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:

Altered perivascular fibroblast activity precedes ALS disease onset

A Publisher Correction to this article was published on 02 June 2021

This article has been updated

Abstract

Apart from well-defined factors in neuronal cells1, only a few reports consider that the variability of sporadic amyotrophic lateral sclerosis (ALS) progression can depend on less-defined contributions from glia2,3 and blood vessels4. In this study we use an expression-weighted cell-type enrichment method to infer cell activity in spinal cord samples from patients with sporadic ALS and mouse models of this disease. Here we report that patients with sporadic ALS present cell activity patterns consistent with two mouse models in which enrichments of vascular cell genes preceded microglial response. Notably, during the presymptomatic stage, perivascular fibroblast cells showed the strongest gene enrichments, and their marker proteins SPP1 and COL6A1 accumulated in enlarged perivascular spaces in patients with sporadic ALS. Moreover, in plasma of 574 patients with ALS from four independent cohorts, increased levels of SPP1 at disease diagnosis repeatedly predicted shorter survival with stronger effect than the established risk factors of bulbar onset or neurofilament levels in cerebrospinal fluid. We propose that the activity of the recently discovered perivascular fibroblast can predict survival of patients with ALS and provide a new conceptual framework to re-evaluate definitions of ALS etiology.

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: Patients with ALS show increased transcriptional activity of perivascular fibroblast cell gene markers that occur at presymptomatic disease stage in SOD1G93A and TARDBPQ331K mice.
Fig. 2: Perivascular fibroblast marker proteins COL6A1 and SPP1 accumulate in enlarged perivascular spaces during ALS progression.
Fig. 3: Prognostic value of SPP1 protein in plasma of patients with ALS.

Similar content being viewed by others

Data availability

Transcriptome datasets and analysis scripts from human patients with ALS are available at GitHub (https://github.com/NathanSkene/ALS_Human_EWCE), SOD1G93A mouse transcriptome datasets and analysis scripts are available at https://github.com/NathanSkene/ALS_Mouse_EWCE, and TARDBPQ331K/ Q331K mouse transcriptome datasets and analysis scripts are available at https://github.com/szczepinskaa/ALS_TDP-43.git. Images and analysis scripts for human histology are deposited and available under the following links: SPP1 in Red (https://doi.org/10.6084/m9.figshare.11632794), COL6A1 in Red (https://doi.org/10.6084/m9.figshare.11635887), SPP1 in DAB (https://doi.org/10.6084/m9.figshare.11628891), and COL6A1 in DAB (https://doi.org/10.6084/m9.figshare.11628585). Additional data tables are available at https://doi.org/10.6084/m9.figshare.14035616. Patient plasma data and analysis scripts for cutoff optimization and Cox proportional hazards model are available at GitHub (https://github.com/lewandowskilab/PVF_Manuscript).

Code availability

Transcriptome analysis scripts from human patients with ALS are available at GitHub (https://github.com/NathanSkene/ALS_Human_EWCE), SOD1G93A mouse transcriptome analysis scripts are available at https://github.com/NathanSkene/ALS_Mouse_EWCE, and TARDBPQ331K/ Q331K mouse transcriptome analysis scripts are available at https://github.com/szczepinskaa/ALS_TDP-43.git. Images and analysis scripts for human histology are deposited and available under the following links: SPP1 in Red training model (https://doi.org/10.6084/m9.figshare.11632794), COL6A1 in Red training model (https://doi.org/10.6084/m9.figshare.11635887), SPP1 in DAB training model (https://doi.org/10.6084/m9.figshare.11628891), and COL6A1 in DAB training model and macro (https://doi.org/10.6084/m9.figshare.11628585). Patient plasma analysis scripts for cutoff optimization and Cox proportional hazards model are available at GitHub (https://github.com/lewandowskilab/PVF_Manuscript).

Change history

References

  1. Cook, C. & Petrucelli, L. Genetic convergence brings clarity to the enigmatic red line in ALS. Neuron 101, 1057–1069 (2019).

    Article  CAS  PubMed  Google Scholar 

  2. Spiller, K. J. et al. Microglia-mediated recovery from ALS-relevant motor neuron degeneration in a mouse model of TDP-43 proteinopathy. Nat. Neurosci. 21, 329–340 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    PubMed  Google Scholar 

  4. Lewandowski, S. A. et al. Presymptomatic activation of the PDGF-CC pathway accelerates onset of ALS neurodegeneration. Acta Neuropathol. 131, 453–464 (2016).

