1
|
Li J, Huang HY, Lin YCD, Zuo H, Tang Y, Huang HD. Cinnamomi ramulus inhibits cancer cells growth by inducing G2/M arrest. Front Pharmacol 2023; 14:1121799. [PMID: 37007025 PMCID: PMC10063822 DOI: 10.3389/fphar.2023.1121799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 02/13/2023] [Indexed: 03/19/2023] Open
Abstract
Introduction: Cinnamomi ramulus (CR) is one of the most widely used traditional Chinese medicine (TCM) with anti-cancer effects. Analyzing transcriptomic responses of different human cell lines to TCM treatment is a promising approach to understand the unbiased mechanism of TCM. Methods: This study treated ten cancer cell lines with different CR concentrations, followed by mRNA sequencing. Differential expression (DE) analysis and gene set enrichment analysis (GSEA) were utilized to analyze transcriptomic data. Finally, the in silico screening results were verified by in vitro experiments. Results: Both DE and GSEA analysis suggested the Cell cycle pathway was the most perturbated pathway by CR across these cell lines. By analyzing the clinical significance and prognosis of G2/M related genes (PLK1, CDK1, CCNB1, and CCNB2) in various cancer tissues, we found that they were up-regulated in most cancer types, and their down-regulation showed better overall survival rates in cancer patients. Finally, in vitro experiments validation on A549, Hep G2, and HeLa cells suggested that CR can inhibit cell growth by suppressing the PLK1/CDK1/ Cyclin B axis. Discussion: This is the first study to apply transcriptomic analysis to investigate the cancer cell growth inhibition of CR on various human cancer cell lines. The core effect of CR on ten cancer cell lines is to induce G2/M arrest by inhibiting the PLK1/CDK1/Cyclin B axis.
Collapse
Affiliation(s)
- Jing Li
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Hsi-Yuan Huang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Yang-Chi-Dung Lin
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Huali Zuo
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Yun Tang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Hsien-Da Huang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| |
Collapse
|
2
|
Shakola F, Palejev D, Ivanov I. A Framework for Comparison and Assessment of Synthetic RNA-Seq Data. Genes (Basel) 2022; 13:2362. [PMID: 36553629 PMCID: PMC9778097 DOI: 10.3390/genes13122362] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/05/2022] [Accepted: 12/06/2022] [Indexed: 12/16/2022] Open
Abstract
The ever-growing number of methods for the generation of synthetic bulk and single cell RNA-seq data have multiple and diverse applications. They are often aimed at benchmarking bioinformatics algorithms for purposes such as sample classification, differential expression analysis, correlation and network studies and the optimization of data integration and normalization techniques. Here, we propose a general framework to compare synthetically generated RNA-seq data and select a data-generating tool that is suitable for a set of specific study goals. As there are multiple methods for synthetic RNA-seq data generation, researchers can use the proposed framework to make an informed choice of an RNA-seq data simulation algorithm and software that are best suited for their specific scientific questions of interest.
Collapse
Affiliation(s)
- Felitsiya Shakola
- GATE Institute, Sofia University, 125 Tsarigradsko Shosse, Bl. 2, 1113 Sofia, Bulgaria
| | - Dean Palejev
- Institute of Mathematics and Informatics, Bulgarian Academy of Sciences, Acad. G. Bonchev St., Bl. 8, 1113 Sofia, Bulgaria
| | - Ivan Ivanov
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
3
|
Das S, Rai SN. Statistical Approach of Gene Set Analysis with Quantitative Trait Loci for Crop Gene Expression Studies. ENTROPY (BASEL, SWITZERLAND) 2021; 23:945. [PMID: 34441085 PMCID: PMC8391627 DOI: 10.3390/e23080945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/19/2021] [Accepted: 07/21/2021] [Indexed: 11/16/2022]
Abstract
Genome-wide expression study is a powerful genomic technology to quantify expression dynamics of genes in a genome. In gene expression study, gene set analysis has become the first choice to gain insights into the underlying biology of diseases or stresses in plants. It also reduces the complexity of statistical analysis and enhances the explanatory power of the obtained results from the primary downstream differential expression analysis. The gene set analysis approaches are well developed in microarrays and RNA-seq gene expression data analysis. These approaches mainly focus on analyzing the gene sets with gene ontology or pathway annotation data. However, in plant biology, such methods may not establish any formal relationship between the genotypes and the phenotypes, as most of the traits are quantitative and controlled by polygenes. The existing Quantitative Trait Loci (QTL)-based gene set analysis approaches only focus on the over-representation analysis of the selected genes while ignoring their associated gene scores. Therefore, we developed an innovative statistical approach, GSQSeq, to analyze the gene sets with trait enriched QTL data. This approach considers the associated differential expression scores of genes while analyzing the gene sets. The performance of the developed method was tested on five different crop gene expression datasets obtained from real crop gene expression studies. Our analytical results indicated that the trait-specific analysis of gene sets was more robust and successful through the proposed approach than existing techniques. Further, the developed method provides a valuable platform for integrating the gene expression data with QTL data.
