1
|
Cheng S, Yang J, Wang Y, Xian L, Hu Z, Zou L. The function and regulation of CCAAT/enhancer binding protein ε. EUR J INFLAMM 2023. [DOI: 10.1177/1721727x231153322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
In recent years, studies on the structure, function, and regulation of the C/EBPε gene have become an essential topic in the field of many diseases. CCAAT/enhancer-binding protein ε (C/EBPε) is the fifth member of the transcription factor CCAAT/C/EBP family of transcription factors. It plays crucial roles in cell proliferation, differentiation, immunity, energy metabolism, and hematopoiesis. C/EBPε plays essential roles in regulating the hematopoietic system, including myeloid cell development and maturation, participation in the body’s immune responses, and prevention of infections. C/EBPε function is regulated by phosphorylation, acetylation, methylation, and other types of genes. This review related to C/EBPε structure, function and regulation provides a theoretical basis for subsequent research in this area. C/EBPε is an emerging therapeutic target and thus provides new strategies for disease prevention and control.
Collapse
Affiliation(s)
- Shaowen Cheng
- Department of Emergency and Traumatology, First Affiliated Hospital of Hainan Medical University, Haikou, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
- Research Unit of Island Emergency Medicine, Chinese Academy of Medical Sciences (No. 2019RU013), Hainan Medical University, Haikou, China
| | - Jian Yang
- Department of Emergency and Traumatology, First Affiliated Hospital of Hainan Medical University, Haikou, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
| | - Yudie Wang
- Emergency and Trauma College, Hainan Medical University, Haikou, China
| | - Lina Xian
- Intensive Care Unit, Hainan Medical University, Haikou, China
| | - Zhihua Hu
- Intensive Care Unit, Hainan Medical University, Haikou, China
| | - Lingyun Zou
- Center for Clinical Data Research, Chongqing University Central Hospital, Chongqing, China
| |
Collapse
|
2
|
Balog BM, Sonti A, Zigmond RE. Neutrophil biology in injuries and diseases of the central and peripheral nervous systems. Prog Neurobiol 2023; 228:102488. [PMID: 37355220 PMCID: PMC10528432 DOI: 10.1016/j.pneurobio.2023.102488] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 05/24/2023] [Accepted: 06/16/2023] [Indexed: 06/26/2023]
Abstract
The role of inflammation in nervous system injury and disease is attracting increased attention. Much of that research has focused on microglia in the central nervous system (CNS) and macrophages in the peripheral nervous system (PNS). Much less attention has been paid to the roles played by neutrophils. Neutrophils are part of the granulocyte subtype of myeloid cells. These cells, like macrophages, originate and differentiate in the bone marrow from which they enter the circulation. After tissue damage or infection, neutrophils are the first immune cells to infiltrate into tissues and are directed there by specific chemokines, which act on chemokine receptors on neutrophils. We have reviewed here the basic biology of these cells, including their differentiation, the types of granules they contain, the chemokines that act on them, the subpopulations of neutrophils that exist, and their functions. We also discuss tools available for identification and further study of neutrophils. We then turn to a review of what is known about the role of neutrophils in CNS and PNS diseases and injury, including stroke, Alzheimer's disease, multiple sclerosis, amyotrophic lateral sclerosis, spinal cord and traumatic brain injuries, CNS and PNS axon regeneration, and neuropathic pain. While in the past studies have focused on neutrophils deleterious effects, we will highlight new findings about their benefits. Studies on their actions should lead to identification of ways to modify neutrophil effects to improve health.
Collapse
Affiliation(s)
- Brian M Balog
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106-4975, USA
| | - Anisha Sonti
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106-4975, USA
| | - Richard E Zigmond
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106-4975, USA.
| |
Collapse
|
3
|
Wang L, Feng J, Deng Y, Yang Q, Wei Q, Ye D, Rong X, Guo J. CCAAT/Enhancer-Binding Proteins in Fibrosis: Complex Roles Beyond Conventional Understanding. RESEARCH (WASHINGTON, D.C.) 2022; 2022:9891689. [PMID: 36299447 PMCID: PMC9575473 DOI: 10.34133/2022/9891689] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/18/2022] [Indexed: 07/29/2023]
Abstract
CCAAT/enhancer-binding proteins (C/EBPs) are a family of at least six identified transcription factors that contain a highly conserved basic leucine zipper domain and interact selectively with duplex DNA to regulate target gene expression. C/EBPs play important roles in various physiological processes, and their abnormal function can lead to various diseases. Recently, accumulating evidence has demonstrated that aberrant C/EBP expression or activity is closely associated with the onset and progression of fibrosis in several organs and tissues. During fibrosis, various C/EBPs can exert distinct functions in the same organ, while the same C/EBP can exert distinct functions in different organs. Modulating C/EBP expression or activity could regulate various molecular processes to alleviate fibrosis in multiple organs; therefore, novel C/EBPs-based therapeutic methods for treating fibrosis have attracted considerable attention. In this review, we will explore the features of C/EBPs and their critical functions in fibrosis in order to highlight new avenues for the development of novel therapies targeting C/EBPs.
Collapse
Affiliation(s)
- Lexun Wang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China
- Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, China
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jiaojiao Feng
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China
- Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, China
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yanyue Deng
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China
- Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, China
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Qianqian Yang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China
- Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, China
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Quxing Wei
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China
- Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, China
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Dewei Ye
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China
- Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, China
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xianglu Rong
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China
- Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, China
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jiao Guo
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China
- Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, China
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
4
|
Park SD, Saunders AS, Reidy MA, Bender DE, Clifton S, Morris KT. A review of granulocyte colony-stimulating factor receptor signaling and regulation with implications for cancer. Front Oncol 2022; 12:932608. [PMID: 36033452 PMCID: PMC9402976 DOI: 10.3389/fonc.2022.932608] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 07/15/2022] [Indexed: 12/29/2022] Open
Abstract
Granulocyte colony-stimulating factor receptor (GCSFR) is a critical regulator of granulopoiesis. Studies have shown significant upregulation of GCSFR in a variety of cancers and cell types and have recognized GCSFR as a cytokine receptor capable of influencing both myeloid and non-myeloid immune cells, supporting pro-tumoral actions. This systematic review aims to summarize the available literature examining the mechanisms that control GCSFR signaling, regulation, and surface expression with emphasis on how these mechanisms may be dysregulated in cancer. Experiments with different cancer cell lines from breast cancer, bladder cancer, glioma, and neuroblastoma are used to review the biological function and underlying mechanisms of increased GCSFR expression with emphasis on actions related to tumor proliferation, migration, and metastasis, primarily acting through the JAK/STAT pathway. Evidence is also presented that demonstrates a differential physiological response to aberrant GCSFR signal transduction in different organs. The lifecycle of the receptor is also reviewed to support future work defining how this signaling axis becomes dysregulated in malignancies.
Collapse
Affiliation(s)
- Sungjin David Park
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK, United States
| | - Apryl S. Saunders
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK, United States
| | - Megan A. Reidy
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK, United States
| | - Dawn E. Bender
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK, United States
| | - Shari Clifton
- Department of Information Management, University of Oklahoma Health Science Center, Oklahoma City, OK, United States
| | - Katherine T. Morris
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK, United States
- *Correspondence: Katherine T. Morris,
| |
Collapse
|
5
|
CCAAT/Enhancer-Binding Protein ε 27 Antagonism of GATA-1 Transcriptional Activity in the Eosinophil Is Mediated by a Unique N-Terminal Repression Domain, Is Independent of Sumoylation and Does Not Require DNA Binding. Int J Mol Sci 2021; 22:ijms222312689. [PMID: 34884493 PMCID: PMC8657826 DOI: 10.3390/ijms222312689] [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: 10/14/2021] [Revised: 11/18/2021] [Accepted: 11/19/2021] [Indexed: 11/16/2022] Open
Abstract
CCAAT/enhancer binding protein epsilon (C/EBPε) is required for eosinophil differentiation, lineage-specific gene transcription, and expression of C/EBPε32 and shorter 27kD and 14kD isoforms is developmentally regulated during this process. We previously defined the 27kD isoform (C/EBPε27) as an antagonist of GATA-1 transactivation of the eosinophil's major basic protein-1 (MBP1) P2-promoter, showing C/EBPε27 and GATA-1 physically interact. In the current study, we used a Tat-C/EBPε27 fusion protein for cell/nuclear transduction of an eosinophil myelocyte cell line to demonstrate that C/EBPε27 is a potent repressor of MBP1 transcription. We performed structure-function analyses of C/EBPε27 mapping its repressor domains, comparing it to C/EBPε32 and C/EBPε14, using GATA-1 co-transactivation of the MBP1-P2 promoter. Results show C/EBPε27 repression of GATA-1 is mediated by its unique 68aa N-terminus combined with previously identified RDI domain. This repressor activity does not require, but is enhanced by, DNA binding via the basic region of C/EBPε27 but independent of sumoylation of the RDI core "VKEEP" sumoylation site. These findings identify the N-terminus of C/EBPε27 as the minimum repressor domain required for antagonism of GATA-1 in the eosinophil. C/EBPε27 repression of GATA-1 occurs via a combination of both C/EBPε27-GATA-1 protein-protein interaction and C/EBPε27 binding to a C/EBP site in the MBP1 promoter. The C/EBPε27 isoform may serve to titrate and/or turn off eosinophil granule protein genes like MBP1 during eosinophil differentiation, as these genes are ultimately silenced in the mature cell. Understanding the functionality of C/EBPε27 in eosinophil development may prove promising in developing therapeutics that reduce eosinophil proliferation in allergic diseases.
Collapse
|
6
|
Al Madhoun A, Haddad D, Al Tarrah M, Jacob S, Al-Ali W, Nizam R, Miranda L, Al-Rashed F, Sindhu S, Ahmad R, Bitar MS, Al-Mulla F. Microarray analysis reveals ONC201 mediated differential mechanisms of CHOP gene regulation in metastatic and nonmetastatic colorectal cancer cells. Sci Rep 2021; 11:11893. [PMID: 34088951 PMCID: PMC8178367 DOI: 10.1038/s41598-021-91092-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 05/20/2021] [Indexed: 01/03/2023] Open
Abstract
The imipramine ONC201 has antiproliferative effects in several cancer cell types and activates integrated stress response pathway associated with the induction of Damage Inducible Transcript 3 (DDIT3, also known as C/EBP homologous protein or CHOP). We investigated the signaling pathways through which ONC201/CHOP crosstalk is regulated in ONC201-treated nonmetastatic and metastatic cancer cell lines (Dukes' type B colorectal adenocarcinoma nonmetastatic SW480 and metastatic LS-174T cells, respectively). Cell proliferation and apoptosis were evaluated by MTT assays and flow cytometry, gene expression was assessed by Affymetrix microarray, signaling pathway perturbations were assessed in silico, and key regulatory proteins were validated by Western blotting. Unlike LS-174T cells, SW480 cells were resistant to ONC201 treatment; Gene Ontology analysis of differentially expressed genes showed that cellular responsiveness to ONC201 treatment also differed substantially. In both ONC201-treated cell lines, CHOP expression was upregulated; however, its upstream regulatory mechanisms were perturbed. Although, PERK, ATF6 and IRE1 ER-stress pathways upregulated CHOP in both cell types, the Bak/Bax pathway regulated CHOP only LS-174T cells. Additionally, CHOP RNA splicing profiles varied between cell lines; these were further modified by ONC201 treatment. In conclusion, we delineated the signaling mechanisms by which CHOP expression is regulated in ONC201-treated non-metastatic and metastatic colorectal cell lines. The observed differences could be related to cellular plasticity and metabolic reprogramming, nevertheless, detailed mechanistic studies are required for further validations.
Collapse
Affiliation(s)
- Ashraf Al Madhoun
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, 15462, Dasman, Kuwait.