    Article  CAS  PubMed  Google Scholar 

  5. Brown, R. H. & Al-Chalabi, A. Amyotrophic lateral sclerosis. N. Engl. J. Med. 377, 162–172 (2017).

    Article  CAS  PubMed  Google Scholar 

  6. Al-Chalabi, A. et al. Analysis of amyotrophic lateral sclerosis as a multistep process: a population-based modelling study. Lancet Neurol. 13, 1108–1113 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  7. 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 

  8. Sreedharan, J. et al. TDP-43 mutations in familial and sporadic amyotrophic lateral sclerosis. Science 319, 1668–1672 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Wu, C.-H. et al. Mutations in the profilin 1 gene cause familial amyotrophic lateral sclerosis. Nature 488, 499–503 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Kang, S. H. et al. Degeneration and impaired regeneration of gray matter oligodendrocytes in amyotrophic lateral sclerosis. Nat. Neurosci. 16, 571–579 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Zhong, Z. et al. ALS-causing SOD1 mutants generate vascular changes prior to motor neuron degeneration. Nat. Neurosci. 11, 420–422 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Zeisel, A. et al. Cell types in the mouse cortex and hippocampus revealed by single-cell RNA-seq. Science 347, 1138–1142 (2015).

    Article  CAS  PubMed  Google Scholar 

  13. Marques, S. et al. Oligodendrocyte heterogeneity in the mouse juvenile and adult central nervous system. Science 352, 1326–1329 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Zeisel, A. et al. Molecular architecture of the mouse nervous system. Cell 174, 999–1014 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Skene, N. G. & Grant, S. G. N. Identification of vulnerable cell types in major brain disorders using single cell transcriptomes and expression weighted cell type enrichment. Front. Neurosci. 10, 16 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Clark, J. A., Southam, K. A., Blizzard, C. A., King, A. E. & Dickson, T. C. Axonal degeneration, distal collateral branching and neuromuscular junction architecture alterations occur prior to symptom onset in the SOD1G93A mouse model of amyotrophic lateral sclerosis. J. Chem. Neuroanat. 76, 35–47 (2016).

    Article  CAS  PubMed  Google Scholar 

  17. Hall, E. D., Oostveen, J. A. & Gurney, M. E. Relationship of microglial and astrocytic activation to disease onset and progression in a transgenic model of familial ALS. Glia 23, 249–256 (1998).

    Article  CAS  PubMed  Google Scholar 

  18. Weydt, P., Hong, S. Y., Kliot, M. & Möller, T. Assessing disease onset and progression in the SOD1 mouse model of ALS. Neuroreport 14, 1051–1054 (2003).

    PubMed  Google Scholar 

  19. White, M. A. et al. TDP-43 gains function due to perturbed autoregulation in a Tardbp knock-in mouse model of ALS-FTD. Nat. Neurosci. 21, 552–563 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Chiu, I. M. et al. A neurodegeneration-specific gene-expression signature of acutely isolated microglia from an amyotrophic lateral sclerosis mouse model. Cell Rep. 4, 385–401 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Sun, S. et al. Translational profiling identifies a cascade of damage initiated in motor neurons and spreading to glia in mutant SOD1-mediated ALS. Proc. Natl Acad. Sci. USA 112, E6993–E7002 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Vanlandewijck, M. et al. A molecular atlas of cell types and zonation in the brain vasculature. Nature 554, 475–480 (2018).

    Article  CAS  PubMed  Google Scholar 

  23. Ma, Q., Ineichen, B. V., Detmar, M. & Proulx, S. T. Outflow of cerebrospinal fluid is predominantly through lymphatic vessels and is reduced in aged mice. Nat. Commun. https://doi.org/10.1038/s41467-017-01484-6 (2017).

  24. Kapoor, K. G., Katz, S. E., Grzybowski, D. M. & Lubow, M. Cerebrospinal fluid outflow: an evolving perspective. Brain Res. Bull. 77, 327–334 (2008).

    Article  PubMed  Google Scholar 

  25. Tam, O. H. et al. Postmortem cortex samples identify distinct molecular subtypes of ALS: retrotransposon activation, oxidative stress, and activated glia. Cell Rep. 29, 1164–1177 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Westeneng, H.-J. et al. Prognosis for patients with amyotrophic lateral sclerosis: development and validation of a personalised prediction model. Lancet Neurol. 17, 423–433 (2018).

