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:

Large-scale associations between the leukocyte transcriptome and BOLD responses to speech differ in autism early language outcome subtypes

Abstract

Heterogeneity in early language development in autism spectrum disorder (ASD) is clinically important and may reflect neurobiologically distinct subtypes. Here, we identified a large-scale association between multiple coordinated blood leukocyte gene coexpression modules and the multivariate functional neuroimaging (fMRI) response to speech. Gene coexpression modules associated with the multivariate fMRI response to speech were different for all pairwise comparisons between typically developing toddlers and toddlers with ASD and poor versus good early language outcome. Associated coexpression modules were enriched in genes that are broadly expressed in the brain and many other tissues. These coexpression modules were also enriched in ASD-associated, prenatal, human-specific, and language-relevant genes. This work highlights distinctive neurobiology in ASD subtypes with different early language outcomes that is present well before such outcomes are known. Associations between neuroimaging measures and gene expression levels in blood leukocytes may offer a unique in vivo window into identifying brain-relevant molecular mechanisms in ASD.

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: Clinical behavioral trajectories over the first 4 years of life in TD toddlers and toddlers with ASD plus good or poor early language outcome.
Fig. 2: Decreased fMRI response to speech in toddlers with ASD and poor early language outcome.
Fig. 3: Multivariate gene coexpression–fMRI association in ASD with good or poor early language outcome and typically developing control toddlers.
Fig. 4: Tissue-class enrichments with sets of nonzero or zero association modules.
Fig. 5: Vocal learning, human-specific, and ASD-associated enrichments with sets of broadly expressed genes and nonzero or zero association modules.

Similar content being viewed by others

Data availability

The raw data that support the findings from this study are publicly available from the NIH National Database for Autism Research (NDAR). Raw blood leukocyte gene expression data are publicly available via Gene Expression Omnibus (GEO) (GSE42133; GSE111175). Songbird Area X gene expression data are publicly available via GEO (GSE34819). GTEx data are publicly available at https://gtexportal.org/. ASD postmortem cortical gene expression can be found at https://github.com/dhglab/Genome-wide-changes-in-lncRNA-alternative-splicing-and-cortical-patterning-in-autism/.

References

  1. Geschwind, D. H. & Levitt, P. Autism spectrum disorders: developmental disconnection syndromes. Curr. Opin. Neurobiol. 17, 103–111 (2007).

    Article  CAS  Google Scholar 

  2. Happé, F., Ronald, A. & Plomin, R. Time to give up on a single explanation for autism. Nat. Neurosci. 9, 1218–1220 (2006).

    Article  Google Scholar 

  3. Lai, M. C., Lombardo, M. V., Chakrabarti, B. & Baron-Cohen, S. Subgrouping the autism “spectrum”: reflections on DSM-5. PLoS Biol. 11, e1001544 (2013).

    Article  CAS  Google Scholar 

  4. Kapur, S., Phillips, A. G. & Insel, T. R. Why has it taken so long for biological psychiatry to develop clinical tests and what to do about it? Mol. Psychiatry 17, 1174–1179 (2012).

    Article  CAS  Google Scholar 

  5. Lombardo, M. V., Lai, M. C. & Baron-Cohen, S. Big data approaches to decomposing heterogeneity across the autism spectrum. Preprint at https://www.biorxiv.org/content/early/2018/03/17/278788 (2018).

  6. Kjelgaard, M. M. & Tager-Flusberg, H. An investigation of language impairment in autism: implications for genetic subgroups. Lang. Cogn. Process. 16, 287–308 (2001).

    Article  Google Scholar 

  7. Tager-Flusberg, H. & Kasari, C. Minimally verbal school-aged children with autism spectrum disorder: the neglected end of the spectrum. Autism Res. 6, 468–478 (2013).

    Article  Google Scholar 

  8. Howlin, P. Outcome in high-functioning adults with autism with and without early language delays: implications for the differentiation between autism and Asperger syndrome. J. Autism. Dev. Disord. 33, 3–13 (2003).

    Article  Google Scholar 

  9. Perry, A. et al. Predictors of outcome for children receiving intensive behavioral intervention in a large, community-based program. Res. Autism Spectr. Disord. 5, 592–603 (2011).

