1
|
Samy A, Hassan A, Hegazi NM, Farid M, Elshafei M. Network pharmacology, molecular docking, and dynamics analyses to predict the antiviral activity of ginger constituents against coronavirus infection. Sci Rep 2024; 14:12059. [PMID: 38802394 PMCID: PMC11130167 DOI: 10.1038/s41598-024-60721-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 04/26/2024] [Indexed: 05/29/2024] Open
Abstract
COVID-19 is a global pandemic that caused a dramatic loss of human life worldwide, leading to accelerated research for antiviral drug discovery. Herbal medicine is one of the most commonly used alternative medicine for the prevention and treatment of many conditions including respiratory system diseases. In this study, a computational pipeline was employed, including network pharmacology, molecular docking simulations, and molecular dynamics simulations, to analyze the common phytochemicals of ginger rhizomes and identify candidate constituents as viral inhibitors. Furthermore, experimental assays were performed to analyze the volatile and non-volatile compounds of ginger and to assess the antiviral activity of ginger oil and hydroalcoholic extract. Network pharmacology analysis showed that ginger compounds target human genes that are involved in related cellular processes to the viral infection. Docking analysis highlighted five pungent compounds and zingiberenol as potential inhibitors for the main protease (Mpro), spike receptor-binding domain (RBD), and human angiotensin-converting enzyme 2 (ACE2). Then, (6)-gingerdiacetate was selected for molecular dynamics (MD) simulations as it exhibited the best binding interactions and free energies over the three target proteins. Trajectories analysis of the three complexes showed that RBD and ACE2 complexes with the ligand preserved similar patterns of root mean square deviation (RMSD) and radius of gyration (Rg) values to their respective native structures. Finally, experimental validation of the ginger hydroalcoholic extract confirmed the existence of (6)-gingerdiacetate and revealed the strong antiviral activity of the hydroalcoholic extract with IC50 of 2.727 μ g / ml . Our study provides insights into the potential antiviral activity of (6)-gingerdiacetate that may enhance the host immune response and block RBD binding to ACE2, thereby, inhibiting SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Asmaa Samy
- Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Afnan Hassan
- Biomedical Sciences Program, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Nesrine M Hegazi
- Department of Phytochemistry and Plant Systematics, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Cairo, 12622, Egypt
| | - Mai Farid
- Department of Phytochemistry and Plant Systematics, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Cairo, 12622, Egypt
| | | |
Collapse
|
2
|
Gujjala VA, Klimek I, Abyadeh M, Tyshkovskiy A, Oz N, Castro JP, Gladyshev VN, Newton J, Kaya A. A disease similarity approach identifies short-lived Niemann-Pick type C disease mice with accelerated brain aging as a novel mouse model for Alzheimer's disease and aging research. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.19.590328. [PMID: 38712089 PMCID: PMC11071364 DOI: 10.1101/2024.04.19.590328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Since its first description in 1906 by Dr. Alois Alzheimer, Alzheimer's disease (AD) has been the most common type of dementia. Initially thought to be caused by age-associated accumulation of plaques, in recent years, research has increasingly associated AD with lysosomal storage and metabolic disorders, and the explanation of its pathogenesis has shifted from amyloid and tau accumulation to oxidative stress and impaired lipid and glucose metabolism aggravated by hypoxic conditions. However, the underlying mechanisms linking those cellular processes and conditions to disease progression have yet to be defined. Here, we applied a disease similarity approach to identify unknown molecular targets of AD by using transcriptomic data from congenital diseases known to increase AD risk, namely Down Syndrome, Niemann Pick Disease Type C (NPC), and Mucopolysaccharidoses I. We uncovered common pathways, hub genes, and miRNAs across in vitro and in vivo models of these diseases as potential molecular targets for neuroprotection and amelioration of AD pathology, many of which have never been associated with AD. We then investigated common molecular alterations in brain samples from an NPC disease mouse model by juxtaposing them with brain samples of both human and mouse models of AD. Detailed phenotypic and molecular analyses revealed that the NPC mut mouse model can serve as a potential short-lived in vivo model for AD research and for understanding molecular factors affecting brain aging. This research represents the first comprehensive approach to congenital disease association with neurodegeneration and a new perspective on AD research while highlighting shortcomings and lack of correlation in diverse in vitro models. Considering the lack of an AD mouse model that recapitulates the physiological hallmarks of brain aging, the characterization of a short-lived NPC mouse model will further accelerate the research in these fields and offer a unique model for understanding the molecular mechanisms of AD from a perspective of accelerated brain aging.
Collapse
|
3
|
Olmos V, Thompson EN, Gogia N, Luttik K, Veeranki V, Ni L, Sim S, Chen K, Krause DS, Lim J. Dysregulation of alternative splicing in spinocerebellar ataxia type 1. Hum Mol Genet 2024; 33:138-149. [PMID: 37802886 PMCID: PMC10979408 DOI: 10.1093/hmg/ddad170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/29/2023] [Accepted: 10/02/2023] [Indexed: 10/08/2023] Open
Abstract
Spinocerebellar ataxia type 1 is caused by an expansion of the polyglutamine tract in ATAXIN-1. Ataxin-1 is broadly expressed throughout the brain and is involved in regulating gene expression. However, it is not yet known if mutant ataxin-1 can impact the regulation of alternative splicing events. We performed RNA sequencing in mouse models of spinocerebellar ataxia type 1 and identified that mutant ataxin-1 expression abnormally leads to diverse splicing events in the mouse cerebellum of spinocerebellar ataxia type 1. We found that the diverse splicing events occurred in a predominantly cell autonomous manner. A majority of the transcripts with misregulated alternative splicing events were previously unknown, thus allowing us to identify overall new biological pathways that are distinctive to those affected by differential gene expression in spinocerebellar ataxia type 1. We also provide evidence that the splicing factor Rbfox1 mediates the effect of mutant ataxin-1 on misregulated alternative splicing and that genetic manipulation of Rbfox1 expression modifies neurodegenerative phenotypes in a Drosophila model of spinocerebellar ataxia type 1 in vivo. Together, this study provides novel molecular mechanistic insight into the pathogenesis of spinocerebellar ataxia type 1 and identifies potential therapeutic strategies for spinocerebellar ataxia type 1.
Collapse
Affiliation(s)
- Victor Olmos
- Department of Genetics, Yale School of Medicine, 295 Congress Avenue, New Haven, CT 06510, United States
| | - Evrett N Thompson
- Department of Cell Biology, Yale School of Medicine, 10 Amistad Street, New Haven, CT 06510, United States
- Yale Stem Cell Center, Yale School of Medicine, 10 Amistad Street, New Haven, CT 06510, United States
| | - Neha Gogia
- Department of Genetics, Yale School of Medicine, 295 Congress Avenue, New Haven, CT 06510, United States
| | - Kimberly Luttik
- Interdepartmental Neuroscience Program, Yale School of Medicine, 295 Congress Avenue, New Haven, CT 06510, United States
- Department of Neuroscience, Yale School of Medicine, 295 Congress Avenue, New Haven, CT 06510, USA
| | - Vaishnavi Veeranki
- Department of Genetics, Yale School of Medicine, 295 Congress Avenue, New Haven, CT 06510, United States
| | - Luhan Ni
- Department of Genetics, Yale School of Medicine, 295 Congress Avenue, New Haven, CT 06510, United States
| | - Serena Sim
- Yale College, 433 Temple Street, New Haven, CT 06510, United States
| | - Kelly Chen
- Yale College, 433 Temple Street, New Haven, CT 06510, United States
| | - Diane S Krause
- Department of Cell Biology, Yale School of Medicine, 10 Amistad Street, New Haven, CT 06510, United States
- Yale Stem Cell Center, Yale School of Medicine, 10 Amistad Street, New Haven, CT 06510, United States
- Department of Pathology, Yale School of Medicine, 10 Amistad Street, New Haven, CT 06510, United States
- Department of Laboratory Medicine, Yale School of Medicine, 10 Amistad Street, New Haven, CT 06510, United States
| | - Janghoo Lim
- Department of Genetics, Yale School of Medicine, 295 Congress Avenue, New Haven, CT 06510, United States
- Yale Stem Cell Center, Yale School of Medicine, 10 Amistad Street, New Haven, CT 06510, United States
- Interdepartmental Neuroscience Program, Yale School of Medicine, 295 Congress Avenue, New Haven, CT 06510, United States
- Department of Neuroscience, Yale School of Medicine, 295 Congress Avenue, New Haven, CT 06510, USA
- Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale School of Medicine, 295 Congress Avenue, New Haven, CT 06510, United States
- Wu Tsai Institute, Yale School of Medicine, 100 College, New Haven, CT 06510, United States
| |
Collapse
|
4
|
Sinha R, Pal RK, De RK. ENLIGHTENMENT: A Scalable Annotated Database of Genomics and NGS-Based Nucleotide Level Profiles. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2024; 21:155-168. [PMID: 38055361 DOI: 10.1109/tcbb.2023.3340067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/08/2023]
Abstract
The revolution in sequencing technologies has enabled human genomes to be sequenced at a very low cost and time leading to exponential growth in the availability of whole-genome sequences. However, the complete understanding of our genome and its association with cancer is a far way to go. Researchers are striving hard to detect new variants and find their association with diseases, which further gives rise to the need for aggregation of this Big Data into a common standard scalable platform. In this work, a database named Enlightenment has been implemented which makes the availability of genomic data integrated from eight public databases, and DNA sequencing profiles of H. sapiens in a single platform. Annotated results with respect to cancer specific biomarkers, pharmacogenetic biomarkers and its association with variability in drug response, and DNA profiles along with novel copy number variants are computed and stored, which are accessible through a web interface. In order to overcome the challenge of storage and processing of NGS technology-based whole-genome DNA sequences, Enlightenment has been extended and deployed to a flexible and horizontally scalable database HBase, which is distributed over a hadoop cluster, which would enable the integration of other omics data into the database for enlightening the path towards eradication of cancer.
Collapse
|
5
|
Chabosseau P, Yong F, Delgadillo-Silva LF, Lee EY, Melhem R, Li S, Gandhi N, Wastin J, Noriega LL, Leclerc I, Ali Y, Hughes JW, Sladek R, Martinez-Sanchez A, Rutter GA. Molecular phenotyping of single pancreatic islet leader beta cells by "Flash-Seq". Life Sci 2023; 316:121436. [PMID: 36706832 DOI: 10.1016/j.lfs.2023.121436] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 01/11/2023] [Accepted: 01/20/2023] [Indexed: 01/26/2023]
Abstract
AIMS Spatially-organized increases in cytosolic Ca2+ within pancreatic beta cells in the pancreatic islet underlie the stimulation of insulin secretion by high glucose. Recent data have revealed the existence of subpopulations of beta cells including "leaders" which initiate Ca2+ waves. Whether leader cells possess unique molecular features, or localisation, is unknown. MAIN METHODS High speed confocal Ca2+ imaging was used to identify leader cells and connectivity analysis, running under MATLAB and Python, to identify highly connected "hub" cells. To explore transcriptomic differences between beta cell sub-groups, individual leaders or followers were labelled by photo-activation of the cryptic fluorescent protein PA-mCherry and subjected to single cell RNA sequencing ("Flash-Seq"). KEY FINDINGS Distinct Ca2+ wave types were identified in individual islets, with leader cells present in 73 % (28 of 38 islets imaged). Scale-free, power law-adherent behaviour was also observed in 29 % of islets, though "hub" cells in these islets did not overlap with leaders. Transcripts differentially expressed (295; padj < 0.05) between leader and follower cells included genes involved in cilium biogenesis and transcriptional regulation. Providing some support for these findings, ADCY6 immunoreactivity tended to be higher in leader than follower cells, whereas cilia number and length tended to be lower in the former. Finally, leader cells were located significantly closer to delta, but not alpha, cells in Euclidian space than were follower cells. SIGNIFICANCE The existence of both a discrete transcriptome and unique localisation implies a role for these features in defining the specialized function of leaders. These data also raise the possibility that localised signalling between delta and leader cells contributes to the initiation and propagation of islet Ca2+ waves.
Collapse
Affiliation(s)
- Pauline Chabosseau
- Centre de Recherche du CHUM, Faculté de Médicine, Université de Montréal, Montréal, QC, Canada
| | - Fiona Yong
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London W12 0NN, United Kingdom; Lee Kong Chian Imperial Medical School, Nanyang Technological University, Singapore
| | - Luis F Delgadillo-Silva
- Centre de Recherche du CHUM, Faculté de Médicine, Université de Montréal, Montréal, QC, Canada
| | - Eun Young Lee
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, United States; Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | - Rana Melhem
- Centre de Recherche du CHUM, Faculté de Médicine, Université de Montréal, Montréal, QC, Canada
| | - Shiying Li
- Centre de Recherche du CHUM, Faculté de Médicine, Université de Montréal, Montréal, QC, Canada
| | - Nidhi Gandhi
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London W12 0NN, United Kingdom
| | - Jules Wastin
- Centre de Recherche du CHUM, Faculté de Médicine, Université de Montréal, Montréal, QC, Canada
| | - Livia Lopez Noriega
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London W12 0NN, United Kingdom
| | - Isabelle Leclerc
- Centre de Recherche du CHUM, Faculté de Médicine, Université de Montréal, Montréal, QC, Canada
| | - Yusuf Ali
- Lee Kong Chian Imperial Medical School, Nanyang Technological University, Singapore
| | - Jing W Hughes
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, United States
| | - Robert Sladek
- Departments of Medicine and Human Genetics, McGill University and Genome Quebec Innovation Centre, Montreal, QC, Canada
| | - Aida Martinez-Sanchez
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London W12 0NN, United Kingdom
| | - Guy A Rutter
- Centre de Recherche du CHUM, Faculté de Médicine, Université de Montréal, Montréal, QC, Canada; Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London W12 0NN, United Kingdom; Lee Kong Chian Imperial Medical School, Nanyang Technological University, Singapore.
| |
Collapse
|
6
|
Yarani R, Palasca O, Doncheva NT, Anthon C, Pilecki B, Svane CAS, Mirza AH, Litman T, Holmskov U, Bang-Berthelsen CH, Vilien M, Jensen LJ, Gorodkin J, Pociot F. Cross-species high-resolution transcriptome profiling suggests biomarkers and therapeutic targets for ulcerative colitis. Front Mol Biosci 2023; 9:1081176. [PMID: 36685283 PMCID: PMC9850088 DOI: 10.3389/fmolb.2022.1081176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/08/2022] [Indexed: 01/07/2023] Open
Abstract
Background: Ulcerative colitis (UC) is a disorder with unknown etiology, and animal models play an essential role in studying its molecular pathophysiology. Here, we aim to identify common conserved pathological UC-related gene expression signatures between humans and mice that can be used as treatment targets and/or biomarker candidates. Methods: To identify differentially regulated protein-coding genes and non-coding RNAs, we sequenced total RNA from the colon and blood of the most widely used dextran sodium sulfate Ulcerative colitis mouse. By combining this with public human Ulcerative colitis data, we investigated conserved gene expression signatures and pathways/biological processes through which these genes may contribute to disease development/progression. Results: Cross-species integration of human and mouse Ulcerative colitis data resulted in the identification of 1442 genes that were significantly differentially regulated in the same direction in the colon and 157 in blood. Of these, 51 genes showed consistent differential regulation in the colon and blood. Less known genes with importance in disease pathogenesis, including SPI1, FPR2, TYROBP, CKAP4, MCEMP1, ADGRG3, SLC11A1, and SELPLG, were identified through network centrality ranking and validated in independent human and mouse cohorts. Conclusion: The identified Ulcerative colitis conserved transcriptional signatures aid in the disease phenotyping and future treatment decisions, drug discovery, and clinical trial design.
