1
|
Kim SH, White Z, Gainullina A, Kang S, Kim J, Dominguez JR, Choi Y, Cabrera I, Plaster M, Takahama M, Czepielewski RS, Yeom J, Gunzer M, Hay N, David O, Chevrier N, Sano T, Kim KW. IL-10 sensing by lung interstitial macrophages prevents bacterial dysbiosis-driven pulmonary inflammation and maintains immune homeostasis. Immunity 2025; 58:1306-1326.e7. [PMID: 40306274 DOI: 10.1016/j.immuni.2025.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 10/02/2024] [Accepted: 04/03/2025] [Indexed: 05/02/2025]
Abstract
Crosstalk between the immune system and the microbiome is critical for maintaining immune homeostasis. Here, we examined this communication and the impact of immune-suppressive IL-10 signaling on pulmonary homeostasis. We found that IL-10 sensing by interstitial macrophages (IMs) is required to prevent spontaneous lung inflammation. Loss of IL-10 signaling in IMs initiated an inflammatory cascade through the activation of classical monocytes and CD4+ T cell subsets, leading to chronic lung inflammation with age. Analyses of antibiotic-treated and germ-free mice established that lung inflammation in the animals lacking IL-10 signaling was triggered by commensal bacteria. 16S rRNA sequencing revealed Delftia acidovorans and Rhodococcus erythropolis as potential drivers of lung inflammation. Intranasal administration of these bacteria or transplantation of human fecal microbiota elicited lung inflammation in gnotobiotic Il10-deficient mice. These findings highlight that IL-10 sensing by IMs contributes to pulmonary homeostasis by preventing lung inflammation caused by commensal dysbiosis.
Collapse
Affiliation(s)
- Seung Hyeon Kim
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Zachary White
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL, USA
| | | | - Soeun Kang
- Department of Biochemistry and Genetics, University of Illinois College of Medicine, Chicago, IL, USA
| | - Jiseon Kim
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Joseph R Dominguez
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Yeonwoo Choi
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Ivan Cabrera
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL, USA
| | - Madison Plaster
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Michihiro Takahama
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Rafael S Czepielewski
- Immunology Center of Georgia, Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA; Georgia Cancer Center, Augusta University, Augusta, GA, USA
| | - Jinki Yeom
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul, Republic of Korea; Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Matthias Gunzer
- Institute for Experimental Immunology and Imaging, University Hospital Essen, University of Duisburg-Essen, Essen, Germany; Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
| | - Nissim Hay
- Department of Biochemistry and Genetics, University of Illinois College of Medicine, Chicago, IL, USA; University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, USA
| | - Odile David
- University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, USA; Department of Pathology, University of Illinois College of Medicine, Chicago, IL, USA
| | - Nicolas Chevrier
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Teruyuki Sano
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL, USA; University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, USA.
| | - Ki-Wook Kim
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA; University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
2
|
Sun F, Li H, Sun D, Fu S, Gu L, Shao X, Wang Q, Dong X, Duan B, Xing F, Wu J, Xiao M, Zhao F, Han JDJ, Liu Q, Fan X, Li C, Wang C, Shi T. Single-cell omics: experimental workflow, data analyses and applications. SCIENCE CHINA. LIFE SCIENCES 2025; 68:5-102. [PMID: 39060615 DOI: 10.1007/s11427-023-2561-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 04/18/2024] [Indexed: 07/28/2024]
Abstract
Cells are the fundamental units of biological systems and exhibit unique development trajectories and molecular features. Our exploration of how the genomes orchestrate the formation and maintenance of each cell, and control the cellular phenotypes of various organismsis, is both captivating and intricate. Since the inception of the first single-cell RNA technology, technologies related to single-cell sequencing have experienced rapid advancements in recent years. These technologies have expanded horizontally to include single-cell genome, epigenome, proteome, and metabolome, while vertically, they have progressed to integrate multiple omics data and incorporate additional information such as spatial scRNA-seq and CRISPR screening. Single-cell omics represent a groundbreaking advancement in the biomedical field, offering profound insights into the understanding of complex diseases, including cancers. Here, we comprehensively summarize recent advances in single-cell omics technologies, with a specific focus on the methodology section. This overview aims to guide researchers in selecting appropriate methods for single-cell sequencing and related data analysis.
Collapse
Affiliation(s)
- Fengying Sun
- Department of Clinical Laboratory, the Affiliated Wuhu Hospital of East China Normal University (The Second People's Hospital of Wuhu City), Wuhu, 241000, China
| | - Haoyan Li
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Dongqing Sun
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Shaliu Fu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Research Institute of Intelligent Computing, Zhejiang Lab, Hangzhou, 311121, China
- Shanghai Research Institute for Intelligent Autonomous Systems, Shanghai, 201210, China
| | - Lei Gu
- Center for Single-cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xin Shao
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314103, China
| | - Qinqin Wang
- Center for Single-cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xin Dong
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Bin Duan
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Research Institute of Intelligent Computing, Zhejiang Lab, Hangzhou, 311121, China
- Shanghai Research Institute for Intelligent Autonomous Systems, Shanghai, 201210, China
| | - Feiyang Xing
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Jun Wu
- Center for Bioinformatics and Computational Biology, Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Minmin Xiao
- Department of Clinical Laboratory, the Affiliated Wuhu Hospital of East China Normal University (The Second People's Hospital of Wuhu City), Wuhu, 241000, China.
| | - Fangqing Zhao
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Jing-Dong J Han
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology (CQB), Peking University, Beijing, 100871, China.
| | - Qi Liu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China.
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China.
- Research Institute of Intelligent Computing, Zhejiang Lab, Hangzhou, 311121, China.
- Shanghai Research Institute for Intelligent Autonomous Systems, Shanghai, 201210, China.
| | - Xiaohui Fan
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314103, China.
- Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
| | - Chen Li
- Center for Single-cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Chenfei Wang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China.
- Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Tieliu Shi
- Department of Clinical Laboratory, the Affiliated Wuhu Hospital of East China Normal University (The Second People's Hospital of Wuhu City), Wuhu, 241000, China.
- Center for Bioinformatics and Computational Biology, Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
- Key Laboratory of Advanced Theory and Application in Statistics and Data Science-MOE, School of Statistics, East China Normal University, Shanghai, 200062, China.
| |
Collapse
|
3
|
Santiago KCL, Shrestha AMS. DNA-protein quasi-mapping for rapid differential gene expression analysis in non-model organisms. BMC Bioinformatics 2024; 25:335. [PMID: 39448913 PMCID: PMC11515663 DOI: 10.1186/s12859-024-05924-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 09/05/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Conventional differential gene expression analysis pipelines for non-model organisms require computationally expensive transcriptome assembly. We recently proposed an alternative strategy of directly aligning RNA-seq reads to a protein database, and demonstrated drastic improvements in speed, memory usage, and accuracy in identifying differentially expressed genes. RESULT Here we report a further speed-up by replacing DNA-protein alignment by quasi-mapping, making our pipeline > 1000× faster than assembly-based approach, and still more accurate. We also compare quasi-mapping to other mapping techniques, and show that it is faster but at the cost of sensitivity. CONCLUSION We provide a quick-and-dirty differential gene expression analysis pipeline for non-model organisms without a reference transcriptome, which directly quasi-maps RNA-seq reads to a reference protein database, avoiding computationally expensive transcriptome assembly.
Collapse
Affiliation(s)
- Kyle Christian L Santiago
- Bioinformatics Lab, Advanced Research Institute for Informatics, Computing, and Networking, De La Salle University Manila, 2401 Taft Avenue, Manila, Philippines
- Department of Software Technology, College of Computer Studies, De La Salle University Manila, 2401 Taft Avenue, Manila, Philippines
| | - Anish M S Shrestha
- Bioinformatics Lab, Advanced Research Institute for Informatics, Computing, and Networking, De La Salle University Manila, 2401 Taft Avenue, Manila, Philippines.
- Department of Software Technology, College of Computer Studies, De La Salle University Manila, 2401 Taft Avenue, Manila, Philippines.
| |
Collapse
|
4
|
Mantas I, Flais I, Masarapu Y, Ionescu T, Frapard S, Jung F, Le Merre P, Saarinen M, Tiklova K, Salmani BY, Gillberg L, Zhang X, Chergui K, Carlén M, Giacomello S, Hengerer B, Perlmann T, Svenningsson P. Claustrum and dorsal endopiriform cortex complex cell-identity is determined by Nurr1 and regulates hallucinogenic-like states in mice. Nat Commun 2024; 15:8176. [PMID: 39289358 PMCID: PMC11408527 DOI: 10.1038/s41467-024-52429-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 09/04/2024] [Indexed: 09/19/2024] Open
Abstract
The Claustrum/dorsal endopiriform cortex complex (CLA) is an enigmatic brain region with extensive glutamatergic projections to multiple cortical areas. The transcription factor Nurr1 is highly expressed in the CLA, but its role in this region is not understood. By using conditional gene-targeted mice, we show that Nurr1 is a crucial regulator of CLA neuron identity. Although CLA neurons remain intact in the absence of Nurr1, the distinctive gene expression pattern in the CLA is abolished. CLA has been hypothesized to control hallucinations, but little is known of how the CLA responds to hallucinogens. After the deletion of Nurr1 in the CLA, both hallucinogen receptor expression and signaling are lost. Furthermore, functional ultrasound and Neuropixel electrophysiological recordings revealed that the hallucinogenic-receptor agonists' effects on functional connectivity between prefrontal and sensorimotor cortices are altered in Nurr1-ablated mice. Our findings suggest that Nurr1-targeted strategies provide additional avenues for functional studies of the CLA.
Collapse
Affiliation(s)
- Ioannis Mantas
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Ivana Flais
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- CNSDR, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
- Department of Neuroimaging King's College London, London, UK
| | - Yuvarani Masarapu
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Tudor Ionescu
- CNSDR, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Solène Frapard
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Felix Jung
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Pierre Le Merre
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Marcus Saarinen
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Katarina Tiklova
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | | | - Linda Gillberg
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Xiaoqun Zhang
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Karima Chergui
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Marie Carlén
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Stefania Giacomello
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Bastian Hengerer
- CNSDR, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Thomas Perlmann
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Per Svenningsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
5
|
Wang L, Ma X, Liu Y, Liu G, Wei H, Luo Z, Liu H, Yan M, Zhang A, Yu X, Xia H, Luo L. Flexibility of parental-like or maternal-like gene expression under diverse environments contributes to combined drought avoidance and drought tolerance in a water-saving and drought-resistance rice hybrid. TAG. THEORETICAL AND APPLIED GENETICS. THEORETISCHE UND ANGEWANDTE GENETIK 2024; 137:221. [PMID: 39271558 DOI: 10.1007/s00122-024-04735-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024]
Abstract
KEY MESSAGE The hybrid rice variety (Hanyou73) exhibits the maternal-like (HH7A) gene expression in roots and parental-like (HH3) gene expression in leaves to obtain both advantages of drought avoidance and drought tolerance from its two parents. BACKGROUND Rice is one of the most important crops in the world. Rice production consumes lots of water and significantly suffers from the water deficiency and drought stress. The water-saving and drought-resistance rice (WDR) confers good drought resistance and performs well in the water-saving cultivation. MAIN FINDINGS A hybrid WDR variety Hanyou73 (HY73) exhibited superior drought resistance compared with its parents Hanhui3 (HH3) and Huhan7A (HH7A). Studies on drought resistance related traits revealed that HY73 performed like HH3 and HH7A on drought tolerance and drought avoidance, respectively. Transcriptomes were analyzed for samples with various phytohormone treatments and abiotic stresses, in which HY73 was closer to HH3 in leaf samples while HH7A in root samples. HY73 and its parents differed largely in DEGs and GO analysis for DEGs suggested the different pathways of drought response in HH3 and HH7A. Parent-like expression analysis revealed that the higher-parent-like expression pattern was prevailing in HY73. In addition, patterns of the parent-like expression significantly transformed between abiotic-stressed/phytohormone-treated and control samples, which might help HY73 to adapt to different environments. WGCNA analysis for those parent-like expression genes revealed some drought resistant genes that should contribute to the superior drought resistance of HY73. Genetic variation on the promotor sequence was confirmed as the reason for the flexible parent-like gene expression in HY73. CONCLUSION Our study uncovered the important roles of complementation of beneficial traits from parents and flexible gene expressions in drought resistance of HY73, which could facilitate the development of new WDR varieties.
Collapse
Affiliation(s)
- Lei Wang
- Shanghai Agrobiological Gene Center, Shanghai, 201106, China
- Key Laboratory of Grain Crop Genetic Resources Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Shanghai, 201106, China
| | - Xiaosong Ma
- Shanghai Agrobiological Gene Center, Shanghai, 201106, China
- Key Laboratory of Grain Crop Genetic Resources Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Shanghai, 201106, China
- National Key Laboratory of Crop Genetic Improvement and National Center of Plant Gene Research (Wuhan), Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yi Liu
- Shanghai Agrobiological Gene Center, Shanghai, 201106, China
- Key Laboratory of Grain Crop Genetic Resources Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Shanghai, 201106, China
- National Key Laboratory of Crop Genetic Improvement and National Center of Plant Gene Research (Wuhan), Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China
| | - Guolan Liu
- Shanghai Agrobiological Gene Center, Shanghai, 201106, China
- Key Laboratory of Grain Crop Genetic Resources Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Shanghai, 201106, China
| | - Haibin Wei
- Shanghai Agrobiological Gene Center, Shanghai, 201106, China
- Key Laboratory of Grain Crop Genetic Resources Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Shanghai, 201106, China
| | - Zhi Luo
- Shanghai Agrobiological Gene Center, Shanghai, 201106, China
| | - Hongyan Liu
- Shanghai Agrobiological Gene Center, Shanghai, 201106, China
- Key Laboratory of Grain Crop Genetic Resources Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Shanghai, 201106, China
| | - Ming Yan
- Shanghai Agrobiological Gene Center, Shanghai, 201106, China
- Key Laboratory of Grain Crop Genetic Resources Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Shanghai, 201106, China
| | - Anning Zhang
- Shanghai Agrobiological Gene Center, Shanghai, 201106, China
- Key Laboratory of Grain Crop Genetic Resources Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Shanghai, 201106, China
- National Key Laboratory of Crop Genetic Improvement and National Center of Plant Gene Research (Wuhan), Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xinqiao Yu
- Shanghai Agrobiological Gene Center, Shanghai, 201106, China
- Key Laboratory of Grain Crop Genetic Resources Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Shanghai, 201106, China
- College of Agriculture, South China Agricultural University, Guangzhou, 510642, China
| | - Hui Xia
- Shanghai Agrobiological Gene Center, Shanghai, 201106, China.
- Key Laboratory of Grain Crop Genetic Resources Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Shanghai, 201106, China.
- National Key Laboratory of Crop Genetic Improvement and National Center of Plant Gene Research (Wuhan), Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China.
- College of Agriculture, South China Agricultural University, Guangzhou, 510642, China.
| | - Lijun Luo
- Shanghai Agrobiological Gene Center, Shanghai, 201106, China.
- Key Laboratory of Grain Crop Genetic Resources Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Shanghai, 201106, China.
- National Key Laboratory of Crop Genetic Improvement and National Center of Plant Gene Research (Wuhan), Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China.