    Article  Google Scholar 

  10. Gotham, K., Pickles, A. & Lord, C. Trajectories of autism severity in children using standardized ADOS scores. Pediatrics 130, e1278–e1284 (2012).

    Article  Google Scholar 

  11. Venter, A., Lord, C. & Schopler, E. A follow-up study of high-functioning autistic children. J. Child Psychol. Psychiatry 33, 489–507 (1992).

    Article  CAS  Google Scholar 

  12. Szatmari, P. et al. Similar developmental trajectories in autism and Asperger syndrome: from early childhood to adolescence. J. Child Psychol. Psychiatry 50, 1459–1467 (2009).

    Article  Google Scholar 

  13. Courchesne, E. et al. TheASD Living Biology: from cell proliferation to clinical phenotype. Mol. Psychiatry https://doi.org/10.1038/s41380-018-0056-y (2018).

  14. Pramparo, T. et al. Cell cycle networks link gene expression dysregulation, mutation, and brain maldevelopment in autistic toddlers. Mol. Syst. Biol. 11, 841 (2015).

    Article  Google Scholar 

  15. Pramparo, T. et al. Prediction of autism by translation and immune/inflammation coexpressed genes in toddlers from pediatric community practices. JAMA Psychiatry 72, 386–394 (2015).

    Article  Google Scholar 

  16. Geschwind, D. H. & State, M. W. Gene hunting in autism spectrum disorder: on the path to precision medicine. Lancet. Neurol. 14, 1109–1120 (2015).

    Article  Google Scholar 

  17. Sandin, S. et al. The heritability of autism spectrum disorder. J. Am. Med. Assoc. 318, 1182–1184 (2017).

    Article  Google Scholar 

  18. Battle, A., Brown, C. D., Engelhardt, B. E. & Montgomery, S. B. Genetic effects on gene expression across human tissues. Nature 550, 204–213 (2017).

    Article  Google Scholar 

  19. Lombardo, M. V. et al. Different functional neural substrates for good and poor language outcome in autism. Neuron 86, 567–577 (2015).

    Article  CAS  Google Scholar 

  20. Boyle, E. A., Li, Y. I. & Pritchard, J. K. An expanded view of complex traits: from polygenic to omnigenic. Cell 169, 1177–1186 (2017).

    Article  CAS  Google Scholar 

  21. Wray, N. R., Wijmenga, C., Sullivan, P. F., Yang, J. & Visscher, P. M. Common disease is more complex than implied by the core gene omnigenic model. Cell 173, 1573–1580 (2018).

    Article  CAS  Google Scholar 

  22. Cremers, H. R., Wager, T. D. & Yarkoni, T. The relation between statistical power and inference in fMRI. PLoS ONE. 12, e0184923 (2017).

    Article  Google Scholar 

  23. Langfelder, P. & Horvath, S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinformatics 9, 559 (2008).

    Article  Google Scholar 

  24. Hickok, G. & Poeppel, D. The cortical organization of speech processing. Nat. Rev. Neurosci. 8, 393–402 (2007).

    Article  CAS  Google Scholar 

  25. Konopka, G. & Roberts, T. F. Insights into the neural and genetic basis of vocal communication. Cell 164, 1269–1276 (2016).

    Article  CAS  Google Scholar 

  26. Pfenning, A. R. et al. Convergent transcriptional specializations in the brains of humans and song-learning birds. Science 346, 1256846 (2014).

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

  28. De Rubeis, S. et al. Synaptic, transcriptional and chromatin genes disrupted in autism. Nature 515, 209–215 (2014).

    Article  Google Scholar 

  29. Bernier, R. et al. Disruptive CHD8 mutations define a subtype of autism early in development. Cell 158, 263–276 (2014).

    Article  CAS  Google Scholar 

  30. Morgan, J. T. et al. Microglial activation and increased microglial density observed in the dorsolateral prefrontal cortex in autism. Biol. Psychiatry 68, 368–376 (2010).

    Article  Google Scholar 

  31. Vargas, D. L., Nascimbene, C., Krishnan, C., Zimmerman, A. W. & Pardo, C. A. Neuroglial activation and neuroinflammation in the brain of patients with autism. Ann. Neurol. 57, 67–81 (2005).

    Article  CAS  Google Scholar 

  32. Voineagu, I. et al. Transcriptomic analysis of autistic brain reveals convergent molecular pathology. Nature 474, 380–384 (2011).

