Although oncogenic mutations have been found in nondiseased, proliferative nonneural tissues, their prevalence in the human brain is unknown. Targeted sequencing of genes implicated in brain tumors in 418 samples derived from 110 individuals of varying ages, without tumor diagnoses, detected oncogenic somatic single-nucleotide variants (sSNV) in 5.4% of the brains, including IDH1R132H. These mutations were largely present in subcortical white matter and enriched in glial cells and, surprisingly, were less common in older individuals. A depletion of high-allele frequency sSNVs representing macroscopic clones with age was replicated by analysis of bulk RNA sequencing data from 1,816 nondiseased brain samples ranging from fetal to old age. We also describe large clonal copy number variants and that sSNVs show mutational signatures resembling those found in gliomas, suggesting that mutational processes of the normal brain drive early glial oncogenesis. This study helps understand the origin and early evolution of brain tumors.

Significance:

In the nondiseased brain, clonal oncogenic mutations are enriched in white matter and are less common in older individuals. We revealed early steps in acquiring oncogenic variants, which are essential to understanding brain tumor origins and building new mutational baselines for diagnostics.

This article is highlighted in the In This Issue feature, p. 1

Tumors result from the clonal expansion of cells due to the presence of somatic driver gene mutations in stem cells (1). Even in normal individuals, self-renewing tissues such as skin, blood, esophagus, endometrium, bladder, colon, and liver harbor cancer-associated somatic mutations, which increase with age (2–11). Recent work examining postmortem brains from a small cohort of 14 nondiseased aged individuals found mutations in cancer-associated genes, although none of the identified mutant alleles have a known role in oncogenesis (12). Consequently, the prevalence of oncogenic driver mutations in the nondiseased human brain remains largely unknown.

Primary gliomas and other brain tumors are considered to occur mostly within the white matter (WM; refs. 13, 14). WM is highly enriched in glial cells (approximately 75% oligodendrocyte-lineage cells, 20% astrocytes, and 5% microglia; ref. 15), whereas gray matter (GM) represents a combination of neurons and glial cells (15). One factor impeding assessing the contribution of clonal oncogenic mutations in the brain is the abundance of nonproliferating neurons concentrated in the GM (15). In contrast, brain-derived glial cells, including oligodendrocyte precursor cells (OPC; ref. 16) and astrocytes, to a lower extent (17), retain the ability to proliferate in the postnatal brain. Proliferation is a significant source of somatic mutations (18, 19); thus, prior conventional bulk analyses with mixtures of neurons and glial cells may have low sensitivity to discover oncogenic variants if they are present in glial cells, which represent just a fraction of the total cells (15).

To overcome this challenge, we performed targeted deep sequencing of WM areas to identify clonal somatic oncogenic variants and compared these with adjacent gray matter from the same brain region (Fig. 1; Supplementary Fig. S1). We also investigated 1,816 nondiseased brain RNA-sequencing (RNA-seq) data sets obtained from two independent cohorts, Genotype–Tissue Expression (GTEx; ref. 20) and BrainVar (21), representing different brain regions and ages, with methods that allow identification of clonal somatic variants. With our approach, we observe that the normal human brain harbors somatic single-nucleotide variants (sSNV) and large somatic copy number variants (sCNV) with oncogenic potential, suggesting glial susceptibility to acquire or further expand existing variants. In contrast with other tissues (3–8), the burden of clonal mutations representing macroscopic clones [variant allele frequency (VAF) >7%; ref. 22] in the brain does not detectably increase with age, so that the mutations are less common in older individuals. The patterns of nucleotide substitution for these sSNVs resemble those previously reported in brain tumors, suggesting that the mutational processes that give rise to brain tumors preexist in normal tissue.

Figure 1.

Experimental strategy. General scheme of our methodologic pipeline using targeted sequencing for variant discovery and in silico analyses of large independent cohorts spanning from fetal to older ages (BrainVar and GTEx databases). DNA obtained from 418 samples derived from normal brain and non–brain tissue were analyzed using MIPs, capturing genes associated with brain tumors, pan-cancer, and focal cortical dysplasia. Samples were deep-sequenced, and called variants were validated using Ion Torrent ultra-deep sequencing. Brain samples from BrainVar (n = 166) and GTEx (n = 1,640) databases were analyzed to discover oncogenic variants and to evaluate CNVs, mutational signatures, and aging correlations.

Figure 1.

Experimental strategy. General scheme of our methodologic pipeline using targeted sequencing for variant discovery and in silico analyses of large independent cohorts spanning from fetal to older ages (BrainVar and GTEx databases). DNA obtained from 418 samples derived from normal brain and non–brain tissue were analyzed using MIPs, capturing genes associated with brain tumors, pan-cancer, and focal cortical dysplasia. Samples were deep-sequenced, and called variants were validated using Ion Torrent ultra-deep sequencing. Brain samples from BrainVar (n = 166) and GTEx (n = 1,640) databases were analyzed to discover oncogenic variants and to evaluate CNVs, mutational signatures, and aging correlations.

Close modal

Experimental Scheme

We analyzed a total of 418 samples derived from 110 individuals spanning different ages (0–108 years; Supplementary Table S1A), with no history of neuro-oncologic or other neurologic diagnoses. We designed molecular inversion probes (MIP; ref. 23) that target all exons and adjacent intronic sequence (to capture splice site mutations) of 121 genes directly associated with brain tumors and other cancer types (Fig. 1; Supplementary Table S1B and S1C). Our panel represented multiple pathways implicated in disease and different classes of proto-oncogenes and tumor suppressor genes. First, to evaluate the presence and accumulation of oncogenic variants during aging in the normal brain, for each of the 110 participants, we analyzed data from at least two different brain regions and one nonbrain sample (Supplementary Table S1A). We primarily focused on the brain's frontal lobe because it is the most prevalent location for malignant tumor emergence, followed by temporal, parietal, and occipital (24). Brain samples consisted of hippocampus (HC), cerebellum (CER), and prefrontal cortex that was subdivided into gray matter (CXG) and white matter (CXW; Supplementary Fig. S1A). When no clear anatomic distinction between CXW and CXG was possible due to tissue size or frosting, the sample was labeled CX. Second, to test whether sampling more brain locations from one individual would increase our sensitivity for oncogenic variants, we also evaluated 91 samples derived from 17 different organs and the entire left hemisphere from one 17-year-old individual (UMB1465; Fig. 1; Supplementary Table S1A).

Identification of Variants from Deep-Targeted Sequencing

Somatic mutations associated with cancer can be either driver mutations, promoting clonal expansions in some cases, or passenger mutations with a less clear effect. To investigate the potential clonal variant accumulation in cancer genes within the nondiseased brain, we focused on low allele frequency variants with oncogenic potential. We define oncogenic variants as (i) previously reported pathogenic variants in cancer or (ii) predicted to be damaging by in silico analysis in a known cancer driver gene (25). We generated deep-targeted sequencing data (average coverage of 590× per sample across targeted regions; Supplementary Fig. S1B) and used the CLCbio Low Frequency Variant Detection algorithm (QIAGEN) to call somatic variants with a probabilistic error model to account for sequencing errors (see Methods). No significant difference in sequencing coverage across ages or tissue types was observed (Supplementary Fig. S1C and S1D).

We obtained a total of 51,036 raw calls (Supplementary Table S1D) and conservatively filtered out variants to obtain high-confidence mutations (see Methods), focusing only on variants with 0.5% to 15% allele frequency (VAF) due to germline contamination at higher VAFs (Supplementary Fig. S1E). Experimental sensitivity analyses using 165 spike-in somatic mutations (108 heterozygous and 57 homozygous SNPs) showed that this computational approach and filtration was optimized for low false-positive rates and achieved specificities of >99% with sensitivities comparable to other studies (12) at different mosaic fractions (Supplementary Fig. S1F). Thirty-five variants (average depth of 1,086×, median VAF of 1.86%) passed our filtering criteria, and of those, 28 were unique, whereas 7 were seen in different samples within the same individual (Supplementary Table S1E). One of these was discovered to be a germline event during validation. We used ultra-deep Ion Torrent sequencing (MIPP-Seq; Thermo Fisher Scientific—US; ref. 26) to validate 19 candidate mosaic variants, including all the 13 variants predicted to affect protein structure and 6 random synonymous, intronic, and promoter variants. With an average 92,757× per site, we achieved a validation rate of 89% with a high correlation of the VAFs between discovery and validation sequencing (r2 = 0.93, Pearson coefficient, Fig. 2A; Supplementary Table S1F).

Figure 2.

Nondiseased brains harbor low allele frequency cancer reported mutations. A, Correlation between MIPs and Ion Torrent VAFs of 12 unique variants detected in the normal brain. B, List of validated brain-specific somatic variants showing general information such as variant allele frequency, pathogenicity prediction, and presence in cancer databases. In addition, we describe detected germline variants with functional impact for each individual. N/R, not reported; NSYND3, highest score in predicted pathogenicity; UNCERTAIN, uncertain pathogenicity of variants with clinical significance, CLINSIG. C, Distribution frequency of genes affected by the detected oncogenic variants found in the brain. D, Distribution frequency of the most affected driver genes found in LGG with pathogenicity relevance (Intogen database); black arrows indicate overlap with our discovered genes. E, Number of oncogenic mutations found in CXW (circles, two-tailed Fisher exact test, P = 0.025) as a function of age (years). F, Comparison of the number of pathogenic mutations found in CX (n = 53), CXG (n = 92), and CXW (n = 94) and HC (n = 69); Fisher exact test, CXW versus CXG P = 0.028, HC versus CXG P = 0.182, CXW versus HC P = 0.4.

