1
|
Wu J, Fan C, Kabir AU, Krchma K, Kim M, Kwon Y, Xing X, Wang T, Choi K. Baf155 controls hematopoietic differentiation and regeneration through chromatin priming. Cell Rep 2024; 43:114558. [PMID: 39088321 PMCID: PMC11465209 DOI: 10.1016/j.celrep.2024.114558] [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: 10/12/2023] [Revised: 05/06/2024] [Accepted: 07/14/2024] [Indexed: 08/03/2024] Open
Abstract
Chromatin priming promotes cell-type-specific gene expression, lineage differentiation, and development. The mechanism of chromatin priming has not been fully understood. Here, we report that mouse hematopoietic stem and progenitor cells (HSPCs) lacking the Baf155 subunit of the BAF (BRG1/BRM-associated factor) chromatin remodeling complex produce a significantly reduced number of mature blood cells, leading to a failure of hematopoietic regeneration upon transplantation and 5-fluorouracil (5-FU) injury. Baf155-deficient HSPCs generate particularly fewer neutrophils, B cells, and CD8+ T cells at homeostasis, supporting a more immune-suppressive tumor microenvironment and enhanced tumor growth. Single-nucleus multiomics analysis reveals that Baf155-deficient HSPCs fail to establish accessible chromatin in selected regions that are enriched for putative enhancers and binding motifs of hematopoietic lineage transcription factors. Our study provides a fundamental mechanistic understanding of the role of Baf155 in hematopoietic lineage chromatin priming and the functional consequences of Baf155 deficiency in regeneration and tumor immunity.
Collapse
Affiliation(s)
- Jun Wu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Changxu Fan
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA; Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ashraf Ul Kabir
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Karen Krchma
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Minseo Kim
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yoojung Kwon
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Xiaoyun Xing
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA; Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ting Wang
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA; Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kyunghee Choi
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
2
|
Wu ML, Wheeler K, Silasi R, Lupu F, Griffin CT. Endothelial Chromatin-Remodeling Enzymes Regulate the Production of Critical ECM Components During Murine Lung Development. Arterioscler Thromb Vasc Biol 2024; 44:1784-1798. [PMID: 38868942 PMCID: PMC11624602 DOI: 10.1161/atvbaha.124.320881] [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: 02/23/2024] [Accepted: 05/29/2024] [Indexed: 06/14/2024]
Abstract
BACKGROUND The chromatin-remodeling enzymes BRG1 (brahma-related gene 1) and CHD4 (chromodomain helicase DNA-binding protein 4) independently regulate the transcription of genes critical for vascular development, but their coordinated impact on vessels in late-stage embryos has not been explored. METHODS In this study, we genetically deleted endothelial Brg1 and Chd4 in mixed background mice (Brg1fl/fl;Chd4fl/fl;VE-Cadherin-Cre), and littermates that were negative for Cre recombinase were used as controls. Tissues were analyzed by immunostaining, immunoblot, and flow cytometry. Quantitative reverse transcription polymerase chain reaction was used to determine gene expression, and chromatin immunoprecipitation revealed gene targets of BRG1 and CHD4 in cultured endothelial cells. RESULTS We found Brg1/Chd4 double mutants grew normally but died soon after birth with small and compact lungs. Despite having normal cellular composition, distal air sacs of the mutant lungs displayed diminished ECM (extracellular matrix) components and TGFβ (transforming growth factor-β) signaling, which typically promotes ECM synthesis. Transcripts for collagen- and elastin-related genes and the TGFβ ligand Tgfb1 were decreased in mutant lung endothelial cells, but genetic deletion of endothelial Tgfb1 failed to recapitulate the small lungs and ECM defects seen in Brg1/Chd4 mutants. We instead found several ECM genes to be direct targets of BRG1 and CHD4 in cultured endothelial cells. CONCLUSIONS Collectively, our data highlight essential roles for endothelial chromatin-remodeling enzymes in promoting ECM deposition in the distal lung tissue during the saccular stage of embryonic lung development.
Collapse
Affiliation(s)
- Meng-Ling Wu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Kate Wheeler
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Robert Silasi
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Florea Lupu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Courtney T. Griffin
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
3
|
Ma X, Zhao J, Feng Y. Epicardial SMARCA4 deletion exacerbates cardiac injury in myocardial infarction and is related to the inhibition of epicardial epithelial-mesenchymal transition. J Mol Cell Cardiol 2024; 191:76-87. [PMID: 38718920 DOI: 10.1016/j.yjmcc.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 05/01/2024] [Accepted: 05/04/2024] [Indexed: 05/12/2024]
Abstract
The reactivated adult epicardium produces epicardium-derived cells (EPDCs) via epithelial-mesenchymal transition (EMT) to benefit the recovery of the heart after myocardial infarction (MI). SMARCA4 is the core catalytic subunit of the chromatin re-modeling complex, which has the potential to target some reactivated epicardial genes in MI. However, the effects of epicardial SMARCA4 on MI remain uncertain. This study found that SMARCA4 was activated over time in epicardial cells following MI, and some of activated cells belonged to downstream differentiation types of EPDCs. This study used tamoxifen to induce lineage tracing and SMARCA4 deletion from epicardial cells in Wt1-CreER;Smarca4fl/fl;Rosa26-RFP adult mice. Epicardial SMARCA4 deletion reduces the number of epicardial cells in adult mice, which was related to changes in the activation, proliferation, and apoptosis of epicardial cells. Epicardial SMARCA4 deletion reduced collagen deposition and angiogenesis in the infarcted area, exacerbated cardiac injury in MI. The exacerbation of cardiac injury was related to the inhibition of generation and differentiation of EPDCs. The alterations in EPDCs were associated with inhibited transition between E-CAD and N-CAD during the epicardial EMT, coupled with the down-regulation of WT1, SNAIL1, and PDGF signaling. In conclusion, this study suggests that Epicardial SMARCA4 plays a critical role in cardiac injury caused by MI, and its regulatory mechanism is related to epicardial EMT. Epicardial SMARCA4 holds potential as a novel molecular target for treating MI.
Collapse
Affiliation(s)
- Xingyu Ma
- College of Life Science and Technology, Jinan University, Guangzhou, China.
| | - Jianjun Zhao
- College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Yi Feng
- College of Life Science and Technology, Jinan University, Guangzhou, China
| |
Collapse
|
4
|
Jolly AJ, Lu S, Dubner AM, Strand KA, Mutryn MF, Pilotti-Riley A, Danis EP, Nemenoff RA, Moulton KS, Majesky MW, Weiser-Evans MC. Redistribution of the chromatin remodeler Brg1 directs smooth muscle-derived adventitial progenitor-to-myofibroblast differentiation and vascular fibrosis. JCI Insight 2023; 8:e164862. [PMID: 36976650 PMCID: PMC10243795 DOI: 10.1172/jci.insight.164862] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
Vascular smooth muscle-derived Sca1+ adventitial progenitor (AdvSca1-SM) cells are tissue-resident, multipotent stem cells that contribute to progression of vascular remodeling and fibrosis. Upon acute vascular injury, AdvSca1-SM cells differentiate into myofibroblasts and are embedded in perivascular collagen and the extracellular matrix. While the phenotypic properties of AdvSca1-SM-derived myofibroblasts have been defined, the underlying epigenetic regulators driving the AdvSca1-SM-to-myofibroblast transition are unclear. We show that the chromatin remodeler Smarca4/Brg1 facilitates AdvSca1-SM myofibroblast differentiation. Brg1 mRNA and protein were upregulated in AdvSca1-SM cells after acute vascular injury, and pharmacological inhibition of Brg1 by the small molecule PFI-3 attenuated perivascular fibrosis and adventitial expansion. TGF-β1 stimulation of AdvSca1-SM cells in vitro reduced expression of stemness genes while inducing expression of myofibroblast genes that was associated with enhanced contractility; PFI blocked TGF-β1-induced phenotypic transition. Similarly, genetic knockdown of Brg1 in vivo reduced adventitial remodeling and fibrosis and reversed AdvSca1-SM-to-myofibroblast transition in vitro. Mechanistically, TGF-β1 promoted redistribution of Brg1 from distal intergenic sites of stemness genes and recruitment to promoter regions of myofibroblast-related genes, which was blocked by PFI-3. These data provide insight into epigenetic regulation of resident vascular progenitor cell differentiation and support that manipulating the AdvSca1-SM phenotype will provide antifibrotic clinical benefits.
Collapse
Affiliation(s)
- Austin J. Jolly
- Department of Medicine, Division of Renal Diseases and Hypertension
- Medical Scientist Training Program
| | - Sizhao Lu
- Department of Medicine, Division of Renal Diseases and Hypertension
- School of Medicine, Consortium for Fibrosis Research and Translation
| | | | - Keith A. Strand
- Department of Medicine, Division of Renal Diseases and Hypertension
| | - Marie F. Mutryn
- Department of Medicine, Division of Renal Diseases and Hypertension
| | | | | | - Raphael A. Nemenoff
- Department of Medicine, Division of Renal Diseases and Hypertension
- School of Medicine, Consortium for Fibrosis Research and Translation
- Cardiovascular Pulmonary Research Program, and
| | - Karen S. Moulton
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Mark W. Majesky
- Center for Developmental Biology & Regenerative Medicine, Seattle Children’s Research Institute, Seattle, Washington, USA
- Departments of Pediatrics and Pathology, University of Washington, Seattle, Washington, USA
| | - Mary C.M. Weiser-Evans
- Department of Medicine, Division of Renal Diseases and Hypertension
- School of Medicine, Consortium for Fibrosis Research and Translation
- Cardiovascular Pulmonary Research Program, and
| |
Collapse
|
5
|
Xiao C, Hou J, Wang F, Song Y, Zheng J, Luo L, Wang J, Ding W, Zhu X, Xiong JW. Endothelial Brg1 fine-tunes Notch signaling during zebrafish heart regeneration. NPJ Regen Med 2023; 8:21. [PMID: 37029137 PMCID: PMC10082087 DOI: 10.1038/s41536-023-00293-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 03/17/2023] [Indexed: 04/09/2023] Open
Abstract
Myocardial Brg1 is essential for heart regeneration in zebrafish, but it remains unknown whether and how endothelial Brg1 plays a role in heart regeneration. Here, we found that both brg1 mRNA and protein were induced in cardiac endothelial cells after ventricular resection and endothelium-specific overexpression of dominant-negative Xenopus Brg1 (dn-xbrg1) inhibited myocardial proliferation and heart regeneration and increased cardiac fibrosis. RNA-seq and ChIP-seq analysis revealed that endothelium-specific overexpression of dn-xbrg1 changed the levels of H3K4me3 modifications in the promoter regions of the zebrafish genome and induced abnormal activation of Notch family genes upon injury. Mechanistically, Brg1 interacted with lysine demethylase 7aa (Kdm7aa) to fine-tune the level of H3K4me3 within the promoter regions of Notch family genes and thus regulated notch gene transcription. Together, this work demonstrates that the Brg1-Kdm7aa-Notch axis in cardiac endothelial cells, including the endocardium, regulates myocardial proliferation and regeneration via modulating the H3K4me3 of the notch promoters in zebrafish.
Collapse
Affiliation(s)
- Chenglu Xiao
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, 100871, Beijing, China
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, 100193, Beijing, China
| | - Junjie Hou
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, 100871, Beijing, China
| | - Fang Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, 310014, Hangzhou, China
| | - Yabing Song
- School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Jiyuan Zheng
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, 100871, Beijing, China
| | - Lingfei Luo
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, 400715, Chongqing, China
| | - Jianbin Wang
- School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Wanqiu Ding
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, 100871, Beijing, China.
| | - Xiaojun Zhu
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, 100871, Beijing, China.
| | - Jing-Wei Xiong
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, 100871, Beijing, China.
| |
Collapse
|
6
|
Andrades A, Peinado P, Alvarez-Perez JC, Sanjuan-Hidalgo J, García DJ, Arenas AM, Matia-González AM, Medina PP. SWI/SNF complexes in hematological malignancies: biological implications and therapeutic opportunities. Mol Cancer 2023; 22:39. [PMID: 36810086 PMCID: PMC9942420 DOI: 10.1186/s12943-023-01736-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 01/30/2023] [Indexed: 02/23/2023] Open
Abstract
Hematological malignancies are a highly heterogeneous group of diseases with varied molecular and phenotypical characteristics. SWI/SNF (SWItch/Sucrose Non-Fermentable) chromatin remodeling complexes play significant roles in the regulation of gene expression, being essential for processes such as cell maintenance and differentiation in hematopoietic stem cells. Furthermore, alterations in SWI/SNF complex subunits, especially in ARID1A/1B/2, SMARCA2/4, and BCL7A, are highly recurrent across a wide variety of lymphoid and myeloid malignancies. Most genetic alterations cause a loss of function of the subunit, suggesting a tumor suppressor role. However, SWI/SNF subunits can also be required for tumor maintenance or even play an oncogenic role in certain disease contexts. The recurrent alterations of SWI/SNF subunits highlight not only the biological relevance of SWI/SNF complexes in hematological malignancies but also their clinical potential. In particular, increasing evidence has shown that mutations in SWI/SNF complex subunits confer resistance to several antineoplastic agents routinely used for the treatment of hematological malignancies. Furthermore, mutations in SWI/SNF subunits often create synthetic lethality relationships with other SWI/SNF or non-SWI/SNF proteins that could be exploited therapeutically. In conclusion, SWI/SNF complexes are recurrently altered in hematological malignancies and some SWI/SNF subunits may be essential for tumor maintenance. These alterations, as well as their synthetic lethal relationships with SWI/SNF and non-SWI/SNF proteins, may be pharmacologically exploited for the treatment of diverse hematological cancers.
Collapse
Affiliation(s)
- Alvaro Andrades
- grid.4489.10000000121678994Department of Biochemistry and Molecular Biology I. Faculty of Sciences, University of Granada, Granada, Spain ,grid.470860.d0000 0004 4677 7069GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, Spain ,grid.507088.2Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Paola Peinado
- grid.4489.10000000121678994Department of Biochemistry and Molecular Biology I. Faculty of Sciences, University of Granada, Granada, Spain ,grid.470860.d0000 0004 4677 7069GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, Spain ,grid.507088.2Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain ,grid.451388.30000 0004 1795 1830Present Address: The Francis Crick Institute, London, UK
| | - Juan Carlos Alvarez-Perez
- grid.4489.10000000121678994Department of Biochemistry and Molecular Biology I. Faculty of Sciences, University of Granada, Granada, Spain ,grid.470860.d0000 0004 4677 7069GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, Spain ,grid.507088.2Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Juan Sanjuan-Hidalgo
- grid.4489.10000000121678994Department of Biochemistry and Molecular Biology I. Faculty of Sciences, University of Granada, Granada, Spain ,grid.470860.d0000 0004 4677 7069GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, Spain
| | - Daniel J. García
- grid.470860.d0000 0004 4677 7069GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, Spain ,grid.4489.10000000121678994Department of Biochemistry and Molecular Biology III and Immunology, University of Granada, Granada, Spain
| | - Alberto M. Arenas
- grid.4489.10000000121678994Department of Biochemistry and Molecular Biology I. Faculty of Sciences, University of Granada, Granada, Spain ,grid.470860.d0000 0004 4677 7069GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, Spain ,grid.507088.2Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Ana M. Matia-González
- grid.4489.10000000121678994Department of Biochemistry and Molecular Biology I. Faculty of Sciences, University of Granada, Granada, Spain ,grid.470860.d0000 0004 4677 7069GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, Spain ,grid.507088.2Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Pedro P. Medina
- grid.4489.10000000121678994Department of Biochemistry and Molecular Biology I. Faculty of Sciences, University of Granada, Granada, Spain ,grid.470860.d0000 0004 4677 7069GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, Spain ,grid.507088.2Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| |
Collapse
|
7
|
Gong W, Das S, Sierra-Pagan JE, Skie E, Dsouza N, Larson TA, Garry MG, Luzete-Monteiro E, Zaret KS, Garry DJ. ETV2 functions as a pioneer factor to regulate and reprogram the endothelial lineage. Nat Cell Biol 2022; 24:672-684. [PMID: 35550615 PMCID: PMC11827897 DOI: 10.1038/s41556-022-00901-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 03/17/2022] [Indexed: 12/14/2022]
Abstract
The vasculature is an essential organ for the delivery of blood and oxygen to all tissues of the body and is thus relevant to the treatment of ischaemic diseases, injury-induced regeneration and solid tumour growth. Previously, we demonstrated that ETV2 is an essential transcription factor for the development of cardiac, endothelial and haematopoietic lineages. Here we report that ETV2 functions as a pioneer factor that relaxes closed chromatin and regulates endothelial development. By comparing engineered embryonic stem cell differentiation and reprogramming models with multi-omics techniques, we demonstrated that ETV2 was able to bind nucleosomal DNA and recruit BRG1. BRG1 recruitment remodelled chromatin around endothelial genes and helped to maintain an open configuration, resulting in increased H3K27ac deposition. Collectively, these results will serve as a platform for the development of therapeutic initiatives directed towards cardiovascular diseases and solid tumours.
Collapse
Affiliation(s)
- Wuming Gong
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Satyabrata Das
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Javier E Sierra-Pagan
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Erik Skie
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Nikita Dsouza
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Thijs A Larson
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Mary G Garry
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
- Paul and Sheila Wellstone Muscular Dystrophy Center, University of Minnesota, Minneapolis, MN, USA
| | - Edgar Luzete-Monteiro
- Institute for Regenerative Medicine, Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Kenneth S Zaret
- Institute for Regenerative Medicine, Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Daniel J Garry
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN, USA.
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA.
- Paul and Sheila Wellstone Muscular Dystrophy Center, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
8
|
Hemogen /BRG1 cooperativity modulates promoter and enhancer activation during erythropoiesis. Blood 2022; 139:3532-3545. [PMID: 35297980 DOI: 10.1182/blood.2021014308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 03/15/2022] [Indexed: 11/20/2022] Open
Abstract
Hemogen, also known as EDAG, is a hematopoietic tissue-specific gene that regulates the proliferation and differentiation of hematopoietic cells. However, the mechanism underlying hemogen function in erythropoiesis is unknown. We found that depletion of hemogen in human CD34+ erythroid progenitor cells and HUDEP2 cells significantly reduced the expression of genes associated with heme and hemoglobin synthesis, supporting a positive role of hemogen in erythroid maturation. In human K562 cells, hemogen antagonized the occupancy of co-repressors NuRD complex and facilitated LDB1 complex-mediated chromatin looping. Hemogen recruited SWI/SNF complex ATPase BRG1 as a co-activator to regulate nucleosome accessibility and H3K27ac enrichment for promoter and enhancer activity. To ask if hemogen/BRG1 cooperativity is conserved in mammalian systems, we generated hemogen KO/KI mice and investigated hemogen/BRG1 function in murine erythropoiesis. Loss of hemogen in E12.5-E16.5 fetal liver cells impeded erythroid differentiation through reducing the production of mature erythroblasts. ChIP-seq in WT and hemogen KO animal revealed BRG1 is largely dependent on hemogen to regulate chromatin accessibility at erythroid gene promoters and enhancers. In summary, hemogen/BRG1 interaction in mammals is essential for fetal erythroid maturation and hemoglobin production through its active role in promoter and enhancer activity and chromatin organization.
Collapse
|
9
|
Bell RAV, Al-Khalaf MH, Brunette S, Alsowaida D, Chu A, Bandukwala H, Dechant G, Apostolova G, Dilworth FJ, Megeney LA. Chromatin Reorganization during Myoblast Differentiation Involves the Caspase-Dependent Removal of SATB2. Cells 2022; 11:966. [PMID: 35326417 PMCID: PMC8946544 DOI: 10.3390/cells11060966] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 02/27/2022] [Accepted: 03/09/2022] [Indexed: 11/17/2022] Open
Abstract
The induction of lineage-specific gene programs are strongly influenced by alterations in local chromatin architecture. However, key players that impact this genome reorganization remain largely unknown. Here, we report that the removal of the special AT-rich binding protein 2 (SATB2), a nuclear protein known to bind matrix attachment regions, is a key event in initiating myogenic differentiation. The deletion of myoblast SATB2 in vitro initiates chromatin remodeling and accelerates differentiation, which is dependent on the caspase 7-mediated cleavage of SATB2. A genome-wide analysis indicates that SATB2 binding within chromatin loops and near anchor points influences both loop and sub-TAD domain formation. Consequently, the chromatin changes that occur with the removal of SATB2 lead to the derepression of differentiation-inducing factors while also limiting the expression of genes that inhibit this cell fate change. Taken together, this study demonstrates that the temporal control of the SATB2 protein is critical in shaping the chromatin environment and coordinating the myogenic differentiation program.