    Article  CAS  Google Scholar 

  33. Hilliard, A. T., Miller, J. E., Fraley, E. R., Horvath, S. & White, S. A. Molecular microcircuitry underlies functional specification in a basal ganglia circuit dedicated to vocal learning. Neuron 73, 537–552 (2012).

    Article  CAS  Google Scholar 

  34. Konopka, G. et al. Human-specific transcriptional networks in the brain. Neuron 75, 601–617 (2012).

    Article  CAS  Google Scholar 

  35. Liu, X. et al. Disruption of an evolutionarily novel synaptic expression pattern in autism. PLoS Biol. 14, e1002558 (2016).

    Article  Google Scholar 

  36. Eising, E. et al. A set of regulatory genes co-expressed in embryonic human brain is implicated in disrupted speech development. Mol. Psychiatry https://doi.org/10.1038/s41380-018-0020-x (2018).

  37. Parikshak, N. N. et al. Integrative functional genomic analyses implicate specific molecular pathways and circuits in autism. Cell 155, 1008–1021 (2013).

    Article  CAS  Google Scholar 

  38. Willsey, A. J. et al. Coexpression networks implicate human midfetal deep cortical projection neurons in the pathogenesis of autism. Cell 155, 997–1007 (2013).

    Article  CAS  Google Scholar 

  39. Miller, J. A. et al. Transcriptional landscape of the prenatal human brain. Nature 508, 199–206 (2014).

    Article  CAS  Google Scholar 

  40. Parikshak, N. N. et al. Genome-wide changes in lncRNA, splicing, and regional gene expression patterns in autism. Nature 540, 423–427 (2016).

    Article  CAS  Google Scholar 

  41. Rossignol, D. A. & Frye, R. E. Mitochondrial dysfunction in autism spectrum disorders: a systematic review and meta-analysis. Mol. Psychiatry 17, 290–314 (2012).

    Article  CAS  Google Scholar 

  42. Sanders, S. J. et al. Insights into autism spectrum disorder genomic architecture and biology from 71 risk loci. Neuron 87, 1215–1233 (2015).

    Article  CAS  Google Scholar 

  43. Marchetto, M. C. N. et al. Differential L1 regulation in pluripotent stem cells of humans and apes. Nature 503, 525–529 (2013).

    Article  CAS  Google Scholar 

  44. Nuttle, X. et al. Emergence of a Homo sapiens–specific gene family and chromosome 16p11.2 CNV susceptibility. Nature 536, 205–209 (2016).

    Article  CAS  Google Scholar 

  45. Hippolyte, L. et al. The number of genomic copies at the 16p11.2 locus modulates language, verbal memory, and inhibition. Biol. Psychiatry 80, 129–139 (2016).

    Article  CAS  Google Scholar 

  46. Demopoulos, C. et al. Abnormal speech motor control in individuals with 16p11.2 deletions. Sci. Rep. 8, 1274 (2018).

    Article  Google Scholar 

  47. Berman, J. I. et al. Abnormal auditory and language pathways in children with 16p11.2 deletion. Neuroimage Clin. 9, 50–57 (2015).

    Article  Google Scholar 

  48. Kosmicki, J. A. et al. Refining the role of de novo protein-truncating variants in neurodevelopmental disorders by using population reference samples. Nat. Genet. 49, 504–510 (2017).

    Article  CAS  Google Scholar 

  49. Lek, M. et al. Analysis of protein-coding genetic variation in 60,706 humans. Nature 536, 285–291 (2016).

    Article  CAS  Google Scholar 

  50. Abrahams, B. S. et al. SFARI Gene 2.0: a community-driven knowledgebase for the autism spectrum disorders (ASDs). Mol. Autism 4, 36 (2013).

    Article  Google Scholar 

  51. Pierce, K., Conant, D., Hazin, R., Stoner, R. & Desmond, J. Preference for geometric patterns early in life as a risk factor for autism. Arch. Gen. Psychiatry 68, 101–109 (2011).

    Article  Google Scholar 

  52. Pierce, K. et al. Eye tracking reveals abnormal visual preference for geometric images as an early biomarker of an autism spectrum disorder subtype associated with increased symptom severity. Biol. Psychiatry 79, 657–666 (2016).

