Stanford Medicine Professor of Pathology, Professor of Genetics and of Biomedical Data Science

Publications

  • RNA Sequencing in Disease Diagnosis. Annual review of genomics and human genetics Smail, C., Montgomery, S. B. 2024

    Abstract

    RNA sequencing (RNA-seq) enables the accurate measurement of multiple transcriptomic phenotypes for modeling the impacts of disease variants. Advances in technologies, experimental protocols, and analysis strategies are rapidly expanding the application of RNA-seq to identify disease biomarkers, tissue- and cell-type-specific impacts, and the spatial localization of disease-associated mechanisms. Ongoing international efforts to construct biobank-scale transcriptomic repositories with matched genomic data across diverse population groups are further increasing the utility of RNA-seq approaches by providing large-scale normative reference resources. The availability of these resources, combined with improved computational analysis pipelines, has enabled the detection of aberrant transcriptomic phenotypes underlying rare diseases. Further expansion of these resources, across both somatic and developmental tissues, is expected to soon provide unprecedented insights to resolve disease origin, mechanism of action, and causal gene contributions, suggesting the continued high utility of RNA-seq in disease diagnosis. Expected final online publication date for the Annual Review of Genomics and Human Genetics, Volume 25 is August 2024. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.

    View details for DOI 10.1146/annurev-genom-021623-121812

    View details for PubMedID 38360541

  • Impact of genome build on RNA-seq interpretation and diagnostics. medRxiv : the preprint server for health sciences Ungar, R. A., Goddard, P. C., Jensen, T. D., Degalez, F., Smith, K. S., Jin, C. A., Bonner, D. E., Bernstein, J. A., Wheeler, M. T., Montgomery, S. B. 2024

    Abstract

    Transcriptomics is a powerful tool for unraveling the molecular effects of genetic variants and disease diagnosis. Prior studies have demonstrated that choice of genome build impacts variant interpretation and diagnostic yield for genomic analyses. To identify the extent genome build also impacts transcriptomics analyses, we studied the effect of the hg19, hg38, and CHM13 genome builds on expression quantification and outlier detection in 386 rare disease and familial control samples from both the Undiagnosed Diseases Network (UDN) and Genomics Research to Elucidate the Genetics of Rare Disease (GREGoR) Consortium. We identified 2,800 genes with build-dependent quantification across six routinely-collected biospecimens, including 1,391 protein-coding genes and 341 known rare disease genes. We further observed multiple genes that only have detectable expression in a subset of genome builds. Finally, we characterized how genome build impacts the detection of outlier transcriptomic events. Combined, we provide a database of genes impacted by build choice, and recommend that transcriptomics-guided analyses and diagnoses are cross-referenced with these data for robustness.

    View details for DOI 10.1101/2024.01.11.24301165

    View details for PubMedID 38260490

    View details for PubMedCentralID PMC10802764

  • Genetic architecture of cardiac dynamic flow volumes. Nature genetics Gomes, B., Singh, A., O'Sullivan, J. W., Schnurr, T. M., Goddard, P. C., Loong, S., Amar, D., Hughes, J. W., Kostur, M., Haddad, F., Salerno, M., Foo, R., Montgomery, S. B., Parikh, V. N., Meder, B., Ashley, E. A. 2023

    Abstract

    Cardiac blood flow is a critical determinant of human health. However, the definition of its genetic architecture is limited by the technical challenge of capturing dynamic flow volumes from cardiac imaging at scale. We present DeepFlow, a deep-learning system to extract cardiac flow and volumes from phase-contrast cardiac magnetic resonance imaging. A mixed-linear model applied to 37,653 individuals from the UK Biobank reveals genome-wide significant associations across cardiac dynamic flow volumes spanning from aortic forward velocity to aortic regurgitation fraction. Mendelian randomization reveals a causal role for aortic root size in aortic valve regurgitation. Among the most significant contributing variants, localizing genes (near ELN, PRDM6 and ADAMTS7) are implicated in connective tissue and blood pressure pathways. Here we show that DeepFlow cardiac flow phenotyping at scale, combined with genotyping data, reinforces the contribution of connective tissue genes, blood pressure and root size to aortic valve function.