Collapse
Affiliation(s)
- Ryan A. V. Bell
- Regenerative Medicine Program, Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada; (R.A.V.B.); (M.H.A.-K.); (S.B.); (D.A.); (A.C.); (H.B.); (F.J.D.)
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Mohammad H. Al-Khalaf
- Regenerative Medicine Program, Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada; (R.A.V.B.); (M.H.A.-K.); (S.B.); (D.A.); (A.C.); (H.B.); (F.J.D.)
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- University of Ottawa Heart Institute, Ottawa, ON K1Y 4W7, Canada
| | - Steve Brunette
- Regenerative Medicine Program, Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada; (R.A.V.B.); (M.H.A.-K.); (S.B.); (D.A.); (A.C.); (H.B.); (F.J.D.)
| | - Dalal Alsowaida
- Regenerative Medicine Program, Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada; (R.A.V.B.); (M.H.A.-K.); (S.B.); (D.A.); (A.C.); (H.B.); (F.J.D.)
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Alphonse Chu
- Regenerative Medicine Program, Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada; (R.A.V.B.); (M.H.A.-K.); (S.B.); (D.A.); (A.C.); (H.B.); (F.J.D.)
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Hina Bandukwala
- Regenerative Medicine Program, Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada; (R.A.V.B.); (M.H.A.-K.); (S.B.); (D.A.); (A.C.); (H.B.); (F.J.D.)
| | - Georg Dechant
- Institute of Neuroscience, Medical University of Innsbruck, A-6020 Innsbruck, Austria; (G.D.); (G.A.)
| | - Galina Apostolova
- Institute of Neuroscience, Medical University of Innsbruck, A-6020 Innsbruck, Austria; (G.D.); (G.A.)
| | - F. Jeffrey Dilworth
- Regenerative Medicine Program, Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada; (R.A.V.B.); (M.H.A.-K.); (S.B.); (D.A.); (A.C.); (H.B.); (F.J.D.)
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Lynn A. Megeney
- Regenerative Medicine Program, Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada; (R.A.V.B.); (M.H.A.-K.); (S.B.); (D.A.); (A.C.); (H.B.); (F.J.D.)
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
10
|
Naidu SR, Capitano M, Ropa J, Cooper S, Huang X, Broxmeyer HE. Chromatin remodeling subunit BRM and valine regulate hematopoietic stem/progenitor cell function and self-renewal via intrinsic and extrinsic effects. Leukemia 2022; 36:821-833. [PMID: 34599272 PMCID: PMC9212754 DOI: 10.1038/s41375-021-01426-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 09/07/2021] [Accepted: 09/14/2021] [Indexed: 02/08/2023]
Abstract
Little is known of hematopoietic stem (HSC) and progenitor (HPC) cell self-renewal. The role of Brahma (BRM), a chromatin remodeler, in HSC function is unknown. Bone marrow (BM) from Brm-/- mice manifested increased numbers of long- and short-term HSCs, GMPs, and increased numbers and cycling of functional HPCs. However, increased Brm-/- BM HSC numbers had decreased secondary and tertiary engraftment, suggesting BRM enhances HSC self-renewal. Valine was elevated in lineage negative Brm-/- BM cells, linking intracellular valine with Brm expression. Valine enhanced HPC colony formation, replating of human cord blood (CB) HPC-derived colonies, mouse BM and human CB HPC survival in vitro, and ex vivo expansion of normal mouse BM HSCs and HPCs. Valine increased oxygen consumption rates of WT cells. BRM through CD98 was linked to regulated import of branched chain amino acids, such as valine, in HPCs. Brm-/- LSK cells exhibited upregulated interferon response/cell cycle gene programs. Effects of BRM depletion are less apparent on isolated HSCs compared to HSCs in the presence of HPCs, suggesting cell extrinsic effects on HSCs. Thus, intracellular valine is regulated by BRM expression in HPCs, and the BRM/valine axis regulates HSC and HPC self-renewal, proliferation, and possibly differentiation fate decisions.
Collapse
Affiliation(s)
- Samisubbu R. Naidu
- Medicine, Indiana University School of Medicine,Indiana University School of Medicine, Department of Microbiology/Immunology, 950 West Walnut Street, Bldg. R2, Room 302, Indianapolis, IN 46202, Indiana University School of Medicine, Department of Medicine, 950 West Walnut Street, Bldg. R2, Room E435,These authors contributed equally to this work
| | - Maegan Capitano
- Departments of Microbiology and Immunology, Indiana University School of Medicine,These authors contributed equally to this work
| | - James Ropa
- Departments of Microbiology and Immunology, Indiana University School of Medicine,These authors contributed equally to this work
| | - Scott Cooper
- Departments of Microbiology and Immunology, Indiana University School of Medicine
| | - Xinxin Huang
- Departments of Microbiology and Immunology, Indiana University School of Medicine
| | - Hal E. Broxmeyer
- Departments of Microbiology and Immunology, Indiana University School of Medicine,Indiana University School of Medicine, Department of Microbiology/Immunology, 950 West Walnut Street, Bldg. R2, Room 302, Indianapolis, IN 46202, Indiana University School of Medicine, Department of Medicine, 950 West Walnut Street, Bldg. R2, Room E435
| |
Collapse
|
11
|
Ding Y, Li Y, Zhao Z, Cliff Zhang Q, Liu F. The chromatin-remodeling enzyme Smarca5 regulates erythrocyte aggregation via Keap1-Nrf2 signaling. eLife 2021; 10:72557. [PMID: 34698638 PMCID: PMC8594921 DOI: 10.7554/elife.72557] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 10/23/2021] [Indexed: 12/30/2022] Open
Abstract
Although thrombosis has been extensively studied using various animal models, our understanding of the underlying mechanism remains elusive. Here, using zebrafish model, we demonstrated that smarca5-deficient red blood cells (RBCs) formed blood clots in the caudal vein plexus. We further used the anti-thrombosis drugs to treat smarca5zko1049a embryos and found that a thrombin inhibitor, argatroban, partially prevented blood clot formation in smarca5zko1049a. To explore the regulatory mechanism of smarca5 in RBC homeostasis, we profiled the chromatin accessibility landscape and transcriptome features in RBCs from smarca5zko1049a and their siblings and found that both the chromatin accessibility at the keap1a promoter and expression of keap1a were decreased. Keap1 is a suppressor protein of Nrf2, which is a major regulator of oxidative responses. We further identified that the expression of hmox1a, a downstream target of Keap1-Nrf2 signaling pathway, was markedly increased upon smarca5 deletion. Importantly, overexpression of keap1a or knockdown of hmox1a partially rescued the blood clot formation, suggesting that the disrupted Keap1-Nrf2 signaling is responsible for the RBC aggregation in smarca5 mutants. Together, our study using zebrafish smarca5 mutants characterizes a novel role for smarca5 in RBC aggregation, which may provide a new venous thrombosis animal model to support drug screening and pre-clinical therapeutic assessments to treat thrombosis.
Collapse
Affiliation(s)
- Yanyan Ding
- The Max-Planck Center for Tissue Stem Cell Research and Regenerative Medicine, Bioland Laboratory, Guangzhou, China.,State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yuzhe Li
- MOE Key Laboratory of Bioinformatics, Beijing Advanced Innovation Center for Structural Biology & Frontier Research Center for Biological Structure, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing, China.,Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Ziqian Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Qiangfeng Cliff Zhang
- MOE Key Laboratory of Bioinformatics, Beijing Advanced Innovation Center for Structural Biology & Frontier Research Center for Biological Structure, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Feng Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
12
|
Yoshikawa T, Fukuda A, Omatsu M, Namikawa M, Sono M, Fukunaga Y, Masuda T, Araki O, Nagao M, Ogawa S, Masuo K, Goto N, Hiramatsu Y, Muta Y, Tsuda M, Maruno T, Nakanishi Y, Kawada K, Takaishi S, Seno H. Brg1 is required to maintain colorectal cancer stem cells. J Pathol 2021; 255:257-269. [PMID: 34415580 DOI: 10.1002/path.5759] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 06/08/2021] [Accepted: 07/13/2021] [Indexed: 01/09/2023]
Abstract
Tumor cells capable of self-renewal and continuous production of progeny cells are called tumor stem cells (TSCs) and are considered to be potential therapeutic targets. However, the mechanisms underlying the survival and function of TSCs are not fully understood. We previously reported that chromatin remodeling regulator Brg1 is essential for intestinal stem cells in mice and Dclk1 is an intestinal TSC marker. In this study, we investigated the role of Brg1 in Dclk1+ intestinal tumor cells for the maintenance of intestinal tumors in mice. Specific ablation of Brg1 in Dclk1+ intestinal tumor cells reduced intestinal tumors in ApcMin mice, and continuous ablation of Brg1 maintained the reduction of intestinal tumors. Lineage tracing in the context of Brg1 ablation in Dclk1+ intestinal tumor cells revealed that Brg1-null Dclk1+ intestinal tumor cells did not give rise to their descendent tumor cells, indicating that Brg1 is essential for the self-renewal of Dclk1+ intestinal tumor cells. Five days after Brg1 ablation, we observed increased apoptosis in Dclk1+ tumor cells. Furthermore, Brg1 was crucial for the stemness of intestinal tumor cells in a spheroid culture system. BRG1 knockdown also impaired cell proliferation and increased apoptosis in human colorectal cancer (CRC) cells. Microarray analysis revealed that apoptosis-related genes were upregulated and stem cell-related genes were downregulated in human CRC cells by BRG1 suppression. Consistently, high BRG1 expression correlated with poor disease-specific survival in human CRC patients. These data indicate that Brg1 plays a crucial role in intestinal TSCs in mice by inhibiting apoptosis and is critical for cell survival and stem cell features in human CRC cells. Thus, BRG1 represents a new therapeutic target for human CRC. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Takaaki Yoshikawa
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Akihisa Fukuda
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Mayuki Omatsu
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Mio Namikawa
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Makoto Sono
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuichi Fukunaga
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Department of Drug Discovery Medicine, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tomonori Masuda
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Osamu Araki
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Munemasa Nagao
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Satoshi Ogawa
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kenji Masuo
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Laboratory for Malignancy Control Research (DSK Project), Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Norihiro Goto
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yukiko Hiramatsu
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yu Muta
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Motoyuki Tsuda
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takahisa Maruno
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuki Nakanishi
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kenji Kawada
- Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shigeo Takaishi
- Laboratory for Malignancy Control Research (DSK Project), Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiroshi Seno
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
13
|
Quillien A, Gilbert G, Boulet M, Ethuin S, Waltzer L, Vandel L. Prmt5 promotes vascular morphogenesis independently of its methyltransferase activity. PLoS Genet 2021; 17:e1009641. [PMID: 34153034 PMCID: PMC8248709 DOI: 10.1371/journal.pgen.1009641] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 07/01/2021] [Accepted: 06/02/2021] [Indexed: 01/02/2023] Open
Abstract
During development, the vertebrate vasculature undergoes major growth and remodeling. While the transcriptional cascade underlying blood vessel formation starts to be better characterized, little is known concerning the role and mode of action of epigenetic enzymes during this process. Here, we explored the role of the Protein Arginine Methyl Transferase Prmt5 in blood vessel formation as well as hematopoiesis using zebrafish as a model system. Through the combination of different prmt5 loss-of-function approaches we highlighted a key role of Prmt5 in both processes. Notably, we showed that Prmt5 promotes vascular morphogenesis through the transcriptional control of ETS transcription factors and adhesion proteins in endothelial cells. Interestingly, using a catalytic dead mutant of Prmt5 and a specific drug inhibitor, we found that while Prmt5 methyltransferase activity was required for blood cell formation, it was dispensable for vessel formation. Analyses of chromatin architecture impact on reporter genes expression and chromatin immunoprecipitation experiments led us to propose that Prmt5 regulates transcription by acting as a scaffold protein that facilitates chromatin looping to promote vascular morphogenesis. Blood vessel formation is an essential developmental process required for the survival of all vertebrates. The vascular anatomy and the mechanisms involved in vessel formation are highly conserved among vertebrates. Hence, we used zebrafish as a model, to decipher the role and the mode of action of Prmt5, an enzyme known to regulate gene expression, in vascular morphogenesis and in blood cell formation in vivo. Using different approaches, we highlighted a key role of Prmt5 during both processes. However, we found that while blood cell formation required Prmt5 enzymatic activity, vascular morphogenesis was independent on its activity. Prmt5 has been proposed as a therapeutic target in many diseases, including cancer. Yet, we show here that Prmt5 acts at least in part independently of its methyltransferase activity to regulate vascular morphogenesis. By shedding light on a mechanism of action of Prmt5 that will be insensitive to enzymatic inhibitors, our data calls forth the design of alternative drugs. In addition, this non-canonical function of Prmt5 may have a more pervasive role than previously thought in physiological conditions, i.e. during development, but also in pathological situations such as in tumor angiogenesis and certainly deserves more attention in the future.
Collapse
Affiliation(s)
- Aurélie Quillien
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
- RESTORE, INSERM UMR1301, CNRS UMR5070, Université Paul Sabatier, Université de Toulouse, Toulouse, France
- * E-mail: (AQ); (LV)
| | - Guerric Gilbert
- Université Clermont Auvergne, CNRS, INSERM, iGReD, Clermont-Ferrand, France
| | - Manon Boulet
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
- Université Clermont Auvergne, CNRS, INSERM, iGReD, Clermont-Ferrand, France
| | - Séverine Ethuin
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Lucas Waltzer
- Université Clermont Auvergne, CNRS, INSERM, iGReD, Clermont-Ferrand, France
| | - Laurence Vandel
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
- Université Clermont Auvergne, CNRS, INSERM, iGReD, Clermont-Ferrand, France
- * E-mail: (AQ); (LV)
| |
Collapse
|
14
|
Wanior M, Preuss F, Ni X, Krämer A, Mathea S, Göbel T, Heidenreich D, Simonyi S, Kahnt AS, Joerger AC, Knapp S. Pan-SMARCA/PB1 Bromodomain Inhibitors and Their Role in Regulating Adipogenesis. J Med Chem 2020; 63:14680-14699. [PMID: 33216538 DOI: 10.1021/acs.jmedchem.0c01242] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Accessibility of the human genome is modulated by the ATP-driven SWI/SNF chromatin remodeling multiprotein complexes BAF (BRG1/BRM-associated factor) and PBAF (polybromo-associated BAF factor), which involves reading of acetylated histone tails by the bromodomain-containing proteins SMARCA2 (BRM), SMARCA4 (BRG1), and polybromo-1. Dysregulation of chromatin remodeling leads to aberrant cell proliferation and differentiation. Here, we have characterized a set of potent and cell-active bromodomain inhibitors with pan-selectivity for canonical family VIII bromodomains. Targeted SWI/SNF bromodomain inhibition blocked the expression of key genes during adipogenesis, including the transcription factors PPARγ and C/EBPα, and impaired the differentiation of 3T3-L1 murine fibroblasts into adipocytes. Our data highlight the role of SWI/SNF bromodomains in adipogenesis and provide a framework for the development of SWI/SNF bromodomain inhibitors for indirect targeting of key transcription factors regulating cell differentiation.
Collapse
Affiliation(s)
- Marek Wanior
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany.,Structural Genomics Consortium (SGC), Buchmann Institute for Molecular Life Sciences (BMLS), Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Franziska Preuss
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany.,Structural Genomics Consortium (SGC), Buchmann Institute for Molecular Life Sciences (BMLS), Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Xiaomin Ni
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany.,Structural Genomics Consortium (SGC), Buchmann Institute for Molecular Life Sciences (BMLS), Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Andreas Krämer
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany.,Structural Genomics Consortium (SGC), Buchmann Institute for Molecular Life Sciences (BMLS), Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany.,Frankfurt Cancer Institute (FCI), Paul-Ehrlich-Str. 42-44, 60596 Frankfurt am Main, Germany
| | - Sebastian Mathea
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany.,Structural Genomics Consortium (SGC), Buchmann Institute for Molecular Life Sciences (BMLS), Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Tamara Göbel
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - David Heidenreich
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany.,Structural Genomics Consortium (SGC), Buchmann Institute for Molecular Life Sciences (BMLS), Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Svenja Simonyi
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Astrid S Kahnt
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Andreas C Joerger
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany.,Structural Genomics Consortium (SGC), Buchmann Institute for Molecular Life Sciences (BMLS), Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany.,German Translational Cancer Network (DKTK), Frankfurt/Mainz Site, 60438 Frankfurt am Main, Germany
| | - Stefan Knapp
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany.,Structural Genomics Consortium (SGC), Buchmann Institute for Molecular Life Sciences (BMLS), Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany.,German Translational Cancer Network (DKTK), Frankfurt/Mainz Site, 60438 Frankfurt am Main, Germany.,Frankfurt Cancer Institute (FCI), Paul-Ehrlich-Str. 42-44, 60596 Frankfurt am Main, Germany
| |
Collapse
|
15
|
Wu J, Krchma K, Lee HJ, Prabhakar S, Wang X, Zhao H, Xing X, Seong RH, Fremont DH, Artyomov MN, Wang T, Choi K. Requisite Chromatin Remodeling for Myeloid and Erythroid Lineage Differentiation from Erythromyeloid Progenitors. Cell Rep 2020; 33:108395. [PMID: 33207205 PMCID: PMC7694876 DOI: 10.1016/j.celrep.2020.108395] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 09/25/2020] [Accepted: 10/26/2020] [Indexed: 12/21/2022] Open
Abstract
The mammalian SWitch/Sucrose Non-Fermentable (SWI/SNF) chromatin-remodeling BAF (BRG1/BRM-associated factor) complex plays an essential role in developmental and pathological processes. We show that the deletion of Baf155, which encodes a subunit of the BAF complex, in the Tie2(+) lineage (Baf155 (CKO) leads to defects in yolk sac myeloid and definitive erythroid (EryD) lineage differentiation from erythromyeloid progenitors (EMPs). The chromatin of myeloid gene loci in Baf155 CKO EMPs is mostly inaccessible and enriched mainly by the ETS binding motif. BAF155 interacts with PU.1 and is recruited to PU.1 target gene loci together with p300 and KDM6a. Treatment of Baf155 CKO embryos with GSK126, an H3K27me2/3 methyltransferase EZH2 inhibitor, rescues myeloid lineage gene expression. This study uncovers indispensable BAF-mediated chromatin remodeling of myeloid gene loci at the EMP stage. Future studies exploiting epigenetics in the generation and application of EMP derivatives for tissue repair, regeneration, and disease are warranted. The mammalian chromatin-remodeling BAF (BRG1/BRM-associated factor) complex has an essential role in developmental and pathological processes. Wu et al. show that BAF-mediated chromatin remodeling and activation of the myeloid and definitive erythroid transcriptional program at the EMP stage is critical for myeloid and definitive erythroid lineage development.
Collapse
Affiliation(s)
- Jun Wu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Karen Krchma
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Hyung Joo Lee
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA; Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Sairam Prabhakar
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Xiaoli Wang
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Haiyong Zhao
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Xiaoyun Xing
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA; Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Rho H Seong
- Department of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Korea
| | - Daved H Fremont
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Maxim N Artyomov
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Ting Wang
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA; Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA; McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
| | - Kyunghee Choi
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA; Graduate School of Biotechnology, Kyung Hee University, Yong In, Korea.
| |
Collapse
|
16
|
Leisegang MS, Bibli SI, Günther S, Pflüger-Müller B, Oo JA, Höper C, Seredinski S, Yekelchyk M, Schmitz-Rixen T, Schürmann C, Hu J, Looso M, Sigala F, Boon RA, Fleming I, Brandes RP. Pleiotropic effects of laminar flow and statins depend on the Krüppel-like factor-induced lncRNA MANTIS. Eur Heart J 2020; 40:2523-2533. [PMID: 31222221 DOI: 10.1093/eurheartj/ehz393] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/28/2018] [Accepted: 05/24/2019] [Indexed: 11/12/2022] Open
Abstract
AIMS To assess the functional relevance and therapeutic potential of the pro-angiogenic long non-coding RNA MANTIS in vascular disease development. METHODS AND RESULTS RNA sequencing, CRISPR activation, overexpression, and RNAi demonstrated that MANTIS, especially its Alu-element, limits endothelial ICAM-1 expression in different types of endothelial cells. Loss of MANTIS increased endothelial monocyte adhesion in an ICAM-1-dependent manner. MANTIS reduced the binding of the SWI/SNF chromatin remodelling factor BRG1 at the ICAM-1 promoter. The expression of MANTIS was induced by laminar flow and HMG-CoA-reductase inhibitors (statins) through mechanisms involving epigenetic rearrangements and the transcription factors KLF2 and KLF4. Mutation of the KLF binding motifs in the MANTIS promoter blocked the flow-induced MANTIS expression. Importantly, the expression of MANTIS in human carotid artery endarterectomy material was lower compared with healthy vessels and this effect was prevented by statin therapy. Interestingly, the protective effects of statins were mediated in part through MANTIS, which was required to facilitate the atorvastatin-induced changes in endothelial gene expression. Moreover, the beneficial endothelial effects of statins in culture models (spheroid outgrowth, proliferation, telomerase activity, and vascular organ culture) were lost upon knockdown of MANTIS. CONCLUSION MANTIS is tightly regulated by the transcription factors KLF2 and KLF4 and limits the ICAM-1 mediated monocyte adhesion to endothelial cells and thus potentially atherosclerosis development in humans. The beneficial effects of statin treatment and laminar flow are dependent on MANTIS.