- College of Agriculture, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
6
|
Jackson DJ, Cerveau N, Posnien N. De novo assembly of transcriptomes and differential gene expression analysis using short-read data from emerging model organisms - a brief guide. Front Zool 2024; 21:17. [PMID: 38902827 PMCID: PMC11188175 DOI: 10.1186/s12983-024-00538-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 06/12/2024] [Indexed: 06/22/2024] Open
Abstract
Many questions in biology benefit greatly from the use of a variety of model systems. High-throughput sequencing methods have been a triumph in the democratization of diverse model systems. They allow for the economical sequencing of an entire genome or transcriptome of interest, and with technical variations can even provide insight into genome organization and the expression and regulation of genes. The analysis and biological interpretation of such large datasets can present significant challenges that depend on the 'scientific status' of the model system. While high-quality genome and transcriptome references are readily available for well-established model systems, the establishment of such references for an emerging model system often requires extensive resources such as finances, expertise and computation capabilities. The de novo assembly of a transcriptome represents an excellent entry point for genetic and molecular studies in emerging model systems as it can efficiently assess gene content while also serving as a reference for differential gene expression studies. However, the process of de novo transcriptome assembly is non-trivial, and as a rule must be empirically optimized for every dataset. For the researcher working with an emerging model system, and with little to no experience with assembling and quantifying short-read data from the Illumina platform, these processes can be daunting. In this guide we outline the major challenges faced when establishing a reference transcriptome de novo and we provide advice on how to approach such an endeavor. We describe the major experimental and bioinformatic steps, provide some broad recommendations and cautions for the newcomer to de novo transcriptome assembly and differential gene expression analyses. Moreover, we provide an initial selection of tools that can assist in the journey from raw short-read data to assembled transcriptome and lists of differentially expressed genes.
Collapse
Affiliation(s)
- Daniel J Jackson
- University of Göttingen, Department of Geobiology, Goldschmidtstr.3, Göttingen, 37077, Germany.
| | - Nicolas Cerveau
- University of Göttingen, Department of Geobiology, Goldschmidtstr.3, Göttingen, 37077, Germany
| | - Nico Posnien
- University of Göttingen, Department of Developmental Biology, GZMB, Justus-Von-Liebig-Weg 11, Göttingen, 37077, Germany.
| |
Collapse
|
7
|
Takahama M, Patil A, Richey G, Cipurko D, Johnson K, Carbonetto P, Plaster M, Pandey S, Cheronis K, Ueda T, Gruenbaum A, Kawamoto T, Stephens M, Chevrier N. A pairwise cytokine code explains the organism-wide response to sepsis. Nat Immunol 2024; 25:226-239. [PMID: 38191855 PMCID: PMC10834370 DOI: 10.1038/s41590-023-01722-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 11/29/2023] [Indexed: 01/10/2024]
Abstract
Sepsis is a systemic response to infection with life-threatening consequences. Our understanding of the molecular and cellular impact of sepsis across organs remains rudimentary. Here, we characterize the pathogenesis of sepsis by measuring dynamic changes in gene expression across organs. To pinpoint molecules controlling organ states in sepsis, we compare the effects of sepsis on organ gene expression to those of 6 singles and 15 pairs of recombinant cytokines. Strikingly, we find that the pairwise effects of tumor necrosis factor plus interleukin (IL)-18, interferon-gamma or IL-1β suffice to mirror the impact of sepsis across tissues. Mechanistically, we map the cellular effects of sepsis and cytokines by computing changes in the abundance of 195 cell types across 9 organs, which we validate by whole-mouse spatial profiling. Our work decodes the cytokine cacophony in sepsis into a pairwise cytokine message capturing the gene, cell and tissue responses of the host to the disease.
Collapse
Affiliation(s)
- Michihiro Takahama
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
- Laboratory of Bioresponse Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | | | - Gabriella Richey
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Denis Cipurko
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Katherine Johnson
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Peter Carbonetto
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
- Research Computing Center, University of Chicago, Chicago, IL, USA
| | - Madison Plaster
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Surya Pandey
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Katerina Cheronis
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Tatsuki Ueda
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Adam Gruenbaum
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | | | - Matthew Stephens
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
- Department of Statistics, University of Chicago, Chicago, IL, USA
| | - Nicolas Chevrier
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
8
|
Decano JL, Maiorino E, Matamalas JT, Chelvanambi S, Tiemeijer BM, Yanagihara Y, Mukai S, Jha PK, Pestana DV, D’Souza E, Whelan M, Ge R, Asano T, Sharma A, Libby P, Singh SA, Aikawa E, Aikawa M. Cellular Heterogeneity of Activated Primary Human Macrophages and Associated Drug-Gene Networks: From Biology to Precision Therapeutics. Circulation 2023; 148:1459-1478. [PMID: 37850387 PMCID: PMC10624416 DOI: 10.1161/circulationaha.123.064794] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 08/24/2023] [Indexed: 10/19/2023]
Abstract
BACKGROUND Interferon-γ (IFNγ) signaling plays a complex role in atherogenesis. IFNγ stimulation of macrophages permits in vitro exploration of proinflammatory mechanisms and the development of novel immune therapies. We hypothesized that the study of macrophage subpopulations could lead to anti-inflammatory interventions. METHODS Primary human macrophages activated by IFNγ (M(IFNγ)) underwent analyses by single-cell RNA sequencing, time-course cell-cluster proteomics, metabolite consumption, immunoassays, and functional tests (phagocytic, efferocytotic, and chemotactic). RNA-sequencing data were analyzed in LINCS (Library of Integrated Network-Based Cellular Signatures) to identify compounds targeting M(IFNγ) subpopulations. The effect of compound BI-2536 was tested in human macrophages in vitro and in a murine model of atherosclerosis. RESULTS Single-cell RNA sequencing identified 2 major clusters in M(IFNγ): inflammatory (M(IFNγ)i) and phagocytic (M(IFNγ)p). M(IFNγ)i had elevated expression of inflammatory chemokines and higher amino acid consumption compared with M(IFNγ)p. M(IFNγ)p were more phagocytotic and chemotactic with higher Krebs cycle activity and less glycolysis than M(IFNγ)i. Human carotid atherosclerotic plaques contained 2 such macrophage clusters. Bioinformatic LINCS analysis using our RNA-sequencing data identified BI-2536 as a potential compound to decrease the M(IFNγ)i subpopulation. BI-2536 in vitro decreased inflammatory chemokine expression and secretion in M(IFNγ) by shrinking the M(IFNγ)i subpopulation while expanding the M(IFNγ)p subpopulation. BI-2536 in vivo shifted the phenotype of macrophages, modulated inflammation, and decreased atherosclerosis and calcification. CONCLUSIONS We characterized 2 clusters of macrophages in atherosclerosis and combined our cellular data with a cell-signature drug library to identify a novel compound that targets a subset of macrophages in atherosclerosis. Our approach is a precision medicine strategy to identify new drugs that target atherosclerosis and other inflammatory diseases.
Collapse
Affiliation(s)
- Julius L. Decano
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (J.L.D., J.T.M., S.C., B.M.T., Y.Y., S.M., D.V.S.P., E.D., M.W., R.G., T.A., A.S., S.A.S., E.A., M.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Enrico Maiorino
- Channing Division of Network Medicine (E.M., A.S., M.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Joan T. Matamalas
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (J.L.D., J.T.M., S.C., B.M.T., Y.Y., S.M., D.V.S.P., E.D., M.W., R.G., T.A., A.S., S.A.S., E.A., M.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Sarvesh Chelvanambi
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (J.L.D., J.T.M., S.C., B.M.T., Y.Y., S.M., D.V.S.P., E.D., M.W., R.G., T.A., A.S., S.A.S., E.A., M.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Bart M. Tiemeijer
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (J.L.D., J.T.M., S.C., B.M.T., Y.Y., S.M., D.V.S.P., E.D., M.W., R.G., T.A., A.S., S.A.S., E.A., M.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Yoshihiro Yanagihara
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (J.L.D., J.T.M., S.C., B.M.T., Y.Y., S.M., D.V.S.P., E.D., M.W., R.G., T.A., A.S., S.A.S., E.A., M.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Shin Mukai
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (J.L.D., J.T.M., S.C., B.M.T., Y.Y., S.M., D.V.S.P., E.D., M.W., R.G., T.A., A.S., S.A.S., E.A., M.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Prabhash Kumar Jha
- Department of Medicine, and Center for Excellence in Vascular Biology (P.K.J., P.L., E.A., M.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Diego V.S. Pestana
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (J.L.D., J.T.M., S.C., B.M.T., Y.Y., S.M., D.V.S.P., E.D., M.W., R.G., T.A., A.S., S.A.S., E.A., M.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Edwin D’Souza
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (J.L.D., J.T.M., S.C., B.M.T., Y.Y., S.M., D.V.S.P., E.D., M.W., R.G., T.A., A.S., S.A.S., E.A., M.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Mary Whelan
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (J.L.D., J.T.M., S.C., B.M.T., Y.Y., S.M., D.V.S.P., E.D., M.W., R.G., T.A., A.S., S.A.S., E.A., M.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Rile Ge
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (J.L.D., J.T.M., S.C., B.M.T., Y.Y., S.M., D.V.S.P., E.D., M.W., R.G., T.A., A.S., S.A.S., E.A., M.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Takaharu Asano
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (J.L.D., J.T.M., S.C., B.M.T., Y.Y., S.M., D.V.S.P., E.D., M.W., R.G., T.A., A.S., S.A.S., E.A., M.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Amitabh Sharma
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (J.L.D., J.T.M., S.C., B.M.T., Y.Y., S.M., D.V.S.P., E.D., M.W., R.G., T.A., A.S., S.A.S., E.A., M.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Channing Division of Network Medicine (E.M., A.S., M.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Peter Libby
- Department of Medicine, and Center for Excellence in Vascular Biology (P.K.J., P.L., E.A., M.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Sasha A. Singh
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (J.L.D., J.T.M., S.C., B.M.T., Y.Y., S.M., D.V.S.P., E.D., M.W., R.G., T.A., A.S., S.A.S., E.A., M.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (J.L.D., J.T.M., S.C., B.M.T., Y.Y., S.M., D.V.S.P., E.D., M.W., R.G., T.A., A.S., S.A.S., E.A., M.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Medicine, and Center for Excellence in Vascular Biology (P.K.J., P.L., E.A., M.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (J.L.D., J.T.M., S.C., B.M.T., Y.Y., S.M., D.V.S.P., E.D., M.W., R.G., T.A., A.S., S.A.S., E.A., M.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Channing Division of Network Medicine (E.M., A.S., M.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Medicine, and Center for Excellence in Vascular Biology (P.K.J., P.L., E.A., M.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
9
|
Nguyen DT. An integrative pipeline for circular RNA quantitative trait locus discovery with application in human T cells. Bioinformatics 2023; 39:btad667. [PMID: 37929995 PMCID: PMC10636286 DOI: 10.1093/bioinformatics/btad667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 10/25/2023] [Accepted: 10/30/2023] [Indexed: 11/07/2023] Open
Abstract
MOTIVATION Molecular quantitative trait locus (QTL) mapping has proven to be a powerful approach for prioritizing genetic regulatory variants and causal genes identified by genome-wide association studies. Recently, this success has been extended to circular RNA (circRNA), a potential group of RNAs that can serve as markers for the diagnosis, prognosis, or therapeutic targets of various human diseases. However, a well-developed computational pipeline for circRNA QTL (circQTL) discovery is still lacking. RESULTS We introduce an integrative method for circQTL mapping and implement it as an automated pipeline based on Nextflow, named cscQTL. The proposed method has two main advantages. Firstly, cscQTL improves the specificity by systematically combining outputs of multiple circRNA calling algorithms to obtain highly confident circRNA annotations. Secondly, cscQTL improves the sensitivity by accurately quantifying circRNA expression with the help of pseudo references. Compared to the single method approach, cscQTL effectively identifies circQTLs with an increase of 20%-100% circQTLs detected and recovered all circQTLs that are highly supported by the single method approach. We apply cscQTL to a dataset of human T cells and discover genetic variants that control the expression of 55 circRNAs. By colocalization tests, we further identify circBACH2 and circYY1AP1 as potential candidates for immune disease regulation. AVAILABILITY AND IMPLEMENTATION cscQTL is freely available at: https://github.com/datngu/cscQTL and https://doi.org/10.5281/zenodo.7851982.
Collapse
Affiliation(s)
- Dat Thanh Nguyen
- Centre for Integrative Genetics, Faculty of Biosciences, Norwegian University of Life Sciences, 1432 Ås, Norway
| |
Collapse
|
10
|
Borozan L, Rojas Ringeling F, Kao SY, Nikonova E, Monteagudo-Mesas P, Matijević D, Spletter ML, Canzar S. Counting pseudoalignments to novel splicing events. Bioinformatics 2023; 39:btad419. [PMID: 37432342 PMCID: PMC10348833 DOI: 10.1093/bioinformatics/btad419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 04/21/2023] [Accepted: 07/10/2023] [Indexed: 07/12/2023] Open
Abstract
MOTIVATION Alternative splicing (AS) of introns from pre-mRNA produces diverse sets of transcripts across cell types and tissues, but is also dysregulated in many diseases. Alignment-free computational methods have greatly accelerated the quantification of mRNA transcripts from short RNA-seq reads, but they inherently rely on a catalog of known transcripts and might miss novel, disease-specific splicing events. By contrast, alignment of reads to the genome can effectively identify novel exonic segments and introns. Event-based methods then count how many reads align to predefined features. However, an alignment is more expensive to compute and constitutes a bottleneck in many AS analysis methods. RESULTS Here, we propose fortuna, a method that guesses novel combinations of annotated splice sites to create transcript fragments. It then pseudoaligns reads to fragments using kallisto and efficiently derives counts of the most elementary splicing units from kallisto's equivalence classes. These counts can be directly used for AS analysis or summarized to larger units as used by other widely applied methods. In experiments on synthetic and real data, fortuna was around 7× faster than traditional align and count approaches, and was able to analyze almost 300 million reads in just 15 min when using four threads. It mapped reads containing mismatches more accurately across novel junctions and found more reads supporting aberrant splicing events in patients with autism spectrum disorder than existing methods. We further used fortuna to identify novel, tissue-specific splicing events in Drosophila. AVAILABILITY AND IMPLEMENTATION fortuna source code is available at https://github.com/canzarlab/fortuna.
