytes and microglia from embryonic mouse spinal cord. J. Neurosci. Methods 163, 111–118 (2007).

    Article  CAS  PubMed  Google Scholar 

  54. Raoul, C. et al. Lentiviral-mediated silencing of SOD1 through RNA interference retards disease onset and progression in a mouse model of ALS. Nat. Med. 11, 423–428 (2005).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We thank members of the Bonni laboratory for helpful discussions. We thank L. Zinman (University of Toronto) for providing human patient tissue samples. This work was supported by a grant from the Edward R. and Anne G. Lefler Foundation (A.B.) and The Ruth L. Kirschstein National Research Service Awards T32 5T32AG00222 (G.G.). Human spinal cord material provided from Northwestern University autopsy program is partially funded from the Les Turner ALS Foundation. Additional human tissue samples were obtained from the Human Brain and Spinal Fluid Resource Center, which is sponsored by the National Institute of Neurological Disorders and Stroke and the US National Institutes of Health, National Multiple Sclerosis Society, and Department of Veterans Affairs.

Author information

Authors and Affiliations

Authors

Contributions

A.B. directed and coordinated the project. G.G. designed and performed or participated in all experiments. J.B. performed mouse husbandry and survival studies. J.B.L. provided α2-Na/K ATPase knockout mice. H.S. performed mass spectrometry analysis. J. Ravits, T.S. and J. Robertson provided human tissue samples. The manuscript was written by G.G. and A.B. and commented on by all authors.

Corresponding author

Correspondence to Azad Bonni.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 α-Adducin in spinal cord is upregulated in symptomatic SOD1G93A spinal cord within astrocytes.

(a) Immunoblots for α-Adducin protein at 60 and 90 day old SOD1G93A mice show α-Adducin is upregulated at 90 days. (b) Immunohistochemistry with in sections of the lumbar spinal cord from symptomatic SOD1G93A mice displays Ser436-phosphorylated α-Adducin does not co-localize with the motor neuron marker (SMi32). Arrowheads indicate motor neurons; scale bar 50μm. (c) Immunohistochemistry from sections of control wild type lumbar spinal cord displays Ser436-phosphorylated α-Adducin does not co-localize with the motor neuron marker (SMi32); upper panels. Arrowheads indicate motor neurons. Immunohistochemistry from sections of control wild type lumbar spinal cord displays Ser436-phosphorylated α-Adducin co-localize with the astrocyte marker (GFAP) lower panels. Scale bar 50μm. (a) are cropped; full length images are presented in Supplementary Figure 11.

Supplementary Figure 2 Expression of a RNAi-resistant form of α-Adducin in SOD1G93A astrocytes restores the ability of SOD1G93A astrocytes to induce non-cell autonomous motor neuron cell death.

(a) Knockdown of α-Adducin relative to control U6 in astrocytes (b) Co-cultured astrocytes and motor neurons were subjected to immunocytochemistry with the motor neuron nuclear protein Islet1 (red) and the dendrite protein MAP2 (green); scale bar 50μm. Wild type astrocytes transfected with the control U6 or α-Adducin RNAi plasmid had little or no effect on motor primary motor neurons cell death or dendrite abnormalities (upper left panel); quantified (c and d). Control U6 SOD1G93A astrocytes induced non-cell autonomous motor neuron cell death and dendrite abnormalities (upper right panel); quantified (c and d). Knockdown of α-Adducin in SOD1G93A astrocytes protected motor neurons against the non-cell autonomous cell death and dendrite abnormalities (lower left panel); quantified (c and d). Expressions of an RNAi-resistant form of α-Adducin (Add-Res) in the background of α-Adducin RNAi in SOD1G93A astrocytes restored the ability of the SOD1G93A astrocytes to induce non-cell autonomous cell death and dendrite abnormalities in motor neurons (lower right panel); quantified (d and e). All data in bar charts show mean ± s.e.m (***p<0.001; unpaired t-test). (a) are cropped; full length images are presented in Supplementary Figure 11.

Supplementary Figure 3 Lentivirial mediated knockdown in vivo predominately target astrocytes.

(a) Immunohistochemistry with GFP in sections of the lumbar spinal cord from SOD1G93A mice displaying percent GFP positive astrocytes (GFAP), microglia (Iba1) and motor neurons (SMi32) 30 days post-injection. Arrowheads indicate motor neurons; scale bar 50μm. (b) Quantifications of percent GFP positive cells revealed lentivirus predominately target astrocytes; n=~300 per cell type/three. All data in bar charts show ± s.e.m (***p<0.001; ANOVA). (c) Immunohistochemistry with GFP in sections of the lumbar spinal cord from wild type mice displaying percent GFP positive astrocytes (GFAP), microglia (Iba1) and motor neurons (SMi32) 30 days post-injection. Arrowheads indicate motor neurons; scale bar 50μm. (d) Quantifications of percent GFP positive cells revealed lentivirus predominately target astrocytes; n=~300 per cell type/three. All data in bar charts show ± s.e.m (***p<0.001; unpaired t-test).

Supplementary Figure 4 Knockdown of α-Adducin in SOD1G93A mice decreases immunoreactivity for phosphorylated Ser436-α-Adducin.

Spinal cord from SOD1G93A mice injected intraspinally with lentivirus expressing short hairpin RNAs targeting α-Adducin and encoding GFP (LV-Addi) or the corresponding control U6 (LV-U6) were subjected to immunohistochemistry using GFP and phospho-α-Adducin (red) antibodies. Knockdown of α-Adducin (LV-Addi) led to a decreased in immunoreactivity of phospho-α-Adducin within the GFP-labeled ventral horn as compared to control U6 (LV-U6) injected ventral horn; scale bar 50μm.

