1
|
Pirini F, Calzari L, Tedaldi G, Tebaldi M, Zampiga V, Cangini I, Danesi R, Ravegnani M, Arcangeli V, Passardi A, Petracci E, Bravaccini S, Marisi G, Viel A, Barana D, Pedroni M, Roncucci L, Calistri D, Gentilini D. Comprehensive genetic and epigenetic characterization of Lynch-like syndrome patients. Int J Cancer 2025. [PMID: 40259440 DOI: 10.1002/ijc.35451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 02/18/2025] [Accepted: 04/04/2025] [Indexed: 04/23/2025]
Abstract
Lynch-like syndrome (LLS) presents very similar clinicopathological characteristics to Lynch syndrome (LS) but the mechanism for cancer predisposition remains unknown. The present study aims to investigate the causal mechanism of LLS by a comprehensive genetic and epigenetic approach. Thirty-two LLS and 34 LS patients with colorectal cancer (CRC) fitting the Amsterdam and Bethesda criteria were included, along with 29 CRC sporadic patients, and analyzed for the presence of pathogenic variants in 94 genes associated with hereditary tumors. The cohorts were also characterized for the methylation profile and examined through a sample group analysis and a Stochastic Epigenetic Mutations (SEMs) analysis in comparison with 29 age-matched healthy controls. The multigene panel analysis revealed the presence of pathogenic variants in non-mismatch repair (MMR) genes and three variants classified as pathogenic/likely pathogenic possibly predisposing to LLS. The epigenetic analysis showed epivariations targeting genes associated with LS or DNA repair, most of them associated with the Fanconi Anemia pathway, which could explain the susceptibility to cancer. Our results highlight the need for using extended genetic and epigenetic analyses to understand the causal mechanism of LLS.
Collapse
Affiliation(s)
- Francesca Pirini
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Luciano Calzari
- Bioinformatics and Statistical Genomic Unit, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Gianluca Tedaldi
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Michela Tebaldi
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Valentina Zampiga
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Ilaria Cangini
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Rita Danesi
- Romagna Cancer Registry, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Mila Ravegnani
- Romagna Cancer Registry, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Valentina Arcangeli
- Romagna Cancer Registry, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Alessandro Passardi
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Elisabetta Petracci
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Sara Bravaccini
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
- Faculty of Medicine and Surgery, University of Enna "Kore", Enna, Italy
| | - Giorgia Marisi
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Alessandra Viel
- Unit of Oncogenetics and Functional Oncogenomics, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Daniela Barana
- Oncology Unit, Local Health and Social Care Unit, ULSS8 Berica, Vicenza, Italy
| | - Monica Pedroni
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Luca Roncucci
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Daniele Calistri
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Davide Gentilini
- Bioinformatics and Statistical Genomic Unit, IRCCS Istituto Auxologico Italiano, Milan, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
2
|
Nishino J, Miya F, Kato M. Gene-based Hardy-Weinberg equilibrium test using genotype count data: application to six types of cancers. BMC Genomics 2025; 26:124. [PMID: 39930364 PMCID: PMC11809088 DOI: 10.1186/s12864-025-11321-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 02/04/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND An alternative approach to investigate associations between genetic variants and disease is to examine deviations from the Hardy-Weinberg equilibrium (HWE) in genotype frequencies within a case population, instead of case-control association analysis. The HWE analysis requires disease cases and demonstrates a notable ability in mapping recessive variants. Allelic heterogeneity is a common phenomenon in diseases. While gene-based case-control association analysis successfully incorporates this heterogeneity, there are no such approaches for HWE analysis. Therefore, we proposed a gene-based HWE test (gene-HWT) by aggregating single-nucleotide polymorphism (SNP)-level HWE test statistics in a gene to address allelic heterogeneity. RESULTS This method used only genotype count data and publicly available linkage disequilibrium information and has a very low computational cost. Extensive simulations demonstrated that gene-HWT effectively controls the type I error at a low significance level and outperforms SNP-level HWE test in power when there are multiple causal variants within a gene. Using gene-HWT, we analyzed genotype count data from a genome-wide association study of six cancer types in Japanese individuals and suggest DGKE and ANO3 as potential germline factors in colorectal cancer. Furthermore, FSTL4 was suggested through a combined analysis across the six cancer types, with particularly notable associations observed in colorectal and prostate cancers. CONCLUSIONS These findings indicate the potential of gene-HWT to elucidate the genetic basis of complex diseases, including cancer.
Collapse
Affiliation(s)
- Jo Nishino
- Division of Bioinformatics, National Cancer Center Research Institute, Tokyo, Japan.
| | - Fuyuki Miya
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| | - Mamoru Kato
- Division of Bioinformatics, National Cancer Center Research Institute, Tokyo, Japan
| |
Collapse
|
3
|
Aliev F, De Sa Nogueira D, Aston-Jones G, Dick DM. Genetic associations between orexin genes and phenotypes related to behavioral regulation in humans, including substance use. Mol Psychiatry 2025:10.1038/s41380-025-02895-4. [PMID: 39880903 DOI: 10.1038/s41380-025-02895-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 08/23/2024] [Accepted: 01/14/2025] [Indexed: 01/31/2025]
Abstract
The hypothalamic neuropeptide system of orexin (hypocretin) neurons provides projections throughout the neuraxis and has been linked to sleep regulation, feeding and motivation for salient rewards including drugs of abuse. However, relatively little has been done to examine genes associated with orexin signaling and specific behavioral phenotypes in humans. Here, we tested for association of twenty-seven genes involved in orexin signaling with behavioral phenotypes in humans. We tested the full gene set, functional subsets, and individual genes involved in orexin signaling. Our primary phenotype of interest was Externalizing, a composite factor comprised of behaviors and disorders associated with reward-seeking, motivation, and behavioral regulation. We also tested for association with additional phenotypes that have been related to orexin regulation in model organism studies, including alcohol consumption, problematic alcohol use, daytime sleepiness, insomnia, cigarettes per day, smoking initiation, and body mass index. The composite set of 27 genes corresponding to orexin function was highly associated with Externalizing, as well as with alcohol consumption, insomnia, cigarettes per day, smoking initiation and BMI. In addition, all gene subsets (except the OXR2/HCRTR2 subset) were associated with Externalizing. BMI was significantly associated with all gene subsets. The "validated factors for PPOX/HCRT" and "PPOX/HCRT upregulation" gene subsets also were associated with alcohol consumption. Individually, 8 genes showed a strong association with Externalizing, 12 with BMI, 7 with smoking initiation, 3 with alcohol consumption, and 2 with problematic alcohol use, after correction for multiple testing. This study indicates that orexin genes are associated with multiple behaviors and disorders related to self-regulation in humans. This is consistent with prior work in animals that implicated orexin signaling in motivational activation induced by salient stimuli, and supports the hypothesis that orexin signaling is an important potential therapeutic target for numerous behavioral disorders.
Collapse
Affiliation(s)
- Fazil Aliev
- Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA
- Rutgers Addiction Research Center, Brain Health Institute, Rutgers University and Rutgers Health, Piscataway, NJ, 08854, USA
| | - David De Sa Nogueira
- Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA
- Rutgers Addiction Research Center, Brain Health Institute, Rutgers University and Rutgers Health, Piscataway, NJ, 08854, USA
| | - Gary Aston-Jones
- Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA
- Rutgers Addiction Research Center, Brain Health Institute, Rutgers University and Rutgers Health, Piscataway, NJ, 08854, USA
| | - Danielle M Dick
- Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA.
- Rutgers Addiction Research Center, Brain Health Institute, Rutgers University and Rutgers Health, Piscataway, NJ, 08854, USA.
| |
Collapse
|
4
|
Gillespie NA, Bell TR, Hearn GC, Hess JL, Tsuang MT, Lyons MJ, Franz CE, Kremen WS, Glatt SJ. A twin analysis to estimate genetic and environmental factors contributing to variation in weighted gene co-expression network module eigengenes. Am J Med Genet B Neuropsychiatr Genet 2025; 198:e33003. [PMID: 39126209 PMCID: PMC11778624 DOI: 10.1002/ajmg.b.33003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 06/18/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024]
Abstract
Multivariate network-based analytic methods such as weighted gene co-expression network analysis are frequently applied to human and animal gene-expression data to estimate the first principal component of a module, or module eigengene (ME). MEs are interpreted as multivariate summaries of correlated gene-expression patterns and network connectivity across genes within a module. As such, they have the potential to elucidate the mechanisms by which molecular genomic variation contributes to individual differences in complex traits. Although increasingly used to test for associations between modules and complex traits, the genetic and environmental etiology of MEs has not been empirically established. It is unclear if, and to what degree, individual differences in blood-derived MEs reflect random variation versus familial aggregation arising from heritable or shared environmental influences. We used biometrical genetic analyses to estimate the contribution of genetic and environmental influences on MEs derived from blood lymphocytes collected on a sample of N = 661 older male twins from the Vietnam Era Twin Study of Aging (VETSA) whose mean age at assessment was 67.7 years (SD = 2.6 years, range = 62-74 years). Of the 26 detected MEs, 14 (56%) had statistically significant additive genetic variation with an average heritability of 44% (SD = 0.08, range = 35%-64%). Despite the relatively small sample size, this demonstration of significant family aggregation including estimates of heritability in 14 of the 26 MEs suggests that blood-based MEs are reliable and merit further exploration in terms of their associations with complex traits and diseases.
Collapse
Affiliation(s)
- Nathan A. Gillespie
- Virginia Institute for Psychiatric and Behavior Genetics, Virginia Commonwealth University, Virginia, USA
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Tyler R. Bell
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, California, USA
| | - Gentry C. Hearn
- Department of Psychological and Brain Sciences, Boston University, Boston, Massachusetts, USA
| | - Jonathan L. Hess
- Department of Psychological and Brain Sciences, Boston University, Boston, Massachusetts, USA
| | - Ming T. Tsuang
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA
| | - Michael J. Lyons
- Department of Psychological and Brain Sciences, Boston University, Boston, Massachusetts, USA
| | - Carol E. Franz
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, California, USA
| | - William S. Kremen
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, California, USA
| | - Stephen J. Glatt
- Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, New York, USA
| |
Collapse
|
5
|
Alekseeva AO, Zolotovskaia MA, Sorokin MI, Suntsova MV, Zakharova GS, Pugacheva PA, Tinkov AA, Korobeinikova TV, Sekacheva MI, Poddubskaya EV, Skalny AV, Kushlinskii NE, Buzdin AA. The First Multiomics Association Study of Trace Element and Mineral Concentration and RNA Sequencing Profiles in Human Cancers. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:2274-2286. [PMID: 39865039 DOI: 10.1134/s0006297924120150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 01/28/2025]
Abstract
Integration of various types of omics data is an important trend in contemporary molecular oncology. In this regard, high-throughput analysis of trace and essential elements in cancer biosamples is an emerging field that has not yet been sufficiently addressed. For the first time, we simultaneously obtained gene expression profiles (RNA sequencing) and essential and trace element profiles (inductively coupled plasma mass spectrometry) for a set of human cancer samples. The biosamples were formalin-fixed, paraffin-embedded primary tumor tissue blocks: 67 for colorectal cancer patients and 18 for other solid cancer types (16 types). Mass spectrometry profiles were obtained for 45 chemical elements: Ag, Al, As, Au, B, Ba, Be, Bi, Ca, Cd, Co, Cr, Cu, Fe, Ga, Ge, Hg, I, K, La, Li, Mg, Mn, Mo, Na, Ni, P, Pb, Pd, Pt, Rb, Sb, Sc, Se, Si, Sn, Sr, Te, Ti, Tl, Zn, U, V, W, and Zr. The expression levels were profiled for 36,596 known human genes, and the activation levels were assessed for 10,520 human intracellular molecular pathways. For the concentrations of essential elements Ca, Cu, Fe, K, Mg, Na, P, and Zn we detected statistically significant correlations on both gene expression and pathway activation levels for both colorectal cancer samples and at the pan-cancer level. In total, 222/137, 122/220, 1/0, 239/186, 71/44, 1/0, 354/294, 69/82 gene/pathway biomarkers were detected for Ca, Cu, Fe, K, Mg, Na, P, and Zn, respectively. We believe that this first-in-class database provided here will be valuable for multiomics cancer research.
