51
|
Mercatelli D, Scalambra L, Triboli L, Ray F, Giorgi FM. Gene regulatory network inference resources: A practical overview. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2019; 1863:194430. [PMID: 31678629 DOI: 10.1016/j.bbagrm.2019.194430] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/06/2019] [Accepted: 09/09/2019] [Indexed: 02/08/2023]
Abstract
Transcriptional regulation is a fundamental molecular mechanism involved in almost every aspect of life, from homeostasis to development, from metabolism to behavior, from reaction to stimuli to disease progression. In recent years, the concept of Gene Regulatory Networks (GRNs) has grown popular as an effective applied biology approach for describing the complex and highly dynamic set of transcriptional interactions, due to its easy-to-interpret features. Since cataloguing, predicting and understanding every GRN connection in all species and cellular contexts remains a great challenge for biology, researchers have developed numerous tools and methods to infer regulatory processes. In this review, we catalogue these methods in six major areas, based on the dominant underlying information leveraged to infer GRNs: Coexpression, Sequence Motifs, Chromatin Immunoprecipitation (ChIP), Orthology, Literature and Protein-Protein Interaction (PPI) specifically focused on transcriptional complexes. The methods described here cover a wide range of user-friendliness: from web tools that require no prior computational expertise to command line programs and algorithms for large scale GRN inferences. Each method for GRN inference described herein effectively illustrates a type of transcriptional relationship, with many methods being complementary to others. While a truly holistic approach for inferring and displaying GRNs remains one of the greatest challenges in the field of systems biology, we believe that the integration of multiple methods described herein provides an effective means with which experimental and computational biologists alike may obtain the most complete pictures of transcriptional relationships. This article is part of a Special Issue entitled: Transcriptional Profiles and Regulatory Gene Networks edited by Dr. Federico Manuel Giorgi and Dr. Shaun Mahony.
Collapse
Affiliation(s)
- Daniele Mercatelli
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Laura Scalambra
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Luca Triboli
- Centre for Integrative Biology (CIBIO), University of Trento, Italy
| | - Forest Ray
- Department of Systems Biology, Columbia University Medical Center, New York, NY, United States
| | - Federico M Giorgi
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy.
| |
Collapse
|
52
|
Inference of plant gene regulatory networks using data-driven methods: A practical overview. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2019; 1863:194447. [PMID: 31678628 DOI: 10.1016/j.bbagrm.2019.194447] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 10/08/2019] [Accepted: 10/31/2019] [Indexed: 11/20/2022]
Abstract
Transcriptional regulation is a complex and dynamic process that plays a vital role in plant growth and development. A key component in the regulation of genes is transcription factors (TFs), which coordinate the transcriptional control of gene activity. A gene regulatory network (GRN) is a collection of regulatory interactions between TFs and their target genes. The accurate delineation of GRNs offers a significant contribution to our understanding about how plant cells are organized and function, and how individual genes are regulated in various conditions, organs or cell types. During the past decade, important progress has been made in the identification of GRNs using experimental and computational approaches. However, a detailed overview of available platforms supporting the analysis of GRNs in plants is missing. Here, we review current databases, platforms and tools that perform data-driven analyses of gene regulation in Arabidopsis. The platforms are categorized into two sections, 1) promoter motif analysis tools that use motif mapping approaches to find TF motifs in the regulatory sequences of genes of interest and 2) network analysis tools that identify potential regulators for a set of input genes using a range of data types in order to generate GRNs. We discuss the diverse datasets integrated and highlight the strengths and caveats of different platforms. Finally, we shed light on the limitations of the above approaches and discuss future perspectives, including the need for integrative approaches to unravel complex GRNs in plants.
Collapse
|
53
|
Yamada Y, Sasaki S. A method for identifying allele-specific hydroxymethylation. Epigenetics 2019; 15:231-250. [PMID: 31533538 DOI: 10.1080/15592294.2019.1664228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
We previously identified sequence-dependent allele-specific methylation (sd-ASM) in adult human peripheral blood leukocytes, in which ASM occurs in cis depending on adjacent polymorphic sequences. A number of groups have identified sd-ASM sites in the human and mouse genomes, illustrating the prevalence of sd-ASM in mammalian genomes. In addition, sd-ASM can lead to sequence-dependent allele-specific expression of neighbouring genes. Imprinted genes also often exhibit parent-of-origin-dependent allele-specific methylation (pd-ASM), which causes parent-of-origin-dependent allele-specific expression. However, whether most of the already known sd-ASM and pd-ASM sites are methylated or hydroxymethylated remains unclear due to technical restrictions. Accordingly, a novel method that enables examination of allelic methylation and hydroxymethylation status and also overcomes the drawbacks of conventional methods is needed. Such a method could also be used to elucidate the mechanisms underlying polymorphism-associated inter-individual differences in disease susceptibility and the mechanism of genomic imprinting. Here, we developed a simple method to determine allelic hydroxymethylation status and identified novel sequence- and parent-of-origin-dependent allele-specific hydroxymethylation sites. Correlation analyses of TF binding sequences and methylation or hydroxymethylation between three mouse strains revealed the involvement of Pax5 in strain-specific methylation and hydroxymethylation in exon 7 of Pdgfrb.
Collapse
Affiliation(s)
- Yoichi Yamada
- Faculty of Electrical and Computer Engineering, Institute of Science and Engineering, Kanazawa University, Kanazawa, Japan
| | - Sho Sasaki
- Division of Electrical and Computer Engineering, Graduate School of Natural Science and Technology, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
54
|
Piekarowicz K, Bertrand AT, Azibani F, Beuvin M, Julien L, Machowska M, Bonne G, Rzepecki R. A Muscle Hybrid Promoter as a Novel Tool for Gene Therapy. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 15:157-169. [PMID: 31660418 PMCID: PMC6807297 DOI: 10.1016/j.omtm.2019.09.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 09/03/2019] [Indexed: 01/29/2023]
Abstract
Gene therapy is a promising strategy to cure rare diseases. The lack of regulatory sequences ensuring specific and robust expression in skeletal and cardiac muscle is a substantial limitation of gene therapy efficiency targeting the muscle tissue. Here we describe a novel muscle hybrid (MH) promoter that is highly active in both skeletal and cardiac muscle cells. It has an easily exchangeable modular structure, including an intronic module that highly enhances the expression of the gene driven by it. In cultured myoblasts, myotubes, and cardiomyocytes, the MH promoter gives relatively stable expression as well as higher activity and protein levels than the standard CMV and desmin gene promoters or the previously developed synthetic or CKM-based promoters. Combined with AAV2/9, the MH promoter also provides a high in vivo expression level in skeletal muscle and the heart after both intramuscular and systemic delivery. It is much more efficient than the desmin-encoding gene promoter, and it maintains the same specificity. This novel promoter has potential for gene therapy in muscle cells. It can provide stable transgene expression, ensuring high levels of therapeutic protein, and limited side effects because of its specificity. This constitutes an improvement in the efficiency of genetic disease therapy.
Collapse
Affiliation(s)
- Katarzyna Piekarowicz
- Laboratory of Nuclear Proteins, Faculty of Biotechnology, University of Wroclaw, Wroclaw 50-383, Poland
| | - Anne T Bertrand
- Sorbonne Université, INSERM UMRS974, Center of Research in Myology, Institute of Myology, Paris 75 651, France
| | - Feriel Azibani
- Sorbonne Université, INSERM UMRS974, Center of Research in Myology, Institute of Myology, Paris 75 651, France
| | - Maud Beuvin
- Sorbonne Université, INSERM UMRS974, Center of Research in Myology, Institute of Myology, Paris 75 651, France
| | - Laura Julien
- Sorbonne Université, INSERM UMRS974, Center of Research in Myology, Institute of Myology, Paris 75 651, France
| | - Magdalena Machowska
- Laboratory of Nuclear Proteins, Faculty of Biotechnology, University of Wroclaw, Wroclaw 50-383, Poland
| | - Gisèle Bonne
- Sorbonne Université, INSERM UMRS974, Center of Research in Myology, Institute of Myology, Paris 75 651, France
| | - Ryszard Rzepecki
- Laboratory of Nuclear Proteins, Faculty of Biotechnology, University of Wroclaw, Wroclaw 50-383, Poland
| |
Collapse
|
55
|
Li J, Xie Y, Zhang C, Wang J, Wu Y, Yang Y, Xie Y, Lv Z. A network-based analysis for mining the risk pathways in glioblastoma. Oncol Lett 2019; 18:2712-2717. [PMID: 31402957 PMCID: PMC6676740 DOI: 10.3892/ol.2019.10598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 06/13/2019] [Indexed: 11/05/2022] Open
Abstract
The most malignant type of brain tumour is glioblastoma multiforme (GBM). Patients with GBM often have a poor prognosis, as a result of incomplete or inaccurate diagnoses. Regulatory pathways have been demonstrated to serve important roles in complex human diseases. Therefore, deciphering these risk pathways may shed light on the molecular mechanisms underlying GBM progression. In the present study, differentially expressed genes and microRNAs (miRNAs) in a publicly available database were identified between normal and tumour samples. To determine the pathophysiology and molecular mechanisms underlying GBM, integrated network analysis was performed to mine GBM-specific risk pathways. Specifically, a GBM-specific regulatory network was constructed that integrated manually curated GBM-associated transcription and post-transcriptional data resources, including transcription factors and miRNAs. A total of 1,827 differentially expressed genes and 30 miRNAs were identified. The differentially expressed genes were significantly enriched in a number of immune response-associated functions. Based on the GBM-specific regulatory network, 15 risk regulatory pathways containing not only known regulators, but also potential novel targets that might be involved in tumourigenesis were identified. Network analysis provides a strategy for leveraging genomic data to identify potential oncogenic pathways and molecular targets for GBM.
Collapse
Affiliation(s)
- Jing Li
- Department of Hepatobiliary Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Yujie Xie
- Department of Rehabilitation Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Chi Zhang
- Department of Rehabilitation Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Jianxiong Wang
- Department of Rehabilitation Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Yong Wu
- Department of Neurology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Yuan Yang
- Department of Neurology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Yang Xie
- Department of Neurology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Zhiyu Lv
- Department of Neurology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
56
|
Wang G, Luo X, Wang J, Wan J, Xia S, Zhu H, Qian J, Wang Y. MeDReaders: a database for transcription factors that bind to methylated DNA. Nucleic Acids Res 2019; 46:D146-D151. [PMID: 29145608 PMCID: PMC5753207 DOI: 10.1093/nar/gkx1096] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 10/21/2017] [Indexed: 12/29/2022] Open
Abstract
Understanding the molecular principles governing interactions between transcription factors (TFs) and DNA targets is one of the main subjects for transcriptional regulation. Recently, emerging evidence demonstrated that some TFs could bind to DNA motifs containing highly methylated CpGs both in vitro and in vivo. Identification of such TFs and elucidation of their physiological roles now become an important stepping-stone toward understanding the mechanisms underlying the methylation-mediated biological processes, which have crucial implications for human disease and disease development. Hence, we constructed a database, named as MeDReaders, to collect information about methylated DNA binding activities. A total of 731 TFs, which could bind to methylated DNA sequences, were manually curated in human and mouse studies reported in the literature. In silico approaches were applied to predict methylated and unmethylated motifs of 292 TFs by integrating whole genome bisulfite sequencing (WGBS) and ChIP-Seq datasets in six human cell lines and one mouse cell line extracted from ENCODE and GEO database. MeDReaders database will provide a comprehensive resource for further studies and aid related experiment designs. The database implemented unified access for users to most TFs involved in such methylation-associated binding actives. The website is available at http://medreader.org/.
Collapse
Affiliation(s)
- Guohua Wang
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China.,The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ximei Luo
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Jianan Wang
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Jun Wan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Shuli Xia
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Heng Zhu
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jiang Qian
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yadong Wang
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| |
Collapse
|
57
|
Teixeira MC, Monteiro PT, Palma M, Costa C, Godinho CP, Pais P, Cavalheiro M, Antunes M, Lemos A, Pedreira T, Sá-Correia I. YEASTRACT: an upgraded database for the analysis of transcription regulatory networks in Saccharomyces cerevisiae. Nucleic Acids Res 2019; 46:D348-D353. [PMID: 29036684 PMCID: PMC5753369 DOI: 10.1093/nar/gkx842] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 09/18/2017] [Indexed: 01/15/2023] Open
Abstract
The YEAst Search for Transcriptional Regulators And Consensus Tracking (YEASTRACT—www.yeastract.com) information system has been, for 11 years, a key tool for the analysis and prediction of transcription regulatory associations at the gene and genomic levels in Saccharomyces cerevisiae. Since its last update in June 2017, YEASTRACT includes approximately 163000 regulatory associations between transcription factors (TF) and target genes in S. cerevisiae, based on more than 1600 bibliographic references; it also includes 247 specific DNA binding consensus recognized by 113 TFs. This release of the YEASTRACT database provides new visualization tools to visualize each regulatory network in an interactive fashion, enabling the user to select and observe subsets of the network such as: (i) considering only DNA binding evidence or both DNA binding and expression evidence; (ii) considering only either positive or negative regulatory associations; or (iii) considering only one set of related environmental conditions. A further tool to observe TF regulons is also offered, enabling a clear-cut understanding of the exact meaning of the available data. We believe that with this new version, YEASTRACT will improve its role as an open web resource instrumental for Yeast Biologists and Systems Biology researchers.