Collapse
Affiliation(s)
- Matthias S Leisegang
- Institute for Cardiovascular Physiology, Goethe University, Theodor-Stern-Kai 7, Frankfurt, Germany.,German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Theodor Stern-Kai 7, Frankfurt, Germany
| | - Sofia-Iris Bibli
- German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Theodor Stern-Kai 7, Frankfurt, Germany.,Institute for Vascular Signalling, Goethe University, Theodor Stern-Kai 7, Frankfurt, Germany
| | - Stefan Günther
- Max-Planck-Institute for Heart and Lung Research, ECCPS Bioinformatics and Sequencing Facility, Ludwigstr. 43, Bad Nauheim, Germany
| | - Beatrice Pflüger-Müller
- Institute for Cardiovascular Physiology, Goethe University, Theodor-Stern-Kai 7, Frankfurt, Germany.,German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Theodor Stern-Kai 7, Frankfurt, Germany
| | - James A Oo
- Institute for Cardiovascular Physiology, Goethe University, Theodor-Stern-Kai 7, Frankfurt, Germany.,German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Theodor Stern-Kai 7, Frankfurt, Germany
| | - Cindy Höper
- Institute for Cardiovascular Physiology, Goethe University, Theodor-Stern-Kai 7, Frankfurt, Germany.,German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Theodor Stern-Kai 7, Frankfurt, Germany
| | - Sandra Seredinski
- Institute for Cardiovascular Physiology, Goethe University, Theodor-Stern-Kai 7, Frankfurt, Germany.,German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Theodor Stern-Kai 7, Frankfurt, Germany
| | - Michail Yekelchyk
- Max-Planck-Institute for Heart and Lung Research, ECCPS Bioinformatics and Sequencing Facility, Ludwigstr. 43, Bad Nauheim, Germany
| | - Thomas Schmitz-Rixen
- Department of Vascular and Endovascular Surgery, Goethe University, Theodor-Stern-Kai 7, Frankfurt, Germany
| | - Christoph Schürmann
- Institute for Cardiovascular Physiology, Goethe University, Theodor-Stern-Kai 7, Frankfurt, Germany.,German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Theodor Stern-Kai 7, Frankfurt, Germany
| | - Jiong Hu
- German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Theodor Stern-Kai 7, Frankfurt, Germany.,Institute for Vascular Signalling, Goethe University, Theodor Stern-Kai 7, Frankfurt, Germany
| | - Mario Looso
- Max-Planck-Institute for Heart and Lung Research, ECCPS Bioinformatics and Sequencing Facility, Ludwigstr. 43, Bad Nauheim, Germany
| | - Fragiska Sigala
- 1st Department of Propaedeutic Surgery, University of Athens Medical School, Hippocration Hospital, Etheros 7-9, Athens, Greece
| | - Reinier A Boon
- German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Theodor Stern-Kai 7, Frankfurt, Germany.,Institute of Cardiovascular Regeneration, Goethe University, Frankfurt, Theodor-Stern-Kai 7, Germany.,Department of Physiology, VU University Medical Center, De Boelelaan 1118, HV Amsterdam, the Netherlands
| | - Ingrid Fleming
- German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Theodor Stern-Kai 7, Frankfurt, Germany.,Institute for Vascular Signalling, Goethe University, Theodor Stern-Kai 7, Frankfurt, Germany
| | - Ralf P Brandes
- Institute for Cardiovascular Physiology, Goethe University, Theodor-Stern-Kai 7, Frankfurt, Germany.,German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Theodor Stern-Kai 7, Frankfurt, Germany
| |
Collapse
|
17
|
Tu J, Liu X, Jia H, Reilly J, Yu S, Cai C, Liu F, Lv Y, Huang Y, Lu Z, Han S, Jiang T, Shu X, Wu X, Tang Z, Lu Q, Liu M. The chromatin remodeler Brg1 is required for formation and maintenance of hematopoietic stem cells. FASEB J 2020; 34:11997-12008. [PMID: 32738093 DOI: 10.1096/fj.201903168rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 06/21/2020] [Accepted: 06/24/2020] [Indexed: 11/11/2022]
Abstract
Hematopoietic stem and progenitor cells (HSPCs) have the ability to self-renew and differentiate into various blood cells, thus playing an important role in maintenance of lifelong hematopoiesis. Brahma-related gene 1 (BRG1), which acts as the ATP subunit of mammalian SWI-SNF-related chromatin remodeling complexes, is involved in human acute myeloid leukemia and highly expresses in short-term HSPCs. But its role and regulatory mechanism for HSPC development have not yet been well established. Here, we generated a brg1 knockout zebrafish model using TALEN technology. We found that in brg1-/- embryo, the primitive hematopoiesis remained well, while definitive hematopoiesis formation was significantly impaired. The number of hemogenic endothelial cells was decreased, further affecting definitive hematopoiesis with reduced myeloid and lymphoid cells. During embryogenesis, the nitric oxide (NO) microenvironment in brg1-/- embryo was seriously damaged and the reduction of HSPCs could be partially rescued by a NO donor. Chromatin immunoprecipitation (ChIP) assays showed that BRG1 could bind to the promoter of KLF2 and trigger its transcriptional activity of NO synthase. Our findings show that Brg1 promotes klf2a expression in hemogenic endothelium and highlight a novel mechanism for HSPC formation and maintenance.
Collapse
Affiliation(s)
- Jiayi Tu
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, PR China
| | - Xiliang Liu
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, PR China
| | - Haibo Jia
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, PR China
| | - James Reilly
- Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Glasgow, Scotland
| | - Shanshan Yu
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, PR China
| | - Chen Cai
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, PR China
| | - Fei Liu
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, PR China
| | - Yuexia Lv
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, PR China
| | - Yuwen Huang
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, PR China
| | - Zhaojing Lu
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, PR China
| | - Shanshan Han
- Medical College, China Three Gorges University, Yichang, China
| | - Tao Jiang
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, PR China
| | - Xinhua Shu
- Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Glasgow, Scotland
| | - Xiaoyan Wu
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhaohui Tang
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, PR China
| | - Qunwei Lu
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, PR China
| | - Mugen Liu
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, PR China
| |
Collapse
|
18
|
Li Z, Lv F, Dai C, Wang Q, Jiang C, Fang M, Xu Y. Activation of Galectin-3 (LGALS3) Transcription by Injurious Stimuli in the Liver Is Commonly Mediated by BRG1. Front Cell Dev Biol 2019; 7:310. [PMID: 31850346 PMCID: PMC6901944 DOI: 10.3389/fcell.2019.00310] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 11/13/2019] [Indexed: 01/13/2023] Open
Abstract
Galectin-3 (encoded by LGALS3) is a glycan-binding protein that regulates a diverse range of pathophysiological processes contributing to the pathogenesis of human diseases. Previous studies have found that galectin-3 levels are up-regulated in the liver by a host of different injurious stimuli. The underlying epigenetic mechanism, however, is unclear. Here we report that conditional knockout of Brahma related gene (BRG1), a chromatin remodeling protein, in hepatocytes attenuated induction of galectin-3 expression in several different animal models of liver injury. Similarly, BRG1 depletion or pharmaceutical inhibition in cultured hepatocytes suppressed the induction of galectin-3 expression by treatment with LPS plus free fatty acid (palmitate). Further analysis revealed that BRG1 interacted with AP-1 to bind to the proximal galectin-3 promoter and activate transcription. Mechanistically, DNA demethylation surrounding the galectin-3 promoter appeared to be a rate-limiting step in BRG1-mediated activation of galectin-3 transcription. BRG1 recruited the DNA 5-methylcytosine dioxygenase TET1 to the galectin-3 to promote active DNA demethylation thereby activating galectin-3 transcription. Finally, TET1 silencing abrogated induction of galectin-3 expression by LPS plus palmitate in cultured hepatocytes. In conclusion, our data unveil a novel epigenetic pathway that contributes to injury-associated activation of galectin-3 transcription in hepatocytes.
Collapse
Affiliation(s)
- Zilong Li
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Fangqiao Lv
- Department of Cell Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Congxin Dai
- Department of Neurosurgery, Peking Union Medical College Hospital, Beijing, China
| | - Qiong Wang
- Department of Surgical Oncology, the Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Chao Jiang
- Department of Surgical Oncology, the Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Mingming Fang
- Department of Clinical Medicine, Laboratory Center for Basic Medical Sciences, Jiangsu Health Vocational College, Nanjing, China.,Institute of Biomedical Research, Liaocheng University, Liaocheng, China
| | - Yong Xu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China.,Institute of Biomedical Research, Liaocheng University, Liaocheng, China
| |
Collapse
|
19
|
Hildebrandt MR, Wang Y, Li L, Yasmin L, Glubrecht DD, Godbout R. Cytoplasmic aggregation of DDX1 in developing embryos: Early embryonic lethality associated with Ddx1 knockout. Dev Biol 2019; 455:420-433. [DOI: 10.1016/j.ydbio.2019.07.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 07/04/2019] [Accepted: 07/19/2019] [Indexed: 01/12/2023]
|
20
|
Li Z, Chen B, Dong W, Kong M, Shao Y, Fan Z, Yu L, Wu D, Lu J, Guo J, Xu Y. The Chromatin Remodeler Brg1 Integrates ROS Production and Endothelial-Mesenchymal Transition to Promote Liver Fibrosis in Mice. Front Cell Dev Biol 2019; 7:245. [PMID: 31750301 PMCID: PMC6842935 DOI: 10.3389/fcell.2019.00245] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 10/07/2019] [Indexed: 12/23/2022] Open
Abstract
Trans-differentiation of endothelial cells to myofibroblast contributes to liver fibrosis. Reactive oxygen species (ROS) plays a key role in endothelial-mesenchymal transition (EndMT) although the underlying epigenetic mechanism is unclear. Here we report that endothelial conditional knockout of Brg1, a chromatin remodeling protein, attenuated liver fibrosis in mice. Brg1 deficiency in endothelial cells was paralleled by a decrease in ROS production and blockade of EndMT both in vivo and in vitro. The ability of BRG1 to regulate ROS production and EndMT was abolished by NOX4 depletion or inhibition. Further analysis revealed that BRG1 interacted with SMAD3 and AP-1 to mediate TGF-β induced NOX4 transcription in endothelial cells. Mechanistically, BRG1 recruited various histone modifying enzymes to alter the chromatin structure surrounding the NOX4 locus thereby activating its transcription. In conclusion, our data uncover a novel epigenetic mechanism that links NOX4-dependent ROS production to EndMT and liver fibrosis. Targeting the BRG1-NOX4 axis may yield novel therapeutics against liver fibrosis.
Collapse
Affiliation(s)
- Zilong Li
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China.,Institute of Biomedical Research, Liaocheng University, Liaocheng, China
| | - Baoyu Chen
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Wenhui Dong
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Ming Kong
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Yang Shao
- Cardiovascular Disease and Research Institute, Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Zhiwen Fan
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Liming Yu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Dongmei Wu
- Key Laboratory of Biotechnology on Medical Plants of Jiangsu Province and School of Life Sciences, Jiangsu Normal University, Xuzhou, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, China
| | - Jun Lu
- Key Laboratory of Biotechnology on Medical Plants of Jiangsu Province and School of Life Sciences, Jiangsu Normal University, Xuzhou, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, China
| | - Junli Guo
- Cardiovascular Disease and Research Institute, Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Yong Xu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China.,Institute of Biomedical Research, Liaocheng University, Liaocheng, China
| |
Collapse
|
21
|
Abstract
Supplemental Digital Content is available in the text. If unifying principles could be revealed for how the same genome encodes different eukaryotic cells and for how genetic variability and environmental input are integrated to impact cardiovascular health, grand challenges in basic cell biology and translational medicine may succumb to experimental dissection. A rich body of work in model systems has implicated chromatin-modifying enzymes, DNA methylation, noncoding RNAs, and other transcriptome-shaping factors in adult health and in the development, progression, and mitigation of cardiovascular disease. Meanwhile, deployment of epigenomic tools, powered by next-generation sequencing technologies in cardiovascular models and human populations, has enabled description of epigenomic landscapes underpinning cellular function in the cardiovascular system. This essay aims to unpack the conceptual framework in which epigenomes are studied and to stimulate discussion on how principles of chromatin function may inform investigations of cardiovascular disease and the development of new therapies.
Collapse
Affiliation(s)
- Manuel Rosa-Garrido
- From the Departments of Anesthesiology, Medicine, and Physiology, David Geffen School of Medicine, University of California, Los Angeles
| | - Douglas J Chapski
- From the Departments of Anesthesiology, Medicine, and Physiology, David Geffen School of Medicine, University of California, Los Angeles
| | - Thomas M Vondriska
- From the Departments of Anesthesiology, Medicine, and Physiology, David Geffen School of Medicine, University of California, Los Angeles.
| |
Collapse
|
22
|
Crosswhite PL. ATP-dependent chromatin remodeling complexes in embryonic vascular development and hypertension. Am J Physiol Heart Circ Physiol 2019; 317:H575-H580. [PMID: 31398060 DOI: 10.1152/ajpheart.00147.2019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hypertension, a chronic elevation in blood pressure, is the largest single contributing factor to mortality worldwide and the most common preventable risk factor for cardiovascular disease. High blood pressure increases the risk for someone to experience a number of adverse cardiovascular events including heart failure, stroke, or aneurysm. Despite advancements in understanding factors that contribute to hypertension, the etiology remains elusive and there remains a critical need to develop innovative study approaches to develop more effective therapeutics. ATP-dependent chromatin remodelers are dynamic regulators of DNA-histone bonds and thus gene expression. The goal of this review is to highlight and summarize reports of ATP-dependent chromatin remodelers contribution to the development or maintenance of hypertension. Emerging evidence from hypertensive animal models suggests that induction of chromatin remodeler activity increases proinflammatory genes and increases blood pressure, whereas human studies demonstrate how chromatin remodelers may act as stress-response sensors to harmful physiological stimuli. Importantly, genomic studies have linked patients with hypertension to mutations in chromatin remodeler genes. Collectively, evidence linking chromatin remodelers and hypertension warrants additional research and ultimately could reveal novel therapeutic approaches for treating this complex and devastating disease.
Collapse
|
23
|
Yang Y, Liu L, Li M, Cheng X, Fang M, Zeng Q, Xu Y. The chromatin remodeling protein BRG1 links ELOVL3 trans-activation to prostate cancer metastasis. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2019; 1862:834-845. [PMID: 31154107 DOI: 10.1016/j.bbagrm.2019.05.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/20/2019] [Accepted: 05/25/2019] [Indexed: 10/26/2022]
Abstract
Prostate cancer malignancies are intimately correlated with deregulated fatty acid metabolism. The underlying epigenetic mechanism is not fully understood. In the present study we investigated the mechanism whereby the chromatin remodeling protein BRG1 regulates the transcription of long-chain fatty acid elongase 3 (Elovl3) in prostate cancer cells. We report that in response to pro-metastatic cues (androgen and TGF-β) BRG1 expression was up-regulated along with Elvol3 in prostate cancer cells. BRG1 over-expression potentiated whereas BRG1 knockdown attenuated prostate cancer cell migration and invasion. Coincidently, Elovl3 was up-regulated following BRG1 over-expression and down-regulated after BRG1 knockdown in prostate cancer cells. Further analysis revealed that BRG1 interacted with and was recruited by retinoic acid receptor-related orphan receptor (RORγ) to the Elovl3 promoter to activate transcription. Chromatin immunoprecipitation (ChIP) profiling demonstrated that BRG1 interacted with histone acetyltransferase p300 to activate Elovl3 transcription. Depletion of p300 by siRNA or inhibition of p300 by curcumin attenuated Elovl3 trans-activation in prostate cancer cells. Together, our data identify a novel epigenetic pathway that links Elovl3 transcription to prostate cancer cell migration and invasion.
Collapse
Affiliation(s)
- Yuyu Yang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China; Institute of Biomedical Research, Liaocheng University, Liaocheng, China
| | - Li Liu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Min Li
- Center for Male Reproductive Medicine, Department of Clinical Medicine, Jiangsu Health Vocational College, Nanjing, China
| | - Xian Cheng
- Jiangsu Institute of Nuclear Medicine, Wuxi, China
| | - Mingming Fang
- Center for Male Reproductive Medicine, Department of Clinical Medicine, Jiangsu Health Vocational College, Nanjing, China
| | - Qingqi Zeng
- Center for Male Reproductive Medicine, Department of Clinical Medicine, Jiangsu Health Vocational College, Nanjing, China.
| | - Yong Xu
- Institute of Biomedical Research, Liaocheng University, Liaocheng, China; Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
24
|
Deletion of Brg1 causes stereocilia bundle fusion and cuticular plate loss in vestibular hair cells. Hear Res 2019; 377:247-259. [PMID: 31003036 DOI: 10.1016/j.heares.2019.04.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 03/16/2019] [Accepted: 04/03/2019] [Indexed: 11/22/2022]
Abstract
Brg1 is an ATPase subunit of the SWI/SNF chromatin-remodeling complex, and it is indispensable for the development and homeostasis of various organs. Conditional deletion of Brg1 in cochlea hair cells (HCs) leads to multiple structural defects and profound deafness. However, the premature death of Brg1-deficient cochlea HCs hindered further study of the role of Brg1. In contrast to cochlea HCs, Brg1-deficient vestibular HCs survived for a long time. Therefore, HC apical structure and vestibular function were examined in inner HC-specific conditional Brg1 knockout mice. Vestibular HCs exhibited fused and elongated stereocilia bundles after deletion of Brg1, and the cuticular plate was absent in most HCs with fused stereocilia bundles. HC loss was observed in conditional Brg1 knockout mice at the age of 12 months. Morphological defects and HC loss were primarily restricted in the striolar region of the utricle and saccule and in the central region of ampulla. The behavioral tests revealed that Brg1 deletion in HCs caused vestibular dysfunction in older adult mice. These results suggest that Brg1 may play specific roles in the maintenance of the HC stereocilia bundle and the cuticular plate.
Collapse
|
25
|
Braadland PR, Urbanucci A. Chromatin reprogramming as an adaptation mechanism in advanced prostate cancer. Endocr Relat Cancer 2019; 26:R211-R235. [PMID: 30844748 DOI: 10.1530/erc-18-0579] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 02/15/2019] [Indexed: 12/13/2022]
Abstract
Tumor evolution is based on the ability to constantly mutate and activate different pathways under the selective pressure of targeted therapies. Epigenetic alterations including those of the chromatin structure are associated with tumor initiation, progression and drug resistance. Many cancers, including prostate cancer, present enlarged nuclei, and chromatin appears altered and irregular. These phenotypic changes are likely to result from epigenetic dysregulation. High-throughput sequencing applied to bulk samples and now to single cells has made it possible to study these processes in unprecedented detail. It is therefore timely to review the impact of chromatin relaxation and increased DNA accessibility on prostate cancer growth and drug resistance, and their effects on gene expression. In particular, we focus on the contribution of chromatin-associated proteins such as the bromodomain-containing proteins to chromatin relaxation. We discuss the consequence of this for androgen receptor transcriptional activity and briefly summarize wider gain-of-function effects on other oncogenic transcription factors and implications for more effective prostate cancer treatment.