Collapse
Affiliation(s)
- Samarendra Das
- Division of Statistical Genetics, ICAR-Indian Agricultural Statistics Research Institute, PUSA, New Delhi 110012, India;
- Biostatistics and Bioinformatics Facility, JG Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
- School of Interdisciplinary and Graduate Studies, University of Louisville, Louisville, KY 40292, USA
| | - Shesh N. Rai
- Biostatistics and Bioinformatics Facility, JG Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
- School of Interdisciplinary and Graduate Studies, University of Louisville, Louisville, KY 40292, USA
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA
- Alcohol Research Center, University of Louisville, Louisville, KY 40202, USA
- Hepatobiology and Toxicology Center, University of Louisville, Louisville, KY 40202, USA
- Department of Bioinformatics and Biostatistics, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
4
|
Data-driven detection of subtype-specific differentially expressed genes. Sci Rep 2021; 11:332. [PMID: 33432005 PMCID: PMC7801594 DOI: 10.1038/s41598-020-79704-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 12/11/2020] [Indexed: 11/08/2022] Open
Abstract
Among multiple subtypes of tissue or cell, subtype-specific differentially-expressed genes (SDEGs) are defined as being most-upregulated in only one subtype but not in any other. Detecting SDEGs plays a critical role in the molecular characterization and deconvolution of multicellular complex tissues. Classic differential analysis assumes a null hypothesis whose test statistic is not subtype-specific, thus can produce a high false positive rate and/or lower detection power. Here we first introduce a One-Versus-Everyone Fold Change (OVE-FC) test for detecting SDEGs. We then propose a scaled test statistic (OVE-sFC) for assessing the statistical significance of SDEGs that applies a mixture null distribution model and a tailored permutation test. The OVE-FC/sFC test was validated on both type 1 error rate and detection power using extensive simulation data sets generated from real gene expression profiles of purified subtype samples. The OVE-FC/sFC test was then applied to two benchmark gene expression data sets of purified subtype samples and detected many known or previously unknown SDEGs. Subsequent supervised deconvolution results on synthesized bulk expression data, obtained using the SDEGs detected from the independent purified expression data by the OVE-FC/sFC test, showed superior performance in deconvolution accuracy when compared with popular peer methods.
Collapse
|
5
|
Fanidis D, Moulos P. Integrative, normalization-insusceptible statistical analysis of RNA-Seq data, with improved differential expression and unbiased downstream functional analysis. Brief Bioinform 2020; 22:5890504. [PMID: 32778872 DOI: 10.1093/bib/bbaa156] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/04/2020] [Accepted: 06/19/2020] [Indexed: 12/11/2022] Open
Abstract
The study of differential gene expression patterns through RNA-Seq comprises a routine task in the daily lives of molecular bioscientists, who produce vast amounts of data requiring proper management and analysis. Despite widespread use, there are still no widely accepted golden standards for the normalization and statistical analysis of RNA-Seq data, and critical biases, such as gene lengths and problems in the detection of certain types of molecules, remain largely unaddressed. Stimulated by these unmet needs and the lack of in-depth research into the potential of combinatorial methods to enhance the analysis of differential gene expression, we had previously introduced the PANDORA P-value combination algorithm while presenting evidence for PANDORA's superior performance in optimizing the tradeoff between precision and sensitivity. In this article, we present the next generation of the algorithm along with a more in-depth investigation of its capabilities to effectively analyze RNA-Seq data. In particular, we show that PANDORA-reported lists of differentially expressed genes are unaffected by biases introduced by different normalization methods, while, at the same time, they comprise a reliable input option for downstream pathway analysis. Additionally, PANDORA outperforms other methods in detecting differential expression patterns in certain transcript types, including long non-coding RNAs.
Collapse
|
6
|
Fifteen Years of Gene Set Analysis for High-Throughput Genomic Data: A Review of Statistical Approaches and Future Challenges. ENTROPY 2020; 22:e22040427. [PMID: 33286201 PMCID: PMC7516904 DOI: 10.3390/e22040427] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/18/2020] [Accepted: 04/03/2020] [Indexed: 12/22/2022]
Abstract
Over the last decade, gene set analysis has become the first choice for gaining insights into underlying complex biology of diseases through gene expression and gene association studies. It also reduces the complexity of statistical analysis and enhances the explanatory power of the obtained results. Although gene set analysis approaches are extensively used in gene expression and genome wide association data analysis, the statistical structure and steps common to these approaches have not yet been comprehensively discussed, which limits their utility. In this article, we provide a comprehensive overview, statistical structure and steps of gene set analysis approaches used for microarrays, RNA-sequencing and genome wide association data analysis. Further, we also classify the gene set analysis approaches and tools by the type of genomic study, null hypothesis, sampling model and nature of the test statistic, etc. Rather than reviewing the gene set analysis approaches individually, we provide the generation-wise evolution of such approaches for microarrays, RNA-sequencing and genome wide association studies and discuss their relative merits and limitations. Here, we identify the key biological and statistical challenges in current gene set analysis, which will be addressed by statisticians and biologists collectively in order to develop the next generation of gene set analysis approaches. Further, this study will serve as a catalog and provide guidelines to genome researchers and experimental biologists for choosing the proper gene set analysis approach based on several factors.