Collapse
Affiliation(s)
- Arsenia O Alekseeva
- Digital Biodesign and Personalized Healthcare Research Center, Sechenov University, Moscow, 119991, Russia
| | - Marianna A Zolotovskaia
- Digital Biodesign and Personalized Healthcare Research Center, Sechenov University, Moscow, 119991, Russia
| | - Maksim I Sorokin
- Digital Biodesign and Personalized Healthcare Research Center, Sechenov University, Moscow, 119991, Russia
| | - Maria V Suntsova
- Digital Biodesign and Personalized Healthcare Research Center, Sechenov University, Moscow, 119991, Russia
| | - Galina S Zakharova
- Digital Biodesign and Personalized Healthcare Research Center, Sechenov University, Moscow, 119991, Russia
| | - Polina A Pugacheva
- Digital Biodesign and Personalized Healthcare Research Center, Sechenov University, Moscow, 119991, Russia
| | - Aleksey A Tinkov
- Center of Bioelementology and Human Ecology, Sechenov University, Moscow, 119435, Russia
| | | | - Marina I Sekacheva
- Digital Biodesign and Personalized Healthcare Research Center, Sechenov University, Moscow, 119991, Russia
| | - Elena V Poddubskaya
- Digital Biodesign and Personalized Healthcare Research Center, Sechenov University, Moscow, 119991, Russia
- Vitamed Clinic, Moscow, 117312, Russia
| | - Anatoly V Skalny
- Center of Bioelementology and Human Ecology, Sechenov University, Moscow, 119435, Russia
| | | | - Anton A Buzdin
- Digital Biodesign and Personalized Healthcare Research Center, Sechenov University, Moscow, 119991, Russia.
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia
- PathoBiology Group, European Organization for Research and Treatment of Cancer (EORTC), Brussels, Belgium
| |
Collapse
|
6
|
Hussain MA, Grant PE, Ou Y. Inferring neurocognition using artificial intelligence on brain MRIs. FRONTIERS IN NEUROIMAGING 2024; 3:1455436. [PMID: 39664769 PMCID: PMC11631947 DOI: 10.3389/fnimg.2024.1455436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 11/07/2024] [Indexed: 12/13/2024]
Abstract
Brain magnetic resonance imaging (MRI) offers a unique lens to study neuroanatomic support of human neurocognition. A core mystery is the MRI explanation of individual differences in neurocognition and its manifestation in intelligence. The past four decades have seen great advancement in studying this century-long mystery, but the sample size and population-level studies limit the explanation at the individual level. The recent rise of big data and artificial intelligence offers novel opportunities. Yet, data sources, harmonization, study design, and interpretation must be carefully considered. This review aims to summarize past work, discuss rising opportunities and challenges, and facilitate further investigations on artificial intelligence inferring human neurocognition.
Collapse
Affiliation(s)
- Mohammad Arafat Hussain
- Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Patricia Ellen Grant
- Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
- Department of Radiology, Harvard Medical School, Boston, MA, United States
| | - Yangming Ou
- Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
- Department of Radiology, Harvard Medical School, Boston, MA, United States
- Computational Health Informatics Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
7
|
Shenhav L, Fehr K, Reyna ME, Petersen C, Dai DLY, Dai R, Breton V, Rossi L, Smieja M, Simons E, Silverman MA, Levy M, Bode L, Field CJ, Marshall JS, Moraes TJ, Mandhane PJ, Turvey SE, Subbarao P, Surette MG, Azad MB. Microbial colonization programs are structured by breastfeeding and guide healthy respiratory development. Cell 2024; 187:5431-5452.e20. [PMID: 39303691 PMCID: PMC11531244 DOI: 10.1016/j.cell.2024.07.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 03/02/2024] [Accepted: 07/12/2024] [Indexed: 09/22/2024]
Abstract
Breastfeeding and microbial colonization during infancy occur within a critical time window for development, and both are thought to influence the risk of respiratory illness. However, the mechanisms underlying the protective effects of breastfeeding and the regulation of microbial colonization are poorly understood. Here, we profiled the nasal and gut microbiomes, breastfeeding characteristics, and maternal milk composition of 2,227 children from the CHILD Cohort Study. We identified robust colonization patterns that, together with milk components, predict preschool asthma and mediate the protective effects of breastfeeding. We found that early cessation of breastfeeding (before 3 months) leads to the premature acquisition of microbial species and functions, including Ruminococcus gnavus and tryptophan biosynthesis, which were previously linked to immune modulation and asthma. Conversely, longer exclusive breastfeeding supports a paced microbial development, protecting against asthma. These findings underscore the importance of extended breastfeeding for respiratory health and highlight potential microbial targets for intervention.
Collapse
Affiliation(s)
- Liat Shenhav
- Institute for Systems Genetics, New York Grossman School of Medicine, New York University, New York, NY, USA; Department of Microbiology, New York Grossman School of Medicine, New York University, New York, NY, USA; Department of Computer Science, Courant Institute of Mathematical Sciences, New York University, New York, NY, USA.
| | - Kelsey Fehr
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada; Manitoba Interdisciplinary Lactation Centre (MILC), Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
| | - Myrtha E Reyna
- Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Charisse Petersen
- Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Darlene L Y Dai
- Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Ruixue Dai
- Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Vanessa Breton
- Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Laura Rossi
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Marek Smieja
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Elinor Simons
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada; Manitoba Interdisciplinary Lactation Centre (MILC), Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
| | - Michael A Silverman
- Division of Infectious Disease, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA; Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Maayan Levy
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lars Bode
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA; Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (MOMI CORE), University of California, San Diego, La Jolla, CA, USA; Human Milk Institute (HMI), University of California, San Diego, La Jolla, CA, USA
| | - Catherine J Field
- Department of Agriculture, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Jean S Marshall
- Department of Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada
| | - Theo J Moraes
- Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Piush J Mandhane
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Stuart E Turvey
- Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Padmaja Subbarao
- Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada; Department of Physiology, University of Toronto, Toronto, ON, Canada.
| | | | - Meghan B Azad
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada; Manitoba Interdisciplinary Lactation Centre (MILC), Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
8
|
Kontou PI, Bagos PG. The goldmine of GWAS summary statistics: a systematic review of methods and tools. BioData Min 2024; 17:31. [PMID: 39238044 PMCID: PMC11375927 DOI: 10.1186/s13040-024-00385-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 08/27/2024] [Indexed: 09/07/2024] Open
Abstract
Genome-wide association studies (GWAS) have revolutionized our understanding of the genetic architecture of complex traits and diseases. GWAS summary statistics have become essential tools for various genetic analyses, including meta-analysis, fine-mapping, and risk prediction. However, the increasing number of GWAS summary statistics and the diversity of software tools available for their analysis can make it challenging for researchers to select the most appropriate tools for their specific needs. This systematic review aims to provide a comprehensive overview of the currently available software tools and databases for GWAS summary statistics analysis. We conducted a comprehensive literature search to identify relevant software tools and databases. We categorized the tools and databases by their functionality, including data management, quality control, single-trait analysis, and multiple-trait analysis. We also compared the tools and databases based on their features, limitations, and user-friendliness. Our review identified a total of 305 functioning software tools and databases dedicated to GWAS summary statistics, each with unique strengths and limitations. We provide descriptions of the key features of each tool and database, including their input/output formats, data types, and computational requirements. We also discuss the overall usability and applicability of each tool for different research scenarios. This comprehensive review will serve as a valuable resource for researchers who are interested in using GWAS summary statistics to investigate the genetic basis of complex traits and diseases. By providing a detailed overview of the available tools and databases, we aim to facilitate informed tool selection and maximize the effectiveness of GWAS summary statistics analysis.
Collapse
Affiliation(s)
| | - Pantelis G Bagos
- Department of Computer Science and Biomedical Informatics, University of Thessaly, 35131, Lamia, Greece.
| |
Collapse
|
9
|
Calzari L, Dragani DF, Zanotti L, Inglese E, Danesi R, Cavagnola R, Brusati A, Ranucci F, Di Blasio AM, Persani L, Campi I, De Martino S, Farsetti A, Barbi V, Gottardi Zamperla M, Baldrighi GN, Gaetano C, Parati G, Gentilini D. Epigenetic patterns, accelerated biological aging, and enhanced epigenetic drift detected 6 months following COVID-19 infection: insights from a genome-wide DNA methylation study. Clin Epigenetics 2024; 16:112. [PMID: 39164752 PMCID: PMC11337605 DOI: 10.1186/s13148-024-01724-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 08/08/2024] [Indexed: 08/22/2024] Open
Abstract
BACKGROUND The epigenetic status of patients 6-month post-COVID-19 infection remains largely unexplored. The existence of long-COVID, or post-acute sequelae of SARS-CoV-2 infection (PASC), suggests potential long-term changes. Long-COVID includes symptoms like fatigue, neurological issues, and organ-related problems, regardless of initial infection severity. The mechanisms behind long-COVID are unclear, but virus-induced epigenetic changes could play a role. METHODS AND RESULTS Our study explores the lasting epigenetic impacts of SARS-CoV-2 infection. We analyzed genome-wide DNA methylation patterns in an Italian cohort of 96 patients 6 months after COVID-19 exposure, comparing them to 191 healthy controls. We identified 42 CpG sites with significant methylation differences (FDR < 0.05), primarily within CpG islands and gene promoters. Dysregulated genes highlighted potential links to glutamate/glutamine metabolism, which may be relevant to PASC symptoms. Key genes with potential significance to COVID-19 infection and long-term effects include GLUD1, ATP1A3, and ARRB2. Furthermore, Horvath's epigenetic clock showed a slight but significant age acceleration in post-COVID-19 patients. We also observed a substantial increase in stochastic epigenetic mutations (SEMs) in the post-COVID-19 group, implying potential epigenetic drift. SEM analysis identified 790 affected genes, indicating dysregulation in pathways related to insulin resistance, VEGF signaling, apoptosis, hypoxia response, T-cell activation, and endothelin signaling. CONCLUSIONS Our study provides valuable insights into the epigenetic consequences of COVID-19. Results suggest possible associations with accelerated aging, epigenetic drift, and the disruption of critical biological pathways linked to insulin resistance, immune response, and vascular health. Understanding these epigenetic changes could be crucial for elucidating the complex mechanisms behind long-COVID and developing targeted therapeutic interventions.
Collapse
Affiliation(s)
- Luciano Calzari
- Bioinformatics and Statistical Genomics Unit, IRCCS Istituto Auxologico Italiano, Cusano Milanino, Milan, Italy
| | - Davide Fernando Dragani
- Bioinformatics and Statistical Genomics Unit, IRCCS Istituto Auxologico Italiano, Cusano Milanino, Milan, Italy
| | - Lucia Zanotti
- Department of Cardiology, S. Luca Hospital, IRCCS, Istituto Auxologico Italiano, Milan, Italy
| | - Elvira Inglese
- Clinical Chemistry Unit, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Via Bassi 21, Pavia, Italy
| | - Romano Danesi
- Clinical Chemistry Unit, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milano, Milan, Italy
| | - Rebecca Cavagnola
- Department of Brain and Behavioral Sciences, University of Pavia, Via Bassi 21, Pavia, Italy
| | - Alberto Brusati
- Department of Brain and Behavioral Sciences, University of Pavia, Via Bassi 21, Pavia, Italy
| | - Francesco Ranucci
- Department of Brain and Behavioral Sciences, University of Pavia, Via Bassi 21, Pavia, Italy
| | - Anna Maria Di Blasio
- Molecular Biology Laboratory, IRCCS Istituto Auxologico Italiano, Cusano Milanino, Milan, Italy
| | - Luca Persani
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
- Department of Endocrine and Metabolic Diseases, Lab of Endocrine and Metabolic Research, San Luca Hospital, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Irene Campi
- Department of Endocrine and Metabolic Diseases, Lab of Endocrine and Metabolic Research, San Luca Hospital, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Sara De Martino
- Consiglio Nazionale delle Ricerche (CNR) - IASI, Rome, Italy
| | | | - Veronica Barbi
- Laboratorio di Epigenetica, Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, 27100, Pavia, Italy
| | - Michela Gottardi Zamperla
- Laboratorio di Epigenetica, Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, 27100, Pavia, Italy
| | - Giulia Nicole Baldrighi
- Department of Brain and Behavioral Sciences, University of Pavia, Via Bassi 21, Pavia, Italy
| | - Carlo Gaetano
- Laboratorio di Epigenetica, Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, 27100, Pavia, Italy
| | - Gianfranco Parati
- Department of Cardiology, S. Luca Hospital, IRCCS, Istituto Auxologico Italiano, Milan, Italy
- Department of Medicine and Surgery, University of Milan-Bicocca, Milan, Italy
| | - Davide Gentilini
- Bioinformatics and Statistical Genomics Unit, IRCCS Istituto Auxologico Italiano, Cusano Milanino, Milan, Italy.