Collapse
Affiliation(s)
- Peder Rustøen Braadland
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Alfonso Urbanucci
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
- Centre for Molecular Medicine Norway, Nordic European Molecular Biology Laboratory Partnership, Forskningsparken, University of Oslo, Oslo, Norway
| |
Collapse
|
26
|
Duran D, Zeng X, Jin SC, Choi J, Nelson-Williams C, Yatsula B, Gaillard J, Furey CG, Lu Q, Timberlake AT, Dong W, Sorscher MA, Loring E, Klein J, Allocco A, Hunt A, Conine S, Karimy JK, Youngblood MW, Zhang J, DiLuna ML, Matouk CC, Mane S, Tikhonova IR, Castaldi C, López-Giráldez F, Knight J, Haider S, Soban M, Alper SL, Komiyama M, Ducruet AF, Zabramski JM, Dardik A, Walcott BP, Stapleton CJ, Aagaard-Kienitz B, Rodesch G, Jackson E, Smith ER, Orbach DB, Berenstein A, Bilguvar K, Vikkula M, Gunel M, Lifton RP, Kahle KT. Mutations in Chromatin Modifier and Ephrin Signaling Genes in Vein of Galen Malformation. Neuron 2019; 101:429-443.e4. [PMID: 30578106 PMCID: PMC10292091 DOI: 10.1016/j.neuron.2018.11.041] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 10/12/2018] [Accepted: 11/20/2018] [Indexed: 02/05/2023]
Abstract
Normal vascular development includes the formation and specification of arteries, veins, and intervening capillaries. Vein of Galen malformations (VOGMs) are among the most common and severe neonatal brain arterio-venous malformations, shunting arterial blood into the brain's deep venous system through aberrant direct connections. Exome sequencing of 55 VOGM probands, including 52 parent-offspring trios, revealed enrichment of rare damaging de novo mutations in chromatin modifier genes that play essential roles in brain and vascular development. Other VOGM probands harbored rare inherited damaging mutations in Ephrin signaling genes, including a genome-wide significant mutation burden in EPHB4. Inherited mutations showed incomplete penetrance and variable expressivity, with mutation carriers often exhibiting cutaneous vascular abnormalities, suggesting a two-hit mechanism. The identified mutations collectively account for ∼30% of studied VOGM cases. These findings provide insight into disease biology and may have clinical implications for risk assessment.
Collapse
Affiliation(s)
- Daniel Duran
- Department of Neurosurgery, Yale School of Medicine, New Haven CT, USA
- These authors contributed equally
| | - Xue Zeng
- Department of Genetics, Yale School of Medicine, New Haven CT, USA
- These authors contributed equally
| | - Sheng Chih Jin
- Department of Genetics, Yale School of Medicine, New Haven CT, USA
- Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, NY, USA
- These authors contributed equally
| | - Jungmin Choi
- Department of Genetics, Yale School of Medicine, New Haven CT, USA
- Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, NY, USA
- These authors contributed equally
| | | | - Bogdan Yatsula
- Department of Surgery, Yale School of Medicine, New Haven CT, USA
| | - Jonathan Gaillard
- Department of Neurosurgery, Yale School of Medicine, New Haven CT, USA
| | | | - Qiongshi Lu
- Department of Biostatistics & Medical Informatics, University of Wisconsin-Madison, Madison WI, USA
| | | | - Weilai Dong
- Department of Genetics, Yale School of Medicine, New Haven CT, USA
| | - Michelle A. Sorscher
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York NY, USA
| | - Erin Loring
- Department of Genetics, Yale School of Medicine, New Haven CT, USA
| | - Jennifer Klein
- Department of Neurosurgery, Boston Children’s Hospital, Boston MA, USA
| | - August Allocco
- Department of Neurosurgery, Yale School of Medicine, New Haven CT, USA
| | - Ava Hunt
- Department of Neurosurgery, Yale School of Medicine, New Haven CT, USA
| | - Sierra Conine
- Department of Neurosurgery, Yale School of Medicine, New Haven CT, USA
| | - Jason K. Karimy
- Department of Neurosurgery, Yale School of Medicine, New Haven CT, USA
| | - Mark W. Youngblood
- Department of Neurosurgery, Yale School of Medicine, New Haven CT, USA
- Department of Genetics, Yale School of Medicine, New Haven CT, USA
| | - Jinwei Zhang
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratory, Exeter, UK
| | - Michael L. DiLuna
- Department of Neurosurgery, Yale School of Medicine, New Haven CT, USA
| | - Charles C. Matouk
- Department of Neurosurgery, Yale School of Medicine, New Haven CT, USA
| | | | | | | | | | - James Knight
- Yale Center for Genome Analysis, West Haven CT, USA
| | - Shozeb Haider
- University College London, School of Pharmacy, London, England
| | - Mariya Soban
- University College London, School of Pharmacy, London, England
- Department of Biochemistry, Aligarh Muslim University, Aligarh, India
| | - Seth L. Alper
- Division of Nephrology and Center for Vascular Biology Research, Beth Israel Deaconess Medical Center; and Department of Medicine, Harvard Medical School, Boston, MA USA
| | - Masaki Komiyama
- Department of Neurointervention, Osaka City General Hospital, Osaka, Japan
| | - Andrew F. Ducruet
- Department of Neurosurgery, Barrow Neurological Institute, Phoenix AZ, USA
| | | | - Alan Dardik
- Department of Surgery, Yale School of Medicine, New Haven CT, USA
| | - Brian P. Walcott
- Department of Neurological Surgery, University of Southern California, Los Angeles, CA, USA
| | - Christopher J. Stapleton
- Department of Neurological Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | | | - Georges Rodesch
- Service de Neuroradiologie Diagnostique et Thérapeutique, Hôpital Foch, Suresnes, France
| | - Eric Jackson
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Edward R. Smith
- Department of Neurointerventional Radiology, Boston Children’s Hospital, Boston MA, USA
| | - Darren B. Orbach
- Department of Neurosurgery, Boston Children’s Hospital, Boston MA, USA
- Department of Neurointerventional Radiology, Boston Children’s Hospital, Boston MA, USA
| | - Alejandro Berenstein
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York NY, USA
| | - Kaya Bilguvar
- Department of Genetics, Yale School of Medicine, New Haven CT, USA
- Yale Center for Genome Analysis, West Haven CT, USA
| | - Miikka Vikkula
- Human Molecular Genetics, de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Murat Gunel
- Department of Neurosurgery, Yale School of Medicine, New Haven CT, USA
- Department of Genetics, Yale School of Medicine, New Haven CT, USA
| | - Richard P. Lifton
- Department of Genetics, Yale School of Medicine, New Haven CT, USA
- Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, NY, USA
- Senior author
| | - Kristopher T. Kahle
- Department of Neurosurgery, Yale School of Medicine, New Haven CT, USA
- Department of Pediatrics, Yale School of Medicine, New Haven CT, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven CT, USA
- Senior author
- Lead contact
| |
Collapse
|
27
|
Gao S, Silasi-Mansat R, Behar M, Lupu F, Griffin CT. Excessive Plasmin Compromises Hepatic Sinusoidal Vascular Integrity After Acetaminophen Overdose. Hepatology 2018; 68:1991-2003. [PMID: 29729197 PMCID: PMC6204085 DOI: 10.1002/hep.30070] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 04/20/2018] [Accepted: 04/29/2018] [Indexed: 12/19/2022]
Abstract
The serine protease plasmin degrades extracellular matrix (ECM) components both directly and indirectly through activation of matrix metalloproteinases. Excessive plasmin activity and subsequent ECM degradation cause hepatic sinusoidal fragility and hemorrhage in developing embryos. We report here that excessive plasmin activity in a murine acetaminophen (APAP) overdose model likewise compromises hepatic sinusoidal vascular integrity in adult animals. We found that hepatic plasmin activity is up-regulated significantly at 6 hours after APAP overdose. This plasmin up-regulation precedes both degradation of the ECM component fibronectin around liver vasculature and bleeding from centrilobular sinusoids. Importantly, administration of the pharmacological plasmin inhibitor tranexamic acid or genetic reduction of plasminogen, the circulating zymogen of plasmin, ameliorates APAP-induced hepatic fibronectin degradation and sinusoidal bleeding. Conclusion: These studies demonstrate that reduction of plasmin stabilizes hepatic sinusoidal vascular integrity after APAP overdose. (Hepatology 2018; 00:1-13).
Collapse
Affiliation(s)
- Siqi Gao
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Robert Silasi-Mansat
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Mandi Behar
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Florea Lupu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Courtney T. Griffin
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| |
Collapse
|
28
|
Angiotensin II induced CSF1 transcription is mediated by a crosstalk between different epigenetic factors in vascular endothelial cells. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2018; 1862:1-11. [PMID: 30317027 DOI: 10.1016/j.bbagrm.2018.10.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 10/04/2018] [Accepted: 10/05/2018] [Indexed: 01/15/2023]
Abstract
Endothelium-derived colony stimulating factor (CSF1) plays a key role in a range of human pathologies. Angiotensin II (Ang II) has been documented to stimulate CSF1 transcription although the underlying epigenetic mechanism remains unclear. Here we report that induction of CSF1 transcription by Ang II in vascular endothelial cells paralleled alterations of signature histone modifications surrounding the CSF1 promoter. Specifically, ChIP assays indicated that there was a simultaneous up-regulation of both acetylated H3 and trimethylated H3K4, indicative of transcriptional activation, and down-regulation of dimethyl H3K9, implicated in transcriptional repression, surrounding the proximal CSF1 promoter. Further analysis revealed that silencing of brahma related gene 1 (BRG1), a chromatin remodeling protein, abrogated CSF1 induction by Ang II. In the meantime, BRG1 silencing erased H3 acetylation and H3K4 trimethylation and restored H3K9 dimethylation. Mechanistically, BRG1 interacted with and recruited SET1A, a histone H3K4 methyltransferase, and JMJD1A, a histone H3K9 demethylase, to the CSF1 promoter to alter chromatin structure thereby promoting CSF1 trans-activation in response to Ang II stimulation. Knockdown of either SET1A or JMJD1A blocked CSF1 induction by Ang II. Finally, we demonstrate that the crosstalk between BRG1 and histone modifying enzymes was mediated by the transcription factor AP-1. In conclusion, our data unveil a novel epigenetic mechanism whereby a BRG1-centered complex mediates transcriptional activation of CSF1 by Ang II in vascular endothelial cells.
Collapse
|
29
|
Portelli SS, Robertson EN, Malecki C, Liddy KA, Hambly BD, Jeremy RW. Epigenetic influences on genetically triggered thoracic aortic aneurysm. Biophys Rev 2018; 10:1241-1256. [PMID: 30267337 PMCID: PMC6233334 DOI: 10.1007/s12551-018-0460-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Accepted: 09/17/2018] [Indexed: 12/14/2022] Open
Abstract
Genetically triggered thoracic aortic aneurysms (TAAs) account for 30% of all TAAs and can result in early morbidity and mortality in affected individuals. Epigenetic factors are now recognised to influence the phenotype of many genetically triggered conditions and have become an area of interest because of the potential for therapeutic manipulation. Major epigenetic modulators include DNA methylation, histone modification and non-coding RNA. This review examines epigenetic modulators that have been significantly associated with genetically triggered TAAs and their potential utility for translation to clinical practice.
Collapse
Affiliation(s)
- Stefanie S Portelli
- Discipline of Pathology and Bosch Institute, The University of Sydney, Sydney, NSW, 2006, Australia.
| | - Elizabeth N Robertson
- Discipline of Pathology and Bosch Institute, The University of Sydney, Sydney, NSW, 2006, Australia
- Cardiology Department, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Cassandra Malecki
- Discipline of Pathology and Bosch Institute, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Kiersten A Liddy
- Discipline of Pathology and Bosch Institute, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Brett D Hambly
- Discipline of Pathology and Bosch Institute, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Richmond W Jeremy
- Discipline of Pathology and Bosch Institute, The University of Sydney, Sydney, NSW, 2006, Australia
- Cardiology Department, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| |
Collapse
|
30
|
Li N, Kong M, Zeng S, Xu Z, Li M, Hong W, Chu X, Sun X, Zhu M, Xu Y. The chromatin remodeling protein BRG1 regulates APAP-induced liver injury by modulating CYP3A11 transcription in hepatocyte. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3487-3495. [PMID: 30293568 DOI: 10.1016/j.bbadis.2018.08.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 07/12/2018] [Accepted: 08/01/2018] [Indexed: 12/22/2022]
Abstract
Acetaminophen (APAP) overdose represents the most frequent cause of acute liver failure. The underlying epigenetic mechanism is not fully understood. In the present study we investigated the mechanism whereby the chromatin remodeling protein brahma related gene 1 (Brg1) regulates APAP induced liver injury in mice. We report that hepatocyte-specific deletion of Brg1 attenuated APAP induced liver injury in mice as evidenced by reduced plasma ALT and AST levels, decreased liver necrosis, amelioration of GSH depletion, and prolonged survival. Brg1 regulated APAP-induced liver injury likely by stimulating the transcription of Cyp3a11, a key cytochrome enzyme involved in APAP metabolism. Immunoprecipitation coupled with DNA affinity microarray identified hepatocyte nuclear factor 4 (HNF4) as a novel binding partner for Brg1. HNF4 recruited Brg1 to the Cyp3a11 promoter and formed a complex with Brg1 to trans-activate Cyp3a11. In contrast, BRG1 deficiency attenuated HNF4 binding to the Cyp3a11 promoter and dampened Cyp3a11 transcription. Therefore, our data suggest that Brg1 might play an essential role mediating APAP induced liver injury in vivo.
Collapse
Affiliation(s)
- Nan Li
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Ming Kong
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Sheng Zeng
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Zheng Xu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Min Li
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Wenxuan Hong
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Xuehui Chu
- Department of General Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China
| | - Xitai Sun
- Department of General Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China
| | - Min Zhu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China; Department of Anatomy, Nanjing Medical University, Nanjing, China.
| | - Yong Xu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China; Institute of Biomedical Research, Liaocheng University, Liaocheng, China.
| |
Collapse
|
31
|
Li N, Kong M, Zeng S, Hao C, Li M, Li L, Xu Z, Zhu M, Xu Y. Brahma related gene 1 (Brg1) contributes to liver regeneration by epigenetically activating the Wnt/β-catenin pathway in mice. FASEB J 2018; 33:327-338. [PMID: 30001167 DOI: 10.1096/fj.201800197r] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Liver regeneration is a complicated pathophysiologic process that is regulated by a myriad of signaling pathways and transcription factors. The interaction among these pathways and factors, either cooperatively or antagonistically, may ultimately lead to recovery and restoration of liver function or permanent loss of liver function and liver failure. In the present study, we investigated the mechanism whereby the chromatin remodeling protein brahma related gene 1 (Brg1) regulates liver regeneration in mice. The Smarca4-Flox strain of mice was crossbred with the Alb-Cre strain to generate hepatocyte-specific Brg1 knockout mice. Liver injury was induced by partial hepatectomy (PHx). We report that Brg1 deletion in hepatocyte compromised liver regeneration and dampened survival after PHx in mice. Brg1 interacted with β-catenin to potentiate Wnt signaling and promote hepatocyte proliferation. Mechanistically, Brg1 recruited lysine demethylase 4 (KDM4) to activate β-catenin target genes. Our data suggest that Brg1 might play an essential role maintaining hepatic homeostasis and contributing to liver repair.-Li, N., Kong, M., Zeng, S., Hao, C., Li, M., Li, L., Xu, Z., Zhu, M., Xu, Y. Brahma related gene 1 (Brg1) contributes to liver regeneration by epigenetically activating the Wnt/β-catenin pathway in mice.
Collapse
Affiliation(s)
- Nan Li
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaboration Center for Cardiovascular Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Ming Kong
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaboration Center for Cardiovascular Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Sheng Zeng
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaboration Center for Cardiovascular Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Chenzhi Hao
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaboration Center for Cardiovascular Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Min Li
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaboration Center for Cardiovascular Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Luyang Li
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaboration Center for Cardiovascular Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Zheng Xu
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaboration Center for Cardiovascular Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Min Zhu
- Department of Anatomy, Nanjing Medical University, Nanjing, China
| | - Yong Xu
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaboration Center for Cardiovascular Translational Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
32
|
Schiattarella GG, Madonna R, Van Linthout S, Thum T, Schulz R, Ferdinandy P, Perrino C. Epigenetic modulation of vascular diseases: Assessing the evidence and exploring the opportunities. Vascul Pharmacol 2018; 107:S1537-1891(17)30468-8. [PMID: 29548901 DOI: 10.1016/j.vph.2018.02.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 01/27/2018] [Accepted: 02/22/2018] [Indexed: 02/09/2023]
Abstract
Vascular adaptations to either physiological or pathophysiological conditions commonly require gene expression modifications in the most represented cellular elements of the vessel wall, i.e. endothelial and smooth muscle cells. In addition to transcription factors, a number of mechanisms contribute to the regulation of gene expression in these cells including noncoding RNAs, histone and DNA modifications, collectively indicated as epigenetic modifications. Here, we summarize the state of art regarding the role of epigenetic changes in major vascular diseases, and discuss the potential diagnostic and therapeutic applications of epigenetic modulation in this context.
Collapse
Affiliation(s)
| | - Rosalinda Madonna
- Center for Aging Sciences and Translational Medicine - CESI-MeT, Institute of Cardiology, Department of Neurosciences, Imaging and Clinical Sciences, "G. D'Annunzio" University, Chiety, Italy; Center for Cardiovascular Biology and Atherosclerosis Research, Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Sophie Van Linthout
- Berlin-Brandenburg Center for Regenerative Therapies, Charité, University Medicine Berlin, Campus Virchow, Berlin, Germany; Charité University Medicine Berlin, Campus Rudolf Virchow, Department of Cardiology, Berlin, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Rainer Schulz
- Institute of Physiology, Justus-Liebig University of Giessen, Giessen, Germany
| | - Peter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Szeged, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Cinzia Perrino
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy.
| |
Collapse
|
33
|
Liu L, Wan X, Zhou P, Zhou X, Zhang W, Hui X, Yuan X, Ding X, Zhu R, Meng G, Xiao H, Ma F, Huang H, Song X, Zhou B, Xiong S, Zhang Y. The chromatin remodeling subunit Baf200 promotes normal hematopoiesis and inhibits leukemogenesis. J Hematol Oncol 2018; 11:27. [PMID: 29482581 PMCID: PMC5828314 DOI: 10.1186/s13045-018-0567-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 02/05/2018] [Indexed: 11/10/2022] Open
Abstract
Background Adenosine triphosphate (ATP)-dependent chromatin remodeling SWI/SNF-like BAF and PBAF complexes have been implicated in the regulation of stem cell function and cancers. Several subunits of BAF or PBAF, including BRG1, BAF53a, BAF45a, BAF180, and BAF250a, are known to be involved in hematopoiesis. Baf200, a subunit of PBAF complex, plays a pivotal role in heart morphogenesis and coronary artery angiogenesis. However, little is known on the importance of Baf200 in normal and malignant hematopoiesis. Methods Utilizing Tie2-Cre-, Vav-iCre-, and Mx1-Cre-mediated Baf200 gene deletion combined with fetal liver/bone marrow transplantation, we investigated the function of Baf200 in fetal and adult hematopoiesis. In addition, a mouse model of MLL-AF9-driven leukemogenesis was used to study the role of Baf200 in malignant hematopoiesis. We also explored the potential mechanism by using RNA-seq, RT-qPCR, cell cycle, and apoptosis assays. Results Tie2-Cre-mediated loss of Baf200 causes perinatal death due to defective erythropoiesis and impaired hematopoietic stem cell expansion in the fetal liver. Vav-iCre-mediated loss of Baf200 causes only mild anemia and enhanced extramedullary hematopoiesis. Fetal liver hematopoietic stem cells from Tie2-Cre+, Baf200f/f or Vav-iCre+, Baf200f/f embryos and bone marrow hematopoietic stem cells from Vav-iCre+, Baf200f/f mice exhibited impaired long-term reconstitution potential in vivo. A cell-autonomous requirement of Baf200 for hematopoietic stem cell function was confirmed utilizing the interferon-inducible Mx1-Cre mouse strain. Transcriptomes analysis revealed that expression of several erythropoiesis- and hematopoiesis-associated genes were regulated by Baf200. In addition, loss of Baf200 in a mouse model of MLL-AF9-driven leukemogenesis accelerates the tumor burden and shortens the host survival. Conclusion Our current studies uncover critical roles of Baf200 in both normal and malignant hematopoiesis and provide a potential therapeutic target for suppressing the progression of leukemia without interfering with normal hematopoiesis. Electronic supplementary material The online version of this article (10.1186/s13045-018-0567-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lulu Liu
- Institute of Biology and Medical Sciences, Soochow University, No. 199 Ren'ai Rd, Suzhou, China.,Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, China
| | - Xiaoling Wan
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Peipei Zhou
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xiaoyuan Zhou
- University of Chinese Academy of Sciences, Beijing, China.,CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Wei Zhang
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, China.,School of Life Sciences, Shanghai University, Shanghai, China
| | - Xinhui Hui
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, China.,School of Life Sciences, Shanghai University, Shanghai, China
| | - Xiujie Yuan
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xiaodan Ding
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Ruihong Zhu
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Guangxun Meng
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Hui Xiao
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Feng Ma
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, China
| | - He Huang
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xianmin Song
- Department of Hematology, Shanghai Jiao Tong University Affiliated Shanghai General Hospital, Shanghai, China
| | - Bin Zhou
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, China.
| | - Sidong Xiong
- Institute of Biology and Medical Sciences, Soochow University, No. 199 Ren'ai Rd, Suzhou, China.
| | - Yan Zhang
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, China. .,University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
34
|
Abstract
Animal models of erythropoiesis have been, and will continue to be, important tools for understanding molecular mechanisms underlying the development of this cell lineage and the pathophysiology associated with various human erythropoietic diseases. In this regard, the mouse is probably the most valuable animal model available to investigators. The physiology and short gestational period of mice make them ideal for studying developmental processes and modeling human diseases. These attributes, coupled with cutting-edge genetic tools such as transgenesis, gene knockouts, conditional gene knockouts, and genome editing, provide a significant resource to the research community to test a plethora of hypotheses. This review summarizes the mouse models available for studying a wide variety of erythroid-related questions, as well as the properties inherent in each one.