    View details for DOI 10.1038/s41588-023-01587-5

    View details for PubMedID 38082205

    View details for PubMedCentralID 7612636

  • Organ aging signatures in the plasma proteome track health and disease. Nature Oh, H. S., Rutledge, J., Nachun, D., Pálovics, R., Abiose, O., Moran-Losada, P., Channappa, D., Urey, D. Y., Kim, K., Sung, Y. J., Wang, L., Timsina, J., Western, D., Liu, M., Kohlfeld, P., Budde, J., Wilson, E. N., Guen, Y., Maurer, T. M., Haney, M., Yang, A. C., He, Z., Greicius, M. D., Andreasson, K. I., Sathyan, S., Weiss, E. F., Milman, S., Barzilai, N., Cruchaga, C., Wagner, A. D., Mormino, E., Lehallier, B., Henderson, V. W., Longo, F. M., Montgomery, S. B., Wyss-Coray, T. 2023; 624 (7990): 164-172

    Abstract

    Animal studies show aging varies between individuals as well as between organs within an individual1-4, but whether this is true in humans and its effect on age-related diseases is unknown. We utilized levels of human blood plasma proteins originating from specific organs to measure organ-specific aging differences in living individuals. Using machine learning models, we analysed aging in 11 major organs and estimated organ age reproducibly in five independent cohorts encompassing 5,676 adults across the human lifespan. We discovered nearly 20% of the population show strongly accelerated age in one organ and 1.7% are multi-organ agers. Accelerated organ aging confers 20-50% higher mortality risk, and organ-specific diseases relate to faster aging of those organs. We find individuals with accelerated heart aging have a 250% increased heart failure risk and accelerated brain and vascular aging predict Alzheimer's disease (AD) progression independently from and as strongly as plasma pTau-181 (ref. 5), the current best blood-based biomarker for AD. Our models link vascular calcification, extracellular matrix alterations and synaptic protein shedding to early cognitive decline. We introduce a simple and interpretable method to study organ aging using plasma proteomics data, predicting diseases and aging effects.

    View details for DOI 10.1038/s41586-023-06802-1

    View details for PubMedID 38057571

    View details for PubMedCentralID PMC10700136

  • Transcriptomics and chromatin accessibility in multiple African population samples. bioRxiv : the preprint server for biology DeGorter, M. K., Goddard, P. C., Karakoc, E., Kundu, S., Yan, S. M., Nachun, D., Abell, N., Aguirre, M., Carstensen, T., Chen, Z., Durrant, M., Dwaracherla, V. R., Feng, K., Gloudemans, M. J., Hunter, N., Moorthy, M. P., Pomilla, C., Rodrigues, K. B., Smith, C. J., Smith, K. S., Ungar, R. A., Balliu, B., Fellay, J., Flicek, P., McLaren, P. J., Henn, B., McCoy, R. C., Sugden, L., Kundaje, A., Sandhu, M. S., Gurdasani, D., Montgomery, S. B. 2023

    Abstract

    Mapping the functional human genome and impact of genetic variants is often limited to European-descendent population samples. To aid in overcoming this limitation, we measured gene expression using RNA sequencing in lymphoblastoid cell lines (LCLs) from 599 individuals from six African populations to identify novel transcripts including those not represented in the hg38 reference genome. We used whole genomes from the 1000 Genomes Project and 164 Maasai individuals to identify 8,881 expression and 6,949 splicing quantitative trait loci (eQTLs/sQTLs), and 2,611 structural variants associated with gene expression (SV-eQTLs). We further profiled chromatin accessibility using ATAC-Seq in a subset of 100 representative individuals, to identity chromatin accessibility quantitative trait loci (caQTLs) and allele-specific chromatin accessibility, and provide predictions for the functional effect of 78.9 million variants on chromatin accessibility. Using this map of eQTLs and caQTLs we fine-mapped GWAS signals for a range of complex diseases. Combined, this work expands global functional genomic data to identify novel transcripts, functional elements and variants, understand population genetic history of molecular quantitative trait loci, and further resolve the genetic basis of multiple human traits and disease.