Figure 2.

Nondiseased brains harbor low allele frequency cancer reported mutations. A, Correlation between MIPs and Ion Torrent VAFs of 12 unique variants detected in the normal brain. B, List of validated brain-specific somatic variants showing general information such as variant allele frequency, pathogenicity prediction, and presence in cancer databases. In addition, we describe detected germline variants with functional impact for each individual. N/R, not reported; NSYND3, highest score in predicted pathogenicity; UNCERTAIN, uncertain pathogenicity of variants with clinical significance, CLINSIG. C, Distribution frequency of genes affected by the detected oncogenic variants found in the brain. D, Distribution frequency of the most affected driver genes found in LGG with pathogenicity relevance (Intogen database); black arrows indicate overlap with our discovered genes. E, Number of oncogenic mutations found in CXW (circles, two-tailed Fisher exact test, P = 0.025) as a function of age (years). F, Comparison of the number of pathogenic mutations found in CX (n = 53), CXG (n = 92), and CXW (n = 94) and HC (n = 69); Fisher exact test, CXW versus CXG P = 0.028, HC versus CXG P = 0.182, CXW versus HC P = 0.4.

Close modal

Presence of Brain Tumor–Associated Oncogenic Variants in Normal Brain

Among the validated variants, 12 occurred in cancer driver genes. These variants were predicted damaging and pathogenic by multiple algorithms (see Methods) using criteria similar to a recent study (27). Nine of these 12 variants were found in the brain and 3 in nonbrain tissue. The brain-specific variants were all exonic and had a median VAF of 1.2% (Supplementary Fig. S1G). These variants distributed similarly between proto-oncogenes and tumor suppressor genes (25, 28) and had the highest score in our predicted pathogenicity scale (NSYND3; see Methods; ref. 27; Fig. 2B). Importantly, the variants we detected did not occur in genes most frequently mutated in clonal hematopoiesis that were included in our panel, indicating that these variants are not likely derived from blood contaminants, as observed in a recent study (12). All of our identified genes were among the most frequently mutated genes in lower-grade gliomas (LGG) and glioblastoma (GBM) but not medulloblastoma (ref. 25; Fig. 2C and D). Of the nine brain-specific variants, six were previously reported pathogenic mutations in cancer (COSMIC, Clinvar, HGMD, and ICGC; Fig. 2B). Among these variants, IDH1, PTPN11, NF1, and PTEN gene variants are of particular interest due to their high prevalence and established pathogenic effects in brain tumors.

For each participant, we evaluated germline variants with predicted deleterious impact to assess possible interactions with somatic mutations or double-hit events (somatic + germline). We identified 240 germline variants (median VAF 50.3% ± 3.3%) predicted to be damaging, and of those 51 were unique over 41 individuals (see Methods, Supplementary Table S1G). We did not detect biallelic double-hit events in our cohort, although our method would be unable to detect mosaic loss of heterozygosity (LOH) or deletions (29), and none of these variants had been previously related to disease, as expected from a nondiseased cohort (Fig. 2B; Supplementary Table S1G). We found no enrichment in predicted damaging germline variants (Fisher exact test, P = 0.621) among the individuals with somatic oncogenic mutations.

Oncogenic Brain Variants Are Not Detectably More Common in Older Individuals

We detected seven previously reported oncogenic variants over 6 different brains out of 110 total; therefore, 5.4% of the evaluated brains carried a reported pathogenic variant in a cancer driver gene. We did not find evidence that older (>30 years) individual brains were enriched with known oncogenic variants in cancer genes (Fig. 2E) at the level of mosaicism detectable with this approach, which contrasts with many other tissues that show accumulation of variants with age (3–8). On the contrary, we observed a depletion of oncogenic variants in the CXW samples of individuals older than 30 years without a cancer diagnosis (6/52 vs. 0/42, Fisher exact test, P = 0.025; 5/43 vs. 0/42 maintaining only one sample of the oversampled individual, P = 0.029). This observation was also true using 19 predicted pathogenic and nonpathogenic brain variants (13/114 vs. 6/143, Fisher exact test, P = 0.025) from our filtered call set (89% validation rate). We interpret these observations as suggestive of lack of an age-related increase in oncogenic variants in the normal brain. Importantly, this behavior was further confirmed in two orthogonal data sets presented in the following sections.

Oncogenic Brain Variants Are More Prevalent in the White Matter

Oncogenic brain variants exhibited increased occurrence in WM (6 mutations in 94 CXW samples tested) compared with no variants in adjacent GM samples (92 CXG samples; Fisher exact test, P = 0.028; Fig. 2F). There were no significant differences in sequencing coverage across tissue types that might explain this difference (Supplementary Fig. S1D). CX samples, which we could not distinguish as WM or GM, also had zero variants (53 samples). HC samples came second after CXW, with 2 detected variants over 69 samples tested (ratio, 0.03; Fisher exact test, CXW vs. HC, P = 0.4, CXG vs. HC, P = 0.182). For CER, we observed 1 mutation (1/10; ratio, 0.1), but given the small sample size, we cannot derive conclusions on this region.

IDH1R132H Mutation Is Enriched in Glial Cells

The most prevalent somatic mutation in glioma is IDH1R132H, present in more than 70% of World Health Organization grade II and III astrocytomas and oligodendrogliomas (30). We observed three mutations in IDH1 in two different individuals. The first individual (UMB1465, 17 years old) had two IDH1 reported mutations, R132H and R100Q, detected in cortical WM from distinct distant regions of the brain, corresponding to prefrontal cortex (PFC) and primary motor area, respectively (Fig. 3A). IDH1R100Q has been infrequently reported in gliomas, and its contribution to oncogenesis is still being determined (31, 32). The second individual (UMB 5621, 37 years old) bore the IDH1R132H mutation in the HC at a VAF of 0.8%. Interestingly, the R132H mutation observed in the PFC of UMB1465 was called in two adjacent WM samples of the same brain section with different VAFs (0.9 and 5.0%; Fig. 2B). For this individual, we sequenced >50 additional brain samples derived from the entire left hemisphere and most organs (Fig. 1; Supplementary Table S1), and we did not detect IDH1R132H in any of these with at least 0.1% VAF at >3,500× coverage (Fig. 3B). The difference in VAFs between the two adjacent R132H-bearing samples (5% vs. 0.9%) may reflect the distance from the center of the mutant clone (Fig. 3B). Mutations with a 5% VAF are often shared between tissues or within an organ (33), and therefore one would not expect such relatively high VAF variants to be restricted to one organ subregion. Because heterozygous IDH1 mutations confer a proliferative advantage in human astrocytes and remodel chromatin into a neural progenitor-like state (34, 35), the sharply different VAFs of this mutation are most consistent with a clonal event with enhanced proliferation, present in a ≈5-mm3 sample within the WM. This mutation's high VAF and focal nature suggest that it was acquired very late in development or postnatally, then further amplified by glial proliferation. Similar mechanisms can be proposed for the PTPN11, PTEN, and NF1 mutations we found (36–38).

Figure 3.

Oncogenic mutations are enriched in the white matter and glial cells. A, Schematic illustrating the discovery of the IDH1R132H mutation in the normal PFC of a 17-year-old individual (UMB1465). The mutation was identified in two adjacent WM samples and not present elsewhere, including GM from the same section or GM/WM from the following brain section. White matter is mainly composed of neuronal axons, astrocytes, oligodendrocytes, and OPCs, whereas GM is a combination of neurons with glial cells. B, Illustration of the two focal and distant pathogenic mutations found within the same brain. C, Schematic of nuclear sorting protocol to isolate neuronal (NEUN+) and nonneuronal cells (NEUN). Nuclei were evaluated using single-cell RNA-seq. t-distributed stochastic neighbor embedding plot of 3,700 NEUN+ nuclei, showing an exclusive presence of excitatory and inhibitory neurons but not glia (top). Evaluation of 1,800 NEUN nuclei showing the presence of glial cells but not neurons (bottom). D, Fold-change gene expression of NEUN versus NEUN+ nuclei subdivided by different brain cell types. E, Genotyping of the IDH1R132H mutation by digital droplet PCR (ddPCR). Graph shows the ratio of mutant/wild-type droplets analyzed in different sorted populations (each data point corresponds to 300 sorted nuclei), showing a nominal enrichment in the NEUN glial fraction. Genomic DNA without the IDH1 R132H mutation was used as a control for the ddPCR reaction (CTRL).

Figure 3.