Collapse
|
35
|
Marathe HG, Watkins-Chow DE, Weider M, Hoffmann A, Mehta G, Trivedi A, Aras S, Basuroy T, Mehrotra A, Bennett DC, Wegner M, Pavan WJ, de la Serna IL. BRG1 interacts with SOX10 to establish the melanocyte lineage and to promote differentiation. Nucleic Acids Res 2017; 45:6442-6458. [PMID: 28431046 PMCID: PMC5499657 DOI: 10.1093/nar/gkx259] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 04/04/2017] [Indexed: 12/30/2022] Open
Abstract
Mutations in SOX10 cause neurocristopathies which display varying degrees of hypopigmentation. Using a sensitized mutagenesis screen, we identified Smarca4 as a modifier gene that exacerbates the phenotypic severity of Sox10 haplo-insufficient mice. Conditional deletion of Smarca4 in SOX10 expressing cells resulted in reduced numbers of cranial and ventral trunk melanoblasts. To define the requirement for the Smarca4 -encoded BRG1 subunit of the SWI/SNF chromatin remodeling complex, we employed in vitro models of melanocyte differentiation in which induction of melanocyte-specific gene expression is closely linked to chromatin alterations. We found that BRG1 was required for expression of Dct, Tyrp1 and Tyr, genes that are regulated by SOX10 and MITF and for chromatin remodeling at distal and proximal regulatory sites. SOX10 was found to physically interact with BRG1 in differentiating melanocytes and binding of SOX10 to the Tyrp1 distal enhancer temporally coincided with recruitment of BRG1. Our data show that SOX10 cooperates with MITF to facilitate BRG1 binding to distal enhancers of melanocyte-specific genes. Thus, BRG1 is a SOX10 co-activator, required to establish the melanocyte lineage and promote expression of genes important for melanocyte function.
Collapse
Affiliation(s)
- Himangi G Marathe
- Department of Biochemistry and Cancer Biology, University of Toledo College of Medicine and Life Sciences, 3035 Arlington Ave, Toledo, OH 43614, USA
| | - Dawn E Watkins-Chow
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892-4472, USA
| | - Matthias Weider
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Alana Hoffmann
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Gaurav Mehta
- Department of Biochemistry and Cancer Biology, University of Toledo College of Medicine and Life Sciences, 3035 Arlington Ave, Toledo, OH 43614, USA
| | - Archit Trivedi
- Department of Biochemistry and Cancer Biology, University of Toledo College of Medicine and Life Sciences, 3035 Arlington Ave, Toledo, OH 43614, USA
| | - Shweta Aras
- Department of Biochemistry and Cancer Biology, University of Toledo College of Medicine and Life Sciences, 3035 Arlington Ave, Toledo, OH 43614, USA
| | - Tupa Basuroy
- Department of Biochemistry and Cancer Biology, University of Toledo College of Medicine and Life Sciences, 3035 Arlington Ave, Toledo, OH 43614, USA
| | - Aanchal Mehrotra
- Department of Biochemistry and Cancer Biology, University of Toledo College of Medicine and Life Sciences, 3035 Arlington Ave, Toledo, OH 43614, USA
| | - Dorothy C Bennett
- Molecular and Clinical Sciences Research Institute, St George's, University of London, London SW17 0RE, UK
| | - Michael Wegner
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - William J Pavan
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892-4472, USA
| | - Ivana L de la Serna
- Department of Biochemistry and Cancer Biology, University of Toledo College of Medicine and Life Sciences, 3035 Arlington Ave, Toledo, OH 43614, USA
| |
Collapse
|
36
|
Hota SK, Bruneau BG. ATP-dependent chromatin remodeling during mammalian development. Development 2017; 143:2882-97. [PMID: 27531948 DOI: 10.1242/dev.128892] [Citation(s) in RCA: 166] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Precise gene expression ensures proper stem and progenitor cell differentiation, lineage commitment and organogenesis during mammalian development. ATP-dependent chromatin-remodeling complexes utilize the energy from ATP hydrolysis to reorganize chromatin and, hence, regulate gene expression. These complexes contain diverse subunits that together provide a multitude of functions, from early embryogenesis through cell differentiation and development into various adult tissues. Here, we review the functions of chromatin remodelers and their different subunits during mammalian development. We discuss the mechanisms by which chromatin remodelers function and highlight their specificities during mammalian cell differentiation and organogenesis.
Collapse
Affiliation(s)
- Swetansu K Hota
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, CA 94158, USA
| | - Benoit G Bruneau
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, CA 94158, USA Department of Pediatrics, University of California, San Francisco, CA 94143, USA Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| |
Collapse
|
37
|
Menendez MT, Ong EC, Shepherd BT, Muthukumar V, Silasi-Mansat R, Lupu F, Griffin CT. BRG1 (Brahma-Related Gene 1) Promotes Endothelial Mrtf Transcription to Establish Embryonic Capillary Integrity. Arterioscler Thromb Vasc Biol 2017; 37:1674-1682. [PMID: 28729363 DOI: 10.1161/atvbaha.117.309785] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 07/10/2017] [Indexed: 01/23/2023]
Abstract
OBJECTIVE The chromatin remodeling enzyme BRG1 (brahma-related gene 1) transcriptionally regulates target genes important for early blood vessel development and primitive hematopoiesis. However, because Brg1 deletion in vascular progenitor cells results in lethal anemia by embryonic day 10.5 (E10.5), roles for BRG1 in embryonic vascular development after midgestation are unknown. In this study, we sought to determine whether endothelial cell BRG1 regulates genes important for vascular development or maintenance later in embryonic development. APPROACH AND RESULTS Using mice with temporally inducible deletion of endothelial BRG1 (Brg1fl/fl;Cdh5(PAC)-CreERT2 ), we found that Brg1 excision between E9.5 and 11.5 results in capillary dilation and lethal hemorrhage by E14.5. This phenotype strongly resembles that seen when the SRF (serum response factor) transcription factor is deleted from embryonic endothelial cells. Although expression of Srf and several of its known endothelial cell target genes are downregulated in BRG1-depleted endothelial cells, we did not detect binding of BRG1 at these gene promoters, indicating that they are not direct BRG1 target genes. Instead, we found that BRG1 binds to the promoters of the SRF cofactors Mrtfa and Mrtfb (myocardin-related transcription factors A and B) in endothelial cells, and these genes are downregulated in Brg1-deficient endothelial cells. CONCLUSIONS BRG1 promotes transcription of endothelial Mrtfa and Mrtfb, which elevates expression of SRF and SRF target genes that establish embryonic capillary integrity. These data highlight a new and temporally specific role for BRG1 in embryonic vasculature and provide novel information about epigenetic regulation of Mrtf expression and SRF signaling in developing blood vessels.
Collapse
Affiliation(s)
- Matthew T Menendez
- From the Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City (M.T.M., E.-C.O., B.T.S, V.M., R.S.-M., F.L., C.T.G.); and Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City (F.L., C.T.G.)
| | - E-Ching Ong
- From the Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City (M.T.M., E.-C.O., B.T.S, V.M., R.S.-M., F.L., C.T.G.); and Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City (F.L., C.T.G.)
| | - Brian T Shepherd
- From the Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City (M.T.M., E.-C.O., B.T.S, V.M., R.S.-M., F.L., C.T.G.); and Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City (F.L., C.T.G.)
| | - Vijay Muthukumar
- From the Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City (M.T.M., E.-C.O., B.T.S, V.M., R.S.-M., F.L., C.T.G.); and Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City (F.L., C.T.G.)
| | - Robert Silasi-Mansat
- From the Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City (M.T.M., E.-C.O., B.T.S, V.M., R.S.-M., F.L., C.T.G.); and Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City (F.L., C.T.G.)
| | - Florea Lupu
- From the Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City (M.T.M., E.-C.O., B.T.S, V.M., R.S.-M., F.L., C.T.G.); and Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City (F.L., C.T.G.)
| | - Courtney T Griffin
- From the Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City (M.T.M., E.-C.O., B.T.S, V.M., R.S.-M., F.L., C.T.G.); and Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City (F.L., C.T.G.).
| |
Collapse
|
38
|
Singh AP, Foley J, Tandon A, Phadke D, Karimi Kinyamu H, Archer TK. A role for BRG1 in the regulation of genes required for development of the lymphatic system. Oncotarget 2017; 8:54925-54938. [PMID: 28903392 PMCID: PMC5589631 DOI: 10.18632/oncotarget.18976] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 06/16/2017] [Indexed: 12/02/2022] Open
Abstract
Lymphatic vasculature is an important part of the cardiovascular system with multiple functions, including regulation of the return of interstitial fluid (lymph) to the bloodstream, immune responses, and fat absorption. Consequently, lymphatic vasculature defects are involved in many pathological processes, including tumor metastasis and lymphedema. BRG1 is an important player in the developmental window when the lymphatic system is initiated. In the current study, we used tamoxifen inducible Rosa26CreERT2-BRG1floxed/floxed mice that allowed temporal analysis of the impact of BRG1 inactivation in the embryo. The BRG1floxed/floxed/Cre-TM embryos exhibited edema and hemorrhage at embryonic day-13 and began to die. BRG1 deficient embryos had abnormal lymphatic sac linings with fewer LYVE1 positive lymphatic endothelial cells. Indeed, loss of BRG1 attenuated expression of a subset of lymphatic genes in-vivo. Furthermore, BRG1 binds at the promoters of COUP-TFII and LYVE1, suggesting that BRG1 modulates expression of these genes in the developing embryos. Conversely, re-expression of BRG1 in cells lacking endogenous BRG1 resulted in induction of lymphatic gene expression in-vitro, suggesting that BRG1 was both required and sufficient for lymphatic gene expression. These studies provide important insights into intrinsic regulation of BRG1-mediated lymphatic-gene expression, and further an understanding of lymphatic gene dysregulation in lymphedema and other disease conditions.
Collapse
Affiliation(s)
- Ajeet Pratap Singh
- Chromatin and Gene Expression Section, Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA.,Present address: Cornell University, College of Veterinary Medicine, Ithaca, New York, USA
| | - Julie Foley
- Special Techniques Group, Cellular and Molecular Pathology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Arpit Tandon
- Sciome.com, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Dhiral Phadke
- Sciome.com, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - H Karimi Kinyamu
- Chromatin and Gene Expression Section, Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Trevor K Archer
- Chromatin and Gene Expression Section, Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| |
Collapse
|
39
|
Witzel M, Petersheim D, Fan Y, Bahrami E, Racek T, Rohlfs M, Puchałka J, Mertes C, Gagneur J, Ziegenhain C, Enard W, Stray-Pedersen A, Arkwright PD, Abboud MR, Pazhakh V, Lieschke GJ, Krawitz PM, Dahlhoff M, Schneider MR, Wolf E, Horny HP, Schmidt H, Schäffer AA, Klein C. Chromatin-remodeling factor SMARCD2 regulates transcriptional networks controlling differentiation of neutrophil granulocytes. Nat Genet 2017; 49:742-752. [PMID: 28369036 DOI: 10.1038/ng.3833] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 03/10/2017] [Indexed: 02/06/2023]
Abstract
We identify SMARCD2 (SWI/SNF-related, matrix-associated, actin-dependent regulator of chromatin, subfamily D, member 2), also known as BAF60b (BRG1/Brahma-associated factor 60b), as a critical regulator of myeloid differentiation in humans, mice, and zebrafish. Studying patients from three unrelated pedigrees characterized by neutropenia, specific granule deficiency, myelodysplasia with excess of blast cells, and various developmental aberrations, we identified three homozygous loss-of-function mutations in SMARCD2. Using mice and zebrafish as model systems, we showed that SMARCD2 controls early steps in the differentiation of myeloid-erythroid progenitor cells. In vitro, SMARCD2 interacts with the transcription factor CEBPɛ and controls expression of neutrophil proteins stored in specific granules. Defective expression of SMARCD2 leads to transcriptional and chromatin changes in acute myeloid leukemia (AML) human promyelocytic cells. In summary, SMARCD2 is a key factor controlling myelopoiesis and is a potential tumor suppressor in leukemia.
Collapse
Affiliation(s)
- Maximilian Witzel
- Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München, Munich, Germany.,Gene Center, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Daniel Petersheim
- Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Yanxin Fan
- Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Ehsan Bahrami
- Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Tomas Racek
- Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Meino Rohlfs
- Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Jacek Puchałka
- Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Christian Mertes
- Gene Center, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Julien Gagneur
- Gene Center, Ludwig-Maximilians-Universität München, Munich, Germany.,Department of Informatics, Technical University of Munich, Munich, Germany
| | - Christoph Ziegenhain
- Anthropology and Human Genomics, Department of Biology II, Faculty of Biology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Wolfgang Enard
- Anthropology and Human Genomics, Department of Biology II, Faculty of Biology, Ludwig-Maximilians-Universität München, Munich, Germany
| | | | - Peter D Arkwright
- Department of Paediatric Allergy and Immunology, University of Manchester, Royal Manchester Children's Hospital, Manchester, UK
| | - Miguel R Abboud
- Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Vahid Pazhakh
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Graham J Lieschke
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Peter M Krawitz
- Medical Genetics and Human Genetic, Charite University Hospital, Berlin, Germany
| | - Maik Dahlhoff
- Molecular Animal Breeding and Biotechnology, Gene Center Ludwig-Maximilians-Universität München, Munich, Germany
| | - Marlon R Schneider
- Molecular Animal Breeding and Biotechnology, Gene Center Ludwig-Maximilians-Universität München, Munich, Germany
| | - Eckhard Wolf
- Molecular Animal Breeding and Biotechnology, Gene Center Ludwig-Maximilians-Universität München, Munich, Germany
| | - Hans-Peter Horny
- Pathology Institute, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Heinrich Schmidt
- Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Alejandro A Schäffer
- National Center for Biotechnology Information, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA
| | - Christoph Klein
- Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München, Munich, Germany.,Gene Center, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
40
|
Leisegang MS, Fork C, Josipovic I, Richter FM, Preussner J, Hu J, Miller MJ, Epah J, Hofmann P, Günther S, Moll F, Valasarajan C, Heidler J, Ponomareva Y, Freiman TM, Maegdefessel L, Plate KH, Mittelbronn M, Uchida S, Künne C, Stellos K, Schermuly RT, Weissmann N, Devraj K, Wittig I, Boon RA, Dimmeler S, Pullamsetti SS, Looso M, Miller FJ, Brandes RP. Long Noncoding RNA MANTIS Facilitates Endothelial Angiogenic Function. Circulation 2017; 136:65-79. [PMID: 28351900 PMCID: PMC5491227 DOI: 10.1161/circulationaha.116.026991] [Citation(s) in RCA: 172] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 03/17/2017] [Indexed: 12/16/2022]
Abstract
Supplemental Digital Content is available in the text. Background: The angiogenic function of endothelial cells is regulated by numerous mechanisms, but the impact of long noncoding RNAs (lncRNAs) has hardly been studied. We set out to identify novel and functionally important endothelial lncRNAs. Methods: Epigenetically controlled lncRNAs in human umbilical vein endothelial cells were searched by exon-array analysis after knockdown of the histone demethylase JARID1B. Molecular mechanisms were investigated by RNA pulldown and immunoprecipitation, mass spectrometry, microarray, several knockdown approaches, CRISPR-Cas9, assay for transposase-accessible chromatin sequencing, and chromatin immunoprecipitation in human umbilical vein endothelial cells. Patient samples from lung and tumors were studied for MANTIS expression. Results: A search for epigenetically controlled endothelial lncRNAs yielded lncRNA n342419, here termed MANTIS, as the most strongly regulated lncRNA. Controlled by the histone demethylase JARID1B, MANTIS was downregulated in patients with idiopathic pulmonary arterial hypertension and in rats treated with monocrotaline, whereas it was upregulated in carotid arteries of Macaca fascicularis subjected to atherosclerosis regression diet, and in endothelial cells isolated from human glioblastoma patients. CRISPR/Cas9-mediated deletion or silencing of MANTIS with small interfering RNAs or GapmeRs inhibited angiogenic sprouting and alignment of endothelial cells in response to shear stress. Mechanistically, the nuclear-localized MANTIS lncRNA interacted with BRG1, the catalytic subunit of the switch/sucrose nonfermentable chromatin-remodeling complex. This interaction was required for nucleosome remodeling by keeping the ATPase function of BRG1 active. Thereby, the transcription of key endothelial genes such as SOX18, SMAD6, and COUP-TFII was regulated by ensuring efficient RNA polymerase II machinery binding. Conclusion: MANTIS is a differentially regulated novel lncRNA facilitating endothelial angiogenic function.
Collapse
Affiliation(s)
- Matthias S Leisegang
- From Institute for Cardiovascular Physiology (M.S.L., C.F., I.J., M.J.M., J.E., F.M., R.P.B.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (F.M.R., J.H., I.W.), Institute of Vascular Signalling (J.H.), Institute of Cardiovascular Regeneration (P.H. Y.P., S.U., K.S., R.A.B., S.D.), Department of Neurosurgery (T.M.F.), Pharmazentrum Frankfurt, Institute of General Pharmacology and Toxicology (K.D.), Goethe University, Germany; ECCPS Bioinformatics and Sequencing Facility (J.P., S.G., C.K., M.L.) and Department of Lung Development and Remodeling (C.V., S.S.P.), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany; Institute of Neurology (K.H.P., M.M., K.D.); Department of Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University Munich, Germany (L.M.); Luxembourg Centre of Neuropathology (M.M.); Laboratoire National de Santé, Dudelange, Luxembourg (M.M.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette (M.M.); NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (M.M.); Cardiovascular Innovation Institute, University of Louisville, KY (S.U.); Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany (R.T.S., N.W., S.S.P.); Department of Medicine, Duke University and Durham VA Medical Center, NC (F.J.M.); and German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (M.S.L., C.F., I.J., J.H., J.E., P.H., F.M., Y.P., K.H.P., K.S., I.W., R.A.B., S.D., R.P.B.)
| | - Christian Fork
- From Institute for Cardiovascular Physiology (M.S.L., C.F., I.J., M.J.M., J.E., F.M., R.P.B.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (F.M.R., J.H., I.W.), Institute of Vascular Signalling (J.H.), Institute of Cardiovascular Regeneration (P.H. Y.P., S.U., K.S., R.A.B., S.D.), Department of Neurosurgery (T.M.F.), Pharmazentrum Frankfurt, Institute of General Pharmacology and Toxicology (K.D.), Goethe University, Germany; ECCPS Bioinformatics and Sequencing Facility (J.P., S.G., C.K., M.L.) and Department of Lung Development and Remodeling (C.V., S.S.P.), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany; Institute of Neurology (K.H.P., M.M., K.D.); Department of Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University Munich, Germany (L.M.); Luxembourg Centre of Neuropathology (M.M.); Laboratoire National de Santé, Dudelange, Luxembourg (M.M.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette (M.M.); NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (M.M.); Cardiovascular Innovation Institute, University of Louisville, KY (S.U.); Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany (R.T.S., N.W., S.S.P.); Department of Medicine, Duke University and Durham VA Medical Center, NC (F.J.M.); and German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (M.S.L., C.F., I.J., J.H., J.E., P.H., F.M., Y.P., K.H.P., K.S., I.W., R.A.B., S.D., R.P.B.)
| | - Ivana Josipovic
- From Institute for Cardiovascular Physiology (M.S.L., C.F., I.J., M.J.M., J.E., F.M., R.P.B.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (F.M.R., J.H., I.W.), Institute of Vascular Signalling (J.H.), Institute of Cardiovascular Regeneration (P.H. Y.P., S.U., K.S., R.A.B., S.D.), Department of Neurosurgery (T.M.F.), Pharmazentrum Frankfurt, Institute of General Pharmacology and Toxicology (K.D.), Goethe University, Germany; ECCPS Bioinformatics and Sequencing Facility (J.P., S.G., C.K., M.L.) and Department of Lung Development and Remodeling (C.V., S.S.P.), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany; Institute of Neurology (K.H.P., M.M., K.D.); Department of Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University Munich, Germany (L.M.); Luxembourg Centre of Neuropathology (M.M.); Laboratoire National de Santé, Dudelange, Luxembourg (M.M.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette (M.M.); NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (M.M.); Cardiovascular Innovation Institute, University of Louisville, KY (S.U.); Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany (R.T.S., N.W., S.S.P.); Department of Medicine, Duke University and Durham VA Medical Center, NC (F.J.M.); and German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (M.S.L., C.F., I.J., J.H., J.E., P.H., F.M., Y.P., K.H.P., K.S., I.W., R.A.B., S.D., R.P.B.)
