The genetic architecture of the human thalamus and its overlap with ten common brain disorders

Torbjørn Elvsåshagen, Alexey Shadrin, Oleksandr Frei, Dennis van der Meer, Shahram Bahrami, Vinod Jangir Kumar, Olav Smeland, Lars T Westlye, Ole A Andreassen, Tobias Kaufmann, Torbjørn Elvsåshagen, Alexey Shadrin, Oleksandr Frei, Dennis van der Meer, Shahram Bahrami, Vinod Jangir Kumar, Olav Smeland, Lars T Westlye, Ole A Andreassen, Tobias Kaufmann

Abstract

The thalamus is a vital communication hub in the center of the brain and consists of distinct nuclei critical for consciousness and higher-order cortical functions. Structural and functional thalamic alterations are involved in the pathogenesis of common brain disorders, yet the genetic architecture of the thalamus remains largely unknown. Here, using brain scans and genotype data from 30,114 individuals, we identify 55 lead single nucleotide polymorphisms (SNPs) within 42 genetic loci and 391 genes associated with volumes of the thalamus and its nuclei. In an independent validation sample (n = 5173) 53 out of the 55 lead SNPs of the discovery sample show the same effect direction (sign test, P = 8.6e-14). We map the genetic relationship between thalamic nuclei and 180 cerebral cortical areas and find overlapping genetic architectures consistent with thalamocortical connectivity. Pleiotropy analyses between thalamic volumes and ten psychiatric and neurological disorders reveal shared variants for all disorders. Together, these analyses identify genetic loci linked to thalamic nuclei and substantiate the emerging view of the thalamus having central roles in cortical functioning and common brain disorders.

Conflict of interest statement

T.E. is a consultant to BrainWaveBank and received speaker’s honoraria from Lundbeck and Janssen Cilag. O.A.A. is a consultant to BrainWaveBank and HealthLytix, and received speaker’s honoraria from Lundbeck. The remaining authors declare no competing interests.