Collapse
Affiliation(s)
- Reza Yarani
- Translational Type 1 Diabetes Research, Department of Clinical Research, Steno Diabetes Center Copenhagen, Gentofte, Denmark,*Correspondence: Reza Yarani, ; Flemming Pociot,
| | - Oana Palasca
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark,Center for non-coding RNA in Technology and Health, University of Copenhagen, Copenhagen, Denmark,Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nadezhda T. Doncheva
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark,Center for non-coding RNA in Technology and Health, University of Copenhagen, Copenhagen, Denmark,Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christian Anthon
- Center for non-coding RNA in Technology and Health, University of Copenhagen, Copenhagen, Denmark,Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bartosz Pilecki
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Cecilie A. S. Svane
- Translational Type 1 Diabetes Research, Department of Clinical Research, Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Aashiq H. Mirza
- Center for non-coding RNA in Technology and Health, University of Copenhagen, Copenhagen, Denmark,Department of Pharmacology, Weill Cornell Medicine, Cornell University, New York, NY, United States
| | - Thomas Litman
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Uffe Holmskov
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Claus H. Bang-Berthelsen
- Research Group for Microbial Biotechnology and Biorefining, National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark,Department of Gastroenterology, North Zealand Hillerød Hospital, Hillerød, Denmark
| | - Mogens Vilien
- Department of Surgery, North Zealand Hospital, Hillerød, Denmark
| | - Lars J. Jensen
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark,Center for non-coding RNA in Technology and Health, University of Copenhagen, Copenhagen, Denmark
| | - Jan Gorodkin
- Center for non-coding RNA in Technology and Health, University of Copenhagen, Copenhagen, Denmark,Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Flemming Pociot
- Translational Type 1 Diabetes Research, Department of Clinical Research, Steno Diabetes Center Copenhagen, Gentofte, Denmark,Center for non-coding RNA in Technology and Health, University of Copenhagen, Copenhagen, Denmark,Copenhagen Diabetes Research Center, Department of Pediatrics, Herlev University Hospital, Herlev, Denmark,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark,*Correspondence: Reza Yarani, ; Flemming Pociot,
| |
Collapse
|
7
|
Halim-Fikri H, Syed-Hassan SNRK, Wan-Juhari WK, Assyuhada MGSN, Hernaningsih Y, Yusoff NM, Merican AF, Zilfalil BA. Central resources of variant discovery and annotation and its role in precision medicine. ASIAN BIOMED 2022; 16:285-298. [PMID: 37551357 PMCID: PMC10392146 DOI: 10.2478/abm-2022-0032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2023]
Abstract
Rapid technological advancement in high-throughput genomics, microarray, and deep sequencing technologies has accelerated the possibility of more complex precision medicine research using large amounts of heterogeneous health-related data from patients, including genomic variants. Genomic variants can be identified and annotated based on the reference human genome either within the sequence as a whole or in a putative functional genomic element. The American College of Medical Genetics and Genomics (ACMG) and the Association for Molecular Pathology (AMP) mutually created standards and guidelines for the appraisal of proof to expand consistency and straightforwardness in clinical variation interpretations. Various efforts toward precision medicine have been facilitated by many national and international public databases that classify and annotate genomic variation. In the present study, several resources are highlighted with recognition and data spreading of clinically important genetic variations.
Collapse
Affiliation(s)
- Hashim Halim-Fikri
- Malaysian Node of the Human Variome Project, School of Medical Sciences, Universiti Sains Malaysia, Kelantan16150, Malaysia
| | | | - Wan-Khairunnisa Wan-Juhari
- Malaysian Node of the Human Variome Project, School of Medical Sciences, Universiti Sains Malaysia, Kelantan16150, Malaysia
- Human Genome Centre, School of Medical Sciences, Universiti Sains Malaysia, Kelantan16150, Malaysia
| | - Mat Ghani Siti Nor Assyuhada
- Malaysian Node of the Human Variome Project, School of Medical Sciences, Universiti Sains Malaysia, Kelantan16150, Malaysia
| | - Yetti Hernaningsih
- Department of Clinical Pathology, Faculty of Medicine Universitas Airlangga, Dr. Soetomo Academic General Hospital, Surabaya, Indonesia
| | - Narazah Mohd Yusoff
- Department of Clinical Pathology, Faculty of Medicine Universitas Airlangga, Dr. Soetomo Academic General Hospital, Surabaya, Indonesia
- Clinical Diagnostic Laboratory, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang13200, Malaysia
| | - Amir Feisal Merican
- Institute of Biological Sciences, Faculty of Science, University of Malaya, Kuala Lumpur50603, Malaysia
- Center of Research for Computational Sciences and Informatics in Biology, Bio Industry, Environment, Agriculture and Healthcare (CRYSTAL), University of Malaya, Kuala Lumpur50603, Malaysia
| | - Bin Alwi Zilfalil
- Malaysian Node of the Human Variome Project, School of Medical Sciences, Universiti Sains Malaysia, Kelantan16150, Malaysia
- Human Genome Centre, School of Medical Sciences, Universiti Sains Malaysia, Kelantan16150, Malaysia
| |
Collapse
|
8
|
Large-scale prediction of key dynamic interacting proteins in multiple cancers. Int J Biol Macromol 2022; 220:1124-1132. [PMID: 36027989 DOI: 10.1016/j.ijbiomac.2022.08.125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 08/15/2022] [Accepted: 08/17/2022] [Indexed: 11/21/2022]
Abstract
Tracking cancer dynamic protein-protein interactions (PPIs) and deciphering their pathogenesis remain a challenge. We presented a dynamic PPIs' hypothesis: permanent and transient interactions might achieve dynamic switchings from normal cells to malignancy, which could cause maintenance functions to be interrupted and transient functions to be sustained. Based on the hypothesis, we first predicted >1400 key cancer genes (KCG) by applying PPI-express we proposed to 18 cancer gene expression datasets. We then further screened out key dynamic interactions (KDI) of cancer based on KCG and transient and permanent interactions under both conditions. Two prominent functional characteristics, "Cell cycle-related" and "Immune-related", were presented for KCG, suggesting that these might be their general characteristics. We found that, compared to permanent to transient KDI pairs (P2T) in the network, transient to permanent (T2P) have significantly higher edge betweenness (EB), and P2T pairs tending to locate intra-functional modules may play roles in maintaining normal biological functions, while T2P KDI pairs tending to locate inter-modules may play roles in biological signal transduction. It was consistent with our hypothesis. Also, we analyzed network characteristics of KDI pairs and their functions. Our findings of KDI may serve to understand and explain a few hallmarks of cancer.
Collapse
|
9
|
Kringel D, Malkusch S, Lötsch J. Drugs and Epigenetic Molecular Functions. A Pharmacological Data Scientometric Analysis. Int J Mol Sci 2021; 22:7250. [PMID: 34298869 PMCID: PMC8311652 DOI: 10.3390/ijms22147250] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/25/2021] [Accepted: 06/30/2021] [Indexed: 12/14/2022] Open
Abstract
Interactions of drugs with the classical epigenetic mechanism of DNA methylation or histone modification are increasingly being elucidated mechanistically and used to develop novel classes of epigenetic therapeutics. A data science approach is used to synthesize current knowledge on the pharmacological implications of epigenetic regulation of gene expression. Computer-aided knowledge discovery for epigenetic implications of current approved or investigational drugs was performed by querying information from multiple publicly available gold-standard sources to (i) identify enzymes involved in classical epigenetic processes, (ii) screen original biomedical scientific publications including bibliometric analyses, (iii) identify drugs that interact with epigenetic enzymes, including their additional non-epigenetic targets, and (iv) analyze computational functional genomics of drugs with epigenetic interactions. PubMed database search yielded 3051 hits on epigenetics and drugs, starting in 1992 and peaking in 2016. Annual citations increased to a plateau in 2000 and show a downward trend since 2008. Approved and investigational drugs in the DrugBank database included 122 compounds that interacted with 68 unique epigenetic enzymes. Additional molecular functions modulated by these drugs included other enzyme interactions, whereas modulation of ion channels or G-protein-coupled receptors were underrepresented. Epigenetic interactions included (i) drug-induced modulation of DNA methylation, (ii) drug-induced modulation of histone conformations, and (iii) epigenetic modulation of drug effects by interference with pharmacokinetics or pharmacodynamics. Interactions of epigenetic molecular functions and drugs are mutual. Recent research activities on the discovery and development of novel epigenetic therapeutics have passed successfully, whereas epigenetic effects of non-epigenetic drugs or epigenetically induced changes in the targets of common drugs have not yet received the necessary systematic attention in the context of pharmacological plasticity.
Collapse
Affiliation(s)
- Dario Kringel
- Institute of Clinical Pharmacology, Goethe-University, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (D.K.); (S.M.)
| | - Sebastian Malkusch
- Institute of Clinical Pharmacology, Goethe-University, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (D.K.); (S.M.)
| | - Jörn Lötsch
- Institute of Clinical Pharmacology, Goethe-University, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (D.K.); (S.M.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| |
Collapse
|
10
|
Wiedemann GM, Santosa EK, Grassmann S, Sheppard S, Le Luduec JB, Adams NM, Dang C, Hsu KC, Sun JC, Lau CM. Deconvoluting global cytokine signaling networks in natural killer cells. Nat Immunol 2021; 22:627-638. [PMID: 33859404 PMCID: PMC8476180 DOI: 10.1038/s41590-021-00909-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 02/24/2021] [Indexed: 01/31/2023]
Abstract
Cytokine signaling via signal transducer and activator of transcription (STAT) proteins is crucial for optimal antiviral responses of natural killer (NK) cells. However, the pleiotropic effects of both cytokine and STAT signaling preclude the ability to precisely attribute molecular changes to specific cytokine-STAT modules. Here, we employed a multi-omics approach to deconstruct and rebuild the complex interaction of multiple cytokine signaling pathways in NK cells. Proinflammatory cytokines and homeostatic cytokines formed a cooperative axis to commonly regulate global gene expression and to further repress expression induced by type I interferon signaling. These cytokines mediated distinct modes of epigenetic regulation via STAT proteins, and collective signaling best recapitulated global antiviral responses. The most dynamically responsive genes were conserved across humans and mice, which included a cytokine-STAT-induced cross-regulatory program. Thus, an intricate crosstalk exists between cytokine signaling pathways, which governs NK cell responses.
Collapse
Affiliation(s)
- Gabriela M. Wiedemann
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA,Department of Internal Medicine II, Technical University of Munich, Munich, Germany
| | - Endi K. Santosa
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Simon Grassmann
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sam Sheppard
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Nicholas M. Adams
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Celeste Dang
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Katharine C. Hsu
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA,Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Joseph C. Sun
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA,Department of Immunology and Microbial Pathogenesis, Weill Cornell Medical College, New York, NY, USA,Correspondence and requests for materials should be addressed to J.C.S. or C.M.L. ;
| | - Colleen M. Lau
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA,Correspondence and requests for materials should be addressed to J.C.S. or C.M.L. ;
| |
Collapse
|
11
|
Zhao M, Havrilla JM, Fang L, Chen Y, Peng J, Liu C, Wu C, Sarmady M, Botas P, Isla J, Lyon GJ, Weng C, Wang K. Phen2Gene: rapid phenotype-driven gene prioritization for rare diseases. NAR Genom Bioinform 2020; 2:lqaa032. [PMID: 32500119 PMCID: PMC7252576 DOI: 10.1093/nargab/lqaa032] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 04/10/2020] [Accepted: 04/28/2020] [Indexed: 02/07/2023] Open
Abstract
Human Phenotype Ontology (HPO) terms are increasingly used in diagnostic settings to aid in the characterization of patient phenotypes. The HPO annotation database is updated frequently and can provide detailed phenotype knowledge on various human diseases, and many HPO terms are now mapped to candidate causal genes with binary relationships. To further improve the genetic diagnosis of rare diseases, we incorporated these HPO annotations, gene-disease databases and gene-gene databases in a probabilistic model to build a novel HPO-driven gene prioritization tool, Phen2Gene. Phen2Gene accesses a database built upon this information called the HPO2Gene Knowledgebase (H2GKB), which provides weighted and ranked gene lists for every HPO term. Phen2Gene is then able to access the H2GKB for patient-specific lists of HPO terms or PhenoPacket descriptions supported by GA4GH (http://phenopackets.org/), calculate a prioritized gene list based on a probabilistic model and output gene-disease relationships with great accuracy. Phen2Gene outperforms existing gene prioritization tools in speed and acts as a real-time phenotype-driven gene prioritization tool to aid the clinical diagnosis of rare undiagnosed diseases. In addition to a command line tool released under the MIT license (https://github.com/WGLab/Phen2Gene), we also developed a web server and web service (https://phen2gene.wglab.org/) for running the tool via web interface or RESTful API queries. Finally, we have curated a large amount of benchmarking data for phenotype-to-gene tools involving 197 patients across 76 scientific articles and 85 patients' de-identified HPO term data from the Children's Hospital of Philadelphia.