| | - Florian Martin Richter
- From Institute for Cardiovascular Physiology (M.S.L., C.F., I.J., M.J.M., J.E., F.M., R.P.B.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (F.M.R., J.H., I.W.), Institute of Vascular Signalling (J.H.), Institute of Cardiovascular Regeneration (P.H. Y.P., S.U., K.S., R.A.B., S.D.), Department of Neurosurgery (T.M.F.), Pharmazentrum Frankfurt, Institute of General Pharmacology and Toxicology (K.D.), Goethe University, Germany; ECCPS Bioinformatics and Sequencing Facility (J.P., S.G., C.K., M.L.) and Department of Lung Development and Remodeling (C.V., S.S.P.), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany; Institute of Neurology (K.H.P., M.M., K.D.); Department of Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University Munich, Germany (L.M.); Luxembourg Centre of Neuropathology (M.M.); Laboratoire National de Santé, Dudelange, Luxembourg (M.M.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette (M.M.); NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (M.M.); Cardiovascular Innovation Institute, University of Louisville, KY (S.U.); Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany (R.T.S., N.W., S.S.P.); Department of Medicine, Duke University and Durham VA Medical Center, NC (F.J.M.); and German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (M.S.L., C.F., I.J., J.H., J.E., P.H., F.M., Y.P., K.H.P., K.S., I.W., R.A.B., S.D., R.P.B.)
| | - Jens Preussner
- From Institute for Cardiovascular Physiology (M.S.L., C.F., I.J., M.J.M., J.E., F.M., R.P.B.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (F.M.R., J.H., I.W.), Institute of Vascular Signalling (J.H.), Institute of Cardiovascular Regeneration (P.H. Y.P., S.U., K.S., R.A.B., S.D.), Department of Neurosurgery (T.M.F.), Pharmazentrum Frankfurt, Institute of General Pharmacology and Toxicology (K.D.), Goethe University, Germany; ECCPS Bioinformatics and Sequencing Facility (J.P., S.G., C.K., M.L.) and Department of Lung Development and Remodeling (C.V., S.S.P.), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany; Institute of Neurology (K.H.P., M.M., K.D.); Department of Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University Munich, Germany (L.M.); Luxembourg Centre of Neuropathology (M.M.); Laboratoire National de Santé, Dudelange, Luxembourg (M.M.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette (M.M.); NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (M.M.); Cardiovascular Innovation Institute, University of Louisville, KY (S.U.); Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany (R.T.S., N.W., S.S.P.); Department of Medicine, Duke University and Durham VA Medical Center, NC (F.J.M.); and German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (M.S.L., C.F., I.J., J.H., J.E., P.H., F.M., Y.P., K.H.P., K.S., I.W., R.A.B., S.D., R.P.B.)
| | - Jiong Hu
- From Institute for Cardiovascular Physiology (M.S.L., C.F., I.J., M.J.M., J.E., F.M., R.P.B.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (F.M.R., J.H., I.W.), Institute of Vascular Signalling (J.H.), Institute of Cardiovascular Regeneration (P.H. Y.P., S.U., K.S., R.A.B., S.D.), Department of Neurosurgery (T.M.F.), Pharmazentrum Frankfurt, Institute of General Pharmacology and Toxicology (K.D.), Goethe University, Germany; ECCPS Bioinformatics and Sequencing Facility (J.P., S.G., C.K., M.L.) and Department of Lung Development and Remodeling (C.V., S.S.P.), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany; Institute of Neurology (K.H.P., M.M., K.D.); Department of Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University Munich, Germany (L.M.); Luxembourg Centre of Neuropathology (M.M.); Laboratoire National de Santé, Dudelange, Luxembourg (M.M.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette (M.M.); NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (M.M.); Cardiovascular Innovation Institute, University of Louisville, KY (S.U.); Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany (R.T.S., N.W., S.S.P.); Department of Medicine, Duke University and Durham VA Medical Center, NC (F.J.M.); and German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (M.S.L., C.F., I.J., J.H., J.E., P.H., F.M., Y.P., K.H.P., K.S., I.W., R.A.B., S.D., R.P.B.)
| | - Matthew J Miller
- From Institute for Cardiovascular Physiology (M.S.L., C.F., I.J., M.J.M., J.E., F.M., R.P.B.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (F.M.R., J.H., I.W.), Institute of Vascular Signalling (J.H.), Institute of Cardiovascular Regeneration (P.H. Y.P., S.U., K.S., R.A.B., S.D.), Department of Neurosurgery (T.M.F.), Pharmazentrum Frankfurt, Institute of General Pharmacology and Toxicology (K.D.), Goethe University, Germany; ECCPS Bioinformatics and Sequencing Facility (J.P., S.G., C.K., M.L.) and Department of Lung Development and Remodeling (C.V., S.S.P.), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany; Institute of Neurology (K.H.P., M.M., K.D.); Department of Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University Munich, Germany (L.M.); Luxembourg Centre of Neuropathology (M.M.); Laboratoire National de Santé, Dudelange, Luxembourg (M.M.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette (M.M.); NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (M.M.); Cardiovascular Innovation Institute, University of Louisville, KY (S.U.); Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany (R.T.S., N.W., S.S.P.); Department of Medicine, Duke University and Durham VA Medical Center, NC (F.J.M.); and German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (M.S.L., C.F., I.J., J.H., J.E., P.H., F.M., Y.P., K.H.P., K.S., I.W., R.A.B., S.D., R.P.B.)
| | - Jeremy Epah
- From Institute for Cardiovascular Physiology (M.S.L., C.F., I.J., M.J.M., J.E., F.M., R.P.B.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (F.M.R., J.H., I.W.), Institute of Vascular Signalling (J.H.), Institute of Cardiovascular Regeneration (P.H. Y.P., S.U., K.S., R.A.B., S.D.), Department of Neurosurgery (T.M.F.), Pharmazentrum Frankfurt, Institute of General Pharmacology and Toxicology (K.D.), Goethe University, Germany; ECCPS Bioinformatics and Sequencing Facility (J.P., S.G., C.K., M.L.) and Department of Lung Development and Remodeling (C.V., S.S.P.), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany; Institute of Neurology (K.H.P., M.M., K.D.); Department of Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University Munich, Germany (L.M.); Luxembourg Centre of Neuropathology (M.M.); Laboratoire National de Santé, Dudelange, Luxembourg (M.M.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette (M.M.); NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (M.M.); Cardiovascular Innovation Institute, University of Louisville, KY (S.U.); Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany (R.T.S., N.W., S.S.P.); Department of Medicine, Duke University and Durham VA Medical Center, NC (F.J.M.); and German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (M.S.L., C.F., I.J., J.H., J.E., P.H., F.M., Y.P., K.H.P., K.S., I.W., R.A.B., S.D., R.P.B.)
| | - Patrick Hofmann
- From Institute for Cardiovascular Physiology (M.S.L., C.F., I.J., M.J.M., J.E., F.M., R.P.B.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (F.M.R., J.H., I.W.), Institute of Vascular Signalling (J.H.), Institute of Cardiovascular Regeneration (P.H. Y.P., S.U., K.S., R.A.B., S.D.), Department of Neurosurgery (T.M.F.), Pharmazentrum Frankfurt, Institute of General Pharmacology and Toxicology (K.D.), Goethe University, Germany; ECCPS Bioinformatics and Sequencing Facility (J.P., S.G., C.K., M.L.) and Department of Lung Development and Remodeling (C.V., S.S.P.), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany; Institute of Neurology (K.H.P., M.M., K.D.); Department of Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University Munich, Germany (L.M.); Luxembourg Centre of Neuropathology (M.M.); Laboratoire National de Santé, Dudelange, Luxembourg (M.M.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette (M.M.); NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (M.M.); Cardiovascular Innovation Institute, University of Louisville, KY (S.U.); Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany (R.T.S., N.W., S.S.P.); Department of Medicine, Duke University and Durham VA Medical Center, NC (F.J.M.); and German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (M.S.L., C.F., I.J., J.H., J.E., P.H., F.M., Y.P., K.H.P., K.S., I.W., R.A.B., S.D., R.P.B.)
| | - Stefan Günther
- From Institute for Cardiovascular Physiology (M.S.L., C.F., I.J., M.J.M., J.E., F.M., R.P.B.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (F.M.R., J.H., I.W.), Institute of Vascular Signalling (J.H.), Institute of Cardiovascular Regeneration (P.H. Y.P., S.U., K.S., R.A.B., S.D.), Department of Neurosurgery (T.M.F.), Pharmazentrum Frankfurt, Institute of General Pharmacology and Toxicology (K.D.), Goethe University, Germany; ECCPS Bioinformatics and Sequencing Facility (J.P., S.G., C.K., M.L.) and Department of Lung Development and Remodeling (C.V., S.S.P.), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany; Institute of Neurology (K.H.P., M.M., K.D.); Department of Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University Munich, Germany (L.M.); Luxembourg Centre of Neuropathology (M.M.); Laboratoire National de Santé, Dudelange, Luxembourg (M.M.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette (M.M.); NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (M.M.); Cardiovascular Innovation Institute, University of Louisville, KY (S.U.); Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany (R.T.S., N.W., S.S.P.); Department of Medicine, Duke University and Durham VA Medical Center, NC (F.J.M.); and German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (M.S.L., C.F., I.J., J.H., J.E., P.H., F.M., Y.P., K.H.P., K.S., I.W., R.A.B., S.D., R.P.B.)
| | - Franziska Moll
- From Institute for Cardiovascular Physiology (M.S.L., C.F., I.J., M.J.M., J.E., F.M., R.P.B.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (F.M.R., J.H., I.W.), Institute of Vascular Signalling (J.H.), Institute of Cardiovascular Regeneration (P.H. Y.P., S.U., K.S., R.A.B., S.D.), Department of Neurosurgery (T.M.F.), Pharmazentrum Frankfurt, Institute of General Pharmacology and Toxicology (K.D.), Goethe University, Germany; ECCPS Bioinformatics and Sequencing Facility (J.P., S.G., C.K., M.L.) and Department of Lung Development and Remodeling (C.V., S.S.P.), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany; Institute of Neurology (K.H.P., M.M., K.D.); Department of Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University Munich, Germany (L.M.); Luxembourg Centre of Neuropathology (M.M.); Laboratoire National de Santé, Dudelange, Luxembourg (M.M.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette (M.M.); NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (M.M.); Cardiovascular Innovation Institute, University of Louisville, KY (S.U.); Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany (R.T.S., N.W., S.S.P.); Department of Medicine, Duke University and Durham VA Medical Center, NC (F.J.M.); and German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (M.S.L., C.F., I.J., J.H., J.E., P.H., F.M., Y.P., K.H.P., K.S., I.W., R.A.B., S.D., R.P.B.)
| | - Chanil Valasarajan
- From Institute for Cardiovascular Physiology (M.S.L., C.F., I.J., M.J.M., J.E., F.M., R.P.B.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (F.M.R., J.H., I.W.), Institute of Vascular Signalling (J.H.), Institute of Cardiovascular Regeneration (P.H. Y.P., S.U., K.S., R.A.B., S.D.), Department of Neurosurgery (T.M.F.), Pharmazentrum Frankfurt, Institute of General Pharmacology and Toxicology (K.D.), Goethe University, Germany; ECCPS Bioinformatics and Sequencing Facility (J.P., S.G., C.K., M.L.) and Department of Lung Development and Remodeling (C.V., S.S.P.), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany; Institute of Neurology (K.H.P., M.M., K.D.); Department of Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University Munich, Germany (L.M.); Luxembourg Centre of Neuropathology (M.M.); Laboratoire National de Santé, Dudelange, Luxembourg (M.M.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette (M.M.); NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (M.M.); Cardiovascular Innovation Institute, University of Louisville, KY (S.U.); Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany (R.T.S., N.W., S.S.P.); Department of Medicine, Duke University and Durham VA Medical Center, NC (F.J.M.); and German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (M.S.L., C.F., I.J., J.H., J.E., P.H., F.M., Y.P., K.H.P., K.S., I.W., R.A.B., S.D., R.P.B.)
| | - Juliana Heidler
- From Institute for Cardiovascular Physiology (M.S.L., C.F., I.J., M.J.M., J.E., F.M., R.P.B.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (F.M.R., J.H., I.W.), Institute of Vascular Signalling (J.H.), Institute of Cardiovascular Regeneration (P.H. Y.P., S.U., K.S., R.A.B., S.D.), Department of Neurosurgery (T.M.F.), Pharmazentrum Frankfurt, Institute of General Pharmacology and Toxicology (K.D.), Goethe University, Germany; ECCPS Bioinformatics and Sequencing Facility (J.P., S.G., C.K., M.L.) and Department of Lung Development and Remodeling (C.V., S.S.P.), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany; Institute of Neurology (K.H.P., M.M., K.D.); Department of Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University Munich, Germany (L.M.); Luxembourg Centre of Neuropathology (M.M.); Laboratoire National de Santé, Dudelange, Luxembourg (M.M.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette (M.M.); NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (M.M.); Cardiovascular Innovation Institute, University of Louisville, KY (S.U.); Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany (R.T.S., N.W., S.S.P.); Department of Medicine, Duke University and Durham VA Medical Center, NC (F.J.M.); and German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (M.S.L., C.F., I.J., J.H., J.E., P.H., F.M., Y.P., K.H.P., K.S., I.W., R.A.B., S.D., R.P.B.)
| | - Yuliya Ponomareva
- From Institute for Cardiovascular Physiology (M.S.L., C.F., I.J., M.J.M., J.E., F.M., R.P.B.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (F.M.R., J.H., I.W.), Institute of Vascular Signalling (J.H.), Institute of Cardiovascular Regeneration (P.H. Y.P., S.U., K.S., R.A.B., S.D.), Department of Neurosurgery (T.M.F.), Pharmazentrum Frankfurt, Institute of General Pharmacology and Toxicology (K.D.), Goethe University, Germany; ECCPS Bioinformatics and Sequencing Facility (J.P., S.G., C.K., M.L.) and Department of Lung Development and Remodeling (C.V., S.S.P.), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany; Institute of Neurology (K.H.P., M.M., K.D.); Department of Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University Munich, Germany (L.M.); Luxembourg Centre of Neuropathology (M.M.); Laboratoire National de Santé, Dudelange, Luxembourg (M.M.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette (M.M.); NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (M.M.); Cardiovascular Innovation Institute, University of Louisville, KY (S.U.); Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany (R.T.S., N.W., S.S.P.); Department of Medicine, Duke University and Durham VA Medical Center, NC (F.J.M.); and German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (M.S.L., C.F., I.J., J.H., J.E., P.H., F.M., Y.P., K.H.P., K.S., I.W., R.A.B., S.D., R.P.B.)
| | - Thomas M Freiman
- From Institute for Cardiovascular Physiology (M.S.L., C.F., I.J., M.J.M., J.E., F.M., R.P.B.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (F.M.R., J.H., I.W.), Institute of Vascular Signalling (J.H.), Institute of Cardiovascular Regeneration (P.H. Y.P., S.U., K.S., R.A.B., S.D.), Department of Neurosurgery (T.M.F.), Pharmazentrum Frankfurt, Institute of General Pharmacology and Toxicology (K.D.), Goethe University, Germany; ECCPS Bioinformatics and Sequencing Facility (J.P., S.G., C.K., M.L.) and Department of Lung Development and Remodeling (C.V., S.S.P.), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany; Institute of Neurology (K.H.P., M.M., K.D.); Department of Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University Munich, Germany (L.M.); Luxembourg Centre of Neuropathology (M.M.); Laboratoire National de Santé, Dudelange, Luxembourg (M.M.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette (M.M.); NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (M.M.); Cardiovascular Innovation Institute, University of Louisville, KY (S.U.); Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany (R.T.S., N.W., S.S.P.); Department of Medicine, Duke University and Durham VA Medical Center, NC (F.J.M.); and German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (M.S.L., C.F., I.J., J.H., J.E., P.H., F.M., Y.P., K.H.P., K.S., I.W., R.A.B., S.D., R.P.B.)
| | - Lars Maegdefessel
- From Institute for Cardiovascular Physiology (M.S.L., C.F., I.J., M.J.M., J.E., F.M., R.P.B.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (F.M.R., J.H., I.W.), Institute of Vascular Signalling (J.H.), Institute of Cardiovascular Regeneration (P.H. Y.P., S.U., K.S., R.A.B., S.D.), Department of Neurosurgery (T.M.F.), Pharmazentrum Frankfurt, Institute of General Pharmacology and Toxicology (K.D.), Goethe University, Germany; ECCPS Bioinformatics and Sequencing Facility (J.P., S.G., C.K., M.L.) and Department of Lung Development and Remodeling (C.V., S.S.P.), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany; Institute of Neurology (K.H.P., M.M., K.D.); Department of Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University Munich, Germany (L.M.); Luxembourg Centre of Neuropathology (M.M.); Laboratoire National de Santé, Dudelange, Luxembourg (M.M.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette (M.M.); NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (M.M.); Cardiovascular Innovation Institute, University of Louisville, KY (S.U.); Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany (R.T.S., N.W., S.S.P.); Department of Medicine, Duke University and Durham VA Medical Center, NC (F.J.M.); and German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (M.S.L., C.F., I.J., J.H., J.E., P.H., F.M., Y.P., K.H.P., K.S., I.W., R.A.B., S.D., R.P.B.)
| | - Karl H Plate
- From Institute for Cardiovascular Physiology (M.S.L., C.F., I.J., M.J.M., J.E., F.M., R.P.B.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (F.M.R., J.H., I.W.), Institute of Vascular Signalling (J.H.), Institute of Cardiovascular Regeneration (P.H. Y.P., S.U., K.S., R.A.B., S.D.), Department of Neurosurgery (T.M.F.), Pharmazentrum Frankfurt, Institute of General Pharmacology and Toxicology (K.D.), Goethe University, Germany; ECCPS Bioinformatics and Sequencing Facility (J.P., S.G., C.K., M.L.) and Department of Lung Development and Remodeling (C.V., S.S.P.), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany; Institute of Neurology (K.H.P., M.M., K.D.); Department of Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University Munich, Germany (L.M.); Luxembourg Centre of Neuropathology (M.M.); Laboratoire National de Santé, Dudelange, Luxembourg (M.M.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette (M.M.); NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (M.M.); Cardiovascular Innovation Institute, University of Louisville, KY (S.U.); Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany (R.T.S., N.W., S.S.P.); Department of Medicine, Duke University and Durham VA Medical Center, NC (F.J.M.); and German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (M.S.L., C.F., I.J., J.H., J.E., P.H., F.M., Y.P., K.H.P., K.S., I.W., R.A.B., S.D., R.P.B.)
| | - Michel Mittelbronn
- From Institute for Cardiovascular Physiology (M.S.L., C.F., I.J., M.J.M., J.E., F.M., R.P.B.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (F.M.R., J.H., I.W.), Institute of Vascular Signalling (J.H.), Institute of Cardiovascular Regeneration (P.H. Y.P., S.U., K.S., R.A.B., S.D.), Department of Neurosurgery (T.M.F.), Pharmazentrum Frankfurt, Institute of General Pharmacology and Toxicology (K.D.), Goethe University, Germany; ECCPS Bioinformatics and Sequencing Facility (J.P., S.G., C.K., M.L.) and Department of Lung Development and Remodeling (C.V., S.S.P.), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany; Institute of Neurology (K.H.P., M.M., K.D.); Department of Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University Munich, Germany (L.M.); Luxembourg Centre of Neuropathology (M.M.); Laboratoire National de Santé, Dudelange, Luxembourg (M.M.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette (M.M.); NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (M.M.); Cardiovascular Innovation Institute, University of Louisville, KY (S.U.); Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany (R.T.S., N.W., S.S.P.); Department of Medicine, Duke University and Durham VA Medical Center, NC (F.J.M.); and German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (M.S.L., C.F., I.J., J.H., J.E., P.H., F.M., Y.P., K.H.P., K.S., I.W., R.A.B., S.D., R.P.B.)
| | - Shizuka Uchida
- From Institute for Cardiovascular Physiology (M.S.L., C.F., I.J., M.J.M., J.E., F.M., R.P.B.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (F.M.R., J.H., I.W.), Institute of Vascular Signalling (J.H.), Institute of Cardiovascular Regeneration (P.H. Y.P., S.U., K.S., R.A.B., S.D.), Department of Neurosurgery (T.M.F.), Pharmazentrum Frankfurt, Institute of General Pharmacology and Toxicology (K.D.), Goethe University, Germany; ECCPS Bioinformatics and Sequencing Facility (J.P., S.G., C.K., M.L.) and Department of Lung Development and Remodeling (C.V., S.S.P.), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany; Institute of Neurology (K.H.P., M.M., K.D.); Department of Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University Munich, Germany (L.M.); Luxembourg Centre of Neuropathology (M.M.); Laboratoire National de Santé, Dudelange, Luxembourg (M.M.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette (M.M.); NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (M.M.); Cardiovascular Innovation Institute, University of Louisville, KY (S.U.); Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany (R.T.S., N.W., S.S.P.); Department of Medicine, Duke University and Durham VA Medical Center, NC (F.J.M.); and German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (M.S.L., C.F., I.J., J.H., J.E., P.H., F.M., Y.P., K.H.P., K.S., I.W., R.A.B., S.D., R.P.B.)
