Long-term neural and physiological phenotyping of a single human

Russell A Poldrack, Timothy O Laumann, Oluwasanmi Koyejo, Brenda Gregory, Ashleigh Hover, Mei-Yen Chen, Krzysztof J Gorgolewski, Jeffrey Luci, Sung Jun Joo, Ryan L Boyd, Scott Hunicke-Smith, Zack Booth Simpson, Thomas Caven, Vanessa Sochat, James M Shine, Evan Gordon, Abraham Z Snyder, Babatunde Adeyemo, Steven E Petersen, David C Glahn, D Reese Mckay, Joanne E Curran, Harald H H Göring, Melanie A Carless, John Blangero, Robert Dougherty, Alexander Leemans, Daniel A Handwerker, Laurie Frick, Edward M Marcotte, Jeanette A Mumford, Russell A Poldrack, Timothy O Laumann, Oluwasanmi Koyejo, Brenda Gregory, Ashleigh Hover, Mei-Yen Chen, Krzysztof J Gorgolewski, Jeffrey Luci, Sung Jun Joo, Ryan L Boyd, Scott Hunicke-Smith, Zack Booth Simpson, Thomas Caven, Vanessa Sochat, James M Shine, Evan Gordon, Abraham Z Snyder, Babatunde Adeyemo, Steven E Petersen, David C Glahn, D Reese Mckay, Joanne E Curran, Harald H H Göring, Melanie A Carless, John Blangero, Robert Dougherty, Alexander Leemans, Daniel A Handwerker, Laurie Frick, Edward M Marcotte, Jeanette A Mumford

Abstract

Psychiatric disorders are characterized by major fluctuations in psychological function over the course of weeks and months, but the dynamic characteristics of brain function over this timescale in healthy individuals are unknown. Here, as a proof of concept to address this question, we present the MyConnectome project. An intensive phenome-wide assessment of a single human was performed over a period of 18 months, including functional and structural brain connectivity using magnetic resonance imaging, psychological function and physical health, gene expression and metabolomics. A reproducible analysis workflow is provided, along with open access to the data and an online browser for results. We demonstrate dynamic changes in brain connectivity over the timescales of days to months, and relations between brain connectivity, gene expression and metabolites. This resource can serve as a testbed to study the joint dynamics of human brain and metabolic function over time, an approach that is critical for the development of precision medicine strategies for brain disorders.

Figures

Figure 1. Overview of the MyConnectome study…
Figure 1. Overview of the MyConnectome study design and analysis pipelines.
Top: a timeline of measurements obtained in the study for fMRI, behavioural measurements and blood samples. Each tick represents a single measurement. Middle: an overview of the resting-state fMRI analysis pipeline. Bottom: an overview of the RNA-sequencing pipeline.
Figure 2. Connectome-wide connectivity across methods.
Figure 2. Connectome-wide connectivity across methods.
Parcellated connectome matrices for (a) full correlation, (b) L2-regularized partial correlation, (c) meta-analytic task connectivity modelling and (d) diffusion tractography (binarized). Networks are sorted by network modules identified from the full correlation connectome. Module labels: none: unassigned, DMN:default mode network, V2: second visual network, FP1: fronto-parietal network, V1: primary visual network, DA: dorsal attention network, VA: ventral attention network, Sal: salience network, CO:cingulo-opercular network, SM:somatomotor network, FP2: secondary fronto-parietal network, MPar:medial parietal network, ParOcc: parieto-occipital network, subcort: subcortical regions.
Figure 3. Longitudinal variability in brain connectivity.
Figure 3. Longitudinal variability in brain connectivity.
(a) Similarity between connectome-wide connectivity patterns across sessions, computed as the Pearson correlation between the connectivity values across the parcellated connectivity matrix. Values on the diagonal as well as the lower plot represent the similarity between each session and the mean across sessions; off-diagonal elements reflect the similarity between each pair of sessions. (b) Time series of connectivity within modules (upper panel) and between modules (lower panel). Notations to the right of each row mark the presence of significant linear (L) and polynomial (P) trends.
Figure 4. Effects of feeding/caffeine on large-scale…
Figure 4. Effects of feeding/caffeine on large-scale network structure.
Networks were generated by binarizing the correlation matrices between parcels at a 1% density threshold, separately for Tuesdays (fasted) and Thursdays (fed/caffeinated). Network visualization was performed using yFiles organic layout in Cytoscape. Hubs are shown as larger nodes, with provincial hubs depicted as circles and connector hubs depicted as triangles. Network module membership is coded by node colour; major networks are shaded, including somatomotor (red), second visual (blue), cingulo-opercular (purple), fronto-parietal (yellow) and default mode (black).
Figure 5. Comparison of fMRI and diffusion…
Figure 5. Comparison of fMRI and diffusion connectivity measures.
(a) Functional connectomes were thresholded at varying densities, and the resulting connections were assessed to identify the proportion of those connections that had non-zero structural connectivity identified using probabilistic diffusion tractography (thresholded at 10% density). The dashed line represents the proportion expected by chance, based on randomization of the structural connections. (b) The proportion of connections surviving at each given density that were interhemispheric, at a range of densities, for each measure. (c) Functional connectomes were thresholded at 0.25% density and presented in three-dimensional stereotactic space. Red connections had non-zero tractography connections, whereas blue connections had zero tractography connections across 500,000 samples.
Figure 6. Diffusion tractography results.
Figure 6. Diffusion tractography results.
Diffusion-weighted imaging identified an anomalous feature with the subject's corpus callosum. Glyphs (left top) reflect the underlying dominant-fibre orientation peaks, and tractography image on the right (inferior view) highlights the region with crossing fibres.
Figure 7. Relations between gene expression and…
Figure 7. Relations between gene expression and resting-state connectivity.
Image shows a heatmap for associations between between-module connectivity in resting-state networks (rows) and gene expression in WGCNA modules (columns). The colour scale reflects the t-statistic for association between each pair of variables. Plus signs indicate those sets that are significant at q<0.1.
Figure 8. Phenome-wide network analysis.
Figure 8. Phenome-wide network analysis.
A ‘phenome-wide network' was generated by treating each significant association (FDR q<0.05) as an edge in a network. The node shape denotes the variable class, node colour denotes network modules as determined using Infomap clustering and the edge colour represents a sign of association (red: positive, blue:negative).