Figures

Fig. 1. Analysis of the GWAS discovery…
Fig. 1. Analysis of the GWAS discovery sample identifies 42 loci associated with thalamic volumes.
a The thalamus was segmented into six nuclei groups—anterior, lateral, ventral, intralaminar, medial, and posterior nuclei—using Bayesian thalamus segmentation, . All data sets were assessed by visually inspecting axial view figures of the segmentations for each participant and we removed sets with segmentation errors, insufficient data quality, and pathologies. The nuclei volumes in the left and right thalamus were summed and these were used in the analyses. b Heritability estimates for the thalamic volumes in the discovery sample of n = 30,114 participants from the UK Biobank. All thalamic volumes showed substantial heritability. Data are presented as mean ± SE. cQQ plots for the thalamic volumes of the discovery sample. d Circular Manhattan plots of GWAS for thalamus volumes of the discovery sample. The innermost plot reflects the GWAS of whole thalamus volume, whereas from center to the periphery, the plots indicate the GWAS of the anterior, lateral, ventral, intralaminar, medial, and posterior nuclei, respectively. Black circular dashed lines indicate genome-wide significance (two-sided P < 7e − 9). Horizontal Manhattan plots for the seven volumes are shown in Supplementary Fig. 2. The colors in a, c, and d indicate the same volumes, i.e., red color reflects whole thalamus; orange, yellow, and light green indicate the anterior, lateral, and ventral nuclei, respectively; whereas dark green, blue, and magenta reflect intralaminar, medial, and posterior nuclei volumes, respectively. GWAS, genome-wide association studies; h2, heritability.
Fig. 2. GWGAS identifies 127 unique genes…
Fig. 2. GWGAS identifies 127 unique genes associated with thalamic volumes.
Nineteen genes were associated with whole thalamus; 4, 29, and 17 genes were associated with volumes of the anterior, lateral, and ventral nuclei; and 37, 11, and 21 genes were associated with intralaminar, medial, and posterior nuclei volumes, respectively. Lighter font color and higher position for gene names indicate greater Z-score. Underlined gene names designate genes that were significantly associated with more than one volume, whereas gene names not underlined indicate genes associated with only one volume. Additional results of the GWGAS are presented in Supplementary Data 12. GWGAS, genome-wide gene-based association analysis.
Fig. 3. Thalamocortical genetic relationships.
Fig. 3. Thalamocortical genetic relationships.
We found significant genetic correlations between all thalamic nuclei and distinct cortical regions. There were significant positive genetic correlations between anterior nuclei and medial premotor cortex; between lateral nuclei and mainly medial prefrontal, anterior cingulate, parietal, and visual cortices; between medial nuclei and prefrontal and temporal cortices; and between posterior nuclei and visual cortices. We also found significant negative genetic associations between ventral nuclei and visual, prefrontal, and temporal cortical regions, and between intralaminar nuclei and rostral medial prefrontal cortex. Warm and cool colors in cortical regions indicate significant positive and negative genetic correlations, respectively, after adjusting for analyses of 180 cortical regions and 6 volumes (two-sided P < FDR). Corresponding statistics are provided in Supplementary Data 15. FDR, false discovery rate; Rg, genetic correlation.
Fig. 4. LD-score regression-based genetic correlations between…
Fig. 4. LD-score regression-based genetic correlations between thalamic volumes and ten brain disorders.
We assessed genetic correlations between thalamic volumes and ten brain disorders using LD-score regression. Warm and cool colors indicate positive and negative genetic associations, respectively. There were significant positive correlations between the whole thalamus and PD, between posterior nuclei and BD, and between intralaminar nuclei and MS, as indicated by a black frame, after FDR correcting across all 70 analyses (7 volumes x 10 disorders; two-sided P < FDR). AD; Alzheimer’s disease. ADHD; attention-deficit hyperactivity disorder. ASD, autism spectrum disorder; BD, bipolar disorder; FDR, false discovery rate; FEP, focal epilepsy; GEP, generalized epilepsy; MD, major depression; MS, multiple sclerosis; PD, Parkinson’s disease; SCZ, schizophrenia.
Fig. 5. Genetic loci shared between thalamic…
Fig. 5. Genetic loci shared between thalamic volumes and ten brain disorders.
Conjunctional FDR analysis detected shared genetic loci across thalamic volumes and the ten disorders. The figure shows results with FDR thresholds of both 0.05 (circles) and 0.01 (triangles). We found the largest number of overlapping loci for SCZ (66), PD (26), and BD (15), when applying a conjunctional FDR threshold of 0.05. For ASD, ADHD, MD, MS, GEP, FEP, and MS, there were 8, 8, 17, 10, 14, 4, and 14 genetic loci jointly associated with thalamic volumes and disorders when using a threshold of 0.05, respectively. When using a conjunctional FDR threshold of 0.01, there were overlapping loci associated with thalamic volumes and SCZ (17), PD (14), BD (5), ASD (2), ADHD (1), MDD (3), MS (2), and AD (2), and no shared locus for GEP or FEP. AD, Alzheimer’s disease; ADHD, attention-deficit hyperactivity disorder; ASD, autism spectrum disorder; BD, bipolar disorder; FDR, false discovery rate; FEP, focal epilepsy; GEP, generalized epilepsy; MD, major depression; MS, multiple sclerosis; PD, Parkinson’s disease; SCZ, schizophrenia.

