Next Article in Journal
Treatment Sequences in Patients with Recurrent or Metastatic Head and Neck Squamous Cell Carcinoma: Cetuximab Followed by Immunotherapy or Vice Versa
Next Article in Special Issue
Bias and Class Imbalance in Oncologic Data—Towards Inclusive and Transferrable AI in Large Scale Oncology Data Sets
Previous Article in Journal
Health Anxiety and Its Relationship to Thyroid-Hormone-Suppression Therapy in Patients with Differentiated Thyroid Cancer
Previous Article in Special Issue
A Bioinformatics Evaluation of the Role of Dual-Specificity Tyrosine-Regulated Kinases in Colorectal Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Whole-Genome Sequencing Identifies PPARGC1A as a Putative Modifier of Cancer Risk in BRCA1/2 Mutation Carriers

by
Qianqian Zhu
1,*,†,
Jie Wang
1,†,
Han Yu
1,
Qiang Hu
1,
Nicholas W. Bateman
2,3,
Mark Long
1,
Spencer Rosario
1,
Emily Schultz
1,
Clifton L. Dalgard
4,5,
Matthew D. Wilkerson
4,5,
Gauthaman Sukumar
3,5,
Ruea-Yea Huang
6,
Jasmine Kaur
7,
Shashikant B. Lele
7,
Emese Zsiros
7,
Jeannine Villella
8,
Amit Lugade
6,
Kirsten Moysich
9,
Thomas P. Conrads
2,10,
George L. Maxwell
2,10 and
Kunle Odunsi
6,7,11,12,*
add Show full author list remove Hide full author list
1
Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
2
Gynecologic Cancer Center of Excellence, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University and Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD 20889, USA
3
Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., 6720A Rockledge Dr., Suite 100, Bethesda, MD 20817, USA
4
The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
5
Department of Anatomy Physiology and Genetics, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
6
Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
7
Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
8
Division of Gynecologic Oncology, Lenox Hill Hospital/Northwell Health Cancer Institute, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New York, NY 11549, USA
9
Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
10
Women’s Health Integrated Research Center, Women’s Service Line, Inova Health System, 3289 Woodburn Rd, Annandale, VA 22003, USA
11
Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL 60637, USA
12
University of Chicago Medicine Comprehensive Cancer Center, Chicago, IL 60637, USA
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2022, 14(10), 2350; https://doi.org/10.3390/cancers14102350
Submission received: 18 March 2022 / Revised: 3 May 2022 / Accepted: 6 May 2022 / Published: 10 May 2022
(This article belongs to the Special Issue Bioinformatics, Big Data and Cancer)

Abstract

:

Simple Summary

In search of genetic factors that affect cancer risks in BRCA carriers, we carried out the first whole-genome sequencing study in a unique registry of familial ovarian cancer, selected to enrich with BRCA1/2 carriers. We are the first to survey rare variants, particularly the non-coding variants for BRCA modifier genes and identified PPARGC1A, a master regulator of mitochondrial biogenesis, as a novel putative BRCA modifier. This finding can help improve cancer risk prediction and provide personalized preventive care for BRCA carriers.

Abstract

While BRCA1 and BRCA2 mutations are known to confer the largest risk of breast cancer and ovarian cancer, the incomplete penetrance of the mutations and the substantial variability in age at cancer onset among carriers suggest additional factors modifying the risk of cancer in BRCA1/2 mutation carriers. To identify genetic modifiers of BRCA1/2, we carried out a whole-genome sequencing study of 66 ovarian cancer patients that were enriched with BRCA carriers, followed by validation using data from the Pan-Cancer Analysis of Whole Genomes Consortium. We found PPARGC1A, a master regulator of mitochondrial biogenesis and function, to be highly mutated in BRCA carriers, and patients with both PPARGC1A and BRCA1/2 mutations were diagnosed with breast or ovarian cancer at significantly younger ages, while the mutation status of each gene alone did not significantly associate with age of onset. Our study suggests PPARGC1A as a possible BRCA modifier gene. Upon further validation, this finding can help improve cancer risk prediction and provide personalized preventive care for BRCA carriers.

1. Introduction

BRCA1 and BRCA2 are the two most well-known cancer predisposition genes. Inheritance of a BRCA1 or BRCA2 mutation greatly increases lifetime risk of breast cancer and ovarian cancer [1,2]. While lifetime risk of developing ovarian cancer for women in the general population is about 1.2% [3], the risk was estimated to be 39–59% and 11–17% for women who carry BRCA1 and BRCA2 mutations, respectively, by age 70–80 [4,5,6,7]. Preventive strategies have therefore been implemented to reduce cancer risk in BRCA1/2 carriers [8,9,10]. On the other hand, the penetrance of BRCA1/2 mutations is not complete, and there is substantial variability in age of cancer onset among carriers. These observations support the hypothesis that cancer risks in BRCA1/2 mutation carriers are modified by other factors. Over the past two decades, significant efforts have been invested in search of these modifying factors, among the most influential of which is the Consortium of Investigators of Modifiers of BRCA1/2 (CIMBA) [11]. These efforts resulted in the discovery of a number of risk modifiers for BRCA1/2 mutations, including environmental, reproductive, and genetic factors [12,13,14,15].
To date, studies to identify genetic modifiers of BRCA1/2 were mainly carried out in three ways: candidate gene studies, investigation of specific variants discovered by genome-wide association studies (GWAS) to associate with cancer risks in the general population, and GWAS carried out specifically in BRCA1 or BRCA2 carriers [13,16,17,18,19]. The potential BRCA1/2 genetic modifiers identified in these studies are all common variants with relatively small effect sizes. On the other hand, the contribution of rare variants, which are more likely to have large effect sizes and/or direct functional consequences, in modifying cancer risks of BRCA1/2 carriers has not been systematically explored. Therefore, we employed whole-genome sequencing (WGS) technology to identify genetic modifiers of BRCA1/2 that are driven by rare variants.
To maximize the likelihood of discovering genetic modifiers of BRCA1/2, we performed WGS on a total of 66 ovarian cancer (OC) patients that were enriched with BRCA carriers. A total of 49 of these patients were selected from the Familial Ovarian Cancer Registry (FOCR) [20,21] to have a strong family history of OC. The discovered candidates were evaluated using independent WGS data of 247 ovarian and breast cancer patients from the Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium [22]. To our knowledge, this is the first and largest WGS study of BRCA1/2 modifiers to date, and we report PPARGC1A as a novel putative genetic modifier of ovarian and breast cancer risk for BRCA1/2 carriers.