Collapse
Affiliation(s)
- Mengge Zhao
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - James M Havrilla
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Li Fang
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Ying Chen
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jacqueline Peng
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Cong Liu
- Department of Biomedical Informatics, Columbia University Medical Center, New York, NY 10032, USA
| | - Chao Wu
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Mahdi Sarmady
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Pablo Botas
- Foundation 29, Pozuelo de Alarcon, 28223 Madrid, Spain
| | - Julián Isla
- Foundation 29, Pozuelo de Alarcon, 28223 Madrid, Spain.,Dravet Syndrome European Federation, 29200 Brest, France
| | - Gholson J Lyon
- Institute for Basic Research in Developmental Disabilities (IBR), Staten Island, NY 10314, USA
| | - Chunhua Weng
- Department of Biomedical Informatics, Columbia University Medical Center, New York, NY 10032, USA
| | - Kai Wang
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
12
|
Brown CC, Gudjonson H, Pritykin Y, Deep D, Lavallée VP, Mendoza A, Fromme R, Mazutis L, Ariyan C, Leslie C, Pe'er D, Rudensky AY. Transcriptional Basis of Mouse and Human Dendritic Cell Heterogeneity. Cell 2019; 179:846-863.e24. [PMID: 31668803 PMCID: PMC6838684 DOI: 10.1016/j.cell.2019.09.035] [Citation(s) in RCA: 361] [Impact Index Per Article: 60.2] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 07/12/2019] [Accepted: 09/27/2019] [Indexed: 12/24/2022]
Abstract
Dendritic cells (DCs) play a critical role in orchestrating adaptive immune responses due to their unique ability to initiate T cell responses and direct their differentiation into effector lineages. Classical DCs have been divided into two subsets, cDC1 and cDC2, based on phenotypic markers and their distinct abilities to prime CD8 and CD4 T cells. While the transcriptional regulation of the cDC1 subset has been well characterized, cDC2 development and function remain poorly understood. By combining transcriptional and chromatin analyses with genetic reporter expression, we identified two principal cDC2 lineages defined by distinct developmental pathways and transcriptional regulators, including T-bet and RORγt, two key transcription factors known to define innate and adaptive lymphocyte subsets. These novel cDC2 lineages were characterized by distinct metabolic and functional programs. Extending our findings to humans revealed conserved DC heterogeneity and the presence of the newly defined cDC2 subsets in human cancer. Single-cell analyses reveal novel dendritic cell subsets Major cDC2 subsets differentially express T-bet and RORγt Distinct pro- and anti-inflammatory potential of T-bet+ and Tbet– cDC2s Transcriptional basis for cDC2 heterogeneity conserved across mouse and human
Collapse
Affiliation(s)
- Chrysothemis C Brown
- Howard Hughes Medical Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Infection, Inflammation and Rheumatology Section, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK.
| | - Herman Gudjonson
- Infection, Inflammation and Rheumatology Section, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
| | - Yuri Pritykin
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Deeksha Deep
- Howard Hughes Medical Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Vincent-Philippe Lavallée
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Alejandra Mendoza
- Howard Hughes Medical Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Rachel Fromme
- Howard Hughes Medical Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Linas Mazutis
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Charlotte Ariyan
- Howard Hughes Medical Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Ludwig Center at Memorial Sloan Kettering Cancer Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Christina Leslie
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Dana Pe'er
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Alexander Y Rudensky
- Howard Hughes Medical Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Ludwig Center at Memorial Sloan Kettering Cancer Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
13
|
Tan Y, Cahan P. SingleCellNet: A Computational Tool to Classify Single Cell RNA-Seq Data Across Platforms and Across Species. Cell Syst 2019; 9:207-213.e2. [PMID: 31377170 DOI: 10.1016/j.cels.2019.06.004] [Citation(s) in RCA: 191] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 04/18/2019] [Accepted: 06/12/2019] [Indexed: 11/28/2022]
Abstract
Single-cell RNA-seq has emerged as a powerful tool in diverse applications, from determining the cell-type composition of tissues to uncovering regulators of developmental programs. A near-universal step in the analysis of single-cell RNA-seq data is to hypothesize the identity of each cell. Often, this is achieved by searching for combinations of genes that have previously been implicated as being cell-type specific, an approach that is not quantitative and does not explicitly take advantage of other single-cell RNA-seq studies. Here, we describe our tool, SingleCellNet, which addresses these issues and enables the classification of query single-cell RNA-seq data in comparison to reference single-cell RNA-seq data. SingleCellNet compares favorably to other methods in sensitivity and specificity, and it is able to classify across platforms and species. We highlight SingleCellNet's utility by classifying previously undetermined cells, and by assessing the outcome of a cell fate engineering experiment.
Collapse
Affiliation(s)
- Yuqi Tan
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Patrick Cahan
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
14
|
Heithoff AJ, Totusek SA, Le D, Barwick L, Gensler G, Franklin DR, Dye AC, Pandey S, Sherman S, Guda C, Fox HS. The integrated National NeuroAIDS Tissue Consortium database: a rich platform for neuroHIV research. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2019; 2019:5277250. [PMID: 30624650 PMCID: PMC6323298 DOI: 10.1093/database/bay134] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 11/29/2018] [Indexed: 01/22/2023]
Abstract
Herein we present major updates to the National NeuroAIDS Tissue Consortium (NNTC) database. The NNTC's ongoing multisite clinical research study was established to facilitate access to ante-mortem and post-mortem data, tissues and biofluids for the neurohuman immunodeficiency virus (HIV) research community. Recently, the NNTC has expanded to include data from the central nervous system HIV Antiretroviral Therapy Effects Research (CHARTER) study. The data and biospecimens from CHARTER and NNTC cohorts are available to qualified researchers upon request. Data generated by requestors using NNTC biospecimens and tissues are returned to the NNTC upon the conclusion of requestors' work, and this external, experimental data are annotated and curated in the publically accessible NNTC database, thereby extending the utility of each case. A flexible and extensible database ontology allows the integration of disparate data sets, including external experimental data, clinical neuropsychological and neuromedical testing data, tissue pathology and neuroimaging data.
Collapse
Affiliation(s)
- Abigail J Heithoff
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Steven A Totusek
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Duc Le
- Bioinformatics and Systems Biology Core, University of Nebraska Medical Center, Omaha, NE, USA
| | | | | | - Donald R Franklin
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Allison C Dye
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sanjit Pandey
- Bioinformatics and Systems Biology Core, University of Nebraska Medical Center, Omaha, NE, USA
| | | | - Chittibabu Guda
- Bioinformatics and Systems Biology Core, University of Nebraska Medical Center, Omaha, NE, USA.,Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Howard S Fox
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
15
|
Lippmann C, Kringel D, Ultsch A, Lötsch J. Computational functional genomics-based approaches in analgesic drug discovery and repurposing. Pharmacogenomics 2018; 19:783-797. [DOI: 10.2217/pgs-2018-0036] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Persistent pain is a major healthcare problem affecting a fifth of adults worldwide with still limited treatment options. The search for new analgesics increasingly includes the novel research area of functional genomics, which combines data derived from various processes related to DNA sequence, gene expression or protein function and uses advanced methods of data mining and knowledge discovery with the goal of understanding the relationship between the genome and the phenotype. Its use in drug discovery and repurposing for analgesic indications has so far been performed using knowledge discovery in gene function and drug target-related databases; next-generation sequencing; and functional proteomics-based approaches. Here, we discuss recent efforts in functional genomics-based approaches to analgesic drug discovery and repurposing and highlight the potential of computational functional genomics in this field including a demonstration of the workflow using a novel R library ‘dbtORA’.
Collapse
Affiliation(s)
- Catharina Lippmann
- Fraunhofer Institute of Molecular Biology & Applied Ecology – Project Group Translational Medicine & Pharmacology (IME–TMP), Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Dario Kringel
- Institute of Clinical Pharmacology, Goethe-University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Alfred Ultsch
- DataBionics Research Group, University of Marburg, Hans-Meerwein-Straße 6, 35032 Marburg, Germany
| | - Jörn Lötsch
- Fraunhofer Institute of Molecular Biology & Applied Ecology – Project Group Translational Medicine & Pharmacology (IME–TMP), Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
- Institute of Clinical Pharmacology, Goethe-University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| |
Collapse
|
16
|
Anderson D, Lassmann T. A phenotype centric benchmark of variant prioritisation tools. NPJ Genom Med 2018; 3:5. [PMID: 29423277 PMCID: PMC5799157 DOI: 10.1038/s41525-018-0044-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 01/09/2018] [Accepted: 01/10/2018] [Indexed: 01/08/2023] Open
Abstract
Next generation sequencing is a standard tool used in clinical diagnostics. In Mendelian diseases the challenge is to discover the single etiological variant among thousands of benign or functionally unrelated variants. After calling variants from aligned sequencing reads, variant prioritisation tools are used to examine the conservation or potential functional consequences of variants. We hypothesised that the performance of variant prioritisation tools may vary by disease phenotype. To test this we created benchmark data sets for variants associated with different disease phenotypes. We found that performance of 24 tested tools is highly variable and differs by disease phenotype. The task of identifying a causative variant amongst a large number of benign variants is challenging for all tools, highlighting the need for further development in the field. Based on our observations, we recommend use of five top performers found in this study (FATHMM, M-CAP, MetaLR, MetaSVM and VEST3). In addition we provide tables indicating which analytical approach works best in which disease context. Variant prioritisation tools are best suited to investigate variants associated with well-studied genetic diseases, as these variants are more readily available during algorithm development than variants associated with rare diseases. We anticipate that further development into disease focussed tools will lead to significant improvements.
Collapse
Affiliation(s)
- Denise Anderson
- Telethon Kids Institute, The University of Western Australia, Subiaco, WA 6008 Australia
| | - Timo Lassmann
- Telethon Kids Institute, The University of Western Australia, Subiaco, WA 6008 Australia
| |
Collapse
|
17
|
WDR5 high expression and its effect on tumorigenesis in leukemia. Oncotarget 2018; 7:37740-37754. [PMID: 27192115 PMCID: PMC5122345 DOI: 10.18632/oncotarget.9312] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 04/27/2016] [Indexed: 01/21/2023] Open
Abstract
WD repeat domain 5 (WDR5) plays an important role in various biological functions through the epigenetic regulation of gene transcription. However, the oncogenic effect of WDR5 in leukemia remains largely unknown. Here, we found WDR5 expression is increased in leukemia patients. High expression of WDR5 is associated with high risk leukemia; Patients with WDR5 and MLL1 high expression have poor complete remission rate. We further identified the global genomic binding of WDR5 in leukemic cells and found the genomic co-localization of WDR5 binding with H3K4me3 enrichment. Moreover, WDR5 knockdown by shRNA suppresses cell proliferation, induces apoptosis, inhibits the expression of WDR5 targets, and blocks the H3K4me3 enrichment on the promoter of its targets. We also observed the positive correlation of WDR5 expression with these targets in the cohort study of leukemia patients. Our data reveal that WDR5 may have oncogenic effect and WDR5-mediated H3K4 methylation plays an important role in leukemogenesis.
Collapse
|
18
|
Perez-Rey D, Alonso-Calvo R, Paraiso-Medina S, Munteanu CR, Garcia-Remesal M. SNOMED2HL7: A tool to normalize and bind SNOMED CT concepts to the HL7 Reference Information Model. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2017; 149:1-9. [PMID: 28802325 DOI: 10.1016/j.cmpb.2017.06.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 04/19/2017] [Accepted: 06/28/2017] [Indexed: 06/07/2023]
Abstract
BACKGROUND Current clinical research and practice requires interoperability among systems in a complex and highly dynamic domain. There has been a significant effort in recent years to develop integrative common data models and domain terminologies. Such efforts have not completely solved the challenges associated with clinical data that are distributed among different and heterogeneous institutions with different systems to encode the information. Currently, when providing homogeneous interfaces to exploit clinical data, certain transformations still involve manual and time-consuming processes that could be automated. OBJECTIVES There is a lack of tools to support data experts adopting clinical standards. This absence is especially significant when links between data model and vocabulary are required. The objective of this work is to present SNOMED2HL7, a novel tool to automatically link biomedical concepts from widely used terminologies, and the corresponding clinical context, to the HL7 Reference Information Model (RIM). METHODS Based on the recommendations of the International Health Terminology Standards Development Organisation (IHTSDO), the SNOMED Normal Form has been implemented within SNOMED2HL7 to decompose and provide a method to reduce the number of options to store the same information. The binding of clinical terminologies to HL7 RIM components is the core of SNOMED2HL7, where terminology concepts have been annotated with the corresponding options within the interoperability standard. A web-based tool has been developed to automatically provide information from the normalization mechanisms and the terminology binding. RESULTS SNOMED2HL7 binding coverage includes the majority of the concepts used to annotate legacy systems. It follows HL7 recommendations to solve binding overlaps and provides the binding of the normalized version of the concepts. The first version of the tool, available at http://kandel.dia.fi.upm.es:8078, has been validated in EU funded projects to integrate real world data for clinical research with an 88.47% of accuracy. CONCLUSIONS This paper presents the first initiative to automatically retrieve concept-centered information required to transform legacy data into widely adopted interoperability standards. Although additional functionality will extend capabilities to automate data transformations, SNOMED2HL7 already provides the functionality required for the clinical interoperability community.
Collapse
Affiliation(s)
- D Perez-Rey
- Biomedical Informatics Group, School of Computer Science, Universidad Politecnica de Madrid. Campus de Montegancedo, s/n, 28660, Boadilla del Monte, Madrid, Spain.
| | - R Alonso-Calvo
- Biomedical Informatics Group, School of Computer Science, Universidad Politecnica de Madrid. Campus de Montegancedo, s/n, 28660, Boadilla del Monte, Madrid, Spain
| | - S Paraiso-Medina
- Biomedical Informatics Group, School of Computer Science, Universidad Politecnica de Madrid. Campus de Montegancedo, s/n, 28660, Boadilla del Monte, Madrid, Spain
| | - C R Munteanu
- RNASA-IMEDIR, Computer Science Faculty, University of A Coruna, Campus de Elviña s/n, 15071, A Coruña, Spain; Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), A Coruña, Spain
| | - M Garcia-Remesal
- Biomedical Informatics Group, School of Computer Science, Universidad Politecnica de Madrid. Campus de Montegancedo, s/n, 28660, Boadilla del Monte, Madrid, Spain
| |
Collapse
|
19
|
Sisino G, Zhou AX, Dahr N, Sabirsh A, Soundarapandian MM, Perera R, Larsson-Lekholm E, Magnone MC, Althage M, Tyrberg B. Long noncoding RNAs are dynamically regulated during β-cell mass expansion in mouse pregnancy and control β-cell proliferation in vitro. PLoS One 2017; 12:e0182371. [PMID: 28796801 PMCID: PMC5552087 DOI: 10.1371/journal.pone.0182371] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 07/17/2017] [Indexed: 11/25/2022] Open
Abstract
Pregnancy is associated with increased β-cell proliferation driven by prolactin. Long noncoding RNAs (lncRNA) are the most abundant RNA species in the mammalian genome, yet, their functional importance is mainly elusive. Aims/hypothesis: This study tests the hypothesis that lncRNAs regulate β-cell proliferation in response to prolactin in the context of β-cell mass compensation in pregnancy. Methods: The expression profile of lncRNAs in mouse islets at day 14.5 of pregnancy was explored by a bioinformatics approach, further confirmed by quantitative PCR at different days of pregnancy, and islet specificity was evaluated by comparing expression in islets versus other tissues. In order to establish the role of the candidate lncRNAs we studied cell proliferation in mouse islets and the MIN6 β-cell line by EdU incorporation and cell count. Results: We found that a group of lncRNAs is differentially regulated in mouse islets at 14.5 days of pregnancy. At different stages of pregnancy, these lncRNAs are dynamically expressed, and expression is prolactin dependent in mouse islets and MIN6 cells. One of those lncRNAs, Gm16308 (Lnc03), is dynamically regulated during pregnancy, prolactin-dependent and islet-enriched. Silencing Lnc03 in primary β-cells and MIN6 cells inhibits, whereas over-expression stimulates, proliferation even in the absence of prolactin, demonstrating that Lnc03 regulates β-cell growth. Conclusions/interpretation: During pregnancy mouse islet proliferation is correlated with dynamic changes of lncRNA expression. In particular, Lnc03 regulates mouse β-cell proliferation and may be a crucial component of β-cell proliferation in β-cell mass adaptation in both health and disease.