- Department of Brain and Behavioral Sciences, University of Pavia, Via Bassi 21, Pavia, Italy.
| |
Collapse
|
10
|
Visonà G, Bouzigon E, Demenais F, Schweikert G. Network propagation for GWAS analysis: a practical guide to leveraging molecular networks for disease gene discovery. Brief Bioinform 2024; 25:bbae014. [PMID: 38340090 PMCID: PMC10858647 DOI: 10.1093/bib/bbae014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/28/2023] [Accepted: 01/08/2024] [Indexed: 02/12/2024] Open
Abstract
MOTIVATION Genome-wide association studies (GWAS) have enabled large-scale analysis of the role of genetic variants in human disease. Despite impressive methodological advances, subsequent clinical interpretation and application remains challenging when GWAS suffer from a lack of statistical power. In recent years, however, the use of information diffusion algorithms with molecular networks has led to fruitful insights on disease genes. RESULTS We present an overview of the design choices and pitfalls that prove crucial in the application of network propagation methods to GWAS summary statistics. We highlight general trends from the literature, and present benchmark experiments to expand on these insights selecting as case study three diseases and five molecular networks. We verify that the use of gene-level scores based on GWAS P-values offers advantages over the selection of a set of 'seed' disease genes not weighted by the associated P-values if the GWAS summary statistics are of sufficient quality. Beyond that, the size and the density of the networks prove to be important factors for consideration. Finally, we explore several ensemble methods and show that combining multiple networks may improve the network propagation approach.
Collapse
Affiliation(s)
- Giovanni Visonà
- Empirical Inference, Max-Planck Institute for Intelligent Systems, Tübingen 72076, Germany
| | | | | | | |
Collapse
|
11
|
Stone TC, Ward V, Hogan A, Alexander Ho KM, Wilson A, McBain H, Duku M, Wolfson P, Cheung S, Rosenfeld A, Lovat LB. Using saliva epigenetic data to develop and validate a multivariable predictor of esophageal cancer status. Epigenomics 2024; 16:109-125. [PMID: 38226541 PMCID: PMC10825730 DOI: 10.2217/epi-2023-0248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 01/04/2024] [Indexed: 01/17/2024] Open
Abstract
Background: Salivary epigenetic biomarkers may detect esophageal cancer. Methods: A total of 256 saliva samples from esophageal adenocarcinoma patients and matched volunteers were analyzed with Illumina EPIC methylation arrays. Three datasets were created, using 64% for discovery, 16% for testing and 20% for validation. Modules of gene-based methylation probes were created using weighted gene coexpression network analysis. Module significance to disease and gene importance to module were determined and a random forest classifier generated using best-scoring gene-related epigenetic probes. A cost-sensitive wrapper algorithm maximized cancer diagnosis. Results: Using age, sex and seven probes, esophageal adenocarcinoma was detected with area under the curve of 0.72 in discovery, 0.73 in testing and 0.75 in validation datasets. Cancer sensitivity was 88% with specificity of 31%. Conclusion: We have demonstrated a potentially clinically viable classifier of esophageal cancer based on saliva methylation.
Collapse
Affiliation(s)
- Timothy C Stone
- Division of Surgery & Interventional Science, University College London, Charles Bell House, 43-45 Foley Street, London, W1W 7TY, UK
| | - Vanessa Ward
- Division of Surgery & Interventional Science, University College London, Charles Bell House, 43-45 Foley Street, London, W1W 7TY, UK
| | - Aine Hogan
- Division of Surgery & Interventional Science, University College London, Charles Bell House, 43-45 Foley Street, London, W1W 7TY, UK
| | - Kai Man Alexander Ho
- Division of Surgery & Interventional Science, University College London, Charles Bell House, 43-45 Foley Street, London, W1W 7TY, UK
- Wellcome/EPSRC Centre for Interventional & Surgical Sciences (WEISS), University College London, Charles Bell House, 43-45 Foley Street, London, W1W 7TY, UK
| | - Ash Wilson
- Division of Surgery & Interventional Science, University College London, Charles Bell House, 43-45 Foley Street, London, W1W 7TY, UK
| | - Hazel McBain
- Wellcome/EPSRC Centre for Interventional & Surgical Sciences (WEISS), University College London, Charles Bell House, 43-45 Foley Street, London, W1W 7TY, UK
| | - Margaret Duku
- Wellcome/EPSRC Centre for Interventional & Surgical Sciences (WEISS), University College London, Charles Bell House, 43-45 Foley Street, London, W1W 7TY, UK
| | - Paul Wolfson
- Division of Surgery & Interventional Science, University College London, Charles Bell House, 43-45 Foley Street, London, W1W 7TY, UK
| | - Sharon Cheung
- Division of Surgery & Interventional Science, University College London, Charles Bell House, 43-45 Foley Street, London, W1W 7TY, UK
| | - Avi Rosenfeld
- Department of Computer Science, Jerusalem College of Technology, Havaad Haleumi 21, Givat Mordechai, 91160, Jerusalem, Israel
| | - Laurence B Lovat
- Division of Surgery & Interventional Science, University College London, Charles Bell House, 43-45 Foley Street, London, W1W 7TY, UK
- Wellcome/EPSRC Centre for Interventional & Surgical Sciences (WEISS), University College London, Charles Bell House, 43-45 Foley Street, London, W1W 7TY, UK
- Department of Gastrointestinal Services, University College London Hospital, 235 Euston Road, London, NW1 2BU, UK
| |
Collapse
|
12
|
Cao F, Liu Y, Cheng Y, Wang Y, He Y, Xu Y. Multi-omics characteristics of tumor-associated macrophages in the tumor microenvironment of gastric cancer and their exploration of immunotherapy potential. Sci Rep 2023; 13:18265. [PMID: 37880233 PMCID: PMC10600170 DOI: 10.1038/s41598-023-38822-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/15/2023] [Indexed: 10/27/2023] Open
Abstract
The incidence and mortality rate of gastric cancer (GC) have remained high worldwide. Although some progress has been made in immunotargeted therapy, the treatment effect remains limited. With more attention has been paid to the immune potential of tumor-associated macrophages (TAMs), but the specific mechanisms of tumor immunity are still unclear. Thus, we screened marker genes in TAMs differentiation (MDMs) through single-cell RNA sequencing, and combined with GC transcriptome data from TCGA and GEO databases, the clinical and TME characteristics, prognostic differences, immune infiltration, and drug sensitivity among different subtypes of patients with GC in different data sets were analyzed. A prognostic model of GC was constructed to evaluate the prognosis and immunotherapy response of patients with GC. In this study, we extensively studied the mutations in MDMs such as CGN, S100A6, and C1QA, and found differences in the infiltration of immune cells and immune checkpoints including M2 TAMs, T cells, CD274, and CTLA4 in different GC subtypes. In the model, we constructed a predictive scoring system with high accuracy and screened out key MDMs-related genes associated with prognosis and M2 TAMs, among which VKORC1 may be involved in GC progression and iron death in tumor cells. Therefore, this study explores the therapeutic strategy of TAMs reprogramming in-depth, providing new ideas for the clinical diagnosis, treatment, and prognosis assessment of GC.
Collapse
Affiliation(s)
- Feng Cao
- Department of General Surgery, University Hospital RWTH Aachen, 52074, Aachen, Germany
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Yanwei Liu
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Yunsheng Cheng
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Yong Wang
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
| | - Yan He
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
| | - Yanyan Xu
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
| |
Collapse
|
13
|
Saito Y, Kamagata K, Uchida W, Takabayashi K, Aoki S. The improvement technique for reproducibility of diffusion tensor image analysis along the perivascular space (DTI-ALPS) for evaluating interstitial fluid diffusivity and glymphatic function. Jpn J Radiol 2023; 41:1029-1030. [PMID: 37162691 DOI: 10.1007/s11604-023-01421-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 03/28/2023] [Indexed: 05/11/2023]
Affiliation(s)
- Yuya Saito
- Department of Radiology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyoku, Tokyo, 113-8421, Japan
| | - Koji Kamagata
- Department of Radiology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyoku, Tokyo, 113-8421, Japan.
| | - Wataru Uchida
- Department of Radiology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyoku, Tokyo, 113-8421, Japan
| | - Kaito Takabayashi
- Department of Radiology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyoku, Tokyo, 113-8421, Japan
| | - Shigeki Aoki
- Department of Radiology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyoku, Tokyo, 113-8421, Japan
| |
Collapse
|
14
|
Defo J, Awany D, Ramesar R. From SNP to pathway-based GWAS meta-analysis: do current meta-analysis approaches resolve power and replication in genetic association studies? Brief Bioinform 2023; 24:6972298. [PMID: 36611240 DOI: 10.1093/bib/bbac600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/30/2022] [Accepted: 12/06/2022] [Indexed: 01/09/2023] Open
Abstract
Genome-wide association studies (GWAS) have benefited greatly from enhanced high-throughput technology in recent decades. GWAS meta-analysis has become increasingly popular to highlight the genetic architecture of complex traits, informing about the replicability and variability of effect estimations across human ancestries. A wealth of GWAS meta-analysis methodologies have been developed depending on the input data and the outcome information of interest. We present a survey of current approaches from SNP to pathway-based meta-analysis by acknowledging the range of resources and methodologies in the field, and we provide a comprehensive review of different categories of Genome-Wide Meta-analysis methods employed. These methods highlight different levels at which GWAS meta-analysis may be done, including Single Nucleotide Polymorphisms, Genes and Pathways, for which we describe their framework outline. We also discuss the strengths and pitfalls of each approach and make suggestions regarding each of them.
Collapse
Affiliation(s)
- Joel Defo
- Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, 7925, Observatory, South Africa.,South African Medical Research Council Genomic and Personalized Medicine Research Unit
| | - Denis Awany
- South African Tuberculosis Vaccine Initiative (SATVI), University of Cape Town, 7925, South Africa
| | - Raj Ramesar
- Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, 7925, Observatory, South Africa.,South African Medical Research Council Genomic and Personalized Medicine Research Unit
| |
Collapse
|
15
|
Gao Y, Felsky D, Reyes-Dumeyer D, Sariya S, Rentería MA, Ma Y, Klein HU, Cosentino S, De Jager PL, Bennett DA, Brickman AM, Schellenberg GD, Mayeux R, Barral S. Integration of GWAS and brain transcriptomic analyses in a multiethnic sample of 35,245 older adults identifies DCDC2 gene as predictor of episodic memory maintenance. Alzheimers Dement 2022; 18:1797-1811. [PMID: 34873813 PMCID: PMC9170841 DOI: 10.1002/alz.12524] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 10/03/2021] [Accepted: 10/12/2021] [Indexed: 01/28/2023]
Abstract
Identifying genes underlying memory function will help characterize cognitively resilient and high-risk declining subpopulations contributing to precision medicine strategies. We estimated episodic memory trajectories in 35,245 ethnically diverse older adults representing eight independent cohorts. We conducted apolipoprotein E (APOE)-stratified genome-wide association study (GWAS) analyses and combined individual cohorts' results via meta-analysis. Three independent transcriptomics datasets were used to further interpret GWAS signals. We identified DCDC2 gene significantly associated with episodic memory (Pmeta = 3.3 x 10-8 ) among non-carriers of APOE ε4 (N = 24,941). Brain transcriptomics revealed an association between episodic memory maintenance and (1) increased dorsolateral prefrontal cortex DCDC2 expression (P = 3.8 x 10-4 ) and (2) lower burden of pathological Alzheimer's disease (AD) hallmarks (paired helical fragment tau P = .003, and amyloid beta load P = .008). Additional transcriptomics results comparing AD and cognitively healthy brain samples showed a downregulation of DCDC2 levels in superior temporal gyrus (P = .007) and inferior frontal gyrus (P = .013). Our work identified DCDC2 gene as a novel predictor of memory maintenance.