Collapse
|
7
|
Geistlinger L, Csaba G, Santarelli M, Ramos M, Schiffer L, Turaga N, Law C, Davis S, Carey V, Morgan M, Zimmer R, Waldron L. Toward a gold standard for benchmarking gene set enrichment analysis. Brief Bioinform 2020; 22:545-556. [PMID: 32026945 PMCID: PMC7820859 DOI: 10.1093/bib/bbz158] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 10/11/2019] [Accepted: 11/09/2019] [Indexed: 12/22/2022] Open
Abstract
MOTIVATION Although gene set enrichment analysis has become an integral part of high-throughput gene expression data analysis, the assessment of enrichment methods remains rudimentary and ad hoc. In the absence of suitable gold standards, evaluations are commonly restricted to selected datasets and biological reasoning on the relevance of resulting enriched gene sets. RESULTS We develop an extensible framework for reproducible benchmarking of enrichment methods based on defined criteria for applicability, gene set prioritization and detection of relevant processes. This framework incorporates a curated compendium of 75 expression datasets investigating 42 human diseases. The compendium features microarray and RNA-seq measurements, and each dataset is associated with a precompiled GO/KEGG relevance ranking for the corresponding disease under investigation. We perform a comprehensive assessment of 10 major enrichment methods, identifying significant differences in runtime and applicability to RNA-seq data, fraction of enriched gene sets depending on the null hypothesis tested and recovery of the predefined relevance rankings. We make practical recommendations on how methods originally developed for microarray data can efficiently be applied to RNA-seq data, how to interpret results depending on the type of gene set test conducted and which methods are best suited to effectively prioritize gene sets with high phenotype relevance. AVAILABILITY http://bioconductor.org/packages/GSEABenchmarkeR. CONTACT ludwig.geistlinger@sph.cuny.edu.
Collapse
Affiliation(s)
- Ludwig Geistlinger
- Graduate School of Public Health and Health Policy, City University of New York, New York, NY 10027, USA
| | - Gergely Csaba
- Institute for Implementation Science and Population Health, City University of New York, New York, NY 10027, USA
| | - Mara Santarelli
- Institute for Bioinformatics, Ludwig-Maximilians-Universität München, 80333 Munich, Germany
| | - Marcel Ramos
- Roswell Park Cancer Institute, Buffalo, NY 14203, USA
| | - Lucas Schiffer
- Graduate School of Arts and Sciences, Boston University, Boston, MA 02215, USA
| | - Nitesh Turaga
- Epigenetics and Development Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Charity Law
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Sean Davis
- Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | | | | | | | - Levi Waldron
- Graduate School of Public Health and Health Policy, City University of New York, New York, NY 10027, USA
| |
Collapse
|
8
|
Glazko G, Zybailov B, Emmert-Streib F, Baranova A, Rahmatallah Y. Proteome-transcriptome alignment of molecular portraits achieved by self-contained gene set analysis: Consensus colon cancer subtypes case study. PLoS One 2019; 14:e0221444. [PMID: 31437237 PMCID: PMC6705791 DOI: 10.1371/journal.pone.0221444] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 08/06/2019] [Indexed: 01/10/2023] Open
Abstract
Gene set analysis (GSA) has become the common methodology for analyzing transcriptomics data. However, self-contained GSA techniques are rarely, if ever, used for proteomics data analysis. Here we present a self-contained proteome level GSA of four consensus molecular subtypes (CMSs) previously established by transcriptome dissection of colon carcinoma specimens. Despite notable difference in structure of proteomics and transcriptomics data, many pathway-wide characteristic features of CMSs found at the mRNA level were reproduced at the protein level. In particular, CMS1 features show heavy involvement of immune system as well as the pathways related to mismatch repair, DNA replication and functioning of proteasome, while CMS4 tumors upregulate complement pathway and proteins participating in epithelial-to-mesenchymal transition (EMT). In addition, protein level GSA yielded a set of novel observations visible at the proteome, but not at the transcriptome level, including possible involvement of major histocompatibility complex II (MHC-II) antigens in the known immunogenicity of CMS1 and a connection between cholesterol trafficking and the regulation of Integrin-linked kinase (ILK) in CMS3. Overall, this study proves utility of self-contained GSA approaches as a critical tool for analyzing proteomics data in general and dissecting protein-level molecular portraits of human tumors in particular.
