1
|
Danaher P, McGuire D, Wu L, Patrick M, Kroeppler D, Zhai H, Olgun DG, Gong D, Cao J, Hwang WL, Schmid J, Beechem JM. InSituCor: exploring spatially correlated genes conditional on the cell type landscape. Genome Biol 2025; 26:105. [PMID: 40275395 PMCID: PMC12020328 DOI: 10.1186/s13059-025-03554-1] [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: 09/25/2023] [Accepted: 03/21/2025] [Indexed: 04/26/2025] Open
Abstract
In spatial transcriptomics data, spatially correlated genes promise to reveal high-interest phenomena like cell-cell interactions and latent variables. But in practice, most spatial correlations arise from the spatial arrangement of cell types, obscuring the more interesting relationships we hope to discover. We introduce InSituCor, a toolkit for discovering modules of spatially correlated genes. InSituCor returns only correlations not explainable by already-known factors like the cell type landscape; this spares precious analyst effort. InSituCor supports both unbiased discovery of whole-dataset correlations and knowledge-driven exploration of genes of interest. As a special case, it evaluates ligand-receptor pairs for spatial co-regulation.
Collapse
Affiliation(s)
| | | | - Lidan Wu
- Bruker Spatial Biology, Seattle, WA, USA
| | | | | | | | - Deniz G Olgun
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiation Oncology, Massachusetts General Hospital, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Dennis Gong
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiation Oncology, Massachusetts General Hospital, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard-MIT Health Sciences and Technology Program, Cambridge, MA, USA
| | - Jingyi Cao
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women'S Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - William L Hwang
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiation Oncology, Massachusetts General Hospital, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | |
Collapse
|
2
|
Selvam PK, Elavarasu SM, Dhanushkumar T, Vasudevan K, George Priya Doss C. Exploring the role of estrogen and progestins in breast cancer: A genomic approach to diagnosis. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 142:25-43. [PMID: 39059987 DOI: 10.1016/bs.apcsb.2023.12.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
Breast cancer (BC) is the most common cancer among women and a major cause of death from cancer. The role of estrogen and progestins, including synthetic hormones like R5020, in the development of BC has been highlighted in numerous studies. In our study, we employed machine learning and advanced bioinformatics to identify genes that could serve as diagnostic markers for BC. We thoroughly analyzed the transcriptomic data of two BC cell lines, T47D and UDC4, and performed differential gene expression analysis. We also conducted functional enrichment analysis to understand the biological functions influenced by these genes. Our study identified several diagnostic genes strongly associated with BC, including MIR6728, ENO1-IT1, ENO1-AS1, RNU6-304P, HMGN2P17, RP3-477M7.5, RP3-477M7.6, and CA6. The genes MIR6728, ENO1-IT1, ENO1-AS1, and HMGN2P17 are involved in cancer control, glycolysis, and DNA-related processes, while CA6 is associated with apoptosis and cancer development. These genes could potentially serve as predictors for BC, paving the way for more precise diagnostic methods and personalized treatment plans. This research enhances our understanding of BC and offers promising avenues for improving patient care in the future.
Collapse
Affiliation(s)
- Prasanna Kumar Selvam
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru, India; Institute of Bioinformatics, International Technology Park, Bangalore, India
| | | | - T Dhanushkumar
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru, India
| | - Karthick Vasudevan
- Institute of Bioinformatics, International Technology Park, Bangalore, India; Manipal Academy of Higher Education (MAHE), Manipal, India
| | - C George Priya Doss
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of BioSciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India.
| |
Collapse
|
3
|
Yao J, Dai S, Zhu R, Tan J, Zhao Q, Yin Y, Sun J, Du X, Ge L, Xu J, Hou C, Li N, Li J, Ji W, Zhu C, Zhang R, Li T. Deciphering molecular heterogeneity and dynamics of human hippocampal neural stem cells at different ages and injury states. eLife 2024; 12:RP89507. [PMID: 38607670 PMCID: PMC11014727 DOI: 10.7554/elife.89507] [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] [Indexed: 04/13/2024] Open
Abstract
While accumulated publications support the existence of neurogenesis in the adult human hippocampus, the homeostasis and developmental potentials of neural stem cells (NSCs) under different contexts remain unclear. Based on our generated single-nucleus atlas of the human hippocampus across neonatal, adult, aging, and injury, we dissected the molecular heterogeneity and transcriptional dynamics of human hippocampal NSCs under different contexts. We further identified new specific neurogenic lineage markers that overcome the lack of specificity found in some well-known markers. Based on developmental trajectory and molecular signatures, we found that a subset of NSCs exhibit quiescent properties after birth, and most NSCs become deep quiescence during aging. Furthermore, certain deep quiescent NSCs are reactivated following stroke injury. Together, our findings provide valuable insights into the development, aging, and reactivation of the human hippocampal NSCs, and help to explain why adult hippocampal neurogenesis is infrequently observed in humans.
