101
|
Lopes I, Altab G, Raina P, de Magalhães JP. Gene Size Matters: An Analysis of Gene Length in the Human Genome. Front Genet 2021; 12:559998. [PMID: 33643374 PMCID: PMC7905317 DOI: 10.3389/fgene.2021.559998] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 01/06/2021] [Indexed: 12/23/2022] Open
Abstract
While it is expected for gene length to be associated with factors such as intron number and evolutionary conservation, we are yet to understand the connections between gene length and function in the human genome. In this study, we show that, as expected, there is a strong positive correlation between gene length, transcript length, and protein size as well as a correlation with the number of genetic variants and introns. Among tissue-specific genes, we find that the longest transcripts tend to be expressed in the blood vessels, nerves, thyroid, cervix uteri, and the brain, while the smallest transcripts tend to be expressed in the pancreas, skin, stomach, vagina, and testis. We report, as shown previously, that natural selection suppresses changes for genes with longer transcripts and promotes changes for genes with smaller transcripts. We also observe that genes with longer transcripts tend to have a higher number of co-expressed genes and protein-protein interactions, as well as more associated publications. In the functional analysis, we show that bigger transcripts are often associated with neuronal development, while smaller transcripts tend to play roles in skin development and in the immune system. Furthermore, pathways related to cancer, neurons, and heart diseases tend to have genes with longer transcripts, with smaller transcripts being present in pathways related to immune responses and neurodegenerative diseases. Based on our results, we hypothesize that longer genes tend to be associated with functions that are important in the early development stages, while smaller genes tend to play a role in functions that are important throughout the whole life, like the immune system, which requires fast responses.
Collapse
Affiliation(s)
| | | | | | - João Pedro de Magalhães
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
102
|
Li ZL, Buck M. Beyond history and "on a roll": The list of the most well-studied human protein structures and overall trends in the protein data bank. Protein Sci 2021; 30:745-760. [PMID: 33550681 DOI: 10.1002/pro.4038] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 02/02/2021] [Accepted: 02/05/2021] [Indexed: 12/17/2022]
Abstract
Of the roughly 20,000 canonical human protein sequences, as of January 20, 2021, 7,077 proteins have had their full or partial, medium- to high-resolution structures determined by x-ray crystallography or other methods. Which of these proteins dominate the protein data bank (the PDB) and why? In this paper, we list the 273 top human protein structures based on the number of their PDB entries. This set of proteins accounts for more than 40% of all available human PDB entries and represent past trends as well as current status for protein structural biology. We briefly discuss the relationship which some of the prominent protein structures have with protein research as a whole and mention their relevance to human diseases. The top-10 soluble and membrane proteins are all well-known (most of their first structures being deposited more than 30 years ago). Overall, there is no dramatic change in recent trends in the PDB. Remarkably, the number of structure depositions has grown nearly exponentially over the last 10 or more years (with a doubling time of 7 years for proteins, obtained from any organism). Growth in human protein structures is slightly faster (at 5.9 years). The information in this paper may be informative to senior scientists but also inspire researchers who are new to protein science, providing the year 2021 snap-shot for the state of protein structural biology.
Collapse
Affiliation(s)
- Zhen-Lu Li
- Department of Physiology and Biophysics, Case Western Reserve University, School of Medicine, Cleveland, Ohio, USA
| | - Matthias Buck
- Department of Physiology and Biophysics, Case Western Reserve University, School of Medicine, Cleveland, Ohio, USA.,Department of Pharmacology; Department of Neurosciences and Case Comprehensive Cancer Center, Case Western Reserve University, School of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
103
|
Sastry AV, Hu A, Heckmann D, Poudel S, Kavvas E, Palsson BO. Independent component analysis recovers consistent regulatory signals from disparate datasets. PLoS Comput Biol 2021; 17:e1008647. [PMID: 33529205 PMCID: PMC7888660 DOI: 10.1371/journal.pcbi.1008647] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 02/17/2021] [Accepted: 12/18/2020] [Indexed: 01/03/2023] Open
Abstract
The availability of bacterial transcriptomes has dramatically increased in recent years. This data deluge could result in detailed inference of underlying regulatory networks, but the diversity of experimental platforms and protocols introduces critical biases that could hinder scalable analysis of existing data. Here, we show that the underlying structure of the E. coli transcriptome, as determined by Independent Component Analysis (ICA), is conserved across multiple independent datasets, including both RNA-seq and microarray datasets. We subsequently combined five transcriptomics datasets into a large compendium containing over 800 expression profiles and discovered that its underlying ICA-based structure was still comparable to that of the individual datasets. With this understanding, we expanded our analysis to over 3,000 E. coli expression profiles and predicted three high-impact regulons that respond to oxidative stress, anaerobiosis, and antibiotic treatment. ICA thus enables deep analysis of disparate data to uncover new insights that were not visible in the individual datasets. Cells adapt to diverse environments by regulating gene expression. Genome-wide measurements of gene expression levels have exponentially increased in recent years, but successful integration and analysis of these datasets are limited. Recently, we showed that independent component analysis (ICA), a signal deconvolution algorithm, can separate a large bacterial gene expression dataset into groups of co-regulated genes. This previous study focused on data generated by a standardized pipeline and did not address whether ICA extracts the same quantitative co-expression signals across expression profiling platforms. In this study, we show that ICA finds similar co-regulation patterns underlying multiple gene expression datasets and can be used as a tool to integrate and interpret diverse datasets. Using a dataset containing over 3,000 expression profiles, we predicted three new regulons and characterized their activities. Since large, standardized expression datasets only exist for a few bacterial strains, these results broaden the possible applications of this tool to better understand transcriptional regulation across a wide range of microbes.
Collapse
Affiliation(s)
- Anand V. Sastry
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States of America
| | - Alyssa Hu
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States of America
| | - David Heckmann
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States of America
| | - Saugat Poudel
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States of America
| | - Erol Kavvas
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States of America
| | - Bernhard O. Palsson
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States of America
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark
- * E-mail:
| |
Collapse
|
104
|
Kim H, Kim E, Lee I, Bae B, Park M, Nam H. Artificial Intelligence in Drug Discovery: A Comprehensive Review of Data-driven and Machine Learning Approaches. BIOTECHNOL BIOPROC E 2021; 25:895-930. [PMID: 33437151 PMCID: PMC7790479 DOI: 10.1007/s12257-020-0049-y] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 05/27/2020] [Accepted: 06/03/2020] [Indexed: 02/07/2023]
Abstract
As expenditure on drug development increases exponentially, the overall drug discovery process requires a sustainable revolution. Since artificial intelligence (AI) is leading the fourth industrial revolution, AI can be considered as a viable solution for unstable drug research and development. Generally, AI is applied to fields with sufficient data such as computer vision and natural language processing, but there are many efforts to revolutionize the existing drug discovery process by applying AI. This review provides a comprehensive, organized summary of the recent research trends in AI-guided drug discovery process including target identification, hit identification, ADMET prediction, lead optimization, and drug repositioning. The main data sources in each field are also summarized in this review. In addition, an in-depth analysis of the remaining challenges and limitations will be provided, and proposals for promising future directions in each of the aforementioned areas.
Collapse
Affiliation(s)
- Hyunho Kim
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005 Korea
| | - Eunyoung Kim
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005 Korea
| | - Ingoo Lee
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005 Korea
| | - Bongsung Bae
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005 Korea
| | - Minsu Park
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005 Korea
| | - Hojung Nam
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005 Korea
| |
Collapse
|
105
|
Norris JM, Simpson BS, Ball R, Freeman A, Kirkham A, Parry MA, Moore CM, Whitaker HC, Emberton M. A Modified Newcastle-Ottawa Scale for Assessment of Study Quality in Genetic Urological Research. Eur Urol 2020; 79:325-326. [PMID: 33375994 DOI: 10.1016/j.eururo.2020.12.017] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 12/10/2020] [Indexed: 11/16/2022]
Abstract
Our modification of the traditional Newcastle-Ottawa scale enables urological researchers to effectively appraise and communicate the quality of genetic-based research in urology.
Collapse
Affiliation(s)
- Joseph M Norris
- UCL Division of Surgery & Interventional Science, University College London, London, UK; Department of Urology, University College London Hospitals NHS Foundation Trust, London, UK.
| | - Benjamin S Simpson
- UCL Division of Surgery & Interventional Science, University College London, London, UK
| | - Rhys Ball
- Department of Pathology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Alex Freeman
- Department of Pathology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Alex Kirkham
- Department of Radiology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Marina A Parry
- UCL Cancer Institute, University College London, London, UK
| | - Caroline M Moore
- UCL Division of Surgery & Interventional Science, University College London, London, UK; Department of Urology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Hayley C Whitaker
- UCL Division of Surgery & Interventional Science, University College London, London, UK
| | - Mark Emberton
- UCL Division of Surgery & Interventional Science, University College London, London, UK; Department of Urology, University College London Hospitals NHS Foundation Trust, London, UK
| |
Collapse
|
106
|
Stoeger T, Nunes Amaral LA. COVID-19 research risks ignoring important host genes due to pre-established research patterns. eLife 2020; 9:e61981. [PMID: 33231169 PMCID: PMC7685703 DOI: 10.7554/elife.61981] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 11/07/2020] [Indexed: 01/08/2023] Open
Abstract
It is known that research into human genes is heavily skewed towards genes that have been widely studied for decades, including many genes that were being studied before the productive phase of the Human Genome Project. This means that the genes most frequently investigated by the research community tend to be only marginally more important to human physiology and disease than a random selection of genes. Based on an analysis of 10,395 research publications about SARS-CoV-2 that mention at least one human gene, we report here that the COVID-19 literature up to mid-October 2020 follows a similar pattern. This means that a large number of host genes that have been implicated in SARS-CoV-2 infection by four genome-wide studies remain unstudied. While quantifying the consequences of this neglect is not possible, they could be significant.
Collapse
Affiliation(s)
- Thomas Stoeger
- Successful Clinical Response in Pneumonia Therapy (SCRIPT) Systems Biology Center, Northwestern University, Evanston, United States
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, United States
- Center for Genetic Medicine, Northwestern University School of Medicine, Chicago, United States
| | - Luís A Nunes Amaral
- Successful Clinical Response in Pneumonia Therapy (SCRIPT) Systems Biology Center, Northwestern University, Evanston, United States
- Northwestern Institute on Complex Systems (NICO), Northwestern University, Evanston, United States
- Department of Molecular Biosciences, Northwestern University, Evanston, United States
- Department of Physics and Astronomy, Northwestern University, Evanston, United States
- Department of Medicine, Northwestern University School of Medicine, Chicago, United States
| |
Collapse
|
107
|
Abstract
One of the grand challenges in contemporary chemical biology is the generation of a probe for every member of the human proteome. Probe selection and optimization strategies typically rely on experimental bioactivity data to determine the potency and selectivity of candidate molecules. However, this approach is profoundly limited by the sparsity of the known data, the annotation bias often found in the literature, and the cost of physical screening. Recent advancements in predictive pharmacology, such as the application of multitask and transfer learning, as well as the use of biologically motivated, structure-agnostic features to characterize molecules, should serve to mitigate these issues. Computational modeling likely offers the only cost-effective approach to substantially increasing the bioactivity annotation density both on the local and global scale and thus, we argue, will need to make a substantial contribution if the ambitious goals of probing the human proteome are to be realized in the foreseeable future.
Collapse
Affiliation(s)
- Tim James
- Evotec (U.K.) Ltd. 114 Innovation Drive, Milton Park, Abingdon, Oxfordshire OX14 4RZ, U.K
| | - Adam Sardar
- Evotec (U.K.) Ltd. 114 Innovation Drive, Milton Park, Abingdon, Oxfordshire OX14 4RZ, U.K
| | - Andrew Anighoro
- Evotec (U.K.) Ltd. 114 Innovation Drive, Milton Park, Abingdon, Oxfordshire OX14 4RZ, U.K
| |
Collapse
|
108
|
Locsin RC, Pepito JA, Juntasopeepun P, Constantino RE. Transcending human frailties with technological enhancements and replacements: Transhumanist perspective in nursing and healthcare. Nurs Inq 2020; 28:e12391. [PMID: 33159824 DOI: 10.1111/nin.12391] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 12/12/2022]
Abstract
As human beings age, they become weak, fragile, and feeble. It is a slowly progressing yet complex syndrome in which old age or some disabilities are not prerequisites; neither does loss of human parts lead to frailty among the physically fit older persons. This paper aims to describe the influences of transhumanist perspectives on human-technology enhancements and replacements in the transcendence of human frailties, including those of older persons, in which technology is projected to deliver solutions toward transcending these frailties. Through technologies including genetic screening and other technological manipulations, intelligent machines and augmented humans improve, maintain, and remedy human-linked susceptibilities. Furthermore, other technologies replace parts fabricated through inorganic-mechanical processes such as 3D-printing. Advancing technologies are reaching the summit of technological sophistication contributing to the transhumanist views of being human in a technological world. Technologies enhance the transcendence of human frailties as essential expressions of the symbiosis between human beings and technology in a transcendental world.