Collapse
Affiliation(s)
- Miguel C Teixeira
- Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal.,iBB-Institute for BioEngineering and Biosciences, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Pedro T Monteiro
- Department of Computer Science and Engineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal.,INESC-ID, SW Algorithms and Tools for Constraint Solving Group, R. Alves Redol 9, 1000-029 Lisbon, Portugal
| | - Margarida Palma
- Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal.,iBB-Institute for BioEngineering and Biosciences, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Catarina Costa
- Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal.,iBB-Institute for BioEngineering and Biosciences, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Cláudia P Godinho
- Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal.,iBB-Institute for BioEngineering and Biosciences, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Pedro Pais
- Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal.,iBB-Institute for BioEngineering and Biosciences, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Mafalda Cavalheiro
- Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal.,iBB-Institute for BioEngineering and Biosciences, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Miguel Antunes
- Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal.,iBB-Institute for BioEngineering and Biosciences, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Alexandre Lemos
- Department of Computer Science and Engineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal.,INESC-ID, SW Algorithms and Tools for Constraint Solving Group, R. Alves Redol 9, 1000-029 Lisbon, Portugal
| | - Tiago Pedreira
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, 2780-156 Oeiras, Portugal
| | - Isabel Sá-Correia
- Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal.,iBB-Institute for BioEngineering and Biosciences, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| |
Collapse
|
58
|
Stubbs BJ, Gopaulakrishnan S, Glass K, Pochet N, Everaert C, Raby B, Carey V. TFutils: Data structures for transcription factor bioinformatics. F1000Res 2019; 8:152. [PMID: 31297189 PMCID: PMC6600865 DOI: 10.12688/f1000research.17976.2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/08/2019] [Indexed: 01/16/2023] Open
Abstract
DNA transcription is intrinsically complex. Bioinformatic work with transcription factors (TFs) is complicated by a multiplicity of data resources and annotations. The Bioconductor package TFutils includes data structures and functions to enhance the precision and utility of integrative analyses that have components involving TFs. TFutils provides catalogs of human TFs from three reference sources (CISBP, HOCOMOCO, and GO), a catalog of TF targets derived from MSigDb, and multiple approaches to enumerating TF binding sites, including an interface to results of 690 ENCODE experiments. Aspects of integration of TF binding patterns and genome-wide association study results are explored in examples.
Collapse
Affiliation(s)
- Benjamin J Stubbs
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Shweta Gopaulakrishnan
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Kimberly Glass
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Nathalie Pochet
- Broad Institute, Cambridge, MA, 02142, USA.,Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Celine Everaert
- Broad Institute, Cambridge, MA, 02142, USA.,Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Benjamin Raby
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Pulmonary Genetics Center, Children's Hospital Boston, Boston, MA, 02115, USA
| | - Vincent Carey
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
59
|
Chen Y, Zhao G, Li N, Luo Z, Wang X, Gu J. Role of 4‑aminobutyrate aminotransferase (ABAT) and the lncRNA co‑expression network in the development of myelodysplastic syndrome. Oncol Rep 2019; 42:509-520. [PMID: 31173260 PMCID: PMC6610043 DOI: 10.3892/or.2019.7175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 05/20/2019] [Indexed: 12/17/2022] Open
Abstract
IncRNAs play an important role in the regulation of gene expression. The present study profiled differentially expressed lncRNAs (DELs) and mRNAs (DEMs) in myelodysplastic syndrome (MDS) to construct a 4-aminobutyrate aminotransferase (ABAT)-DEL-DEM co-expression network in MDS development using the Agilent human BeadChips and Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway and network analyses. Compared with controls, there were 543 DELs and 2,705 DEMs in MDS patients, among which 285 (52.5%) DELs were downregulated and 258 (47.5%) DELs were upregulated, whereas 1,521 (56.2%) DEMs were downregulated and 1,184 (43.70%) DEMs were upregulated in MDS patients. The ABAT-DEL-DEM co-expression network contained six DELs that were co-expressed with ABAT in MDS. The GO analysis revealed that the co-expression network mainly participated in response to organic cyclic compound, cell proliferation, cell part morphogenesis, regulation of cell proliferation and enzyme-linked receptor protein signaling pathways, while the KEGG database showed that the co-expression network was involved in various pathways, such as phagosome and metabolic pathways. Furthermore, the expression of a selected DEL (lncENST00000444102) and ABAT was shown to be significantly downregulated in MDS patients, and in SKM-1 and THP-1 cells. The selected lncENST00000444102 was then overexpressed and ABAT expression was knocked down in the MDS cell lines using lentiviral transfection. In addition, lncENST00000444102 overexpression reduced the viability and increased the apoptosis of MDS cells, ABAT expression was upregulated by lncENST00000444102.
Collapse
Affiliation(s)
- Yanzhen Chen
- WorldWide Medical Center, Huashan Hospital of Fudan University, Shanghai 200040, P.R. China
| | - Guangjie Zhao
- Department of Hematology, Huashan Hospital of Fudan University, Shanghai 200040, P.R. China
| | - Nianyi Li
- Department of Hematology, Huashan Hospital of Fudan University, Shanghai 200040, P.R. China
| | - Zhongguang Luo
- Department of Digestive Diseases, Huashan Hospital of Fudan University, Shanghai 200040, P.R. China
| | - Xiaoqin Wang
- Department of Hematology, Huashan Hospital of Fudan University, Shanghai 200040, P.R. China
| | - Jingwen Gu
- WorldWide Medical Center, Huashan Hospital of Fudan University, Shanghai 200040, P.R. China
| |
Collapse
|
60
|
The Transcription Factor Deaf1 Modulates Engrailed-1 Expression to Regulate Skin Appendage Fate. J Invest Dermatol 2019; 139:2378-2381.e4. [PMID: 31145909 DOI: 10.1016/j.jid.2019.05.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 04/29/2019] [Accepted: 05/10/2019] [Indexed: 12/23/2022]
|
61
|
Christensen AH, Andersen CB, Wassilew K, Svendsen JH, Bundgaard H, Brand SM, Schmitz B. Rare non-coding Desmoglein-2 variant contributes to Arrhythmogenic right ventricular cardiomyopathy. J Mol Cell Cardiol 2019; 131:164-170. [PMID: 31051180 DOI: 10.1016/j.yjmcc.2019.04.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 04/28/2019] [Indexed: 11/19/2022]
Abstract
Arrhythmogenic right ventricular cardiomyopathy (ARVC) has been linked to variants in the coding sequence of desmosomal genes. The potential contribution of non-coding desmoglein-2 (DSG2) variants for development of ARVC is undescribed. We sequenced 1450 base pairs upstream of ATG in the DSG2 gene in 65 unrelated patients diagnosed with ARVC (10 borderline cases). Identified variants was evaluated by cosegregation and allele population frequency analysis, in silico tools, immunohistological investigations of myocardial biopsies, gene reporter assays, electrophoretic mobility shift assays (EMSA), and chromatin immunoprecipitation. The genetic analysis identified one novel, rare heterozygous DSG2 upstream variant (-317G > A) in a genetically unexplained ARVC patient. The variant segregated with signs of disease, was absent in publicly available databases, and affected a predicted binding site for activating protein-1 (AP-1). Immunohistochemical analysis of a myocardial biopsy from the -317G > A patient showed a marked reduction in DSG2 protein levels compared to healthy controls. Luciferase reporter gene assays showed promoter activity of the identified DSG2 upstream region and a general reduction in transcriptional activity in the presence of the minor DSG2_A allele (p < .01). Moreover, the DSG2_A allele reduced DSG2 activation by TGF-beta1 and a protein kinase C pathway activator (PMA; all p < .001 vs. DSG2_G). EMSAs showed altered transcription factor binding in presence of the DSG2_A allele. Chromatin immunoprecipitation assays in wild type epithelial cells identified AP-1 components c-FOS and c-JUN at the -317 locus. In conclusion, the non-coding DSG2 promoter variant -317G > A reduces DSG2 transcription in vitro and reduced myocardial DSG2 protein levels were observed in vivo. Our data support a contribution of non-coding DSG2 variants to the pathogenesis of ARVC.
Collapse
Affiliation(s)
- Alex Hørby Christensen
- Department of Cardiology, The Heart Centre, Rigshospitalet, Copenhagen University Hospital, Denmark; Department of Cardiology, Herlev-Gentofte Hospital, Copenhagen University Hospital, Denmark.
| | - Claus B Andersen
- Department of Pathology, Rigshospitalet, Copenhagen University Hospital, Denmark
| | - Katharina Wassilew
- Department of Pathology, Rigshospitalet, Copenhagen University Hospital, Denmark
| | - Jesper Hastrup Svendsen
- Department of Cardiology, The Heart Centre, Rigshospitalet, Copenhagen University Hospital, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Henning Bundgaard
- Department of Cardiology, The Heart Centre, Rigshospitalet, Copenhagen University Hospital, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Stefan-Martin Brand
- Institute of Sports Medicine, Molecular Genetics of Cardiovascular Disease, University Hospital Münster, 48149 Münster, Germany
| | - Boris Schmitz
- Institute of Sports Medicine, Molecular Genetics of Cardiovascular Disease, University Hospital Münster, 48149 Münster, Germany
| |
Collapse
|
62
|
Falcon T, Pinheiro DG, Ferreira-Caliman MJ, Turatti ICC, de Abreu FCP, Galaschi-Teixeira JS, Martins JR, Elias-Neto M, Soares MPM, Laure MB, Figueiredo VLC, Lopes NP, Simões ZLP, Garófalo CA, Bitondi MMG. Exploring integument transcriptomes, cuticle ultrastructure, and cuticular hydrocarbons profiles in eusocial and solitary bee species displaying heterochronic adult cuticle maturation. PLoS One 2019; 14:e0213796. [PMID: 30870522 PMCID: PMC6417726 DOI: 10.1371/journal.pone.0213796] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 02/28/2019] [Indexed: 12/26/2022] Open
Abstract
Differences in the timing of exoskeleton melanization and sclerotization are evident when comparing eusocial and solitary bees. This cuticular maturation heterochrony may be associated with life style, considering that eusocial bees remain protected inside the nest for many days after emergence, while the solitary bees immediately start outside activities. To address this issue, we characterized gene expression using large-scale RNA sequencing (RNA-seq), and quantified cuticular hydrocarbon (CHC) through gas chromatography-mass spectrometry in comparative studies of the integument (cuticle plus its underlying epidermis) of two eusocial and a solitary bee species. In addition, we used transmission electron microscopy (TEM) for studying the developing cuticle of these and other three bee species also differing in life style. We found 13,200, 55,209 and 30,161 transcript types in the integument of the eusocial Apis mellifera and Frieseomelitta varia, and the solitary Centris analis, respectively. In general, structural cuticle proteins and chitin-related genes were upregulated in pharate-adults and newly-emerged bees whereas transcripts for odorant binding proteins, cytochrome P450 and antioxidant proteins were overrepresented in foragers. Consistent with our hypothesis, a distance correlation analysis based on the differentially expressed genes suggested delayed cuticle maturation in A. mellifera in comparison to the solitary bee. However, this was not confirmed in the comparison with F. varia. The expression profiles of 27 of 119 genes displaying functional attributes related to cuticle formation/differentiation were positively correlated between A. mellifera and F. varia, and negatively or non-correlated with C. analis, suggesting roles in cuticular maturation heterochrony. However, we also found transcript profiles positively correlated between each one of the eusocial species and C. analis. Gene co-expression networks greatly differed between the bee species, but we identified common gene interactions exclusively between the eusocial species. Except for F. varia, the TEM analysis is consistent with cuticle development timing adapted to the social or solitary life style. In support to our hypothesis, the absolute quantities of n-alkanes and unsaturated CHCs were significantly higher in foragers than in the earlier developmental phases of the eusocial bees, but did not discriminate newly-emerged from foragers in C. analis. By highlighting differences in integument gene expression, cuticle ultrastructure, and CHC profiles between eusocial and solitary bees, our data provided insights into the process of heterochronic cuticle maturation associated to the way of life.
Collapse
Affiliation(s)
- Tiago Falcon
- Departamento de Genética, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
- Núcleo de Bioinformática, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Daniel G. Pinheiro
- Departamento de Tecnologia, Faculdade de Ciências Agrárias e Veterinárias, Universidade Estadual Paulista “Júlio de Mesquita Filho”, Jaboticabal, Brazil
| | - Maria Juliana Ferreira-Caliman
- Departamento de Biologia, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Izabel C. C. Turatti
- Departamento de Física e Química, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Fabiano C. Pinto de Abreu
- Departamento de Genética, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Juliana S. Galaschi-Teixeira
- Departamento de Biologia, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Juliana R. Martins
- Departamento de Genética, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Moysés Elias-Neto
- Departamento de Biologia, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Michelle P. M. Soares
- Departamento de Genética, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Marcela B. Laure
- Departamento de Genética, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Vera L. C. Figueiredo
- Departamento de Biologia, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Norberto Peporine Lopes
- Departamento de Física e Química, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Zilá L. P. Simões
- Departamento de Biologia, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Carlos A. Garófalo
- Departamento de Biologia, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Márcia M. G. Bitondi
- Departamento de Biologia, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
63
|
Cavalli M, Baltzer N, Umer HM, Grau J, Lemnian I, Pan G, Wallerman O, Spalinskas R, Sahlén P, Grosse I, Komorowski J, Wadelius C. Allele specific chromatin signals, 3D interactions, and motif predictions for immune and B cell related diseases. Sci Rep 2019; 9:2695. [PMID: 30804403 PMCID: PMC6389883 DOI: 10.1038/s41598-019-39633-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 01/24/2019] [Indexed: 12/20/2022] Open
Abstract
Several Genome Wide Association Studies (GWAS) have reported variants associated to immune diseases. However, the identified variants are rarely the drivers of the associations and the molecular mechanisms behind the genetic contributions remain poorly understood. ChIP-seq data for TFs and histone modifications provide snapshots of protein-DNA interactions allowing the identification of heterozygous SNPs showing significant allele specific signals (AS-SNPs). AS-SNPs can change a TF binding site resulting in altered gene regulation and are primary candidates to explain associations observed in GWAS and expression studies. We identified 17,293 unique AS-SNPs across 7 lymphoblastoid cell lines. In this set of cell lines we interrogated 85% of common genetic variants in the population for potential regulatory effect and we identified 237 AS-SNPs associated to immune GWAS traits and 714 to gene expression in B cells. To elucidate possible regulatory mechanisms we integrated long-range 3D interactions data to identify putative target genes and motif predictions to identify TFs whose binding may be affected by AS-SNPs yielding a collection of 173 AS-SNPs associated to gene expression and 60 to B cell related traits. We present a systems strategy to find functional gene regulatory variants, the TFs that bind differentially between alleles and novel strategies to detect the regulated genes.
