1
|
Cuartero S, Stik G, Stadhouders R. Three-dimensional genome organization in immune cell fate and function. Nat Rev Immunol 2022; 23:206-221. [PMID: 36127477 DOI: 10.1038/s41577-022-00774-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2022] [Indexed: 11/09/2022]
Abstract
Immune cell development and activation demand the precise and coordinated control of transcriptional programmes. Three-dimensional (3D) organization of the genome has emerged as an important regulator of chromatin state, transcriptional activity and cell identity by facilitating or impeding long-range genomic interactions among regulatory elements and genes. Chromatin folding thus enables cell type-specific and stimulus-specific transcriptional responses to extracellular signals, which are essential for the control of immune cell fate, for inflammatory responses and for generating a diverse repertoire of antigen receptor specificities. Here, we review recent findings connecting 3D genome organization to the control of immune cell differentiation and function, and discuss how alterations in genome folding may lead to immune dysfunction and malignancy.
Collapse
Affiliation(s)
- Sergi Cuartero
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Spain. .,Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain.
| | - Grégoire Stik
- Centre for Genomic Regulation (CRG), Institute of Science and Technology (BIST), Barcelona, Spain. .,Universitat Pompeu Fabra (UPF), Barcelona, Spain.
| | - Ralph Stadhouders
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands. .,Department of Cell Biology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
2
|
Antczak M, Cañete PF, Chen Z, Belle C, Yu D. Evolution of γ chain cytokines: Mechanisms, methods and applications. Comput Struct Biotechnol J 2022; 20:4746-4755. [PMID: 36147674 PMCID: PMC9465101 DOI: 10.1016/j.csbj.2022.08.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/21/2022] [Accepted: 08/22/2022] [Indexed: 11/12/2022] Open
Abstract
The common γ chain family of cytokines and their receptors play fundamental roles in the immune system. Evolutionary studies of γ chain cytokines have elegantly illustrated how the immune system adapts to ever-changing environmental conditions. Indeed, these studies have revealed the uniqueness of cytokine evolution, which exhibits strong positive selection pressure needed to adapt to rapidly evolving threats whilst still conserving their receptor binding capabilities. In this review, we summarise the evolutionary mechanisms that gave rise to the characteristically diverse family of γ chain cytokines. We also speculate on the benefits of studying cytokine evolution, which may provide alternative ways to design novel cytokine therapeutic strategies. Additionally, we discuss current evolutionary models that elucidate the emergence of distinct cytokines (IL-4 and IL-13) and cytokine receptors (IL-2Rα and IL-15Rα). Finally, we address and reflect on the difficulties associated with evolutionary studies of rapidly evolving genes and describe a variety of computational methods that have revealed numerous aspects of cytokine evolution.
Collapse
Affiliation(s)
- Magdalena Antczak
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Pablo F. Cañete
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Zhian Chen
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Clémence Belle
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Di Yu
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
- Ian Frazer Centre for Children’s Immunotherapy Research, Child Health Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| |
Collapse
|
3
|
Choi Y, Song MJ, Jung WJ, Jeong H, Park S, Yang B, Lee EC, Joo JS, Choi D, Koo SH, Kim EK, Nam KT, Kim HP. Liver-Specific Deletion of Mouse CTCF Leads to Hepatic Steatosis via Augmented PPARγ Signaling. Cell Mol Gastroenterol Hepatol 2021; 12:1761-1787. [PMID: 34358714 PMCID: PMC8551791 DOI: 10.1016/j.jcmgh.2021.07.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS The liver is the major organ for metabolizing lipids, and malfunction of the liver leads to various diseases. Nonalcoholic fatty liver disease is rapidly becoming a major health concern worldwide and is characterized by abnormal retention of excess lipids in the liver. CCCTC-binding factor (CTCF) is a highly conserved zinc finger protein that regulates higher-order chromatin organization and is involved in various gene regulation processes. Here, we sought to determine the physiological role of CTCF in hepatic lipid metabolism. METHODS We generated liver-specific, CTCF-ablated and/or CD36 whole-body knockout mice. Overexpression or knockdown of peroxisome proliferator-activated receptor (PPAR)γ in the liver was achieved using adenovirus. Mice were examined for development of hepatic steatosis and inflammation. RNA sequencing was performed to identify genes affected by CTCF depletion. Genome-wide occupancy of H3K27 acetylation, PPARγ, and CTCF were analyzed by chromatin immunoprecipitation sequencing. Genome-wide chromatin interactions were analyzed by in situ Hi-C. RESULTS Liver-specific, CTCF-deficient mice developed hepatic steatosis and inflammation when fed a standard chow diet. Global analysis of the transcriptome and enhancer landscape revealed that CTCF-depleted liver showed enhanced accumulation of PPARγ in the nucleus, which leads to increased expression of its downstream target genes, including fat storage-related gene CD36, which is involved in the lipid metabolic process. Hepatic steatosis developed in liver-specific, CTCF-deficient mice was ameliorated by repression of PPARγ via pharmacologic blockade or adenovirus-mediated knockdown, but hardly rescued by additional knockout of CD36. CONCLUSIONS Our data indicate that liver-specific deletion of CTCF leads to hepatosteatosis through augmented PPARγ DNA-binding activity, which up-regulates its downstream target genes associated with the lipid metabolic process.
Collapse
Affiliation(s)
- Yeeun Choi
- Department of Environmental Medical Biology, Institute of Tropical Medicine, Seoul, Korea; Brain Korea 21 Plus Project for Medical Science, Seoul, Korea
| | - Min-Ji Song
- Department of Environmental Medical Biology, Institute of Tropical Medicine, Seoul, Korea
| | - Woong-Jae Jung
- Department of Environmental Medical Biology, Institute of Tropical Medicine, Seoul, Korea; Department of Bioinformatics, Graduate School of Soongsil University, Seoul, Korea
| | - Haengdueng Jeong
- Brain Korea 21 Plus Project for Medical Science, Seoul, Korea; Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Seokjae Park
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea; Neurometabolomics Research Center, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea
| | - Bobae Yang
- Department of Environmental Medical Biology, Institute of Tropical Medicine, Seoul, Korea; Brain Korea 21 Plus Project for Medical Science, Seoul, Korea
| | - Eun-Chong Lee
- Department of Environmental Medical Biology, Institute of Tropical Medicine, Seoul, Korea; Brain Korea 21 Plus Project for Medical Science, Seoul, Korea
| | - Jung-Sik Joo
- Department of Environmental Medical Biology, Institute of Tropical Medicine, Seoul, Korea; Brain Korea 21 Plus Project for Medical Science, Seoul, Korea
| | - Dahee Choi
- Division of Life Sciences, Korea University, Seoul, Korea
| | - Seung-Hoi Koo
- Division of Life Sciences, Korea University, Seoul, Korea
| | - Eun-Kyoung Kim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea; Neurometabolomics Research Center, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea
| | - Ki Taek Nam
- Brain Korea 21 Plus Project for Medical Science, Seoul, Korea; Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Hyoung-Pyo Kim
- Department of Environmental Medical Biology, Institute of Tropical Medicine, Seoul, Korea; Brain Korea 21 Plus Project for Medical Science, Seoul, Korea.
| |
Collapse
|
4
|
Scourzic L, Salataj E, Apostolou E. Deciphering the Complexity of 3D Chromatin Organization Driving Lymphopoiesis and Lymphoid Malignancies. Front Immunol 2021; 12:669881. [PMID: 34054841 PMCID: PMC8160312 DOI: 10.3389/fimmu.2021.669881] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 04/26/2021] [Indexed: 12/18/2022] Open
Abstract
Proper lymphopoiesis and immune responses depend on the spatiotemporal control of multiple processes, including gene expression, DNA recombination and cell fate decisions. High-order 3D chromatin organization is increasingly appreciated as an important regulator of these processes and dysregulation of genomic architecture has been linked to various immune disorders, including lymphoid malignancies. In this review, we present the general principles of the 3D chromatin topology and its dynamic reorganization during various steps of B and T lymphocyte development and activation. We also discuss functional interconnections between architectural, epigenetic and transcriptional changes and introduce major key players of genomic organization in B/T lymphocytes. Finally, we present how alterations in architectural factors and/or 3D genome organization are linked to dysregulation of the lymphopoietic transcriptional program and ultimately to hematological malignancies.
Collapse
Affiliation(s)
| | | | - Effie Apostolou
- Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
5
|
Iqubal A, Iqubal MK, Fazal SA, Pottoo FH, Haque SE. Nutraceuticals and their Derived Nano-formulations for the Prevention and Treatment of Alzheimer's disease. Curr Mol Pharmacol 2021; 15:23-50. [PMID: 33687906 DOI: 10.2174/1874467214666210309115605] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/22/2020] [Accepted: 12/02/2020] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease is one of the common chronic neurological disorders and associated with cognitive dysfunction, depression and progressive dementia. Presence of β-amyloid or senile plaques, hyper-phosphorylated tau proteins, neurofibrillary tangle, oxidative-nitrative stress, mitochondrial dysfunction, endoplasmic reticulum stress, neuroinflammation and derailed neurotransmitter status are the hallmark of AD. Currently, donepezil, memantine, rivastigmine and galantamine are approved by the FDA for symptomatic management. It is well-known that these approved drugs only exert symptomatic relief and possess poor patient-compliance. Additionally, various published evidence shows the neuroprotective potential of various nutraceuticals via their antioxidant, anti-inflammatory and anti-apoptotic effects in the preclinical and clinical studies. These nutraceuticals possess a significant neuroprotective potential and hence, can be a future pharmacotherapeutic for the management and treatment of AD. However, nutraceutical suffers from certain major limitations such as poor solubility, low bioavailability, low stability, fast hepatic-metabolism and larger particle size. These pharmacokinetic attributes restrict their entry into the brain via the blood-brain barrier. Therefore, to over such issues, various nanoformulation of nutraceuticals was developed, that allows their effective delivery into brain owning to reduced particle size, increased lipophilicity increased bioavailability and avoidance of fast hepatic metabolism. Thus, in this review, we have discussed the etiology of AD, focused on the pharmacotherapeutics of nutraceuticals with preclinical and clinical evidence, discussed pharmaceutical limitation and regulatory aspects of nutraceuticals to ensure safety and efficacy. We further explored the latitude of various nanoformulation of nutraceuticals as a novel approach to overcome the existing pharmaceutical limitation and for effective delivery into the brain.
Collapse
Affiliation(s)
- Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi-110062. India
| | - Mohammad Kashif Iqubal
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi-110062. India
| | - Syed Abul Fazal
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi-110062. India
| | - Faheem Hyder Pottoo
- Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal, University, P.O.BOX 1982, Damman, 31441. Saudi Arabia
| | - Syed Ehtaishamul Haque
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi-110062. India
| |
Collapse
|
6
|
Yang Q, Zhang R, Tang P, Sun Y, Johnson C, Saredy J, Wu S, Wang J, Lu Y, Saaoud F, Shao Y, Drummer C, Xu K, Yu D, Li R, Ge S, Jiang X, Wang H, Yang X. Ultrasound May Suppress Tumor Growth, Inhibit Inflammation, and Establish Tolerogenesis by Remodeling Innatome via Pathways of ROS, Immune Checkpoints, Cytokines, and Trained Immunity/Tolerance. J Immunol Res 2021; 2021:6664453. [PMID: 33628851 PMCID: PMC7889351 DOI: 10.1155/2021/6664453] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/27/2020] [Accepted: 12/16/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The immune mechanisms underlying low-intensity ultrasound- (LIUS-) mediated suppression of inflammation and tumorigenesis remain poorly determined. METHODS We used microarray datasets from the NCBI GEO DataSet repository and conducted comprehensive data-mining analyses, where we examined the gene expression of 1376 innate immune regulators (innatome genes (IGs) in cells treated with LIUS. RESULTS We made the following findings: (1) LIUS upregulates proinflammatory IGs and downregulates metastasis genes in cancer cells, and LIUS upregulates adaptive immunity pathways but inhibits danger-sensing and inflammation pathways and promote tolerogenic differentiation in bone marrow (BM) cells. (2) LIUS upregulates IGs encoded for proteins localized in the cytoplasm, extracellular space, and others, but downregulates IG proteins localized in nuclear and plasma membranes, and LIUS downregulates phosphatases. (3) LIUS-modulated IGs act partially via several important pathways of reactive oxygen species (ROS), reverse signaling of immune checkpoint receptors B7-H4 and BTNL2, inflammatory cytokines, and static or oscillatory shear stress and heat generation, among which ROS is a dominant mechanism. (4) LIUS upregulates trained immunity enzymes in lymphoma cells and downregulates trained immunity enzymes and presumably establishes trained tolerance in BM cells. (5) LIUS modulates chromatin long-range interactions to differentially regulate IGs expression in cancer cells and noncancer cells. CONCLUSIONS Our analysis suggests novel molecular mechanisms that are utilized by LIUS to induce tumor suppression and inflammation inhibition. Our findings may lead to development of new treatment protocols for cancers and chronic inflammation.
Collapse
Affiliation(s)
- Qian Yang
- Centers for Cardiovascular Research and Inflammation, Translational, & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
- Department of Ultrasonic Diagnosis and Treatment Center, XiAn International Medical Center Hospital, XiAn, China
- Heart Center, St. Christopher's Hospital for Children, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Ruijing Zhang
- Centers for Cardiovascular Research and Inflammation, Translational, & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
- Department of Nephrology, Second Hospital of Shanxi Medical University, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China
| | - Peng Tang
- Department of Orthopedics, Beijing Charity Hospital of China Rehabilitation Research Center, Beijing, China
| | - Yu Sun
- Centers for Cardiovascular Research and Inflammation, Translational, & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Candice Johnson
- Centers for Cardiovascular Research and Inflammation, Translational, & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Jason Saredy
- Metabolic Disease Research & Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Susu Wu
- Centers for Cardiovascular Research and Inflammation, Translational, & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Jiwei Wang
- Centers for Cardiovascular Research and Inflammation, Translational, & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Yifan Lu
- Centers for Cardiovascular Research and Inflammation, Translational, & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Fatma Saaoud
- Centers for Cardiovascular Research and Inflammation, Translational, & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Ying Shao
- Centers for Cardiovascular Research and Inflammation, Translational, & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Charles Drummer
- Centers for Cardiovascular Research and Inflammation, Translational, & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Keman Xu
- Centers for Cardiovascular Research and Inflammation, Translational, & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Daohai Yu
- Department of Clinical Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Rongshan Li
- Department of Nephrology, Second Hospital of Shanxi Medical University, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China
| | - Shuping Ge
- Heart Center, St. Christopher's Hospital for Children, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Xiaohua Jiang
- Centers for Cardiovascular Research and Inflammation, Translational, & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
- Metabolic Disease Research & Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Hong Wang
- Metabolic Disease Research & Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Xiaofeng Yang
- Centers for Cardiovascular Research and Inflammation, Translational, & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
- Metabolic Disease Research & Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
7
|
Yang CM, Kang MK, Jung WJ, Joo JS, Kim YJ, Choi Y, Kim HP. p53 expression confers sensitivity to 5-fluorouracil via distinct chromatin accessibility dynamics in human colorectal cancer. Oncol Lett 2021; 21:226. [PMID: 33613715 PMCID: PMC7856692 DOI: 10.3892/ol.2021.12487] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 12/03/2020] [Indexed: 12/12/2022] Open
Abstract
One of the most commonly used drugs in chemotherapy, 5-fluorouracil (5-FU) has been shown to be effective in only 10-15% of patients with colon cancer. Thus, studies of the mechanisms affecting 5-FU sensitivity in these patients are necessary. The tumor suppressor protein p53 is a transcription factor that serves important roles in cell apoptosis by regulating the cell cycle. It has also been characterized as a key factor influencing drug sensitivity. Furthermore, accessible chromatin is a hallmark of active DNA regulatory elements and functions as a crucial epigenetic factor regulating cancer mechanisms. The present study assessed the genetic regulatory landscape in colon cancer by performing RNA sequencing and Assay for Transposase-Accessible Chromatin sequencing, and investigated the effects of 5-FU on chromatin accessibility and gene expression. Notably, while treatment with 5-FU mediated global increases in chromatin accessibility, chromatin organization in several genomic regions differed depending on the expression status of p53. Since the occupancy of p53 does not overlap with accessible chromatin regions, the 5-FU-mediated changes in chromatin accessibility were not regulated by direct binding of p53. In the p53-expressing condition, the 5-FU-mediated accessible chromatin region was primarily associated with genes encoding cell death pathways. Additionally, 5-FU was revealed to induce open chromatin conformation at regions containing binding motifs for AP-1 family transcription factors, which may drive expression of apoptosis pathway genes. In conclusion, expression of p53 may confer 5-FU sensitivity by regulating chromatin accessibility of distinct genes associated with cell apoptosis in a transcription-independent manner.
Collapse
Affiliation(s)
- Chul Min Yang
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea
| | - Moo-Koo Kang
- Department of Environmental Medical Biology, Institute of Tropical Medicine, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea
| | - Woong-Jae Jung
- Department of Environmental Medical Biology, Institute of Tropical Medicine, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea
| | - Jung-Sik Joo
- Department of Environmental Medical Biology, Institute of Tropical Medicine, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea
| | - Yong-Jin Kim
- Department of Environmental Medical Biology, Institute of Tropical Medicine, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea
| | - Yeeun Choi
- Department of Environmental Medical Biology, Institute of Tropical Medicine, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea
| | - Hyoung-Pyo Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea.,Department of Environmental Medical Biology, Institute of Tropical Medicine, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea
| |
Collapse
|
8
|
Pham D, Moseley CE, Gao M, Savic D, Winstead CJ, Sun M, Kee BL, Myers RM, Weaver CT, Hatton RD. Batf Pioneers the Reorganization of Chromatin in Developing Effector T Cells via Ets1-Dependent Recruitment of Ctcf. Cell Rep 2020; 29:1203-1220.e7. [PMID: 31665634 PMCID: PMC7182170 DOI: 10.1016/j.celrep.2019.09.064] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 06/28/2019] [Accepted: 09/20/2019] [Indexed: 11/28/2022] Open
Abstract
The basic leucine zipper transcription factor activating transcription factor-like (Batf) contributes to transcriptional programming of multiple effector T cells and is required for T helper 17 (Th17) and T follicular helper (Tfh) cell development. Here, we examine mechanisms by which Batf initiates gene transcription in developing effector CD4 T cells. We find that, in addition to its pioneering function, Batf controls developmentally regulated recruitment of the architectural factor Ctcf to promote chromatin looping that is associated with lineage-specific gene transcription. The chromatin-organizing actions of Batf are largely dependent on Ets1, which appears to be indispensable for the Batf-dependent recruitment of Ctcf. Moreover, most of the Batf-dependent sites to which Ctcf is recruited lie outside of activating protein-1-interferon regulatory factor (Ap-1-Irf) composite elements (AICEs), indicating that direct involvement of Batf-Irf complexes is not required. These results identify a cooperative role for Batf, Ets1, and Ctcf in chromatin reorganization that underpins the transcriptional programming of effector T cells.
Collapse
Affiliation(s)
- Duy Pham
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Carson E Moseley
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Min Gao
- Informatics Institute, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Daniel Savic
- Human Genomics and Genetics, HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | - Colleen J Winstead
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mengxi Sun
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Barbara L Kee
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Richard M Myers
- Human Genomics and Genetics, HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | - Casey T Weaver
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Robin D Hatton
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
9
|
Olson WJ, Jakic B, Labi V, Schoeler K, Kind M, Klepsch V, Baier G, Hermann-Kleiter N. Orphan Nuclear Receptor NR2F6 Suppresses T Follicular Helper Cell Accumulation through Regulation of IL-21. Cell Rep 2020; 28:2878-2891.e5. [PMID: 31509749 PMCID: PMC6791812 DOI: 10.1016/j.celrep.2019.08.024] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 07/08/2019] [Accepted: 08/05/2019] [Indexed: 12/24/2022] Open
Abstract
CD4 T follicular helper (Tfh) cells are specialized in helping B cells during the germinal center (GC) reaction and ultimately promote long-term humoral immunity. Here we report that loss of the nuclear orphan receptor NR2F6 causes enhanced survival and accumulation of Tfh cells, GC B cells, and plasma cells (PCs) following T cell-dependent immunization. Nr2f6-deficient CD4 T cell dysfunction is the primary cause of cell accumulation. Cytokine expression in Nr2f6-deficient Tfh cells is dysregulated, and Il21 expression is enhanced. Mechanistically, NR2F6 binds directly to the interleukin 21 (IL-21) promoter and a conserved noncoding sequence (CNS) near the Il21 gene in resting CD4+ T cells. During Tfh cell differentiation, this direct NR2F6 DNA interaction is abolished. Enhanced Tfh cell accumulation in Nr2f6-deficient mice can be reverted by blocking IL-21R signaling. Thus, NR2F6 is a critical negative regulator of IL-21 cytokine production in Tfh cells and prevents excessive Tfh cell accumulation. Loss of NR2F6 results in accumulation of Tfh, GC B, and plasma cells after immunization Increased GC populations depend on Nr2f6 loss within the CD4 compartment NR2F6 directly binds to several sites within the Il21 promoter and CNS −36 NR2F6 restrains Il21 expression in CD4 cells; IL-21R blockade reduces Tfh accumulation
Collapse
Affiliation(s)
- William J Olson
- Institute of Cell Genetics, Department for Genetics and Pharmacology, Medical University of Innsbruck, Peter Mayr Str. 1a, 6020 Innsbruck, Austria
| | - Bojana Jakic
- Institute of Cell Genetics, Department for Genetics and Pharmacology, Medical University of Innsbruck, Peter Mayr Str. 1a, 6020 Innsbruck, Austria
| | - Verena Labi
- Division of Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Innrain 80, 6020 Innsbruck, Austria
| | - Katia Schoeler
- Division of Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Innrain 80, 6020 Innsbruck, Austria
| | - Michaela Kind
- Institute of Cell Genetics, Department for Genetics and Pharmacology, Medical University of Innsbruck, Peter Mayr Str. 1a, 6020 Innsbruck, Austria
| | - Victoria Klepsch
- Institute of Cell Genetics, Department for Genetics and Pharmacology, Medical University of Innsbruck, Peter Mayr Str. 1a, 6020 Innsbruck, Austria
| | - Gottfried Baier
- Institute of Cell Genetics, Department for Genetics and Pharmacology, Medical University of Innsbruck, Peter Mayr Str. 1a, 6020 Innsbruck, Austria
| | - Natascha Hermann-Kleiter
- Institute of Cell Genetics, Department for Genetics and Pharmacology, Medical University of Innsbruck, Peter Mayr Str. 1a, 6020 Innsbruck, Austria.
| |
Collapse
|
10
|
Madera-Salcedo IK, Sánchez-Hernández BE, Svyryd Y, Esquivel-Velázquez M, Rodríguez-Rodríguez N, Trejo-Zambrano MI, García-González HB, Hernández-Molina G, Mutchinick OM, Alcocer-Varela J, Rosetti F, Crispín JC. PPP2R2B hypermethylation causes acquired apoptosis deficiency in systemic autoimmune diseases. JCI Insight 2019; 5:126457. [PMID: 31335320 DOI: 10.1172/jci.insight.126457] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Chronic inflammation causes target organ damage in patients with systemic autoimmune diseases. The factors that allow this protracted response are poorly understood. We analyzed the transcriptional regulation of PPP2R2B (B55ß), a molecule necessary for the termination of the immune response, in patients with autoimmune diseases. Altered expression of B55ß conditioned resistance to cytokine withdrawal-induced death (CWID) in patients with autoimmune diseases. The impaired upregulation of B55ß was caused by inflammation-driven hypermethylation of specific cytosines located within a regulatory element of PPP2R2B preventing CTCF binding. This phenotype could be induced in healthy T cells by exposure to TNF-α. Our results reveal a gene whose expression is affected by an acquired defect, through an epigenetic mechanism, in the setting of systemic autoimmunity. Because failure to remove activated T cells through CWID could contribute to autoimmune pathology, this mechanism illustrates a vicious cycle through which autoimmune inflammation contributes to its own perpetuation.
Collapse
Affiliation(s)
| | - Beatriz E Sánchez-Hernández
- Department of Genetics, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Yevgeniya Svyryd
- Department of Genetics, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | | | | | | | | | | | - Osvaldo M Mutchinick
- Department of Genetics, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | | | | | | |
Collapse
|
11
|
Placek K, Schultze JL, Aschenbrenner AC. Epigenetic reprogramming of immune cells in injury, repair, and resolution. J Clin Invest 2019; 129:2994-3005. [PMID: 31329166 DOI: 10.1172/jci124619] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Immune cells are pivotal in the reaction to injury, whereupon, under ideal conditions, repair and resolution phases restore homeostasis following initial acute inflammation. Immune cell activation and reprogramming require transcriptional changes that can only be initiated if epigenetic alterations occur. Recently, accelerated deciphering of epigenetic mechanisms has extended knowledge of epigenetic regulation, including long-distance chromatin remodeling, DNA methylation, posttranslational histone modifications, and involvement of small and long noncoding RNAs. Epigenetic changes have been linked to aspects of immune cell development, activation, and differentiation. Furthermore, genome-wide epigenetic landscapes have been established for some immune cells, including tissue-resident macrophages, and blood-derived cells including T cells. The epigenetic mechanisms underlying developmental steps from hematopoietic stem cells to fully differentiated immune cells led to development of epigenetic technologies and insights into general rules of epigenetic regulation. Compared with more advanced research areas, epigenetic reprogramming of immune cells in injury remains in its infancy. While the early epigenetic mechanisms supporting activation of the immune response to injury have been studied, less is known about resolution and repair phases and cell type-specific changes. We review prominent recent findings concerning injury-mediated epigenetic reprogramming, particularly in stroke and myocardial infarction. Lastly, we illustrate how single-cell technologies will be crucial to understanding epigenetic reprogramming in the complex sequential processes following injury.
Collapse
Affiliation(s)
- Katarzyna Placek
- Immunology and Metabolism, LIMES Institute, University of Bonn, Bonn, Germany
| | - Joachim L Schultze
- Platform for Single Cell Genomics and Epigenomics at the German Center for Neurodegenerative Diseases and the University of Bonn, Bonn, Germany.,Genomics and Immunoregulation, LIMES Institute, University of Bonn, Bonn, Germany
| | - Anna C Aschenbrenner
- Genomics and Immunoregulation, LIMES Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
12
|
Wang J, Lai B, Nanayakkara G, Yang Q, Sun Y, Lu Y, Shao Y, Yu D, Yang WY, Cueto R, Fu H, Zeng H, Shen W, Wu S, Zhang C, Liu Y, Choi ET, Wang H, Yang X. Experimental Data-Mining Analyses Reveal New Roles of Low-Intensity Ultrasound in Differentiating Cell Death Regulatome in Cancer and Non-cancer Cells via Potential Modulation of Chromatin Long-Range Interactions. Front Oncol 2019; 9:600. [PMID: 31355136 PMCID: PMC6640725 DOI: 10.3389/fonc.2019.00600] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 06/18/2019] [Indexed: 12/17/2022] Open
Abstract
Background: The mechanisms underlying low intensity ultrasound (LIUS) mediated suppression of inflammation and tumorigenesis remain poorly determined. Methods: We used microarray datasets from NCBI GEO Dataset databases and conducted a comprehensive data mining analyses, where we studied the gene expression of 299 cell death regulators that regulate 13 different cell death types (cell death regulatome) in cells treated with LIUS. Results: We made the following findings: (1) LIUS exerts a profound effect on the expression of cell death regulatome in cancer cells and non-cancer cells. Of note, LIUS has the tendency to downregulate the gene expression of cell death regulators in non-cancer cells. Most of the cell death regulator genes downregulated by LIUS in non-cancer cells are responsible for mediating inflammatory signaling pathways; (2) LIUS activates different cell death transcription factors in cancer and non-cancer cells. Transcription factors TP-53 and SRF- were induced by LIUS exposure in cancer cells and non-cancer cells, respectively; (3) As two well-accepted mechanisms of LIUS, mild hyperthermia and oscillatory shear stress induce changes in the expression of cell death regulators, therefore, may be responsible for inducing LIUS mediated changes in gene expression patterns of cell death regulators in cells; (4) LIUS exposure may change the redox status of the cells. LIUS may induce more of antioxidant effects in non-cancer cells compared to cancer cells; and (5) The genes modulated by LIUS in cancer cells have distinct chromatin long range interaction (CLRI) patterns to that of non-cancer cells. Conclusions: Our analysis suggests novel molecular mechanisms that may be utilized by LIUS to induce tumor suppression and inflammation inhibition. Our findings may lead to development of new treatment protocols for cancers and chronic inflammation.
Collapse
Affiliation(s)
- Jiwei Wang
- Department of Pharmacology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Microbiology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Immunology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Bin Lai
- Department of Pharmacology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Microbiology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Immunology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Gayani Nanayakkara
- Department of Pharmacology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Microbiology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Immunology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
| | - Qian Yang
- Department of Pharmacology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Microbiology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Immunology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
| | - Yu Sun
- Department of Pharmacology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Microbiology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Immunology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
| | - Yifan Lu
- Department of Pharmacology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Microbiology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Immunology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
| | - Ying Shao
- Department of Pharmacology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Microbiology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Immunology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
| | - Daohai Yu
- Department of Clinical Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - William Y. Yang
- Department of Pharmacology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Microbiology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Immunology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
| | - Ramon Cueto
- Department of Pharmacology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Microbiology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Immunology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
| | - Hangfei Fu
- Department of Pharmacology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Microbiology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Immunology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
| | - Huihong Zeng
- Department of Pharmacology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Microbiology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Immunology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
| | - Wen Shen
- Department of Pharmacology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Microbiology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Immunology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
| | - Susu Wu
- Department of Pharmacology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Microbiology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Immunology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
| | - Chunquan Zhang
- Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yanna Liu
- Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Eric T. Choi
- Department of Pharmacology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Microbiology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Immunology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Division of Vascular and Endovascular Surgery, Department of Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Hong Wang
- Department of Pharmacology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Microbiology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Immunology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
| | - Xiaofeng Yang
- Department of Pharmacology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Microbiology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
- Department of Immunology, Centers for Metabolic Disease Research, Inflammation, Translational and Clinical Lung Research, Cardiovascular Research, Thrombosis Research, Philadelphia, PA, United States
| |
Collapse
|
13
|
Lu Y, Sun Y, Drummer C, Nanayakkara GK, Shao Y, Saaoud F, Johnson C, Zhang R, Yu D, Li X, Yang WY, Yu J, Jiang X, Choi ET, Wang H, Yang X. Increased acetylation of H3K14 in the genomic regions that encode trained immunity enzymes in lysophosphatidylcholine-activated human aortic endothelial cells - Novel qualification markers for chronic disease risk factors and conditional DAMPs. Redox Biol 2019; 24:101221. [PMID: 31153039 PMCID: PMC6543097 DOI: 10.1016/j.redox.2019.101221] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 05/06/2019] [Accepted: 05/14/2019] [Indexed: 12/14/2022] Open
Abstract
To test our hypothesis that proatherogenic lysophosphatidylcholine (LPC) upregulates trained immunity pathways (TIPs) in human aortic endothelial cells (HAECs), we conducted an intensive analyses on our RNA-Seq data and histone 3 lysine 14 acetylation (H3K14ac)-CHIP-Seq data, both performed on HAEC treated with LPC. Our analysis revealed that: 1) LPC induces upregulation of three TIPs including glycolysis enzymes (GE), mevalonate enzymes (ME), and acetyl-CoA generating enzymes (ACE); 2) LPC induces upregulation of 29% of 31 histone acetyltransferases, three of which acetylate H3K14; 3) LPC induces H3K14 acetylation (H3K14ac) in the genomic DNA that encodes LPC-induced TIP genes (79%) in comparison to that of in LPC-induced effector genes (43%) including ICAM-1; 4) TIP pathways are significantly different from that of EC activation effectors including adhesion molecule ICAM-1; 5) reactive oxygen species generating enzyme NOX2 deficiency decreases, but antioxidant transcription factor Nrf2 deficiency increases, the expressions of a few TIP genes and EC activation effector genes; and 6) LPC induced TIP genes(81%) favor inter-chromosomal long-range interactions (CLRI, trans-chromatin interaction) while LPC induced effector genes (65%) favor intra-chromosomal CLRIs (cis-chromatin interaction). Our findings demonstrated that proatherogenic lipids upregulate TIPs in HAECs, which are a new category of qualification markers for chronic disease risk factors and conditional DAMPs and potential mechanisms for acute inflammation transition to chronic ones. These novel insights may lead to identifications of new cardiovascular risk factors in upregulating TIPs in cardiovascular cells and novel therapeutic targets for the treatment of metabolic cardiovascular diseases, inflammation, and cancers. (total words: 245).
Collapse
Affiliation(s)
- Yifan Lu
- Centers for Inflammation, Translational & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Yu Sun
- Centers for Inflammation, Translational & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Charles Drummer
- Centers for Inflammation, Translational & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Gayani K Nanayakkara
- Centers for Inflammation, Translational & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Ying Shao
- Centers for Inflammation, Translational & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Fatma Saaoud
- Centers for Inflammation, Translational & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Candice Johnson
- Centers for Inflammation, Translational & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Ruijing Zhang
- Centers for Inflammation, Translational & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Daohai Yu
- Department of Clinical Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Xinyuan Li
- Centers for Inflammation, Translational & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - William Y Yang
- Centers for Inflammation, Translational & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Jun Yu
- Metabolic Disease Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Xiaohua Jiang
- Centers for Inflammation, Translational & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA; Metabolic Disease Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Eric T Choi
- Metabolic Disease Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA; Division of Vascular & Endovascular Surgery, Department of Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Hong Wang
- Metabolic Disease Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA; Department of Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Xiaofeng Yang
- Centers for Inflammation, Translational & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA; Metabolic Disease Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA; Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA; Department of Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA.
| |
Collapse
|
14
|
Li A, Sun Y, Drummer C, Lu Y, Yu D, Zhou Y, Li X, Pearson SJ, Johnson C, Yu C, Yang WY, Mastascusa K, Jiang X, Sun J, Rogers T, Hu W, Wang H, Yang X. Increasing Upstream Chromatin Long-Range Interactions May Favor Induction of Circular RNAs in LysoPC-Activated Human Aortic Endothelial Cells. Front Physiol 2019; 10:433. [PMID: 31057422 PMCID: PMC6482593 DOI: 10.3389/fphys.2019.00433] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 03/28/2019] [Indexed: 01/10/2023] Open
Abstract
Circular RNAs (circRNAs) are non-coding RNAs that form covalently closed continuous loops, and act as gene regulators in physiological and disease conditions. To test our hypothesis that proatherogenic lipid lysophosphatidylcholine (LPC) induce a set of circRNAs in human aortic endothelial cell (HAEC) activation, we performed circRNA analysis by searching our RNA-Seq data from LPC-activated HAECs, and found: (1) LPC induces significant modulation of 77 newly characterized cirRNAs, among which 47 circRNAs (61%) are upregulated; (2) 34 (72%) out of 47 upregulated circRNAs are upregulated when the corresponding mRNAs are downregulated, suggesting that the majority of circRNAs are upregulated presumably via LPC-induced “abnormal splicing” when the canonical splicing for generation of corresponding mRNAs is suppressed; (3) Upregulation of 47 circRNAs is temporally associated with mRNAs-mediated LPC-upregulated cholesterol synthesis-SREBP2 pathway and LPC-downregulated TGF-β pathway; (4) Increase in upstream chromatin long-range interaction sites to circRNA related genes is associated with preferred circRNA generation over canonical splicing for mRNAs, suggesting that shifting chromatin long-range interaction sites from downstream to upstream may promote induction of a list of circRNAs in lysoPC-activated HAECs; (5) Six significantly changed circRNAs may have sponge functions for miRNAs; and (6) 74% significantly changed circRNAs contain open reading frames, suggesting that putative short proteins may interfere with the protein interaction-based signaling. Our findings have demonstrated for the first time that a new set of LPC-induced circRNAs may contribute to homeostasis in LPC-induced HAEC activation. These novel insights may lead to identifications of new therapeutic targets for treating metabolic cardiovascular diseases, inflammations, and cancers.
Collapse
Affiliation(s)
- Angus Li
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States.,Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, United States
| | - Yu Sun
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Charles Drummer
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Yifan Lu
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Daohai Yu
- Department of Clinical Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Yan Zhou
- Biostatistics and Bioinformatics Facility, Fox Chase Cancer Center, Temple Health, Philadelphia, PA, United States
| | - Xinyuan Li
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Simone J Pearson
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Candice Johnson
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Catherine Yu
- Geisinger Commonwealth School of Medicine, Scranton, PA, United States
| | - William Y Yang
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Kevin Mastascusa
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Xiaohua Jiang
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Jianxin Sun
- Center for Translational Medicine, Department of Medicine, Sidney Kimmel Medical College, Philadelphia University - Thomas Jefferson University, Philadelphia, PA, United States
| | - Thomas Rogers
- Center for Inflammation, Translational and Clinical Lung Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Wenhui Hu
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Hong Wang
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Xiaofeng Yang
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States.,Center for Inflammation, Translational and Clinical Lung Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
15
|
Shin JO, Lee JJ, Kim M, Chung YW, Min H, Kim JY, Kim HP, Bok J. CTCF Regulates Otic Neurogenesis via Histone Modification in the Neurog1 Locus. Mol Cells 2018; 41:695-702. [PMID: 30008200 PMCID: PMC6078853 DOI: 10.14348/molcells.2018.0230] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 06/04/2018] [Indexed: 11/27/2022] Open
Abstract
The inner ear is a complex sensory organ responsible for hearing and balance. Formation of the inner ear is dependent on tight regulation of spatial and temporal expression of genes that direct a series of developmental processes. Recently, epigenetic regulation has emerged as a crucial regulator of the development of various organs. However, what roles higher-order chromatin organization and its regulator molecules play in inner ear development are unclear. CCCTC-binding factor (CTCF) is a highly conserved 11-zinc finger protein that regulates the three-dimensional architecture of chromatin, and is involved in various gene regulation processes. To delineate the role of CTCF in inner ear development, the present study investigated inner ear-specific Ctcf knockout mouse embryos (Pax2-Cre; Ctcffl/fl ). The loss of Ctcf resulted in multiple defects of inner ear development and severely compromised otic neurogenesis, which was partly due to a loss of Neurog1 expression. Furthermore, reduced Neurog1 gene expression by CTCF knockdown was found to be associated with changes in histone modification at the gene's promoter, as well as its upstream enhancer. The results of the present study demonstrate that CTCF plays an essential role in otic neurogenesis by modulating histone modification in the Neurog1 locus.
Collapse
Affiliation(s)
- Jeong-Oh Shin
- Department of Anatomy, Yonsei University College of Medicine, Seoul 03722,
Korea
| | - Jong-Joo Lee
- Department of Environmental Medical Biology, Yonsei University College of Medicine, Seoul 03722,
Korea
- BK21 PLUS project for Medical Science, Yonsei University College of Medicine, Seoul 03722,
Korea
| | - Mikyoung Kim
- Department of Environmental Medical Biology, Yonsei University College of Medicine, Seoul 03722,
Korea
| | - Youn Wook Chung
- Department of Environmental Medical Biology, Yonsei University College of Medicine, Seoul 03722,
Korea
| | - Hyehyun Min
- Department of Anatomy, Yonsei University College of Medicine, Seoul 03722,
Korea
| | - Jae-Yoon Kim
- Department of Anatomy, Yonsei University College of Medicine, Seoul 03722,
Korea
- BK21 PLUS project for Medical Science, Yonsei University College of Medicine, Seoul 03722,
Korea
| | - Hyoung-Pyo Kim
- Department of Environmental Medical Biology, Yonsei University College of Medicine, Seoul 03722,
Korea
- BK21 PLUS project for Medical Science, Yonsei University College of Medicine, Seoul 03722,
Korea
| | - Jinwoong Bok
- Department of Anatomy, Yonsei University College of Medicine, Seoul 03722,
Korea
- BK21 PLUS project for Medical Science, Yonsei University College of Medicine, Seoul 03722,
Korea
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722,
Korea
| |
Collapse
|
16
|
Li R, Wang J, Zhu F, Li R, Liu B, Xu W, He G, Cao H, Wang Y, Yang J. HMGB1 regulates T helper 2 and T helper17 cell differentiation both directly and indirectly in asthmatic mice. Mol Immunol 2018; 97:45-55. [PMID: 29567318 DOI: 10.1016/j.molimm.2018.02.014] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 02/12/2018] [Accepted: 02/19/2018] [Indexed: 12/15/2022]
Abstract
The Th (T helper) 2 response is characteristic of allergic asthma, and Th17 cells are involved in more severe asthma. Recent studies demonstrated that HMGB1 (High mobility group box 1 protein) regulates airway inflammation and the Th2, Th17 inflammatory response in asthma. HMGB1 can interact with Toll-like receptors (TLR) 2 and 4, and the receptor for advanced glycation end products (RAGE), activating the NF-κB (nuclear factor kappa B) signaling pathway and inducing the release of downstream inflammatory mediators. Both Th cells and dendritic cells express TLR2, TLR4, and RAGE receptors. Therefore, we speculate that HMGB1 could regulate the differentiation of Th2, Th17 cells in asthma through direct and indirect mechanisms. An ovalbumin (OVA)-induced mouse asthmatic model was established. Anti-HMGB1 antibody or rHMGB1 was administered to OVA-sensitized mice 30 min prior to each challenge. For in vitro studies, magnetically separated CD4+ naive T cells were stimulated with or without rHMGB1 and/or anti-HMGB1 antibody. BMDCs (bone marrow-derived dendritic cells)-stimulated with or without rHMGB1 and/or anti-HMGB1 antibody were cocultured with CD4+ naive T cells. Our study showed that administration of rHMGB1 aggravated airway inflammation and mucus production, and induced Th2, Th17 polarization in asthmatic mice, and that anti-HMGB1 antibody weakened characteristic features of asthma and blocked the Th2, Th17 inflammatory responses. HMGB1 could directly act on naive T cells to induce differentiation of Th2, Th17 cells in vitro through activating the TLR2, TLR4, RAGE-NF-κB signal pathway in CD4+ naive T cells. HMGB1 could also indirectly promote Th2, Th17 differentiation via activating the TLR2, TLR4, RAGE-NF-κB signal pathway in DCs to mediate their maturation and antigen-presenting ability in vitro.
Collapse
Affiliation(s)
- Ruiting Li
- Department of Respiratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, PR China
| | - Jing Wang
- Department of Intensive Care Unit, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, PR China
| | - Fangfang Zhu
- Department of Intensive Care Unit, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, PR China
| | - Ruifang Li
- Department of Neurology, Hubei Third People's Hospital, Wuhan, Hubei 430033, PR China
| | - Bing Liu
- Department of Respiratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, PR China
| | - Wenjuan Xu
- Department of Respiratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, PR China
| | - Guangzhen He
- Department of Respiratory Medicine, Taihe Hospital of Hubei University of Medicine, Shiyan, 442000, PR China
| | - Huan Cao
- Department of Respiratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, PR China
| | - Yimin Wang
- Department of Respiratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, PR China
| | - Jiong Yang
- Department of Respiratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, PR China.
| |
Collapse
|
17
|
Abnormal Microglia and Enhanced Inflammation-Related Gene Transcription in Mice with Conditional Deletion of Ctcf in Camk2a-Cre-Expressing Neurons. J Neurosci 2017; 38:200-219. [PMID: 29133437 DOI: 10.1523/jneurosci.0936-17.2017] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 10/21/2017] [Accepted: 11/09/2017] [Indexed: 01/22/2023] Open
Abstract
CCCTC-binding factor (CTCF) is an 11 zinc finger DNA-binding domain protein that regulates gene expression by modifying 3D chromatin structure. Human mutations in CTCF cause intellectual disability and autistic features. Knocking out Ctcf in mouse embryonic neurons is lethal by neonatal age, but the effects of CTCF deficiency in postnatal neurons are less well studied. We knocked out Ctcf postnatally in glutamatergic forebrain neurons under the control of Camk2a-Cre. CtcfloxP/loxP;Camk2a-Cre+ (Ctcf CKO) mice of both sexes were viable and exhibited profound deficits in spatial learning/memory, impaired motor coordination, and decreased sociability by 4 months of age. Ctcf CKO mice also had reduced dendritic spine density in the hippocampus and cerebral cortex. Microarray analysis of mRNA from Ctcf CKO mouse hippocampus identified increased transcription of inflammation-related genes linked to microglia. Separate microarray analysis of mRNA isolated specifically from Ctcf CKO mouse hippocampal neurons by ribosomal affinity purification identified upregulation of chemokine signaling genes, suggesting crosstalk between neurons and microglia in Ctcf CKO hippocampus. Finally, we found that microglia in Ctcf CKO mouse hippocampus had abnormal morphology by Sholl analysis and increased immunostaining for CD68, a marker of microglial activation. Our findings confirm that Ctcf KO in postnatal neurons causes a neurobehavioral phenotype in mice and provide novel evidence that CTCF depletion leads to overexpression of inflammation-related genes and microglial dysfunction.SIGNIFICANCE STATEMENT CCCTC-binding factor (CTCF) is a DNA-binding protein that organizes nuclear chromatin topology. Mutations in CTCF cause intellectual disability and autistic features in humans. CTCF deficiency in embryonic neurons is lethal in mice, but mice with postnatal CTCF depletion are less well studied. We find that mice lacking Ctcf in Camk2a-expressing neurons (Ctcf CKO mice) have spatial learning/memory deficits, impaired fine motor skills, subtly altered social interactions, and decreased dendritic spine density. We demonstrate that Ctcf CKO mice overexpress inflammation-related genes in the brain and have microglia with abnormal morphology that label positive for CD68, a marker of microglial activation. Our findings suggest that inflammation and dysfunctional neuron-microglia interactions are factors in the pathology of CTCF deficiency.
Collapse
|
18
|
Spolski R, Gromer D, Leonard WJ. The γ c family of cytokines: fine-tuning signals from IL-2 and IL-21 in the regulation of the immune response. F1000Res 2017; 6:1872. [PMID: 29123649 PMCID: PMC5657018 DOI: 10.12688/f1000research.12202.1] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/24/2017] [Indexed: 01/08/2023] Open
Abstract
Interleukin (IL)-2, IL-4, IL-7, IL-9, IL-15, and IL-21 form a family of cytokines based on the sharing of a receptor component, the common cytokine receptor γ chain, γ
c, which is encoded by the gene mutated in humans with X-linked severe combined immunodeficiency (XSCID). Together, these cytokines play critical roles in lymphoid development, differentiation, growth, and survival as well as mediating effector function. Here, we provide an overview of the main actions of members of this cytokine family but then primarily focus on IL-2 and IL-21, discussing their dynamic interplay and contributions to a fine-tuned immune response. Moreover, we discuss the therapeutic utility of modulating their actions, particularly for autoimmunity and cancer.
Collapse
Affiliation(s)
- Rosanne Spolski
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Daniel Gromer
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Warren J Leonard
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
19
|
Mirza AH, Kaur S, Pociot F. Long non-coding RNAs as novel players in β cell function and type 1 diabetes. Hum Genomics 2017; 11:17. [PMID: 28738846 PMCID: PMC5525349 DOI: 10.1186/s40246-017-0113-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 07/18/2017] [Indexed: 12/15/2022] Open
Abstract
Background Long non-coding RNAs (lncRNAs) are a sub-class within non-coding RNA repertoire that have emerged as crucial regulators of the gene expression in various pathophysiological conditions. lncRNAs display remarkable versatility and wield their functions through interactions with RNA, DNA, or proteins. Accumulating body of evidence based on multitude studies has highlighted the role of lncRNAs in many autoimmune and inflammatory diseases, including type 1 diabetes (T1D). Main body of abstract This review highlights emerging roles of lncRNAs in immune and islet β cell function as well as some of the challenges and opportunities in understanding the pathogenesis of T1D and its complications. Conclusion We accentuate that the lncRNAs within T1D-loci regions in consort with regulatory variants and enhancer clusters orchestrate the chromatin remodeling in β cells and thereby act as cis/trans-regulatory determinants of islet cell transcriptional programs.
Collapse
Affiliation(s)
- Aashiq H Mirza
- CPH-DIRECT, Department of Pediatrics, Herlev University Hospital, Herlev Ringvej 75, DK-2730, Herlev, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Center for non-coding RNA in Technology and Health, University of Copenhagen, Copenhagen, Denmark
| | - Simranjeet Kaur
- CPH-DIRECT, Department of Pediatrics, Herlev University Hospital, Herlev Ringvej 75, DK-2730, Herlev, Denmark
| | - Flemming Pociot
- CPH-DIRECT, Department of Pediatrics, Herlev University Hospital, Herlev Ringvej 75, DK-2730, Herlev, Denmark. .,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark. .,Center for non-coding RNA in Technology and Health, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
20
|
Song MJ, Kim M, Choi Y, Yi MH, Kim J, Park SJ, Yong TS, Kim HP. Epigenome mapping highlights chromatin-mediated gene regulation in the protozoan parasite Trichomonas vaginalis. Sci Rep 2017; 7:45365. [PMID: 28345651 PMCID: PMC5366954 DOI: 10.1038/srep45365] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 02/22/2017] [Indexed: 02/06/2023] Open
Abstract
Trichomonas vaginalis is an extracellular flagellated protozoan parasite that causes trichomoniasis, one of the most common non-viral sexually transmitted diseases. To survive and to maintain infection, T. vaginalis adapts to a hostile host environment by regulating gene expression. However, the mechanisms of transcriptional regulation are poorly understood for this parasite. Histone modification has a marked effect on chromatin structure and directs the recruitment of transcriptional machinery, thereby regulating essential cellular processes. In this study, we aimed to outline modes of chromatin-mediated gene regulation in T. vaginalis. Inhibition of histone deacetylase (HDAC) alters global transcriptional responses and induces hyperacetylation of histones and hypermethylation of H3K4. Analysis of the genome of T. vaginalis revealed that a number of enzymes regulate histone modification, suggesting that epigenetic mechanisms are important to controlling gene expression in this organism. Additionally, we describe the genome-wide localization of two histone H3 modifications (H3K4me3 and H3K27Ac), which we found to be positively associated with active gene expression in both steady and dynamic transcriptional states. These results provide the first direct evidence that histone modifications play an essential role in transcriptional regulation of T. vaginalis, and may help guide future epigenetic research into therapeutic intervention strategies against this parasite.
Collapse
Affiliation(s)
- Min-Ji Song
- Department of Environmental Medical Biology, Institute of Tropical Medicine, Yonsei University College of Medicine, Seoul, 03722, Korea.,Graduate Program of Nano Science and Technology, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Mikyoung Kim
- Department of Environmental Medical Biology, Institute of Tropical Medicine, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Yeeun Choi
- Department of Environmental Medical Biology, Institute of Tropical Medicine, Yonsei University College of Medicine, Seoul, 03722, Korea.,BK21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Myung-Hee Yi
- Department of Environmental Medical Biology, Institute of Tropical Medicine, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Juri Kim
- Department of Environmental Medical Biology, Institute of Tropical Medicine, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Soon-Jung Park
- Department of Environmental Medical Biology, Institute of Tropical Medicine, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Tai-Soon Yong
- Department of Environmental Medical Biology, Institute of Tropical Medicine, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Hyoung-Pyo Kim
- Department of Environmental Medical Biology, Institute of Tropical Medicine, Yonsei University College of Medicine, Seoul, 03722, Korea.,Graduate Program of Nano Science and Technology, Yonsei University College of Medicine, Seoul, 03722, Korea.,BK21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Korea
| |
Collapse
|
21
|
Stratigi K, Chatzidoukaki O, Garinis GA. DNA damage-induced inflammation and nuclear architecture. Mech Ageing Dev 2016; 165:17-26. [PMID: 27702596 DOI: 10.1016/j.mad.2016.09.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 09/20/2016] [Accepted: 09/25/2016] [Indexed: 12/12/2022]
Abstract
Nuclear architecture and the chromatin state affect most-if not all- DNA-dependent transactions, including the ability of cells to sense DNA lesions and restore damaged DNA back to its native form. Recent evidence points to functional links between DNA damage sensors, DNA repair mechanisms and the innate immune responses. The latter raises the question of how such seemingly disparate processes operate within the intrinsically complex nuclear landscape and the chromatin environment. Here, we discuss how DNA damage-induced immune responses operate within chromatin and the distinct sub-nuclear compartments highlighting their relevance to chronic inflammation.
Collapse
Affiliation(s)
- Kalliopi Stratigi
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Nikolaou Plastira 100, 70013, Heraklion, Crete, Greece
| | - Ourania Chatzidoukaki
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Nikolaou Plastira 100, 70013, Heraklion, Crete, Greece
| | - George A Garinis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Nikolaou Plastira 100, 70013, Heraklion, Crete, Greece; Department of Biology, University of Crete, Vassilika Vouton, GR71409, Heraklion, Crete, Greece.
| |
Collapse
|
22
|
Zhang J, Han B, Li X, Bies J, Jiang P, Koller RP, Wolff L. Distal regulation of c-myb expression during IL-6-induced differentiation in murine myeloid progenitor M1 cells. Cell Death Dis 2016; 7:e2364. [PMID: 27607579 PMCID: PMC5059869 DOI: 10.1038/cddis.2016.267] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 07/25/2016] [Accepted: 07/29/2016] [Indexed: 12/31/2022]
Abstract
The c-Myb transcription factor is a major regulator that controls differentiation and proliferation of hematopoietic progenitor cells, which is frequently deregulated in hematological diseases, such as lymphoma and leukemia. Understanding of the mechanisms regulating the transcription of c-myb gene is challenging as it lacks a typical promoter and multiple factors are involved. Our previous studies identified some distal regulatory elements in the upstream regions of c-myb gene in murine myeloid progenitor M1 cells, but the detailed mechanisms still remain unclear. In the present study, we found that a cell differentiation-related DNase1 hypersensitive site is located at a -28k region upstream of c-myb gene and that transcription factors Hoxa9, Meis1 and PU.1 bind to the -28k region. Circular chromosome conformation capture (4C) assay confirmed the interaction between the -28k region and the c-myb promoter, which is supported by the enrichment of CTCF and Cohesin. Our analysis also points to a critical role for Hoxa9 and PU.1 in distal regulation of c-myb expression in murine myeloid cells and cell differentiation. Overexpression of Hoxa9 disrupted the IL-6-induced differentiation of M1 cells and upregulated c-myb expression through binding of the -28k region. Taken together, our results provide an evidence for critical role of the -28k region in distal regulatory mechanism for c-myb gene expression during differentiation of myeloid progenitor M1 cells.
Collapse
Affiliation(s)
- Junfang Zhang
- Key Laboratory of Aquacultural Resources and Utilization, Ministry of Education, College of Fishery and Life Science, Shanghai Ocean University, No.999 Huchenghuan Road, Pudong New District, Shanghai 201306, China
| | - Bingshe Han
- Key Laboratory of Aquacultural Resources and Utilization, Ministry of Education, College of Fishery and Life Science, Shanghai Ocean University, No.999 Huchenghuan Road, Pudong New District, Shanghai 201306, China
| | - Xiaoxia Li
- Key Laboratory of Aquacultural Resources and Utilization, Ministry of Education, College of Fishery and Life Science, Shanghai Ocean University, No.999 Huchenghuan Road, Pudong New District, Shanghai 201306, China
| | - Juraj Bies
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Penglei Jiang
- Key Laboratory of Aquacultural Resources and Utilization, Ministry of Education, College of Fishery and Life Science, Shanghai Ocean University, No.999 Huchenghuan Road, Pudong New District, Shanghai 201306, China
| | - Richard P Koller
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Linda Wolff
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| |
Collapse
|