Collapse
Affiliation(s)
- Galina Glazko
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR, United States of America
| | - Boris Zybailov
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, United States of America
| | - Frank Emmert-Streib
- Computational Medicine and Statistical Learning Laboratory, Tampere University of Technology, Korkeakoulunkatu, Tampere, Finland FI
| | - Ancha Baranova
- School of Systems Biology, George Mason University, Manassas VA, United States of America
- Research Center for Medical Genetics, Moscow, Russia
| | - Yasir Rahmatallah
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR, United States of America
| |
Collapse
|
9
|
Ebrahimpoor M, Spitali P, Hettne K, Tsonaka R, Goeman J. Simultaneous Enrichment Analysis of all Possible Gene-sets: Unifying Self-Contained and Competitive Methods. Brief Bioinform 2019; 21:1302-1312. [PMID: 31297505 PMCID: PMC7373179 DOI: 10.1093/bib/bbz074] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 05/28/2019] [Accepted: 05/28/2019] [Indexed: 01/23/2023] Open
Abstract
Studying sets of genomic features is increasingly popular in genomics, proteomics and metabolomics since analyzing at set level not only creates a natural connection to biological knowledge but also offers more statistical power. Currently, there are two gene-set testing approaches, self-contained and competitive, both of which have their advantages and disadvantages, but neither offers the final solution. We introduce simultaneous enrichment analysis (SEA), a new approach for analysis of feature sets in genomics and other omics based on a new unified null hypothesis, which includes the self-contained and competitive null hypotheses as special cases. We employ closed testing using Simes tests to test this new hypothesis. For every feature set, the proportion of active features is estimated, and a confidence bound is provided. Also, for every unified null hypotheses, a \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{upgreek}
\usepackage{mathrsfs}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
}{}$P$\end{document}-value is calculated, which is adjusted for family-wise error rate. SEA does not need to assume that the features are independent. Moreover, users are allowed to choose the feature set(s) of interest after observing the data. We develop a novel pipeline and apply it on RNA-seq data of dystrophin-deficient mdx mice, showcasing the flexibility of the method. Finally, the power properties of the method are evaluated through simulation studies.
Collapse
Affiliation(s)
- Mitra Ebrahimpoor
- Medical statistics, Department of Biomedical Data Science, Leiden University Medical Center, Leiden, The Netherlands
| | - Pietro Spitali
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Kristina Hettne
- Medical statistics, Department of Biomedical Data Science, Leiden University Medical Center, Leiden, The Netherlands
| | - Roula Tsonaka
- Medical statistics, Department of Biomedical Data Science, Leiden University Medical Center, Leiden, The Netherlands
| | - Jelle Goeman
- Medical statistics, Department of Biomedical Data Science, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
10
|
Lin SJ, Lu TP, Yu QY, Hsiao CK. Probabilistic prioritization of candidate pathway association with pathway score. BMC Bioinformatics 2018; 19:391. [PMID: 30355338 PMCID: PMC6201593 DOI: 10.1186/s12859-018-2411-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 10/05/2018] [Indexed: 01/12/2023] Open
Abstract
Background Current methods for gene-set or pathway analysis are usually designed to test the enrichment of a single gene-set. Once the analysis is carried out for each of the sets under study, a list of significant sets can be obtained. However, if one wishes to further prioritize the importance or strength of association of these sets, no such quantitative measure is available. Using the magnitude of p-value to rank the pathways may not be appropriate because p-value is not a measure for strength of significance. In addition, when testing each pathway, these analyses are often implicitly affected by the number of differentially expressed genes included in the set and/or affected by the dependence among genes. Results Here we propose a two-stage procedure to prioritize the pathways/gene-sets. In the first stage we develop a pathway-level measure with three properties. First, it contains all genes (differentially expressed or not) in the same set, and summarizes the collective effect of all genes per sample. Second, this pathway score accounts for the correlation between genes by synchronizing their correlation directions. Third, the score includes a rank transformation to enhance the variation among samples as well as to avoid the influence of extreme heterogeneity among genes. In the second stage, all scores are included simultaneously in a Bayesian logistic regression model which can evaluate the strength of association for each set and rank the sets based on posterior probabilities. Simulations from Gaussian distributions and human microarray data, and a breast cancer study with RNA-Seq are considered for demonstration and comparison with other existing methods. Conclusions The proposed summary pathway score provides for each sample an overall evaluation of gene expression in a gene-set. It demonstrates the advantages of including all genes in the set and the synchronization of correlation direction. The simultaneous utilization of all pathway-level scores in a Bayesian model not only offers a probabilistic evaluation and ranking of the pathway association but also presents good accuracy in identifying the top-ranking pathways. The resulting recommendation list of ranked pathways can be a reference for potential target therapy or for future allocation of research resources. Electronic supplementary material The online version of this article (10.1186/s12859-018-2411-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shu-Ju Lin
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, 10055, Taiwan
| | - Tzu-Pin Lu
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, 10055, Taiwan.,Bioinformatics and Biostatistics Core, Center of Genomic Medicine, National Taiwan University, Taipei, 10055, Taiwan
| | - Qi-You Yu
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, 10055, Taiwan
| | - Chuhsing Kate Hsiao
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, 10055, Taiwan. .,Bioinformatics and Biostatistics Core, Center of Genomic Medicine, National Taiwan University, Taipei, 10055, Taiwan.