Collapse
Affiliation(s)
- Junjun Yao
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and TechnologyKunmingChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| | - Shaoxing Dai
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and TechnologyKunmingChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| | - Ran Zhu
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and TechnologyKunmingChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| | - Ju Tan
- Department of Anatomy, National and Regional Engineering Laboratory of Tissue Engineering, State Key Laboratory of Trauma, Burn and Combined Injury, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Third Military Medical UniversityChongqingChina
| | - Qiancheng Zhao
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and TechnologyKunmingChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| | - Yu Yin
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and TechnologyKunmingChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| | - Jiansen Sun
- Zhong-Zhi- Yi-Gu Research InstituteChongqingChina
| | - Xuewei Du
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and TechnologyKunmingChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| | - Longjiao Ge
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and TechnologyKunmingChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| | - Jianhua Xu
- Department of Anatomy, National and Regional Engineering Laboratory of Tissue Engineering, State Key Laboratory of Trauma, Burn and Combined Injury, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Third Military Medical UniversityChongqingChina
| | - Chunli Hou
- Department of Anatomy, National and Regional Engineering Laboratory of Tissue Engineering, State Key Laboratory of Trauma, Burn and Combined Injury, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Third Military Medical UniversityChongqingChina
| | - Nan Li
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and TechnologyKunmingChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| | - Jun Li
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| | - Weizhi Ji
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and TechnologyKunmingChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| | - Chuhong Zhu
- Department of Anatomy, National and Regional Engineering Laboratory of Tissue Engineering, State Key Laboratory of Trauma, Burn and Combined Injury, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Third Military Medical UniversityChongqingChina
| | - Runrui Zhang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and TechnologyKunmingChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| | - Tianqing Li
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and TechnologyKunmingChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| |
Collapse
|
4
|
Sun H, Niu Q, Yang J, Zhao Y, Tian Z, Fan J, Zhang Z, Wang Y, Geng S, Zhang Y, Guan G, Williams DT, Luo J, Yin H, Liu Z. Transcriptome Profiling Reveals Features of Immune Response and Metabolism of Acutely Infected, Dead and Asymptomatic Infection of African Swine Fever Virus in Pigs. Front Immunol 2022; 12:808545. [PMID: 34975923 PMCID: PMC8714921 DOI: 10.3389/fimmu.2021.808545] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 11/29/2021] [Indexed: 12/16/2022] Open
Abstract
African swine fever virus (ASFV) infection can result in lethal disease in pigs. ASFV encodes 150-167 proteins, of which only approximately 50 encoded viral structure proteins are functionally known. ASFV also encodes some nonstructural proteins that are involved in the regulation of viral transcription, viral replication and evasion from host defense. However, the understanding of the molecular correlates of the severity of these infections is still limited. The purpose of this study was to compare host and viral gene expression differences and perform functional analysis in acutely infected, dead and cohabiting asymptomatic pigs infected with ASFV by using RNA-Seq technique; healthy pigs were used as controls. A total of 3,760 and 2,874 upregulated genes and 4,176 and 2,899 downregulated genes were found in healthy pigs vs. acutely infected, dead pigs or asymptomatic pigs, respectively. Additionally, 941 upregulated genes and 956 downregulated genes were identified in asymptomatic vs. acutely infected, dead pigs. Different alternative splicing (AS) events were also analyzed, as were gene chromosome locations, and protein-protein interaction (PPI) network prediction analysis was performed for significantly differentially expressed genes (DEGs). In addition, 30 DEGs were validated by RT-qPCR, and the results were consistent with the RNA-Seq results. We further analyzed the interaction between ASFV and its host at the molecular level and predicted the mechanisms responsible for asymptomatic pigs based on the selected DEGs. Interestingly, we found that some viral genes in cohabiting asymptomatic pigs might integrate into host genes (DP96R, I73R and L83L) or remain in the tissues of cohabiting asymptomatic pigs. In conclusion, the data obtained in the present study provide new evidence for further elucidating ASFV-host interactions and the ASFV infection mechanism and will facilitate the implementation of integrated strategies for controlling ASF spread.