Collapse
Affiliation(s)
- Marco Cavalli
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Nicholas Baltzer
- Department of Cell and Molecular Biology, Computational Biology and Bioinformatics, Uppsala University, Uppsala, Sweden
| | - Husen M Umer
- Department of Cell and Molecular Biology, Computational Biology and Bioinformatics, Uppsala University, Uppsala, Sweden
| | - Jan Grau
- Institute of Computer Science, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Ioana Lemnian
- Institute of Computer Science, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Gang Pan
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Ola Wallerman
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Rapolas Spalinskas
- Science for Life Laboratory, Division of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Pelin Sahlén
- Science for Life Laboratory, Division of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Ivo Grosse
- Institute of Computer Science, Martin Luther University Halle-Wittenberg, Halle, Germany.,German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig, Leipzig, Germany
| | - Jan Komorowski
- Department of Cell and Molecular Biology, Computational Biology and Bioinformatics, Uppsala University, Uppsala, Sweden.,Institute of Computer Science, Polish Academy of Sciences, Warszawa, Poland
| | - Claes Wadelius
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
64
|
Khatri I, Ganguly K, Sharma S, Carmicheal J, Kaur S, Batra SK, Bhasin MK. Systems Biology Approach to Identify Novel Genomic Determinants for Pancreatic Cancer Pathogenesis. Sci Rep 2019; 9:123. [PMID: 30644396 PMCID: PMC6333820 DOI: 10.1038/s41598-018-36328-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 11/05/2018] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy with a 5-year survival rate of <8%. Its dismal prognosis stems from inefficient therapeutic modalities owing to the lack of understanding about pancreatic cancer pathogenesis. Considering the molecular complexity and heterogeneity of PDAC, identification of novel molecular contributors involved in PDAC onset and progression using global "omics" analysis will pave the way to improved strategies for disease prevention and therapeutic targeting. Meta-analysis of multiple miRNA microarray datasets containing healthy controls (HC), chronic pancreatitis (CP) and PDAC cases, identified 13 miRNAs involved in the progression of PDAC. These miRNAs showed dysregulation in both tissue as well as blood samples, along with progressive decrease in expression from HC to CP to PDAC. Gene-miRNA interaction analysis further elucidated 5 miRNAs (29a/b, 27a, 130b and 148a) that are significantly downregulated in conjunction with concomitant upregulation of their target genes throughout PDAC progression. Among these, miRNA-29a/b targeted genes were found to be most significantly altered in comparative profiling of HC, CP and PDAC, indicating its involvement in malignant evolution. Further, pathway analysis suggested direct involvement of miRNA-29a/b in downregulating the key pathways associated with PDAC development and metastasis including focal adhesion signaling and extracellular matrix organization. Our systems biology data analysis, in combination with real-time PCR validation indicates direct functional involvement of miRNA-29a in PDAC progression and is a potential prognostic marker and therapeutic candidate for patients with progressive disease.
Collapse
Affiliation(s)
- Indu Khatri
- BIDMC Genomics, Proteomics, Bioinformatics and Systems Biology Center, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Koelina Ganguly
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Sunandini Sharma
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Joseph Carmicheal
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Sukhwinder Kaur
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA.
| | - Manoj K Bhasin
- BIDMC Genomics, Proteomics, Bioinformatics and Systems Biology Center, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| |
Collapse
|
65
|
Ma S, Jiang T, Jiang R. Constructing tissue-specific transcriptional regulatory networks via a Markov random field. BMC Genomics 2018; 19:884. [PMID: 30598101 PMCID: PMC6311931 DOI: 10.1186/s12864-018-5277-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Recent advances in sequencing technologies have enabled parallel assays of chromatin accessibility and gene expression for major human cell lines. Such innovation provides a great opportunity to decode phenotypic consequences of genetic variation via the construction of predictive gene regulatory network models. However, there still lacks a computational method to systematically integrate chromatin accessibility information with gene expression data to recover complicated regulatory relationships between genes in a tissue-specific manner. RESULTS We propose a Markov random field (MRF) model for constructing tissue-specific transcriptional regulatory networks via integrative analysis of DNase-seq and RNA-seq data. Our method, named CSNets (cell-line specific regulatory networks), first infers regulatory networks for individual cell lines using chromatin accessibility information, and then fine-tunes these networks using the MRF based on pairwise similarity between cell lines derived from gene expression data. Using this method, we constructed regulatory networks specific to 110 human cell lines and 13 major tissues with the use of ENCODE data. We demonstrated the high quality of these networks via comprehensive statistical analysis based on ChIP-seq profiles, functional annotations, taxonomic analysis, and literature surveys. We further applied these networks to analyze GWAS data of Crohn's disease and prostate cancer. Results were either consistent with the literature or provided biological insights into regulatory mechanisms of these two complex diseases. The website of CSNets is freely available at http://bioinfo.au.tsinghua.edu.cn/jianglab/CSNETS/ . CONCLUSIONS CSNets demonstrated the power of joint analysis on epigenomic and transcriptomic data towards the accurate construction of gene regulatory network. Our work provides not only a useful resource of regulatory networks to the community, but also valuable experiences in methodology development for multi-omics data integration.
Collapse
Affiliation(s)
- Shining Ma
- Department of Statistics, Department of Biomedical Data Science, Bio-X Program Stanford University, Stanford, CA 94305 USA
| | - Tao Jiang
- Ministry of Education Key Laboratory of Bioinformatics; Bioinformatics Division, Beijing National Research Center for Information Science and Technology; Department of Automation, Tsinghua University, Beijing, 100084 China
- Department of Computer Science and Engineering, University of California, Riverside, CA 92521 USA
| | - Rui Jiang
- Ministry of Education Key Laboratory of Bioinformatics; Bioinformatics Division, Beijing National Research Center for Information Science and Technology; Department of Automation, Tsinghua University, Beijing, 100084 China
| |
Collapse
|
66
|
Dey KK, Xie D, Stephens M. A new sequence logo plot to highlight enrichment and depletion. BMC Bioinformatics 2018; 19:473. [PMID: 30526486 PMCID: PMC6288878 DOI: 10.1186/s12859-018-2489-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 11/12/2018] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Sequence logo plots have become a standard graphical tool for visualizing sequence motifs in DNA, RNA or protein sequences. However standard logo plots primarily highlight enrichment of symbols, and may fail to highlight interesting depletions. Current alternatives that try to highlight depletion often produce visually cluttered logos. RESULTS We introduce a new sequence logo plot, the EDLogo plot, that highlights both enrichment and depletion, while minimizing visual clutter. We provide an easy-to-use and highly customizable R package Logolas to produce a range of logo plots, including EDLogo plots. This software also allows elements in the logo plot to be strings of characters, rather than a single character, extending the range of applications beyond the usual DNA, RNA or protein sequences. And the software includes new Empirical Bayes methods to stabilize estimates of enrichment and depletion, and thus better highlight the most significant patterns in data. We illustrate our methods and software on applications to transcription factor binding site motifs, protein sequence alignments and cancer mutation signature profiles. CONCLUSIONS Our new EDLogo plots and flexible software implementation can help data analysts visualize both enrichment and depletion of characters (DNA sequence bases, amino acids, etc.) across a wide range of applications.
Collapse
Affiliation(s)
- Kushal K Dey
- Department of Statistics, University of Chicago, Chicago, 60637, USA.
| | - Dongyue Xie
- Department of Statistics, University of Chicago, Chicago, 60637, USA
| | - Matthew Stephens
- Department of Statistics, University of Chicago, Chicago, 60637, USA.,Department of Human Genetics, university of Chicago, Chicago, 60637, USA
| |
Collapse
|
67
|
Schlotterer A, Kolibabka M, Lin J, Acunman K, Dietrich N, Sticht C, Fleming T, Nawroth P, Hammes HP. Methylglyoxal induces retinopathy-type lesions in the absence of hyperglycemia: studies in a rat model. FASEB J 2018; 33:4141-4153. [PMID: 30485119 DOI: 10.1096/fj.201801146rr] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The aim of this study was to evaluate whether damage to the neurovascular unit in diabetes depends on reactive metabolites such as methylglyoxal (MG), and to assess its impact on retinal gene expression. Male Wistar rats were supplied with MG (50 mM) by drinking water and compared with age-matched streptozotocin-diabetic animals and untreated controls. Retinal damage was evaluated for the accumulation of MG-derived advanced glycation end products, changes in hexosamine and PKC pathway activation, microglial activation, vascular alterations (pericyte loss and vasoregression), neuroretinal function assessed by electroretinogram, and neurodegeneration. Retinal gene regulation was studied by microarray analysis, and transcription factor involvement was identified by upstream regulator analysis. Systemic application of MG by drinking water increased retinal MG to levels comparable with diabetic animals. Elevated retinal MG resulted in MG-derived hydroimidazolone modifications in the ganglion cell layer, inner nuclear layer, and outer nuclear layer, a moderate activation of the hexosamine pathway, a pan-retinal activation of microglia, loss of pericytes, increased formation of acellular capillaries, decreased function of bipolar cells, and increased expression of the crystallin gene family. MG mimics important aspects of diabetic retinopathy and plays a pathogenic role in microglial activation, vascular damage, and neuroretinal dysfunction. In response to MG, the retina induces expression of neuroprotective crystallins.-Schlotterer, A., Kolibabka, M., Lin, J., Acunman, K., Dietrich, N., Sticht, C., Fleming, T., Nawroth, P., Hammes, H.-P. Methylglyoxal induces retinopathy-type lesions in the absence of hyperglycemia: studies in a rat model.
Collapse
Affiliation(s)
- Andrea Schlotterer
- Fifth Medical Department, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Matthias Kolibabka
- Fifth Medical Department, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Jihong Lin
- Fifth Medical Department, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Kübra Acunman
- Fifth Medical Department, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Nadine Dietrich
- Fifth Medical Department, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Carsten Sticht
- Medical Research Center, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany; and
| | - Thomas Fleming
- Department of Medicine I and Clinical Chemistry, Heidelberg University, Heidelberg, Germany
| | - Peter Nawroth
- Department of Medicine I and Clinical Chemistry, Heidelberg University, Heidelberg, Germany
| | - Hans-Peter Hammes
- Fifth Medical Department, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
68
|
Khan FM, Gupta SK, Wolkenhauer O. Integrative workflows for network analysis. Essays Biochem 2018; 62:549-561. [PMID: 30366988 DOI: 10.1042/ebc20180005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Revised: 09/10/2018] [Accepted: 09/14/2018] [Indexed: 01/03/2025]
Abstract
Due to genetic heterogeneity across patients, the identification of effective disease signatures and therapeutic targets is challenging. Addressing this challenge, we have previously developed a network-based approach, which integrates heterogeneous sources of biological information to identify disease specific core-regulatory networks. In particular, our workflow uses a multi-objective optimization function to calculate a ranking score for network components (e.g. feedback/feedforward loops) based on network properties, biomedical and high-throughput expression data. High ranked network components are merged to identify the core-regulatory network(s) that is then subjected to dynamical analysis using stimulus-response and in silico perturbation experiments for the identification of disease gene signatures and therapeutic targets. In a case study, we implemented our workflow to identify bladder and breast cancer specific core-regulatory networks underlying epithelial-mesenchymal transition from the E2F1 molecular interaction map.In this study, we review our workflow and described how it has developed over time to understand the mechanisms underlying disease progression and prediction of signatures for clinical decision making.
Collapse
Affiliation(s)
- Faiz M Khan
- Department of Systems Biology and Bioinformatics, University of Rostock, Rostock, Germany
| | - Shailendra K Gupta
- Department of Systems Biology and Bioinformatics, University of Rostock, Rostock, Germany
- Chhattisgarh Swami Vivekanand Technical University, Bhilai, Chhattisgarh, India
| | - Olaf Wolkenhauer
- Department of Systems Biology and Bioinformatics, University of Rostock, Rostock, Germany
- Chhattisgarh Swami Vivekanand Technical University, Bhilai, Chhattisgarh, India
- Stellenbosch Institute of Advanced Study (STIAS), Wallenberg Research Centre at Stellenbosch University, South Africa
| |
Collapse
|
69
|
de Souza MM, Zerlotini A, Geistlinger L, Tizioto PC, Taylor JF, Rocha MIP, Diniz WJS, Coutinho LL, Regitano LCA. A comprehensive manually-curated compendium of bovine transcription factors. Sci Rep 2018; 8:13747. [PMID: 30213987 PMCID: PMC6137171 DOI: 10.1038/s41598-018-32146-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 08/29/2018] [Indexed: 01/28/2023] Open
Abstract
Transcription factors (TFs) are pivotal regulatory proteins that control gene expression in a context-dependent and tissue-specific manner. In contrast to human, where comprehensive curated TF collections exist, bovine TFs are only rudimentary recorded and characterized. In this article, we present a manually-curated compendium of 865 sequence-specific DNA-binding bovines TFs, which we analyzed for domain family distribution, evolutionary conservation, and tissue-specific expression. In addition, we provide a list of putative transcription cofactors derived from known interactions with the identified TFs. Since there is a general lack of knowledge concerning the regulation of gene expression in cattle, the curated list of TF should provide a basis for an improved comprehension of regulatory mechanisms that are specific to the species.