| | - Carsten Künne
- From Institute for Cardiovascular Physiology (M.S.L., C.F., I.J., M.J.M., J.E., F.M., R.P.B.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (F.M.R., J.H., I.W.), Institute of Vascular Signalling (J.H.), Institute of Cardiovascular Regeneration (P.H. Y.P., S.U., K.S., R.A.B., S.D.), Department of Neurosurgery (T.M.F.), Pharmazentrum Frankfurt, Institute of General Pharmacology and Toxicology (K.D.), Goethe University, Germany; ECCPS Bioinformatics and Sequencing Facility (J.P., S.G., C.K., M.L.) and Department of Lung Development and Remodeling (C.V., S.S.P.), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany; Institute of Neurology (K.H.P., M.M., K.D.); Department of Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University Munich, Germany (L.M.); Luxembourg Centre of Neuropathology (M.M.); Laboratoire National de Santé, Dudelange, Luxembourg (M.M.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette (M.M.); NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (M.M.); Cardiovascular Innovation Institute, University of Louisville, KY (S.U.); Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany (R.T.S., N.W., S.S.P.); Department of Medicine, Duke University and Durham VA Medical Center, NC (F.J.M.); and German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (M.S.L., C.F., I.J., J.H., J.E., P.H., F.M., Y.P., K.H.P., K.S., I.W., R.A.B., S.D., R.P.B.)
| | - Konstantinos Stellos
- From Institute for Cardiovascular Physiology (M.S.L., C.F., I.J., M.J.M., J.E., F.M., R.P.B.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (F.M.R., J.H., I.W.), Institute of Vascular Signalling (J.H.), Institute of Cardiovascular Regeneration (P.H. Y.P., S.U., K.S., R.A.B., S.D.), Department of Neurosurgery (T.M.F.), Pharmazentrum Frankfurt, Institute of General Pharmacology and Toxicology (K.D.), Goethe University, Germany; ECCPS Bioinformatics and Sequencing Facility (J.P., S.G., C.K., M.L.) and Department of Lung Development and Remodeling (C.V., S.S.P.), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany; Institute of Neurology (K.H.P., M.M., K.D.); Department of Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University Munich, Germany (L.M.); Luxembourg Centre of Neuropathology (M.M.); Laboratoire National de Santé, Dudelange, Luxembourg (M.M.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette (M.M.); NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (M.M.); Cardiovascular Innovation Institute, University of Louisville, KY (S.U.); Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany (R.T.S., N.W., S.S.P.); Department of Medicine, Duke University and Durham VA Medical Center, NC (F.J.M.); and German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (M.S.L., C.F., I.J., J.H., J.E., P.H., F.M., Y.P., K.H.P., K.S., I.W., R.A.B., S.D., R.P.B.)
| | - Ralph T Schermuly
- From Institute for Cardiovascular Physiology (M.S.L., C.F., I.J., M.J.M., J.E., F.M., R.P.B.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (F.M.R., J.H., I.W.), Institute of Vascular Signalling (J.H.), Institute of Cardiovascular Regeneration (P.H. Y.P., S.U., K.S., R.A.B., S.D.), Department of Neurosurgery (T.M.F.), Pharmazentrum Frankfurt, Institute of General Pharmacology and Toxicology (K.D.), Goethe University, Germany; ECCPS Bioinformatics and Sequencing Facility (J.P., S.G., C.K., M.L.) and Department of Lung Development and Remodeling (C.V., S.S.P.), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany; Institute of Neurology (K.H.P., M.M., K.D.); Department of Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University Munich, Germany (L.M.); Luxembourg Centre of Neuropathology (M.M.); Laboratoire National de Santé, Dudelange, Luxembourg (M.M.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette (M.M.); NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (M.M.); Cardiovascular Innovation Institute, University of Louisville, KY (S.U.); Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany (R.T.S., N.W., S.S.P.); Department of Medicine, Duke University and Durham VA Medical Center, NC (F.J.M.); and German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (M.S.L., C.F., I.J., J.H., J.E., P.H., F.M., Y.P., K.H.P., K.S., I.W., R.A.B., S.D., R.P.B.)
| | - Norbert Weissmann
- From Institute for Cardiovascular Physiology (M.S.L., C.F., I.J., M.J.M., J.E., F.M., R.P.B.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (F.M.R., J.H., I.W.), Institute of Vascular Signalling (J.H.), Institute of Cardiovascular Regeneration (P.H. Y.P., S.U., K.S., R.A.B., S.D.), Department of Neurosurgery (T.M.F.), Pharmazentrum Frankfurt, Institute of General Pharmacology and Toxicology (K.D.), Goethe University, Germany; ECCPS Bioinformatics and Sequencing Facility (J.P., S.G., C.K., M.L.) and Department of Lung Development and Remodeling (C.V., S.S.P.), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany; Institute of Neurology (K.H.P., M.M., K.D.); Department of Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University Munich, Germany (L.M.); Luxembourg Centre of Neuropathology (M.M.); Laboratoire National de Santé, Dudelange, Luxembourg (M.M.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette (M.M.); NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (M.M.); Cardiovascular Innovation Institute, University of Louisville, KY (S.U.); Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany (R.T.S., N.W., S.S.P.); Department of Medicine, Duke University and Durham VA Medical Center, NC (F.J.M.); and German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (M.S.L., C.F., I.J., J.H., J.E., P.H., F.M., Y.P., K.H.P., K.S., I.W., R.A.B., S.D., R.P.B.)
| | - Kavi Devraj
- From Institute for Cardiovascular Physiology (M.S.L., C.F., I.J., M.J.M., J.E., F.M., R.P.B.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (F.M.R., J.H., I.W.), Institute of Vascular Signalling (J.H.), Institute of Cardiovascular Regeneration (P.H. Y.P., S.U., K.S., R.A.B., S.D.), Department of Neurosurgery (T.M.F.), Pharmazentrum Frankfurt, Institute of General Pharmacology and Toxicology (K.D.), Goethe University, Germany; ECCPS Bioinformatics and Sequencing Facility (J.P., S.G., C.K., M.L.) and Department of Lung Development and Remodeling (C.V., S.S.P.), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany; Institute of Neurology (K.H.P., M.M., K.D.); Department of Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University Munich, Germany (L.M.); Luxembourg Centre of Neuropathology (M.M.); Laboratoire National de Santé, Dudelange, Luxembourg (M.M.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette (M.M.); NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (M.M.); Cardiovascular Innovation Institute, University of Louisville, KY (S.U.); Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany (R.T.S., N.W., S.S.P.); Department of Medicine, Duke University and Durham VA Medical Center, NC (F.J.M.); and German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (M.S.L., C.F., I.J., J.H., J.E., P.H., F.M., Y.P., K.H.P., K.S., I.W., R.A.B., S.D., R.P.B.)
| | - Ilka Wittig
- From Institute for Cardiovascular Physiology (M.S.L., C.F., I.J., M.J.M., J.E., F.M., R.P.B.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (F.M.R., J.H., I.W.), Institute of Vascular Signalling (J.H.), Institute of Cardiovascular Regeneration (P.H. Y.P., S.U., K.S., R.A.B., S.D.), Department of Neurosurgery (T.M.F.), Pharmazentrum Frankfurt, Institute of General Pharmacology and Toxicology (K.D.), Goethe University, Germany; ECCPS Bioinformatics and Sequencing Facility (J.P., S.G., C.K., M.L.) and Department of Lung Development and Remodeling (C.V., S.S.P.), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany; Institute of Neurology (K.H.P., M.M., K.D.); Department of Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University Munich, Germany (L.M.); Luxembourg Centre of Neuropathology (M.M.); Laboratoire National de Santé, Dudelange, Luxembourg (M.M.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette (M.M.); NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (M.M.); Cardiovascular Innovation Institute, University of Louisville, KY (S.U.); Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany (R.T.S., N.W., S.S.P.); Department of Medicine, Duke University and Durham VA Medical Center, NC (F.J.M.); and German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (M.S.L., C.F., I.J., J.H., J.E., P.H., F.M., Y.P., K.H.P., K.S., I.W., R.A.B., S.D., R.P.B.)
| | - Reinier A Boon
- From Institute for Cardiovascular Physiology (M.S.L., C.F., I.J., M.J.M., J.E., F.M., R.P.B.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (F.M.R., J.H., I.W.), Institute of Vascular Signalling (J.H.), Institute of Cardiovascular Regeneration (P.H. Y.P., S.U., K.S., R.A.B., S.D.), Department of Neurosurgery (T.M.F.), Pharmazentrum Frankfurt, Institute of General Pharmacology and Toxicology (K.D.), Goethe University, Germany; ECCPS Bioinformatics and Sequencing Facility (J.P., S.G., C.K., M.L.) and Department of Lung Development and Remodeling (C.V., S.S.P.), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany; Institute of Neurology (K.H.P., M.M., K.D.); Department of Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University Munich, Germany (L.M.); Luxembourg Centre of Neuropathology (M.M.); Laboratoire National de Santé, Dudelange, Luxembourg (M.M.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette (M.M.); NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (M.M.); Cardiovascular Innovation Institute, University of Louisville, KY (S.U.); Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany (R.T.S., N.W., S.S.P.); Department of Medicine, Duke University and Durham VA Medical Center, NC (F.J.M.); and German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (M.S.L., C.F., I.J., J.H., J.E., P.H., F.M., Y.P., K.H.P., K.S., I.W., R.A.B., S.D., R.P.B.)
| | - Stefanie Dimmeler
- From Institute for Cardiovascular Physiology (M.S.L., C.F., I.J., M.J.M., J.E., F.M., R.P.B.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (F.M.R., J.H., I.W.), Institute of Vascular Signalling (J.H.), Institute of Cardiovascular Regeneration (P.H. Y.P., S.U., K.S., R.A.B., S.D.), Department of Neurosurgery (T.M.F.), Pharmazentrum Frankfurt, Institute of General Pharmacology and Toxicology (K.D.), Goethe University, Germany; ECCPS Bioinformatics and Sequencing Facility (J.P., S.G., C.K., M.L.) and Department of Lung Development and Remodeling (C.V., S.S.P.), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany; Institute of Neurology (K.H.P., M.M., K.D.); Department of Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University Munich, Germany (L.M.); Luxembourg Centre of Neuropathology (M.M.); Laboratoire National de Santé, Dudelange, Luxembourg (M.M.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette (M.M.); NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (M.M.); Cardiovascular Innovation Institute, University of Louisville, KY (S.U.); Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany (R.T.S., N.W., S.S.P.); Department of Medicine, Duke University and Durham VA Medical Center, NC (F.J.M.); and German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (M.S.L., C.F., I.J., J.H., J.E., P.H., F.M., Y.P., K.H.P., K.S., I.W., R.A.B., S.D., R.P.B.)
| | - Soni Savai Pullamsetti
- From Institute for Cardiovascular Physiology (M.S.L., C.F., I.J., M.J.M., J.E., F.M., R.P.B.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (F.M.R., J.H., I.W.), Institute of Vascular Signalling (J.H.), Institute of Cardiovascular Regeneration (P.H. Y.P., S.U., K.S., R.A.B., S.D.), Department of Neurosurgery (T.M.F.), Pharmazentrum Frankfurt, Institute of General Pharmacology and Toxicology (K.D.), Goethe University, Germany; ECCPS Bioinformatics and Sequencing Facility (J.P., S.G., C.K., M.L.) and Department of Lung Development and Remodeling (C.V., S.S.P.), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany; Institute of Neurology (K.H.P., M.M., K.D.); Department of Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University Munich, Germany (L.M.); Luxembourg Centre of Neuropathology (M.M.); Laboratoire National de Santé, Dudelange, Luxembourg (M.M.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette (M.M.); NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (M.M.); Cardiovascular Innovation Institute, University of Louisville, KY (S.U.); Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany (R.T.S., N.W., S.S.P.); Department of Medicine, Duke University and Durham VA Medical Center, NC (F.J.M.); and German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (M.S.L., C.F., I.J., J.H., J.E., P.H., F.M., Y.P., K.H.P., K.S., I.W., R.A.B., S.D., R.P.B.)
| | - Mario Looso
- From Institute for Cardiovascular Physiology (M.S.L., C.F., I.J., M.J.M., J.E., F.M., R.P.B.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (F.M.R., J.H., I.W.), Institute of Vascular Signalling (J.H.), Institute of Cardiovascular Regeneration (P.H. Y.P., S.U., K.S., R.A.B., S.D.), Department of Neurosurgery (T.M.F.), Pharmazentrum Frankfurt, Institute of General Pharmacology and Toxicology (K.D.), Goethe University, Germany; ECCPS Bioinformatics and Sequencing Facility (J.P., S.G., C.K., M.L.) and Department of Lung Development and Remodeling (C.V., S.S.P.), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany; Institute of Neurology (K.H.P., M.M., K.D.); Department of Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University Munich, Germany (L.M.); Luxembourg Centre of Neuropathology (M.M.); Laboratoire National de Santé, Dudelange, Luxembourg (M.M.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette (M.M.); NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (M.M.); Cardiovascular Innovation Institute, University of Louisville, KY (S.U.); Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany (R.T.S., N.W., S.S.P.); Department of Medicine, Duke University and Durham VA Medical Center, NC (F.J.M.); and German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (M.S.L., C.F., I.J., J.H., J.E., P.H., F.M., Y.P., K.H.P., K.S., I.W., R.A.B., S.D., R.P.B.)
| | - Francis J Miller
- From Institute for Cardiovascular Physiology (M.S.L., C.F., I.J., M.J.M., J.E., F.M., R.P.B.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (F.M.R., J.H., I.W.), Institute of Vascular Signalling (J.H.), Institute of Cardiovascular Regeneration (P.H. Y.P., S.U., K.S., R.A.B., S.D.), Department of Neurosurgery (T.M.F.), Pharmazentrum Frankfurt, Institute of General Pharmacology and Toxicology (K.D.), Goethe University, Germany; ECCPS Bioinformatics and Sequencing Facility (J.P., S.G., C.K., M.L.) and Department of Lung Development and Remodeling (C.V., S.S.P.), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany; Institute of Neurology (K.H.P., M.M., K.D.); Department of Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University Munich, Germany (L.M.); Luxembourg Centre of Neuropathology (M.M.); Laboratoire National de Santé, Dudelange, Luxembourg (M.M.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette (M.M.); NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (M.M.); Cardiovascular Innovation Institute, University of Louisville, KY (S.U.); Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany (R.T.S., N.W., S.S.P.); Department of Medicine, Duke University and Durham VA Medical Center, NC (F.J.M.); and German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (M.S.L., C.F., I.J., J.H., J.E., P.H., F.M., Y.P., K.H.P., K.S., I.W., R.A.B., S.D., R.P.B.)
| | - Ralf P Brandes
- From Institute for Cardiovascular Physiology (M.S.L., C.F., I.J., M.J.M., J.E., F.M., R.P.B.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (F.M.R., J.H., I.W.), Institute of Vascular Signalling (J.H.), Institute of Cardiovascular Regeneration (P.H. Y.P., S.U., K.S., R.A.B., S.D.), Department of Neurosurgery (T.M.F.), Pharmazentrum Frankfurt, Institute of General Pharmacology and Toxicology (K.D.), Goethe University, Germany; ECCPS Bioinformatics and Sequencing Facility (J.P., S.G., C.K., M.L.) and Department of Lung Development and Remodeling (C.V., S.S.P.), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany; Institute of Neurology (K.H.P., M.M., K.D.); Department of Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University Munich, Germany (L.M.); Luxembourg Centre of Neuropathology (M.M.); Laboratoire National de Santé, Dudelange, Luxembourg (M.M.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette (M.M.); NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (M.M.); Cardiovascular Innovation Institute, University of Louisville, KY (S.U.); Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany (R.T.S., N.W., S.S.P.); Department of Medicine, Duke University and Durham VA Medical Center, NC (F.J.M.); and German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (M.S.L., C.F., I.J., J.H., J.E., P.H., F.M., Y.P., K.H.P., K.S., I.W., R.A.B., S.D., R.P.B.).
| |
Collapse
|
41
|
Chen Z, Li S, Subramaniam S, Shyy JYJ, Chien S. Epigenetic Regulation: A New Frontier for Biomedical Engineers. Annu Rev Biomed Eng 2017; 19:195-219. [PMID: 28301736 DOI: 10.1146/annurev-bioeng-071516-044720] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Gene expression in mammalian cells depends on the epigenetic status of the chromatin, including DNA methylation, histone modifications, promoter-enhancer interactions, and noncoding RNA-mediated regulation. The coordinated actions of these multifaceted regulations determine cell development, cell cycle regulation, cell state and fate, and the ultimate responses in health and disease. Therefore, studies of epigenetic modulations are critical for our understanding of gene regulation mechanisms at the molecular, cellular, tissue, and organ levels. The aim of this review is to provide biomedical engineers with an overview of the principles of epigenetics, methods of study, recent findings in epigenetic regulation in health and disease, and computational and sequencing tools for epigenetics analysis, with an emphasis on the cardiovascular system. This review concludes with the perspectives of the application of bioengineering to advance epigenetics and the utilization of epigenetics to translate bioengineering research into clinical medicine.
Collapse
Affiliation(s)
- Zhen Chen
- Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of Hope, Duarte, California 91016; .,Department of Medicine, University of California at San Diego, La Jolla, California 92093; ,
| | - Shuai Li
- Department of Medicine, University of California at San Diego, La Jolla, California 92093; ,
| | - Shankar Subramaniam
- Department of Bioengineering and Institute of Engineering in Medicine, University of California at San Diego, La Jolla, California 92093; ,
| | - John Y-J Shyy
- Department of Medicine, University of California at San Diego, La Jolla, California 92093; ,
| | - Shu Chien
- Department of Medicine, University of California at San Diego, La Jolla, California 92093; , .,Department of Bioengineering and Institute of Engineering in Medicine, University of California at San Diego, La Jolla, California 92093; ,
| |
Collapse
|
42
|
Willis MS, Holley DW, Wang Z, Chen X, Quintana M, Jensen BC, Tannu M, Parker J, Jeyaraj D, Jain MK, Wolfram JA, Lee HG, Bultman SJ. BRG1 and BRM function antagonistically with c-MYC in adult cardiomyocytes to regulate conduction and contractility. J Mol Cell Cardiol 2017; 105:99-109. [PMID: 28232072 DOI: 10.1016/j.yjmcc.2017.02.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 01/27/2017] [Accepted: 02/15/2017] [Indexed: 12/15/2022]
Abstract
RATIONALE The contractile dysfunction that underlies heart failure involves perturbations in multiple biological processes ranging from metabolism to electrophysiology. Yet the epigenetic mechanisms that are altered in this disease state have not been elucidated. SWI/SNF chromatin-remodeling complexes are plausible candidates based on mouse knockout studies demonstrating a combined requirement for the BRG1 and BRM catalytic subunits in adult cardiomyocytes. Brg1/Brm double mutants exhibit metabolic and mitochondrial defects and are not viable although their cause of death has not been ascertained. OBJECTIVE To determine the cause of death of Brg1/Brm double-mutant mice, to test the hypothesis that BRG1 and BRM are required for cardiac contractility, and to identify relevant downstream target genes. METHODS AND RESULTS A tamoxifen-inducible gene-targeting strategy utilizing αMHC-Cre-ERT was implemented to delete both SWI/SNF catalytic subunits in adult cardiomyocytes. Brg1/Brm double-mutant mice were monitored by echocardiography and electrocardiography, and they underwent rapidly progressive ventricular dysfunction including conduction defects and arrhythmias that culminated in heart failure and death within 3weeks. Mechanistically, BRG1/BRM repressed c-Myc expression, and enforced expression of a DOX-inducible c-MYC trangene in mouse cardiomyocytes phenocopied the ventricular conduction defects observed in Brg1/Brm double mutants. BRG1/BRM and c-MYC had opposite effects on the expression of cardiac conduction genes, and the directionality was consistent with their respective loss- and gain-of-function phenotypes. To support the clinical relevance of this mechanism, BRG1/BRM occupancy was diminished at the same target genes in human heart failure cases compared to controls, and this correlated with increased c-MYC expression and decreased CX43 and SCN5A expression. CONCLUSION BRG1/BRM and c-MYC have an antagonistic relationship regulating the expression of cardiac conduction genes that maintain contractility, which is reminiscent of their antagonistic roles as a tumor suppressor and oncogene in cancer.