Collapse
Affiliation(s)
- Yizhe Gao
- Taub Institute for Research on Alzheimer’s Disease
and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia
University, New York, NY, USA
| | - Daniel Felsky
- Krembil Centre for Neuroinformatics, Centre for Addiction
and Mental Health, Toronto, ON, Canada.,Department of Psychiatry & Institute of Medical
Science, University of Toronto, Toronto, ON, Canada
| | - Dolly Reyes-Dumeyer
- Taub Institute for Research on Alzheimer’s Disease
and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia
University, New York, NY, USA.,G.H. Sergievsky Center, Vagelos College of Physicians and
Surgeons, Columbia University, New York, NY, USA.,Department of Neurology, Vagelos College of Physicians and
Surgeons, New York-Presbyterian Hospital, Columbia University Medical Center, New
York, NY, USA
| | - Sanjeev Sariya
- Taub Institute for Research on Alzheimer’s Disease
and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia
University, New York, NY, USA
| | - Miguel Arce Rentería
- Taub Institute for Research on Alzheimer’s Disease
and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia
University, New York, NY, USA.,Department of Neurology, Vagelos College of Physicians and
Surgeons, New York-Presbyterian Hospital, Columbia University Medical Center, New
York, NY, USA
| | - Yiyi Ma
- Center for Translational & Computational
Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center,
New York, NY, 10032, USA
| | - Hans-Ulrich Klein
- Taub Institute for Research on Alzheimer’s Disease
and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia
University, New York, NY, USA.,Center for Translational & Computational
Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center,
New York, NY, 10032, USA
| | - Stephanie Cosentino
- Taub Institute for Research on Alzheimer’s Disease
and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia
University, New York, NY, USA.,G.H. Sergievsky Center, Vagelos College of Physicians and
Surgeons, Columbia University, New York, NY, USA.,Department of Neurology, Vagelos College of Physicians and
Surgeons, New York-Presbyterian Hospital, Columbia University Medical Center, New
York, NY, USA
| | - Philip L. De Jager
- Taub Institute for Research on Alzheimer’s Disease
and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia
University, New York, NY, USA.,Center for Translational & Computational
Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center,
New York, NY, 10032, USA.,Cell Circuits Program, Broad Institute, Cambridge, MA,
USA
| | - David A. Bennett
- Rush University Medical Center, Rush Alzheimer’s
Disease Center, Chicago, IL, USA.,Rush University Medical Center, Department of Neurological
Sciences, Chicago, IL, USA
| | - Adam M. Brickman
- Taub Institute for Research on Alzheimer’s Disease
and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia
University, New York, NY, USA.,G.H. Sergievsky Center, Vagelos College of Physicians and
Surgeons, Columbia University, New York, NY, USA.,Department of Neurology, Vagelos College of Physicians and
Surgeons, New York-Presbyterian Hospital, Columbia University Medical Center, New
York, NY, USA
| | - Gerard D. Schellenberg
- Department of Pathology and Laboratory Medicine,
University of Pennsylvania, Philadelphia, PA, USA
| | - Richard Mayeux
- Taub Institute for Research on Alzheimer’s Disease
and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia
University, New York, NY, USA.,G.H. Sergievsky Center, Vagelos College of Physicians and
Surgeons, Columbia University, New York, NY, USA.,Department of Neurology, Vagelos College of Physicians and
Surgeons, New York-Presbyterian Hospital, Columbia University Medical Center, New
York, NY, USA
| | - Sandra Barral
- Taub Institute for Research on Alzheimer’s Disease
and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia
University, New York, NY, USA.,G.H. Sergievsky Center, Vagelos College of Physicians and
Surgeons, Columbia University, New York, NY, USA.,Department of Neurology, Vagelos College of Physicians and
Surgeons, New York-Presbyterian Hospital, Columbia University Medical Center, New
York, NY, USA
| | -
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| |
Collapse
|
16
|
Aliev F, Barr PB, Davies AG, Dick DM, Bettinger J. Genes regulating levels of ω-3 long-chain polyunsaturated fatty acids are associated with alcohol use disorder and consumption, and broader externalizing behavior in humans. Alcohol Clin Exp Res 2022; 46:1657-1664. [PMID: 35904282 PMCID: PMC9509483 DOI: 10.1111/acer.14916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 06/07/2022] [Accepted: 07/19/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND Individual variation in the physiological response to alcohol is predictive of an individual's likelihood to develop alcohol use disorder (AUD). Evidence from diverse model organisms indicates that the levels of long-chain polyunsaturated omega-3 fatty acids (ω-3 LC-PUFAs) can modulate the behavioral response to ethanol and therefore may impact the propensity to develop AUD. While most ω-3 LC-PUFAs come from diet, humans can produce these fatty acids from shorter chain precursors through a series of enzymatic steps. Natural variation in the genes encoding these enzymes has been shown to affect ω-3 LC-PUFA levels. We hypothesized that variation in these genes could contribute to the susceptibility to develop AUD. METHODS We identified nine genes (FADS1, FADS2, FADS3, ELOVL2, GCKR, ELOVL1, ACOX1, APOE, and PPARA) that are required to generate ω-3 LC-PUFAs and/or have been shown or predicted to affect ω-3 LC-PUFA levels. Using both set-based and gene-based analyses we examined their association with AUD and two AUD-related phenotypes, alcohol consumption, and an externalizing phenotype. RESULTS We found that the set of nine genes is associated with all three phenotypes. When examined individually, GCKR, FADS2, and ACOX1 showed significant association signals with alcohol consumption. GCKR was significantly associated with AUD. ELOVL1 and APOE were associated with externalizing. CONCLUSIONS Taken together with observations that dietary ω-3 LC-PUFAs can affect ethanol-related phenotypes, this work suggests that these fatty acids provide a link between the environmental and genetic influences on the risk of developing AUD.
Collapse
Affiliation(s)
- Fazil Aliev
- Department of PsychologyVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Peter B. Barr
- Department of PsychologyVirginia Commonwealth UniversityRichmondVirginiaUSA
- Department of Psychiatry & Behavioral SciencesSUNY Downstate Health Sciences UniversityBrooklynNew YorkUSA
| | - Andrew G. Davies
- Department of Pharmacology and ToxicologyVirginia Commonwealth UniversityRichmondVirginiaUSA
- Virginia Commonwealth University Alcohol Research CenterRichmondVirginiaUSA
| | - Danielle M. Dick
- Department of PsychologyVirginia Commonwealth UniversityRichmondVirginiaUSA
- Virginia Commonwealth University Alcohol Research CenterRichmondVirginiaUSA
| | - Jill C. Bettinger
- Department of Pharmacology and ToxicologyVirginia Commonwealth UniversityRichmondVirginiaUSA
- Virginia Commonwealth University Alcohol Research CenterRichmondVirginiaUSA
| |
Collapse
|
17
|
Belonogova NM, Svishcheva GR, Kirichenko AV, Zorkoltseva IV, Tsepilov YA, Axenovich TI. sumSTAAR: A flexible framework for gene-based association studies using GWAS summary statistics. PLoS Comput Biol 2022; 18:e1010172. [PMID: 35653402 PMCID: PMC9197066 DOI: 10.1371/journal.pcbi.1010172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 06/14/2022] [Accepted: 05/05/2022] [Indexed: 11/19/2022] Open
Abstract
Gene-based association analysis is an effective gene-mapping tool. Many gene-based methods have been proposed recently. However, their power depends on the underlying genetic architecture, which is rarely known in complex traits, and so it is likely that a combination of such methods could serve as a universal approach. Several frameworks combining different gene-based methods have been developed. However, they all imply a fixed set of methods, weights and functional annotations. Moreover, most of them use individual phenotypes and genotypes as input data. Here, we introduce sumSTAAR, a framework for gene-based association analysis using summary statistics obtained from genome-wide association studies (GWAS). It is an extended and modified version of STAAR framework proposed by Li and colleagues in 2020. The sumSTAAR framework offers a wider range of gene-based methods to combine. It allows the user to arbitrarily define a set of these methods, weighting functions and probabilities of genetic variants being causal. The methods used in the framework were adapted to analyse genes with large number of SNPs to decrease the running time. The framework includes the polygene pruning procedure to guard against the influence of the strong GWAS signals outside the gene. We also present new improved matrices of correlations between the genotypes of variants within genes. These matrices estimated on a sample of 265,000 individuals are a state-of-the-art replacement of widely used matrices based on the 1000 Genomes Project data.
Collapse
Affiliation(s)
- Nadezhda M. Belonogova
- Laboratory of Segregation and Recombination Analyses, Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Gulnara R. Svishcheva
- Laboratory of Segregation and Recombination Analyses, Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
- Laboratory of Animal Genetics, Vavilov Institute of General Genetics, the Russian Academy of Sciences, Moscow, Russia
| | - Anatoly V. Kirichenko
- Laboratory of Segregation and Recombination Analyses, Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Irina V. Zorkoltseva
- Laboratory of Segregation and Recombination Analyses, Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Yakov A. Tsepilov
- Laboratory of Segregation and Recombination Analyses, Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
- Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russia
| | - Tatiana I. Axenovich
- Laboratory of Segregation and Recombination Analyses, Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
18
|
Chimusa ER, Defo J. Dissecting Meta-Analysis in GWAS Era: Bayesian Framework for Gene/Subnetwork-Specific Meta-Analysis. Front Genet 2022; 13:838518. [PMID: 35664319 PMCID: PMC9159898 DOI: 10.3389/fgene.2022.838518] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 04/07/2022] [Indexed: 11/13/2022] Open
Abstract
Over the past decades, advanced high-throughput technologies have continuously contributed to genome-wide association studies (GWASs). GWAS meta-analysis has been increasingly adopted, has cross-ancestry replicability, and has power to illuminate the genetic architecture of complex traits, informing about the reliability of estimation effects and their variability across human ancestries. However, detecting genetic variants that have low disease risk still poses a challenge. Designing a meta-analysis approach that combines the effect of various SNPs within genes or genes within pathways from multiple independent population GWASs may be helpful in identifying associations with small effect sizes and increasing the association power. Here, we proposed ancMETA, a Bayesian graph-based framework, to perform the gene/pathway-specific meta-analysis by combining the effect size of multiple SNPs within genes, and genes within subnetwork/pathways across multiple independent population GWASs to deconvolute the interactions between genes underlying the pathogenesis of complex diseases across human populations. We assessed the proposed framework on simulated datasets, and the results show that the proposed model holds promise for increasing statistical power for meta-analysis of genetic variants underlying the pathogenesis of complex diseases. To illustrate the proposed meta-analysis framework, we leverage seven different European bipolar disorder (BD) cohorts, and we identify variants in the angiotensinogen (AGT) gene to be significantly associated with BD across all 7 studies. We detect a commonly significant BD-specific subnetwork with the ESR1 gene as the main hub of a subnetwork, associated with neurotrophin signaling (p = 4e−14) and myometrial relaxation and contraction (p = 3e−08) pathways. ancMETA provides a new contribution to post-GWAS methodologies and holds promise for comprehensively examining interactions between genes underlying the pathogenesis of genetic diseases and also underlying ethnic differences.
