1
|
Wittorf KJ, Weber KK, Swenson SA, Buckley SM. Ubiquitin E3 ligase FBXO21 regulates cytokine-mediated signaling pathways, but is dispensable for steady-state hematopoiesis. Exp Hematol 2022; 114:33-42.e3. [PMID: 35987460 DOI: 10.1016/j.exphem.2022.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/03/2022] [Accepted: 08/04/2022] [Indexed: 11/24/2022]
Abstract
Hematopoietic cell fate decisions such as self-renewal and differentiation are highly regulated through multiple molecular pathways. One pathway, the ubiquitin proteasome system (UPS), controls protein levels by tagging them with polyubiquitin chains and promoting their degradation through the proteasome. Ubiquitin E3 ligases serve as the substrate-recognition component of the UPS. By investigating the FBOX family of E3 ligases, we discovered that Fbxo21 was highly expressed in the hematopoietic stem and progenitor cell (HSPC) population, and exhibited low to no expression in mature myeloid populations. To determine the role of FBXO21 on HSPC maintenance, self-renewal, and differentiation, we generated shRNAs against FBXO21 and a hematopoiesis-specific Fbxo21 conditional knockout (cKO) mouse model. We found that silencing FBXO21 in HSPCs led to a loss in colony formation and an increase in cell differentiation in vitro. Additionally, stressing the HSPC populations in our Fbxo21 cKO mouse with 5-fluorouracil injections resulted in a decrease in survival, despite these populations exhibiting minimal alterations during steady-state hematopoiesis. Although FBXO21 has previously been proposed to regulate cytokine signaling via ASK and p38, our results indicate that depletion of FBXO21 led to altered ERK signaling in vitro. Together, these findings suggest ubiquitin E3 ligase FBXO21 regulates HSPCs through cytokine-mediated pathways.
Collapse
Affiliation(s)
- Karli J Wittorf
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE
| | - Kasidy K Weber
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE
| | - Samantha A Swenson
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE
| | - Shannon M Buckley
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE.
| |
Collapse
|
2
|
Sasaki Y, Guo YM, Goto T, Ubukawa K, Asanuma K, Kobayashi I, Sawada K, Wakui H, Takahashi N. IL-6 Generated from Human Hematopoietic Stem and Progenitor Cells through TLR4 Signaling Promotes Emergency Granulopoiesis by Regulating Transcription Factor Expression. THE JOURNAL OF IMMUNOLOGY 2021; 207:1078-1086. [PMID: 34341172 DOI: 10.4049/jimmunol.2100168] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 06/16/2021] [Indexed: 11/19/2022]
Abstract
Emergency granulopoiesis, also known as demand-adapted granulopoiesis, is defined as the response of an organism to systemic bacterial infections, and it results in neutrophil mobilization from reservoir pools and increased myelopoiesis in the bone marrow. Indirect and direct initiating mechanisms of emergency granulopoiesis have been hypothesized. However, the detailed mechanism of hyperactive myelopoiesis in the bone marrow, which leads to granulocyte left shift, remains unknown. In this study, we report that TLR4 is expressed on granulo-monocytic progenitors, as well as mobilized human peripheral blood CD34+ cells, which account for 0.2% of monocytes in peripheral blood, and ∼ 10% in bone marrow. LPS, a component of Gram-negative bacteria that results in a systemic bacterial infection, induces the differentiation of peripheral blood CD34+ cells into myelocytes and monocytes in vitro via the TLR4 signaling pathway. Moreover, CD34+ cells directly responded to LPS stimulation by activating the MAPK and NF-κB signaling pathways, and they produced IL-6 that promotes emergency granulopoiesis by phosphorylating C/EBPα and C/EBPβ, and this effect was suppressed by the action of an IL-6 receptor inhibitor. This work supports the finding that TLR is expressed on human hematopoietic stem and progenitor cells, and it provides evidence that human hematopoietic stem and progenitor cells can directly sense pathogens and produce cytokines exerting autocrine and/or paracrine effects, thereby promoting differentiation.
Collapse
Affiliation(s)
- Yumi Sasaki
- Department of Life Science, Graduate School of Engineering Science, Akita University, Akita, Japan
| | - Yong-Mei Guo
- Department of Hematology, Nephrology, and Rheumatology, Akita University Graduate School of Medicine, Akita, Japan;
| | - Tatsufumi Goto
- Department of Life Science, Graduate School of Engineering Science, Akita University, Akita, Japan
| | - Kumi Ubukawa
- Department of Hematology, Nephrology, and Rheumatology, Akita University Graduate School of Medicine, Akita, Japan
| | - Ken Asanuma
- Division of Radio Isotope, Bioscience Education and Research Support Center, Akita University School of Medicine, Akita, Japan; and
| | - Isuzu Kobayashi
- Department of Hematology, Nephrology, and Rheumatology, Akita University Graduate School of Medicine, Akita, Japan
| | - Kenichi Sawada
- Medical Corporation Hokubukai Utsukushigaoka Hospital, Hokkaido, Japan
| | - Hideki Wakui
- Department of Life Science, Graduate School of Engineering Science, Akita University, Akita, Japan
| | - Naoto Takahashi
- Department of Hematology, Nephrology, and Rheumatology, Akita University Graduate School of Medicine, Akita, Japan
| |
Collapse
|
3
|
Nie Y, Su L, Li W, Gao S. Novel insights of acute myeloid leukemia with CEBPA deregulation: Heterogeneity dissection and re-stratification. Crit Rev Oncol Hematol 2021; 163:103379. [PMID: 34087345 DOI: 10.1016/j.critrevonc.2021.103379] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 03/21/2021] [Accepted: 05/29/2021] [Indexed: 12/17/2022] Open
Abstract
Acute myeloid leukemia with bi-allelic CEBPA mutation was categorized as an independent disease entity with favorable prognosis, however, recent researches have revealed huge heterogeneity within this disease group, and for some patients, relapse remained a major cause of treatment failure. Further risk stratification is essentially needed. Here by reviewing the latest literature, we summarized the characteristics of CEBPA mutation profiles and clinical features, with a special intention of dissecting the heterogeneity within the seemingly homogeneous AML with bi-allelic CEBPA mutations. Specifically, non-classical CEBPA mutation, miscellaneous companion genetic aberrations and the presence of germline CEBPA mutation are three major sources of heterogeneity. Identifying these factors can help us predict patients at a higher risk of relapse, for whom aggressive treatment may be recommended. Novel therapeutic approaches regarding manipulating potentially druggable targets as well as the debate over post remission consolidation regimens has also been discussed.
Collapse
Affiliation(s)
- Yuanyuan Nie
- Department of Hematology, The First Hospital of Jilin University, Changchun, 130012, China
| | - Long Su
- Department of Hematology, The First Hospital of Jilin University, Changchun, 130012, China
| | - Wei Li
- Department of Hematology, The First Hospital of Jilin University, Changchun, 130012, China; Stem Cell and Cancer Center, The First Hospital of Jilin University, Changchun, 130012, China
| | - Sujun Gao
- Department of Hematology, The First Hospital of Jilin University, Changchun, 130012, China.
| |
Collapse
|
4
|
Identification of Potential Key lncRNAs in the Context of Mouse Myeloid Differentiation by Systematic Transcriptomics Analysis. Genes (Basel) 2021; 12:genes12050630. [PMID: 33922442 PMCID: PMC8146222 DOI: 10.3390/genes12050630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 04/09/2021] [Accepted: 04/21/2021] [Indexed: 11/17/2022] Open
Abstract
Hematopoietic differentiation is a well-orchestrated process by many regulators such as transcription factor and long non-coding RNAs (lncRNAs). However, due to the large number of lncRNAs and the difficulty in determining their roles, the study of lncRNAs is a considerable challenge in hematopoietic differentiation. Here, through gene co-expression network analysis over RNA-seq data generated from representative types of mouse myeloid cells, we obtained a catalog of potential key lncRNAs in the context of mouse myeloid differentiation. Then, employing a widely used in vitro cell model, we screened a novel lncRNA, named Gdal1 (Granulocytic differentiation associated lncRNA 1), from this list and demonstrated that Gdal1 was required for granulocytic differentiation. Furthermore, knockdown of Cebpe, a principal transcription factor of granulocytic differentiation regulation, led to down-regulation of Gdal1, but not vice versa. In addition, expression of genes involved in myeloid differentiation and its regulation, such as Cebpa, were influenced in Gdal1 knockdown cells with differentiation blockage. We thus systematically identified myeloid differentiation associated lncRNAs and substantiated the identification by investigation of one of these lncRNAs on cellular phenotype and gene regulation levels. This study promotes our understanding of the regulation of myeloid differentiation and the characterization of roles of lncRNAs in hematopoietic system.
Collapse
|
5
|
Polak A, Bialopiotrowicz E, Krzymieniewska B, Wozniak J, Stojak M, Cybulska M, Kaniuga E, Mikula M, Jablonska E, Gorniak P, Noyszewska-Kania M, Szydlowski M, Piechna K, Piwocka K, Bugajski L, Lech-Maranda E, Barankiewicz J, Kolkowska-Lesniak A, Patkowska E, Glodkowska-Mrowka E, Baran N, Juszczynski P. SYK inhibition targets acute myeloid leukemia stem cells by blocking their oxidative metabolism. Cell Death Dis 2020; 11:956. [PMID: 33159047 PMCID: PMC7648638 DOI: 10.1038/s41419-020-03156-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 10/17/2020] [Accepted: 10/19/2020] [Indexed: 12/15/2022]
Abstract
Spleen tyrosine kinase (SYK) is an important oncogene and signaling mediator activated by cell surface receptors crucial for acute myeloid leukemia (AML) maintenance and progression. Genetic or pharmacologic inhibition of SYK in AML cells leads to increased differentiation, reduced proliferation, and cellular apoptosis. Herein, we addressed the consequences of SYK inhibition to leukemia stem-cell (LSC) function and assessed SYK-associated pathways in AML cell biology. Using gain-of-function MEK kinase mutant and constitutively active STAT5A, we demonstrate that R406, the active metabolite of a small-molecule SYK inhibitor fostamatinib, induces differentiation and blocks clonogenic potential of AML cells through the MEK/ERK1/2 pathway and STAT5A transcription factor, respectively. Pharmacological inhibition of SYK with R406 reduced LSC compartment defined as CD34+CD38−CD123+ and CD34+CD38−CD25+ in vitro, and decreased viability of LSCs identified by a low abundance of reactive oxygen species. Primary leukemic blasts treated ex vivo with R406 exhibited lower engraftment potential when xenotransplanted to immunodeficient NSG/J mice. Mechanistically, these effects are mediated by disturbed mitochondrial biogenesis and suppression of oxidative metabolism (OXPHOS) in LSCs. These mechanisms appear to be partially dependent on inhibition of STAT5 and its target gene MYC, a well-defined inducer of mitochondrial biogenesis. In addition, inhibition of SYK increases the sensitivity of LSCs to cytarabine (AraC), a standard of AML induction therapy. Taken together, our findings indicate that SYK fosters OXPHOS and participates in metabolic reprogramming of AML LSCs in a mechanism that at least partially involves STAT5, and that SYK inhibition targets LSCs in AML. Since active SYK is expressed in a majority of AML patients and confers inferior prognosis, the combination of SYK inhibitors with standard chemotherapeutics such as AraC constitutes a new therapeutic modality that should be evaluated in future clinical trials.
Collapse
Affiliation(s)
- Anna Polak
- Department of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Emilia Bialopiotrowicz
- Department of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Beata Krzymieniewska
- Department of Diagnostic Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Jolanta Wozniak
- Department of Diagnostic Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Marta Stojak
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Kraków, Poland
| | - Magdalena Cybulska
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Ewelina Kaniuga
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Michał Mikula
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Ewa Jablonska
- Department of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Patryk Gorniak
- Department of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Monika Noyszewska-Kania
- Department of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Maciej Szydlowski
- Department of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Karolina Piechna
- Department of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Katarzyna Piwocka
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Lukasz Bugajski
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Ewa Lech-Maranda
- Department of Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Joanna Barankiewicz
- Department of Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | | | - Elzbieta Patkowska
- Department of Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Eliza Glodkowska-Mrowka
- Department of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Natalia Baran
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Przemyslaw Juszczynski
- Department of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland.
| |
Collapse
|
6
|
Caiment F, Wolters J, Smit E, Schrooders Y, Kleinjans J, van den Beucken T. Valproic acid promotes mitochondrial dysfunction in primary human hepatocytes in vitro; impact of C/EBPα-controlled gene expression. Arch Toxicol 2020; 94:3463-3473. [PMID: 32623605 PMCID: PMC7502062 DOI: 10.1007/s00204-020-02835-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 06/30/2020] [Indexed: 11/29/2022]
Abstract
Valproic acid (VPA) is a frequently prescribed anti-epileptic drug which is known to cause liver toxicity and steatosis through mitochondrial dysfunction. Nevertheless the mechanisms underlying these adverse effects are incompletely understood. In this study, we determined the effect of relatively short (3 h) or prolonged (72 h) exposure to VPA on mitochondrial function in primary human hepatocytes (PHHs). While 3 h VPA exposure did not affect oxygen consumption rates (OCRs) in PHHs, prolonged exposure (24–72 h) significantly reduced basal and maximal OCRs. Given that in particular prolonged VPA exposure is required to cause mitochondrial dysfunction, we investigated gene expression data after VPA exposure for 24, 48, 72 h and 72 h VPA followed by a 72 h washout period. We were able to reduce the comprehensive gene expression changes into a more comprehensible set of 18 TFs that were predicted to be persistently activated after 72 h of VPA exposure. Lentiviral knock-down of one of the candidate TFs, C/EBPα, partly rescued VPA-induced mitochondrial dysfunction. Furthermore, RNA-Seq analysis of shC/EBPα and shGFP control PHHs identified 24 genuine C/EBPα target genes that are regulated in response to prolonged VPA exposure in PHHs. Altogether this provides new insights on the involvement of C/EBPα in driving VPA-induced mitochondrial dysfunction in human liver cells. This hub gene, with its downstream regulators involved in this deregulation, thus represent potential new biomarkers for VPA-induced mitochondrial dysfunction.
Collapse
Affiliation(s)
- F Caiment
- Maastricht University, Maastricht, The Netherlands
| | - J Wolters
- Maastricht University, Maastricht, The Netherlands
| | - E Smit
- Maastricht University, Maastricht, The Netherlands
| | - Y Schrooders
- Maastricht University, Maastricht, The Netherlands
| | - J Kleinjans
- Maastricht University, Maastricht, The Netherlands
| | | |
Collapse
|
7
|
E3 ligase SCF SKP2 ubiquitinates and degrades tumor suppressor C/EBPα in acute myeloid leukemia. Life Sci 2020; 257:118041. [PMID: 32622945 DOI: 10.1016/j.lfs.2020.118041] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 06/18/2020] [Accepted: 06/29/2020] [Indexed: 12/23/2022]
Abstract
AIM Transcription factor CCAAT/Enhancer binding protein alpha (C/EBPα) is a key regulator of myeloid differentiation, granulopoiesis in particular. Although CEBPA mutations are found in more than 10% in AML, functional inhibition of C/EBPα protein is also widely observed in AML. Here, we sought to examine if SKP2, an aberrantly enhanced E3 ubiquitin ligase in primary AMLs inhibits C/EBPα stability to induce differentiation block. MAIN METHODS Here we employed cell based assays such as transfections, immunoblotting, co-immunoprecipitation, luciferase and gel shift assays along with differentiation assays to investigate SKP2 regulated C/EBPα protein stability in acute myeloid leukemia. KEY FINDINGS Here we discovered that oncogenic E3 ubiquitin ligase SCFskp2 ubiquitinates and destabilizes C/EBPα in a proteasome-dependent manner. Our data demonstrates that SKP2 physically interacts with C-terminal of C/EBPα and promotes its K48-linked ubiquitination-mediated degradation leading to its reduced transactivation potential, DNA binding ability and cellular functions. We further show that while overexpression of SKP2 inhibits both ectopic as well as endogenous C/EBPα in heterologous (HEK293T) as well as myeloid leukemia cells respectively, SKP2 depletion restores endogenous C/EBPα leading to reduced colony formation and enhanced myeloid differentiation of myeloid leukemia cells. Using Estradiol-inducible K562-C/EBPα-ER cells as yet another model of granulocytic differentiation, we further confirmed that SKP2 overexpression indeed inhibits granulocytic differentiation by mitigating C/EBPα stability. SIGNIFICANCE Our findings identify SKP2 as a potential negative regulator of C/EBPα stability and function in AML which suggests that SKP2 can be potentially targeted in AML to restore C/EBPα and overcome differentiation block.
Collapse
|
8
|
Nacht AS, Ferrari R, Zaurin R, Scabia V, Carbonell-Caballero J, Le Dily F, Quilez J, Leopoldi A, Brisken C, Beato M, Vicent GP. C/EBPα mediates the growth inhibitory effect of progestins on breast cancer cells. EMBO J 2019; 38:e101426. [PMID: 31373033 DOI: 10.15252/embj.2018101426] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 06/21/2019] [Accepted: 06/24/2019] [Indexed: 01/19/2023] Open
Abstract
Steroid hormones are key gene regulators in breast cancer cells. While estrogens stimulate cell proliferation, progestins activate a single cell cycle followed by proliferation arrest. Here, we use biochemical and genome-wide approaches to show that progestins achieve this effect via a functional crosstalk with C/EBPα. Using ChIP-seq, we identify around 1,000 sites where C/EBPα binding precedes and helps binding of progesterone receptor (PR) in response to hormone. These regions exhibit epigenetic marks of active enhancers, and C/EBPα maintains an open chromatin conformation that facilitates loading of ligand-activated PR. Prior to hormone exposure, C/EBPα favors promoter-enhancer contacts that assure hormonal regulation of key genes involved in cell proliferation by facilitating binding of RAD21, YY1, and the Mediator complex. Knockdown of C/EBPα disrupts enhancer-promoter contacts and decreases the presence of these architectural proteins, highlighting its key role in 3D chromatin looping. Thus, C/EBPα fulfills a previously unknown function as a potential growth modulator in hormone-dependent breast cancer.
Collapse
Affiliation(s)
- A Silvina Nacht
- Center for Genomic Regulation (CRG), Barcelona, Spain.,Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Roberto Ferrari
- Center for Genomic Regulation (CRG), Barcelona, Spain.,Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Roser Zaurin
- Center for Genomic Regulation (CRG), Barcelona, Spain.,Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Valentina Scabia
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - José Carbonell-Caballero
- Center for Genomic Regulation (CRG), Barcelona, Spain.,Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Francois Le Dily
- Center for Genomic Regulation (CRG), Barcelona, Spain.,Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Javier Quilez
- Center for Genomic Regulation (CRG), Barcelona, Spain.,Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Alexandra Leopoldi
- Center for Genomic Regulation (CRG), Barcelona, Spain.,Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Cathrin Brisken
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Miguel Beato
- Center for Genomic Regulation (CRG), Barcelona, Spain.,Barcelona Institute for Science and Technology (BIST), Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Guillermo P Vicent
- Center for Genomic Regulation (CRG), Barcelona, Spain.,Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| |
Collapse
|
9
|
Zhou L, Zhang T, Zhu Q, Zhang P, Yu L, Shen B, Yi W, Qiu M, Zhu C. Mina53 regulates the differentiation and proliferation of leukemia cells. Hematol Oncol 2019; 37:513-515. [DOI: 10.1002/hon.2621] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 04/11/2019] [Indexed: 11/06/2022]
Affiliation(s)
- Lixiao Zhou
- College of Life Sciences Zhejiang University Hangzhou China
- Key Laboratory of Organ Development and Regeneration of Zhejiang Province, Institute of Life Sciences Hangzhou Normal University Hangzhou China
| | - Ting Zhang
- College of Medical Technology Zhejiang Chinese Medical University Hangzhou China
| | - Qiang Zhu
- College of Life Sciences Zhejiang University Hangzhou China
| | | | - Liyang Yu
- College of Life Sciences Zhejiang University Hangzhou China
| | - Binghui Shen
- Department of Cancer Genetics and Epigenetics Beckman Research Institute, City of Hope Duarte California
| | - Wen Yi
- College of Life Sciences Zhejiang University Hangzhou China
| | - Mengsheng Qiu
- College of Life Sciences Zhejiang University Hangzhou China
- Key Laboratory of Organ Development and Regeneration of Zhejiang Province, Institute of Life Sciences Hangzhou Normal University Hangzhou China
| | - Chenggang Zhu
- College of Life Sciences Zhejiang University Hangzhou China
| |
Collapse
|
10
|
Liu LM, Sun WZ, Fan XZ, Xu YL, Cheng MB, Zhang Y. Methylation of C/EBPα by PRMT1 Inhibits Its Tumor-Suppressive Function in Breast Cancer. Cancer Res 2019; 79:2865-2877. [PMID: 31015230 DOI: 10.1158/0008-5472.can-18-3211] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 01/24/2019] [Accepted: 04/17/2019] [Indexed: 11/16/2022]
Abstract
C/EBPα is an essential transcription factor involved in regulating the expression or function of certain cell-cycle regulators, including in breast cancer cells. Although protein arginine methyltransferases have been shown to play oncogenic roles in a variety of cancers, little is known about the role of arginine methylation in regulating the antiproliferation activity of C/EBPα. Here, we report that the protein arginine methyltransferase 1 (PRMT1) is overexpressed in human breast cancer and that elevated PRMT1 correlates with cancer malignancy. RNA-sequencing analysis revealed that knockdown of PRMT1 in breast cancer cells is accompanied by a decrease in the expression of pro-proliferative genes, including cyclin D1. Furthermore, tandem affinity purification followed by mass spectrometry identified PRMT1 as a component of the C/EBPα complex. C/EBPα associated with and was methylated by PRMT1 at three arginine residues (R35, R156, and R165). PRMT1-dependent methylation of C/EBPα promoted the expression of cyclin D1 by blocking the interaction between C/EBPα and its corepressor HDAC3, which resulted in rapid growth of tumor cells during the pathogenesis of breast cancer. Inhibition of PRMT1 significantly impeded the growth of cancer cells from patients with triple-negative breast cancer. This evidence that PRMT1 mediates C/EBPα methylation sheds light on a novel pathway and potential therapeutic target in breast cancer. SIGNIFICANCE: This study provides novel mechanistic insight of the role of the arginine methyltransferase PRMT1 in breast cancer pathogenesis.
Collapse
Affiliation(s)
- Li-Ming Liu
- State Key Laboratory of Medical Molecular Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Wen-Zheng Sun
- State Key Laboratory of Medical Molecular Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xue-Zhe Fan
- State Key Laboratory of Medical Molecular Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ya-Li Xu
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Mo-Bin Cheng
- State Key Laboratory of Medical Molecular Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China. .,Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ye Zhang
- State Key Laboratory of Medical Molecular Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China. .,Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
11
|
Hashimoto A, Gao C, Mastio J, Kossenkov A, Abrams SI, Purandare AV, Desilva H, Wee S, Hunt J, Jure-Kunkel M, Gabrilovich DI. Inhibition of Casein Kinase 2 Disrupts Differentiation of Myeloid Cells in Cancer and Enhances the Efficacy of Immunotherapy in Mice. Cancer Res 2018; 78:5644-5655. [PMID: 30139814 DOI: 10.1158/0008-5472.can-18-1229] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 07/09/2018] [Accepted: 07/31/2018] [Indexed: 12/23/2022]
Abstract
The role of myeloid cells as regulators of tumor progression that significantly impact the efficacy of cancer immunotherapies makes them an attractive target for inhibition. Here we explore the effect of a novel, potent, and selective inhibitor of serine/threonine protein kinase casein kinase 2 (CK2) on modulating myeloid cells in the tumor microenvironment. Although inhibition of CK2 caused only a modest effect on dendritic cells in tumor-bearing mice, it substantially reduced the amount of polymorphonuclear myeloid-derived suppressor cells and tumor-associated macrophages. This effect was not caused by the induction of apoptosis, but rather by a block of differentiation. Our results implicated downregulation of CCAAT-enhancer binding protein-α in this effect. Although CK2 inhibition did not directly affect tumor cells, it dramatically enhanced the antitumor activity of immune checkpoint receptor blockade using anti-CTLA-4 antibody. These results suggest a potential role of CK2 inhibitors in combination therapies against cancer.Significance: These findings demonstrate the modulatory effects of casein kinase 2 inhibitors on myeloid cell differentiation in the tumor microenvironment, which subsequently synergize with the antitumor effects of checkpoint inhibitor CTLA4. Cancer Res; 78(19); 5644-55. ©2018 AACR.
Collapse
Affiliation(s)
| | - Chan Gao
- Bristol-Myers Squibb, Princeton, New Jersey
| | | | | | - Scott I Abrams
- Roswell Park Comprehensive Cancer Center, Department of Immunology, Buffalo, New York, Medimmune, Gaithersburg, Maryland
| | | | | | - Susan Wee
- Bristol-Myers Squibb, Princeton, New Jersey
| | | | | | | |
Collapse
|
12
|
Qiu J, Wang W, Hu S, Wang Y, Sun W, Hu J, Gan X, Wang J. Molecular cloning, characterization and expression analysis of C/EBP α, β and δ in adipose-related tissues and adipocyte of duck ( Anas platyrhynchos ). Comp Biochem Physiol B Biochem Mol Biol 2018; 221-222:29-43. [DOI: 10.1016/j.cbpb.2018.04.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 04/14/2018] [Accepted: 04/16/2018] [Indexed: 12/17/2022]
|
13
|
Zha JS, Zhu BL, Liu L, Lai YJ, Long Y, Hu XT, Deng XJ, Wang XF, Yan Z, Chen GJ. Phorbol esters dPPA/dPA promote furin expression involving transcription factor CEBPβ in neuronal cells. Oncotarget 2017; 8:60159-60172. [PMID: 28947961 PMCID: PMC5601129 DOI: 10.18632/oncotarget.18569] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 06/10/2017] [Indexed: 02/03/2023] Open
Abstract
Using high-throughput small molecule screening targeting furin gene, we identified that phorbol esters dPPA (12-Deoxyphorbol 13-phenylacetate 20-acetate) and dPA (12-Deoxyphorbol 13-acetate) significantly increased furin protein and mRNA expression in SH-SY5Y cells. This effect was prevented by PKC (protein kinase C) inhibitor calphostin C but not Ro318220, suggesting that the C1 domain, rather than the catalytic domain of PKC plays an important role. Luciferase assay revealed that nucleotides -7925 to -7426 were sufficient to mediate dPPA/dPA enhancement of furin P1 promoter activity. RNA interference of transcriptional factors CEBPβ (CCAAT/enhancer-binding protein β) and GATA1 revealed that knockdown of CEBPβ significantly attenuated the effect of dPPA on furin expression. Pharmacological inhibition of ERK and PI3K but not TGFβ receptor diminished the up-regulation of furin by dPPA. These results suggested that in neuronal cells, transcriptional activation of furin by dPPA/dPA may be initiated by C1 domain containing proteins including PKC; the intracellular signaling involves ERK and PI3K and transcription factor CEBPβ.
Collapse
Affiliation(s)
- Jing-Si Zha
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - Bing-Lin Zhu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - Lu Liu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - Yu-Jie Lai
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - Yan Long
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - Xiao-Tong Hu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - Xiao-Juan Deng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - Xue-Feng Wang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - Zhen Yan
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, 14214, USA
| | - Guo-Jun Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| |
Collapse
|
14
|
EVI2B is a C/EBPα target gene required for granulocytic differentiation and functionality of hematopoietic progenitors. Cell Death Differ 2017; 24:705-716. [PMID: 28186500 DOI: 10.1038/cdd.2017.6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 12/16/2016] [Accepted: 01/04/2017] [Indexed: 12/29/2022] Open
Abstract
Development of hematopoietic populations through the process of differentiation is critical for proper hematopoiesis. The transcription factor CCAAT/enhancer binding protein alpha (C/EBPα) is a master regulator of myeloid differentiation, and the identification of C/EBPα target genes is key to understand this process. Here we identified the Ecotropic Viral Integration Site 2B (EVI2B) gene as a direct target of C/EBPα. We showed that the product of the gene, the transmembrane glycoprotein EVI2B (CD361), is abundantly expressed on the surface of primary hematopoietic cells, the highest levels of expression being reached in mature granulocytes. Using shRNA-mediated downregulation of EVI2B in human and murine cell lines and in primary hematopoietic stem and progenitor cells, we demonstrated impaired myeloid lineage development and altered progenitor functions in EVI2B-silenced cells. We showed that the compromised progenitor functionality in Evi2b-depleted cells can be in part explained by deregulation of cell proliferation and apoptosis. In addition, we generated an Evi2b knockout murine model and demonstrated altered properties of hematopoietic progenitors, as well as impaired G-CSF dependent myeloid colony formation in the knockout cells. Remarkably, we found that EVI2B is significantly downregulated in human acute myeloid leukemia samples characterized by defects in CEBPA. Altogether, our data demonstrate that EVI2B is a downstream target of C/EBPα, which regulates myeloid differentiation and functionality of hematopoietic progenitors.
Collapse
|
15
|
Gao N, Yu WZ, Guo NJ, Wang XX, Sun JR. Clinical significance of galectin-3 in patients with adult acute myeloid leukemia: a retrospective cohort study with long-term follow-up and formulation of risk scoring system. Leuk Lymphoma 2016; 58:1394-1402. [PMID: 27736291 DOI: 10.1080/10428194.2016.1243677] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Galectin-3 plays an increasingly important role in development and progression of tumor. However, little is known about the clinical impact of galectin-3 in non-acute promyelocytic leukemia (non-M3 AML). Peripheral blood of 298 patients with primary non-M3 AML and 30 normal donors was collected for measurement of galectin-3. Galectin-3 levels were significantly higher compared with the control group (p < .001). Patients with higher galectin-3 levels had lower CR rates (p = .001) and 1-year overall survival (OS) rates (p = .002). The Kaplan-Meier survival analysis showed that higher galectin-3 levels group had significantly shorter OS. Cox regression model revealed high galectin-3 level was an independent poor prognostic factor. A scoring system incorporating galectin-3 and other prognostic factors (age, WBC, karyotype, NPM1/FLT3-ITD, CEBPAdouble-mutation and c-KIT, WT1) was formulated to predict prognosis. In conclusion, galectin-3 may be a reliable prognostic marker in AML patients. The multifactorial scoring system was more powerful than a single factor to predict clinical outcome.
Collapse
Affiliation(s)
- Na Gao
- a Department of hematology , Binzhou Medical University Hospital , Binzhou , Shandong , PR China
| | - Wen-Zheng Yu
- a Department of hematology , Binzhou Medical University Hospital , Binzhou , Shandong , PR China
| | - Nong-Jian Guo
- b Department of hematology, Central Hospital of Jinan , Shandong University School of Medicine , Jinan , Shandong , PR China
| | - Xue-Xia Wang
- a Department of hematology , Binzhou Medical University Hospital , Binzhou , Shandong , PR China
| | - Jian-Rong Sun
- a Department of hematology , Binzhou Medical University Hospital , Binzhou , Shandong , PR China
| |
Collapse
|
16
|
Yan W, Ding A, Kim HJ, Zheng H, Wei F, Ma X. Progranulin Controls Sepsis via C/EBPα-Regulated Il10 Transcription and Ubiquitin Ligase/Proteasome-Mediated Protein Degradation. THE JOURNAL OF IMMUNOLOGY 2016; 197:3393-3405. [PMID: 27619993 DOI: 10.4049/jimmunol.1600862] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 08/15/2016] [Indexed: 11/19/2022]
Abstract
Progranulin (PGRN) is a widely expressed, pleiotropic protein that is involved in diverse biological processes, including cellular proliferation, neuron development, and wound healing. However, the role of PGRN in the regulation of pathogen-induced systemic inflammation and the mechanisms involved have not been established. In this study, we show that PGRN-deficient mice display heightened mortality in models of polymicrobial sepsis and endotoxinemia, with increased tissue levels of inflammatory cytokines and reduced IL-10 production. Conversely, administration of rPGRN decreases the susceptibility of PGRN-deficient mice to LPS-induced endotoxemic shock and augments IL-10 production by LPS-activated macrophages in a TNFR-dependent manner. Molecular analysis reveals a direct role of the transcription factor C/EBPα in PGRN-regulated IL-10 expression. C/EBPα-deficient macrophages produce less IL-10 in response to LPS. Furthermore, mice deficient in C/EBPα in hematopoietic cells are highly vulnerable to LPS-induced septic shock. Lastly, the defective IL-10 production by PGRN-deficient cells is primarily due to reduced C/EBPα protein stability via the E3 ubiquitin-conjugating enzyme E6AP and proteasome-mediated degradation. To our knowledge, this study provides the first evidence that PGRN is a nonredundant regulator of systemic inflammation via modulating the levels and activity of C/EBPα, IL-10, and the ubiquitin-proteasome proteolysis pathway. The results bear strong and profound implications for PGRN insufficiency and its mutation-associated systemic and organ-specific inflammatory human diseases.
Collapse
Affiliation(s)
- Wenjun Yan
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Aihao Ding
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY 10065; and
| | - Ha-Jeong Kim
- Department of Physiology, Kyungpook National University School of Medicine, Jung-gu, Daegu 41944, Republic of Korea
| | - Hua Zheng
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Fang Wei
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiaojing Ma
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China; .,Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY 10065; and
| |
Collapse
|
17
|
Liao YJ, Lee YH, Chang FL, Ho H, Huang CH, Twu YC. The SHP2-ERK2 signaling pathway regulates branched I antigen formation by controlling the binding of CCAAT/enhancer binding protein α to the IGnTC promoter region during erythroid differentiation. Transfusion 2016; 56:2691-2702. [PMID: 27600951 DOI: 10.1111/trf.13796] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 06/20/2016] [Accepted: 06/25/2016] [Indexed: 11/29/2022]
Abstract
BACKGROUND Phosphorylation status of the transcription factor CCAAT/enhancer binding protein α (C/EBPα) has been demonstrated in a human hematopoietic cell model to regulate the formation of branched I antigen by affecting its binding affinity to the promoter region of the IGnTC gene during erythroid and granulocytic differentiation. STUDY DESIGN AND METHODS The K-562 cell line was induced to differentiate into red blood cells (RBCs) or granulocytes by sodium butyrate or retinoic acid, respectively, to study the involvement of three MAP kinase pathways in I antigen synthesis. The regulatory effects of the extracellular signal-regulated kinase (ERK)2-Src homology region 2 domain-containing phosphatase 2 (SHP2) pathway on phosphorylation status and binding affinities of C/EBPα as well as the subsequent activation of IGnTC and synthesis of surface I formation were studied in wild-type K-562 cells and in mutant cells that overexpress ERK2 and SHP2. RESULTS We found that SHP2-ERK2 signaling regulates the phosphorylation status of C/EBPα to alter its binding affinity onto the IGnTC promoter region, thereby activating the synthesis of cell surface I antigen formation during erythropoiesis. CONCLUSION SHP2-ERK2 signaling acts upstream of C/EBPα as a regulator of cell surface I antigen synthesis. Such regulation is specific for RBC but not for granulocyte differentiation.
Collapse
Affiliation(s)
- Yi-Jen Liao
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University
| | - Yen-Hua Lee
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Medical Research, Mackay Memorial Hospital, New Taipei City, Taiwan
| | - Fu-Ling Chang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Hsun Ho
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chin-Han Huang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yuh-Ching Twu
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
18
|
Alachkar H, Mutonga M, Malnassy G, Park JH, Fulton N, Woods A, Meng L, Kline J, Raca G, Odenike O, Takamatsu N, Miyamoto T, Matsuo Y, Stock W, Nakamura Y. T-LAK cell-originated protein kinase presents a novel therapeutic target in FLT3-ITD mutated acute myeloid leukemia. Oncotarget 2016; 6:33410-25. [PMID: 26450903 PMCID: PMC4741775 DOI: 10.18632/oncotarget.5418] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 09/22/2015] [Indexed: 11/25/2022] Open
Abstract
Gain-of-function mutations of FLT3 (FLT3-ITD), comprises up to 30% of normal karyotype acute myeloid leukemia (AML) and is associated with an adverse prognosis. Current FLT3 kinase inhibitors have been tested extensively, but have not yet resulted in a survival benefit and novel therapies are awaited. Here we show that T-LAK cell-originated protein kinase (TOPK), a mitotic kinase highly expressed in and correlated with more aggressive phenotype in several types of cancer, is expressed in AML but not in normal CD34+ cells and that TOPK knockdown decreased cell viability and induced apoptosis. Treatment of AML cells with TOPK inhibitor (OTS514) resulted in a dose-dependent decrease in cell viability with lower IC50 in FLT3-mutated cells, including blasts obtained from patients relapsed after FLT3-inhibitor treatment. Using a MV4-11-engrafted mouse model, we found that mice treated with 7.5 mg/kg IV daily for 3 weeks survived significantly longer than vehicle treated mice (median survival 46 vs 29 days, P < 0.001). Importantly, we identified TOPK as a FLT3-ITD and CEBPA regulated kinase, and that modulating TOPK expression or activity resulted in significant decrease of FLT3 expression and CEBPA phosphorylation. Thus, targeting TOPK in FLT3-ITD AML represents a novel therapeutic approach for this adverse risk subset of AML.
Collapse
Affiliation(s)
- Houda Alachkar
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | - Martin Mutonga
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | - Gregory Malnassy
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | - Jae-Hyun Park
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | - Noreen Fulton
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | - Alex Woods
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | - Liping Meng
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | - Justin Kline
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | - Gordana Raca
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | - Olatoyosi Odenike
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | | | | | - Yo Matsuo
- OncoTherapy Science, Inc., Kanagawa, Japan
| | - Wendy Stock
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | - Yusuke Nakamura
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| |
Collapse
|
19
|
Wang X, Li M, Gao Y, Gao J, Yang W, Liang H, Ji Q, Li Y, Liu H, Huang J, Cheng T, Yuan W. Rheb1-mTORC1 maintains macrophage differentiation and phagocytosis in mice. Exp Cell Res 2016; 344:219-28. [DOI: 10.1016/j.yexcr.2016.04.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 04/21/2016] [Accepted: 04/28/2016] [Indexed: 11/25/2022]
|
20
|
Zhang G, Dong F, Luan C, Zhang X, Shao H, Liu J, Sun C. Overexpression of CCAAT Enhancer-Binding Protein α Inhibits the Growth of K562 Cells via the Foxo3a-Bim Pathway. Acta Haematol 2016; 136:65-70. [PMID: 27161125 DOI: 10.1159/000444517] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 02/07/2016] [Indexed: 11/19/2022]
Abstract
We aimed to investigate the role of CCAAT enhancer-binding protein α (C/EBPα) in the pathogenesis of chronic myeloid leukemia (CML) and the mechanism underlying its effect. Bone marrow specimens from 50 patients with CML and peripheral blood specimens from 20 healthy individuals were collected. K562 cells were treated with imatinib. Subsequently, a stable cell line, K562-C/EBPα, was constructed. Cell proliferation was assayed with cell counting kit-8, and mRNA levels of C/EBPα, forkhead transcription factor FKHRL1 (Foxo3a) and Bim were detected by semiquantitative PCR. The correlation of C/EBPα and BCR-ABL was assessed by Spearman's correlation analysis. The results showed that C/EBPα mRNA levels were significantly reduced in CML patients compared with healthy subjects (p < 0.001) and were negatively correlated with BCR-ABL1 (r = -0.5046, p < 0.01). Additionally, imatinib enhanced the expression of C/EBPα in K562 cells compared with untreated cells (p < 0.05). Overexpression of C/EBPα significantly decreased cell proliferation and upregulated the expressions of the apoptosis-related genes Foxo3a (p < 0.01) and Bim (p < 0.05) in K562 cells. In conclusion, C/EBPα expression was decreased in patients with CML. Imatinib enhances the expression of C/EBPα in K562 cells, and the overexpression of C/EBPα inhibits cell proliferation and increases apoptosis via the Foxo3a-Bim pathway.
Collapse
Affiliation(s)
- Guili Zhang
- Department of Laboratory, Yantai Yuhuangding Hospital, Medical College, Qingdao University, Yantai, China
| | | | | | | | | | | | | |
Collapse
|
21
|
Acetylation of C/EBPα inhibits its granulopoietic function. Nat Commun 2016; 7:10968. [PMID: 27005833 PMCID: PMC4814574 DOI: 10.1038/ncomms10968] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 02/07/2016] [Indexed: 01/01/2023] Open
Abstract
CCAAT/enhancer-binding protein alpha (C/EBPα) is an essential transcription factor for myeloid lineage commitment. Here we demonstrate that acetylation of C/EBPα at lysine residues K298 and K302, mediated at least in part by general control non-derepressible 5 (GCN5), impairs C/EBPα DNA-binding ability and modulates C/EBPα transcriptional activity. Acetylated C/EBPα is enriched in human myeloid leukaemia cell lines and acute myeloid leukaemia (AML) samples, and downregulated upon granulocyte-colony stimulating factor (G-CSF)- mediated granulocytic differentiation of 32Dcl3 cells. C/EBPα mutants that mimic acetylation failed to induce granulocytic differentiation in C/EBPα-dependent assays, in both cell lines and in primary hematopoietic cells. Our data uncover GCN5 as a negative regulator of C/EBPα and demonstrate the importance of C/EBPα acetylation in myeloid differentiation. C/EBPα is an essential transcription factor for myeloid lineage commitment. Here, the authors show that acetylation of C/EBPα at K298 and K302, mediated at least in part by GCN5, impairs C/EBPα DNA binding ability and modulates C/EBPα transcriptional activity.
Collapse
|
22
|
Zhang G, Dong F, Luan C, Zhang X, Shao H, Liu J, Sun C. [The expression and role of the transcription factor C/EBPα in chronic myeloid leukemia]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2015; 36:947-50. [PMID: 26632469 PMCID: PMC7342430 DOI: 10.3760/cma.j.issn.0253-2727.2015.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Indexed: 11/05/2022]
Abstract
OBJECTIVE To investigate the expression and the possible mechanism of the transcription factor C/EBPα in chronic myeloid leukemia(CML). METHODS Bone marrow samples from 50 CML patients(including 33 patients in chronic phase, 7 in accelerated phase and 10 in blast crisis)and peripheral blood specimens of 20 healthy donors were collected. The expression of C/EBPα gene and the effect of Imatinib on its expression was detected by RT- PCR. C/EBPα gene was inserted into lentivirus expression vector pLVX- EGFP- 3FLAG- Puro by recombinant DNA technology to construct C/EBPα stable expression in K562 cells. Cell proliferation was assayed by CCK-8. The expressions of Foxo3a and Bim genes were detected by RT-PCR. RESULTS The level of C/EBPα expression was significantly declined in CML patients compared with that of normal control group(P<0.01)and had negative correlation with bcr- abl expression(Spearman r=- 0.505, P<0.01). The stable K562- C/EBPα cell line was successfully established and confirmed by RT-PCR and Western blot. Cell proliferation ability was lower in the K562- C/EBPα group than that in the non- transfection and mock-vehicle groups. The expressions of Foxo3a and Bim genes were 1.06 ± 0.06 and 0.53 ± 0.07, respectively, which was higher than that of nontransfection and mock-vehicle groups(P<0.01, P<0.05). CONCLUSION C/EBPα expression was decreased in CML patients, overexpression of C/EBPα could inhibit K562 cell growth.
Collapse
Affiliation(s)
- Guili Zhang
- Department of Laboratory, Yantai Yuhuangding Hospital, Medical College, Qingdao University, Yantai 264000, China
| | - Fei Dong
- Department of Laboratory, Yantai Yuhuangding Hospital, Medical College, Qingdao University, Yantai 264000, China
| | - Caifu Luan
- Department of Laboratory, Yantai Yuhuangding Hospital, Medical College, Qingdao University, Yantai 264000, China
| | - Xia Zhang
- Department of Laboratory, Yantai Yuhuangding Hospital, Medical College, Qingdao University, Yantai 264000, China
| | - Huiyuan Shao
- Department of Laboratory, Yantai Yuhuangding Hospital, Medical College, Qingdao University, Yantai 264000, China
| | - Jie Liu
- Department of Laboratory, Yantai Yuhuangding Hospital, Medical College, Qingdao University, Yantai 264000, China
| | - Chengming Sun
- Department of Laboratory, Yantai Yuhuangding Hospital, Medical College, Qingdao University, Yantai 264000, China
| |
Collapse
|
23
|
The Multiple Roles of Microrna-223 in Regulating Bone Metabolism. Molecules 2015; 20:19433-48. [PMID: 26512640 PMCID: PMC6332311 DOI: 10.3390/molecules201019433] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 10/13/2015] [Accepted: 10/20/2015] [Indexed: 12/23/2022] Open
Abstract
Bone metabolism is a lifelong process for maintaining skeletal system homeostasis, which is regulated by bone-resorbing osteoclasts and bone-forming osteoblasts. Aberrant differentiation of osteoclasts and osteoblasts leads to imbalanced bone metabolism, resulting in ossification and osteolysis diseases. MicroRNAs (miRNAs) are pivotal factors in regulating bone metabolism via post-transcriptional inhibition of target genes. Recent studies have revealed that miR-223 exerts multiple effects on bone metabolism, especially in the processes of osteoclast and osteoblasts differentiation. In this review, we highlight the roles of miR-223 during the processes of osteoclast and osteoblast differentiation, as well as the potential clinical applications of miR-223 in bone metabolism disorders.
Collapse
|
24
|
Hu N, Qiu Y, Dong F. Role of Erk1/2 signaling in the regulation of neutrophil versus monocyte development in response to G-CSF and M-CSF. J Biol Chem 2015; 290:24561-73. [PMID: 26296889 DOI: 10.1074/jbc.m115.668871] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Indexed: 01/11/2023] Open
Abstract
Lineage specification in the hematopoietic system depends on the expression of lineage specific transcription factors. However, the role of hematopoietic cytokines in this process has been controversial and little is known about the intracellular signaling mechanisms by which cytokines instruct lineage choice. G-CSF and M-CSF are two lineage-specific cytokines that play a dominant role in granulopoiesis and monopoiesis, respectively. We show here that a G-CSFR mutant in which tyrosine 729 had been mutated to phenylalanine (Y729F) promoted monocyte rather than neutrophil development in myeloid precursors, which was associated with prolonged activation of Erk1/2 and augmented activation of downstream targets c-Fos and Egr1. Inhibition of Erk1/2 activation or knockdown of c-Fos or Egr1 largely rescued neutrophil development in cells expressing G-CSFR Y729F. We also show that M-CSF, but not G-CSF, stimulated strong and sustained activation of Erk1/2 in mouse lineage marker negative (Lin(-)) bone marrow cells. Significantly, inhibition of Erk1/2 signaling in these cells favored neutrophil over monocyte development in response to M-CSF. Thus, prolonged Erk1/2 activation resulted in monocyte development following G-CSF induction whereas inhibition of Erk1/2 signaling promoted neutrophil development at the expense of monocyte formation in response to M-CSF. These results reveal an important mechanism by which G-CSF and M-CSF instruct neutrophil versus monocyte lineage choice, i.e. differential activation of Erk1/2 pathway.
Collapse
Affiliation(s)
- Nan Hu
- From the Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| | - Yaling Qiu
- From the Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| | - Fan Dong
- From the Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| |
Collapse
|
25
|
Muto M, Baghdadi M, Maekawa R, Wada H, Seino KI. Myeloid molecular characteristics of human γδ T cells support their acquisition of tumor antigen-presenting capacity. Cancer Immunol Immunother 2015; 64:941-9. [PMID: 25904200 PMCID: PMC11028926 DOI: 10.1007/s00262-015-1700-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 04/15/2015] [Indexed: 10/23/2022]
Abstract
Human T cells expressing γδ T cell receptor have a potential to show antigen-presenting cell-like phenotype and function upon their activation. However, the mechanisms that underlie the alterations in human γδ T cells remain largely unclear. In this study, we have investigated the molecular characteristics of human γδ T cells related to their acquisition of antigen-presenting capacity in comparison with activated αβ T cells. We found that activated γδ but not αβ T cells upregulated cell surface expression of a scavenger receptor, CD36, which seemed to be mediated by signaling through mitogen-activated protein kinase and/or NF-κB pathways. Confocal microscopical analysis revealed that activated γδ T cells can phagocytose protein antigens. Activated γδ T cells could induce tumor antigen-specific CD8(+) T cells using both apoptotic and live tumor cells as antigen resources. Furthermore, we detected that C/EBPα, a critical transcription factor for the development of myeloid-lineage cells, is expressed much higher in γδ T cells than in αβ T cells. These results unveiled the molecular mechanisms for the elicitation of antigen-presenting functions in γδ T cells and would also help designing new approaches for γδ T cell-mediated human cancer immunotherapy.
Collapse
Affiliation(s)
- Masato Muto
- Institute for Genetic Medicine, Hokkaido University, Kita15 Nishi7, Sapporo, Hokkaido 060-0815 Japan
- Medinet Medical Institute, MEDINET Co., Ltd., Tokyo, Japan
| | - Muhammad Baghdadi
- Institute for Genetic Medicine, Hokkaido University, Kita15 Nishi7, Sapporo, Hokkaido 060-0815 Japan
| | - Ryuji Maekawa
- Medinet Medical Institute, MEDINET Co., Ltd., Tokyo, Japan
| | - Haruka Wada
- Institute for Genetic Medicine, Hokkaido University, Kita15 Nishi7, Sapporo, Hokkaido 060-0815 Japan
| | - Ken-ichiro Seino
- Institute for Genetic Medicine, Hokkaido University, Kita15 Nishi7, Sapporo, Hokkaido 060-0815 Japan
| |
Collapse
|
26
|
Aleem E, Arceci RJ. Targeting cell cycle regulators in hematologic malignancies. Front Cell Dev Biol 2015; 3:16. [PMID: 25914884 PMCID: PMC4390903 DOI: 10.3389/fcell.2015.00016] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 02/25/2015] [Indexed: 12/20/2022] Open
Abstract
Hematologic malignancies represent the fourth most frequently diagnosed cancer in economically developed countries. In hematologic malignancies normal hematopoiesis is interrupted by uncontrolled growth of a genetically altered stem or progenitor cell (HSPC) that maintains its ability of self-renewal. Cyclin-dependent kinases (CDKs) not only regulate the mammalian cell cycle, but also influence other vital cellular processes, such as stem cell renewal, differentiation, transcription, epigenetic regulation, apoptosis, and DNA repair. Chromosomal translocations, amplification, overexpression and altered CDK activities have been described in different types of human cancer, which have made them attractive targets for pharmacological inhibition. Mouse models deficient for one or more CDKs have significantly contributed to our current understanding of the physiological functions of CDKs, as well as their roles in human cancer. The present review focuses on selected cell cycle kinases with recent emerging key functions in hematopoiesis and in hematopoietic malignancies, such as CDK6 and its role in MLL-rearranged leukemia and acute lymphocytic leukemia, CDK1 and its regulator WEE-1 in acute myeloid leukemia (AML), and cyclin C/CDK8/CDK19 complexes in T-cell acute lymphocytic leukemia. The knowledge gained from gene knockout experiments in mice of these kinases is also summarized. An overview of compounds targeting these kinases, which are currently in clinical development in various solid tumors and hematopoietic malignances, is presented. These include the CDK4/CDK6 inhibitors (palbociclib, LEE011, LY2835219), pan-CDK inhibitors that target CDK1 (dinaciclib, flavopiridol, AT7519, TG02, P276-00, terampeprocol and RGB 286638) as well as the WEE-1 kinase inhibitor, MK-1775. The advantage of combination therapy of cell cycle inhibitors with conventional chemotherapeutic agents used in the treatment of AML, such as cytarabine, is discussed.
Collapse
Affiliation(s)
- Eiman Aleem
- Department of Child Health, The Ronald A. Matricaria Institute of Molecular Medicine at Phoenix Children's Hospital, University of Arizona College of Medicine-Phoenix Phoenix, AZ, USA ; Department of Zoology, Faculty of Science, Alexandria University Alexandria, Egypt
| | - Robert J Arceci
- Department of Child Health, The Ronald A. Matricaria Institute of Molecular Medicine at Phoenix Children's Hospital, University of Arizona College of Medicine-Phoenix Phoenix, AZ, USA
| |
Collapse
|
27
|
Gocek E, Studzinski GP. The Potential of Vitamin D-Regulated Intracellular Signaling Pathways as Targets for Myeloid Leukemia Therapy. J Clin Med 2015; 4:504-34. [PMID: 26239344 PMCID: PMC4470153 DOI: 10.3390/jcm4040504] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Revised: 01/06/2015] [Accepted: 03/06/2015] [Indexed: 02/06/2023] Open
Abstract
The current standard regimens for the treatment of acute myeloid leukemia (AML) are curative in less than half of patients; therefore, there is a great need for innovative new approaches to this problem. One approach is to target new treatments to the pathways that are instrumental to cell growth and survival with drugs that are less harmful to normal cells than to neoplastic cells. In this review, we focus on the MAPK family of signaling pathways and those that are known to, or potentially can, interact with MAPKs, such as PI3K/AKT/FOXO and JAK/STAT. We exemplify the recent studies in this field with specific relevance to vitamin D and its derivatives, since they have featured prominently in recent scientific literature as having anti-cancer properties. Since microRNAs also are known to be regulated by activated vitamin D, this is also briefly discussed here, as are the implications of the emerging acquisition of transcriptosome data and potentiation of the biological effects of vitamin D by other compounds. While there are ongoing clinical trials of various compounds that affect signaling pathways, more studies are needed to establish the clinical utility of vitamin D in the treatment of cancer.
Collapse
Affiliation(s)
- Elzbieta Gocek
- Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland.
| | - George P Studzinski
- Department of Pathology, New Jersey Medical School, Rutgers, The State University of New Jersey, 185 South Orange Ave., Newark, NJ 17101, USA.
| |
Collapse
|
28
|
Friedman AD. C/EBPα in normal and malignant myelopoiesis. Int J Hematol 2015; 101:330-41. [PMID: 25753223 DOI: 10.1007/s12185-015-1764-6] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 02/18/2015] [Accepted: 02/19/2015] [Indexed: 12/22/2022]
Abstract
CCAAT/enhancer binding protein α (C/EBPα) dimerizes via its leucine zipper (LZ) domain to bind DNA via its basic region and activate transcription via N-terminal trans-activation domains. The activity of C/EBPα is modulated by several serine/threonine kinases and via sumoylation, its gene is activated by RUNX1 and additional transcription factors, its mRNA stability is modified by miRNAs, and its mRNA is subject to translation control that affects AUG selection. In addition to inducing differentiation, C/EBPα inhibits cell cycle progression and apoptosis. Within hematopoiesis, C/EBPα levels increase as long-term stem cells progress to granulocyte-monocyte progenitors (GMP). Absence of C/EBPα prevents GMP formation, and higher levels are required for granulopoiesis compared to monopoiesis. C/EBPα interacts with AP-1 proteins to bind hybrid DNA elements during monopoiesis, and induction of Gfi-1, C/EBPε, KLF5, and miR-223 by C/EBPα enables granulopoiesis. The CEBPA ORF is mutated in approximately 10 % of acute myeloid leukemias (AML), leading to expression of N-terminally truncated C/EBPαp30 and C-terminal, in-frame C/EBPαLZ variants, which inhibit C/EBPα activities but also play additional roles during myeloid transformation. RUNX1 mutation, CEBPA promoter methylation, Trib1 or Trib2-mediated C/EBPαp42 degradation, and signaling pathways leading to C/EBPα serine 21 phosphorylation reduce C/EBPα expression or activity in additional AML cases.
Collapse
Affiliation(s)
- Alan D Friedman
- Division of Pediatric Oncology, Johns Hopkins University, Cancer Research Building I, Room 253, 1650 Orleans Street, Baltimore, MD, 21231, USA,
| |
Collapse
|
29
|
Imran M, Park JS, Lim IK. Stress-induced NF-κB activation differentiates promyelocytic leukemia cells to macrophages in response to all-trans-retinoic acid. Cell Signal 2015; 27:694-706. [DOI: 10.1016/j.cellsig.2014.11.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 11/15/2014] [Accepted: 11/21/2014] [Indexed: 12/24/2022]
|
30
|
Benet M, Guzmán C, Pisonero-Vaquero S, García-Mediavilla MV, Sánchez-Campos S, Martínez-Chantar ML, Donato MT, Castell JV, Jover R. Repression of the nuclear receptor small heterodimer partner by steatotic drugs and in advanced nonalcoholic fatty liver disease. Mol Pharmacol 2015; 87:582-94. [PMID: 25576488 DOI: 10.1124/mol.114.096313] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The small heterodimer partner (SHP) (NR0B2) is an atypical nuclear receptor that lacks a DNA-binding domain. It interacts with and inhibits many transcription factors, affecting key metabolic processes, including bile acid, cholesterol, fatty acid, and drug metabolism. Our aim was to determine the influence of steatotic drugs and nonalcoholic fatty liver disease (NAFLD) on SHP expression and investigate the potential mechanisms. SHP was found to be repressed by steatotic drugs (valproate, doxycycline, tetracycline, and cyclosporin A) in cultured hepatic cells and the livers of different animal models of NAFLD: iatrogenic (tetracycline-treated rats), genetic (glycine N-methyltransferase-deficient mice), and nutritional (mice fed a methionine- and choline-deficient diet). Among the different transcription factors investigated, CCAAT-enhancer-binding protein α (C/EBPα) showed the strongest dominant-repressive effect on SHP expression in HepG2 and human hepatocytes. Reporter assays revealed that the inhibitory effect of C/EBPα and steatotic drugs colocalize between -340 and -509 base pair of the SHP promoter, and mutation of a predicted C/EBPα response element at -473 base pair abolished SHP repression by both C/EBPα and drugs. Moreover, inhibition of major stress signaling pathways demonstrated that the mitogen-activated protein kinase kinase 1/2 pathway activates, while the phosphatidylinositol 3 kinase pathway represses SHP in a C/EBP-dependent manner. We conclude that SHP is downregulated by several steatotic drugs and in advanced NAFLD. These conditions can activate signals that target C/EBPα and consequently repress SHP, thus favoring the progression and severity of NAFLD.
Collapse
Affiliation(s)
- Marta Benet
- Experimental Hepatology Unit, IIS Hospital La Fe, Valencia (M.B., C.G., M.T.D., J.V.C., R.J.); CIBERehd, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona (M.B., M.V.G.-M., S.S.-C., M.L.M.-C., M.T.D., J.V.C., R.J.); Institute of Biomedicine, University of León, León (S.P.-V., M.V.G.-M., S.S.-C.); CIC bioGUNE, Technology Park of Bizkaia, Derio (M.L.M.-C.); and Department of Biochemistry and Molecular Biology, University of Valencia, Valencia, Spain (M.T.D., J.V.C., R.J.)
| | - Carla Guzmán
- Experimental Hepatology Unit, IIS Hospital La Fe, Valencia (M.B., C.G., M.T.D., J.V.C., R.J.); CIBERehd, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona (M.B., M.V.G.-M., S.S.-C., M.L.M.-C., M.T.D., J.V.C., R.J.); Institute of Biomedicine, University of León, León (S.P.-V., M.V.G.-M., S.S.-C.); CIC bioGUNE, Technology Park of Bizkaia, Derio (M.L.M.-C.); and Department of Biochemistry and Molecular Biology, University of Valencia, Valencia, Spain (M.T.D., J.V.C., R.J.)
| | - Sandra Pisonero-Vaquero
- Experimental Hepatology Unit, IIS Hospital La Fe, Valencia (M.B., C.G., M.T.D., J.V.C., R.J.); CIBERehd, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona (M.B., M.V.G.-M., S.S.-C., M.L.M.-C., M.T.D., J.V.C., R.J.); Institute of Biomedicine, University of León, León (S.P.-V., M.V.G.-M., S.S.-C.); CIC bioGUNE, Technology Park of Bizkaia, Derio (M.L.M.-C.); and Department of Biochemistry and Molecular Biology, University of Valencia, Valencia, Spain (M.T.D., J.V.C., R.J.)
| | - M Victoria García-Mediavilla
- Experimental Hepatology Unit, IIS Hospital La Fe, Valencia (M.B., C.G., M.T.D., J.V.C., R.J.); CIBERehd, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona (M.B., M.V.G.-M., S.S.-C., M.L.M.-C., M.T.D., J.V.C., R.J.); Institute of Biomedicine, University of León, León (S.P.-V., M.V.G.-M., S.S.-C.); CIC bioGUNE, Technology Park of Bizkaia, Derio (M.L.M.-C.); and Department of Biochemistry and Molecular Biology, University of Valencia, Valencia, Spain (M.T.D., J.V.C., R.J.)
| | - Sonia Sánchez-Campos
- Experimental Hepatology Unit, IIS Hospital La Fe, Valencia (M.B., C.G., M.T.D., J.V.C., R.J.); CIBERehd, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona (M.B., M.V.G.-M., S.S.-C., M.L.M.-C., M.T.D., J.V.C., R.J.); Institute of Biomedicine, University of León, León (S.P.-V., M.V.G.-M., S.S.-C.); CIC bioGUNE, Technology Park of Bizkaia, Derio (M.L.M.-C.); and Department of Biochemistry and Molecular Biology, University of Valencia, Valencia, Spain (M.T.D., J.V.C., R.J.)
| | - M Luz Martínez-Chantar
- Experimental Hepatology Unit, IIS Hospital La Fe, Valencia (M.B., C.G., M.T.D., J.V.C., R.J.); CIBERehd, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona (M.B., M.V.G.-M., S.S.-C., M.L.M.-C., M.T.D., J.V.C., R.J.); Institute of Biomedicine, University of León, León (S.P.-V., M.V.G.-M., S.S.-C.); CIC bioGUNE, Technology Park of Bizkaia, Derio (M.L.M.-C.); and Department of Biochemistry and Molecular Biology, University of Valencia, Valencia, Spain (M.T.D., J.V.C., R.J.)
| | - M Teresa Donato
- Experimental Hepatology Unit, IIS Hospital La Fe, Valencia (M.B., C.G., M.T.D., J.V.C., R.J.); CIBERehd, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona (M.B., M.V.G.-M., S.S.-C., M.L.M.-C., M.T.D., J.V.C., R.J.); Institute of Biomedicine, University of León, León (S.P.-V., M.V.G.-M., S.S.-C.); CIC bioGUNE, Technology Park of Bizkaia, Derio (M.L.M.-C.); and Department of Biochemistry and Molecular Biology, University of Valencia, Valencia, Spain (M.T.D., J.V.C., R.J.)
| | - José Vicente Castell
- Experimental Hepatology Unit, IIS Hospital La Fe, Valencia (M.B., C.G., M.T.D., J.V.C., R.J.); CIBERehd, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona (M.B., M.V.G.-M., S.S.-C., M.L.M.-C., M.T.D., J.V.C., R.J.); Institute of Biomedicine, University of León, León (S.P.-V., M.V.G.-M., S.S.-C.); CIC bioGUNE, Technology Park of Bizkaia, Derio (M.L.M.-C.); and Department of Biochemistry and Molecular Biology, University of Valencia, Valencia, Spain (M.T.D., J.V.C., R.J.)
| | - Ramiro Jover
- Experimental Hepatology Unit, IIS Hospital La Fe, Valencia (M.B., C.G., M.T.D., J.V.C., R.J.); CIBERehd, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona (M.B., M.V.G.-M., S.S.-C., M.L.M.-C., M.T.D., J.V.C., R.J.); Institute of Biomedicine, University of León, León (S.P.-V., M.V.G.-M., S.S.-C.); CIC bioGUNE, Technology Park of Bizkaia, Derio (M.L.M.-C.); and Department of Biochemistry and Molecular Biology, University of Valencia, Valencia, Spain (M.T.D., J.V.C., R.J.)
| |
Collapse
|
31
|
Abstract
The hematopoietic stem cell (HSC) is a unique cell positioned highest in the hematopoietic hierarchical system. The HSC has the ability to stay in quiescence, to self-renew, or to differentiate and generate all lineages of blood cells. The path to be actualized is influenced by signals that derive from the cell's microenvironment, which activate molecular pathways inside the cell. Signaling pathways are commonly organized through inducible protein-protein interactions, mediated by adaptor proteins that link activated receptors to cytoplasmic effectors. This review will focus on the signaling molecules and how they work in concert to determine the HSC's fate.
Collapse
Affiliation(s)
- Igal Louria-Hayon
- Department of Hematology, Rambam Health Care Campus, Haifa, Israel ; Department of Biotechnology, Hadassah Academic College, Jerusalem, Israel
| |
Collapse
|
32
|
Instruction of hematopoietic lineage choice by cytokine signaling. Exp Cell Res 2014; 329:207-13. [PMID: 25046868 DOI: 10.1016/j.yexcr.2014.07.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 07/08/2014] [Accepted: 07/10/2014] [Indexed: 01/06/2023]
Abstract
Hematopoiesis is the cumulative consequence of finely tuned signaling pathways activated through extrinsic factors, such as local niche signals and systemic hematopoietic cytokines. Whether extrinsic factors actively instruct the lineage choice of hematopoietic stem and progenitor cells or are only selectively allowing survival and proliferation of already intrinsically lineage-committed cells has been debated over decades. Recent results demonstrated that cytokines can instruct lineage choice. However, the precise function of individual cytokine-triggered signaling molecules in inducing cellular events like proliferation, lineage choice, and differentiation remains largely elusive. Signal transduction pathways activated by different cytokine receptors are highly overlapping, but support the production of distinct hematopoietic lineages. Cellular context, signaling dynamics, and the crosstalk of different signaling pathways determine the cellular response of a given extrinsic signal. New tools to manipulate and continuously quantify signaling events at the single cell level are therefore required to thoroughly interrogate how dynamic signaling networks yield a specific cellular response.
Collapse
|
33
|
Kang JW, Park YS, Kim MS, Lee DH, Bak Y, Ham SY, Song YS, Hong JT, Yoon DY. IL-32α down-regulates β2 integrin (CD18) expression by suppressing PU.1 expression in myeloid cells. Cell Signal 2014; 26:1514-22. [DOI: 10.1016/j.cellsig.2014.03.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 03/06/2014] [Accepted: 03/25/2014] [Indexed: 11/26/2022]
|
34
|
Bartels M, Calgarotto AK, Martens AC, Maso V, da Silva SL, Bierings MB, de Souza Queiroz ML, Coffer PJ. Differential effects of nitrostyrene derivatives on myelopoiesis involve regulation of C/EBPα and p38MAPK activity. PLoS One 2014; 9:e90586. [PMID: 24614182 PMCID: PMC3948686 DOI: 10.1371/journal.pone.0090586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Accepted: 02/03/2014] [Indexed: 11/18/2022] Open
Abstract
Bone marrow failure syndromes and MDS represent a heterogenous group of diseases, characterized by ineffective myelopoiesis, the risk of clonal evolution and a generally poor response to chemotherapy-based treatment regimen. Nitrostyrene derivatives have been studied as protein phosphatase inhibitors in various tumor models. Pharmacological studies have identified nitrostyrene as the structural core underlying a pro-apoptotic effect in tumor cells, yet their effects on normal cells, including those of the hematopoietic system, are largely unknown. In this study, utilizing umbilical cord blood-derived myeloid progenitor cells, patient-derived bone marrow cells, and a (BALB/c) mouse model; we investigated the effects of treatment with two nitrostyrene derivatives (NTS1 and NTS2) on myeloid development. We demonstrate that these compounds stimulate the expansion and differentiation of myeloid progenitors in vitro and improve myeloid reconstitution after chemotherapy-induced bone marrow depletion in vitro and in vivo. These effects were accompanied by increased C/EBPα expression and activity and inhibition of the p38MAPK signalling pathway. Together, our data suggest that nitrostyrenes improve myelopoiesis and represent potential new treatment strategies for patients suffering from bone marrow failure syndromes, hypocellular myelodysplastic syndrome and chemotherapy-induced aplasia.
Collapse
Affiliation(s)
- Marije Bartels
- Department of Cell Biology, University Medical Center Utrecht, Utrecht, The Netherlands; Division of Pediatrics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Andrana K Calgarotto
- Departamento de Farmacologica, Universidade Estadual de Campinas, Campinas/SP, Brazil
| | - Anton C Martens
- Department of Cell Biology, University Medical Center Utrecht, Utrecht, The Netherlands; Department of Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Victor Maso
- Departamento de Farmacologica, Universidade Estadual de Campinas, Campinas/SP, Brazil
| | - Saulo L da Silva
- Departamento de Química, Universidade Federal de São João Del-Rei, Ouro Branco/MG, Brazil
| | - Marc B Bierings
- Division of Pediatrics, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Paul J Coffer
- Department of Cell Biology, University Medical Center Utrecht, Utrecht, The Netherlands; Division of Pediatrics, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
35
|
Young DJ, Stoddart A, Nakitandwe J, Chen SC, Qian Z, Downing JR, Le Beau MM. Knockdown of Hnrnpa0, a del(5q) gene, alters myeloid cell fate in murine cells through regulation of AU-rich transcripts. Haematologica 2014; 99:1032-40. [PMID: 24532040 DOI: 10.3324/haematol.2013.098657] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The control of mRNA stability plays a central role in orchestrating gene-regulatory networks in hematopoietic cell growth, differentiation and tumorigenesis. HNRNPA0, which encodes an RNA-binding protein shown to regulate transcript stability via binding to the AU-rich elements of mRNAs, is located within the commonly deleted segment of 5q31.2 in myeloid neoplasms with a del(5q), and is expressed at haploinsufficient levels in these patients. We show that HNRNPA0 is normally highly expressed in hematopoietic stem cells and exhibits dynamic changes in expression during the course of differentiation. To model HNRNPA0 haploinsufficiency, we used RNAi interference in primary murine cells and an experimental cell system, and found that reduced Hnrnpa0 expression leads to a shift from monocytic towards granulocytic differentiation. Microarray-based global expression profiling revealed that Hnrnpa0 knockdown disproportionally impacts AU-rich containing transcripts and alters expression of myeloid specification genes. In therapy-related myeloid neoplasms with a del(5q), AU-rich containing mRNAs are enriched in transcripts that encode proteins associated with increased growth and proliferation. Our findings implicate haploinsufficiency of HNRNPA0 as one of the key initiating mutations in the pathogenesis of myeloid neoplasms with a del(5q), and suggest that therapies that target AU-rich elements warrant consideration in efforts to develop new mechanism-based treatment strategies.
Collapse
Affiliation(s)
- David J Young
- Department of Pediatrics, Division of Oncology, Johns Hopkins University, Baltimora, MD
| | - Angela Stoddart
- Department of Medicine and the Comprehensive Cancer Center, University of Chicago, IL
| | - Joy Nakitandwe
- St. Jude Children's Research Hospital, Memphis, Tennessee
| | | | - Zhijian Qian
- University of Illinois Cancer Center, Chicago, IL, USA
| | | | - Michelle M Le Beau
- Department of Medicine and the Comprehensive Cancer Center, University of Chicago, IL
| |
Collapse
|
36
|
Abstract
Tribbles are members of the pseudokinase family of proteins, with no associated kinase activity detectable to date. As tribbles appear not to function as kinases, there has been debate surrounding their functional classification. Tribbles have been proposed to function as adaptor molecules facilitating degradation of their target proteins. Tribbles have also been proposed to mediate signalling changes to MAPK (mitogen-activated protein kinase) cascades and also to function as decoy kinases interfering with the activity of known kinases. The present review discusses the functionally divergent roles of tribbles as molecular adaptors mediating degradation, changes to signalling cascades and action as decoy kinases.
Collapse
|
37
|
Shimokawa T, Nunomura S, Fujisawa D, Ra C. Identification of the C/EBPα C-terminal tail residues involved in the protein interaction with GABP and their potency in myeloid differentiation of K562 cells. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2013; 1829:1207-17. [PMID: 24076158 DOI: 10.1016/j.bbagrm.2013.09.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Revised: 09/18/2013] [Accepted: 09/19/2013] [Indexed: 11/17/2022]
Abstract
The CCAAT/enhancer-binding protein α (C/EBPα) is the member of a family of related basic leucine zipper (bZIP) transcription factors and is critical for granulopoiesis. We previously demonstrated that C/EBPα interacts with the ETS domain of widely expressed GABPα, which leads to cooperative transcriptional activation of the myeloid-specific promoter for human FCAR encoding the Fc receptor for IgA (FcαR, CD89) in part by facilitating recruitment of C/EBPα to the promoter. The C/EBPα molecule contains transactivation domains (TADs) at its N-terminus and a DNA-binding and dimerization bZIP structure at its C-terminus. We demonstrate here that GABPα interacts with the last 18 residues of the C/EBPα C-terminus beyond the bZIP DNA-binding and dimerizing region. Deletion of this C-terminus resulted in loss of GABPα interaction but not affecting its DNA binding ability, indicating that it is not required for homodimer formation. Moreover, the C-terminus confers the ability to functionally synergize with GABP on a heterologous TAD when fused to the C-terminus of the VP16 TAD. We identified a three-amino acid stretch (amino acids 341-343) that is important for both functional and protein interactions with GABP. Ectopic expression in K562 cells of C/EBPα mutant incapable of interacting with GABPα does not induce expression of granulocytic differentiation markers including CD15, CD11b, GCSF-R and C/EBPε, and does not inhibit proliferation, whereas wild type does. These results demonstrate the functional importance of the C/EBPα C-terminus beyond the bZIP DNA-binding and dimerization region, which may mediate cooperative activation by C/EBPα and GABP of myeloid-specific genes involved in C/EBPα-dependent granulopoiesis.
Collapse
Affiliation(s)
- Toshibumi Shimokawa
- Division of Molecular Cell Immunology and Allergology, Department of Biomedical Sciences, Nihon University School of Medicine, Itabashi-ku, Tokyo 173-8610, Japan.
| | | | | | | |
Collapse
|
38
|
Kang JW, Park YS, Kim MS, Lee DH, Bak Y, Ham SY, Park SH, Hong JT, Yoon DY. Interleukin (IL)-32β-mediated CCAAT/enhancer-binding protein α (C/EBPα) phosphorylation by protein kinase Cδ (PKCδ) abrogates the inhibitory effect of C/EBPα on IL-10 production. J Biol Chem 2013; 288:23650-8. [PMID: 23814099 DOI: 10.1074/jbc.m113.465575] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We previously reported that IL-32β promotes IL-10 production in myeloid cells. However, the underlying mechanism remains elusive. In this study, we demonstrated that IL-32β abrogated the inhibitory effect of CCAAT/enhancer-binding protein α (C/EBPα) on IL-10 expression in U937 cells. We observed that the phosphorylation of C/EBPα Ser-21 was inhibited by a PKCδ-specific inhibitor, rottlerin, or IL-32β knockdown by siRNA and that IL-32β shifted to the membrane from the cytosol upon phorbol 12-myristate 13-acetate treatment. We revealed that IL-32β suppressed the binding of C/EBPα to IL-10 promoter by using ChIP assay. These data suggest that PKCδ and IL-32β may modulate the effect of C/EBPα on IL-10 expression. We next demonstrated by immunoprecipitation that IL-32β interacted with PKCδ and C/EBPα, thereby mediating C/EBPα Ser-21 phosphorylation by PKCδ. We showed that IL-32β suppressed the inhibitory effect of C/EBPα on IL-10 promoter activity. However, the IL-10 promoter activity was reduced to the basal level by rottlerin treatment. When C/EBPα serine 21 was mutated to glycine (S21G), the inhibitory effect of C/EBPα S21G on IL-10 promoter activity was not modulated by IL-32β. Taken together, our results show that IL-32β-mediated C/EBPα Ser-21 phosphorylation by PKCδ suppressed C/EBPα binding to IL-10 promoter, which promoted IL-10 production in U937 cells.
Collapse
Affiliation(s)
- Jeong-Woo Kang
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Steinmann S, Coulibaly A, Ohnheiser J, Jakobs A, Klempnauer KH. Interaction and cooperation of the CCAAT-box enhancer-binding protein β (C/EBPβ) with the homeodomain-interacting protein kinase 2 (Hipk2). J Biol Chem 2013; 288:22257-69. [PMID: 23782693 DOI: 10.1074/jbc.m113.487769] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
CCAAT box/enhancer-binding protein β (C/EBPβ) is a bZip transcription factor that plays crucial roles in important cellular processes such as differentiation and proliferation of specific cell types. Previously, we showed that C/EBPβ cooperates with the coactivator p300 through a novel mechanism that involves the C/EBPβ-induced phosphorylation of multiple sites in the carboxyl-terminal domain of p300 by protein kinase Hipk2. We have now examined the interaction and cooperation of C/EBPβ, p300, and Hipk2 in more detail. We show that Hipk2 and C/EBPβ are direct physical binding partners whose interaction is mediated by sequences located in the amino-terminal and central domains of Hipk2 and the amino-terminal part of C/EBPβ. In addition to phosphorylating p300 recruited to C/EBPβ, Hipk2 also phosphorylates C/EBPβ at sites that have previously been shown to plays key roles in the regulation of C/EBPβ activity. Silencing of Hipk2 expression disrupts adipocyte differentiation of 3T3-L1 cells, a physiological C/EBPβ-dependent differentiation process indicating that the cooperation of C/EBPβ and Hipk2 is functionally relevant. Finally, we demonstrate that C/EBPα, a related C/EBP family member whose amino-terminal sequences differ significantly from that of C/EBPβ, is unable to interact and cooperate with Hipk2. Instead, our data suggest that C/EBPα cooperates with the protein kinase Jnk to induce phosphorylation of p300. Overall, our data identify Hipk2 as a novel regulator of C/EBPβ and implicate different protein kinases in the cooperation of p300 with C/EBPβ and C/EBPα.
Collapse
Affiliation(s)
- Simone Steinmann
- Institut für Biochemie, Westfälische-Wilhelms-Universität Münster, Wilhelm-Klemm-Strasse 2, D-48149 Münster, Germany
| | | | | | | | | |
Collapse
|
40
|
E6AP, an E3 ubiquitin ligase negatively regulates granulopoiesis by targeting transcription factor C/EBPα for ubiquitin-mediated proteasome degradation. Cell Death Dis 2013; 4:e590. [PMID: 23598402 PMCID: PMC3641343 DOI: 10.1038/cddis.2013.120] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
CCAAT/enhancer-binding protein alpha (C/EBPα) is an important transcription factor involved in granulocytic differentiation. Here, for the first time we demonstrate that E6-associated protein (E6AP), an E3 ubiquitin ligase targets C/EBPα for ubiquitin-mediated proteasome degradation and thereby negatively modulates its functions. Wild-type E6AP promotes ubiquitin dependent proteasome degradation of C/EBPα, while catalytically inactive E6-associated protein having cysteine replaced with alanine at amino-acid position 843 (E6AP-C843A) rather stabilizes it. Further, these two proteins physically associate both in non-myeloid (overexpressed human embryonic kidney epithelium) and myeloid cells. We show that E6AP-mediated degradation of C/EBPα protein expression curtails its transactivation potential on its target genes. Noticeably, E6AP degrades both wild-type 42 kDa CCAAT-enhancer-binding protein alpha (p42C/EBPα) and mutant isoform 30 kDa CCAAT-enhancer-binding protein alpha (p30C/EBPα), this may explain perturbed p42C/EBPα/p30C/EBPα ratio often observed in acute myeloid leukemia (AML). We show that overexpression of catalytically inactive E6AP-C843A in C/EBPα inducible K562- p42C/EBPα-estrogen receptor (ER) cells inhibits β-estradiol (E2)-induced C/EBPα degradation leading to enhanced granulocytic differentiation. This enhanced granulocytic differentiation upon E2-induced activation of C/EBPα in C/EBPα stably transfected cells (β-estradiol inducible K562 cells stably expressing p42C/EBPα-ER (K562-C/EBPα-p42-ER)) was further substantiated by siE6AP-mediated knockdown of E6AP in both K562-C/EBPα-p42-ER and 32dcl3 (32D clone 3, a cell line widely used model for in vitro study of hematopoietic cell proliferation, differentiation, and apoptosis) cells. Taken together, our data suggest that E6AP targeted C/EBPα protein degradation may provide a possible explanation for both loss of expression and/or functional inactivation of C/EBPα often experienced in myeloid leukemia.
Collapse
|
41
|
PKCδ enhances C/EBPα degradation via inducing its phosphorylation and cytoplasmic translocation. Biochem Biophys Res Commun 2013; 433:220-5. [DOI: 10.1016/j.bbrc.2013.02.087] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Accepted: 02/26/2013] [Indexed: 11/24/2022]
|
42
|
Inhibition of the receptor tyrosine kinase Axl impedes activation of the FLT3 internal tandem duplication in human acute myeloid leukemia: implications for Axl as a potential therapeutic target. Blood 2013; 121:2064-73. [PMID: 23321254 DOI: 10.1182/blood-2012-07-444018] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Approximately 20% to 25% of patients with acute myeloid leukemia (AML) have a constitutively activated FLT3-internal tandem duplication (FLT3-ITD), and these patients exhibit a poor prognosis. Here, we report that Axl, a receptor tyrosine kinase (RTK) overexpressed and constitutively active in human AML, targets the RTK FLT3 in FLT3-ITD(+) AML. Abrogation of Axl activation by soluble Axl chimeric protein (Axl-Fc) or small interfering RNA (siRNA) diminishes constitutive FLT3 phosphorylation in FLT3-ITD(+) AML. In addition, inhibition of Axl activation by Axl-Fc interferes with the physical interaction between Axl and FLT3. We found that Axl-Fc, a pharmacologic Axl inhibitor, or siRNA targeting Axl inhibits cell growth, induces cell-cycle arrest and apoptosis, and relieves a block in myeloid differentiation of FLT3-ITD(+) AML in vitro. Axl-Fc also suppresses the growth of human FLT3-ITD(+) AML in vivo. Collectively, our data suggest that Axl contributes to the pathogenesis of FLT3-ITD(+) AML through, at least in part, positive regulation of constitutive FLT3 activation. This also suggests that Axl should be pursued as a potential target for the treatment of FLT3-ITD(+) AML.
Collapse
|
43
|
Abstract
Macrophages not only are prominent effector cells of the immune system that are critical in inflammation and innate immune responses but also fulfill important functions in tissue homeostasis. Transcription factors can define macrophage identity and control their numbers and functions through the induction and maintenance of specific transcriptional programs. Here, we review the mechanisms employed by lineage-specific transcription factors to shape macrophage identity during the development from hematopoietic stem and progenitor cells. We also present current insight into how specific transcription factors control macrophage numbers, by regulating coordinated proliferation and differentiation of myeloid progenitor cells and self-renewal of mature macrophages. We finally discuss how functional specialization of mature macrophages in response to environmental stimuli can be induced through synergistic activity of lineage- and stimulus-specific transcription factors that plug into preexisting transcriptional programs. Understanding the mechanisms that define macrophage identity, numbers, and functions will provide important insights into the differential properties of macrophage populations under various physiological and pathological conditions.
Collapse
Affiliation(s)
- Kaaweh Molawi
- Centre d'Immunologie de Marseille-Luminy, UM2 Aix-Marseille Université, Marseille cedex 9; INSERM, Marseille, France; CNRS, Marseille, France; Max-Delbrück-Centrum für Molekulare Medizin, Berlin, Germany
| | | |
Collapse
|
44
|
Staser K, Park SJ, Rhodes SD, Zeng Y, He YZ, Shew MA, Gehlhausen JR, Cerabona D, Menon K, Chen S, Sun Z, Yuan J, Ingram DA, Nalepa G, Yang FC, Clapp DW. Normal hematopoiesis and neurofibromin-deficient myeloproliferative disease require Erk. J Clin Invest 2012; 123:329-34. [PMID: 23221339 DOI: 10.1172/jci66167] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Accepted: 10/18/2012] [Indexed: 11/17/2022] Open
Abstract
Neurofibromatosis type 1 (NF1) predisposes individuals to the development of juvenile myelomonocytic leukemia (JMML), a fatal myeloproliferative disease (MPD). In genetically engineered murine models, nullizygosity of Nf1, a tumor suppressor gene that encodes a Ras-GTPase-activating protein, results in hyperactivity of Raf/Mek/Erk in hematopoietic stem and progenitor cells (HSPCs). Activated Erk1/2 phosphorylate kinases and transcription factors with myriad mitogenic roles in diverse cell types. However, genetic studies examining Erk1/2's differential and/or combined control of normal and Nf1-deficient myelopoiesis are lacking. Moreover, prior studies relying on chemical Mek/Erk inhibitors have reached conflicting conclusions in normal and Nf1-deficient mice. Here, we show that while single Erk1 or Erk2 disruption did not grossly compromise myelopoiesis, dual Erk1/2 disruption rapidly ablated granulocyte and monocyte production in vivo, diminished progenitor cell number, and prevented HSPC proliferation in vitro. Genetic disruption of Erk1/2 in the context of Nf1 nullizygosity (Mx1Cre(+)Nf1(flox/flox)Erk1(-/-)Erk2(flox/flox)) fully protects against the development of MPD. Collectively, we identified a fundamental requirement for Erk1/2 signaling in normal and Nf1-deficient hematopoiesis, elucidating a critical hematopoietic function for Erk1/2 while genetically validating highly selective Mek/Erk inhibitors in a leukemia that is otherwise resistant to traditional therapy.
Collapse
Affiliation(s)
- Karl Staser
- Herman Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
A novel role of a lipid species, sphingosine-1-phosphate, in epithelial innate immunity. Mol Cell Biol 2012; 33:752-62. [PMID: 23230267 DOI: 10.1128/mcb.01103-12] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
A variety of external perturbations can induce endoplasmic reticulum (ER) stress, followed by stimulation of epithelial cells to produce an innate immune element, the cathelicidin antimicrobial peptide (CAMP). ER stress also increases production of the proapoptotic lipid ceramide and its antiapoptotic metabolite, sphingosine-1-phosphate (S1P). We demonstrate here that S1P mediates ER stress-induced CAMP generation. Cellular ceramide and S1P levels rose in parallel with CAMP levels following addition of either exogenous cell-permeating ceramide (C2Cer), which increases S1P production, or thapsigargin (an ER stressor), applied to cultured human skin keratinocytes or topically to mouse skin. Knockdown of S1P lyase, which catabolizes S1P, enhanced ER stress-induced CAMP production in cultured cells and mouse skin. These and additional inhibitor studies show that S1P is responsible for ER stress-induced upregulation of CAMP expression. Increased CAMP expression is likely mediated via S1P-dependent NF-κB-C/EBPα activation. Finally, lysates of both ER-stressed and S1P-stimulated cells blocked growth of virulent Staphylococcus aureus in vitro, and topical C2Cer and LL-37 inhibited invasion of Staphylococcus aureus into murine skin. These studies suggest that S1P generation resulting in increased CAMP production comprises a novel regulatory mechanism of epithelial innate immune responses to external perturbations, pointing to a new therapeutic approach to enhance antimicrobial defense.
Collapse
|
46
|
Mononuclear phagocyte miRNome analysis identifies miR-142 as critical regulator of murine dendritic cell homeostasis. Blood 2012; 121:1016-27. [PMID: 23212522 DOI: 10.1182/blood-2012-07-445999] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The mononuclear phagocyte system comprises cells as diverse as monocytes, macrophages, and dendritic cells (DCs), which collectively play key roles in innate immune responses and the triggering of adaptive immunity. Recent studies have highlighted the role of growth and transcription factors in defining developmental pathways and lineage relations within this cellular compartment. However, contributions of miRNAs to the development of mononuclear phagocytes remain largely unknown. In the present study, we report a comprehensive map of miRNA expression profiles for distinct myeloid populations, including BM-resident progenitors, monocytes, and mature splenic DCs. Each of the analyzed cell populations displayed a distinctive miRNA profile, suggesting a role for miRNAs in defining myeloid cell identities. Focusing on DC development, we found miR-142 to be highly expressed in classic FLT3-L–dependent CD4+ DCs, whereas reduced expression was observed in closely related CD8α+ or CD4- CD8α- DCs. Moreover, mice deficient for miR-142 displayed an impairment of CD4+ DC homeostasis both in vitro and in vivo. Furthermore, loss of miR-142–dependent CD4+ DCs was accompanied by a severe and specific defect in the priming of CD4+ T cells. The results of our study establish a novel role for miRNAs in myeloid cell specification and define miR-142 as a pivotal genetic component in the maintenance of CD4+ DCs.
Collapse
|
47
|
Alberich-Jordà M, Wouters B, Balastik M, Shapiro-Koss C, Zhang H, Di Ruscio A, DiRuscio A, Radomska HS, Ebralidze AK, Amabile G, Ye M, Zhang J, Lowers I, Avellino R, Melnick A, Figueroa ME, Valk PJM, Delwel R, Tenen DG. C/EBPγ deregulation results in differentiation arrest in acute myeloid leukemia. J Clin Invest 2012; 122:4490-504. [PMID: 23160200 DOI: 10.1172/jci65102] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 09/20/2012] [Indexed: 11/17/2022] Open
Abstract
C/EBPs are a family of transcription factors that regulate growth control and differentiation of various tissues. We found that C/EBPγ is highly upregulated in a subset of acute myeloid leukemia (AML) samples characterized by C/EBPα hypermethylation/silencing. Similarly, C/EBPγ was upregulated in murine hematopoietic stem/progenitor cells lacking C/EBPα, as C/EBPα mediates C/EBPγ suppression. Studies in myeloid cells demonstrated that CEBPG overexpression blocked neutrophilic differentiation. Further, downregulation of Cebpg in murine Cebpa-deficient stem/progenitor cells or in human CEBPA-silenced AML samples restored granulocytic differentiation. In addition, treatment of these leukemias with demethylating agents restored the C/EBPα-C/EBPγ balance and upregulated the expression of myeloid differentiation markers. Our results indicate that C/EBPγ mediates the myeloid differentiation arrest induced by C/EBPα deficiency and that targeting the C/EBPα-C/EBPγ axis rescues neutrophilic differentiation in this unique subset of AMLs.
Collapse
|
48
|
Lenalidomide-mediated enhanced translation of C/EBPα-p30 protein up-regulates expression of the antileukemic microRNA-181a in acute myeloid leukemia. Blood 2012; 121:159-69. [PMID: 23100311 DOI: 10.1182/blood-2012-05-428573] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Recently, we showed that increased miR-181a expression was associated with improved outcomes in cytogenetically normal acute myeloid leukemia (CN-AML). Interestingly, miR-181a expression was increased in CN-AML patients harboring CEBPA mutations, which are usually biallelic and associate with better prognosis. CEBPA encodes the C/EBPα transcription factor. We demonstrate here that the presence of N-terminal CEBPA mutations and miR-181a expression are linked. Indeed, the truncated C/EBPα-p30 isoform, which is produced from the N-terminal mutant CEBPA gene or from the differential translation of wild-type CEBPA mRNA and is commonly believed to have no transactivation activity, binds to the miR-181a-1 promoter and up-regulates the microRNA expression. Furthermore, we show that lenalidomide, a drug approved for myelodysplastic syndromes and multiple myeloma, enhances translation of the C/EBPα-p30 isoform, resulting in higher miR-181a levels. In xenograft mouse models, ectopic miR-181a expression inhibits tumor growth. Similarly, lenalidomide exhibits antitumorigenic activity paralleled by increased miR-181a expression. This regulatory pathway may explain an increased sensitivity to apoptosis-inducing chemotherapy in subsets of AML patients. Altogether, our data provide a potential explanation for the improved clinical outcomes observed in CEBPA-mutated CN-AML patients, and suggest that lenalidomide treatment enhancing the C/EBPα-p30 protein levels and in turn miR-181a may sensitize AML blasts to chemotherapy.
Collapse
|
49
|
Fragliasso V, Chiodo Y, Ferrari-Amorotti G, Soliera AR, Manzotti G, Cattelani S, Candini O, Grisendi G, Vergalli J, Mariani SA, Guerzoni C, Calabretta B. Phosphorylation of serine 21 modulates the proliferation inhibitory more than the differentiation inducing effects of C/EBPα in K562 cells. J Cell Biochem 2012; 113:1704-13. [PMID: 22212957 DOI: 10.1002/jcb.24040] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The CCAAT/enhancer binding protein α (C/EBPα) is a transcription factor required for differentiation of myeloid progenitors. In acute myeloid leukemia (AML) cells expressing the constitutively active FLT3-ITD receptor tyrosine kinase, MAP kinase-dependent phosphorylation of serine 21 (S21) inhibits the ability of C/EBPα to induce granulocytic differentiation. To assess whether this post-translational modification also modulates the activity of C/EBPα in BCR/ABL-expressing cells, we tested the biological effects of wild-type and mutant C/EBPα mimicking phosphorylated or non-phosphorylatable serine 21 (S21D and S21A, respectively) in K562 cells ectopically expressing tamoxifen-regulated C/EBPα-ER chimeric proteins. We show here that S21D C/EBPα-ER induced terminal granulocytic differentiation of K562 cells almost as well as wild-type C/EBPα-ER, while S21A C/EBPα-ER was less efficient. Furthermore, wild-type C/EBPα suppressed the proliferation and colony formation of K562 cells vigorously, while S21D and S21A C/EBPα mutants had more modest anti-proliferative effects. Both mutants were less effective than wild-type C/EBPα in suppressing endogenous E2F-dependent transactivation and bound less E2F-2 and/or E2F-3 proteins in anti-C/EBPα immunoprecipitates. Together, these findings suggest that mutation of S21 more than its phosphorylation inhibits the anti-proliferative effects of C/EBPα due to reduced interaction with or impaired regulation of the activity of E2F proteins. By contrast, phosphorylation of serine 21 appears to have a modest role in modulating the differentiation-inducing effects of C/EBPα in K562 cells.
Collapse
Affiliation(s)
- Valentina Fragliasso
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Kapoor I, Pal P, Lochab S, Kanaujiya JK, Trivedi AK. Proteomics approaches for myeloid leukemia drug discovery. Expert Opin Drug Discov 2012; 7:1165-75. [DOI: 10.1517/17460441.2012.724055] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|