    Article  Google Scholar 

  53. Pierce, K. et al. Detecting, studying, and treating autism early: the One-Year Well-Baby Check-Up Approach. J. Pediatr. 159, 458–465.e6 (2011).

    Article  Google Scholar 

  54. Wetherby, A. & Prizant, B. Communication and Symbolic Behavior Scales Developmental Profile, First Normed Edition (Paul H. Brookes, Baltimore, 2002).

  55. Wetherby, A. M., Brosnan-Maddox, S., Peace, V. & Newton, L. Validation of the Infant-Toddler Checklist as a broadband screener for autism spectrum disorders from 9 to 24 months of age. Autism 12, 487–511 (2008).

    Article  Google Scholar 

  56. Lord, C. et al. The autism diagnostic observation schedule-generic: a standard measure of social and communication deficits associated with the spectrum of autism. J. Autism. Dev. Disord. 30, 205–223 (2000).

    Article  CAS  Google Scholar 

  57. Mullen, E. M. Mullen Scales of Early Learning (American Guidance Service, Circle Pine, MN, USA, 1995).

  58. Sparrow, S., Cicchetti, D. & Balla, D. Vineland-II Scales Of Adaptive Behavior: Survey Form Manual (American Guidance Service, Circle Pines, MN, USA, 2005).

  59. Schroeder, A. et al. The RIN: an RNA integrity number for assigning integrity values to RNA measurements. BMC. Mol. Biol. 7, 3 (2006).

    Article  Google Scholar 

  60. Du, P., Kibbe, W. A. & Lin, S. M. lumi: a pipeline for processing Illumina microarray. Bioinformatics 24, 1547–1548 (2008).

    Article  CAS  Google Scholar 

  61. Gentleman, R. C. et al. Bioconductor: open software development for computational biology and bioinformatics. Genome. Biol. 5, R80 (2004).

    Article  Google Scholar 

  62. Chikina, M., Zaslavsky, E. & Sealfon, S. C. CellCODE: a robust latent variable approach to differential expression analysis for heterogeneous cell populations. Bioinformatics 31, 1584–1591 (2015).

    Article  CAS  Google Scholar 

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

    Article  Google Scholar 

  64. Storey, J. D. A direct approach to false discovery rates. J. R. Statist. Soc. B 64, 479–498 (2002).

    Article  Google Scholar 

  65. Zhang, B. & Horvath, S. A general framework for weighted gene co-expression network analysis. Stat. Appl. Genet. Mol. Biol. 4, Article17 (2005).

  66. Oldham, M. C., Horvath, S. & Geschwind, D. H. Conservation and evolution of gene coexpression networks in human and chimpanzee brains. Proc. Natl Acad. Sci. USA 103, 17973–17978 (2006).

    Article  CAS  Google Scholar 

  67. Langfelder, P., Luo, R., Oldham, M. C. & Horvath, S. Is my network module preserved and reproducible? PLoS. Comput. Biol. 7, e1001057 (2011).

    Article  CAS  Google Scholar 

  68. Eyler, L. T., Pierce, K. & Courchesne, E. A failure of left temporal cortex to specialize for language is an early emerging and fundamental property of autism. Brain 135, 949–960 (2012).

    Article  Google Scholar 

  69. Redcay, E. & Courchesne, E. Deviant functional magnetic resonance imaging patterns of brain activity to speech in 2-3-year-old children with autism spectrum disorder. Biol. Psychiatry 64, 589–598 (2008).

    Article  Google Scholar 

  70. Redcay, E., Haist, F. & Courchesne, E. Functional neuroimaging of speech perception during a pivotal period in language acquisition. Dev. Sci. 11, 237–252 (2008).

    Article  Google Scholar 

  71. Dehaene-Lambertz, G., Dehaene, S. & Hertz-Pannier, L. Functional neuroimaging of speech perception in infants. Science 298, 2013–2015 (2002).

    Article  CAS  Google Scholar 

  72. Genovese, C. R., Lazar, N. A. & Nichols, T. Thresholding of statistical maps in functional neuroimaging using the false discovery rate. Neuroimage 15, 870–878 (2002).