Collapse
Affiliation(s)
- Luka Borozan
- Department of Mathematics, Josip Juraj Strossmayer University of Osijek, Osijek 31000, Croatia
| | - Francisca Rojas Ringeling
- Gene Center, Ludwig-Maximilians-Universität München, Munich 81377, Germany
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, United States
| | - Shao-Yen Kao
- Biomedical Center, Department of Physiological Chemistry, Ludwig-Maximilians-Universität München, Planegg-Martinsried 82152, Germany
| | - Elena Nikonova
- Biomedical Center, Department of Physiological Chemistry, Ludwig-Maximilians-Universität München, Planegg-Martinsried 82152, Germany
| | | | - Domagoj Matijević
- Department of Mathematics, Josip Juraj Strossmayer University of Osijek, Osijek 31000, Croatia
| | - Maria L Spletter
- Biomedical Center, Department of Physiological Chemistry, Ludwig-Maximilians-Universität München, Planegg-Martinsried 82152, Germany
- School of Science and Engineering, Division of Biological & Biomedical Systems, University of Missouri Kansas City, Kansas City, MO 64110, United States
| | - Stefan Canzar
- Gene Center, Ludwig-Maximilians-Universität München, Munich 81377, Germany
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, United States
- Department of Computer Science and Engineering, The Pennsylvania State University, University Park, PA 16802, United States
| |
Collapse
|
11
|
Koslow M, Zhu P, McCabe C, Xu X, Lin X. Kidney transcriptome and cystic kidney disease genes in zebrafish. Front Physiol 2023; 14:1184025. [PMID: 37256068 PMCID: PMC10226271 DOI: 10.3389/fphys.2023.1184025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 04/20/2023] [Indexed: 06/01/2023] Open
Abstract
Introduction: Polycystic kidney disease (PKD) is a condition where fluid filled cysts form on the kidney which leads to overall renal failure. Zebrafish has been recently adapted to study polycystic kidney disease, because of its powerful embryology and genetics. However, there are concerns on the conservation of this lower vertebrate in modeling polycystic kidney disease. Methods: Here, we aim to assess the molecular conservation of zebrafish by searching homologues polycystic kidney disease genes and carrying transcriptome studies in this animal. Results and Discussion: We found that out of 82 human cystic kidney disease genes, 81 have corresponding zebrafish homologs. While 75 of the genes have a single homologue, only 6 of these genes have two homologs. Comparison of the expression level of the transcripts enabled us to identify one homolog over the other homolog with >70% predominance, which would be prioritized for future experimental studies. Prompted by sexual dimorphism in human and rodent kidneys, we studied transcriptome between different sexes and noted significant differences in male vs. female zebrafish, indicating that sex dimorphism also occurs in zebrafish. Comparison between zebrafish and mouse identified 10% shared genes and 38% shared signaling pathways. String analysis revealed a cluster of genes differentially expressed in male vs. female zebrafish kidneys. In summary, this report demonstrated remarkable molecular conservation, supporting zebrafish as a useful animal model for cystic kidney disease.
Collapse
Affiliation(s)
- Matthew Koslow
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| | - Ping Zhu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| | - Chantal McCabe
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, United States
| | - Xiaolei Xu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Xueying Lin
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
12
|
Rebolledo C, Silva JP, Saavedra N, Maracaja-Coutinho V. Computational approaches for circRNAs prediction and in silico characterization. Brief Bioinform 2023; 24:7150741. [PMID: 37139555 DOI: 10.1093/bib/bbad154] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 03/20/2023] [Accepted: 03/30/2023] [Indexed: 05/05/2023] Open
Abstract
Circular RNAs (circRNAs) are single-stranded and covalently closed non-coding RNA molecules originated from RNA splicing. Their functions include regulatory potential over other RNA species, such as microRNAs, messenger RNAs and RNA binding proteins. For circRNA identification, several algorithms are available and can be classified in two major types: pseudo-reference-based and split-alignment-based approaches. In general, the data generated from circRNA transcriptome initiatives is deposited on public specific databases, which provide a large amount of information on different species and functional annotations. In this review, we describe the main computational resources for the identification and characterization of circRNAs, covering the algorithms and predictive tools to evaluate its potential role in a particular transcriptomics project, including the public repositories containing relevant data and information for circRNAs, recapitulating their characteristics, reliability and amount of data reported.
Collapse
Affiliation(s)
- Camilo Rebolledo
- Center of Molecular Biology & Pharmacogenetics, Department of Basic Sciences, Scientific and Technological Resources, Universidad de La Frontera, Temuco, Chile
- Advanced Center for Chronic Diseases - ACCDiS, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
- Centro de Modelamiento Molecular, Biofísica y Bioinformática - CM2B2, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Juan Pablo Silva
- Centro de Modelamiento Molecular, Biofísica y Bioinformática - CM2B2, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
- ANID Anillo ACT210004 SYSTEMIX, Rancagua, Chile
| | - Nicolás Saavedra
- Center of Molecular Biology & Pharmacogenetics, Department of Basic Sciences, Scientific and Technological Resources, Universidad de La Frontera, Temuco, Chile
| | - Vinicius Maracaja-Coutinho
- Advanced Center for Chronic Diseases - ACCDiS, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
- Centro de Modelamiento Molecular, Biofísica y Bioinformática - CM2B2, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
- ANID Anillo ACT210004 SYSTEMIX, Rancagua, Chile
- Anillo Inflammation in HIV/AIDS - InflammAIDS, Santiago, Chile
| |
Collapse
|
13
|
Deshpande D, Chhugani K, Chang Y, Karlsberg A, Loeffler C, Zhang J, Muszyńska A, Munteanu V, Yang H, Rotman J, Tao L, Balliu B, Tseng E, Eskin E, Zhao F, Mohammadi P, P. Łabaj P, Mangul S. RNA-seq data science: From raw data to effective interpretation. Front Genet 2023; 14:997383. [PMID: 36999049 PMCID: PMC10043755 DOI: 10.3389/fgene.2023.997383] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 02/24/2023] [Indexed: 03/14/2023] Open
Abstract
RNA sequencing (RNA-seq) has become an exemplary technology in modern biology and clinical science. Its immense popularity is due in large part to the continuous efforts of the bioinformatics community to develop accurate and scalable computational tools to analyze the enormous amounts of transcriptomic data that it produces. RNA-seq analysis enables genes and their corresponding transcripts to be probed for a variety of purposes, such as detecting novel exons or whole transcripts, assessing expression of genes and alternative transcripts, and studying alternative splicing structure. It can be a challenge, however, to obtain meaningful biological signals from raw RNA-seq data because of the enormous scale of the data as well as the inherent limitations of different sequencing technologies, such as amplification bias or biases of library preparation. The need to overcome these technical challenges has pushed the rapid development of novel computational tools, which have evolved and diversified in accordance with technological advancements, leading to the current myriad of RNA-seq tools. These tools, combined with the diverse computational skill sets of biomedical researchers, help to unlock the full potential of RNA-seq. The purpose of this review is to explain basic concepts in the computational analysis of RNA-seq data and define discipline-specific jargon.
Collapse
Affiliation(s)
- Dhrithi Deshpande
- Department of Pharmacology and Pharmaceutical Sciences, USC Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, Los Angeles, CA, United States
| | - Karishma Chhugani
- Department of Pharmacology and Pharmaceutical Sciences, USC Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, Los Angeles, CA, United States
| | - Yutong Chang
- Department of Pharmacology and Pharmaceutical Sciences, USC Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, Los Angeles, CA, United States
| | - Aaron Karlsberg
- Department of Clinical Pharmacy, USC Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, Los Angeles, CA, United States
| | - Caitlin Loeffler
- Department of Computer Science, University of California, Los Angeles, CA, United States
| | - Jinyang Zhang
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
| | - Agata Muszyńska
- Małopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
- Institute of Automatic Control, Electronics and Computer Science, Silesian University of Technology, Gliwice, Poland
| | - Viorel Munteanu
- Department of Computers, Informatics and Microelectronics, Technical University of Moldova, Chisinau, Moldova
| | - Harry Yang
- Department of Microbiology, Immunology and Molecular Genetics, University of California Los Angeles, Los Angeles, CA, United States
| | - Jeremy Rotman
- Department of Clinical Pharmacy, USC Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, Los Angeles, CA, United States
| | - Laura Tao
- Department of Computational Medicine, David Geffen School of Medicine at UCLA, CHS, Los Angeles, CA, United States
| | - Brunilda Balliu
- Department of Computational Medicine, David Geffen School of Medicine at UCLA, CHS, Los Angeles, CA, United States
| | | | - Eleazar Eskin
- Department of Computer Science, University of California, Los Angeles, CA, United States
- Department of Computational Medicine, David Geffen School of Medicine at UCLA, CHS, Los Angeles, CA, United States
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Fangqing Zhao
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Pejman Mohammadi
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, United States
| | - Paweł P. Łabaj
- Małopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
- Department of Biotechnology, Boku University Vienna, Vienna, Austria
| | - Serghei Mangul
- Department of Clinical Pharmacy, USC Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, Los Angeles, CA, United States
- Department of Quantitative and Computational Biology, USC Dornsife College of Letters, Arts and Sciences, Los Angeles, CA, United States
- *Correspondence: Serghei Mangul,
| |
Collapse
|
14
|
Lonsdale A, Halman A, Brown L, Kosasih H, Ekert P, Oshlack A. Toblerone: detecting exon deletion events in cancer using RNA-seq. F1000Res 2023; 12:130. [PMID: 37767021 PMCID: PMC10521068 DOI: 10.12688/f1000research.129490.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/27/2023] [Indexed: 09/29/2023] Open
Abstract
Cancer is driven by mutations of the genome that can result in the activation of oncogenes or repression of tumour suppressor genes. In acute lymphoblastic leukemia (ALL) focal deletions in IKAROS family zinc finger 1 (IKZF1) result in the loss of zinc-finger DNA-binding domains and a dominant negative isoform that is associated with higher rates of relapse and poorer patient outcomes. Clinically, the presence of IKZF1 deletions informs prognosis and treatment options. In this work we developed a method for detecting exon deletions in genes using RNA-seq with application to IKZF1. We developed a pipeline that first uses a custom transcriptome reference consisting of transcripts with exon deletions. Next, RNA-seq reads are mapped using a pseudoalignment algorithm to identify reads that uniquely support deletions. These are then evaluated for evidence of the deletion with respect to gene expression and other samples. We applied the algorithm, named Toblerone, to a cohort of 99 B-ALL paediatric samples including validated IKZF1 deletions. Furthermore, we developed a graphical desktop app for non-bioinformatics users that can quickly and easily identify and report deletions in IKZF1 from RNA-seq data with informative graphical outputs.
Collapse
Affiliation(s)
- Andrew Lonsdale
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, 3010, Australia
- Murdoch Children’s Research Institute, Parkville, VIC, 3052, Australia
- Peter MacCallum Cancer Centre, Parkville, VIC, 3052, Australia
| | - Andreas Halman
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, 3010, Australia
- Peter MacCallum Cancer Centre, Parkville, VIC, 3052, Australia
| | - Lauren Brown
- Murdoch Children’s Research Institute, Parkville, VIC, 3052, Australia
- School of Women’s and Children’s Health, UNSW Sydney, Sydney, NSW, 2052, Australia
- Children's Cancer Institute Australia, Sydney, NSW, 2052, Australia
| | - Hansen Kosasih
- Murdoch Children’s Research Institute, Parkville, VIC, 3052, Australia
| | - Paul Ekert
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, 3010, Australia
- Murdoch Children’s Research Institute, Parkville, VIC, 3052, Australia
- Peter MacCallum Cancer Centre, Parkville, VIC, 3052, Australia
- School of Women’s and Children’s Health, UNSW Sydney, Sydney, NSW, 2052, Australia
- Children's Cancer Institute Australia, Sydney, NSW, 2052, Australia
| | - Alicia Oshlack
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, 3010, Australia
- Peter MacCallum Cancer Centre, Parkville, VIC, 3052, Australia
- School of Mathematics and Statistics, University of Melbourne, Parkville, VIC, 3010, Australia
| |
Collapse
|
15
|
Takahama M, Patil A, Johnson K, Cipurko D, Miki Y, Taketomi Y, Carbonetto P, Plaster M, Richey G, Pandey S, Cheronis K, Ueda T, Gruenbaum A, Dudek SM, Stephens M, Murakami M, Chevrier N. Organism-Wide Analysis of Sepsis Reveals Mechanisms of Systemic Inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.30.526342. [PMID: 36778287 PMCID: PMC9915512 DOI: 10.1101/2023.01.30.526342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Sepsis is a systemic response to infection with life-threatening consequences. Our understanding of the impact of sepsis across organs of the body is rudimentary. Here, using mouse models of sepsis, we generate a dynamic, organism-wide map of the pathogenesis of the disease, revealing the spatiotemporal patterns of the effects of sepsis across tissues. These data revealed two interorgan mechanisms key in sepsis. First, we discover a simplifying principle in the systemic behavior of the cytokine network during sepsis, whereby a hierarchical cytokine circuit arising from the pairwise effects of TNF plus IL-18, IFN-γ, or IL-1β explains half of all the cellular effects of sepsis on 195 cell types across 9 organs. Second, we find that the secreted phospholipase PLA2G5 mediates hemolysis in blood, contributing to organ failure during sepsis. These results provide fundamental insights to help build a unifying mechanistic framework for the pathophysiological effects of sepsis on the body.
Collapse
|
16
|
Shen W, Xiang H, Huang T, Tang H, Peng M, Cai D, Hu P, Ren H. KMCP: accurate metagenomic profiling of both prokaryotic and viral populations by pseudo-mapping. Bioinformatics 2023; 39:btac845. [PMID: 36579886 PMCID: PMC9828150 DOI: 10.1093/bioinformatics/btac845] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 12/17/2022] [Accepted: 12/28/2022] [Indexed: 12/30/2022] Open
Abstract
MOTIVATION The growing number of microbial reference genomes enables the improvement of metagenomic profiling accuracy but also imposes greater requirements on the indexing efficiency, database size and runtime of taxonomic profilers. Additionally, most profilers focus mainly on bacterial, archaeal and fungal populations, while less attention is paid to viral communities. RESULTS We present KMCP (K-mer-based Metagenomic Classification and Profiling), a novel k-mer-based metagenomic profiling tool that utilizes genome coverage information by splitting the reference genomes into chunks and stores k-mers in a modified and optimized Compact Bit-Sliced Signature Index for fast alignment-free sequence searching. KMCP combines k-mer similarity and genome coverage information to reduce the false positive rate of k-mer-based taxonomic classification and profiling methods. Benchmarking results based on simulated and real data demonstrate that KMCP, despite a longer running time than all other methods, not only allows the accurate taxonomic profiling of prokaryotic and viral populations but also provides more confident pathogen detection in clinical samples of low depth. AVAILABILITY AND IMPLEMENTATION The software is open-source under the MIT license and available at https://github.com/shenwei356/kmcp. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Wei Shen
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Hongyan Xiang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Tianquan Huang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Hui Tang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Mingli Peng
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Dachuan Cai
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Peng Hu
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Hong Ren
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
17
|
Costa-Silva J, Domingues DS, Menotti D, Hungria M, Lopes FM. Temporal progress of gene expression analysis with RNA-Seq data: A review on the relationship between computational methods. Comput Struct Biotechnol J 2022; 21:86-98. [PMID: 36514333 PMCID: PMC9730150 DOI: 10.1016/j.csbj.2022.11.051] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/25/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022] Open
Abstract
Analysis of differential gene expression from RNA-seq data has become a standard for several research areas. The steps for the computational analysis include many data types and file formats, and a wide variety of computational tools that can be applied alone or together as pipelines. This paper presents a review of the differential expression analysis pipeline, addressing its steps and the respective objectives, the principal methods available in each step, and their properties, therefore introducing an organized overview to this context. This review aims to address mainly the aspects involved in the differentially expressed gene (DEG) analysis from RNA sequencing data (RNA-seq), considering the computational methods. In addition, a timeline of the computational methods for DEG is shown and discussed, and the relationships existing between the most important computational tools are presented by an interaction network. A discussion on the challenges and gaps in DEG analysis is also highlighted in this review. This paper will serve as a tutorial for new entrants into the field and help established users update their analysis pipelines.