    Article  PubMed  Google Scholar 

  27. Braak, H. et al. Amyotrophic lateral sclerosis—a model of corticofugal axonal spread. Nat. Rev. Neurol. 9, 708–714 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Chiu, A. Y. et al. Age-dependent penetrance of disease in a transgenic mouse model of familial amyotrophic lateral sclerosis. Mol. Cell. Neurosci. 6, 349–362 (1995).

    Article  CAS  PubMed  Google Scholar 

  29. Gille, B. et al. Inflammatory markers in cerebrospinal fluid: independent prognostic biomarkers in amyotrophic lateral sclerosis? J. Neurol. Neurosurg. Psychiatry https://doi.org/10.1136/jnnp-2018-319586 (2019).

  30. Turner, M. R., Goldacre, R., Talbot, K. & Goldacre, M. J. Cerebrovascular injury as a risk factor for amyotrophic lateral sclerosis. J. Neurol. Neurosurg. Psychiatry 87, 244–246 (2016).

    Article  PubMed  Google Scholar 

  31. Garton, F. C., Trabjerg, B. B., Wray, N. R. & Agerbo, E. Cardiovascular disease, psychiatric diagnosis and sex differences in the multistep hypothesis of amyotrophic lateral sclerosis. Eur. J. Neurol. https://doi.org/10.1111/ene.14554 (2020).

  32. Rule, R. R., Schuff, N., Miller, R. G. & Weiner, M. W. Gray matter perfusion correlates with disease severity in ALS. Neurology 74, 821–827 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Murphy, M. J. et al. Widespread cerebral haemodynamics disturbances occur early in amyotrophic lateral sclerosis. Amyotroph. Lateral Scler. 13, 202–209 (2012).

    Article  PubMed  Google Scholar 

  34. Van Laere, K. et al. Value of 18fluorodeoxyglucose-positron-emission tomography in amyotrophic lateral sclerosis: a prospective study. JAMA Neurol. 71, 553–561 (2014).

    Article  PubMed  Google Scholar 

  35. Wolak, T. Osteopontin—a multi-modal marker and mediator in atherosclerotic vascular disease. Atherosclerosis 236, 327–337 (2014).

    Article  CAS  PubMed  Google Scholar 

  36. Chiocchetti, A. et al. Osteopontin bridging innate and adaptive immunity in autoimmune diseases. J. Immunol. Res. 2016, 7675437 (2016).

    PubMed  PubMed Central  Google Scholar 

  37. Zhao, H. et al. The role of osteopontin in the progression of solid organ tumour. Cell Death Dis. 9, 356 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Ding, J. et al. Large perivascular spaces visible on magnetic resonance imaging, cerebral small vessel disease progression, and risk of dementia. JAMA Neurol. 74, 1105 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Smeijer, D., Ikram, M. K. & Hilal, S. Enlarged perivascular spaces and dementia: a systematic review. J. Alzheimers Dis. 72, 247–256 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Brown, R. et al. Understanding the role of the perivascular space in cerebral small vessel disease. Cardiovasc. Res. 114, 1462–1473 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Poesen, K. et al. Neurofilament markers for ALS correlate with extent of upper and lower motor neuron disease. Neurology 88, 2302–2309 (2017).

    Article  CAS  PubMed  Google Scholar 

  42. Lerman, B. J. et al. Deletion of galectin-3 exacerbates microglial activation and accelerates disease progression and demise in a SOD1G93A mouse model of amyotrophic lateral sclerosis. Brain Behav. 2, 563–575 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Gautier, L., Cope, L., Bolstad, B. M. & Irizarry, R. A. affy—analysis of Affymetrix GeneChip data at the probe level. Bioinformatics 20, 307–315 (2004).

    Article  CAS  PubMed  Google Scholar 

  44. Durinck, S., Spellman, P. T., Birney, E. & Huber, W. Mapping identifiers for the integration of genomic datasets with the R/Bioconductor package biomaRt. Nat. Protoc. 4, 1184–1191 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Rabin, S. J. et al. Sporadic ALS has compartment-specific aberrant exon splicing and altered cell-matrix adhesion biology. Hum. Mol. Genet. 19, 313–328 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Leitner, M., Menzies, S. & Lutz, C. Working with ALS Mice: Guidelines for Preclinical Testing and Colony Management. (Prize4Life & The Jackson Laboratory, 2009).