    Article  Google Scholar 

  73. Yarkoni, T., Poldrack, R. A., Nichols, T. E., Van Essen, D. C. & Wager, T. D. Large-scale automated synthesis of human functional neuroimaging data. Nat. Methods 8, 665–670 (2011).

    Article  CAS  Google Scholar 

  74. Krishnan, A., Williams, L. J., McIntosh, A. R. & Abdi, H. Partial Least Squares (PLS) methods for neuroimaging: a tutorial and review. Neuroimage 56, 455–475 (2011).

    Article  Google Scholar 

  75. McIntosh, A. R. & Lobaugh, N. J. Partial least squares analysis of neuroimaging data: applications and advances. Neuroimage 23, S250–S263 (2004).

    Article  Google Scholar 

  76. Cotney, J. et al. The autism-associated chromatin modifier CHD8 regulates other autism risk genes during human neurodevelopment. Nat. Commun. 6, 6404 (2015).

    Article  CAS  Google Scholar 

  77. Darnell, J. C. et al. FMRP stalls ribosomal translocation on mRNAs linked to synaptic function and autism. Cell 146, 247–261 (2011).

    Article  CAS  Google Scholar 

  78. Sugathan, A. et al. CHD8 regulates neurodevelopmental pathways associated with autism spectrum disorder in neural progenitors. Proc. Natl Acad. Sci. USA 111, E4468–E4477 (2014).

    Article  CAS  Google Scholar 

  79. Greenblatt, E. J. & Spradling, A. C. Fragile X mental retardation 1 gene enhances the translation of large autism-related proteins. Science 361, 709–712 (2018).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This research was supported by grants to E.C. and K.P. (NIMH R01-MH080134 (K.P.), NIMH R01-MH104446 (K.P.), NFAR grant (K.P.), NIMH Autism Center of Excellence grant P50-MH081755 (E.C. and K.P.), NIMH R01-MH036840 (E.C.), NIMH R01-MH110558 (E.C. and N.E.L.), NIMH U01-MH108898 (E.C.), NIDCD R01-DC016385 (E.C., K.P., and M.V.L.), CDMRP AR130409 (E.C.), and Simons Foundation 176540 (E.C.)). The work was additionally supported by an ERC Starting Grant (ERC-2017-STG; 755816) to M.V.L. and a grant from the Brain & Behavior Research Foundation (NARSAD) to T.P. We thank R. Znamirowski, C. Ahrens-Barbeau, S. Solso, K. Campbell, M. Mayo, and J. Young for help with data collection, and S. Spendlove and M. Weinfeld for assistance with clinical characterization of subjects.

Author information

Authors and Affiliations

Authors

Contributions

E.C., K.P., L.E., M.V.L., and T.P. conceived the idea and designed the study. M.V.L. conceived and performed all analyses. T.P., V.G., V.W., R.A.I.B., and N.E.L. aided in data analyses. E.C., K.P., L.E., L.L., and C.C.B. collected data. E.C., K.P., L.E., N.E.L., T.P., and M.V.L. obtained grant funding. M.V.L. and E.C. wrote the manuscript. All authors contributed to editing the manuscript.

Corresponding authors

Correspondence to Michael V. Lombardo or Eric Courchesne.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Integrated supplementary information

Supplementary Figure 1 Preservation of ASD blood leukocyte modules in ASD frontal and temporal cortical tissue.

Zsummary preservation statistics for all ASD blood leukocyte modules. Zsummary > 10 indicates strong preservation, while 2 < Zsummary < 10 indicates moderate preservation. Non-zero modules M2, M8, and M11 are annotated on this plot, since they show Zsummary > 2. M2 has Zsummary = 8.1, which indicates high-moderate preservation.

Supplementary Figure 2 Enrichments between non-zero and zero modules with vocal learning, human-specific, and ASD-associated gene lists after broadly expressed genes are removed.

Numbers within the cells indicate the enrichment odds ratio, while the color indicates the -log10(p-value). Cells highlighted in green pass multiple comparison correction at FDR q < 0.05.

Supplementary Figure 3 Soft power, TOM dendrogram, and module preservation plots for comparisons of each group.

Panel A shows the soft power plot for the main WGCNA analysis including data from all groups. A horizontal red line depicts soft power topology model fit R2 of 0.9, where the chosen soft power of 16 is located. Panel B shows the TOM dendrogram with modules labeled at the bottom. Panel C shows the module preservation Zsummary statistic for WGCNA analyses run separately on each group in order to show that networks are highly preserved (Zsummary > 10) across groups.