Collapse
Affiliation(s)
- Juliana Costa-Silva
- Department of Informatics – Federal University of Paraná, Rua Coronel Francisco Heráclito dos Santos, 100, 81531-990 Curitiba, Paraná, Brazil
| | - Douglas S. Domingues
- Department of Genetics, “Luiz de Queiroz” College of Agriculture, University of São Paulo, Av. Pádua Dias, 11, 13418-900 Piracicaba, São Paulo, Brazil
| | - David Menotti
- Department of Informatics – Federal University of Paraná, Rua Coronel Francisco Heráclito dos Santos, 100, 81531-990 Curitiba, Paraná, Brazil
| | - Mariangela Hungria
- Department of Soil Biotecnology - Embrapa Soybean, Cx. Postal 231, 86000-970 Londrina, Paraná, Brazil
| | - Fabrício Martins Lopes
- Department of Computer Science, Universidade Tecnológica Federal do Paraná – UTFPR, Av. Alberto Carazzai, 1640, 86300-000, Cornélio Procópio, Paraná, Brazil
| |
Collapse
|
18
|
Taguchi YH, Turki T. Adapted tensor decomposition and PCA based unsupervised feature extraction select more biologically reasonable differentially expressed genes than conventional methods. Sci Rep 2022; 12:17438. [PMID: 36261574 PMCID: PMC9580456 DOI: 10.1038/s41598-022-21474-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 09/27/2022] [Indexed: 01/12/2023] Open
Abstract
Tensor decomposition- and principal component analysis-based unsupervised feature extraction were proposed almost 5 and 10 years ago, respectively; although these methods have been successfully applied to a wide range of genome analyses, including drug repositioning, biomarker identification, and disease-causing genes' identification, some fundamental problems have been identified: the number of genes identified was too small to assume that there were no false negatives, and the histogram of P values derived was not fully coincident with the null hypothesis that principal component and singular value vectors follow the Gaussian distribution. Optimizing the standard deviation such that the histogram of P values is as much as possible coincident with the null hypothesis results in an increase in the number and biological reliability of the selected genes. Our contribution was that we improved these methods so as to be able to select biologically more reasonable differentially expressed genes than the state of art methods that must empirically assume negative binomial distributions and dispersion relation, which is required for the selecting more expressed genes than less expressed ones, which can be achieved by the proposed methods that do not have to assume these.
Collapse
Affiliation(s)
- Y-H Taguchi
- Department of Physics, Chuo University, 1-13-27 Kasuga, Bunkyo-ku, Tokyo, 112-8551, Japan.
| | - Turki Turki
- Department of Computer Science, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| |
Collapse
|
19
|
Fang S, Chen B, Zhang Y, Sun H, Liu L, Liu S, Li Y, Xu X. Computational Approaches and Challenges in Spatial Transcriptomics. GENOMICS, PROTEOMICS & BIOINFORMATICS 2022:S1672-0229(22)00129-2. [PMID: 36252814 PMCID: PMC10372921 DOI: 10.1016/j.gpb.2022.10.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 09/08/2022] [Accepted: 10/09/2022] [Indexed: 01/19/2023]
Abstract
The development of spatial transcriptomics (ST) technologies has transformed genetic research from a single-cell data level to a two-dimensional spatial coordinate system and facilitated the study of the composition and function of various cell subsets in different environments and organs. The large-scale data generated by these ST technologies, which contain spatial gene expression information, have elicited the need for spatially resolved approaches to meet the requirements of computational and biological data interpretation. These requirements include dealing with the explosive growth of data to determine the cell-level and gene-level expression, correcting the inner batch effect and loss of expression to improve the data quality, conducting efficient interpretation and in-depth knowledge mining both at the single-cell and tissue-wide levels, and conducting multi-omics integration analysis to provide an extensible framework toward the in-depth understanding of biological processes. However, algorithms designed specifically for ST technologies to meet these requirements are still in their infancy. Here, we review computational approaches to these problems in light of corresponding issues and challenges, and present forward-looking insights into algorithm development.
Collapse
|
20
|
Deng W, Mou T, Pawitan Y, Vu TN. Quantification of mutant–allele expression at isoform level in cancer from RNA-seq data. NAR Genom Bioinform 2022; 4:lqac052. [PMID: 35855322 PMCID: PMC9278039 DOI: 10.1093/nargab/lqac052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/26/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
Even though the role of DNA mutations in cancer is well recognized, current quantification of the RNA expression, performed either at gene or isoform level, typically ignores the mutation status. Standard methods for estimating allele-specific expression (ASE) consider gene-level expression, but the functional impact of a mutation is best assessed at isoform level. Hence our goal is to quantify the mutant–allele expression at isoform level. We have developed and implemented a method, named MAX, for quantifying mutant–allele expression given a list of mutations. For a gene of interest, a mutant reference is constructed by incorporating all possible mutant versions of the wild-type isoforms in the transcriptome annotation. The mutant reference is then used for the RNA-seq reads mapping, which in principle works similarly for any quantification tool. We apply an alternating EM algorithm to the read-count data from the mapping step. In a simulation study, MAX performs well against standard isoform-quantification methods. Also, MAX achieves higher accuracy than conventional gene-based ASE methods such as ASEP. An analysis of a real dataset of acute myeloid leukemia reveals a subgroup of NPM1-mutated patients responding well to a kinase inhibitor. Our findings indicate that quantification of mutant–allele expression at isoform level is feasible and has potential added values for assessing the functional impact of DNA mutations in cancers.
Collapse
Affiliation(s)
- Wenjiang Deng
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet , Stockholm, Sweden
| | - Tian Mou
- School of Biomedical Engineering, Shenzhen University , Shenzhen, China
| | - Yudi Pawitan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet , Stockholm, Sweden
| | - Trung Nghia Vu
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet , Stockholm, Sweden
| |
Collapse
|
21
|
Yeo AT, Rawal S, Delcuze B, Christofides A, Atayde A, Strauss L, Balaj L, Rogers VA, Uhlmann EJ, Varma H, Carter BS, Boussiotis VA, Charest A. Single-cell RNA sequencing reveals evolution of immune landscape during glioblastoma progression. Nat Immunol 2022; 23:971-984. [PMID: 35624211 PMCID: PMC9174057 DOI: 10.1038/s41590-022-01215-0] [Citation(s) in RCA: 146] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 04/18/2022] [Indexed: 01/22/2023]
Abstract
Glioblastoma (GBM) is an incurable primary malignant brain cancer hallmarked with a substantial protumorigenic immune component. Knowledge of the GBM immune microenvironment during tumor evolution and standard of care treatments is limited. Using single-cell transcriptomics and flow cytometry, we unveiled large-scale comprehensive longitudinal changes in immune cell composition throughout tumor progression in an epidermal growth factor receptor-driven genetic mouse GBM model. We identified subsets of proinflammatory microglia in developing GBMs and anti-inflammatory macrophages and protumorigenic myeloid-derived suppressors cells in end-stage tumors, an evolution that parallels breakdown of the blood-brain barrier and extensive growth of epidermal growth factor receptor+ GBM cells. A similar relationship was found between microglia and macrophages in patient biopsies of low-grade glioma and GBM. Temozolomide decreased the accumulation of myeloid-derived suppressor cells, whereas concomitant temozolomide irradiation increased intratumoral GranzymeB+ CD8+T cells but also increased CD4+ regulatory T cells. These results provide a comprehensive and unbiased immune cellular landscape and its evolutionary changes during GBM progression.
Collapse
Affiliation(s)
- Alan T Yeo
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Sackler School of Graduate Studies, Tufts University School of Medicine, Boston, MA, USA
| | - Shruti Rawal
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Bethany Delcuze
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Sackler School of Graduate Studies, Tufts University School of Medicine, Boston, MA, USA
| | - Anthos Christofides
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Agata Atayde
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Laura Strauss
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Leonora Balaj
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Vaughn A Rogers
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Erik J Uhlmann
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Hemant Varma
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Bob S Carter
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Vassiliki A Boussiotis
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| | - Al Charest
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| |
Collapse
|
22
|
Fu Z, Yan W, Chen CT, Nilsson AK, Bull E, Allen W, Yang J, Ko M, SanGiovanni JP, Akula JD, Talukdar S, Hellström A, Smith LEH. Omega-3/Omega-6 Long-Chain Fatty Acid Imbalance in Phase I Retinopathy of Prematurity. Nutrients 2022; 14:1333. [PMID: 35405946 PMCID: PMC9002570 DOI: 10.3390/nu14071333] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 11/17/2022] Open
Abstract
There is a gap in understanding the effect of the essential ω-3 and ω-6 long-chain polyunsaturated fatty acids (LCPUFA) on Phase I retinopathy of prematurity (ROP), which precipitates proliferative ROP. Postnatal hyperglycemia contributes to Phase I ROP by delaying retinal vascularization. In mouse neonates with hyperglycemia-associated Phase I retinopathy, dietary ω-3 (vs. ω-6 LCPUFA) supplementation promoted retinal vessel development. However, ω-6 (vs. ω-3 LCPUFA) was also developmentally essential, promoting neuronal growth and metabolism as suggested by a strong metabolic shift in almost all types of retinal neuronal and glial cells identified with single-cell transcriptomics. Loss of adiponectin (APN) in mice (mimicking the low APN levels in Phase I ROP) decreased LCPUFA levels (including ω-3 and ω-6) in retinas under normoglycemic and hyperglycemic conditions. ω-3 (vs. ω-6) LCPUFA activated the APN pathway by increasing the circulating APN levels and inducing expression of the retinal APN receptor. Our findings suggested that both ω-3 and ω-6 LCPUFA are crucial in protecting against retinal neurovascular dysfunction in a Phase I ROP model; adequate ω-6 LCPUFA levels must be maintained in addition to ω-3 supplementation to prevent retinopathy. Activation of the APN pathway may further enhance the ω-3 and ω-6 LCPUFA's protection against ROP.
Collapse
Affiliation(s)
- Zhongjie Fu
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (E.B.); (W.A.); (J.Y.); (M.K.); (J.D.A.)
| | - Wenjun Yan
- Center for Brain Science, Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA;
| | - Chuck T. Chen
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD 20814, USA;
| | - Anders K. Nilsson
- Section for Ophthalmology, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 412 96 Gothenburg, Sweden; (A.K.N.); (A.H.)
| | - Edward Bull
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (E.B.); (W.A.); (J.Y.); (M.K.); (J.D.A.)
| | - William Allen
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (E.B.); (W.A.); (J.Y.); (M.K.); (J.D.A.)
| | - Jay Yang
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (E.B.); (W.A.); (J.Y.); (M.K.); (J.D.A.)
| | - Minji Ko
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (E.B.); (W.A.); (J.Y.); (M.K.); (J.D.A.)
| | - John Paul SanGiovanni
- BIO5 Institute, Department of Nutritional Sciences, The University of Arizona, Tucson, AZ 85721, USA;
| | - James D. Akula
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (E.B.); (W.A.); (J.Y.); (M.K.); (J.D.A.)
| | - Saswata Talukdar
- Cardiometabolic Diseases, Merck Research Laboratories, Boston, MA 02115, USA;
| | - Ann Hellström
- Section for Ophthalmology, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 412 96 Gothenburg, Sweden; (A.K.N.); (A.H.)
| | - Lois E. H. Smith
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (E.B.); (W.A.); (J.Y.); (M.K.); (J.D.A.)
| |
Collapse
|
23
|
Alevin-fry unlocks rapid, accurate and memory-frugal quantification of single-cell RNA-seq data. Nat Methods 2022; 19:316-322. [PMID: 35277707 PMCID: PMC8933848 DOI: 10.1038/s41592-022-01408-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 01/27/2022] [Indexed: 01/19/2023]
Abstract
The rapid growth of high-throughput single-cell and single-nucleus RNA-sequencing (sc/snRNA-seq) technologies has produced a wealth of data over the past few years. The size, volume, and distinctive characteristics of these data necessitate the development of new computational methods to accurately and efficiently quantify sc/snRNA-seq data into count matrices that constitute the input to downstream analyses. We introduce the alevin-fry framework for quantifying sc/snRNA-seq data. In addition to being faster and more memory frugal than other accurate quantification approaches, alevin-fry ameliorates the memory scalability and false-positive expression issues that are exhibited by other lightweight tools. We demonstrate how alevin-fry can be effectively used to quantify sc/snRNA-seq data, and also how the spliced and unspliced molecule quantification required as input for RNA velocity analyses can be seamlessly extracted from the same preprocessed data used to generate regular gene expression count matrices.
Collapse
|
24
|
Nguyen DT, Trac QT, Nguyen TH, Nguyen HN, Ohad N, Pawitan Y, Vu TN. Circall: fast and accurate methodology for discovery of circular RNAs from paired-end RNA-sequencing data. BMC Bioinformatics 2021; 22:495. [PMID: 34645386 PMCID: PMC8513298 DOI: 10.1186/s12859-021-04418-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 09/29/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Circular RNA (circRNA) is an emerging class of RNA molecules attracting researchers due to its potential for serving as markers for diagnosis, prognosis, or therapeutic targets of cancer, cardiovascular, and autoimmune diseases. Current methods for detection of circRNA from RNA sequencing (RNA-seq) focus mostly on improving mapping quality of reads supporting the back-splicing junction (BSJ) of a circRNA to eliminate false positives (FPs). We show that mapping information alone often cannot predict if a BSJ-supporting read is derived from a true circRNA or not, thus increasing the rate of FP circRNAs. RESULTS We have developed Circall, a novel circRNA detection method from RNA-seq. Circall controls the FPs using a robust multidimensional local false discovery rate method based on the length and expression of circRNAs. It is computationally highly efficient by using a quasi-mapping algorithm for fast and accurate RNA read alignments. We applied Circall on two simulated datasets and three experimental datasets of human cell-lines. The results show that Circall achieves high sensitivity and precision in the simulated data. In the experimental datasets it performs well against current leading methods. Circall is also substantially faster than the other methods, particularly for large datasets. CONCLUSIONS With those better performances in the detection of circRNAs and in computational time, Circall facilitates the analyses of circRNAs in large numbers of samples. Circall is implemented in C++ and R, and available for use at https://www.meb.ki.se/sites/biostatwiki/circall and https://github.com/datngu/Circall.