  48. Ludolph, A. et al. A revision of the El Escorial criteria—2015. Amyotroph. Lateral Scler. Front. Degener. 16, 291–292 (2015).

    Article  Google Scholar 

  49. van Vliet, E. A. et al. Expression and cellular distribution of P-glycoprotein and breast cancer resistance protein in amyotrophic lateral sclerosis patients. J. Neuropathol. Exp. Neurol. 79, 266–276 (2020).

    Article  PubMed  CAS  Google Scholar 

  50. Casula, M. et al. Toll-like receptor signaling in amyotrophic lateral sclerosis spinal cord tissue. Neuroscience 179, 233–243 (2011).

    Article  CAS  PubMed  Google Scholar 

  51. Huisman, M. H. B. et al. Population based epidemiology of amyotrophic lateral sclerosis using capture–recapture methodology. J. Neurol. Neurosurg. Psychiatry 82, 1165–1170 (2011).

    Article  PubMed  Google Scholar 

  52. 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 Mot. Neuron Disord. 1, 293–299 (2000).

    Article  CAS  Google Scholar 

  53. Wallin, N. et al. The Swedish motor neuron disease quality registry. https://doi.org/10.1080/21678421.2018.1497065 (2018).

  54. Schwenk, J. M., Gry, M., Rimini, R., Uhlén, M. & Nilsson, P. Antibody suspension bead arrays within serum proteomics. J. Proteome Res. 7, 3168–3179 (2008).

    Article  CAS  PubMed  Google Scholar 

  55. Häggmark, A. et al. Plasma profiling reveals three proteins associated to amyotrophic lateral sclerosis. Ann. Clin. Transl. Neurol. 1, 544–553 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Pin, E. et al. Array-based profiling of proteins and autoantibody repertoires in CSF. Methods Mol. Biol. 2044, 303–318 (2019).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

S.A.L. is supported by the Olle Engkvist Byggmästare Foundation (SLS-499431); Ulla-Carin Lindquists Stiftelse för ALS-forskning, Åhléns Foundation (mA2/h17, 203074); the Thierry Latran Foundation (FIB-ALS); and Neuroförbundet. N.S. was supported by the Wellcome Trust (108726/Z/15/Z), Edmond J. Safra Foundation, Lily Safra and UK Dementia Research Institute. We thank the ALS Stichting grant ‘The Dutch ALS Tissue Bank’ (E.A.) and Netherlands Brain Bank (E.H.) for providing ALS tissue samples. We acknowledge the team who helped in the collection of ALS tissue samples (D. Troost, M. de Visser, A.J. van der Kooi and J. Raaphorst). U.K. and C.I. are supported by Björklunds Fund, the Ulla-Carin Lindquist Foundation, Neuro Sweden and SLL Hälsa Medicin och Teknik. E.R.-V. is supported by the Swedish Alzheimer Foundation (Alzheimerfonden), Swedish Dementia Association (Demensfonden), Gun & Bertil Stohne’s Foundation and Gamla Tjänarinnor Foundation. This project has received funding from the European Research Council under the European Union’s Horizon 2020 Research and Innovation Program (grant agreement no. 772376 – EScORIAL) awarded to J.V. R.A.H. is supported by Alltid Litt Sterkere, AlzheimerFonden, Swedish Medical Research Council, Swedish Brain Foundation and the Karolinska Institutet. M.L. was supported by a grant from the Knut and Alice Wallenberg Foundation (2012.0091). This study was also supported by grants to P.N. from the Swedish FTD Initiative funded by the Schörling Family Foundation and the KTH Center for Applied Precision Medicine funded by the Erling-Persson Family Foundation. P.V.D. holds a senior clinical investigatorship of FWO-Vlaanderen and is supported by the E. von Behring Chair for Neuromuscular and Neurodegenerative Disorders, the ALS Liga België and the KU Leuven funds ‘Een Hart voor ALS’, ‘Laeversfonds voor ALS Onderzoek’ and the ‘Valéry Perrier Race against ALS Fund’. Several authors of this publication are members of the European Reference Network for Rare Neuromuscular Diseases. We also thank K. Hultenby at the Karolinska Institutet’s electron microscopy facility; G. Ella Thorlacius, M.-G. Hong, S. Bergström, J. Yousef, H. Sarlus and A. Manouchehrinia for support with data analysis; and T. Brännström and M. Marklund for assistance with selection and collection of SOD1G93A mouse tissues. S.A.L. thanks the unpaid interns on Erasmus scholarships for their efforts and contributions.