- Department of Animal and Imaging Core Facilities, Dasman Diabetes Institute, 15462, Dasman, Kuwait.
| | - Dania Haddad
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, 15462, Dasman, Kuwait
| | - Mustafa Al Tarrah
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, 15462, Dasman, Kuwait
| | - Sindhu Jacob
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, 15462, Dasman, Kuwait
| | - Waleed Al-Ali
- Department of Pharmacology and Toxicology, Faculty of Medicine, Kuwait University, 046302, Jabriya, Kuwait
| | - Rasheeba Nizam
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, 15462, Dasman, Kuwait
| | - Lavina Miranda
- Department of Animal and Imaging Core Facilities, Dasman Diabetes Institute, 15462, Dasman, Kuwait
| | - Fatema Al-Rashed
- Department of Immunology and Microbiology, Dasman Diabetes Institute, 15462, Dasman, Kuwait
| | - Sardar Sindhu
- Department of Animal and Imaging Core Facilities, Dasman Diabetes Institute, 15462, Dasman, Kuwait
- Department of Immunology and Microbiology, Dasman Diabetes Institute, 15462, Dasman, Kuwait
| | - Rasheed Ahmad
- Department of Immunology and Microbiology, Dasman Diabetes Institute, 15462, Dasman, Kuwait
| | - Milad S Bitar
- Department of Pharmacology and Toxicology, Faculty of Medicine, Kuwait University, 046302, Jabriya, Kuwait
| | - Fahd Al-Mulla
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, 15462, Dasman, Kuwait.
| |
Collapse
|
7
|
Abstract
PURPOSE OF REVIEW Lineage commitment is governed by instructive and stochastic signals, which drive both active induction of the lineage program and repression of alternative fates. Eosinophil lineage commitment is driven by the ordered interaction of transcription factors, supported by cytokine signals. This review summarizes key findings in the study of eosinophil lineage commitment and examines new data investigating the factors that regulate this process. RECENT FINDINGS Recent and past studies highlight how intrinsic and extrinsic signals modulate transcription factor network and lineage decisions. Early action of the transcription factors C/EBPα and GATA binding protein-1 along with C/EBPε supports lineage commitment and eosinophil differentiation. This process is regulated and enforced by the pseudokinase Trib1, a regulator of C/EBPα levels. The cytokines interleukin (IL)-5 and IL-33 also support early eosinophil development. However, current studies suggest that these cytokines are not specifically required for lineage commitment. SUMMARY Together, recent evidence suggests a model where early transcription factor activity drives expression of key eosinophil genes and cytokine receptors to prime lineage commitment. Understanding the factors and signals that control eosinophil lineage commitment may guide therapeutic development for eosinophil-mediated diseases and provide examples for fate choices in other lineages.
Collapse
|
8
|
EVI1 in Leukemia and Solid Tumors. Cancers (Basel) 2020; 12:cancers12092667. [PMID: 32962037 PMCID: PMC7564095 DOI: 10.3390/cancers12092667] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/02/2020] [Accepted: 09/13/2020] [Indexed: 11/30/2022] Open
Abstract
Simple Summary Ecotropic viral integration site 1 (EVI1) is transcriptionally activated in a subset of myeloid leukemias. Since its discovery, other isoforms of EVI1 have been identified. It has been shown that EVI1 and its isoforms mainly function as transcription factors and to play important roles not only in leukemia but also in a variety of solid tumors. To provide a comprehensive understanding of this family of proteins, we summarize the currently available knowledge of expression and function of EVI1 and its isoforms in leukemia and solid tumors and provide insights of future studies. Abstract The EVI1 gene encodes for a transcription factor with two zinc finger domains and is transcriptionally activated in a subset of myeloid leukemias. In leukemia, the transcriptional activation of EVI1 usually results from chromosomal rearrangements. Besides leukemia, EVI1 has also been linked to solid tumors including breast cancer, lung cancer, ovarian cancer and colon cancer. The MDS1/EVI1 gene is encoded by the same locus as EVI1. While EVI1 functions as a transcription repressor, MDS1/EVI1 acts as a transcription activator. The fusion protein encoded by the AML1/MDS1/EVI1 chimeric gene, resulting from chromosomal translocations in a subset of chronic myeloid leukemia, exhibits a similar function to EVI1. EVI1 has been shown to regulate cell proliferation, differentiation and apoptosis, whereas the functions of MDS1/EVI1 and AML1/MDS1/EVI1 remain elusive. In this review, we summarize the genetic structures, biochemical properties and biological functions of these proteins in cancer.
Collapse
|
9
|
Hsu BE, Tabariès S, Johnson RM, Andrzejewski S, Senecal J, Lehuédé C, Annis MG, Ma EH, Völs S, Ramsay L, Froment R, Monast A, Watson IR, Granot Z, Jones RG, St-Pierre J, Siegel PM. Immature Low-Density Neutrophils Exhibit Metabolic Flexibility that Facilitates Breast Cancer Liver Metastasis. Cell Rep 2020; 27:3902-3915.e6. [PMID: 31242422 DOI: 10.1016/j.celrep.2019.05.091] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 02/13/2019] [Accepted: 05/22/2019] [Indexed: 02/07/2023] Open
Abstract
Neutrophils are phenotypically heterogeneous and exert either anti- or pro-metastatic functions. We show that cancer-cell-derived G-CSF is necessary, but not sufficient, to mobilize immature low-density neutrophils (iLDNs) that promote liver metastasis. In contrast, mature high-density neutrophils inhibit the formation of liver metastases. Transcriptomic and metabolomic analyses of high- and low-density neutrophils reveal engagement of numerous metabolic pathways specifically in low-density neutrophils. iLDNs exhibit enhanced global bioenergetic capacity, through their ability to engage mitochondrial-dependent ATP production, and remain capable of executing pro-metastatic neutrophil functions, including NETosis, under nutrient-deprived conditions. We demonstrate that NETosis is an important neutrophil function that promotes breast cancer liver metastasis. iLDNs rely on the catabolism of glutamate and proline to support mitochondrial-dependent metabolism in the absence of glucose, which enables sustained NETosis. These data reveal that distinct pro-metastatic neutrophil populations exhibit a high degree of metabolic flexibility, which facilitates the formation of liver metastases.
Collapse
Affiliation(s)
- Brian E Hsu
- Goodman Cancer Research Centre, McGill University, Montreal, Québec, QC H3A 1A3, Canada; Department of Medicine, McGill University, Montreal, Québec, QC H3G 1Y6, Canada
| | - Sébastien Tabariès
- Goodman Cancer Research Centre, McGill University, Montreal, Québec, QC H3A 1A3, Canada
| | | | - Sylvia Andrzejewski
- Goodman Cancer Research Centre, McGill University, Montreal, Québec, QC H3A 1A3, Canada
| | - Julien Senecal
- Goodman Cancer Research Centre, McGill University, Montreal, Québec, QC H3A 1A3, Canada; Department of Medicine, McGill University, Montreal, Québec, QC H3G 1Y6, Canada
| | - Camille Lehuédé
- Goodman Cancer Research Centre, McGill University, Montreal, Québec, QC H3A 1A3, Canada
| | - Matthew G Annis
- Goodman Cancer Research Centre, McGill University, Montreal, Québec, QC H3A 1A3, Canada
| | - Eric H Ma
- Goodman Cancer Research Centre, McGill University, Montreal, Québec, QC H3A 1A3, Canada; Department of Physiology, McGill University, Montreal, Québec, QC H3G 1Y6, Canada
| | - Sandra Völs
- Department of Developmental Biology and Cancer Research, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - LeeAnn Ramsay
- Goodman Cancer Research Centre, McGill University, Montreal, Québec, QC H3A 1A3, Canada
| | - Remi Froment
- Department of Pathology and Microbiology, Université de Montréal, Saint Hyacinth, Québec, QC J2S 2M2, Canada
| | - Anie Monast
- Goodman Cancer Research Centre, McGill University, Montreal, Québec, QC H3A 1A3, Canada
| | - Ian R Watson
- Goodman Cancer Research Centre, McGill University, Montreal, Québec, QC H3A 1A3, Canada
| | - Zvi Granot
- Department of Developmental Biology and Cancer Research, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Russell G Jones
- Goodman Cancer Research Centre, McGill University, Montreal, Québec, QC H3A 1A3, Canada; Department of Physiology, McGill University, Montreal, Québec, QC H3G 1Y6, Canada
| | - Julie St-Pierre
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Peter M Siegel
- Goodman Cancer Research Centre, McGill University, Montreal, Québec, QC H3A 1A3, Canada; Department of Medicine, McGill University, Montreal, Québec, QC H3G 1Y6, Canada.
| |
Collapse
|
10
|
The "Janus" Role of C/EBPs Family Members in Cancer Progression. Int J Mol Sci 2020; 21:ijms21124308. [PMID: 32560326 PMCID: PMC7352866 DOI: 10.3390/ijms21124308] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/11/2020] [Accepted: 06/15/2020] [Indexed: 12/13/2022] Open
Abstract
CCAAT/enhancer-binding proteins (C/EBPs) constitute a family of transcription factors composed of six members that are critical for normal cellular differentiation in a variety of tissues. They promote the expression of genes through interaction with their promoters. Moreover, they have a key role in regulating cellular proliferation through interaction with cell cycle proteins. C/EBPs are considered to be tumor suppressor factors due to their ability to arrest cell growth (contributing to the terminal differentiation of several cell types) and for their role in cellular response to DNA damage, nutrient deprivation, hypoxia, and genotoxic agents. However, C/EBPs can elicit completely opposite effects on cell proliferation and cancer development and they have been described as both tumor promoters and tumor suppressors. This "Janus" role of C/EBPs depends on different factors, such as the type of tumor, the isoform/s expressed in cells, the type of dimerization (homo- or heterodimerization), the presence of inhibitory elements, and the ability to inhibit the expression of other tumor suppressors. In this review, we discuss the implication of the C/EBPs family in cancer, focusing on the molecular aspects that make these transcription factors tumor promoters or tumor suppressors.
Collapse
|
11
|
Paramonova N, Trapina I, Dokane K, Kalnina J, Sjakste T, Sjakste N. An Intergenic rs9275596 Polymorphism on Chr. 6p21 Is Associated with Multiple Sclerosis in Latvians. MEDICINA-LITHUANIA 2020; 56:medicina56040154. [PMID: 32244438 PMCID: PMC7230508 DOI: 10.3390/medicina56040154] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/24/2020] [Accepted: 03/26/2020] [Indexed: 12/30/2022]
Abstract
Background and objectives: Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system, leading to demyelination of neurons and potentially debilitating physical and mental symptoms. The disease is more prevalent in women than in men. The major histocompatibility complex (MHC) region has been identified as a major genetic determinant for autoimmune diseases, and its role in some neurological disorders including MS was evaluated. An intergenic single-nucleotide polymorphism (SNP), rs9275596, located between the HLA-DQB1 and HLA-DQA2 genes, is in significant association with various autoimmune diseases according to genome-wide association studies (GWASs). A cumulative effect of this SNP with other polymorphisms from this region was revealed. The aim of the study was to verify the data on rs9275596 association in multiple sclerosis in a case/control study of the Latvian population and to evaluate eventual functional significance of allele substitutions. Materials and Methods: rs9275596 (chr6:32713854; GRCh38.p12) was genotyped in 273 MS patients and 208 controls on main and sex-specific associations. Eventual functional significance of allele substitutions was evaluated in silico using publicly available tools. Results: The rs9275596 rare alleles were identified as a disease susceptibility factor in association with the MS main group and in affected females (p < 0.001 and p < 0.01, respectively). Risk factor genotypes with rare alleles included were associated with the MS common cohort (p < 0.002) and female cohort (odds ratio, OR = 2.24) and were identified as disease susceptible in males (OR = 2.41). It was shown that structural changes of rs9275596 affect the secondary structure of DNA. Functional significance of allele substitutions was evaluated on the eventual sequence affinity to transcription factors (TFs) and splicing signals similarity. A possible impact of the particular polymorphisms on the transcription and splicing efficiency is discussed. Conclusions: Our results suggest susceptibility of rs9275596 to multiple sclerosis in Latvians.
Collapse
Affiliation(s)
- Natalia Paramonova
- Genomics and Bioinformatics, Institute of Biology of the University of Latvia, LV-1004 Riga, Latvia; (N.P.); (K.D.); (J.K.); (T.S.); (N.S.)
| | - Ilva Trapina
- Genomics and Bioinformatics, Institute of Biology of the University of Latvia, LV-1004 Riga, Latvia; (N.P.); (K.D.); (J.K.); (T.S.); (N.S.)
- Correspondence: ; Tel.: +371-29354786
| | - Kristine Dokane
- Genomics and Bioinformatics, Institute of Biology of the University of Latvia, LV-1004 Riga, Latvia; (N.P.); (K.D.); (J.K.); (T.S.); (N.S.)
| | - Jolanta Kalnina
- Genomics and Bioinformatics, Institute of Biology of the University of Latvia, LV-1004 Riga, Latvia; (N.P.); (K.D.); (J.K.); (T.S.); (N.S.)
| | - Tatjana Sjakste
- Genomics and Bioinformatics, Institute of Biology of the University of Latvia, LV-1004 Riga, Latvia; (N.P.); (K.D.); (J.K.); (T.S.); (N.S.)
| | - Nikolajs Sjakste
- Genomics and Bioinformatics, Institute of Biology of the University of Latvia, LV-1004 Riga, Latvia; (N.P.); (K.D.); (J.K.); (T.S.); (N.S.)