Collapse
Affiliation(s)
- Giorgia Sisino
- Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden
| | - Alex-Xianghua Zhou
- Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden
| | - Niklas Dahr
- Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Alan Sabirsh
- Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden
| | | | - Ranjan Perera
- Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, United States of America
| | | | - Maria Chiara Magnone
- Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden
| | - Magnus Althage
- Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden
| | - Björn Tyrberg
- Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden
- * E-mail:
| |
Collapse
|
20
|
Lötsch J, Lippmann C, Kringel D, Ultsch A. Integrated Computational Analysis of Genes Associated with Human Hereditary Insensitivity to Pain. A Drug Repurposing Perspective. Front Mol Neurosci 2017; 10:252. [PMID: 28848388 PMCID: PMC5550731 DOI: 10.3389/fnmol.2017.00252] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Accepted: 07/26/2017] [Indexed: 12/31/2022] Open
Abstract
Genes causally involved in human insensitivity to pain provide a unique molecular source of studying the pathophysiology of pain and the development of novel analgesic drugs. The increasing availability of “big data” enables novel research approaches to chronic pain while also requiring novel techniques for data mining and knowledge discovery. We used machine learning to combine the knowledge about n = 20 genes causally involved in human hereditary insensitivity to pain with the knowledge about the functions of thousands of genes. An integrated computational analysis proposed that among the functions of this set of genes, the processes related to nervous system development and to ceramide and sphingosine signaling pathways are particularly important. This is in line with earlier suggestions to use these pathways as therapeutic target in pain. Following identification of the biological processes characterizing hereditary insensitivity to pain, the biological processes were used for a similarity analysis with the functions of n = 4,834 database-queried drugs. Using emergent self-organizing maps, a cluster of n = 22 drugs was identified sharing important functional features with hereditary insensitivity to pain. Several members of this cluster had been implicated in pain in preclinical experiments. Thus, the present concept of machine-learned knowledge discovery for pain research provides biologically plausible results and seems to be suitable for drug discovery by identifying a narrow choice of repurposing candidates, demonstrating that contemporary machine-learned methods offer innovative approaches to knowledge discovery from available evidence.
Collapse
Affiliation(s)
- Jörn Lötsch
- Institute of Clinical Pharmacology, Goethe-UniversityFrankfurt am Main, Germany.,Fraunhofer Institute of Molecular Biology and Applied Ecology-Project Group, Translational Medicine and Pharmacology (IME-TMP)Frankfurt am Main, Germany
| | - Catharina Lippmann
- Fraunhofer Institute of Molecular Biology and Applied Ecology-Project Group, Translational Medicine and Pharmacology (IME-TMP)Frankfurt am Main, Germany
| | - Dario Kringel
- Institute of Clinical Pharmacology, Goethe-UniversityFrankfurt am Main, Germany
| | - Alfred Ultsch
- DataBionics Research Group, University of MarburgMarburg, Germany
| |
Collapse
|
21
|
Wang YY, Chen WH, Xiao PP, Xie WB, Luo Q, Bork P, Zhao XM. GEAR: A database of Genomic Elements Associated with drug Resistance. Sci Rep 2017; 7:44085. [PMID: 28294141 PMCID: PMC5353689 DOI: 10.1038/srep44085] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 02/02/2017] [Indexed: 12/28/2022] Open
Abstract
Drug resistance is becoming a serious problem that leads to the failure of standard treatments, which is generally developed because of genetic mutations of certain molecules. Here, we present GEAR (A database of Genomic Elements Associated with drug Resistance) that aims to provide comprehensive information about genomic elements (including genes, single-nucleotide polymorphisms and microRNAs) that are responsible for drug resistance. Right now, GEAR contains 1631 associations between 201 human drugs and 758 genes, 106 associations between 29 human drugs and 66 miRNAs, and 44 associations between 17 human drugs and 22 SNPs. These relationships are firstly extracted from primary literature with text mining and then manually curated. The drug resistome deposited in GEAR provides insights into the genetic factors underlying drug resistance. In addition, new indications and potential drug combinations can be identified based on the resistome. The GEAR database can be freely accessed through http://gear.comp-sysbio.org.
Collapse
Affiliation(s)
- Yin-Ying Wang
- Department of Computer Science and Technology, Tongji University, Shanghai 201804, China.,Department of Electronic Engineering, City University of Hong Kong, Kowloon 999077, Hong Kong
| | - Wei-Hua Chen
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular-imaging, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan, Hubei 430074, China
| | - Pei-Pei Xiao
- Department of Computer Science and Technology, Tongji University, Shanghai 201804, China
| | - Wen-Bin Xie
- Department of Computer Science and Technology, Tongji University, Shanghai 201804, China
| | - Qibin Luo
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Peer Bork
- European Molecular Biology Laboratory (EMBL), Heidelberg, 69117, Germany
| | - Xing-Ming Zhao
- Department of Computer Science and Technology, Tongji University, Shanghai 201804, China
| |
Collapse
|
22
|
Abstract
Systems biology is an approach to study all genes, gene transcripts, proteins, metabolites, and their interactions in specific cells, tissues, organs, or the whole organism. It is based on data derived from high-throughput analytical technologies and bioinformatics tools to analyze these data, and aims to understand the whole system rather than individual aspects of it. Systems biology can be applied to virtually all conditions and diseases and therefore also to hypertension and its underlying vascular disorders. Unlike other methods in this book there is no clear-cut protocol to explain a systems biology approach. We will instead outline some of the most important and common steps in the generation and analysis of systems biology data.
Collapse
Affiliation(s)
- Christian Delles
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, 126 University Place, Glasgow, G12 8TA, UK.
| | - Holger Husi
- School of Natural Sciences, University of Stirling, Stirling, UK
| |
Collapse
|
23
|
Ng FS, Sengupta S, Huang Y, Yu AM, You S, Roberts MA, Iyer LK, Yang Y, Jackson FR. TRAP-seq Profiling and RNAi-Based Genetic Screens Identify Conserved Glial Genes Required for Adult Drosophila Behavior. Front Mol Neurosci 2016; 9:146. [PMID: 28066175 PMCID: PMC5177635 DOI: 10.3389/fnmol.2016.00146] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 11/30/2016] [Indexed: 01/06/2023] Open
Abstract
Although, glial cells have well characterized functions in the developing and mature brain, it is only in the past decade that roles for these cells in behavior and plasticity have been delineated. Glial astrocytes and glia-neuron signaling, for example, are now known to have important modulatory functions in sleep, circadian behavior, memory and plasticity. To better understand mechanisms of glia-neuron signaling in the context of behavior, we have conducted cell-specific, genome-wide expression profiling of adult Drosophila astrocyte-like brain cells and performed RNA interference (RNAi)-based genetic screens to identify glial factors that regulate behavior. Importantly, our studies demonstrate that adult fly astrocyte-like cells and mouse astrocytes have similar molecular signatures; in contrast, fly astrocytes and surface glia-different classes of glial cells-have distinct expression profiles. Glial-specific expression of 653 RNAi constructs targeting 318 genes identified multiple factors associated with altered locomotor activity, circadian rhythmicity and/or responses to mechanical stress (bang sensitivity). Of interest, 1 of the relevant genes encodes a vesicle recycling factor, 4 encode secreted proteins and 3 encode membrane transporters. These results strongly support the idea that glia-neuron communication is vital for adult behavior.
Collapse
Affiliation(s)
- Fanny S Ng
- Department of Neuroscience, Sackler Program in Biomedical Sciences, Tufts University School of Medicine Boston, MA, USA
| | - Sukanya Sengupta
- Department of Neuroscience, Sackler Program in Biomedical Sciences, Tufts University School of Medicine Boston, MA, USA
| | - Yanmei Huang
- Department of Neuroscience, Sackler Program in Biomedical Sciences, Tufts University School of Medicine Boston, MA, USA
| | - Amy M Yu
- Department of Neuroscience, Sackler Program in Biomedical Sciences, Tufts University School of Medicine Boston, MA, USA
| | - Samantha You
- Department of Neuroscience, Sackler Program in Biomedical Sciences, Tufts University School of Medicine Boston, MA, USA
| | - Mary A Roberts
- Department of Neuroscience, Sackler Program in Biomedical Sciences, Tufts University School of Medicine Boston, MA, USA
| | - Lakshmanan K Iyer
- Department of Neuroscience, Sackler Program in Biomedical Sciences, Tufts University School of Medicine Boston, MA, USA
| | - Yongjie Yang
- Department of Neuroscience, Sackler Program in Biomedical Sciences, Tufts University School of Medicine Boston, MA, USA
| | - F Rob Jackson
- Department of Neuroscience, Sackler Program in Biomedical Sciences, Tufts University School of Medicine Boston, MA, USA
| |
Collapse
|
24
|
Lötsch J, Ultsch A. A machine-learned computational functional genomics-based approach to drug classification. Eur J Clin Pharmacol 2016; 72:1449-1461. [DOI: 10.1007/s00228-016-2134-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 09/16/2016] [Indexed: 12/20/2022]
|
25
|
Wang SJ, Laulederkind SJF, Hayman GT, Petri V, Smith JR, Tutaj M, Nigam R, Dwinell MR, Shimoyama M. Comprehensive coverage of cardiovascular disease data in the disease portals at the Rat Genome Database. Physiol Genomics 2016; 48:589-600. [PMID: 27287925 DOI: 10.1152/physiolgenomics.00046.2016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 06/08/2016] [Indexed: 01/18/2023] Open
Abstract
Cardiovascular diseases are complex diseases caused by a combination of genetic and environmental factors. To facilitate progress in complex disease research, the Rat Genome Database (RGD) provides the community with a disease portal where genome objects and biological data related to cardiovascular diseases are systematically organized. The purpose of this study is to present biocuration at RGD, including disease, genetic, and pathway data. The RGD curation team uses controlled vocabularies/ontologies to organize data curated from the published literature or imported from disease and pathway databases. These organized annotations are associated with genes, strains, and quantitative trait loci (QTLs), thus linking functional annotations to genome objects. Screen shots from the web pages are used to demonstrate the organization of annotations at RGD. The human cardiovascular disease genes identified by annotations were grouped according to data sources and their annotation profiles were compared by in-house tools and other enrichment tools available to the public. The analysis results show that the imported cardiovascular disease genes from ClinVar and OMIM are functionally different from the RGD manually curated genes in terms of pathway and Gene Ontology annotations. The inclusion of disease genes from other databases enriches the collection of disease genes not only in quantity but also in quality.
Collapse
Affiliation(s)
- Shur-Jen Wang
- Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin; and
| | | | - G Thomas Hayman
- Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin; and
| | - Victoria Petri
- Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin; and
| | - Jennifer R Smith
- Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin; and
| | - Marek Tutaj
- Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin; and
| | - Rajni Nigam
- Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin; and
| | - Melinda R Dwinell
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Mary Shimoyama
- Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin; and
| |
Collapse
|
26
|
Zhong Q, Pevzner SJ, Hao T, Wang Y, Mosca R, Menche J, Taipale M, Taşan M, Fan C, Yang X, Haley P, Murray RR, Mer F, Gebreab F, Tam S, MacWilliams A, Dricot A, Reichert P, Santhanam B, Ghamsari L, Calderwood MA, Rolland T, Charloteaux B, Lindquist S, Barabási AL, Hill DE, Aloy P, Cusick ME, Xia Y, Roth FP, Vidal M. An inter-species protein-protein interaction network across vast evolutionary distance. Mol Syst Biol 2016; 12:865. [PMID: 27107014 PMCID: PMC4848758 DOI: 10.15252/msb.20156484] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 02/22/2016] [Accepted: 03/04/2016] [Indexed: 12/20/2022] Open
Abstract
In cellular systems, biophysical interactions between macromolecules underlie a complex web of functional interactions. How biophysical and functional networks are coordinated, whether all biophysical interactions correspond to functional interactions, and how such biophysical-versus-functional network coordination is shaped by evolutionary forces are all largely unanswered questions. Here, we investigate these questions using an "inter-interactome" approach. We systematically probed the yeast and human proteomes for interactions between proteins from these two species and functionally characterized the resulting inter-interactome network. After a billion years of evolutionary divergence, the yeast and human proteomes are still capable of forming a biophysical network with properties that resemble those of intra-species networks. Although substantially reduced relative to intra-species networks, the levels of functional overlap in the yeast-human inter-interactome network uncover significant remnants of co-functionality widely preserved in the two proteomes beyond human-yeast homologs. Our data support evolutionary selection against biophysical interactions between proteins with little or no co-functionality. Such non-functional interactions, however, represent a reservoir from which nascent functional interactions may arise.