Author information

Authors and Affiliations

Authors

Contributions

N.S. and S.A.L. designed transcriptomics enrichment experiments. N.S. and A.S. performed the computational analysis of enrichment experiments. M.T., I.S.A., A.D. and P.L. performed additional transcriptomics analysis. M.L., J.D.G. and S.A.L. facilitated and performed mouse histology and immunostaining. I.V.G.A. performed histology validation experiments. L.E., M.T. and S.A.L. performed electron microscopy imaging and quantifications. R.A.H., E.H., J.A., C.M. and E.A. facilitated and performed human histology staining. Clinical centers in Ulm (A.H. and A.L.), Utrecht (H.V.B. and J.V.), Leuven (J.D.V., M.D.S., K.P. and P.V.D.) and Stockholm (C.I. and U.K.) designed patient and control cohorts and provided plasma or CSF samples. S.A.L., A.M. and J.R. designed and chose protein targets for plasma profiling. A.M., J.R., J.O., F.S., S.W., M.U., P.N., E.R.-W. and C.I. facilitated and performed plasma profiling, survival associations and clinical parameter statistics. S.A.L. wrote the manuscript with input from the co-authors. P.N. and S.A.L. oversaw all aspects of the study.

Corresponding author

Correspondence to Sebastian A. Lewandowski.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer reviewer information Nature Medicine thanks Robert Baloh and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Jerome Staal 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 Cell type-specific transcripts regulated in SOD1G93A mice identified by EWCE rankings.

RNA expression dynamics graphs show genes activated at selected timepoints of ALS progression in spinal cords of SOD1G93A mice. Expression is shown as fold of change ± SEM. Cell type RNA specificity graphs show data from single cell transcriptomics of mouse cortex and oligodendrocyte lineage (Zeisel et al. 2015, Marques et al. 2016). Bars represent relative count of RNA per cell ± SEM and median values from the following n number of cells: Perivascular fibroblasts (PVF=76), Vascular smooth muscle (vSMC=62), Endothelium (EC=137), Pericytes (PC=21), Microglia (MG=98), Astrocytes (AC=224), Interneurons (IN=290), Excitatory neurons (EN=1338), Oligodendrocytes (OD=4528) and Oligodendrocyte precursors (OPC=449).

Extended Data Fig. 2 Vascular cell-enriched transcripts regulated in SOD1G93A mice identified by EWCE rankings.

a, RNA expression dynamics graphs show genes activated at selected timepoints of ALS progression in spinal cords of SOD1G93A mice. Cell type RNA specificity graphs show data from single cell transcriptomics of mouse cortex with oligodendrocyte lineage (set 1: Zeisel et al. 2015, Marques et al. 2016) and brain vascular resource (set 2: Vanlandewijck et al. 2018). Bars represent median count of RNA per cell ± SEM. b, P-values for two -tailed ANOVA and t-test for genes presented in Fig. 1e. c, P-values for EWCE scores for graphs presented in Fig. 1b.

Extended Data Fig. 3 Enlarged perivascular spaces coincide with separation of astrocyte and mural basement membranes in sporadic ALS patients.

a, COL6A1 histochemistry indicates enlarged perivascular spaces in sALS patient spinal cord grey matter (ROI 1). Suggested location of endothelial cell (EC - grey arrow), mural cell (MC - black arrow) and perivascular fibroblast (PVF - red arrow) in relation to COL6A1 layers (ROI 2,3), bar: 200 μm (overview), 10 μm (ROIs). b, Two color histochemistry indicates separation of astrocyte basement membrane (LAMA1) from mural basement membrane (COL4A1) in sALS patients, bar: 10 μm.

Extended Data Fig. 4 COL6A1 and SPP1 proteins accumulate in enlarged perivascular spaces in sporadic ALS patients’ spinal cords.

a, Two-color histochemistry for COL6A1 or SPP1 (red) relative to mural basement membrane outlined with COL6A1 (blue). Four sporadic ALS cases are represented with non neurological control spinal cords, bar: 100 μm. b, Correlation of SPP1 and COL6A1 histochemistry with pathological scores for GFAP, HLA-DR and motor neuron (MN) loss. Spearman test R-rho and two-tailed p-vales are indicated in each graph.