2. Materials and Methods

2.1. Study Population

A total of 50 hereditary OC patients from 48 families were selected from FOCR (formerly known as the Gilda Familial Ovarian Cancer Registry) for WGS based on DNA availability, prior genetic test results of BRCA1/2, and strong family history. The FOCR housed at Roswell Park Comprehensive Cancer Center (RPCCC) recruits families with two or more cases of OC, families with three or more cases of cancer on the same side of family with at least one being OC, families with at least one female having two or more primary cancers and one of the primaries being OC, and families with two or more cases of cancer with at least one being OC diagnosed at an early age of onset (45 years old or younger) [21]. Families provide written informed consent under an institutional protocol CIC95-27. Cases are verified by medical record and/or death certificate when required, and a registry pathologist verifies stage and histology. The registry comprises 50,401 individuals including 5614 ovarian cancers from 2636 unique families. A total of 27 of the 50 FOCR patients were known carriers of BRCA from prior genetic testing performed by Myriad or inhouse [21]. An additional 18 RPCCC patients with sporadic OC were also included for WGS.

2.2. Whole-Genome Sequencing, Variant Calling, and Variant Filtering

Sequencing library preparation and whole-genome sequencing was performed at The American Genome Center at the Uniformed Services University, Bethesda, MD (Supplementary Methods). The GATK data pre-processing workflow was used to generate analysis-ready alignments (Supplementary Methods). DeepVariant (v0.5) was used to call single nucleotide variants (SNVs) and small insertions and deletions (indels) in standard VCF format for each sample with a convolutional neural network model [23]. Structural variants (SV) were detected using a structural variant calling workflow developed by bcbio, which used an integrative caller MetaSV [24] that combines the results from four separate methods, CNVkit [25], Manta [26], LUMPY [27], and Wham [28]. We only considered SVs longer than 50 bp and required SVs to be detected by at least two of the four methods with ≥3 supporting reads (split read or disconcordant read) in each method. Using the genotypes of SNVs, we performed sample level quality assessment using the Bioconductor package SeqSQC [29]. One FOCR patient and one sporadic OC patient were identified as population outliers and hence were excluded from further analyses.
A series of filters were applied to keep only rare and functional variants in our analysis (Supplementary Methods). Variants in the eight genes with significantly higher mutation rate in BRCA carriers of our WGS discovery cohort (Table 1) were manually inspected to ensure reliable variant calls.

2.3. Statistical Analysis

One-sided Fisher’s exact test was used to test whether the gene mutation frequency is higher in BRCA carriers than non-carriers.

2.4. Network Propagation and Pathway Enrichment Analysis

Using HotNet2 [30] and the Reactome functional interaction networks [31], we applied a network propagation analysis on the −log10 scores of the one-sided Fisher’s exact test p-values calculated by comparing gene mutation frequencies between BRCA carriers and non-carriers within the 49 hereditary OC patients. Only genes with Fisher’s exact test p-values ≤ 0.6 were included in the analysis. The statistical significance of the identified sub-networks was based on the number and size of the identified sub-networks compared to those found using a permutation test. We used 100 permutations and a minimum network size of 2 for statistical testing.
To examine the biological functions of each significant gene sub-network, pathway enrichment analysis of genes in each sub-network was performed using hypergeometric testing based on the Reactome pathway database [31]. Multiple testing was corrected using the Benjamini–Hochberg method. For each sub-network, pathways with adjusted p-values < 0.05 were considered significantly enriched.

2.5. Validation Using PCAWG Breast and Ovarian Cancer Cohorts

We obtained germline genetic variants called by PCAWG and kept only breast and ovarian cancer patients that were of European ancestry. Sample level quality assessment was performed using the Bioconductor package SeqSQC [29], which resulted in the removal of nine problematic samples. Germline variants of the remaining PCAWG samples were filtered and annotated in the same way as described above for the germline variants in our WGS data. We kept only the variants whose target genes were BRCA1, BRCA2, PPARGC1A, and PBX1 in our analysis.

2.6. Study Approval

The study was approved by Institutional Review Boards. All participants provided written informed consent.

3. Results

3.1. Study Population in the Discovery Stage

Our discovery WGS cohort consisted of 49 OC patients from the FOCR (formerly known as the Gilda Familial Ovarian Cancer Registry) with a strong family history of OC [20,21] and 17 sporadic OC patients (Figure 1, Tables S1–S3); all were of European descent. Among the 49 hereditary OC patients, 27 were known BRCA carriers from prior genetic testing and were purposely included to enrich for BRCA carriers in our discovery cohort (Materials and Methods).