Collapse
Affiliation(s)
- Rozzano C Locsin
- Department of Nursing, Faculty of Nursing, Chiang Mai University, Chiang Mai, Thailand.,Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan.,Florida Atlantic University, Christine E. Lynn College of Nursing, Boca Raton, FL, USA
| | - Joseph Andrew Pepito
- College of Allied Medical Sciences, Cebu Doctors' University, Cebu City, Philippines
| | - Phanida Juntasopeepun
- Department of Policy, Planning, and IT Management, Faculty of Nursing, Chiang Mai University, Chiang Mai, Thailand
| | | |
Collapse
|
109
|
Cardoso-Moreira M, Sarropoulos I, Velten B, Mort M, Cooper DN, Huber W, Kaessmann H. Developmental Gene Expression Differences between Humans and Mammalian Models. Cell Rep 2020; 33:108308. [PMID: 33113372 PMCID: PMC7610014 DOI: 10.1016/j.celrep.2020.108308] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 08/16/2020] [Accepted: 10/05/2020] [Indexed: 11/21/2022] Open
Abstract
Identifying the molecular programs underlying human organ development and how they differ from model species is key for understanding human health and disease. Developmental gene expression profiles provide a window into the genes underlying organ development and a direct means to compare them across species. We use a transcriptomic resource covering the development of seven organs to characterize the temporal profiles of human genes associated with distinct disease classes and to determine, for each human gene, the similarity of its spatiotemporal expression with its orthologs in rhesus macaque, mouse, rat, and rabbit. We find clear associations between spatiotemporal profiles and the phenotypic manifestations of diseases. We also find that half of human genes differ from their mouse orthologs in their temporal trajectories in at least one of the organs. These include more than 200 genes associated with brain, heart, and liver disease for which mouse models should undergo extra scrutiny.
Collapse
Affiliation(s)
- Margarida Cardoso-Moreira
- Center for Molecular Biology (ZMBH), DKFZ-ZMBH Alliance, Heidelberg University, 69120 Heidelberg, Germany.
| | - Ioannis Sarropoulos
- Center for Molecular Biology (ZMBH), DKFZ-ZMBH Alliance, Heidelberg University, 69120 Heidelberg, Germany
| | - Britta Velten
- Genome Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Matthew Mort
- Institute of Medical Genetics, Cardiff University, Cardiff CF14 4XN, UK
| | - David N Cooper
- Institute of Medical Genetics, Cardiff University, Cardiff CF14 4XN, UK
| | - Wolfgang Huber
- Genome Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Henrik Kaessmann
- Center for Molecular Biology (ZMBH), DKFZ-ZMBH Alliance, Heidelberg University, 69120 Heidelberg, Germany.
| |
Collapse
|
110
|
Cheng F, Ma Y, Uzzi B, Loscalzo J. Importance of scientific collaboration in contemporary drug discovery and development: a detailed network analysis. BMC Biol 2020; 18:138. [PMID: 33050894 PMCID: PMC7556984 DOI: 10.1186/s12915-020-00868-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 09/15/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Growing evidence shows that scientific collaboration plays a crucial role in transformative innovation in the life sciences. For example, contemporary drug discovery and development reflects the work of teams of individuals from academic centers, the pharmaceutical industry, the regulatory science community, health care providers, and patients. However, public understanding of how collaborations between academia and industry catalyze novel target identification and first-in-class drug discovery is limited. RESULTS We perform a comprehensive network analysis on a large scientific corpus of collaboration and citations (97,688 papers with 1,862,500 citations from 170 million scientific records) to quantify the success trajectory of innovative drug development. By focusing on four types of cardiovascular drugs, we demonstrate how knowledge flows between institutions to highlight the underlying contributions of many different institutions in the development of a new drug. We highlight how such network analysis could help to increase industrial and governmental support, and improve the efficiency or accelerate decision-making in drug discovery and development. CONCLUSION We demonstrate that network analysis of large public databases can identify and quantify investigator and institutional relationships in drug discovery and development. If broadly applied, this type of network analysis may help to enhance public understanding of and support for biomedical research, and could identify factors that facilitate decision-making in first-in-class drug discovery among academia, the pharmaceutical industry, and healthcare systems.
Collapse
Affiliation(s)
- Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Yifang Ma
- Department of Statistics and Data Science, Southern University of Science and Technology, Shenzhen, 518055, China
- Northwestern Institute on Complex Systems (NICO) and Kellogg School of Management, Northwestern University, Evanston, IL, 60208, USA
| | - Brian Uzzi
- Northwestern Institute on Complex Systems (NICO) and Kellogg School of Management, Northwestern University, Evanston, IL, 60208, USA
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St., Boston, MA, 02115, USA.
| |
Collapse
|
111
|
Whitt L, Ricachenevsky FK, Ziegler GZ, Clemens S, Walker E, Maathuis FJM, Kear P, Baxter I. A curated list of genes that affect the plant ionome. PLANT DIRECT 2020; 4:e00272. [PMID: 33103043 PMCID: PMC7576880 DOI: 10.1002/pld3.272] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 08/26/2020] [Accepted: 08/28/2020] [Indexed: 05/07/2023]
Abstract
Understanding the mechanisms underlying plants' adaptation to their environment will require knowledge of the genes and alleles underlying elemental composition. Modern genetics is capable of quickly, and cheaply indicating which regions of DNA are associated with particular phenotypes in question, but most genes remain poorly annotated, hindering the identification of candidate genes. To help identify candidate genes underlying elemental accumulations, we have created the known ionome gene (KIG) list: a curated collection of genes experimentally shown to change uptake, accumulation, and distribution of elements. We have also created an automated computational pipeline to generate lists of KIG orthologs in other plant species using the PhytoMine database. The current version of KIG consists of 176 known genes covering 5 species, 23 elements, and their 1588 orthologs in 10 species. Analysis of the known genes demonstrated that most were identified in the model plant Arabidopsis thaliana, and that transporter coding genes and genes altering the accumulation of iron and zinc are overrepresented in the current list.
Collapse
Affiliation(s)
- Lauren Whitt
- Donald Danforth Plant Science CenterSaint LouisMOUSA
| | - Felipe Klein Ricachenevsky
- Departamento de Botânica Programa de Pós‐Graduação em Biologia Celular e MolecularUniversidade Federal do Rio Grande do SulPorto AlegreBrazil
| | | | | | | | | | | | - Ivan Baxter
- Donald Danforth Plant Science CenterSaint LouisMOUSA
| |
Collapse
|
112
|
Schnable JC. Genes and gene models, an important distinction. THE NEW PHYTOLOGIST 2020; 228:50-55. [PMID: 31241760 DOI: 10.1111/nph.16011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 06/07/2019] [Indexed: 05/22/2023]
Abstract
Genome sequencing has fundamentally changed how plant biologists think about genes. All or nearly all genes can ultimately be associated with a gene model. However, many gene models appear to play little or no role in the traits of an organism. A range of structural, molecular, population and evolutionary features all show a separation between genes with known phenotypes and the overall set of annotated gene models. These different features could be combined to develop models to distinguish the genes that determine the traits of plants from the subset gene other annotated gene models which are unlikely to play a role in doing so. Efforts to identify the subset of annotated gene models likely involved in specifying the characteristics of plants would help aid a wide range of researchers.
Collapse
Affiliation(s)
- James C Schnable
- Department of Agronomy and Horticulture and Center for Plant Science Innovation, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| |
Collapse
|
113
|
Baral K, Rotwein P. ZMAT2 in Humans and Other Primates: A Highly Conserved and Understudied Gene. Evol Bioinform Online 2020; 16:1176934320941500. [PMID: 32952394 PMCID: PMC7485168 DOI: 10.1177/1176934320941500] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 06/18/2020] [Indexed: 12/18/2022] Open
Abstract
Recent advances in genetics present unique opportunities for enhancing our
understanding of human physiology and disease predisposition through detailed
analysis of gene structure, expression, and population variation via examination
of data in publicly accessible genome and gene expression repositories. Yet, the
vast majority of human genes remain understudied. Here, we show the scope of
these genomic and genetic resources by evaluating ZMAT2, a
member of a 5-gene family that through May 2020 had been the focus of only 4
peer-reviewed scientific publications. Using analysis of information extracted
from public databases, we show that human ZMAT2 is a 6-exon
gene and find that it exhibits minimal genetic variation in human populations
and in disease states, including cancer. We further demonstrate that the gene
and its encoded protein are highly conserved among nonhuman primates and define
a cohort of ZMAT2 pseudogenes in the marmoset genome.
Collectively, our investigations illustrate how complementary use of genomic,
gene expression, and population genetic resources can lead to new insights about
human and mammalian biology and evolution, and when coupled with data supporting
key roles for ZMAT2 in keratinocyte differentiation and pre-RNA splicing argue
that this gene is worthy of further study.
Collapse
Affiliation(s)
- Kabita Baral
- Graduate School, College of Science, The University of Texas at El Paso, El Paso, TX, USA.,Department of Microbiology, University of Calgary, Calgary, AB, Canada
| | - Peter Rotwein
- Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, USA
| |
Collapse
|
114
|
Pividori M, Rajagopal PS, Barbeira A, Liang Y, Melia O, Bastarache L, Park Y, Consortium GTE, Wen X, Im HK. PhenomeXcan: Mapping the genome to the phenome through the transcriptome. SCIENCE ADVANCES 2020; 6:eaba2083. [PMID: 32917697 PMCID: PMC11206444 DOI: 10.1126/sciadv.aba2083] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 07/29/2020] [Indexed: 05/02/2023]
Abstract
Large-scale genomic and transcriptomic initiatives offer unprecedented insight into complex traits, but clinical translation remains limited by variant-level associations without biological context and lack of analytic resources. Our resource, PhenomeXcan, synthesizes 8.87 million variants from genome-wide association study summary statistics on 4091 traits with transcriptomic data from 49 tissues in Genotype-Tissue Expression v8 into a gene-based, queryable platform including 22,515 genes. We developed a novel Bayesian colocalization method, fast enrichment estimation aided colocalization analysis (fastENLOC), to prioritize likely causal gene-trait associations. We successfully replicate associations from the phenome-wide association studies (PheWAS) catalog Online Mendelian Inheritance in Man, and an evidence-based curated gene list. Using PhenomeXcan results, we provide examples of novel and underreported genome-to-phenome associations, complex gene-trait clusters, shared causal genes between common and rare diseases via further integration of PhenomeXcan with ClinVar, and potential therapeutic targets. PhenomeXcan (phenomexcan.org) provides broad, user-friendly access to complex data for translational researchers.
Collapse
Affiliation(s)
- Milton Pividori
- Section of Genetic Medicine, Department of Medicine, The University of Chicago, Chicago, IL, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Padma S Rajagopal
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Alvaro Barbeira
- Section of Genetic Medicine, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Yanyu Liang
- Section of Genetic Medicine, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Owen Melia
- Section of Genetic Medicine, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Lisa Bastarache
- Department of Biomedical Informatics, Department of Medicine, Vanderbilt University, Nashville, TN, USA
- Center for Human Genetics Research, Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - YoSon Park
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | | | - Xiaoquan Wen
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, USA.
| | - Hae K Im
- Section of Genetic Medicine, Department of Medicine, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
115
|
Gene Expression Comparison between Sézary Syndrome and Lymphocytic-Variant Hypereosinophilic Syndrome Refines Biomarkers for Sézary Syndrome. Cells 2020; 9:cells9091992. [PMID: 32872487 PMCID: PMC7563155 DOI: 10.3390/cells9091992] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/26/2020] [Accepted: 08/27/2020] [Indexed: 02/07/2023] Open
Abstract
Sézary syndrome (SS), an aggressive cutaneous T-cell lymphoma (CTCL) with poor prognosis, is characterized by the clinical hallmarks of circulating malignant T cells, erythroderma and lymphadenopathy. However, highly variable clinical skin manifestations and similarities with benign mimickers can lead to significant diagnostic delay and inappropriate therapy that can lead to disease progression and mortality. SS has been the focus of numerous transcriptomic-profiling studies to identify sensitive and specific diagnostic and prognostic biomarkers. Benign inflammatory disease controls (e.g., psoriasis, atopic dermatitis) have served to identify chronic inflammatory phenotypes in gene expression profiles, but provide limited insight into the lymphoproliferative and oncogenic roles of abnormal gene expression in SS. This perspective was recently clarified by a transcriptome meta-analysis comparing SS and lymphocytic-variant hypereosinophilic syndrome, a benign yet often clonal T-cell lymphoproliferation, with clinical features similar to SS. Here we review the rationale for selecting lymphocytic-variant hypereosinophilic syndrome (L-HES) as a disease control for SS, and discuss differentially expressed genes that may distinguish benign from malignant lymphoproliferative phenotypes, including additional context from prior gene expression studies to improve understanding of genes important in SS.