Oncogenic mutations are enriched in the white matter and glial cells. A, Schematic illustrating the discovery of the IDH1R132H mutation in the normal PFC of a 17-year-old individual (UMB1465). The mutation was identified in two adjacent WM samples and not present elsewhere, including GM from the same section or GM/WM from the following brain section. White matter is mainly composed of neuronal axons, astrocytes, oligodendrocytes, and OPCs, whereas GM is a combination of neurons with glial cells. B, Illustration of the two focal and distant pathogenic mutations found within the same brain. C, Schematic of nuclear sorting protocol to isolate neuronal (NEUN+) and nonneuronal cells (NEUN). Nuclei were evaluated using single-cell RNA-seq. t-distributed stochastic neighbor embedding plot of 3,700 NEUN+ nuclei, showing an exclusive presence of excitatory and inhibitory neurons but not glia (top). Evaluation of 1,800 NEUN nuclei showing the presence of glial cells but not neurons (bottom). D, Fold-change gene expression of NEUN versus NEUN+ nuclei subdivided by different brain cell types. E, Genotyping of the IDH1R132H mutation by digital droplet PCR (ddPCR). Graph shows the ratio of mutant/wild-type droplets analyzed in different sorted populations (each data point corresponds to 300 sorted nuclei), showing a nominal enrichment in the NEUN glial fraction. Genomic DNA without the IDH1 R132H mutation was used as a control for the ddPCR reaction (CTRL).

Close modal

To investigate the lineage of origin of the IDH1R132H mutation, we evaluated the presence of this mutation in neuronal and nonneuronal nuclei (see Methods). NEUN-positive nuclei were analyzed using single-nucleus RNA-seq to confirm the identity of the sorted cells, demonstrating that this population was all neuronal nuclei, broadly subclassified into excitatory and inhibitory neurons, without any further contamination (Fig. 3C). As expected, the NEUN-negative fraction lacked neurons and represented a mixture of glial cells, including OPCs, astrocytes, oligodendrocytes, and a small fraction of microglial cells (Fig. 3C). NEUN-positive and NEUN-negative populations showed gene expression profiles consistent with neurons and glia (oligodendrocyte, astrocyte, and OPCs), respectively (Fig. 3D). The remaining sample containing the IDH1R132H mutation (individual UMB1465) was subjected to nuclear sorting and genotyping using digital droplet PCR targeting the R132H mutation. Our results demonstrate that the IDH1R132H mutation was enriched in the NEUN-negative population (Fig. 3E), consistent with our initial observation of its presence only in WM and suggesting a glial localization of this mutation. Interestingly, the R132H mutation appears to be detectable also at very low levels in the NEUN-positive population, although we cannot rule out low-level contamination in generating this signal or an early event in neural precursors. The shared presence of the R132H mutation in neurons and glia would suggest a congenital origin of this mutation because cortical neurons are virtually all generated prenatally.

Clonal Brain Variants at High Allele Fraction Do Not Increase with Age

To confirm the existence of oncogenic variants, we evaluated two independent cohorts of nondiseased human brains, including 1,640 and 166 bulk RNA-seq samples obtained from the GTEx project (20–79 years; ref. 20) and BrainVar database (fetal to 19 years; ref. 21), respectively, also representing different brain regions (Supplementary Fig. S2A and S2B). RNA-MuTect (22) was used to identify somatic variants (Supplementary Fig. S2C), and suspected RNA editing bases were removed (A>G, T>C). RNA-MuTect sensitivity was tested in normal samples with coextracted DNA and RNA data and was able to detect DNA mutations with allele fractions of >7% in the RNA, in cases where the gene was sufficiently highly expressed (22), and these were defined as macroscopic clones. In the GTEx cohort, we found a total of 590 variants, including 325 missense, of which 62 variants (19%) overlap with the exact amino acid change reported in the Catalogue of Somatic Mutations in Cancer (COSMIC; CMC v92), and 27 others are disruptive (splice site or nonsense, 5 with COSMIC overlap, 19%). In BrainVar, we found a total of 746 variants, including 493 missense (56 with COSMIC overlap, 11%) and 70 disruptive (5 with COSMIC overlap, 7%; Supplementary Table S2A–S2D). Within variants with COSMIC overlap, we identified reported variants in cancer driver genes (VAF ≤10%) associated with brain tumors, such as DDX3X and MAX, with DDX3X being mutated in 8% of medulloblastomas (25). We also detected predicted pathogenic variants (with a high score of pathogenicity NSYND3 and LOF) in other brain tumor driver genes highly represented in LGGs (FGFR1, PDGFRA, MTOR), but these exact base substitutions were not previously reported. Interestingly, we observed several variants in PDGFRA in both GTEx and BrainVar (VAF ≤10%) data sets, suggesting that this gene, which is of importance in all gliomas, is frequently mutated (25).

We used the BrainVar (n = 166, fetal to 19 years) and GTEx (n = 1,640, 20–79 years) mutation calls to further investigate our initial observation about the lack of age-related accumulation of detectable clonal variants in the brain with a less biased approach, including all expressed genes rather than only those previously implicated in brain tumors. By integrating both data sets and modeling the mutation counts per sample using mixed-effect negative binomial regression, while adjusting for a standardized RNA integrity score, standardized ischemic time, and standardized total mapped reads, we observed depletion of all [mean ratio = 0.241; 95% confidence interval (CI), 0.149–0.391; P = 3.5e-09), predicted pathogenic (mean ratio = 0.28; 95% CI, 0.155–0.506 95%; P = 1.7e-05), and disruptive variants (mean ratio = 0.366; 95% CI, 0.2–0.66; P = 0.001) with age (Fig. 4AC; Supplementary Fig. S2D–S2F; Supplementary Fig. S3), a discovery consistent with our panel findings. The negative association with age was also significant by analyzing all mutations from each data set independently (BrainVar, P = 0.00029; GTEx, P = 0.043) and also in GTEx for pathogenic and disruptive mutations (Supplementary Fig. S3A–S3E). For BrainVar, the regression model was not able to converge due to the low number of pathogenic and disruptive mutations. As a control, we did not observe a similar significant depletion for the T>C variants, removed as potential RNA editing events, in the combined data set (mean ratio = 0.765, P = 0.282) and also in each cohort independently (Supplementary Fig. S3F and S3G). Furthermore, analysis of all variants from both cohorts after only filtering known RNA editing sites in databases also showed a significant decrease with age (mean ratio = 0.411, P = 5.1e-06), demonstrating that T>C removal does not affect our observed aging trend (Supplementary Fig. S3H). The effect with age may vary for different brain subregions. Among 13 evaluated regions, cortex showed a nominal depletion of disruptive and pathogenic variants with age (P = 0.035 and P = 0.032, respectively), whereas cerebellum also showed significant depletion for pathogenic variants, P = 0.01 (and nominal for disruptive, P = 0.07; Supplementary Fig. S4A). Because we observed a general negative association when combining GTEx and BrainVar, and none of the evaluated brain regions showed a significant increase with age, we conclude that there is no age-related increase of clonal somatic mutations that reaches the level of detection of this method in normal brain.

Figure 4.

Somatic mutations detectable by RNA-seq do not accumulate with age in the normal brain. Evaluation of somatic mutations using RNA-MuTect in 1,640 GTEx and 166 BrainVar nondiseased brain samples. Suspected RNA editing bases A>G and T>C were removed to reduce false positives as well as variants with VAF >40%. Dot plots showing the proportion of samples with at least one mutation across age and forest plots of the aging incidence rate ratio for all mutations (A), predicted pathogenic and nonpathogenic mutations (B), and disruptive (nonsense, splice site) and nondisruptive mutations (3′ UTR, 5′ UTR, 5′ flank, or nonstop; C). Error bars are the Clopper–Pearson 95% confidence interval of the sample proportion. Forest plots also include standardized RNA integrity score and standardized total mappable read count, with horizontal lines indicating 95% confidence intervals. Incidence rate ratio was estimated using mixed-effects negative binomial model with donor ID as a random effect.

Figure 4.

Somatic mutations detectable by RNA-seq do not accumulate with age in the normal brain. Evaluation of somatic mutations using RNA-MuTect in 1,640 GTEx and 166 BrainVar nondiseased brain samples. Suspected RNA editing bases A>G and T>C were removed to reduce false positives as well as variants with VAF >40%. Dot plots showing the proportion of samples with at least one mutation across age and forest plots of the aging incidence rate ratio for all mutations (A), predicted pathogenic and nonpathogenic mutations (B), and disruptive (nonsense, splice site) and nondisruptive mutations (3′ UTR, 5′ UTR, 5′ flank, or nonstop; C). Error bars are the Clopper–Pearson 95% confidence interval of the sample proportion. Forest plots also include standardized RNA integrity score and standardized total mappable read count, with horizontal lines indicating 95% confidence intervals. Incidence rate ratio was estimated using mixed-effects negative binomial model with donor ID as a random effect.

Close modal

To learn more about how different brain regions compare with each other in their mutational burden, we used the negative binomial model to rank regions based on comparing region-specific mutation incidence rate to the overall mutation incidence rate (Supplementary Fig. S4B). When assessing all variants, only caudate (basal ganglia) had nominally fewer mutations than average (P = 0.039), whereas cerebellum exhibited a very significant increase in the relative mutation count (P = 4.2e-08; Supplementary Fig. S4B). The same pattern was true when assessing pathogenic and nonpathogenic variants, with the exception that in the cortex, pathogenic mutations also showed a nominal increase in the relative mutation burden compared with the overall brain mean (P = 0.024), with hypothalamus showing a decrease in overall mutation count (P = 0.034; Supplementary Fig. S4B).