Extended Data Fig. 5 Basement membrane gene ontology groups are enriched in presymptomatic stages of ALS.

Enrichment of gene sets from gene ontology groups (basement membrane - GO 0005604, basement membrane organization - GO 0071711 and collagen metabolic process - GO 0032963) in sporadic ALS patients and SOD1G93A mice. Significantly regulated genes are shown as red circles (). Mroast test analysis from the limma R package. Multiple test (FDR) adjusted gene expression enrichment is shown as blue bar. p - FDR adjusted two-tailed mroast test p-value.

Extended Data Fig. 6 ALS patient cohort summary statistics.

a, Proportions between ALS patient and control samples with age and gender balance. n numbers are indicated within bars. b, Proportions within ALS patients for onset location, survival status and genetic vs. sporadic disease. c, Distributions within ALS patients for plasma sampling delay, age at symptom onset, proportion of known survival and ALS-FRS-R scale at sampling.

Extended Data Fig. 7 Antibodies against perivascular fibroblast enriched proteins used in the plasma profiling.

a, Antibodies for PVF cell proteins ranked by the survival estimate p-value (Kaplan-Meier log rank test) between protein value quartiles in plasma from ALS patients in the Utrecht cohort. Age at onset, site of onset and gender were used as covariates. b, Aminoacid sequences of E.coli expressed protein epitope signature tags (PrESTs) for COL6A1 (UniProt ID P12109) and SPP1 (UniProt ID P10451) used for immunisation.

Extended Data Fig. 8 Prognostic value of SPP1 and COL6A1 proteins in plasma of ALS patients within individual country cohorts.

a, SPP1 (HPA027541), (b) SPP1 (HPA005562) - technical valifdation antibody and (c) COL6A1 (HPA01914). Relative levels of protein levels in ALS patients and age-matched controls. Red color indicates thresholded protein level. Automated threshold selection using maximally selected log rank statistics. Boxplots show median, 2nd-3rd quartile and whiskers show ±1.5 of the IQR. COL6A1 protein values in the Belgian cohort did not indicate an optimized threshold and were separated at the median value. Kaplan-Meier survival estimate of high and low protein groups with two-tailed K-M logrank test p-values.

Extended Data Fig. 9 Prognostic value of SPP1 (validation antibody HPA005562) or COL6A1 proteins in plasma and CSF of ALS patients.

a, Relative levels of COL6A1 and SPP1 proteins in plasma, threshold selection and Kaplan-Meier survival estimates of ALS patients in discovery (n=452) and replication (n=122) cohorts. Red color indicates thresholded protein level. Thresholds are established using maximally selected log rank statistics. All boxplots show median, 2nd-3rd quartile and whiskers show ±1.5 of the IQR. COL6A1 protein values in the replication cohort did not indicate an optimized threshold and were separated at the median. Two-tailed Kaplan-Meier logrank test p-values. b, Cox proportional hazard models for continuous increase of SPP1 (HPA005562) and COL6A1 relative to hazard ratios indicated by gender, bulbar onset type plasma sampling age and cohort identity. Hazard ratios for Neurofilament light in CSF are indicated in the replication cohort. Whiskers represent 95% CI. c, Relative levels of SPP1 protein in CSF from the Swedish cohort from the two antibodies (HPA027541 and HPA005562). Threshold selection and Kaplan-Meier survival estimates of ALS patients. Two-tailed Kaplan-Meier logrank test p-values.

Extended Data Fig. 10 Plasma and CSF content of COL6A1 and SPP1 proteins in the Swedish cohort.

a, Correlations between the COL6A1 and SPP1 antibodies in the early sampling. Two-tailed Spearman test Rho and p-values are indicated within each graph. Error bands represent 95% CI. b, Relative levels of COL6A1 (HPA019142 antibody) and SPP1 protein (HPA027541 and HPA005562 antibodies) in plasma and CSF as referenced to sibling or spouse controls. All boxplots show median, 2nd-3rd quartile and whiskers show ±1.5 of the IQR. Wilcoxon test p-values are indicated above brackets. c, COL6A1 and SPP1 protein values during longitudinal sampling of plasma and CSF. Two-tailed Wilcoxon test p-values are indicated above brackets.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Månberg, A., Skene, N., Sanders, F. et al. Altered perivascular fibroblast activity precedes ALS disease onset. Nat Med 27, 640–646 (2021). https://doi.org/10.1038/s41591-021-01295-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-021-01295-9

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