Collapse
Affiliation(s)
- Marcela M de Souza
- Post-graduation Program of Evolutionary Genetics and Molecular Biology, Federal University of São Carlos, São Carlos, São Paulo, 13560-970, Brazil.,Animal Biotechnology, Embrapa Pecuária Sudeste, São Carlos, São Paulo, 13560-970, Brazil
| | - Adhemar Zerlotini
- Bioinformatic Multi-user Laboratory, Embrapa Informática Agropecuária, Campinas, São Paulo, 70770-901, Brazil
| | - Ludwig Geistlinger
- Animal Biotechnology, Embrapa Pecuária Sudeste, São Carlos, São Paulo, 13560-970, Brazil
| | | | - Jeremy F Taylor
- Division of Animal Science, University of Missouri, Columbia, Missouri, 65211-5300, USA
| | - Marina I P Rocha
- Post-graduation Program of Evolutionary Genetics and Molecular Biology, Federal University of São Carlos, São Carlos, São Paulo, 13560-970, Brazil
| | - Wellison J S Diniz
- Post-graduation Program of Evolutionary Genetics and Molecular Biology, Federal University of São Carlos, São Carlos, São Paulo, 13560-970, Brazil
| | - Luiz L Coutinho
- Functional Genomic Center, University of São Paulo, Piracicaba, São Paulo, 13418-900, Brazil
| | - Luciana C A Regitano
- Animal Biotechnology, Embrapa Pecuária Sudeste, São Carlos, São Paulo, 13560-970, Brazil.
| |
Collapse
|
70
|
Wang K, Ma C, Xing C, Chen CL, Chen Z, Yao Y, Wang J, Tao C. Burkitt lymphoma-associated network construction and important network motif analysis. Oncol Lett 2018; 16:3054-3062. [PMID: 30127896 PMCID: PMC6096059 DOI: 10.3892/ol.2018.9010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 05/11/2018] [Indexed: 12/13/2022] Open
Abstract
Biological and medical researchers have discovered numerous transcription factors (TFs), microRNAs (miRNAs) and genes associated with Burkitt lymphoma (BL) through individual experiments; however, their regulatory mechanisms remain unclear. In the present study, BL-dysregulated and BL-associated networks were constructed to investigate these mechanisms. All data and regulatory associations were from known data resources and literature. The dysregulated network consisted of dysregulated TFs, miRNAs and genes, and partially determined the pathogenesis mechanisms underlying BL. The BL-associated network consisted of BL-associated TFs, miRNAs and genes. It has been indicated that the network motif consisted of TFs, miRNAs and genes serve potential functions in numerous biological processes within cancer. Two of the most studied network motifs are feedback loop (FBL) and feed-forward loop (FFL). The important network motifs were extracted, including the FBL motif, 3-nodes FFL motif and 4-nodes motif, from BL-dysregulated and BL-associated networks, and 10 types of motifs were identified from BL-associated network. Finally, 26/31 FBL motifs, 45/75 3-nodes FFL motifs and 54/94 4-nodes motifs were obtained from the dysregulated/associated networks. A total of four TFs (E2F1, NFKB1, E2F4 and TCF3) exhibit complicated regulation associations in BL-associated networks. The biological network does not demonstrate the dysregulated status for healthy people. When the individual becomes unwell, their biological network exhibits a dysregulated status. If the dysregulated status is regulated to a normal status by a number of medical methods, the diseases may be treated successfully. BL-dysregulated networks serve important roles in pathogenesis mechanisms underlying BL regulation of the dysregulated network, which may be an effective strategy that contributes to gene therapy for BL.
Collapse
Affiliation(s)
- Kunhao Wang
- School of Information Engineering, Changchun University of Science and Technology, Changchun, Jilin 130600, P.R. China
| | - Chao Ma
- College of Digital Media, Shenzhen Institute of Information Technology, Shenzhen, Guangdong 518172, P.R. China
| | - Chong Xing
- Department of Information and Technology, Changchun Finance College, Changchun, Jilin 130028, P.R. China
| | - Chin-Ling Chen
- Department of Computer Science and Information Engineering, Chaoyang University of Technology, Taichung 41349, Taiwan, R.O.C
| | - Zhigang Chen
- School of Information Engineering, Changchun University of Science and Technology, Changchun, Jilin 130600, P.R. China
| | - Yuxia Yao
- School of Information Engineering, Changchun University of Science and Technology, Changchun, Jilin 130600, P.R. China
| | - Jianan Wang
- School of Information Engineering, Changchun University of Science and Technology, Changchun, Jilin 130600, P.R. China
| | - Chunyu Tao
- School of Information Engineering, Changchun University of Science and Technology, Changchun, Jilin 130600, P.R. China
| |
Collapse
|
71
|
Serrano-Candelas E, Ainsua-Enrich E, Navinés-Ferrer A, Rodrigues P, García-Valverde A, Bazzocco S, Macaya I, Arribas J, Serrano C, Sayós J, Arango D, Martin M. Silencing of adaptor protein SH3BP2 reduces KIT/PDGFRA receptors expression and impairs gastrointestinal stromal tumors growth. Mol Oncol 2018; 12:1383-1397. [PMID: 29885053 PMCID: PMC6068349 DOI: 10.1002/1878-0261.12332] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 05/18/2018] [Accepted: 05/28/2018] [Indexed: 12/13/2022] Open
Abstract
Gastrointestinal stromal tumors (GISTs) represent about 80% of the mesenchymal neoplasms of the gastrointestinal tract. Most GISTs contain oncogenic KIT (85%) or PDGFRA (5%) receptors. The kinase inhibitor imatinib mesylate is the preferential treatment for these tumors; however, the development of drug resistance has highlighted the need for novel therapeutic strategies. Recently, we reported that the adaptor molecule SH3 Binding Protein 2 (SH3BP2) regulates KIT expression and signaling in human mast cells. Our current study shows that SH3BP2 is expressed in primary tumors and cell lines from GIST patients and that SH3BP2 silencing leads to a downregulation of oncogenic KIT and PDGFRA expression and an increase in apoptosis in imatinib-sensitive and imatinib-resistant GIST cells. The microphthalmia-associated transcription factor (MITF), involved in KIT expression in mast cells and melanocytes, is expressed in GISTs. Interestingly, MITF is reduced after SH3BP2 silencing. Importantly, reconstitution of both SH3BP2 and MITF restores cell viability. Furthermore, SH3BP2 silencing significantly reduces cell migration and tumor growth of imatinib-sensitive and imatinib-resistant cells in vivo. Altogether, SH3BP2 regulates KIT and PDGFRA expression and cell viability, indicating a role as a potential target in imatinib-sensitive and imatinib-resistant GISTs.
Collapse
Affiliation(s)
- Eva Serrano-Candelas
- Biochemistry Unit, Biomedicine Department, Faculty of Medicine, University of Barcelona, Spain.,Laboratory of Clinic and Experimental Immunoallergy, IDIBAPS, Barcelona, Spain
| | - Erola Ainsua-Enrich
- Biochemistry Unit, Biomedicine Department, Faculty of Medicine, University of Barcelona, Spain.,Laboratory of Clinic and Experimental Immunoallergy, IDIBAPS, Barcelona, Spain
| | - Arnau Navinés-Ferrer
- Biochemistry Unit, Biomedicine Department, Faculty of Medicine, University of Barcelona, Spain.,Laboratory of Clinic and Experimental Immunoallergy, IDIBAPS, Barcelona, Spain
| | - Paulo Rodrigues
- Group of Biomedical Research in Digestive Tract Tumors, CIBBIM-Nanomedicine, Vall d'Hebron University Hospital, Research Institute (VHIR), Autonomous University of Barcelona, Spain
| | | | - Sarah Bazzocco
- Group of Biomedical Research in Digestive Tract Tumors, CIBBIM-Nanomedicine, Vall d'Hebron University Hospital, Research Institute (VHIR), Autonomous University of Barcelona, Spain
| | - Irati Macaya
- Group of Biomedical Research in Digestive Tract Tumors, CIBBIM-Nanomedicine, Vall d'Hebron University Hospital, Research Institute (VHIR), Autonomous University of Barcelona, Spain
| | - Joaquín Arribas
- Preclinical Research Program, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.,The Catalan Institute of Research and Advanced Studies (ICREA), Barcelona, Spain.,CIBERONC, Barcelona, Spain.,Department of Biochemistry and Molecular Biology, Universitat Autónoma de Barcelona, Bellaterra, Spain
| | - César Serrano
- Preclinical Research Program, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.,Vall d'Hebron University Hospital, Barcelona, Spain
| | - Joan Sayós
- Immune Regulation and Immunotherapy Group, CIBBIM-Nanomedicine, Vall d'Hebron University Hospital, Research Institute (VHIR), Autonomous University of Barcelona, Spain
| | - Diego Arango
- Group of Biomedical Research in Digestive Tract Tumors, CIBBIM-Nanomedicine, Vall d'Hebron University Hospital, Research Institute (VHIR), Autonomous University of Barcelona, Spain
| | - Margarita Martin
- Biochemistry Unit, Biomedicine Department, Faculty of Medicine, University of Barcelona, Spain.,Laboratory of Clinic and Experimental Immunoallergy, IDIBAPS, Barcelona, Spain
| |
Collapse
|
72
|
Rodriguez N, Maili L, Chiquet BT, Blanton SH, Hecht JT, Letra A. BRCA1 and BRCA2 gene variants and nonsyndromic cleft lip/palate. Birth Defects Res 2018; 110:1043-1048. [PMID: 29921024 DOI: 10.1002/bdr2.1346] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 04/10/2018] [Accepted: 04/16/2018] [Indexed: 01/12/2023]
Abstract
BACKGROUND Nonsyndromic cleft lip with or without cleft palate (NSCL/P) is a debilitating condition that not only affects the individual, but the entire family. The purpose of this study was to investigate the association of BRCA1 and BRCA2 genes with NSCL/P. METHODS Twelve polymorphisms in/nearby BRCA1 and BRCA2 were genotyped using Taqman chemistry. Our data set consisted of 3,473 individuals including 2,191 nonHispanic white (NHW) individuals (from 151 multiplex and 348 simplex families) and 1,282 Hispanic individuals (from 92 multiplex and 216 simplex families). Data analysis was performed using Family-Based Association Test (FBAT), stratified by ethnicity and family history of NSCL/P. RESULTS Nominal associations were found between NSCL/P and BRCA1 in Hispanics and BRCA2 in NHW and Hispanics (p < .05). Significant haplotype associations were found between NSCL/P and both BRCA1 and BRCA2 (p ≤ .004). CONCLUSIONS Our results suggest a modest association between BRCA1 and BRCA2 and NSCL/P. Further studies in additional populations and functional studies are needed to elucidate the role of these genes in developmental processes and signaling pathways contributing to NSCL/P.
Collapse
Affiliation(s)
- Nicholas Rodriguez
- Center for Craniofacial Research, UTHealth School of Dentistry at Houston, Houston TX, USA
| | - Lorena Maili
- Department of Pediatrics, UTHealth McGovern Medical School, Houston TX, USA.,Pediatric Research Center, UTHealth McGovern Medical School, Houston TX, USA
| | - Brett T Chiquet
- Center for Craniofacial Research, UTHealth School of Dentistry at Houston, Houston TX, USA.,Department of Pediatric Dentistry, UTHealth School of Dentistry at Houston, Houston TX, USA.,Pediatric Research Center, UTHealth McGovern Medical School, Houston TX, USA
| | - Susan H Blanton
- Department of Human Genetics and John P. Hussman Institute of Human Genomics, University of Miami Miller School of Medicine, Miami FL, USA
| | - Jacqueline T Hecht
- Center for Craniofacial Research, UTHealth School of Dentistry at Houston, Houston TX, USA.,Department of Pediatrics, UTHealth McGovern Medical School, Houston TX, USA.,Pediatric Research Center, UTHealth McGovern Medical School, Houston TX, USA
| | - Ariadne Letra
- Center for Craniofacial Research, UTHealth School of Dentistry at Houston, Houston TX, USA.,Department of Diagnostic and Biomedical Sciences, UTHealth School of Dentistry at Houston, Houston TX, USA.,Pediatric Research Center, UTHealth McGovern Medical School, Houston TX, USA
| |
Collapse
|
73
|
Romero-Garmendia I, Garcia-Etxebarria K, Hernandez-Vargas H, Santin I, Jauregi-Miguel A, Plaza-Izurieta L, Cros MP, Legarda M, Irastorza I, Herceg Z, Fernandez-Jimenez N, Bilbao JR. Transcription Factor Binding Site Enrichment Analysis in Co-Expression Modules in Celiac Disease. Genes (Basel) 2018; 9:E245. [PMID: 29748492 PMCID: PMC5977185 DOI: 10.3390/genes9050245] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 05/07/2018] [Accepted: 05/08/2018] [Indexed: 12/12/2022] Open
Abstract
The aim of this study was to construct celiac co-expression patterns at a whole genome level and to identify transcription factors (TFs) that could drive the gliadin-related changes in coordination of gene expression observed in celiac disease (CD). Differential co-expression modules were identified in the acute and chronic responses to gliadin using expression data from a previous microarray study in duodenal biopsies. Transcription factor binding site (TFBS) and Gene Ontology (GO) annotation enrichment analyses were performed in differentially co-expressed genes (DCGs) and selection of candidate regulators was performed. Expression of candidates was measured in clinical samples and the activation of the TFs was further characterized in C2BBe1 cells upon gliadin challenge. Enrichment analyses of the DCGs identified 10 TFs and five were selected for further investigation. Expression changes related to active CD were detected in four TFs, as well as in several of their in silico predicted targets. The activation of TFs was further characterized in C2BBe1 cells upon gliadin challenge, and an increase in nuclear translocation of CAMP Responsive Element Binding Protein 1 (CREB1) and IFN regulatory factor-1 (IRF1) in response to gliadin was observed. Using transcriptome-wide co-expression analyses we are able to propose novel genes involved in CD pathogenesis that respond upon gliadin stimulation, also in non-celiac models.
Collapse
Affiliation(s)
- Irati Romero-Garmendia
- University of the Basque Country (UPV-EHU) and Biocruces Health Research Institute, 48940 Leioa, Spain.
| | - Koldo Garcia-Etxebarria
- University of the Basque Country (UPV-EHU) and Biocruces Health Research Institute, 48940 Leioa, Spain.
| | - Hector Hernandez-Vargas
- Epigenetics Group, International Agency for Research on Cancer (IARC), 69372 Lyon CEDEX 08, France.
| | - Izortze Santin
- University of the Basque Country (UPV-EHU) and Biocruces Health Research Institute, 48940 Leioa, Spain.