References

    1. Acsády L. The thalamic paradox. Nat. Neurosci. 2017;20:901–902. doi: 10.1038/nn.4583.
    1. Nakajima M, Halassa MM. Thalamic control of functional cortical connectivity. Curr. Opin. Neurobiol. 2017;44:127–131. doi: 10.1016/j.conb.2017.04.001.
    1. Hwang K, Bertolero MA, Liu WB, D’Esposito M. The human thalamus is an integrative hub for functional brain networks. J. Neurosci. 2017;37:5594–5607. doi: 10.1523/JNEUROSCI.0067-17.2017.
    1. Sherman SM. Thalamus plays a central role in ongoing cortical functioning. Nat. Neurosci. 2016;19:533–541. doi: 10.1038/nn.4269.
    1. Guo ZV, et al. Maintenance of persistent activity in a frontal thalamocortical loop. Nature. 2017;545:181–186. doi: 10.1038/nature22324.
    1. Bolkan SS, et al. Thalamic projections sustain prefrontal activity during working memory maintenance. Nat. Neurosci. 2017;20:987–996. doi: 10.1038/nn.4568.
    1. Schmitt LI, et al. Thalamic amplification of cortical connectivity sustains attentional control. Nature. 2017;545:219–223. doi: 10.1038/nature22073.
    1. Arcaro MJ, Pinsk MA, Chen J, Kastner S. Organizing principles of pulvino-cortical functional coupling in humans. Nat. Commun. 2018;9:5382. doi: 10.1038/s41467-018-07725-6.
    1. Aggleton JP, Pralus A, Nelson AJ, Hornberger M. Thalamic pathology and memory loss in early Alzheimer’s disease: moving the focus from the medial temporal lobe to Papez circuit. Brain. 2016;139:1877–1890. doi: 10.1093/brain/aww083.
    1. Parker PR, Lalive AL, Kreitzer AC. Pathway-specific remodeling of thalamostriatal synapses in Parkinsonian mice. Neuron. 2016;89:734–740. doi: 10.1016/j.neuron.2015.12.038.
    1. Woodward ND, Karbasforoushan H, Heckers S. Thalamocortical dysconnectivity in schizophrenia. Am. J. Psychiatry. 2012;169:1092–1099. doi: 10.1176/appi.ajp.2012.12010056.
    1. Hibar DP, et al. Subcortical volumetric abnormalities in bipolar disorder. Mol. Psychiatry. 2016;21:1710–1716. doi: 10.1038/mp.2015.227.
    1. Zhao B, et al. Genome-wide association analysis of 19,629 individuals identifies variants influencing regional brain volumes and refines their genetic co-architecture with cognitive and mental health traits. Nat. Genet. 2019;51:1637–1644. doi: 10.1038/s41588-019-0516-6.
    1. Satizabal CL, et al. Genetic architecture of subcortical brain structures in 38,851 individuals. Nat. Genet. 2019;51:1624–1636. doi: 10.1038/s41588-019-0511-y.
    1. Sudlow C, et al. UK biobank: an open access resource for identifying the causes of a wide range of complex diseases of middle and old age. PLoS Med. 2015;12:e1001779. doi: 10.1371/journal.pmed.1001779.
    1. Fischl B, et al. Whole brain segmentation: automated labeling of neuroanatomical structures in the human brain. Neuron. 2002;33:341–355. doi: 10.1016/S0896-6273(02)00569-X.
    1. Iglesias JE, et al. A probabilistic atlas of the human thalamic nuclei combining ex vivo MRI and histology. Neuroimage. 2018;183:314–326. doi: 10.1016/j.neuroimage.2018.08.012.
    1. Purcell S, et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am. J. Hum. Genet. 2007;81:559–575. doi: 10.1086/519795.
    1. van der Meer, D., Rokicki, J., Kaufmann, T., Córdova-Palomera, A. & Moberget, T. Brain scans from 21,297 individuals reveal the genetic architecture of hippocampal subfield volumes. Mol Psychiatry, 10.1038/s41380-41018-40262-41387 (2018).
    1. Elvsashagen T, et al. The genetic architecture of human brainstem structures and their involvement in common brain disorders. Nat. Commun. 2020;11:4016. doi: 10.1038/s41467-020-17376-1.
    1. Bulik-Sullivan B, et al. An atlas of genetic correlations across human diseases and traits. Nat. Genet. 2015;47:1236–1241. doi: 10.1038/ng.3406.
    1. Kircher M, et al. A general framework for estimating the relative pathogenicity of human genetic variants. Nat. Genet. 2014;46:310–315. doi: 10.1038/ng.2892.