Collapse
|
19
|
Cao X, Wang X, Zhang S, Sha Q. Gene-based association tests using GWAS summary statistics and incorporating eQTL. Sci Rep 2022; 12:3553. [PMID: 35241742 PMCID: PMC8894384 DOI: 10.1038/s41598-022-07465-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 02/11/2022] [Indexed: 01/29/2023] Open
Abstract
Although genome-wide association studies (GWAS) have been successfully applied to a variety of complex diseases and identified many genetic variants underlying complex diseases via single marker tests, there is still a considerable heritability of complex diseases that could not be explained by GWAS. One alternative approach to overcome the missing heritability caused by genetic heterogeneity is gene-based analysis, which considers the aggregate effects of multiple genetic variants in a single test. Another alternative approach is transcriptome-wide association study (TWAS). TWAS aggregates genomic information into functionally relevant units that map to genes and their expression. TWAS is not only powerful, but can also increase the interpretability in biological mechanisms of identified trait associated genes. In this study, we propose a powerful and computationally efficient gene-based association test, called Overall. Using extended Simes procedure, Overall aggregates information from three types of traditional gene-based association tests and also incorporates expression quantitative trait locus (eQTL) information into a gene-based association test using GWAS summary statistics. We show that after a small number of replications to estimate the correlation among the integrated gene-based tests, the p values of Overall can be calculated analytically. Simulation studies show that Overall can control type I error rates very well and has higher power than the tests that we compared with. We also apply Overall to two schizophrenia GWAS summary datasets and two lipids GWAS summary datasets. The results show that this newly developed method can identify more significant genes than other methods we compared with.
Collapse
Affiliation(s)
- Xuewei Cao
- Department of Mathematical Sciences, Michigan Technological University, Houghton, MI, 49931, USA
| | - Xuexia Wang
- Department of Mathematics, University of North Texas, Denton, TX, USA
| | - Shuanglin Zhang
- Department of Mathematical Sciences, Michigan Technological University, Houghton, MI, 49931, USA
| | - Qiuying Sha
- Department of Mathematical Sciences, Michigan Technological University, Houghton, MI, 49931, USA.
| |
Collapse
|
20
|
Identification of key biomarkers and signaling pathways and analysis of their association with immune cells in immunoglobulin A nephropathy. Cent Eur J Immunol 2022; 47:189-205. [PMID: 36817268 PMCID: PMC9896983 DOI: 10.5114/ceji.2022.119867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/10/2022] [Indexed: 11/17/2022] Open
Abstract
Introduction Immunoglobulin A nephropathy (IgAN) is the most common glomerular disease worldwide, with a poor prognosis. The aim of our study was to identify key biomarkers and their associations with immune cells to aid in the study of IgAN pathology and immunotherapy. Material and methods The data of IgAN were downloaded from a public database. The metaMA package and limma package were used to identify differentially expressed mRNAs (DEmRNAs) and differentially expressed miRNAs (DEmiRNAs), respectively. Biological functions of the DEmRNAs were analyzed. Machine learning was used to screen the mRNA biomarkers of IgAN. Pearson's correlation coefficient was used to analyze the correlation between mRNA biomarkers, immune cells and signaling pathways. Moreover, we constructed a miRNAs-mRNAs targeted regulatory network. Finally, we performed in vitro validation of the identified miRNAs and mRNAs. Results 1205 DEmRNAs and 125 DEmiRNAs were identified. In gene set enrichment analysis (GSEA), tumor necrosis factor α (TNF-α) signaling via nuclear factor κB (NF-κB), apoptosis and MTORC-1 signaling were inhibited in IgAN. 8 mRNA biomarkers were screened by machine learning. In addition, the distribution of 8 immune cell types was found to be significantly different between normal controls and IgAN by difference analysis. Pearson correlation coefficient analysis demonstrated that AKAP8L was significantly negatively correlated with CD4+ memory T-cells. AKAP8L was also significantly negatively correlated with TNF-α signaling via NF-κB, apoptosis, and MTORC-1 signaling. Subsequently, 5 mRNA biomarkers predicted corresponding negative regulatory miRNAs. Conclusions The identification of 8 important biomarkers and their correlation with immune cells and biological signaling pathways provides new ideas for further study of IgAN.
Collapse
|
21
|
Maddipati R, Norgard RJ, Baslan T, Rathi KS, Zhang A, Saeid A, Higashihara T, Wu F, Kumar A, Annamalai V, Bhattacharya S, Raman P, Adkisson CA, Pitarresi JR, Wengyn MD, Yamazoe T, Li J, Balli D, LaRiviere MJ, Ngo TVC, Folkert IW, Millstein ID, Bermeo J, Carpenter EL, McAuliffe JC, Oktay MH, Brekken RA, Lowe SW, Iacobuzio-Donahue CA, Notta F, Stanger BZ. MYC levels regulate metastatic heterogeneity in pancreatic adenocarcinoma. Cancer Discov 2021; 12:542-561. [PMID: 34551968 PMCID: PMC8831468 DOI: 10.1158/2159-8290.cd-20-1826] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 07/26/2021] [Accepted: 09/17/2021] [Indexed: 11/16/2022]
Abstract
The degree of metastatic disease varies widely amongst cancer patients and impacts clinical outcomes. However, the biological and functional differences that drive the extent of metastasis are poorly understood. We analyzed primary tumors and paired metastases using a multi-fluorescent lineage-labeled mouse model of pancreatic ductal adenocarcinoma (PDAC) - a tumor type where most patients present with metastases. Genomic and transcriptomic analysis revealed an association between metastatic burden and gene amplification or transcriptional upregulation of MYC and its downstream targets. Functional experiments showed that MYC promotes metastasis by recruiting tumor associated macrophages (TAMs), leading to greater bloodstream intravasation. Consistent with these findings, metastatic progression in human PDAC was associated with activation of MYC signaling pathways and enrichment for MYC amplifications specifically in metastatic patients. Collectively, these results implicate MYC activity as a major determinant of metastatic burden in advanced PDAC.
Collapse
Affiliation(s)
| | - Robert J Norgard
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Timour Baslan
- Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center
| | - Komal S Rathi
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia
| | - Amy Zhang
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research
| | - Asal Saeid
- The University of Texas Southwestern Medical Center
| | | | - Feng Wu
- The University of Texas Southwestern Medical Center
| | - Angad Kumar
- Internal Medicine, The University of Texas Southwestern Medical Center
| | - Valli Annamalai
- Department of Internal Medicine, The University of Texas Southwestern Medical Center
| | | | | | | | | | | | - Taiji Yamazoe
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Jinyang Li
- School of Medicine, University of Pennsylvania
| | - David Balli
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | | | - Tuong-Vi C Ngo
- Division of Surgical Oncology, Department of Surgery, and Hamon Center for Therapeutic Oncology Research, The University of Texas Southwestern Medical Center
| | | | - Ian D Millstein
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Jonathan Bermeo
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center
| | | | - John C McAuliffe
- Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center
| | | | - Rolf A Brekken
- Hamon Center for Therapeutic Oncology Research, Departments of Surgery and Pharmacology, UT Southwestern Medical Center at Dallas
| | - Scott W Lowe
- Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center
| | | | | | - Ben Z Stanger
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania
| |
Collapse
|
22
|
Blériot C, Barreby E, Dunsmore G, Ballaire R, Chakarov S, Ficht X, De Simone G, Andreata F, Fumagalli V, Guo W, Wan G, Gessain G, Khalilnezhad A, Zhang XM, Ang N, Chen P, Morgantini C, Azzimato V, Kong WT, Liu Z, Pai R, Lum J, Shihui F, Low I, Xu C, Malleret B, Kairi MFM, Balachander A, Cexus O, Larbi A, Lee B, Newell EW, Ng LG, Phoo WW, Sobota RM, Sharma A, Howland SW, Chen J, Bajenoff M, Yvan-Charvet L, Venteclef N, Iannacone M, Aouadi M, Ginhoux F. A subset of Kupffer cells regulates metabolism through the expression of CD36. Immunity 2021; 54:2101-2116.e6. [PMID: 34469775 DOI: 10.1016/j.immuni.2021.08.006] [Citation(s) in RCA: 132] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 04/27/2021] [Accepted: 08/09/2021] [Indexed: 12/11/2022]
Abstract
Tissue macrophages are immune cells whose phenotypes and functions are dictated by origin and niches. However, tissues are complex environments, and macrophage heterogeneity within the same organ has been overlooked so far. Here, we used high-dimensional approaches to characterize macrophage populations in the murine liver. We identified two distinct populations among embryonically derived Kupffer cells (KCs) sharing a core signature while differentially expressing numerous genes and proteins: a major CD206loESAM- population (KC1) and a minor CD206hiESAM+ population (KC2). KC2 expressed genes involved in metabolic processes, including fatty acid metabolism both in steady-state and in diet-induced obesity and hepatic steatosis. Functional characterization by depletion of KC2 or targeted silencing of the fatty acid transporter Cd36 highlighted a crucial contribution of KC2 in the liver oxidative stress associated with obesity. In summary, our study reveals that KCs are more heterogeneous than anticipated, notably describing a subpopulation wired with metabolic functions.
Collapse
Affiliation(s)
- Camille Blériot
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore; Inserm U1015, Gustave Roussy, Villejuif 94800, France.
| | - Emelie Barreby
- Center for Infectious Medicine, Department of Medicine, Karolinska Institute, Huddinge 14157, Sweden
| | | | | | - Svetoslav Chakarov
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore; Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xenia Ficht
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Giorgia De Simone
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy; Vita-Salute San Raffaele University, Milan 20132, Italy
| | - Francesco Andreata
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Valeria Fumagalli
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy; Vita-Salute San Raffaele University, Milan 20132, Italy
| | - Wei Guo
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Guochen Wan
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Gregoire Gessain
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Ahad Khalilnezhad
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore; Department of Microbiology and Immunology, Immunology Translational Research Program, Yong Loo Lin School of Medicine, Immunology Program, Life Sciences Institute, National University of Singapore, Singapore 117543, Singapore
| | - Xiao Meng Zhang
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Nicholas Ang
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Ping Chen
- Center for Infectious Medicine, Department of Medicine, Karolinska Institute, Huddinge 14157, Sweden
| | - Cecilia Morgantini
- Center for Infectious Medicine, Department of Medicine, Karolinska Institute, Huddinge 14157, Sweden
| | - Valerio Azzimato
- Center for Infectious Medicine, Department of Medicine, Karolinska Institute, Huddinge 14157, Sweden
| | - Wan Ting Kong
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Zhaoyuan Liu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Rhea Pai
- Genome Institute of Singapore, A(∗)STAR, Singapore 138672, Singapore
| | - Josephine Lum
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Foo Shihui
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Ivy Low
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Connie Xu
- Center for Infectious Medicine, Department of Medicine, Karolinska Institute, Huddinge 14157, Sweden
| | - Benoit Malleret
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore; Department of Microbiology and Immunology, Immunology Translational Research Program, Yong Loo Lin School of Medicine, Immunology Program, Life Sciences Institute, National University of Singapore, Singapore 117543, Singapore
| | - Muhammad Faris Mohd Kairi
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Akhila Balachander
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Olivier Cexus
- Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, United Kingdom
| | - Anis Larbi
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Bernett Lee
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Evan W Newell
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Lai Guan Ng
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore; Department of Microbiology and Immunology, Immunology Translational Research Program, Yong Loo Lin School of Medicine, Immunology Program, Life Sciences Institute, National University of Singapore, Singapore 117543, Singapore
| | - Wint Wint Phoo
- Functional Proteomics Laboratory, SingMass National Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A(∗)STAR), Singapore 138673, Singapore
| | - Radoslaw M Sobota
- Functional Proteomics Laboratory, SingMass National Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A(∗)STAR), Singapore 138673, Singapore
| | - Ankur Sharma
- Genome Institute of Singapore, A(∗)STAR, Singapore 138672, Singapore
| | - Shanshan W Howland
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Jinmiao Chen
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Marc Bajenoff
- Aix Marseille University, CNRS, INSERM, CIML, Marseille 13288, France
| | | | - Nicolas Venteclef
- Centre de Recherche des Cordeliers, INSERM, Université de Paris, Sorbonne Université, IMMEDIAB Laboratory, Paris 75006, France
| | - Matteo Iannacone
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy; Vita-Salute San Raffaele University, Milan 20132, Italy; Experimental Imaging Centre, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Myriam Aouadi
- Center for Infectious Medicine, Department of Medicine, Karolinska Institute, Huddinge 14157, Sweden
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore; Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Microbiology and Immunology, Immunology Translational Research Program, Yong Loo Lin School of Medicine, Immunology Program, Life Sciences Institute, National University of Singapore, Singapore 117543, Singapore; Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore 169856, Singapore.