3.2. Discovery of Candidate Genes That Modify Cancer Risks of BRCA1/2

The germline genetic variants were detected from the discovery WGS data using a deep learning variant calling algorithm [23], which has been shown to achieve higher sensitivity and specificity in pathogenic variant detection than standard methods [32]. A series of stringent variant filtering steps were carried out to retain only rare (MAF < 0.5% in European population) and functional variants for subsequent analyses (Materials and Methods). The 27 known BRCA carriers were confirmed to carry BRCA1/2 mutations from our WGS analysis, and 12 more patients in the discovery cohort were found to be BRCA carriers, including 9 hereditary OC patients and 3 sporadic OC patients.
To identify BRCA modifiers that increase cancer risks in BRCA carriers, we utilized three different approaches. First, we compared mutation frequency of each gene between BRCA carriers and non-carriers in our discovery cohort using Fisher’s exact test and focused on genes that are more frequently mutated in BRCA carriers (Table 1 and Table S4), requiring uncorrected Fisher’s exact test p-value ≤ 0.05 in consideration of our relatively small sample size. In addition, we also required the genes to have significantly higher mutation frequency in BRCA carriers in the analysis of the 49 hereditary OC patients, assuming the effect of BRCA modifier is most enriched in cancer patients with family history. Eight genes satisfied both criteria, including BRCA1/2 and a known cancer gene PBX1.
Table 1. Comparison of gene mutation frequency between BRCA carriers and non-carriers in the discovery cohort.
Table 1. Comparison of gene mutation frequency between BRCA carriers and non-carriers in the discovery cohort.
GeneHereditary OC
(36 BRCA Carriers vs. 13 Non-Carriers)
Hereditary OC + Sporadic OC
(39 BRCA Carriers vs. 27 Non-Carriers)
# and Fraction of Mutated BRCA Carriers# and Fraction of Mutated
Non-Carriers
p-Value *# and Fraction of Mutated BRCA Carriers# and Fraction of Mutated
Non-Carriers
p-Value *
BRCA1290.8100.002.95 × 10−7300.7700.003.84 × 10−11
BRCA2100.2800.003.09 × 10−2120.3100.007.94 × 10−4
PPARGC1A110.3100.002.06 × 10−2110.2810.049.99 × 10−3
PBX1140.3910.083.49 × 10−2140.3630.112.15 × 10−2
LMNTD190.2500.004.58 × 10−290.2300.005.73 × 10−3
AHDC190.2500.004.58 × 10−290.2310.043.01 × 10−2
MADD90.2500.004.58 × 10−290.2310.043.01 × 10−2
TRERF190.2500.004.58 × 10−2100.2610.041.75 × 10−2
* Raw p-value from one-sided Fisher’s exact test (Ha: gene mutation frequency in BRCA carriers ≥ the frequency in non-carriers).
Next, we adopted a network-based approach to identify the pathways that are altered in BRCA carriers based on the hypothesis that malfunction of certain biological processes increases cancer risk in BRCA carriers, and within those processes, multiple genes instead of a unique gene can be targeted by germline genetic mutations. Specifically, we mapped genes that showed elevated mutation rates in BRCA carriers within the hereditary OC cohort onto Reactome functional interaction networks [31] and used the network propagation method HotNet2 [30] to detect sub-networks that contain multiple contributing neighboring genes (Materials and Methods). A total of 15 significant sub-networks were identified (HotNet2 permutation p-value = 0.01). The largest sub-network contained BRCA1/2 and 45 other genes (Figure 2 and Figure S1). The biological pathways that were significantly enriched in this sub-network included transcriptional regulation of white adipocyte differentiation, transcriptional regulation by Notch3 and Notch1, circadian clock, transcriptional activation of mitochondrial biogenesis, and SUMOylation (Table S5). Assuming the BRCA modifiers alter the same biological processes as BRCA1/2, we focused on the 47 genes within the same sub-network as BRCA1/2 in further analysis.
Finally, under our prior hypothesis that any BRCA modifier gene would be more highly mutated in BRCA carriers, we would expect the expression of any such modifier gene to differ between BRCA carriers and non-carriers, assuming the genetic mutation affects its gene expression. Therefore, we utilized both RNAseq and whole-exome sequencing (WES) data from The Cancer Genome Atlas (TCGA) Pan-Cancer analysis project to detect differentially expressed genes (DEGs) between BRCA carriers and non-carriers in breast or ovarian cancer (Supplementary Methods). A total of 4600 DEGs were identified in breast cancer by comparing 15 BRCA carriers with 416 non-carriers, while 189 DEGs were identified in OC by comparing 16 BRCA carriers with 63 non-carriers. Among these DEGs, only one, PPARGC1A, was identified from our WGS analysis as more highly mutated in BRCA carriers than non-carriers. It was significantly up-regulated in BRCA carriers of breast cancer (log2 fold change (carriers vs. non-carriers) = 1.86, adjusted p = 6.10 × 10−3).

3.3. Independent Validation of BRCA Modifier Candidate Genes

Using the three different approaches described above, we found two potential BRCA modifier candidates, PPARGC1A and PBX1, which were identified by three and two approaches, respectively, (Figure 1). We then sought to validate these two genes using independent WGS cohorts of breast and ovarian cancer from the Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium [22], consisting of both the TCGA and the International Cancer Genome Consortium (ICGC). Among the 156 and 91 breast cancer and OC patients from PCAWG, only 17% and 19% were BRCA carriers, respectively, which was similar to the value in the sporadic OC cases of our discovery cohort but much lower than in the hereditary OC cases of our discovery cohort, where 73% were BRCA carriers.
In PCAWG’s breast cancer cohort, we observed a trend of higher PPARGC1A mutation frequency in BRCA carriers than non-carriers (Figure 3a, Table S6). This trend was also found in PCAWG’s ovarian cancer cases from TCGA, but not in those from ICGC (Table S6). When combining the discovery and validation cohorts, the PPARGC1A gene in BRCA carriers possessed more deleterious mutations than in non-carriers with borderline significance (p = 0.055, Figure 3b). The other candidate gene, PBX1, did not show a higher mutation rate in BRCA carriers in either PCAWG breast or ovarian cancer cohort (Table S7).

3.4. PPARGC1A as a Novel BRCA Modifier Candidate Gene

We then investigated whether PPARGC1A mutations affect age of onset of breast and ovarian cancer in the PCAWG patients. We found that the interaction between PPARGC1A mutation status and BRCA status significantly associated with an earlier age of cancer onset (p = 0.03), while the main effect of each gene alone was not significant (p = 0.33 and 0.96, respectively, for PPARGC1A status and BRCA status). Patients who carried both BRCA1/2 mutations and PPARGC1A mutations were diagnosed with breast or ovarian cancer at a significantly younger age (Figure 4). The median age of onset was 48, 55.5, 60.5, and 58 respectively for patients carrying mutations in both PPARGC1A and BRCA1/2 genes, in BRCA1/2 genes only, in PPARGC1A only, or in none of the three genes. The interaction term remained significant when restricting on breast cancer patients, but not in a regression model on the smaller cohort of ovarian cancer patients, where only one patient carried both BRCA and PPARGC1A mutations (Table S8). Consistent patterns were observed when BRCA1 and BRCA2 status were included in the regression model (Table S9).
The majority of the PPARGC1A mutations we identified in both the discovery and the validation cohorts were non-coding variants (Figure 5). To investigate how these non-coding variants affect PPARGC1A expression, we compared PPARGC1A expression between the carriers of non-coding variants and the non-carriers using PCAWG RNA-seq data [33] and observed a trend of lower expression in the carriers (p = 0.09 and 0.44 for ovarian cancer and breast cancer, respectively, Figure S2).