| |
Collapse
|
11
|
Gover-Proaktor A, Granot G, Pasmanik-Chor M, Pasvolsky O, Shapira S, Raz O, Raanani P, Leader A. Bosutinib, dasatinib, imatinib, nilotinib, and ponatinib differentially affect the vascular molecular pathways and functionality of human endothelial cells. Leuk Lymphoma 2018; 60:189-199. [PMID: 29741440 DOI: 10.1080/10428194.2018.1466294] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
The tyrosine kinase inhibitors (TKIs), nilotinib, ponatinib, and dasatinib (but not bosutinib or imatinib), are associated with vascular adverse events (VAEs) in chronic myeloid leukemia (CML). Though the mechanism is inadequately understood, an effect on vascular cells has been suggested. We investigated the effect of imatinib, nilotinib, dasatinib, bosutinib, and ponatinib on tube formation, cell viability, and gene expression of human vascular endothelial cells (HUVECs). We found a distinct genetic profile in HUVECs treated with dasatinib, ponatinib, and nilotinib compared to bosutinib and imatinib, who resembled untreated samples. However, unique gene expression and molecular pathway alterations were detected between dasatinib, ponatinib, and nilotinib. Angiogenesis/blood vessel-related pathways and HUVEC function (tube formation/viability) were adversely affected by dasatinib, ponatinib, and nilotinib but not by imatinib or bosutinib. These results correspond to the differences in VAE profiles of these TKIs, support a direct effect on vascular cells, and provide direction for future research.
Collapse
Affiliation(s)
- Ayala Gover-Proaktor
- a Felsenstein Medical Research Center , Beilinson Hospital, Rabin Medical Center , Petah-Tikva , Israel
| | - Galit Granot
- a Felsenstein Medical Research Center , Beilinson Hospital, Rabin Medical Center , Petah-Tikva , Israel
| | - Metsada Pasmanik-Chor
- b Bioinformatics Unit, G.S.W. Faculty of Life Sciences , Tel Aviv University , Tel Aviv , Israel
| | - Oren Pasvolsky
- c Institute of Hematology, Davidoff Cancer Center , Beilinson Hospital, Rabin Medical Center , Petah-Tikva , Israel.,d Sackler School of Medicine , Tel Aviv University , Tel Aviv , Israel
| | - Saar Shapira
- a Felsenstein Medical Research Center , Beilinson Hospital, Rabin Medical Center , Petah-Tikva , Israel.,d Sackler School of Medicine , Tel Aviv University , Tel Aviv , Israel
| | - Oshrat Raz
- a Felsenstein Medical Research Center , Beilinson Hospital, Rabin Medical Center , Petah-Tikva , Israel
| | - Pia Raanani
- c Institute of Hematology, Davidoff Cancer Center , Beilinson Hospital, Rabin Medical Center , Petah-Tikva , Israel.,d Sackler School of Medicine , Tel Aviv University , Tel Aviv , Israel
| | - Avi Leader
- c Institute of Hematology, Davidoff Cancer Center , Beilinson Hospital, Rabin Medical Center , Petah-Tikva , Israel.,d Sackler School of Medicine , Tel Aviv University , Tel Aviv , Israel
| |
Collapse
|
12
|
Abstract
The analysis of gene sets (in a form of functionally related genes or pathways) has become the method of choice for extracting the strongest signals from omics data. The motivation behind using gene sets instead of individual genes is two-fold. First, this approach incorporates pre-existing biological knowledge into the analysis and facilitates the interpretation of experimental results. Second, it employs a statistical hypotheses testing framework. Here, we briefly review main Gene Set Analysis (GSA) approaches for testing differential expression of gene sets and several GSA approaches for testing statistical hypotheses beyond differential expression that allow extracting additional biological information from the data. We distinguish three major types of GSA approaches testing: (1) differential expression (DE), (2) differential variability (DV), and (3) differential co-expression (DC) of gene sets between two phenotypes. We also present comparative power analysis and Type I error rates for different approaches in each major type of GSA on simulated data. Our evaluation presents a concise guideline for selecting GSA approaches best performing under particular experimental settings. The value of the three major types of GSA approaches is illustrated with real data example. While being applied to the same data set, major types of GSA approaches result in complementary biological information.
Collapse
|
13
|
Yoon S, Nam D. Gene dispersion is the key determinant of the read count bias in differential expression analysis of RNA-seq data. BMC Genomics 2017; 18:408. [PMID: 28545404 PMCID: PMC5445461 DOI: 10.1186/s12864-017-3809-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 05/21/2017] [Indexed: 01/05/2023] Open
Abstract
Background In differential expression analysis of RNA-sequencing (RNA-seq) read count data for two sample groups, it is known that highly expressed genes (or longer genes) are more likely to be differentially expressed which is called read count bias (or gene length bias). This bias had great effect on the downstream Gene Ontology over-representation analysis. However, such a bias has not been systematically analyzed for different replicate types of RNA-seq data. Results We show that the dispersion coefficient of a gene in the negative binomial modeling of read counts is the critical determinant of the read count bias (and gene length bias) by mathematical inference and tests for a number of simulated and real RNA-seq datasets. We demonstrate that the read count bias is mostly confined to data with small gene dispersions (e.g., technical replicates and some of genetically identical replicates such as cell lines or inbred animals), and many biological replicate data from unrelated samples do not suffer from such a bias except for genes with some small counts. It is also shown that the sample-permuting GSEA method yields a considerable number of false positives caused by the read count bias, while the preranked method does not. Conclusion We showed the small gene variance (similarly, dispersion) is the main cause of read count bias (and gene length bias) for the first time and analyzed the read count bias for different replicate types of RNA-seq data and its effect on gene-set enrichment analysis. Electronic supplementary material The online version of this article (doi:10.1186/s12864-017-3809-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sora Yoon
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Dougu Nam
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea. .,Department of Mathematical Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea.