Collapse
|
116
|
Ichino N, Serres MR, Urban RM, Urban MD, Treichel AJ, Schaefbauer KJ, Greif LE, Varshney GK, Skuster KJ, McNulty MS, Daby CL, Wang Y, Liao HK, El-Rass S, Ding Y, Liu W, Anderson JL, Wishman MD, Sabharwal A, Schimmenti LA, Sivasubbu S, Balciunas D, Hammerschmidt M, Farber SA, Wen XY, Xu X, McGrail M, Essner JJ, Burgess SM, Clark KJ, Ekker SC. Building the vertebrate codex using the gene breaking protein trap library. eLife 2020; 9:54572. [PMID: 32779569 PMCID: PMC7486118 DOI: 10.7554/elife.54572] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 08/07/2020] [Indexed: 12/14/2022] Open
Abstract
One key bottleneck in understanding the human genome is the relative under-characterization of 90% of protein coding regions. We report a collection of 1200 transgenic zebrafish strains made with the gene-break transposon (GBT) protein trap to simultaneously report and reversibly knockdown the tagged genes. Protein trap-associated mRFP expression shows previously undocumented expression of 35% and 90% of cloned genes at 2 and 4 days post-fertilization, respectively. Further, investigated alleles regularly show 99% gene-specific mRNA knockdown. Homozygous GBT animals in ryr1b, fras1, tnnt2a, edar and hmcn1 phenocopied established mutants. 204 cloned lines trapped diverse proteins, including 64 orthologs of human disease-associated genes with 40 as potential new disease models. Severely reduced skeletal muscle Ca2+ transients in GBT ryr1b homozygous animals validated the ability to explore molecular mechanisms of genetic diseases. This GBT system facilitates novel functional genome annotation towards understanding cellular and molecular underpinnings of vertebrate biology and human disease.
Collapse
Affiliation(s)
- Noriko Ichino
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - MaKayla R Serres
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Rhianna M Urban
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Mark D Urban
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Anthony J Treichel
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Kyle J Schaefbauer
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Lauren E Greif
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Gaurav K Varshney
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, United States.,Functional & Chemical Genomics Program, Oklahoma Medical Research Foundation, Oklahoma City, United States
| | - Kimberly J Skuster
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Melissa S McNulty
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Camden L Daby
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Ying Wang
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, United States
| | - Hsin-Kai Liao
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, United States
| | - Suzan El-Rass
- Zebrafish Centre for Advanced Drug Discovery & Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto & University of Toronto, Toronto, Canada
| | - Yonghe Ding
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States.,Department of Cardiovascular Medicine, Mayo Clinic, Rochester, United States
| | - Weibin Liu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States.,Department of Cardiovascular Medicine, Mayo Clinic, Rochester, United States
| | - Jennifer L Anderson
- Department of Embryology, Carnegie Institution for Science, Baltimore, United States
| | - Mark D Wishman
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Ankit Sabharwal
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Lisa A Schimmenti
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States.,Department of Clinical Genomics, Mayo Clinic, Rochester, United States.,Department of Otorhinolaryngology, Mayo Clinic, Rochester, United States
| | - Sridhar Sivasubbu
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology, Delhi, India
| | - Darius Balciunas
- Department of Biology, Temple University, Philadelphia, United States
| | - Matthias Hammerschmidt
- Institute of Zoology, Developmental Biology Unit, University of Cologne, Cologne, Germany
| | - Steven Arthur Farber
- Department of Embryology, Carnegie Institution for Science, Baltimore, United States
| | - Xiao-Yan Wen
- Zebrafish Centre for Advanced Drug Discovery & Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto & University of Toronto, Toronto, Canada
| | - Xiaolei Xu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States.,Department of Cardiovascular Medicine, Mayo Clinic, Rochester, United States
| | - Maura McGrail
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, United States
| | - Jeffrey J Essner
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, United States
| | - Shawn M Burgess
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, United States
| | - Karl J Clark
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Stephen C Ekker
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| |
Collapse
|
117
|
Madsen EB, Aagaard K. Concentration of Danish research funding on individual researchers and research topics: Patterns and potential drivers. QUANTITATIVE SCIENCE STUDIES 2020. [DOI: 10.1162/qss_a_00077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
The degree of concentration in research funding has long been a principal matter of contention in science policy. Strong concentration has been seen as a tool for optimizing and focusing research investments but also as a damaging path towards hypercompetition, diminished diversity, and conservative topic selection. While several studies have documented funding concentration linked to individual funding organizations, few have looked at funding concentration from a systemic perspective. In this article, we examine nearly 20,000 competitive grants allocated by 15 major Danish research funders. Our results show a strongly skewed allocation of funding towards a small elite of individual researchers, and towards a select group of research areas and topics. We discuss potential drivers and highlight that funding concentration likely results from a complex interplay between funders’ overlapping priorities, excellence-dominated evaluation criteria, and lack of coordination between both public and private research funding bodies.
Collapse
Affiliation(s)
- Emil Bargmann Madsen
- Danish Centre for Studies in Research and Research Policy, Department of Political Science, Aarhus University, Bartholins allé 7, 8000 Aarhus C
| | - Kaare Aagaard
- Danish Centre for Studies in Research and Research Policy, Department of Political Science, Aarhus University, Bartholins allé 7, 8000 Aarhus C
| |
Collapse
|
118
|
Abstract
Understanding the etiology of congenital disorders requires interdisciplinary research and close collaborations between clinicians, geneticists and developmental biologists. The pace of gene discovery has quickened due to advances in sequencing technology, resulting in a wealth of publicly available sequence data but also a gap between gene discovery and crucial mechanistic insights provided by studies in model systems. In this Spotlight, I highlight the opportunities for developmental biologists to engage with human geneticists and genetic resources to advance the study of congenital disorders.
Collapse
|
119
|
Finkle JD, Bagheri N. Hybrid analysis of gene dynamics predicts context-specific expression and offers regulatory insights. Bioinformatics 2020; 35:4671-4678. [PMID: 30994899 PMCID: PMC6853664 DOI: 10.1093/bioinformatics/btz256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 03/07/2019] [Accepted: 04/11/2019] [Indexed: 12/02/2022] Open
Abstract
Motivation To understand the regulatory pathways underlying diseases, studies often investigate the differential gene expression between genetically or chemically differing cell populations. Differential expression analysis identifies global changes in transcription and enables the inference of functional roles of applied perturbations. This approach has transformed the discovery of genetic drivers of disease and possible therapies. However, differential expression analysis does not provide quantitative predictions of gene expression in untested conditions. We present a hybrid approach, termed Differential Expression in Python (DiffExPy), that uniquely combines discrete, differential expression analysis with in silico differential equation simulations to yield accurate, quantitative predictions of gene expression from time-series data. Results To demonstrate the distinct insight provided by DiffExpy, we applied it to published, in vitro, time-series RNA-seq data from several genetic PI3K/PTEN variants of MCF10a cells stimulated with epidermal growth factor. DiffExPy proposed ensembles of several minimal differential equation systems for each differentially expressed gene. These systems provide quantitative models of expression for several previously uncharacterized genes and uncover new regulation by the PI3K/PTEN pathways. We validated model predictions on expression data from conditions that were not used for model training. Our discrete, differential expression analysis also identified SUZ12 and FOXA1 as possible regulators of specific groups of genes that exhibit late changes in expression. Our work reveals how DiffExPy generates quantitatively predictive models with testable, biological hypotheses from time-series expression data. Availability and implementation DiffExPy is available on GitHub (https://github.com/bagherilab/diffexpy). Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Justin D Finkle
- Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL 60208, USA
| | - Neda Bagheri
- Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL 60208, USA.,Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208, USA.,Center for Synthetic Biology, Northwestern University, Evanston, IL 60208, USA.,Chemistry of Life Processes, Northwestern University, Evanston, IL 60208, USA
| |
Collapse
|
120
|
Dai X, Xu Z, Liang Z, Tu X, Zhong S, Schnable JC, Li P. Non-homology-based prediction of gene functions in maize (Zea mays ssp. mays). THE PLANT GENOME 2020; 13:e20015. [PMID: 33016608 DOI: 10.1002/tpg2.20015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 12/22/2019] [Accepted: 02/12/2020] [Indexed: 06/11/2023]
Abstract
Advances in genome sequencing and annotation have eased the difficulty of identifying new gene sequences. Predicting the functions of these newly identified genes remains challenging. Genes descended from a common ancestral sequence are likely to have common functions. As a result, homology is widely used for gene function prediction. This means functional annotation errors also propagate from one species to another. Several approaches based on machine learning classification algorithms were evaluated for their ability to accurately predict gene function from non-homology gene features. Among the eight supervised classification algorithms evaluated, random-forest-based prediction consistently provided the most accurate gene function prediction. Non-homology-based functional annotation provides complementary strengths to homology-based annotation, with higher average performance in Biological Process GO terms, the domain where homology-based functional annotation performs the worst, and weaker performance in Molecular Function GO terms, the domain where the accuracy of homology-based functional annotation is highest. GO prediction models trained with homology-based annotations were able to successfully predict annotations from a manually curated "gold standard" GO annotation set. Non-homology-based functional annotation based on machine learning may ultimately prove useful both as a method to assign predicted functions to orphan genes which lack functionally characterized homologs, and to identify and correct functional annotation errors which were propagated through homology-based functional annotations.
Collapse
Affiliation(s)
- Xiuru Dai
- State Key Laboratory of Crop Biology, Shandong Agricultural University, Taian, 273100, China
- Quantitative Life Sciences Initiative, Center for Plant Science Innovation, and Department of Agronomy and Horticulture, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Zheng Xu
- Department of Mathematics and Statistics, Wright State University, Dayton, OH, 45435, USA
| | - Zhikai Liang
- Quantitative Life Sciences Initiative, Center for Plant Science Innovation, and Department of Agronomy and Horticulture, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Xiaoyu Tu
- State Key Laboratory of Agrobiotechnology, School of Life Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Silin Zhong
- State Key Laboratory of Agrobiotechnology, School of Life Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - James C Schnable
- Quantitative Life Sciences Initiative, Center for Plant Science Innovation, and Department of Agronomy and Horticulture, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Pinghua Li
- State Key Laboratory of Crop Biology, Shandong Agricultural University, Taian, 273100, China
| |
Collapse
|
121
|
Choudhari R, Yang B, Rotwein P, Gadad SS. Structure and expression of the long noncoding RNA gene MIR503 in humans and non-human primates. Mol Cell Endocrinol 2020; 510:110819. [PMID: 32311422 DOI: 10.1016/j.mce.2020.110819] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/07/2020] [Accepted: 04/07/2020] [Indexed: 12/27/2022]
Abstract
Recent technical and other advances in genomics provide unique opportunities to improve our understanding of human physiology and disease predisposition through a detailed analysis of gene structure and expression by examining data in public genome and gene-expression repositories. Yet, the vast majority of human genes remain understudied. This is particularly true of genes for long noncoding RNAs (lncRNAs). Here, we describe the detailed characterization of MIR503HG, a lncRNA gene found on the X chromosome in humans. Using information extracted from public databases, we show that human MIR503HG is a 5-exon gene, and that it is highly conserved among 5 non-human primates spanning over 85 million years ago of evolutionary diversification. MIR503HG is transcribed and processed into multiple distinct RNAs in each of these species through differential exon use and alternative RNA splicing, with a higher abundance of transcripts being found in reproductive tissues, especially during the early stages of ovary and testis development, indicating a possible role in reproductive biology. Furthermore, in select reproductive system cancers, MIR503HG transcripts are downregulated, with higher levels of RNA expression being associated with clinical outcomes. Collectively, these investigations show how the use of genomic, gene expression, and other genetic resources can lead to new insights about human biology and disease, and argue that MIR503HG is worthy of additional study.