Interestingly, we found one outlier sample (6-year-old female) in the BrainVar data set with a high mutation load (139 mutations called), including 23 mutations overlapping reported COSMIC events, suggesting a potential preneoplastic clonal expansion. The only drivers detected were GPC5 and FAT3, although neither of these have been directly associated with brain tumors. However, these types of mutations might increase proliferative fitness, leading to subsequent mutation accumulation, as reflected in the high mutational load of this sample.

Somatic Copy Number Alterations in Nondiseased Brain

Because sCNVs are the most frequent and important driver events in the oncogenesis of multiple brain tumors, we next assessed the prevalence of sCNVs in 1,636 brain samples across 253 participants from the GTEx (v7) consortium. We used a recently developed algorithm called superFreq (39) that leverages allele frequency information across germline heterozygous sites and read depth to identify sCNVs from RNA-seq data. Although this algorithm was designed for cancer samples, it can provide lower-bound estimates of the sCNV landscape in normal tissues. The initial raw call set consisted of 1,242 variants across 213 samples (Supplementary Table S2E). Due to the noisy nature of RNA-seq data, we implemented a stringent filtering strategy (see Methods). Briefly, we removed variants that overlapped fewer than 100 genes, so that the precision for those events is expected to be 80% to 90% (39), and we filtered variants whose log-fold change (LFC) and clonality were too noisy to be reliably estimated via visual inspection. The final call set was 37 sCNVs across 20 participants, consisting of 15 gains, 13 copy number neutral loss of heterozygosity (CN-LOH), and 9 losses (Fig. 5A). The mosaic fraction of these events ranged from 13.4% to 48.0%, with a median of 29.8%. From this sample size, we estimated the percentage of normal adult individuals to have at least one sCNV in a brain sample to be 7.9% (95% CI, 4.90–11.94; Fig. 5B). No sCNV rate differences were detected between brain regions, and no evidence of age-related change was observed (Fig. 5A). We also analyzed 147 participants from BrainVar and obtained 262 initial calls, of which 7 remained after filtering (2 losses, 1 gain, and 4 CN-LOHs), across 5 prenatal participants (Fig. 5A; Supplementary Table S2E), suggesting that 4.8% (95% CI, 1.11–7.78) of young brain samples have at least one sCNV detectable with this approach.

Figure 5.

CNVs are found in the normal brain. Visualization of detected sCNVs in the GTEx and BrainVar databases (A). Color codes indicate different age ranges, brain regions, and types of alteration (gain, loss, or LOH). Upper bar plot summarizes the counts of the different alteration types, and the bar plot on the right side also summarizes the alteration types but sorted by chromosome. Labels along the left y-axis refer to chromosome arms, and the percentages displayed in the right y-axis represent the frequency of events in each chromosome arm. B, Distribution of sCNV events across different ages from prenatal to elder. The number of individuals under each age range is described under the age label. C, Graphic representation of two representative sCNVs and the genes located in that region, one involving the whole chromosome 19 found at 19% clonality and the second involving the q-arm of chromosome 22 found at 24% clonality.

Figure 5.

CNVs are found in the normal brain. Visualization of detected sCNVs in the GTEx and BrainVar databases (A). Color codes indicate different age ranges, brain regions, and types of alteration (gain, loss, or LOH). Upper bar plot summarizes the counts of the different alteration types, and the bar plot on the right side also summarizes the alteration types but sorted by chromosome. Labels along the left y-axis refer to chromosome arms, and the percentages displayed in the right y-axis represent the frequency of events in each chromosome arm. B, Distribution of sCNV events across different ages from prenatal to elder. The number of individuals under each age range is described under the age label. C, Graphic representation of two representative sCNVs and the genes located in that region, one involving the whole chromosome 19 found at 19% clonality and the second involving the q-arm of chromosome 22 found at 24% clonality.

Close modal

Some of the sCNV overlap reported events in glioma and other brain tumors. Among the copy number losses in both data sets (n = 11), we observed four events in chromosome (Chr) 22q, four in Chr19, two in Chr16, and one in Chr2q (Fig. 5A). LOH22q has been reported in brain tumors, including astrocytoma (9%–30%), GBM (24%), and meningioma (65%; ref. 40). In two of the four 22q events, we detected loss of NF2 and SMARCB1 (Fig. 5C; Supplementary Fig. S5), which are highly involved in meningioma (40) and atypical teratoid rhabdoid tumors (41). Events in Chr19 were characterized by one 19q-arm loss and two 19p-arm losses. The 19q LOH and loss events have been frequently reported in oligodendrogliomas (100%), astrocytomas (30%–40%), and GBM (30%; ref. 42). We observed CN-LOH events overlapping CDKN2A and SMARCA4, two important genes in brain tumors (40), but the effect of these is less clear. Among copy number gains (n = 16), we detected five events in Chr6q, all of them gaining half of the q-arm, which includes relevant genes such as MYB, involved in pediatric gliomas (43). Chr1 had four partial gains of the q-arm, and 1q gains were reported in high-grade gliomas (44). We also detected partial and whole gains in Chr12p, Chr13q, Chr15q, Chr17q, and Chr18.

Clonal Variants in Normal Brain Share Mutational Mechanisms Seen in Brain Tumors

To understand specific mutagenic processes underlying the accumulation of clonal point mutations in the brain compared with other tissues, we performed mutational signature analyses using clonal sSNVs obtained from a recent study using the GTEx database (22). Using this data set allowed us to compare brain spectra with other organs using variants called with a consistent pipeline across tissues. We performed our analyses using various VAF cutoffs of 5% to 40% and obtained consistent results throughout this range (Fig. 6; Supplementary Fig. S6A). We focused on sSNVs with a VAF of less than 15% because they are most likely to reflect the sSNVs we targeted in our panel data. We found that the estimated mutational signatures from normal brains were similar to those from brain tumors. Normal brain sSNVs statistically decomposed into several signatures (SBS 39, 5, 23, 1, 30, and 2), each reported in COSMIC as present in brain tumors (Fig. 6) and consistent with previous findings (45). Our analysis confirmed that the mutational signatures found in normal brains are indeed enriched for brain cancer signatures (permutation test, P = 0.00018). The brain tumor signature enrichment was not observed in any other non-brain tissue tested using a Bonferroni-corrected P value of 0.01, except for pancreas (permutation test: pancreas, P = 0.00236; skin, P = 0.07895; and heart, P = 0.4; Fig. 6; Supplementary Fig. S6B). To validate our analysis, we processed skin sSNVs from the same study (22) and found that those signatures were enriched for skin cancer signatures (P = 0.00025; Fig. 6), consistent with previous findings (2). Normal pancreas sSNVs also showed a good correlation with those found in pancreatic cancer (permutation test, P = 0.0075; Supplementary Fig. S6B). The significant overlap of pancreas sSNVs with brain cancer signatures suggests similar mutational processes in these tissues, perhaps reflecting similarities in transcriptional and developmental programs (46). Brain mutational signatures reflect a combination of processes, including replication and transcription-induced mutations and their respective repair mechanisms. Interestingly, the COSMIC signature SBS1 observed in normal brain and in all tumor types is associated with cell division and proliferation (18), reflecting developmental processes or postnatal glial proliferation.

Figure 6.

Mutations found in normal brain exhibit signatures present in brain tumors. Mutational signature analysis of normal brain and skin (VAF <15%). Number of mutations evaluated is described next to the tissue label, and graphs show bases substitution, signatures, and spectrum obtained for each tissue. Colored circles next to each signature represent that the signature was observed in cancer (green, brain cancer; purple, skin cancer).

Figure 6.

Mutations found in normal brain exhibit signatures present in brain tumors. Mutational signature analysis of normal brain and skin (VAF <15%). Number of mutations evaluated is described next to the tissue label, and graphs show bases substitution, signatures, and spectrum obtained for each tissue. Colored circles next to each signature represent that the signature was observed in cancer (green, brain cancer; purple, skin cancer).

Close modal

In this study, we observed oncogenic variants in the brain of individuals without diagnosed cancer at a rate higher than the brain tumor prevalence (24), indicating that the mere presence of these events in the brain is not equivalent to clinical progression to cancer. This may have diagnostic implications because knowing the occurrence of oncogenic variants in normal tissue may help establish baselines for more accurate diagnosis.

Evaluation of 1,816 normal brain samples from two orthogonal studies allowed us to independently confirm the existence of oncogenic variants in the normal human brain, in concordance with a previous study (47). Although we do not see the same pattern of affected genes in our panel data (DNA sequencing) and the BrainVar- and GTEx-based analysis (RNA-seq), such as NF1 and IDH1 recurrences, this difference may relate to limitations of RNA-based mutation calling, such as tissue-specific and expression bias (WM enriched or GM enriched), coverage, and removal of RNA-editing bases (22). We also describe that sSNVs mutational signatures associated with brain tumors can be observed in normal brains, reflecting transcription and replication-induced mutations. Our data suggest that many signatures previously reported in brain tumors include many passenger mutations present in the normal brain and are not necessarily all tumor specific or strictly associated with malignancy. Based on our data, we believe that replication-induced mutations are likely a result of prenatal development or postnatal glial proliferation in concordance with previous etiologic factors contributing to brain cancer (1, 19). The contribution of signatures we see in normal tissue and brain tumors is different likely due to tissue sampling differences and because during tumor development, particular mutational mechanisms, such as SBS1, can diverge from those observed in the low-proliferating normal brain.