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28029 Madrid, Spain.
| | - Amaia Jauregi-Miguel
- University of the Basque Country (UPV-EHU) and Biocruces Health Research Institute, 48940 Leioa, Spain.
| | - Leticia Plaza-Izurieta
- University of the Basque Country (UPV-EHU) and Biocruces Health Research Institute, 48940 Leioa, Spain.
| | - Marie-Pierre Cros
- Epigenetics Group, International Agency for Research on Cancer (IARC), 69372 Lyon CEDEX 08, France.
| | - Maria Legarda
- Pediatric Gastroenterology Unit, Cruces University Hospital, University of the Basque Country-(UPV/EHU) and Biocruces Health Research Institute, 48903 Barakaldo, Spain.
| | - Iñaki Irastorza
- Pediatric Gastroenterology Unit, Cruces University Hospital, University of the Basque Country-(UPV/EHU) and Biocruces Health Research Institute, 48903 Barakaldo, Spain.
| | - Zdenko Herceg
- Epigenetics Group, International Agency for Research on Cancer (IARC), 69372 Lyon CEDEX 08, France.
| | - Nora Fernandez-Jimenez
- University of the Basque Country (UPV-EHU) and Biocruces Health Research Institute, 48940 Leioa, Spain.
- Epigenetics Group, International Agency for Research on Cancer (IARC), 69372 Lyon CEDEX 08, France.
| | - Jose Ramon Bilbao
- University of the Basque Country (UPV-EHU) and Biocruces Health Research Institute, 48940 Leioa, Spain.
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28029 Madrid, Spain.
| |
Collapse
|
74
|
MiR-23a transcriptional activated by Runx2 increases metastatic potential of mouse hepatoma cell via directly targeting Mgat3. Sci Rep 2018; 8:7366. [PMID: 29743543 PMCID: PMC5943354 DOI: 10.1038/s41598-018-25768-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/27/2018] [Indexed: 12/19/2022] Open
Abstract
MicroRNAs (miRNAs) and aberrant glycosylation both play important roles in tumor metastasis. In this study, the role of miR-23a in N-glycosylation and the metastasis of mouse hepatocellular carcinoma (HCC) cells was investigated. The miRNA expression array profiles that were confirmed by qPCR and Western blot analyses revealed higher miR-23a expression levels in Hca-P cells (with lymphatic metastasis potential) than in Hepa1-6 cells (with no lymphatic metastasis potential), while the expression of mannoside acetylglucosaminyltransferase 3 (Mgat3) was negatively associated with metastasis potential. Mgat3 is a key glycosyltransferase in the synthesis of the bisecting (β1,4GlcNAc branching) N-glycan structure. Bioinformatics analysis indicated that Mgat3 may be a target of miR-23a, and this hypothesis was verified by dual-luciferase reporter gene assays. Furthermore, we found that the transcription factor Runx2 can directly bind to the miR-23a gene promoter and promote its expression, as shown in dual-luciferase reporter gene assays and ChIP assays. Collectively, these results indicate that miR-23a might increase the metastatic potential of mouse HCC by affecting the branch formation of N-glycan chains presented on the cell surface through the targeting of the glycosyltransferase Mgat3. These findings may provide insight into the relationship between abnormal miRNA expression and aberrant glycosylation during tumor lymphatic metastasis.
Collapse
|
75
|
Takenaka Y, Mikami K, Seno S, Matsuda H. Automated transition analysis of activated gene regulation during diauxic nutrient shift in Escherichia coli and adipocyte differentiation in mouse cells. BMC Bioinformatics 2018; 19:89. [PMID: 29745848 PMCID: PMC5998889 DOI: 10.1186/s12859-018-2072-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Background Comprehensively understanding the dynamics of biological systems is among the biggest current challenges in biology and medicine. To acquire this understanding, researchers have measured the time-series expression profiles of cell lines of various organisms. Biological technologies have also drastically improved, providing a huge amount of information with support from bioinformatics and systems biology. However, the transitions between the activation and inactivation of gene regulations, at the temporal resolution of single time points, are difficult to extract from time-course gene expression profiles. Results Our proposed method reports the activation period of each gene regulation from gene expression profiles and a gene regulatory network. The correctness and effectiveness of the method were validated by analyzing the diauxic shift from glucose to lactose in Escherichia coli. The method completely detected the three periods of the shift; 1) consumption of glucose as nutrient source, 2) the period of seeking another nutrient source and 3) consumption of lactose as nutrient source. We then applied the method to mouse adipocyte differentiation data. Cell differentiation into adipocytes is known to involve two waves of the gene regulation cascade, and sub-waves are predicted. From the gene expression profiles of the cell differentiation process from ES to adipose cells (62 time points), our method acquired four periods; three periods covering the two known waves of the cascade, and a final period of gene regulations when the differentiation to adipocytes was completed. Conclusions Our proposed method identifies the transitions of gene regulations from time-series gene expression profiles. Dynamic analyses are essential for deep understanding of biological systems and for identifying the causes of the onset of diseases such as diabetes and osteoporosis. The proposed method can greatly contribute to the progress of biology and medicine.
Collapse
Affiliation(s)
- Yoichi Takenaka
- Faculty of Informatics, Kansai University, Ryousenji 2-1-1, Takatsuki, Osaka, Japan. .,Graduate School of Information Science and Technology, Osaka University, Yamadaoka 1-5, Suita, Osaka, Japan. .,Graduate School of Medicine, Osaka University, Yamadaoka 2, Suita, Osaka, Japan.
| | - Kazuma Mikami
- Recruit Holdings Co. Ltd., Marunouchi 1-9-2, Chiyoda, Tokyo, Japan
| | - Shigeto Seno
- Graduate School of Information Science and Technology, Osaka University, Yamadaoka 1-5, Suita, Osaka, Japan
| | - Hideo Matsuda
- Graduate School of Information Science and Technology, Osaka University, Yamadaoka 1-5, Suita, Osaka, Japan
| |
Collapse
|
76
|
Mohamed H, Gould D. PCR Assembly of Synthetic Promoters. Methods Mol Biol 2018; 1651:147-156. [PMID: 28801905 DOI: 10.1007/978-1-4939-7223-4_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
In this chapter, we describe a two-step assembly PCR method to construct synthetic promoters. Essentially, this method takes advantage of specific annealing between complimentary DNA sequences to build random TFBS combinations within the assembled PCR products. A DNA polymerase is then employed to fill in the unpaired nucleotides in the generated sequences and also to amplify the assembled PCR products. We have used this method to generate synthetic promoters whereby the orientation of the TFBS can be controlled, the spacing between TFBS can be predetermined, and also the full diversity of the consensus TFBS can be covered through the use of degenerate oligonucleotides .
Collapse
Affiliation(s)
- Hodan Mohamed
- Public Health England, 61 Colindale Avenue, NW9 5EQ, London, UK
| | - David Gould
- Department of Biochemical Pharmacology, William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK.
| |
Collapse
|
77
|
Song Z, Zhang C, He L, Sui Y, Lin X, Pan J. Uncovering transcription factor and microRNA risk regulatory pathways associated with osteoarthritis by network analysis. Biochem Biophys Res Commun 2018; 500:902-906. [PMID: 29709470 DOI: 10.1016/j.bbrc.2018.04.189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 04/24/2018] [Indexed: 12/31/2022]
Abstract
Osteoarthritis (OA) is the most common form of joint disease. The development of inflammation have been considered to play a key role during the progression of OA. Regulatory pathways are known to play crucial roles in many pathogenic processes. Thus, deciphering these risk regulatory pathways is critical for elucidating the mechanisms underlying OA. We constructed an OA-specific regulatory network by integrating comprehensive curated transcription and post-transcriptional resource involving transcription factor (TF) and microRNA (miRNA). To deepen our understanding of underlying molecular mechanisms of OA, we developed an integrated systems approach to identify OA-specific risk regulatory pathways. In this study, we identified 89 significantly differentially expressed genes between normal and inflamed areas of OA patients. We found the OA-specific regulatory network was a standard scale-free network with small-world properties. It significant enriched many immune response-related functions including leukocyte differentiation, myeloid differentiation and T cell activation. Finally, 141 risk regulatory pathways were identified based on OA-specific regulatory network, which contains some known regulator of OA. The risk regulatory pathways may provide clues for the etiology of OA and be a potential resource for the discovery of novel OA-associated disease genes.
Collapse
Affiliation(s)
- Zhenhua Song
- Central South University, Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, Hainan, China.
| | - Chi Zhang
- Rehabilitation Medicine Department, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Lingxiao He
- XuZhou Medical University, Xuzhou, Jiangsu, China
| | - Yanfang Sui
- Central South University, Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, Hainan, China
| | - Xiafei Lin
- Central South University, Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, Hainan, China
| | - Jingjing Pan
- Central South University, Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, Hainan, China
| |
Collapse
|
78
|
Rabachini T, Boccardo E, Andrade R, Perez KR, Nonogaki S, Cuccovia IM, Villa LL. HPV-16 E7 expression up-regulates phospholipase D activity and promotes rapamycin resistance in a pRB-dependent manner. BMC Cancer 2018; 18:485. [PMID: 29703186 PMCID: PMC5923196 DOI: 10.1186/s12885-018-4392-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 04/17/2018] [Indexed: 01/04/2023] Open
Abstract
Background Human Papillomavirus (HPV) infection is the main risk factor for the development and progression of cervical cancer. HPV-16 E6 and E7 expression is essential for induction and maintenance of the transformed phenotype. These oncoproteins interfere with the function of several intracellular proteins, including those controlling the PI3K/AKT/mTOR pathway in which Phospolipase D (PLD) and Phosphatidic acid (PA) play a critical role. Methods PLD activity was measured in primary human keratinocytes transduced with retroviruses expressing HPV-16 E6, E7 or E7 mutants. The cytostatic effect of rapamycin, a well-known mTOR inhibitor with potential clinical applications, was evaluated in monolayer and organotypic cultures. Results HPV-16 E7 expression in primary human keratinocytes leads to an increase in PLD expression and activity. Moreover, this activation is dependent on the ability of HPV-16 E7 to induce retinoblastoma protein (pRb) degradation. We also show that cells expressing HPV-16 E7 or silenced for pRb acquire resistance to the antiproliferative effect of rapamycin. Conclusion This is the first indication that HPV oncoproteins can affect PLD activity. Since PA can interfere with the ability of rapamycin to bind mTOR, the use of combined strategies to target mTOR and PLD activity might be considered to treat HPV-related malignancies. Electronic supplementary material The online version of this article (10.1186/s12885-018-4392-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tatiana Rabachini
- Ludwig Institute for Cancer Research - Hospital Sírio-Libanês, São Paulo, SP, Brazil.
| | - Enrique Boccardo
- Ludwig Institute for Cancer Research - Hospital Sírio-Libanês, São Paulo, SP, Brazil.,Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Rubiana Andrade
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Katia Regina Perez
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil.,Departamento de Biofísica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Suely Nonogaki
- Centro de Patologia do Instituto Adolfo Lutz, São Paulo, SP, Brazil
| | - Iolanda Midea Cuccovia
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Luisa Lina Villa
- Ludwig Institute for Cancer Research - Hospital Sírio-Libanês, São Paulo, SP, Brazil.,Faculdade de Medicina da Universidade de São Paulo, Instituto do Câncer do Estado de São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
79
|
Evolution of DNAase I Hypersensitive Sites in MHC Regulatory Regions of Primates. Genetics 2018; 209:579-589. [PMID: 29669733 DOI: 10.1534/genetics.118.301028] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 04/16/2018] [Indexed: 01/08/2023] Open
Abstract
It has been challenging to determine the disease-causing variant(s) for most major histocompatibility complex (MHC)-associated diseases. However, it is becoming increasingly clear that regulatory variation is pervasive and a fundamentally important mechanism governing phenotypic diversity and disease susceptibility. We gathered DNase I data from 136 human cells to characterize the regulatory landscape of the MHC region, including 4867 DNase I hypersensitive sites (DHSs). We identified thousands of regulatory elements that have been gained or lost in the human or chimpanzee genomes since their evolutionary divergence. We compared alignments of the DHS across six primates and found 149 DHSs with convincing evidence of positive and/or purifying selection. Of these DHSs, compared to neutral sequences, 24 evolved rapidly in the human lineage. We identified 15 instances of transcription-factor-binding motif gains, such as USF, MYC, MAX, MAFK, STAT1, PBX3, etc, and observed 16 GWAS (genome-wide association study) SNPs associated with diseases within these 24 DHSs using FIMO (Find Individual Motif Occurrences) and UCSC (University of California, Santa Cruz) ChIP-seq data. Combining eQTL and Hi-C data, our results indicated that there were five SNPs located in human gains motifs affecting the corresponding gene's expression, two of which closely matched DHS target genes. In addition, a significant SNP, rs7756521, at genome-wide significant level likely affects DDR expression and represents a causal genetic variant for HIV-1 control. These results indicated that species-specific motif gains or losses of rapidly evolving DHSs in the primate genomes might play a role during adaptation evolution and provided some new evidence for a potentially causal role for these GWAS SNPs.