4. Discussion

In this first WGS study to discover putative BRCA genetic modifiers, we performed WGS on 66 OC patients that were enriched with BRCA carriers and identified two genes, PPARGC1A and PBX1, to be highly mutated in BRCA carriers and within the same gene subnetwork with BRCA1/2. In addition, PPARGC1A was found to be differentially expressed between BRCA carriers and non-carriers within the TCGA breast cancer cohort. Our independent validation in PCAWG observed a similar trend of a higher PPARGC1A mutation rate in BRCA carriers than non-carriers for patients with breast cancer. Importantly, we found that patients with both PPARGC1A and BRCA1/2 mutations were diagnosed with breast or ovarian cancer at a significantly younger age, while the effect of each gene alone was not significant. Therefore, our results suggest PPARGC1A to be a potential new BRCA modifier. While previous candidate gene studies of common genetic variants linked PPARGC1A with ovarian cancer and familial breast cancer risk [34,35], our WGS study, focusing on rare and functional variants, was the first to reveal the effect of PPARGC1A in the context of BRCA1/2 mutation. The PPARGC1A mutations we identified through WGS were dominantly non-coding variants, which highlights the importance of going beyond just the gene coding regions in search of BRCA modifiers and cancer predisposition genes. It is worth noting that the pathways involving PPARGC1A, such as transcriptional regulation of white adipocyte differentiation and transcriptional activation of mitochondrial biogenesis, were found to be disturbed in BRCA carriers (Table S3). Future studies targeting these pathways may allow identification of additional BRCA modifiers.
PPARGC1A, also known as Peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), is a coactivator of PPARγ, which is a crucial gene regulating BRCA1 gene expression [36]. In addition, PPARGC1A is a master regulator of mitochondrial biogenesis and function. It is essential for cancer cells to rapidly adapt to energy-demanding situations. Both increased and decreased PPARGC1A expression have been reported in a range of cancer types and associated with a worse prognosis [37,38,39]. These contradicting observations are now thought to result from cancer cells exploiting PPARGC1A to provide them metabolic plasticity to support their evolving needs along the course of cancer development [37,38,40,41]. During early tumorigenesis, PPARGC1A may be downregulated to facilitate the increased consumption of glucose and glutamine in cancer cells [37,40]. This is consistent with our findings of significantly earlier cancer development in carriers with mutations in both PPARGC1A and BRCA1/2, as well as lower PPARGC1A expression in the PPARGC1A mutation carriers. On the other hand, a recent study demonstrated inhibition of PPARGC1A in tumor infiltrating T cells leads to T cell exhaustion and tumor immune escape [42]. BRCA1 and BRCA2 are key homologous recombination genes. Defects in BRCA1/2 result in tumors with extensive genomic instability that stimulates inflammatory signaling [43,44,45,46,47,48,49]. Therefore, cancer cells that are genomically unstable must evolve to escape immune surveillance in order to avoid being cleared by the immune system [43]. Because tumor-infiltrating T cells in PPARGC1A mutation carriers experience PPARGC1A inhibition and T cell exhaustion due to metabolic insufficiency [42], BRCA1/2-mutant cancer cells in PPARGC1A mutation carriers have an advantage in escaping immune surveillance and thus can develop into tumors earlier than in individuals without PPARGC1A mutations. In summary, loss of PPARGC1A due to germline PPARGC1A variants might stimulate tumor development by giving tumor cells a metabolic advantage or weakening immune surveillance, or both.
A major strength of our study is the enrichment for BRCA carriers from a familial ovarian cancer registry. However, there are limitations to our study. While we have leveraged the largest publicly available WGS collection of breast and ovarian cancer patients from PCAWG, our validation study is limited by the relatively small sample size. Furthermore, the validation power is reduced due to over-representation of sporadic cancer and the small number of BRCA carriers in these publicly available WGS cohorts. A future sequencing study of the entire PPARGC1A locus in large ovarian and breast cancer cohorts, particularly in cancer patients with family history, is warranted to further validate our finding of PPARGC1A as a BRCA1/2 genetic modifier. Knocking out PPARGC1A in breast or ovarian cancer mouse models with mutated BRCA1/2 [50,51,52] will also help to investigate the effect of PPARGC1A in increasing cancer risk in the context of BRCA1/2 mutations and to reveal the underlying biological mechanism.

5. Conclusions

We conducted the first WGS study of hereditary OC patients enriched with BRCA carriers and followed with a validation study using the largest WGS collection of OC and BC patients to date to identify PPARGC1A as a possible BRCA modifier gene. Given the impact of PPARGC1A on the age of onset of OC and BC among BRCA mutation carriers, our results could have significant implications for cancer risk prediction and personalized preventive care for BRCA carriers. Future follow-up studies including additional sequencing and functional experiments are warranted to confirm these findings.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/cancers14102350/s1, Supplementary Methods; Figure S1: The remaining 14 gene sub-networks significantly altered in BRCA carriers. Genes that were significantly highly mutated in BRCA carriers (Table 1) are highlighted by red underscores.; Figure S2: PPARGC1A expression between carriers of non-coding mutations and non-carriers in PCAWG. Gene expression levels (FPKM-UQ) were estimated using the FPKM metric, based on alignments from the TopHat and STAR algorithms and normalized with the Upper Quartile method. FPKM, fragments per kilobase of transcript per million mapped reads. p-values were calculated using one-sided Wilcoxon rank sum test between 11 carriers and 41 non-carriers in breast cancer and between 7 carriers and 60 non-carriers in ovarian cancer; Table S1: Characteristics of the discovery cohort; Table S2: The sequencing coverage and quality statistics of whole-genome-sequenced FOCR patients; Table S3: The sequencing coverage and quality statistics of whole-genome sequenced sporadic OC patients; Table S4: The PPARGC1A variants included in our analyses; Table S5: The pathways significantly enriched in the genes within the largest gene sub-network significantly altered in BRCA carriers; Table S6: Comparison of PPARGC1A mutation frequency between BRCA carriers and non-carriers in the validation cohorts; Table S7: Comparison of PBX1 mutation frequency between BRCA carriers and non-carriers in the validation cohorts; Table S8: Linear regression between cancer age onset and gene mutation status (BRCA, PPARGC1A) in PCAWG; Table S9: Linear regression between cancer age onset and gene mutation status (BRCA1, BRCA2, PPARGC1A) in PCAWG. (References [53,54,55,56,57,58,59,60,61,62,63,64] cited in the supplementary materials)

Author Contributions

Q.Z. and K.O. conceived the study and wrote the manuscript; N.W.B., C.L.D., M.D.W., G.S., T.P.C. and G.L.M. generated the FOCR WGS data; Q.Z., J.W., H.Y., Q.H., M.L., S.R. and E.S. performed data analyses; R.-Y.H., J.K., S.B.L., E.Z., J.V., A.L., K.M. and K.O. coordinated samples and clinical data; Q.Z., J.W., H.Y., M.L., S.R. and K.O. interpreted data. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Institutes of Health under award numbers U24CA232979, P50CA159981 and P30CA016056 as well as the American Cancer Society Research Scholar Grant RSG-14-214-01-TBE and the Roswell Park Alliance Foundation.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki and approved by the Institutional Review Board of Roswell Park Comprehensive Cancer Center (protocol I215512).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

Data are restricted due to ethical concerns in keeping with the institute’s policies on germline variation data and the level of patient consent gained. Data are available from the Familial Ovarian Cancer Registry (ovarianregistry@roswellpark.org) for researchers who meet the criteria for access to confidential data. The results published here are in part based upon data generated by The Cancer Genome Atlas (dbGaP Study Accession: phs000178.v10.p8) managed by the NCI and NHGRI. Information about TCGA can be found at http://cancergenome.nih.gov.

Acknowledgments

The authors would like to thank and acknowledge the Familial Ovarian Cancer Registry participants, donors and past researchers.