    1. Lee JJ, et al. Gene discovery and polygenic prediction from a genome-wide association study of educational attainment in 1.1 million individuals. Nat. Genet. 2018;50:1112–1121. doi: 10.1038/s41588-018-0147-3.
    1. Pickrell JK, et al. Detection and interpretation of shared genetic influences on 42 human traits. Nat. Genet. 2016;48:709–717. doi: 10.1038/ng.3570.
    1. Lasagna-Reeves CA, et al. Reduction of Nuak1 decreases tau and reverses phenotypes in a tauopathy mouse model. Neuron. 2016;92:407–418. doi: 10.1016/j.neuron.2016.09.022.
    1. Wang Y, Mandelkow E. Tau in physiology and pathology. Nat. Rev. Neurosci. 2015;17:22. doi: 10.1038/nrn.2015.1.
    1. Chang D, et al. A meta-analysis of genome-wide association studies identifies 17 new Parkinson’s disease risk loci. Nat. Genet. 2017;49:1511–1516. doi: 10.1038/ng.3955.
    1. Finucane HK, Reshef YA. Heritability enrichment of specifically expressed genes identifies disease-relevant tissues and cell types. Nat. Genet. 2018;50:621–629. doi: 10.1038/s41588-018-0081-4.
    1. Watanabe K, Taskesen E, van Bochoven A, Posthuma D. Functional mapping and annotation of genetic associations with FUMA. Nat. Commun. 2017;8:1826. doi: 10.1038/s41467-017-01261-5.
    1. de Leeuw CA, Mooij JM, Heskes T, Posthuma D. MAGMA: generalized gene-set analysis of GWAS data. PLoS Comput. Biol. 2015;11:e1004219. doi: 10.1371/journal.pcbi.1004219.
    1. Lam M, et al. Large-scale cognitive GWAS meta-analysis reveals tissue-specific neural expression and potential nootropic drug targets. Cell Rep. 2017;21:2597–2613. doi: 10.1016/j.celrep.2017.11.028.
    1. Zeng L, et al. Identification of a novel human doublecortin-domain-containing gene (DCDC1) expressed mainly in testis. J. Hum. Gene.t. 2003;48:393–396. doi: 10.1007/s10038-003-0033-3.
    1. Sherman, S. M. & Guillery, R. W. Exploring the Thalamus and its Role in Cortical Function 2nd edn (MIT Press, Boston, 2006).
    1. Hawrylycz M, et al. Canonical genetic signatures of the adult human brain. Nat. Neurosci. 2015;18:1832–1844. doi: 10.1038/nn.4171.
    1. Richiardi J, et al. BRAIN NETWORKS. Correlated gene expression supports synchronous activity in brain networks. Science. 2015;348:1241–1244. doi: 10.1126/science.1255905.
    1. Glasser MF, et al. A multi-modal parcellation of human cerebral cortex. Nature. 2016;536:171–178. doi: 10.1038/nature18933.
    1. Bezdudnaya T, Keller A. Laterodorsal nucleus of the thalamus: a processor of somatosensory inputs. J. Comp. Neurol. 2008;507:1979–1989. doi: 10.1002/cne.21664.
    1. Conte WL, Kamishina H, Corwin JV, Reep RL. Topography in the projections of lateral posterior thalamus with cingulate and medial agranular cortex in relation to circuitry for directed attention and neglect. Brain Res. 2008;1240:87–95. doi: 10.1016/j.brainres.2008.09.013.
    1. Wang Z, et al. Community-informed connectomics of the thalamocortical system in generalized epilepsy. Neurology. 2019;93:e1112–e1122. doi: 10.1212/WNL.0000000000008096.
    1. Woodward ND, Giraldo-Chica M, Rogers B, Cascio CJ. Thalamocortical dysconnectivity in autism spectrum disorder: an analysis of the Autism Brain Imaging Data Exchange. Biol. Psychiatry Cogn. Neurosci. Neuroimaging. 2017;2:76–84.
    1. Demontis D, et al. Discovery of the first genome-wide significant risk loci for attention deficit/hyperactivity disorder. Nat. Genet. 2019;51:63–75. doi: 10.1038/s41588-018-0269-7.
    1. Autism Spectrum Disorders Working Group of The Psychiatric Genomics Consortium. Meta-analysis of GWAS of over 16,000 individuals with autism spectrum disorder highlights a novel locus at 10q24.32 and a significant overlap with schizophrenia. Mol. Autism8, 21 (2017).
    1. Stahl EA, et al. Genome-wide association study identifies 30 loci associated with bipolar disorder. Nat. Genet. 2019;51:793–803. doi: 10.1038/s41588-019-0397-8.