| |
Collapse
|
23
|
Raj S, Thalamuthu A, Armstrong NJ, Wright MJ, Kwok JB, Trollor JN, Ames D, Schofield PR, Brodaty H, Sachdev PS, Mather KA. Investigating Olfactory Gene Variation and Odour Identification in Older Adults. Genes (Basel) 2021; 12:genes12050669. [PMID: 33946865 PMCID: PMC8145954 DOI: 10.3390/genes12050669] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/27/2021] [Accepted: 04/27/2021] [Indexed: 11/16/2022] Open
Abstract
Ageing is associated with a decrease in odour identification. Additionally, deficits in olfaction have been linked to age-related disease and mortality. Heritability studies suggest genetic variation contributes to olfactory identification. The olfactory receptor (OR) gene family is the largest in the human genome and responsible for overall odour identification. In this study, we sought to find olfactory gene family variants associated with individual and overall odour identification and to examine the relationships between polygenic risk scores (PRS) for olfactory-related phenotypes and olfaction. Participants were Caucasian older adults from the Sydney Memory and Ageing Study and the Older Australian Twins Study with genome-wide genotyping data (n = 1395, mean age = 75.52 ± 6.45). The Brief-Smell Identification Test (BSIT) was administered in both cohorts. PRS were calculated from independent GWAS summary statistics for Alzheimer’s disease (AD), white matter hyperintensities (WMH), Parkinson’s disease (PD), hippocampal volume and smoking. Associations with olfactory receptor genes (n = 967), previously identified candidate olfaction-related SNPs (n = 36) and different PRS with BSIT scores (total and individual smells) were examined. All of the relationships were analysed using generalised linear mixed models (GLMM), adjusted for age and sex. Genes with suggestive evidence for odour identification were found for 8 of the 12 BSIT items. Thirteen out of 36 candidate SNPs previously identified from the literature were suggestively associated with several individual BSIT items but not total score. PRS for smoking, WMH and PD were negatively associated with chocolate identification. This is the first study to conduct genetic analyses with individual odorant identification, which found suggestive olfactory-related genes and genetic variants for multiple individual BSIT odours. Replication in independent and larger cohorts is needed.
Collapse
Affiliation(s)
- Siddharth Raj
- Centre for Healthy Brain Ageing, Faculty of Medicine, School of Psychiatry, University of New South Wales (UNSW), Sydney, NSW 2031, Australia; (S.R.); (A.T.); (J.N.T.); (H.B.); (P.S.S.)
| | - Anbupalam Thalamuthu
- Centre for Healthy Brain Ageing, Faculty of Medicine, School of Psychiatry, University of New South Wales (UNSW), Sydney, NSW 2031, Australia; (S.R.); (A.T.); (J.N.T.); (H.B.); (P.S.S.)
| | - Nicola J Armstrong
- Department of Mathematics and Statistics, Curtin University, Perth, WA 6102, Australia;
| | - Margaret J Wright
- Queensland Brain Institute, University of Queensland, St. Lucia, QLD 4072, Australia;
- Centre for Advanced Imaging, University of Queensland, St. Lucia, QLD 4072, Australia
| | - John B Kwok
- School of Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia;
| | - Julian N Trollor
- Centre for Healthy Brain Ageing, Faculty of Medicine, School of Psychiatry, University of New South Wales (UNSW), Sydney, NSW 2031, Australia; (S.R.); (A.T.); (J.N.T.); (H.B.); (P.S.S.)
- Department of Developmental Disability Neuropsychiatry, UNSW, Sydney, NSW 2031, Australia
| | - David Ames
- National Ageing Research Institute, Parkville, VIC 3052, Australia;
- Academic Unit for Psychiatry of Old Age, University of Melbourne, St George’s Hospital, Kew, Melbourne, VIC 3010, Australia
| | - Peter R Schofield
- Neuroscience Research Australia, Sydney, NSW 2031, Australia;
- School of Medical Sciences, UNSW, Sydney, NSW 2031, Australia
| | - Henry Brodaty
- Centre for Healthy Brain Ageing, Faculty of Medicine, School of Psychiatry, University of New South Wales (UNSW), Sydney, NSW 2031, Australia; (S.R.); (A.T.); (J.N.T.); (H.B.); (P.S.S.)
- Dementia Collaborative Research Centre Assessment and Better Care, UNSW, Sydney, NSW 2031, Australia
| | - Perminder S Sachdev
- Centre for Healthy Brain Ageing, Faculty of Medicine, School of Psychiatry, University of New South Wales (UNSW), Sydney, NSW 2031, Australia; (S.R.); (A.T.); (J.N.T.); (H.B.); (P.S.S.)
- Neuropsychiatric Institute, the Prince of Wales Hospital, Sydney, NSW 2031, Australia
| | - Karen A Mather
- Centre for Healthy Brain Ageing, Faculty of Medicine, School of Psychiatry, University of New South Wales (UNSW), Sydney, NSW 2031, Australia; (S.R.); (A.T.); (J.N.T.); (H.B.); (P.S.S.)
- Neuroscience Research Australia, Sydney, NSW 2031, Australia;
- Correspondence: ; Tel.: +61-(2)-9065-1347
| |
Collapse
|
24
|
Swanzey E, O'Connor C, Reinholdt LG. Mouse Genetic Reference Populations: Cellular Platforms for Integrative Systems Genetics. Trends Genet 2021; 37:251-265. [PMID: 33010949 PMCID: PMC7889615 DOI: 10.1016/j.tig.2020.09.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/28/2020] [Accepted: 09/02/2020] [Indexed: 02/07/2023]
Abstract
Interrogation of disease-relevant cellular and molecular traits exhibited by genetically diverse cell populations enables in vitro systems genetics approaches for uncovering the basic properties of cellular function and identity. Primary cells, stem cells, and organoids derived from genetically diverse mouse strains, such as Collaborative Cross and Diversity Outbred populations, offer the opportunity for parallel in vitro/in vivo screening. These panels provide genetic resolution for variant discovery and functional characterization, as well as disease modeling and in vivo validation capabilities. Here we review mouse cellular systems genetics approaches for characterizing the influence of genetic variation on signaling networks and phenotypic diversity, and we discuss approaches for data integration and cross-species validation.
Collapse
Affiliation(s)
| | - Callan O'Connor
- The Jackson Laboratory, Bar Harbor, ME, USA; Tufts Graduate School of Biomedical Sciences, Boston, MA, USA
| | | |
Collapse
|
25
|
Climente-González H, Lonjou C, Lesueur F, Stoppa-Lyonnet D, Andrieu N, Azencott CA. Boosting GWAS using biological networks: A study on susceptibility to familial breast cancer. PLoS Comput Biol 2021; 17:e1008819. [PMID: 33735170 PMCID: PMC8009366 DOI: 10.1371/journal.pcbi.1008819] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 03/30/2021] [Accepted: 02/18/2021] [Indexed: 11/20/2022] Open
Abstract
Genome-wide association studies (GWAS) explore the genetic causes of complex diseases. However, classical approaches ignore the biological context of the genetic variants and genes under study. To address this shortcoming, one can use biological networks, which model functional relationships, to search for functionally related susceptibility loci. Many such network methods exist, each arising from different mathematical frameworks, pre-processing steps, and assumptions about the network properties of the susceptibility mechanism. Unsurprisingly, this results in disparate solutions. To explore how to exploit these heterogeneous approaches, we selected six network methods and applied them to GENESIS, a nationwide French study on familial breast cancer. First, we verified that network methods recovered more interpretable results than a standard GWAS. We addressed the heterogeneity of their solutions by studying their overlap, computing what we called the consensus. The key gene in this consensus solution was COPS5, a gene related to multiple cancer hallmarks. Another issue we observed was that network methods were unstable, selecting very different genes on different subsamples of GENESIS. Therefore, we proposed a stable consensus solution formed by the 68 genes most consistently selected across multiple subsamples. This solution was also enriched in genes known to be associated with breast cancer susceptibility (BLM, CASP8, CASP10, DNAJC1, FGFR2, MRPS30, and SLC4A7, P-value = 3 × 10-4). The most connected gene was CUL3, a regulator of several genes linked to cancer progression. Lastly, we evaluated the biases of each method and the impact of their parameters on the outcome. In general, network methods preferred highly connected genes, even after random rewirings that stripped the connections of any biological meaning. In conclusion, we present the advantages of network-guided GWAS, characterize their shortcomings, and provide strategies to address them. To compute the consensus networks, implementations of all six methods are available at https://github.com/hclimente/gwas-tools.
Collapse
Affiliation(s)
- Héctor Climente-González
- Institut Curie, PSL Research University, Paris, France
- INSERM, U900, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, Paris, France
- RIKEN Center for Advanced Intelligence Project (AIP), Tokyo, Japan
| | - Christine Lonjou
- Institut Curie, PSL Research University, Paris, France
- INSERM, U900, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, Paris, France
| | - Fabienne Lesueur
- Institut Curie, PSL Research University, Paris, France
- INSERM, U900, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, Paris, France
| | | | - Dominique Stoppa-Lyonnet
- Service de Génétique, Institut Curie, Paris, France
- INSERM, U830, Paris, France
- Université Paris Descartes, Paris, France
| | - Nadine Andrieu
- Institut Curie, PSL Research University, Paris, France
- INSERM, U900, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, Paris, France
| | - Chloé-Agathe Azencott
- Institut Curie, PSL Research University, Paris, France
- INSERM, U900, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, Paris, France
| |
Collapse
|
26
|
Zhang M, Yang Q, Yuan X, Yan X, Wang J, Cheng T, Zhang Q. Integrating Genome-Wide Association Analysis With Transcriptome Sequencing to Identify Candidate Genes Related to Blooming Time in Prunus mume. FRONTIERS IN PLANT SCIENCE 2021; 12:690841. [PMID: 34335659 PMCID: PMC8319914 DOI: 10.3389/fpls.2021.690841] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 05/28/2021] [Indexed: 05/12/2023]
Abstract
Prunus mume is one of the most important woody perennials for edible and ornamental use. Despite a substantial variation in the flowering phenology among the P. mume germplasm resources, the genetic control for flowering time remains to be elucidated. In this study, we examined five blooming time-related traits of 235 P. mume landraces for 2 years. Based on the phenotypic data, we performed genome-wide association studies, which included a combination of marker- and gene-based association tests, and identified 1,445 candidate genes that are consistently linked with flowering time across multiple years. Furthermore, we assessed the global transcriptome change of floral buds from the two P. mume cultivars exhibiting contrasting bloom dates and detected 617 associated genes that were differentially expressed during the flowering process. By integrating a co-expression network analysis, we screened out 191 gene candidates of conserved transcriptional pattern during blooming across cultivars. Finally, we validated the temporal expression profiles of these candidates and highlighted their putative roles in regulating floral bud break and blooming time in P. mume. Our findings are important to expand the understanding of flowering time control in woody perennials and will boost the molecular breeding of novel varieties in P. mume.