Conflicts of Interest

K.O. is a co-founder of Tactiva Therapeutics and receives research support from AstraZeneca and Tesaro. T.P.C. is a scientific advisory board member for ThermoFisher Scientific, Inc. G.L.M. is a consultant for Kiyatec, GSK, and Merck. The other authors have declared no conflict of interest exists. The contents of this publication are the sole responsibility of the author(s) and do not necessarily reflect the views, opinions or policies of Uniformed Services University of the Health Sciences (USUHS), the Department of Defense (DoD), The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., or the U.S. Government.

References

  1. Miki, Y.; Swensen, J.; Shattuck-Eidens, D.; Futreal, P.; Harshman, K.; Tavtigian, S.; Liu, Q.; Cochran, C.; Bennett, L.; Ding, W.; et al. A strong candidate for the breast and ovarian cancer susceptibility gene BRCA1. Science 1994, 266, 66–71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Wooster, R.; Bignell, G.; Lancaster, J.; Swift, S.; Seal, S.; Mangion, J.; Collins, N.; Gregory, S.; Gumbs, C.; Micklem, G. Identification of the breast cancer susceptibility gene BRCA2. Nature 1995, 378, 789–792. [Google Scholar] [CrossRef] [PubMed]
  3. Howlader, N.; Noone, A.M.; Krapcho, M.; Miller, D.; Brest, A.; Yu, M.; Ruhl, J.; Tatalovich, Z.; Mariotto, A.; Lewis, D.R.; et al. SEER Cancer Statistics Review, 1975–2017; National Cancer Institute: Bethesda, MD, USA, 2020. [Google Scholar]
  4. Kuchenbaecker, K.B.; Hopper, J.L.; Barnes, D.R.; Phillips, K.A.; Mooij, T.M.; Roos-Blom, M.J.; Jervis, S.; van Leeuwen, F.E.; Milne, R.L.; Andrieu, N.; et al. Risks of Breast, Ovarian, and Contralateral Breast Cancer for BRCA1 and BRCA2 Mutation Carriers. JAMA 2017, 317, 2402–2416. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Antoniou, A.; Pharoah, P.D.; Narod, S.; Risch, H.A.; Eyfjord, J.E.; Hopper, J.L.; Loman, N.; Olsson, H.; Johannsson, O.; Borg, A.; et al. Average risks of breast and ovarian cancer associated with BRCA1 or BRCA2 mutations detected in case Series unselected for family history: A combined analysis of 22 studies. Am. J. Hum. Genet. 2003, 72, 1117–1130. [Google Scholar] [CrossRef] [Green Version]
  6. Chen, S.; Parmigiani, G. Meta-analysis of BRCA1 and BRCA2 penetrance. J. Clin. Oncol. 2007, 25, 1329–1333. [Google Scholar] [CrossRef] [Green Version]
  7. Mavaddat, N.; Peock, S.; Frost, D.; Ellis, S.; Platte, R.; Fineberg, E.; Evans, D.G.; Izatt, L.; Eeles, R.A.; Adlard, J.; et al. Cancer Risks for BRCA1 and BRCA2 Mutation Carriers: Results from Prospective Analysis of EMBRACE. J. Natl. Cancer Inst. 2013, 105, 812–822. [Google Scholar] [CrossRef] [Green Version]
  8. LeVasseur, N.; Chia, S. Cancer screening and prevention in BRCA mutation carriers: A missed opportunity? Br. J. Cancer 2019, 121, 1–2. [Google Scholar] [CrossRef] [Green Version]
  9. Daly, M.B.; Pilarski, R.; Yurgelun, M.B.; Berry, M.P.; Buys, S.S.; Dickson, P.; Domchek, S.M.; Elkhanany, A.; Friedman, S.; Garber, J.E.; et al. NCCN Guidelines Insights: Genetic/Familial High-Risk Assessment: Breast, Ovarian, and Pancreatic, Version 1.2020. J. Natl. Compr. Cancer Netw. 2020, 18, 380–391. [Google Scholar] [CrossRef]
  10. Paluch-Shimon, S.; Cardoso, F.; Sessa, C.; Balmana, J.; Cardoso, M.J.; Gilbert, F.; Senkus, E. Prevention and screening in BRCA mutation carriers and other breast/ovarian hereditary cancer syndromes: ESMO Clinical Practice Guidelines for cancer prevention and screening. Ann. Oncol. 2016, 27, v103–v110. [Google Scholar] [CrossRef]
  11. Chenevix-Trench, G.; Milne, R.L.; Antoniou, A.C.; Couch, F.J.; Easton, D.F.; Goldgar, D.E. An international initiative to identify genetic modifiers of cancer risk in BRCA1 and BRCA2 mutation carriers: The Consortium of Investigators of Modifiers of BRCA1 and BRCA2 (CIMBA). Breast Cancer Res. 2007, 9, 104. [Google Scholar] [CrossRef] [Green Version]
  12. Friebel, T.M.; Domchek, S.M.; Rebbeck, T.R. Modifiers of Cancer Risk in BRCA1 and BRCA2 Mutation Carriers: A Systematic Review and Meta-Analysis. J. Natl. Cancer Inst. 2014, 106, dju091. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Milne, R.L.; Antoniou, A.C. Genetic modifiers of cancer risk for BRCA1 and BRCA2 mutation carriers. Ann. Oncol. 2011, 22 (Suppl. S1), i11–i17. [Google Scholar] [CrossRef] [PubMed]
  14. Kotsopoulos, J.; Lubinski, J.; Lynch, H.T.; Kim-Sing, C.; Neuhausen, S.; Demsky, R.; Foulkes, W.D.; Ghadirian, P.; Tung, N.; Ainsworth, P.; et al. Oophorectomy after menopause and the risk of breast cancer in BRCA1 and BRCA2 mutation carriers. Cancer Epidemiol. Biomark. Prev. 2012, 21, 1089–1096. [Google Scholar] [CrossRef] [Green Version]
  15. Narod, S.A. Modifiers of risk of hereditary breast and ovarian cancer. Nat. Rev. Cancer 2002, 2, 113–123. [Google Scholar] [CrossRef] [PubMed]