Collapse
Affiliation(s)
- Ramesh Choudhari
- Center of Emphasis in Cancer, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, Texas, 79905, United States.
| | - Barbara Yang
- Center of Emphasis in Cancer, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, Texas, 79905, United States.
| | - Peter Rotwein
- Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, Texas, 79905, United States.
| | - Shrikanth S Gadad
- Center of Emphasis in Cancer, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, Texas, 79905, United States; Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, Texas, 79905, United States; Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, Texas, 79905, United States; Cecil H. and Ida Green Center for Reproductive Biology Sciences and Division of Basic Reproductive Biology Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, United States.
| |
Collapse
|
122
|
Zlotnik A. Perspective: Insights on the Nomenclature of Cytokines and Chemokines. Front Immunol 2020; 11:908. [PMID: 32499780 PMCID: PMC7243804 DOI: 10.3389/fimmu.2020.00908] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 04/20/2020] [Indexed: 12/02/2022] Open
Affiliation(s)
- Albert Zlotnik
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
123
|
Abstract
Cardiovascular diseases are the leading cause of death worldwide. Complex diseases with highly heterogenous disease progression among patient populations, cardiovascular diseases feature multifactorial contributions from both genetic and environmental stressors. Despite significant effort utilizing multiple approaches from molecular biology to genome-wide association studies, the genetic landscape of cardiovascular diseases, particularly for the nonfamilial forms of heart failure, is still poorly understood. In the past decade, systems-level approaches based on omics technologies have become an important approach for the study of complex traits in large populations. These advances create opportunities to integrate genetic variation with other biological layers to identify and prioritize candidate genes, understand pathogenic pathways, and elucidate gene-gene and gene-environment interactions. In this review, we will highlight some of the recent progress made using systems genetics approaches to uncover novel mechanisms and molecular bases of cardiovascular pathophysiological manifestations. The key technology and data analysis platforms necessary to implement systems genetics will be described, and the current major challenges and future directions will also be discussed. For complex cardiovascular diseases, such as heart failure, systems genetics represents a powerful strategy to obtain mechanistic insights and to develop individualized diagnostic and therapeutic regiments, paving the way for precision cardiovascular medicine.
Collapse
Affiliation(s)
- Christoph D. Rau
- Departments of Anesthesiology, Medicine, Physiology
- Current address: Department of Genetics, University of North Carolina School of Medicine, Chapel Hill, NC 27599
| | - Aldons J. Lusis
- Department of Human Genetics and Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Yibin Wang
- Departments of Anesthesiology, Medicine, Physiology
| |
Collapse
|
124
|
Schwinn MK, Steffen LS, Zimmerman K, Wood KV, Machleidt T. A Simple and Scalable Strategy for Analysis of Endogenous Protein Dynamics. Sci Rep 2020; 10:8953. [PMID: 32488146 PMCID: PMC7265437 DOI: 10.1038/s41598-020-65832-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 05/08/2020] [Indexed: 11/20/2022] Open
Abstract
The ability to analyze protein function in a native context is central to understanding cellular physiology. This study explores whether tagging endogenous proteins with a reporter is a scalable strategy for generating cell models that accurately quantitate protein dynamics. Specifically, it investigates whether CRISPR-mediated integration of the HiBiT luminescent peptide tag can easily be accomplished on a large-scale and whether integrated reporter faithfully represents target biology. For this purpose, a large set of proteins representing diverse structures and functions, some of which are known or potential drug targets, were targeted for tagging with HiBiT in multiple cell lines. Successful insertion was detected for 86% of the targets, as determined by luminescence-based plate assays, blotting, and imaging. In order to determine whether endogenously tagged proteins yield more representative models, cells expressing HiBiT protein fusions either from endogenous loci or plasmids were directly compared in functional assays. In the tested cases, only the edited lines were capable of accurately reproducing the anticipated biology. This study provides evidence that cell lines expressing HiBiT fusions from endogenous loci can be rapidly generated for many different proteins and that these cellular models provide insight into protein function that may be unobtainable using overexpression-based approaches.
Collapse
Affiliation(s)
- Marie K Schwinn
- Promega Corporation, Madison, Wisconsin, 53711, United States.
| | - Leta S Steffen
- Promega Corporation, Madison, Wisconsin, 53711, United States
| | - Kris Zimmerman
- Promega Corporation, Madison, Wisconsin, 53711, United States
| | - Keith V Wood
- Light Bio, Inc., Madison, Wisconsin, 53711, United States
| | | |
Collapse
|
125
|
Rotwein P. The Zmat2 gene in non-mammalian vertebrates: Organizational simplicity within a divergent locus in fish. PLoS One 2020; 15:e0233081. [PMID: 32463827 PMCID: PMC7255616 DOI: 10.1371/journal.pone.0233081] [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: 01/16/2020] [Accepted: 04/25/2020] [Indexed: 11/22/2022] Open
Abstract
ZMAT2 is among the least-studied of mammalian proteins and genes, even though it is the ortholog of Snu23, a protein involved in pre-mRNA splicing in yeast. Here we have used data from genomic and gene expression repositories to examine the Zmat2 gene and locus in 8 terrestrial vertebrates, 10 ray-finned fish, and 1 lobe-finned fish representing > 500 million years of evolutionary diversification. The analyses revealed that vertebrate Zmat2 genes are similar to their mammalian counterparts, as in 16/19 species studied they contain 6 exons, and in 18/19 encode a single conserved protein. However, unlike in mammals, no Zmat2 pseudogenes were identified in these vertebrates, although an expressed Zmat2 paralog was characterized in flycatcher that resembled a DNA copy of a processed and retro-transposed mRNA, and thus could be a proto-pseudogene captured during its evolutionary journey from active to inert. The Zmat2 locus in terrestrial vertebrates, and in spotted gar and coelacanth, also shares additional genes with its mammalian counterparts, including Histidyl-tRNA synthetase (Hars), Hars2, and others, but these are absent from the Zmat2 locus in teleost fish, in which Stem-loop-binding protein 2 (Slbp2) and Lymphocyte cytosolic protein 2a (Lcp2a) are present instead. Taken together, these observations argue that a recognizable Zmat2 was present in the earliest vertebrate ancestors, and postulate that during chromosomal tetraploidization and subsequent re-diploidization during modern teleost evolution, the duplicated Zmat2 gene was retained and the original lost. This study also highlights how information from genomic resources can be leveraged to reveal new biologically significant insights.
Collapse
Affiliation(s)
- Peter Rotwein
- Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech Health University Health Sciences Center, El Paso, Texas, United States of America
| |
Collapse
|
126
|
Abstract
Surprisingly we remain ignorant of the function of the majority of genes in the human and mouse genomes. The dark genome is a major obstacle to the interpretation of the function of human genetic variation and its impact on disease. At the same time, pleiotropy, how individual variants influence multiple phenotypes, is key to understanding gene function and the role of genes and genetic networks in disease systems. Both understanding the genetics of disease and developing new therapeutic approaches and advances in precision medicine are all compromised by our limited knowledge of gene function and pleiotropic effects. Illuminating the dark genome and revealing pleiotropy across the genome requires a highly coordinated and international effort to acquire and analyse high-dimensional phenotype data from model organisms. We describe briefly how the International Mouse Phenotyping Consortium is addressing these challenges and the novel features of the pleiotropic landscape that are revealed by functional genomics programmes at genome-wide scale.
Collapse
Affiliation(s)
| | - Heena V Lad
- MRC Harwell Institute, Harwell, OX11 0RD, UK
| |
Collapse
|
127
|
Sung AY, Floyd BJ, Pagliarini DJ. Systems Biochemistry Approaches to Defining Mitochondrial Protein Function. Cell Metab 2020; 31:669-678. [PMID: 32268114 PMCID: PMC7176052 DOI: 10.1016/j.cmet.2020.03.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 03/06/2020] [Accepted: 03/13/2020] [Indexed: 02/07/2023]
Abstract
Defining functions for the full complement of proteins is a grand challenge in the post-genomic era and is essential for our understanding of basic biology and disease pathogenesis. In recent times, this endeavor has benefitted from a combination of modern large-scale and classical reductionist approaches-a process we refer to as "systems biochemistry"-that helps surmount traditional barriers to the characterization of poorly understood proteins. This strategy is proving to be particularly effective for mitochondria, whose well-defined proteome has enabled comprehensive analyses of the full mitochondrial system that can position understudied proteins for fruitful mechanistic investigations. Recent systems biochemistry approaches have accelerated the identification of new disease-related mitochondrial proteins and of long-sought "missing" proteins that fulfill key functions. Collectively, these studies are moving us toward a more complete understanding of mitochondrial activities and providing a molecular framework for the investigation of mitochondrial pathogenesis.
Collapse
Affiliation(s)
- Andrew Y Sung
- Morgridge Institute for Research, Madison, WI, USA; Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA; School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Brendan J Floyd
- Morgridge Institute for Research, Madison, WI, USA; Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA; Department of Pediatrics, Stanford School of Medicine, Stanford, CA, USA
| | - David J Pagliarini
- Morgridge Institute for Research, Madison, WI, USA; Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
128
|
Baxter I. We aren't good at picking candidate genes, and it's slowing us down. CURRENT OPINION IN PLANT BIOLOGY 2020; 54:57-60. [PMID: 32106014 DOI: 10.1016/j.pbi.2020.01.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/05/2020] [Accepted: 01/28/2020] [Indexed: 05/26/2023]
Abstract
In order to gain a molecular understanding of the genetic basis for plant traits, we need to be able to identify the underlying gene and the causal allele for genetic loci. This process usually involves a step where a researcher selects likely candidate genes from a list. The process of picking candidate genes is inherently prone to distortion due to human bias, and this is slowing down our research enterprise.
Collapse
Affiliation(s)
- Ivan Baxter
- Donald Danforth Plant Science Center, United States.
| |
Collapse
|
129
|
Abstract
A key goal of cancer systems biology is to use big data to elucidate the molecular networks by which cancer develops. However, to date there has been no systematic evaluation of how far these efforts have progressed. In this Analysis, we survey six major systems biology approaches for mapping and modelling cancer pathways with attention to how well their resulting network maps cover and enhance current knowledge. Our sample of 2,070 systems biology maps captures all literature-curated cancer pathways with significant enrichment, although the strong tendency is for these maps to recover isolated mechanisms rather than entire integrated processes. Systems biology maps also identify previously underappreciated functions, such as a potential role for human papillomavirus-induced chromosomal alterations in ovarian tumorigenesis, and they add new genes to known cancer pathways, such as those related to metabolism, Hippo signalling and immunity. Notably, we find that many cancer networks have been provided only in journal figures and not for programmatic access, underscoring the need to deposit network maps in community databases to ensure they can be readily accessed. Finally, few of these findings have yet been clinically translated, leaving ample opportunity for future translational studies. Periodic surveys of cancer pathway maps, such as the one reported here, are critical to assess progress in the field and identify underserved areas of methodology and cancer biology.
Collapse
Affiliation(s)
- Brent M Kuenzi
- Division of Genetics, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Trey Ideker
- Division of Genetics, Department of Medicine, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
130
|
Nagy LG, Merényi Z, Hegedüs B, Bálint B. Novel phylogenetic methods are needed for understanding gene function in the era of mega-scale genome sequencing. Nucleic Acids Res 2020; 48:2209-2219. [PMID: 31943056 PMCID: PMC7049691 DOI: 10.1093/nar/gkz1241] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 12/15/2019] [Accepted: 12/31/2019] [Indexed: 12/21/2022] Open
Abstract
Ongoing large-scale genome sequencing projects are forecasting a data deluge that will almost certainly overwhelm current analytical capabilities of evolutionary genomics. In contrast to population genomics, there are no standardized methods in evolutionary genomics for extracting evolutionary and functional (e.g. gene-trait association) signal from genomic data. Here, we examine how current practices of multi-species comparative genomics perform in this aspect and point out that many genomic datasets are under-utilized due to the lack of powerful methodologies. As a result, many current analyses emphasize gene families for which some functional data is already available, resulting in a growing gap between functionally well-characterized genes/organisms and the universe of unknowns. This leaves unknown genes on the 'dark side' of genomes, a problem that will not be mitigated by sequencing more and more genomes, unless we develop tools to infer functional hypotheses for unknown genes in a systematic manner. We provide an inventory of recently developed methods capable of predicting gene-gene and gene-trait associations based on comparative data, then argue that realizing the full potential of whole genome datasets requires the integration of phylogenetic comparative methods into genomics, a rich but underutilized toolbox for looking into the past.