Collapse
Affiliation(s)
- Hualin Sun
- African Swine Fever Regional Laboratory, China (Lanzhou) and State Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Qingli Niu
- African Swine Fever Regional Laboratory, China (Lanzhou) and State Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Jifei Yang
- African Swine Fever Regional Laboratory, China (Lanzhou) and State Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yaru Zhao
- African Swine Fever Regional Laboratory, China (Lanzhou) and State Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Zhancheng Tian
- African Swine Fever Regional Laboratory, China (Lanzhou) and State Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Jie Fan
- African Swine Fever Regional Laboratory, China (Lanzhou) and State Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Zhonghui Zhang
- African Swine Fever Regional Laboratory, China (Lanzhou) and State Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yiwang Wang
- African Swine Fever Regional Laboratory, China (Lanzhou) and State Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Shuxian Geng
- African Swine Fever Regional Laboratory, China (Lanzhou) and State Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yulong Zhang
- African Swine Fever Regional Laboratory, China (Lanzhou) and State Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Guiquan Guan
- African Swine Fever Regional Laboratory, China (Lanzhou) and State Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - David T Williams
- Commonwealth Scientific and Industrial Research Organization (CSIRO) Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Jianxun Luo
- African Swine Fever Regional Laboratory, China (Lanzhou) and State Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Hong Yin
- African Swine Fever Regional Laboratory, China (Lanzhou) and State Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Zhijie Liu
- African Swine Fever Regional Laboratory, China (Lanzhou) and State Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| |
Collapse
|
5
|
You Z, Li Y, Wang Q, Zhao Z, Li Y, Qian Q, Li B, Zhang J, Huang B, Liang J, Chen R, Lyu Z, Chen Y, Lian M, Xiao X, Miao Q, Fang J, Lian Z, Eric Gershwin M, Tang R, Ma X. The Clinical Significance of Hepatic CD69 + CD103 + CD8 + Resident-Memory T Cells in Autoimmune Hepatitis. Hepatology 2021; 74:847-863. [PMID: 33554350 DOI: 10.1002/hep.31739] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 12/01/2020] [Accepted: 01/05/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS The diverse inflammatory response found in the liver of patients with autoimmune hepatitis (AIH) is well established, but identification of potentially pathogenic subpopulations has proven enigmatic. APPROACH AND RESULTS We report herein that CD69+ CD103+ CD8+ tissue-resident memory T cells (TRM ) are significantly increased in the liver of patients with AIH compared to chronic hepatitis B, NAFLD, and healthy control tissues. In addition, there was a significant statistical correlation between elevation of CD8+ TRM cells and AIH disease severity. Indeed, in patients with successful responses to immunosuppression, the frequencies of such hepatic CD8+ TRM cells decreased significantly. CD69+ CD8+ and CD69+ CD103+ CD8+ T cells, also known as CD8+ TRM cells, reflect tissue residency and are well known to provide intense immune antigenic responses. Hence, it was particularly interesting that patients with AIH also manifest an elevated expression of IL-15 and TGF-β on inflammatory cells, and extensive hepatic expression of E-cadherin; these factors likely contribute to the development and localization of CD8+ TRM cells. Based on these data and, in particular, the relationships between disease severity and CD8+ TRM cells, we studied the mechanisms involved with glucocorticoid (GC) modulation of CD8+ TRM cell expansion. Our data reflect that GCs in vitro inhibit the expansion of CD8+ TRM cells induced by IL-15 and TGF-β and with direct down-regulation of the nuclear factor Blimp1 of CD8+ TRM cells. CONCLUSIONS Our data suggest that CD8+ TRM cells play a critical role in the pathogenesis of AIH, and GCs attenuate hepatic inflammation through direct inhibition of CD8+ TRM cell expansion.