| |
Collapse
|
14
|
Lei M, Xu J, Huang LC, Wang L, Li J. Network module-based model in the differential expression analysis for RNA-seq. Bioinformatics 2017; 33:2699-2705. [DOI: 10.1093/bioinformatics/btx214] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 04/11/2017] [Indexed: 12/16/2022] Open
Affiliation(s)
- Mingli Lei
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Jia Xu
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Li-Ching Huang
- Center for Quantitative Sciences, Vanderbilt University, Nashville, TN, USA
| | - Lily Wang
- Department of Public Health Sciences, Division of Biostatistics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jing Li
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| |
Collapse
|
15
|
Rahmatallah Y, Zybailov B, Emmert-Streib F, Glazko G. GSAR: Bioconductor package for Gene Set analysis in R. BMC Bioinformatics 2017; 18:61. [PMID: 28118818 PMCID: PMC5259853 DOI: 10.1186/s12859-017-1482-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 01/10/2017] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Gene set analysis (in a form of functionally related genes or pathways) has become the method of choice for analyzing omics data in general and gene expression data in particular. There are many statistical methods that either summarize gene-level statistics for a gene set or apply a multivariate statistic that accounts for intergene correlations. Most available methods detect complex departures from the null hypothesis but lack the ability to identify the specific alternative hypothesis that rejects the null. RESULTS GSAR (Gene Set Analysis in R) is an open-source R/Bioconductor software package for gene set analysis (GSA). It implements self-contained multivariate non-parametric statistical methods testing a complex null hypothesis against specific alternatives, such as differences in mean (shift), variance (scale), or net correlation structure. The package also provides a graphical visualization tool, based on the union of two minimum spanning trees, for correlation networks to examine the change in the correlation structures of a gene set between two conditions and highlight influential genes (hubs). CONCLUSIONS Package GSAR provides a set of multivariate non-parametric statistical methods that test a complex null hypothesis against specific alternatives. The methods in package GSAR are applicable to any type of omics data that can be represented in a matrix format. The package, with detailed instructions and examples, is freely available under the GPL (> = 2) license from the Bioconductor web site.
Collapse
Affiliation(s)
- Yasir Rahmatallah
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.
| | - Boris Zybailov
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Frank Emmert-Streib
- Computational Medicine and Statistical Learning Laboratory, Tampere University of Technology, Korkeakoulunkatu 1, Tampere, FI-33720, Finland
| | - Galina Glazko
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| |
Collapse
|
16
|
Zoh RS, Mallick B, Ivanov I, Baladandayuthapani V, Manyam G, Chapkin RS, Lampe JW, Carroll RJ. PCAN: Probabilistic correlation analysis of two non-normal data sets. Biometrics 2016; 72:1358-1368. [PMID: 27037601 DOI: 10.1111/biom.12516] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 12/01/2015] [Accepted: 02/01/2016] [Indexed: 11/29/2022]
Abstract
Most cancer research now involves one or more assays profiling various biological molecules, e.g., messenger RNA and micro RNA, in samples collected on the same individuals. The main interest with these genomic data sets lies in the identification of a subset of features that are active in explaining the dependence between platforms. To quantify the strength of the dependency between two variables, correlation is often preferred. However, expression data obtained from next-generation sequencing platforms are integer with very low counts for some important features. In this case, the sample Pearson correlation is not a valid estimate of the true correlation matrix, because the sample correlation estimate between two features/variables with low counts will often be close to zero, even when the natural parameters of the Poisson distribution are, in actuality, highly correlated. We propose a model-based approach to correlation estimation between two non-normal data sets, via a method we call Probabilistic Correlations ANalysis, or PCAN. PCAN takes into consideration the distributional assumption about both data sets and suggests that correlations estimated at the model natural parameter level are more appropriate than correlations estimated directly on the observed data. We demonstrate through a simulation study that PCAN outperforms other standard approaches in estimating the true correlation between the natural parameters. We then apply PCAN to the joint analysis of a microRNA (miRNA) and a messenger RNA (mRNA) expression data set from a squamous cell lung cancer study, finding a large number of negative correlation pairs when compared to the standard approaches.
