1
|
Wu M, Liu Y, Zhu X, Zhang X, Kong Q, Lu W, Yuan X, Liu Y, Lu K, Dai Y, Zhang B. Advances in i-motif structures: Stability, gene expression, and therapeutic applications. Int J Biol Macromol 2025:143555. [PMID: 40294675 DOI: 10.1016/j.ijbiomac.2025.143555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 04/12/2025] [Accepted: 04/25/2025] [Indexed: 04/30/2025]
Abstract
The i-motif, a cytosine-rich DNA structure formed under acidic conditions, plays a pivotal role in regulating gene expression and holds significant therapeutic potential across various diseases. Found in the promoter regions of oncogenes such as Bcl-2, C-MYC, and KRAS, i-motifs dynamically interact with transcription factors and ligands to modulate oncogene activity. Their pH-sensitive nature enables innovative applications, including cellular pH sensors like the "i-switch" and drug delivery platforms such as DNA hydrogels that release therapeutics in acidic tumor microenvironments. However, challenges remain in developing specific ligands and detection methods. Advances in nanotechnology and multi-target therapies highlight the transformative potential of i-motifs in precision medicine. This review underscores the importance of i-motifs as therapeutic targets and tools, bridging fundamental research with clinical applications in oncology, metabolic disorders, and neurodegenerative diseases.
Collapse
Affiliation(s)
- Mengqing Wu
- College of Basic Medicine, Zunyi Medical University, Zunyi 563000, Guizhou Province, China
| | - Yang Liu
- College of Basic Medicine, Zunyi Medical University, Zunyi 563000, Guizhou Province, China; School of Public Health, Zunyi Medical University, Zunyi 563000, Guizhou Province, China
| | - Xiao Zhu
- College of Basic Medicine, Zunyi Medical University, Zunyi 563000, Guizhou Province, China
| | - Xiaoke Zhang
- College of Basic Medicine, Zunyi Medical University, Zunyi 563000, Guizhou Province, China
| | - Qinghong Kong
- College of Basic Medicine, Zunyi Medical University, Zunyi 563000, Guizhou Province, China
| | - Wei Lu
- College of Basic Medicine, Zunyi Medical University, Zunyi 563000, Guizhou Province, China
| | - Xiao Yuan
- College of Basic Medicine, Zunyi Medical University, Zunyi 563000, Guizhou Province, China
| | - Yunlai Liu
- College of Basic Medicine, Zunyi Medical University, Zunyi 563000, Guizhou Province, China
| | - Keyu Lu
- College of Basic Medicine, Zunyi Medical University, Zunyi 563000, Guizhou Province, China.
| | - Yangxue Dai
- College of Basic Medicine, Zunyi Medical University, Zunyi 563000, Guizhou Province, China.
| | - Bo Zhang
- College of Basic Medicine, Zunyi Medical University, Zunyi 563000, Guizhou Province, China.
| |
Collapse
|
2
|
Lemma RB, Fuglerud BM, Frampton J, Gabrielsen OS. MYB: A Key Transcription Factor in the Hematopoietic System Subject to Many Levels of Control. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1459:3-29. [PMID: 39017837 DOI: 10.1007/978-3-031-62731-6_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
MYB is a master regulator and pioneer factor highly expressed in hematopoietic progenitor cells (HPCs) where it contributes to the reprogramming processes operating during hematopoietic development. MYB plays a complex role being involved in several lineages of the hematopoietic system. At the molecular level, the MYB gene is subject to intricate regulation at many levels through several enhancer and promoter elements, through transcriptional elongation control, as well as post-transcriptional regulation. The protein is modulated by post-translational modifications (PTMs) such as SUMOylation restricting the expression of its downstream targets. Together with a range of interaction partners, cooperating transcription factors (TFs) and epigenetic regulators, MYB orchestrates a fine-tuned symphony of genes expressed during various stages of haematopoiesis. At the same time, the complex MYB system is vulnerable, being a target for unbalanced control and cancer development.
Collapse
Affiliation(s)
- Roza Berhanu Lemma
- Department of Biosciences, University of Oslo, Oslo, Norway
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo, Oslo, Norway
| | | | - Jon Frampton
- Department of Cancer & Genomic Sciences, College of Medicine & Health, University of Birmingham, Edgbaston, Birmingham, UK
| | | |
Collapse
|
3
|
Lemma RB, Ledsaak M, Fuglerud BM, Rodríguez-Castañeda F, Eskeland R, Gabrielsen OS. MYB regulates the SUMO protease SENP1 and its novel interaction partner UXT, modulating MYB target genes and the SUMO landscape. J Biol Chem 2023; 299:105062. [PMID: 37468105 PMCID: PMC10463205 DOI: 10.1016/j.jbc.2023.105062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 07/05/2023] [Accepted: 07/07/2023] [Indexed: 07/21/2023] Open
Abstract
SUMOylation is a post-translational modification frequently found on nuclear proteins, including transcription factors (TFs) and coactivators. By controlling the activity of several TFs, SUMOylation may have far-reaching effects. MYB is an example of a developmental TF subjected to SUMO-mediated regulation, through both SUMO conjugation and SUMO binding. How SUMO affects MYB target genes is unknown. Here, we explored the global effect of reduced SUMOylation of MYB on its downstream gene programs. RNA-Seq in K562 cells after MYB knockdown and rescue with mutants having an altered SUMO status revealed a number of differentially regulated genes and distinct gene ontology term enrichments. Clearly, the SUMO status of MYB both quantitatively and qualitatively affects its regulome. The transcriptome data further revealed that MYB upregulates the SUMO protease SENP1, a key enzyme that removes SUMO conjugation from SUMOylated proteins. Given this role of SENP1 in the MYB regulome, we expanded the analysis, mapped interaction partners of SENP1, and identified UXT as a novel player affecting the SUMO system by acting as a repressor of SENP1. MYB inhibits the expression of UXT suggesting that MYB is able not only to control a specific gene program directly but also indirectly by affecting the SUMO landscape through SENP1 and UXT. These findings suggest an autoactivation loop whereby MYB, through enhancing SENP1 and reducing UXT, is itself being activated by a reduced level of repressive SUMOylation. We propose that overexpressed MYB, seen in multiple cancers, may drive this autoactivation loop and contribute to oncogenic activation of MYB.
Collapse
Affiliation(s)
- Roza Berhanu Lemma
- Department of Biosciences, University of Oslo, Oslo, Norway; Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo, Oslo, Norway.
| | - Marit Ledsaak
- Department of Biosciences, University of Oslo, Oslo, Norway; Faculty of Medicine, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | | | | | - Ragnhild Eskeland
- Department of Biosciences, University of Oslo, Oslo, Norway; Faculty of Medicine, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway; Faculty of Medicine, Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | |
Collapse
|
4
|
Næs G, Storesund JO, Udayakumar P, Ledsaak M, Gabrielsen OS. Dissecting the transactivation domain (tAD) of the transcription factor c-Myb to assess recent models of tAD function. FEBS Open Bio 2020; 10:2329-2342. [PMID: 32937031 PMCID: PMC7609802 DOI: 10.1002/2211-5463.12978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/25/2020] [Accepted: 09/10/2020] [Indexed: 11/29/2022] Open
Abstract
Transcription factors use a DNA-binding domain to localize their action and a transactivation domain (tAD) to stimulate activation of the associated gene. Recent work has renewed interest in how tADs activate genes, which remains poorly understood. Key features in the new models are exposure of short linear motifs (SLMs) and liquid-liquid phase separation (LLPS). Inspired by the new models for tAD function, we decided to revisit the tAD of the haematopoietic transcription factor c-Myb by performing a mutational analysis to see how these new models fit and potentially explain the tAD behaviour of this master regulator. We know that c-Myb has an acidic tAD, which contains a well-characterized SLM in the form of a LxxLL motif. By testing 12 alanine-scanning mutants and three mutants with major reorganization of its tAD in two mammalian reporter systems, we found a pattern of effects very close to what would be expected from the SLM-exposure model, with strong effects exerted by both acidic replacements and SLM mutation. When the same mutants were tested in a yeast system, the pattern of effects was dramatically different, with the SLM mutation exerting no effect, and tAD behaviour was much less affected by small alterations, as would be expected from a LLPS model. These observations are discussed in the light of the two new tAD models, and a two-step hypothesis for transactivation, combining both models, is proposed.
Collapse
Affiliation(s)
- Guro Næs
- Department of BiosciencesUniversity of OsloNorway
| | | | | | | | | |
Collapse
|
5
|
Abstract
The c-Myb gene encodes a transcription factor that regulates cell proliferation, differentiation, and apoptosis through protein-protein interaction and transcriptional regulation of signaling pathways. The protein is frequently overexpressed in human leukemias, breast cancers, and other solid tumors suggesting that it is a bona fide oncogene. c-MYB is often overexpressed by translocation in human tumors with t(6;7)(q23;q34) resulting in c-MYB-TCRβ in T cell ALL, t(X;6)(p11;q23) with c-MYB-GATA1 in acute basophilic leukemia, and t(6;9)(q22-23;p23-24) with c-MYB-NF1B in adenoid cystic carcinoma. Antisense oligonucleotides to c-MYB were developed to purge bone marrow cells to eliminate tumor cells in leukemias. Recently, small molecules that inhibit c-MYB activity have been developed to disrupt its interaction with p300. The Dmp1 (cyclin D binding myb-like protein 1; Dmtf1) gene was isolated through its virtue for binding to cyclin D2. It is a transcription factor that has a Myb-like repeat for DNA binding. The Dmtf1 protein directly binds to the Arf promoter for transactivation and physically interacts with p53 to activate the p53 pathway. The gene is hemizygously deleted in 35-42% of human cancers and is associated with longer survival. The significances of aberrant expression of c-MYB and DMTF1 proteins in human cancers and their clinical significances are discussed.
Collapse
Affiliation(s)
- Elizabeth A. Fry
- The Department of Pathology, Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157 USA
| | - Kazushi Inoue
- The Department of Pathology, Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157 USA
| |
Collapse
|
6
|
Wang X, Angelis N, Thein SL. MYB - A regulatory factor in hematopoiesis. Gene 2018; 665:6-17. [PMID: 29704633 PMCID: PMC10764194 DOI: 10.1016/j.gene.2018.04.065] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 04/06/2018] [Accepted: 04/23/2018] [Indexed: 01/07/2023]
Abstract
MYB is a transcription factor which was identified in birds as a viral oncogene (v-MYB). Its cellular counterpart was subsequently isolated as c-MYB which has three functional domains - DNA binding domain, transactivation domain and negative regulatory domain. c-MYB is essential for survival, and deletion of both alleles of the gene results in embryonic death. It is highly expressed in hematopoietic cells, thymus and neural tissue, and required for T and B lymphocyte development and erythroid maturation. Additionally, aberrant MYB expression has been found in numerous solid cancer cells and human leukemia. Recent studies have also implicated c-MYB in the regulation of expression of fetal hemoglobin which is highly beneficial to the β-hemoglobinopathies (beta thalassemia and sickle cell disease). These findings suggest that MYB could be a potential therapeutic target in leukemia, and possibly also a target for therapeutic increase of fetal hemoglobin in the β-hemoglobinopathies.
Collapse
Affiliation(s)
- Xunde Wang
- National Heart, Lung and Blood Institute/NIH, Sickle Cell Branch, Bethesda, USA
| | - Nikolaos Angelis
- National Heart, Lung and Blood Institute/NIH, Sickle Cell Branch, Bethesda, USA
| | - Swee Lay Thein
- National Heart, Lung and Blood Institute/NIH, Sickle Cell Branch, Bethesda, USA.
| |
Collapse
|
7
|
Fuglerud BM, Lemma RB, Wanichawan P, Sundaram AYM, Eskeland R, Gabrielsen OS. A c-Myb mutant causes deregulated differentiation due to impaired histone binding and abrogated pioneer factor function. Nucleic Acids Res 2017; 45:7681-7696. [PMID: 28472346 PMCID: PMC5570105 DOI: 10.1093/nar/gkx364] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 04/26/2017] [Indexed: 12/21/2022] Open
Abstract
The transcription factor c-Myb is involved in early differentiation and proliferation of haematopoietic cells, where it operates as a regulator of self-renewal and multi-lineage differentiation. Deregulated c-Myb plays critical roles in leukaemias and other human cancers. Due to its role as a master regulator, we hypothesized it might function as a pioneer transcription factor. Our approach to test this was to analyse a mutant of c-Myb, D152V, previously reported to cause haematopoietic defects in mice by an unknown mechanism. Our transcriptome data from K562 cells indicates that this mutation specifically affects c-Myb's ability to regulate genes involved in differentiation, causing failure in c-Myb's ability to block differentiation. Furthermore, we see a major effect of this mutation in assays where chromatin opening is involved. We show that each repeat in the minimal DNA-binding domain of c-Myb binds to histones and that D152V disrupts histone binding of the third repeat. ATAC-seq data indicates this mutation impairs the ability of c-Myb to cause chromatin opening at specific sites. Taken together, our findings support that c-Myb acts as a pioneer factor and show that D152V impairs this function. The D152V mutant is the first mutant of a transcription factor specifically destroying pioneer factor function.
Collapse
Affiliation(s)
- Bettina M Fuglerud
- Department of Biosciences, University of Oslo, P.O.Box 1066 Blindern, N-0316 Oslo, Norway
| | - Roza B Lemma
- Department of Biosciences, University of Oslo, P.O.Box 1066 Blindern, N-0316 Oslo, Norway
| | - Pimthanya Wanichawan
- Department of Biosciences, University of Oslo, P.O.Box 1066 Blindern, N-0316 Oslo, Norway.,Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, P.O.Box 4956 Nydalen, N-0424 Oslo, Norway.,Center for Heart Failure Research, Oslo University Hospital and University of Oslo, P.O.Box 4956 Nydalen, N-0424 Oslo, Norway
| | - Arvind Y M Sundaram
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, P.O.Box 4950 Nydalen, N-0424 Oslo, Norway
| | - Ragnhild Eskeland
- Department of Biosciences, University of Oslo, P.O.Box 1066 Blindern, N-0316 Oslo, Norway.,Norwegian Center for Stem Cell Research, Department of Immunology, Oslo University Hospital, P.O.Box 1112 Blindern, N-0317 Oslo, Norway
| | - Odd S Gabrielsen
- Department of Biosciences, University of Oslo, P.O.Box 1066 Blindern, N-0316 Oslo, Norway
| |
Collapse
|
8
|
CHOI GUNHO, KO KIHAN, KIM JUNGOH, KIM JINKWON, OH SEUNGHUN, HAN INBO, CHO KYUNGGI, KIM OKJOON, BAE JINKUN, KIM NAMKEUN. Association of miR-34a, miR-130a, miR-150 and miR-155 polymorphisms with the risk of ischemic stroke. Int J Mol Med 2016; 38:345-56. [DOI: 10.3892/ijmm.2016.2609] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 05/11/2016] [Indexed: 11/06/2022] Open
|
9
|
Shikatani EA, Chandy M, Besla R, Li CC, Momen A, El-Mounayri O, Robbins CS, Husain M. c-Myb Regulates Proliferation and Differentiation of Adventitial Sca1+ Vascular Smooth Muscle Cell Progenitors by Transactivation of Myocardin. Arterioscler Thromb Vasc Biol 2016; 36:1367-76. [PMID: 27174098 DOI: 10.1161/atvbaha.115.307116] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 04/29/2016] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Vascular smooth muscle cells (VSMCs) are believed to dedifferentiate and proliferate in response to vessel injury. Recently, adventitial progenitor cells were implicated as a source of VSMCs involved in vessel remodeling. c-Myb is a transcription factor known to regulate VSMC proliferation in vivo and differentiation of VSMCs from mouse embryonic stem cell-derived progenitors in vitro. However, the role of c-Myb in regulating specific adult vascular progenitor cell populations was not known. Our objective was to examine the role of c-Myb in the proliferation and differentiation of Sca1(+) adventitial VSMC progenitor cells. APPROACH AND RESULTS Using mice with wild-type or hypomorphic c-myb (c-myb(h/h)), BrdU (bromodeoxyuridine) uptake and flow cytometry revealed defective proliferation of Sca1(+) adventitial VSMC progenitor cells at 8, 14, and 28 days post carotid artery denudation injury in c-myb(h/h) arteries. c-myb(h/h) cKit(+)CD34(-)Flk1(-)Sca1(+)CD45(-)Lin(-) cells failed to proliferate, suggesting that c-myb regulates the activation of specific Sca1(+) progenitor cells in vivo and in vitro. Although expression levels of transforming growth factor-β1 did not vary between wild-type and c-myb(h/h) carotid arteries, in vitro differentiation of c-myb(h/h) Sca1(+) cells manifested defective transforming growth factor-β1-induced VSMC differentiation. This is mediated by reduced transcriptional activation of myocardin because chromatin immunoprecipitation revealed c-Myb binding to the myocardin promoter only during differentiation of Sca1(+) cells, myocardin promoter mutagenesis identified 2 specific c-Myb-responsive binding sites, and adenovirus-mediated expression of myocardin rescued the phenotype of c-myb(h/h) progenitors. CONCLUSIONS These data support a role for c-Myb in the regulation of VSMC progenitor cells and provide novel insight into how c-myb regulates VSMC differentiation through myocardin.
Collapse
Affiliation(s)
- Eric A Shikatani
- From the Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada (E.A.S., M.C., R.B., A.M., O.E.-M., C.S.R., M.H.); and Heart and Stroke Richard Lewar Centre of Excellence, Ted Rogers Centre for Heart Research, McEwen Centre for Regenerative Medicine, and Peter Munk Cardiac Centre (E.A.S., M.C., R.B., C.S.R., M.H.), Department of Laboratory Medicine and Pathobiology (E.A.S., R.B., C.S.R., M.H.), Department of Immunology (C.C.L., C.S.R.), and Department of Medicine (M.C., M.H.), University of Toronto, Ontario, Canada
| | - Mark Chandy
- From the Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada (E.A.S., M.C., R.B., A.M., O.E.-M., C.S.R., M.H.); and Heart and Stroke Richard Lewar Centre of Excellence, Ted Rogers Centre for Heart Research, McEwen Centre for Regenerative Medicine, and Peter Munk Cardiac Centre (E.A.S., M.C., R.B., C.S.R., M.H.), Department of Laboratory Medicine and Pathobiology (E.A.S., R.B., C.S.R., M.H.), Department of Immunology (C.C.L., C.S.R.), and Department of Medicine (M.C., M.H.), University of Toronto, Ontario, Canada
| | - Rickvinder Besla
- From the Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada (E.A.S., M.C., R.B., A.M., O.E.-M., C.S.R., M.H.); and Heart and Stroke Richard Lewar Centre of Excellence, Ted Rogers Centre for Heart Research, McEwen Centre for Regenerative Medicine, and Peter Munk Cardiac Centre (E.A.S., M.C., R.B., C.S.R., M.H.), Department of Laboratory Medicine and Pathobiology (E.A.S., R.B., C.S.R., M.H.), Department of Immunology (C.C.L., C.S.R.), and Department of Medicine (M.C., M.H.), University of Toronto, Ontario, Canada
| | - Cedric C Li
- From the Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada (E.A.S., M.C., R.B., A.M., O.E.-M., C.S.R., M.H.); and Heart and Stroke Richard Lewar Centre of Excellence, Ted Rogers Centre for Heart Research, McEwen Centre for Regenerative Medicine, and Peter Munk Cardiac Centre (E.A.S., M.C., R.B., C.S.R., M.H.), Department of Laboratory Medicine and Pathobiology (E.A.S., R.B., C.S.R., M.H.), Department of Immunology (C.C.L., C.S.R.), and Department of Medicine (M.C., M.H.), University of Toronto, Ontario, Canada
| | - Abdul Momen
- From the Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada (E.A.S., M.C., R.B., A.M., O.E.-M., C.S.R., M.H.); and Heart and Stroke Richard Lewar Centre of Excellence, Ted Rogers Centre for Heart Research, McEwen Centre for Regenerative Medicine, and Peter Munk Cardiac Centre (E.A.S., M.C., R.B., C.S.R., M.H.), Department of Laboratory Medicine and Pathobiology (E.A.S., R.B., C.S.R., M.H.), Department of Immunology (C.C.L., C.S.R.), and Department of Medicine (M.C., M.H.), University of Toronto, Ontario, Canada
| | - Omar El-Mounayri
- From the Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada (E.A.S., M.C., R.B., A.M., O.E.-M., C.S.R., M.H.); and Heart and Stroke Richard Lewar Centre of Excellence, Ted Rogers Centre for Heart Research, McEwen Centre for Regenerative Medicine, and Peter Munk Cardiac Centre (E.A.S., M.C., R.B., C.S.R., M.H.), Department of Laboratory Medicine and Pathobiology (E.A.S., R.B., C.S.R., M.H.), Department of Immunology (C.C.L., C.S.R.), and Department of Medicine (M.C., M.H.), University of Toronto, Ontario, Canada
| | - Clinton S Robbins
- From the Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada (E.A.S., M.C., R.B., A.M., O.E.-M., C.S.R., M.H.); and Heart and Stroke Richard Lewar Centre of Excellence, Ted Rogers Centre for Heart Research, McEwen Centre for Regenerative Medicine, and Peter Munk Cardiac Centre (E.A.S., M.C., R.B., C.S.R., M.H.), Department of Laboratory Medicine and Pathobiology (E.A.S., R.B., C.S.R., M.H.), Department of Immunology (C.C.L., C.S.R.), and Department of Medicine (M.C., M.H.), University of Toronto, Ontario, Canada
| | - Mansoor Husain
- From the Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada (E.A.S., M.C., R.B., A.M., O.E.-M., C.S.R., M.H.); and Heart and Stroke Richard Lewar Centre of Excellence, Ted Rogers Centre for Heart Research, McEwen Centre for Regenerative Medicine, and Peter Munk Cardiac Centre (E.A.S., M.C., R.B., C.S.R., M.H.), Department of Laboratory Medicine and Pathobiology (E.A.S., R.B., C.S.R., M.H.), Department of Immunology (C.C.L., C.S.R.), and Department of Medicine (M.C., M.H.), University of Toronto, Ontario, Canada.
| |
Collapse
|
10
|
Pan JH, Adair-Kirk TL, Patel AC, Huang T, Yozamp NS, Xu J, Reddy EP, Byers DE, Pierce RA, Holtzman MJ, Brody SL. Myb permits multilineage airway epithelial cell differentiation. Stem Cells 2015; 32:3245-56. [PMID: 25103188 DOI: 10.1002/stem.1814] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Accepted: 07/14/2014] [Indexed: 12/12/2022]
Abstract
The epithelium of the pulmonary airway is specially differentiated to provide defense against environmental insults, but also subject to dysregulated differentiation that results in lung disease. The current paradigm for airway epithelial differentiation is a one-step program whereby a p63(+) basal epithelial progenitor cell generates a ciliated or secretory cell lineage, but the cue for this transition and whether there are intermediate steps are poorly defined. Here, we identify transcription factor Myb as a key regulator that permits early multilineage differentiation of airway epithelial cells. Myb(+) cells were identified as p63(-) and therefore distinct from basal progenitor cells, but were still negative for markers of differentiation. Myb RNAi treatment of primary-culture airway epithelial cells and Myb gene deletion in mice resulted in a p63(-) population with failed maturation of Foxj1(+) ciliated cells as well as Scbg1a1(+) and Muc5ac(+) secretory cells. Consistent with these findings, analysis of whole genome expression of Myb-deficient cells identified Myb-dependent programs for ciliated and secretory cell differentiation. Myb(+) cells were rare in human airways but were increased in regions of ciliated cells and mucous cell hyperplasia in samples from subjects with chronic obstructive pulmonary disease. Together, the results show that a p63(-) Myb(+) population of airway epithelial cells represents a distinct intermediate stage of differentiation that is required under normal conditions and may be heightened in airway disease.
Collapse
Affiliation(s)
- Jie-Hong Pan
- Department of Medicine, Washington University, St. Louis, Missouri, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Sampurno S, Cross R, Pearson H, Kaur P, Malaterre J, Ramsay RG. Myb via TGFβ is required for collagen type 1 production and skin integrity. Growth Factors 2015; 33:102-12. [PMID: 25807069 DOI: 10.3109/08977194.2015.1016222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Skin integrity requires an ongoing replacement and repair orchestrated by several cell types. We previously investigated the architecture of the skin of avian myeloblastosis viral oncogene homolog (Myb) knock-out (KO) embryos and wound repair in Myb(+/)(-) mice revealing a need for Myb in the skin, attributed to fibroblast-dependent production of collagen type 1. Here, using targeted Myb deletion in keratin-14 (K14) positive cells we reveal further Myb-specific defects in epidermal cell proliferation, thickness and ultrastructural morphology. This was associated with a severe deficit in collagen type 1 production, reminiscent of that observed in patients with ichthyosis vulgaris and Ehlers-Danlos syndrome. Since collagen type 1 is a product of fibroblasts, the collagen defect observed was unexpected and appears to be directed by the loss of Myb with significantly reduced tumor growth factor beta 1 (Tgfβ-1) expression by primary keratinocytes. Our findings support a specific role for Myb in K14+ epithelial cells in the preservation of adult skin integrity and function.
Collapse
Affiliation(s)
- Shienny Sampurno
- Trescowthick Research Laboratories, Peter MacCallum Cancer Centre , East Melbourne , Australia
| | | | | | | | | | | |
Collapse
|
12
|
Ahmad SM, Busser BW, Huang D, Cozart EJ, Michaud S, Zhu X, Jeffries N, Aboukhalil A, Bulyk ML, Ovcharenko I, Michelson AM. Machine learning classification of cell-specific cardiac enhancers uncovers developmental subnetworks regulating progenitor cell division and cell fate specification. Development 2014; 141:878-88. [PMID: 24496624 PMCID: PMC3912831 DOI: 10.1242/dev.101709] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The Drosophila heart is composed of two distinct cell types, the contractile cardial cells (CCs) and the surrounding non-muscle pericardial cells (PCs), development of which is regulated by a network of conserved signaling molecules and transcription factors (TFs). Here, we used machine learning with array-based chromatin immunoprecipitation (ChIP) data and TF sequence motifs to computationally classify cell type-specific cardiac enhancers. Extensive testing of predicted enhancers at single-cell resolution revealed the added value of ChIP data for modeling cell type-specific activities. Furthermore, clustering the top-scoring classifier sequence features identified novel cardiac and cell type-specific regulatory motifs. For example, we found that the Myb motif learned by the classifier is crucial for CC activity, and the Myb TF acts in concert with two forkhead domain TFs and Polo kinase to regulate cardiac progenitor cell divisions. In addition, differential motif enrichment and cis-trans genetic studies revealed that the Notch signaling pathway TF Suppressor of Hairless [Su(H)] discriminates PC from CC enhancer activities. Collectively, these studies elucidate molecular pathways used in the regulatory decisions for proliferation and differentiation of cardiac progenitor cells, implicate Su(H) in regulating cell fate decisions of these progenitors, and document the utility of enhancer modeling in uncovering developmental regulatory subnetworks.
Collapse
Affiliation(s)
- Shaad M Ahmad
- Laboratory of Developmental Systems Biology, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Pieraccioli M, Imbastari F, Antonov A, Melino G, Raschellà G. Activation of miR200 by c-Myb depends on ZEB1 expression and miR200 promoter methylation. Cell Cycle 2014; 12:2309-20. [PMID: 24067373 DOI: 10.4161/cc.25405] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Tumor progression to metastasis is a complex, sequential process that requires proliferation, resistance to apoptosis, motility and invasion to colonize at distant sites. The acquisition of these features implies a phenotypic plasticity by tumor cells that must adapt to different conditions by modulating several signaling pathways (1) during the journey to the final site of metastasis. Several transcription factors and microRNA play a role in tumor progression, but less is known about the control of their expression during this process. Here, we demonstrate by ectopic expression and gene silencing that the proto-oncogene c-Myb activates the expression of the 5 members of miR200 family (miR200b, miR200a, miR429, miR200c and miR141) that are involved in the control of epithelial-mesenchymal transition (EMT) and metastasis in many types of cancers. Transcriptional activation of miR200 by c-Myb occurs through binding to myb binding sites located in the promoter regions of miR200 genes on human chromosomes 1 and 12. Furthermore, when c-Myb and the transcriptional repressor ZEB1 are co-expressed, as at the onset EMT, the repression by ZEB1 prevails over the activation by c-Myb, and the expression of miR200 is inhibited. We also demonstrate that during EMT induced by TGF-β, the promoters of miR200 genes are methylated, and their transcription is repressed regardless of the presence of repressors such as ZEB1 and activators such as c-Myb. Finally, we find a correlation between the expression of c-Myb and that of four out of 5 miR200 in a data set of 207 breast cancer patients.
Collapse
Affiliation(s)
- Marco Pieraccioli
- ENEA Research Center Casaccia, Radiation Biology and Human Health Unit, Rome, Italy
| | | | | | | | | |
Collapse
|
14
|
MicroRNA-195 inhibits non-small cell lung cancer cell proliferation, migration and invasion by targeting MYB. Cancer Lett 2014; 347:65-74. [PMID: 24486218 DOI: 10.1016/j.canlet.2014.01.019] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 01/20/2014] [Accepted: 01/22/2014] [Indexed: 12/14/2022]
Abstract
MicroRNA-195 (miR-195) has been implicated in several other cancers; however, its role in non-small cell lung cancer (NSCLC) remains unclear. In this study, we demonstrated that miR-195 was significantly down-regulated in NSCLC samples and cell lines compared with corresponding normal counterparts. In vitro and in vivo functional assays demonstrated that modulation of miR-195 expression affected NSCLC cell proliferation, migration and invasion. Using miRNA target prediction algorithms and reporter assays, we demonstrated that miR-195 suppressed the expression of MYB both at the mRNA and protein level, and was directly bound to the 3'untranslated region of MYB mRNA. Overexpression of MYB in NSCLC cells using an ectopic expression vector restored the decreased cell proliferation, migration and invasion effects induced by miR-195. Finally, we observed an inverse correlation between MYB and miR-195 in NSCLC. Taken together, our findings indicated that miR-195 functions as tumour suppressor in NSCLC, and the miR-195/MYB axis might represent a potential therapeutic target for NSCLC intervention.
Collapse
|
15
|
Gu A, Ji G, Yan L, Zhou Y. The 8-oxoguanine DNA glycosylase 1 (ogg1) decreases the vulnerability of the developing brain to DNA damage. DNA Repair (Amst) 2013; 12:1094-104. [DOI: 10.1016/j.dnarep.2013.08.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 08/17/2013] [Accepted: 08/27/2013] [Indexed: 12/17/2022]
|
16
|
Myb and the Regulation of Stem Cells in the Intestine and Brain: A Tale of Two Niches. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 786:353-68. [DOI: 10.1007/978-94-007-6621-1_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
17
|
Lorenzo PI, Brendeford EM, Gilfillan S, Gavrilov AA, Leedsak M, Razin SV, Eskeland R, Sæther T, Gabrielsen OS. Identification of c-Myb Target Genes in K562 Cells Reveals a Role for c-Myb as a Master Regulator. Genes Cancer 2012; 2:805-17. [PMID: 22393465 DOI: 10.1177/1947601911428224] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Accepted: 10/08/2011] [Indexed: 11/17/2022] Open
Abstract
The c-Myb transcription factor is an important regulator of hematopoietic cell development. c-Myb is expressed in immature hematopoietic cells and plays a direct role in lineage fate selection, cell cycle progression, and differentiation of myeloid as well as B- and T-lymphoid progenitor cells. As a DNA-binding transcription factor, c-Myb regulates specific gene programs through activation of target genes. Still, our understanding of these programs is incomplete. Here, we report a set of novel c-Myb target genes, identified using a combined approach: specific c-Myb knockdown by 2 different siRNAs and subsequent global expression profiling, combined with the confirmation of direct binding of c-Myb to the target promoters by ChIP assays. The combination of these 2 approaches, as well as additional validation such as cloning and testing the promoters in reporter assays, confirmed that MYADM, LMO2, GATA2, STAT5A, and IKZF1 are target genes of c-Myb. Additional studies, using chromosome conformation capture, demonstrated that c-Myb target genes may directly interact with each other, indicating that these genes may be coordinately regulated. Of the 5 novel target genes identified, 3 are transcription factors, and one is a transcriptional co-regulator, supporting a role of c-Myb as a master regulator controlling the expression of other transcriptional regulators in the hematopoietic system.
Collapse
|
18
|
Busser BW, Taher L, Kim Y, Tansey T, Bloom MJ, Ovcharenko I, Michelson AM. A machine learning approach for identifying novel cell type-specific transcriptional regulators of myogenesis. PLoS Genet 2012; 8:e1002531. [PMID: 22412381 PMCID: PMC3297574 DOI: 10.1371/journal.pgen.1002531] [Citation(s) in RCA: 34] [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: 03/30/2011] [Accepted: 12/23/2011] [Indexed: 12/22/2022] Open
Abstract
Transcriptional enhancers integrate the contributions of multiple classes of transcription factors (TFs) to orchestrate the myriad spatio-temporal gene expression programs that occur during development. A molecular understanding of enhancers with similar activities requires the identification of both their unique and their shared sequence features. To address this problem, we combined phylogenetic profiling with a DNA-based enhancer sequence classifier that analyzes the TF binding sites (TFBSs) governing the transcription of a co-expressed gene set. We first assembled a small number of enhancers that are active in Drosophila melanogaster muscle founder cells (FCs) and other mesodermal cell types. Using phylogenetic profiling, we increased the number of enhancers by incorporating orthologous but divergent sequences from other Drosophila species. Functional assays revealed that the diverged enhancer orthologs were active in largely similar patterns as their D. melanogaster counterparts, although there was extensive evolutionary shuffling of known TFBSs. We then built and trained a classifier using this enhancer set and identified additional related enhancers based on the presence or absence of known and putative TFBSs. Predicted FC enhancers were over-represented in proximity to known FC genes; and many of the TFBSs learned by the classifier were found to be critical for enhancer activity, including POU homeodomain, Myb, Ets, Forkhead, and T-box motifs. Empirical testing also revealed that the T-box TF encoded by org-1 is a previously uncharacterized regulator of muscle cell identity. Finally, we found extensive diversity in the composition of TFBSs within known FC enhancers, suggesting that motif combinatorics plays an essential role in the cellular specificity exhibited by such enhancers. In summary, machine learning combined with evolutionary sequence analysis is useful for recognizing novel TFBSs and for facilitating the identification of cognate TFs that coordinate cell type-specific developmental gene expression patterns.
Collapse
Affiliation(s)
- Brian W. Busser
- Laboratory of Developmental Systems Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Leila Taher
- Computational Biology Branch, National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Yongsok Kim
- Laboratory of Developmental Systems Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Terese Tansey
- Laboratory of Developmental Systems Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Molly J. Bloom
- Laboratory of Developmental Systems Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ivan Ovcharenko
- Computational Biology Branch, National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail: (IO); (AMM)
| | - Alan M. Michelson
- Laboratory of Developmental Systems Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail: (IO); (AMM)
| |
Collapse
|
19
|
Affiliation(s)
- Huiyuan Li
- State Key Laboratory of Experimental Haematology, Institute of Haematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin
| | - Haifeng Zhao
- State Key Laboratory of Experimental Haematology, Institute of Haematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin
| | - Donghai Wang
- Department of Haematology, Peking University First Hospital, Beijing, China
| | - Renchi Yang
- State Key Laboratory of Experimental Haematology, Institute of Haematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin
| |
Collapse
|
20
|
Whitney IE, Raven MA, Lu L, Williams RW, Reese BE. A QTL on chromosome 10 modulates cone photoreceptor number in the mouse retina. Invest Ophthalmol Vis Sci 2011; 52:3228-36. [PMID: 21330668 DOI: 10.1167/iovs.10-6693] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
PURPOSE This investigation examines the genetic sources of marked variation in cone photoreceptor number among inbred lines of mice, identifying candidate genes that may control the proliferation, differentiation, or survival of this neuronal population. METHODS Cone photoreceptor populations were counted in C57BL/6J (B6/J) and A/J strains, and 26 recombinant inbred (RI) strains derived from them. Eyes from RI strains were also collected for microarray analysis. Quantitative trait locus (QTL) analysis was carried out by simple and composite interval mapping and validated using a consomic line. Candidate genes were evaluated based on genetic variance between the parental strains and analysis of gene expression. Expression data, deposited in GeneNetwork (www.GeneNetwork.org), were used to generate a coexpression network of established cone photoreceptor genes as a reference standard. RESULTS B6/J has 70% more cone photoreceptors than A/J. A significant QTL was mapped to chromosome 10 (Chr 10) and confirmed using B6.A<10> mice. Of 19 positional candidate genes, one-the myeloblastosis oncogene (Myb)-stood out. Myb has a potentially damaging missense mutation, high retinal expression, and a known role in cell proliferation. The ectonucleotide pyrophosphatase/phosphodiesterase 1 gene (Enpp1) was a second strong candidate, with an expression pattern that covaried with cone photoreceptors and that was differentially expressed between the parental strains. Enpp1 and several other candidate genes covaried with multiple genes within the cone photoreceptor gene network. CONCLUSIONS The mouse retina shows marked variation in cone photoreceptor number, some of which must be controlled by polymorphisms in a gene or genes on Chr 10.
Collapse
Affiliation(s)
- Irene E Whitney
- Neuroscience Research Institute and Department of Molecular, Cell, and Developmental Biology, University of California, Santa Barbara, California 93106-5060, USA
| | | | | | | | | |
Collapse
|
21
|
Chen RX, Xia YH, Xue TC, Ye SL. Transcription factor c-Myb promotes the invasion of hepatocellular carcinoma cells via increasing osteopontin expression. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2010; 29:172. [PMID: 21190594 PMCID: PMC3023683 DOI: 10.1186/1756-9966-29-172] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2010] [Accepted: 12/30/2010] [Indexed: 01/04/2023]
Abstract
Background Specific gene expression is tightly regulated by various transcription factors. Osteopontin (OPN) is a phosphoprotein that mediates hepatocellular carcinoma (HCC) progression and metastasis. However, the mechanism of OPN up-regulation in HCC metastasis remains to be clarified. Methods Oligonucleotide array-based transcription factor assays were applied to compare different activities of transcription factors in two human HCC cell lines with different OPN expression levels. The effects of one selected transcription factor on OPN expression were further evaluated. Results Eleven transcription factors were over-expressed in metastatic HCC cell line HCCLM6 cells whereas twelve transcription factors were down-regulated. Electrophoretic mobility shift assays (EMSA) and reporter gene assays showed that one of up-regulated transcription factors c-Myb could bind the OPN promoter and increase its transcription activity. In addition, small interfering RNA targeting c-Myb could inhibit OPN expression and significantly decrease migration and invasion of HCCLM6 cells in vitro. Conclusion Our data first demonstrate that c-Myb has a functionally important role in the regulation of OPN expression in HCC cells, suggesting that c-Myb might be a new target to control HCC metastasis.
Collapse
Affiliation(s)
- Rong-Xin Chen
- Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | | | | | | |
Collapse
|
22
|
Guillon-Munos A, Dambrine G, Richerioux N, Coupeau D, Muylkens B, Rasschaert D. The chicken miR-150 targets the avian orthologue of the functional zebrafish MYB 3'UTR target site. BMC Mol Biol 2010; 11:67. [PMID: 20813039 PMCID: PMC2940766 DOI: 10.1186/1471-2199-11-67] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2009] [Accepted: 09/02/2010] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The c-myb proto-oncogene is the founding member of a family of transcription factors involved principally in haematopoiesis, in diverse organisms, from zebrafish to mammals. Its deregulation has been implicated in human leukaemogenesis and other cancers. The expression of c-myb is tightly regulated by post-transcriptional mechanisms involving microRNAs. MicroRNAs are small, highly conserved non-coding RNAs that inhibit translation and decrease mRNA stability by binding to regulatory motifs mostly located in the 3'UTR of target mRNAs conserved throughout evolution. MYB is an evolutionarily conserved miR-150 target experimentally validated in mice, humans and zebrafish. However, the functional miR-150 sites of humans and mice are orthologous, whereas that of zebrafish is different. RESULTS We identified the avian mature miRNA-150-5P, Gallus gallus gga-miR-150 from chicken leukocyte small-RNA libraries and showed that, as expected, the gga-miR-150 sequence was highly conserved, including the seed region sequence present in the other miR-150 sequences listed in miRBase. Reporter assays showed that gga-miR-150 acted on the avian MYB 3'UTR and identified the avian MYB target site involved in gga-miR-150 binding. A comparative in silico analysis of the miR-150 target sites of MYB 3'UTRs from different species led to the identification of a single set of putative target sites in amphibians and zebrafish, whereas two sets of putative target sites were identified in chicken and mammals. However, only the target site present in the chicken MYB 3'UTR that was identical to that in zebrafish was functional, despite the additional presence of mammalian target sites in chicken. This specific miR-150 site usage was not cell-type specific and persisted when the chicken c-myb 3'UTR was used in the cell system to identify mammalian target sites, showing that this miR-150 target site usage was intrinsic to the chicken c-myb 3'UTR. CONCLUSION Our study of the avian MYB/gga-miR-150 interaction shows a conservation of miR-150 target site functionality between chicken and zebrafish that does not extend to mammals.
Collapse
Affiliation(s)
- Audrey Guillon-Munos
- Université François Rabelais, Equipe Transcription, Lymphome Viro-induit, UFR Sciences et Techniques, Parc de Grandmont, F-37200 Tours, France
| | | | | | | | | | | |
Collapse
|
23
|
Mitochondrial Hep27 is a c-Myb target gene that inhibits Mdm2 and stabilizes p53. Mol Cell Biol 2010; 30:3981-93. [PMID: 20547751 DOI: 10.1128/mcb.01284-09] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The ever-expanding knowledge of the role of p53 in cellular metabolism, apoptosis, and cell cycle control has led to increasing interest in defining the stress response pathways that regulate Mdm2. In an effort to identify novel Mdm2 binding partners, we performed a large-scale immunoprecipitation of Mdm2 in the osteosarcoma U2OS cell line. One significant binding protein identified was Hep27, a member of the short-chain alcohol dehydrogenase/reductase (SDR) family of enzymes. Here, we demonstrate that the Hep27 preprotein contains an N-terminal mitochondrial targeting signal that is cleaved following mitochondrial import, resulting in mitochondrial matrix accumulation of mature Hep27. A fraction of the mitochondrial Hep27 translocates to the nucleus, where it binds to Mdm2 in the central domain, resulting in the attenuation of Mdm2-mediated p53 degradation. In addition, Hep27 is regulated at the transcriptional level by the proto-oncogene c-Myb and is required for c-Myb-induced p53 stabilization. Breast cancer gene expression analysis correlated estrogen receptor (ER) status with Hep27 expression and p53 function, providing a potential in vivo link between estrogen receptor signaling and p53 activity. Our data demonstrate a unique c-Myb-Hep27-Mdm2-p53 mitochondria-to-nucleus signaling pathway that may have functional significance for ER-positive breast cancers.