We adapted SuperFreq (39) for cloud computing to evaluate sCNVs in 1,783 normal brain samples, which to our knowledge comprises the largest normal brain cohort examined in this context. We report large chromosomal alterations in line with previous studies in single neurons (48–50) with some overlapping events reported in brain tumors such as 22q and 19q deletions (40). We only focused on large events to improve calling precision, limiting our discovery of smaller events. Gains were more frequent than losses, and losses mostly affected Chr22 and Chr19, whereas gains most commonly involved Chr6 and Chr1. sCNV events occurred at surprisingly high frequency in our cohorts (7.9% GTEx, 4.8% BrainVar) with a median mosaic fraction of 29.8% (14.9% VAF). Despite the high level of mosaicism of these sCNVs, they were often not shared between multiple brain regions, suggestive of restricted events arising during development or postnatally due to local clonal expansion. Given the low number of sCNV events we found, we cannot draw conclusions about any regional or aging trends. Our estimated rates of frequency in the cohort and mosaic fraction are reasonable compared with those found in a recent study using bulk whole-genome sequencing of postmortem brains (51). However, larger samples sizes and more sensitive techniques will be needed to more definitively determine rates of sCNV.

All the clonal oncogenic sSNVs found in the WM were detected in younger individuals in our targeted panel (<30 years), and we failed to find evidence of an age-related accumulation of oncogenic events. Given the relatively young age of the participants and postmortem nature of the data, we do not know whether those same individuals may develop cancer in the future from those mutant clones. In our panel data, we found a surprising lack of age-related oncogenic variant accumulation in the brain, which differs from findings in other tissues (3–8). It is worth noting that our targeted-sequencing approach evaluated only oncogenic variants, which differs from those studies in blood, skin, and esophagus, among others, that evaluated all mutations in known cancer genes (2, 4–9). Our BrainVar and GTEx analysis allowed us to look for all mutations in brain-expressed genes, thus resembling more closely previous studies. In this case, we also found a stable number, or even depletion, of all clonal, disruptive, and predicted pathogenic variants with age considering all brain samples from all ages. These results confirm our panel data finding and further contrast the mutational dynamics between the brain and other tissues. Two recent reports using GTEx data also included brain samples within a broader study. The first report indicates that some brain regions may have high correlation between mutations and age, even more than sun-exposed skin or blood, whereas other regions seem to be negatively associated (47). Nonetheless, all the evaluated brain regions do not achieve a high level of statistical support (showing a FDR >0.1) and hence are not inconsistent with a lack of age-related increase. In the second study (45), they report negative values of age correlation with brain mutations, but these are also insignificant, supporting a lack of age-related increase and a trend consistent with our findings.

The relative stability of oncogenic brain mutations with age, combined with the ability of RNA-seq analysis to detect only those clonal mutations with high mosaic fraction, suggests that some oncogenic mutations at a young age may be congenital. The modest reduction of oncogenic mutations with age may then reflect postnatal elimination of mutant clones from an individual, perhaps by immune surveillance, or postnatal elimination of individuals carrying mutant clones from the healthy cohort. Indeed, we evaluated prenatal and postnatal brain samples and found that during brain development, the mutation count and the frequency of nondiseased individuals with mutations are highest prenatally and then decline with age. Because the brain is an organ with low overall proliferation in postnatal stages, oncogenic clonal expansions over time can directly result in disease.

Both of our methods only have sensitivity to detect clones with a relatively large mosaic fraction. Although our targeted sequencing approach has a similar sensitivity to other methods (12), detecting ultra-low clonal events remains challenging. For example, the sensitivity for events with 0.5% VAF is ∼10%; hence, our rates may be underestimated. Similarly, the RNA-seq approach only detects macroscopic clones with VAF >7% (22). Therefore, we cannot by any means rule out, and in fact it seems plausible, that “micro” somatic variants at lower mosaic fraction may indeed show age-related accumulation at levels below our sensitivity to detect them.

All the pathogenic variants found in the cerebral cortex occurred in the WM. Two scenarios may explain this observation: (i) these are derived from active glial proliferation or (ii) subcortical WM is closer to the ventricles, and clones arising there can reach the WM more readily than the GM. Also, GM harbors large numbers of neurons, and these may further dilute such mutations, which might only be detectable by deeper sequencing. A follow-up study of WM of varying depths could test the second scenario. Future studies evaluating cancer variants in nondiseased brains should evaluate large cohorts of WM samples and GM to a higher depth.

We detected the IDH1R132H variant to be enriched in the nonneuronal population, which is 60% OPCs, and this is the most highly proliferative endogenous cell type of the brain (52). Brain tumors are thought to originate mainly from progenitor cells in neurogenic niches (53); however, the effect of oncogenic mutations in progenitors, such as OPCs residing in nonneurogenic niches such as the cortical WM, remains elusive. OPCs can produce tumors and have been identified in several reports as the most common cell of origin for gliomas (54–57). Thus, the IDH1R132H mutation detected in our glial fraction may constitute an early event in a pathogenic progression toward infiltrating glioma. In our case, we did not find more than one mutation per sample, but our limits of sensitivity would likely preclude the identification of emerging subclones.

Although our study represents a comprehensive survey of those sSNVs and sCNVs identifiable at high allele frequency from fetal to the old ages, a universe of events at lower mosaic fraction remains to be explored. Until now, the differential mutational burden between WM and GM remained largely unexplored, and this proved to be critical for discovering oncogenic variants in a normal brain. Although our findings also provide important information of early processes in the acquisition of oncogenic events in the brain, future studies addressing the accumulation of somatic variants in single glial cells may provide another layer of information to continue dissecting early mechanisms of brain oncogenesis.

Study Design

We analyzed a total of 418 samples derived from 110 individuals with no history of neuro-oncologic or other brain diseases, spanning different ages (0–108 years; Supplementary Table S1). We used MIPs (23) to evaluate mutations in 121 genes implicated in brain tumors, other cancers, and focal cortical dysplasia (Fig. 1; Supplementary Table S1). For each of the 110 participants, we analyzed at least two different brain regions and one nonbrain sample (Supplementary Table S1). Brain samples consisted of HC, CER, and prefrontal cortex that was subdivided into gray matter (CXG) and white matter (CXW; Supplementary Fig. S1A). When no clear distinction between CXW and CXG was possible, the sample was labeled as CX. Furthermore, we also evaluated 91 samples derived from 17 different organs and the entire left hemisphere from one 17-year-old individual (UMB1465; Fig. 1; Supplementary Table S1). We also evaluated the presence of somatic mutations in two large independent cohorts of 1,640 and 167 brain samples obtained from the GTEx project (20) and BrainVar (21), respectively, using RNA-MuTect (ref. 22; Supplementary Table S2A and S2B). GTEx provided samples from 13 different brain regions, and BrainVar provided samples from the dorsolateral PFC (mainly from Brodmann area 46) or from the frontal cerebral wall (for donors younger than 10 postconception weeks).

Variant Calling

Sample FASTQs were first subjected to a local realignment step using CLCbio (QIAGEN). Variant calling was performed using the CLCbio–Low Frequency Variant Detection mode that relies on statistical models for evaluating the sequencing error rate based on parameters defined by each batched analysis (http://resources.qiagenbioinformatics.com/manuals/clccancerresearchworkbench/200/index.php?manual=Low_Frequency_Variant_Detection.html). An error model is assumed and estimated for each nucleotide quality score. Error model parameters are all estimated from the data set being analyzed, so it will adapt to the sequencing technology used and the characteristics of the particular sequencing run. Samples with average coverage below 396× (1 SD from the mean) were not considered for the analysis. Resulting called variants were filtered out if they had a maximum allele frequency on the population greater than 0.1% (Gnomad; ref. 58), they occurred in more than three individuals from our cohort, the call quality score was less than 200, they were found in homopolymeric regions greater than 1 in length, they were covered by fewer than two different MIPs, they had fewer than 12 alternate reads covering the variant, a reference read depth was less than 200, the variant was present in SNP clusters, and they were not in a targeted region and not predicted to have functional impact on the protein function (see below, pathogenic classification). Only variants with a VAF between 0.5% and 15% were analyzed.

For germline calling, we evaluated variants that were between 40% and 60% allele frequency, were present in fewer than four individuals to avoid common variants, had a site coverage of at least 200×, had a call quality score of 200, were covered with more than one MIP, and were not present in SNP clusters or found in homopolymeric regions greater than 1 in length. In addition, because in most cases, we had multiple tissues from the same individual, the germline variants were required to be present in at least two samples to be considered for our analysis. Only variants with potential impact on protein function were included (see below, pathogenic classification; Supplementary Table S1).