    View details for DOI 10.1101/2023.11.04.564839

    View details for PubMedID 37986808

    View details for PubMedCentralID PMC10659267

  • Multi- Omic Profiling of Macrophages Lacking Tet2 or Dnmt3a Reveals Mechanisms of Hyper-Inflammation in Clonal Hematopoiesis Rodrigues, K. B., Gopakumar, J., Weng, Z., Mitchell, S., Maurer, M., Nachun, D., Eulalio, T., Estrada, D., Mazumder, T., Ma, L., Montgomery, S., Jaiswal, S. AMER SOC HEMATOLOGY. 2023
  • Integrative analyses highlight functional regulatory variants associated with neuropsychiatric diseases. Nature genetics Guo, M. G., Reynolds, D. L., Ang, C. E., Liu, Y., Zhao, Y., Donohue, L. K., Siprashvili, Z., Yang, X., Yoo, Y., Mondal, S., Hong, A., Kain, J., Meservey, L., Fabo, T., Elfaki, I., Kellman, L. N., Abell, N. S., Pershad, Y., Bayat, V., Etminani, P., Holodniy, M., Geschwind, D. H., Montgomery, S. B., Duncan, L. E., Urban, A. E., Altman, R. B., Wernig, M., Khavari, P. A. 2023

    Abstract

    Noncoding variants of presumed regulatory function contribute to the heritability of neuropsychiatric disease. A total of 2,221 noncoding variants connected to risk for ten neuropsychiatric disorders, including autism spectrum disorder, attention deficit hyperactivity disorder, bipolar disorder, borderline personality disorder, major depression, generalized anxiety disorder, panic disorder, post-traumatic stress disorder, obsessive-compulsive disorder and schizophrenia, were studied in developing human neural cells. Integrating epigenomic and transcriptomic data with massively parallel reporter assays identified differentially-active single-nucleotide variants (daSNVs) in specific neural cell types. Expression-gene mapping, network analyses and chromatin looping nominated candidate disease-relevant target genes modulated by these daSNVs. Follow-up integration of daSNV gene editing with clinical cohort analyses suggested that magnesium transport dysfunction may increase neuropsychiatric disease risk and indicated that common genetic pathomechanisms may mediate specific symptoms that are shared across multiple neuropsychiatric diseases.

    View details for DOI 10.1038/s41588-023-01533-5

    View details for PubMedID 37857935

    View details for PubMedCentralID 4112379

  • The functional impact of rare variation across the regulatory cascade. Cell genomics Li, T., Ferraro, N., Strober, B. J., Aguet, F., Kasela, S., Arvanitis, M., Ni, B., Wiel, L., Hershberg, E., Ardlie, K., Arking, D. E., Beer, R. L., Brody, J., Blackwell, T. W., Clish, C., Gabriel, S., Gerszten, R., Guo, X., Gupta, N., Johnson, W. C., Lappalainen, T., Lin, H. J., Liu, Y., Nickerson, D. A., Papanicolaou, G., Pritchard, J. K., Qasba, P., Shojaie, A., Smith, J., Sotoodehnia, N., Taylor, K. D., Tracy, R. P., Van Den Berg, D., Wheeler, M. T., Rich, S. S., Rotter, J. I., Battle, A., Montgomery, S. B. 2023; 3 (10): 100401

    Abstract

    Each human genome has tens of thousands of rare genetic variants; however, identifying impactful rare variants remains a major challenge. We demonstrate how use of personal multi-omics can enable identification of impactful rare variants by using the Multi-Ethnic Study of Atherosclerosis, which included several hundred individuals, with whole-genome sequencing, transcriptomes, methylomes, and proteomes collected across two time points, 10 years apart. We evaluated each multi-omics phenotype's ability to separately and jointly inform functional rare variation. By combining expression and protein data, we observed rare stop variants 62 times and rare frameshift variants 216 times as frequently as controls, compared to 13-27 times as frequently for expression or protein effects alone. We extended a Bayesian hierarchical model, "Watershed," to prioritize specific rare variants underlying multi-omics signals across the regulatory cascade. With this approach, we identified rare variants that exhibited large effect sizes on multiple complex traits including height, schizophrenia, and Alzheimer's disease.