Collapse
|
80
|
Lee KS, Chatterjee P, Choi EY, Sung MK, Oh J, Won H, Park SM, Kim YJ, Yi SV, Choi JK. Selection on the regulation of sympathetic nervous activity in humans and chimpanzees. PLoS Genet 2018; 14:e1007311. [PMID: 29672586 PMCID: PMC5908061 DOI: 10.1371/journal.pgen.1007311] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 03/17/2018] [Indexed: 12/31/2022] Open
Abstract
Adrenergic α2C receptor (ADRA2C) is an inhibitory modulator of the sympathetic nervous system. Knockout mice for this gene show physiological and behavioural alterations that are associated with the fight-or-flight response. There is evidence of positive selection on the regulation of this gene during chicken domestication. Here, we find that the neuronal expression of ADRA2C is lower in human and chimpanzee than in other primates. On the basis of three-dimensional chromatin structure, we identified a cis-regulatory region whose DNA sequences have been significantly accelerated in human and chimpanzee. Active histone modification marks this region in rhesus macaque but not in human and chimpanzee; instead, repressive marks are enriched in various human brain samples. This region contains two neuron-restrictive silencer factor (NRSF) binding motifs, each of which harbours a polymorphism. Our genotyping and analysis of population genome data indicate that at both polymorphic sites, the derived allele has reached fixation in humans and chimpanzees but not in bonobos, whereas only the ancestral allele is present among macaques. Our CRISPR/Cas9 genome editing and reporter assays show that both derived nucleotides repress ADRA2C, most likely by increasing NRSF binding. In addition, we detected signatures of recent positive selection for lower neuronal ADRA2C expression in humans. Our findings indicate that there has been selective pressure for enhanced sympathetic nervous activity in the evolution of humans and chimpanzees.
Collapse
Affiliation(s)
- Kang Seon Lee
- Department of Bio and Brain Engineering, KAIST, Daejeon, Republic of Korea
| | - Paramita Chatterjee
- School of Biology, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Eun-Young Choi
- Specific Organs Cancer Branch, Research Institute, National Cancer Center, Ilsan, Gyeonggi, Republic of Korea
| | - Min Kyung Sung
- Department of Bio and Brain Engineering, KAIST, Daejeon, Republic of Korea
| | - Jaeho Oh
- Department of Bio and Brain Engineering, KAIST, Daejeon, Republic of Korea
| | - Hyejung Won
- Department of Neurology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Seong-Min Park
- Specific Organs Cancer Branch, Research Institute, National Cancer Center, Ilsan, Gyeonggi, Republic of Korea
| | - Youn-Jae Kim
- Specific Organs Cancer Branch, Research Institute, National Cancer Center, Ilsan, Gyeonggi, Republic of Korea
| | - Soojin V. Yi
- School of Biology, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Jung Kyoon Choi
- Department of Bio and Brain Engineering, KAIST, Daejeon, Republic of Korea
| |
Collapse
|
81
|
Ahmad H, Requena T, Frejo L, Cobo M, Gallego-Martinez A, Martin F, Lopez-Escamez JA, Bronstein AM. Clinical and Functional Characterization of a Missense ELF2 Variant in a CANVAS Family. Front Genet 2018; 9:85. [PMID: 29628936 PMCID: PMC5876245 DOI: 10.3389/fgene.2018.00085] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 02/28/2018] [Indexed: 12/30/2022] Open
Abstract
Cerebellar ataxia with neuropathy and bilateral vestibular areflexia syndrome (CANVAS) is a rare disorder with an unknown etiology. We present a British family with presumed autosomal dominant CANVAS with incomplete penetrance and variable expressivity. Exome sequencing identified a rare missense variant in the ELF2 gene at chr4:g.140058846 C > T, c.10G > A, p.A4T which segregated in all affected patients. By using transduced BE (2)-M17 cells, we found that the mutated ELF2 (mt-ELF2) gene increased ATXN2 and reduced ELOVL5 gene expression, the causal genes of type 2 and type 38 spinocerebellar ataxias. Both, western blot and confocal microscopy confirmed an increase of ataxin-2 in BE(2)-M17 cells transduced with lentivirus expressing mt-ELF2 (CEE-mt-ELF2), which was not observed in cells transduced with lentivirus expressing wt-ELF2 (CEE-wt-ELF2). Moreover, we observed a significant decrease in the number and size of lipid droplets in the CEE-mt-ELF2-transduced BE (2)-M17 cells, but not in the CEE-wt-ELF2-transduced BE (2)-M17. Furthermore, changes in the expression of ELOVL5 could be related with the reduction of lipid droplets in BE (2)-M17 cells. This work supports that ELF2 gene regulates the expression of ATXN2 and ELOVL5 genes, and defines new molecular links in the pathophysiology of cerebellar ataxias.
Collapse
Affiliation(s)
- Hena Ahmad
- Division of Brain Sciences, Imperial College, Charing Cross Hospital, London, United Kingdom.,National Hospital for Neurology & Neurosurgery, London, United Kingdom
| | - Teresa Requena
- Otology and Neurotology Group CTS495, Department of Genomic Medicine, Centro de Genómica e Investigación Oncológica (GENYO), Pfizer-Universidad de Granada-Junta de Andalucía, Granada, Spain
| | - Lidia Frejo
- Otology and Neurotology Group CTS495, Department of Genomic Medicine, Centro de Genómica e Investigación Oncológica (GENYO), Pfizer-Universidad de Granada-Junta de Andalucía, Granada, Spain
| | - Marien Cobo
- Gene and Cell Therapy Group, Department of Genomic Medicine, Centro de Genómica e Investigación Oncológica (GENYO), Pfizer-Universidad de Granada-Junta de Andalucía, Granada, Spain
| | - Alvaro Gallego-Martinez
- Otology and Neurotology Group CTS495, Department of Genomic Medicine, Centro de Genómica e Investigación Oncológica (GENYO), Pfizer-Universidad de Granada-Junta de Andalucía, Granada, Spain
| | - Francisco Martin
- Gene and Cell Therapy Group, Department of Genomic Medicine, Centro de Genómica e Investigación Oncológica (GENYO), Pfizer-Universidad de Granada-Junta de Andalucía, Granada, Spain
| | - Jose A Lopez-Escamez
- Otology and Neurotology Group CTS495, Department of Genomic Medicine, Centro de Genómica e Investigación Oncológica (GENYO), Pfizer-Universidad de Granada-Junta de Andalucía, Granada, Spain.,Department of Otolaryngology, Instituto de Investigación Biosanitaria ibs.GRANADA, Hospital Virgen de las Nieves, Universidad de Granada, Granada, Spain
| | - Adolfo M Bronstein
- Division of Brain Sciences, Imperial College, Charing Cross Hospital, London, United Kingdom.,National Hospital for Neurology & Neurosurgery, London, United Kingdom
| |
Collapse
|
82
|
TACC3 transcriptionally upregulates E2F1 to promote cell growth and confer sensitivity to cisplatin in bladder cancer. Cell Death Dis 2018; 9:72. [PMID: 29358577 PMCID: PMC5833822 DOI: 10.1038/s41419-017-0112-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 08/28/2017] [Accepted: 09/18/2017] [Indexed: 12/11/2022]
Abstract
Accumulating evidence has shown that transforming acidic coiled-coil 3 (TACC3) is deregulated in a broad spectrum of cancers. In the present study, we reported that TACC3 was markedly elevated in bladder cancer, especially in muscle-invasive bladder cancers (MIBCs). The upregulation of TACC3 was positively associated with tumor invasiveness, grade, T stage, and progression in patients with bladder cancer. Furthermore, a Kaplan-Meier survival analysis showed that patients with bladder cancer whose tumors had high TACC3 expression experienced a dismal prognosis compared with patients whose tumors had low TACC3 expression. Functional studies have found that TACC3 is a prerequisite for the development of malignant characteristics of bladder cancer cells, including cell proliferation and invasion. Moreover, TACC3 promoted G1/S transition, which was mediated via activation of the transcription of E2F1, eventually enhancing cell proliferation. Notably, the overexpression of TACC3 or E2F1 indicates a high sensitivity to cisplatin. Taken together, these findings define a tumor-supportive role for TACC3, which may also serve as a prognostic and therapeutic indicator in bladder cancers.
Collapse
|
83
|
Ullmark T, Montano G, Gullberg U. DNA and RNA binding by the Wilms' tumour gene 1 (WT1) protein +KTS and −KTS isoforms-From initial observations to recent global genomic analyses. Eur J Haematol 2018; 100:229-240. [DOI: 10.1111/ejh.13010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2017] [Indexed: 12/17/2022]
Affiliation(s)
- Tove Ullmark
- Department of Haematology and Transfusion Medicine; Lund University; Lund Sweden
| | - Giorgia Montano
- Department of Haematology and Transfusion Medicine; Lund University; Lund Sweden
| | - Urban Gullberg
- Department of Haematology and Transfusion Medicine; Lund University; Lund Sweden
| |
Collapse
|
84
|
Hu J, Hu Z, Wang X, Gu M, Gao Z, Liang Y, Ma C, Liu X, Hu S, Chen S, Peng D, Jiao X, Liu X. Deep sequencing of the mouse lung transcriptome reveals distinct long non-coding RNAs expression associated with the high virulence of H5N1 avian influenza virus in mice. Virulence 2018; 9:1092-1111. [PMID: 30052469 PMCID: PMC6086314 DOI: 10.1080/21505594.2018.1475795] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 05/08/2018] [Indexed: 01/22/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) play multiple key regulatory roles in various biological processes. However, their function in influenza A virus (IAV) pathogenicity remains largely unexplored. Here, using next generation sequencing, we systemically compared the whole-transcriptome response of the mouse lung infected with either the highly pathogenic (A/Chicken/Jiangsu/k0402/2010, CK10) or the nonpathogenic (A/Goose/Jiangsu/k0403/2010, GS10) H5N1 virus. A total of 126 significantly differentially expressed (SDE) lncRNAs from three replicates were identified to be associated with the high virulence of CK10, whereas 94 SDE lncRNAs were related with GS10. Functional category analysis suggested that the SDE lncRNAs-coexpressed mRNAs regulated by CK10 were highly related with aberrant and uncontrolled inflammatory responses. Further canonical pathway analysis also confirmed that these targets were highly enriched for inflammatory-related pathways. Moreover, 9 lncRNAs and 17 lncRNAs-coexpressed mRNAs associated with a large number of targeted genes were successfully verified by qRT-PCR. One targeted lncRNA (NONMMUT011061) that was markedly activated and correlated with a great number of mRNAs was selected for further in-depth analysis, including predication of transcription factors, potential interacting proteins, genomic location, coding ability and construction of the secondary structure. More importantly, NONMMUT011061 was also distinctively stimulated during the highly pathogenic H5N8 virus infection in mice, suggesting a potential universal role of NONMMUT011061 in the pathogenesis of different H5 IAV. Altogether, these results provide a subset of lncRNAs that might play important roles in the pathogenesis of influenza virus and add the lncRNAs to the vast repertoire of host factors utilized by IAV for infection and persistence.
Collapse
Affiliation(s)
- Jiao Hu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-food Safety and Quality, Ministry of Agriculture of China (26116120), Yangzhou University, Yangzhou, China
| | - Zenglei Hu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-food Safety and Quality, Ministry of Agriculture of China (26116120), Yangzhou University, Yangzhou, China
| | - Xiaoquan Wang
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-food Safety and Quality, Ministry of Agriculture of China (26116120), Yangzhou University, Yangzhou, China
| | - Min Gu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-food Safety and Quality, Ministry of Agriculture of China (26116120), Yangzhou University, Yangzhou, China
| | - Zhao Gao
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-food Safety and Quality, Ministry of Agriculture of China (26116120), Yangzhou University, Yangzhou, China
| | - Yanyan Liang
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-food Safety and Quality, Ministry of Agriculture of China (26116120), Yangzhou University, Yangzhou, China
| | - Chunxi Ma
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-food Safety and Quality, Ministry of Agriculture of China (26116120), Yangzhou University, Yangzhou, China
| | - Xiaowen Liu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-food Safety and Quality, Ministry of Agriculture of China (26116120), Yangzhou University, Yangzhou, China
| | - Shunlin Hu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-food Safety and Quality, Ministry of Agriculture of China (26116120), Yangzhou University, Yangzhou, China
| | - Sujuan Chen
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-food Safety and Quality, Ministry of Agriculture of China (26116120), Yangzhou University, Yangzhou, China
| | - Daxing Peng
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-food Safety and Quality, Ministry of Agriculture of China (26116120), Yangzhou University, Yangzhou, China
| | - Xinan Jiao
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-food Safety and Quality, Ministry of Agriculture of China (26116120), Yangzhou University, Yangzhou, China
| | - Xiufan Liu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-food Safety and Quality, Ministry of Agriculture of China (26116120), Yangzhou University, Yangzhou, China
| |
Collapse
|
85
|
Luo J, Huang W, Cao B. A novel approach to identify the miRNA-mRNA causal regulatory modules in Cancer. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2018; 15:309-315. [PMID: 28113985 DOI: 10.1109/tcbb.2016.2612199] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
MicroRNAs (miRNAs) play an essential role in many biological processes by regulating the target genes, especially in the initiation and development of cancers. Therefore, the identification of the miRNA-mRNA regulatory modules is important for understanding the regulatory mechanisms. Most computational methods only used statistical correlations in predicting miRNA-mRNA modules, and neglected the fact there are causal relationships between miRNAs and their target genes. In this paper, we propose a novel approach called CALM(the causal regulatory modules) to identify the miRNA-mRNA regulatory modules through integrating the causal interactions and statistical correlations between the miRNAs and their target genes. Our algorithm largely consists of three steps: it first forms the causal regulatory relationships of miRNAs and genes from gene expression profiles and detects the miRNA clusters according to the GO function information of their target genes, then expands each miRNA cluster by greedy adding(discarding) the target genes to maximize the modularity score. To show the performance of our method, we apply CALM on four datasets including EMT, breast, ovarian, thyroid cancer and validate our results. The experiment results show that our method can not only outperform the compared method, but also achieve ideal overall performance in terms of the functional enrichment.