Collapse
Affiliation(s)
- Zhengrui You
- Division of Gastroenterology and HepatologyKey Laboratory of Gastroenterology and HepatologyMinistry of HealthState Key Laboratory for Oncogenes and Related GenesRenji HospitalShanghai Institute of Digestive DiseaseSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - You Li
- Division of Gastroenterology and HepatologyKey Laboratory of Gastroenterology and HepatologyMinistry of HealthState Key Laboratory for Oncogenes and Related GenesRenji HospitalShanghai Institute of Digestive DiseaseSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Qixia Wang
- Division of Gastroenterology and HepatologyKey Laboratory of Gastroenterology and HepatologyMinistry of HealthState Key Laboratory for Oncogenes and Related GenesRenji HospitalShanghai Institute of Digestive DiseaseSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Zhibin Zhao
- Chronic Disease LaboratoryInstitutes for Life Sciences and School of MedicineSouth China University of TechnologyGuangzhouChina
| | - Yikang Li
- Division of Gastroenterology and HepatologyKey Laboratory of Gastroenterology and HepatologyMinistry of HealthState Key Laboratory for Oncogenes and Related GenesRenji HospitalShanghai Institute of Digestive DiseaseSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Qiwei Qian
- Division of Gastroenterology and HepatologyKey Laboratory of Gastroenterology and HepatologyMinistry of HealthState Key Laboratory for Oncogenes and Related GenesRenji HospitalShanghai Institute of Digestive DiseaseSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Bo Li
- Division of Gastroenterology and HepatologyKey Laboratory of Gastroenterology and HepatologyMinistry of HealthState Key Laboratory for Oncogenes and Related GenesRenji HospitalShanghai Institute of Digestive DiseaseSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Jun Zhang
- Division of Gastroenterology and HepatologyKey Laboratory of Gastroenterology and HepatologyMinistry of HealthState Key Laboratory for Oncogenes and Related GenesRenji HospitalShanghai Institute of Digestive DiseaseSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Bingyuan Huang
- Division of Gastroenterology and HepatologyKey Laboratory of Gastroenterology and HepatologyMinistry of HealthState Key Laboratory for Oncogenes and Related GenesRenji HospitalShanghai Institute of Digestive DiseaseSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Jubo Liang
- Division of Gastroenterology and HepatologyKey Laboratory of Gastroenterology and HepatologyMinistry of HealthState Key Laboratory for Oncogenes and Related GenesRenji HospitalShanghai Institute of Digestive DiseaseSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Ruiling Chen
- Division of Gastroenterology and HepatologyKey Laboratory of Gastroenterology and HepatologyMinistry of HealthState Key Laboratory for Oncogenes and Related GenesRenji HospitalShanghai Institute of Digestive DiseaseSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Zhuwan Lyu
- Division of Gastroenterology and HepatologyKey Laboratory of Gastroenterology and HepatologyMinistry of HealthState Key Laboratory for Oncogenes and Related GenesRenji HospitalShanghai Institute of Digestive DiseaseSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Yong Chen
- Division of Gastroenterology and HepatologyKey Laboratory of Gastroenterology and HepatologyMinistry of HealthState Key Laboratory for Oncogenes and Related GenesRenji HospitalShanghai Institute of Digestive DiseaseSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Min Lian
- Division of Gastroenterology and HepatologyKey Laboratory of Gastroenterology and HepatologyMinistry of HealthState Key Laboratory for Oncogenes and Related GenesRenji HospitalShanghai Institute of Digestive DiseaseSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Xiao Xiao
- Division of Gastroenterology and HepatologyKey Laboratory of Gastroenterology and HepatologyMinistry of HealthState Key Laboratory for Oncogenes and Related GenesRenji HospitalShanghai Institute of Digestive DiseaseSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Qi Miao
- Division of Gastroenterology and HepatologyKey Laboratory of Gastroenterology and HepatologyMinistry of HealthState Key Laboratory for Oncogenes and Related GenesRenji HospitalShanghai Institute of Digestive DiseaseSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Jingyuan Fang
- Division of Gastroenterology and HepatologyKey Laboratory of Gastroenterology and HepatologyMinistry of HealthState Key Laboratory for Oncogenes and Related GenesRenji HospitalShanghai Institute of Digestive DiseaseSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Zhexiong Lian
- Chronic Disease LaboratoryInstitutes for Life Sciences and School of MedicineSouth China University of TechnologyGuangzhouChina
| | - M Eric Gershwin
- Division of RheumatologyDepartment of Medicine, Allergy and Clinical ImmunologyUniversity of California at DavisDavisCA
| | - Ruqi Tang
- Division of Gastroenterology and HepatologyKey Laboratory of Gastroenterology and HepatologyMinistry of HealthState Key Laboratory for Oncogenes and Related GenesRenji HospitalShanghai Institute of Digestive DiseaseSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Xiong Ma
- Division of Gastroenterology and HepatologyKey Laboratory of Gastroenterology and HepatologyMinistry of HealthState Key Laboratory for Oncogenes and Related GenesRenji HospitalShanghai Institute of Digestive DiseaseSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
6
|
Jia S, Hu P. ChrNet: A re-trainable chromosome-based 1D convolutional neural network for predicting immune cell types. Genomics 2021; 113:2023-2031. [PMID: 33932523 DOI: 10.1016/j.ygeno.2021.04.037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 04/10/2021] [Accepted: 04/26/2021] [Indexed: 10/21/2022]
Abstract
Cells from our immune system detect and kill pathogens to protect our body against various diseases. However, current methods for determining cell types have some major limitations, such as being time-consuming and with low throughput, etc. Immune cells that are associated with cancer tissues play a critical role in revealing tumor development. Identifying the immune composition within tumor microenvironment in a timely manner will be helpful in improving clinical prognosis and therapeutic management for cancer. Although unsupervised clustering approaches have been prevailing to process scRNA-seq datasets, their results vary among studies with different input parameters and sizes, and the identification of the cell types of the clusters is still very challenging. Genes in human genome can be aligned to chromosomes with specific orders. Hence, we hypothesize incorporating this information into our learning model will potentially improve the cell type classification performance. In order to utilize gene positional information, we introduced ChrNet, a novel chromosome-specific re-trainable supervised learning method based on one-dimensional convolutional neural network (1D-CNN). By benchmarking with several models, our model shows superior performance in immune cell type profiling with larger than 90% accuracy. It is expected that this approach can become a reference architecture for other cell type classification methods. Our ChrNet tool is available online at: https://github.com/Krisloveless/ChrNet.
Collapse
Affiliation(s)
- Shuo Jia
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada
| | - Pingzhao Hu
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada; Department of Computer Science, University of Manitoba, Winnipeg, MB, Canada; Research Institute in Oncology and Hematology, CancerCare Manitoba, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
7
|
Watters JM, Wright G, Smith MA, Shah B, Wright KL. Histone deacetylase 8 inhibition suppresses mantle cell lymphoma viability while preserving natural killer cell function. Biochem Biophys Res Commun 2020; 534:773-779. [PMID: 33190829 DOI: 10.1016/j.bbrc.2020.11.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 11/02/2020] [Indexed: 12/11/2022]
Abstract
Mantle Cell Lymphoma (MCL) is a non-Hodgkin lymphoma with a median survival rate of five years. Standard treatment with high-dose chemotherapy plus rituximab (anti-CD20 antibody) has extended overall survival although, the disease remains incurable. Histone deacetylases (HDAC) are a family of enzymes that regulate multiple proteins and cellular pathways through post-translational modification. Broad spectrum HDAC inhibitors have shown some therapeutic promise, inducing cell cycle inhibition and apoptosis in leukemia and non-Hodgkin's lymphoma. However, the therapeutic effects of these broad-spectrum HDAC inhibitors can detrimentally dampen Natural Killer (NK) cell cytotoxicity, reduce NK viability, and downregulate activation receptors important for NK mediated anti-tumor responses. Impairment of NK function in MCL patients during therapy potentially limits therapeutic activity of rituximab. Thus, there is an unmet need to decipher specific roles of individual HDACs in order to preserve and/or enhance NK function, while, directly impairing MCL viability. We investigated the impact of HDAC8 in MCL cell lines. Inhibition or genetic loss of HDAC8 caused MCL cells to undergo apoptosis. In contrast, exposure of primary human NK cells to an HDAC8 inhibitor does not alter viability, receptor expression, or antibody dependent cellular cytotoxicity (ADCC). However, an increase in effector cytokine interferon-gamma (IFNγ) producing NK cells was observed in response to HDAC8 inhibition. Taken together these data suggest that selective HDAC8 inhibitors may simultaneously preserve NK functional activity, while impairing MCL tumor growth, establishing a rationale for future clinical evaluation.
Collapse
Affiliation(s)
- January M Watters
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA; Cancer Biology Ph.D. Program, University of South Florida, Tampa, USA
| | - Gabriela Wright
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Matthew A Smith
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Bijal Shah
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Kenneth L Wright
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| |
Collapse
|
8
|
Priebe V, Sartori G, Napoli S, Chung EYL, Cascione L, Kwee I, Arribas AJ, Mensah AA, Rinaldi A, Ponzoni M, Zucca E, Rossi D, Efremov D, Lenz G, Thome M, Bertoni F. Role of ETS1 in the Transcriptional Network of Diffuse Large B Cell Lymphoma of the Activated B Cell-Like Type. Cancers (Basel) 2020; 12:cancers12071912. [PMID: 32679859 PMCID: PMC7409072 DOI: 10.3390/cancers12071912] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/09/2020] [Accepted: 07/11/2020] [Indexed: 01/08/2023] Open
Abstract
Diffuse large B cell lymphoma (DLBCL) is a heterogenous disease that has been distinguished into at least two major molecular entities, the germinal center-like B cell (GCB) DLBCL and activated-like B cell (ABC) DLBCL, based on transcriptome expression profiling. A recurrent ch11q24.3 gain is observed in roughly a fourth of DLBCL cases resulting in the overexpression of two ETS transcription factor family members, ETS1 and FLI1. Here, we knocked down ETS1 expression by siRNA and analyzed expression changes integrating them with ChIP-seq data to identify genes directly regulated by ETS1. ETS1 silencing affected expression of genes involved in B cell signaling activation, B cell differentiation, cell cycle, and immune processes. Integration of RNA-Seq (RNA sequencing) data and ChIP-Seq (chromatin immunoprecipitation sequencing) identified 97 genes as bona fide, positively regulated direct targets of ETS1 in ABC-DLBCL. Among these was the Fc receptor for IgM, FCMR (also known as FAIM3 or Toso), which showed higher expression in ABC- than GCB-DLBCL clinical specimens. These findings show that ETS1 is contributing to the lymphomagenesis in a subset of DLBCL and identifies FCMR as a novel target of ETS1, predominantly expressed in ABC-DLBCL.
Collapse
Affiliation(s)
- Valdemar Priebe
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland; (V.P.); (G.S.); (S.N.); (E.Y.L.C.); (L.C.); (I.K.); (A.J.A.); (A.A.M.); (A.R.); (E.Z.); (D.R.)
| | - Giulio Sartori
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland; (V.P.); (G.S.); (S.N.); (E.Y.L.C.); (L.C.); (I.K.); (A.J.A.); (A.A.M.); (A.R.); (E.Z.); (D.R.)
| | - Sara Napoli
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland; (V.P.); (G.S.); (S.N.); (E.Y.L.C.); (L.C.); (I.K.); (A.J.A.); (A.A.M.); (A.R.); (E.Z.); (D.R.)
| | - Elaine Yee Lin Chung
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland; (V.P.); (G.S.); (S.N.); (E.Y.L.C.); (L.C.); (I.K.); (A.J.A.); (A.A.M.); (A.R.); (E.Z.); (D.R.)
| | - Luciano Cascione
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland; (V.P.); (G.S.); (S.N.); (E.Y.L.C.); (L.C.); (I.K.); (A.J.A.); (A.A.M.); (A.R.); (E.Z.); (D.R.)
- Swiss Institute of Bioinformatics (SIB), 1015 Lausanne, Switzerland
| | - Ivo Kwee
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland; (V.P.); (G.S.); (S.N.); (E.Y.L.C.); (L.C.); (I.K.); (A.J.A.); (A.A.M.); (A.R.); (E.Z.); (D.R.)
- Swiss Institute of Bioinformatics (SIB), 1015 Lausanne, Switzerland
- Dalle Molle Institute for Artificial Intelligence (IDSIA), 6928 Manno, Switzerland
| | - Alberto Jesus Arribas
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland; (V.P.); (G.S.); (S.N.); (E.Y.L.C.); (L.C.); (I.K.); (A.J.A.); (A.A.M.); (A.R.); (E.Z.); (D.R.)
- Swiss Institute of Bioinformatics (SIB), 1015 Lausanne, Switzerland
| | - Afua Adjeiwaa Mensah
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland; (V.P.); (G.S.); (S.N.); (E.Y.L.C.); (L.C.); (I.K.); (A.J.A.); (A.A.M.); (A.R.); (E.Z.); (D.R.)
| | - Andrea Rinaldi
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland; (V.P.); (G.S.); (S.N.); (E.Y.L.C.); (L.C.); (I.K.); (A.J.A.); (A.A.M.); (A.R.); (E.Z.); (D.R.)
| | - Maurilio Ponzoni
- San Raffaele Scientific Institute, Vita Salute University, 20132 Milan, Italy;
| | - Emanuele Zucca
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland; (V.P.); (G.S.); (S.N.); (E.Y.L.C.); (L.C.); (I.K.); (A.J.A.); (A.A.M.); (A.R.); (E.Z.); (D.R.)
- Oncology Institute of Southern Switzerland, 6500 Bellinzona, Switzerland
| | - Davide Rossi
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland; (V.P.); (G.S.); (S.N.); (E.Y.L.C.); (L.C.); (I.K.); (A.J.A.); (A.A.M.); (A.R.); (E.Z.); (D.R.)
- Oncology Institute of Southern Switzerland, 6500 Bellinzona, Switzerland
| | - Dimitar Efremov
- Molecular Hematology, International Centre for Genetic Engineering and Biotechnology, 34149 Trieste, Italy;
| | - Georg Lenz
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Münster, 48149 Münster, Germany;
| | - Margot Thome
- Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland;
| | - Francesco Bertoni
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland; (V.P.); (G.S.); (S.N.); (E.Y.L.C.); (L.C.); (I.K.); (A.J.A.); (A.A.M.); (A.R.); (E.Z.); (D.R.)
- Oncology Institute of Southern Switzerland, 6500 Bellinzona, Switzerland
- Correspondence: ; Tel.: +41-91-8200-367; Fax: +41-91-8200-397
| |
Collapse
|
9
|
Bibby JA, Purvis HA, Hayday T, Chandra A, Okkenhaug K, Rosenzweig S, Aksentijevich I, Wood M, Lachmann HJ, Kemper C, Cope AP, Perucha E. Cholesterol metabolism drives regulatory B cell IL-10 through provision of geranylgeranyl pyrophosphate. Nat Commun 2020; 11:3412. [PMID: 32641742 PMCID: PMC7343868 DOI: 10.1038/s41467-020-17179-4] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 06/04/2020] [Indexed: 02/07/2023] Open
Abstract
Regulatory B cells restrict immune and inflammatory responses across a number of contexts. This capacity is mediated primarily through the production of IL-10. Here we demonstrate that the induction of a regulatory program in human B cells is dependent on a metabolic priming event driven by cholesterol metabolism. Synthesis of the metabolic intermediate geranylgeranyl pyrophosphate (GGPP) is required to specifically drive IL-10 production, and to attenuate Th1 responses. Furthermore, GGPP-dependent protein modifications control signaling through PI3Kδ-AKT-GSK3, which in turn promote BLIMP1-dependent IL-10 production. Inherited gene mutations in cholesterol metabolism result in a severe autoinflammatory syndrome termed mevalonate kinase deficiency (MKD). Consistent with our findings, B cells from MKD patients induce poor IL-10 responses and are functionally impaired. Moreover, metabolic supplementation with GGPP is able to reverse this defect. Collectively, our data define cholesterol metabolism as an integral metabolic pathway for the optimal functioning of human IL-10 producing regulatory B cells.
Collapse
Affiliation(s)
- Jack A Bibby
- Centre for Inflammation Biology and Cancer Immunology, School of Immunology and Microbial Sciences, King's College London, London, SE1 1UL, UK. .,Complement and Inflammation Research Section (CIRS), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Harriet A Purvis
- Centre for Inflammation Biology and Cancer Immunology, School of Immunology and Microbial Sciences, King's College London, London, SE1 1UL, UK
| | - Thomas Hayday
- Centre for Inflammation Biology and Cancer Immunology, School of Immunology and Microbial Sciences, King's College London, London, SE1 1UL, UK
| | - Anita Chandra
- Division of Immunology, Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK
| | - Klaus Okkenhaug
- Division of Immunology, Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK
| | - Sofia Rosenzweig
- Inflammatory Disease Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ivona Aksentijevich
- Inflammatory Disease Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Michael Wood
- National Amyloidosis Centre, Division of Medicine, University College London and Royal Free Hospital London NHS Foundation Trust, London, NW3 2PF, UK
| | - Helen J Lachmann
- National Amyloidosis Centre, Division of Medicine, University College London and Royal Free Hospital London NHS Foundation Trust, London, NW3 2PF, UK
| | - Claudia Kemper
- Centre for Inflammation Biology and Cancer Immunology, School of Immunology and Microbial Sciences, King's College London, London, SE1 1UL, UK.,Complement and Inflammation Research Section (CIRS), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA.,Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Andrew P Cope
- Centre for Inflammation Biology and Cancer Immunology, School of Immunology and Microbial Sciences, King's College London, London, SE1 1UL, UK. .,Centre for Rheumatic Diseases, King's College London, London, SE1 1UL, UK.
| | - Esperanza Perucha
- Centre for Inflammation Biology and Cancer Immunology, School of Immunology and Microbial Sciences, King's College London, London, SE1 1UL, UK. .,Centre for Rheumatic Diseases, King's College London, London, SE1 1UL, UK.
| |
Collapse
|
10
|
Wang Z, Pascal LE, Chandran UR, Chaparala S, Lv S, Ding H, Qi L, Wang Z. ELL2 Is Required for the Growth and Survival of AR-Negative Prostate Cancer Cells. Cancer Manag Res 2020; 12:4411-4427. [PMID: 32606936 PMCID: PMC7294050 DOI: 10.2147/cmar.s248854] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 05/01/2020] [Indexed: 12/20/2022] Open
Abstract
Background Elongation factor for RNA polymerase II 2 (ELL2) was reported as a putative tumor suppressor in the prostate. ELL2 is frequently down-regulated in prostatic adenocarcinoma specimens, and loss of ELL2 induced murine prostatic intraepithelial neoplasia and enhanced AR-positive prostate cancer cell proliferation. However, the ELL2 gene appears to be amplified in AR-negative neuroendocrine prostate tumors, suggesting a potential oncogenic role for ELL2 in AR-negative prostate cancer cells. In this study, we explored the potential function of ELL2 in PC-3 and DU145, two AR-negative prostate cancer cell lines. Materials and Methods The role of ELL2 in PC-3 and DU145 cells was studied using siRNA-mediated ELL2 knockdown. Genes regulated by ELL2 knockdown in PC-3 cells were identified and analyzed using RNA-Seq and bioinformatics. The expression of representative genes was confirmed by Western blot and/or quantitative PCR. Cell growth was determined by BrdU, MTT and colony formation assays. Cell death was analyzed by 7-AAD/Annexin V staining and trypan blue exclusion staining. Cell cycle was determined by PI staining and flow cytometry. Results ELL2 knockdown inhibited the proliferation of PC-3 and DU145 cells. RNA-Seq analysis showed an enrichment in genes associated with cell death and survival following ELL2 knockdown. The interferon-γ pathway was identified as the top canonical pathway comprising of 55.6% of the genes regulated by ELL2. ELL2 knockdown induced an increase in STAT1 and IRF1 mRNA and an induction of total STAT1 and phosphorylated STAT1 protein. Inhibition of cell proliferation by ELL2 knockdown was partly abrogated by STAT1 knockdown. ELL2 knockdown inhibited colony formation and induced apoptosis in both PC-3 and DU145 cells. Furthermore, knockdown of ELL2 caused S-phase cell cycle arrest, inhibition of CDK2 phosphorylation and cyclin D1 expression, and increased expression of cyclin E. Conclusion ELL2 knockdown in PC-3 and DU145 cells induced S-phase cell cycle arrest and profound apoptosis, which was accompanied by the induction of genes associated with cell death and survival pathways. These observations suggest that ELL2 is a potential oncogenic protein required for survival and proliferation in AR-negative prostate cancer cells.
Collapse
Affiliation(s)
- Zhi Wang
- Department of Urology, Xiangya Hospital of Central South University, Changsha, People's Republic of China.,Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Laura E Pascal
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Uma R Chandran
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Srilakshmi Chaparala
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shidong Lv
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Hui Ding
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Lin Qi
- Department of Urology, Xiangya Hospital of Central South University, Changsha, People's Republic of China
| | - Zhou Wang
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
11
|
The H2B ubiquitin-protein ligase RNF40 is required for somatic cell reprogramming. Cell Death Dis 2020; 11:287. [PMID: 32341358 PMCID: PMC7184622 DOI: 10.1038/s41419-020-2482-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 04/06/2020] [Accepted: 04/07/2020] [Indexed: 12/11/2022]
Abstract
Direct reprogramming of somatic cells to induced pluripotent stem cells (iPSCs) requires a resetting of the epigenome in order to facilitate a cell fate transition. Previous studies have shown that epigenetic modifying enzymes play a central role in controlling induced pluripotency and the generation of iPSC. Here we show that RNF40, a histone H2B lysine 120 E3 ubiquitin-protein ligase, is specifically required for early reprogramming during induced pluripotency. Loss of RNF40-mediated H2B monoubiquitination (H2Bub1) impaired early gene activation in reprogramming. We further show that RNF40 contributes to tissue-specific gene suppression via indirect effects by controlling the expression of the polycomb repressive complex-2 histone methyltransferase component EZH2, as well as through more direct effects by promoting the resolution of H3K4me3/H3K27me3 bivalency on H2Bub1-occupied pluripotency genes. Thus, we identify RNF40 as a central epigenetic mediator of cell state transition with distinct functions in resetting somatic cell state to pluripotency.
Collapse
|
12
|
Data supporting the functional role of Eleven-nineteen Lysine-rich Leukemia 3 (ELL3) in B cell lymphoma cell line cells. Data Brief 2017; 15:222-227. [PMID: 29022001 PMCID: PMC5633249 DOI: 10.1016/j.dib.2017.09.042] [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: 08/21/2017] [Revised: 09/15/2017] [Accepted: 09/20/2017] [Indexed: 11/22/2022] Open
Abstract
The data presented here are related to the research article entitled “Selective expression of the transcription elongation factor ELL3 in B cells prior to ELL2 drives proliferation and survival” (Alexander et al., 2017) [1]. The cited research article characterizes Eleven-nineteen Lysine-rich Leukemia 3 (ELL3) expression in the B cell compartment and functional dependence in B lymphoma cell lines. This data report describes the mRNA expression pattern in a panel of cell lines representing the B cell compartment, supplementing the protein expression data presented in the associated research report. In addition, a reanalysis is presented of publicly available mRNA expression data from primary murine B cells to reveal dynamic regulation of the ELL family members post LPS stimulation (Barwick et al., 2016) [2]. The effect of ELL3 depletion on cell morphology, latent Epstein Barr Virus (EBV) lytic replication and differentiation markers in a Burkitt's lymphoma (BL) cell line cells are presented.
Collapse
|