    View details for DOI 10.1016/j.xgen.2023.100401

    View details for PubMedID 37868038

    View details for PubMedCentralID PMC10589633

  • Integrated single-cell multiome analysis reveals muscle fiber-type gene regulatory circuitry modulated by endurance exercise. bioRxiv : the preprint server for biology Rubenstein, A. B., Smith, G. R., Zhang, Z., Chen, X., Chambers, T. L., Ruf-Zamojski, F., Mendelev, N., Cheng, W. S., Zamojski, M., Amper, M. A., Nair, V. D., Marderstein, A. R., Montgomery, S. B., Troyanskaya, O. G., Zaslavsky, E., Trappe, T., Trappe, S., Sealfon, S. C. 2023

    Abstract

    Endurance exercise is an important health modifier. We studied cell-type specific adaptations of human skeletal muscle to acute endurance exercise using single-nucleus (sn) multiome sequencing in human vastus lateralis samples collected before and 3.5 hours after 40 min exercise at 70% VO2max in four subjects, as well as in matched time of day samples from two supine resting circadian controls. High quality same-cell RNA-seq and ATAC-seq data were obtained from 37,154 nuclei comprising 14 cell types. Among muscle fiber types, both shared and fiber-type specific regulatory programs were identified. Single-cell circuit analysis identified distinct adaptations in fast, slow and intermediate fibers as well as LUM-expressing FAP cells, involving a total of 328 transcription factors (TFs) acting at altered accessibility sites regulating 2,025 genes. These data and circuit mapping provide single-cell insight into the processes underlying tissue and metabolic remodeling responses to exercise.

    View details for DOI 10.1101/2023.09.26.558914

    View details for PubMedID 37808658

    View details for PubMedCentralID PMC10557702

  • Author Correction: Africa-specific human genetic variation near CHD1L associates with HIV-1 load. Nature McLaren, P. J., Porreca, I., Iaconis, G., Mok, H. P., Mukhopadhyay, S., Karakoc, E., Cristinelli, S., Pomilla, C., Bartha, I., Thorball, C. W., Tough, R. H., Angelino, P., Kiar, C. S., Carstensen, T., Fatumo, S., Porter, T., Jarvis, I., Skarnes, W. C., Bassett, A., DeGorter, M. K., Sathya Moorthy, M. P., Tuff, J. F., Kim, E. Y., Walter, M., Simons, L. M., Bashirova, A., Buchbinder, S., Carrington, M., Cossarizza, A., De Luca, A., Goedert, J. J., Goldstein, D. B., Haas, D. W., Herbeck, J. T., Johnson, E. O., Kaleebu, P., Kilembe, W., Kirk, G. D., Kootstra, N. A., Kral, A. H., Lambotte, O., Luo, M., Mallal, S., Martinez-Picado, J., Meyer, L., Miro, J. M., Moodley, P., Motala, A. A., Mullins, J. I., Nam, K., Obel, N., Pirie, F., Plummer, F. A., Poli, G., Price, M. A., Rauch, A., Theodorou, I., Trkola, A., Walker, B. D., Winkler, C. A., Zagury, J. F., Montgomery, S. B., Ciuffi, A., Hultquist, J. F., Wolinsky, S. M., Dougan, G., Lever, A. M., Gurdasani, D., Groom, H., Sandhu, M. S., Fellay, J. 2023

    View details for DOI 10.1038/s41586-023-06591-7

    View details for PubMedID 37670157

  • Beyond the exome: What's next in diagnostic testing for Mendelian conditions. American journal of human genetics Wojcik, M. H., Reuter, C. M., Marwaha, S., Mahmoud, M., Duyzend, M. H., Barseghyan, H., Yuan, B., Boone, P. M., Groopman, E. E., Délot, E. C., Jain, D., Sanchis-Juan, A., Starita, L. M., Talkowski, M., Montgomery, S. B., Bamshad, M. J., Chong, J. X., Wheeler, M. T., Berger, S. I., O'Donnell-Luria, A., Sedlazeck, F. J., Miller, D. E. 2023; 110 (8): 1229-1248

    Abstract

    Despite advances in clinical genetic testing, including the introduction of exome sequencing (ES), more than 50% of individuals with a suspected Mendelian condition lack a precise molecular diagnosis. Clinical evaluation is increasingly undertaken by specialists outside of clinical genetics, often occurring in a tiered fashion and typically ending after ES. The current diagnostic rate reflects multiple factors, including technical limitations, incomplete understanding of variant pathogenicity, missing genotype-phenotype associations, complex gene-environment interactions, and reporting differences between clinical labs. Maintaining a clear understanding of the rapidly evolving landscape of diagnostic tests beyond ES, and their limitations, presents a challenge for non-genetics professionals. Newer tests, such as short-read genome or RNA sequencing, can be challenging to order, and emerging technologies, such as optical genome mapping and long-read DNA sequencing, are not available clinically. Furthermore, there is no clear guidance on the next best steps after inconclusive evaluation. Here, we review why a clinical genetic evaluation may be negative, discuss questions to be asked in this setting, and provide a framework for further investigation, including the advantages and disadvantages of new approaches that are nascent in the clinical sphere. We present a guide for the next best steps after inconclusive molecular testing based upon phenotype and prior evaluation, including when to consider referral to research consortia focused on elucidating the underlying cause of rare unsolved genetic disorders.