Collapse
Affiliation(s)
- Quan Zhong
- Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA Department of Genetics, Harvard Medical School, Boston, MA, USA Department of Biological Sciences, Wright State University, Dayton, OH, USA
| | - Samuel J Pevzner
- Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA Department of Genetics, Harvard Medical School, Boston, MA, USA Department of Biomedical Engineering, Boston University, Boston, MA, USA Boston University School of Medicine, Boston, MA, USA
| | - Tong Hao
- Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Yang Wang
- Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Roberto Mosca
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona Catalonia, Spain
| | - Jörg Menche
- Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA Center for Complex Network Research (CCNR) and Department of Physics, Northeastern University, Boston, MA, USA
| | - Mikko Taipale
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Murat Taşan
- Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA Departments of Molecular Genetics and Computer Science, University of Toronto, Toronto, ON, Canada Donnelly Centre, University of Toronto, Toronto, ON, Canada Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, ON, Canada
| | - Changyu Fan
- Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Xinping Yang
- Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Patrick Haley
- Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Ryan R Murray
- Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Flora Mer
- Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Fana Gebreab
- Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Stanley Tam
- Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Andrew MacWilliams
- Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Amélie Dricot
- Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Patrick Reichert
- Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Balaji Santhanam
- Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Lila Ghamsari
- Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Michael A Calderwood
- Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Thomas Rolland
- Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Benoit Charloteaux
- Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Susan Lindquist
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Albert-László Barabási
- Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA Center for Complex Network Research (CCNR) and Department of Physics, Northeastern University, Boston, MA, USA Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - David E Hill
- Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Patrick Aloy
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona Catalonia, Spain Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Michael E Cusick
- Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Yu Xia
- Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA Department of Bioengineering, McGill University, Montreal, QC, Canada
| | - Frederick P Roth
- Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA Departments of Molecular Genetics and Computer Science, University of Toronto, Toronto, ON, Canada Donnelly Centre, University of Toronto, Toronto, ON, Canada Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, ON, Canada Canadian Institute for Advanced Research, Toronto, ON, Canada
| | - Marc Vidal
- Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA Department of Genetics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
27
|
Xu J, Lee HJ, Zeng J, Wu Y, Zhang Y, Huang LC, Johnson A, Holla V, Bailey AM, Cohen T, Meric-Bernstam F, Bernstam EV, Xu H. Extracting genetic alteration information for personalized cancer therapy from ClinicalTrials.gov. J Am Med Inform Assoc 2016; 23:750-7. [PMID: 27013523 DOI: 10.1093/jamia/ocw009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 01/13/2016] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVE Clinical trials investigating drugs that target specific genetic alterations in tumors are important for promoting personalized cancer therapy. The goal of this project is to create a knowledge base of cancer treatment trials with annotations about genetic alterations from ClinicalTrials.gov. METHODS We developed a semi-automatic framework that combines advanced text-processing techniques with manual review to curate genetic alteration information in cancer trials. The framework consists of a document classification system to identify cancer treatment trials from ClinicalTrials.gov and an information extraction system to extract gene and alteration pairs from the Title and Eligibility Criteria sections of clinical trials. By applying the framework to trials at ClinicalTrials.gov, we created a knowledge base of cancer treatment trials with genetic alteration annotations. We then evaluated each component of the framework against manually reviewed sets of clinical trials and generated descriptive statistics of the knowledge base. RESULTS AND DISCUSSION The automated cancer treatment trial identification system achieved a high precision of 0.9944. Together with the manual review process, it identified 20 193 cancer treatment trials from ClinicalTrials.gov. The automated gene-alteration extraction system achieved a precision of 0.8300 and a recall of 0.6803. After validation by manual review, we generated a knowledge base of 2024 cancer trials that are labeled with specific genetic alteration information. Analysis of the knowledge base revealed the trend of increased use of targeted therapy for cancer, as well as top frequent gene-alteration pairs of interest. We expect this knowledge base to be a valuable resource for physicians and patients who are seeking information about personalized cancer therapy.
Collapse
Affiliation(s)
- Jun Xu
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Hee-Jin Lee
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jia Zeng
- Institute for Personalized Cancer Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yonghui Wu
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yaoyun Zhang
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Liang-Chin Huang
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Amber Johnson
- Institute for Personalized Cancer Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vijaykumar Holla
- Institute for Personalized Cancer Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ann M Bailey
- Institute for Personalized Cancer Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Trevor Cohen
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Funda Meric-Bernstam
- Institute for Personalized Cancer Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, USA Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elmer V Bernstam
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, USA Division of General Internal Medicine, Department of Internal Medicine, Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Hua Xu
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
28
|
Lötsch J, Ultsch A. A computational functional genomics based self-limiting self-concentration mechanism of cell specialization as a biological role of jumping genes. Integr Biol (Camb) 2016; 8:91-103. [DOI: 10.1039/c5ib00203f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
LINE-1 retrotransposition may result in silencing of genes. This is more likely with genes not carrying active LINE-1 as those are about 10 times more frequent in the given set of genes. Over time this leads to self-specialization of the cell toward processes associated with gene carrying active LINE-1, which then functionally prevail in the chronified situation.
Collapse
Affiliation(s)
- Jörn Lötsch
- Institute of Clinical Pharmacology
- Goethe-University
- Theodor-Stern-Kai 7
- 60590 Frankfurt am Main
- Germany
| | - Alfred Ultsch
- DataBionics Research Group, University of Marburg
- Hans-Meerwein-Straβe
- D-35032 Marburg
- Germany
| |
Collapse
|
29
|
Wang LH, Yen CJ, Li TN, Elowe S, Wang WC, Wang LHC. Sgo1 is a potential therapeutic target for hepatocellular carcinoma. Oncotarget 2015; 6:2023-33. [PMID: 25638162 PMCID: PMC4385833 DOI: 10.18632/oncotarget.2764] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Accepted: 11/17/2014] [Indexed: 02/07/2023] Open
Abstract
Shugoshin-like protein 1 (Sgo1) is an essential protein in mitosis; it protects sister chromatid cohesion and thereby ensures the fidelity of chromosome separation. We found that the expression of Sgo1 mRNA was relatively low in normal tissues, but was upregulated in 82% of hepatocellular carcinoma (HCC), and correlated with elevated alpha-fetoprotein and early disease onset of HCC. The depletion of Sgo1 reduced cell viability of hepatoma cell lines including HuH7, HepG2, Hep3B, and HepaRG. Using time-lapse microscopy, we showed that hepatoma cells were delayed and ultimately die in mitosis in the absence of Sgo1. In contrast, cell viability and mitotic progression of immortalized cells were not significantly affected. Notably, mitotic cell death induced upon Sgo1 depletion was suppressed upon inhibitions of cyclin-dependent kinase-1 and Aurora kinase-B, or the depletion of mitotic arrest deficient-2. Thus, mitotic cell death induced upon Sgo1 depletion in hepatoma cells is mediated by persistent activation of the spindle assembly checkpoint. Together, these results highlight the essential role of Sgo1 in the maintenance of a proper mitotic progression in hepatoma cells and suggest that Sgo1 is a promising oncotarget for HCC.
Collapse
Affiliation(s)
- Lyu-Han Wang
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Chia-Jui Yen
- Institute of Clinical Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tian-Neng Li
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Sabine Elowe
- Université Laval, Faculty of Medicine, Department of Pediatrics, and Reproduction, Perinatal Health, and Infant Health, Québec, Canada
| | - Wen-Ching Wang
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Lily Hui-Ching Wang
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan.,Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
30
|
Skinner BM, Sargent CA, Churcher C, Hunt T, Herrero J, Loveland JE, Dunn M, Louzada S, Fu B, Chow W, Gilbert J, Austin-Guest S, Beal K, Carvalho-Silva D, Cheng W, Gordon D, Grafham D, Hardy M, Harley J, Hauser H, Howden P, Howe K, Lachani K, Ellis PJI, Kelly D, Kerry G, Kerwin J, Ng BL, Threadgold G, Wileman T, Wood JMD, Yang F, Harrow J, Affara NA, Tyler-Smith C. The pig X and Y Chromosomes: structure, sequence, and evolution. Genome Res 2015; 26:130-9. [PMID: 26560630 PMCID: PMC4691746 DOI: 10.1101/gr.188839.114] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 11/09/2015] [Indexed: 12/19/2022]
Abstract
We have generated an improved assembly and gene annotation of the pig X Chromosome, and a first draft assembly of the pig Y Chromosome, by sequencing BAC and fosmid clones from Duroc animals and incorporating information from optical mapping and fiber-FISH. The X Chromosome carries 1033 annotated genes, 690 of which are protein coding. Gene order closely matches that found in primates (including humans) and carnivores (including cats and dogs), which is inferred to be ancestral. Nevertheless, several protein-coding genes present on the human X Chromosome were absent from the pig, and 38 pig-specific X-chromosomal genes were annotated, 22 of which were olfactory receptors. The pig Y-specific Chromosome sequence generated here comprises 30 megabases (Mb). A 15-Mb subset of this sequence was assembled, revealing two clusters of male-specific low copy number genes, separated by an ampliconic region including the HSFY gene family, which together make up most of the short arm. Both clusters contain palindromes with high sequence identity, presumably maintained by gene conversion. Many of the ancestral X-related genes previously reported in at least one mammalian Y Chromosome are represented either as active genes or partial sequences. This sequencing project has allowed us to identify genes--both single copy and amplified--on the pig Y Chromosome, to compare the pig X and Y Chromosomes for homologous sequences, and thereby to reveal mechanisms underlying pig X and Y Chromosome evolution.
Collapse
Affiliation(s)
- Benjamin M Skinner
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, United Kingdom
| | - Carole A Sargent
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, United Kingdom
| | - Carol Churcher
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Toby Hunt
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Javier Herrero
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, Cambridge CB10 1SD, United Kingdom; Bill Lyons Informatics Centre, UCL Cancer Institute, University College London, London WC1E 6BT, United Kingdom
| | - Jane E Loveland
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Matt Dunn
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Sandra Louzada
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Beiyuan Fu
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - William Chow
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - James Gilbert
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| | | | - Kathryn Beal
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, Cambridge CB10 1SD, United Kingdom
| | - Denise Carvalho-Silva
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, Cambridge CB10 1SD, United Kingdom
| | - William Cheng
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Daria Gordon
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Darren Grafham
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Matt Hardy
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Jo Harley
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Heidi Hauser
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Philip Howden
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, United Kingdom; Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Kerstin Howe
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Kim Lachani
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, United Kingdom
| | - Peter J I Ellis
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, United Kingdom
| | - Daniel Kelly
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Giselle Kerry
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - James Kerwin
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Bee Ling Ng
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Glen Threadgold
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Thomas Wileman
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Jonathan M D Wood
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Fengtang Yang
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Jen Harrow
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Nabeel A Affara
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, United Kingdom
| | - Chris Tyler-Smith
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| |
Collapse
|
31
|
Kelsen JR, Dawany N, Moran CJ, Petersen BS, Sarmady M, Sasson A, Pauly-Hubbard H, Martinez A, Maurer K, Soong J, Rappaport E, Franke A, Keller A, Winter HS, Mamula P, Piccoli D, Artis D, Sonnenberg GF, Daly M, Sullivan KE, Baldassano RN, Devoto M. Exome sequencing analysis reveals variants in primary immunodeficiency genes in patients with very early onset inflammatory bowel disease. Gastroenterology 2015; 149:1415-24. [PMID: 26193622 PMCID: PMC4853027 DOI: 10.1053/j.gastro.2015.07.006] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 07/08/2015] [Accepted: 07/13/2015] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Very early onset inflammatory bowel disease (VEO-IBD), IBD diagnosed at 5 years of age or younger, frequently presents with a different and more severe phenotype than older-onset IBD. We investigated whether patients with VEO-IBD carry rare or novel variants in genes associated with immunodeficiencies that might contribute to disease development. METHODS Patients with VEO-IBD and parents (when available) were recruited from the Children's Hospital of Philadelphia from March 2013 through July 2014. We analyzed DNA from 125 patients with VEO-IBD (age, 3 wk to 4 y) and 19 parents, 4 of whom also had IBD. Exome capture was performed by Agilent SureSelect V4, and sequencing was performed using the Illumina HiSeq platform. Alignment to human genome GRCh37 was achieved followed by postprocessing and variant calling. After functional annotation, candidate variants were analyzed for change in protein function, minor allele frequency less than 0.1%, and scaled combined annotation-dependent depletion scores of 10 or less. We focused on genes associated with primary immunodeficiencies and related pathways. An additional 210 exome samples from patients with pediatric IBD (n = 45) or adult-onset Crohn's disease (n = 20) and healthy individuals (controls, n = 145) were obtained from the University of Kiel, Germany, and used as control groups. RESULTS Four hundred genes and regions associated with primary immunodeficiency, covering approximately 6500 coding exons totaling more than 1 Mbp of coding sequence, were selected from the whole-exome data. Our analysis showed novel and rare variants within these genes that could contribute to the development of VEO-IBD, including rare heterozygous missense variants in IL10RA and previously unidentified variants in MSH5 and CD19. CONCLUSIONS In an exome sequence analysis of patients with VEO-IBD and their parents, we identified variants in genes that regulate B- and T-cell functions and could contribute to pathogenesis. Our analysis could lead to the identification of previously unidentified IBD-associated variants.
Collapse
Affiliation(s)
- Judith R. Kelsen
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia
| | - Noor Dawany
- Department of Biomedical Health Informatics, The Children's Hospital of Philadelphia
| | - Christopher J. Moran
- Division of Pediatric Gastroenterology, Hepatology, & Nutrition, Massachusetts General Hospital for Children
| | - Britt-Sabina Petersen
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Germany
| | - Mahdi Sarmady
- Department of Biomedical Health Informatics, The Children's Hospital of Philadelphia
| | - Ariella Sasson
- Department of Biomedical Health Informatics, The Children's Hospital of Philadelphia
| | - Helen Pauly-Hubbard
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia
| | - Alejandro Martinez
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia
| | - Kelly Maurer
- Division of Immunology and Allergy, The Children's Hospital of Philadelphia
| | - Joanne Soong
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology and Hepatology, Department of Microbiology & Immunology, and The Jill Robert's Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medical College, New York, New York, USA
| | - Eric Rappaport
- Nucleic Acid/PCR Core, The Children's Hospital of Philadelphia
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Germany
| | - Andreas Keller
- Department of Clinical Bioinformatics, Saarland University, Germany
| | - Harland S. Winter
- Division of Pediatric Gastroenterology, Hepatology, & Nutrition, Massachusetts General Hospital for Children
| | - Petar Mamula
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia
| | - David Piccoli
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia
| | - David Artis
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology and Hepatology, Department of Microbiology & Immunology, and The Jill Robert's Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medical College, New York, New York, USA
| | - Gregory F. Sonnenberg
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology and Hepatology, Department of Microbiology & Immunology, and The Jill Robert's Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medical College, New York, New York, USA
| | - Mark Daly
- Analytic and Translational Unit Center for Human Genetic Research Department of Medicine, Massachusetts General Hospital,The Broad Institute of MIT and Harvard
| | | | - Robert N. Baldassano
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia
| | - Marcella Devoto
- Division of Human Genetics, The Children's Hospital of Philadelphia, Department of Pediatrics, Department of Biostatistics and Epidemiology, Perelman School of Medicine, University of Pennsylvania; Department of Molecular Medicine, University Sapienza, Rome, Italy
| |
Collapse
|
32
|
Computational functional genomics based analysis of pain-relevant micro-RNAs. Hum Genet 2015; 134:1221-38. [DOI: 10.1007/s00439-015-1600-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 09/01/2015] [Indexed: 02/07/2023]
|
33
|
Yang H, Robinson PN, Wang K. Phenolyzer: phenotype-based prioritization of candidate genes for human diseases. Nat Methods 2015; 12:841-3. [PMID: 26192085 PMCID: PMC4718403 DOI: 10.1038/nmeth.3484] [Citation(s) in RCA: 282] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 05/18/2015] [Indexed: 12/21/2022]
Abstract
Prior biological knowledge and phenotype information may help to identify disease genes from human whole-genome and whole-exome sequencing studies. We developed Phenolyzer (http://phenolyzer.usc.edu), a tool that uses prior information to implicate genes involved in diseases. Phenolyzer exhibits superior performance over competing methods for prioritizing Mendelian and complex disease genes, based on disease or phenotype terms entered as free text.