Pathogenic Classification

Pathogenic classification of damaging missense variants was performed following a method reported in a previous study (27), and categorizing the predicted pathogenicity relied on six different prediction algorithms (SIFT51, PolyPhen2_HDIV52, PolyPhen2_HVAR, MutationTaster53, MutationAssessor54, and LRT55; refs. 59–61), damaging status and conservation sites. For example, NSYND3, which had the highest pathogenic score, was given if a variant was predicted to be pathogenic by at least five of six of the prediction tools above, considered damaging by CADD, DANN, or FATHMM, and affected a conserved site (42, 62, 63).

GTEx and BrainVar Mutation Calling and Signatures

We implemented RNA-MuTect (22) in Terra's Google Cloud Platform to call somatic mutations in 1,640 bulk RNA-seq brain samples retrieved from the GTEx project (release v7) and 167 bulk RNA-seq brain samples retrieved from BrainVar (21). RNA-MuTect was run using both the provided DNA panel-of-normals (PoN) based on ∼7,000 normal samples from The Cancer Genome Atlas and the provided RNA PoN based on a panel of ∼6,500 GTEx samples. The threshold for the minimum number of reads supporting the alternative allele was set to 4 as recommended by the pipeline authors (22). All A>G and T>C variants were removed from the call set to reduce false positives from RNA editing artifacts using the maftools package (release 3.11) in R (64) from all analysis unless otherwise specified. Variants greater than 40% VAF were removed from all analysis unless otherwise specified. In addition, variants were annotated as oncogenic if the protein change was present in the Cancer Mutation Census (v92; ref. 65). BrainVar sample “HSB498” was removed from all analysis as an outlier due to abnormally high mutation count and presence of several oncogenic mutations. In addition, BrainVar variants present in more than one sample (based on exact cDNA change) were removed from all analysis.

For age trend analyses, statistical regression was performed by fitting a mixed-effects negative binomial model on all sample mutation counts using the lme4 package (version 1.1–23) in R. For regression on the GTEx samples, the fixed effects included age (years) while adjusting for standardized RNA integrity score, standardized ischemic time, and standardized total mapped reads. GTEx donor ID and brain subregion were set as a random effect to adjust for donor-specific and region-specific variation. To compare the aging trend for each brain region, the brain region was removed as a random effect, and instead, the model was fit for each brain region separately. To compare the average mutation count for each brain region, the brain region was changed to a fixed effect, where each brain region variable compares its region-specific incidence rate with the overall incidence rate. For regression on the BrainVar samples, the fixed effects included stage (prenatal or postnatal) and sex (male or female) while adjusting for standardized RNA integrity score and standardized total mapped reads. To perform regression on the GTEx and BrainVar samples together, only the GTEx samples from the frontal cortex (BA9) were included.

For mutational signature analysis, we used variants from different organs obtained from a recent published study (22). We performed analyses using VAF filtering thresholds of 5% to 40%, which were relatively consistent. Mutations were filtered to retain only variants with less than 15% VAF (to match the variants we targeted with DNA sequencing), and suspected RNA editing bases were removed (A>G, T>C). Lists of mutations were analyzed using Mutalisk software (66). Enrichment analysis was performed with a permutation test, where we identified the number of mutational signatures present in a particular cancer type according to the COSMIC signature database (artifact signatures were not included) and then obtained random uniform samples from the total of 50 signatures with replacement and counted the number of signatures that were related to the given cancer type. The number of signatures sampled at each simulation was determined by the number of signatures that were estimated to contribute to a particular mutational spectrum by Mutalisk.

Somatic Copy Alteration Calling of Normal Brains

We identified putative sCNVs from nondiseased human brain samples of the GTEx consortium v7 (20) and BrainVar (21) using superFreq (39). The algorithm of superFreq employs an error propagation framework to leverage information from B-allele frequencies and read depth to identify sCNVs from RNA-seq data. Due to the large sample size (n = ∼1,700) samples, which were stored in the cloud, we adapted the superFreq workflow to be run in the cloud. The code to deploy the superFreq workflow and usage instruction were made publicly available through a github repository (https://github.com/emauryg/superFreq_cloud). On the basis of discussions with the developers of superFreq, we used 10 random samples as quality control reference samples for each cohort. These samples are used to remove variation in the data that originate from technical variability. The GTEx and BrainVar cohorts were run separately.

The raw call set was then filtered to obtain a final call set that minimized potential false positives. We focused our analyses on the autosomes and filtered sCNVs that overlapped fewer than 100 genes. Based on validation studies of the original superFreq manuscript, events that overlapped at least 100 genes had a high precision of 80% to 90% and a recall of 60%. We further filtered variants with predicted breakpoints in the MHC (6: 27486711-33448264, GRCh37) and KIR (9: 54574747-55504099) regions. We also filtered out events that had a clonality of >0.80 because these events would be more likely to be germline events in the noncancer setting. Last, we filtered out variants that did not pass visual inspection based on diagnostic plots.

Statistical analysis

Statistical analyses were performed as described in the main text with uncorrected P values, and the P value necessary for significance after multiple hypothesis testing was provided for comparison where relevant. The calculations were done with custom scripts in the R computing language (http://www.r-project.org).

Data and Materials Availability

Tables of the data are included in supplementary data. Cloud pipeline for RNA MuTect is available at https://github.com/CodingBash/rna_mutect_cloud. Cloud pipeline for superFreq is available at https://github.com/emauryg/superFreq_cloud. Other materials, including analysis scripts, are available through the authors upon reasonable request.

Further methodologic descriptions can be found in the Supplementary Information.

J. Ganz reports grants from American Brain Tumor Association and grants from NCI-SPORE during the conduct of the study. B. Becerra reports grants from 5T32HG002295-18 outside the submitted work. R.N. Doan reports a patent for Cost-Effective Detection of Low Frequency Genetic Variation pending. K.L. Ligon reports grants from the NIH during the conduct of the study. C.A. Walsh reports grants from Howard Hughes Medical Institute, grants from the NIH, and grants from Paul G. Allen Frontiers Program during the conduct of the study; personal fees from Third Rock Ventures, personal fees from Maze Therapeutics, personal fees from Annals of Neurology, and personal fees from NeuroDevelopments outside the submitted work. No disclosures were reported by the other authors.

J. Ganz: Conceptualization, resources, data curation, software, formal analysis, supervision, funding acquisition, validation, investigation, visualization, methodology, writing–original draft, project administration, writing–review and editing. E.A. Maury: Conceptualization, resources, data curation, software, formal analysis, supervision, funding acquisition, validation, investigation, visualization, methodology, writing–original draft, project administration, writing–review and editing. B. Becerra: Software, formal analysis, validation, investigation, visualization, methodology, writing–review and editing. S. Bizzotto: Software, formal analysis, validation, investigation, visualization, methodology, writing–review and editing. R.N. Doan: Data curation, software, formal analysis, validation, methodology, writing–review and editing. C.J. Kenny: Resources, methodology. T. Shin: Software, formal analysis, methodology. J. Kim: Software, formal analysis, funding acquisition, methodology, writing–review and editing. Z. Zhou: Software, formal analysis, funding acquisition, methodology, writing–review and editing. K.L. Ligon: Methodology, writing–review and editing. E.A. Lee: Conceptualization, resources, data curation, supervision, investigation, methodology, writing–original draft, writing–review and editing. C.A. Walsh: Conceptualization, resources, supervision, funding acquisition, investigation, methodology, writing–original draft, writing–review and editing.

J. Ganz was supported by a Basic Research Fellowship from the American Brain Tumor Association (BRF1900016) and by the NCI-Brain Cancer SPORE (grant P50CA165962). E.A. Maury is supported by the Harvard/MIT MD-PhD MSTP program (T32GM007753), the Biomedical Informatics and Data Science Training Program (T15LM007092), and the Ruth L. Kirschstein NRSA F31 Fellowship (F31MH124292). B. Becerra is supported by the NHGRI T32 Training Grant (5T32HG002295-18). K.L. Ligon is supported by the NIH (R01CA188228; R01CA215489; P50CA165962), Pediatric Brain Tumor Foundation, and the PLGA Foundation. E.A. Lee is supported by the NIH (K01 AG051791; DP2 AG072437) and Suh Kyungbae Foundation. C.A. Walsh is supported by the NINDS (R01 NS032457), the NIMH (U01 MH106883), and the NIA (R01AG070921). E.A. Lee and C.A. Walsh are supported by the Allen Discovery Center program through The Paul G. Allen Frontiers Group. C.A. Walsh is an investigator for the Howard Hughes Medical Institute. We thank W. Bainter and K. Stafstrom for performing the Ion Torrent sequencing and J. Neil and the University of Maryland Brain Bank for helping with the tissue collection. We thank the donors and their families for their invaluable donations for the advancement of science, as well as the Boston Children's Hospital IDDRC Molecular Genetics Core Facility supported by NIH award U54HD090255 from the National Institute of Child Health and Human Development. We specially thank Diane Shao and Mike Miller for scientific discussion, as well as Sean Hill, Michael Lodato, and Sonia Kim for valuable technical help and discussion. We also thank Nenad Sestan, Christoffer Flensburg, Hunter Fraser, Gad Getz, and Keren Yizhak and collaborators for facilitating access to their data sets and methodologic advice. Illustrations were created with BioRender.com.