    View details for DOI 10.1016/j.ajhg.2023.06.009

    View details for PubMedID 37541186

  • Africa-specific human genetic variation near CHD1L associates with HIV-1 load. Nature McLaren, P. J., Porreca, I., Iaconis, G., Mok, H. P., Mukhopadhyay, S., Karakoc, E., Cristinelli, S., Pomilla, C., Bartha, I., Thorball, C. W., Tough, R. H., Angelino, P., Kiar, C. S., Carstensen, T., Fatumo, S., Porter, T., Jarvis, I., Skarnes, W. C., Bassett, A., DeGorter, M. K., Sathya Moorthy, M. P., Tuff, J. F., Kim, E. Y., Walter, M., Simons, L. M., Bashirova, A., Buchbinder, S., Carrington, M., Cossarizza, A., De Luca, A., Goedert, J. J., Goldstein, D. B., Haas, D. W., Herbeck, J. T., Johnson, E. O., Kaleebu, P., Kilembe, W., Kirk, G. D., Kootstra, N. A., Kral, A. H., Lambotte, O., Luo, M., Mallal, S., Martinez-Picado, J., Meyer, L., Miro, J. M., Moodley, P., Motala, A. A., Mullins, J. I., Nam, K., Obel, N., Pirie, F., Plummer, F. A., Poli, G., Price, M. A., Rauch, A., Theodorou, I., Trkola, A., Walker, B. D., Winkler, C. A., Zagury, J. F., Montgomery, S. B., Ciuffi, A., Hultquist, J. F., Wolinsky, S. M., Dougan, G., Lever, A. M., Gurdasani, D., Groom, H., Sandhu, M. S., Fellay, J. 2023

    Abstract

    HIV-1 remains a global health crisis1, highlighting the need to identify new targets for therapies. Here, given the disproportionate HIV-1 burden and marked human genome diversity in Africa2, we assessed the genetic determinants of control of set-point viral load in 3,879 people of African ancestries living with HIV-1 participating in the international collaboration for the genomics of HIV3. We identify a previously undescribed association signal on chromosome 1 where the peak variant associates with an approximately 0.3 log10-transformed copies per ml lower set-point viral load per minor allele copy and is specific to populations of African descent. The top associated variant is intergenic and lies between a long intergenic non-coding RNA (LINC00624) and the coding gene CHD1L, which encodes a helicase that is involved in DNA repair4. Infection assays in iPS cell-derived macrophages and other immortalized cell lines showed increased HIV-1 replication in CHD1L-knockdown and CHD1L-knockout cells. We provide evidence from population genetic studies that Africa-specific genetic variation near CHD1L associates with HIV replication in vivo. Although experimental studies suggest that CHD1L is able to limit HIV infection in some cell types in vitro, further investigation is required to understand the mechanisms underlying our observations, including any potential indirect effects of CHD1L on HIV spread in vivo that our cell-based assays cannot recapitulate.

    View details for DOI 10.1038/s41586-023-06370-4

    View details for PubMedID 37532928

    View details for PubMedCentralID 3723635

  • Molecular quantitative trait loci NATURE REVIEWS METHODS PRIMERS Aguet, F., Alasoo, K., Li, Y., Battle, A., Im, H., Montgomery, S. B., Lappalainen, T. 2023; 3 (1)
  • Beyond the exome: what's next in diagnostic testing for Mendelian conditions. ArXiv Wojcik, M. H., Reuter, C. M., Marwaha, S., Mahmoud, M., Duyzend, M. H., Barseghyan, H., Yuan, B., Boone, P. M., Groopman, E. E., Délot, E. C., Jain, D., Sanchis-Juan, A., Starita, L. M., Talkowski, M., Montgomery, S. B., Bamshad, M. J., Chong, J. X., Wheeler, M. T., Berger, S. I., O'Donnell-Luria, A., Sedlazeck, F. J., Miller, D. E. 2023