  16. Couch, F.J.; Wang, X.; McGuffog, L.; Lee, A.; Olswold, C.; Kuchenbaecker, K.B.; Soucy, P.; Fredericksen, Z.; Barrowdale, D.; Dennis, J.; et al. Genome-Wide Association Study in BRCA1 Mutation Carriers Identifies Novel Loci Associated with Breast and Ovarian Cancer Risk. PLoS Genet. 2013, 9, e1003212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Gaudet, M.M.; Kuchenbaecker, K.B.; Vijai, J.; Klein, R.J.; Kirchhoff, T.; McGuffog, L.; Barrowdale, D.; Dunning, A.M.; Lee, A.; Dennis, J.; et al. Identification of a BRCA2-specific modifier locus at 6p24 related to breast cancer risk. PLoS Genet. 2013, 9, e1003173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Antoniou, A.C.; Wang, X.; Fredericksen, Z.S.; McGuffog, L.; Tarrell, R.; Sinilnikova, O.M.; Healey, S.; Morrison, J.; Kartsonaki, C.; Lesnick, T.; et al. A locus on 19p13 modifies risk of breast cancer in BRCA1 mutation carriers and is associated with hormone receptor-negative breast cancer in the general population. Nat. Genet. 2010, 42, 885–892. [Google Scholar] [CrossRef] [PubMed]
  19. Ramus, S.J.; Kartsonaki, C.; Gayther, S.A.; Pharoah, P.D.P.; Sinilnikova, O.M.; Beesley, J.; Chen, X.; McGuffog, L.; Healey, S.; Couch, F.J.; et al. Genetic variation at 9p22.2 and ovarian cancer risk for BRCA1 and BRCA2 mutation carriers. J. Natl. Cancer Inst. 2011, 103, 105–116. [Google Scholar] [CrossRef] [Green Version]
  20. Zhu, Q.; Zhang, J.; Chen, Y.; Hu, Q.; Shen, H.; Huang, R.-Y.; Liu, Q.; Kaur, J.; Long, M.; Battaglia, S.; et al. Whole-exome sequencing of ovarian cancer families uncovers putative predisposition genes. Int. J. Cancer 2020, 146, 2147–2155. [Google Scholar] [CrossRef]
  21. Tayo, B.O.; DiCioccio, R.A.; Liang, Y.; Trevisan, M.; Cooper, R.S.; Lele, S.; Sucheston, L.; Piver, S.M.; Odunsi, K. Complex Segregation Analysis of Pedigrees from the Gilda Radner Familial Ovarian Cancer Registry Reveals Evidence for Mendelian Dominant Inheritance. PLoS ONE 2009, 4, e5939. [Google Scholar] [CrossRef] [Green Version]
  22. Campbell, P.J.; Getz, G.; Korbel, J.O.; Stuart, J.M.; Jennings, J.L.; Stein, L.D.; Perry, M.D.; Nahal-Bose, H.K.; Ouellette, B.F.F.; Li, C.H.; et al. Pan-cancer analysis of whole genomes. Nature 2020, 578, 82–93. [Google Scholar] [CrossRef] [Green Version]
  23. Poplin, R.; Chang, P.-C.; Alexander, D.; Schwartz, S.; Colthurst, T.; Ku, A.; Newburger, D.; Dijamco, J.; Nguyen, N.; Afshar, P.T.; et al. A universal SNP and small-indel variant caller using deep neural networks. Nat. Biotechnol. 2018, 36, 983–987. [Google Scholar] [CrossRef] [PubMed]
  24. Mohiyuddin, M.; Mu, J.C.; Li, J.; Bani Asadi, N.; Gerstein, M.B.; Abyzov, A.; Wong, W.H.; Lam, H.Y.K. MetaSV: An accurate and integrative structural-variant caller for next generation sequencing. Bioinformatics 2015, 31, 2741–2744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Talevich, E.; Shain, A.H.; Botton, T.; Bastian, B.C. CNVkit: Genome-Wide Copy Number Detection and Visualization from Targeted DNA Sequencing. PLoS Comput. Biol. 2016, 12, e1004873. [Google Scholar] [CrossRef] [PubMed]
  26. Chen, X.; Schulz-Trieglaff, O.; Shaw, R.; Barnes, B.; Schlesinger, F.; Källberg, M.; Cox, A.J.; Kruglyak, S.; Saunders, C.T. Manta: Rapid detection of structural variants and indels for germline and cancer sequencing applications. Bioinformatics 2016, 32, 1220–1222. [Google Scholar] [CrossRef] [PubMed]
  27. Layer, R.M.; Chiang, C.; Quinlan, A.R.; Hall, I.M. LUMPY: A probabilistic framework for structural variant discovery. Genome Biol. 2014, 15, R84. [Google Scholar] [CrossRef] [Green Version]
  28. Kronenberg, Z.N.; Osborne, E.J.; Cone, K.R.; Kennedy, B.J.; Domyan, E.T.; Shapiro, M.D.; Elde, N.C.; Yandell, M. Wham: Identifying Structural Variants of Biological Consequence. PLoS Comput. Biol. 2015, 11, e1004572. [Google Scholar] [CrossRef]
  29. Liu, Q.; Hu, Q.; Yao, S.; Kwan, M.L.; Roh, J.M.; Zhao, H.; Ambrosone, C.B.; Kushi, L.H.; Liu, S.; Zhu, Q. SeqSQC: A Bioconductor Package for Evaluating the Sample Quality of Next-generation Sequencing Data. Genom. Proteom. Bioinform. 2019, 17, 211–218. [Google Scholar] [CrossRef]
  30. Leiserson, M.D.M.; Vandin, F.; Wu, H.-T.; Dobson, J.R.; Eldridge, J.V.; Thomas, J.L.; Papoutsaki, A.; Kim, Y.; Niu, B.; McLellan, M.; et al. Pan-cancer network analysis identifies combinations of rare somatic mutations across pathways and protein complexes. Nat. Genet. 2015, 47, 106–114. [Google Scholar] [CrossRef]
  31. Croft, D.; O’Kelly, G.; Wu, G.; Haw, R.; Gillespie, M.; Matthews, L.; Caudy, M.; Garapati, P.; Gopinath, G.; Jassal, B.; et al. Reactome: A database of reactions, pathways and biological processes. Nucleic Acids Res. 2011, 39, D691–D697. [Google Scholar] [CrossRef]
  32. AlDubayan, S.H.; Conway, J.R.; Camp, S.Y.; Witkowski, L.; Kofman, E.; Reardon, B.; Han, S.; Moore, N.; Elmarakeby, H.; Salari, K.; et al. Detection of Pathogenic Variants with Germline Genetic Testing Using Deep Learning vs Standard Methods in Patients with Prostate Cancer and Melanoma. JAMA 2020, 324, 1957–1969. [Google Scholar] [CrossRef] [PubMed]
  33. Calabrese, C.; Davidson, N.R.; Demircioğlu, D.; Fonseca, N.A.; He, Y.; Kahles, A.; Lehmann, K.-V.; Liu, F.; Shiraishi, Y.; Soulette, C.M.; et al. Genomic basis for RNA alterations in cancer. Nature 2020, 578, 129–136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Wirtenberger, M.; Tchatchou, S.; Hemminki, K.; Schmutzhard, J.; Sutter, C.; Schmutzler, R.K.; Meindl, A.; Wappenschmidt, B.; Kiechle, M.; Arnold, N.; et al. Associations of genetic variants in the estrogen receptor coactivators PPARGC1A, PPARGC1B and EP300 with familial breast cancer. Carcinogenesis 2006, 27, 2201–2208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Permuth-Wey, J.; Chen, Y.A.; Tsai, Y.-Y.; Chen, Z.; Qu, X.; Lancaster, J.M.; Stockwell, H.; Dagne, G.; Iversen, E.; Risch, H.; et al. Inherited variants in mitochondrial biogenesis genes may influence epithelial ovarian cancer risk. Cancer Epidemiol. Biomark. Prev. 2011, 20, 1131–1145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Pignatelli, M.; Cocca, C.; Santos, A.; Perez-Castillo, A. Enhancement of BRCA1 gene expression by the peroxisome proliferator-activated receptor γ in the MCF-7 breast cancer cell line. Oncogene 2003, 22, 5446–5450. [Google Scholar] [CrossRef] [Green Version]