Collapse
Affiliation(s)
- Hui Yang
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
- Neuroscience Graduate Program, University of Southern California, Los Angeles, California, USA
| | - Peter N Robinson
- Institute for Medical and Human Genetics, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Max Planck Institute for Molecular Genetics, Berlin, Germany
- Berlin Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Berlin, Germany
- Institute for Bioinformatics, Department of Mathematics and Computer Science, Freie Universität Berlin, Berlin, Germany
| | - Kai Wang
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
- Department of Psychiatry, University of Southern California, Los Angeles, California, USA
- Division of Bioinformatics, Department of Preventive Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
34
|
Cserhati MF, Pandey S, Beaudoin JJ, Baccaglini L, Guda C, Fox HS. The National NeuroAIDS Tissue Consortium (NNTC) Database: an integrated database for HIV-related studies. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2015; 2015:bav074. [PMID: 26228431 PMCID: PMC4520230 DOI: 10.1093/database/bav074] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Accepted: 06/30/2015] [Indexed: 11/13/2022]
Abstract
We herein present the National NeuroAIDS Tissue Consortium-Data Coordinating Center (NNTC-DCC) database, which is the only available database for neuroAIDS studies that contains data in an integrated, standardized form. This database has been created in conjunction with the NNTC, which provides human tissue and biofluid samples to individual researchers to conduct studies focused on neuroAIDS. The database contains experimental datasets from 1206 subjects for the following categories (which are further broken down into subcategories): gene expression, genotype, proteins, endo-exo-chemicals, morphometrics and other (miscellaneous) data. The database also contains a wide variety of downloadable data and metadata for 95 HIV-related studies covering 170 assays from 61 principal investigators. The data represent 76 tissue types, 25 measurement types, and 38 technology types, and reaches a total of 33 017 407 data points. We used the ISA platform to create the database and develop a searchable web interface for querying the data. A gene search tool is also available, which searches for NCBI GEO datasets associated with selected genes. The database is manually curated with many user-friendly features, and is cross-linked to the NCBI, HUGO and PubMed databases. A free registration is required for qualified users to access the database. Database URL: http://nntc-dcc.unmc.edu
Collapse
Affiliation(s)
- Matyas F Cserhati
- Department of Genetics, Cell Biology and Anatomy, Bioinformatics and Systems Biology Core
| | - Sanjit Pandey
- Department of Genetics, Cell Biology and Anatomy, Bioinformatics and Systems Biology Core
| | - James J Beaudoin
- Department of Pharmacology and Experimental Neuroscience, College of Medicine
| | | | - Chittibabu Guda
- Department of Genetics, Cell Biology and Anatomy, Bioinformatics and Systems Biology Core, Fred and Pamela Buffet Cancer Center, Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Howard S Fox
- Department of Pharmacology and Experimental Neuroscience, College of Medicine,
| |
Collapse
|
35
|
Lee PF, Soo VW. An ensemble rank learning approach for gene prioritization. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2015; 2013:3507-10. [PMID: 24110485 DOI: 10.1109/embc.2013.6610298] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Several different computational approaches have been developed to solve the gene prioritization problem. We intend to use the ensemble boosting learning techniques to combine variant computational approaches for gene prioritization in order to improve the overall performance. In particular we add a heuristic weighting function to the Rankboost algorithm according to: 1) the absolute ranks generated by the adopted methods for a certain gene, and 2) the ranking relationship between all gene-pairs from each prioritization result. We select 13 known prostate cancer genes in OMIM database as training set and protein coding gene data in HGNC database as test set. We adopt the leave-one-out strategy for the ensemble rank boosting learning. The experimental results show that our ensemble learning approach outperforms the four gene-prioritization methods in ToppGene suite in the ranking results of the 13 known genes in terms of mean average precision, ROC and AUC measures.
Collapse
|
36
|
Ibekwe TS, Bhimrao SK, Westerberg BD, Kozak FK. A meta-analysis and systematic review of the prevalence of mitochondrially encoded 12S RNA in the general population: Is there a role for screening neonates requiring aminoglycosides? Afr J Paediatr Surg 2015; 12:105-13. [PMID: 26168747 PMCID: PMC4955414 DOI: 10.4103/0189-6725.160342] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND This was a meta-analysis and systematic review to determine the global prevalence of the mitochondrially encoded 12S RNA (MT-RNR1) genetic mutation in order to assess the need for neonatal screening prior to aminoglycoside therapy. MATERIALS AND METHODS A comprehensive search of MEDLINE, EMBASE, Ovid, Database of Abstracts of Reviews of Effect, Cochrane Library, Clinical Evidence and Cochrane Central Register of Trials was performed including cross-referencing independently by 2 assessors. Selections were restricted to human studies in English. Meta-analysis was done with MetaXL 2013. RESULTS Forty-five papers out of 295 met the criteria. Pooled prevalence in the general population for MT-RNR1 gene mutations (A1555G, C1494T, A7445G) was 2% (1-4%) at 99%. CONCLUSION Routine screening for MT-RNR1 mutations in the general population prior to treatment with aminoglycosides appear desirable but poorly supported by the weak level of evidence available in the literature. Routine screening in high-risk (Chinese and Spanish) populations appear justified.
Collapse
Affiliation(s)
- Titus S Ibekwe
- Department of ENT, University of Abuja Teaching Hospital and College of Health Sciences, University of Abuja, Abuja, Nigeria
| | | | | | | |
Collapse
|
37
|
Mesbah-Uddin M, Elango R, Banaganapalli B, Shaik NA, Al-Abbasi FA. In-silico analysis of inflammatory bowel disease (IBD) GWAS loci to novel connections. PLoS One 2015; 10:e0119420. [PMID: 25786114 PMCID: PMC4364731 DOI: 10.1371/journal.pone.0119420] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 01/13/2015] [Indexed: 12/19/2022] Open
Abstract
Genome-wide association studies (GWASs) for many complex diseases, including inflammatory bowel disease (IBD), produced hundreds of disease-associated loci—the majority of which are noncoding. The number of GWAS loci is increasing very rapidly, but the process of translating single nucleotide polymorphisms (SNPs) from these loci to genomic medicine is lagging. In this study, we investigated 4,734 variants from 152 IBD associated GWAS loci (IBD associated 152 lead noncoding SNPs identified from pooled GWAS results + 4,582 variants in strong linkage-disequilibrium (LD) (r2 ≥0.8) for EUR population of 1K Genomes Project) using four publicly available bioinformatics tools, e.g. dbPSHP, CADD, GWAVA, and RegulomeDB, to annotate and prioritize putative regulatory variants. Of the 152 lead noncoding SNPs, around 11% are under strong negative selection (GERP++ RS ≥2); and ~30% are under balancing selection (Tajima’s D score >2) in CEU population (1K Genomes Project)—though these regions are positively selected (GERP++ RS <0) in mammalian evolution. The analysis of 4,734 variants using three integrative annotation tools produced 929 putative functional SNPs, of which 18 SNPs (from 15 GWAS loci) are in concordance with all three classifiers. These prioritized noncoding SNPs may contribute to IBD pathogenesis by dysregulating the expression of nearby genes. This study showed the usefulness of integrative annotation for prioritizing fewer functional variants from a large number of GWAS markers.
Collapse
Affiliation(s)
- Md. Mesbah-Uddin
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- * E-mail: (MMU); (FAA)
| | - Ramu Elango
- Princess Al-Jawhara Al-Brahim Center of Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Babajan Banaganapalli
- Princess Al-Jawhara Al-Brahim Center of Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Noor Ahmad Shaik
- Princess Al-Jawhara Al-Brahim Center of Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Genetic Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Fahad A. Al-Abbasi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- * E-mail: (MMU); (FAA)
| |
Collapse
|
38
|
Alonso-Calvo R, Perez-Rey D, Paraiso-Medina S, Claerhout B, Hennebert P, Bucur A. Enabling semantic interoperability in multi-centric clinical trials on breast cancer. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2015; 118:322-329. [PMID: 25682737 DOI: 10.1016/j.cmpb.2015.01.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 12/10/2014] [Accepted: 01/23/2015] [Indexed: 06/04/2023]
Abstract
BACKGROUND AND OBJECTIVES Post-genomic clinical trials require the participation of multiple institutions, and collecting data from several hospitals, laboratories and research facilities. This paper presents a standard-based solution to provide a uniform access endpoint to patient data involved in current clinical research. METHODS The proposed approach exploits well-established standards such as HL7 v3 or SPARQL and medical vocabularies such as SNOMED CT, LOINC and HGNC. A novel mechanism to exploit semantic normalization among HL7-based data models and biomedical ontologies has been created by using Semantic Web technologies. RESULTS Different types of queries have been used for testing the semantic interoperability solution described in this paper. The execution times obtained in the tests enable the development of end user tools within a framework that requires efficient retrieval of integrated data. CONCLUSIONS The proposed approach has been successfully tested by applications within the INTEGRATE and EURECA EU projects. These applications have been deployed and tested for: (i) patient screening, (ii) trial recruitment, and (iii) retrospective analysis; exploiting semantically interoperable access to clinical patient data from heterogeneous data sources.
Collapse
Affiliation(s)
- Raul Alonso-Calvo
- Biomedical Informatics Group, DLSIIS & DIA, Facultad de Informática, Universidad Politécnica de Madrid, Campus de Montegancedo S/N, 28660 Boadilla del Monte, Madrid, Spain.
| | - David Perez-Rey
- Biomedical Informatics Group, DLSIIS & DIA, Facultad de Informática, Universidad Politécnica de Madrid, Campus de Montegancedo S/N, 28660 Boadilla del Monte, Madrid, Spain
| | - Sergio Paraiso-Medina
- Biomedical Informatics Group, DLSIIS & DIA, Facultad de Informática, Universidad Politécnica de Madrid, Campus de Montegancedo S/N, 28660 Boadilla del Monte, Madrid, Spain
| | - Brecht Claerhout
- Custodix NV, Kortrijksesteenweg 214b3, Sint-Martens-Latem, Belgium
| | | | - Anca Bucur
- PHILIPS Research Europe, High Tech Campus 34, Eindhoven, Netherlands
| |
Collapse
|
39
|
Lötsch J, Daiker H, Hähner A, Ultsch A, Hummel T. Drug-target based cross-sectional analysis of olfactory drug effects. Eur J Clin Pharmacol 2015; 71:461-71. [DOI: 10.1007/s00228-015-1814-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 01/23/2015] [Indexed: 10/24/2022]
|
40
|
Oulas A, Karathanasis N, Louloupi A, Pavlopoulos GA, Poirazi P, Kalantidis K, Iliopoulos I. Prediction of miRNA targets. Methods Mol Biol 2015; 1269:207-29. [PMID: 25577381 DOI: 10.1007/978-1-4939-2291-8_13] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Computational methods for miRNA target prediction are currently undergoing extensive review and evaluation. There is still a great need for improvement of these tools and bioinformatics approaches are looking towards high-throughput experiments in order to validate predictions. The combination of large-scale techniques with computational tools will not only provide greater credence to computational predictions but also lead to the better understanding of specific biological questions. Current miRNA target prediction tools utilize probabilistic learning algorithms, machine learning methods and even empirical biologically defined rules in order to build models based on experimentally verified miRNA targets. Large-scale protein downregulation assays and next-generation sequencing (NGS) are now being used to validate methodologies and compare the performance of existing tools. Tools that exhibit greater correlation between computational predictions and protein downregulation or RNA downregulation are considered the state of the art. Moreover, efficiency in prediction of miRNA targets that are concurrently verified experimentally provides additional validity to computational predictions and further highlights the competitive advantage of specific tools and their efficacy in extracting biologically significant results. In this review paper, we discuss the computational methods for miRNA target prediction and provide a detailed comparison of methodologies and features utilized by each specific tool. Moreover, we provide an overview of current state-of-the-art high-throughput methods used in miRNA target prediction.
Collapse
Affiliation(s)
- Anastasis Oulas
- Institute of Marine Biology, Biotechnology and Aquaculture-HCMR, Heraklion, Crete, Greece
| | | | | | | | | | | | | |
Collapse
|
41
|
Kenyon EJ, Campos I, Bull JC, Williams PH, Stemple DL, Clark MD. Zebrafish Rab5 proteins and a role for Rab5ab in nodal signalling. Dev Biol 2014; 397:212-24. [PMID: 25478908 PMCID: PMC4294769 DOI: 10.1016/j.ydbio.2014.11.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 11/03/2014] [Accepted: 11/11/2014] [Indexed: 01/08/2023]
Abstract
The RAB5 gene family is the best characterised of all human RAB families and is essential for in vitro homotypic fusion of early endosomes. In recent years, the disruption or activation of Rab5 family proteins has been used as a tool to understand growth factor signal transduction in whole animal systems such as Drosophila melanogaster and zebrafish. In this study we have examined the functions for four rab5 genes in zebrafish. Disruption of rab5ab expression by antisense morpholino oligonucleotide (MO) knockdown abolishes nodal signalling in early zebrafish embryos, whereas overexpression of rab5ab mRNA leads to ectopic expression of markers that are normally downstream of nodal signalling. By contrast MO disruption of other zebrafish rab5 genes shows little or no effect on expression of markers of dorsal organiser development. We conclude that rab5ab is essential for nodal signalling and organizer specification in the developing zebrafish embryo. We have examined the activities of each of the zebrafish Rab5 genes using morpholino knockdowns. Loss of one Rab5 isoform, Rab5ab, affects formation of the dorsal organizer. Rab5ab overexpression leads to ectopic expression of dorsal markers.