    Abstract

    Despite advances in clinical genetic testing, including the introduction of exome sequencing (ES), more than 50% of individuals with a suspected Mendelian condition lack a precise molecular diagnosis. Clinical evaluation is increasingly undertaken by specialists outside of clinical genetics, often occurring in a tiered fashion and typically ending after ES. The current diagnostic rate reflects multiple factors, including technical limitations, incomplete understanding of variant pathogenicity, missing genotype-phenotype associations, complex gene-environment interactions, and reporting differences between clinical labs. Maintaining a clear understanding of the rapidly evolving landscape of diagnostic tests beyond ES, and their limitations, presents a challenge for non-genetics professionals. Newer tests, such as short-read genome or RNA sequencing, can be challenging to order and emerging technologies, such as optical genome mapping and long-read DNA or RNA sequencing, are not available clinically. Furthermore, there is no clear guidance on the next best steps after inconclusive evaluation. Here, we review why a clinical genetic evaluation may be negative, discuss questions to be asked in this setting, and provide a framework for further investigation, including the advantages and disadvantages of new approaches that are nascent in the clinical sphere. We present a guide for the next best steps after inconclusive molecular testing based upon phenotype and prior evaluation, including when to consider referral to a consortium such as GREGoR, which is focused on elucidating the underlying cause of rare unsolved genetic disorders.

    View details for DOI 10.1002/ajmg.a.63053

    View details for PubMedID 36713248

    View details for PubMedCentralID PMC9882576

  • The mitochondrial multi-omic response to exercise training across tissues. bioRxiv : the preprint server for biology Amar, D., Gay, N. R., Jimenez-Morales, D., Beltran, P. M., Ramaker, M. E., Raja, A. N., Zhao, B., Sun, Y., Marwaha, S., Gaul, D., Hershman, S. G., Xia, A., Lanza, I., Fernandez, F. M., Montgomery, S. B., Hevener, A. L., Ashley, E. A., Walsh, M. J., Sparks, L. M., Burant, C. F., Rector, R. S., Thyfault, J., Wheeler, M. T., Goodpaster, B. H., Coen, P. M., Schenk, S., Bodine, S. C., Lindholm, M. E. 2023

    Abstract

    Mitochondria are adaptable organelles with diverse cellular functions critical to whole-body metabolic homeostasis. While chronic endurance exercise training is known to alter mitochondrial activity, these adaptations have not yet been systematically characterized. Here, the Molecular Transducers of Physical Activity Consortium (MoTrPAC) mapped the longitudinal, multi-omic changes in mitochondrial analytes across 19 tissues in male and female rats endurance trained for 1, 2, 4 or 8 weeks. Training elicited substantial changes in the adrenal gland, brown adipose, colon, heart and skeletal muscle, while we detected mild responses in the brain, lung, small intestine and testes. The colon response was characterized by non-linear dynamics that resulted in upregulation of mitochondrial function that was more prominent in females. Brown adipose and adrenal tissues were characterized by substantial downregulation of mitochondrial pathways. Training induced a previously unrecognized robust upregulation of mitochondrial protein abundance and acetylation in the liver, and a concomitant shift in lipid metabolism. The striated muscles demonstrated a highly coordinated response to increase oxidative capacity, with the majority of changes occurring in protein abundance and post-translational modifications. We identified exercise upregulated networks that are downregulated in human type 2 diabetes and liver cirrhosis. In both cases HSD17B10, a central dehydrogenase in multiple metabolic pathways and mitochondrial tRNA maturation, was the main hub. In summary, we provide a multi-omic, cross-tissue atlas of the mitochondrial response to training and identify candidates for prevention of disease-associated mitochondrial dysfunction.

    View details for DOI 10.1101/2023.01.13.523698

    View details for PubMedID 36711881

    View details for PubMedCentralID PMC9882193

  • Multiomic identification of key transcriptional regulatory programs during endurance exercise training. bioRxiv : the preprint server for biology Smith, G. R., Zhao, B., Lindholm, M. E., Raja, A., Viggars, M., Pincas, H., Gay, N. R., Sun, Y., Ge, Y., Nair, V. D., Sanford, J. A., S Amper, M. A., Vasoya, M., Smith, K. S., Montgomery, S., Zaslavsky, E., Bodine, S. C., Esser, K. A., Walsh, M. J., Snyder, M. P., Sealfon, S. C., MoTrPAC Study Group 2023