References

    1. Kupper Z. & Hoffmann H. Course patterns of psychosocial functioning in schizophrenia patients attending a vocational rehabilitation program. Schizophr. Bull. 26, 681–698 (2000).
    1. Chen R. et al.. Personal omics profiling reveals dynamic molecular and medical phenotypes. Cell 148, 1293–1307 (2012).
    1. Freimer N. & Sabatti C. The human phenome project. Nat. Genet. 34, 15–21 (2003).
    1. Bilder R. M. et al.. Phenomics: the systematic study of phenotypes on a genome-wide scale. Neuroscience 164, 30–42 (2009).
    1. Smith S. M. et al.. Correspondence of the brain's functional architecture during activation and rest. Proc. Natl Acad. Sci. USA 106, 13040–13045 (2009).
    1. Gordon E. M. et al.. Generation and evaluation of a cortical area parcellation from resting-state correlations. Cereb. Cortex doi:10.1093/cercor/bhu239 (2014).
    1. Laumann T. et al.. Functional network and areal organization of a densely-sampled individual human brain. Neuron 87, 657–670 (2015).
    1. Rosvall M. & Bergstrom C. T. Maps of random walks on complex networks reveal community structure. Proc. Natl Acad. Sci. USA 105, 1118–1123 (2008).
    1. Mueller S. et al.. Individual variability in functional connectivity architecture of the human brain. Neuron 77, 586–595 (2013).
    1. Rubinov M. & Sporns O. Weight-conserving characterization of complex functional brain networks. Neuroimage 56, 2068–2079 (2011).
    1. Guimer à R. & Nunes Amaral L. A. Functional cartography of complex metabolic networks. Nature 433, 895–900 (2005).
    1. Sporns O., Honey C. J. & Kötter R. Identification and classification of hubs in brain networks. PLoS ONE 2, e1049 (2007).
    1. Eickhoff S. B. et al.. Co-activation patterns distinguish cortical modules, their connectivity and functional differentiation. Neuroimage 57, 938–949 (2011).
    1. Smith S. M. et al.. Network modelling methods for fMRI. Neuroimage 54, 875–891 (2011).
    1. Reveley C. et al.. Superficial white matter fiber systems impede detection of long-range cortical connections in diffusion mr tractography. Proc. Natl Acad. Sci. USA 112, E2820–E2828 (2015).
    1. Van Essen D. C. et al.. Chapter 16—Mapping Connections in Humans and Non-Human Primates: Aspirations and Challenges for Diffusion Imaging 2nd edn Elsevier (2013).
    1. Kohane I. Deeper, longer phenotyping to accelerate the discovery of the genetic architectures of diseases. Genome. Biol. 15, 115 (2014).
    1. Hodes G. E. et al.. Individual differences in the peripheral immune system promote resilience versus susceptibility to social stress. Proc. Natl Acad. Sci. USA 111, 16136–16141 (2014).
    1. Rosenblat J. D., Cha D. S., Mansur R. B. & McIntyre R. S. Inflamed moods: A review of the interactions between inflammation and mood disorders. Prog. Neuropsychopharmacol. Biol. Psychiatry 53C, 23–34 (2014).
    1. Crum A. J., Corbin W. R., Brownell K. D. & Salovey P. Mind over milkshakes: mindsets, not just nutrients, determine ghrelin response. Health Psychol 30, 424–429 discussion 430–431 (2011).
    1. Kemeny M. E. & Schedlowski M. Understanding the interaction between psychosocial stress and immune-related diseases: a stepwise progression. Brain Behav. Immun. 21, 1009–1018 (2007).
    1. Podnar J., Deiderick H., Huerta G. & Hunicke-Smith S. Next-generation sequencing rna-seq library construction. Curr. Protoc. Mol. Biol. 106, 4.21.1–4.21.19 (2014).
    1. Zhang B. & Horvath S. A general framework for weighted gene co-expression network analysis. Stat. Appl. Genet. Mol. Biol. 4,, Article 17 (2005).
    1. Huang D. W., Sherman B. T. & Lempicki R. A. Systematic and integrative analysis of large gene lists using david bioinformatics resources. Nat. Protoc. 4, 44–57 (2009).
    1. Flisiak I., Chodynicka B., Porebski P. & Flisiak R. Association between psoriasis severity and transforming growth factor beta(1) and beta (2) in plasma and scales from psoriatic lesions. Cytokine 19, 121–125 (2002).
    1. Thompson P. M., Ge T., Glahn D. C., Jahanshad N. & Nichols T. E. Genetics of the connectome. Neuroimage 80, 475–488 (2013).
    1. Mamdani F. et al.. Coding and noncoding gene expression biomarkers in mood disorders and schizophrenia. Dis. Markers 35, 11–21 (2013).
    1. Glahn D. C. et al.. Genetic control over the resting brain. Proc. Natl Acad. Sci. USA 107, 1223–1228 (2010).
    1. Sprooten E. et al.. Common genetic variants and gene expression associated with white matter microstructure in the human brain. Neuroimage 97, 252–261 (2014).
    1. Frey B. J. & Dueck D. Clustering by passing messages between data points. Science 315, 972–976 (2007).
    1. Kamburov A., Cavill R., Ebbels T. M. D., Herwig R. & Keun H. C. Integrated pathway-level analysis of transcriptomics and metabolomics data with impala. Bioinformatics 27, 2917–2918 (2011).
    1. Hyndman R. J. & Khandakar Y. Automatic time series forecasting: the forecast package for r. J. Stat. Softw. 27, 1–22 (2008).
    1. Bopp J. M. et al.. The longitudinal course of bipolar disorder as revealed through weekly text messaging: a feasibility study. Bipolar Disord. 12, 327–334 (2010).
    1. Van Essen D. C. et al.. The human connectome project: a data acquisition perspective. Neuroimage 62, 2222–2231 (2012).
    1. Moeller S. et al.. Multiband multislice GE-EPI at 7 tesla, with 16-fold acceleration using partial parallel imaging with application to high spatial and temporal whole-brain fMRI. Magn. Reson. Med. 63, 1144–1153 (2010).
    1. Sotiropoulos S. N. et al.. Advances in diffusion mri acquisition and processing in the human connectome project. Neuroimage 80, 125–143 (2013).
    1. Logan G., Dagenbach D. & Carr T. in Inhibitory Processes in Attention, Memory, and Language (eds Dagenbach D., Carr T. H. 189–240Academic Press (1994).
    1. Fedorenko E., Hsieh P.-J., Nieto-Castañón A., Whitfield-Gabrieli S. & Kanwisher N. New method for fMRI investigations of language: defining rois functionally in individual subjects. J. Neurophysiol. 104, 1177–1194 (2010).
    1. Fedorenko E., Duncan J. & Kanwisher N. Broad domain generality in focal regions of frontal and parietal cortex. Proc. Natl Acad. Sci. USA 110, 16616–16621 (2013).
    1. Kastrup A., Li T. Q., Takahashi A., Glover G. H. & Moseley M. E. Functional magnetic resonance imaging of regional cerebral blood oxygenation changes during breath holding. Stroke 29, 2641–2645 (1998).
    1. Fiehn O. & Kind T. Metabolite profiling in blood plasma. Methods Mol. Biol. 358, 3–17 (2007).
    1. Fiehn O. et al.. Quality control for plant metabolomics: reporting MSIcompliant studies. Plant J. 53, 691–704 (2008).
    1. Boyd R. L. Riot scan: recursive inspection of text scanner (version 1.8.2). (2013).
    1. Pennebaker J. W., Chung C. K., Frazee J., Lavergne G. M. & Beaver D. I. When Small Words Foretell Academic Success: The Case of College Admissions Essays. PLOS ONE 9, e115844 doi:10.1371/journal.pone.0115844 (2014).
    1. Power J. D. et al.. Methods to detect, characterize, and remove motion artifact in resting state fMRI. Neuroimage 84, 320–341 (2014).
    1. Jenkinson M., Beckmann C. F., Behrens T. E. J., Woolrich M. W. & Smith S. M. Fsl. Neuroimage 62, 782–790 (2012).
    1. Eisenstein S. A. et al.. Characterization of extrastriatal d2 in vivo specific binding of [f](n-methyl)benperidol using pet. Synapse 66, 770–780 (2012).
    1. Friston K. J., Williams S., Howard R., Frackowiak R. S. & Turner R. Movement-related effects in fMRI time-series. Magn. Reson. Med. 35, 346–355 (1996).