Collapse
Affiliation(s)
- Emma J Kenyon
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Isabel Campos
- Champalimaud Centre for the Unknown, Fundação Champalimaud, Lisboa, Portugal
| | - James C Bull
- Department of Biosciences, Swansea University, Swansea SA2 8PP, United Kingdom
| | - P Huw Williams
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Derek L Stemple
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, United Kingdom.
| | - Matthew D Clark
- Sequencing Technology Development, The Genome Analysis Centre, Norwich Research Park, Colney, Norwich NR4 7UH, United Kingdom
| |
Collapse
|
42
|
Ultsch A, Lötsch J. What do all the (human) micro-RNAs do? BMC Genomics 2014; 15:976. [PMID: 25404408 PMCID: PMC4289375 DOI: 10.1186/1471-2164-15-976] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 10/13/2014] [Indexed: 12/23/2022] Open
Abstract
Background Micro-RNAs (miRNA) are attributed to the systems biological role of a regulatory mechanism of the expression of protein coding genes. Research has identified miRNAs dysregulations in several but distinct pathophysiological processes, which hints at distinct systems-biology functions of miRNAs. The present analysis approached the role of miRNAs from a genomics perspective and assessed the biological roles of 2954 genes and 788 human miRNAs, which can be considered to interact, based on empirical evidence and computational predictions of miRNA versus gene interactions. Results From a genomics perspective, the biological processes in which the genes that are influenced by miRNAs are involved comprise of six major topics comprising biological regulation, cellular metabolism, information processing, development, gene expression and tissue homeostasis. The usage of this knowledge as a guidance for further research is sketched for two genetically defined functional areas: cell death and gene expression. Results suggest that the latter points to a fundamental role of miRNAs consisting of hyper-regulation of gene expression, i.e., the control of the expression of such genes which control specifically the expression of genes. Conclusions Laboratory research identified contributions of miRNA regulation to several distinct biological processes. The present analysis transferred this knowledge to a systems-biology level. A comprehensible and precise description of the biological processes in which the genes that are influenced by miRNAs are notably involved could be made. This knowledge can be employed to guide future research concerning the biological role of miRNA (dys-) regulations. The analysis also suggests that miRNAs especially control the expression of genes that control the expression of genes. Electronic supplementary material The online version of this article (doi:10.1186/1471-2164-15-976) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Jörn Lötsch
- Institute of Clinical Pharmacology, Goethe - University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.
| |
Collapse
|
43
|
Beccuti M, Carrara M, Cordero F, Lazzarato F, Donatelli S, Nadalin F, Policriti A, Calogero RA. Chimera: a Bioconductor package for secondary analysis of fusion products. ACTA ACUST UNITED AC 2014; 30:3556-7. [PMID: 25286921 PMCID: PMC4253834 DOI: 10.1093/bioinformatics/btu662] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Summary:Chimera is a Bioconductor package that organizes, annotates, analyses and validates fusions reported by different fusion detection tools; current implementation can deal with output from bellerophontes, chimeraScan, deFuse, fusionCatcher, FusionFinder, FusionHunter, FusionMap, mapSplice, Rsubread, tophat-fusion and STAR. The core of Chimera is a fusion data structure that can store fusion events detected with any of the aforementioned tools. Fusions are then easily manipulated with standard R functions or through the set of functionalities specifically developed in Chimera with the aim of supporting the user in managing fusions and discriminating false-positive results. Availability and implementation:Chimera is implemented as a Bioconductor package in R. The package and the vignette can be downloaded at bioconductor.org. Contact:raffaele.calogero@unito.it Supplementary information:Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Marco Beccuti
- Department of Computer Sciences, University of Torino, C.so Svizzera 185, 10149 Torino, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Department of Translational Medicine, University of Piemonte Orientale Avogadro, Novara, Unit of Cancer Epidemiology, Department of Medical Sciences, University of Turin, Turin, Department of Computational and Quantitative Biology, UMR 7238 CNRS - Université Pierre et Marie Curie, Paris, France and Department of Mathematics and Computer Science, University of Udine, Italy
| | - Matteo Carrara
- Department of Computer Sciences, University of Torino, C.so Svizzera 185, 10149 Torino, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Department of Translational Medicine, University of Piemonte Orientale Avogadro, Novara, Unit of Cancer Epidemiology, Department of Medical Sciences, University of Turin, Turin, Department of Computational and Quantitative Biology, UMR 7238 CNRS - Université Pierre et Marie Curie, Paris, France and Department of Mathematics and Computer Science, University of Udine, Italy
| | - Francesca Cordero
- Department of Computer Sciences, University of Torino, C.so Svizzera 185, 10149 Torino, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Department of Translational Medicine, University of Piemonte Orientale Avogadro, Novara, Unit of Cancer Epidemiology, Department of Medical Sciences, University of Turin, Turin, Department of Computational and Quantitative Biology, UMR 7238 CNRS - Université Pierre et Marie Curie, Paris, France and Department of Mathematics and Computer Science, University of Udine, Italy
| | - Fulvio Lazzarato
- Department of Computer Sciences, University of Torino, C.so Svizzera 185, 10149 Torino, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Department of Translational Medicine, University of Piemonte Orientale Avogadro, Novara, Unit of Cancer Epidemiology, Department of Medical Sciences, University of Turin, Turin, Department of Computational and Quantitative Biology, UMR 7238 CNRS - Université Pierre et Marie Curie, Paris, France and Department of Mathematics and Computer Science, University of Udine, Italy Department of Computer Sciences, University of Torino, C.so Svizzera 185, 10149 Torino, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Department of Translational Medicine, University of Piemonte Orientale Avogadro, Novara, Unit of Cancer Epidemiology, Department of Medical Sciences, University of Turin, Turin, Department of Computational and Quantitative Biology, UMR 7238 CNRS - Université Pierre et Marie Curie, Paris, France and Department of Mathematics and Computer Science, University of Udine, Italy
| | - Susanna Donatelli
- Department of Computer Sciences, University of Torino, C.so Svizzera 185, 10149 Torino, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Department of Translational Medicine, University of Piemonte Orientale Avogadro, Novara, Unit of Cancer Epidemiology, Department of Medical Sciences, University of Turin, Turin, Department of Computational and Quantitative Biology, UMR 7238 CNRS - Université Pierre et Marie Curie, Paris, France and Department of Mathematics and Computer Science, University of Udine, Italy
| | - Francesca Nadalin
- Department of Computer Sciences, University of Torino, C.so Svizzera 185, 10149 Torino, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Department of Translational Medicine, University of Piemonte Orientale Avogadro, Novara, Unit of Cancer Epidemiology, Department of Medical Sciences, University of Turin, Turin, Department of Computational and Quantitative Biology, UMR 7238 CNRS - Université Pierre et Marie Curie, Paris, France and Department of Mathematics and Computer Science, University of Udine, Italy
| | - Alberto Policriti
- Department of Computer Sciences, University of Torino, C.so Svizzera 185, 10149 Torino, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Department of Translational Medicine, University of Piemonte Orientale Avogadro, Novara, Unit of Cancer Epidemiology, Department of Medical Sciences, University of Turin, Turin, Department of Computational and Quantitative Biology, UMR 7238 CNRS - Université Pierre et Marie Curie, Paris, France and Department of Mathematics and Computer Science, University of Udine, Italy
| | - Raffaele A Calogero
- Department of Computer Sciences, University of Torino, C.so Svizzera 185, 10149 Torino, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Department of Translational Medicine, University of Piemonte Orientale Avogadro, Novara, Unit of Cancer Epidemiology, Department of Medical Sciences, University of Turin, Turin, Department of Computational and Quantitative Biology, UMR 7238 CNRS - Université Pierre et Marie Curie, Paris, France and Department of Mathematics and Computer Science, University of Udine, Italy
| |
Collapse
|
44
|
Paraiso-Medina S, Perez-Rey D, Bucur A, Claerhout B, Alonso-Calvo R. Semantic Normalization and Query Abstraction Based on SNOMED-CT and HL7: Supporting Multicentric Clinical Trials. IEEE J Biomed Health Inform 2014; 19:1061-7. [PMID: 25248204 DOI: 10.1109/jbhi.2014.2357025] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Advances in the use of omic data and other biomarkers are increasing the number of variables in clinical research. Additional data have stratified the population of patients and require that current studies be performed among multiple institutions. Semantic interoperability and standardized data representation are a crucial task in the management of modern clinical trials. In the past few years, different efforts have focused on integrating biomedical information. Due to the complexity of this domain and the specific requirements of clinical research, the majority of data integration tasks are still performed manually. This paper presents a semantic normalization process and a query abstraction mechanism to facilitate data integration and retrieval. A process based on well-established standards from the biomedical domain and the latest semantic web technologies has been developed. Methods proposed in this paper have been tested within the EURECA EU research project, where clinical scenarios require the extraction of semantic knowledge from biomedical vocabularies. The aim of this paper is to provide a novel method to abstract from the data model and query syntax. The proposed approach has been compared with other initiatives in the field by storing the same dataset with each of those solutions. Results show an extended functionality and query capabilities at the cost of slightly worse performance in query execution. Implementations in real settings have shown that following this approach, usable interfaces can be developed to exploit clinical trial data outcomes.
Collapse
|
45
|
Ulveling D, Dinger ME, Francastel C, Hubé F. Identification of a dinucleotide signature that discriminates coding from non-coding long RNAs. Front Genet 2014; 5:316. [PMID: 25250049 PMCID: PMC4158813 DOI: 10.3389/fgene.2014.00316] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 08/22/2014] [Indexed: 11/13/2022] Open
Abstract
To date, the main criterion by which long ncRNAs (lncRNAs) are discriminated from mRNAs is based on the capacity of the transcripts to encode a protein. However, it becomes important to identify non-ORF-based sequence characteristics that can be used to parse between ncRNAs and mRNAs. In this study, we first established an extremely selective workflow to define a highly refined database of lncRNAs which was used for comparison with mRNAs. Then using this highly selective collection of lncRNAs, we found the CG dinucleotide frequencies were clearly distinct. In addition, we showed that the bias in CG dinucleotide frequency was conserved in human and mouse genomes. We propose that this sequence feature will serve as a useful classifier in transcript classification pipelines. We also suggest that our refined database of "bona fide" lncRNAs will be valuable for the discovery of other sequence characteristics distinct to lncRNAs.
Collapse
Affiliation(s)
- Damien Ulveling
- CNRS UMR7216, Epigenetics and Cell Fate, Université Paris Diderot, Sorbonne Paris Cité Paris, France
| | - Marcel E Dinger
- The University of Queensland Diamantina Institute, The University of Queensland Brisbane, QLD, Australia
| | - Claire Francastel
- CNRS UMR7216, Epigenetics and Cell Fate, Université Paris Diderot, Sorbonne Paris Cité Paris, France
| | - Florent Hubé
- CNRS UMR7216, Epigenetics and Cell Fate, Université Paris Diderot, Sorbonne Paris Cité Paris, France
| |
Collapse
|
46
|
DICER1 mutations in childhood cystic nephroma and its relationship to DICER1-renal sarcoma. Mod Pathol 2014; 27:1267-80. [PMID: 24481001 PMCID: PMC4117822 DOI: 10.1038/modpathol.2013.242] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Revised: 10/18/2013] [Accepted: 10/20/2013] [Indexed: 01/13/2023]
Abstract
The pathogenesis of cystic nephroma of the kidney has interested pathologists for over 50 years. Emerging from its initial designation as a type of unilateral multilocular cyst, cystic nephroma has been considered as either a developmental abnormality or a neoplasm or both. Many have viewed cystic nephroma as the benign end of the pathologic spectrum with cystic partially differentiated nephroblastoma and Wilms tumor, whereas others have considered it a mixed epithelial and stromal tumor. We hypothesize that cystic nephroma, like the pleuropulmonary blastoma in the lung, represents a spectrum of abnormal renal organogenesis with risk for malignant transformation. Here we studied DICER1 mutations in a cohort of 20 cystic nephromas and 6 cystic partially differentiated nephroblastomas, selected independently of a familial association with pleuropulmonary blastoma and describe four cases of sarcoma arising in cystic nephroma, which have a similarity to the solid areas of type II or III pleuropulmonary blastoma. The genetic analyses presented here confirm that DICER1 mutations are the major genetic event in the development of cystic nephroma. Further, cystic nephroma and pleuropulmonary blastoma have similar DICER1 loss of function and 'hotspot' missense mutation rates, which involve specific amino acids in the RNase IIIb domain. We propose an alternative pathway with the genetic pathogenesis of cystic nephroma and DICER1-renal sarcoma paralleling that of type I to type II/III malignant progression of pleuropulmonary blastoma.