  37. Mastropasqua, F.; Girolimetti, G.; Shoshan, M. PGC1α: Friend or Foe in Cancer? Genes 2018, 9, 48. [Google Scholar] [CrossRef] [Green Version]
  38. Gravel, S.-P. Deciphering the Dichotomous Effects of PGC-1α on Tumorigenesis and Metastasis. Front. Oncol. 2018, 8, 75. [Google Scholar] [CrossRef] [Green Version]
  39. Gentric, G.; Kieffer, Y.; Mieulet, V.; Goundiam, O.; Bonneau, C.; Nemati, F.; Hurbain, I.; Raposo, G.; Popova, T.; Stern, M.-H.; et al. PML-Regulated Mitochondrial Metabolism Enhances Chemosensitivity in Human Ovarian Cancers. Cell Metab. 2019, 29, 156–173. [Google Scholar] [CrossRef] [Green Version]
  40. McGuirk, S.; Audet-Delage, Y.; St-Pierre, J. Metabolic Fitness and Plasticity in Cancer Progression. Trends Cancer 2020, 6, 49–61. [Google Scholar] [CrossRef]
  41. Tan, Z.; Luo, X.; Xiao, L.; Tang, M.; Bode, A.M.; Dong, Z.; Cao, Y. The Role of PGC1α in Cancer Metabolism and its Therapeutic Implications. Mol. Cancer Ther. 2016, 15, 774–782. [Google Scholar] [CrossRef] [Green Version]
  42. Scharping, N.E.; Menk, A.V.; Moreci, R.S.; Whetstone, R.D.; Dadey, R.E.; Watkins, S.C.; Ferris, R.L.; Delgoffe, G.M. The Tumor Microenvironment Represses T Cell Mitochondrial Biogenesis to Drive Intratumoral T Cell Metabolic Insufficiency and Dysfunction. Immunity 2016, 45, 374–388. [Google Scholar] [CrossRef] [Green Version]
  43. Van Vugt, M.A.T.M.; Parkes, E.E. When breaks get hot: Inflammatory signaling in BRCA1/2-mutant cancers. Trends Cancer 2022, 8, 174–189. [Google Scholar] [CrossRef] [PubMed]
  44. Lord, C.J.; Ashworth, A. BRCAness revisited. Nat. Rev. Cancer 2016, 16, 110–120. [Google Scholar] [CrossRef] [PubMed]
  45. Strickland, K.C.; Howitt, B.E.; Shukla, S.A.; Rodig, S.; Ritterhouse, L.L.; Liu, J.F.; Garber, J.E.; Chowdhury, D.; Wu, C.J.; D’Andrea, A.D.; et al. Association and prognostic significance of BRCA1/2-mutation status with neoantigen load, number of tumor-infiltrating lymphocytes and expression of PD-1/PD-L1 in high grade serous ovarian cancer. Oncotarget 2016, 7, 13587–13598. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. George, J.; Alsop, K.; Etemadmoghadam, D.; Hondow, H.; Mikeska, T.; Dobrovic, A.; deFazio, A.; Smyth, G.K.; Levine, D.A.; Mitchell, G.; et al. Nonequivalent gene expression and copy number alterations in high-grade serous ovarian cancers with BRCA1 and BRCA2 mutations. Clin. Cancer Res. 2013, 19, 3474–3484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. McAlpine, J.N.; Porter, H.; Köbel, M.; Nelson, B.H.; Prentice, L.M.; Kalloger, S.E.; Senz, J.; Milne, K.; Ding, J.; Shah, S.P.; et al. BRCA1 and BRCA2 mutations correlate with TP53 abnormalities and presence of immune cell infiltrates in ovarian high-grade serous carcinoma. Mod. Pathol. 2012, 25, 740–750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Wieser, V.; Gaugg, I.; Fleischer, M.; Shivalingaiah, G.; Wenzel, S.; Sprung, S.; Lax, S.F.; Zeimet, A.G.; Fiegl, H.; Marth, C. BRCA1/2 and TP53 mutation status associates with PD-1 and PD-L1 expression in ovarian cancer. Oncotarget 2018, 9, 17501–17511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Bruand, M.; Barras, D.; Mina, M.; Ghisoni, E.; Morotti, M.; Lanitis, E.; Fahr, N.; Desbuisson, M.; Grimm, A.; Zhang, H.; et al. Cell-autonomous inflammation of BRCA1-deficient ovarian cancers drives both tumor-intrinsic immunoreactivity and immune resistance via STING. Cell Rep. 2021, 36, 109412. [Google Scholar] [CrossRef]
  50. McCarthy, A.; Savage, K.; Gabriel, A.; Naceur, C.; Reis-Filho, J.S.; Ashworth, A. A mouse model of basal-like breast carcinoma with metaplastic elements. J. Pathol. 2007, 211, 389–398. [Google Scholar] [CrossRef]
  51. Perets, R.; Wyant, G.A.; Muto, K.W.; Bijron, J.G.; Poole, B.B.; Chin, K.T.; Chen, J.Y.H.; Ohman, A.W.; Stepule, C.D.; Kwak, S.; et al. Transformation of the Fallopian Tube Secretory Epithelium Leads to High-Grade Serous Ovarian Cancer in Brca;Tp53;Pten Models. Cancer Cell 2013, 24, 751–765. [Google Scholar] [CrossRef] [Green Version]
  52. Szabova, L.; Yin, C.; Bupp, S.; Guerin, T.M.; Schlomer, J.J.; Householder, D.B.; Baran, M.L.; Yi, M.; Song, Y.; Sun, W.; et al. Perturbation of Rb, p53, and Brca1 or Brca2 Cooperate in Inducing Metastatic Serous Epithelial Ovarian Cancer. Cancer Res. 2012, 72, 4141–4153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Kent, W.J.; Sugnet, C.W.; Furey, T.S.; Roskin, K.M.; Pringle, T.H.; Zahler, A.M.; Haussler, D. The Human Genome Browser at UCSC. Genome Res. 2002, 12, 996–1006. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Karczewski, K.J.; Francioli, L.C.; Tiao, G.; Cummings, B.B.; Alföldi, J.; Wang, Q.; Collins, R.L.; Laricchia, K.M.; Ganna, A.; Birnbaum, D.P.; et al. The mutational constraint spectrum quantified from variation in 141,456 humans. Nature 2020, 581, 434–443. [Google Scholar] [CrossRef] [PubMed]