    Abstract

    Transcription factors (TFs) play a key role in regulating gene expression and responses to stimuli. We conducted an integrated analysis of chromatin accessibility and RNA expression across various rat tissues following endurance exercise training (EET) to map epigenomic changes to transcriptional changes and determine key TFs involved. We uncovered tissue-specific changes across both omic layers, including highly correlated differentially accessible regions (DARs) and differentially expressed genes (DEGs). We identified open chromatin regions associated with DEGs (DEGaPs) and found tissue-specific and genomic feature-specific TF motif enrichment patterns among both DARs and DEGaPs. Accessible promoters of up-vs. down-regulated DEGs per tissue showed distinct TF enrichment patterns. Further, some EET-induced TFs in skeletal muscle were either validated at the proteomic level (MEF2C and NUR77) or correlated with exercise-related phenotypic changes. We provide an in-depth analysis of the epigenetic and trans-factor-dependent processes governing gene expression during EET.

    View details for DOI 10.1101/2023.01.10.523450

    View details for PubMedID 36711841

  • RNAget: an API to securely retrieve RNA quantifications. Bioinformatics (Oxford, England) Upchurch, S., Palumbo, E., Adams, J., Bujold, D., Bourque, G., Nedzel, J., Graham, K., Kagda, M. S., Assis, P., Hitz, B., Righi, E., Guigo, R., Wold, B. J., GA4GH RNA-Seq Task Team, Adams, J., Brazma, A., Bujold, D., Burchard, J., Capka, J., Cherry, M., Clarke, L., Craft, B., Dermitzakis, M., Diekhans, M., Dursi, J., Fitzsimons, M. S., Flaming, Z., Garrido, R., Gil, A., Godden, P., Green, M., Guigo, R., Guttman, M., Haas, B., Haeussler, M., Hitz, B., Li, B., Linnarsson, S., Lipski, A., Liu, D., Longerich, S., Lougheed, D., Manning, J., Marioni, J., Meyer, C., Montgomery, S., Morrow, A., Munoz-Power Fuentes, A., Nedzel, J., Nguyen, D., Osborn, K., Ouellette, F., Palumbo, E., Papatheodorou, I., Pervouchine, D., Ramani, A., Rambla, J., Sadjad, B., Steinberg, D., Talkar, J., Tickle, T., Tzeng, K., Upchurch, S., Vaisipour, S., Watford, S., Wold, B., Zhang, Z., Zhu, J. 2023; 39 (4)

    Abstract

    SUMMARY: Large-scale sharing of genomic quantification data requires standardized access interfaces. In this Global Alliance for Genomics and Health project, we developed RNAget, an API for secure access to genomic quantification data in matrix form. RNAget provides for slicing matrices to extract desired subsets of data and is applicable to all expression matrix-format data, including RNA sequencing and microarrays. Further, it generalizes to quantification matrices of other sequence-based genomics such as ATAC-seq and ChIP-seq.AVAILABILITY AND IMPLEMENTATION: https://ga4gh-rnaseq.github.io/schema/docs/index.html.

    View details for DOI 10.1093/bioinformatics/btad126

    View details for PubMedID 36897015

  • Methylation differences in Alzheimer's disease neuropathologic change in the aged human brain. Acta neuropathologica communications Lang, A. L., Eulalio, T., Fox, E., Yakabi, K., Bukhari, S. A., Kawas, C. H., Corrada, M. M., Montgomery, S. B., Heppner, F. L., Capper, D., Nachun, D., Montine, T. J. 2022; 10 (1): 174

    Abstract

    Alzheimer's disease (AD) is the most common cause of dementia with advancing age as its strongest risk factor. AD neuropathologic change (ADNC) is known to be associated with numerous DNA methylation changes in the human brain, but the oldest old (> 90 years) have so far been underrepresented in epigenetic studies of ADNC. Our study participants were individuals aged over 90 years (n = 47) from The 90+ Study. We analyzed DNA methylation from bulk samples in eight precisely dissected regions of the human brain: middle frontal gyrus, cingulate gyrus, entorhinal cortex, dentate gyrus, CA1, substantia nigra, locus coeruleus and cerebellar cortex. We deconvolved our bulk data into cell-type-specific (CTS) signals using computational methods. CTS methylation differences were analyzed across different levels of ADNC. The highest amount of ADNC related methylation differences was found in the dentate gyrus, a region that has so far been underrepresented in large scale multi-omic studies. In neurons of the dentate gyrus, DNA methylation significantly differed with increased burden of amyloid beta (Aβ) plaques at 5897 promoter regions of protein-coding genes. Amongst these, higher Aβ plaque burden was associated with promoter hypomethylation of the Presenilin enhancer 2 (PEN-2) gene, one of the rate limiting genes in the formation of gamma-secretase, a multicomponent complex that is responsible in part for the endoproteolytic cleavage of amyloid precursor protein into Aβ peptides. In addition to novel ADNC related DNA methylation changes, we present the most detailed array-based methylation survey of the old aged human brain to date. Our open-sourced dataset can serve as a brain region reference panel for future studies and help advance research in aging and neurodegenerative diseases.