Collapse
Affiliation(s)
- Dat Thanh Nguyen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Quang Thinh Trac
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Thi-Hau Nguyen
- University of Engineering and Technology, Vietnam National University in Hanoi, Hanoi, Vietnam
| | - Ha-Nam Nguyen
- Information Technology Institute, Vietnam National University in Hanoi, Hanoi, Vietnam
| | - Nir Ohad
- School of Plant Sciences and Food Security, Tel Aviv University, Tel Aviv, Israel
| | - Yudi Pawitan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Trung Nghia Vu
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
25
|
Pereira JD, DuBreuil DM, Devlin AC, Held A, Sapir Y, Berezovski E, Hawrot J, Dorfman K, Chander V, Wainger BJ. Human sensorimotor organoids derived from healthy and amyotrophic lateral sclerosis stem cells form neuromuscular junctions. Nat Commun 2021; 12:4744. [PMID: 34362895 PMCID: PMC8346474 DOI: 10.1038/s41467-021-24776-4] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 07/06/2021] [Indexed: 02/07/2023] Open
Abstract
Human induced pluripotent stem cells (iPSC) hold promise for modeling diseases in individual human genetic backgrounds and thus for developing precision medicine. Here, we generate sensorimotor organoids containing physiologically functional neuromuscular junctions (NMJs) and apply the model to different subgroups of amyotrophic lateral sclerosis (ALS). Using a range of molecular, genomic, and physiological techniques, we identify and characterize motor neurons and skeletal muscle, along with sensory neurons, astrocytes, microglia, and vasculature. Organoid cultures derived from multiple human iPSC lines generated from individuals with ALS and isogenic lines edited to harbor familial ALS mutations show impairment at the level of the NMJ, as detected by both contraction and immunocytochemical measurements. The physiological resolution of the human NMJ synapse, combined with the generation of major cellular cohorts exerting autonomous and non-cell autonomous effects in motor and sensory diseases, may prove valuable to understand the pathophysiological mechanisms of ALS.
Collapse
Affiliation(s)
- João D Pereira
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniel M DuBreuil
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Anna-Claire Devlin
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Aaron Held
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yechiam Sapir
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Eugene Berezovski
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - James Hawrot
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Katherine Dorfman
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Vignesh Chander
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Brian J Wainger
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
- Broad Institute of Harvard University and MIT, Cambridge, MA, USA.
| |
Collapse
|
26
|
Denti L, Pirola Y, Previtali M, Ceccato T, Della Vedova G, Rizzi R, Bonizzoni P. Shark: fishing relevant reads in an RNA-Seq sample. Bioinformatics 2021; 37:464-472. [PMID: 32926128 PMCID: PMC8088329 DOI: 10.1093/bioinformatics/btaa779] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 08/17/2020] [Accepted: 09/02/2020] [Indexed: 11/19/2022] Open
Abstract
Motivation Recent advances in high-throughput RNA-Seq technologies allow to produce massive datasets. When a study focuses only on a handful of genes, most reads are not relevant and degrade the performance of the tools used to analyze the data. Removing irrelevant reads from the input dataset leads to improved efficiency without compromising the results of the study. Results We introduce a novel computational problem, called gene assignment and we propose an efficient alignment-free approach to solve it. Given an RNA-Seq sample and a panel of genes, a gene assignment consists in extracting from the sample, the reads that most probably were sequenced from those genes. The problem becomes more complicated when the sample exhibits evidence of novel alternative splicing events. We implemented our approach in a tool called Shark and assessed its effectiveness in speeding up differential splicing analysis pipelines. This evaluation shows that Shark is able to significantly improve the performance of RNA-Seq analysis tools without having any impact on the final results. Availability and implementation The tool is distributed as a stand-alone module and the software is freely available at https://github.com/AlgoLab/shark. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Luca Denti
- Department of Informatics, Systems and Communication, University of Milano-Bicocca, Milano 20126, Italy
| | - Yuri Pirola
- Department of Informatics, Systems and Communication, University of Milano-Bicocca, Milano 20126, Italy
| | - Marco Previtali
- Department of Informatics, Systems and Communication, University of Milano-Bicocca, Milano 20126, Italy
| | - Tamara Ceccato
- Department of Informatics, Systems and Communication, University of Milano-Bicocca, Milano 20126, Italy
| | - Gianluca Della Vedova
- Department of Informatics, Systems and Communication, University of Milano-Bicocca, Milano 20126, Italy
| | - Raffaella Rizzi
- Department of Informatics, Systems and Communication, University of Milano-Bicocca, Milano 20126, Italy
| | - Paola Bonizzoni
- Department of Informatics, Systems and Communication, University of Milano-Bicocca, Milano 20126, Italy
| |
Collapse
|
27
|
Zolfaghari Emameh R, Hosseini SN, Parkkila S. Application of beta and gamma carbonic anhydrase sequences as tools for identification of bacterial contamination in the whole genome sequence of inbred Wuzhishan minipig (Sus scrofa) annotated in databases. Database (Oxford) 2021; 2021:baab029. [PMID: 34003248 PMCID: PMC8130508 DOI: 10.1093/database/baab029] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/19/2021] [Accepted: 05/11/2021] [Indexed: 11/13/2022]
Abstract
Sus scrofa or pig was domesticated thousands of years ago. Through various indigenous breeds, different phenotypes were produced such as Chinese inbred miniature minipig or Wuzhishan pig (WZSP), which is broadly used in the life and medical sciences. The whole genome of WZSP was sequenced in 2012. Through a bioinformatics study of pig carbonic anhydrase (CA) sequences, we detected some β- and γ-class CAs among the WZSP CAs annotated in databases, while β- or γ-CAs had not previously been described in vertebrates. This finding urged us to analyze the quality of whole genome sequence of WZSP for the possible bacterial contamination. In this study, we used bioinformatics methods and web tools such as UniProt, European Bioinformatics Institute, National Center for Biotechnology Information, Ensembl Genome Browser, Ensembl Bacteria, RSCB PDB and Pseudomonas Genome Database. Our analysis defined that pig has 12 classical α-CAs and 3 CA-related proteins. Meanwhile, it was approved that the detected CAs in WZSP are categorized in the β- and γ-CA families, which belong to Pseudomonas spp. and Acinetobacter spp. The protein structure study revealed that the identified β-CA sequence from WZSP belongs to Pseudomonas aeruginosa with PDB ID: 5JJ8, and the identified γ-CA sequence from WZSP belongs to P. aeruginosa with PDB ID: 3PMO. Bioinformatics and computational methods accompanied with bacterial-specific markers, such as 16S rRNA and β- and γ-class CA sequences, can be used to identify bacterial contamination in mammalian DNA samples.
Collapse
Affiliation(s)
- Reza Zolfaghari Emameh
- Department of Energy and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), 14965/161, Tehran, Iran
| | - Seyed Nezamedin Hosseini
- Department of Recombinant Hepatitis B Vaccine, Production and Research Complex, Pasteur Institute of Iran, Tehran, Iran
| | - Seppo Parkkila
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Fimlab Ltd, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
28
|
Rodriguez S, Hug C, Todorov P, Moret N, Boswell SA, Evans K, Zhou G, Johnson NT, Hyman BT, Sorger PK, Albers MW, Sokolov A. Machine learning identifies candidates for drug repurposing in Alzheimer's disease. Nat Commun 2021; 12:1033. [PMID: 33589615 PMCID: PMC7884393 DOI: 10.1038/s41467-021-21330-0] [Citation(s) in RCA: 139] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 01/21/2021] [Indexed: 01/31/2023] Open
Abstract
Clinical trials of novel therapeutics for Alzheimer's Disease (AD) have consumed a large amount of time and resources with largely negative results. Repurposing drugs already approved by the Food and Drug Administration (FDA) for another indication is a more rapid and less expensive option. We present DRIAD (Drug Repurposing In AD), a machine learning framework that quantifies potential associations between the pathology of AD severity (the Braak stage) and molecular mechanisms as encoded in lists of gene names. DRIAD is applied to lists of genes arising from perturbations in differentiated human neural cell cultures by 80 FDA-approved and clinically tested drugs, producing a ranked list of possible repurposing candidates. Top-scoring drugs are inspected for common trends among their targets. We propose that the DRIAD method can be used to nominate drugs that, after additional validation and identification of relevant pharmacodynamic biomarker(s), could be readily evaluated in a clinical trial.
Collapse
Affiliation(s)
- Steve Rodriguez
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Clemens Hug
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA
| | - Petar Todorov
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA
| | - Nienke Moret
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA
| | - Sarah A Boswell
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA
| | - Kyle Evans
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - George Zhou
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Nathan T Johnson
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Peter K Sorger
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA
| | - Mark W Albers
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA.
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA.
| | - Artem Sokolov
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
29
|
Reiter T, Brooks† PT, Irber† L, Joslin† SEK, Reid† CM, Scott† C, Brown CT, Pierce-Ward NT. Streamlining data-intensive biology with workflow systems. Gigascience 2021; 10:giaa140. [PMID: 33438730 PMCID: PMC8631065 DOI: 10.1093/gigascience/giaa140] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 11/06/2020] [Accepted: 11/13/2020] [Indexed: 11/14/2022] Open
Abstract
As the scale of biological data generation has increased, the bottleneck of research has shifted from data generation to analysis. Researchers commonly need to build computational workflows that include multiple analytic tools and require incremental development as experimental insights demand tool and parameter modifications. These workflows can produce hundreds to thousands of intermediate files and results that must be integrated for biological insight. Data-centric workflow systems that internally manage computational resources, software, and conditional execution of analysis steps are reshaping the landscape of biological data analysis and empowering researchers to conduct reproducible analyses at scale. Adoption of these tools can facilitate and expedite robust data analysis, but knowledge of these techniques is still lacking. Here, we provide a series of strategies for leveraging workflow systems with structured project, data, and resource management to streamline large-scale biological analysis. We present these practices in the context of high-throughput sequencing data analysis, but the principles are broadly applicable to biologists working beyond this field.
Collapse
Affiliation(s)
- Taylor Reiter
- Department of Population Health and Reproduction, University of California, Davis, 1 Shields Avenue, Davis, CA 95616, USA
| | - Phillip T Brooks†
- Department of Population Health and Reproduction, University of California, Davis, 1 Shields Avenue, Davis, CA 95616, USA
| | - Luiz Irber†
- Department of Population Health and Reproduction, University of California, Davis, 1 Shields Avenue, Davis, CA 95616, USA
| | - Shannon E K Joslin†
- Department of Animal Science, University of California, Davis, 1 Shields Avenue, Davis, CA 95616, USA
| | - Charles M Reid†
- Department of Population Health and Reproduction, University of California, Davis, 1 Shields Avenue, Davis, CA 95616, USA
| | - Camille Scott†
- Department of Population Health and Reproduction, University of California, Davis, 1 Shields Avenue, Davis, CA 95616, USA
| | - C Titus Brown
- Department of Population Health and Reproduction, University of California, Davis, 1 Shields Avenue, Davis, CA 95616, USA
| | - N Tessa Pierce-Ward
- Department of Population Health and Reproduction, University of California, Davis, 1 Shields Avenue, Davis, CA 95616, USA
| |
Collapse
|
30
|
Binkley MS, Jeon YJ, Nesselbush M, Moding EJ, Nabet BY, Almanza D, Kunder C, Stehr H, Yoo CH, Rhee S, Xiang M, Chabon JJ, Hamilton E, Kurtz DM, Gojenola L, Owen SG, Ko RB, Shin JH, Maxim PG, Lui NS, Backhus LM, Berry MF, Shrager JB, Ramchandran KJ, Padda SK, Das M, Neal JW, Wakelee HA, Alizadeh AA, Loo BW, Diehn M. KEAP1/NFE2L2 Mutations Predict Lung Cancer Radiation Resistance That Can Be Targeted by Glutaminase Inhibition. Cancer Discov 2020; 10:1826-1841. [PMID: 33071215 PMCID: PMC7710558 DOI: 10.1158/2159-8290.cd-20-0282] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 08/12/2020] [Accepted: 09/16/2020] [Indexed: 11/16/2022]
Abstract
Tumor genotyping is not routinely performed in localized non-small cell lung cancer (NSCLC) due to lack of associations of mutations with outcome. Here, we analyze 232 consecutive patients with localized NSCLC and demonstrate that KEAP1 and NFE2L2 mutations are predictive of high rates of local recurrence (LR) after radiotherapy but not surgery. Half of LRs occurred in tumors with KEAP1/NFE2L2 mutations, indicating that they are major molecular drivers of clinical radioresistance. Next, we functionally evaluate KEAP1/NFE2L2 mutations in our radiotherapy cohort and demonstrate that only pathogenic mutations are associated with radioresistance. Furthermore, expression of NFE2L2 target genes does not predict LR, underscoring the utility of tumor genotyping. Finally, we show that glutaminase inhibition preferentially radiosensitizes KEAP1-mutant cells via depletion of glutathione and increased radiation-induced DNA damage. Our findings suggest that genotyping for KEAP1/NFE2L2 mutations could facilitate treatment personalization and provide a potential strategy for overcoming radioresistance conferred by these mutations. SIGNIFICANCE: This study shows that mutations in KEAP1 and NFE2L2 predict for LR after radiotherapy but not surgery in patients with NSCLC. Approximately half of all LRs are associated with these mutations and glutaminase inhibition may allow personalized radiosensitization of KEAP1/NFE2L2-mutant tumors.This article is highlighted in the In This Issue feature, p. 1775.
Collapse
Affiliation(s)
- Michael S Binkley
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Young-Jun Jeon
- Stanford Cancer Institute, Stanford, California
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | | | - Everett J Moding
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Barzin Y Nabet
- Department of Radiation Oncology, Stanford University, Stanford, California
- Stanford Cancer Institute, Stanford, California
| | - Diego Almanza
- Cancer Biology Program, Stanford University, Stanford, California
| | - Christian Kunder
- Department of Pathology, Stanford University, Stanford, California
| | - Henning Stehr
- Department of Pathology, Stanford University, Stanford, California
| | - Christopher H Yoo
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Siyeon Rhee
- Department of Biology, Stanford University, Stanford, California
| | - Michael Xiang
- Department of Radiation Oncology, University of California, Los Angeles, Los Angeles, California
| | | | - Emily Hamilton
- Cancer Biology Program, Stanford University, Stanford, California
| | - David M Kurtz
- Division of Oncology, Department of Medicine, Stanford University, Stanford, California
| | - Linda Gojenola
- Department of Pathology, Stanford University, Stanford, California
| | - Susie Grant Owen
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Ryan B Ko
- Department of Radiation Oncology, Stanford University, Stanford, California
| | | | - Peter G Maxim
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Natalie S Lui
- Division of Thoracic Surgery, Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, California
| | - Leah M Backhus
- Division of Thoracic Surgery, Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, California
| | - Mark F Berry
- Division of Thoracic Surgery, Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, California
| | - Joseph B Shrager
- Division of Thoracic Surgery, Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, California
| | - Kavitha J Ramchandran
- Stanford Cancer Institute, Stanford, California
- Division of Oncology, Department of Medicine, Stanford University, Stanford, California
| | - Sukhmani K Padda
- Stanford Cancer Institute, Stanford, California
- Division of Oncology, Department of Medicine, Stanford University, Stanford, California
| | - Millie Das
- Stanford Cancer Institute, Stanford, California
- Division of Oncology, Department of Medicine, Stanford University, Stanford, California
| | - Joel W Neal
- Stanford Cancer Institute, Stanford, California
- Division of Oncology, Department of Medicine, Stanford University, Stanford, California
| | - Heather A Wakelee
- Stanford Cancer Institute, Stanford, California
- Division of Oncology, Department of Medicine, Stanford University, Stanford, California
| | - Ash A Alizadeh
- Stanford Cancer Institute, Stanford, California
- Division of Oncology, Department of Medicine, Stanford University, Stanford, California
| | - Billy W Loo
- Department of Radiation Oncology, Stanford University, Stanford, California
- Stanford Cancer Institute, Stanford, California
| | - Maximilian Diehn
- Department of Radiation Oncology, Stanford University, Stanford, California.