The publication costs of this article were defrayed in part by the payment of publication fees. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

Note: Supplementary data for this article are available at Cancer Discovery Online (http://cancerdiscovery.aacrjournals.org/).

1.
Tomasetti
C
,
Vogelstein
B
,
Parmigiani
G
.
Half or more of the somatic mutations in cancers of self-renewing tissues originate prior to tumor initiation
.
Proc Natl Acad Sci U S A
2013
;
110
:
1999
2004
.
2.
Martincorena
I
,
Roshan
A
,
Gerstung
M
,
Ellis
P
,
Van Loo
P
,
McLaren
S
et al
.
Tumor evolution: high burden and pervasive positive selection of somatic mutations in normal human skin
.
Science
2015
;
348
:
880
6
.
3.
Genovese
G
,
Kahler
AK
,
Handsaker
RE
,
Lindberg
J
,
Rose
SA
,
Bakhoum
SF
et al
.
Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence
.
N Engl J Med
2014
;
371
:
2477
87
.
4.
Jaiswal
S
,
Fontanillas
P
,
Flannick
J
,
Manning
A
,
Grauman
PV
,
Mar
BG
et al
.
Age-related clonal hematopoiesis associated with adverse outcomes
.
N Engl J Med
2014
;
371
:
2488
98
.
5.
Moore
L
,
Leongamornlert
D
,
Coorens
THH
,
Sanders
MA
,
Ellis
P
,
Dentro
SC
et al
.
The mutational landscape of normal human endometrial epithelium
.
Nature
2020
;
580
:
640
6
.
6.
Lac
V
,
Nazeran
TM
,
Tessier-Cloutier
B
,
Aguirre-Hernandez
R
,
Albert
A
,
Lum
A
et al
.
Oncogenic mutations in histologically normal endometrium: the new normal?
J Pathol
2019
;
249
:
173
81
.
7.
Yokoyama
A
,
Kakiuchi
N
,
Yoshizato
T
,
Nannya
Y
,
Suzuki
H
,
Takeuchi
Y
et al
.
Age-related remodelling of oesophageal epithelia by mutated cancer drivers
.
Nature
2019
;
565
:
312
7
.
8.
Martincorena
I
,
Fowler
JC
,
Wabik
A
,
Lawson
ARJ
,
Abascal
F
,
Hall
MWJ
et al
.
Somatic mutant clones colonize the human esophagus with age
.
Science
2018
;
362
:
911
7
.
9.
Lee-Six
H
,
Olafsson
S
,
Ellis
P
,
Osborne
RJ
,
Sanders
MA
,
Moore
L
et al
.
The landscape of somatic mutation in normal colorectal epithelial cells
.
Nature
2019
;
574
:
532
7
.
10.
Lawson
ARJ
,
Abascal
F
,
Coorens
THH
,
Hooks
Y
,
O'Neill
L
,
Latimer
C
et al
.
Extensive heterogeneity in somatic mutation and selection in the human bladder
.
Science
2020
;
370
:
75
82
.
11.
Brunner
SF
,
Roberts
ND
,
Wylie
LA
,
Moore
L
,
Aitken
SJ
,
Davies
SE
et al
.
Somatic mutations and clonal dynamics in healthy and cirrhotic human liver
.
Nature
2019
;
574
:
538
42
.
12.
Keogh
MJ
,
Wei
W
,
Aryaman
J
,
Walker
L
,
van den Ameele
J
,
Coxhead
J
et al
.
High prevalence of focal and multi-focal somatic genetic variants in the human brain
.
Nat Commun
2018
;
9
:
4257
.
13.
Laug
D
,
Glasgow
SM
,
Deneen
B
.
A glial blueprint for gliomagenesis
.
Nat Rev Neurosci
2018
;
19
:
393
403
.
14.
Bohman
LE
,
Swanson
KR
,
Moore
JL
,
Rockne
R
,
Mandigo
C
,
Hankinson
T
et al
.
Magnetic resonance imaging characteristics of glioblastoma multiforme: implications for understanding glioma ontogeny
.
Neurosurgery
2010
;
67
:
1319
27
;
discussion 27–8
.
15.
von Bartheld
CS
,
Bahney
J
,
Herculano-Houzel
S
.
The search for true numbers of neurons and glial cells in the human brain: a review of 150 years of cell counting
.
J Comp Neurol
2016
;
524
:
3865
95
.
16.
Yeung
MS
,
Zdunek
S
,
Bergmann
O
,
Bernard
S
,
Salehpour
M
,
Alkass
K
et al
.
Dynamics of oligodendrocyte generation and myelination in the human brain
.
Cell
2014
;
159
:
766
74
.
17.
Ge
WP
,
Jia
JM
.
Local production of astrocytes in the cerebral cortex
.
Neuroscience
2016
;
323
:
3
9
.
18.
Alexandrov
LB
,
Jones
PH
,
Wedge
DC
,
Sale
JE
,
Campbell
PJ
,
Nik-Zainal
S
et al
.
Clock-like mutational processes in human somatic cells
.
Nat Genet
2015
;
47
:
1402
7
.
19.
Tomasetti
C
,
Li
L
,
Vogelstein
B
.
Stem cell divisions, somatic mutations, cancer etiology, and cancer prevention
.
Science
2017
;
355
:
1330
4
.
20.
Consortium
GT
.
The GTEx Consortium atlas of genetic regulatory effects across human tissues
.
Science
2020
;
369
:
1318
30
.
21.
Werling
DM
,
Pochareddy
S
,
Choi
J
,
An
JY
,
Sheppard
B
,
Peng
M
et al
.
Whole-genome and RNA sequencing reveal variation and transcriptomic coordination in the developing human prefrontal cortex
.
Cell Rep
2020
;
31
:
107489
.
22.
Yizhak
K
,
Aguet
F
,
Kim
J
,
Hess
JM
,
Kubler
K
,
Grimsby
J
et al
.
RNA sequence analysis reveals macroscopic somatic clonal expansion across normal tissues
.
Science
2019
;
364
:
eaaw0726
.
23.
Hardenbol
P
,
Baner
J
,
Jain
M
,
Nilsson
M
,
Namsaraev
EA
,
Karlin-Neumann
GA
et al
.
Multiplexed genotyping with sequence-tagged molecular inversion probes
.
Nat Biotechnol
2003
;
21
:
673
8
.
24.
Ostrom
QT
,
Gittleman
H
,
Liao
P
,
Vecchione-Koval
T
,
Wolinsky
Y
,
Kruchko
C
et al
.
CBTRUS Statistical Report: primary brain and other central nervous system tumors diagnosed in the United States in 2010–2014
.
Neuro Oncol
2017
;
19
:
v1
v88
.
25.
Gonzalez-Perez
A
,
Perez-Llamas
C
,
Deu-Pons
J
,
Tamborero
D
,
Schroeder
MP
,
Jene-Sanz
A
et al
.
IntOGen-mutations identifies cancer drivers across tumor types
.
Nat Methods
2013
;
10
:
1081
2
.
26.
Doan
RN
,
Miller
MB
,
Kim
SN
,
Rodin
RE
,
Ganz
J
,
Bizzotto
S
et al
.
MIPP-Seq: ultra-sensitive rapid detection and validation of low-frequency mosaic mutations
.
BMC Med Genet
2021
;
14
:
47
.
27.
Doan
RN
,
Lim
ET
,
De Rubeis
S
,
Betancur
C
,
Cutler
DJ
,
Chiocchetti
AG
et al
.
Recessive gene disruptions in autism spectrum disorder
.
Nat Genet
2019
;
51
:
1092
8
.
28.
Chakravarty
D
,
Gao
J
,
Phillips
S
,
Kundra
R
,
Zhang
H
,
Wang
J
et al
.
OncoKB: a precision oncology knowledge base
.
JCO Precis Oncol
2017
:
1
16
.
29.
Tischfield
JA
.
Loss of heterozygosity or: how I learned to stop worrying and love mitotic recombination
.
Am J Hum Genet
1997
;
61
:
995
9
.
30.
Yan
H
,
Parsons
DW
,
Jin
G
,
McLendon
R
,
Rasheed
BA
,
Yuan
W
et al
.
IDH1 and IDH2 mutations in gliomas
.
N Engl J Med
2009
;
360
:
765
73
.
31.
Avellaneda Matteo
D
,
Grunseth
AJ
,
Gonzalez
ER
,
Anselmo
SL
,
Kennedy
MA
,
Moman
P
et al
.
Molecular mechanisms of isocitrate dehydrogenase 1 (IDH1) mutations identified in tumors: the role of size and hydrophobicity at residue 132 on catalytic efficiency
.
J Biol Chem
2017
;
292
:
7971
83
.
32.
Gupta
R
,
Flanagan
S
,
Li
CC
,
Lee
M
,
Shivalingham
B
,
Maleki
S
et al
.
Expanding the spectrum of IDH1 mutations in gliomas
.
Mod Pathol
2013
;
26
:
619
25
.
33.
Lodato
MA
,
Woodworth
MB
,
Lee
S
,
Evrony
GD
,
Mehta
BK
,
Karger
A
et al
.