  55. Liu, X.; Jian, X.; Boerwinkle, E. dbNSFP: A lightweight database of human nonsynonymous SNPs and their functional predictions. Hum. Mutat. 2011, 32, 894–899. [Google Scholar] [CrossRef]
  56. Wang, K.; Li, M.; Hakonarson, H. ANNOVAR: Functional annotation of genetic variants from highthroughput sequencing data. Nucleic Acids Res. 2010, 38, e164. [Google Scholar] [CrossRef]
  57. Liu, X.; White, S.; Peng, B.; Johnson, A.D.; Brody, J.A.; Li, A.H.; Huang, Z.; Carroll, A.; Wei, P.; Gibbs, R.; et al. WGSA: An annotation pipeline for human genome sequencing studies. J. Med. Genet. 2016, 53, 111–112. [Google Scholar] [CrossRef] [Green Version]
  58. Fishilevich, S.; Nudel, R.; Rappaport, N.; Hadar, R.; Plaschkes, I.; Iny Stein, T.; Rosen, N.; Kohn, A.; Twik, M.; Safran, M.; et al. GeneHancer: Genome-wide integration of enhancers and target genes in GeneCards. Database 2017, 2017, bax028. [Google Scholar] [CrossRef] [Green Version]
  59. Moore, J.E.; Pratt, H.E.; Purcaro, M.J.; Weng, Z. A curated benchmark of enhancer-gene interactions for evaluating enhancer-target gene prediction methods. Genome Biol. 2020, 21, 17. [Google Scholar] [CrossRef] [Green Version]
  60. MacDonald, J.R.; Ziman, R.; Yuen, R.K.; Feuk, L.; Scherer, S.W. The Database of Genomic Variants: A curated collection of structural variation in the human genome. Nucleic. Acids Res. 2014, 42, D986–D992. [Google Scholar] [CrossRef] [Green Version]
  61. Aran, D.; Sirota, M.; Butte, A.J. Systematic pan-cancer analysis of tumour purity. Nat. Commun. 2015, 6, 8971. [Google Scholar] [CrossRef] [Green Version]
  62. Yoshihara, K.; Shahmoradgoli, M.; Martínez, E.; Vegesna, R.; Kim, H.; Torres-Garcia, W.; Treviño, V.; Shen, H.; Laird, P.W.; Levine, D.A.; et al. Inferring tumour purity and stromal and immune cell admixture from expression data. Nat. Commun. 2013, 4, 2612. [Google Scholar] [CrossRef] [PubMed]
  63. Love, M.I.; Huber, W.; Anders, S. Moderated estimation of fold change and dispersion for RNAseq data with DESeq2. Genome Biol. 2014, 15, 550. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Huang, K.L.; Mashl, R.J.; Wu, Y.; Ritter, D.I.; Wang, J.; Oh, C.; Paczkowska, M.; Reynolds, S.; Wyczalkowski, M.A.; Oak, N.; et al. Pathogenic Germline Variants in 10,389 Adult Cancers. Cell 2018, 173, 355–370.e314. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Schema of the analyses to identify genetic modifiers of ovarian and breast cancer risks in BRCA carriers.
Figure 1. Schema of the analyses to identify genetic modifiers of ovarian and breast cancer risks in BRCA carriers.
Cancers 14 02350 g001
Figure 2. The largest gene sub-network significantly altered in BRCA carriers. Genes that were significantly highly mutated in BRCA carriers (Table 1) were highlighted by red underscores.
Figure 2. The largest gene sub-network significantly altered in BRCA carriers. Genes that were significantly highly mutated in BRCA carriers (Table 1) were highlighted by red underscores.
Cancers 14 02350 g002
Figure 3. Fraction of BRCA carriers and non-carriers that contained PPARGC1A mutations: (a) analysis within the PCAWG validation cohorts; (b) analysis within our discovery cohort, the PCAWG validation cohorts, and all cohorts combined. The numbers inside the bars are the numbers of BRCA carriers and non-carriers.
Figure 3. Fraction of BRCA carriers and non-carriers that contained PPARGC1A mutations: (a) analysis within the PCAWG validation cohorts; (b) analysis within our discovery cohort, the PCAWG validation cohorts, and all cohorts combined. The numbers inside the bars are the numbers of BRCA carriers and non-carriers.
Cancers 14 02350 g003
Figure 4. The distribution of cancer age onset by BRCA and PPARGC1A mutation status in PCAWG breast and ovarian cancer patients. The number in parenthesis is the sample size of each group.
Figure 4. The distribution of cancer age onset by BRCA and PPARGC1A mutation status in PCAWG breast and ovarian cancer patients. The number in parenthesis is the sample size of each group.
Cancers 14 02350 g004
Figure 5. The PPARGC1A variants identified in the discovery stage (a); and validation stage (b); respectively.
Figure 5. The PPARGC1A variants identified in the discovery stage (a); and validation stage (b); respectively.
Cancers 14 02350 g005
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Zhu, Q.; Wang, J.; Yu, H.; Hu, Q.; Bateman, N.W.; Long, M.; Rosario, S.; Schultz, E.; Dalgard, C.L.; Wilkerson, M.D.; et al. Whole-Genome Sequencing Identifies PPARGC1A as a Putative Modifier of Cancer Risk in BRCA1/2 Mutation Carriers. Cancers 2022, 14, 2350. https://doi.org/10.3390/cancers14102350

AMA Style

Zhu Q, Wang J, Yu H, Hu Q, Bateman NW, Long M, Rosario S, Schultz E, Dalgard CL, Wilkerson MD, et al. Whole-Genome Sequencing Identifies PPARGC1A as a Putative Modifier of Cancer Risk in BRCA1/2 Mutation Carriers. Cancers. 2022; 14(10):2350. https://doi.org/10.3390/cancers14102350

Chicago/Turabian Style

Zhu, Qianqian, Jie Wang, Han Yu, Qiang Hu, Nicholas W. Bateman, Mark Long, Spencer Rosario, Emily Schultz, Clifton L. Dalgard, Matthew D. Wilkerson, and et al. 2022. "Whole-Genome Sequencing Identifies PPARGC1A as a Putative Modifier of Cancer Risk in BRCA1/2 Mutation Carriers" Cancers 14, no. 10: 2350. https://doi.org/10.3390/cancers14102350

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop