51
|
Abstract
UNLABELLED Parkinson's disease (PD) is characterized by the progressive loss of select neuronal populations, but the prodeath genes mediating the neurodegenerative processes remain to be fully elucidated. Trib3 (tribbles pseudokinase 3) is a stress-induced gene with proapoptotic activity that was previously described as highly activated at the transcriptional level in a 6-hydroxydopamine (6-OHDA) cellular model of PD. Here, we report that Trib3 immunostaining is elevated in dopaminergic neurons of the substantia nigra pars compacta (SNpc) of human PD patients. Trib3 protein is also upregulated in cellular models of PD, including neuronal PC12 cells and rat dopaminergic ventral midbrain neurons treated with 6-OHDA, 1-methyl-4-phenylpyridinium (MPP+), or α-synuclein fibrils (αSYN). In the toxin models, Trib3 induction is substantially mediated by the transcription factors CHOP and ATF4. Trib3 overexpression is sufficient to promote neuronal death; conversely, Trib3 knockdown protects neuronal PC12 cells as well as ventral midbrain dopaminergic neurons from 6-OHDA, MPP+, or αSYN. Mechanism studies revealed that Trib3 physically interacts with Parkin, a prosurvival protein whose loss of function is associated with PD. Elevated Trib3 reduces Parkin expression in cultured cells; and in the SNpc of PD patients, Parkin levels are reduced in a subset of dopaminergic neurons expressing high levels of Trib3. Loss of Parkin at least partially mediates the prodeath actions of Trib3 in that Parkin knockdown in cellular PD models abolishes the protective effect of Trib3 downregulation. Together, these findings identify Trib3 and its regulatory pathways as potential targets to suppress the progression of neuron death and degeneration in PD. SIGNIFICANCE STATEMENT Parkinson's disease (PD) is the most common neurodegenerative movement disorder. Current treatments ameliorate symptoms, but not the underlying neuronal death. Understanding the core neurodegenerative processes in PD is a prerequisite for identifying new therapeutic targets and, ultimately, curing this disease. Here, we describe a novel pathway involving the proapoptotic protein Trib3 in neuronal death associated with PD. These findings are supported by data from multiple cellular models of PD and by immunostaining of postmortem PD brains. Upstream, Trib3 is induced by the transcription factors ATF4 and CHOP; and downstream, Trib3 interferes with the PD-associated prosurvival protein Parkin to mediate death. These findings establish this new pathway as a potential and promising therapeutic target for treatment of PD.
Collapse
|
52
|
Wanquet A, Prebet T, Berthon C, Sebert M, Roux C, Kulasekararaj A, Micol JB, Esterni B, Itzykson R, Thepot S, Recher C, Delaunay J, Dreyfus F, Mufti G, Fenaux P, Vey N. Azacitidine treatment for patients with myelodysplastic syndrome and acute myeloid leukemia with chromosome 3q abnormalities. Am J Hematol 2015; 90:859-63. [PMID: 26113240 DOI: 10.1002/ajh.24099] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 06/22/2015] [Indexed: 12/26/2022]
Abstract
Acute Myeloid Leukemia (AML) and myelodysplasia (MDS) with chromosome 3q abnormalities have a dismal outcome either untreated or with conventional treatments. Azacitidine (AZA) is now considered as the standard of care in high-risk MDS and oligoblastic AML patients. The objective of this study was to evaluate the impact of azacitine treatment in this cytogenetic subgroup. We report here a multicentre retrospective study of 157 patients treated with AZA for AML/MDS with chromosome 3q abnormalities and 27 patients with isolated EVI-1 overexpression. Median age was 65 years, 40 patients (25%) had inv(3)(q21q26.2) or t(3;3)(q21;q26.2), 36 patients (23%) had other balanced 3q26 rearrangements, 8 patients (5%) had balanced 3q21 rearrangements and 73 patients (46%) had other 3q abnormalities. The overall response rate was 50% (29% CR). Median overall survival was 10.6 months. By multivariate analysis, patients with lower bone marrow blast counts, higher platelet counts, non-complex cytogenetics, and absence of prior treatment with intensive chemotherapy had a better outcome. 27 patients were allo-transplanted and achieved a 21-month median OS. Balanced 3q21 translocations were associated with a better response rate and overall survival. Outcome of patients with isolated EVI-1 overexpression was comparable to that of patients with chromosome 3q lesions. Thus, AML/MDS patients with 3q abnormalities appear to be a heterogeneous group in their response to AZA, and AZA may represent a suitable option in particular as a bridge to allogeneic transplantation.
Collapse
Affiliation(s)
- Anne Wanquet
- Hematology Department; Institut Paoli-Calmettes; Marseille France
| | - Thomas Prebet
- Hematology Department; Institut Paoli-Calmettes; Marseille France
| | - Céline Berthon
- Hematology Department; Centre Hospitalo Universitaire De Lille; Lille France
| | - Marie Sebert
- Hematology Department; Assistance Publique-Hôpitaux De Paris (APHP), Hopital Saint Louis, Paris and Paris 7 University; France
| | - Clémence Roux
- Hematology Department; Centre Hospitalo Universitaire De Nice; Nice France
| | | | | | - Benjamin Esterni
- Biostatistics Department; Institut paoli-Calmettes; Marseille France
| | - Raphael Itzykson
- Hematology Department; Assistance Publique-Hôpitaux De Paris (APHP), Hopital Saint Louis, Paris and Paris 7 University; France
| | - Sylvain Thepot
- Hematology Department; Hopital Avicenne (APHP) and Paris 13 University; Bobigny France
| | - Christian Recher
- Hematology Department; Centre Hospitalo Universitaire De Toulouse; Toulouse France
| | - Jacques Delaunay
- Hematology Department; Centre Hospitalo Universitaire De Nantes; Nantes France
| | - François Dreyfus
- Hematology Department; Hopital Cochin (APHP) and Paris 5 University; Paris France
| | - Ghulam Mufti
- Hematology Department; King's College; London United Kingdom
| | - Pierre Fenaux
- Hematology Department; Assistance Publique-Hôpitaux De Paris (APHP), Hopital Saint Louis, Paris and Paris 7 University; France
| | - Norbert Vey
- Hematology Department; Institut Paoli-Calmettes; Marseille France
- Aix Marseille University; Marseille France
| |
Collapse
|
53
|
Lentiviral vector system for coordinated constitutive and drug controlled tetracycline-regulated gene co-expression. Biomaterials 2015; 63:189-201. [DOI: 10.1016/j.biomaterials.2015.06.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 06/10/2015] [Accepted: 06/12/2015] [Indexed: 12/15/2022]
|
54
|
Steinmetz B, Hackl H, Slabáková E, Schwarzinger I, Smějová M, Spittler A, Arbesu I, Shehata M, Souček K, Wieser R. The oncogene EVI1 enhances transcriptional and biological responses of human myeloid cells to all-trans retinoic acid. Cell Cycle 2015; 13:2931-43. [PMID: 25486480 PMCID: PMC4613657 DOI: 10.4161/15384101.2014.946869] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The product of the ecotropic virus integration site 1 (EVI1) gene, whose overexpression is associated with a poor prognosis in myeloid leukemias and some epithelial tumors, regulates gene transcription both through direct DNA binding and through modulation of the activity of other sequence specific transcription factors. Previous results from our laboratory have shown that EVI1 influenced transcription regulation in response to the myeloid differentiation inducing agent, all-trans retinoic acid (ATRA), in a dual manner: it enhanced ATRA induced transcription of the RARβ gene, but repressed the ATRA induction of the EVI1 gene itself. In the present study, we asked whether EVI1 would modulate the ATRA regulation of a larger number of genes, as well as biological responses to this agent, in human myeloid cells. U937 and HL-60 cells ectopically expressing EVI1 through retroviral transduction were subjected to microarray based gene expression analysis, and to assays measuring cellular proliferation, differentiation, and apoptosis. These experiments showed that EVI1 modulated the ATRA response of several dozens of genes, and in fact reinforced it in the vast majority of cases. A particularly strong synergy between EVI1 and ATRA was observed for GDF15, which codes for a member of the TGF-β superfamily of cytokines. In line with the gene expression results, EVI1 enhanced cell cycle arrest, differentiation, and apoptosis in response to ATRA, and knockdown of GDF15 counteracted some of these effects. The potential clinical implications of these findings are discussed.
Collapse
Key Words
- AML, acute myeloid leukemia
- APL, acute promyelocytic leukemia
- ATRA, all-trans retinoic acid
- Ar, ATRA regulation
- DMSO, dimethyl sulfoxide
- EVI1
- Em, EVI1 modulation
- Er, EVI1 regulation
- FBS, fetal bovine serum
- FC, fold change
- FDR, false discovery rate
- GDF15
- GFP, green fluorescent protein
- MDS, myelodysplastic syndrome
- PSG, penicillin streptomycin glutamine
- RAR, retinoic acid receptor
- RARE, retinoic acid response element
- SE, standard error
- all-trans retinoic acid
- apoptosis
- cell cycle
- gene expression profiling
- mcoEvi1, murine codon optimized Evi1
- myeloid differentiation
Collapse
Affiliation(s)
- Birgit Steinmetz
- a Department of Medicine I ; Medical University of Vienna ; Währinger Gürtel, Vienna , Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
55
|
Nagiec MM, Skepner AP, Negri J, Eichhorn M, Kuperwasser N, Comer E, Muncipinto G, Subramanian A, Clish C, Musunuru K, Duvall JR, Foley M, Perez JR, Palmer MAJ. Modulators of hepatic lipoprotein metabolism identified in a search for small-molecule inducers of tribbles pseudokinase 1 expression. PLoS One 2015; 10:e0120295. [PMID: 25811180 PMCID: PMC4374785 DOI: 10.1371/journal.pone.0120295] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 01/28/2015] [Indexed: 12/04/2022] Open
Abstract
Recent genome wide association studies have linked tribbles pseudokinase 1 (TRIB1) to the risk of coronary artery disease (CAD). Based on the observations that increased expression of TRIB1 reduces secretion of VLDL and is associated with lower plasma levels of LDL cholesterol and triglycerides, higher plasma levels of HDL cholesterol and reduced risk for myocardial infarction, we carried out a high throughput phenotypic screen based on quantitative RT-PCR assay to identify compounds that induce TRIB1 expression in human HepG2 hepatoma cells. In a screen of a collection of diversity-oriented synthesis (DOS)-derived compounds, we identified a series of benzofuran-based compounds that upregulate TRIB1 expression and phenocopy the effects of TRIB1 cDNA overexpression, as they inhibit triglyceride synthesis and apoB secretion in cells. In addition, the compounds downregulate expression of MTTP and APOC3, key components of the lipoprotein assembly pathway. However, CRISPR-Cas9 induced chromosomal disruption of the TRIB1 locus in HepG2 cells, while confirming its regulatory role in lipoprotein metabolism, demonstrated that the effects of benzofurans persist in TRIB1-null cells indicating that TRIB1 is sufficient but not necessary to transmit the effects of the drug. Remarkably, active benzofurans, as well as natural products capable of TRIB1 upregulation, also modulate hepatic cell cholesterol metabolism by elevating the expression of LDLR transcript and LDL receptor protein, while reducing the levels of PCSK9 transcript and secreted PCSK9 protein and stimulating LDL uptake. The effects of benzofurans are not masked by cholesterol depletion and are independent of the SREBP-2 regulatory circuit, indicating that these compounds represent a novel class of chemically tractable small-molecule modulators that shift cellular lipoprotein metabolism in HepG2 cells from lipogenesis to scavenging.
Collapse
Affiliation(s)
- Marek M. Nagiec
- Therapeutics Platform, Center for the Science of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Adam P. Skepner
- Therapeutics Platform, Center for the Science of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Joseph Negri
- Therapeutics Platform, Center for the Science of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Michelle Eichhorn
- Therapeutics Platform, Center for the Science of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Nicolas Kuperwasser
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, United States of America
| | - Eamon Comer
- Therapeutics Platform, Center for the Science of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Giovanni Muncipinto
- Therapeutics Platform, Center for the Science of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Aravind Subramanian
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Clary Clish
- Metabolite Profiling Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Kiran Musunuru
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, United States of America
| | - Jeremy R. Duvall
- Therapeutics Platform, Center for the Science of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Michael Foley
- Therapeutics Platform, Center for the Science of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Jose R. Perez
- Therapeutics Platform, Center for the Science of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Michelle A. J. Palmer
- Therapeutics Platform, Center for the Science of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| |
Collapse
|
56
|
EVI1 promotes tumor growth via transcriptional repression of MS4A3. J Hematol Oncol 2015; 8:28. [PMID: 25886616 PMCID: PMC4389965 DOI: 10.1186/s13045-015-0124-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 02/26/2015] [Indexed: 01/07/2023] Open
Abstract
Background The transcription factor Ecotropic Virus Integration site 1 (EVI1) regulates cellular proliferation, differentiation, and apoptosis, and its overexpression contributes to an aggressive course of disease in myeloid leukemias and other malignancies. Notwithstanding, knowledge about the target genes mediating its biological and pathological functions remains limited. We therefore aimed to identify and characterize novel EVI1 target genes in human myeloid cells. Methods U937T_EVI1, a human myeloid cell line expressing EVI1 in a tetracycline regulable manner, was subjected to gene expression profiling. qRT-PCR was used to confirm the regulation of membrane-spanning-4-domains subfamily-A member-3 (MS4A3) by EVI1. Reporter constructs containing various parts of the MS4A3 upstream region were employed in luciferase assays, and binding of EVI1 to the MS4A3 promoter was investigated by chromatin immunoprecipitation. U937 derivative cell lines experimentally expressing EVI1 and/or MS4A3 were generated by retroviral transduction, and tested for their tumorigenicity by subcutaneous injection into severe combined immunodeficient mice. Results Gene expression microarray analysis identified 27 unique genes that were up-regulated, and 29 unique genes that were down-regulated, in response to EVI1 induction in the human myeloid cell line U937T. The most strongly repressed gene was MS4A3, and its down-regulation by EVI1 was confirmed by qRT-PCR in additional, independent experimental model systems. MS4A3 mRNA levels were also negatively correlated with those of EVI1 in several published AML data sets. Reporter gene assays and chromatin immunoprecipitation showed that EVI1 regulated MS4A3 via direct binding to a promoter proximal region. Experimental re-expression of MS4A3 in an EVI1 overexpressing cell line counteracted the tumor promoting effect of EVI1 in a murine xenograft model by increasing the rate of apoptosis. Conclusions Our data reveal MS4A3 as a novel direct target of EVI1 in human myeloid cells, and show that its repression plays a role in EVI1 mediated tumor aggressiveness. Electronic supplementary material The online version of this article (doi:10.1186/s13045-015-0124-6) contains supplementary material, which is available to authorized users.
Collapse
|
57
|
Abstract
Protein phosphorylation lies at the heart of cell signalling, and somatic mutation(s) in kinases drives and sustains a multitude of human diseases, including cancer. The human protein kinase superfamily (the kinome) encodes approximately 50 'pseudokinases', which were initially predicted to be incapable of dynamic cell signalling when compared with canonical enzymatically active kinases. This assumption was supported by bioinformatics, which showed that amino acid changes at one or more key loci, making up the nucleotide-binding site or phosphotransferase machinery, were conserved in multiple vertebrate and non-vertebrate pseudokinase homologues. Protein kinases are highly attractive targets for drug discovery, as evidenced by the approval of almost 30 kinase inhibitors in oncology, and the successful development of the dual JAK1/2 (Janus kinase 1/2) inhibitor ruxolitinib for inflammatory indications. However, for such a large (>550) protein family, a remarkable number have still not been analysed at the molecular level, and only a surprisingly small percentage of kinases have been successfully targeted clinically. This is despite evidence that many are potential candidates for the development of new therapeutics. Indeed, several recent reports confirm that disease-associated pseudokinases can bind to nucleotide co-factors at concentrations achievable in the cell. Together, these findings suggest that drug targeting using either ATP-site or unbiased ligand-discovery approaches should now be attempted using the validation technology currently employed to evaluate their classic protein kinase counterparts. In the present review, we discuss members of the human pseudokinome repertoire, and catalogue somatic amino acid pseudokinase mutations that are emerging as the depth and clinical coverage of the human cancer pseudokinome expand.
Collapse
|
58
|
Tanaka M, Aisaki KI, Kitajima S, Igarashi K, Kanno J, Nakamura T. Gene expression response to EWS-FLI1 in mouse embryonic cartilage. GENOMICS DATA 2014; 2:296-8. [PMID: 26484113 PMCID: PMC4535656 DOI: 10.1016/j.gdata.2014.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 09/03/2014] [Accepted: 09/07/2014] [Indexed: 01/12/2023]
Abstract
Ewing's sarcoma is a rare bone tumor that affects children and adolescents. We have recently succeeded to induce Ewing's sarcoma-like small round cell tumor in mice by expression of EWS-ETS fusion genes in murine embryonic osteochondrogenic progenitors. The Ewing's sarcoma precursors are enriched in embryonic superficial zone (eSZ) cells of long bone. To get insights into the mechanisms of Ewing's sarcoma development, gene expression profiles between EWS-FLI1-sensitive eSZ cells and EWS-FLI1-resistant embryonic growth plate (eGP) cells were compared using DNA microarrays. Gene expression of eSZ and eGP cells (total, 30 samples) was evaluated with or without EWS-FLI1 expression 0, 8 or 48 h after gene transduction. Our data provide useful information for gene expression responses to fusion oncogenes in human sarcoma.
Collapse
Affiliation(s)
- Miwa Tanaka
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Ken-Ichi Aisaki
- Division of Cellular and Molecular Toxicology, Biosafety Research Center, National Institute of Health Science, Tokyo, Japan
| | - Satoshi Kitajima
- Division of Cellular and Molecular Toxicology, Biosafety Research Center, National Institute of Health Science, Tokyo, Japan
| | - Katsuhide Igarashi
- Division of Cellular and Molecular Toxicology, Biosafety Research Center, National Institute of Health Science, Tokyo, Japan
| | - Jun Kanno
- Division of Cellular and Molecular Toxicology, Biosafety Research Center, National Institute of Health Science, Tokyo, Japan
| | - Takuro Nakamura
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| |
Collapse
|
59
|
Salazar M, Lorente M, García-Taboada E, Pérez Gómez E, Dávila D, Zúñiga-García P, María Flores J, Rodríguez A, Hegedus Z, Mosén-Ansorena D, Aransay AM, Hernández-Tiedra S, López-Valero I, Quintanilla M, Sánchez C, Iovanna JL, Dusetti N, Guzmán M, Francis SE, Carracedo A, Kiss-Toth E, Velasco G. Loss of Tribbles pseudokinase-3 promotes Akt-driven tumorigenesis via FOXO inactivation. Cell Death Differ 2014; 22:131-44. [PMID: 25168244 DOI: 10.1038/cdd.2014.133] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 07/11/2014] [Accepted: 07/24/2014] [Indexed: 01/08/2023] Open
Abstract
Tribbles pseudokinase-3 (TRIB3) has been proposed to act as an inhibitor of AKT although the precise molecular basis of this activity and whether the loss of TRIB3 contributes to cancer initiation and progression remain to be clarified. In this study, by using a wide array of in vitro and in vivo approaches, including a Trib3 knockout mouse, we demonstrate that TRIB3 has a tumor-suppressing role. We also find that the mechanism by which TRIB3 loss enhances tumorigenesis relies on the dysregulation of the phosphorylation of AKT by the mTORC2 complex, which leads to an enhanced phosphorylation of AKT on Ser473 and the subsequent hyperphosphorylation and inactivation of the transcription factor FOXO3. These observations support the notion that loss of TRIB3 is associated with a more aggressive phenotype in various types of tumors by enhancing the activity of the mTORC2/AKT/FOXO axis.
Collapse
Affiliation(s)
- M Salazar
- 1] Department of Biochemistry and Molecular Biology I, School of Biology, Complutense University, Madrid, Spain [2] Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, Spain
| | - M Lorente
- 1] Department of Biochemistry and Molecular Biology I, School of Biology, Complutense University, Madrid, Spain [2] Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, Spain
| | - E García-Taboada
- Department of Biochemistry and Molecular Biology I, School of Biology, Complutense University, Madrid, Spain
| | - E Pérez Gómez
- 1] Department of Biochemistry and Molecular Biology I, School of Biology, Complutense University, Madrid, Spain [2] Instituto de Investigación Hospital 12 de Octubre (I+12), Madrid, Spain
| | - D Dávila
- 1] Department of Biochemistry and Molecular Biology I, School of Biology, Complutense University, Madrid, Spain [2] Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, Spain
| | | | - J María Flores
- Department of Animal Surgery and Medicine, School of Veterinary, Complutense University, Madrid, Spain
| | - A Rodríguez
- Department of Animal Surgery and Medicine, School of Veterinary, Complutense University, Madrid, Spain
| | - Z Hegedus
- Institute of Biophysics, Hungarian Academy of Sciences, Szeged, Hungary
| | | | - A M Aransay
- CIC bioGUNE-CIBERehd, Bizkaia Technology Park, Derio, Spain
| | - S Hernández-Tiedra
- 1] Department of Biochemistry and Molecular Biology I, School of Biology, Complutense University, Madrid, Spain [2] Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, Spain
| | - I López-Valero
- 1] Department of Biochemistry and Molecular Biology I, School of Biology, Complutense University, Madrid, Spain [2] Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, Spain
| | - M Quintanilla
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
| | - C Sánchez
- 1] Department of Biochemistry and Molecular Biology I, School of Biology, Complutense University, Madrid, Spain [2] Instituto de Investigación Hospital 12 de Octubre (I+12), Madrid, Spain
| | - J L Iovanna
- Centre de Recherche en Carcérologie de Marseille (CRCM), INSERM UMR, CNRS UMR 7258, Aix Marseille Université and Institut Paoli Calmette, Marseille, France
| | - N Dusetti
- Centre de Recherche en Carcérologie de Marseille (CRCM), INSERM UMR, CNRS UMR 7258, Aix Marseille Université and Institut Paoli Calmette, Marseille, France
| | - M Guzmán
- 1] Department of Biochemistry and Molecular Biology I, School of Biology, Complutense University, Madrid, Spain [2] Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) and Instituto Ramón y Cajal de Investigaciones Sanitarias (IRYCIS), Madrid, Spain
| | - S E Francis
- Department of Cardiovascular Science, University of Sheffield, Sheffield, UK
| | - A Carracedo
- 1] CIC bioGUNE, Bizkaia Technology Park, Derio, Spain [2] Ikerbasque, Basque Foundation for Science, Bilbao, Spain [3] Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU), Bilbao, Spain
| | - E Kiss-Toth
- Department of Cardiovascular Science, University of Sheffield, Sheffield, UK
| | - G Velasco
- 1] Department of Biochemistry and Molecular Biology I, School of Biology, Complutense University, Madrid, Spain [2] Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, Spain
| |
Collapse
|
60
|
Su G, Lian X, Tan D, Tao H, Liu H, Chen S, Yin H, Wu D, Yin B. Aberrant expression of ecotropic viral integration site-1 in acute myeloid leukemia and acute lymphoblastic leukemia. Leuk Lymphoma 2014; 56:472-9. [DOI: 10.3109/10428194.2014.924118] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
61
|
Mashima T, Soma-Nagae T, Migita T, Kinoshita R, Iwamoto A, Yuasa T, Yonese J, Ishikawa Y, Seimiya H. TRIB1 supports prostate tumorigenesis and tumor-propagating cell survival by regulation of endoplasmic reticulum chaperone expression. Cancer Res 2014; 74:4888-97. [PMID: 24962028 DOI: 10.1158/0008-5472.can-13-3718] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Endocrine therapy is the standard treatment for advanced prostate cancer; however, relapse occurs in most patients with few treatment options available after recurrence. To overcome this therapeutic hurdle, the identification of new molecular targets is a critical issue. The capability to proliferate in three-dimensional (3D) conditions is a characteristic property of cancer cells. Therefore, factors that regulate 3D growth are considered rational targets for cancer therapy. Here, we applied a functional genomic approach to the 3D spheroid cell culture model and identified TRIB1, a member of the Trib family of serine/threonine kinase-like proteins, as an essential factor for prostate cancer cell growth and survival. RNAi-mediated silencing of TRIB1 suppressed prostate cancer cell growth selectively under the 3D conditions. This effect was rescued by ectopic expression of an RNAi-resistant TRIB1 exogene. Gene signature-based analysis revealed that TRIB1 was related to endoplasmic reticulum (ER) pathways in prostate cancer and was required for expression of the ER chaperone GRP78, which is critical for prostate tumorigenesis. Of note, GRP78 was expressed preferentially in a subpopulation of prostate cancer cells that possess tumor-propagating potential, and these tumor-propagating cells were highly sensitive to TRIB1 and GRP78 depletion. In a xenograft model of human prostate cancer, TRIB1 depletion strongly inhibited tumor formation. Supporting these observations, we documented frequent overexpression of TRIB1 in clinical specimens of prostate cancer. Overall, our results indicated that the TRIB1-ER chaperone axis drives prostate tumorigenesis and the survival of the tumor-propagating cells.
Collapse
Affiliation(s)
- Tetsuo Mashima
- Division of Molecular Biotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Taeko Soma-Nagae
- Division of Molecular Biotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Toshiro Migita
- Division of Molecular Biotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan. Department of Molecular Medicine, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Ryoko Kinoshita
- Division of Molecular Biotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan. Graduate School of Pharmaceutical Sciences, Meiji Pharmaceutical University, Tokyo, Japan
| | - Atsushi Iwamoto
- Division of Molecular Biotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan. Department of Medical Genome Sciences, Graduate School of Frontier Sciences, the University of Tokyo, Tokyo, Japan
| | - Takeshi Yuasa
- Department of Urology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Junji Yonese
- Department of Urology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yuichi Ishikawa
- Division of Pathology, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Hiroyuki Seimiya
- Division of Molecular Biotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan. Graduate School of Pharmaceutical Sciences, Meiji Pharmaceutical University, Tokyo, Japan. Department of Medical Genome Sciences, Graduate School of Frontier Sciences, the University of Tokyo, Tokyo, Japan.
| |
Collapse
|
62
|
Tanaka M, Yamazaki Y, Kanno Y, Igarashi K, Aisaki KI, Kanno J, Nakamura T. Ewing's sarcoma precursors are highly enriched in embryonic osteochondrogenic progenitors. J Clin Invest 2014; 124:3061-74. [PMID: 24911143 DOI: 10.1172/jci72399] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 04/10/2014] [Indexed: 12/18/2022] Open
Abstract
Ewing's sarcoma is a highly malignant bone tumor found in children and adolescents, and the origin of this malignancy is not well understood. Here, we introduced a Ewing's sarcoma-associated genetic fusion of the genes encoding the RNA-binding protein EWS and the transcription factor ETS (EWS-ETS) into a fraction of cells enriched for osteochondrogenic progenitors derived from the embryonic superficial zone (eSZ) of long bones collected from late gestational murine embryos. EWS-ETS fusions efficiently induced Ewing's sarcoma-like small round cell sarcoma formation by these cells. Analysis of the eSZ revealed a fraction of a precursor cells that express growth/differentiation factor 5 (Gdf5), the transcription factor Erg, and parathyroid hormone-like hormone (Pthlh), and selection of the Pthlh-positive fraction alone further enhanced EWS-ETS-dependent tumor induction. Genes downstream of the EWS-ETS fusion protein were quite transcriptionally active in eSZ cells, especially in regions in which the chromatin structure of the ETS-responsive locus was open. Inhibition of β-catenin, poly (ADP-ribose) polymerase 1 (PARP1), or enhancer of zeste homolog 2 (EZH2) suppressed cell growth in a murine model of Ewing's sarcoma, suggesting the utility of the current system as a preclinical model. These results indicate that eSZ cells are highly enriched in precursors to Ewing's sarcoma and provide clues to the histogenesis of Ewing's sarcoma in bone.
Collapse
|
63
|
Sakai Y, Fukamachi K, Futakuchi M, Miyoshi I, Tsuda H, Suzui M, Hayashi H. A novel transgenic mouse model carrying human Tribbles related protein 3 (TRB3) gene and its site specific phenotype. Biol Pharm Bull 2014; 37:1068-74. [PMID: 24882419 DOI: 10.1248/bpb.b14-00260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Tribbles related protein 3 (TRB3) pseudokinase plays a crucial role in cell proliferation, migration and morphogenesis during development. In our recent study, an introduction of human TRB3 gene into mouse mammary tumor cells caused an increase of proliferation of tumor cells and their nuclear size. In the current study, to examine whether this gene causes de novo morphological changes in a specific organ site we have developed a novel variation of the transgenic mouse model that conditionally expresses human TRB3 (hTRB3) gene using Cre-recombinase (Cre)/loxP recombination system. By injecting hTRB3 transgene construct into pronuclei of mouse embryo, we eventually obtained four hTRB3 mice. The gene expression was controlled by infection of adenovirus-expressing Cre via the tail vein of hTRB3 mouse. In Cre-mediated hTRB3 mouse, expression of the hTRB3 protein was detected in the cytoplasm of hepatocytes in the liver. Expression of this protein was also seen in lymphocytes in the spleen, glomerular endothelial cells, and epithelial cells of collecting duct of the kidney. In hepatocytes of the hTRB3 mouse, nuclear size was significantly greater than that of the wild type mouse, indicating that hTRB3 can play a role at least in part in hepatic morphogenesis. The present animal model may provide a system for evaluation of de novo morphological changes induced by a specific transgene in a specific organ site.
Collapse
Affiliation(s)
- Yuto Sakai
- Department of Drug Metabolism and Disposition, Graduate School of Pharmaceutical Sciences, Nagoya City University
| | | | | | | | | | | | | |
Collapse
|
64
|
Glass C, Wilson M, Gonzalez R, Zhang Y, Perkins AS. The role of EVI1 in myeloid malignancies. Blood Cells Mol Dis 2014; 53:67-76. [PMID: 24495476 DOI: 10.1016/j.bcmd.2014.01.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 12/26/2013] [Indexed: 01/01/2023]
Abstract
The EVI1 oncogene at human chr 3q26 is rearranged and/or overexpressed in a subset of acute myeloid leukemias and myelodysplasias. The EVI1 protein is a 135 kDa transcriptional regulator with DNA-binding zinc finger domains. Here we provide a critical review of the current state of research into the molecular mechanisms by which this gene plays a role in myeloid malignancies. The major pertinent cellular effects are blocking myeloid differentiation and preventing cellular apoptosis, and several potential mechanisms for these phenomena have been identified. Evidence supports a role for EVI1 in inducing cellular quiescence, and this may contribute to the resistance to chemotherapy seen in patients with neoplasms that overexpress EVI1. Another isoform, MDS1-EVI1 (or PRDM3), encoded by the same locus as EVI1, harbors an N-terminal histone methyltransferase(HMT) domain; experimental findings indicate that this protein and its HMT activity are critical for the progression of a subset of AMLs, and this provides a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Carolyn Glass
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine, Rochester, NY 14642, USA
| | - Michael Wilson
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine, Rochester, NY 14642, USA
| | - Ruby Gonzalez
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine, Rochester, NY 14642, USA
| | - Yi Zhang
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine, Rochester, NY 14642, USA
| | - Archibald S Perkins
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine, Rochester, NY 14642, USA.
| |
Collapse
|
65
|
Lin ZY, Huang YQ, Zhang YQ, Han ZD, He HC, Ling XH, Fu X, Dai QS, Cai C, Chen JH, Liang YX, Jiang FN, Zhong WD, Wang F, Wu CL. MicroRNA-224 inhibits progression of human prostate cancer by downregulating TRIB1. Int J Cancer 2014; 135:541-50. [PMID: 24382668 DOI: 10.1002/ijc.28707] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 12/18/2013] [Indexed: 12/18/2022]
Abstract
Our previous microarray data showed that microRNA-224 (miR-224) was downregulated in human prostate cancer (PCa) tissues compared with adjacent benign tissues. However, the underlying mechanisms by which miR-224 is involved in PCa remain unclear. In this study, we identified TRIB1 as a target gene of miR-224. Forced expression of miR-224 suppressed PCa cell proliferation, invasion and migration, and promoted cell apoptosis by downregulating TRIB1. Moreover, the expression level of miR-224 in PCa tissues was negatively correlated with that of TRIB1. miR-224 downregulation was frequently found in PCa tissues with metastasis, higher PSA level and clinical stage, whereas TRIB1 upregulation was significantly associated with metastasis. Both miR-224 downregulation and TRIB1 upregulation were significantly associated with poor biochemical recurrence-free survival of patients with PCa. In conclusion, these findings reveal that the aberrant expression of miR-224 and TRIB1 may promote PCa progression and have potentials to serve as novel biomarkers for PCa prognosis.
Collapse
Affiliation(s)
- Zhuo-Yuan Lin
- Guangdong Provincial Institute of Nephrology, Southern Medical University, Guangzhou, China; Department of Urology Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
66
|
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
|
67
|
Soubeyrand S, Naing T, Martinuk A, McPherson R. ERK1/2 regulates hepatocyte Trib1 in response to mitochondrial dysfunction. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:3405-3414. [PMID: 24161842 DOI: 10.1016/j.bbamcr.2013.10.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 09/13/2013] [Accepted: 10/02/2013] [Indexed: 01/10/2023]
Abstract
The TRIB1 locus (8q24.13) is a novel locus identified and replicated by several genome-wide association studies for associations with plasma triglycerides, apolipoprotein B and coronary artery disease. The TRIB1 protein product, tribbles-like protein 1 (Trib1), regulates MAPK activity. MAP kinases transduce a large variety of external signals, leading to a wide range of cellular responses, including growth, differentiation, inflammation and apoptosis. Importantly, Trib1 has been shown to regulate hepatic lipogenesis and very low density lipoprotein production. Despite the relevance of hepatocyte Trib1 to lipid metabolism and atherosclerosis, little is known about the mechanisms regulating Trib1 itself. Here, we identify the mitochondria axis as a regulator of Trib1. Treatment of HepG2 cells with a short pulse of a low oligomycin concentration led to a potent and prolonged increase in the Trib1 mRNA, an effect that was shared with other mitochondria stressors. HuH7 cells as well murine hepatocytes were also responsive albeit to a weaker extent. The upregulation appeared largely independent of reactive oxygen species generation or metabolic stress and was mainly under transcriptional control, with ERK1/2 playing an important regulating role in the process. While the presence of the Trib1 protein could be inferred, attempts to correlate the increased mRNA to changes in protein level were unsuccessful due to the lack of recognizable Trib1 signal. Our data enrich the current paradigm of Trib1 as an activator of the MAPK pathway by uncovering a role for MAPK in regulating Trib1.
Collapse
Affiliation(s)
| | - Thet Naing
- Atherogenomics Laboratory, University of Ottawa Heart Institute, Ottawa, Canada
| | - Amy Martinuk
- Atherogenomics Laboratory, University of Ottawa Heart Institute, Ottawa, Canada
| | - Ruth McPherson
- Atherogenomics Laboratory, University of Ottawa Heart Institute, Ottawa, Canada; Division of Cardiology, University of Ottawa Heart Institute, Ottawa, Canada.
| |
Collapse
|
68
|
Nagamachi A, Matsui H, Asou H, Ozaki Y, Aki D, Kanai A, Takubo K, Suda T, Nakamura T, Wolff L, Honda H, Inaba T. Haploinsufficiency of SAMD9L, an endosome fusion facilitator, causes myeloid malignancies in mice mimicking human diseases with monosomy 7. Cancer Cell 2013; 24:305-17. [PMID: 24029230 DOI: 10.1016/j.ccr.2013.08.011] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Revised: 12/25/2012] [Accepted: 08/15/2013] [Indexed: 01/21/2023]
Abstract
Monosomy 7 and interstitial deletion of 7q (-7/7q-) are well-recognized nonrandom chromosomal abnormalities frequently found among patients with myelodysplastic syndromes (MDSs) and myeloid leukemias. We previously identified candidate myeloid tumor suppressor genes (SAMD9, SAMD9-like = SAMD9L, and Miki) in the 7q21.3 subband. We established SAMD9L-deficient mice and found that SAMD9L(+/-) mice as well as SAMD9L(-/-) mice develop myeloid diseases resembling human diseases associated with -7/7q-. SAMD9L-deficient hematopoietic stem cells showed enhanced colony formation potential and in vivo reconstitution ability. SAMD9L localizes in early endosomes. SAMD9L-deficient cells showed delays in homotypic endosome fusion, resulting in persistence of ligand-bound cytokine receptors. These findings suggest that haploinsufficiency of SAMD9L and/or SAMD9 gene(s) contributes to myeloid transformation.
Collapse
Affiliation(s)
- Akiko Nagamachi
- Department of Molecular Oncology and Leukemia Program Project, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
69
|
Akira S, Misawa T, Satoh T, Saitoh T. Macrophages control innate inflammation. Diabetes Obes Metab 2013; 15 Suppl 3:10-8. [PMID: 24003916 DOI: 10.1111/dom.12151] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 05/22/2013] [Indexed: 12/18/2022]
Abstract
Macrophages play a critical role in the pathogenesis of metabolic diseases including gout and type 2 diabetes. The Nod-like receptor (NLR) family, pyrin domain containing 3 (NLRP3) forms the inflammasome with apoptosis-associated speck-like protein containing a CARD (ASC), the adaptor protein, and mediates inflammatory responses by macrophages. By compound screening, we found that tubulin polymerization inhibitors suppress NLRP3 inflammasome activation. NLRP3 inflammasome inducers reduce the NAD(+) level to inactivate the α-tubulin deacetylase Sirtuin 2, resulting in accumulation of acetylated α-tubulin. Acetylated α-tubulin mediates mitochondrial transport and subsequent proximity of ASC on mitochondria to NLRP3 on the endoplasmic reticulum. Thus, microtubule-driven transport of mitochondria is required for NLRP3 inflammasome activation. Macrophages are comprised of two subsets, M1 (inflammatory) and M2 (anti-inflammatory). Trib1 is an adaptor protein involved in protein degradation of immune-related transcription factors. We found that Trib1 is critical for the differentiation of F4/80(+) MR(+) tissue-resident M2-like macrophages. Mice lacking Trib1 in haematopoietic cells show severe lipodystrophy owing to increased lipolysis, even on a normal diet. In response to a high-fat diet, the mice show hypertriglyceridaemia and insulin resistance, together with increased proinflammatory cytokine production. Thus, Trib1 is critical for adipose tissue maintenance and suppression of metabolic disorders by controlling the differentiation of tissue-resident M2-like macrophages.
Collapse
Affiliation(s)
- S Akira
- Laboratory of Host Defense, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.
| | | | | | | |
Collapse
|
70
|
Abstract
The ubiquitin ligase constitutively photomorphogenic 1 (COP1) is involved in many biological responses in mammalian cells, but its role in tumorigenesis remains unclear. Here we show that COP1 is a ubiquitin ligase for the tumor suppressor CCAAT/enhancer-binding protein (C/EBPα) and promotes its degradation in vivo, thereby blocking myeloid differentiation of hematopoietic cells for tumorigenesis. In this process, mammalian homolog of Tribbles, Trib1, which contains a COP1-binding motif, is essential for down-regulation of C/EBPα expression. Murine bone marrow transplantation experiments showed that coexpression of COP1 accelerates development of acute myeloid leukemia induced by Trib1, which pathologically resembles that of p42C/EBPα-deficient mice. Interestingly, coexpression of ligase activity-deficient COP1 mutant abrogated Trib1-induced leukemogenesis. These results indicate that COP1 and Trib1 act as an oncoprotein complex functioning upstream of C/EBPα, and its ligase activity is crucial for leukemogenesis.
Collapse
|
71
|
Glass C, Wuertzer C, Cui X, Bi Y, Davuluri R, Xiao YY, Wilson M, Owens K, Zhang Y, Perkins A. Global Identification of EVI1 Target Genes in Acute Myeloid Leukemia. PLoS One 2013; 8:e67134. [PMID: 23826213 PMCID: PMC3694976 DOI: 10.1371/journal.pone.0067134] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 05/19/2013] [Indexed: 12/20/2022] Open
Abstract
The ecotropic virus integration site 1 (EVI1) transcription factor is associated with human myeloid malignancy of poor prognosis and is overexpressed in 8-10% of adult AML and strikingly up to 27% of pediatric MLL-rearranged leukemias. For the first time, we report comprehensive genomewide EVI1 binding and whole transcriptome gene deregulation in leukemic cells using a combination of ChIP-Seq and RNA-Seq expression profiling. We found disruption of terminal myeloid differentiation and cell cycle regulation to be prominent in EVI-induced leukemogenesis. Specifically, we identified EVI1 directly binds to and downregulates the master myeloid differentiation gene Cebpe and several of its downstream gene targets critical for terminal myeloid differentiation. We also found EVI1 binds to and downregulates Serpinb2 as well as numerous genes involved in the Jak-Stat signaling pathway. Finally, we identified decreased expression of several ATP-dependent P2X purinoreceptors genes involved in apoptosis mechanisms. These findings provide a foundation for future study of potential therapeutic gene targets for EVI1-induced leukemia.
Collapse
Affiliation(s)
- Carolyn Glass
- Department of Pathology and Lab Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Charles Wuertzer
- Department of Pathology and Lab Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Xiaohui Cui
- Department of Pathology and Lab Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Yingtao Bi
- Molecular and Cellular Oncogenesis Program, Center for Systems and Computational Biology The Wistar Institute, Philadelphia, Pennsylvania, United States of America
| | - Ramana Davuluri
- Molecular and Cellular Oncogenesis Program, Center for Systems and Computational Biology The Wistar Institute, Philadelphia, Pennsylvania, United States of America
| | - Ying-Yi Xiao
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Michael Wilson
- Department of Pathology and Lab Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Kristina Owens
- Department of Pathology and Lab Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Yi Zhang
- Department of Pathology and Lab Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Archibald Perkins
- Department of Pathology and Lab Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
| |
Collapse
|
72
|
Phosphorylation of the leukemic oncoprotein EVI1 on serine 196 modulates DNA binding, transcriptional repression and transforming ability. PLoS One 2013; 8:e66510. [PMID: 23776681 PMCID: PMC3680417 DOI: 10.1371/journal.pone.0066510] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Accepted: 05/10/2013] [Indexed: 11/26/2022] Open
Abstract
The EVI1 (ecotropic viral integration site 1) gene at 3q26 codes for a transcriptional regulator with an essential role in haematopoiesis. Overexpression of EVI1 in acute myeloid leukaemia (AML) is frequently associated with 3q26 rearrangements and confers extremely poor prognosis. EVI1 mediates transcriptional regulation, signalling, and epigenetic modifications by interacting with DNA, proteins and protein complexes. To explore to what extent protein phosphorylation impacts on EVI1 functions, we analysed endogenous EVI1 protein from a high EVI1 expressing Fanconi anaemia (FA) derived AML cell line. Mass spectrometric analysis of immunoprecipitated EVI1 revealed phosphorylation at serine 196 (S196) in the sixth zinc finger of the N-terminal zinc finger domain. Mutated EVI1 with an aspartate substitution at serine 196 (S196D), which mimics serine phosphorylation of this site, exhibited reduced DNA-binding and transcriptional repression from a gene promotor selectively targeted by the N-terminal zinc finger domain. Forced expression of the S196D mutant significantly reduced EVI1 mediated transformation of Rat1 fibroblasts. While EVI1-mediated serial replating of murine haematopoietic progenitors was maintained by EVI1-S196D, this was associated with significantly higher Evi1-trancript levels compared with WT-EVI1 or EVI1-S196A, mimicking S196 non-phosphorylated EVI1. These data suggest that EVI1 function is modulated by phosphorylation of the first zinc finger domain.
Collapse
|
73
|
Sakai Y, Fukamachi K, Futakuchi M, Hayashi H, Suzui M. Promotive effects of cell proliferation and chromosomal instability induced by tribbles-related protein 3 in mouse mammary tumor cells. Oncol Rep 2013; 30:64-70. [PMID: 23633152 DOI: 10.3892/or.2013.2441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 03/28/2013] [Indexed: 11/05/2022] Open
Abstract
Tribbles-related protein 3 (TRB3) has been shown to be a crucial modulator of tumorigenesis. However, the precise role and the functional morphology of TRB3 are not clearly understood. To elucidate these enigmas we established the cell line, M2TRB3, by introducing the human TRB3 gene and protein in Cl66M2 (M2) mouse mammary tumor cells. This cell line stably expressed the TRB3 gene and protein. After 72 h of cell culture, there was a 34% increase in the growth of M2TRB3 cells compared to the control M2 mock cells. The mean volume of the tumors originating from the M2TRB3 cells was significantly increased by 38% when compared to the mean volume of the M2 mock tumors, and the proliferating cell nuclear antigen (PCNA) labeling index in the M2TRB3 tumors was higher when compared to that of the M2 and M2 mock cells. In the tumor tissue samples, the mean diameter of nuclei in the M2TRB3 tumor cells (9.4±0.3 µm) showed a significant increase compared to that of the M2 mock tumor cells (7.0±0.2 µm). M2TRB3 cells also showed a marked increase in the population of tetraploid or octaploid nuclei compared to M2 mock cells bearing mainly either diploid or tetraploid nuclei. Western blot analysis revealed the overexpression of cyclin B1 and cyclin D1 in M2TRB3 cells when compared to that in the M2 mock cells. These novel findings provide further evidence that TRB3 promotes cell proliferation and chromosomal instability by causing polyploidization during development.
Collapse
Affiliation(s)
- Yuto Sakai
- Department of Molecular Toxicology, Nagoya City University Graduate School of Medical Sciences and Medical School, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | | | | | | | | |
Collapse
|
74
|
Abstract
There is growing research interest in the mammalian Tribbles (Trib) family of serine/threonine pseudokinases and their oncogenic association with acute leukemias. This review is to understand the role of Trib genes in hematopoietic malignancies and their potential as targets for novel therapeutic strategies in acute myeloid leukemia and acute lymphoblastic leukemia. We discuss the role of Tribs as central signaling mediators in different subtypes of acute leukemia and propose that inhibition of dysregulated Trib signaling may be therapeutically beneficial.
Collapse
|
75
|
Satoh T, Kidoya H, Naito H, Yamamoto M, Takemura N, Nakagawa K, Yoshioka Y, Morii E, Takakura N, Takeuchi O, Akira S. Critical role of Trib1 in differentiation of tissue-resident M2-like macrophages. Nature 2013; 495:524-8. [PMID: 23515163 DOI: 10.1038/nature11930] [Citation(s) in RCA: 236] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Accepted: 01/22/2013] [Indexed: 01/09/2023]
Abstract
Macrophages consist of at least two subgroups, M1 and M2 (refs 1-3). Whereas M1 macrophages are proinflammatory and have a central role in host defence against bacterial and viral infections, M2 macrophages are associated with responses to anti-inflammatory reactions, helminth infection, tissue remodelling, fibrosis and tumour progression. Trib1 is an adaptor protein involved in protein degradation by interacting with COP1 ubiquitin ligase. Genome-wide association studies in humans have implicated TRIB1 in lipid metabolism. Here we show that Trib1 is critical for the differentiation of F4/80(+)MR(+) tissue-resident macrophages--that share characteristics with M2 macrophages (which we term M2-like macrophages)--and eosinophils but not for the differentiation of M1 myeloid cells. Trib1 deficiency results in a severe reduction of M2-like macrophages in various organs, including bone marrow, spleen, lung and adipose tissues. Aberrant expression of C/EBPα in Trib1-deficient bone marrow cells is responsible for the defects in macrophage differentiation. Unexpectedly, mice lacking Trib1 in haematopoietic cells show diminished adipose tissue mass accompanied by evidence of increased lipolysis, even when fed a normal diet. Supplementation of M2-like macrophages rescues the pathophysiology, indicating that a lack of these macrophages is the cause of lipolysis. In response to a high-fat diet, mice lacking Trib1 in haematopoietic cells develop hypertriglyceridaemia and insulin resistance, together with increased proinflammatory cytokine gene induction. Collectively, these results demonstrate that Trib1 is critical for adipose tissue maintenance and suppression of metabolic disorders by controlling the differentiation of tissue-resident M2-like macrophages.
Collapse
Affiliation(s)
- Takashi Satoh
- Laboratory of Host Defense, WPI Immunology Frontier Research Center, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Dugast E, Kiss-Toth E, Docherty L, Danger R, Chesneau M, Pichard V, Judor JP, Pettré S, Conchon S, Soulillou JP, Brouard S, Ashton-Chess J. Identification of tribbles-1 as a novel binding partner of Foxp3 in regulatory T cells. J Biol Chem 2013; 288:10051-10060. [PMID: 23417677 DOI: 10.1074/jbc.m112.448654] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
In a previous study, we identified TRIB1, a serine-threonine kinase-like molecule, as a biomarker of chronic antibody-mediated rejection of human kidneys when measured in peripheral blood mononuclear cells. Here, we focused our analysis on a specific subset of peripheral blood mononuclear cells that play a dominant role in regulating immune responses in health and disease, so-called CD4(+)CD25(+)Foxp3(+) regulatory T cells (Tregs). We isolated both human and murine Treg and non-Treg counterparts and analyzed TRIB1 and Foxp3 mRNA expression by quantitative PCR on the freshly isolated cells or following 24 h of activation. Physical interaction between the human TRIB1 and Foxp3 proteins was analyzed in live cell lines by protein complementation assay using both flow cytometry and microscopy and confirmed in primary freshly isolated human CD4(+)CD25(hi)CD127(-) Tregs by co-immunoprecipitation. Both TRIB1 and Foxp3 were expressed at significantly higher levels in Tregs than in their CD4(+)CD25(-) counterparts (p < 0.001). Moreover, TRIB1 and Foxp3 mRNA levels correlated tightly in Tregs (Spearman r = 1.0; p < 0.001, n = 7), but not in CD4(+)CD25(-) T cells. The protein complementation assay revealed a direct physical interaction between TRIB1 and Foxp3 in live cells. This interaction was impaired upon deletion of the TRIB1 N-terminal but not the C-terminal domain, suggesting an interaction in the nucleus. This direct interaction within the nucleus was confirmed in primary human Tregs by co-immunoprecipitation. These data show a direct relationship between TRIB1 and Foxp3 in terms of their expression and physical interaction and highlight Tribbles-1 as a novel binding partner of Foxp3 in Tregs.
Collapse
Affiliation(s)
- Emilie Dugast
- UMR1064, Institut National de la Santé et de la Recherche Médicale, Nantes 44000, France; Faculté de Médecine Université de Nantes, Nantes 44000, France; TcLand Expression, 21 rue de la Noue Bras de Fer, 44200 Nantes, France; Institut de Recherche en Transplantation, Institut de Transplantation Urologie-Néphrologie, Nantes 44000, France
| | - Endre Kiss-Toth
- Department of Cardiovascular Science, University of Sheffield, Sheffield S10 2TN, United Kingdom
| | - Louise Docherty
- Department of Cardiovascular Science, University of Sheffield, Sheffield S10 2TN, United Kingdom
| | - Richard Danger
- UMR1064, Institut National de la Santé et de la Recherche Médicale, Nantes 44000, France; Faculté de Médecine Université de Nantes, Nantes 44000, France; Institut de Recherche en Transplantation, Institut de Transplantation Urologie-Néphrologie, Nantes 44000, France
| | - Mélanie Chesneau
- UMR1064, Institut National de la Santé et de la Recherche Médicale, Nantes 44000, France; Faculté de Médecine Université de Nantes, Nantes 44000, France; Institut de Recherche en Transplantation, Institut de Transplantation Urologie-Néphrologie, Nantes 44000, France
| | - Virginie Pichard
- UMR1064, Institut National de la Santé et de la Recherche Médicale, Nantes 44000, France; Faculté de Médecine Université de Nantes, Nantes 44000, France; Institut de Recherche en Transplantation, Institut de Transplantation Urologie-Néphrologie, Nantes 44000, France
| | - Jean-Paul Judor
- UMR1064, Institut National de la Santé et de la Recherche Médicale, Nantes 44000, France; Faculté de Médecine Université de Nantes, Nantes 44000, France; Institut de Recherche en Transplantation, Institut de Transplantation Urologie-Néphrologie, Nantes 44000, France
| | - Ségolène Pettré
- UMR1064, Institut National de la Santé et de la Recherche Médicale, Nantes 44000, France; Faculté de Médecine Université de Nantes, Nantes 44000, France; Institut de Recherche en Transplantation, Institut de Transplantation Urologie-Néphrologie, Nantes 44000, France
| | - Sophie Conchon
- UMR1064, Institut National de la Santé et de la Recherche Médicale, Nantes 44000, France; Faculté de Médecine Université de Nantes, Nantes 44000, France; Institut de Recherche en Transplantation, Institut de Transplantation Urologie-Néphrologie, Nantes 44000, France
| | - Jean-Paul Soulillou
- UMR1064, Institut National de la Santé et de la Recherche Médicale, Nantes 44000, France; Institut de Recherche en Transplantation, Institut de Transplantation Urologie-Néphrologie, Nantes 44000, France; Centre Hospitalier Universitaire de Nantes, Nantes 44000, France
| | - Sophie Brouard
- UMR1064, Institut National de la Santé et de la Recherche Médicale, Nantes 44000, France; Institut de Recherche en Transplantation, Institut de Transplantation Urologie-Néphrologie, Nantes 44000, France; Centre Hospitalier Universitaire de Nantes, Nantes 44000, France.
| | | |
Collapse
|
77
|
Cunard R. Mammalian tribbles homologs at the crossroads of endoplasmic reticulum stress and Mammalian target of rapamycin pathways. SCIENTIFICA 2013; 2013:750871. [PMID: 24490110 PMCID: PMC3892554 DOI: 10.1155/2013/750871] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 11/20/2013] [Indexed: 05/03/2023]
Abstract
In 2000, investigators discovered Tribbles, a Drosophila protein that coordinates morphogenesis by inhibiting mitosis. Further work has delineated Xenopus (Xtrb2), Nematode (Nipi-3), and mammalian homologs of Drosophila tribbles, which include TRB1, TRB2, and TRB3. The sequences of tribbles homologs are highly conserved, and despite their protein kinase structure, to date they have not been shown to have kinase activity. TRB family members play a role in the differentiation of macrophages, lymphocytes, muscle cells, adipocytes, and osteoblasts. TRB isoforms also coordinate a number of critical cellular processes including glucose and lipid metabolism, inflammation, cellular stress, survival, apoptosis, and tumorigenesis. TRB family members modulate multiple complex signaling networks including mitogen activated protein kinase cascades, protein kinase B/AKT signaling, mammalian target of rapamycin, and inflammatory pathways. The following review will discuss metazoan homologs of Drosophila tribbles, their structure, expression patterns, and functions. In particular, we will focus on TRB3 function in the kidney in podocytes. This review will also discuss the key signaling pathways with which tribbles proteins interact and provide a rationale for developing novel therapeutics that exploit these interactions to provide better treatment options for both acute and chronic kidney disease.
Collapse
Affiliation(s)
- Robyn Cunard
- Research Service and Division of Nephrology-Hypertension, Veterans Affairs San Diego Healthcare System, Veterans Medical Research Foundation, Mail Code 151, 3350 La Jolla Village Drive, San Diego, CA 92161, USA
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- *Robyn Cunard:
| |
Collapse
|
78
|
Abstract
Bone marrow (BM) cells depend on their niche for growth and survival. However, the genes modulated by niche stimuli have not been discriminated yet. For this purpose, we investigated BM aspirations from patients with various hematological malignancies. Each aspirate was fractionated, and the various samples were fixed at different time points and analyzed by microarray. Identification of niche-modulated genes relied on sustained change in expression following loss of niche regulation. Compared with the reference (‘authentic') samples, which were fixed immediately following aspiration, the BM samples fixed after longer stay out-of-niche acquired numerous changes in gene-expression profile (GEP). The overall genes modulated included a common subset of functionally diverse genes displaying prompt and sustained ‘switch' in expression irrespective of the tumor type. Interestingly, the ‘switch' in GEP was reversible and turned ‘off-and-on' again in culture conditions, resuming cell–cell–matrix contact versus respread into suspension, respectively. Moreover, the resuming of contact prolonged the survival of tumor cells out-of-niche, and the regression of the ‘contactless switch' was followed by induction of a new set of genes, this time mainly encoding extracellular proteins including angiogenic factors and extracellular matrix proteins. Our data set, being unique in authentic expression design, uncovered niche-modulated and niche-modulating genes capable of controlling homing, expansion and angiogenesis.
Collapse
|
79
|
Wolf S, Rudolph C, Morgan M, Büsche G, Salguero G, Stripecke R, Schlegelberger B, Baum C, Modlich U. Selection for Evi1 activation in myelomonocytic leukemia induced by hyperactive signaling through wild-type NRas. Oncogene 2012; 32:3028-38. [PMID: 22847614 DOI: 10.1038/onc.2012.329] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Activation of NRas signaling is frequently found in human myeloid leukemia and can be induced by activating mutations as well as by mutations in receptors or signaling molecules upstream of NRas. To study NRas-induced leukemogenesis, we retrovirally overexpressed wild-type NRas in a murine bone marrow transplantation (BMT) model in C57BL/6J mice. Overexpression of wild-type NRas caused myelomonocytic leukemias ∼3 months after BMT in the majority of mice. A subset of mice (30%) developed malignant histiocytosis similar to mice that received mutationally activated NRas(G12D)-expressing bone marrow. Aberrant Ras signaling was demonstrated in cells expressing mutationally active or wild-type NRas, as increased activation of Erk and Akt was observed in both models. However, more NRas(G12D) were found to be in the activated, GTP-bound state in comparison with wild-type NRas. Consistent with observations reported for primary human myelomonocytic leukemia cells, Stat5 activation was also detected in murine leukemic cells. Furthermore, clonal evolution was detected in NRas wild-type-induced leukemias, including expansion of clones containing activating vector insertions in known oncogenes, such as Evi1 and Prdm16. In vitro cooperation of NRas and Evi1 improved long-term expansion of primary murine bone marrow cells. Evi1-positive cells upregulated Bcl-2 and may, therefore, provide anti-apoptotic signals that collaborate with the NRas-induced proliferative effects. As activation of Evi1 has been shown to coincide with NRAS mutations in human acute myeloid leukemia, our murine model recapitulates crucial events in human leukemogenesis.
Collapse
Affiliation(s)
- S Wolf
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
80
|
EVI-1 modulates leukemogenic potential and apoptosis sensitivity in human acute lymphoblastic leukemia. Leukemia 2012; 27:56-65. [DOI: 10.1038/leu.2012.211] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
81
|
Esmailzadeh S, Jiang X. AHI-1: a novel signaling protein and potential therapeutic target in human leukemia and brain disorders. Oncotarget 2012; 2:918-34. [PMID: 22248740 PMCID: PMC3282096 DOI: 10.18632/oncotarget.405] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Progress in the understanding of the molecular and cellular mechanisms of human cancer, including human leukemia and lymphomas, has been spurred by cloning of fusion genes created by chromosomal translocations or by retroviral insertional mutagenesis; a number of oncogenes and tumor suppressors involved in development of a number of malignancies have been identified in this manner. The BCR-ABL fusion gene, originating in a multipotent hematopoietic stem cell, is the molecular signature of chronic myeloid leukemia (CML). Discovery of this fusion gene has led to the development of one of the first successful targeted molecular therapies for cancer (Imatinib). It illustrates the advances that can result from an understanding of the molecular basis of disease. However, there still remain many as yet unidentified mutations that may influence the initiation or progression of human diseases. Thus, identification and characterization of the mechanism of action of genes that contribute to human diseases is an important and opportune area of current research. One promising candidate as a potential therapeutic target is Abelson helper integration site-1(Ahi-1/AHI-1) that was identified by retroviral insertional mutagenesis in murine models of leukemia/lymphomas and is highly elevated in certain human lymphoma and leukemia stem/progenitor cells. It encodes a unique protein with a SH3 domain, multiple SH3 binding sites and a WD40-repeat domain, suggesting that the normal protein has novel signaling activities. A new AHI-1-BCR-ABL-JAK2 interaction complex has recently been identified and this complex regulates transforming activities and drug resistance in CML stem/progenitor cells. Importantly, AHI-1 has recently been identified as a susceptibility gene involved in a number of brain disorders, including Joubert syndrome. Therefore, understanding molecular functions of the AHI-1 gene could lead to important and novel insights into disease processes involved in specific types of diseases. Ultimately, this knowledge will set the stage for translation into new and more effective diagnostic and treatment strategies.
Collapse
Affiliation(s)
- Sharmin Esmailzadeh
- Terry Fox Laboratory, British Columbia Cancer Agency and Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | | |
Collapse
|
82
|
Zhang H, Photiou A, Grothey A, Stebbing J, Giamas G. The role of pseudokinases in cancer. Cell Signal 2012; 24:1173-84. [PMID: 22330072 DOI: 10.1016/j.cellsig.2012.01.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Accepted: 01/27/2012] [Indexed: 01/12/2023]
Abstract
Kinases play a critical role in regulating many cellular functions including development, differentiation and proliferation. To date, over 518 proteins with kinase activity, comprising ~2-3% of total cellular proteins, have been identified from within the human kinome. Interestingly, approximately 10% of kinases are categorised as pseudokinases since they lack one or more conserved catalytic residues within their kinase domain and were originally thought to have no enzymatic activity. Recently, there has been strong evidence to suggest that some pseudokinsases can not only function as scaffold proteins, but may also possess kinase activity leading to modulation of cell signalling pathways. Altered activity of these pseudokinases can result in impaired cellular function, particularly in malignancies. In this review we are discussing recent evidence that apart from a scaffolding role, pseudokinases also orchestrate cellular processes as active kinases per se in signalling pathways of malignant cells.
Collapse
Affiliation(s)
- Hua Zhang
- Department of Cancer and Surgery, Imperial College, Hammersmith Hospital Campus, Du Cane Road, London, W12 ONN, UK
| | | | | | | | | |
Collapse
|
83
|
Kataoka K, Kurokawa M. Ecotropic viral integration site 1, stem cell self-renewal and leukemogenesis. Cancer Sci 2012; 103:1371-7. [PMID: 22494115 DOI: 10.1111/j.1349-7006.2012.02303.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Revised: 04/02/2012] [Accepted: 04/08/2012] [Indexed: 12/27/2022] Open
Abstract
It has become evident that acute myeloid leukemia (AML) is organized as a cellular hierarchy initiated and maintained by a subset of self-renewing leukemia stem cells. Recent gene expression profile analysis of human leukemia stem cells and hematopoietic stem cell (HSC) populations identified a key transcriptional program shared by leukemia stem cells and HSC, which is associated with adverse outcomes in AML patients. One molecule that has been established as a pivotal regulator in fine-tuning of stem cell properties as well as a potent oncogenic determinant is ecotropic viral integration site 1 (EVI1). EVI1 is a transcription factor that has stem cell-specific expression pattern and is essential for the regulation of HSC self-renewal. This gene is notorious for its involvement in AML, as its activation confers extremely poor prognosis in patients with AML. Molecular analysis has identified a variety of gene products that are involved in HSC regulation as downstream targets or interacting proteins of EVI1. Thus, exploration of the molecular pathogenesis underlying EVI1-related leukemogenesis provides insight into how shared stemness transcriptional programs contribute to leukemia progression and therapeutic resistance in AML. Here, we review the current knowledge regarding the role of EVI1 in HSC self-renewal and leukemogenesis and highlight the relationship between stem cell self-renewal properties and adverse outcome in myeloid malignancies.
Collapse
Affiliation(s)
- Keisuke Kataoka
- Department of Hematology and Oncology, University of Tokyo, Tokyo, Japan
| | | |
Collapse
|
84
|
Identification of TRIB1 R107L gain-of-function mutation in human acute megakaryocytic leukemia. Blood 2012; 119:2608-11. [DOI: 10.1182/blood-2010-12-324806] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Abstract
Trib1 has been identified as a myeloid oncogene in a murine leukemia model. Here we identified a TRIB1 somatic mutation in a human case of Down syndrome–related acute megakaryocytic leukemia. The mutation was observed at well-conserved arginine 107 residue in the pseudokinase domain. This R107L mutation remained in leukocytes of the remission stage in which GATA1 mutation disappeared, suggesting the TRIB1 mutation is an earlier genetic event in leukemogenesis. The bone marrow transfer experiment showed that acute myeloid leukemia development was accelerated by transducing murine bone marrow cells with the R107L mutant in which enhancement of ERK phosphorylation and C/EBPα degradation by Trib1 expression was even greater than in those expressing wild-type. These results suggest that TRIB1 may be a novel important oncogene for Down syndrome–related acute megakaryocytic leukemia.
Collapse
|
85
|
Fog CK, Galli GG, Lund AH. PRDM proteins: important players in differentiation and disease. Bioessays 2011; 34:50-60. [PMID: 22028065 DOI: 10.1002/bies.201100107] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The PRDM family has recently spawned considerable interest as it has been implicated in fundamental aspects of cellular differentiation and exhibits expanding ties to human diseases. The PRDMs belong to the SET domain family of histone methyltransferases, however, enzymatic activity has been determined for only few PRDMs suggesting that they act by recruiting co-factors or, more speculatively, confer methylation of non-histone targets. Several PRDM family members are deregulated in human diseases, most prominently in hematological malignancies and solid cancers, where they can act as both tumor suppressors or drivers of oncogenic processes. The molecular mechanisms have been delineated for only few PRDMs and little is known about functional redundancy within the family. Future studies should identify target genes of PRDM proteins and the protein complexes in which PRDM proteins reside to provide a more comprehensive understanding of the biological and biochemical functions of this important protein family.
Collapse
Affiliation(s)
- Cathrine K Fog
- Biotech Research and Innovation Centre and Centre for Epigenetics, University of Copenhagen, Denmark
| | | | | |
Collapse
|
86
|
Hua F, Mu R, Liu J, Xue J, Wang Z, Lin H, Yang H, Chen X, Hu Z. TRB3 interacts with SMAD3 promoting tumor cell migration and invasion. J Cell Sci 2011; 124:3235-46. [DOI: 10.1242/jcs.082875] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Tribbles homolog 3 (TRB3, also known as TRIB3, NIPK and SKIP3), a human homolog of Drosophila Tribbles, has been found to interact with a variety of signaling molecules to regulate diverse cellular functions. Here, we report that TRB3 is a novel SMAD3-interacting protein. Expression of exogenous TRB3 enhanced the transcriptional activity of SMAD3, whereas knocking down endogenous TRB3 reduced the transcriptional activity of SMAD3. The kinase-like domain (KD) of TRB3 was responsible for the interaction with SMAD3 and the regulation of SMAD3-mediated transcriptional activity. In addition, TGF-β1 stimulation or overexpression of SMAD3 enhanced the TRB3 promoter activity and expression, suggesting that there is a positive feedback loop between TRB3 and TGF-β–SMAD3 signaling. Mechanistically, TRB3 was found to trigger the degradation of SMAD ubiquitin regulatory factor 2 (Smurf2), which resulted in a decrease in the degradation of SMAD2 and phosphorylated SMAD3. Moreover, TRB3–SMAD3 interaction promoted the nuclear localization of SMAD3 because of the interaction of TRB3 with the MH2 domain of SMAD3. These effects of TRB3 were responsible for potentiating the SMAD3-mediated activity. Furthermore, knockdown of endogenous TRB3 expression inhibited the migration and invasion of tumor cells in vitro, which were associated with an increase in the expression of E-cadherin and a decrease in the expression of Twist-1 and Snail, two master regulators of epithelial-to-mesenchymal transition, suggesting a crucial role for TRB3 in maintaining the mesenchymal status of tumor cells. These results demonstrate that TRB3 acts as a novel SMAD3-interacting protein to participate in the positive regulation of TGF-β–SMAD-mediated cellular biological functions.
Collapse
Affiliation(s)
- Fang Hua
- Molecular Immunology and Pharmacology Laboratory, State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, P R China
| | - Rong Mu
- Molecular Immunology and Pharmacology Laboratory, State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, P R China
| | - Jinwen Liu
- Molecular Immunology and Pharmacology Laboratory, State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, P R China
| | - Jianfei Xue
- Molecular Immunology and Pharmacology Laboratory, State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, P R China
| | - Ziyan Wang
- Molecular Immunology and Pharmacology Laboratory, State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, P R China
| | - Heng Lin
- Molecular Immunology and Pharmacology Laboratory, State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, P R China
| | - Hongzhen Yang
- Molecular Immunology and Pharmacology Laboratory, State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, P R China
| | - Xiaoguang Chen
- Molecular Immunology and Pharmacology Laboratory, State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, P R China
| | - Zhuowei Hu
- Molecular Immunology and Pharmacology Laboratory, State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, P R China
| |
Collapse
|
87
|
Abstract
Cooperation of multiple mutations is thought to be required for cancer development. In previous studies, murine myeloid leukemias induced by transducing wild-type bone marrow progenitors with a SRY sex determining region Y-box 4 (Sox4)-expressing retrovirus frequently carried proviral insertions at Sfpi1, decreasing its mRNA levels, suggesting that reduced Sfpi1 expression cooperates with Sox4 in myeloid leukemia induction. In support of this hypothesis, we show here that mice receiving Sox4 virus-infected Sfpi1(ko/+) bone marrow progenitors developed myeloid leukemia with increased penetrance and shortened latency. Interestingly, Sox4 expression further decreased Sfpi1 transcription. Ectopic SOX4 expression reduced endogenous PU.1 mRNA levels in HL60 promyelocytes, and decreased Sfpi1 mRNA levels were also observed in the spleens of leukemic and preleukemic mice receiving Sox4 virus-infected wild-type bone marrow cells. In addition, Sox4 protein bound to a critical upstream regulatory element of Sfpi1 in ChIP assays. Such cooperation probably occurs in de novo human acute myeloid leukemias, as an analysis of 285 acute myeloid leukemia patient samples found a significant negative correlation between SOX4 and PU.1 expression. Our results establish a novel cooperation between Sox4 and reduced Sfpi1 expression in myeloid leukemia development and suggest that SOX4 could be an important new therapeutic target in human acute myeloid leukemia.
Collapse
|
88
|
Bhushan L, Kandpal RP. EphB6 receptor modulates micro RNA profile of breast carcinoma cells. PLoS One 2011; 6:e22484. [PMID: 21811619 PMCID: PMC3139643 DOI: 10.1371/journal.pone.0022484] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Accepted: 06/22/2011] [Indexed: 12/30/2022] Open
Abstract
Breast carcinoma cells have a specific pattern of expression for Eph receptors and ephrin ligands. EphB6 has previously been characterized as a signature molecule for invasive breast carcinoma cells. The transcription of EphB6 is silenced in breast carcinoma cells and its re-expression leads to decreased invasiveness of MDA-MB-231 cells. Such differences in phenotypes of native and EphB6 expressing MDA-MB-231 cells relate to an altered profile of micro RNAs. Comparative hybridization of total RNA to slides containing all known miRNAs by using locked nucleic acid (LNA) miRCURY platform yielded a significantly altered profile of miRNAs in MDA-MB-231 cells stably transfected with EphB6. After applying a threshold of change and a p-value of <0.001, the list of significantly altered miRNAs included miR-16, miR-23a, miR-24, miR-26a, miR-29a, miR-100, miRPlus-E1172 and miRPlus-E1258. The array-based changes were validated by real-time qPCR of miR-16, miR-23a, miR-24 and miR-100. Except miRPlus-E1172 and miRPlus-E1258, the remaining six miRNAs have been observed in a variety of cancers. The biological relevance of target mRNAs was predicted by using a common-target selection approach that allowed the identification of SMARCA5, SMARCC1, eIF2C2, eIF2C4, eIF4EBP2, FKABP5, FKBP1A, TRIB1, TRIB2, TRIB3, BMPR2, BMPR1A and BMPR1B as important targets of a subset of significantly altered miRNAs. Quantitative PCR revealed that the levels of SMARCC1, eIFC4, eIF4EB2, FKBP1a, FKBP5, TRIB1, TRIB3, BMPR1a and BMPR2 transcripts were significantly decreased in MDA-MB-231 cells transfected with EphB6. These observations confirm targeting of specific mRNAs by miR-100, miR-23a, miR-16 and miR-24, and suggest that the kinase-deficient EphB6 receptor is capable of initiating signal transduction from the cell surface to the nucleus resulting in the altered expression of a variety of genes involved in tumorigenesis and invasion. The alterations in miRNAs and their target mRNAs also suggest indirect involvement of EphB6 in PI3K/Akt/mTOR pathways.
Collapse
Affiliation(s)
- Lokesh Bhushan
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, California, United States of America
| | - Raj P. Kandpal
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, California, United States of America
- * E-mail:
| |
Collapse
|
89
|
Abstract
Sensing and interpreting extracellular signals in response to changes in the environment has been a fundamental feature of all life forms from the very beginning of evolution. To fulfil this function, networks of proteins have evolved, forming the intracellular signal transduction machinery. Whereas the appropriate control of these signal transduction systems is essential to homoeostasis, dysregulation of signalling leads to disease and often the death of the organism. The tribbles family of pseudokinases have emerged in recent years as key controllers of signal transduction via their interactions with several key kinases, ubiquitin ligases and transcription factors. In line with their role in regulating fundamentally important signalling pathways, members of the tribbles family have been implicated in the development of a range of human diseases. Whereas our mechanistic understanding of how these proteins contribute to disease is far from complete, the present paper attempts to summarize some of the most important recent developments in this field of research.
Collapse
|
90
|
Sugawara W, Arai Y, Kasai F, Fujiwara Y, Haruta M, Hosaka R, Nishida K, Kurosumi M, Kobayashi Y, Akagi K, Kaneko Y. Association of germline or somatic TP53 missense mutation with oncogene amplification in tumors developed in patients with Li-Fraumeni or Li-Fraumeni-like syndrome. Genes Chromosomes Cancer 2011; 50:535-45. [DOI: 10.1002/gcc.20878] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Accepted: 03/18/2011] [Indexed: 11/11/2022] Open
|
91
|
Yokoyama T, Nakamura T. Tribbles in disease: Signaling pathways important for cellular function and neoplastic transformation. Cancer Sci 2011; 102:1115-22. [PMID: 21338441 DOI: 10.1111/j.1349-7006.2011.01914.x] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The tribbles family of genes encodes pseudokinase proteins that are highly conserved in evolution. Instead of direct phosphorylation of target proteins, tribbles act as adaptors in signaling pathways for important cellular processes. These include mitogen-activated protein kinase kinase (MAPKK), CCAAT/enhancer-binding protein (C/EBP), activating transcription factor 4 (ATF4) and C/EBP-homologous protein (CHOP). Trib1 and Trib2 have been identified as myeloid oncogenes, and both may be involved in human leukemia. Tribbles proteins are also involved in a series of non-neoplastic disorders including metabolic and neurological diseases. The RAS/mitogen-activated protein kinase (MAPK) pathway molecules (in particular MAPK/ERK kinase 1 (MEK1) and C/EBP transcription factors) include tribbles-binding proteins that are involved in leukemogenesis, and the role of Trib1 as a linker between MAPK signaling and C/EBP degradation is proposed. Although the molecular function of tribbles is still under investigation, the research on tribbles in cellular processes, homeostasis of organisms and human diseases will provide valuable information for therapy of cancer as well as non-neoplastic disorders.
Collapse
Affiliation(s)
- Takashi Yokoyama
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | | |
Collapse
|
92
|
Evi1 represses PTEN expression and activates PI3K/AKT/mTOR via interactions with polycomb proteins. Blood 2011; 117:3617-28. [PMID: 21289308 DOI: 10.1182/blood-2009-12-261602] [Citation(s) in RCA: 111] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Evi1 (ecotropic viral integration site 1) is essential for proliferation of hematopoietic stem cells and implicated in the development of myeloid disorders. Particularly, high Evi1 expression defines one of the largest clusters in acute myeloid leukemia and is significantly associated with extremely poor prognosis. However, mechanistic basis of Evi1-mediated leukemogenesis has not been fully elucidated. Here, we show that Evi1 directly represses phosphatase and tensin homologue deleted on chromosome 10 (PTEN) transcription in the murine bone marrow, which leads to activation of AKT/mammalian target of rapamycin (mTOR) signaling. In a murine bone marrow transplantation model, Evi1 leukemia showed modestly increased sensitivity to an mTOR inhibitor rapamycin. Furthermore, we found that Evi1 binds to several polycomb group proteins and recruits polycomb repressive complexes for PTEN down-regulation, which shows a novel epigenetic mechanism of AKT/mTOR activation in leukemia. Expression analyses and ChIPassays with human samples indicate that our findings in mice models are recapitulated in human leukemic cells. Dependence of Evi1-expressing leukemic cells on AKT/mTOR signaling provides the first example of targeted therapeutic modalities that suppress the leukemogenic activity of Evi1. The PTEN/AKT/mTOR signaling pathway and the Evi1-polycomb interaction can be promising therapeutic targets for leukemia with activated Evi1.
Collapse
|
93
|
Evi-1 is a transcriptional target of mixed-lineage leukemia oncoproteins in hematopoietic stem cells. Blood 2010; 117:6304-14. [PMID: 21190993 DOI: 10.1182/blood-2009-07-234310] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Ecotropic viral integration site-1 (Evi-1) is a nuclear transcription factor that plays an essential role in the regulation of hematopoietic stem cells. Aberrant expression of Evi-1 has been reported in up to 10% of patients with acute myeloid leukemia and is a diagnostic marker that predicts a poor outcome. Although chromosomal rearrangement involving the Evi-1 gene is one of the major causes of Evi-1 activation, overexpression of Evi-1 is detected in a subgroup of acute myeloid leukemia patients without any chromosomal abnormalities, which indicates the presence of other mechanisms for Evi-1 activation. In this study, we found that Evi-1 is frequently up-regulated in bone marrow cells transformed by the mixed-lineage leukemia (MLL) chimeric genes MLL-ENL or MLL-AF9. Analysis of the Evi-1 gene promoter region revealed that MLL-ENL activates transcription of Evi-1. MLL-ENL-mediated up-regulation of Evi-1 occurs exclusively in the undifferentiated hematopoietic population, in which Evi-1 particularly contributes to the propagation of MLL-ENL-immortalized cells. Furthermore, gene-expression analysis of human acute myeloid leukemia cases demonstrated the stem cell-like gene-expression signature of MLL-rearranged leukemia with high levels of Evi-1. Our findings indicate that Evi-1 is one of the targets of MLL oncoproteins and is selectively activated in hematopoietic stem cell-derived MLL leukemic cells.
Collapse
|
94
|
Keeshan K, Bailis W, Dedhia PH, Vega ME, Shestova O, Xu L, Toscano K, Uljon SN, Blacklow SC, Pear WS. Transformation by Tribbles homolog 2 (Trib2) requires both the Trib2 kinase domain and COP1 binding. Blood 2010; 116:4948-57. [PMID: 20805362 PMCID: PMC3012589 DOI: 10.1182/blood-2009-10-247361] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2009] [Accepted: 08/07/2010] [Indexed: 01/20/2023] Open
Abstract
Tribbles homolog 2 (Trib2) is a pseudokinase that induces acute myelogenous leukemia (AML) in mice and is highly expressed in a subset of human AML. Trib2 has 3 distinct regions, a proline-rich N-terminus, a serine/threonine kinase homology domain, and a C-terminal constitutive photomorphogenesis 1 (COP1)-binding domain. We performed a structure-function analysis of Trib2 using in vitro and in vivo assays. The N-terminus was not required for Trib2-induced AML. Deletion or mutation of the COP1-binding site abrogated the ability of Trib2 to degrade CCAAT/enhancer-binding protein-α (C/EBP-α), block granulocytic differentiation, and to induce AML in vivo. Furthermore, COP1 knockdown inhibited the ability of Trib2 to degrade C/EBP-α, showing that it is important for mediating Trib2 activity. We also show that the Trib2 kinase domain is essential for its function. Trib2 contains variant catalytic loop sequences, compared with conventional kinases, that we show are necessary for Trib2 activity. The kinase domain mutants bind, but cannot efficiently degrade, C/EBP-α. Together, our data demonstrate that Trib2 can bind both COP1 and C/EBP-α, leading to degradation of C/EBP-α. Identification of the functional regions of Trib2 that are essential to its oncogenic role provides the basis for developing inhibitors that will block Trib functions in cancer.
Collapse
Affiliation(s)
- Karen Keeshan
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Jones L, Wei G, Sevcikova S, Phan V, Jain S, Shieh A, Wong JCY, Li M, Dubansky J, Maunakea ML, Ochoa R, Zhu G, Tennant TR, Shannon KM, Lowe SW, Le Beau MM, Kogan SC. Gain of MYC underlies recurrent trisomy of the MYC chromosome in acute promyelocytic leukemia. ACTA ACUST UNITED AC 2010; 207:2581-94. [PMID: 21059853 PMCID: PMC2989761 DOI: 10.1084/jem.20091071] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The leukemogenic effects of Myc drive recurrent trisomy in a mouse model of acute myeloid leukemia. Gain of chromosome 8 is the most common chromosomal gain in human acute myeloid leukemia (AML). It has been hypothesized that gain of the MYC protooncogene is of central importance in trisomy 8, but the experimental data to support this are limited and controversial. In a mouse model of promyelocytic leukemia in which the MRP8 promoter drives expression of the PML-RARA fusion gene in myeloid cells, a Myc allele is gained in approximately two-thirds of cases as a result of trisomy for mouse chromosome 15. We used this model to test the idea that MYC underlies acquisition of trisomy in AML. We used a retroviral vector to drive expression of wild-type, hypermorphic, or hypomorphic MYC in bone marrow that expressed the PML-RARA transgene. MYC retroviruses cooperated in myeloid leukemogenesis and suppressed gain of chromosome 15. When the PML-RARA transgene was expressed in a Myc haploinsufficient background, we observed selection for increased copies of the wild-type Myc allele concomitant with leukemic transformation. In addition, we found that human myeloid leukemias with trisomy 8 have increased MYC. These data show that gain of MYC can contribute to the pathogenic effect of the most common trisomy of human AML.
Collapse
Affiliation(s)
- Letetia Jones
- Helen Diller Family Comprehensive Cancer Center and Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Abstract
Trib1 is a myeloid oncogene that cooperates with Hoxa9 and Meis1. Although the MAPK pathway and C/EBP transcription factors are known to interact with Trib proteins, the mechanisms by which Trib1 contributes to myeloid leukemogenesis remains to be clarified. Here we report that interaction between Trib1 and MEK1 is required for Trib1-induced leukemogenesis. The C-terminal ILLHPWF motif that is well conserved among Trib family proteins is required for MEK1 binding, enhancement of ERK phosphorylation, enhanced self-renewal activity of bone marrow cells and leukemogenic activity by Trib1. The motif is also important for Trib1-induced C/EBPα degradation though interaction between Trib1 and C/EBPα is not necessary. Inhibition of ERK phosphorylation suppressed Trib1-induced C/EBPα degradation, indicating an important role for Trib1/MEK1 interaction. These results suggest that Trib1 may be a key mediator between the RTK-MAPK pathway and the C/EBP transcription factor in myeloid leukemogenesis.
Collapse
|
97
|
Goyama S, Kurokawa M. Evi-1 as a critical regulator of leukemic cells. Int J Hematol 2010; 91:753-7. [PMID: 20532840 DOI: 10.1007/s12185-010-0618-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2009] [Accepted: 05/21/2010] [Indexed: 11/25/2022]
Abstract
Ecotropic viral integration site-1 (EVI-1) has been recognized as one of the dominant oncogenes associated with murine and human myeloid leukemia. Recent clinical studies demonstrated that high EVI-1 expression was an independent negative prognostic indicator of survival in leukemia patients. In addition, gene-targeting studies in mice reveal that Evi-1 is preferentially expressed in hematopoietic stem cells (HSCs) and plays an essential role in proliferation/maintenance of HSCs. Proteins associated with EVI-1, signaling pathways regulated by EVI-1, and downstream mediators of EVI-1 transcriptional regulation have been described and characterized. In this study, we summarize current knowledge regarding biochemical properties and biological functions of EVI-1, which provides a foundation for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Susumu Goyama
- Department of Hematology and Oncology, Graduate School of Medicine, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | | |
Collapse
|
98
|
PU.1-mediated upregulation of CSF1R is crucial for leukemia stem cell potential induced by MOZ-TIF2. Nat Med 2010; 16:580-5, 1p following 585. [PMID: 20418886 DOI: 10.1038/nm.2122] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2010] [Accepted: 02/18/2010] [Indexed: 01/07/2023]
Abstract
Leukemias and other cancers possess self-renewing stem cells that help to maintain the cancer. Cancer stem cell eradication is thought to be crucial for successful anticancer therapy. Using an acute myeloid leukemia (AML) model induced by the leukemia-associated monocytic leukemia zinc finger (MOZ)-TIF2 fusion protein, we show here that AML can be cured by the ablation of leukemia stem cells. The MOZ fusion proteins MOZ-TIF2 and MOZ-CBP interacted with the transcription factor PU.1 to stimulate the expression of macrophage colony-stimulating factor receptor (CSF1R, also known as M-CSFR, c-FMS or CD115). Studies using PU.1-deficient mice showed that PU.1 is essential for the ability of MOZ-TIF2 to establish and maintain AML stem cells. Cells expressing high amounts of CSF1R (CSF1R(high) cells), but not those expressing low amounts of CSF1R (CSF1R(low) cells), showed potent leukemia-initiating activity. Using transgenic mice expressing a drug-inducible suicide gene controlled by the CSF1R promoter, we cured AML by ablation of CSF1R(high) cells. Moreover, induction of AML was suppressed in CSF1R-deficient mice and CSF1R inhibitors slowed the progression of MOZ-TIF2-induced leukemia. Thus, in this subtype of AML, leukemia stem cells are contained within the CSF1R(high) cell population, and we suggest that targeting of PU.1-mediated upregulation of CSF1R expression might be a useful therapeutic approach.
Collapse
|
99
|
Differential ability of Tribbles family members to promote degradation of C/EBPalpha and induce acute myelogenous leukemia. Blood 2010; 116:1321-8. [PMID: 20410507 DOI: 10.1182/blood-2009-07-229450] [Citation(s) in RCA: 117] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Trib1, Trib2, and Trib3 are mammalian homologs of Tribbles, an evolutionarily conserved Drosophila protein family that mediates protein degradation. Tribbles proteins function as adapters to recruit E3 ubiquitin ligases and enhance ubiquitylation of the target protein to promote its degradation. Increased Trib1 and Trib2 mRNA expression occurs in human myeloid leukemia and induces acute myeloid leukemia in mice, whereas Trib3 has not been associated with leukemia. Given the high degree of structural conservation among Tribbles family members, we directly compared the 3 mammalian Tribbles in hematopoietic cells by reconstituting mice with hematopoietic stem cells retrovirally expressing these proteins. All mice receiving Trib1 or Trib2 transduced hematopoietic stem cells developed acute myeloid leukemia, whereas Trib3 mice did not. Our previous data indicated that Trib2-mediated degradation of the transcription factor, CCAAT/enhancer-binding protein-alpha (C/EBPalpha), is important for leukemogenesis. Similar to Trib2, Trib1 induced C/EBPalpha degradation and inhibited its function. In contrast, Trib3 failed to inactivate or promote efficient degradation of C/EBPalpha. These data reveal that the 3 Tribbles homologs differ in their ability to promote degradation of C/EBPalpha, which account for their differential ability to induce leukemia.
Collapse
|
100
|
Camargo A, Ruano J, Fernandez JM, Parnell LD, Jimenez A, Santos-Gonzalez M, Marin C, Perez-Martinez P, Uceda M, Lopez-Miranda J, Perez-Jimenez F. Gene expression changes in mononuclear cells in patients with metabolic syndrome after acute intake of phenol-rich virgin olive oil. BMC Genomics 2010; 11:253. [PMID: 20406432 PMCID: PMC2874810 DOI: 10.1186/1471-2164-11-253] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2009] [Accepted: 04/20/2010] [Indexed: 11/10/2022] Open
Abstract
Background Previous studies have shown that acute intake of high-phenol virgin olive oil reduces pro-inflammatory, pro-oxidant and pro-thrombotic markers compared with low phenols virgin olive oil, but it still remains unclear whether effects attributed to its phenolic fraction are exerted at transcriptional level in vivo. To achieve this goal, we aimed at identifying expression changes in genes which could be mediated by virgin olive oil phenol compounds in the human. Results Postprandial gene expression microarray analysis was performed on peripheral blood mononuclear cells during postprandial period. Two virgin olive oil-based breakfasts with high (398 ppm) and low (70 ppm) content of phenolic compounds were administered to 20 patients suffering from metabolic syndrome following a double-blinded, randomized, crossover design. To eliminate the potential effect that might exist in their usual dietary habits, all subjects followed a similar low-fat, carbohydrate rich diet during the study period. Microarray analysis identified 98 differentially expressed genes (79 underexpressed and 19 overexpressed) when comparing the intake of phenol-rich olive oil with low-phenol olive oil. Many of these genes seem linked to obesity, dyslipemia and type 2 diabetes mellitus. Among these, several genes seem involved in inflammatory processes mediated by transcription factor NF-κB, activator protein-1 transcription factor complex AP-1, cytokines, mitogen-activated protein kinases MAPKs or arachidonic acid pathways. Conclusion This study shows that intake of virgin olive oil based breakfast, which is rich in phenol compounds is able to repress in vivo expression of several pro-inflammatory genes, thereby switching activity of peripheral blood mononuclear cells to a less deleterious inflammatory profile. These results provide at least a partial molecular basis for reduced risk of cardiovascular disease observed in Mediterranean countries, where virgin olive oil represents a main source of dietary fat. Admittedly, other lifestyle factors are also likely to contribute to lowered risk of cardiovascular disease in this region.
Collapse
Affiliation(s)
- Antonio Camargo
- IMIBIC (Instituto Maimonides de Investigacion Biomedica de Cordoba), Reina Sofia University Hospital, University of Cordoba, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|