Collapse
Affiliation(s)
- Roger S Zoh
- Department of Epidemiology and Biostatistics, Texas A&M University, College Station, Texas, U.S.A
| | - Bani Mallick
- Department of Statistics, Texas A&M University, College Station, Texas, U.S.A
| | - Ivan Ivanov
- Department of Veterinary Medicine and Biomedical Sciences, Texas A&M University, Texas, U.S.A
| | | | - Ganiraju Manyam
- The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, Texas, U.S.A
| | - Robert S Chapkin
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, Texas, U.S.A
| | - Johanna W Lampe
- Department of Epidemiology, University of Washington and the Fred Hutchinson Cancer Research Center Seattle, Washington, U.S.A
| | - Raymond J Carroll
- Department of Statistics, Texas A&M University, College Station, Texas, U.S.A
| |
Collapse
|
17
|
Han Y, Gao S, Muegge K, Zhang W, Zhou B. Advanced Applications of RNA Sequencing and Challenges. Bioinform Biol Insights 2015; 9:29-46. [PMID: 26609224 PMCID: PMC4648566 DOI: 10.4137/bbi.s28991] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 09/30/2015] [Accepted: 10/02/2015] [Indexed: 12/18/2022] Open
Abstract
Next-generation sequencing technologies have revolutionarily advanced sequence-based research with the advantages of high-throughput, high-sensitivity, and high-speed. RNA-seq is now being used widely for uncovering multiple facets of transcriptome to facilitate the biological applications. However, the large-scale data analyses associated with RNA-seq harbors challenges. In this study, we present a detailed overview of the applications of this technology and the challenges that need to be addressed, including data preprocessing, differential gene expression analysis, alternative splicing analysis, variants detection and allele-specific expression, pathway analysis, co-expression network analysis, and applications combining various experimental procedures beyond the achievements that have been made. Specifically, we discuss essential principles of computational methods that are required to meet the key challenges of the RNA-seq data analyses, development of various bioinformatics tools, challenges associated with the RNA-seq applications, and examples that represent the advances made so far in the characterization of the transcriptome.
Collapse
Affiliation(s)
- Yixing Han
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Shouguo Gao
- Bioinformatics and Systems Biology Core, National Heart Lung Blood Institute, National Institutes of Health, Rockville Pike, Bethesda, MD, USA
| | - Kathrin Muegge
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA. ; Leidos Biomedical Research, Inc., Basic Science Program, Frederick National Laboratory, Frederick, MD, USA
| | - Wei Zhang
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Bing Zhou
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
18
|
Richards AJ, Herrel A, Bonneaud C. htsint: a Python library for sequencing pipelines that combines data through gene set generation. BMC Bioinformatics 2015; 16:307. [PMID: 26399714 PMCID: PMC4581156 DOI: 10.1186/s12859-015-0729-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 09/08/2015] [Indexed: 11/10/2022] Open
Abstract
Background Sequencing technologies provide a wealth of details in terms of genes, expression, splice variants, polymorphisms, and other features. A standard for sequencing analysis pipelines is to put genomic or transcriptomic features into a context of known functional information, but the relationships between ontology terms are often ignored. For RNA-Seq, considering genes and their genetic variants at the group level enables a convenient way to both integrate annotation data and detect small coordinated changes between experimental conditions, a known caveat of gene level analyses. Results We introduce the high throughput data integration tool, htsint, as an extension to the commonly used gene set enrichment frameworks. The central aim of htsint is to compile annotation information from one or more taxa in order to calculate functional distances among all genes in a specified gene space. Spectral clustering is then used to partition the genes, thereby generating functional modules. The gene space can range from a targeted list of genes, like a specific pathway, all the way to an ensemble of genomes. Given a collection of gene sets and a count matrix of transcriptomic features (e.g. expression, polymorphisms), the gene sets produced by htsint can be tested for ‘enrichment’ or conditional differences using one of a number of commonly available packages. Conclusion The database and bundled tools to generate functional modules were designed with sequencing pipelines in mind, but the toolkit nature of htsint allows it to also be used in other areas of genomics. The software is freely available as a Python library through GitHub at https://github.com/ajrichards/htsint.
Collapse
Affiliation(s)
- Adam J Richards
- Station d'Ecologie Expérimentale du CNRS, USR 2936, Route du CNRS, Moulis, 09200, France.
| | - Anthony Herrel
- UMR 7179 CNRS/MNHN, Département d'Ecologie et de Gestion de la Biodiversité 57 rue Cuvier, Case postale 55, Paris, 75231, France. .,Ghent University, Evolutionary Morphology of Vertebrates, K.L. Ledeganckstraat 35, Ghent, B-9000, Belgium.
| | - Camille Bonneaud
- Station d'Ecologie Expérimentale du CNRS, USR 2936, Route du CNRS, Moulis, 09200, France. .,Centre for Ecology & Conservation, College of Life and Environmental Sciences, University of Exeter, Penryn TR10 9FE, Cornwall, UK.