    1. Glasser M. F. et al.. The minimal preprocessing pipelines for the human connectome project. Neuroimage 80, 105–124 (2013).
    1. Wig G. S. et al.. Parcellating an individual subject's cortical and subcortical brain structures using snowball sampling of resting-state correlations. Cereb. Cortex 24, 2036–2054 (2014).
    1. Dale A. M., Fischl B. & Sereno M. I. Cortical surface-based analysis. I. segmentation and surface reconstruction. Neuroimage 9, 179–194 (1999).
    1. Fischl B., Sereno M. I. & Dale A. M. Cortical surface-based analysis. II: inflation, flattening, and a surface-based coordinate system. Neuroimage 9, 195–207 (1999).
    1. Śegonne F. et al.. A hybrid approach to the skull stripping problem in MRI. Neuroimage 22, 1060–1075 (2004).
    1. Śegonne F., Pacheco J. & Fischl B. Geometrically accurate topology-correction of cortical surfaces using nonseparating loops. IEEE. Trans. Med. Imaging. 26, 518–529 (2007).
    1. Van Essen D. C., Glasser M. F., Dierker D. L., Harwell J. & Coalson T. Parcellations and hemispheric asymmetries of human cerebral cortex analyzed on surface-based atlases. Cereb. Cortex. 22, 2241–2262 (2012).
    1. Van Essen D. C. et al.. An integrated software suite for surface-based analyses of cerebral cortex. J. Am. Med. Inform. Assoc. 8, 443–459 (2001).
    1. Glasser M. F. & Van Essen D. C. Mapping human cortical areas in vivo based on myelin content as revealed by t1- and t2-weighted mri. J. Neurosci. 31, 11597–11616 (2011).
    1. Beucher S. & Lantúejoul C. Use of watersheds in contour detection. International Workshop on Image Processing: Real-time Edge and motion detection/estimation. Rennes, France (1979).
    1. Power J. D. et al.. Functional network organization of the human brain. Neuron 72, 665–678 (2011).
    1. Hsieh C.-J., Sustik M. A., Dhillon I. S. & Ravikumar P. Quic: quadratic approximation for sparse inverse covariance estimation. J. Mach. Learn. Res. 15, 2911–2947 (2014).
    1. van Wieringen W. N. & Peeters C. F. W. Ridge estimation of inverse covariance matrices from high-dimensional data. Preprint at arXiv: 1403.0904 (2014).
    1. Andersson J. L. R., Skare S. & Ashburner J. How to correct susceptibility distortions in spin-echo echo-planar images: application to diffusion tensor imaging. Neuroimage 20, 870–888 (2003).
    1. Li H. Aligning sequence reads, clone sequences and assembly contigs with bwa-mem. Preprint at arXiv: 1303.3997v1 (2013).
    1. Langmead B. & Salzberg S. L. Fast gapped-read alignment with bowtie 2. Nat. Methods 9, 357–359 (2012).
    1. Anders S. & Huber W. Differential expression analysis for sequence count data. Genome Biol. 11, R106 (2010).
    1. Ashurst J. L. et al.. The vertebrate genome annotation (vega) database. Nucleic Acids Res. 33, D459–D465 (2005).
    1. Langfelder P. & Horvath S. Wgcna: an r package for weighted correlation network analysis. BMC Bioinformatics 9, 559 (2008).
    1. Croft D. et al.. The reactome pathway knowledgebase. Nucleic Acids Res. 42, D472–D477 (2014).
    1. Mi H., Muruganujan A. & Thomas P. D. Panther in 2013: modeling the evolution of gene function, and other gene attributes, in the context of phylogenetic trees. Nucleic Acids Res. 41, D377–D386 (2013).
    1. Rhee S. Y., Wood V., Dolinski K. & Draghici S. Use and misuse of the gene ontology annotations. Nat. Rev. Genet. 9, 509–515 (2008).
    1. Shannon P. et al.. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 13, 2498–2504 (2003).
    1. Miller J. A. et al.. Strategies for aggregating gene expression data: the collapserows r function. BMC Bioinformatics 12, 322 (2011).
    1. Almasy L. & Blangero J. Multipoint quantitative-trait linkage analysis in general pedigrees. Am. J. Hum. Genet. 62, 1198–1211 (1998).

Source: PubMed

3
Abonneren