Collapse
Affiliation(s)
- Man Zhang
- National Engineering Research Center for Floriculture, Beijing Key Laboratory of Ornamental Plants Germplasm Innovation & Molecular Breeding, Beijing Laboratory of Urban and Rural Ecological Environment, Key Laboratory of Genetics and Breeding in Forest Trees and Ornamental Plants of Ministry of Education, School of Landscape Architecture, Beijing Forestry University, Beijing, China
| | - Qingqing Yang
- National Engineering Research Center for Floriculture, Beijing Key Laboratory of Ornamental Plants Germplasm Innovation & Molecular Breeding, Beijing Laboratory of Urban and Rural Ecological Environment, Key Laboratory of Genetics and Breeding in Forest Trees and Ornamental Plants of Ministry of Education, School of Landscape Architecture, Beijing Forestry University, Beijing, China
| | - Xi Yuan
- National Engineering Research Center for Floriculture, Beijing Key Laboratory of Ornamental Plants Germplasm Innovation & Molecular Breeding, Beijing Laboratory of Urban and Rural Ecological Environment, Key Laboratory of Genetics and Breeding in Forest Trees and Ornamental Plants of Ministry of Education, School of Landscape Architecture, Beijing Forestry University, Beijing, China
| | | | - Jia Wang
- National Engineering Research Center for Floriculture, Beijing Key Laboratory of Ornamental Plants Germplasm Innovation & Molecular Breeding, Beijing Laboratory of Urban and Rural Ecological Environment, Key Laboratory of Genetics and Breeding in Forest Trees and Ornamental Plants of Ministry of Education, School of Landscape Architecture, Beijing Forestry University, Beijing, China
| | - Tangren Cheng
- National Engineering Research Center for Floriculture, Beijing Key Laboratory of Ornamental Plants Germplasm Innovation & Molecular Breeding, Beijing Laboratory of Urban and Rural Ecological Environment, Key Laboratory of Genetics and Breeding in Forest Trees and Ornamental Plants of Ministry of Education, School of Landscape Architecture, Beijing Forestry University, Beijing, China
| | - Qixiang Zhang
- National Engineering Research Center for Floriculture, Beijing Key Laboratory of Ornamental Plants Germplasm Innovation & Molecular Breeding, Beijing Laboratory of Urban and Rural Ecological Environment, Key Laboratory of Genetics and Breeding in Forest Trees and Ornamental Plants of Ministry of Education, School of Landscape Architecture, Beijing Forestry University, Beijing, China
- *Correspondence: Qixiang Zhang
| |
Collapse
|
27
|
Svishcheva GR, Belonogova NM, Zorkoltseva IV, Kirichenko AV, Axenovich TI. Gene-based association tests using GWAS summary statistics. Bioinformatics 2020; 35:3701-3708. [PMID: 30860568 DOI: 10.1093/bioinformatics/btz172] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 02/12/2019] [Accepted: 03/11/2019] [Indexed: 01/09/2023] Open
Abstract
MOTIVATION A huge number of genome-wide association studies (GWAS) summary statistics freely available in databases provide a new material for gene-based association analysis aimed at identifying rare genetic variants. Only a few of the many popular gene-based methods developed for individual genotype and phenotype data are adapted for the practical use of the GWAS summary statistics as input. RESULTS We analytically prove and numerically illustrate that all popular powerful methods developed for gene-based association analysis of individual phenotype and genotype data can be modified to utilize GWAS summary statistics. We have modified and implemented all of the popular methods, including burden and kernel machine-based tests, multiple and functional linear regression, principal components analysis and others, in the R package sumFREGAT. Using real summary statistics for coronary artery disease, we show that the new package is able to detect genes not found by the existing packages. AVAILABILITY AND IMPLEMENTATION The R package sumFREGAT is freely and publicly available at: https://CRAN.R-project.org/package=sumFREGAT. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Gulnara R Svishcheva
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia.,Vavilov Institute of General Genetics, the Russian Academy of Sciences, Moscow, Russia
| | - Nadezhda M Belonogova
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Irina V Zorkoltseva
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Anatoly V Kirichenko
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Tatiana I Axenovich
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia.,Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russia.,Department of Biotechnology, L.K. Ernst Federal Center for Animal Husbandry, Dubrovitsy, Russia
| |
Collapse
|
28
|
Cheng W, Ramachandran S, Crawford L. Estimation of non-null SNP effect size distributions enables the detection of enriched genes underlying complex traits. PLoS Genet 2020; 16:e1008855. [PMID: 32542026 PMCID: PMC7316356 DOI: 10.1371/journal.pgen.1008855] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 06/25/2020] [Accepted: 05/13/2020] [Indexed: 12/22/2022] Open
Abstract
Traditional univariate genome-wide association studies generate false positives and negatives due to difficulties distinguishing associated variants from variants with spurious nonzero effects that do not directly influence the trait. Recent efforts have been directed at identifying genes or signaling pathways enriched for mutations in quantitative traits or case-control studies, but these can be computationally costly and hampered by strict model assumptions. Here, we present gene-ε, a new approach for identifying statistical associations between sets of variants and quantitative traits. Our key insight is that enrichment studies on the gene-level are improved when we reformulate the genome-wide SNP-level null hypothesis to identify spurious small-to-intermediate SNP effects and classify them as non-causal. gene-ε efficiently identifies enriched genes under a variety of simulated genetic architectures, achieving greater than a 90% true positive rate at 1% false positive rate for polygenic traits. Lastly, we apply gene-ε to summary statistics derived from six quantitative traits using European-ancestry individuals in the UK Biobank, and identify enriched genes that are in biologically relevant pathways.
Collapse
Affiliation(s)
- Wei Cheng
- Department of Ecology and Evolutionary Biology, Brown University, Providence, Rhode Island, United States of America
- Center for Computational Molecular Biology, Brown University, Providence, Rhode Island, United States of America
| | - Sohini Ramachandran
- Department of Ecology and Evolutionary Biology, Brown University, Providence, Rhode Island, United States of America
- Center for Computational Molecular Biology, Brown University, Providence, Rhode Island, United States of America
| | - Lorin Crawford
- Center for Computational Molecular Biology, Brown University, Providence, Rhode Island, United States of America
- Department of Biostatistics, Brown University, Providence, Rhode Island, United States of America
- Center for Statistical Sciences, Brown University, Providence, Rhode Island, United States of America
| |
Collapse
|
29
|
McGuirl MR, Smith SP, Sandstede B, Ramachandran S. Detecting Shared Genetic Architecture Among Multiple Phenotypes by Hierarchical Clustering of Gene-Level Association Statistics. Genetics 2020; 215:511-529. [PMID: 32245788 PMCID: PMC7268989 DOI: 10.1534/genetics.120.303096] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 03/31/2020] [Indexed: 12/31/2022] Open
Abstract
Emerging large-scale biobanks pairing genotype data with phenotype data present new opportunities to prioritize shared genetic associations across multiple phenotypes for molecular validation. Past research, by our group and others, has shown gene-level tests of association produce biologically interpretable characterization of the genetic architecture of a given phenotype. Here, we present a new method, Ward clustering to identify Internal Node branch length outliers using Gene Scores (WINGS), for identifying shared genetic architecture among multiple phenotypes. The objective of WINGS is to identify groups of phenotypes, or "clusters," sharing a core set of genes enriched for mutations in cases. We validate WINGS using extensive simulation studies and then combine gene-level association tests with WINGS to identify shared genetic architecture among 81 case-control and seven quantitative phenotypes in 349,468 European-ancestry individuals from the UK Biobank. We identify eight prioritized phenotype clusters and recover multiple published gene-level associations within prioritized clusters.
Collapse
Affiliation(s)
- Melissa R McGuirl
- Division of Applied Mathematics, Brown University, Providence, Rhode Island 02912
| | - Samuel Pattillo Smith
- Center for Computational Molecular Biology, Brown University, Providence, Rhode Island 02912
- Department of Ecology and Evolutionary Biology, Brown University, Providence, Rhode Island 02912
| | - Björn Sandstede
- Division of Applied Mathematics, Brown University, Providence, Rhode Island 02912
- Data Science Initiative, Brown University, Providence, Rhode Island 02912
| | - Sohini Ramachandran
- Center for Computational Molecular Biology, Brown University, Providence, Rhode Island 02912
- Department of Ecology and Evolutionary Biology, Brown University, Providence, Rhode Island 02912
| |
Collapse
|
30
|
Zorkoltseva IV, Belonogova NM, Svishcheva GR, Kirichenko AV, Axenovich TI. <i>In silico</i> mapping of coronary artery disease genes. Vavilovskii Zhurnal Genet Selektsii 2020. [DOI: 10.18699/vj19.585] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
To date, more than 100 loci associated with coronary artery disease (CAD) have been detected in large-scale genome-wide studies. For some of the several hundreds of genes located in these loci, roles in the pathogenesis of the disease have been shown. However, the genetic mechanisms and specific genes controlling this disease are still not fully understood. This study is aimed at in silico search for new CAD genes. We performed a gene-based association analysis, where all polymorphic variants within a gene are analyzed simultaneously. The analysis was based on the results of the genome-wide association studies (GWAS) available from the open databases MICAD (120,575 people, 85,112 markers) and UK Biobank (337,199 people, 10,894,597 markers). We used the sumFREGAT package implementing a wide range of new methods for gene-based association analysis using summary statistics. We found 88 genes demonstrating significant gene-based associations. Forty-four of the identified genes were already known as CAD genes. Furthermore, we identified 28 additional genes in the known CAD loci. They can be considered as new candidate genes. Finally, we identified sixteen new genes (AGPAT4, ARHGEF12, BDP1, DHX58, EHBP1, FBF1, HSPB9, NPBWR2, PDLIM5, PLCB3, PLEKHM2, POU2F3, PRKD2, TMEM136, TTC29 and UTP20) outside the known loci. Information about the functional role of these genes allows us to consider many of them as candidates for CAD. The 41 identified genes did not have significant GWAS signals and they were identified only due to simultaneous consideration of all variants within the gene in the framework of gene-based analysis. These results demonstrate that gene-based association analysis is a powerful tool for gene mapping. The method can utilize huge amounts of GWAS results accumulated in the world to map different traits and diseases. This type of studies is widely available, as it does not require additional material costs.
Collapse
Affiliation(s)
| | | | - G. R. Svishcheva
- Institute of Cytology and Genetics, SB RAS; Vavilov Institute of General Genetics, RAS
| | | | - T. I. Axenovich
- Institute of Cytology and Genetics, SB RAS; Novosibirsk State University
| |
Collapse
|
31
|
Network-Based Functional Prediction Augments Genetic Association To Predict Candidate Genes for Histamine Hypersensitivity in Mice. G3-GENES GENOMES GENETICS 2019; 9:4223-4233. [PMID: 31645420 PMCID: PMC6893195 DOI: 10.1534/g3.119.400740] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Genetic mapping is a primary tool of genetics in model organisms; however, many quantitative trait loci (QTL) contain tens or hundreds of positional candidate genes. Prioritizing these genes for validation is often ad hoc and biased by previous findings. Here we present a technique for prioritizing positional candidates based on computationally inferred gene function. Our method uses machine learning with functional genomic networks, whose links encode functional associations among genes, to identify network-based signatures of functional association to a trait of interest. We demonstrate the method by functionally ranking positional candidates in a large locus on mouse Chr 6 (45.9 Mb to 127.8 Mb) associated with histamine hypersensitivity (Histh). Histh is characterized by systemic vascular leakage and edema in response to histamine challenge, which can lead to multiple organ failure and death. Although Histh risk is strongly influenced by genetics, little is known about its underlying molecular or genetic causes, due to genetic and physiological complexity of the trait. To dissect this complexity, we ranked genes in the Histh locus by predicting functional association with multiple Histh-related processes. We integrated these predictions with new single nucleotide polymorphism (SNP) association data derived from a survey of 23 inbred mouse strains and congenic mapping data. The top-ranked genes included Cxcl12, Ret, Cacna1c, and Cntn3, all of which had strong functional associations and were proximal to SNPs segregating with Histh. These results demonstrate the power of network-based computational methods to nominate highly plausible quantitative trait genes even in challenging cases involving large QTL and extreme trait complexity.