Collapse
Affiliation(s)
- László G Nagy
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Temesvari krt 62. Szeged 6726, Hungary
| | - Zsolt Merényi
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Temesvari krt 62. Szeged 6726, Hungary
| | - Botond Hegedüs
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Temesvari krt 62. Szeged 6726, Hungary
| | - Balázs Bálint
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Temesvari krt 62. Szeged 6726, Hungary
| |
Collapse
|
131
|
Haselimashhadi H, Mason JC, Munoz-Fuentes V, López-Gómez F, Babalola K, Acar EF, Kumar V, White J, Flenniken AM, King R, Straiton E, Seavitt JR, Gaspero A, Garza A, Christianson AE, Hsu CW, Reynolds CL, Lanza DG, Lorenzo I, Green JR, Gallegos JJ, Bohat R, Samaco RC, Veeraragavan S, Kim JK, Miller G, Fuchs H, Garrett L, Becker L, Kang YK, Clary D, Cho SY, Tamura M, Tanaka N, Soo KD, Bezginov A, About GB, Champy MF, Vasseur L, Leblanc S, Meziane H, Selloum M, Reilly PT, Spielmann N, Maier H, Gailus-Durner V, Sorg T, Hiroshi M, Yuichi O, Heaney JD, Dickinson ME, Wolfgang W, Tocchini-Valentini GP, Lloyd KCK, McKerlie C, Seong JK, Yann H, de Angelis MH, Brown SDM, Smedley D, Flicek P, Mallon AM, Parkinson H, Meehan TF. Soft windowing application to improve analysis of high-throughput phenotyping data. Bioinformatics 2020; 36:1492-1500. [PMID: 31591642 PMCID: PMC7115897 DOI: 10.1093/bioinformatics/btz744] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 08/20/2019] [Accepted: 10/04/2019] [Indexed: 11/14/2022] Open
Abstract
Motivation High-throughput phenomic projects generate complex data from small treatment and large control groups that increase the power of the analyses but introduce variation over time. A method is needed to utlize a set of temporally local controls that maximizes analytic power while minimizing noise from unspecified environmental factors. Results Here we introduce ‘soft windowing’, a methodological approach that selects a window of time that includes the most appropriate controls for analysis. Using phenotype data from the International Mouse Phenotyping Consortium (IMPC), adaptive windows were applied such that control data collected proximally to mutants were assigned the maximal weight, while data collected earlier or later had less weight. We applied this method to IMPC data and compared the results with those obtained from a standard non-windowed approach. Validation was performed using a resampling approach in which we demonstrate a 10% reduction of false positives from 2.5 million analyses. We applied the method to our production analysis pipeline that establishes genotype–phenotype associations by comparing mutant versus control data. We report an increase of 30% in significant P-values, as well as linkage to 106 versus 99 disease models via phenotype overlap with the soft-windowed and non-windowed approaches, respectively, from a set of 2082 mutant mouse lines. Our method is generalizable and can benefit large-scale human phenomic projects such as the UK Biobank and the All of Us resources. Availability and implementation The method is freely available in the R package SmoothWin, available on CRAN http://CRAN.R-project.org/package=SmoothWin. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Hamed Haselimashhadi
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | - Jeremy C Mason
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | - Violeta Munoz-Fuentes
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | - Federico López-Gómez
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | - Kolawole Babalola
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | - Elif F Acar
- The Centre for Phenogenomics.,The Hospital for Sick Children, Toronto, Canada.,Department of Statistics, University of Manitoba, Winnipeg, MB R3T 2N2 Canada
| | - Vivek Kumar
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - Jacqui White
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - Ann M Flenniken
- The Centre for Phenogenomics.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | - Ritu Bohat
- Baylor College of Medicine, Houston, TX, USA
| | | | | | - Jong Kyoung Kim
- Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu, Korea
| | | | | | | | - Lore Becker
- Helmholtz Center Munich, Neuherberg, Germany
| | | | - David Clary
- Mouse Biology Program, University of California Davis, Davis, CA, USA
| | - Soo Young Cho
- National Cancer Center (NCC) & Korea Mouse Phenotyping Center (KMPC), Korea
| | | | | | - Kyung Dong Soo
- Seoul National University & Korea Mouse Phenotyping Center (KMPC), Korea
| | - Alexandr Bezginov
- The Centre for Phenogenomics.,The Hospital for Sick Children, Toronto, Canada
| | - Ghina Bou About
- Université de Strasbourg, CNRS, INSERM, Institut Clinique de la Souris, PHENOMIN-ICS, 67404 Illkirch, France
| | - Marie-France Champy
- Université de Strasbourg, CNRS, INSERM, Institut Clinique de la Souris, PHENOMIN-ICS, 67404 Illkirch, France
| | - Laurent Vasseur
- Université de Strasbourg, CNRS, INSERM, Institut Clinique de la Souris, PHENOMIN-ICS, 67404 Illkirch, France
| | - Sophie Leblanc
- Université de Strasbourg, CNRS, INSERM, Institut Clinique de la Souris, PHENOMIN-ICS, 67404 Illkirch, France
| | - Hamid Meziane
- Université de Strasbourg, CNRS, INSERM, Institut Clinique de la Souris, PHENOMIN-ICS, 67404 Illkirch, France
| | - Mohammed Selloum
- Université de Strasbourg, CNRS, INSERM, Institut Clinique de la Souris, PHENOMIN-ICS, 67404 Illkirch, France
| | - Patrick T Reilly
- Université de Strasbourg, CNRS, INSERM, Institut Clinique de la Souris, PHENOMIN-ICS, 67404 Illkirch, France
| | | | | | | | - Tania Sorg
- Université de Strasbourg, CNRS, INSERM, Institut Clinique de la Souris, PHENOMIN-ICS, 67404 Illkirch, France
| | | | - Obata Yuichi
- RIKEN BioResource Research Center, Tsukuba, Japan
| | | | | | - Wurst Wolfgang
- Institute of Developmental Genetics, Helmholtz Centre Munich, Munich, Germany
| | | | | | - Colin McKerlie
- The Centre for Phenogenomics.,The Hospital for Sick Children, Toronto, Canada
| | - Je Kyung Seong
- Seoul National University & Korea Mouse Phenotyping Center (KMPC), Korea
| | - Herault Yann
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique, Biologie Moléculaire et Cellulaire, Institut Clinique de la Souris, IGBMC, PHENOMIN-ICS, 67404 Illkirch, France
| | | | | | - Damian Smedley
- William Harvey Research Institute, Charterhouse Square Barts and the London School of Medicine and Dentistry Queen Mary University of London, London EC1M 6BQ, UK
| | - Paul Flicek
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | | | - Helen Parkinson
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | - Terrence F Meehan
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| |
Collapse
|
132
|
Byrne JA, Christopher J. Digital magic, or the dark arts of the 21 st century-how can journals and peer reviewers detect manuscripts and publications from paper mills? FEBS Lett 2020; 594:583-589. [PMID: 32067229 DOI: 10.1002/1873-3468.13747] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In recent years, it has been proposed that unrealistic requirements for academics and medical doctors to publish in scientific journals, combined with monetary publication rewards, have led to forms of contract cheating offered by organizations known as paper mills. Paper mills are alleged to offer products ranging from research data through to ghostwritten fraudulent or fabricated manuscripts and submission services. While paper mill operations remain poorly understood, it seems likely that paper mills need to balance product quantity and quality, such that they produce or contribute to large numbers of manuscripts that will be accepted for publication. Producing manuscripts at scale may be facilitated by the use of manuscript templates, which could give rise to shared features such as textual and organizational similarities, the description of highly generic study hypotheses and experimental approaches, digital images that show evidence of manipulation and/or reuse, and/or errors affecting verifiable experimental reagents. Based on these features, we propose practical steps that editors, journal staff, and peer reviewers can take to recognize and respond to research manuscripts and publications that may have been produced with undeclared assistance from paper mills.
Collapse
Affiliation(s)
- Jennifer A Byrne
- NSW Health Statewide Biobank, Camperdown, NSW, Australia.,School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, NSW, Australia
| | - Jana Christopher
- FEBS Letters Editorial Office, Heidelberg University Biochemistry Center, Germany
| |
Collapse
|
133
|
Boycott KM, Campeau PM, Howley HE, Pavlidis P, Rogic S, Oriel C, Berman JN, Hamilton RM, Hicks GG, Lipshitz HD, Masson JY, Shoubridge EA, Junker A, Leroux MR, McMaster CR, Michaud JL, Turvey SE, Dyment D, Innes AM, van Karnebeek CD, Lehman A, Cohn RD, MacDonald IM, Rachubinski RA, Frosk P, Vandersteen A, Wozniak RW, Pena IA, Wen XY, Lacaze-Masmonteil T, Rankin C, Hieter P. The Canadian Rare Diseases Models and Mechanisms (RDMM) Network: Connecting Understudied Genes to Model Organisms. Am J Hum Genet 2020; 106:143-152. [PMID: 32032513 DOI: 10.1016/j.ajhg.2020.01.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 01/10/2020] [Indexed: 01/14/2023] Open
Abstract
Advances in genomics have transformed our ability to identify the genetic causes of rare diseases (RDs), yet we have a limited understanding of the mechanistic roles of most genes in health and disease. When a novel RD gene is first discovered, there is minimal insight into its biological function, the pathogenic mechanisms of disease-causing variants, and how therapy might be approached. To address this gap, the Canadian Rare Diseases Models and Mechanisms (RDMM) Network was established to connect clinicians discovering new disease genes with Canadian scientists able to study equivalent genes and pathways in model organisms (MOs). The Network is built around a registry of more than 500 Canadian MO scientists, representing expertise for over 7,500 human genes. RDMM uses a committee process to identify and evaluate clinician-MO scientist collaborations and approve 25,000 Canadian dollars in catalyst funding. To date, we have made 85 clinician-MO scientist connections and funded 105 projects. These collaborations help confirm variant pathogenicity and unravel the molecular mechanisms of RD, and also test novel therapies and lead to long-term collaborations. To expand the impact and reach of this model, we made the RDMM Registry open-source, portable, and customizable, and we freely share our committee structures and processes. We are currently working with emerging networks in Europe, Australia, and Japan to link international RDMM networks and registries and enable matches across borders. We will continue to create meaningful collaborations, generate knowledge, and advance RD research locally and globally for the benefit of patients and families living with RD.
Collapse
Affiliation(s)
- Kym M Boycott
- CHEO Research Institute, University of Ottawa, Ottawa, ON K1H 8L1, Canada.
| | - Philippe M Campeau
- Centre de Recherche du CHU Ste-Justine, Department of Pediatrics, Université de Montréal, Montréal, QC H3T 1C5, Canada
| | - Heather E Howley
- CHEO Research Institute, University of Ottawa, Ottawa, ON K1H 8L1, Canada
| | - Paul Pavlidis
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Department of Psychiatry, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Sanja Rogic
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Department of Psychiatry, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Christine Oriel
- Maternal Infant Child and Youth Research Network (MICYRN), Vancouver, BC V5Z 4H4, Canada
| | - Jason N Berman
- CHEO Research Institute, University of Ottawa, Ottawa, ON K1H 8L1, Canada
| | - Robert M Hamilton
- Labatt Family Heart Centre and Translational Medicine, Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Geoffrey G Hicks
- Regenerative Medicine Program, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 3P5, Canada; Department of Biochemistry and Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 3P5, Canada
| | - Howard D Lipshitz
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Jean-Yves Masson
- Oncology Division, CHU de Québec-Université Laval, Laval University Cancer Research Center, Quebec City, QC, G1R 3S3, Canada
| | - Eric A Shoubridge
- Department of Human Genetics, Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
| | - Anne Junker
- Department of Pediatrics, British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, BC V6H 3N1, Canada
| | - Michel R Leroux
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | | | - Jaques L Michaud
- Centre de Recherche du CHU Ste-Justine, Department of Pediatrics, Université de Montréal, Montréal, QC H3T 1C5, Canada
| | - Stuart E Turvey
- Department of Human Genetics, Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
| | - David Dyment
- CHEO Research Institute, University of Ottawa, Ottawa, ON K1H 8L1, Canada
| | - A Micheil Innes
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Alberta Children's Hospital, Calgary, AB T2N 4N1, Canada
| | - Clara D van Karnebeek
- Department of Human Genetics, Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada; Department of Pediatrics, Amsterdam University Medical Centres, Amsterdam, the Netherlands; Department of Clinical Genetics, Amsterdam University Medical Centres, Amsterdam, the Netherlands
| | - Anna Lehman
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6H 3N1, Canada
| | - Ronald D Cohn
- Genetics and Genome Biology Program, SickKids Research Institute, Department of Paediatrics and Molecular Genetics, University of Toronto, Toronto, ON M5G 0A4, Canada
| | - Ian M MacDonald
- Department of Ophthalmology and Visual Sciences, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R7, Canada
| | - Richard A Rachubinski
- Genetics and Genome Biology Program, SickKids Research Institute, Department of Paediatrics and Molecular Genetics, University of Toronto, Toronto, ON M5G 0A4, Canada
| | - Patrick Frosk
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB R3A 1S1, Canada
| | - Anthony Vandersteen
- Department of Pediatrics, Maritime Medical Genetics Service, Dalhousie University, IWK Health Centre, Halifax, NS B3K 6R8, Canada
| | - Richard W Wozniak
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Izabella A Pena
- CHEO Research Institute, University of Ottawa, Ottawa, ON K1H 8L1, Canada
| | - Xiao-Yan Wen
- Zebrafish Centre for Advanced Drug Discovery, Keenan Research Centre for Biomedical Science, St Michael's Hospital, Unity Health Toronto, Department of Medicine, University of Toronto, Toronto, ON M5B 1T8
| | - Thierry Lacaze-Masmonteil
- Maternal Infant Child and Youth Research Network (MICYRN), Vancouver, BC V5Z 4H4, Canada; Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Catharine Rankin
- Department of Psychology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Philip Hieter
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada.