- Stanford Cancer Institute, Stanford, California
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California
| |
Collapse
|
31
|
Ha CWY, Martin A, Sepich-Poore GD, Shi B, Wang Y, Gouin K, Humphrey G, Sanders K, Ratnayake Y, Chan KSL, Hendrick G, Caldera JR, Arias C, Moskowitz JE, Ho Sui SJ, Yang S, Underhill D, Brady MJ, Knott S, Kaihara K, Steinbaugh MJ, Li H, McGovern DPB, Knight R, Fleshner P, Devkota S. Translocation of Viable Gut Microbiota to Mesenteric Adipose Drives Formation of Creeping Fat in Humans. Cell 2020; 183:666-683.e17. [PMID: 32991841 PMCID: PMC7521382 DOI: 10.1016/j.cell.2020.09.009] [Citation(s) in RCA: 262] [Impact Index Per Article: 52.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 07/19/2020] [Accepted: 09/01/2020] [Indexed: 02/08/2023]
Abstract
A mysterious feature of Crohn's disease (CD) is the extra-intestinal manifestation of "creeping fat" (CrF), defined as expansion of mesenteric adipose tissue around the inflamed and fibrotic intestine. In the current study, we explore whether microbial translocation in CD serves as a central cue for CrF development. We discovered a subset of mucosal-associated gut bacteria that consistently translocated and remained viable in CrF in CD ileal surgical resections, and identified Clostridium innocuum as a signature of this consortium with strain variation between mucosal and adipose isolates, suggesting preference for lipid-rich environments. Single-cell RNA sequencing characterized CrF as both pro-fibrotic and pro-adipogenic with a rich milieu of activated immune cells responding to microbial stimuli, which we confirm in gnotobiotic mice colonized with C. innocuum. Ex vivo validation of expression patterns suggests C. innocuum stimulates tissue remodeling via M2 macrophages, leading to an adipose tissue barrier that serves to prevent systemic dissemination of bacteria.
Collapse
Affiliation(s)
- Connie W Y Ha
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Anthony Martin
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Gregory D Sepich-Poore
- Department of Bioengineering, University of California San Diego, La Jolla, California 92093, USA
| | - Baochen Shi
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Yizhou Wang
- Applied Genomics, Computation and Translational Core, Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Kenneth Gouin
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Applied Genomics, Computation and Translational Core, Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Gregory Humphrey
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
| | - Karenina Sanders
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
| | | | | | - Gustaf Hendrick
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - J R Caldera
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Christian Arias
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jacob E Moskowitz
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Shannan J Ho Sui
- Harvard Chan Bioinformatics Core, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Shaohong Yang
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - David Underhill
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Matthew J Brady
- Department of Medicine, Section of Endocrinology and Metabolism, The University of Chicago, Chicago, IL 60637, USA
| | - Simon Knott
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Applied Genomics, Computation and Translational Core, Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | | | - Michael J Steinbaugh
- Harvard Chan Bioinformatics Core, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Huiying Li
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Dermot P B McGovern
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Rob Knight
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA; Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Phillip Fleshner
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Division of Colorectal Surgery, Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Suzanne Devkota
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| |
Collapse
|
32
|
Srivastava A, Malik L, Sarkar H, Zakeri M, Almodaresi F, Soneson C, Love MI, Kingsford C, Patro R. Alignment and mapping methodology influence transcript abundance estimation. Genome Biol 2020; 21:239. [PMID: 32894187 PMCID: PMC7487471 DOI: 10.1186/s13059-020-02151-8] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 08/19/2020] [Indexed: 01/23/2023] Open
Abstract
Background The accuracy of transcript quantification using RNA-seq data depends on many factors, such as the choice of alignment or mapping method and the quantification model being adopted. While the choice of quantification model has been shown to be important, considerably less attention has been given to comparing the effect of various read alignment approaches on quantification accuracy. Results We investigate the influence of mapping and alignment on the accuracy of transcript quantification in both simulated and experimental data, as well as the effect on subsequent differential expression analysis. We observe that, even when the quantification model itself is held fixed, the effect of choosing a different alignment methodology, or aligning reads using different parameters, on quantification estimates can sometimes be large and can affect downstream differential expression analyses as well. These effects can go unnoticed when assessment is focused too heavily on simulated data, where the alignment task is often simpler than in experimentally acquired samples. We also introduce a new alignment methodology, called selective alignment, to overcome the shortcomings of lightweight approaches without incurring the computational cost of traditional alignment. Conclusion We observe that, on experimental datasets, the performance of lightweight mapping and alignment-based approaches varies significantly, and highlight some of the underlying factors. We show this variation both in terms of quantification and downstream differential expression analysis. In all comparisons, we also show the improved performance of our proposed selective alignment method and suggest best practices for performing RNA-seq quantification.
Collapse
Affiliation(s)
- Avi Srivastava
- Department of Computer Science, Stony Brook University, Stony Brook, USA
| | - Laraib Malik
- Department of Computer Science, Stony Brook University, Stony Brook, USA
| | - Hirak Sarkar
- Department of Computer Science, University of Maryland, College Park, USA
| | - Mohsen Zakeri
- Department of Computer Science, University of Maryland, College Park, USA
| | - Fatemeh Almodaresi
- Department of Computer Science, University of Maryland, College Park, USA
| | - Charlotte Soneson
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland.,SIB Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Michael I Love
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, USA.,Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Carl Kingsford
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, USA
| | - Rob Patro
- Department of Computer Science, University of Maryland, College Park, USA.
| |
Collapse
|
33
|
Saxena S, Lokhande H, Gombolay G, Raheja R, Rooney T, Chitnis T. Identification of TNFAIP3 as relapse biomarker and potential therapeutic target for MOG antibody associated diseases. Sci Rep 2020; 10:12405. [PMID: 32709905 PMCID: PMC7381621 DOI: 10.1038/s41598-020-69182-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 07/02/2020] [Indexed: 12/23/2022] Open
Abstract
MOG-antibody associated disease (MOG-AAD) is a recently recognized demyelinating disorder predominantly affecting children but also occurs in adults, with a relapsing course in approximately 50% of patients. We evaluated peripheral blood mononuclear cells from MOG-AAD patients by flow cytometry and found a strong antigen specific central memory cell (CMC) response with increased Th1 and Th17 cells at the time of a relapse. Transcriptomic analysis of CMCs by three independent sequencing platforms revealed TNFAIP3 as a relapse biomarker, whose expression was down regulated at a relapse compared to remission in MOG-AAD patients. Serum in an additional cohort of patients showed decreased TNFAIP3 levels at relapse compared to remission state in MOG-AAD patients. Our studies suggest that alterations in TNFAIP3 levels are associated with relapses in MOG-AAD patients, which may have clinical utility as a disease course biomarker and therapeutic target.
Collapse
Affiliation(s)
- Shrishti Saxena
- Ann Romney Center for Neurologic Disease, Brigham and Women's Hospital, Harvard Medical School, 60 Fenwood Road, Office 9002K, Boston, MA, 02115-6128, USA
| | - Hrishikesh Lokhande
- Ann Romney Center for Neurologic Disease, Brigham and Women's Hospital, Harvard Medical School, 60 Fenwood Road, Office 9002K, Boston, MA, 02115-6128, USA
| | - Grace Gombolay
- Ann Romney Center for Neurologic Disease, Brigham and Women's Hospital, Harvard Medical School, 60 Fenwood Road, Office 9002K, Boston, MA, 02115-6128, USA.,Emory University and Children's Healthcare of Atlanta, Atlanta, GA, 30329, USA.,Department of Neurology, Partners Pediatric Multiple Sclerosis Center, Massachusetts General Hospital, Boston, MA, USA
| | - Radhika Raheja
- Ann Romney Center for Neurologic Disease, Brigham and Women's Hospital, Harvard Medical School, 60 Fenwood Road, Office 9002K, Boston, MA, 02115-6128, USA
| | - Timothy Rooney
- Ann Romney Center for Neurologic Disease, Brigham and Women's Hospital, Harvard Medical School, 60 Fenwood Road, Office 9002K, Boston, MA, 02115-6128, USA
| | - Tanuja Chitnis
- Ann Romney Center for Neurologic Disease, Brigham and Women's Hospital, Harvard Medical School, 60 Fenwood Road, Office 9002K, Boston, MA, 02115-6128, USA. .,Department of Neurology, Partners Pediatric Multiple Sclerosis Center, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
34
|
Gao M, Ling M, Tang X, Wang S, Xiao X, Qiao Y, Yang W, Yu R. Comparison of high-throughput single-cell RNA sequencing data processing pipelines. Brief Bioinform 2020; 22:5868074. [PMID: 34020539 DOI: 10.1093/bib/bbaa116] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 04/23/2020] [Accepted: 05/17/2020] [Indexed: 11/13/2022] Open
Abstract
With the development of single-cell RNA sequencing (scRNA-seq) technology, it has become possible to perform large-scale transcript profiling for tens of thousands of cells in a single experiment. Many analysis pipelines have been developed for data generated from different high-throughput scRNA-seq platforms, bringing a new challenge to users to choose a proper workflow that is efficient, robust and reliable for a specific sequencing platform. Moreover, as the amount of public scRNA-seq data has increased rapidly, integrated analysis of scRNA-seq data from different sources has become increasingly popular. However, it remains unclear whether such integrated analysis would be biassed if the data were processed by different upstream pipelines. In this study, we encapsulated seven existing high-throughput scRNA-seq data processing pipelines with Nextflow, a general integrative workflow management framework, and evaluated their performance in terms of running time, computational resource consumption and data analysis consistency using eight public datasets generated from five different high-throughput scRNA-seq platforms. Our work provides a useful guideline for the selection of scRNA-seq data processing pipelines based on their performance on different real datasets. In addition, these guidelines can serve as a performance evaluation framework for future developments in high-throughput scRNA-seq data processing.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Rongshan Yu
- Digital Fujian Institute of Healthcare and Biomedical Big Data, School of Informatic, Xiamen University
| |
Collapse
|
35
|
Niebler S, Müller A, Hankeln T, Schmidt B. RainDrop: Rapid activation matrix computation for droplet-based single-cell RNA-seq reads. BMC Bioinformatics 2020; 21:274. [PMID: 32611394 PMCID: PMC7329424 DOI: 10.1186/s12859-020-03593-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 06/09/2020] [Indexed: 12/19/2022] Open
Abstract
Background Obtaining data from single-cell transcriptomic sequencing allows for the investigation of cell-specific gene expression patterns, which could not be addressed a few years ago. With the advancement of droplet-based protocols the number of studied cells continues to increase rapidly. This establishes the need for software tools for efficient processing of the produced large-scale datasets. We address this need by presenting RainDrop for fast gene-cell count matrix computation from single-cell RNA-seq data produced by 10x Genomics Chromium technology. Results RainDrop can process single-cell transcriptomic datasets consisting of 784 million reads sequenced from around 8.000 cells in less than 40 minutes on a standard workstation. It significantly outperforms the established Cell Ranger pipeline and the recently introduced Alevin tool in terms of runtime by a maximal (average) speedup of 30.4 (22.6) and 3.5 (2.4), respectively, while keeping high agreements of the generated results. Conclusions RainDrop is a software tool for highly efficient processing of large-scale droplet-based single-cell RNA-seq datasets on standard workstations written in C++. It is available at https://gitlab.rlp.net/stnieble/raindrop.
Collapse
Affiliation(s)
- Stefan Niebler
- Department of Computer Science, Johannes Gutenberg University, Mainz, 55099, Germany
| | - André Müller
- Department of Computer Science, Johannes Gutenberg University, Mainz, 55099, Germany
| | - Thomas Hankeln
- Molecular Genetics and Genome Analysis, Institute of Organismal and Molecular Evolution, Johannes Gutenberg University, Mainz, 55099, Germany
| | - Bertil Schmidt
- Department of Computer Science, Johannes Gutenberg University, Mainz, 55099, Germany.
| |
Collapse
|
36
|
Tattikota SG, Cho B, Liu Y, Hu Y, Barrera V, Steinbaugh MJ, Yoon SH, Comjean A, Li F, Dervis F, Hung RJ, Nam JW, Ho Sui S, Shim J, Perrimon N. A single-cell survey of Drosophila blood. eLife 2020; 9:e54818. [PMID: 32396065 PMCID: PMC7237219 DOI: 10.7554/elife.54818] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 05/08/2020] [Indexed: 12/30/2022] Open
Abstract
Drosophila blood cells, called hemocytes, are classified into plasmatocytes, crystal cells, and lamellocytes based on the expression of a few marker genes and cell morphologies, which are inadequate to classify the complete hemocyte repertoire. Here, we used single-cell RNA sequencing (scRNA-seq) to map hemocytes across different inflammatory conditions in larvae. We resolved plasmatocytes into different states based on the expression of genes involved in cell cycle, antimicrobial response, and metabolism together with the identification of intermediate states. Further, we discovered rare subsets within crystal cells and lamellocytes that express fibroblast growth factor (FGF) ligand branchless and receptor breathless, respectively. We demonstrate that these FGF components are required for mediating effective immune responses against parasitoid wasp eggs, highlighting a novel role for FGF signaling in inter-hemocyte crosstalk. Our scRNA-seq analysis reveals the diversity of hemocytes and provides a rich resource of gene expression profiles for a systems-level understanding of their functions.
Collapse
Affiliation(s)
| | - Bumsik Cho
- Department of Life Science, Hanyang UniversitySeoulRepublic of Korea
| | - Yifang Liu
- Department of Genetics, Blavatnik Institute, Harvard Medical SchoolBostonUnited States
| | - Yanhui Hu
- Department of Genetics, Blavatnik Institute, Harvard Medical SchoolBostonUnited States
| | | | | | - Sang-Ho Yoon
- Department of Life Science, Hanyang UniversitySeoulRepublic of Korea
| | - Aram Comjean
- Department of Genetics, Blavatnik Institute, Harvard Medical SchoolBostonUnited States
| | - Fangge Li
- Department of Genetics, Blavatnik Institute, Harvard Medical SchoolBostonUnited States
| | - Franz Dervis
- Department of Genetics, Blavatnik Institute, Harvard Medical SchoolBostonUnited States
| | - Ruei-Jiun Hung
- Department of Genetics, Blavatnik Institute, Harvard Medical SchoolBostonUnited States
| | - Jin-Wu Nam
- Department of Life Science, Hanyang UniversitySeoulRepublic of Korea
| | | | - Jiwon Shim
- Department of Life Science, Hanyang UniversitySeoulRepublic of Korea
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical SchoolBostonUnited States
- Howard Hughes Medical InstituteBostonUnited States
| |
Collapse
|
37
|
Asghari H, Lin YY, Xu Y, Haghshenas E, Collins CC, Hach F. CircMiner: accurate and rapid detection of circular RNA through splice-aware pseudo-alignment scheme. Bioinformatics 2020; 36:3703-3711. [DOI: 10.1093/bioinformatics/btaa232] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 03/23/2020] [Accepted: 04/01/2020] [Indexed: 12/14/2022] Open
Abstract
Abstract
Motivation
The ubiquitous abundance of circular RNAs (circRNAs) has been revealed by performing high-throughput sequencing in a variety of eukaryotes. circRNAs are related to some diseases, such as cancer in which they act as oncogenes or tumor-suppressors and, therefore, have the potential to be used as biomarkers or therapeutic targets. Accurate and rapid detection of circRNAs from short reads remains computationally challenging. This is due to the fact that identifying chimeric reads, which is essential for finding back-splice junctions, is a complex process. The sensitivity of discovery methods, to a high degree, relies on the underlying mapper that is used for finding chimeric reads. Furthermore, all the available circRNA discovery pipelines are resource intensive.