Supplementary Figure 5 Knockdown of α-Adducin or α2-Na/K ATPase in SOD1G93A mice do not alter gliosis in the spinal cord.

Spinal cord from SOD1G93A mice injected intraspinally with lentivirus expressing short hairpin RNAs targeting α-Adducin or α2-Na/K ATPase or the corresponding control U6 virus were subjected to immunohistochemistry using GFP and the GFAP (red) antibodies. Knockdown of α-Adducin (LV-Addi) or α2-Na/K ATPase (LV-ATPi) had little or no effect on the presence or abundance of astrocytes within the GFP-labeled ventral horn; scale bar 50μm.

Supplementary Figure 6 Knockdown of α-Adducin or α2-Na/K ATPase in SOD1G93A mice do not alter microgliosis in spinal cord.

Spinal cord from SOD1G93A mice injected intraspinally with lentivirus expressing short hairpin RNAs targeting α-Adducin or α2-Na/K ATPase or the corresponding control U6 virus were subjected to immunohistochemistry using GFP and the Iba1 (red) antibodies. Knockdown of α-Adducin (LV-Addi) or α2-Na/K ATPase (LV-ATPi) had little or no effect on the presence or abundance of microglia within the GFP-labeled ventral horn; scale bar 50μm.

Supplementary Figure 7 α2-Na/K ATPase co-immunoprecipitates with α-Adducin in spinal cord lysates and is specifically upregulated in astrocytes in symptomatic SOD1G93A mice.

(a) Immunoblots show immunoprecipitated α-Adducin from SOD1G93A and control wild type spinal cord lysates subjected to immunoblotting with the α-Adducin and α2-Na/K ATPase antibodies following glycine elution, confirming α2-Na/K ATPase as an interactor of α-Adducin (left panels). Immunoblots show α2-Na/K ATPase is predominately expressed in primary glial cultures relative to primary motor neuron cultures enriched with the neuron marker β-tubulin. 14-3-3β is used as an internal control (right panel). (b) Immunohistochemistry with astrocyte marker GFAP and α2-Na/K ATPase antibody in sections of the lumbar spinal cord from SOD1G93A mice at 60 days displays α2-Na/K ATPase expression within astrocytes; scale bar 50μm. (c) Immunohistochemistry with GFAP and α2-Na/K ATPase antibody in sections of the lumbar spinal cord from symptomatic SOD1G93A mice at 120 days displays upregulation of α2-Na/K ATPase expression within astrocytes; scale bar 50μm. (a) are cropped; full length images are presented in Supplementary Figure 11.

Supplementary Figure 8 Intraspinally injection of control lentivirus in SOD1G93A mice had no effect on motor neuron survival.

(a) Spinal cord from end stage SOD1G93A mice injected at age 90 days intraspinally with the control lentivirus encoding GFP (LV-U6 SOD1G93A) was subjected to immunohistochemistry at end stage. End stage was defined as a time point at which the animal was unable to upright itself within 30s of placement on its side. Immunohistochemistry with GFP in SOD1G93A lumbar sections revealed delivery of control injected virus (LV-U6) into the ventral horn; scale bar 100μm. Alternating GFP positive sections were subjected to immunohistochemistry using the GFP antibody and the neurofilment-SMi32 antibody (red), a motor neuron marker, or Nissl stained (lower panels) for quantification of surviving motor neurons within GFP-labeled injected ventral horn and contralateral non-injected ventral horn (n≥20 sections per animal); scale bar 50μm. Control LV-U6 SOD1G93A mice (n=3) displayed equivalent degeneration of motor neurons within injected GFP-labeled ventral horn and non-injected contralateral ventral horn. Arrowheads indicate surviving motor neurons; quantification shown in (b).

Supplementary Figure 9 Heterozygous disruption of the α2-Na/K ATPase gene in SOD1G93A mice delays motor neuron degeneration.

(a) Nissl stained sections from endstage control SOD1G93A mice (n=5) and aged-matched SOD1G93A littermates heterozygous-null for the α2-Na/K ATPase allele (n=5) displayed more than twice the number of motor neurons in ATPase+/-;SOD1G93A than control SOD1G93A mice. Arrow heads indicate surviving motor neurons; quantification shown in (b); scale bar 50μm (***p<0.001; unpaired t-test).

Supplementary Figure 10 Condition media from heterozygous-null from α2-Na/K ATPase SOD1G93A astrocytes is neuroprotective.

(a) Precondition media from wild type, SOD1G93A, and heterozygous-null α2-Na/K ATPase; SOD1G93A astrocytes were exposed to motor neurons and subjected to immunocytochemistry with antibodies recognizing the motor neuron nuclear protein Islet1 (red) and the dendrite protein MAP2 (green); scale bar 50μm. Precondition media from wild type astrocytes had little or no effect on motor neuron survival (upper panels); quantified (b). Preconditioned medium from SOD1G93A astrocytes induced non-cell autonomous motor neuron cell death (middle); quantified (b). Preconditioned medium from heterozygous-null α2-Na/K ATPase; SOD1G93A astrocytes protected motor neurons against the non-cell autonomous cell death (lower panel); All data in bar charts show mean ± s.e.m (***p<0.001; unpaired t-test).

Supplementary Figure 11 Full scans of key Western blot data. In many experiments, membranes were stripped and reblotted with a second antibody.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gallardo, G., Barowski, J., Ravits, J. et al. An α2-Na/K ATPase/α-adducin complex in astrocytes triggers non–cell autonomous neurodegeneration. Nat Neurosci 17, 1710–1719 (2014). https://doi.org/10.1038/nn.3853

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.3853

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