    View details for DOI 10.1186/s40478-022-01470-0

    View details for PubMedID 36447297

    View details for PubMedCentralID PMC9710143

  • Deep learning-assisted genome-wide characterization of massively parallel reporter assays. Nucleic acids research Lu, F., Sossin, A., Abell, N., Montgomery, S. B., He, Z. 2022

    Abstract

    Massively parallel reporter assay (MPRA) is a high-throughput method that enables the study of the regulatory activities of tens of thousands of DNA oligonucleotides in a single experiment. While MPRA experiments have grown in popularity, their small sample sizes compared to the scale of the human genome limits our understanding of the regulatory effects they detect. To address this, we develop a deep learning model, MpraNet, to distinguish potential MPRA targets from the background genome. This model achieves high discriminative performance (AUROC=0.85) at differentiating MPRA positives from a set of control variants that mimic the background genome when applied to the lymphoblastoid cell line. We observe that existing functional scores represent very distinct functional effects, and most of them fail to characterize the regulatory effect that MPRA detects. Using MpraNet, we predict potential MPRA functional variants across the genome and identify the distributions of MPRA effect relative to other characteristics of genetic variation, including allele frequency, alternative functional annotations specified by FAVOR, and phenome-wide associations. We also observed that the predicted MPRA positives are not uniformly distributed across the genome; instead, they are clumped together in active regions comprising 9.95% of the genome and inactive regions comprising 89.07% of the genome. Furthermore, we propose our model as a screen to filter MPRA experiment candidates at genome-wide scale, enabling future experiments to be more cost-efficient by increasing precision relative to that observed from previous MPRAs.

    View details for DOI 10.1093/nar/gkac990

    View details for PubMedID 36350674

  • RNA editing underlies genetic risk of common inflammatory diseases. Nature Li, Q., Gloudemans, M. J., Geisinger, J. M., Fan, B., Aguet, F., Sun, T., Ramaswami, G., Li, Y. I., Ma, J. B., Pritchard, J. K., Montgomery, S. B., Li, J. B. 2022

    Abstract

    A major challenge in human genetics is to identify the molecular mechanisms of trait-associated and disease-associated variants. To achieve this, quantitative trait locus (QTL) mapping of genetic variants with intermediate molecular phenotypes such as gene expression and splicing have been widely adopted1,2. However, despite successes, the molecular basis for a considerable fraction of trait-associated and disease-associated variants remains unclear3,4. Here we show that ADAR-mediated adenosine-to-inosine RNA editing, a post-transcriptional event vital for suppressing cellular double-stranded RNA (dsRNA)-mediated innate immune interferon responses5-11, is an important potential mechanism underlying genetic variants associated with common inflammatory diseases. We identified and characterized 30,319 cis-RNA editing QTLs (edQTLs) across 49 human tissues. These edQTLs were significantly enriched in genome-wide association study signals for autoimmune and immune-mediated diseases. Colocalization analysis of edQTLs with disease risk loci further pinpointed key, putatively immunogenic dsRNAs formed by expected inverted repeat Alu elements as well as unexpected, highly over-represented cis-natural antisense transcripts. Furthermore, inflammatory disease risk variants, in aggregate, were associated with reduced editing of nearby dsRNAs and induced interferon responses in inflammatory diseases. This unique directional effect agrees with the established mechanism that lack of RNA editing by ADAR1 leads to the specific activation of the dsRNA sensor MDA5 and subsequent interferon responses and inflammation7-9. Our findings implicate cellular dsRNA editing and sensing as a previously underappreciated mechanism of common inflammatory diseases.

    View details for DOI 10.1038/s41586-022-05052-x

    View details for PubMedID 35922514