Results
We introduce CircMiner, a novel stand-alone circRNA detection method that rapidly identifies and filters out linear RNA sequencing reads and detects back-splice junctions. CircMiner employs a rapid pseudo-alignment technique to identify linear reads that originate from transcripts, genes or the genome. CircMiner further processes the remaining reads to identify the back-splice junctions and detect circRNAs with single-nucleotide resolution. We evaluated the efficacy of CircMiner using simulated datasets generated from known back-splice junctions and showed that CircMiner has superior accuracy and speed compared to the existing circRNA detection tools. Additionally, on two RNase R treated cell line datasets, CircMiner was able to detect most of consistent, high confidence circRNAs compared to untreated samples of the same cell line.
Availability and implementation
CircMiner is implemented in C++ and is available online at https://github.com/vpc-ccg/circminer.
Supplementary information
Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Hossein Asghari
- School of Computing Science, Simon Fraser University, Burnaby, BC V5A1S6, Canada
- Vancouver Prostate Centre, Vancouver, BC V6H3Z6, Canada
| | - Yen-Yi Lin
- Vancouver Prostate Centre, Vancouver, BC V6H3Z6, Canada
| | - Yang Xu
- Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Ehsan Haghshenas
- School of Computing Science, Simon Fraser University, Burnaby, BC V5A1S6, Canada
- Vancouver Prostate Centre, Vancouver, BC V6H3Z6, Canada
| | - Colin C Collins
- Vancouver Prostate Centre, Vancouver, BC V6H3Z6, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC V5Z1M9, Canada
| | - Faraz Hach
- Vancouver Prostate Centre, Vancouver, BC V6H3Z6, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC V5Z1M9, Canada
| |
Collapse
|
38
|
Hu G, Grover CE, Arick MA, Liu M, Peterson DG, Wendel JF. Homoeologous gene expression and co-expression network analyses and evolutionary inference in allopolyploids. Brief Bioinform 2020; 22:1819-1835. [PMID: 32219306 PMCID: PMC7986634 DOI: 10.1093/bib/bbaa035] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/06/2020] [Accepted: 02/24/2020] [Indexed: 12/29/2022] Open
Abstract
Polyploidy is a widespread phenomenon throughout eukaryotes. Due to the coexistence of duplicated genomes, polyploids offer unique challenges for estimating gene expression levels, which is essential for understanding the massive and various forms of transcriptomic responses accompanying polyploidy. Although previous studies have explored the bioinformatics of polyploid transcriptomic profiling, the causes and consequences of inaccurate quantification of transcripts from duplicated gene copies have not been addressed. Using transcriptomic data from the cotton genus (Gossypium) as an example, we present an analytical workflow to evaluate a variety of bioinformatic method choices at different stages of RNA-seq analysis, from homoeolog expression quantification to downstream analysis used to infer key phenomena of polyploid expression evolution. In general, EAGLE-RC and GSNAP-PolyCat outperform other quantification pipelines tested, and their derived expression dataset best represents the expected homoeolog expression and co-expression divergence. The performance of co-expression network analysis was less affected by homoeolog quantification than by network construction methods, where weighted networks outperformed binary networks. By examining the extent and consequences of homoeolog read ambiguity, we illuminate the potential artifacts that may affect our understanding of duplicate gene expression, including an overestimation of homoeolog co-regulation and the incorrect inference of subgenome asymmetry in network topology. Taken together, our work points to a set of reasonable practices that we hope are broadly applicable to the evolutionary exploration of polyploids.
Collapse
Affiliation(s)
- Guanjing Hu
- Department of Ecology, Evolution, and Organismal Biology, Iowa State University, Ames, IA 50011, USA
| | - Corrinne E Grover
- Department of Ecology, Evolution, and Organismal Biology, Iowa State University, Ames, IA 50011, USA
| | - Mark A Arick
- Department of Ecology, Evolution, and Organismal Biology, Iowa State University, Ames, IA 50011, USA
| | - Meiling Liu
- Department of Ecology, Evolution, and Organismal Biology, Iowa State University, Ames, IA 50011, USA
| | - Daniel G Peterson
- Department of Ecology, Evolution, and Organismal Biology, Iowa State University, Ames, IA 50011, USA
| | - Jonathan F Wendel
- Department of Ecology, Evolution, and Organismal Biology, Iowa State University, Ames, IA 50011, USA
| |
Collapse
|
39
|
Schauer NJ, Liu X, Magin RS, Doherty LM, Chan WC, Ficarro SB, Hu W, Roberts RM, Iacob RE, Stolte B, Giacomelli AO, Perera S, McKay K, Boswell SA, Weisberg EL, Ray A, Chauhan D, Dhe-Paganon S, Anderson KC, Griffin JD, Li J, Hahn WC, Sorger PK, Engen JR, Stegmaier K, Marto JA, Buhrlage SJ. Selective USP7 inhibition elicits cancer cell killing through a p53-dependent mechanism. Sci Rep 2020; 10:5324. [PMID: 32210275 PMCID: PMC7093416 DOI: 10.1038/s41598-020-62076-x] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 02/25/2020] [Indexed: 12/21/2022] Open
Abstract
Ubiquitin specific peptidase 7 (USP7) is a deubiquitinating enzyme (DUB) that removes ubiquitin tags from specific protein substrates in order to alter their degradation rate and sub-cellular localization. USP7 has been proposed as a therapeutic target in several cancers because it has many reported substrates with a role in cancer progression, including FOXO4, MDM2, N-Myc, and PTEN. The multi-substrate nature of USP7, combined with the modest potency and selectivity of early generation USP7 inhibitors, has presented a challenge in defining predictors of response to USP7 and potential patient populations that would benefit most from USP7-targeted drugs. Here, we describe the structure-guided development of XL177A, which irreversibly inhibits USP7 with sub-nM potency and selectivity across the human proteome. Evaluation of the cellular effects of XL177A reveals that selective USP7 inhibition suppresses cancer cell growth predominantly through a p53-dependent mechanism: XL177A specifically upregulates p53 transcriptional targets transcriptome-wide, hotspot mutations in TP53 but not any other genes predict response to XL177A across a panel of ~500 cancer cell lines, and TP53 knockout rescues XL177A-mediated growth suppression of TP53 wild-type (WT) cells. Together, these findings suggest TP53 mutational status as a biomarker for response to USP7 inhibition. We find that Ewing sarcoma and malignant rhabdoid tumor (MRT), two pediatric cancers that are sensitive to other p53-dependent cytotoxic drugs, also display increased sensitivity to XL177A.
Collapse
Affiliation(s)
- Nathan J Schauer
- Department of Cancer Biology and the Linde Program in Cancer Chemical Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Xiaoxi Liu
- Department of Cancer Biology and the Linde Program in Cancer Chemical Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Robert S Magin
- Department of Cancer Biology and the Linde Program in Cancer Chemical Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Laura M Doherty
- Department of Cancer Biology and the Linde Program in Cancer Chemical Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
- Department of Systems Biology and Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Wai Cheung Chan
- Department of Cancer Biology and the Linde Program in Cancer Chemical Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Scott B Ficarro
- Department of Cancer Biology and the Linde Program in Cancer Chemical Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Oncologic Pathology and Blais Proteomics Center, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Wanyi Hu
- Department of Cancer Biology and the Linde Program in Cancer Chemical Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Rebekka M Roberts
- Department of Cancer Biology and the Linde Program in Cancer Chemical Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Roxana E Iacob
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA, USA
| | - Björn Stolte
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Boston, MA, USA
- Dr. von Hauner Children's Hospital, Department of Pediatrics, University Hospital, LMU Munich, Munich, Germany
- The Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Andrew O Giacomelli
- The Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | | | - Kyle McKay
- Department of Chemistry, University of Vermont, Burlington, VT, USA
| | - Sarah A Boswell
- Department of Systems Biology and Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Ellen L Weisberg
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Arghya Ray
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- The LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Dharminder Chauhan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- The LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sirano Dhe-Paganon
- Department of Cancer Biology and the Linde Program in Cancer Chemical Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Ken C Anderson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- The LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Dana-Farber Cancer Institute, Boston, MA, USA
| | - James D Griffin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jianing Li
- Department of Chemistry, University of Vermont, Burlington, VT, USA
| | - William C Hahn
- The Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Peter K Sorger
- Department of Systems Biology and Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
| | - John R Engen
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA, USA
| | - Kimberly Stegmaier
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Boston, MA, USA
- The Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Jarrod A Marto
- Department of Cancer Biology and the Linde Program in Cancer Chemical Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Oncologic Pathology and Blais Proteomics Center, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Sara J Buhrlage
- Department of Cancer Biology and the Linde Program in Cancer Chemical Biology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
40
|
Comparative Transcriptomic Response of Two Pinus Species to Infection with the Pine Wood Nematode Bursaphelenchus xylophilus. FORESTS 2020. [DOI: 10.3390/f11020204] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Pine wilt disease (PWD) caused by pine wood nematode (PWN), Bursaphelenchus xylophilus, is a serious threat to global forest populations of conifers, in particular Pinus spp. Recently, the presence of PWN was reported in dead Yunnan pine (Pinus yunnanensis) trees under natural conditions. To further understand the potential impact caused by PWN in Yunnan pine populations, a transcriptional profiling analysis was performed over different time points (0 hours (h), 6 h, 24 h, 48 h, and 7 days) after PWN inoculation. A total of 9961 differentially expressed genes were identified after inoculation, which suggested a dynamic response against the pathogen, with a more intense pattern at 48 h after inoculation. The results also highlighted a set of biological mechanisms triggered after inoculation that provide valuable information regarding the response of Yunnan pine to PWN infection. When compared with maritime pine (Pinus pinaster), the Yunnan pine response was less complex and involved a smaller number of differentially expressed genes, which may be associated with the increased degree of resistance to PWN displayed by Yunnan pine. These results revealed different strategies to cope with PWN infection by these two pine species, which display contrasting degrees of susceptibility, especially in the timely perception of the infection and response magnitude.
Collapse
|
41
|
Yu X, Liu X. Mapping RNA-seq reads to transcriptomes efficiently based on learning to hash method. Comput Biol Med 2020; 116:103539. [DOI: 10.1016/j.compbiomed.2019.103539] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 11/10/2019] [Accepted: 11/10/2019] [Indexed: 11/25/2022]
|
42
|
De Vitis C, Corleone G, Salvati V, Ascenzi F, Pallocca M, De Nicola F, Fanciulli M, di Martino S, Bruschini S, Napoli C, Ricci A, Bassi M, Venuta F, Rendina EA, Ciliberto G, Mancini R. B4GALT1 Is a New Candidate to Maintain the Stemness of Lung Cancer Stem Cells. J Clin Med 2019; 8:E1928. [PMID: 31717588 PMCID: PMC6912435 DOI: 10.3390/jcm8111928] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/30/2019] [Accepted: 11/05/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND According to the cancer stem cells (CSCs) hypothesis, a population of cancer cells with stem cell properties is responsible for tumor propagation, drug resistance, and disease recurrence. Study of the mechanisms responsible for lung CSCs propagation is expected to provide better understanding of cancer biology and new opportunities for therapy. METHODS The Lung Adenocarcinoma (LUAD) NCI-H460 cell line was grown either as 2D or as 3D cultures. Transcriptomic and genome-wide chromatin accessibility studies of 2D vs. 3D cultures were carried out using RNA-sequencing and Assay for Transposase Accessible Chromatin with high-throughput sequencing (ATAC-seq), respectively. Reverse transcription polymerase chain reaction (RT-PCR) was also carried out on RNA extracted from primary cultures derived from malignant pleural effusions to validate RNA-seq results. RESULTS RNA-seq and ATAC-seq data disentangled transcriptional and genome accessibility variability of 3D vs. 2D cultures in NCI-H460 cells. The examination of genomic landscape of genes upregulated in 3D vs. 2D cultures led to the identification of 2D cultures led to the identification of Beta-1,4-galactosyltranferase 1 (B4GALT1) as the top candidate. B4GALT1 as the top candidate. B4GALT1 was validated as a stemness factor, since its silencing caused strong inhibition of 3D spheroid formation. CONCLUSION Combined transcriptomic and chromatin accessibility study of 3D vs. 2D LUAD cultures led to the identification of B4GALT1 as a new factor involved in the propagation and maintenance of LUAD CSCs.
Collapse
Affiliation(s)
- Claudia De Vitis
- Department of Clinical and Molecular Medicine, Sant’Andrea Hospital, “Sapienza” University of Rome, 00161 Rome, Italy; (C.D.V.); (R.M.)
| | - Giacomo Corleone
- SAFU Laboratory, Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS “Regina Elena” National Cancer Institute, 00144 Rome, Italy; (G.C.); (M.P.); (F.D.N.); (M.F.)
| | - Valentina Salvati
- Preclinical Models and New Therapeutic Agents Unit, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | - Francesca Ascenzi
- Tumor Immunology and Immunotherapy Unit, Department of Research, Advanced Diagnostic and Technological Innovation, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | - Matteo Pallocca
- SAFU Laboratory, Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS “Regina Elena” National Cancer Institute, 00144 Rome, Italy; (G.C.); (M.P.); (F.D.N.); (M.F.)
| | - Francesca De Nicola
- SAFU Laboratory, Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS “Regina Elena” National Cancer Institute, 00144 Rome, Italy; (G.C.); (M.P.); (F.D.N.); (M.F.)
| | - Maurizio Fanciulli
- SAFU Laboratory, Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS “Regina Elena” National Cancer Institute, 00144 Rome, Italy; (G.C.); (M.P.); (F.D.N.); (M.F.)
| | - Simona di Martino
- Pathology Unit, IRCSS “Regina Elena” National Cancer Institute, 00144 Rome, Italy;
| | - Sara Bruschini
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy;
| | - Christian Napoli
- Department of Medical Surgical Sciences and Translational Medicine, Sant’Andrea Hospital, “Sapienza” University of Rome, 00189 Rome, Italy;
| | - Alberto Ricci
- Department of Clinical and Molecular Medicine, Division of Pneumology, Sapienza University of Rome, Sant’Andrea Hospital, 00189 Rome, Italy;
| | - Massimiliano Bassi
- Department of Thoracic Surgery, University of Rome Sapienza, 00161 Rome, Italy; (M.B.); (F.V.)
| | - Federico Venuta
- Department of Thoracic Surgery, University of Rome Sapienza, 00161 Rome, Italy; (M.B.); (F.V.)
| | - Erino Angelo Rendina
- Department of Thoracic Surgery, Sant’Andrea Hospital, “Sapienza” University of Rome, 00189 Rome, Italy
| | - Gennaro Ciliberto
- Scientific Direction, IRCCS “Regina Elena” National Cancer Institute, 00144 Rome, Italy
| | - Rita Mancini
- Department of Clinical and Molecular Medicine, Sant’Andrea Hospital, “Sapienza” University of Rome, 00161 Rome, Italy; (C.D.V.); (R.M.)