Collapse
Affiliation(s)
- Monte S Willis
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA; Department of Pathology & Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA.
| | - Darcy Wood Holley
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Zhongjing Wang
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Xin Chen
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong University, 250021 Jinan, PR China
| | - Megan Quintana
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Brian C Jensen
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Manasi Tannu
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong University, 250021 Jinan, PR China
| | - Joel Parker
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Darwin Jeyaraj
- Department of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; Harrington Discovery Institute, University Hospitals Harrington Heart & Vascular Institute, Cleveland, OH 44106, USA
| | - Mukesh K Jain
- Department of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; Harrington Discovery Institute, University Hospitals Harrington Heart & Vascular Institute, Cleveland, OH 44106, USA
| | - Julie A Wolfram
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Hyoung-Gon Lee
- Department of Biology, The University of Texas at San Antonio, San Antonio, TX 78249, USA.
| | - Scott J Bultman
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
43
|
Takada Y, Fukuda A, Chiba T, Seno H. Brg1 plays an essential role in development and homeostasis of the duodenum through regulation of Notch signaling. Development 2016; 143:3532-3539. [PMID: 27510977 DOI: 10.1242/dev.141549] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 07/28/2016] [Indexed: 12/16/2022]
Abstract
Brg1, a core subunit of the SWI/SNF chromatin remodeling complex, is essential for development and homeostasis of various organs. However, the functional role of Brg1 in intestinal development and homeostasis, and the underlying molecular mechanism, remain unknown. We found that deletion of Brg1 in the mouse intestine resulted in growth impairment and early death associated with abnormal crypt-villous formation, skewed differentiation into secretory lineage cells, markedly increased apoptosis, and stem cell loss in the duodenum. Furthermore, we found that the Notch signaling pathway was dramatically downregulated in Brg1-deficient duodenum. Remarkably, overexpression of the Notch1 intercellular domain (ICD) partially reversed the prognosis of intestinal Brg1 mutant mice. Notch1 ICD overexpression rescued morphogenesis, prevented over-differentiation into secretory lineage cells, and restored apoptosis to normal levels in Brg1-deficient duodenum, although stem cell loss was not rescued. Our data demonstrate that Brg1 plays an essential role in development and homeostasis, including morphogenesis, stem cell differentiation and cell survival in the duodenum. Mechanistically, the rescue of the intestinal Brg1 mutant phenotype by overexpression of the Notch1 ICD indicates that Notch signaling is a key downstream target that mediates the effects of Brg1.
Collapse
Affiliation(s)
- Yutaka Takada
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
| | - Akihisa Fukuda
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
| | - Tsutomu Chiba
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
| | - Hiroshi Seno
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
| |
Collapse
|
44
|
Jin Y, Ren N, Li S, Fu X, Sun X, Men Y, Xu Z, Zhang J, Xie Y, Xia M, Gao J. Deletion of Brg1 causes abnormal hair cell planer polarity, hair cell anchorage, and scar formation in mouse cochlea. Sci Rep 2016; 6:27124. [PMID: 27255603 PMCID: PMC4891731 DOI: 10.1038/srep27124] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 05/12/2016] [Indexed: 12/28/2022] Open
Abstract
Hair cells (HCs) are mechanosensors that play crucial roles in perceiving sound, acceleration, and fluid motion. The precise architecture of the auditory epithelium and its repair after HC loss is indispensable to the function of organ of Corti (OC). In this study, we showed that Brg1 was highly expressed in auditory HCs. Specific deletion of Brg1 in postnatal HCs resulted in rapid HC degeneration and profound deafness in mice. Further experiments showed that cell-intrinsic polarity of HCs was abolished, docking of outer hair cells (OHCs) by Deiter’s cells (DCs) failed, and scar formation in the reticular lamina was deficient. We demonstrated that Brg1 ablation disrupted the Gαi/Insc/LGN and aPKC asymmetric distributions, without overt effects on the core planer cell polarity (PCP) pathway. We also demonstrated that Brg1-deficient HCs underwent apoptosis, and that leakage in the reticular lamina caused by deficient scar formation shifted the mode of OHC death from apoptosis to necrosis. Together, these data demonstrated a requirement for Brg1 activity in HC development and suggested a role for Brg1 in the proper cellular structure formation of HCs.
Collapse
Affiliation(s)
- Yecheng Jin
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan 250100, China
| | - Naixia Ren
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan 250100, China
| | - Shiwei Li
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan 250100, China
| | - Xiaolong Fu
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan 250100, China
| | - Xiaoyang Sun
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan 250100, China
| | - Yuqin Men
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan 250100, China
| | - Zhigang Xu
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan 250100, China
| | - Jian Zhang
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan 250100, China
| | - Yue Xie
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan 250100, China
| | - Ming Xia
- Department of Otolaryngology-Head and Neck Surgery, The Second Hospital of Shandong University, Jinan 250033, China
| | - Jiangang Gao
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan 250100, China
| |
Collapse
|
45
|
He S, Limi S, McGreal RS, Xie Q, Brennan LA, Kantorow WL, Kokavec J, Majumdar R, Hou H, Edelmann W, Liu W, Ashery-Padan R, Zavadil J, Kantorow M, Skoultchi AI, Stopka T, Cvekl A. Chromatin remodeling enzyme Snf2h regulates embryonic lens differentiation and denucleation. Development 2016; 143:1937-47. [PMID: 27246713 PMCID: PMC4920164 DOI: 10.1242/dev.135285] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 03/21/2016] [Indexed: 12/30/2022]
Abstract
Ocular lens morphogenesis is a model for investigating mechanisms of cellular differentiation, spatial and temporal gene expression control, and chromatin regulation. Brg1 (Smarca4) and Snf2h (Smarca5) are catalytic subunits of distinct ATP-dependent chromatin remodeling complexes implicated in transcriptional regulation. Previous studies have shown that Brg1 regulates both lens fiber cell differentiation and organized degradation of their nuclei (denucleation). Here, we employed a conditional Snf2h(flox) mouse model to probe the cellular and molecular mechanisms of lens formation. Depletion of Snf2h induces premature and expanded differentiation of lens precursor cells forming the lens vesicle, implicating Snf2h as a key regulator of lens vesicle polarity through spatial control of Prox1, Jag1, p27(Kip1) (Cdkn1b) and p57(Kip2) (Cdkn1c) gene expression. The abnormal Snf2h(-/-) fiber cells also retain their nuclei. RNA profiling of Snf2h(-/) (-) and Brg1(-/-) eyes revealed differences in multiple transcripts, including prominent downregulation of those encoding Hsf4 and DNase IIβ, which are implicated in the denucleation process. In summary, our data suggest that Snf2h is essential for the establishment of lens vesicle polarity, partitioning of prospective lens epithelial and fiber cell compartments, lens fiber cell differentiation, and lens fiber cell nuclear degradation.
Collapse
Grants
- R01 EY012200 NEI NIH HHS
- R01 CA079057 NCI NIH HHS
- R01 DK096266 NIDDK NIH HHS
- R01 GM116143 NIGMS NIH HHS
- R01 EY013022 NEI NIH HHS
- R01 CA076329 NCI NIH HHS
- T32 GM007491 NIGMS NIH HHS
- R56 CA079057 NCI NIH HHS
- R01 EY014237 NEI NIH HHS
- 001 World Health Organization
- R01 EY022645 NEI NIH HHS
- Grant support: R01 EY012200 (AC), EY014237 (AC), EY014237-7S1 (AC), EY013022 (MK), CA079057 (AIS), EY022645 (WL), T32 GM007491 (SL), GACR: P305/12/1033 (TS, JK), UNCE: 204021 (TS, JK), and an unrestricted grant from Research to Prevent Blindness to the Department of Ophthalmology and Visual Sciences. TS is member of the BIOCEV ? Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (CZ.1.05/1.1.00/02.0109) supported by the European Regional Development Fund. The Israel Science Foundation 610/10, the Israel Ministry of Science 36494, the Ziegler Foundation and the Binational Science Foundation (2013016) to RAP.
Collapse
Affiliation(s)
- Shuying He
- Department of Ophthalmology & Visual Sciences and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Saima Limi
- Department of Ophthalmology & Visual Sciences and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Rebecca S McGreal
- Department of Ophthalmology & Visual Sciences and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Qing Xie
- Department of Ophthalmology & Visual Sciences and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Lisa A Brennan
- Department of Biomedical Science, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Wanda Lee Kantorow
- Department of Biomedical Science, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Juraj Kokavec
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA First Faculty of Medicine, Charles University, 121 08 Prague, Czech Republic
| | - Romit Majumdar
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Harry Hou
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Winfried Edelmann
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Wei Liu
- Department of Ophthalmology & Visual Sciences and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ruth Ashery-Padan
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine Tel-Aviv University, Ramat Aviv, Tel Aviv 69978, Israel
| | - Jiri Zavadil
- Department of Pathology and NYU Center for Health Informatics and Bioinformatics, New York University Langone Medical Center, New York, NY 10016, USA Mechanisms of Carcinogenesis Section, International Agency for Research on Cancer, Lyon Cedex 08 69372, France
| | - Marc Kantorow
- Department of Biomedical Science, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Arthur I Skoultchi
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Tomas Stopka
- First Faculty of Medicine, Charles University, 121 08 Prague, Czech Republic
| | - Ales Cvekl
- Department of Ophthalmology & Visual Sciences and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
46
|
Crosswhite PL, Podsiadlowska JJ, Curtis CD, Gao S, Xia L, Srinivasan RS, Griffin CT. CHD4-regulated plasmin activation impacts lymphovenous hemostasis and hepatic vascular integrity. J Clin Invest 2016; 126:2254-66. [PMID: 27140400 DOI: 10.1172/jci84652] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 03/10/2016] [Indexed: 12/11/2022] Open
Abstract
The chromatin-remodeling enzyme CHD4 maintains vascular integrity at mid-gestation; however, it is unknown whether this enzyme contributes to later blood vessel or lymphatic vessel development. Here, we addressed this issue in mice harboring a deletion of Chd4 specifically in cells that express lymphatic vessel endothelial hyaluronan receptor 1 (LYVE1), which include lymphatic endothelial cells (LECs) and liver sinusoidal endothelial cells. Chd4 mutant embryos died before birth and exhibited severe edema, blood-filled lymphatics, and liver hemorrhage. CHD4-deficient embryos developed normal lymphovenous (LV) valves, which regulate the return of lymph to the blood circulation; however, these valves lacked the fibrin-rich thrombi that prevent blood from entering the lymphatic system. Transcripts of the urokinase plasminogen activator receptor (uPAR), which facilitates activation of the fibrin-degrading protease plasmin, were upregulated in Chd4 mutant LYVE1+ cells, and plasmin activity was elevated near the LV valves. Genetic reduction of the uPAR ligand urokinase prevented degradation of fibrin-rich thrombi at the LV valves and largely resolved the blood-filled lymphatics in Chd4 mutants. Urokinase reduction also ameliorated liver hemorrhage and prolonged embryonic survival by reducing plasmin-mediated extracellular matrix degradation around sinusoidal blood vessels. These results highlight the susceptibility of LV thrombi and liver sinusoidal vessels to plasmin-mediated damage and demonstrate the importance of CHD4 in regulating embryonic plasmin activation after mid-gestation.
Collapse
|
47
|
Matsumoto S, Banine F, Feistel K, Foster S, Xing R, Struve J, Sherman LS. Brg1 directly regulates Olig2 transcription and is required for oligodendrocyte progenitor cell specification. Dev Biol 2016; 413:173-87. [PMID: 27067865 DOI: 10.1016/j.ydbio.2016.04.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 02/28/2016] [Accepted: 04/04/2016] [Indexed: 01/10/2023]
Abstract
The Olig2 basic-helix-loop-helix transcription factor promotes oligodendrocyte specification in early neural progenitor cells (NPCs), including radial glial cells, in part by recruiting SWI/SNF chromatin remodeling complexes to the enhancers of genes involved in oligodendrocyte differentiation. How Olig2 expression is regulated during oligodendrogliogenesis is not clear. Here, we find that the Brg1 subunit of SWI/SNF complexes interacts with a proximal Olig2 promoter and represses Olig2 transcription in the mouse cortex at E14, when oligodendrocyte progenitors (OPCs) are not yet found in this location. Brg1 does not interact with the Olig2 promoter in the E14 ganglionic eminence, where NPCs differentiate into Olig2-positive OPCs. Consistent with these findings, Brg1-null NPCs demonstrate precocious expression of Olig2 in the cortex. However, these cells fail to differentiate into OPCs. We further find that Brg1 is necessary for neuroepithelial-to-radial glial cell transition, but not neuronal differentiation despite a reduction in expression of the pro-neural transcription factor Pax6. Collectively, these and earlier findings support a model whereby Brg1 promotes neurogenic radial glial progenitor cell specification but is dispensable for neuronal differentiation. Concurrently, Brg1 represses Olig2 expression and the specification of OPCs, but is required for OPC differentiation and oligodendrocyte maturation.
Collapse
Affiliation(s)
- Steven Matsumoto
- Integrative Biosciences Department, School of Dentistry, Oregon Health & Science University, Portland, OR 97239, USA; Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Ave., Beaverton, OR 97006, USA
| | - Fatima Banine
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Ave., Beaverton, OR 97006, USA
| | - Kerstin Feistel
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Ave., Beaverton, OR 97006, USA
| | - Scott Foster
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Ave., Beaverton, OR 97006, USA
| | - Rubing Xing
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Ave., Beaverton, OR 97006, USA
| | - Jaime Struve
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Ave., Beaverton, OR 97006, USA
| | - Larry S Sherman
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Ave., Beaverton, OR 97006, USA; Department of Cell, Developmental and Cancer Biology, School of Medicine, Oregon Health and Science University, Portland, OR 97239, USA.
| |
Collapse
|
48
|
Muscat A, Popovski D, Jayasekara WSN, Rossello FJ, Ferguson M, Marini KD, Alamgeer M, Algar EM, Downie P, Watkins DN, Cain JE, Ashley DM. Low-Dose Histone Deacetylase Inhibitor Treatment Leads to Tumor Growth Arrest and Multi-Lineage Differentiation of Malignant Rhabdoid Tumors. Clin Cancer Res 2016; 22:3560-70. [PMID: 26920892 DOI: 10.1158/1078-0432.ccr-15-2260] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 02/10/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE Malignant rhabdoid tumor (MRT) and atypical teratoid rhabdoid tumors (ATRT) are rare aggressive undifferentiated tumors primarily affecting the kidney and CNS of infants and young children. MRT are almost exclusively characterized by homozygous deletion or inactivation of the chromatin remodeling gene SMARCB1 SMARCB1 protein loss leads to direct impairment of chromatin remodeling and we have previously reported a role for this protein in histone acetylation. This provided the rationale for investigating the therapeutic potential of histone deactylase inhibitors (HDACi) in MRT. EXPERIMENTAL DESIGN Whereas previously HDACis have been used at doses and schedules that induce cytotoxicity, in the current studies we have tested the hypothesis, both in vitro and in vivo, that sustained treatment of human MRT with low-dose HDACi can lead to sustained cell growth arrest and differentiation. RESULTS Sustained low-dose panobinostat (LBH589) treatment led to changes in cellular morphology associated with a marked increase in the induction of neural, renal, and osteoblast differentiation pathways. Genome-wide transcriptional profiling highlighted differential gene expression supporting multilineage differentiation. Using mouse xenograft models, sustained low-dose LBH589 treatment caused tumor growth arrest associated with tumor calcification detectable by X-ray imaging. Histological analysis of LBH589-treated tumors revealed significant regions of ossification, confirmed by Alizarin Red staining. Immunohistochemical analysis showed increased TUJ1 and PAX2 staining suggestive of neuronal and renal differentiation, respectively. CONCLUSIONS Low-dose HDACi treatment can terminally differentiate MRT tumor cells and reduce their ability to self-renew. The use of low-dose HDACi as a novel therapeutic approach warrants further investigation. Clin Cancer Res; 22(14); 3560-70. ©2016 AACR.
Collapse
Affiliation(s)
- Andrea Muscat
- Cancer Services, Barwon Health, Geelong, Victoria, Australia. School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - Dean Popovski
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia. Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| | - W Samantha N Jayasekara
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia. Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| | - Fernando J Rossello
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia. Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Melissa Ferguson
- Cancer Services, Barwon Health, Geelong, Victoria, Australia. School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - Kieren D Marini
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia. Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| | - Muhammad Alamgeer
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia. Department of Medical Oncology, Monash Medical Centre, East Bentleigh, Victoria, Australia
| | - Elizabeth M Algar
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia. Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| | - Peter Downie
- Children's Cancer Centre, Monash Children's Hospital, Monash Health, Victoria, Australia. Department of Paediatrics, Monash University, Clayton, Victoria, Australia
| | - D Neil Watkins
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia. Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia. The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Jason E Cain
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia. Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia.
| | - David M Ashley
- Cancer Services, Barwon Health, Geelong, Victoria, Australia. School of Medicine, Deakin University, Geelong, Victoria, Australia.
| |
Collapse
|
49
|
Chandler RL, Magnuson T. The SWI/SNF BAF-A complex is essential for neural crest development. Dev Biol 2016; 411:15-24. [PMID: 26806701 DOI: 10.1016/j.ydbio.2016.01.015] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 01/17/2016] [Accepted: 01/20/2016] [Indexed: 10/22/2022]
Abstract
Growing evidence indicates that chromatin remodeler mutations underlie the pathogenesis of human neurocristopathies or disorders that affect neural crest cells (NCCs). However, causal relationships among chromatin remodeler subunit mutations and NCC defects remain poorly understood. Here we show that homozygous loss of ARID1A-containing, SWI/SNF chromatin remodeling complexes (BAF-A) in NCCs results in embryonic lethality in mice, with mutant embryos succumbing to heart defects. Strikingly, monoallelic loss of ARID1A in NCCs led to craniofacial defects in adult mice, including shortened snouts and low set ears, and these defects were more pronounced following homozygous loss of ARID1A, with the ventral cranial bones being greatly reduced in size. Early NCC specification and expression of the BRG1 NCC target gene, PLEXINA2, occurred normally in the absence of ARID1A. Nonetheless, mutant embryos displayed incomplete conotruncal septation of the cardiac outflow tract and defects in the posterior pharyngeal arteries, culminating in persistent truncus arteriosus and agenesis of the ductus arteriosus. Consistent with this, migrating cardiac NCCs underwent apoptosis within the circumpharyngeal ridge. Our data support the notion that multiple, distinct chromatin remodeling complexes govern genetically separable events in NCC development and highlight a potential pathogenic role for NCCs in the human BAF complex disorder, Coffin-Siris Syndrome.
Collapse
Affiliation(s)
- Ronald L Chandler
- Department of Genetics, University of North Carolina at Chapel Hill, United States
| | - Terry Magnuson
- Department of Genetics, University of North Carolina at Chapel Hill, United States.
| |
Collapse
|
50
|
Teichroeb JH, Kim J, Betts DH. The role of telomeres and telomerase reverse transcriptase isoforms in pluripotency induction and maintenance. RNA Biol 2016; 13:707-19. [PMID: 26786236 DOI: 10.1080/15476286.2015.1134413] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Telomeres are linear guanine-rich DNA structures at the ends of chromosomes. The length of telomeric DNA is actively regulated by a number of mechanisms in highly proliferative cells such as germ cells, cancer cells, and pluripotent stem cells. Telomeric DNA is synthesized by way of the ribonucleoprotein called telomerase containing a reverse transcriptase (TERT) subunit and RNA component (TERC). TERT is highly conserved across species and ubiquitously present in their respective pluripotent cells. Recent studies have uncovered intricate associations between telomeres and the self-renewal and differentiation properties of pluripotent stem cells. Interestingly, the past decade's work indicates that the TERT subunit also has the capacity to modulate mitochondrial function, to remodel chromatin structure, and to participate in key signaling pathways such as the Wnt/β-catenin pathway. Many of these non-canonical functions do not require TERT's catalytic activity, which hints at possible functions for the extensive number of alternatively spliced TERT isoforms that are highly expressed in pluripotent stem cells. In this review, some of the established and potential routes of pluripotency induction and maintenance are highlighted from the perspectives of telomere maintenance, known TERT isoform functions and their complex regulation.
Collapse
Affiliation(s)
- Jonathan H Teichroeb
- a Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry , The University of Western Ontario , London , Ontario , Canada
| | - Joohwan Kim
- a Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry , The University of Western Ontario , London , Ontario , Canada
| | - Dean H Betts
- a Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry , The University of Western Ontario , London , Ontario , Canada.,b Children's Health Research Institute, Lawson Health Research Institute , London , Ontario , Canada
| |
Collapse
|