Somatic mutation in single human neurons tracks developmental and transcriptional history
.
Science
2015
;
350
:
94
8
.
34.
Koivunen
P
,
Lee
S
,
Duncan
CG
,
Lopez
G
,
Lu
G
,
Ramkissoon
S
et al
.
Transformation by the (R)-enantiomer of 2-hydroxyglutarate linked to EGLN activation
.
Nature
2012
;
483
:
484
8
.
35.
Turcan
S
,
Rohle
D
,
Goenka
A
,
Walsh
LA
,
Fang
F
,
Yilmaz
E
et al
.
IDH1 mutation is sufficient to establish the glioma hypermethylator phenotype
.
Nature
2012
;
483
:
479
83
.
36.
Gutmann
DH
,
Loehr
A
,
Zhang
Y
,
Kim
J
,
Henkemeyer
M
,
Cashen
A
.
Haploinsufficiency for the neurofibromatosis 1 (NF1) tumor suppressor results in increased astrocyte proliferation
.
Oncogene
1999
;
18
:
4450
9
.
37.
Liu
KW
,
Feng
H
,
Bachoo
R
,
Kazlauskas
A
,
Smith
EM
,
Symes
K
et al
.
SHP-2/PTPN11 mediates gliomagenesis driven by PDGFRA and INK4A/ARF aberrations in mice and humans
.
J Clin Invest
2011
;
121
:
905
17
.
38.
Endersby
R
,
Baker
SJ
.
PTEN signaling in brain: neuropathology and tumorigenesis
.
Oncogene
2008
;
27
:
5416
30
.
39.
Flensburg
C
,
Oshlack
A
,
Majewski
IJ
.
Detecting copy number alterations in RNA-Seq using SuperFreq
.
Bioinformatics
2021
Jun 16 [Epub ahead of print]
.
40.
von Deimling
A
,
Fimmers
R
,
Schmidt
MC
,
Bender
B
,
Fassbender
F
,
Nagel
J
et al
.
Comprehensive allelotype and genetic analysis of 466 human nervous system tumors
.
J Neuropathol Exp Neurol
2000
;
59
:
544
58
.
41.
Hasselblatt
M
,
Isken
S
,
Linge
A
,
Eikmeier
K
,
Jeibmann
A
,
Oyen
F
et al
.
High-resolution genomic analysis suggests the absence of recurrent genomic alterations other than SMARCB1 aberrations in atypical teratoid/rhabdoid tumors
.
Genes Chromosomes Cancer
2013
;
52
:
185
90
.
42.
Cooper
GM
,
Stone
EA
,
Asimenos
G
,
Program
NCS
,
Green
ED
,
Batzoglou
S
et al
.
Distribution and intensity of constraint in mammalian genomic sequence
.
Genome Res
2005
;
15
:
901
13
.
43.
Tatevossian
RG
,
Tang
B
,
Dalton
J
,
Forshew
T
,
Lawson
AR
,
Ma
J
et al
.
MYB upregulation and genetic aberrations in a subset of pediatric low-grade gliomas
.
Acta Neuropathol
2010
;
120
:
731
43
.
44.
Jeuken
JW
,
Sprenger
SH
,
Boerman
RH
,
von Deimling
A
,
Teepen
HL
,
van Overbeeke
JJ
et al
.
Subtyping of oligo-astrocytic tumours by comparative genomic hybridization
.
J Pathol
2001
;
194
:
81
7
.
45.
Muyas
F
,
Zapata
L
,
Guigo
R
,
Ossowski
S
.
The rate and spectrum of mosaic mutations during embryogenesis revealed by RNA sequencing of 49 tissues
.
Genome Med
2020
;
12
:
49
.
46.
Arntfield
ME
,
van der Kooy
D
.
Beta-cell evolution: how the pancreas borrowed from the brain: the shared toolbox of genes expressed by neural and pancreatic endocrine cells may reflect their evolutionary relationship
.
Bioessays
2011
;
33
:
582
7
.
47.
Garcia-Nieto
PE
,
Morrison
AJ
,
Fraser
HB
.
The somatic mutation landscape of the human body
.
Genome Biol
2019
;
20
:
298
.
48.
Cai
X
,
Evrony
GD
,
Lehmann
HS
,
Elhosary
PC
,
Mehta
BK
,
Poduri
A
et al
.
Single-cell, genome-wide sequencing identifies clonal somatic copy-number variation in the human brain
.
Cell Rep
2014
;
8
:
1280
9
.
49.
McConnell
MJ
,
Lindberg
MR
,
Brennand
KJ
,
Piper
JC
,
Voet
T
,
Cowing-Zitron
C
et al
.
Mosaic copy number variation in human neurons
.
Science
2013
;
342
:
632
7
.
50.
Chronister
WD
,
Burbulis
IE
,
Wierman
MB
,
Wolpert
MJ
,
Haakenson
MF
,
Smith
ACB
et al
.
Neurons with complex karyotypes are rare in aged human neocortex
.
Cell Rep
2019
;
26
:
825
35
.
51.
Sherman
MA
,
Rodin
RE
,
Genovese
G
,
Dias
C
,
Barton
AR
,
Mukamel
RE
et al
.
Large mosaic copy number variations confer autism risk
.
Nat Neurosci
2021
;
24
:
197
203
.
52.
Huang
W
,
Bhaduri
A
,
Velmeshev
D
,
Wang
S
,
Wang
L
,
Rottkamp
CA
et al
.
Origins and proliferative states of human oligodendrocyte precursor cells
.
Cell
2020
;
182
:
594
608
.
53.
Lee
JH
,
Lee
JE
,
Kahng
JY
,
Kim
SH
,
Park
JS
,
Yoon
SJ
et al
.
Human glioblastoma arises from subventricular zone cells with low-level driver mutations
.
Nature
2018
;
560
:
243
7
.
54.
Assanah
M
,
Lochhead
R
,
Ogden
A
,
Bruce
J
,
Goldman
J
,
Canoll
P
.
Glial progenitors in adult white matter are driven to form malignant gliomas by platelet-derived growth factor-expressing retroviruses
.
J Neurosci
2006
;
26
:
6781
90
.
55.
Zong
H
,
Parada
LF
,
Baker
SJ
.
Cell of origin for malignant gliomas and its implication in therapeutic development
.
Cold Spring Harb Perspect Biol
2015
;
7
:
a020610
.
56.
Suva
ML
,
Tirosh
I
.
The glioma stem cell model in the era of single-cell genomics
.
Cancer Cell
2020
;
37
:
630
6
.
57.
Liu
C
,
Sage
JC
,
Miller
MR
,
Verhaak
RG
,
Hippenmeyer
S
,
Vogel
H
et al
.
Mosaic analysis with double markers reveals tumor cell of origin in glioma
.
Cell
2011
;
146
:
209
21
.
58.
Karczewski
KJ
,
Francioli
LC
,
Tiao
G
,
Cummings
BB
,
Alfoldi
J
,
Wang
Q
et al
.
The mutational constraint spectrum quantified from variation in 141,456 humans
.
Nature
2020
;
581
:
434
43
.
59.
Adzhubei
IA
,
Schmidt
S
,
Peshkin
L
,
Ramensky
VE
,
Gerasimova
A
,
Bork
P
et al
.
A method and server for predicting damaging missense mutations
.
Nat Methods
2010
;
7
:
248
9
.
60.
Schwarz
JM
,
Rodelsperger
C
,
Schuelke
M
,
Seelow
D
.
MutationTaster evaluates disease-causing potential of sequence alterations
.
Nat Methods
2010
;
7
:
575
6
.
61.
Reva
B
,
Antipin
Y
,
Sander
C
.
Predicting the functional impact of protein mutations: application to cancer genomics
.
Nucleic Acids Res
2011
;
39
:
e118
.
62.
Quang
D
,
Chen
Y
,
Xie
X
.
DANN: a deep learning approach for annotating the pathogenicity of genetic variants
.
Bioinformatics
2015
;
31
:
761
3
.
63.
Rentzsch
P
,
Witten
D
,
Cooper
GM
,
Shendure
J
,
Kircher
M
.
CADD: predicting the deleteriousness of variants throughout the human genome
.
Nucleic Acids Res
2019
;
47
:
D886
D94
.
64.
Mayakonda
A
,
Lin
DC
,
Assenov
Y
,
Plass
C
,
Koeffler
HP
.
Maftools: efficient and comprehensive analysis of somatic variants in cancer
.
Genome Res
2018
;
28
:
1747
56
.
65.
Tate
JG
,
Bamford
S
,
Jubb
HC
,
Sondka
Z
,
Beare
DM
,
Bindal
N
et al
.
COSMIC: the catalogue of somatic mutations in cancer
.
Nucleic Acids Res
2019
;
47
:
D941
D7
.
66.
Lee
J
,
Lee
AJ
,
Lee
JK
,
Park
J
,
Kwon
Y
,
Park
S
et al
.
Mutalisk: a web-based somatic MUTation AnaLyIS toolKit for genomic, transcriptional and epigenomic signatures
.
Nucleic Acids Res
2018
;
46
:
W102
W8
.
This open access article is distributed under the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) license.