| |
Collapse
|
43
|
Sun C, Medvedev P. Toward fast and accurate SNP genotyping from whole genome sequencing data for bedside diagnostics. Bioinformatics 2019; 35:415-420. [PMID: 30032192 DOI: 10.1093/bioinformatics/bty641] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Accepted: 07/18/2018] [Indexed: 12/13/2022] Open
Abstract
Motivation Genotyping a set of variants from a database is an important step for identifying known genetic traits and disease-related variants within an individual. The growing size of variant databases as well as the high depth of sequencing data poses an efficiency challenge. In clinical applications, where time is crucial, alignment-based methods are often not fast enough. To fill the gap, Shajii et al. propose LAVA, an alignment-free genotyping method which is able to more quickly genotype single nucleotide polymorphisms (SNPs); however, there remains large room for improvements in running time and accuracy. Results We present the VarGeno method for SNP genotyping from Illumina whole genome sequencing data. VarGeno builds upon LAVA by improving the speed of k-mer querying as well as the accuracy of the genotyping strategy. We evaluate VarGeno on several read datasets using different genotyping SNP lists. VarGeno performs 7-13 times faster than LAVA with similar memory usage, while improving accuracy. Availability and implementation VarGeno is freely available at: https://github.com/medvedevgroup/vargeno. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Chen Sun
- Department of Computer Science and Engineering, Pennsylvania State University, USA
| | - Paul Medvedev
- Department of Computer Science and Engineering, Pennsylvania State University, USA.,Department of Biochemistry and Molecular Biology, Pennsylvania State University, USA.,Center for Computational Biology and Bioinformatics, Pennsylvania State University, USA
| |
Collapse
|
44
|
Langmead B, Wilks C, Antonescu V, Charles R. Scaling read aligners to hundreds of threads on general-purpose processors. Bioinformatics 2019; 35:421-432. [PMID: 30020410 PMCID: PMC6361242 DOI: 10.1093/bioinformatics/bty648] [Citation(s) in RCA: 463] [Impact Index Per Article: 77.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 07/17/2018] [Indexed: 12/27/2022] Open
Abstract
Motivation General-purpose processors can now contain many dozens of processor cores and support hundreds of simultaneous threads of execution. To make best use of these threads, genomics software must contend with new and subtle computer architecture issues. We discuss some of these and propose methods for improving thread scaling in tools that analyze each read independently, such as read aligners. Results We implement these methods in new versions of Bowtie, Bowtie 2 and HISAT. We greatly improve thread scaling in many scenarios, including on the recent Intel Xeon Phi architecture. We also highlight how bottlenecks are exacerbated by variable-record-length file formats like FASTQ and suggest changes that enable superior scaling. Availability and implementation Experiments for this study: https://github.com/BenLangmead/bowtie-scaling. Bowtie http://bowtie-bio.sourceforge.net. Bowtie 2 http://bowtie-bio.sourceforge.net/bowtie2. HISAT http://www.ccb.jhu.edu/software/hisat Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Ben Langmead
- Department of Computer Science, Johns Hopkins University, Baltimore, MD, USA.,Center for Computational Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Christopher Wilks
- Department of Computer Science, Johns Hopkins University, Baltimore, MD, USA.,Center for Computational Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Valentin Antonescu
- Department of Computer Science, Johns Hopkins University, Baltimore, MD, USA
| | - Rone Charles
- Department of Computer Science, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
45
|
Durai DA, Schulz MH. In silico read normalization using set multi-cover optimization. Bioinformatics 2019; 34:3273-3280. [PMID: 29912280 PMCID: PMC6157080 DOI: 10.1093/bioinformatics/bty307] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 04/18/2018] [Indexed: 11/24/2022] Open
Abstract
Motivation De Bruijn graphs are a common assembly data structure for sequencing datasets. But with the advances in sequencing technologies, assembling high coverage datasets has become a computational challenge. Read normalization, which removes redundancy in datasets, is widely applied to reduce resource requirements. Current normalization algorithms, though efficient, provide no guarantee to preserve important k-mers that form connections between regions in the graph. Results Here, normalization is phrased as a set multi-cover problem on reads and a heuristic algorithm, Optimized Read Normalization Algorithm (ORNA), is proposed. ORNA normalizes to the minimum number of reads required to retain all k-mers and their relative k-mer abundances from the original dataset. Hence, all connections from the original graph are preserved. ORNA was tested on various RNA-seq datasets with different coverage values. It was compared to the current normalization algorithms and was found to be performing better. Normalizing error corrected data allows for more accurate assemblies compared to the normalized uncorrected dataset. Further, an application is proposed in which multiple datasets are combined and normalized to predict novel transcripts that would have been missed otherwise. Finally, ORNA is a general purpose normalization algorithm that is fast and significantly reduces datasets with loss of assembly quality in between [1, 30]% depending on reduction stringency. Availability and implementation ORNA is available at https://github.com/SchulzLab/ORNA. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Dilip A Durai
- Cluster of Excellence on Multimodal Computing and Interaction, Saarland University, Saarbrücken, Germany.,Department of Computational Biology and Applied Algorithmics, Max Planck Institute for Informatics, Saarbrücken, Germany.,Saarbrücken Graduate School of Computer Science, Saarland University, Saarbrücken, Germany
| | - Marcel H Schulz
- Cluster of Excellence on Multimodal Computing and Interaction, Saarland University, Saarbrücken, Germany.,Department of Computational Biology and Applied Algorithmics, Max Planck Institute for Informatics, Saarbrücken, Germany
| |
Collapse
|
46
|
Madroñero J, Corredor Rozo ZL, Escobar Pérez JA, Velandia Romero ML. Next generation sequencing and proteomics in plant virology: how is Colombia doing? ACTA BIOLÓGICA COLOMBIANA 2019. [DOI: 10.15446/abc.v24n3.79486] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Crop production and trade are two of the most economically important activities in Colombia, and viral diseases cause a high negative impact to agricultural sector. Therefore, the detection, diagnosis, control, and management of viral diseases are crucial. Currently, Next-Generation Sequencing (NGS) and ‘Omic’ technologies constitute a right-hand tool for the discovery of novel viruses and for studying virus-plant interactions. This knowledge allows the development of new viral diagnostic methods and the discovery of key components of infectious processes, which could be used to generate plants resistant to viral infections. Globally, crop sciences are advancing in this direction. In this review, advancements in ‘omic’ technologies and their different applications in plant virology in Colombia are discussed. In addition, bioinformatics pipelines and resources for omics data analyses are presented. Due to their decreasing prices, NGS technologies are becoming an affordable and promising means to explore many phytopathologies affecting a wide variety of Colombian crops so as to improve their trade potential.
Collapse
|
47
|
Gunady MK, Mount SM, Corrada Bravo H. Yanagi: Fast and interpretable segment-based alternative splicing and gene expression analysis. BMC Bioinformatics 2019; 20:421. [PMID: 31409274 PMCID: PMC6693274 DOI: 10.1186/s12859-019-2947-6] [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] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 06/12/2019] [Indexed: 12/13/2022] Open
Abstract
Background Ultra-fast pseudo-alignment approaches are the tool of choice in transcript-level RNA sequencing (RNA-seq) analyses. Unfortunately, these methods couple the tasks of pseudo-alignment and transcript quantification. This coupling precludes the direct usage of pseudo-alignment to other expression analyses, including alternative splicing or differential gene expression analysis, without including a non-essential transcript quantification step. Results In this paper, we introduce a transcriptome segmentation approach to decouple these two tasks. We propose an efficient algorithm to generate maximal disjoint segments given a transcriptome reference library on which ultra-fast pseudo-alignment can be used to produce per-sample segment counts. We show how to apply these maximally unambiguous count statistics in two specific expression analyses – alternative splicing and gene differential expression – without the need of a transcript quantification step. Our experiments based on simulated and experimental data showed that the use of segment counts, like other methods that rely on local coverage statistics, provides an advantage over approaches that rely on transcript quantification in detecting and correctly estimating local splicing in the case of incomplete transcript annotations. Conclusions The transcriptome segmentation approach implemented in Yanagi exploits the computational and space efficiency of pseudo-alignment approaches. It significantly expands their applicability and interpretability in a variety of RNA-seq analyses by providing the means to model and capture local coverage variation in these analyses. Electronic supplementary material The online version of this article (10.1186/s12859-019-2947-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mohamed K Gunady
- Department of Computer Science, University of Maryland, College Park, Maryland, USA.,Center for Bioinformatics and Computational Biology, University of Maryland, College Park, Maryland, USA
| | - Stephen M Mount
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA
| | - Héctor Corrada Bravo
- Department of Computer Science, University of Maryland, College Park, Maryland, USA. .,Center for Bioinformatics and Computational Biology, University of Maryland, College Park, Maryland, USA.
| |
Collapse
|
48
|
Deng W, Mou T, Kalari KR, Niu N, Wang L, Pawitan Y, Vu TN. Alternating EM algorithm for a bilinear model in isoform quantification from RNA-seq data. Bioinformatics 2019; 36:805-812. [PMID: 31400221 PMCID: PMC9883676 DOI: 10.1093/bioinformatics/btz640] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 06/13/2019] [Accepted: 08/09/2019] [Indexed: 02/02/2023] Open
Abstract
MOTIVATION Estimation of isoform-level gene expression from RNA-seq data depends on simplifying assumptions, such as uniform read distribution, that are easily violated in real data. Such violations typically lead to biased estimates. Most existing methods provide bias correction step(s), which is based on biological considerations-such as GC content-and applied in single samples separately. The main problem is that not all biases are known. RESULTS We have developed a novel method called XAEM based on a more flexible and robust statistical model. Existing methods are essentially based on a linear model Xβ, where the design matrix X is known and is computed based on the simplifying assumptions. In contrast XAEM considers Xβ as a bilinear model with both X and β unknown. Joint estimation of X and β is made possible by a simultaneous analysis of multi-sample RNA-seq data. Compared to existing methods, XAEM automatically performs empirical correction of potentially unknown biases. We use an alternating expectation-maximization (AEM) algorithm, alternating between estimation of X and β. For speed XAEM utilizes quasi-mapping for read alignment, thus leading to a fast algorithm. Overall XAEM performs favorably compared to recent advanced methods. For simulated datasets, XAEM obtains higher accuracy for multiple-isoform genes. In a differential-expression analysis of a real single-cell RNA-seq dataset, XAEM achieves substantially better rediscovery rates in independent validation sets. AVAILABILITY AND IMPLEMENTATION The method and pipeline are implemented as a tool and freely available for use at http://fafner.meb.ki.se/biostatwiki/xaem/. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Wenjiang Deng
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm 17177, Sweden
| | - Tian Mou
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm 17177, Sweden
| | | | - Nifang Niu
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | | | | |
Collapse
|
49
|
Mortzfeld BM, Taubenheim J, Klimovich AV, Fraune S, Rosenstiel P, Bosch TCG. Temperature and insulin signaling regulate body size in Hydra by the Wnt and TGF-beta pathways. Nat Commun 2019; 10:3257. [PMID: 31332174 PMCID: PMC6646324 DOI: 10.1038/s41467-019-11136-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 06/07/2019] [Indexed: 02/03/2023] Open
Abstract
How multicellular organisms assess and control their size is a fundamental question in biology, yet the molecular and genetic mechanisms that control organ or organism size remain largely unsolved. The freshwater polyp Hydra demonstrates a high capacity to adapt its body size to different temperatures. Here we identify the molecular mechanisms controlling this phenotypic plasticity and show that temperature-induced cell number changes are controlled by Wnt- and TGF-β signaling. Further we show that insulin-like peptide receptor (INSR) and forkhead box protein O (FoxO) are important genetic drivers of size determination controlling the same developmental regulators. Thus, environmental and genetic factors directly affect developmental mechanisms in which cell number is the strongest determinant of body size. These findings identify the basic mechanisms as to how size is regulated on an organismic level and how phenotypic plasticity is integrated into conserved developmental pathways in an evolutionary informative model organism.
Collapse
Affiliation(s)
- Benedikt M Mortzfeld
- Zoological Institute, Christian-Albrechts University Kiel, Am Botanischen Garten 1-9, 24118, Kiel, Germany
- Department of Bioengineering, University of Massachusetts Dartmouth, 285 Old Westport Rd, Dartmouth, MA, 02747, USA
| | - Jan Taubenheim
- Zoological Institute, Christian-Albrechts University Kiel, Am Botanischen Garten 1-9, 24118, Kiel, Germany
- Institute for Zoology and Organismic Interactions, Heinrich-Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Alexander V Klimovich
- Zoological Institute, Christian-Albrechts University Kiel, Am Botanischen Garten 1-9, 24118, Kiel, Germany
| | - Sebastian Fraune
- Zoological Institute, Christian-Albrechts University Kiel, Am Botanischen Garten 1-9, 24118, Kiel, Germany
- Institute for Zoology and Organismic Interactions, Heinrich-Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, Christian-Albrechts University Kiel, University Hospital Schleswig-Holstein, Rosalind-Franklin-Straße 12, 24105, Kiel, Germany
| | - Thomas C G Bosch
- Zoological Institute, Christian-Albrechts University Kiel, Am Botanischen Garten 1-9, 24118, Kiel, Germany.
| |
Collapse
|
50
|
Hafner M, Mills CE, Subramanian K, Chen C, Chung M, Boswell SA, Everley RA, Liu C, Walmsley CS, Juric D, Sorger PK. Multiomics Profiling Establishes the Polypharmacology of FDA-Approved CDK4/6 Inhibitors and the Potential for Differential Clinical Activity. Cell Chem Biol 2019; 26:1067-1080.e8. [PMID: 31178407 DOI: 10.1016/j.chembiol.2019.05.005] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 03/14/2019] [Accepted: 05/13/2019] [Indexed: 11/28/2022]
Abstract
The target profiles of many drugs are established early in their development and are not systematically revisited at the time of FDA approval. Thus, it is often unclear whether therapeutics with the same nominal targets but different chemical structures are functionally equivalent. In this paper we use five different phenotypic and biochemical assays to compare approved inhibitors of cyclin-dependent kinases 4/6-collectively regarded as breakthroughs in the treatment of hormone receptor-positive breast cancer. We find that transcriptional, proteomic, and phenotypic changes induced by palbociclib, ribociclib, and abemaciclib differ significantly; abemaciclib in particular has advantageous activities partially overlapping those of alvocidib, an older polyselective CDK inhibitor. In cells and mice, abemaciclib inhibits kinases other than CDK4/6 including CDK2/cyclin A/E-implicated in resistance to CDK4/6 inhibition-and CDK1/cyclin B. The multifaceted experimental and computational approaches described here therefore uncover underappreciated differences in CDK4/6 inhibitor activities with potential importance in treating human patients.
Collapse
Affiliation(s)
- Marc Hafner
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Caitlin E Mills
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Kartik Subramanian
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Chen Chen
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Mirra Chung
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Sarah A Boswell
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Robert A Everley
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Changchang Liu
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Charlotte S Walmsley
- Termeer Center for Targeted Therapies, Massachusetts General Hospital Cancer Center, Boston, MA 02114, USA
| | - Dejan Juric
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA; Termeer Center for Targeted Therapies, Massachusetts General Hospital Cancer Center, Boston, MA 02114, USA.
| | - Peter K Sorger
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|