- Department of Medical Biochemistry of the University of Latvia, LV-1004 Riga, Latvia
| |
Collapse
|
12
|
Göös H, Fogarty CL, Sahu B, Plagnol V, Rajamäki K, Nurmi K, Liu X, Einarsdottir E, Jouppila A, Pettersson T, Vihinen H, Krjutskov K, Saavalainen P, Järvinen A, Muurinen M, Greco D, Scala G, Curtis J, Nordström D, Flaumenhaft R, Vaarala O, Kovanen PE, Keskitalo S, Ranki A, Kere J, Lehto M, Notarangelo LD, Nejentsev S, Eklund KK, Varjosalo M, Taipale J, Seppänen MRJ. Gain-of-function CEBPE mutation causes noncanonical autoinflammatory inflammasomopathy. J Allergy Clin Immunol 2019; 144:1364-1376. [PMID: 31201888 PMCID: PMC11057357 DOI: 10.1016/j.jaci.2019.06.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 05/06/2019] [Accepted: 06/04/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND CCAAT enhancer-binding protein epsilon (C/EBPε) is a transcription factor involved in late myeloid lineage differentiation and cellular function. The only previously known disorder linked to C/EBPε is autosomal recessive neutrophil-specific granule deficiency leading to severely impaired neutrophil function and early mortality. OBJECTIVE The aim of this study was to molecularly characterize the effects of C/EBPε transcription factor Arg219His mutation identified in a Finnish family with previously genetically uncharacterized autoinflammatory and immunodeficiency syndrome. METHODS Genetic analysis, proteomics, genome-wide transcriptional profiling by means of RNA-sequencing, chromatin immunoprecipitation (ChIP) sequencing, and assessment of the inflammasome function of primary macrophages were performed. RESULTS Studies revealed a novel mechanism of genome-wide gain-of-function that dysregulated transcription of 464 genes. Mechanisms involved dysregulated noncanonical inflammasome activation caused by decreased association with transcriptional repressors, leading to increased chromatin occupancy and considerable changes in transcriptional activity, including increased expression of NLR family, pyrin domain-containing 3 protein (NLRP3) and constitutively expressed caspase-5 in macrophages. CONCLUSION We describe a novel autoinflammatory disease with defective neutrophil function caused by a homozygous Arg219His mutation in the transcription factor C/EBPε. Mutated C/EBPε acts as a regulator of both the inflammasome and interferome, and the Arg219His mutation causes the first human monogenic neomorphic and noncanonical inflammasomopathy/immunodeficiency. The mechanism, including widely dysregulated transcription, is likely not unique for C/EBPε. Similar multiomics approaches should also be used in studying other transcription factor-associated diseases.
Collapse
Affiliation(s)
- Helka Göös
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Christopher L Fogarty
- Folkhälsan Research Center, Helsinki, Finland; Abdominal Center Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Diabetes & Obesity Research Program, Research Program's Unit, University of Helsinki, Helsinki, Finland; Institute of Clinical Medicine, University of Helsinki, Helsinki, Finland
| | - Biswajyoti Sahu
- Research Programs Unit, Genome-Scale Biology, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Vincent Plagnol
- University College London Genetics Institute, University College London, London, United Kingdom
| | - Kristiina Rajamäki
- Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Katariina Nurmi
- Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Xiaonan Liu
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Elisabet Einarsdottir
- Folkhälsan Institute of Genetics, Helsinki, Finland; Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden; Research Programs Unit, Molecular Neurology, University of Helsinki, Helsinki, Finland
| | - Annukka Jouppila
- Helsinki University Hospital Research Institute, Helsinki, Finland
| | - Tom Pettersson
- Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Internal Medicine and Rehabilitation, Helsinki University Hospital, Helsinki, Finland
| | - Helena Vihinen
- Electron Microscopy Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Kaarel Krjutskov
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden; Helsinki University Hospital Research Institute, Helsinki, Finland; Competence Centre on Health Technologies, Tartu, Estonia
| | - Päivi Saavalainen
- Research Programs Unit, Immunobiology, University of Helsinki, Helsinki, Finland; Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland
| | - Asko Järvinen
- Adult Immunodeficiency Unit, Infectious Diseases, Inflammation Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Mari Muurinen
- Folkhälsan Institute of Genetics, Helsinki, Finland; Research Programs Unit, Molecular Neurology, University of Helsinki, Helsinki, Finland
| | - Dario Greco
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland; Faculty of Medicine and Life Sciences & Institute of Biosciences and Medical Technology, University of Tampere, Tampere, Finland
| | - Giovanni Scala
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland; Faculty of Medicine and Life Sciences & Institute of Biosciences and Medical Technology, University of Tampere, Tampere, Finland
| | - James Curtis
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Dan Nordström
- Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Rheumatology, Inflammation Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Robert Flaumenhaft
- Beth Israel Deaconess Medical Center, Department of Medicine, Harvard Medical School, Boston, Mass
| | - Outi Vaarala
- Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Respiratory, Inflammation and Autoimmunity, Innovative Medicine, AstraZeneca, Mölndal, Sweden
| | - Panu E Kovanen
- Department of Pathology, University of Helsinki, and HUSLAB, Helsinki University Hospital, Helsinki, Finland
| | - Salla Keskitalo
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Annamari Ranki
- Department of Dermatology, Allergology and Venereal Diseases, Inflammation Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Juha Kere
- Folkhälsan Institute of Genetics, Helsinki, Finland; Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden; Research Programs Unit, Molecular Neurology, University of Helsinki, Helsinki, Finland; School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London, United Kingdom
| | - Markku Lehto
- Folkhälsan Research Center, Helsinki, Finland; Abdominal Center Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Diabetes & Obesity Research Program, Research Program's Unit, University of Helsinki, Helsinki, Finland
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Sergey Nejentsev
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Kari K Eklund
- Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Rheumatology, Inflammation Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Orton Orthopaedic Hospital and Research Institute, Invalid Foundation, Helsinki, Finland
| | - Markku Varjosalo
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Jussi Taipale
- Research Programs Unit, Genome-Scale Biology, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland; Division of Functional Genomics and Systems Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Sweden; Department of Biochemistry, Cambridge University, Cambridge, United Kingdom
| | - Mikko R J Seppänen
- Adult Immunodeficiency Unit, Infectious Diseases, Inflammation Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Rare Diseases Center and Pediatric Research Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
| |
Collapse
|
13
|
Yokoyama N, Kim YJ, Hirabayashi Y, Tabe Y, Takamori K, Ogawa H, Iwabuchi K. Kras promotes myeloid differentiation through Wnt/β-catenin signaling. FASEB Bioadv 2019; 1:435-449. [PMID: 32123842 PMCID: PMC6996383 DOI: 10.1096/fba.2019-00004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 01/07/2019] [Accepted: 05/21/2019] [Indexed: 12/11/2022] Open
Abstract
Wild-type Kras, a small GTPase, inactivates Ras growth-promoting signaling. However, the role of Kras in differentiation of myeloid cells remains unclear. This study showed the involvement of Kras in a novel regulatory mechanism underlying the dimethyl sulfoxide (DMSO)-induced differentiation of human acute myeloid leukemia HL-60 cells. Kras was found to positively regulate DMSO-induced differentiation, with the activity of Kras increasing upon DMSO. Inhibition of Kras attenuated CD11b expression in differentiated HL-60 cells. GSK3β, an important component of Wnt signaling, was found to be a downstream signal of Kras. Phosphorylation of GSK3β was markedly enhanced by DMSO treatment. Moreover, inhibition of GSK3β enhanced CD11b expression and triggered the accumulation in the nucleus of β-catenin and Tcf in response to DMSO. Inhibitors of β-catenin-mediated pathways blocked CD11b expression, further indicating that β-catenin is involved in the differentiation of HL-60 cells. Elevated expression of C/EBPα and C/EBPɛ accompanied by the expression of granulocyte colony-stimulating factor (G-CSF) receptor was observed during differentiation. Taken together, these findings suggest that Kras engages in cross talk with the Wnt/β-catenin pathway upon DMSO treatment of HL-60 cells, thereby regulating the granulocytic differentiation of HL-60 cells. These results indicate that Kras acts as a tumor suppressor during the differentiation of myeloid cells.
Collapse
Affiliation(s)
- Noriko Yokoyama
- Institute for Environmental and Gender Specific Medicine Juntendo University Graduate School of Medicine Urayasu Chiba Japan
| | - Yeon-Jeong Kim
- Laboratory for Neuronal Growth Mechanisms Riken Brain Science Institutes Saitama Japan
| | - Yoshio Hirabayashi
- Institute for Environmental and Gender Specific Medicine Juntendo University Graduate School of Medicine Urayasu Chiba Japan
- Cellular Informatics Laboratory RIKEN Wako Saitama Japan
| | - Yoko Tabe
- Department of Laboratory Medicine Juntendo University School of Medicine Hospital Hongo Tokyo Japan
| | - Kenji Takamori
- Institute for Environmental and Gender Specific Medicine Juntendo University Graduate School of Medicine Urayasu Chiba Japan
| | - Hideoki Ogawa
- Institute for Environmental and Gender Specific Medicine Juntendo University Graduate School of Medicine Urayasu Chiba Japan
| | - Kazuhisa Iwabuchi
- Institute for Environmental and Gender Specific Medicine Juntendo University Graduate School of Medicine Urayasu Chiba Japan
- Infection Control Nursing Juntendo University Graduate School of Health Care and Nursing Urayasu Chiba Japan
- Laboratory of Biochemistry Juntendo University Faculty of Health Care and Nursing Urayasu Chiba Japan
| |
Collapse
|
14
|
Different Faces for Different Places: Heterogeneity of Neutrophil Phenotype and Function. J Immunol Res 2019; 2019:8016254. [PMID: 30944838 PMCID: PMC6421822 DOI: 10.1155/2019/8016254] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 09/22/2018] [Accepted: 01/03/2019] [Indexed: 02/05/2023] Open
Abstract
As the most abundant leukocytes in the circulation, neutrophils are committed to innate and adaptive immune effector function to protect the human body. They are capable of killing intruding microbes through various ways including phagocytosis, release of granules, and formation of extracellular traps. Recent research has revealed that neutrophils are heterogeneous in phenotype and function and can display outstanding plasticity in both homeostatic and disease states. The great flexibility and elasticity arm neutrophils with important regulatory and controlling functions in various disease states such as autoimmunity and inflammation as well as cancer. Hence, this review will focus on recent literature describing neutrophils' variable and diverse phenotypes and functions in different contexts.
Collapse
|
15
|
Splicing dysfunction and disease: The case of granulopoiesis. Semin Cell Dev Biol 2018; 75:23-39. [DOI: 10.1016/j.semcdb.2017.08.048] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/28/2017] [Accepted: 08/28/2017] [Indexed: 12/20/2022]
|
16
|
Fulkerson PC. Transcription Factors in Eosinophil Development and As Therapeutic Targets. Front Med (Lausanne) 2017; 4:115. [PMID: 28791289 PMCID: PMC5522844 DOI: 10.3389/fmed.2017.00115] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 07/06/2017] [Indexed: 12/11/2022] Open
Abstract
Dynamic gene expression is a major regulatory mechanism that directs hematopoietic cell fate and differentiation, including eosinophil lineage commitment and eosinophil differentiation. Though GATA-1 is well established as a critical transcription factor (TF) for eosinophil development, delineating the transcriptional networks that regulate eosinophil development at homeostasis and in inflammatory states is not complete. Yet, recent advances in molecular experimental tools using purified eosinophil developmental stages have led to identifying new regulators of gene expression during eosinophil development. Herein, recent studies that have provided new insight into the mechanisms of gene regulation during eosinophil lineage commitment and eosinophil differentiation are reviewed. A model is described wherein distinct classes of TFs work together via collaborative and hierarchical interactions to direct eosinophil development. In addition, the therapeutic potential for targeting TFs to regulate eosinophil production is discussed. Understanding how specific signals direct distinct patterns of gene expression required for the specialized functions of eosinophils will likely lead to new targets for therapeutic intervention.
Collapse
Affiliation(s)
- Patricia C Fulkerson
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
17
|
Malu K, Garhwal R, Pelletier MGH, Gotur D, Halene S, Zwerger M, Yang ZF, Rosmarin AG, Gaines P. Cooperative Activity of GABP with PU.1 or C/EBPε Regulates Lamin B Receptor Gene Expression, Implicating Their Roles in Granulocyte Nuclear Maturation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 197:910-22. [PMID: 27342846 PMCID: PMC5022553 DOI: 10.4049/jimmunol.1402285] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 05/30/2016] [Indexed: 01/26/2023]
Abstract
Nuclear segmentation is a hallmark feature of mammalian neutrophil differentiation, but the mechanisms that control this process are poorly understood. Gene expression in maturing neutrophils requires combinatorial actions of lineage-restricted and more widely expressed transcriptional regulators. Examples include interactions of the widely expressed ETS transcription factor, GA-binding protein (GABP), with the relatively lineage-restricted E-twenty-six (ETS) factor, PU.1, and with CCAAT enhancer binding proteins, C/EBPα and C/EBPε. Whether such cooperative interactions between these transcription factors also regulate the expression of genes encoding proteins that control nuclear segmentation is unclear. We investigated the roles of ETS and C/EBP family transcription factors in regulating the gene encoding the lamin B receptor (LBR), an inner nuclear membrane protein whose expression is required for neutrophil nuclear segmentation. Although C/EBPε was previously shown to bind the Lbr promoter, surprisingly, we found that neutrophils derived from Cebpe null mice exhibited normal Lbr gene and protein expression. Instead, GABP provided transcriptional activation through the Lbr promoter in the absence of C/EBPε, and activities supported by GABP were greatly enhanced by either C/EBPε or PU.1. Both GABP and PU.1 bound Ets sites in the Lbr promoter in vitro, and in vivo within both early myeloid progenitors and differentiating neutrophils. These findings demonstrate that GABP, PU.1, and C/EBPε cooperate to control transcription of the gene encoding LBR, a nuclear envelope protein that is required for the characteristic lobulated morphology of mature neutrophils.
Collapse
Affiliation(s)
- Krishnakumar Malu
- Department of Biological Sciences, Biomedical Engineering and Biotechnology Program, University of Massachusetts, Lowell, MA 01854
| | - Rahul Garhwal
- Department of Biological Sciences, Biomedical Engineering and Biotechnology Program, University of Massachusetts, Lowell, MA 01854
| | - Margery G H Pelletier
- Department of Biological Sciences, Biomedical Engineering and Biotechnology Program, University of Massachusetts, Lowell, MA 01854
| | - Deepali Gotur
- Department of Biological Sciences, Biomedical Engineering and Biotechnology Program, University of Massachusetts, Lowell, MA 01854
| | - Stephanie Halene
- Section of Hematology, Department of Internal Medicine and Yale Comprehensive Cancer Center, Yale University School of Medicine, New Haven, CT 06520
| | - Monika Zwerger
- Department of Biochemistry, University of Zurich, 8057 Zurich, Switzerland; and
| | - Zhong-Fa Yang
- Division of Hematology-Oncology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655
| | - Alan G Rosmarin
- Division of Hematology-Oncology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655
| | - Peter Gaines
- Department of Biological Sciences, Biomedical Engineering and Biotechnology Program, University of Massachusetts, Lowell, MA 01854;
| |
Collapse
|
18
|
Metabolic role of dipeptidyl peptidase 4 (DPP4) in primary human (pre)adipocytes. Sci Rep 2016; 6:23074. [PMID: 26983599 PMCID: PMC4794806 DOI: 10.1038/srep23074] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 02/25/2016] [Indexed: 12/25/2022] Open
Abstract
Dipeptidyl peptidase 4 (DPP4) is the target of the gliptins, a recent class of oral antidiabetics. DPP4 (also called CD26) was previously characterized in immune cells but also has important metabolic functions which are not yet fully understood. Thus, we investigated the function of DPP4 in human white preadipocytes and adipocytes. We found that both cell types express DPP4 in high amounts; DPP4 release markedly increased during differentiation. In preadipocytes, lentiviral DPP4 knockdown caused significant changes in gene expression as determined by whole-genome DNA-array analysis. Metabolic genes were increased, e.g. PDK4 18-fold and PPARγC1α (=PGC1α) 6-fold, and proliferation-related genes were decreased (e.g. FGF7 5-fold). These effects, contributing to differentiation, were not inhibited by the PPARγ antagonist T0070907. Vice versa, the PPARγ agonist pioglitazone induced a different set of genes (mainly FABP4). DPP4 knockdown also affected growth factor signaling and, accordingly, retarded preadipocyte proliferation. In particular, basal and insulin-induced ERK activation (but not Akt activation) was markedly diminished (by around 60%). This indicates that DPP4 knockdown contributes to adipocyte maturation by mimicking growth factor withdrawal, an early step in fat cell differentiation. In mature adipocytes, DPP4 becomes liberated so that adipose tissue may constitute a relevant source of circulating DPP4.
Collapse
|
19
|
Pulido-Salgado M, Vidal-Taboada JM, Saura J. C/EBPβ and C/EBPδ transcription factors: Basic biology and roles in the CNS. Prog Neurobiol 2015; 132:1-33. [PMID: 26143335 DOI: 10.1016/j.pneurobio.2015.06.003] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 05/08/2015] [Accepted: 06/16/2015] [Indexed: 02/01/2023]
Abstract
CCAAT/enhancer binding protein (C/EBP) β and C/EBPδ are transcription factors of the basic-leucine zipper class which share phylogenetic, structural and functional features. In this review we first describe in depth their basic molecular biology which includes fascinating aspects such as the regulated use of alternative initiation codons in the C/EBPβ mRNA. The physical interactions with multiple transcription factors which greatly opens the number of potentially regulated genes or the presence of at least five different types of post-translational modifications are also remarkable molecular mechanisms that modulate C/EBPβ and C/EBPδ function. In the second part, we review the present knowledge on the localization, expression changes and physiological roles of C/EBPβ and C/EBPδ in neurons, astrocytes and microglia. We conclude that C/EBPβ and C/EBPδ share two unique features related to their role in the CNS: whereas in neurons they participate in memory formation and synaptic plasticity, in glial cells they regulate the pro-inflammatory program. Because of their role in neuroinflammation, C/EBPβ and C/EBPδ in microglia are potential targets for treatment of neurodegenerative disorders. Any strategy to reduce C/EBPβ and C/EBPδ activity in neuroinflammation needs to take into account its potential side-effects in neurons. Therefore, cell-specific treatments will be required for the successful application of this strategy.
Collapse
Affiliation(s)
- Marta Pulido-Salgado
- Biochemistry and Molecular Biology Unit, School of Medicine, University of Barcelona, IDIBAPS, Casanova 143, planta 3, 08036 Barcelona, Spain
| | - Jose M Vidal-Taboada
- Biochemistry and Molecular Biology Unit, School of Medicine, University of Barcelona, IDIBAPS, Casanova 143, planta 3, 08036 Barcelona, Spain
| | - Josep Saura
- Biochemistry and Molecular Biology Unit, School of Medicine, University of Barcelona, IDIBAPS, Casanova 143, planta 3, 08036 Barcelona, Spain.
| |
Collapse
|
20
|
Joshi A, Pooley C, Freeman TC, Lennartsson A, Babina M, Schmidl C, Geijtenbeek T, Michoel T, Severin J, Itoh M, Lassmann T, Kawaji H, Hayashizaki Y, Carninci P, Forrest ARR, Rehli M, Hume DA. Technical Advance: Transcription factor, promoter, and enhancer utilization in human myeloid cells. J Leukoc Biol 2015; 97:985-995. [PMID: 25717144 PMCID: PMC4398258 DOI: 10.1189/jlb.6ta1014-477rr] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 01/23/2015] [Accepted: 01/28/2015] [Indexed: 11/24/2022] Open
Abstract
Analysis of the myeloid transcriptome by integrating 91 samples from the myeloid lineage and AML cell lines to predict novel regulatory interactions, enhancers, miRNAs, and lincRNAs. The generation of myeloid cells from their progenitors is regulated at the level of transcription by combinatorial control of key transcription factors influencing cell-fate choice. To unravel the global dynamics of this process at the transcript level, we generated transcription profiles for 91 human cell types of myeloid origin by use of CAGE profiling. The CAGE sequencing of these samples has allowed us to investigate diverse aspects of transcription control during myelopoiesis, such as identification of novel transcription factors, miRNAs, and noncoding RNAs specific to the myeloid lineage. We further reconstructed a transcription regulatory network by clustering coexpressed transcripts and associating them with enriched cis-regulatory motifs. With the use of the bidirectional expression as a proxy for enhancers, we predicted over 2000 novel enhancers, including an enhancer 38 kb downstream of IRF8 and an intronic enhancer in the KIT gene locus. Finally, we highlighted relevance of these data to dissect transcription dynamics during progressive maturation of granulocyte precursors. A multifaceted analysis of the myeloid transcriptome is made available (www.myeloidome.roslin.ed.ac.uk). This high-quality dataset provides a powerful resource to study transcriptional regulation during myelopoiesis and to infer the likely functions of unannotated genes in human innate immunity.
Collapse
Affiliation(s)
- Anagha Joshi
- *The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Scotland, United Kingdom; RIKEN Preventive Medicine and Diagnosis Innovation Program, Tsurumi-ku, Yokohama, Japan; RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Tsurumi-ku, Yokohama, Japan; Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden; Department of Dermatology and Allergy, Charité Universitätsmedizin Berlin, Germany; Department of Internal Medicine III, University Hospital, University of Regensburg, Germany; Department of Experimental Immunology, Academic Medical Center, Amsterdam, the Netherlands; and **RIKEN Omics Science Center, Tsurumi-ku, Yokohama, Japan
| | - Christopher Pooley
- *The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Scotland, United Kingdom; RIKEN Preventive Medicine and Diagnosis Innovation Program, Tsurumi-ku, Yokohama, Japan; RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Tsurumi-ku, Yokohama, Japan; Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden; Department of Dermatology and Allergy, Charité Universitätsmedizin Berlin, Germany; Department of Internal Medicine III, University Hospital, University of Regensburg, Germany; Department of Experimental Immunology, Academic Medical Center, Amsterdam, the Netherlands; and **RIKEN Omics Science Center, Tsurumi-ku, Yokohama, Japan
| | - Tom C Freeman
- *The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Scotland, United Kingdom; RIKEN Preventive Medicine and Diagnosis Innovation Program, Tsurumi-ku, Yokohama, Japan; RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Tsurumi-ku, Yokohama, Japan; Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden; Department of Dermatology and Allergy, Charité Universitätsmedizin Berlin, Germany; Department of Internal Medicine III, University Hospital, University of Regensburg, Germany; Department of Experimental Immunology, Academic Medical Center, Amsterdam, the Netherlands; and **RIKEN Omics Science Center, Tsurumi-ku, Yokohama, Japan
| | - Andreas Lennartsson
- *The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Scotland, United Kingdom; RIKEN Preventive Medicine and Diagnosis Innovation Program, Tsurumi-ku, Yokohama, Japan; RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Tsurumi-ku, Yokohama, Japan; Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden; Department of Dermatology and Allergy, Charité Universitätsmedizin Berlin, Germany; Department of Internal Medicine III, University Hospital, University of Regensburg, Germany; Department of Experimental Immunology, Academic Medical Center, Amsterdam, the Netherlands; and **RIKEN Omics Science Center, Tsurumi-ku, Yokohama, Japan
| | - Magda Babina
- *The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Scotland, United Kingdom; RIKEN Preventive Medicine and Diagnosis Innovation Program, Tsurumi-ku, Yokohama, Japan; RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Tsurumi-ku, Yokohama, Japan; Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden; Department of Dermatology and Allergy, Charité Universitätsmedizin Berlin, Germany; Department of Internal Medicine III, University Hospital, University of Regensburg, Germany; Department of Experimental Immunology, Academic Medical Center, Amsterdam, the Netherlands; and **RIKEN Omics Science Center, Tsurumi-ku, Yokohama, Japan
| | - Christian Schmidl
- *The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Scotland, United Kingdom; RIKEN Preventive Medicine and Diagnosis Innovation Program, Tsurumi-ku, Yokohama, Japan; RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Tsurumi-ku, Yokohama, Japan; Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden; Department of Dermatology and Allergy, Charité Universitätsmedizin Berlin, Germany; Department of Internal Medicine III, University Hospital, University of Regensburg, Germany; Department of Experimental Immunology, Academic Medical Center, Amsterdam, the Netherlands; and **RIKEN Omics Science Center, Tsurumi-ku, Yokohama, Japan
| | - Teunis Geijtenbeek
- *The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Scotland, United Kingdom; RIKEN Preventive Medicine and Diagnosis Innovation Program, Tsurumi-ku, Yokohama, Japan; RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Tsurumi-ku, Yokohama, Japan; Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden; Department of Dermatology and Allergy, Charité Universitätsmedizin Berlin, Germany; Department of Internal Medicine III, University Hospital, University of Regensburg, Germany; Department of Experimental Immunology, Academic Medical Center, Amsterdam, the Netherlands; and **RIKEN Omics Science Center, Tsurumi-ku, Yokohama, Japan
| | | | - Tom Michoel
- *The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Scotland, United Kingdom; RIKEN Preventive Medicine and Diagnosis Innovation Program, Tsurumi-ku, Yokohama, Japan; RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Tsurumi-ku, Yokohama, Japan; Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden; Department of Dermatology and Allergy, Charité Universitätsmedizin Berlin, Germany; Department of Internal Medicine III, University Hospital, University of Regensburg, Germany; Department of Experimental Immunology, Academic Medical Center, Amsterdam, the Netherlands; and **RIKEN Omics Science Center, Tsurumi-ku, Yokohama, Japan
| | - Jessica Severin
- *The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Scotland, United Kingdom; RIKEN Preventive Medicine and Diagnosis Innovation Program, Tsurumi-ku, Yokohama, Japan; RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Tsurumi-ku, Yokohama, Japan; Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden; Department of Dermatology and Allergy, Charité Universitätsmedizin Berlin, Germany; Department of Internal Medicine III, University Hospital, University of Regensburg, Germany; Department of Experimental Immunology, Academic Medical Center, Amsterdam, the Netherlands; and **RIKEN Omics Science Center, Tsurumi-ku, Yokohama, Japan
| | - Masayoshi Itoh
- *The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Scotland, United Kingdom; RIKEN Preventive Medicine and Diagnosis Innovation Program, Tsurumi-ku, Yokohama, Japan; RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Tsurumi-ku, Yokohama, Japan; Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden; Department of Dermatology and Allergy, Charité Universitätsmedizin Berlin, Germany; Department of Internal Medicine III, University Hospital, University of Regensburg, Germany; Department of Experimental Immunology, Academic Medical Center, Amsterdam, the Netherlands; and **RIKEN Omics Science Center, Tsurumi-ku, Yokohama, Japan
| | - Timo Lassmann
- *The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Scotland, United Kingdom; RIKEN Preventive Medicine and Diagnosis Innovation Program, Tsurumi-ku, Yokohama, Japan; RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Tsurumi-ku, Yokohama, Japan; Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden; Department of Dermatology and Allergy, Charité Universitätsmedizin Berlin, Germany; Department of Internal Medicine III, University Hospital, University of Regensburg, Germany; Department of Experimental Immunology, Academic Medical Center, Amsterdam, the Netherlands; and **RIKEN Omics Science Center, Tsurumi-ku, Yokohama, Japan
| | - Hideya Kawaji
- *The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Scotland, United Kingdom; RIKEN Preventive Medicine and Diagnosis Innovation Program, Tsurumi-ku, Yokohama, Japan; RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Tsurumi-ku, Yokohama, Japan; Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden; Department of Dermatology and Allergy, Charité Universitätsmedizin Berlin, Germany; Department of Internal Medicine III, University Hospital, University of Regensburg, Germany; Department of Experimental Immunology, Academic Medical Center, Amsterdam, the Netherlands; and **RIKEN Omics Science Center, Tsurumi-ku, Yokohama, Japan
| | - Yoshihide Hayashizaki
- *The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Scotland, United Kingdom; RIKEN Preventive Medicine and Diagnosis Innovation Program, Tsurumi-ku, Yokohama, Japan; RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Tsurumi-ku, Yokohama, Japan; Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden; Department of Dermatology and Allergy, Charité Universitätsmedizin Berlin, Germany; Department of Internal Medicine III, University Hospital, University of Regensburg, Germany; Department of Experimental Immunology, Academic Medical Center, Amsterdam, the Netherlands; and **RIKEN Omics Science Center, Tsurumi-ku, Yokohama, Japan
| | - Piero Carninci
- *The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Scotland, United Kingdom; RIKEN Preventive Medicine and Diagnosis Innovation Program, Tsurumi-ku, Yokohama, Japan; RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Tsurumi-ku, Yokohama, Japan; Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden; Department of Dermatology and Allergy, Charité Universitätsmedizin Berlin, Germany; Department of Internal Medicine III, University Hospital, University of Regensburg, Germany; Department of Experimental Immunology, Academic Medical Center, Amsterdam, the Netherlands; and **RIKEN Omics Science Center, Tsurumi-ku, Yokohama, Japan
| | - Alistair R R Forrest
- *The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Scotland, United Kingdom; RIKEN Preventive Medicine and Diagnosis Innovation Program, Tsurumi-ku, Yokohama, Japan; RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Tsurumi-ku, Yokohama, Japan; Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden; Department of Dermatology and Allergy, Charité Universitätsmedizin Berlin, Germany; Department of Internal Medicine III, University Hospital, University of Regensburg, Germany; Department of Experimental Immunology, Academic Medical Center, Amsterdam, the Netherlands; and **RIKEN Omics Science Center, Tsurumi-ku, Yokohama, Japan
| | - Michael Rehli
- *The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Scotland, United Kingdom; RIKEN Preventive Medicine and Diagnosis Innovation Program, Tsurumi-ku, Yokohama, Japan; RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Tsurumi-ku, Yokohama, Japan; Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden; Department of Dermatology and Allergy, Charité Universitätsmedizin Berlin, Germany; Department of Internal Medicine III, University Hospital, University of Regensburg, Germany; Department of Experimental Immunology, Academic Medical Center, Amsterdam, the Netherlands; and **RIKEN Omics Science Center, Tsurumi-ku, Yokohama, Japan
| | - David A Hume
- *The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Scotland, United Kingdom; RIKEN Preventive Medicine and Diagnosis Innovation Program, Tsurumi-ku, Yokohama, Japan; RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Tsurumi-ku, Yokohama, Japan; Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden; Department of Dermatology and Allergy, Charité Universitätsmedizin Berlin, Germany; Department of Internal Medicine III, University Hospital, University of Regensburg, Germany; Department of Experimental Immunology, Academic Medical Center, Amsterdam, the Netherlands; and **RIKEN Omics Science Center, Tsurumi-ku, Yokohama, Japan
| |
Collapse
|
21
|
Lieber S, Scheer F, Finkernagel F, Meissner W, Giehl G, Brendel C, Diederich WE, Müller-Brüsselbach S, Müller R. The inverse agonist DG172 triggers a PPARβ/δ-independent myeloid lineage shift and promotes GM-CSF/IL-4-induced dendritic cell differentiation. Mol Pharmacol 2015; 87:162-73. [PMID: 25398837 DOI: 10.1124/mol.114.094672] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The stilbene derivative (Z)-2-(2-bromophenyl)-3-{[4-(1-methylpiperazine)amino]phenyl}acrylonitrile (DG172) was developed as a highly selective inhibitory peroxisome proliferator-activated receptor (PPAR)β/δ ligand. Here, we describe a novel PPARβ/δ-independent, yet highly specific, effect of DG172 on the differentiation of bone marrow cells (BMCs). DG172 strongly augmented granulocyte-macrophage-colony-stimulating factor (GM-CSF)-induced differentiation of primary BMCs from Ppard null mice into two specific populations, characterized as mature (CD11c(hi)MHCII(hi)) and immature (CD11c(hi)MHCII(lo)) dendritic cells (DCs). IL-4 synergized with DG172 to shift the differentiation from MHCII(lo) cells to mature DCs in vitro. The promotion of DC differentiation occurred at the expense of differentiation to granulocytic Gr1(+)Ly6B(+) cells. In agreement with these findings, transcriptome analyses showed a strong DG172-mediated repression of genes encoding neutrophilic markers in both differentiating wild-type and Ppard null cells, while macrophage/DC marker genes were up-regulated. DG172 also inhibited the expression of transcription factors driving granulocytic differentiation (Cebpe, Gfi1, and Klf5), and increased the levels of transcription factors promoting macrophage/DC differentiation (Irf4, Irf8, Spib, and Spic). DG172 exerted these effects only at an early stage of BMC differentiation induced by GM-CSF, did not affect macrophage-colony-stimulating factor-triggered differentiation to macrophages and had no detectable PPARβ/δ-independent effect on other cell types tested. Structure-function analyses demonstrated that the 4-methylpiperazine moiety in DG172 is required for its effect on DC differentiation, but is dispensable for PPARβ/δ binding. Based on these data we developed a new compound, (Z)-2-(4-chlorophenyl)-3-[4-(4-methylpiperazine-1-yl)phenyl]acrylonitrile (DG228), which enhances DC differentiation in the absence of significant PPARβ/δ binding.
Collapse
Affiliation(s)
- Sonja Lieber
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany (S.L., F.F., W.M., S.M.-B., R.M.); Institute of Pharmaceutical Chemistry, Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany (F.S., W.E.D.); and Clinic for Hematology, Oncology and Immunology (G.G., C.B.); Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany
| | - Frithjof Scheer
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany (S.L., F.F., W.M., S.M.-B., R.M.); Institute of Pharmaceutical Chemistry, Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany (F.S., W.E.D.); and Clinic for Hematology, Oncology and Immunology (G.G., C.B.); Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany
| | - Florian Finkernagel
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany (S.L., F.F., W.M., S.M.-B., R.M.); Institute of Pharmaceutical Chemistry, Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany (F.S., W.E.D.); and Clinic for Hematology, Oncology and Immunology (G.G., C.B.); Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany
| | - Wolfgang Meissner
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany (S.L., F.F., W.M., S.M.-B., R.M.); Institute of Pharmaceutical Chemistry, Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany (F.S., W.E.D.); and Clinic for Hematology, Oncology and Immunology (G.G., C.B.); Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany
| | - Gavin Giehl
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany (S.L., F.F., W.M., S.M.-B., R.M.); Institute of Pharmaceutical Chemistry, Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany (F.S., W.E.D.); and Clinic for Hematology, Oncology and Immunology (G.G., C.B.); Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany
| | - Cornelia Brendel
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany (S.L., F.F., W.M., S.M.-B., R.M.); Institute of Pharmaceutical Chemistry, Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany (F.S., W.E.D.); and Clinic for Hematology, Oncology and Immunology (G.G., C.B.); Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany
| | - Wibke E Diederich
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany (S.L., F.F., W.M., S.M.-B., R.M.); Institute of Pharmaceutical Chemistry, Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany (F.S., W.E.D.); and Clinic for Hematology, Oncology and Immunology (G.G., C.B.); Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany
| | - Sabine Müller-Brüsselbach
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany (S.L., F.F., W.M., S.M.-B., R.M.); Institute of Pharmaceutical Chemistry, Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany (F.S., W.E.D.); and Clinic for Hematology, Oncology and Immunology (G.G., C.B.); Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany
| | - Rolf Müller
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany (S.L., F.F., W.M., S.M.-B., R.M.); Institute of Pharmaceutical Chemistry, Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany (F.S., W.E.D.); and Clinic for Hematology, Oncology and Immunology (G.G., C.B.); Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany
| |
Collapse
|
22
|
CCAAT/enhancer binding protein β in relation to ER stress, inflammation, and metabolic disturbances. BIOMED RESEARCH INTERNATIONAL 2015; 2015:324815. [PMID: 25699273 PMCID: PMC4324884 DOI: 10.1155/2015/324815] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 12/12/2014] [Accepted: 12/12/2014] [Indexed: 12/12/2022]
Abstract
The prevalence of the metabolic syndrome and underlying metabolic disturbances increase rapidly in developed countries. Various molecular targets are currently under investigation to unravel the molecular mechanisms that cause these disturbances. This is done in attempt to counter or prevent the negative health consequences of the metabolic disturbances. Here, we reviewed the current knowledge on the role of C/EBP-β in these metabolic disturbances. C/EBP-β deletion in mice resulted in downregulation of hepatic lipogenic genes and increased expression of β-oxidation genes in brown adipose tissue. Furthermore, C/EBP-β is important in the differentiation and maturation of adipocytes and is increased during ER stress and proinflammatory conditions. So far, studies were only conducted in animals and in cell systems. The results found that C/EBP-β is an important transcription factor within the metabolic disturbances of the metabolic system. Therefore, it is interesting to examine the potential role of C/EBP-β at molecular and physiological level in humans.
Collapse
|
23
|
Abstract
C/EBPε, a member of the CCAAT/enhancer binding protein (C/EBP) family of transcription factors, is exclusively expressed in myeloid cells and regulates transition from the promyelocytic stage to the myelocytic stage of neutrophil development, being indispensable for secondary and tertiary granule formation. Knowledge concerning the functional role of C/EBPε posttranslational modifications is limited to studies concerning phosphorylation and sumoylation. In the current study, using ectopic expression and ex vivo differentiation of CD34(+) hematopoietic progenitor cells, we demonstrate that C/EBPε is acetylated, which was confirmed by mass spectrometry analysis, identifying 4 acetylated lysines in 3 distinct functional domains. Regulation of C/EBPε acetylation levels by the p300 acetyltransferase and the sirtuin 1 deacetylase controls transcriptional activity, which can at least in part be explained by modulation of DNA binding. During neutrophil development, acetylation of lysines 121 and 198 were found to be crucial for terminal neutrophil differentiation and the expression of neutrophil-specific granule proteins, including lactoferrin and collagenase. Taken together, our data illustrate a critical role for acetylation in the functional regulation of C/EBPε activity during terminal neutrophil development.
Collapse
|
24
|
Nitto T, Sawaki K. Molecular mechanisms of the antileukemia activities of retinoid and arsenic. J Pharmacol Sci 2014; 126:179-85. [PMID: 25319615 DOI: 10.1254/jphs.14r15cp] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Acute promyelocytic leukemia (APL) is characterized by the occurrence of translocations between chromosomes 15 and 17, resulting in generation of a fusion protein of promyelocytic leukemia (PML) and retinoid A receptor (RAR) α. APL cells are unable to differentiate into mature granulocytes since PML-RARα functions as a strong transcriptional repressor for a gene involved in granulocyte differentiation. All-trans retinoic acid (ATRA) is the first agent that has been developed to target specific disease-causing molecules, i.e., ATRA suppresses abnormal functions of oncogenic proteins. Moreover, ATRA facilitates the differentiation of APL cells toward mature granulocytes by changing epigenetic modifiers from corepressor complexes to co-activator complexes on target genes after binding to the ligand-binding domain at the RARα moiety of the PML-RARα oncoprotein. On the other hand, arsenic trioxide (ATO), another promising agent used to treat APL, directly binds to the PML moiety of the PML-RARα protein, causing oxidation and multimerization. ATO enhances the conjugation of small ubiquitin-like modifiers to PML-RARα, followed by ubiquitination and degradation, relieving the genes associated with granulocytic differentiation from suppressive restraint by the oncoprotein. Recent clinical studies have demonstrated that combination therapy with both ATRA and ATO is useful to achieve remission.
Collapse
Affiliation(s)
- Takeaki Nitto
- Laboratory of Pharmacotherapy, Yokohama College of Pharmacy, Japan
| | | |
Collapse
|
25
|
Glass C, Wilson M, Gonzalez R, Zhang Y, Perkins AS. The role of EVI1 in myeloid malignancies. Blood Cells Mol Dis 2014; 53:67-76. [PMID: 24495476 DOI: 10.1016/j.bcmd.2014.01.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 12/26/2013] [Indexed: 01/01/2023]
Abstract
The EVI1 oncogene at human chr 3q26 is rearranged and/or overexpressed in a subset of acute myeloid leukemias and myelodysplasias. The EVI1 protein is a 135 kDa transcriptional regulator with DNA-binding zinc finger domains. Here we provide a critical review of the current state of research into the molecular mechanisms by which this gene plays a role in myeloid malignancies. The major pertinent cellular effects are blocking myeloid differentiation and preventing cellular apoptosis, and several potential mechanisms for these phenomena have been identified. Evidence supports a role for EVI1 in inducing cellular quiescence, and this may contribute to the resistance to chemotherapy seen in patients with neoplasms that overexpress EVI1. Another isoform, MDS1-EVI1 (or PRDM3), encoded by the same locus as EVI1, harbors an N-terminal histone methyltransferase(HMT) domain; experimental findings indicate that this protein and its HMT activity are critical for the progression of a subset of AMLs, and this provides a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Carolyn Glass
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine, Rochester, NY 14642, USA
| | - Michael Wilson
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine, Rochester, NY 14642, USA
| | - Ruby Gonzalez
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine, Rochester, NY 14642, USA
| | - Yi Zhang
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine, Rochester, NY 14642, USA
| | - Archibald S Perkins
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine, Rochester, NY 14642, USA.
| |
Collapse
|
26
|
Abstract
CCAAT/enhancer binding protein-ε (C/EBP-ε) is considered a master transcription factor regulating terminal neutrophil maturation. It is essential for expression of secondary granule proteins, but it also regulates proliferation, cell cycle, and maturation during granulopoiesis. Cebpe(-/-) mice have incomplete granulocytic differentiation and increased sensitivity toward bacterial infections. The amount of C/EBP-ε messenger RNA (mRNA) increases with maturation from myeloblasts with peak level in myelocytes (MC)/metamyelocytes (MM), when the cells stop proliferating followed by a decline in more mature cells. In contrast, C/EBP-ε protein is virtually detectable only in the MC/MM population, indicating that expression in more immature cells could be inhibited by microRNAs (miRNAs). We found that miRNA-130a (miR-130a) regulates C/EBP-ε protein expression in both murine and human granulocytic precursors. Overexpression of miR-130a in a murine cell line downregulated C/EBP-ε protein and lactoferrin (Ltf), cathelicidin antimicrobial protein (Camp), and lipocalin-2 (Lcn2) mRNA expression giving rise to cells with a more immature phenotype, as seen in the Cebpe(-/-) mouse. Introduction of a C/EBP-ε mRNA without target site for miR-130a restored both C/EBP-ε production, expression of Camp and Lcn2, and resulted in the cells having a more mature phenotype. We conclude that miR-130a is important for the regulation of the timed expression of C/EBP-ε during granulopoiesis.
Collapse
|
27
|
Lim JYS, Bhatia S, Robison LL, Yang JJ. Genomics of racial and ethnic disparities in childhood acute lymphoblastic leukemia. Cancer 2013; 120:955-62. [PMID: 24382716 DOI: 10.1002/cncr.28531] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 11/01/2013] [Accepted: 11/20/2013] [Indexed: 12/23/2022]
Abstract
Although the cure rates of childhood acute lymphoblastic leukemia (ALL) have improved dramatically in the past 40 years, not all children have benefited equally from this impressive progress. Racial and ethnic disparities in the incidence and treatment outcome of childhood ALL persist, with Hispanic children having an elevated risk of developing ALL and one of the lowest survival rates after ALL therapy. A critical barrier to progress is the lack of an understanding of the causes of ALL disparities, particularly racial and ethnic differences in ALL biology. In this review, the authors summarize the current knowledge on population variation in childhood ALL incidence and treatment outcome, discuss the contributing genetic and nongenetic variables, and highlight possible therapeutic interventions to mitigate disparities in ALL.
Collapse
Affiliation(s)
- Joshua Yew-Suang Lim
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee; Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | | | | |
Collapse
|
28
|
Grech G, Pollacco J, Portelli M, Sacco K, Baldacchino S, Grixti J, Saliba C. Expression of different functional isoforms in haematopoiesis. Int J Hematol 2013; 99:4-11. [PMID: 24293279 DOI: 10.1007/s12185-013-1477-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 11/18/2013] [Accepted: 11/18/2013] [Indexed: 12/26/2022]
Abstract
Haematopoiesis is a complex process regulated at various levels facilitating rapid responses to external factors including stress, modulation of lineage commitment and terminal differentiation of progenitors. Although the transcription program determines the RNA pool of a cell, various mRNA strands can be obtained from the same template, giving rise to multiple protein isoforms. The majority of variants and isoforms co-occur in normal haematopoietic cells or are differentially expressed at various maturity stages of progenitor maturation and cellular differentiation within the same lineage or across lineages. Genetic aberrations or specific cellular states result in the predominant expression of abnormal isoforms leading to deregulation and disease. The presence of upstream open reading frames (uORF) in 5' untranslated regions (UTRs) of a transcript, couples the utilization of start codons with the cellular status and availability of translation initiation factors (eIFs). In addition, tissue-specific and cell lineage-specific alternative promoter use, regulates several transcription factors producing transcript variants with variable 5' exons. In this review, we propose to give a detailed account of the differential isoform formation, causing haematological malignancies.
Collapse
Affiliation(s)
- Godfrey Grech
- Department of Pathology, Medical School, University of Malta, Msida, MSD2090, Malta,
| | | | | | | | | | | | | |
Collapse
|
29
|
Glass C, Wuertzer C, Cui X, Bi Y, Davuluri R, Xiao YY, Wilson M, Owens K, Zhang Y, Perkins A. Global Identification of EVI1 Target Genes in Acute Myeloid Leukemia. PLoS One 2013; 8:e67134. [PMID: 23826213 PMCID: PMC3694976 DOI: 10.1371/journal.pone.0067134] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 05/19/2013] [Indexed: 12/20/2022] Open
Abstract
The ecotropic virus integration site 1 (EVI1) transcription factor is associated with human myeloid malignancy of poor prognosis and is overexpressed in 8-10% of adult AML and strikingly up to 27% of pediatric MLL-rearranged leukemias. For the first time, we report comprehensive genomewide EVI1 binding and whole transcriptome gene deregulation in leukemic cells using a combination of ChIP-Seq and RNA-Seq expression profiling. We found disruption of terminal myeloid differentiation and cell cycle regulation to be prominent in EVI-induced leukemogenesis. Specifically, we identified EVI1 directly binds to and downregulates the master myeloid differentiation gene Cebpe and several of its downstream gene targets critical for terminal myeloid differentiation. We also found EVI1 binds to and downregulates Serpinb2 as well as numerous genes involved in the Jak-Stat signaling pathway. Finally, we identified decreased expression of several ATP-dependent P2X purinoreceptors genes involved in apoptosis mechanisms. These findings provide a foundation for future study of potential therapeutic gene targets for EVI1-induced leukemia.
Collapse
Affiliation(s)
- Carolyn Glass
- Department of Pathology and Lab Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Charles Wuertzer
- Department of Pathology and Lab Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Xiaohui Cui
- Department of Pathology and Lab Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Yingtao Bi
- Molecular and Cellular Oncogenesis Program, Center for Systems and Computational Biology The Wistar Institute, Philadelphia, Pennsylvania, United States of America
| | - Ramana Davuluri
- Molecular and Cellular Oncogenesis Program, Center for Systems and Computational Biology The Wistar Institute, Philadelphia, Pennsylvania, United States of America
| | - Ying-Yi Xiao
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Michael Wilson
- Department of Pathology and Lab Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Kristina Owens
- Department of Pathology and Lab Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Yi Zhang
- Department of Pathology and Lab Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Archibald Perkins
- Department of Pathology and Lab Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
| |
Collapse
|
30
|
Role of C/EBPβ-LAP and C/EBPβ-LIP in early adipogenic differentiation of human white adipose-derived progenitors and at later stages in immature adipocytes. Differentiation 2013; 85:20-31. [DOI: 10.1016/j.diff.2012.11.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Revised: 09/20/2012] [Accepted: 11/26/2012] [Indexed: 12/20/2022]
|
31
|
Alberich-Jordà M, Wouters B, Balastik M, Shapiro-Koss C, Zhang H, Di Ruscio A, DiRuscio A, Radomska HS, Ebralidze AK, Amabile G, Ye M, Zhang J, Lowers I, Avellino R, Melnick A, Figueroa ME, Valk PJM, Delwel R, Tenen DG. C/EBPγ deregulation results in differentiation arrest in acute myeloid leukemia. J Clin Invest 2012; 122:4490-504. [PMID: 23160200 DOI: 10.1172/jci65102] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 09/20/2012] [Indexed: 11/17/2022] Open
Abstract
C/EBPs are a family of transcription factors that regulate growth control and differentiation of various tissues. We found that C/EBPγ is highly upregulated in a subset of acute myeloid leukemia (AML) samples characterized by C/EBPα hypermethylation/silencing. Similarly, C/EBPγ was upregulated in murine hematopoietic stem/progenitor cells lacking C/EBPα, as C/EBPα mediates C/EBPγ suppression. Studies in myeloid cells demonstrated that CEBPG overexpression blocked neutrophilic differentiation. Further, downregulation of Cebpg in murine Cebpa-deficient stem/progenitor cells or in human CEBPA-silenced AML samples restored granulocytic differentiation. In addition, treatment of these leukemias with demethylating agents restored the C/EBPα-C/EBPγ balance and upregulated the expression of myeloid differentiation markers. Our results indicate that C/EBPγ mediates the myeloid differentiation arrest induced by C/EBPα deficiency and that targeting the C/EBPα-C/EBPγ axis rescues neutrophilic differentiation in this unique subset of AMLs.
Collapse
|
32
|
Kyme P, Thoennissen NH, Tseng CW, Thoennissen GB, Wolf AJ, Shimada K, Krug UO, Lee K, Müller-Tidow C, Berdel WE, Hardy WD, Gombart AF, Koeffler HP, Liu GY. C/EBPε mediates nicotinamide-enhanced clearance of Staphylococcus aureus in mice. J Clin Invest 2012; 122:3316-29. [PMID: 22922257 DOI: 10.1172/jci62070] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Accepted: 07/05/2012] [Indexed: 02/06/2023] Open
Abstract
The myeloid-specific transcription factor, CCAAT/enhancer-binding protein ε (C/EBPε) is a critical mediator of myelopoiesis. Mutation of this gene is responsible for neutrophil-specific granule deficiency in humans, a condition that confers susceptibility to Staphylococcus aureus infection. We found that C/EBPε-deficient mice are severely affected by infection with S. aureus, and C/EBPε deficiency in neutrophils contributes to the infectious phenotype. Conversely, exposure to the epigenetic modulator nicotinamide (vitamin B3) increased expression of C/EBPε in WT myeloid cells. Further, nicotinamide increased the activity of C/EBPε and select downstream antimicrobial targets, particularly in neutrophils. In a systemic murine infection model as well as in murine and human peripheral blood, nicotinamide enhanced killing of S. aureus by up to 1,000 fold but had no effect when administered to either C/EBPε-deficient mice or mice depleted of neutrophils. Nicotinamide was efficacious in both prophylactic and therapeutic settings. Our findings suggest that C/EBPε is an important target to boost killing of bacteria by the innate immune system.
Collapse
Affiliation(s)
- Pierre Kyme
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Maturity-dependent fractionation of neutrophil progenitors: A new method to examine in vivo expression profiles of differentiation-regulating genes. Exp Hematol 2012; 40:675-81. [DOI: 10.1016/j.exphem.2012.03.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Revised: 03/06/2012] [Accepted: 03/26/2012] [Indexed: 01/15/2023]
|
34
|
Site-specific CpG methylation in the CCAAT/enhancer binding protein delta (CEBPδ) CpG island in breast cancer is associated with metastatic relapse. Br J Cancer 2012; 107:732-8. [PMID: 22782348 PMCID: PMC3419957 DOI: 10.1038/bjc.2012.308] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background: The CCAAT/enhancer binding protein delta (CEBPδ) is a member of a highly conserved family of basic region leucine zipper transcription factors. It has properties consistent with a tumour suppressor; however, other data suggest that CEBPδ may be involved in the metastatic process. Methods: We analysed the expression of CEBPδ and the methylation status of the CpG island in human breast cancer cell lines, in 107 archival cases of primary breast cancer and in two series of metastatic breast cancers using qPCR and pyrosequencing. Results: Expression of CEBPδ is downregulated in primary breast cancer by site-specific methylation in the CEBPδ CpG island. Expression is also downregulated in 50% of cases during progression from primary carcinoma to metastatic lesions. The CEBPδ CpG island is methylated in 81% metastatic breast cancer lesions, while methylation in the CEBPδ CpG island in primary cancers is associated with increased risk of relapse and metastasis. Conclusion: CCAAT/enhancer binding protein delta CpG island methylation is associated with metastasis in breast cancer. Detection of methylated CEBPδ genomic DNA may have utility as an epigenetic biomarker of primary breast carcinomas at increased risk of relapse and metastasis.
Collapse
|
35
|
Nagy L, Szanto A, Szatmari I, Széles L. Nuclear hormone receptors enable macrophages and dendritic cells to sense their lipid environment and shape their immune response. Physiol Rev 2012; 92:739-89. [PMID: 22535896 DOI: 10.1152/physrev.00004.2011] [Citation(s) in RCA: 169] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
A key issue in the immune system is to generate specific cell types, often with opposing activities. The mechanisms of differentiation and subtype specification of immune cells such as macrophages and dendritic cells are critical to understand the regulatory principles and logic of the immune system. In addition to cytokines and pathogens, it is increasingly appreciated that lipid signaling also has a key role in differentiation and subtype specification. In this review we explore how intracellular lipid signaling via a set of transcription factors regulates cellular differentiation, subtype specification, and immune as well as metabolic homeostasis. We introduce macrophages and dendritic cells and then we focus on a group of transcription factors, nuclear receptors, which regulate gene expression upon receiving lipid signals. The receptors we cover are the ones with a recognized physiological function in these cell types and ones which heterodimerize with the retinoid X receptor. These are as follows: the receptor for a metabolite of vitamin A, retinoic acid: retinoic acid receptor (RAR), the vitamin D receptor (VDR), the fatty acid receptor: peroxisome proliferator-activated receptor γ (PPARγ), the oxysterol receptor liver X receptor (LXR), and their obligate heterodimeric partner, the retinoid X receptor (RXR). We discuss how they can get activated and how ligand is generated and eliminated in these cell types. We also explore how activation of a particular target gene contributes to biological functions and how the regulation of individual target genes adds up to the coordination of gene networks. It appears that RXR heterodimeric nuclear receptors provide these cells with a coordinated and interrelated network of transcriptional regulators for interpreting the lipid milieu and the metabolic changes to bring about gene expression changes leading to subtype and functional specification. We also show that these networks are implicated in various immune diseases and are amenable to therapeutic exploitation.
Collapse
Affiliation(s)
- Laszlo Nagy
- Department of Biochemistry and Molecular Biology, University of Debrecen, Medical and Health Science Center, Egyetem tér 1, Debrecen, Hungary.
| | | | | | | |
Collapse
|
36
|
Mozes-Koch R, Gover O, Tanne E, Peretz Y, Maori E, Chernin L, Sela I. Expression of an entire bacterial operon in plants. PLANT PHYSIOLOGY 2012; 158:1883-92. [PMID: 22353575 PMCID: PMC3320193 DOI: 10.1104/pp.111.186197] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Accepted: 02/06/2012] [Indexed: 05/26/2023]
Abstract
Multigene expression is required for metabolic engineering, i.e. coregulated expression of all genes in a metabolic pathway for the production of a desired secondary metabolite. To that end, several transgenic approaches have been attempted with limited success. Better success has been achieved by transforming plastids with operons. IL-60 is a platform of constructs driven from the geminivirus Tomato yellow leaf curl virus. We demonstrate that IL-60 enables nontransgenic expression of an entire bacterial operon in tomato (Solanum lycopersicum) plants without the need for plastid (or any other) transformation. Delivery to the plant is simple, and the rate of expressing plants is close to 100%, eliminating the need for selectable markers. Using this platform, we show the expression of an entire metabolic pathway in plants and delivery of the end product secondary metabolite (pyrrolnitrin). Expression of this unique secondary metabolite resulted in the appearance of a unique plant phenotype disease resistance. Pyrrolnitrin production was already evident 2 d after application of the operon to plants and persisted throughout the plant's life span. Expression of entire metabolic pathways in plants is potentially beneficial for plant improvement, disease resistance, and biotechnological advances, such as commercial production of desired metabolites.
Collapse
|
37
|
Venza M, Visalli M, Alafaci C, Caffo M, Caruso G, Salpietro FM, Tomasello F, Teti D. Interleukin-8 overexpression in astrocytomas is induced by prostaglandin E2 and is associated with the transcription factors CCAAT/enhancer-binding protein-β and CCAAT/enhancer-binding homologous protein. Neurosurgery 2011; 69:713-21; discussion 721. [PMID: 21471847 DOI: 10.1227/neu.0b013e31821954c6] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The upregulation of microsomal prostaglandin E synthase-1 (mPGES-1) and the overexpression of interleukin-8 (IL-8) have been separately linked to glioma malignancy. OBJECTIVE To evaluate (1) the correlation between the mRNA levels of IL-8, mPGES-1, and the main transcription factors (TFs) activating the IL-8 promoter in human brain tumors of different grades; (2) the role of prostaglandin E2 (PGE2) on IL-8 activation and the expression of these TFs in tumor-derived cells; and (3) the biological impact of PGE2 treatment and mPGES-1 silencing on IL-8 synthesis and tumorigenesis. METHODS Quantitative real-time polymerase chain reaction, transfection experiments, and cell proliferation and apoptosis assays were performed. RESULTS Regardless of histological grade, a significant positive association between IL-8 expression and mPGES-1, CCAAT/enhancer-binding protein-β (C/EBP-β) and C/EBP Homologous Protein (CHOP) mRNA levels was found only in astrogliomas (P < .001). The correlation was not significant in the other brain tumors. PGE2-treated astroglioma cells showed a marked upregulation of IL-8, C/EBP-β, and CHOP, as well as increased proliferation and decreased apoptosis compared with untreated cells. mPGES-1-silenced astroglioma cells displayed decreased IL-8 synthesis, accompanied by reduced cell growth and an increased rate of apoptosis. The other brain tumor cells were unaffected either by PGE2 treatment or by mPGES-1 knockout. CONCLUSION (1) PGE2 is responsible for IL-8 overexpression, independently of the malignancy grade, in astrogliomas only. (2) C/EBP-β and CHOP may be involved in mediating PGE2-induced IL-8 activation in these tumors. (3) mPGES-1 inhibition may have potential as a form of adjuvant therapy for astrogliomas.
Collapse
Affiliation(s)
- Mario Venza
- Department of Neurosciences, Psychiatry and Anaesthesiology, Neurosurgical Clinic, University of Messina, Messina, Italy
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Ceccarelli V, Racanicchi S, Martelli MP, Nocentini G, Fettucciari K, Riccardi C, Marconi P, Di Nardo P, Grignani F, Binaglia L, Vecchini A. Eicosapentaenoic acid demethylates a single CpG that mediates expression of tumor suppressor CCAAT/enhancer-binding protein delta in U937 leukemia cells. J Biol Chem 2011; 286:27092-102. [PMID: 21659508 DOI: 10.1074/jbc.m111.253609] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Polyunsaturated fatty acids (PUFAs) inhibit proliferation and induce differentiation in leukemia cells. To investigate the molecular mechanisms whereby fatty acids affect these processes, U937 leukemia cells were conditioned with stearic, oleic, linolenic, α-linolenic, arachidonic, eicosapentaenoic, and docosahexaenoic acids. PUFAs affected proliferation; eicosapentaenoic acid (EPA) was the most potent on cell cycle progression. EPA enhanced the expression of the myeloid lineage-specific transcription factors CCAAT/enhancer-binding proteins (C/EBPβ and C/EBPδ), PU.1, and c-Jun, resulting in increased expression of the monocyte lineage-specific target gene, the macrophage colony-stimulating factor receptor. Indeed, it is known that PU.1 and C/EBPs interact with their consensus sequences on a small DNA fragment of macrophage colony-stimulating factor receptor promoter, which is a determinant for expression. We demonstrated that C/EBPβ and C/EBPδ bind the same response element as a heterodimer. We focused on the enhanced expression of C/EBPδ, which has been reported to be a tumor suppressor gene silenced by promoter hypermethylation in U937 cells. After U937 conditioning with EPA and bisulfite sequencing of the -370/-20 CpG island on the C/EBPδ promoter region, we found a site-specific CpG demethylation that was a determinant for the binding activity of Sp1, an essential factor for C/EBPδ gene basal expression. Our results provide evidence for a new role of PUFAs in the regulation of gene expression. Moreover, we demonstrated for the first time that re-expression of the tumor suppressor C/EBPδ is controlled by the methylation state of a site-specific CpG dinucleotide.
Collapse
Affiliation(s)
- Veronica Ceccarelli
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia 06126, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Neutrophils are produced in the bone marrow from stem cells that proliferate and differentiate to mature neutrophils fully equipped with an armory of granules. These contain proteins that enable the neutrophil to deliver lethal hits against microorganisms, but also to cause great tissue damage. Neutrophils circulate in the blood as dormant cells. At sites of infection, endothelial cells capture bypassing neutrophils and guide them through the endothelial cell lining whereby the neutrophils are activated and tuned for the subsequent interaction with microbes. Once in tissues, neutrophils kill microorganisms by microbicidal agents liberated from granules or generated by metabolic activation. As a final act, neutrophils can extrude stands of DNA with bactericidal proteins attached that act as extracellular traps for microorganisms.
Collapse
Affiliation(s)
- Niels Borregaard
- The Granulocyte Research Laboratory, Department of Hematology, National University Hospital (Rigshospitalet), University of Copenhagen, DK-2100 Copenhagen, Denmark.
| |
Collapse
|
40
|
Abstract
Conserved upstream open reading frames (uORFs) are found within many eukaryotic transcripts and are known to regulate protein translation. Evidence from genetic and bioinformatic studies implicates disturbed uORF-mediated translational control in the etiology of human diseases. A genetic mouse model has recently provided proof-of-principle support for the physiological relevance of uORF-mediated translational control in mammals. The targeted disruption of the uORF initiation codon within the transcription factor CCAAT/enhancer binding protein β (C/EBPβ) gene resulted in deregulated C/EBPβ protein isoform expression, associated with defective liver regeneration and impaired osteoclast differentiation. The high prevalence of uORFs in the human transcriptome suggests that intensified search for mutations within 5' RNA leader regions may reveal a multitude of alterations affecting uORFs, causing pathogenic deregulation of protein expression.
Collapse
Affiliation(s)
- Klaus Wethmar
- Max Delbrueck Center for Molecular MedicineBerlin, Germany
- Charité, University Medicine BerlinGermany
| | - Jeske J Smink
- Max Delbrueck Center for Molecular MedicineBerlin, Germany
| | - Achim Leutz
- Max Delbrueck Center for Molecular MedicineBerlin, Germany
| |
Collapse
|
41
|
Halene S, Gaines P, Sun H, Zibello T, Lin S, Khanna-Gupta A, Williams SC, Perkins A, Krause D, Berliner N. C/EBPepsilon directs granulocytic-vs-monocytic lineage determination and confers chemotactic function via Hlx. Exp Hematol 2009; 38:90-103. [PMID: 19925846 DOI: 10.1016/j.exphem.2009.11.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2009] [Revised: 11/11/2009] [Accepted: 11/12/2009] [Indexed: 01/19/2023]
Abstract
OBJECTIVE Mutations in the CCAAT enhancer binding protein epsilon (C/EBPepsilon) gene have been identified in the cells of patients with neutrophil specific granule deficiency, a rare congenital disorder marked by recurrent bacterial infections. Their neutrophils, in addition to lacking specific granules required for normal respiratory burst activity, also lack normal phagocytosis and chemotaxis. Although the specific granule deficiency phenotype has been replicated in C/EBPepsilon(-/-) (knockout [KO]) mice, the mechanisms by which C/EBPepsilon mutations act to decrease neutrophil function are not entirely clear. MATERIALS AND METHODS In order to determine the role of C/EBPepsilon in neutrophil differentiation and migration, we generated immortalized progenitor cell lines from C/EBPepsilon KO and wild-type mice and performed expression and flow cytometric analysis and functional studies. RESULTS Expression of lineage-specific cell surface antigens on our in vitro differentiated cell lines revealed persistent expression of monocytic markers on KO granulocytes. We verified this in primary murine peripheral blood and bone marrow cells. In addition, KO bone marrow had an increase in immature myeloid precursors at the common myeloid progenitor and granulocyte/monocyte progenitor levels, suggesting a critical role for C/EBPepsilon not only in granulocyte maturation beyond the promyelocyte stage, but also in the monocyte/granulocyte lineage decision. We found that restoration of Hlx (H2.0-like homeo box 1) expression, which was decreased in C/EBPepsilon KO cells, rescued chemotaxis, but not the other defects of C/EBPepsilon KO neutrophils. CONCLUSIONS We show two new regulatory functions of C/EBPepsilon in myelopoiesis: in the absence of C/EBPepsilon, there is not only incomplete differentiation of granulocytes, but myelopoiesis is disrupted with the appearance of an intermediate cell type with monocyte and granulocyte features, and the neutrophils have abnormal chemotaxis. Restoration of expression of Hlx provides partial recovery of function; it has no effect on neutrophil maturation, but can completely ameliorate the chemotaxis defect in C/EBPepsilon KO cells.
Collapse
Affiliation(s)
- Stephanie Halene
- Section of Hematology, Department of Internal Medicine and Yale Cancer Center, Yale University School of Medicine, New Haven, CT 06520-8073, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Kimura A, Sultana TA. Granulocyte Colony-Stimulating Factor Receptors on CD34++Cells in Patients with Myelodysplastic Syndrome (MDS) and MDS-Acute Myeloid Leukemia. Leuk Lymphoma 2009; 45:1995-2000. [PMID: 15370243 DOI: 10.1080/10428190410001714034] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
We studied surface expression of granulocyte colony-stimulating factor receptor (G-CSFR) on CD34++ progenitor cells of myelodysplastic patients. Late stages of disease showed a higher proportion of high or low G-CSFR expression than early stages. Most of the patients with the low expression had neutropenia. Neutropenia was relatively less present in the normal group, but it reappeared in the high group. All the neutropenic patients in the high group showed response to G-CSF, while response in the normal group was minor. These findings suggest that lowered expression of G-CSFR leads to neutropenia in myelodysplastic patients. This article reviewed the knowledge of the G-CSFR and its role in the disorders of granulopoiesis, including myelodysplastic syndrome (MDS).
Collapse
Affiliation(s)
- Akiro Kimura
- Department of Hematology and Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Kasumi, Minami-ku, Japan.
| | | |
Collapse
|
43
|
Zahnow CA. CCAAT/enhancer-binding protein beta: its role in breast cancer and associations with receptor tyrosine kinases. Expert Rev Mol Med 2009; 11:e12. [PMID: 19351437 PMCID: PMC3095491 DOI: 10.1017/s1462399409001033] [Citation(s) in RCA: 143] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The CCAAT/enhancer-binding proteins (C/EBPs) are a family of leucine-zipper transcription factors that regulate gene expression to control cellular proliferation, differentiation, inflammation and metabolism. Encoded by an intronless gene, C/EBPbeta is expressed as several distinct protein isoforms (LAP1, LAP2, LIP) whose expression is regulated by the differential use of several in-frame translation start sites. LAP1 and LAP2 are transcriptional activators and are associated with differentiation, whereas LIP is frequently elevated in proliferative tissue and acts as a dominant-negative inhibitor of transcription. However, emerging evidence suggests that LIP can serve as a transcriptional activator in some cellular contexts, and that LAP1 and LAP2 might also have unique actions. The LIP:LAP ratio is crucial for the maintenance of normal growth and development, and increases in this ratio lead to aggressive forms of breast cancer. This review discusses the regulation of C/EBPbeta activity by post-translational modification, the individual actions of LAP1, LAP2 and LIP, and the functions and downstream targets that are unique to each isoform. The role of the C/EBPbeta isoforms in breast cancer is discussed and emphasis is placed on their interactions with receptor tyrosine kinases.
Collapse
Affiliation(s)
- Cynthia A Zahnow
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, The Bunting-Blaustein Cancer Research Building, 1650 Orleans St, Baltimore, MD 21231-1000, USA.
| |
Collapse
|
44
|
Liu Y, Nonnemacher MR, Wigdahl B. CCAAT/enhancer-binding proteins and the pathogenesis of retrovirus infection. Future Microbiol 2009; 4:299-321. [PMID: 19327116 PMCID: PMC2710768 DOI: 10.2217/fmb.09.4] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Previous studies indicate that two upstream CCAAT/enhancer-binding protein (C/EBP) sites and C/EBPbeta are required for subtype B HIV-1 gene expression in cells of the monocyte-macrophage lineage. The mechanisms of C/EBP regulation of HIV-1 transcription and replication remain unclear. This review focuses on studies concerning the role of C/EBP factors in HIV-1, human T-cell leukemia virus type 1, and SIV transcription in various cell types and tissues cultured in vitro, animal models and during human infection. The structure and function of the C/EBPbeta gene and the related protein isoforms are discussed along with the transcription factors, coactivators, viral proteins, cytokines and chemokines that affect C/EBP function.
Collapse
Affiliation(s)
- Yujie Liu
- Department of Microbiology & Immunology, Center for Molecular Virology & Neuroimmunology, Center for Cancer Biology, Philadelphia, PA 19129, USA
| | - Michael R Nonnemacher
- Department of Microbiology & Immunology, Center for Molecular Virology & Neuroimmunology, Center for Cancer Biology, Philadelphia, PA 19129, USA
| | - Brian Wigdahl
- Department of Microbiology & Immunology, Center for Molecular Virology & Neuroimmunology, Center for Cancer Biology, Philadelphia, PA 19129, USA, Tel.: +1 215 762 8399, Fax: +1 215 762 1955,
| |
Collapse
|
45
|
Kfoury N, Kapatos G. Identification of neuronal target genes for CCAAT/enhancer binding proteins. Mol Cell Neurosci 2009; 40:313-27. [PMID: 19103292 PMCID: PMC2703816 DOI: 10.1016/j.mcn.2008.11.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2008] [Revised: 11/04/2008] [Accepted: 11/05/2008] [Indexed: 01/19/2023] Open
Abstract
CCAAT/Enhancer Binding Proteins (C/EBPs) play pivotal roles in the development and plasticity of the nervous system. Identification of the physiological targets of C/EBPs (C/EBP target genes) should therefore provide insight into the underlying biology of these processes. We used unbiased genome-wide mapping to identify 115 C/EBPbeta target genes in PC12 cells that include transcription factors, neurotransmitter receptors, ion channels, protein kinases and synaptic vesicle proteins. C/EBPbeta binding sites were located primarily within introns, suggesting novel regulatory functions, and were associated with binding sites for other developmentally important transcription factors. Experiments using dominant negatives showed C/EBPbeta to repress transcription of a subset of target genes. Target genes in rat brain were subsequently found to preferentially bind C/EBPalpha, beta and delta. Analysis of the hippocampal transcriptome of C/EBPbeta knockout mice revealed dysregulation of a high percentage of transcripts identified as C/EBP target genes. These results support the hypothesis that C/EBPs play non-redundant roles in the brain.
Collapse
Affiliation(s)
- Najla Kfoury
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| | | |
Collapse
|
46
|
Bedi R, Du J, Sharma AK, Gomes I, Ackerman SJ. Human C/EBP-epsilon activator and repressor isoforms differentially reprogram myeloid lineage commitment and differentiation. Blood 2009; 113:317-27. [PMID: 18832658 PMCID: PMC2615649 DOI: 10.1182/blood-2008-02-139741] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2008] [Accepted: 09/08/2008] [Indexed: 11/20/2022] Open
Abstract
CCAAT enhancer-binding protein-epsilon (C/EBP-epsilon) is required for the terminal differentiation of neutrophils and eosinophils. Human C/EBP-epsilon is expressed as 4 isoforms (32, 30, 27, and 14 kDa) through differential RNA splicing, and alternative promoters and translational start sites. The C/EBP-epsilon(32/30) isoforms are transcriptional activators, whereas C/EBP-epsilon(27) interacts with and represses GATA-1 transactivation of eosinophil promoters. C/EBP-epsilon(14) contains only DNA-binding and -dimerization domains and may function as a dominant-negative regulator. To define functional activities for these C/EBP-epsilon isoforms in myelopoiesis, human CD34(+) progenitors were transduced with internal ribosomal entry site-enhanced green fluorescent protein retroviral vectors encoding the 32/30, 27, and 14-kDa isoforms, purified by fluorescence-activated cell sorter, and analyzed in colony-forming assays and suspension cultures. Progenitors transduced with C/EBP-epsilon(32/30) default exclusively to eosinophil differentiation and gene expression, independent of interleukin-5, and regardless of inclusion of cytokines to induce other lineages. In contrast, the putative repressor C/EBP-epsilon(27) isoform strongly inhibits eosinophil differentiation and gene expression, including GATA-1, promoting granulocyte (neutrophil)-macrophage differen-tiation. The C/EBP-epsilon(14) repressor isoform strongly inhibits eosinophil development and gene expression, promoting erythroid differentiation, an effect enhanced by erythropoietin. Thus, C/EBP-epsilon isoforms can reprogram myeloid lineage commitment and differentiation consistent with their predicted activities based on activator and repressor domains and in vitro functional activities.
Collapse
Affiliation(s)
- Richa Bedi
- Department of Biochemistry and Molecular Genetics, Section of Hematology-Oncology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | | | | | | | | |
Collapse
|
47
|
Milanovic M, Terszowski G, Struck D, Liesenfeld O, Carstanjen D. IFN consensus sequence binding protein (Icsbp) is critical for eosinophil development. THE JOURNAL OF IMMUNOLOGY 2008; 181:5045-53. [PMID: 18802108 DOI: 10.4049/jimmunol.181.7.5045] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
IFN consensus sequence binding protein (Icsbp) (IFN response factor-8) is a hematopoietic transcription factor with dual functions in myelopoiesis and immunity. In this study, we report a novel role of Icsbp in regulating the development of eosinophils. Loss of Icsbp in mice leads to a reduction of eosinophils in different tissues. During parasite infection with the nematode Nippostrongylus brasiliensis, Icsbp-deficient mice fail to mount eosinophilia despite a vigorous IL-5 response. Numbers of phenotypically defined eosinophil progenitors are decreased and those progenitors have, on a per-cell basis, reduced eosinophil differentiation potential. The transcription factor Gata1, crucial for eosinophil development, is reduced expressed in committed eosinophil progenitors in wells as mature eosinophils. These findings identify Icsbp as a novel transcription factor critical for the development of the eosinophil lineage.
Collapse
Affiliation(s)
- Maja Milanovic
- Leibniz-Forschungsinstitut fuer Molekulare Pharmakologie, Berlin, Germany
| | | | | | | | | |
Collapse
|
48
|
Cloutier A, Guindi C, Larivée P, Dubois CM, Amrani A, McDonald PP. Inflammatory Cytokine Production by Human Neutrophils Involves C/EBP Transcription Factors. THE JOURNAL OF IMMUNOLOGY 2008; 182:563-71. [DOI: 10.4049/jimmunol.182.1.563] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
49
|
Chen S, Han YH, Zheng Y, Zhao M, Yan H, Zhao Q, Chen GQ, Li D. NDRG1 contributes to retinoic acid-induced differentiation of leukemic cells. Leuk Res 2008; 33:1108-13. [PMID: 19046768 DOI: 10.1016/j.leukres.2008.10.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2008] [Revised: 10/17/2008] [Accepted: 10/18/2008] [Indexed: 12/21/2022]
Abstract
N-Myc downstream-regulated gene 1 (NDRG1) protein has been shown to be up-regulated during leukemic cell differentiation induced by some differentiation-inducing agents such as all-trans retinoic acid (ATRA). However, the potential role of up-regulated NDRG1 in the event is greatly unknown. In this work, we show that inducible NDRG1 expression can drive leukemic U937 cells to undergo differentiation, while the knock-down of NDRG1 expression by specific small interfering RNA significantly antagonizes ATRA-induced differentiation of leukemic cells, proposing the role of NDRG1 in leukemic cell differentiation. Furthermore, our work shows that CCAAT/enhancer-binding protein beta (C/EBPbeta) and PU.1, which are important hematopoiesis-related transcription factors, may act as downstream effectors of NDRG1 in leukemic cell differentiation. Taking together, this study provides direct evidence for the role of NDRG1 protein in myeloid leukemic cell differentiation.
Collapse
Affiliation(s)
- Su Chen
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao-Tong University School of Medicine, Luwan, Shanghai, China
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Arcasoy MO, Maun NA, Perez L, Forget BG, Berliner N. Erythropoietin mediates terminal granulocytic differentiation of committed myeloid cells with ectopic erythropoietin receptor expression. Eur J Haematol 2008. [DOI: 10.1034/j.1600-0609.2001.t01-1-00491.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|