1
|
Arnab SP, Campelo dos Santos AL, Fumagalli M, DeGiorgio M. Efficient Detection and Characterization of Targets of Natural Selection Using Transfer Learning. Mol Biol Evol 2025; 42:msaf094. [PMID: 40341942 PMCID: PMC12062966 DOI: 10.1093/molbev/msaf094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 04/16/2025] [Accepted: 04/17/2025] [Indexed: 05/11/2025] Open
Abstract
Natural selection leaves detectable patterns of altered spatial diversity within genomes, and identifying affected regions is crucial for understanding species evolution. Recently, machine learning approaches applied to raw population genomic data have been developed to uncover these adaptive signatures. Convolutional neural networks (CNNs) are particularly effective for this task, as they handle large data arrays while maintaining element correlations. However, shallow CNNs may miss complex patterns due to their limited capacity, while deep CNNs can capture these patterns but require extensive data and computational power. Transfer learning addresses these challenges by utilizing a deep CNN pretrained on a large dataset as a feature extraction tool for downstream classification and evolutionary parameter prediction. This approach reduces extensive training data generation requirements and computational needs while maintaining high performance. In this study, we developed TrIdent, a tool that uses transfer learning to enhance detection of adaptive genomic regions from image representations of multilocus variation. We evaluated TrIdent across various genetic, demographic, and adaptive settings, in addition to unphased data and other confounding factors. TrIdent demonstrated improved detection of adaptive regions compared to recent methods using similar data representations. We further explored model interpretability through class activation maps and adapted TrIdent to infer selection parameters for identified adaptive candidates. Using whole-genome haplotype data from European and African populations, TrIdent effectively recapitulated known sweep candidates and identified novel cancer, and other disease-associated genes as potential sweeps.
Collapse
Affiliation(s)
- Sandipan Paul Arnab
- Department of Electrical Engineering and Computer Science, Florida Atlantic University, Boca Raton, FL, USA
| | | | - Matteo Fumagalli
- School of Biological and Behavioural Sciences, Queen Mary University of London, London, UK
- The Alan Turing Institute, London, UK
| | - Michael DeGiorgio
- Department of Electrical Engineering and Computer Science, Florida Atlantic University, Boca Raton, FL, USA
| |
Collapse
|
2
|
G C B, Du P, Zhang Y, Yang L, Dong F. Bcl-xL is important for the antiapoptotic activity of Gfi1 and is upregulated by Gfi1 through hemgn. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkae066. [PMID: 40262274 DOI: 10.1093/jimmun/vkae066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 12/16/2024] [Indexed: 04/24/2025]
Abstract
Gfi1 is a transcriptional repressor that plays a critical role in hematopoiesis. Gfi1 represses its target genes primarily through interacting with the histone demethylase LSD1 via its SNAG domain. A major function of Gfi1 is to inhibit DNA damage-induced apoptosis through its involvement in post-translational modifications and subsequent inhibition of p53 protein, and in PRMT1-dependent methylation of MRE11 and 53BP1, which is necessary for these proteins to function in DNA repair. We show here that Gfi1 inhibited apoptosis induced not only by DNA damage but also by growth factor withdrawal, inhibitory cytokine TGF-β and MYC activation. We further demonstrate that Gfi1 upregulated the expression of the pro-survival Bcl-2 family member Bcl-xL in a manner that was independent of p53. Bcl-xL overexpression partially rescued the hypersensitivity to DNA damage of Gfi1-knocked down leukemic cells and Gfi1-deficient mouse primary bone marrow (BM) cells. In contrast, Bcl-xL knockdown partially abolished the protective effect of Gfi1 on DNA damage-induced apoptosis. Notably, interaction with LSD1 was required and sufficient for Gfi1-mediaed upregulation of Bcl-xL, suggesting that Gfi1 may augment Bcl-xL expression by an indirect mechanism. We further demonstrate that Bcl-xL upregulation by Gfi1 was dependent on Hemgn upregulation, which results from Gfi1-mediated repression of PU.1. Our data reveal a novel mechanism by which Gfi1 inhibits apoptosis.
Collapse
Affiliation(s)
- Binod G C
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States
| | - Pei Du
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States
| | - Yangyang Zhang
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States
| | - Li Yang
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Fan Dong
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States
| |
Collapse
|
3
|
Arnab SP, Dos Santos ALC, Fumagalli M, DeGiorgio M. Efficient detection and characterization of targets of natural selection using transfer learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.05.641710. [PMID: 40093065 PMCID: PMC11908262 DOI: 10.1101/2025.03.05.641710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Natural selection leaves detectable patterns of altered spatial diversity within genomes, and identifying affected regions is crucial for understanding species evolution. Recently, machine learning approaches applied to raw population genomic data have been developed to uncover these adaptive signatures. Convolutional neural networks (CNNs) are particularly effective for this task, as they handle large data arrays while maintaining element correlations. However, shallow CNNs may miss complex patterns due to their limited capacity, while deep CNNs can capture these patterns but require extensive data and computational power. Transfer learning addresses these challenges by utilizing a deep CNN pre-trained on a large dataset as a feature extraction tool for downstream classification and evolutionary parameter prediction. This approach reduces extensive training data generation requirements and computational needs while maintaining high performance. In this study, we developed TrIdent, a tool that uses transfer learning to enhance detection of adaptive genomic regions from image representations of multilocus variation. We evaluated TrIdent across various genetic, demographic, and adaptive settings, in addition to unphased data and other confounding factors. TrIdent demonstrated improved detection of adaptive regions compared to recent methods using similar data representations. We further explored model interpretability through class activation maps and adapted TrIdent to infer selection parameters for identified adaptive candidates. Using whole-genome haplotype data from European and African populations, TrIdent effectively recapitulated known sweep candidates and identified novel cancer, and other disease-associated genes as potential sweeps.
Collapse
Affiliation(s)
- Sandipan Paul Arnab
- Department of Electrical Engineering and Computer Science, Florida Atlantic University, Boca Raton, FL, USA
| | | | - Matteo Fumagalli
- School of Biological and Behavioural Sciences, Queen Mary University of London, London, UK
- The Alan Turing Institute, London, UK
| | - Michael DeGiorgio
- Department of Electrical Engineering and Computer Science, Florida Atlantic University, Boca Raton, FL, USA
| |
Collapse
|
4
|
G C B, Hoyt LJ, Dovat S, Dong F. Upregulation of nuclear protein Hemgn by transcriptional repressor Gfi1 through repressing PU.1 contributes to the anti-apoptotic activity of Gfi1. J Biol Chem 2024; 300:107860. [PMID: 39374784 PMCID: PMC11550643 DOI: 10.1016/j.jbc.2024.107860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/20/2024] [Accepted: 09/27/2024] [Indexed: 10/09/2024] Open
Abstract
Gfi1 is a transcriptional repressor that plays a critical role in hematopoiesis. The repressive activity of Gfi1 is mediated mainly by its SNAG domain that interacts with and thereby recruits the histone demethylase LSD1 to its target genes. An important function of Gfi1 is to protect hematopoietic cells against stress-induced apoptosis, which has been attributed to its participation in the posttranscriptional modifications of p53 protein, leading to suppression of p53 activity. In this study, we show that Gfi1 upregulated the expression of Hemgn, a nuclear protein, through a 16-bp promoter region spanning from +47 to +63 bp relative to the transcription start site (TSS), which was dependent on its interaction with LSD1. We further demonstrate that Gfi1, Ikaros, and PU.1 are bound to this 16-bp region. However, while Ikaros activated Hemgn and collaborated with Gfi1 to augment Hemgn expression, it was not required for Gfi1-mediated Hemgn upregulation. In contrast, PU.1 repressed Hemgn and inhibited Hemgn upregulation by Gfi1. Notably, PU.1 knockdown and deficiency, while augmenting Hemgn expression, abolished Hemgn upregulation by Gfi1. PU.1 (Spi-1) is repressed by Gfi1. We show here that PU.1 repression by Gfi1 preceded and correlated well with Hemgn upregulation. Thus, our data strongly suggest that Gfi1 upregulates Hemgn by repressing PU.1. In addition, we demonstrate that Hemgn upregulation contributed to the anti-apoptotic activity of Gfi1 in a p53-independent manner.
Collapse
Affiliation(s)
- Binod G C
- Department of Biological Sciences, University of Toledo, Toledo, Ohio, USA
| | - Laney Jia Hoyt
- Department of Biological Sciences, University of Toledo, Toledo, Ohio, USA
| | - Sinisa Dovat
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Fan Dong
- Department of Biological Sciences, University of Toledo, Toledo, Ohio, USA.
| |
Collapse
|
5
|
Venati SR, Uversky VN. Exploring Intrinsic Disorder in Human Synucleins and Associated Proteins. Int J Mol Sci 2024; 25:8399. [PMID: 39125972 PMCID: PMC11313516 DOI: 10.3390/ijms25158399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/27/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
In this work, we explored the intrinsic disorder status of the three members of the synuclein family of proteins-α-, β-, and γ-synucleins-and showed that although all three human synucleins are highly disordered, the highest levels of disorder are observed in γ-synuclein. Our analysis of the peculiarities of the amino acid sequences and modeled 3D structures of the human synuclein family members revealed that the pathological mutations A30P, E46K, H50Q, A53T, and A53E associated with the early onset of Parkinson's disease caused some increase in the local disorder propensity of human α-synuclein. A comparative sequence-based analysis of the synuclein proteins from various evolutionary distant species and evaluation of their levels of intrinsic disorder using a set of commonly used bioinformatics tools revealed that, irrespective of their origin, all members of the synuclein family analyzed in this study were predicted to be highly disordered proteins, indicating that their intrinsically disordered nature represents an evolutionary conserved and therefore functionally important feature. A detailed functional disorder analysis of the proteins in the interactomes of the human synuclein family members utilizing a set of commonly used disorder analysis tools showed that the human α-synuclein interactome has relatively higher levels of intrinsic disorder as compared with the interactomes of human β- and γ- synucleins and revealed that, relative to the β- and γ-synuclein interactomes, α-synuclein interactors are involved in a much broader spectrum of highly diversified functional pathways. Although proteins interacting with three human synucleins were characterized by highly diversified functionalities, this analysis also revealed that the interactors of three human synucleins were involved in three common functional pathways, such as the synaptic vesicle cycle, serotonergic synapse, and retrograde endocannabinoid signaling. Taken together, these observations highlight the critical importance of the intrinsic disorder of human synucleins and their interactors in various neuronal processes.
Collapse
Affiliation(s)
- Sriya Reddy Venati
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA;
| | - Vladimir N. Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA;
- USF Health Byrd Alzheimer’s Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| |
Collapse
|
6
|
James AM, Schmid EW, Walter JC, Farnung L. In silico screening identifies SHPRH as a novel nucleosome acidic patch interactor. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.26.600687. [PMID: 38979307 PMCID: PMC11230416 DOI: 10.1101/2024.06.26.600687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Nucleosomes are the fundamental unit of eukaryotic chromatin. Diverse factors interact with nucleosomes to modulate chromatin architecture and facilitate DNA repair, replication, transcription, and other cellular processes. An important platform for chromatin binding is the H2A-H2B acidic patch. Here, we used AlphaFold-Multimer to screen over 7000 human proteins for nucleosomal acidic patch binding and identify 41 potential acidic patch binders. We determined the cryo-EM structure of one hit, SHPRH, with the nucleosome at 2.8 Å. The structure confirms the predicted acidic patch interaction, reveals that the SHPRH ATPase engages a different nucleosomal DNA location than other SF2-type ATPases, and clarifies the roles of SHPRH's domains in nucleosome recognition. Our results illustrate the use of in silico screening as a high throughput method to identify specific interaction types and expands the set of potential acidic patch binding factors. All the screening data is freely available at https://predictomes.org/view/acidicpatch.
Collapse
|
7
|
Fomukong HA, Kalu M, Aimola IA, Sallau AB, Bello-Manga H, Gouegni FE, Ameloko JU, Bello ZK, David AU, Baba RS. Single-cell RNA seq analysis of erythroid cells reveals a specific sub-population of stress erythroid progenitors. Hematology 2023; 28:2261802. [PMID: 37791839 DOI: 10.1080/16078454.2023.2261802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 09/18/2023] [Indexed: 10/05/2023] Open
Abstract
BACKGROUND : Erythroid cells play important roles in hemostasis and disease. However, there is still significant knowledge gap regarding stress erythropoiesis. METHODS : Two single-cell RNAseq datasets of erythroid cells on GEO with accession numbers GSE149938 and GSE184916 were obtained. The datasets from two sources, bone marrow and peripheral blood were analyzed using Seurat v4.1.1, and other tools in R. QC metrics were performed, data were normalized and scaled. Principal components that capture the variation of the data were determined. In clustering the cells, KNN graph was constructed and Louvain algorithm was applied to optimize the standard modularity function. Clusters were defined via differential expression of features. RESULTS We identified 9 different cell types, with a particular cluster representing the stress erythroids. The clusters showed differentially expressed genes as observed from the gene signature plot. The stress erythroid cluster differentially expressed some genes including ALAS2, HEMGN, and GUK1. CONCLUSION The erythroid population was found to be heterogeneous, with a distinct sub-cell type constituting the stress erythroids; this may have important implications for our knowledge of steady-state and stress erythropoiesis, and the markers found in this cluster may prove useful for future research into the dynamics of stress erythroid progenitor cell differentiation.
Collapse
Affiliation(s)
- Hanneda A Fomukong
- Department of Biochemistry, Ahmadu Bello University, Kaduna, Nigeria
- Africa Center of Excellence for Neglected Tropical Diseases and Forensic Biotechnology, Ahmadu Bello University, Kaduna, Nigeria
| | - Mayen Kalu
- Department of Biochemistry, Ahmadu Bello University, Kaduna, Nigeria
- Africa Center of Excellence for Neglected Tropical Diseases and Forensic Biotechnology, Ahmadu Bello University, Kaduna, Nigeria
| | - Idowu A Aimola
- Department of Biochemistry, Ahmadu Bello University, Kaduna, Nigeria
- Africa Center of Excellence for Neglected Tropical Diseases and Forensic Biotechnology, Ahmadu Bello University, Kaduna, Nigeria
| | - Abdullahi B Sallau
- Department of Biochemistry, Ahmadu Bello University, Kaduna, Nigeria
- Africa Center of Excellence for Neglected Tropical Diseases and Forensic Biotechnology, Ahmadu Bello University, Kaduna, Nigeria
| | | | - Flore E Gouegni
- Department of Biochemistry, Ahmadu Bello University, Kaduna, Nigeria
- Africa Center of Excellence for Neglected Tropical Diseases and Forensic Biotechnology, Ahmadu Bello University, Kaduna, Nigeria
| | - Joy U Ameloko
- Department of Biochemistry, Ahmadu Bello University, Kaduna, Nigeria
- Africa Center of Excellence for Neglected Tropical Diseases and Forensic Biotechnology, Ahmadu Bello University, Kaduna, Nigeria
| | - Zeenat K Bello
- Department of Biochemistry, Ahmadu Bello University, Kaduna, Nigeria
- Africa Center of Excellence for Neglected Tropical Diseases and Forensic Biotechnology, Ahmadu Bello University, Kaduna, Nigeria
| | - Alfa U David
- Department of Biochemistry, Ahmadu Bello University, Kaduna, Nigeria
- Africa Center of Excellence for Neglected Tropical Diseases and Forensic Biotechnology, Ahmadu Bello University, Kaduna, Nigeria
| | - Reuben S Baba
- Department of Biochemistry, Ahmadu Bello University, Kaduna, Nigeria
| |
Collapse
|
8
|
Jiang Y, Yu L, Hu Q, Kang Y, You J, Huang C, Xu X, Chen L. Cancer/testis antigen HEMGN correlated with immune infiltration serves as a prognostic biomarker in lung adenocarcinoma. Mol Immunol 2023; 153:226-237. [PMID: 36563642 DOI: 10.1016/j.molimm.2022.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 12/06/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022]
Abstract
HEMGN belongs to the Cancer/testis antigens (CTAs), which are expressed in various types of human cancers and have received particular attention in cancer immunotherapy. However, the potential function of HEMGN involved in lung cancer and the immune response is not yet elucidated. HEMGN expression in lung adenocarcinoma (LUAD) was estimated via the Tumor Immune Estimation Resource (TIMER), The Cancer Genome Atlas (TCGA), The University of Alabama at Birmingham Cancer data analysis Portal (UALCAN), and Human Protein Atlas databases. The prognostic role of HEMGN was investigated by Gene Expression Profiling Interactive Analysis (GEPIA), PrognoScan, and Kaplan-Meier plotter databases. The associations between HEMGN and clinicopathological parameters were analyzed with UALCAN database. Then, immunohistochemical and Real-Time Quantitative Reverse Transcription PCR (qRT-PCR) analysis were performed to further verify the associations in tissue or serum samples. Serum from patients were detected for HEMGN antibody by enzyme-linked immunosorbent assay (ELISA). Flow cytometry was used to detect immune cell infiltration in peripheral blood of patients with LUAD. In addition, Gene Set Enrichment Analysis (GSEA) was conducted to investigate the functional role of HEMGN. Furthermore, we obtained the somatic mutation data from the TCGA LUAD dataset and analyzed the mutation profiles with "maftools" package. Finally, we evaluated the associations between HEMGN and immune infiltration level and the characteristic markers of immune cells in TIMER, GEPIA, and CIBERSORT. The mRNA and protein expressions of HEMGN were significantly decreased in LUAD patients. High HEMGN expression was remarkably associated with better prognosis in LUAD patients. The concentration levels of anti-HEMGN antibody in LUAD were significantly higher than that in healthy individuals and were closely correlated with clinical stage. In addition, HEMGN was involved in distinct typical genomic alterations in LUAD. GSEA demonstrated that HEMGN was significantly connected with immunity and substance metabolism. Notably, HEMGN was significantly related to immune infiltrates, including B cells, CD8 + T cells, CD4 + T cells, neutrophils, macrophages, dendritic cells (DCs), and various kinds of functional T cells. Furthermore, HEMGN had a significant association with diverse immune gene markers. HEMGN can be considered as a prognostic biomarker of LUAD and is associated with immune infiltration.
Collapse
Affiliation(s)
- Yingfeng Jiang
- Department of Clinical Laboratory, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Lili Yu
- Department of Thoracic Surgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Qingfeng Hu
- Department of Clinical Laboratory, Maternal and Child Health Hospital of Xianyou County, Putian, China
| | - Yanli Kang
- Department of Clinical Laboratory, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Jianbin You
- Department of Clinical Laboratory, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Chen Huang
- Department of Thoracic Surgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Xunyu Xu
- Department of Thoracic Surgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China.
| | - Liangyuan Chen
- Department of Clinical Laboratory, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China.
| |
Collapse
|
9
|
Hemogen /BRG1 cooperativity modulates promoter and enhancer activation during erythropoiesis. Blood 2022; 139:3532-3545. [PMID: 35297980 DOI: 10.1182/blood.2021014308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 03/15/2022] [Indexed: 11/20/2022] Open
Abstract
Hemogen, also known as EDAG, is a hematopoietic tissue-specific gene that regulates the proliferation and differentiation of hematopoietic cells. However, the mechanism underlying hemogen function in erythropoiesis is unknown. We found that depletion of hemogen in human CD34+ erythroid progenitor cells and HUDEP2 cells significantly reduced the expression of genes associated with heme and hemoglobin synthesis, supporting a positive role of hemogen in erythroid maturation. In human K562 cells, hemogen antagonized the occupancy of co-repressors NuRD complex and facilitated LDB1 complex-mediated chromatin looping. Hemogen recruited SWI/SNF complex ATPase BRG1 as a co-activator to regulate nucleosome accessibility and H3K27ac enrichment for promoter and enhancer activity. To ask if hemogen/BRG1 cooperativity is conserved in mammalian systems, we generated hemogen KO/KI mice and investigated hemogen/BRG1 function in murine erythropoiesis. Loss of hemogen in E12.5-E16.5 fetal liver cells impeded erythroid differentiation through reducing the production of mature erythroblasts. ChIP-seq in WT and hemogen KO animal revealed BRG1 is largely dependent on hemogen to regulate chromatin accessibility at erythroid gene promoters and enhancers. In summary, hemogen/BRG1 interaction in mammals is essential for fetal erythroid maturation and hemoglobin production through its active role in promoter and enhancer activity and chromatin organization.
Collapse
|
10
|
Zhao K, Liu J, Zhu Y, Dong X, Yin R, Liu X, Gao H, Xiao F, Gao R, Wang Q, Zhan Y, Yu M, Chen H, Ning H, Zhang C, Yang X, Li C. Hemgn Protects Hematopoietic Stem and Progenitor Cells Against Transplantation Stress Through Negatively Regulating IFN-γ Signaling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103838. [PMID: 34923767 PMCID: PMC8844507 DOI: 10.1002/advs.202103838] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/14/2021] [Indexed: 06/14/2023]
Abstract
Hematopoietic stem and progenitor cells (HSPCs) possess the remarkable ability to regenerate the whole blood system in response to ablated stress demands. Delineating the mechanisms that maintain HSPCs during regenerative stresses is increasingly important. Here, it is shown that Hemgn is significantly induced by hematopoietic stresses including irradiation and bone marrow transplantation (BMT). Hemgn deficiency does not disturb steady-state hematopoiesis in young mice. Hemgn-/- HSPCs display defective engraftment activity during BMT with reduced homing and survival and increased apoptosis. Transcriptome profiling analysis reveals that upregulated genes in transplanted Hemgn-/- HSPCs are enriched for gene sets related to interferon gamma (IFN-γ) signaling. Hemgn-/- HSPCs show enhanced responses to IFN-γ treatment and increased aging over time. Blocking IFN-γ signaling in irradiated recipients either pharmacologically or genetically rescues Hemgn-/- HSPCs engraftment defect. Mechanistical studies reveal that Hemgn deficiency sustain nuclear Stat1 tyrosine phosphorylation via suppressing T-cell protein tyrosine phosphatase TC45 activity. Spermidine, a selective activator of TC45, rescues exacerbated phenotype of HSPCs in IFN-γ-treated Hemgn-/- mice. Collectively, these results identify that Hemgn is a critical regulator for successful engraftment and reconstitution of HSPCs in mice through negatively regulating IFN-γ signaling. Targeted Hemgn may be used to improve conditioning regimens and engraftment during HSPCs transplantation.
Collapse
Affiliation(s)
- Ke Zhao
- State Key Laboratory of ProteomicsBeijing Proteome Research CenterNational Center for Protein Sciences (Beijing)Beijing Institute of LifeomicsBeijing102206China
| | - Jin‐Fang Liu
- State Key Laboratory of ProteomicsBeijing Proteome Research CenterNational Center for Protein Sciences (Beijing)Beijing Institute of LifeomicsBeijing102206China
| | - Ya‐Xin Zhu
- School of Life SciencesHebei UniversityNo. 180 Wusi Dong Road, Lian Chi DistrictBaoding CityHebei Province071000China
| | - Xiao‐Ming Dong
- College of Life SciencesShanxi Normal UniversityNo. 199, South Chang'an Road, Yanta DistrictXi'an710062China
| | - Rong‐Hua Yin
- State Key Laboratory of ProteomicsBeijing Proteome Research CenterNational Center for Protein Sciences (Beijing)Beijing Institute of LifeomicsBeijing102206China
| | - Xian Liu
- State Key Laboratory of ProteomicsBeijing Proteome Research CenterNational Center for Protein Sciences (Beijing)Beijing Institute of LifeomicsBeijing102206China
| | - Hui‐Ying Gao
- State Key Laboratory of ProteomicsBeijing Proteome Research CenterNational Center for Protein Sciences (Beijing)Beijing Institute of LifeomicsBeijing102206China
| | - Feng‐Jun Xiao
- Department of Experimental Hematology and BiochemistryBeijing Institute of Radiation MedicineBeijing100850China
| | - Rui Gao
- State Key Laboratory of ProteomicsBeijing Proteome Research CenterNational Center for Protein Sciences (Beijing)Beijing Institute of LifeomicsBeijing102206China
| | - Qi Wang
- An Hui Medical UniversitySchool of Basic Medical SciencesHefei230032China
| | - Yi‐Qun Zhan
- State Key Laboratory of ProteomicsBeijing Proteome Research CenterNational Center for Protein Sciences (Beijing)Beijing Institute of LifeomicsBeijing102206China
| | - Miao Yu
- State Key Laboratory of ProteomicsBeijing Proteome Research CenterNational Center for Protein Sciences (Beijing)Beijing Institute of LifeomicsBeijing102206China
| | - Hui Chen
- State Key Laboratory of ProteomicsBeijing Proteome Research CenterNational Center for Protein Sciences (Beijing)Beijing Institute of LifeomicsBeijing102206China
| | - Hong‐Mei Ning
- Department of Hematopoietic Stem Cell TransplantationThe Fifth Medical Center of Chinese PLA General HospitalBeijing100071China
| | - Cai‐Bo Zhang
- Department of Life SciencesQilu Normal UniversityNo. 2, Wenbo Road, Zhangqiu DistrictJinanShandong250013China
| | - Xiao‐Ming Yang
- State Key Laboratory of ProteomicsBeijing Proteome Research CenterNational Center for Protein Sciences (Beijing)Beijing Institute of LifeomicsBeijing102206China
| | - Chang‐Yan Li
- State Key Laboratory of ProteomicsBeijing Proteome Research CenterNational Center for Protein Sciences (Beijing)Beijing Institute of LifeomicsBeijing102206China
- School of Life SciencesHebei UniversityNo. 180 Wusi Dong Road, Lian Chi DistrictBaoding CityHebei Province071000China
- An Hui Medical UniversitySchool of Basic Medical SciencesHefei230032China
| |
Collapse
|
11
|
Mughal MR, Koch H, Huang J, Chiaromonte F, DeGiorgio M. Learning the properties of adaptive regions with functional data analysis. PLoS Genet 2020; 16:e1008896. [PMID: 32853200 PMCID: PMC7480868 DOI: 10.1371/journal.pgen.1008896] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 09/09/2020] [Accepted: 05/29/2020] [Indexed: 12/12/2022] Open
Abstract
Identifying regions of positive selection in genomic data remains a challenge in population genetics. Most current approaches rely on comparing values of summary statistics calculated in windows. We present an approach termed SURFDAWave, which translates measures of genetic diversity calculated in genomic windows to functional data. By transforming our discrete data points to be outputs of continuous functions defined over genomic space, we are able to learn the features of these functions that signify selection. This enables us to confidently identify complex modes of natural selection, including adaptive introgression. We are also able to predict important selection parameters that are responsible for shaping the inferred selection events. By applying our model to human population-genomic data, we recapitulate previously identified regions of selective sweeps, such as OCA2 in Europeans, and predict that its beneficial mutation reached a frequency of 0.02 before it swept 1,802 generations ago, a time when humans were relatively new to Europe. In addition, we identify BNC2 in Europeans as a target of adaptive introgression, and predict that it harbors a beneficial mutation that arose in an archaic human population that split from modern humans within the hypothesized modern human-Neanderthal divergence range.
Collapse
Affiliation(s)
- Mehreen R. Mughal
- Bioinformatics and Genomics at the Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Hillary Koch
- Department of Statistics, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Jinguo Huang
- Bioinformatics and Genomics at the Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Francesca Chiaromonte
- Department of Statistics, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Michael DeGiorgio
- Department of Computer and Electrical Engineering and Computer Science, Florida Atlantic University, Boca Raton, Florida, United States of America
| |
Collapse
|
12
|
Dong XM, Zhao K, Zheng WW, Xu CW, Zhang MJ, Yin RH, Gao R, Tang LJ, Liu JF, Chen H, Zhan YQ, Yu M, Ge CH, Gao HY, Li X, Luo T, Ning HM, Yang XM, Li CY. EDAG mediates Hsp70 nuclear localization in erythroblasts and rescues dyserythropoiesis in myelodysplastic syndrome. FASEB J 2020; 34:8416-8427. [PMID: 32350948 DOI: 10.1096/fj.201902946r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 04/06/2020] [Accepted: 04/13/2020] [Indexed: 12/11/2022]
Abstract
During human erythroid maturation, Hsp70 translocates into the nucleus and protects GATA-1 from caspase-3 cleavage. Failure of Hsp70 to localize to the nucleus was found in Myelodysplastic syndrome (MDS) erythroblasts and can induce dyserythropoiesis, with arrest of maturation and death of erythroblasts. However, the mechanism of the nuclear trafficking of Hsp70 in erythroblasts remains unknown. Here, we found the hematopoietic transcriptional regulator, EDAG, to be a novel binding partner of Hsp70 that forms a protein complex with Hsp70 and GATA-1 during human normal erythroid differentiation. EDAG overexpression blocked the cytoplasmic translocation of Hsp70 induced by EPO deprivation, inhibited GATA-1 degradation, thereby promoting erythroid maturation in an Hsp70-dependent manner. Furthermore, in myelodysplastic syndrome (MDS) patients with dyserythropoiesis, EDAG is dramatically down-regulated, and forced expression of EDAG has been found to restore the localization of Hsp70 in the nucleus and elevate the protein level of GATA-1 to a significant extent. In addition, EDAG rescued the dyserythropoiesis of MDS patients by increasing erythroid differentiation and decreasing cell apoptosis. This study demonstrates the molecular mechanism of Hsp70 nuclear sustaining during erythroid maturation and establishes that EDAG might be a suitable therapeutic target for dyserythropoiesis in MDS patients.
Collapse
Affiliation(s)
- Xiao-Ming Dong
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China.,Department of Pharmaceutical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, China.,College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Ke Zhao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China
| | - Wei-Wei Zheng
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China
| | - Cheng-Wang Xu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China
| | - Mei-Jiang Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China.,Department of Pharmaceutical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
| | - Rong-Hua Yin
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China
| | - Rui Gao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China
| | - Liu-Jun Tang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China
| | - Jin-Fang Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China
| | - Hui Chen
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China
| | - Yi-Qun Zhan
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China
| | - Miao Yu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China
| | - Chang-Hui Ge
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, Beijing, China
| | - Hui-Ying Gao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China
| | - Xiu Li
- School of Postgraduate, Anhui Medical University, Hefei, China
| | - Teng Luo
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China
| | - Hong-Mei Ning
- Department of Hematopoietic Stem Cell Transplantation, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiao-Ming Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China.,Department of Pharmaceutical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, China.,School of Postgraduate, Anhui Medical University, Hefei, China
| | - Chang-Yan Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China.,School of Postgraduate, Anhui Medical University, Hefei, China
| |
Collapse
|
13
|
Milardi D, Grande G, Vincenzoni F, Pierconti F, Pontecorvi A. Proteomics for the Identification of Biomarkers in Testicular Cancer-Review. Front Endocrinol (Lausanne) 2019; 10:462. [PMID: 31354629 PMCID: PMC6639829 DOI: 10.3389/fendo.2019.00462] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 06/25/2019] [Indexed: 01/04/2023] Open
Abstract
A large number of biomarkers have been proposed for the diagnosis of testicular cancer, representing putative molecular targets for anticancer treatments. However, no conclusive data have been provided. Proteomics represents a research field recently developed. It evaluates the large-scale analysis of the full protein components of a single cell, of a specific tissue, or of biological fluids. In the last decades, proteomics has been applied in clinical fields, thanks to modern technology and new bioinformatic tools, to identify novel molecular markers of diseases. The aim of this review is to argue the findings of recent studies in the discoveries of putative prognostic and diagnostic markers of testis cancer by proteomic techniques. We present here a panel of proteins identified by proteomics which might be used after validation for early detection and the prognostic evaluation of testicular tumors. In addition, the molecular mechanisms revealed by these proteomic studies might also guide the development of novel treatments in future.
Collapse
Affiliation(s)
- Domenico Milardi
- International Scientific Institute “Paul VI”, Rome, Italy
- Division of Endocrinology, Fondazione Policlinico'A. Gemelli' IRCCS, Rome, Italy
| | - Giuseppe Grande
- International Scientific Institute “Paul VI”, Rome, Italy
- Division of Endocrinology, Fondazione Policlinico'A. Gemelli' IRCCS, Rome, Italy
- *Correspondence: Giuseppe Grande
| | - Federica Vincenzoni
- School of Medicine, Biochemistry and Clinical Biochemistry Institute, Catholic University of Rome, Rome, Italy
- Department of Laboratory Diagnostic and Infectious Diseases, Fondazione Policlinico'A. Gemelli' IRCCS, Rome, Italy
| | - Francesco Pierconti
- Division of Anatomic Pathology and Histology, School of Medicine, Catholic University of Rome, Rome, Italy
| | - Alfredo Pontecorvi
- International Scientific Institute “Paul VI”, Rome, Italy
- Division of Endocrinology, Fondazione Policlinico'A. Gemelli' IRCCS, Rome, Italy
| |
Collapse
|
14
|
Peters MJ, Parker SK, Grim J, Allard CAH, Levin J, Detrich HW. Divergent Hemogen genes of teleosts and mammals share conserved roles in erythropoiesis: analysis using transgenic and mutant zebrafish. Biol Open 2018; 7:bio.035576. [PMID: 30097520 PMCID: PMC6124579 DOI: 10.1242/bio.035576] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Hemogen is a vertebrate transcription factor that performs important functions in erythropoiesis and testicular development and may contribute to neoplasia. Here we identify zebrafish Hemogen and show that it is considerably smaller (∼22 kDa) than its human ortholog (∼55 kDa), a striking difference that is explained by an underlying modular structure. We demonstrate that Hemogens are largely composed of 21-25 amino acid repeats, some of which may function as transactivation domains (TADs). Hemogen expression in embryonic and adult zebrafish is detected in hematopoietic, renal, neural and gonadal tissues. Using Tol2- and CRISPR/Cas9-generated transgenic zebrafish, we show that Hemogen expression is controlled by two Gata1-dependent regulatory sequences that act alone and together to control spatial and temporal expression during development. Partial depletion of Hemogen in embryos by morpholino knockdown reduces the number of erythrocytes in circulation. CRISPR/Cas9-generated zebrafish lines containing either a frameshift mutation or an in-frame deletion in a putative, C-terminal TAD display anemia and embryonic tail defects. This work expands our understanding of Hemogen and provides mutant zebrafish lines for future study of the mechanism of this important transcription factor. Summary: Transgenic and mutant zebrafish lines were created to characterize the expression and functions of Hemogen, a transcription factor involved in the formation of red blood cells and other processes.
Collapse
Affiliation(s)
- Michael J Peters
- Department of Marine and Environmental Sciences, Northeastern University, Nahant, MA 01908, USA
| | - Sandra K Parker
- Department of Marine and Environmental Sciences, Northeastern University, Nahant, MA 01908, USA
| | - Jeffrey Grim
- Department of Marine and Environmental Sciences, Northeastern University, Nahant, MA 01908, USA
| | - Corey A H Allard
- Department of Marine and Environmental Sciences, Northeastern University, Nahant, MA 01908, USA
| | - Jonah Levin
- Department of Marine and Environmental Sciences, Northeastern University, Nahant, MA 01908, USA
| | - H William Detrich
- Department of Marine and Environmental Sciences, Northeastern University, Nahant, MA 01908, USA
| |
Collapse
|
15
|
Zhao K, Zheng WW, Dong XM, Yin RH, Gao R, Li X, Liu JF, Zhan YQ, Yu M, Chen H, Ge CH, Ning HM, Yang XM, Li CY. EDAG promotes the expansion and survival of human CD34+ cells. PLoS One 2018; 13:e0190794. [PMID: 29324880 PMCID: PMC5764277 DOI: 10.1371/journal.pone.0190794] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 12/20/2017] [Indexed: 01/17/2023] Open
Abstract
EDAG is multifunctional transcriptional regulator primarily expressed in the linloc-kit+Sca-1+ hematopoietic stem cells (HSC) and CD34+ progenitor cells. Previous studies indicate that EDAG is required for maintaining hematopoietic lineage commitment balance. Here using ex vivo culture and HSC transplantation models, we report that EDAG enhances the proliferative potential of human cord blood CD34+ cells, increases survival, prevents cell apoptosis and promotes their repopulating capacity. Moreover, EDAG overexpression induces rapid entry of CD34+ cells into the cell cycle. Gene expression profile analysis indicate that EDAG knockdown leads to down-regulation of various positive cell cycle regulators including cyclin A, B, D, and E. Together these data provides novel insights into EDAG in regulation of expansion and survival of human hematopoietic stem/progenitor cells.
Collapse
Affiliation(s)
- Ke Zhao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Wei-Wei Zheng
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Xiao-Ming Dong
- Tianjin University, School of Chemical Engineering and Technology, Department of Pharmaceutical Engineering, Tianjin, China
| | - Rong-Hua Yin
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Rui Gao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Xiu Li
- An Hui Medical University, Hefei, China
| | - Jin-Fang Liu
- Guang Dong Pharmaceutical University, School of Pharmacy, Guangzhou, China
| | - Yi-Qun Zhan
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Miao Yu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Hui Chen
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Chang-Hui Ge
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Hong-Mei Ning
- Department of Hematopoietic Stem Cell Transplantation, Affiliated Hospital to Academy of Military Medical Sciences, Beijing, China
- * E-mail: (HMN); (XMY); (CYL)
| | - Xiao-Ming Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
- Tianjin University, School of Chemical Engineering and Technology, Department of Pharmaceutical Engineering, Tianjin, China
- * E-mail: (HMN); (XMY); (CYL)
| | - Chang-Yan Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
- Guang Dong Pharmaceutical University, School of Pharmacy, Guangzhou, China
- * E-mail: (HMN); (XMY); (CYL)
| |
Collapse
|
16
|
Zhang YC, Ye H, Zeng Z, Chin YE, Huang YN, Fu GH. The NF-κB p65/miR-23a-27a-24 cluster is a target for leukemia treatment. Oncotarget 2016; 6:33554-67. [PMID: 26378023 PMCID: PMC4741785 DOI: 10.18632/oncotarget.5591] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 08/23/2015] [Indexed: 11/25/2022] Open
Abstract
p65 is a transcription factor that is involved in many physiological and pathologic processes. Here we report that p65 strongly binds to the miR-23a-27a-24 cluster promoter to up-regulate its expression. As bone marrow-derived cells differentiate into red blood cells in vitro, p65/miR-23a-27a-24 cluster expression increases sharply and then declines before the appearance of red blood cells, suggesting that this cluster is negatively related to erythroid terminal differentiation. Bioinformatic and molecular biology experiments confirmed that the miR-23a-27a-24 cluster inhibited the expression of the erythroid proteome and contributed to erythroleukemia progression. In addition, high level of the p65/miR-23a-27a-24 cluster was found in APL and AML cell lines and in nucleated peripheral blood cells from leukemia patients. Furthermore, anti-leukemia drugs significantly inhibited the expression of the p65/miR-23a-27a-24 cluster in leukemia cells. Administration of the p65 inhibitor parthenolide significantly improved hematology and myelogram indices while prolonging the life span of erythroleukemia mice. Meanwhile, stable overexpression of these three miRNAs in mouse erythroleukemia cells enhanced cell malignancy. Our findings thus connect a novel regulation pathway of the p65/miR-23a-27a-24 cluster with the erythroid proteome and provide an applicable approach for treating leukemia.
Collapse
Affiliation(s)
- Yong-Chang Zhang
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Ye
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhi Zeng
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Y Eugene Chin
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS) and Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Yu-Ning Huang
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guo-Hui Fu
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
17
|
Xu J, Li Z, Su Q, Zhao J, Ma J. Embryonic develop-associated gene 1 is overexpressed and acts as a tumor promoter in thyroid carcinoma. Biomed Pharmacother 2016; 81:86-92. [DOI: 10.1016/j.biopha.2016.03.052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 03/30/2016] [Accepted: 03/31/2016] [Indexed: 11/15/2022] Open
|
18
|
Chen DL, Hu ZQ, Zheng XF, Wang XY, Xu YZ, Li WQ, Fang HS, Kan L, Wang SY. EDAG-1 promotes proliferation and invasion of human thyroid cancer cells by activating MAPK/Erk and AKT signal pathways. Cancer Biol Ther 2016; 17:414-21. [PMID: 26934676 PMCID: PMC4910939 DOI: 10.1080/15384047.2016.1156259] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 01/08/2016] [Accepted: 02/14/2016] [Indexed: 10/22/2022] Open
Abstract
Erythroid differentiation-associated gene (EDAG) is differentially expressed in normal hematopoietic progenitor/stem cells and a variety of embryonic tissues. High EDAG-1 expression is also found in human thyroid cancer cells and peripheral blood of patients with leukemia, but its functional significance was unclear. Current study aims to further clarify the expression pattern of EDAG-1 and tests its roles in proliferation and invasion of human thyroid cancer cells in vitro and in vivo. To this end, we have performed gain-of-function and loss-of-function studies to clarify how EDAG-1 regulates the proliferation, invasion, and adhesion ability of human thyroid cancer cells SW579cells. We found that overexpression of EDAG-1 promoted the proliferation, invasion, and adhesion of human thyroid cancer cells, whereas silencing of EDAG-1 reversed all these changes and reduced the tumorigenesis risk of nude mice. Mechanistically, we found that overexpression of EDAG-1 activated the MAPK/Erk and AKT signal pathways. These findings provide novel insights of the role of EDAG-1 in thyroid tumors, and may have direct clinical implication.
Collapse
Affiliation(s)
- Dan-lei Chen
- Department of Pathophysiology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, PR China
| | - Zhong-qian Hu
- Department of Pathophysiology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, PR China
- Department of Ultrasound, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, PR China
| | - Xian-fang Zheng
- Department of Pathophysiology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, PR China
| | - Xin-yi Wang
- Department of Pathophysiology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, PR China
- Department of Clinical Medicine, Anhui Medical University, Hefei, Anhui, PR China
| | - You-zhi Xu
- Department of Pathophysiology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, PR China
| | - Wen-qing Li
- Department of Pathophysiology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, PR China
| | - Hao-shu Fang
- Department of Pathophysiology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, PR China
| | - Lixin Kan
- Department of Pathophysiology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, PR China
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Si-ying Wang
- Department of Pathophysiology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, PR China
| |
Collapse
|
19
|
Zheng WW, Dong XM, Yin RH, Xu FF, Ning HM, Zhang MJ, Xu CW, Yang Y, Ding YL, Wang ZD, Zhao WB, Tang LJ, Chen H, Wang XH, Zhan YQ, Yu M, Ge CH, Li CY, Yang XM. EDAG positively regulates erythroid differentiation and modifies GATA1 acetylation through recruiting p300. Stem Cells 2015; 32:2278-89. [PMID: 24740910 DOI: 10.1002/stem.1723] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 03/03/2014] [Accepted: 03/24/2014] [Indexed: 11/11/2022]
Abstract
Erythroid differentiation-associated gene (EDAG) has been considered to be a transcriptional regulator that controls hematopoietic cell differentiation, proliferation, and apoptosis. The role of EDAG in erythroid differentiation of primary erythroid progenitor cells and in vivo remains unknown. In this study, we found that EDAG is highly expressed in CMPs and MEPs and upregulated during the erythroid differentiation of CD34(+) cells following erythropoietin (EPO) treatment. Overexpression of EDAG induced erythroid differentiation of CD34(+) cells in vitro and in vivo using immunodeficient mice. Conversely, EDAG knockdown reduced erythroid differentiation in EPO-treated CD34(+) cells. Detailed mechanistic analysis suggested that EDAG forms complex with GATA1 and p300 and increases GATA1 acetylation and transcriptional activity by facilitating the interaction between GATA1 and p300. EDAG deletion mutants lacking the binding domain with GATA1 or p300 failed to enhance erythroid differentiation, suggesting that EDAG regulates erythroid differentiation partly through forming EDAG/GATA1/p300 complex. In the presence of the specific inhibitor of p300 acetyltransferase activity, C646, EDAG was unable to accelerate erythroid differentiation, indicating an involvement of p300 acetyltransferase activity in EDAG-induced erythroid differentiation. ChIP-PCR experiments confirmed that GATA1 and EDAG co-occupy GATA1-targeted genes in primary erythroid cells and in vivo. ChIP-seq was further performed to examine the global occupancy of EDAG during erythroid differentiation and a total of 7,133 enrichment peaks corresponding to 3,847 genes were identified. Merging EDAG ChIP-Seq and GATA1 ChIP-Seq datasets revealed that 782 genes overlapped. Microarray analysis suggested that EDAG knockdown selectively inhibits GATA1-activated target genes. These data provide novel insights into EDAG in regulation of erythroid differentiation.
Collapse
Affiliation(s)
- Wei-Wei Zheng
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Follistatin-like 1 attenuates differentiation and survival of erythroid cells through Smad2/3 signaling. Biochem Biophys Res Commun 2015; 466:711-6. [PMID: 26365350 DOI: 10.1016/j.bbrc.2015.09.044] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 09/08/2015] [Indexed: 02/03/2023]
Abstract
Hematopoiesis is a complex process tightly controlled by sets of transcription factors in a context-dependent and stage-specific manner. Smad2/3 transcription factor plays a central role in differentiation and survival of erythroid cells. Here we report that follistatin-like 1 (FSTL1) treatment impairs hemin-induced erythroid differentiation and cell survival. FSTL1 differentially regulates transforming growth factor beta (TGF-β) and bone morphogenetic protein (BMP) signaling. Blockade of Smad2/3 signaling with the ALK5/type I TGF-βR kinase inhibitor, SB-525334, was efficacious for rescue of erythroid differentiation blockage and apoptosis. Reversely, activation of Smad1/5/8 signaling with BMP4 cannot rescue FSTL1-mediated erythroid differentiation blockage and apoptosis. Collectively, these data provide mechanistic insight into the regulation of erythropoiesis by FSTL1 signaling and lay a foundation for exploring FSTL1 signaling as a therapeutic target for anemia.
Collapse
|
21
|
Mortlock SA, Wei J, Williamson P. T-cell activation and early gene response in dogs. PLoS One 2015; 10:e0121169. [PMID: 25803042 PMCID: PMC4372360 DOI: 10.1371/journal.pone.0121169] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 01/28/2015] [Indexed: 12/27/2022] Open
Abstract
T-cells play a crucial role in canine immunoregulation and defence against invading pathogens. Proliferation is fundamental to T-cell differentiation, homeostasis and immune response. Initiation of proliferation following receptor mediated stimuli requires a temporally programmed gene response that can be identified as immediate-early, mid- and late phases. The immediate-early response genes in T-cell activation engage the cell cycle machinery and promote subsequent gene activation events. Genes involved in this immediate-early response in dogs are yet to be identified. The present study was undertaken to characterise the early T-cell gene response in dogs to improve understanding of the genetic mechanisms regulating immune function. Gene expression profiles were characterised using canine gene expression microarrays and quantitative reverse transcription PCR (qRT-PCR), and paired samples from eleven dogs. Significant functional annotation clusters were identified following stimulation with phytohemagluttinin (PHA) (5μg/ml), including the Toll-like receptor signaling pathway and phosphorylation pathways. Using strict statistical criteria, 13 individual genes were found to be differentially expressed, nine of which have ontologies that relate to proliferation and cell cycle control. These included, prostaglandin-endoperoxide synthase 2 (PTGS2/COX2), early growth response 1 (EGR1), growth arrest and DNA damage-inducible gene (GADD45B), phorbol-12-myristate-13-acetate-induced protein 1 (PMAIP1), V-FOS FBJ murine osteosarcoma viral oncogene homolog (FOS), early growth response 2 (EGR2), hemogen (HEMGN), polo-like kinase 2 (PLK2) and polo-like kinase 3 (PLK3). Differential gene expression was re-examined using qRT-PCR, which confirmed that EGR1, EGR2, PMAIP1, PTGS2, FOS and GADD45B were significantly upregulated in stimulated cells and ALAS2 downregulated. PTGS2 and EGR1 showed the highest levels of response in these dogs. Both of these genes are involved in cell cycle regulation. This study provides a comprehensive analysis of the early T-cell gene response to activation in dogs.
Collapse
Affiliation(s)
- Sally-Anne Mortlock
- Faculty of Veterinary Science, The University of Sydney, NSW 2006, Australia
| | - Jerry Wei
- Faculty of Veterinary Science, The University of Sydney, NSW 2006, Australia
- Sydney Medical School, The University of Sydney, NSW 2006, Australia
| | - Peter Williamson
- Faculty of Veterinary Science, The University of Sydney, NSW 2006, Australia
| |
Collapse
|
22
|
ABRO1 suppresses tumourigenesis and regulates the DNA damage response by stabilizing p53. Nat Commun 2014; 5:5059. [PMID: 25283148 PMCID: PMC4205886 DOI: 10.1038/ncomms6059] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 08/21/2014] [Indexed: 01/02/2023] Open
Abstract
Abraxas brother 1 (ABRO1) has been reported to be a component of the BRISC complex, a multiprotein complex that specifically cleaves 'Lys-63'-linked ubiquitin. However, current knowledge of the functions of ABRO1 is limited. Here we report that ABRO1 is frequently downregulated in human liver, kidney, breast and thyroid gland tumour tissues. Depletion of ABRO1 in cancer cells reduces p53 levels and enhances clone formation and cellular transformation. Conversely, overexpression of ABRO1 suppresses cell proliferation and tumour formation in a p53-dependent manner. We further show that ABRO1 stabilizes p53 by facilitating the interaction of p53 with USP7. DNA-damage induced accumulation of endogenous ABRO1 as well as translocation of ABRO1 to the nucleus, and the induction of p53 by DNA damage is almost completely attenuated by ABRO1 depletion. Our study shows that ABRO1 is a novel p53 regulator that plays an important role in tumour suppression and the DNA damage response.
Collapse
|
23
|
The over-expression of aquaporin-1 alters erythroid gene expression in human erythroleukemia K562 cells. Tumour Biol 2014; 36:291-302. [PMID: 25252847 DOI: 10.1007/s13277-014-2614-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 09/09/2014] [Indexed: 10/24/2022] Open
Abstract
Aquaporin genes are differentially expressed in primitive versus definitive erythropoiesis. Our previous research results showed that over-expression of aquaporin-1 (AQP1) gene greatly promotes the erythroid differentiation of erythroleukemia K562 cells, using benzidine staining and quantitative reverse-transcriptase polymerase chain reaction (qRT-PCR) analysis for representative erythroid-related genes, including γ-globin. But the molecular mechanisms underlying erythroid-specific gene regulation remain unknown. In this study, we demonstrated that AQP1 induced hemoglobins expression and altered erythroid gene expression by microarray analysis in K562 cells. The retroviral expression vector of AQP1 (pBABE-puro-AQP1) was constructed and infected K562 cells to establish a stable AQP1 over-expression cell line (K562-AQP1). AQP1 over-expression effectively inhibited cell proliferation and induced cell growth arrest in G1 phase of K562 cells. Then microarray profile was applied to analyze the differentially expressed genes which involved the mechanism of AQP1 in erythroid differentiation induction. The DAVID functional annotation clustering tool was used to identify biological functions enriched with the differentially expressed genes (n = 466 genes) and to group genes into clusters based on their functional similarity. Significant enrichment of genes involved in "oxygen transporter activity" (p = 3.8E-7) including hemoglobins (HBD, HBG, HBB, HBE1, and HBQ1), HEMGN, and EBP42 were validated by qRT-PCR. Moreover, silencing of HEMGN by RNA interference in K562-AQP1 cells resulted in down-regulation of these genes. These data provide a better understanding of the role of AQP1 in erythroid differentiation, by promoting HEMGN induction and other potential signaling pathways associated with hemoglobin induction.
Collapse
|
24
|
Jenkins NT, Padilla J, Thorne PK, Martin JS, Rector RS, Davis JW, Laughlin MH. Transcriptome-wide RNA sequencing analysis of rat skeletal muscle feed arteries. I. Impact of obesity. J Appl Physiol (1985) 2014; 116:1017-32. [PMID: 24436298 PMCID: PMC4035791 DOI: 10.1152/japplphysiol.01233.2013] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Accepted: 01/14/2014] [Indexed: 01/13/2023] Open
Abstract
We employed next-generation RNA sequencing (RNA-Seq) technology to determine the influence of obesity on global gene expression in skeletal muscle feed arteries. Transcriptional profiles of the gastrocnemius and soleus muscle feed arteries (GFA and SFA, respectively) and aortic endothelial cell-enriched samples from obese Otsuka Long-Evans Tokushima Fatty (OLETF) and lean Long-Evans Tokushima Otsuka (LETO) rats were examined. Obesity produced 282 upregulated and 133 downregulated genes in SFA and 163 upregulated and 77 downregulated genes in GFA [false discovery rate (FDR) < 10%] with an overlap of 93 genes between the arteries. In LETO rats, there were 89 upregulated and 114 downregulated genes in the GFA compared with the SFA. There were 244 upregulated and 275 downregulated genes in OLETF rats (FDR < 10%) in the GFA compared with the SFA, with an overlap of 76 differentially expressed genes common to both LETO and OLETF rats in both the GFA and SFA. A total of 396 transcripts were found to be differentially expressed between LETO and OLETF in aortic endothelial cell-enriched samples. Overall, we found 1) the existence of heterogeneity in the transcriptional profile of the SFA and GFA within healthy LETO rats, 2) that this between-vessel heterogeneity was markedly exacerbated in the hyperphagic, obese OLETF rat, and 3) a greater number of genes whose expression was altered by obesity in the SFA compared with the GFA. Also, results indicate that in OLETF rats the GFA takes on a relatively more proatherogenic phenotype compared with the SFA.
Collapse
Affiliation(s)
- Nathan T Jenkins
- Department of Kinesiology, University of Georgia, Athens, Georgia
| | | | | | | | | | | | | |
Collapse
|
25
|
Kong XZ, Yin RH, Ning HM, Zheng WW, Dong XM, Yang Y, Xu FF, Li JJ, Zhan YQ, Yu M, Ge CH, Zhang JH, Chen H, Li CY, Yang XM. Effects of THAP11 on erythroid differentiation and megakaryocytic differentiation of K562 cells. PLoS One 2014; 9:e91557. [PMID: 24637716 PMCID: PMC3956667 DOI: 10.1371/journal.pone.0091557] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 02/13/2014] [Indexed: 12/16/2022] Open
Abstract
Hematopoiesis is a complex process regulated by sets of transcription factors in a stage-specific and context-dependent manner. THAP11 is a transcription factor involved in cell growth, ES cell pluripotency, and embryogenesis. Here we showed that THAP11 was down-regulated during erythroid differentiation but up-regulated during megakaryocytic differentiation of cord blood CD34+ cells. Overexpression of THAP11 in K562 cells inhibited the erythroid differentiation induced by hemin with decreased numbers of benzidine-positive cells and decreased mRNA levels of α-globin (HBA) and glycophorin A (GPA), and knockdown of THAP11 enhanced the erythroid differentiation. Conversely, THAP11 overexpression accelerated the megakaryocytic differentiation induced by phorbol myristate acetate (PMA) with increased percentage of CD41+ cells, increased numbers of 4N cells, and elevated CD61 mRNA levels, and THAP11 knockdown attenuated the megakaryocytic differentiation. The expression levels of transcription factors such as c-Myc, c-Myb, GATA-2, and Fli1 were changed by THAP11 overexpression. In this way, our results suggested that THAP11 reversibly regulated erythroid and megakaryocytic differentiation.
Collapse
Affiliation(s)
- Xiang-Zhen Kong
- Department of Pharmaceutical Engineering, Tianjin University, Tianjin, China
- Department of Biochemistry and Molecular Biology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Rong-Hua Yin
- Department of Biochemistry and Molecular Biology, Beijing Institute of Radiation Medicine, Beijing, China
- State Key Laboratory of Proteomics, Beijing, China
| | - Hong-Mei Ning
- Department of Hematopoietic Stem Cell Transplantation, Affiliated Hospital to Academy of Military Medical Sciences, Beijing, China
| | - Wei-Wei Zheng
- Department of Biochemistry and Molecular Biology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Xiao-Ming Dong
- Department of Pharmaceutical Engineering, Tianjin University, Tianjin, China
| | - Yang Yang
- Department of Chemistry, Purdue University, West Lafayette, Indiana, United States of America
| | - Fei-Fei Xu
- Department of Biochemistry and Molecular Biology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Jian-Jie Li
- Department of Pulmonary Neoplasms Internal Medicine, Affiliated Hospital to Academy of Military Medicine Sciences, Beijing, China
| | - Yi-Qun Zhan
- Department of Biochemistry and Molecular Biology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Miao Yu
- Department of Biochemistry and Molecular Biology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Chang-Hui Ge
- Department of Biochemistry and Molecular Biology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Jian-Hong Zhang
- Department of Biochemistry and Molecular Biology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Hui Chen
- Department of Biochemistry and Molecular Biology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Chang-Yan Li
- Department of Biochemistry and Molecular Biology, Beijing Institute of Radiation Medicine, Beijing, China
- State Key Laboratory of Proteomics, Beijing, China
- * E-mail: (XMY); (CYL)
| | - Xiao-Ming Yang
- Department of Pharmaceutical Engineering, Tianjin University, Tianjin, China
- Department of Biochemistry and Molecular Biology, Beijing Institute of Radiation Medicine, Beijing, China
- State Key Laboratory of Proteomics, Beijing, China
- * E-mail: (XMY); (CYL)
| |
Collapse
|
26
|
Zhang MJ, Ding YL, Xu CW, Yang Y, Lian WX, Zhan YQ, Li W, Xu WX, Yu M, Ge CH, Ning HM, Li CY, Yang XM. Erythroid differentiation-associated gene interacts with NPM1 (nucleophosmin/B23) and increases its protein stability, resisting cell apoptosis. FEBS J 2012; 279:2848-62. [PMID: 22712502 DOI: 10.1111/j.1742-4658.2012.08663.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Erythroid differentiation-associated gene (EDAG) is a haematopoietic tissue-specific transcription regulator that plays a key role in maintaining the homeostasis of haematopoietic lineage commitment. In acute myeloid leukaemia (AML) patients, the high expression level of EDAG is associated with poor prognosis. NPM1 (nucleophosmin/B23), a ubiquitous nucleolar phosphoprotein, comprises a multifunctional protein that is involved in several cellular processes, including ribosome biogenesis, centrosome duplication, cell cycle progression, cell growth and transformation. Various studies have implicated NPM1 overexpression in promoting tumour cell proliferation, blocking the differentiation of leukaemia cells and resisting apoptosis. In the present study, using co-immunoprecipitation, we characterized EDAG as a physiological binding partner of NPM1; The N-terminal (amino acids 1-124) region of EDAG interacts with the N-terminal (amino acids 118-187) of NPM1. Under cycloheximide treatment, the stability of NPM1 protein was enhanced by EDAG overexpression, whereas knockdown of EDAG by lentivirus-mediated small interfering RNA resulted in an increased degradation rate of NPM1 in K562 cells. During 4β-phorbol l2-myristate 13-acetate-induced K562 megakaryocytic differentiation, overexpression of EDAG prevented the down-regulation of NPM1 proteins, whereas knockdown of EDAG accelerated the down-regulation of NPM1. EDAG deletion mutant lacking the binding domain with NPM1 lost the ability to stabilize NPM1 protein. Furthermore, knockdown of EDAG in K562 cells led to increased cell apoptosis induced by imatinib, and re-expression of NPM1 attenuated the increased apoptosis. These results suggest that EDAG enhances the protein stability of NPM1 via binding to NPM1, which plays a critical role in the anti-apoptosis of leukaemia cells.
Collapse
Affiliation(s)
- Mei-Jiang Zhang
- Department of Pharmaceutical Engineering, Tianjin University, Tianjin, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Griaud F, Williamson AJK, Taylor S, Potier DN, Spooncer E, Pierce A, Whetton AD. BCR/ABL modulates protein phosphorylation associated with the etoposide-induced DNA damage response. J Proteomics 2012; 77:14-26. [PMID: 22705319 DOI: 10.1016/j.jprot.2012.06.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Revised: 05/22/2012] [Accepted: 06/02/2012] [Indexed: 10/28/2022]
Abstract
BCR/ABL expression is the key characteristic of chronic myeloid leukaemia (CML). The progression of CML is associated with genomic instability. Systematic analysis of DNA damage signalling in the presence of BCR/ABL thus offers opportunities to identify mechanisms of leukaemic progression. We therefore undertook a quantitative phosphoproteomics study to test whether BCR/ABL expression could globally affect the response to genotoxic stress signalling. Etoposide-induced DNA damage was chosen and the concentration and time of exposure determined such that apoptosis was not associated with the study. More than 1100 phosphoentities were identified. BCR/ABL was shown to significantly alter the response to etoposide in many cases. These include sites on MDC1, a key component of DNA repair, and Hemogen. Hemogen is a transcriptional target of HOXB4 and GATA1, two transcription factors involved in haemopoietic development, and is overexpressed in acute myeloid leukaemia. To validate Hemogen phosphorylation, absolute quantification using an isotopomeric standard and selected reaction monitoring was performed. This revealed a strong correlation with isobaric tagging data and offering quantification at about 10 fmol per million cells. Furthermore we found that multiple protein phosphorylation changes mediated by BCR/ABL were connected to the increased activation of NFκB, a key survival transcription factor, after etoposide exposure.
Collapse
Affiliation(s)
- François Griaud
- Stem Cell and Leukaemia Proteomics Laboratory, Manchester Academic Health, Science Centre, The University of Manchester, Wolfson Molecular Imaging, Centre, 27 Palatine Road, Withington, Manchester, M20 3LJ, UK
| | | | | | | | | | | | | |
Collapse
|
28
|
Nuclear factor-kappaB as a switch in regulation of resveratrol-mediated apoptosis and erythrocytic differentiation in human leukaemia cells. Food Chem 2012. [DOI: 10.1016/j.foodchem.2011.12.062] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
29
|
Taaffe JE, Bosinger SE, Del Prete GQ, Else JG, Ratcliffe S, Ward CD, Migone T, Paiardini M, Silvestri G. CCR5 blockade is well tolerated and induces changes in the tissue distribution of CCR5+ and CD25+ T cells in healthy, SIV-uninfected rhesus macaques. J Med Primatol 2011; 41:24-42. [PMID: 22077380 DOI: 10.1111/j.1600-0684.2011.00521.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND CCR5 is a main co-receptor for HIV, but also homes lymphocytes to sites of inflammation. We hypothesized that inhibition of CCR5 signaling would reduce HIV-associated chronic immune activation. METHODS To test this hypothesis, we administered an antagonistic anti-CCR5 monoclonal antibody (HGS101) to five uninfected rhesus macaques (RMs) and monitored lymphocyte dynamics in blood and tissue. RESULTS CCR5 blockade resulted in decreased levels of CCR5+ T cells in blood and, at later timepoints, in lymph nodes. Additionally, the levels of CD25+ T cells increased in lymph nodes, but decreased in blood, bone marrow, and rectal mucosa. Finally, a profile of gene expression from HGS101-treated RMs revealed a subtle, but consistent, in vivo signature of CCR5 blockade that suggests a mild immune-modulatory effect. CONCLUSIONS Treatment with anti-CCR5 antibody induces changes in the tissue distribution of CCR5+ and CD25+ T cells that may impact on the overall levels of immune activation during HIV and SIV infection.
Collapse
Affiliation(s)
- Jessica E Taaffe
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Joehanes R, Johnson AD, Barb JJ, Raghavachari N, Liu P, Woodhouse KA, O'Donnell CJ, Munson PJ, Levy D. Gene expression analysis of whole blood, peripheral blood mononuclear cells, and lymphoblastoid cell lines from the Framingham Heart Study. Physiol Genomics 2011; 44:59-75. [PMID: 22045913 DOI: 10.1152/physiolgenomics.00130.2011] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Despite a growing number of reports of gene expression analysis from blood-derived RNA sources, there have been few systematic comparisons of various RNA sources in transcriptomic analysis or for biomarker discovery in the context of cardiovascular disease (CVD). As a pilot study of the Systems Approach to Biomarker Research (SABRe) in CVD Initiative, this investigation used Affymetrix Exon arrays to characterize gene expression of three blood-derived RNA sources: lymphoblastoid cell lines (LCL), whole blood using PAXgene tubes (PAX), and peripheral blood mononuclear cells (PBMC). Their performance was compared in relation to identifying transcript associations with sex and CVD risk factors, such as age, high-density lipoprotein, and smoking status, and the differential blood cell count. We also identified a set of exons that vary substantially between participants, but consistently in each RNA source. Such exons are thus stable phenotypes of the participant and may potentially become useful fingerprinting biomarkers. In agreement with previous studies, we found that each of the RNA sources is distinct. Unlike PAX and PBMC, LCL gene expression showed little association with the differential blood count. LCL, however, was able to detect two genes related to smoking status. PAX and PBMC identified Y-chromosome probe sets similarly and slightly better than LCL.
Collapse
Affiliation(s)
- Roby Joehanes
- The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, Massachusetts, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Yu M, Li H, Liu Q, Liu F, Tang L, Li C, Yuan Y, Zhan Y, Xu W, Li W, Chen H, Ge C, Wang J, Yang X. Nuclear factor p65 interacts with Keap1 to repress the Nrf2-ARE pathway. Cell Signal 2011; 23:883-92. [PMID: 21262351 DOI: 10.1016/j.cellsig.2011.01.014] [Citation(s) in RCA: 245] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Accepted: 01/14/2011] [Indexed: 12/12/2022]
Abstract
Keap1 is an inhibitor of Nrf2 involved in Nrf2-dependent antioxidant response. However, the mechanisms on how Keap1 regulates Nrf2-ARE signaling pathway remains to be determined. Here, by using a yeast two-hybrid technology, p65 subunit of NF-κB transcription factor was identified as a partner of Keap1. We show that Keap1 physically associated with p65 in vivo and in vitro. Overexpression of p65 inhibited Nrf2-dependent transcription induced by diethylmaleate (DEM) or tert-butyl hydroxyquinone (tBHQ). Knock down of Keap1 by RNA interference partially blocked the repression of Nrf2-mediated activation by p65. It was demonstrated that p65 decreased Nrf2 binding to its cognate DNA sequences and enhanced Nrf2 ubiquitination. The N-terminal region of p65 is necessary for both the interaction with Keap1 and its transcriptional suppression activity. Moreover, nuclear translocation of Keap1 was augmented by p65. Taken together, our findings suggest that NF-κB signaling inhibits Nrf2-ARE pathway through the interaction of p65 and Keap1.
Collapse
Affiliation(s)
- Miao Yu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Ding YL, Xu CW, Wang ZD, Zhan YQ, Li W, Xu WX, Yu M, Ge CH, Li CY, Yang XM. Over-expression of EDAG in the myeloid cell line 32D: induction of GATA-1 expression and erythroid/megakaryocytic phenotype. J Cell Biochem 2010; 110:866-74. [PMID: 20564185 DOI: 10.1002/jcb.22597] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Erythroid differentiation-associated gene (EDAG), a hematopoietic tissue-specific transcription regulator, plays a key role in maintaining the homeostasis of hematopoietic lineage commitment. However, the mechanism and genes regulated by EDAG remain unknown. In this study, we showed that overexpression of EDAG in a myeloid cell line 32D led to an erythroid phenotype with increased number of benzidine-positive cells and up-regulation of erythroid specific surface marker TER119. The megakaryocytic specific marker CD61 was also induced significantly. Using a genome-wide microarray analysis and a twofold change cutoff, we identified 332 genes with reduced expression and 288 genes with increased expression. Among up-regulation genes, transcription factor GATA-1 and its target genes including EKLF, NF-E2, Gfi-1b, hemogen, SCL, hemoglobin alpha, beta and megakaryocytic gene GPIX were increased. Silencing of EDAG by RNA interference in K562 cells resulted in down-regulation of these genes. Taken together, EDAG functions as a positive regulator of erythroid/megakaryocytic differentiation in 32D cells associated with the induction of GATA-1 and its target genes.
Collapse
Affiliation(s)
- Ya-Li Ding
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Zhang W, Wang J, Zhang Y, Yuan Y, Guan W, Jin C, Chen H, Wang X, Yang X, He F. The scaffold protein TANK/I-TRAF inhibits NF-kappaB activation by recruiting polo-like kinase 1. Mol Biol Cell 2010; 21:2500-13. [PMID: 20484576 PMCID: PMC2903677 DOI: 10.1091/mbc.e09-08-0715] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2009] [Revised: 04/12/2010] [Accepted: 05/10/2010] [Indexed: 01/26/2023] Open
Abstract
TANK/I-TRAF is a TRAF-binding protein that negatively regulates NF-kappaB activation. The underlying mechanism of this activity remains unclear. Here we show that TANK directly interacts with PLK1, a conserved cell cycle-regulated kinase. PLK1 inhibits NF-kappaB transcriptional activation induced by TNF-alpha, IL-1beta, or several activators, but not by nuclear transcription factor p65. PLK1 expression reduces the DNA-binding activity of NF-kappaB induced by TNF-alpha. Moreover, endogenous activation of PLK1 reduces the TNF-induced phosphorylation of endogenous IkappaBalpha. PLK1 is bound to NEMO (IKKgamma) through TANK to form a ternary complex in vivo. We describe a new regulatory mechanism for PLK1: PLK1 negatively regulates TNF-induced IKK activation by inhibiting the ubiquitination of NEMO. These findings reveal that the scaffold protein TANK recruits PLK1 to negatively regulate NF-kappaB activation and provide direct evidence that PLK1 is required for the repression function of TANK.
Collapse
Affiliation(s)
- Wanqiao Zhang
- *State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 102206, China; and
| | - Jian Wang
- *State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 102206, China; and
| | - Ying Zhang
- *State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 102206, China; and
| | - Yanzhi Yuan
- *State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 102206, China; and
| | - Wei Guan
- *State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 102206, China; and
| | - Chaozhi Jin
- *State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 102206, China; and
| | - Hui Chen
- *State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 102206, China; and
| | - Xiaohui Wang
- *State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 102206, China; and
| | - Xiaoming Yang
- *State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 102206, China; and
| | - Fuchu He
- *State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 102206, China; and
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|
34
|
Genome-wide identification of TAL1's functional targets: insights into its mechanisms of action in primary erythroid cells. Genome Res 2010; 20:1064-83. [PMID: 20566737 DOI: 10.1101/gr.104935.110] [Citation(s) in RCA: 142] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Coordination of cellular processes through the establishment of tissue-specific gene expression programs is essential for lineage maturation. The basic helix-loop-helix hemopoietic transcriptional regulator TAL1 (formerly SCL) is required for terminal differentiation of red blood cells. To gain insight into TAL1 function and mechanisms of action in erythropoiesis, we performed ChIP-sequencing and gene expression analyses from primary fetal liver erythroid cells. We show that TAL1 coordinates expression of genes in most known red cell-specific processes. The majority of TAL1's genomic targets require direct DNA-binding activity. However, one-fifth of TAL1's target sequences, mainly among those showing high affinity for TAL1, can recruit the factor independently of its DNA binding activity. An unbiased DNA motif search of sequences bound by TAL1 identified CAGNTG as TAL1-preferred E-box motif in erythroid cells. Novel motifs were also characterized that may help distinguish activated from repressed genes and suggest a new mechanism by which TAL1 may be recruited to DNA. Finally, analysis of recruitment of GATA1, a protein partner of TAL1, to sequences occupied by TAL1 suggests that TAL1's binding is necessary prior or simultaneous to that of GATA1. This work provides the framework to study regulatory networks leading to erythroid terminal maturation and to model mechanisms of action of tissue-specific transcription factors.
Collapse
|
35
|
Wang YQ, Chen TM, Lei Y, Feng HL, Wang K, Zhang Y. CGI-100 specific shRNA inhibits proliferation and induces differentiation in leukemia K562 cells. Chin J Cancer Res 2010. [DOI: 10.1007/s11670-010-0126-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
36
|
Hemgn is a direct transcriptional target of HOXB4 and induces expansion of murine myeloid progenitor cells. Blood 2010; 116:711-9. [PMID: 20393131 DOI: 10.1182/blood-2009-07-235341] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
HOXB4, a member of the Homeobox transcription factor family, promotes expansion of hematopoietic stem cells and hematopoietic progenitor cells in vivo and ex vivo when overexpressed. However, the molecular mechanisms underlying this effect are not well understood. To identify direct target genes of HOXB4 in primary murine hematopoietic progenitor cells, we induced HOXB4 function in lineage-negative murine bone marrow cells, using a tamoxifen-inducible HOXB4-ER(T2) fusion protein. Using expression microarrays, 77 probe sets were identified with differentially changed expression in early response to HOXB4 induction. Among them, we show that Hemogen (Hemgn), encoding a hematopoietic-specific nuclear protein of unknown function, is a direct transcriptional target of HOXB4. We show that HOXB4 binds to the promoter region of Hemgn both ex vivo and in vivo. When we overexpressed Hemgn in bone marrow cells, we observed that Hemgn promoted cellular expansion in liquid cultures and increased self-renewal of myeloid colony-forming units in culture, partially recapitulating the effect of HOXB4 overexpression. Furthermore, down-regulation of Hemgn using an shRNA strategy proved that Hemgn contributes to HOXB4-mediated expansion in our myeloid progenitor assays. Our results identify a functionally relevant, direct transcriptional target of HOXB4 and identify other target genes that may also participate in the HOXB4 genetic network.
Collapse
|
37
|
Vande Walle L, Wirawan E, Lamkanfi M, Festjens N, Verspurten J, Saelens X, Vanden Berghe T, Vandenabeele P. The mitochondrial serine protease HtrA2/Omi cleaves RIP1 during apoptosis of Ba/F3 cells induced by growth factor withdrawal. Cell Res 2010; 20:421-33. [PMID: 20125124 DOI: 10.1038/cr.2010.18] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Interleukin-3 (IL-3) deprivation of the mouse pro-B cell line Ba/F3 induces cell death that is abrogated by B-cell lymphoma 2 (Bcl-2) overexpression, but remains unaffected by the pan-caspase inhibitor carbobenzoxy-valyl-analyl-aspartyl-[O-methyl]-fluoromethylketone (zVAD-fmk). IL-3 withdrawal causes receptor-interacting protein (RIP)1 cleavage into C-terminal fragments of 30 and 25 kDa, and only cleavage leading to the former was prevented by zVAD-fmk. siRNA experiments demonstrated that generation of the 25-kDa fragment was due to a Bcl-2-modulated release of the mitochondrial serine protease high temperature requirement protein A2 (HtrA2)/Omi. Accordingly, recombinant HtrA2/Omi efficiently cleaved mouse RIP1 in vitro, generating fragments matching those observed in IL-3-deprived Ba/F3 cells. The HtrA2/Omi cleavage site in mouse RIP1 was mapped to the intermediate domain and the corresponding N- and C-terminal fragments were impaired in their ability to activate nuclear factor-kappaB, c-Jun N-terminal kinase and p38 mitogen-activated protein kinase. Interestingly, knockdown of HtrA2/Omi afforded protection against IL-3 withdrawal-induced death in the presence of zVAD-fmk, demonstrating a role for HtrA2/Omi in caspase-independent cell death during growth factor withdrawal by cleaving RIP1.
Collapse
|
38
|
Marqués-García F, Ferrandiz N, Fernández-Alonso R, González-Cano L, Herreros-Villanueva M, Rosa-Garrido M, Fernández-García B, Vaque JP, Marqués MM, Alonso ME, Segovia JC, León J, Marín MC. p73 plays a role in erythroid differentiation through GATA1 induction. J Biol Chem 2009; 284:21139-56. [PMID: 19509292 DOI: 10.1074/jbc.m109.026849] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The TP73 gene gives rise to transactivation domain-p73 isoforms (TAp73) as well as DeltaNp73 variants with a truncated N terminus. Although TAp73alpha and -beta proteins are capable of inducing cell cycle arrest, apoptosis, and differentiation, DeltaNp73 acts in many cell types as a dominant-negative repressor of p53 and TAp73. It has been proposed that p73 is involved in myeloid differentiation, and its altered expression is involved in leukemic degeneration. However, there is little evidence as to which p73 variants (TA or DeltaN) are expressed during differentiation and whether specific p73 isoforms have the capacity to induce, or hinder, this differentiation in leukemia cells. In this study we identify GATA1 as a direct transcriptional target of TAp73alpha. Furthermore, TAp73alpha induces GATA1 activity, and it is required for erythroid differentiation. Additionally, we describe a functional cooperation between TAp73 and DeltaNp73 in the context of erythroid differentiation in human myeloid cells, K562 and UT-7. Moreover, the impaired expression of GATA1 and other erythroid genes in the liver of p73KO embryos, together with the moderated anemia observed in p73KO young mice, suggests a physiological role for TP73 in erythropoiesis.
Collapse
|
39
|
Sharif-Khatibi L, Kariminia A, Khoei S, Goliaei B. Hyperthermia induces differentiation without apoptosis in permissive temperatures in human erythroleukaemia cells. Int J Hyperthermia 2008; 23:645-55. [PMID: 18097851 DOI: 10.1080/02656730701769833] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
PURPOSE The aim of the present study was to investigate whether induction of differentiation by hyperthermia is accompanied by apoptosis and necrosis to further evaluate the benefits of using hyperthermia as a differentiation inducing physical modality. MATERIALS AND METHOD Differentiation was evaluated in K562 erythroleukaemia cells by measuring haemoglobin synthesis and flow cytometric measurement of glycophorin A expression. Apoptosis was measured by Annexin-V-FITC and Propidium Iodide (PI) double staining assay. Apoptosis and necrosis was also evaluated morphologically using staining with acridine orange/ethidium bromide (AO/EtBr) by fluorescence microscopy. Heat shock protein 70 (HSP70) level was measured by ELISA kit. RESULTS Hyperthermia (43 degrees C) induced differentiation as judged by increased haemoglobin synthesis and glycophorin A expression. No sign of apoptosis or necrosis could be detected at this temperature. Cell viability did not change due to heat treatment, and cellular proliferation was reduced in a dose (heating time) dependent manner. At 45 degrees C, hyperthermia induced apoptosis and necrosis with minimal or no sign of differentiation. HSP70 level was significantly increased at 43 degrees C along with differentiation of leukaemic cells, while at 45 degrees C no significant effect on HSP70 production could be observed. CONCLUSIONS The encouraging results obtained here indicate that by heat treatment at 43 degrees C, hyperthermia can be used alone or in combination with other modalities as a differentiation inducing agent without any detectable apoptotic activity. Positive correlation between HSP70 production and induction of differentiation and lack of apoptosis by hyperthermia confirm the possible role of HSP70 in the heat-induced differentiation and apoptosis in leukaemic cells.
Collapse
|
40
|
Li CY, Zhan YQ, Li W, Xu CW, Xu WX, Yu DH, Peng RY, Cui YF, Yang X, Hou N, Li YH, Dong B, Sun HB, Yang XM. Overexpression of a hematopoietic transcriptional regulator EDAG induces myelopoiesis and suppresses lymphopoiesis in transgenic mice. Leukemia 2007; 21:2277-86. [PMID: 17690693 DOI: 10.1038/sj.leu.2404901] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Erythroid differentiation-associated gene (EDAG) is a hematopoietic tissue-specific gene that is highly expressed in the earliest CD34+ lin- bone marrow (BM) cells and involved in the proliferation and differentiation of hematopoietic cells. To investigate the role of EDAG in hematopoiesis, we established an EDAG transgenic mouse model driven by human CD11a promoter. The transgenic mice showed increased mortality with severe organ infiltration by neutrophils, and the homeostasis of hematopoiesis was broken. The myelopoiesis was enhanced with expansion of myeloid cells in BM, increased peripheral granulocytes and extramedullary myelopoiesis in spleen. In contrast to myeloid cells, the lymphoid commitment was severely impaired with the B lymphopoiesis blocked at the transition from pro/pre-B I to pre-B II stage in BM and T thymocytes development blocked at the most immature stage (DN I). Moreover, we showed that EDAG was a transcriptional regulator which had transactivation activity and regulated the expression of several key transcription factors such as PU.1 and Pax5 in transgenic hematopoietic stem cells. These data suggested that EDAG was a key transcriptional regulator in maintaining the homeostasis of hematopoietic lineage commitment.
Collapse
Affiliation(s)
- C-Y Li
- Department of Biochemistry and Molecular Biology, Beijing Institute of Radiation Medicine, Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Zhao M, Chen R, Li A, Zhou J. Effects of hemin and thermal stress exposure on JWA expression. FRONTIERS OF MEDICINE IN CHINA 2007; 1:104-108. [PMID: 24557627 DOI: 10.1007/s11684-007-0020-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
To investigate the expression of JWA after hemin and (or) thermal stress exposure, we treated K562 (chronic myelogenous leukemia cells) cells with different doses of hemin and thermal stress using different exposure times. The expression of JWA protein was determined by Western blot analysis. Reverse transcription-polymerase chain reaction was carried out to determine JWA mRNA expression. JWA promoter transcription activity analysis was performed by chloramphenicol acetyl transferase-enzyme linked immunosorbent assay (CAT-ELISA). The expression of JWA protein was significantly increased by up to (3.23 ± 0.57) folds compared to the control in K562 cells after hemin treatment (50 μM for one week), and a similar pattern was observed in the cells after treatment with thermal stress (42°C) for 2 hours [increased by (8.00 ± 1.73) folds]. The expression of JWA mRNA was also significantly elevated by up to (1.37 ± 0.06) folds in K562 cells treated with hemin (30 μM for 48 hours), and a similar regulatory pattern [increased by (1.87 ± 0.13) folds] was observed with thermal stress exposure (42°C) for 30 minutes. However, a combined antagonistic effect was observed in the treatment of K562 cells with hemin (30 μM, 48 h) followed by thermal stress (42°C, 30 min). CAT-ELISA further confirmed that either hemin or thermal stress treatment could up-regulate JWA transcription activity, however, the effects could be counteracted partly by treatment with a combination of both. Hemin and thermal stress might regulate JWA expression via distinct intracellular signal transduction pathways.
Collapse
Affiliation(s)
- Ming Zhao
- Department of Molecular Cell Biology and Toxicology, School of Public Health, Nanjing Medical University, Nanjing, 210029, China
| | | | | | | |
Collapse
|
42
|
Yang LV, Wan J, Ge Y, Fu Z, Kim SY, Fujiwara Y, Taub JW, Matherly LH, Eliason J, Li L. The GATA site-dependent hemogen promoter is transcriptionally regulated by GATA1 in hematopoietic and leukemia cells. Leukemia 2006; 20:417-25. [PMID: 16437149 DOI: 10.1038/sj.leu.2404105] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Hemgn (a gene symbol for hemogen in mouse, EDAG in human and RP59 in rat) encodes a nuclear protein that is highly expressed in hematopoietic tissues and acute leukemia. To characterize its regulatory mechanisms, we examined the activities of a Hemgn promoter containing 2975 bp of 5' flanking sequence and 196 bp of 5' untranslated region (5' UTR) sequence both in vitro and in vivo: this promoter is preferentially activated in a hematopoietic cell line, not in nonhematopoietic cell lines, and is sufficient to drive the transcription of a lacZ transgene in hematopoietic tissues in transgenic mice. Mutagenesis analyses showed that the 5' UTR including two highly conserved GATA boxes is critical for the promoter activity. GATA1, not GATA2, binds to the GATA binding sites and transactivates the Hemgn promoter in a dose-dependent manner. Furthermore, the expression of human hemogen (EDAG) transcripts were closely correlated with levels of GATA1 transcripts in primary acute myeloid leukemia specimens. This study suggests that the Hemgn promoter contains critical regulatory elements for its transcription in hematopoietic tissues and Hemgn is a direct target of GATA1 in leukemia cells.
Collapse
Affiliation(s)
- L V Yang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Marcucci G, Baldus CD, Ruppert AS, Radmacher MD, Mrózek K, Whitman SP, Kolitz JE, Edwards CG, Vardiman JW, Powell BL, Baer MR, Moore JO, Perrotti D, Caligiuri MA, Carroll AJ, Larson RA, de la Chapelle A, Bloomfield CD. Overexpression of the ETS-related gene, ERG, predicts a worse outcome in acute myeloid leukemia with normal karyotype: a Cancer and Leukemia Group B study. J Clin Oncol 2005; 23:9234-42. [PMID: 16275934 DOI: 10.1200/jco.2005.03.6137] [Citation(s) in RCA: 198] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE To test the prognostic significance of ETS-related gene (ERG) expression in cytogenetically normal primary acute myeloid leukemia (AML). PATIENTS AND METHODS Pretreatment blood samples from 84 cytogenetically normal AML patients aged less than 60 years, who were characterized for BAALC expression, FLT3 internal tandem duplication (ITD), and MLL partial tandem duplication (PTD) and uniformly treated on Cancer and Leukemia Group B 9621 protocol, were analyzed for ERG expression by real-time reverse transcriptase polymerase chain reaction. Patients were divided into quartiles according to ERG levels and were compared for clinical outcome. High-density oligonucleotide arrays were used to identify genes differentially expressed between high and low ERG expressers. RESULTS With a median follow-up of 5.7 years, patients with the upper 25% of ERG expression values had a worse cumulative incidence of relapse (CIR; P < .001) and overall survival (OS; P = .011) than the remaining patients. In a multivariable analysis, high ERG expression (P < .001) and the presence of MLL PTD (P = .027) predicted worse CIR. With regard to OS, an interaction was observed between expression of ERG and BAALC (P = .013), with ERG overexpression predicting shorter survival only in low BAALC expressers (P = .002). ERG overexpression was an independent prognostic factor even when the unfavorable group of FLT3 ITD patients lacking an FLT3 wild-type allele was included. High ERG expression was associated with upregulation of 112 expressed-sequenced tags and named genes, many of which are involved in cell proliferation, differentiation, and apoptosis. CONCLUSION ERG overexpression in AML patients with normal cytogenetics predicts an adverse clinical outcome and seems to be associated with a specific molecular signature.
Collapse
Affiliation(s)
- Guido Marcucci
- Division of Hematology and Oncology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus OH 43210, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
An LL, Li G, Wu KF, Ma XT, Zheng GG, Qiu LG, Song YH. High expression of EDAG and its significance in AML. Leukemia 2005; 19:1499-502. [PMID: 15920494 DOI: 10.1038/sj.leu.2403808] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|