| |
Collapse
|
134
|
Lloyd KCK, Adams DJ, Baynam G, Beaudet AL, Bosch F, Boycott KM, Braun RE, Caulfield M, Cohn R, Dickinson ME, Dobbie MS, Flenniken AM, Flicek P, Galande S, Gao X, Grobler A, Heaney JD, Herault Y, de Angelis MH, Lupski JR, Lyonnet S, Mallon AM, Mammano F, MacRae CA, McInnes R, McKerlie C, Meehan TF, Murray SA, Nutter LMJ, Obata Y, Parkinson H, Pepper MS, Sedlacek R, Seong JK, Shiroishi T, Smedley D, Tocchini-Valentini G, Valle D, Wang CKL, Wells S, White J, Wurst W, Xu Y, Brown SDM. The Deep Genome Project. Genome Biol 2020; 21:18. [PMID: 32008577 PMCID: PMC6996159 DOI: 10.1186/s13059-020-1931-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 01/08/2020] [Indexed: 12/12/2022] Open
Affiliation(s)
- K. C. Kent Lloyd
- Department of Surgery, School of Medicine, and Mouse Biology Program, University of California, Davis, CA 95618 USA
| | - David J. Adams
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1SA UK
| | - Gareth Baynam
- Western Australian Register of Developmental Anomalies and Genetic Services of Western Australia, Department of Health, Government of Western Australia, Perth, Australia
- Division of Paediatrics and Telethon Kids Institute, Faculty of Health and Medical Sciences, University of Western Australia, Perth, Australia
- Faculty of Science and Engineering, School of Spatial Sciences, Curtin University, Perth, Australia
| | - Arthur L. Beaudet
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030 USA
| | - Fatima Bosch
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma Barcelona, Barcelona, Spain
| | - Kym M. Boycott
- Children’s Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON K1H 8L1 Canada
| | | | - Mark Caulfield
- Genomics England, William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ UK
| | - Ronald Cohn
- The Hospital for Sick Children, Toronto, ON M5G 1X8 Canada
| | - Mary E. Dickinson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030 USA
- Departments of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030 USA
| | - Michael S. Dobbie
- Phenomics Australia, The Australian National University, 131 Garran Road, Acton, ACT 2601 Australia
| | - Ann M. Flenniken
- The Centre for Phenogenomics, Lunenfeld-Tanenbaum Research Institute, Toronto, ON M5T 3H7 Canada
| | - Paul Flicek
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD UK
| | - Sanjeev Galande
- National Facility for Gene Function in Health and Disease, Department of Biology, Indian Institute of Science, Education and Research (IISER) Pune, Pune, Maharashtra 411008 India
| | - Xiang Gao
- SKL of Pharmaceutical Biotechnology and Model Animal Research Center, Collaborative Innovation Center for Genetics and Development, Nanjing Biomedical Research Institute, Nanjing University, Nanjing, 210061 China
| | - Anne Grobler
- DST/NWU Preclinical Drug Development Platform, North-West University, Potchefstroom, 2520 South Africa
| | - Jason D. Heaney
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030 USA
| | - Yann Herault
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique, Biologie Moléculaire et Cellulaire, Institut Clinique de la Souris, IGBMC, PHENOMIN-ICS, 67404 Illkirch, France
| | - Martin Hrabě de Angelis
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Chair of Experimental Genetics, Center of Life and Food Sciences Weihenstephan, Technische Universität München, 85354 Freising-Weihenstephan, Germany
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - James R. Lupski
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030 USA
| | - Stanislas Lyonnet
- Institut Imagine, UMR-1163 INSERM et Université de Paris, Hôpital Universitaire Necker-Enfants Malades, 24, Boulevard du Montparnasse, 75015 Paris, France
| | - Ann-Marie Mallon
- Medical Research Council Harwell Institute (Mammalian Genetics Unit and Mary Lyon Centre), Harwell, Oxfordshire OX11 0RD UK
| | - Fabio Mammano
- Monterotondo Mouse Clinic, Italian National Research Council (CNR), Institute of Biochemistry and Cell Biology (IBBC), Monterotondo Scalo, I-00015 Rome, Italy
| | - Calum A. MacRae
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, USA
| | - Roderick McInnes
- Lady Davis Research Institute, Jewish General Hospital, McGill University, 3999 Côte Ste- Catherine Road, Montreal, Quebec H3T 1E2 Canada
| | - Colin McKerlie
- The Hospital for Sick Children, Toronto, ON M5G 1X8 Canada
- The Centre for Phenogenomics, The Hospital for Sick Children, Toronto, ON M5T 3H7 Canada
| | - Terrence F. Meehan
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD UK
| | | | - Lauryl M. J. Nutter
- The Centre for Phenogenomics, The Hospital for Sick Children, Toronto, ON M5T 3H7 Canada
| | - Yuichi Obata
- RIKEN BioResource Research Center, Tsukuba, Ibaraki, 305-0074 Japan
| | - Helen Parkinson
- National Facility for Gene Function in Health and Disease, Department of Biology, Indian Institute of Science, Education and Research (IISER) Pune, Pune, Maharashtra 411008 India
| | - Michael S. Pepper
- Institute for Cellular and Molecular Medicine, Department Immunology, and SAMRC Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Radislav Sedlacek
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, 252 50 Vestec, Czech Republic
| | - Je Kyung Seong
- Korea Mouse Phenotyping Consortium (KMPC) and BK21 Program for Veterinary Science, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, 599 Gwanangno, Gwanak-gu, Seoul, 08826 South Korea
| | | | - Damian Smedley
- Clinical Pharmacology, William Harvey Research Institute, School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ UK
| | - Glauco Tocchini-Valentini
- Monterotondo Mouse Clinic, Italian National Research Council (CNR), Institute of Biochemistry and Cell Biology (IBBC), Monterotondo Scalo, I-00015 Rome, Italy
| | - David Valle
- McKusick-Nathans Department of Genetic Medicine, The Johns Hopkins University School of Medicine, 519 BRB, 733 N Broadway, Baltimore, MD 21205 USA
| | - Chi-Kuang Leo Wang
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan
| | - Sara Wells
- Medical Research Council Harwell Institute (Mammalian Genetics Unit and Mary Lyon Centre), Harwell, Oxfordshire OX11 0RD UK
| | | | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany
- Chair of Developmental Genetics, Center of Life and Food Sciences Weihenstephan, Technische Universität München, 85354 Freising-Weihenstephan, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Munich Cluster for Systems Neurology (SyNergy), Adolf-Butenandt-Institut, Ludwig Maximillian’s Universitat Munchen, 81377 Munich, Germany
| | - Ying Xu
- Cambridge-Suda Genomic Resource Center, Jiangsu Key Laboratory of Neuropsychiatric Diseases, Medical College of Soochow University, Suzhou, 215123 Jiangsu China
| | - Steve D. M. Brown
- Medical Research Council Harwell Institute (Mammalian Genetics Unit and Mary Lyon Centre), Harwell, Oxfordshire OX11 0RD UK
| |
Collapse
|
135
|
Rotwein P, Baral K. Zmat2 in mammals: conservation and diversification among genes and Pseudogenes. BMC Genomics 2020; 21:113. [PMID: 32005145 PMCID: PMC6995233 DOI: 10.1186/s12864-020-6506-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 01/17/2020] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Recent advances in genetics and genomics present unique opportunities for enhancing our understanding of mammalian biology and evolution through detailed multi-species comparative analysis of gene organization and expression. Yet, of the more than 20,000 protein coding genes found in mammalian genomes, fewer than 10% have been examined in any detail. Here we elucidate the power of data available in publicly-accessible genomic and genetic resources by querying them to evaluate Zmat2, a minimally studied gene whose human ortholog has been implicated in spliceosome function and in keratinocyte differentiation. RESULTS We find extensive conservation in coding regions and overall structure of Zmat2 in 18 mammals representing 13 orders and spanning ~ 165 million years of evolutionary development, and in their encoded proteins. We identify a tandem duplication in the Zmat2 gene and locus in opossum, but not in other monotremes, marsupials, or other mammals, indicating that this event occurred subsequent to the divergence of these species from one another. We also define a collection of Zmat2 pseudogenes in half of the mammals studied, and suggest based on phylogenetic analysis that they each arose independently in the recent evolutionary past. CONCLUSIONS Mammalian Zmat2 genes and ZMAT2 proteins illustrate conservation of structure and sequence, along with the development and diversification of pseudogenes in a large fraction of species. Collectively, these observations also illustrate how the focused identification and interpretation of data found in public genomic and gene expression resources can be leveraged to reveal new insights of potentially high biological significance.
Collapse
Affiliation(s)
- Peter Rotwein
- Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech Health University Health Sciences Center, El Paso, TX, 79905, USA.
| | - Kabita Baral
- Graduate School, College of Science, University of Texas at El Paso, El Paso, TX, 79902, USA
| |
Collapse
|
136
|
McDiarmid TA, Belmadani M, Liang J, Meili F, Mathews EA, Mullen GP, Hendi A, Wong WR, Rand JB, Mizumoto K, Haas K, Pavlidis P, Rankin CH. Systematic phenomics analysis of autism-associated genes reveals parallel networks underlying reversible impairments in habituation. Proc Natl Acad Sci U S A 2020; 117:656-667. [PMID: 31754030 PMCID: PMC6968627 DOI: 10.1073/pnas.1912049116] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
A major challenge facing the genetics of autism spectrum disorders (ASDs) is the large and growing number of candidate risk genes and gene variants of unknown functional significance. Here, we used Caenorhabditis elegans to systematically functionally characterize ASD-associated genes in vivo. Using our custom machine vision system, we quantified 26 phenotypes spanning morphology, locomotion, tactile sensitivity, and habituation learning in 135 strains each carrying a mutation in an ortholog of an ASD-associated gene. We identified hundreds of genotype-phenotype relationships ranging from severe developmental delays and uncoordinated movement to subtle deficits in sensory and learning behaviors. We clustered genes by similarity in phenomic profiles and used epistasis analysis to discover parallel networks centered on CHD8•chd-7 and NLGN3•nlg-1 that underlie mechanosensory hyperresponsivity and impaired habituation learning. We then leveraged our data for in vivo functional assays to gauge missense variant effect. Expression of wild-type NLG-1 in nlg-1 mutant C. elegans rescued their sensory and learning impairments. Testing the rescuing ability of conserved ASD-associated neuroligin variants revealed varied partial loss of function despite proper subcellular localization. Finally, we used CRISPR-Cas9 auxin-inducible degradation to determine that phenotypic abnormalities caused by developmental loss of NLG-1 can be reversed by adult expression. This work charts the phenotypic landscape of ASD-associated genes, offers in vivo variant functional assays, and potential therapeutic targets for ASD.
Collapse
Affiliation(s)
- Troy A McDiarmid
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 2B5, Canada
| | - Manuel Belmadani
- Department of Psychiatry, University of British Columbia, Vancouver, BC V6T 2A1, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Joseph Liang
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 2B5, Canada
| | - Fabian Meili
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 2B5, Canada
| | - Eleanor A Mathews
- Genetic Models of Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
| | - Gregory P Mullen
- Biology Program, Oklahoma City University, Oklahoma City, OK 73106
| | - Ardalan Hendi
- Department of Zoology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Wan-Rong Wong
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - James B Rand
- Genetic Models of Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| | - Kota Mizumoto
- Department of Zoology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Kurt Haas
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 2B5, Canada
| | - Paul Pavlidis
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 2B5, Canada
- Department of Psychiatry, University of British Columbia, Vancouver, BC V6T 2A1, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Catharine H Rankin
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 2B5, Canada;
- Department of Psychology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
137
|
Abstract
The 11 existing FDA-approved osteoporosis drug treatments include hormone replacement therapy, 2 SERMs (raloxifene and bazedoxifene), 5 inhibitors of bone-resorbing osteoclasts (4 bisphosphonates and anti-RANKL denosumab), 2 parathyroid hormone analogues (teriparatide and abaloparatide), and 1 WNT signaling enhancer (romosozumab). These therapies are effective and provide multiple options for patients and physicians. As the genomic revolution continues, potential novel targets for future drug development are identified. This review takes a wide perspective to describe potentially rewarding topics to explore, including knowledge of genes and pathways involved in bone cell metabolism, the utility of animal models, targeting drugs to bone, and ongoing advances in drug design and delivery.