Collapse
|
86
|
Prandi S, Voigt A, Meyerhof W, Behrens M. Expression profiling of Tas2r genes reveals a complex pattern along the mouse GI tract and the presence of Tas2r131 in a subset of intestinal Paneth cells. Cell Mol Life Sci 2018; 75:49-65. [PMID: 28801754 PMCID: PMC11105753 DOI: 10.1007/s00018-017-2621-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 08/02/2017] [Accepted: 08/04/2017] [Indexed: 12/18/2022]
Abstract
The chemical variability of the intestinal lumen requires the presence of molecular receptors detecting the various substances naturally occurring in the diet and as a result of the activity of the microbiota. Despite their early discovery, intestinal bitter taste receptors (Tas2r) have not yet been assigned an unambiguous physiological function. Recently, using a CRE-recombinant approach we showed that the Tas2r131 gene is expressed in a subset of mucin-producing goblet cells in the colon of mice. Moreover, we also demonstrated that the expression of the Tas2r131 locus is not restricted to this region. In the present study we aimed at characterizing the presence of positive cells also in other gastrointestinal regions. Our results show that Tas2r131+ cells appear in the jejunum and the ileum, and are absent from the stomach and the duodenum. We identified the positive cells as a subpopulation of deep-crypt Paneth cells in the ileum, strengthening the notion of a defensive role for Tas2rs in the gut. To get a broader perspective on the expression of bitter taste receptors in the alimentary canal, we quantified the expression of all 35 Tas2r genes along the gastrointestinal tract by qRT-PCR. We discovered that the number and expression level of Tas2r genes profoundly vary along the alimentary canal, with the stomach and the colon expressing the largest subsets.
Collapse
Affiliation(s)
- Simone Prandi
- Department of Molecular Genetics, German Institute for Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Anja Voigt
- Department of Molecular Genetics, German Institute for Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Wolfgang Meyerhof
- Department of Molecular Genetics, German Institute for Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Maik Behrens
- Department of Molecular Genetics, German Institute for Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany.
| |
Collapse
|
87
|
Park D, Kim BC, Kim CH, Choi YJ, Jeong HO, Kim ME, Lee JS, Park MH, Chung KW, Kim DH, Lee J, Im DS, Yoon S, Lee S, Yu BP, Bhak J, Chung HY. RNA-Seq analysis reveals new evidence for inflammation-related changes in aged kidney. Oncotarget 2017; 7:30037-48. [PMID: 27153548 PMCID: PMC5058662 DOI: 10.18632/oncotarget.9152] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 04/18/2016] [Indexed: 01/10/2023] Open
Abstract
Age-related dysregulated inflammation plays an essential role as a major risk factor underlying the pathophysiological aging process. To better understand how inflammatory processes are related to aging at the molecular level, we sequenced the transcriptome of young and aged rat kidney using RNA-Seq to detect known genes, novel genes, and alternative splicing events that are differentially expressed. By comparing young (6 months of age) and old (25 months of age) rats, we detected 722 up-regulated genes and 111 down-regulated genes. In the aged rats, we found 32 novel genes and 107 alternatively spliced genes. Notably, 6.6% of the up-regulated genes were related to inflammation (P < 2.2 × 10-16, Fisher exact t-test); 15.6% were novel genes with functional protein domains (P = 1.4 × 10-5); and 6.5% were genes showing alternative splicing events (P = 3.3 × 10-4). Based on the results of pathway analysis, we detected the involvement of inflammation-related pathways such as cytokines (P = 4.4 × 10-16), which were found up-regulated in the aged rats. Furthermore, an up-regulated inflammatory gene analysis identified the involvement of transcription factors, such as STAT4, EGR1, and FOSL1, which regulate cancer as well as inflammation in aging processes. Thus, RNA changes in these pathways support their involvement in the pro-inflammatory status during aging. We propose that whole RNA-Seq is a useful tool to identify novel genes and alternative splicing events by documenting broadly implicated inflammation-related genes involved in aging processes.
Collapse
Affiliation(s)
- Daeui Park
- Molecular Inflammation Research Center for Aging Intervention, Pusan National University, Busan, Korea.,Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, Korea.,Human and Environmental Toxicology, School of Engineering, University of Science and Technology, Daejeon, Korea
| | - Byoung-Chul Kim
- Molecular Inflammation Research Center for Aging Intervention, Pusan National University, Busan, Korea.,Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, Korea
| | | | - Yeon Ja Choi
- Molecular Inflammation Research Center for Aging Intervention, Pusan National University, Busan, Korea
| | - Hyoung Oh Jeong
- Molecular Inflammation Research Center for Aging Intervention, Pusan National University, Busan, Korea
| | - Mi Eun Kim
- Department of Biology, College of Natural Sciences, Chosun University, Gwangju, Korea
| | - Jun Sik Lee
- Department of Biology, College of Natural Sciences, Chosun University, Gwangju, Korea
| | - Min Hi Park
- Molecular Inflammation Research Center for Aging Intervention, Pusan National University, Busan, Korea
| | - Ki Wung Chung
- Molecular Inflammation Research Center for Aging Intervention, Pusan National University, Busan, Korea
| | - Dae Hyun Kim
- Molecular Inflammation Research Center for Aging Intervention, Pusan National University, Busan, Korea
| | - Jaewon Lee
- Molecular Inflammation Research Center for Aging Intervention, Pusan National University, Busan, Korea
| | - Dong-Soon Im
- Molecular Inflammation Research Center for Aging Intervention, Pusan National University, Busan, Korea
| | - Seokjoo Yoon
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, Korea.,Human and Environmental Toxicology, School of Engineering, University of Science and Technology, Daejeon, Korea
| | - Sunghoon Lee
- Personal Genomics Institute,Genome Research Foundation, Suwon, Korea
| | - Byung Pal Yu
- Department of Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Jong Bhak
- Personal Genomics Institute,Genome Research Foundation, Suwon, Korea
| | - Hae Young Chung
- Molecular Inflammation Research Center for Aging Intervention, Pusan National University, Busan, Korea
| |
Collapse
|
88
|
Young WC, Raftery AE, Yeung KY. Model-Based Clustering With Data Correction For Removing Artifacts In Gene Expression Data. Ann Appl Stat 2017; 11:1998-2026. [PMID: 30740193 DOI: 10.1214/17-aoas1051] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The NIH Library of Integrated Network-based Cellular Signatures (LINCS) contains gene expression data from over a million experiments, using Luminex Bead technology. Only 500 colors are used to measure the expression levels of the 1,000 landmark genes measured, and the data for the resulting pairs of genes are deconvolved. The raw data are sometimes inadequate for reliable deconvolution, leading to artifacts in the final processed data. These include the expression levels of paired genes being flipped or given the same value, and clusters of values that are not at the true expression level. We propose a new method called model-based clustering with data correction (MCDC) that is able to identify and correct these three kinds of artifacts simultaneously. We show that MCDC improves the resulting gene expression data in terms of agreement with external baselines, as well as improving results from subsequent analysis.
Collapse
Affiliation(s)
- William Chad Young
- Department of Statistics, University of Washington, Box 354322, Seattle, WA 98195
| | - Adrian E Raftery
- Department of Statistics, University of Washington, Box 354322, Seattle, WA 98195
| | - Ka Yee Yeung
- Institute of Technology, University of Washington Tacoma, Campus Box 358426, 1900 Commerce Street, Tacoma, WA 98402
| |
Collapse
|
89
|
He DD, Lu Y, Gittelman R, Jin Y, Ling F, Joshua A. Positive selection of the TRIM family regulatory region in primate genomes. Proc Biol Sci 2017; 283:rspb.2016.1602. [PMID: 27733547 DOI: 10.1098/rspb.2016.1602] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Accepted: 09/13/2016] [Indexed: 12/13/2022] Open
Abstract
Viral selection pressure has acted on restriction factors that play an important role in the innate immune system by inhibiting the replication of viruses during primate evolution. Tripartite motif-containing (TRIM) family members are some of these restriction factors. It is becoming increasingly clear that gene expression differences, rather than protein-coding regions changes, could play a vital role in the anti-retroviral immune mechanism. Increasingly, recent studies have created genome-scale catalogues of DNase I hypersensitive sites (DHSs), which demark potentially functional regulatory DNA. To improve our understanding of the molecular evolution mechanism of antiviral differences between species, we leveraged 14 130 DHSs derived from 145 cell types to characterize the regulatory landscape of the TRIM region. Subsequently, we compared the alignments of the DHSs across six primates and found 375 DHSs that are conserved in non-human primates but exhibit significantly accelerated rates of evolution in the human lineage (haDHSs). Furthermore, we discovered 31 human-specific potential transcription factor motifs within haDHSs, including the KROX and SP1, that both interact with HIV-1 Importantly, the corresponding haDHS was correlated with antiviral factor TRIM23 Thus, our results suggested that some viruses may contribute, through regulatory DNA differences, to organismal evolution by mediating TRIM gene expression to escape immune surveillance.
Collapse
Affiliation(s)
- Dan-Dan He
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, People's Republic of China
| | - Yueer Lu
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, People's Republic of China
| | - Rachel Gittelman
- Department of Genome Sciences, University of Washington, Seattle, WA 98125, USA
| | - Yabin Jin
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, People's Republic of China
| | - Fei Ling
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, People's Republic of China
| | - Akey Joshua
- Department of Genome Sciences, University of Washington, Seattle, WA 98125, USA
| |
Collapse
|
90
|
Pei C, Wang SL, Fang J, Zhang W. GSMC: Combining Parallel Gibbs Sampling with Maximal Cliques for Hunting DNA Motif. J Comput Biol 2017; 24:1243-1253. [PMID: 29116820 DOI: 10.1089/cmb.2017.0100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Regulatory elements are responsible for regulating gene transcription. Therefore, identification of these elements is a tremendous challenge in the field of gene expression. Transcription factors (TFs) play a key role in gene regulation by binding to target promoter sequences. A set of conserved sequence patterns with a highly similar structure that is bound by a TF is called a motif. Motif discovery has been a difficult problem over the past decades. Meanwhile, it is a foundation stone in meeting this challenge. Recent advances in obtaining genomic sequences and high-throughput gene expression analysis techniques have enabled the rapid development of computational methods for motif discovery. As a result, a large number of motif-finding algorithms aiming at various motif models have sprung up in the past few years. However, most of them are not suitable for analysis of the large data sets generated by next-generation sequencing. To better handle large-scale ChIP-Seq data and achieve better performance in computational time and motif detection accuracy, we propose an excellent motif-finding algorithm known as GSMC (Combining Parallel Gibbs Sampling with Maximal Cliques for hunting DNA Motif). The GSMC algorithm consists of two steps. First, we employ the commonly used Gibbs sampling to generating initial motifs. Second, we utilize maximal cliques to cluster motifs according to Similarity with Position Information Contents (SPIC). Consequently, we raise the detection accuracy in a great degree, in the meantime holding comparative computation efficiency. In addition, we can find much more credible cofactor interacting motifs.
Collapse
Affiliation(s)
- Chao Pei
- 1 College of Computer Science and Electronics Engineering, Hunan University , Changsha, China
| | - Shu-Lin Wang
- 1 College of Computer Science and Electronics Engineering, Hunan University , Changsha, China
| | - Jianwen Fang
- 2 Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute , Rockville, MD 20850
| | - Wei Zhang
- 1 College of Computer Science and Electronics Engineering, Hunan University , Changsha, China
| |
Collapse
|
91
|
Liu ZW, Li H, Wang WL, Wu ZJ, Cui X, Zhuang J. CsGOGAT Is Important in Dynamic Changes of Theanine Content in Postharvest Tea Plant Leaves under Different Temperature and Shading Spreadings. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:9693-9702. [PMID: 29020770 DOI: 10.1021/acs.jafc.7b04552] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
We analyzed the changes of theanine content in postharvest tea leaves under high temperature (38 °C), low temperature (4 °C), and shading spreadings by using ultrahigh-performance liquid chromatography. The differentially expressed proteins (DEPs), CsFd-GOGAT and CsNADH-GOGAT, which are involved in theanine biosynthesis pathway, were identified from the corresponding proteome data. The protein-protein interactions of CsFd-GOGAT and CsNADH-GOGAT, CsTS1, or CsNiR were verified by yeast two-hybrid technology. The expression profiles of 17 genes in theanine metabolism, including CsFd-GOGAT and CsNADH-GOGAT, were analyzed by quantitative real-time polymerase chain reaction. The correlations between the dynamic changes of theanine content and expression profiles of related genes and DEPs were analyzed. This study preliminarily proved the importance of CsGOGAT in dynamic changes of theanine content in postharvest tea leaves during spreading.
Collapse
Affiliation(s)
- Zhi-Wei Liu
- Tea Science Research Institute, College of Horticulture, Nanjing Agricultural University , Nanjing, 210095, People's Republic of China
| | - Hui Li
- Tea Science Research Institute, College of Horticulture, Nanjing Agricultural University , Nanjing, 210095, People's Republic of China
| | - Wen-Li Wang
- Tea Science Research Institute, College of Horticulture, Nanjing Agricultural University , Nanjing, 210095, People's Republic of China
| | - Zhi-Jun Wu
- Tea Science Research Institute, College of Horticulture, Nanjing Agricultural University , Nanjing, 210095, People's Republic of China
| | - Xin Cui
- Tea Science Research Institute, College of Horticulture, Nanjing Agricultural University , Nanjing, 210095, People's Republic of China
| | - Jing Zhuang
- Tea Science Research Institute, College of Horticulture, Nanjing Agricultural University , Nanjing, 210095, People's Republic of China
| |
Collapse
|
92
|
Kanayama M, Hayano T, Koebis M, Maeda T, Tabe Y, Horie S, Aiba A. Hyperactive mTOR induces neuroendocrine differentiation in prostate cancer cell with concurrent up-regulation of IRF1. Prostate 2017; 77:1489-1498. [PMID: 28905415 DOI: 10.1002/pros.23425] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Accepted: 08/23/2017] [Indexed: 02/02/2023]
Abstract
BACKGROUND Neuroendocrine-differentiated prostate cancer (NEPCa) is refractory to androgen deprivation therapy and shows a poor prognosis. The underlying mechanisms responsible for neuroendocrine differentiation (NED) are yet to be clarified. In this study, we investigated the role of mammalian target of rapamycin (mTOR) in NEPCa. METHODS We utilized a gain-of-function analysis by establishing a human PCa LNCaP stable line that expresses hyperactive mTOR (LNCaP-mTOR). Then, we employed a comprehensive mass spectrometric analysis to identify a key transcription factor in LNCaP-mTOR, followed by a loss-of-function analysis using CRISPR/Cas system. RESULTS The activation of mTOR induced NED. We observed significant cell growth arrest in NED of LNCaP-mTOR, which accompanied increased expression of p21WAF1/CIP1 . A comprehensive mass spectrometric analysis identified interferon regulatory factor 1 (IRF1) as a key transcription factor in growth arrest of LNCaP-mTOR. The disruption of IRF1 gene in LNCaP-mTOR reversed cell growth arrest along with the suppression of its target p21WAF1/CIP1 . These results indicate that the growth arrest in NED is at least in part dependent on IRF1 through the induction of p21WAF1/CIP1 . CONCLUSIONS We identified active mTOR as a novel inducer of NED, and elucidated a mechanism underlying the malignant transformation of NEPCa by recapitulating NED in vitro.