| |
Collapse
|
19
|
Rahmatallah Y, Emmert-Streib F, Glazko G. Gene set analysis approaches for RNA-seq data: performance evaluation and application guideline. Brief Bioinform 2015; 17:393-407. [PMID: 26342128 PMCID: PMC4870397 DOI: 10.1093/bib/bbv069] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Indexed: 11/15/2022] Open
Abstract
Transcriptome sequencing (RNA-seq) is gradually replacing microarrays for high-throughput studies of gene expression. The main challenge of analyzing microarray data is not in finding differentially expressed genes, but in gaining insights into the biological processes underlying phenotypic differences. To interpret experimental results from microarrays, gene set analysis (GSA) has become the method of choice, in particular because it incorporates pre-existing biological knowledge (in a form of functionally related gene sets) into the analysis. Here we provide a brief review of several statistically different GSA approaches (competitive and self-contained) that can be adapted from microarrays practice as well as those specifically designed for RNA-seq. We evaluate their performance (in terms of Type I error rate, power, robustness to the sample size and heterogeneity, as well as the sensitivity to different types of selection biases) on simulated and real RNA-seq data. Not surprisingly, the performance of various GSA approaches depends only on the statistical hypothesis they test and does not depend on whether the test was developed for microarrays or RNA-seq data. Interestingly, we found that competitive methods have lower power as well as robustness to the samples heterogeneity than self-contained methods, leading to poor results reproducibility. We also found that the power of unsupervised competitive methods depends on the balance between up- and down-regulated genes in tested gene sets. These properties of competitive methods have been overlooked before. Our evaluation provides a concise guideline for selecting GSA approaches, best performing under particular experimental settings in the context of RNA-seq.
Collapse
|
20
|
Stupnikov A, Glazko GV, Emmert-Streib F. Effects of subsampling on characteristics of RNA-seq data from triple-negative breast cancer patients. CHINESE JOURNAL OF CANCER 2015; 34:427-38. [PMID: 26253000 PMCID: PMC4593382 DOI: 10.1186/s40880-015-0040-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 07/03/2015] [Indexed: 11/29/2022]
Abstract
Background Data from RNA-seq experiments provide a wealth of information about the transcriptome of an organism. However, the analysis of such data is very demanding. In this study, we aimed to establish robust analysis procedures that can be used in clinical practice. Methods We studied RNA-seq data from triple-negative breast cancer patients. Specifically, we investigated the subsampling of RNA-seq data. Results The main results of our investigations are as follows: (1) the subsampling of RNA-seq data gave biologically realistic simulations of sequencing experiments with smaller sequencing depth but not direct scaling of count matrices; (2) the saturation of results required an average sequencing depth larger than 32 million reads and an individual sequencing depth larger than 46 million reads; and (3) for an abrogated feature selection, higher moments of the distribution of all expressed genes had a higher sensitivity for signal detection than the corresponding mean values. Conclusions Our results reveal important characteristics of RNA-seq data that must be understood before one can apply such an approach to translational medicine.
Collapse
Affiliation(s)
- Alexey Stupnikov
- Computational Biology and Machine Learning Laboratory, Faculty of Medicine, Health and Life Sciences, School of Medicine, Dentistry and Biomedical Sciences, Center for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7JL, UK.
| | - Galina V Glazko
- Division of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.
| | - Frank Emmert-Streib
- Computational Biology and Machine Learning Laboratory, Faculty of Medicine, Health and Life Sciences, School of Medicine, Dentistry and Biomedical Sciences, Center for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7JL, UK. .,Computational Medicine and Statistical Learning Laboratory, Department of Signal Processing, Tampere University of Technology, Korkeakoulunkatu 1, Tampere, 33720, Finland.
| |
Collapse
|
21
|
Linde J, Schulze S, Henkel SG, Guthke R. Data- and knowledge-based modeling of gene regulatory networks: an update. EXCLI JOURNAL 2015; 14:346-78. [PMID: 27047314 PMCID: PMC4817425 DOI: 10.17179/excli2015-168] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 02/10/2015] [Indexed: 02/01/2023]
Abstract
Gene regulatory network inference is a systems biology approach which predicts interactions between genes with the help of high-throughput data. In this review, we present current and updated network inference methods focusing on novel techniques for data acquisition, network inference assessment, network inference for interacting species and the integration of prior knowledge. After the advance of Next-Generation-Sequencing of cDNAs derived from RNA samples (RNA-Seq) we discuss in detail its application to network inference. Furthermore, we present progress for large-scale or even full-genomic network inference as well as for small-scale condensed network inference and review advances in the evaluation of network inference methods by crowdsourcing. Finally, we reflect the current availability of data and prior knowledge sources and give an outlook for the inference of gene regulatory networks that reflect interacting species, in particular pathogen-host interactions.
Collapse
Affiliation(s)
- Jörg Linde
- Research Group Systems Biology / Bioinformatics, Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knöll-Institute, Beutenbergstr. 11a, 07745 Jena, Germany
| | - Sylvie Schulze
- Research Group Systems Biology / Bioinformatics, Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knöll-Institute, Beutenbergstr. 11a, 07745 Jena, Germany
| | | | - Reinhard Guthke
- Research Group Systems Biology / Bioinformatics, Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knöll-Institute, Beutenbergstr. 11a, 07745 Jena, Germany
| |
Collapse
|