Collapse
|
138
|
Paananen J, Fortino V. An omics perspective on drug target discovery platforms. Brief Bioinform 2019; 21:1937-1953. [PMID: 31774113 PMCID: PMC7711264 DOI: 10.1093/bib/bbz122] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/23/2019] [Accepted: 07/27/2019] [Indexed: 01/28/2023] Open
Abstract
The drug discovery process starts with identification of a disease-modifying target. This critical step traditionally begins with manual investigation of scientific literature and biomedical databases to gather evidence linking molecular target to disease, and to evaluate the efficacy, safety and commercial potential of the target. The high-throughput and affordability of current omics technologies, allowing quantitative measurements of many putative targets (e.g. DNA, RNA, protein, metabolite), has exponentially increased the volume of scientific data available for this arduous task. Therefore, computational platforms identifying and ranking disease-relevant targets from existing biomedical data sources, including omics databases, are needed. To date, more than 30 drug target discovery (DTD) platforms exist. They provide information-rich databases and graphical user interfaces to help scientists identify putative targets and pre-evaluate their therapeutic efficacy and potential side effects. Here we survey and compare a set of popular DTD platforms that utilize multiple data sources and omics-driven knowledge bases (either directly or indirectly) for identifying drug targets. We also provide a description of omics technologies and related data repositories which are important for DTD tasks.
Collapse
Affiliation(s)
- Jussi Paananen
- Institute of Biomedicine, University of Eastern Finland, Finland.,Blueprint Genetics Ltd, Finland
| | - Vittorio Fortino
- Institute of Biomedicine, University of Eastern Finland, Finland
| |
Collapse
|
139
|
Li H, Rukina D, David FPA, Li TY, Oh CM, Gao AW, Katsyuba E, Bou Sleiman M, Komljenovic A, Huang Q, Williams RW, Robinson-Rechavi M, Schoonjans K, Morgenthaler S, Auwerx J. Identifying gene function and module connections by the integration of multispecies expression compendia. Genome Res 2019; 29:2034-2045. [PMID: 31754022 PMCID: PMC6886503 DOI: 10.1101/gr.251983.119] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 10/31/2019] [Indexed: 12/13/2022]
Abstract
The functions of many eukaryotic genes are still poorly understood. Here, we developed and validated a new method, termed GeneBridge, which is based on two linked approaches to impute gene function and bridge genes with biological processes. First, Gene-Module Association Determination (G-MAD) allows the annotation of gene function. Second, Module-Module Association Determination (M-MAD) allows predicting connectivity among modules. We applied the GeneBridge tools to large-scale multispecies expression compendia—1700 data sets with over 300,000 samples from human, mouse, rat, fly, worm, and yeast—collected in this study. G-MAD identifies novel functions of genes—for example, DDT in mitochondrial respiration and WDFY4 in T cell activation—and also suggests novel components for modules, such as for cholesterol biosynthesis. By applying G-MAD on data sets from respective tissues, tissue-specific functions of genes were identified—for instance, the roles of EHHADH in liver and kidney, as well as SLC6A1 in brain and liver. Using M-MAD, we identified a list of module-module associations, such as those between mitochondria and proteasome, mitochondria and histone demethylation, as well as ribosomes and lipid biosynthesis. The GeneBridge tools together with the expression compendia are available as an open resource, which will facilitate the identification of connections linking genes, modules, phenotypes, and diseases.
Collapse
Affiliation(s)
- Hao Li
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Daria Rukina
- Institute of Mathematics, École Polytechnique Fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Fabrice P A David
- Gene Expression Core Facility, École Polytechnique Fédérale de Lausanne, Lausanne 1015, Switzerland.,SV-IT, École Polytechnique Fédérale de Lausanne, Lausanne 1015, Switzerland.,Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland
| | - Terytty Yang Li
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Chang-Myung Oh
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Arwen W Gao
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Elena Katsyuba
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Maroun Bou Sleiman
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Andrea Komljenovic
- Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland.,Department of Ecology and Evolution, University of Lausanne, Lausanne 1015, Switzerland
| | - Qingyao Huang
- Laboratory of Metabolic Signaling, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Robert W Williams
- Department of Genetics, Genomics and Informatics, University of Tennessee, Memphis, Tennessee 38163, USA
| | - Marc Robinson-Rechavi
- Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland.,Department of Ecology and Evolution, University of Lausanne, Lausanne 1015, Switzerland
| | - Kristina Schoonjans
- Laboratory of Metabolic Signaling, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Stephan Morgenthaler
- Institute of Mathematics, École Polytechnique Fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne 1015, Switzerland
| |
Collapse
|
140
|
Abstract
The common forms of metabolic diseases are highly complex, involving hundreds of genes, environmental and lifestyle factors, age-related changes, sex differences and gut-microbiome interactions. Systems genetics is a population-based approach to address this complexity. In contrast to commonly used 'reductionist' approaches, such as gain or loss of function, that examine one element at a time, systems genetics uses high-throughput 'omics' technologies to quantitatively assess the many molecular differences among individuals in a population and then to relate these to physiologic functions or disease states. Unlike genome-wide association studies, systems genetics seeks to go beyond the identification of disease-causing genes to understand higher-order interactions at the molecular level. The purpose of this review is to introduce the systems genetics applications in the areas of metabolic and cardiovascular disease. Here, we explain how large clinical and omics-level data and databases from both human and animal populations are available to help researchers place genes in the context of pathways and networks and formulate hypotheses that can then be experimentally examined. We provide lists of such databases and examples of the integration of reductionist and systems genetics data. Among the important applications emerging is the development of improved nutritional and pharmacological strategies to address the rise of metabolic diseases.
Collapse
Affiliation(s)
- Marcus Seldin
- Department of Medicine, Division of Cardiology, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Biological Chemistry and Center for Epigenetics and Metabolism, University of California, Irvine, Irvine, CA, USA
| | - Xia Yang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Aldons J Lusis
- Department of Medicine, Division of Cardiology, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
141
|
Tracking human genes along the translational continuum. NPJ Genom Med 2019; 4:25. [PMID: 31632691 PMCID: PMC6795796 DOI: 10.1038/s41525-019-0100-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 09/03/2019] [Indexed: 12/03/2022] Open
Abstract
Understanding the drivers of research on human genes is a critical component to success of translation efforts of genomics into medicine and public health. Using publicly available curated online databases we sought to identify specific genes that are featured in translational genetic research in comparison to all genomics research publications. Articles in the CDC’s Public Health Genomics and Precision Health Knowledge Base were stratified into studies that have moved beyond basic research to population and clinical epidemiologic studies (T1: clinical and population human genome epidemiology research), and studies that evaluate, implement, and assess impact of genes in clinical and public health areas (T2+: beyond bench to bedside). We examined gene counts and numbers of publications within these phases of translation in comparison to all genes from Medline. We are able to highlight those genes that are moving from basic research to clinical and public health translational research, namely in cancer and a few genetic diseases with high penetrance and clinical actionability. Identifying human genes of translational value is an important step towards determining an evidence-based trajectory of the human genome in clinical and public health practice over time.
Collapse
|
142
|
Hutchins BI, Baker KL, Davis MT, Diwersy MA, Haque E, Harriman RM, Hoppe TA, Leicht SA, Meyer P, Santangelo GM. The NIH Open Citation Collection: A public access, broad coverage resource. PLoS Biol 2019; 17:e3000385. [PMID: 31600197 PMCID: PMC6786512 DOI: 10.1371/journal.pbio.3000385] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Citation data have remained hidden behind proprietary, restrictive licensing agreements, which raises barriers to entry for analysts wishing to use the data, increases the expense of performing large-scale analyses, and reduces the robustness and reproducibility of the conclusions. For the past several years, the National Institutes of Health (NIH) Office of Portfolio Analysis (OPA) has been aggregating and enhancing citation data that can be shared publicly. Here, we describe the NIH Open Citation Collection (NIH-OCC), a public access database for biomedical research that is made freely available to the community. This dataset, which has been carefully generated from unrestricted data sources such as MedLine, PubMed Central (PMC), and CrossRef, now underlies the citation statistics delivered in the NIH iCite analytic platform. We have also included data from a machine learning pipeline that identifies, extracts, resolves, and disambiguates references from full-text articles available on the internet. Open citation links are available to the public in a major update of iCite (https://icite.od.nih.gov).
Collapse
Affiliation(s)
- B. Ian Hutchins
- Office of Portfolio Analysis, Division of Program Coordination, Planning, and Strategic Initiatives, Office of the Director, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Kirk L. Baker
- Office of Portfolio Analysis, Division of Program Coordination, Planning, and Strategic Initiatives, Office of the Director, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Matthew T. Davis
- Office of Portfolio Analysis, Division of Program Coordination, Planning, and Strategic Initiatives, Office of the Director, National Institutes of Health, Bethesda, Maryland, United States of America
| | | | - Ehsanul Haque
- Office of Portfolio Analysis, Division of Program Coordination, Planning, and Strategic Initiatives, Office of the Director, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Robert M. Harriman
- Office of Portfolio Analysis, Division of Program Coordination, Planning, and Strategic Initiatives, Office of the Director, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Travis A. Hoppe
- Office of Portfolio Analysis, Division of Program Coordination, Planning, and Strategic Initiatives, Office of the Director, National Institutes of Health, Bethesda, Maryland, United States of America
| | | | - Payam Meyer
- Office of Portfolio Analysis, Division of Program Coordination, Planning, and Strategic Initiatives, Office of the Director, National Institutes of Health, Bethesda, Maryland, United States of America
| | - George M. Santangelo
- Office of Portfolio Analysis, Division of Program Coordination, Planning, and Strategic Initiatives, Office of the Director, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
143
|
Thompson SL, Welch AC, Ho EV, Bessa JM, Portugal-Nunes C, Morais M, Young JW, Knowles JA, Dulawa SC. Btbd3 expression regulates compulsive-like and exploratory behaviors in mice. Transl Psychiatry 2019; 9:222. [PMID: 31501410 PMCID: PMC6733800 DOI: 10.1038/s41398-019-0558-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 06/20/2019] [Indexed: 12/01/2022] Open
Abstract
BTB/POZ domain-containing 3 (BTBD3) was identified as a potential risk gene in the first genome-wide association study of obsessive-compulsive disorder (OCD). BTBD3 is a putative transcription factor implicated in dendritic pruning in developing primary sensory cortices. We assessed whether BTBD3 also regulates neural circuit formation within limbic cortico-striato-thalamo-cortical circuits and behaviors related to OCD in mice. Behavioral phenotypes associated with OCD that are measurable in animals include compulsive-like behaviors and reduced exploration. We tested Btbd3 wild-type, heterozygous, and knockout mice for compulsive-like behaviors including cage-mate barbering, excessive wheel-running, repetitive locomotor patterns, and reduced goal-directed behavior in the probabilistic learning task (PLT), and for exploratory behavior in the open field, digging, and marble-burying tests. Btbd3 heterozygous and knockout mice showed excessive barbering, wheel-running, impaired goal-directed behavior in the PLT, and reduced exploration. Further, chronic treatment with fluoxetine, but not desipramine, reduced barbering in Btbd3 wild-type and heterozygous, but not knockout mice. In contrast, Btbd3 expression did not alter anxiety-like, depression-like, or sensorimotor behaviors. We also quantified dendritic morphology within anterior cingulate cortex, mediodorsal thalamus, and hippocampus, regions of high Btbd3 expression. Surprisingly, Btbd3 knockout mice only showed modest increases in spine density in the anterior cingulate, while dendritic morphology was unaltered elsewhere. Finally, we virally knocked down Btbd3 expression in whole, or just dorsal, hippocampus during neonatal development and assessed behavior during adulthood. Whole, but not dorsal, hippocampal Btbd3 knockdown recapitulated Btbd3 knockout phenotypes. Our findings reveal that hippocampal Btbd3 expression selectively modulates compulsive-like and exploratory behavior.