Collapse
|
47
|
Papanikolaou N, Pavlopoulos GA, Pafilis E, Theodosiou T, Schneider R, Satagopam VP, Ouzounis CA, Eliopoulos AG, Promponas VJ, Iliopoulos I. BioTextQuest(+): a knowledge integration platform for literature mining and concept discovery. ACTA ACUST UNITED AC 2014; 30:3249-56. [PMID: 25100685 DOI: 10.1093/bioinformatics/btu524] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
SUMMARY The iterative process of finding relevant information in biomedical literature and performing bioinformatics analyses might result in an endless loop for an inexperienced user, considering the exponential growth of scientific corpora and the plethora of tools designed to mine PubMed(®) and related biological databases. Herein, we describe BioTextQuest(+), a web-based interactive knowledge exploration platform with significant advances to its predecessor (BioTextQuest), aiming to bridge processes such as bioentity recognition, functional annotation, document clustering and data integration towards literature mining and concept discovery. BioTextQuest(+) enables PubMed and OMIM querying, retrieval of abstracts related to a targeted request and optimal detection of genes, proteins, molecular functions, pathways and biological processes within the retrieved documents. The front-end interface facilitates the browsing of document clustering per subject, the analysis of term co-occurrence, the generation of tag clouds containing highly represented terms per cluster and at-a-glance popup windows with information about relevant genes and proteins. Moreover, to support experimental research, BioTextQuest(+) addresses integration of its primary functionality with biological repositories and software tools able to deliver further bioinformatics services. The Google-like interface extends beyond simple use by offering a range of advanced parameterization for expert users. We demonstrate the functionality of BioTextQuest(+) through several exemplary research scenarios including author disambiguation, functional term enrichment, knowledge acquisition and concept discovery linking major human diseases, such as obesity and ageing. AVAILABILITY The service is accessible at http://bioinformatics.med.uoc.gr/biotextquest. CONTACT g.pavlopoulos@gmail.com or georgios.pavlopoulos@esat.kuleuven.be SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Nikolas Papanikolaou
- Division of Basic Sciences, University of Crete, Medical School, Heraklion 71110, Greece, Institute of Marine Biology, Biotechnology and Aquaculture (IMBBC), Hellenic Centre for Marine Research (HCMR), Heraklion, Greece, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Campus Belval, 7, avenue des Hauts-Fourneaux, L-4362 Esch sur Alzette, Luxembourg, Biological Computation & Process Laboratory (BCPL), Chemical Process & Energy Resources Institute (CPERI), Centre for Research & Technology Hellas (CERTH), PO Box 361, GR-57001 Thessalonica, Greece, Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, 70013 Heraklion, Crete, Greece and Department of Biological Sciences, Bioinformatics Research Laboratory, University of Cyprus, PO Box 20537, CY 1678, Nicosia, Cyprus
| | - Georgios A Pavlopoulos
- Division of Basic Sciences, University of Crete, Medical School, Heraklion 71110, Greece, Institute of Marine Biology, Biotechnology and Aquaculture (IMBBC), Hellenic Centre for Marine Research (HCMR), Heraklion, Greece, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Campus Belval, 7, avenue des Hauts-Fourneaux, L-4362 Esch sur Alzette, Luxembourg, Biological Computation & Process Laboratory (BCPL), Chemical Process & Energy Resources Institute (CPERI), Centre for Research & Technology Hellas (CERTH), PO Box 361, GR-57001 Thessalonica, Greece, Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, 70013 Heraklion, Crete, Greece and Department of Biological Sciences, Bioinformatics Research Laboratory, University of Cyprus, PO Box 20537, CY 1678, Nicosia, Cyprus
| | - Evangelos Pafilis
- Division of Basic Sciences, University of Crete, Medical School, Heraklion 71110, Greece, Institute of Marine Biology, Biotechnology and Aquaculture (IMBBC), Hellenic Centre for Marine Research (HCMR), Heraklion, Greece, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Campus Belval, 7, avenue des Hauts-Fourneaux, L-4362 Esch sur Alzette, Luxembourg, Biological Computation & Process Laboratory (BCPL), Chemical Process & Energy Resources Institute (CPERI), Centre for Research & Technology Hellas (CERTH), PO Box 361, GR-57001 Thessalonica, Greece, Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, 70013 Heraklion, Crete, Greece and Department of Biological Sciences, Bioinformatics Research Laboratory, University of Cyprus, PO Box 20537, CY 1678, Nicosia, Cyprus
| | - Theodosios Theodosiou
- Division of Basic Sciences, University of Crete, Medical School, Heraklion 71110, Greece, Institute of Marine Biology, Biotechnology and Aquaculture (IMBBC), Hellenic Centre for Marine Research (HCMR), Heraklion, Greece, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Campus Belval, 7, avenue des Hauts-Fourneaux, L-4362 Esch sur Alzette, Luxembourg, Biological Computation & Process Laboratory (BCPL), Chemical Process & Energy Resources Institute (CPERI), Centre for Research & Technology Hellas (CERTH), PO Box 361, GR-57001 Thessalonica, Greece, Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, 70013 Heraklion, Crete, Greece and Department of Biological Sciences, Bioinformatics Research Laboratory, University of Cyprus, PO Box 20537, CY 1678, Nicosia, Cyprus
| | - Reinhard Schneider
- Division of Basic Sciences, University of Crete, Medical School, Heraklion 71110, Greece, Institute of Marine Biology, Biotechnology and Aquaculture (IMBBC), Hellenic Centre for Marine Research (HCMR), Heraklion, Greece, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Campus Belval, 7, avenue des Hauts-Fourneaux, L-4362 Esch sur Alzette, Luxembourg, Biological Computation & Process Laboratory (BCPL), Chemical Process & Energy Resources Institute (CPERI), Centre for Research & Technology Hellas (CERTH), PO Box 361, GR-57001 Thessalonica, Greece, Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, 70013 Heraklion, Crete, Greece and Department of Biological Sciences, Bioinformatics Research Laboratory, University of Cyprus, PO Box 20537, CY 1678, Nicosia, Cyprus
| | - Venkata P Satagopam
- Division of Basic Sciences, University of Crete, Medical School, Heraklion 71110, Greece, Institute of Marine Biology, Biotechnology and Aquaculture (IMBBC), Hellenic Centre for Marine Research (HCMR), Heraklion, Greece, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Campus Belval, 7, avenue des Hauts-Fourneaux, L-4362 Esch sur Alzette, Luxembourg, Biological Computation & Process Laboratory (BCPL), Chemical Process & Energy Resources Institute (CPERI), Centre for Research & Technology Hellas (CERTH), PO Box 361, GR-57001 Thessalonica, Greece, Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, 70013 Heraklion, Crete, Greece and Department of Biological Sciences, Bioinformatics Research Laboratory, University of Cyprus, PO Box 20537, CY 1678, Nicosia, Cyprus
| | - Christos A Ouzounis
- Division of Basic Sciences, University of Crete, Medical School, Heraklion 71110, Greece, Institute of Marine Biology, Biotechnology and Aquaculture (IMBBC), Hellenic Centre for Marine Research (HCMR), Heraklion, Greece, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Campus Belval, 7, avenue des Hauts-Fourneaux, L-4362 Esch sur Alzette, Luxembourg, Biological Computation & Process Laboratory (BCPL), Chemical Process & Energy Resources Institute (CPERI), Centre for Research & Technology Hellas (CERTH), PO Box 361, GR-57001 Thessalonica, Greece, Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, 70013 Heraklion, Crete, Greece and Department of Biological Sciences, Bioinformatics Research Laboratory, University of Cyprus, PO Box 20537, CY 1678, Nicosia, Cyprus Division of Basic Sciences, University of Crete, Medical School, Heraklion 71110, Greece, Institute of Marine Biology, Biotechnology and Aquaculture (IMBBC), Hellenic Centre for Marine Research (HCMR), Heraklion, Greece, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Campus Belval, 7, avenue des Hauts-Fourneaux, L-4362 Esch sur Alzette, Luxembourg, Biological Computation & Process Laboratory (BCPL), Chemical Process & Energy Resources Institute (CPERI), Centre for Research & Technology Hellas (CERTH), PO Box 361, GR-57001 Thessalonica, Greece, Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, 70013 Heraklion, Crete, Greece and Department of Biological Sciences, Bioinformatics Research Laboratory, University of Cyprus, PO Box 20537, CY 1678, Nicosia, Cyprus
| | - Aristides G Eliopoulos
- Division of Basic Sciences, University of Crete, Medical School, Heraklion 71110, Greece, Institute of Marine Biology, Biotechnology and Aquaculture (IMBBC), Hellenic Centre for Marine Research (HCMR), Heraklion, Greece, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Campus Belval, 7, avenue des Hauts-Fourneaux, L-4362 Esch sur Alzette, Luxembourg, Biological Computation & Process Laboratory (BCPL), Chemical Process & Energy Resources Institute (CPERI), Centre for Research & Technology Hellas (CERTH), PO Box 361, GR-57001 Thessalonica, Greece, Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, 70013 Heraklion, Crete, Greece and Department of Biological Sciences, Bioinformatics Research Laboratory, University of Cyprus, PO Box 20537, CY 1678, Nicosia, Cyprus Division of Basic Sciences, University of Crete, Medical School, Heraklion 71110, Greece, Institute of Marine Biology, Biotechnology and Aquaculture (IMBBC), Hellenic Centre for Marine Research (HCMR), Heraklion, Greece, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Campus Belval, 7, avenue des Hauts-Fourneaux, L-4362 Esch sur Alzette, Luxembourg, Biological Computation & Process Laboratory (BCPL), Chemical Process & Energy Resources Institute (CPERI), Centre for Research & Technology Hellas (CERTH), PO Box 361, GR-57001 Thessalonica, Greece, Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, 70013 Heraklion, Crete, Greece and Department of Biological Sciences, Bioinformatics Research Laboratory, University of Cyprus, PO Box 20537, CY 1678, Nicosia, Cyprus
| | - Vasilis J Promponas
- Division of Basic Sciences, University of Crete, Medical School, Heraklion 71110, Greece, Institute of Marine Biology, Biotechnology and Aquaculture (IMBBC), Hellenic Centre for Marine Research (HCMR), Heraklion, Greece, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Campus Belval, 7, avenue des Hauts-Fourneaux, L-4362 Esch sur Alzette, Luxembourg, Biological Computation & Process Laboratory (BCPL), Chemical Process & Energy Resources Institute (CPERI), Centre for Research & Technology Hellas (CERTH), PO Box 361, GR-57001 Thessalonica, Greece, Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, 70013 Heraklion, Crete, Greece and Department of Biological Sciences, Bioinformatics Research Laboratory, University of Cyprus, PO Box 20537, CY 1678, Nicosia, Cyprus
| | - Ioannis Iliopoulos
- Division of Basic Sciences, University of Crete, Medical School, Heraklion 71110, Greece, Institute of Marine Biology, Biotechnology and Aquaculture (IMBBC), Hellenic Centre for Marine Research (HCMR), Heraklion, Greece, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Campus Belval, 7, avenue des Hauts-Fourneaux, L-4362 Esch sur Alzette, Luxembourg, Biological Computation & Process Laboratory (BCPL), Chemical Process & Energy Resources Institute (CPERI), Centre for Research & Technology Hellas (CERTH), PO Box 361, GR-57001 Thessalonica, Greece, Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, 70013 Heraklion, Crete, Greece and Department of Biological Sciences, Bioinformatics Research Laboratory, University of Cyprus, PO Box 20537, CY 1678, Nicosia, Cyprus
| |
Collapse
|
48
|
Esguerra JLS, Eliasson L. Functional implications of long non-coding RNAs in the pancreatic islets of Langerhans. Front Genet 2014; 5:209. [PMID: 25071836 PMCID: PMC4083688 DOI: 10.3389/fgene.2014.00209] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 06/19/2014] [Indexed: 12/14/2022] Open
Abstract
Type-2 diabetes (T2D) is a complex disease characterized by insulin resistance in target tissues and impaired insulin release from pancreatic beta cells. As central tissue of glucose homeostasis, the pancreatic islet continues to be an important focus of research to understand the pathophysiology of the disease. The increased access to human pancreatic islets has resulted in improved knowledge of islet function, and together with advances in RNA sequencing and related technologies, revealed the transcriptional and epigenetic landscape of human islet cells. The discovery of thousands of long non-coding RNA (lncRNA) transcripts highly enriched in the pancreatic islet and/or specifically expressed in the beta-cells, points to yet another layer of gene regulation of many hitherto unknown mechanistic principles governing islet cell functions. Here we review fundamental islet physiology and propose functional implications of the lncRNAs in islet development and endocrine cell functions. We also take into account important differences between rodent and human islets in terms of morphology and function, and suggest how species-specific lncRNAs may partly influence gene regulation to define the unique phenotypic identity of an organism and the functions of its constituent cells. The implication of primate-specific lncRNAs will be far-reaching in all aspects of diabetes research, but most importantly in the identification and development of novel targets to improve pancreatic islet cell functions as a therapeutic approach to treat T2D.
Collapse
Affiliation(s)
- Jonathan L S Esguerra
- Islet Cell Exocytosis, Department of Clinical Sciences-Malmö, Lund University Diabetes Centre, Lund University Malmö, Sweden
| | - Lena Eliasson
- Islet Cell Exocytosis, Department of Clinical Sciences-Malmö, Lund University Diabetes Centre, Lund University Malmö, Sweden
| |
Collapse
|
49
|
Rodrigo-Domingo M, Waagepetersen R, Bødker JS, Falgreen S, Kjeldsen MK, Johnsen HE, Dybkær K, Bøgsted M. Reproducible probe-level analysis of the Affymetrix Exon 1.0 ST array with R/Bioconductor. Brief Bioinform 2014; 15:519-33. [PMID: 23603090 PMCID: PMC4103539 DOI: 10.1093/bib/bbt011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Accepted: 02/15/2013] [Indexed: 12/22/2022] Open
Abstract
The presence of different transcripts of a gene across samples can be analysed by whole-transcriptome microarrays. Reproducing results from published microarray data represents a challenge owing to the vast amounts of data and the large variety of preprocessing and filtering steps used before the actual analysis is carried out. To guarantee a firm basis for methodological development where results with new methods are compared with previous results, it is crucial to ensure that all analyses are completely reproducible for other researchers. We here give a detailed workflow on how to perform reproducible analysis of the GeneChip®Human Exon 1.0 ST Array at probe and probeset level solely in R/Bioconductor, choosing packages based on their simplicity of use. To exemplify the use of the proposed workflow, we analyse differential splicing and differential gene expression in a publicly available dataset using various statistical methods. We believe this study will provide other researchers with an easy way of accessing gene expression data at different annotation levels and with the sufficient details needed for developing their own tools for reproducible analysis of the GeneChip®Human Exon 1.0 ST Array.
Collapse
|
50
|
Identification of gene ontologies linked to prefrontal-hippocampal functional coupling in the human brain. Proc Natl Acad Sci U S A 2014; 111:9657-62. [PMID: 24979789 DOI: 10.1073/pnas.1404082111] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Functional interactions between the dorsolateral prefrontal cortex and hippocampus during working memory have been studied extensively as an intermediate phenotype for schizophrenia. Coupling abnormalities have been found in patients, their unaffected siblings, and carriers of common genetic variants associated with schizophrenia, but the global genetic architecture of this imaging phenotype is unclear. To achieve genome-wide hypothesis-free identification of genes and pathways associated with prefrontal-hippocampal interactions, we combined gene set enrichment analysis with whole-genome genotyping and functional magnetic resonance imaging data from 269 healthy German volunteers. We found significant enrichment of the synapse organization and biogenesis gene set. This gene set included known schizophrenia risk genes, such as neural cell adhesion molecule (NRCAM) and calcium channel, voltage-dependent, beta 2 subunit (CACNB2), as well as genes with well-defined roles in neurodevelopmental and plasticity processes that are dysfunctional in schizophrenia and have mechanistic links to prefrontal-hippocampal functional interactions. Our results demonstrate a readily generalizable approach that can be used to identify the neurogenetic basis of systems-level phenotypes. Moreover, our findings identify gene sets in which genetic variation may contribute to disease risk through altered prefrontal-hippocampal functional interactions and suggest a link to both ongoing and developmental synaptic plasticity.
Collapse
|