Supplementary Figure 4 Plots of module eigengenes for all modules and all groups.

For all comparisons shown below, the sample sizes are TD n = 37 (blue), ASD Good n = 40 (pink), ASD Poor n = 41 (green). The box in the boxplots indicates the interquartile range (IQR; Q1 indicates the 25th, while Q3 indicates the 75th percentile) and the whiskers indicate Q1-(1.5*IQR) or Q3 + (1.5*IQR). The line within the box represents the median. The y-axis indicates the module eigengene, while the x-axis shows each group.

Supplementary Figure 5 Module preservation between groups.

This figure shows the module preservation Zsummary statistic for WGCNA analyses run separately on each group in order to show that networks are highly preserved (Zsummary > 10) across groups.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–5

Reporting Summary

Supplementary Table 1

Statistics for linear mixed effect model analysis of clinical trajectory differences. Sheet 1 (Trajectories_ANOVA_allGrps) shows the F-statistic and p-values for main effects of Age and Group and the Age*Group interaction. Sheets 2–4 are the same F-statistic and p-values for analyses on pairwise group comparisons. Any comparison passing multiple comparison correction at FDR q<0.05 is highlighted in red

Supplementary Table 2

Statistics for ROI analyses. Sheet 1 called “ROI_ANOVAstats” are the ANOVA statistics from ROI analyses. Sheet 2 called “ROI_PairwiseGroupComparisons” are the statistics for the pairwise group-comparisons for ROI analyses

Supplementary Table 3

Differential expression analysis results. Sheet 1 called “DE_subtypes_genes” shows results from differential expression analysis at the gene-level. The main model evaluates the effect of group (TD, ASD Good, ASD Poor) with an F-test and corresponding p-value. The FDR q-value is also shown for each gene. No gene passes for a statistically significant of group at FDR q<0.05. All other columns in Sheet 1 show the pairwise group comparisons (t-statistic, p-value, and FDR q per each gene). Sheet 2 called “DE_subtypes_ME” shows results from differentially expression analysis on module eigengenes from the WGCNA analysis. The main model evaluated is the same model as the gene-level DE analysis, testing an effect of group (TD, ASD Good, ASD Poor), and the remaining columns show statistics for each pairwise group comparison. Just like the DE analysis at the gene-level, no modules show statistically significant effects of group surviving FDR q<0.05

Supplementary Table 4

Statistics from PLS analyses. Sheet 1 (“Subtype_PLS”) shows statistics for the PLS model that included TD and ASD subtypes (ASD Good and ASD Poor) as separate groups. Sheet 2 (“CaseCtrl_PLS”) shows statistics for the PLS model that included TD and ASD as the main group distinction (with no distinction for ASD language outcome subtypes). Sheet 3 (“Subtype_PLS_LV1_moduleCorr”) shows the fMRI-gene co-expression correlations and 95% confidence intervals for each group and each module and indicates which modules are non-zero or not

Supplementary Table 5

Biological process enrichment results for each module. Each sheet shows the results of biological process enrichment tests for each module from MetaCore GeneGO

Supplementary Table 6

Lists of genes within each module along with module membership statistics for each module. Each row shows a gene and each column shows the module membership value of that gene to each module. The column labeled “Module” shows the module label assigned to each gene

Supplementary Table 7

Demographic and clinical behavioral variables summary. This table shows demographic information such as sample size, sex, age, and descriptive statistics (mean with standard deviation in parentheses) for each clinical variable at the intake and outcome assessments

Supplementary Table 8

ANOVA statistics for cell type surrogate proportion variables estimated with CellCODE. Surrogate proportion variables of different leukocyte cell types were estimated with CellCODE and tested for a group difference with an ANOVA. The table reports the F-statistic and p-value for the group effect from these ANOVAs

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lombardo, M.V., Pramparo, T., Gazestani, V. et al. Large-scale associations between the leukocyte transcriptome and BOLD responses to speech differ in autism early language outcome subtypes. Nat Neurosci 21, 1680–1688 (2018). https://doi.org/10.1038/s41593-018-0281-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-018-0281-3

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