    1. Wray NR, et al. Genome-wide association analyses identify 44 risk variants and refine the genetic architecture of major depression. Nat. Genet. 2018;50:668–681. doi: 10.1038/s41588-018-0090-3.
    1. Hyde CL, et al. Identification of 15 genetic loci associated with risk of major depression in individuals of European descent. Nat. Genet. 2016;48:1031–1036. doi: 10.1038/ng.3623.
    1. Ripke S, et al. Biological insights from 108 schizophrenia-associated genetic loci. Nature. 2014;511:421–427. doi: 10.1038/nature13595.
    1. Lambert JC, et al. Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer’s disease. Nat. Genet. 2013;45:1452–1458. doi: 10.1038/ng.2802.
    1. Sawcer S, et al. Genetic risk and a primary role for cell-mediated immune mechanisms in multiple sclerosis. Nature. 2011;476:214–219. doi: 10.1038/nature10251.
    1. Nalls MA, et al. Large-scale meta-analysis of genome-wide association data identifies six new risk loci for Parkinson’s disease. Nat. Genet. 2014;46:989–993. doi: 10.1038/ng.3043.
    1. International League Against Epilepsy Consortium on Complex Epilepsies. Genome-wide mega-analysis identifies 16 loci and highlights diverse biological mechanisms in the common epilepsies. Nat. Commun. 9, 5269 (2018).
    1. Andreassen OA, et al. Improved detection of common variants associated with schizophrenia by leveraging pleiotropy with cardiovascular-disease risk factors. Am. J. Hum. Genet. 2013;92:197–209. doi: 10.1016/j.ajhg.2013.01.001.
    1. Liu JZ, et al. Dense genotyping of immune-related disease regions identifies nine new risk loci for primary sclerosing cholangitis. Nat. Genet. 2013;45:670–675. doi: 10.1038/ng.2616.
    1. Schork AJ, Wang Y, Thompson WK, Dale AM, Andreassen OA. New statistical approaches exploit the polygenic architecture of schizophrenia-implications for the underlying neurobiology. Curr. Opin. Neurobiol. 2016;36:89–98. doi: 10.1016/j.conb.2015.10.008.
    1. Smeland OB, Frei O. Discovery of shared genomic loci using the conditional false discovery rate approach. Hum. Genet. 2020;139:85–94. doi: 10.1007/s00439-019-02060-2.
    1. Lennon MJ, Jones SP, Lovelace MD, Guillemin GJ, Brew BJ. Bcl11b-A critical neurodevelopmental transcription factor-roles in health and disease. Front. Cell Neurosci. 2017;11:89. doi: 10.3389/fncel.2017.00089.
    1. Lessel D, et al. BCL11B mutations in patients affected by a neurodevelopmental disorder with reduced type 2 innate lymphoid cells. Brain. 2018;141:2299–2311. doi: 10.1093/brain/awy173.
    1. Tropea TF, et al. TMEM106B effect on cognition in Parkinson disease and frontotemporal dementia. Ann. Neurol. 2019;85:801–811. doi: 10.1002/ana.25486.
    1. Boyle AP, et al. Annotation of functional variation in personal genomes using RegulomeDB. Genome Res. 2012;22:1790–1797. doi: 10.1101/gr.137323.112.
    1. Ernst J, Kellis M. ChromHMM: automating chromatin-state discovery and characterization. Nat. Methods. 2012;9:215–216. doi: 10.1038/nmeth.1906.
    1. Ernst J, Kellis M. Chromatin-state discovery and genome annotation with ChromHMM. Nat. Protoc. 2017;12:2478–2492. doi: 10.1038/nprot.2017.124.
    1. Subramanian A, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA. 2005;102:15545–15550. doi: 10.1073/pnas.0506580102.
    1. R Core Team. R: A Language and Environment for Statistical Computing (R Foundation for Statistical Computing, 2013).
    1. Andreassen OA, et al. Improved detection of common variants associated with schizophrenia and bipolar disorder using pleiotropy-informed conditional false discovery rate. PLoS Genet. 2013;9:e1003455. doi: 10.1371/journal.pgen.1003455.
    1. Nichols T, Brett M, Andersson J, Wager T, Poline JB. Valid conjunction inference with the minimum statistic. Neuroimage. 2005;25:653–660. doi: 10.1016/j.neuroimage.2004.12.005.

Source: PubMed

3
Sottoscrivi