Collapse
Affiliation(s)
- Summer L Thompson
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
- Committee on Neurobiology, The University of Chicago, Chicago, IL, 60637, USA
| | - Amanda C Welch
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Emily V Ho
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - João M Bessa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Carlos Portugal-Nunes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Mónica Morais
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Jared W Young
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - James A Knowles
- Department of Cell Biology, SUNY Downstate Medical Center College of Medicine, Brooklyn, NY, 11203, USA
| | - Stephanie C Dulawa
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
144
|
Oprea TI. Exploring the dark genome: implications for precision medicine. Mamm Genome 2019; 30:192-200. [PMID: 31270560 DOI: 10.1007/s00335-019-09809-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 06/15/2019] [Indexed: 01/08/2023]
Abstract
The increase in the number of both patients and healthcare practitioners who grew up using the Internet and computers (so-called "digital natives") is likely to impact the practice of precision medicine, and requires novel platforms for data integration and mining, as well as contextualized information retrieval. The "Illuminating the Druggable Genome Knowledge Management Center" (IDG KMC) quantifies data availability from a wide range of chemical, biological, and clinical resources, and has developed platforms that can be used to navigate understudied proteins (the "dark genome"), and their potential contribution to specific pathologies. Using the "Target Importance and Novelty Explorer" (TIN-X) highlights the role of LRRC10 (a dark gene) in dilated cardiomyopathy. Combining mouse and human phenotype data leads to increased strength of evidence, which is discussed for four additional dark genes: SLX4IP and its role in glucose metabolism, the role of HSF2BP in coronary artery disease, the involvement of ELFN1 in attention-deficit hyperactivity disorder and the role of VPS13D in mouse neural tube development and its confirmed role in childhood onset movement disorders. The workflow and tools described here are aimed at guiding further experimental research, particularly within the context of precision medicine.
Collapse
Affiliation(s)
- Tudor I Oprea
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM, USA. .,UNM Comprehensive Cancer Center, Albuquerque, NM, USA. .,Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden. .,Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
145
|
Carter AJ, Kraemer O, Zwick M, Mueller-Fahrnow A, Arrowsmith CH, Edwards AM. Target 2035: probing the human proteome. Drug Discov Today 2019; 24:2111-2115. [PMID: 31278990 DOI: 10.1016/j.drudis.2019.06.020] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 06/11/2019] [Accepted: 06/27/2019] [Indexed: 11/29/2022]
Abstract
Biomedical scientists tend to focus on only a small fraction of the proteins encoded by the human genome despite overwhelming genetic evidence that many understudied proteins are important for human disease. One of the best ways to interrogate the function of a protein and to determine its relevance as a drug target is by using a pharmacological modulator, such as a chemical probe or an antibody. If these tools were available for most human proteins, it should be possible to translate the tremendous advances in genomics into a greater understanding of human health and disease, and catalyze the creation of innovative new medicines. Target 2035 is a global federation for developing and applying new technologies with the goal of creating chemogenomic libraries, chemical probes, and/or functional antibodies for the entire proteome.
Collapse
Affiliation(s)
- Adrian J Carter
- Discovery Research Coordination, Boehringer Ingelheim, 55216 Ingelheim am Rhein, Germany.
| | - Oliver Kraemer
- Discovery Research Coordination, Boehringer Ingelheim, 55216 Ingelheim am Rhein, Germany
| | - Matthias Zwick
- Computational Biology, Boehringer Ingelheim, 88400 Biberach an der Riß, Germany
| | | | - Cheryl H Arrowsmith
- Structural Genomics Consortium, University of Toronto, Toronto, Ontario, M5G 1L7, Canada; Princess Margaret Cancer Centre, Toronto, Ontario, M5G 1L7, Canada
| | - Aled M Edwards
- Structural Genomics Consortium, University of Toronto, Toronto, Ontario, M5G 1L7, Canada
| |
Collapse
|
146
|
Yang C, Ambayo H, Baets BD, Kolsteren P, Thanintorn N, Hawwash D, Bouwman J, Bronselaer A, Pattyn F, Lachat C. An Ontology to Standardize Research Output of Nutritional Epidemiology: From Paper-Based Standards to Linked Content. Nutrients 2019; 11:E1300. [PMID: 31181762 PMCID: PMC6628051 DOI: 10.3390/nu11061300] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/03/2019] [Accepted: 06/06/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The use of linked data in the Semantic Web is a promising approach to add value to nutrition research. An ontology, which defines the logical relationships between well-defined taxonomic terms, enables linking and harmonizing research output. To enable the description of domain-specific output in nutritional epidemiology, we propose the Ontology for Nutritional Epidemiology (ONE) according to authoritative guidance for nutritional epidemiology. METHODS Firstly, a scoping review was conducted to identify existing ontology terms for reuse in ONE. Secondly, existing data standards and reporting guidelines for nutritional epidemiology were converted into an ontology. The terms used in the standards were summarized and listed separately in a taxonomic hierarchy. Thirdly, the ontologies of the nutritional epidemiologic standards, reporting guidelines, and the core concepts were gathered in ONE. Three case studies were included to illustrate potential applications: (i) annotation of existing manuscripts and data, (ii) ontology-based inference, and (iii) estimation of reporting completeness in a sample of nine manuscripts. RESULTS Ontologies for "food and nutrition" (n = 37), "disease and specific population" (n = 100), "data description" (n = 21), "research description" (n = 35), and "supplementary (meta) data description" (n = 44) were reviewed and listed. ONE consists of 339 classes: 79 new classes to describe data and 24 new classes to describe the content of manuscripts. CONCLUSION ONE is a resource to automate data integration, searching, and browsing, and can be used to assess reporting completeness in nutritional epidemiology.
Collapse
Affiliation(s)
- Chen Yang
- Department of Food Technology, Safety and Health, Ghent University, 9000 Ghent, Belgium.
| | - Henry Ambayo
- Department of Food Technology, Safety and Health, Ghent University, 9000 Ghent, Belgium.
| | - Bernard De Baets
- KERMIT, Department of Data Analysis and Mathematical Modelling, Ghent University, 9000 Ghent, Belgium.
| | - Patrick Kolsteren
- Department of Food Technology, Safety and Health, Ghent University, 9000 Ghent, Belgium.
| | - Nattapon Thanintorn
- Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, MO 65203, USA.
| | - Dana Hawwash
- Department of Food Technology, Safety and Health, Ghent University, 9000 Ghent, Belgium.
| | - Jildau Bouwman
- Netherlands Organization for Applied Scientific Research, NL-2509 Zeist, The Netherlands.
| | - Antoon Bronselaer
- Department of Telecommunications and information processing, Ghent University, 9000 Ghent, Belgium.
| | | | - Carl Lachat
- Department of Food Technology, Safety and Health, Ghent University, 9000 Ghent, Belgium.
| |
Collapse
|
147
|
Access to Our Journals. J Oral Maxillofac Surg 2019; 77:1965-1966. [PMID: 31158345 DOI: 10.1016/j.joms.2019.04.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 04/23/2019] [Indexed: 11/21/2022]
|
148
|
Brommage R, Powell DR, Vogel P. Predicting human disease mutations and identifying drug targets from mouse gene knockout phenotyping campaigns. Dis Model Mech 2019; 12:dmm038224. [PMID: 31064765 PMCID: PMC6550044 DOI: 10.1242/dmm.038224] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Two large-scale mouse gene knockout phenotyping campaigns have provided extensive data on the functions of thousands of mammalian genes. The ongoing International Mouse Phenotyping Consortium (IMPC), with the goal of examining all ∼20,000 mouse genes, has examined 5115 genes since 2011, and phenotypic data from several analyses are available on the IMPC website (www.mousephenotype.org). Mutant mice having at least one human genetic disease-associated phenotype are available for 185 IMPC genes. Lexicon Pharmaceuticals' Genome5000™ campaign performed similar analyses between 2000 and the end of 2008 focusing on the druggable genome, including enzymes, receptors, transporters, channels and secreted proteins. Mutants (4654 genes, with 3762 viable adult homozygous lines) with therapeutically interesting phenotypes were studied extensively. Importantly, phenotypes for 29 Lexicon mouse gene knockouts were published prior to observations of similar phenotypes resulting from homologous mutations in human genetic disorders. Knockout mouse phenotypes for an additional 30 genes mimicked previously published human genetic disorders. Several of these models have helped develop effective treatments for human diseases. For example, studying Tph1 knockout mice (lacking peripheral serotonin) aided the development of telotristat ethyl, an approved treatment for carcinoid syndrome. Sglt1 (also known as Slc5a1) and Sglt2 (also known as Slc5a2) knockout mice were employed to develop sotagliflozin, a dual SGLT1/SGLT2 inhibitor having success in clinical trials for diabetes. Clinical trials evaluating inhibitors of AAK1 (neuropathic pain) and SGLT1 (diabetes) are underway. The research community can take advantage of these unbiased analyses of gene function in mice, including the minimally studied 'ignorome' genes.
Collapse
Affiliation(s)
- Robert Brommage
- Department of Metabolism Research, Lexicon Pharmaceuticals, 8800 Technology Forest Place, The Woodlands, TX 77381, USA
| | - David R Powell
- Department of Metabolism Research, Lexicon Pharmaceuticals, 8800 Technology Forest Place, The Woodlands, TX 77381, USA
| | - Peter Vogel
- St. Jude Children's Research Hospital, Pathology, MS 250, Room C5036A, 262 Danny Thomas Place, Memphis, TN 38105, USA
| |
Collapse
|
149
|
Drewry DH, Wells CI, Zuercher WJ, Willson TM. A Perspective on Extreme Open Science: Companies Sharing Compounds without Restriction. SLAS DISCOVERY 2019; 24:505-514. [PMID: 31034310 DOI: 10.1177/2472555219838210] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Although the human genome provides the blueprint for life, most of the proteins it encodes remain poorly studied. This perspective describes how one group of scientists, in seeking new targets for drug discovery, used open science through unrestricted sharing of small molecules to shed light on dark matter of the genome. Starting initially with a single pharmaceutical company before expanding to multiple companies, a precedent was established for sharing published kinase inhibitors as chemical tools. The integration of open science and kinase chemogenomics has supported the study of many new potential drug targets by the scientific community.
Collapse
Affiliation(s)
- David H Drewry
- 1 Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Carrow I Wells
- 1 Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - William J Zuercher
- 1 Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Timothy M Willson
- 1 Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
150
|
Thyme SB, Pieper LM, Li EH, Pandey S, Wang Y, Morris NS, Sha C, Choi JW, Herrera KJ, Soucy ER, Zimmerman S, Randlett O, Greenwood J, McCarroll SA, Schier AF. Phenotypic Landscape of Schizophrenia-Associated Genes Defines Candidates and Their Shared Functions. Cell 2019; 177:478-491.e20. [PMID: 30929901 PMCID: PMC6494450 DOI: 10.1016/j.cell.2019.01.048] [Citation(s) in RCA: 138] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 10/15/2018] [Accepted: 01/27/2019] [Indexed: 01/25/2023]
Abstract
Genomic studies have identified hundreds of candidate genes near loci associated with risk for schizophrenia. To define candidates and their functions, we mutated zebrafish orthologs of 132 human schizophrenia-associated genes. We created a phenotype atlas consisting of whole-brain activity maps, brain structural differences, and profiles of behavioral abnormalities. Phenotypes were diverse but specific, including altered forebrain development and decreased prepulse inhibition. Exploration of these datasets identified promising candidates in more than 10 gene-rich regions, including the magnesium transporter cnnm2 and the translational repressor gigyf2, and revealed shared anatomical sites of activity differences, including the pallium, hypothalamus, and tectum. Single-cell RNA sequencing uncovered an essential role for the understudied transcription factor znf536 in the development of forebrain neurons implicated in social behavior and stress. This phenotypic landscape of schizophrenia-associated genes prioritizes more than 30 candidates for further study and provides hypotheses to bridge the divide between genetic association and biological mechanism.
Collapse
Affiliation(s)
- Summer B Thyme
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA.
| | - Lindsey M Pieper
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Eric H Li
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Shristi Pandey
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Yiqun Wang
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Nathan S Morris
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Carrie Sha
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Joo Won Choi
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Kristian J Herrera
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Edward R Soucy
- Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Steve Zimmerman
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Owen Randlett
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Joel Greenwood
- Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Steven A McCarroll
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Cambridge, MA 02142, USA
| | - Alexander F Schier
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA; Center for Brain Science, Harvard University, Cambridge, MA 02138, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Biozentrum, University of Basel, CH-4056 Basel, Switzerland; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; FAS Center for Systems Biology, Harvard University, MA 02138, USA; Allen Discovery Center for Cell Lineage Tracing, Seattle, WA 98104, USA.
| |
Collapse
|