Collapse
|
32
|
Lupo PJ, Mitchell LE, Jenkins MM. Genome-wide association studies of structural birth defects: A review and commentary. Birth Defects Res 2019; 111:1329-1342. [PMID: 31654503 DOI: 10.1002/bdr2.1606] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 09/21/2019] [Accepted: 10/02/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND While there is strong evidence that genetic risk factors play an important role in the etiologies of structural birth defects, compared to other diseases, there have been relatively few genome-wide association studies (GWAS) of these conditions. We reviewed the current landscape of GWAS conducted for birth defects, noting novel insights, and future directions. METHODS This article reviews the literature with regard to GWAS of structural birth defects. Key defects included in this review include oral clefts, congenital heart defects (CHDs), biliary atresia, pyloric stenosis, hypospadias, craniosynostosis, and clubfoot. Additionally, other issues related to GWAS are considered, including the assessment of polygenic risk scores and issues related to genetic ancestry, as well as utilizing genome-wide single nucleotide polymorphism array data to evaluate gene-environment interactions and Mendelian randomization. RESULTS For some birth defects, including oral clefts and CHDs, several novel susceptibility loci have been identified and replicated through GWAS, including 8q24 for oral clefts, DGKK for hypospadias, and 4p16 for CHDs. Relatively common birth defects for which there are currently no published GWAS include neural tube defects, anotia/microtia, anophthalmia/microphthalmia, gastroschisis, and omphalocele. CONCLUSIONS Overall, GWAS have been successful in identifying several novel susceptibility genes and genomic regions for structural birth defects. These findings have provided new insights into the etiologies of these phenotypes. However, GWAS have been underutilized for understanding the genetic etiologies of several birth defects.
Collapse
Affiliation(s)
- Philip J Lupo
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, Texas
| | - Laura E Mitchell
- Department of Epidemiology, Human Genetics and Environmental Sciences, UTHealth School of Public Health, Houston, Texas
| | - Mary M Jenkins
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, Georgia
| |
Collapse
|
33
|
Bai J, Zhang X, Kang X, Jin L, Wang P, Wang Z. Screening of core genes and pathways in breast cancer development via comprehensive analysis of multi gene expression datasets. Oncol Lett 2019; 18:5821-5830. [PMID: 31788055 PMCID: PMC6865771 DOI: 10.3892/ol.2019.10979] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 08/13/2019] [Indexed: 01/16/2023] Open
Abstract
Breast cancer has been the leading cause of cancer-associated mortality in women worldwide. Perturbation of oncogene and tumor suppressor gene expression is generally considered as the fundamental cause of cancer initiation and progression. In the present study, three gene expression datasets containing information of breast cancer and adjacent normal tissues that were detected using traditional gene microarrays were downloaded and batch effects were removed with R programming software. The differentially expressed genes between breast cancer and normal tissue groups were closely associated with cancer development pathways. Interestingly, five pathways, including ‘extracellular matrix-receptor interaction’, ‘peroxisome proliferator-activated receptors signaling pathway’, ‘propanoate metabolism’, ‘pyruvate metabolism’ and ‘regulation of lipolysis in adipocytes’, were thoroughly connected by 10 genes. Patients with upregulation of six of these hub genes (acetyl-CoA carboxylase β, acyl-CoA dehydrogenase medium chain, adiponectin, C1Q and collagen domain containing, acyl-CoA synthetase short chain family member 2, phosphoenolpyruvate carboxykinase 1 and perilipin 1) exhibited improved breast cancer prognosis. Additionally, breast cancer-specific network analysis identified several gene-gene interaction modules. These gene clusters had strong interactions according to the scoring in the whole network, which may be important to the development of breast cancer. In conclusion, the present study may improve the understanding of the mechanisms of breast cancer and provide several valuable prognosis and treatment signatures.
Collapse
Affiliation(s)
- Jie Bai
- Department of Thyroid and Breast III, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Xiaoyu Zhang
- Department of Thyroid and Breast III, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Xiaoning Kang
- Department of Ultrasound II, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Lijun Jin
- Department of Thyroid and Breast III, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Peng Wang
- Department of Thyroid and Breast III, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Zunyi Wang
- Department of Thyroid and Breast III, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| |
Collapse
|
34
|
Cao Z, Hao Y, Fung CW, Lee YY, Wang P, Li X, Xie K, Lam WJ, Qiu Y, Tang BZ, Shui G, Liu P, Qu J, Kang BH, Mak HY. Dietary fatty acids promote lipid droplet diversity through seipin enrichment in an ER subdomain. Nat Commun 2019; 10:2902. [PMID: 31263173 PMCID: PMC6602954 DOI: 10.1038/s41467-019-10835-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 06/03/2019] [Indexed: 02/06/2023] Open
Abstract
Exogenous metabolites from microbial and dietary origins have profound effects on host metabolism. Here, we report that a sub-population of lipid droplets (LDs), which are conserved organelles for fat storage, is defined by metabolite-modulated targeting of the C. elegans seipin ortholog, SEIP-1. Loss of SEIP-1 function reduces the size of a subset of LDs while over-expression of SEIP-1 has the opposite effect. Ultrastructural analysis reveals SEIP-1 enrichment in an endoplasmic reticulum (ER) subdomain, which co-purifies with LDs. Analyses of C. elegans and bacterial genetic mutants indicate a requirement of polyunsaturated fatty acids (PUFAs) and microbial cyclopropane fatty acids (CFAs) for SEIP-1 enrichment, as confirmed by dietary supplementation experiments. In mammalian cells, heterologously expressed SEIP-1 engages nascent lipid droplets and promotes their subsequent expansion in a conserved manner. Our results suggest that microbial and polyunsaturated fatty acids serve unexpected roles in regulating cellular fat storage by promoting LD diversity. Lipid droplets (LDs) are fat storage organelles that are initiated and expanded by seipins at ER contact sites. Here the authors show that the C. elegans seipin ortholog SEIP-1 is recruited to these sites by certain dietary fatty acids to support the expansion of a subset of LDs.
Collapse
Affiliation(s)
- Zhe Cao
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Yan Hao
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Chun Wing Fung
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Yiu Yiu Lee
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Pengfei Wang
- School of Life Science, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xuesong Li
- Biophotonics Research Laboratory, Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Kang Xie
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Wen Jiun Lam
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Yifei Qiu
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Ben Zhong Tang
- Department of Chemistry, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Guanghou Shui
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Pingsheng Liu
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Jianan Qu
- Biophotonics Research Laboratory, Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Byung-Ho Kang
- School of Life Science, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ho Yi Mak
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong SAR, China.
| |
Collapse
|
35
|
|
36
|
Zhu X, Stephens M. Large-scale genome-wide enrichment analyses identify new trait-associated genes and pathways across 31 human phenotypes. Nat Commun 2018; 9:4361. [PMID: 30341297 PMCID: PMC6195536 DOI: 10.1038/s41467-018-06805-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 09/29/2018] [Indexed: 12/27/2022] Open
Abstract
Genome-wide association studies (GWAS) aim to identify genetic factors associated with phenotypes. Standard analyses test variants for associations individually. However, variant-level associations are hard to identify and can be difficult to interpret biologically. Enrichment analyses help address both problems by targeting sets of biologically related variants. Here we introduce a new model-based enrichment method that requires only GWAS summary statistics. Applying this method to interrogate 4,026 gene sets in 31 human phenotypes identifies many previously-unreported enrichments, including enrichments of endochondral ossification pathway for height, NFAT-dependent transcription pathway for rheumatoid arthritis, brain-related genes for coronary artery disease, and liver-related genes for Alzheimer’s disease. A key feature of our method is that inferred enrichments automatically help identify new trait-associated genes. For example, accounting for enrichment in lipid transport genes highlights association between MTTP and low-density lipoprotein levels, whereas conventional analyses of the same data found no significant variants near this gene. In genome-wide association studies, variant-level associations are hard to identify and can be difficult to interpret biologically. Here, the authors develop a new model-based enrichment analysis method, and apply it to identify new associated genes, pathways and tissues across 31 human phenotypes.
Collapse
Affiliation(s)
- Xiang Zhu
- Department of Statistics, Stanford University, Stanford, 94305, CA, USA. .,Department of Statistics, The University of Chicago, Chicago, 60637, IL, USA.
| | - Matthew Stephens
- Department of Statistics, The University of Chicago, Chicago, 60637, IL, USA. .,Department of Human Genetics, The University of Chicago, Chicago, 60637, IL, USA.
| |
Collapse
|
37
|
Wu C, Pan W. Integration of Enhancer-Promoter Interactions with GWAS Summary Results Identifies Novel Schizophrenia-Associated Genes and Pathways. Genetics 2018; 209:699-709. [PMID: 29728367 PMCID: PMC6028261 DOI: 10.1534/genetics.118.300805] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 05/02/2018] [Indexed: 12/28/2022] Open
Abstract
It remains challenging to boost statistical power of genome-wide association studies (GWASs) to identify more risk variants or loci that can account for "missing heritability." Furthermore, since most identified variants are not in gene-coding regions, a biological interpretation of their function is largely lacking. On the other hand, recent biotechnological advances have made it feasible to experimentally measure the three-dimensional organization of the genome, including enhancer-promoter interactions in high resolutions. Due to the well-known critical roles of enhancer-promoter interactions in regulating gene expression programs, such data have been applied to link GWAS risk variants to their putative target genes, gaining insights into underlying biological mechanisms. However, their direct use in GWAS association testing is yet to be exploited. Here we propose integrating enhancer-promoter interactions into GWAS association analysis to both boost statistical power and enhance interpretability. We demonstrate that through an application to two large-scale schizophrenia (SCZ) GWAS summary data sets, the proposed method could identify some novel SCZ-associated genes and pathways (containing no significant SNPs). For example, after the Bonferroni correction, for the larger SCZ data set with 36,989 cases and 113,075 controls, our method applied to the gene body and enhancer regions identified 27 novel genes and 11 novel KEGG pathways to be significant, all missed by the transcriptome-wide association study (TWAS) approach. We conclude that our proposed method is potentially useful and is complementary to TWAS and other standard gene- and pathway-based methods.
Collapse
Affiliation(s)
- Chong Wu
- Division of Biostatistics, University of Minnesota, Minneapolis, Minnesota 55455
| | - Wei Pan
- Division of Biostatistics, University of Minnesota, Minneapolis, Minnesota 55455
| |
Collapse
|
38
|
Jose AM. Replicating and Cycling Stores of Information Perpetuate Life. Bioessays 2018; 40:e1700161. [PMID: 29493806 PMCID: PMC7303024 DOI: 10.1002/bies.201700161] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 01/06/2018] [Indexed: 12/12/2022]
Abstract
Life is perpetuated through a single-cell bottleneck between generations in many organisms. Here, I highlight that this cell holds information in two distinct stores: in the linear DNA sequence that is replicated during cell divisions, and in the three-dimensional arrangement of molecules that can change during development but is recreated at the start of each generation. These two interdependent stores of information - one replicating with each cell division and the other cycling with a period of one generation - coevolve while perpetuating an organism. Unlike the genome sequence, the arrangement of molecules, including DNA, RNAs, proteins, sugars, lipids, etc., is not well understood. Because this arrangement and the genome sequence are transmitted together from one generation to the next, analysis of both is necessary to understand evolution and origins of inherited diseases. Recent developments suggest that tools are in place to examine how all the information to build an organism is encoded within a single cell, and how this cell code is reproduced in every generation. See also the video abstract here: https://youtu.be/IdWEL-T6TPU.
Collapse
Affiliation(s)
- Antony M. Jose
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|