Collapse
Affiliation(s)
- Mayuko Kanayama
- Department of Urology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Laboratory of Animal Resources, Center for Disease Biology and Integrated Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Toshiya Hayano
- Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Shiga, Japan
| | - Michinori Koebis
- Laboratory of Animal Resources, Center for Disease Biology and Integrated Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tatsuya Maeda
- Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo, Japan
| | - Yoko Tabe
- Department of Clinical Laboratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shigeo Horie
- Department of Urology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Atsu Aiba
- Laboratory of Animal Resources, Center for Disease Biology and Integrated Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
93
|
Cumbo F, Vergni D, Santoni D. Investigating transcription factor synergism in humans. DNA Res 2017; 25:103-112. [PMID: 29069301 PMCID: PMC5824945 DOI: 10.1093/dnares/dsx041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 09/19/2017] [Indexed: 01/13/2023] Open
Abstract
Proteins are the core and the engine of every process in cells thus the study of mechanisms that drive the regulation of protein expression, is essential. Transcription factors play a central role in this extremely complex task and they synergically co-operate in order to provide a fine tuning of protein expressions. In the present study, we designed a mathematically well-founded procedure to investigate the mutual positioning of transcription factors binding sites related to a given couple of transcription factors in order to evaluate the possible association between them. We obtained a list of highly related transcription factors couples, whose binding site occurrences significantly group together for a given set of gene promoters, identifying the biological contexts in which the couples are involved in and the processes they should contribute to regulate.
Collapse
Affiliation(s)
- Fabio Cumbo
- Institute for Systems Analysis and Computer Science 'Antonio Ruberti', National Research Council of Italy, 00185 Rome, Italy.,Department of Engineering, Third University of Rome, 00146 Rome, Italy.,SYSBIO.IT-Centre of Systems Biology, 20126 Milan, Italy
| | - Davide Vergni
- Institute for Applied Mathematics 'Mauro Picone', National Research Council of Italy, 00185 Rome, Italy
| | - Daniele Santoni
- Institute for Systems Analysis and Computer Science 'Antonio Ruberti', National Research Council of Italy, 00185 Rome, Italy
| |
Collapse
|
94
|
NFIA co-localizes with PPARγ and transcriptionally controls the brown fat gene program. Nat Cell Biol 2017; 19:1081-1092. [PMID: 28812581 DOI: 10.1038/ncb3590] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 07/10/2017] [Indexed: 12/17/2022]
Abstract
Brown fat dissipates energy as heat and protects against obesity. Here, we identified nuclear factor I-A (NFIA) as a transcriptional regulator of brown fat by a genome-wide open chromatin analysis of murine brown and white fat followed by motif analysis of brown-fat-specific open chromatin regions. NFIA and the master transcriptional regulator of adipogenesis, PPARγ, co-localize at the brown-fat-specific enhancers. Moreover, the binding of NFIA precedes and facilitates the binding of PPARγ, leading to increased chromatin accessibility and active transcription. Introduction of NFIA into myoblasts results in brown adipocyte differentiation. Conversely, the brown fat of NFIA-knockout mice displays impaired expression of the brown-fat-specific genes and reciprocal elevation of muscle genes. Finally, expression of NFIA and the brown-fat-specific genes is positively correlated in human brown fat. These results indicate that NFIA activates the cell-type-specific enhancers and facilitates the binding of PPARγ to control the brown fat gene program.
Collapse
|
95
|
Unraveling a tumor type-specific regulatory core underlying E2F1-mediated epithelial-mesenchymal transition to predict receptor protein signatures. Nat Commun 2017; 8:198. [PMID: 28775339 PMCID: PMC5543083 DOI: 10.1038/s41467-017-00268-2] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 06/15/2017] [Indexed: 12/18/2022] Open
Abstract
Cancer is a disease of subverted regulatory pathways. In this paper, we reconstruct the regulatory network around E2F, a family of transcription factors whose deregulation has been associated to cancer progression, chemoresistance, invasiveness, and metastasis. We integrate gene expression profiles of cancer cell lines from two E2F1-driven highly aggressive bladder and breast tumors, and use network analysis methods to identify the tumor type-specific core of the network. By combining logic-based network modeling, in vitro experimentation, and gene expression profiles from patient cohorts displaying tumor aggressiveness, we identify and experimentally validate distinctive, tumor type-specific signatures of receptor proteins associated to epithelial-mesenchymal transition in bladder and breast cancer. Our integrative network-based methodology, exemplified in the case of E2F1-induced aggressive tumors, has the potential to support the design of cohort- as well as tumor type-specific treatments and ultimately, to fight metastasis and therapy resistance.Deregulation of E2F family transcription factors is associated with cancer progression and metastasis. Here, the authors construct a map of the regulatory network around the E2F family, and using gene expression profiles, identify tumour type-specific regulatory cores and receptor expression signatures associated with epithelial-mesenchymal transition in bladder and breast cancer.
Collapse
|
96
|
Liu S, Zibetti C, Wan J, Wang G, Blackshaw S, Qian J. Assessing the model transferability for prediction of transcription factor binding sites based on chromatin accessibility. BMC Bioinformatics 2017; 18:355. [PMID: 28750606 PMCID: PMC5530957 DOI: 10.1186/s12859-017-1769-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 07/19/2017] [Indexed: 12/04/2022] Open
Abstract
Background Computational prediction of transcription factor (TF) binding sites in different cell types is challenging. Recent technology development allows us to determine the genome-wide chromatin accessibility in various cellular and developmental contexts. The chromatin accessibility profiles provide useful information in prediction of TF binding events in various physiological conditions. Furthermore, ChIP-Seq analysis was used to determine genome-wide binding sites for a range of different TFs in multiple cell types. Integration of these two types of genomic information can improve the prediction of TF binding events. Results We assessed to what extent a model built upon on other TFs and/or other cell types could be used to predict the binding sites of TFs of interest. A random forest model was built using a set of cell type-independent features such as specific sequences recognized by the TFs and evolutionary conservation, as well as cell type-specific features derived from chromatin accessibility data. Our analysis suggested that the models learned from other TFs and/or cell lines performed almost as well as the model learned from the target TF in the cell type of interest. Interestingly, models based on multiple TFs performed better than single-TF models. Finally, we proposed a universal model, BPAC, which was generated using ChIP-Seq data from multiple TFs in various cell types. Conclusion Integrating chromatin accessibility information with sequence information improves prediction of TF binding.The prediction of TF binding is transferable across TFs and/or cell lines suggesting there are a set of universal “rules”. A computational tool was developed to predict TF binding sites based on the universal “rules”. Electronic supplementary material The online version of this article (doi:10.1186/s12859-017-1769-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sheng Liu
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, 21287, MD, USA
| | - Cristina Zibetti
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, 21287, MD, USA
| | - Jun Wan
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, 21287, MD, USA
| | - Guohua Wang
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, 21287, MD, USA
| | - Seth Blackshaw
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, 21287, MD, USA.,Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, 21287, MD, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, 21287, MD, USA.,Centre for Human Systems Biology, Johns Hopkins University School of Medicine, Baltimore, 21287, MD, USA.,Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, 21287, MD, USA
| | - Jiang Qian
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, 21287, MD, USA.
| |
Collapse
|
97
|
A data analysis framework for biomedical big data: Application on mesoderm differentiation of human pluripotent stem cells. PLoS One 2017; 12:e0179613. [PMID: 28654683 PMCID: PMC5487013 DOI: 10.1371/journal.pone.0179613] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 05/31/2017] [Indexed: 12/16/2022] Open
Abstract
The development of high-throughput biomolecular technologies has resulted in generation of vast omics data at an unprecedented rate. This is transforming biomedical research into a big data discipline, where the main challenges relate to the analysis and interpretation of data into new biological knowledge. The aim of this study was to develop a framework for biomedical big data analytics, and apply it for analyzing transcriptomics time series data from early differentiation of human pluripotent stem cells towards the mesoderm and cardiac lineages. To this end, transcriptome profiling by microarray was performed on differentiating human pluripotent stem cells sampled at eleven consecutive days. The gene expression data was analyzed using the five-stage analysis framework proposed in this study, including data preparation, exploratory data analysis, confirmatory analysis, biological knowledge discovery, and visualization of the results. Clustering analysis revealed several distinct expression profiles during differentiation. Genes with an early transient response were strongly related to embryonic- and mesendoderm development, for example CER1 and NODAL. Pluripotency genes, such as NANOG and SOX2, exhibited substantial downregulation shortly after onset of differentiation. Rapid induction of genes related to metal ion response, cardiac tissue development, and muscle contraction were observed around day five and six. Several transcription factors were identified as potential regulators of these processes, e.g. POU1F1, TCF4 and TBP for muscle contraction genes. Pathway analysis revealed temporal activity of several signaling pathways, for example the inhibition of WNT signaling on day 2 and its reactivation on day 4. This study provides a comprehensive characterization of biological events and key regulators of the early differentiation of human pluripotent stem cells towards the mesoderm and cardiac lineages. The proposed analysis framework can be used to structure data analysis in future research, both in stem cell differentiation, and more generally, in biomedical big data analytics.
Collapse
|
98
|
Ng HP, Jennings S, Wang J, Molina PE, Nelson S, Wang G. Non-canonical Glucocorticoid Receptor Transactivation of gilz by Alcohol Suppresses Cell Inflammatory Response. Front Immunol 2017. [PMID: 28638383 PMCID: PMC5461336 DOI: 10.3389/fimmu.2017.00661] [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] [Indexed: 12/27/2022] Open
Abstract
Acute alcohol exposure suppresses cell inflammatory response. The underlying mechanism has not been fully defined. Here we report that alcohol was able to activate glucocorticoid receptor (GR) signaling in the absence of glucocorticoids (GCs) and upregulated glucocorticoid-induced leucine zipper (gilz), a prominent GC-responsive gene. Such a non-canonical activation of GR was not blocked by mifepristone, a potent GC competitor. The proximal promoter of gilz, encompassing five GC-responsive elements (GREs), was incorporated and tested in a luciferase reporter system. Deletion and/or mutation of the GREs abrogated the promoter responsiveness to alcohol. Thus, the GR–GRE interaction transduced the alcohol action on gilz. Alcohol induced GR nuclear translocation, which was enhanced by the alcohol dehydrogenase inhibitor fomepizole, suggesting that it was alcohol, not its metabolites, that engendered the effect. Gel mobility shift assay showed that unliganded GR was able to bind GREs and such interaction withstood clinically relevant levels of alcohol. GR knockout via CRISPR/Cas9 gene targeting or GILZ depletion via small RNA interference diminished alcohol suppression of cell inflammatory response to LPS. Thus, a previously unrecognized, non-canonical GR activation of gilz is involved in alcohol modulation of cell immune response.
Collapse
Affiliation(s)
- Hang Pong Ng
- Alcohol and Drug Abuse Center, Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Scott Jennings
- Alcohol and Drug Abuse Center, Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Jack Wang
- Alcohol and Drug Abuse Center, Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Patricia E Molina
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Steve Nelson
- Department of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Guoshun Wang
- Alcohol and Drug Abuse Center, Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| |
Collapse
|
99
|
Genome-wide Analysis of STAT3-Mediated Transcription during Early Human Th17 Cell Differentiation. Cell Rep 2017; 19:1888-1901. [DOI: 10.1016/j.celrep.2017.05.013] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 12/23/2016] [Accepted: 05/02/2017] [Indexed: 01/24/2023] Open
|
100
|
Xiao X, Chang H, Li M. Molecular mechanisms underlying noncoding risk variations in psychiatric genetic studies. Mol Psychiatry 2017; 22:497-511. [PMID: 28044063 PMCID: PMC5378805 DOI: 10.1038/mp.2016.241] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 11/08/2016] [Accepted: 11/14/2016] [Indexed: 12/18/2022]
Abstract
Recent large-scale genetic approaches such as genome-wide association studies have allowed the identification of common genetic variations that contribute to risk architectures of psychiatric disorders. However, most of these susceptibility variants are located in noncoding genomic regions that usually span multiple genes. As a result, pinpointing the precise variant(s) and biological mechanisms accounting for the risk remains challenging. By reviewing recent progresses in genetics, functional genomics and neurobiology of psychiatric disorders, as well as gene expression analyses of brain tissues, here we propose a roadmap to characterize the roles of noncoding risk loci in the pathogenesis of psychiatric illnesses (that is, identifying the underlying molecular mechanisms explaining the genetic risk conferred by those genomic loci, and recognizing putative functional causative variants). This roadmap involves integration of transcriptomic data, epidemiological and bioinformatic methods, as well as in vitro and in vivo experimental approaches. These tools will promote the translation of genetic discoveries to physiological mechanisms, and ultimately guide the development of preventive, therapeutic and prognostic measures for psychiatric disorders.
Collapse
Affiliation(s)
- X Xiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China
| | - H Chang
- Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China
| | - M Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China
| |
Collapse
|