Collapse
|
24
|
Mann J, Mann DA. Transcriptional regulation of hepatic stellate cells. Adv Drug Deliv Rev 2009; 61:497-512. [PMID: 19393271 DOI: 10.1016/j.addr.2009.03.011] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2008] [Accepted: 03/10/2009] [Indexed: 02/08/2023]
Abstract
Hepatic stellate cell (HSC) activation is a process of cellular transdifferentiation in which, upon liver injury, the quiescent vitamin A storing perisinusoidal HSC is converted into a wound-healing myofibroblast and acquires potent pro-inflammatory and pro-fibrogenic activities. This remarkable phenotypic transformation is underpinned by changes in the expression of a vast number of genes. In this review we survey current knowledge of the transcription factors that either control HSC activation or which regulate specific fibrogenic functions of the activated HSC such as collagen expression, proliferation and resistance to apoptosis.
Collapse
|
25
|
Polymorphisms of sepiapterin reductase gene alter promoter activity and may influence risk of bipolar disorder. Pharmacogenet Genomics 2009; 19:330-7. [PMID: 19415819 DOI: 10.1097/fpc.0b013e328328f82c] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
OBJECTIVES In a previous investigation, we observed altered expression of sepiapterin reductase (SPR) in cultured neural cells chronically exposed to paroxetine. SPR is an enzyme, which catalyzes the final step in the synthesis of tetrahydrobiopterin (BH4). BH4 is an essential cofactor for synthesis of many neurotransmitters including serotonin. Given the pivotal role of SPR in neurotransmitter production, we sought to test the hypothesis that SPR would influence susceptibility to mood disorders and patient response to antidepressants. METHODS We tested for association of SPR promoter polymorphisms with antidepressant response in a well-characterized triad cohort of mood disorders. We evaluated the functional effect of these variants using the Dual-Luciferase Reporter Gene Assay System in two independent cell lines. RESULTS Two promoter single nucleotide polymorphisms (rs1876487 and rs2421095) in SPR were identified that occurred in three distinct haplotypes. We found a statistically significant association of haplotype pair 2,3 with bipolar I disorder [odds ratio: 5.47; 95% confidence interval: (1.68-17.88); P<0.005] and the personality measure self-transcendence (P = 0.020). Moreover, we found preliminary evidence that individuals with haplotype pair 2,3 responded better to the treatment with selective serotonin reuptake inhibitors. Reporter gene assays revealed a 1.4-fold to 1.6-fold decrease in the transcription rate of the two less common haplotypes (2 and 3) compared with haplotype 1, in the two cell lines investigated. CONCLUSION This reduced transcription rate for SPR promoter haplotypes 2 and 3 may impact on BH4-mediated neurotransmitter production, thus suggesting a biological process through which SPR gene variants might influence antidepressant response and susceptibility to bipolar disorder.
Collapse
|
26
|
Williams BB, Wall M, Miao RY, Williams B, Bertoncello I, Kershaw MH, Mantamadiotis T, Haber M, Norris MD, Gautam A, Darcy PK, Ramsay RG. Induction of T cell-mediated immunity using a c-Myb DNA vaccine in a mouse model of colon cancer. Cancer Immunol Immunother 2008; 57:1635-45. [PMID: 18386000 PMCID: PMC11030567 DOI: 10.1007/s00262-008-0497-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2007] [Accepted: 02/26/2008] [Indexed: 10/22/2022]
Abstract
Overexpression of the proto-oncogene c-Myb occurs in more than 80% of colorectal cancer (CRC) and is associated with aggressive disease and poor prognosis. To test c-Myb as a therapeutic target in CRC we devised a DNA fusion vaccine to generate an anti-CRC immune response. c-Myb, like many tumor antigens, is weakly immunogenic as it is a "self" antigen and subject to tolerance. To break tolerance, a DNA fusion vaccine was generated comprising wild-type c-Myb cDNA flanked by two potent Th epitopes derived from tetanus toxin. Vaccination was performed targeting a highly aggressive, weakly immunogenic, subcutaneous, syngeneic, colon adenocarcinoma cell line MC38 which highly expresses c-Myb. Prophylactic intravenous vaccination significantly suppressed tumor growth, through the induction of anti-tumor immunity for which the tetanus epitopes were essential. Vaccination generated anti-tumor immunity mediated by both CD4+ and CD8+ T cells and increased infiltration of immune effector cells at the tumor site. Importantly, no evidence of autoimmune pathology in endogenous c-Myb expressing tissues was detected as a consequence of breaking tolerance. In summary, these results establish c-Myb as a potential antigen for immune targeting in CRC and serve to provide proof of principle for the continuing development of DNA vaccines targeting c-Myb to bring this approach to the clinic.
Collapse
MESH Headings
- Adenocarcinoma/genetics
- Adenocarcinoma/immunology
- Adenocarcinoma/therapy
- Animals
- Base Sequence
- Blotting, Western
- Bone Marrow/immunology
- Bone Marrow/metabolism
- Cancer Vaccines/genetics
- Cancer Vaccines/immunology
- Cancer Vaccines/therapeutic use
- Colonic Neoplasms/genetics
- Colonic Neoplasms/immunology
- Colonic Neoplasms/therapy
- Disease Models, Animal
- Female
- Flow Cytometry
- Genes, MHC Class I/physiology
- Genes, myb/genetics
- Green Fluorescent Proteins/genetics
- Humans
- Immunity
- Lymphocyte Activation
- Lymphocytes, Tumor-Infiltrating/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Molecular Sequence Data
- Peptide Fragments/immunology
- Proto-Oncogene Mas
- Stem Cells/cytology
- Stem Cells/immunology
- Stem Cells/metabolism
- Survival Rate
- T-Lymphocytes/immunology
- T-Lymphocytes/pathology
- T-Lymphocytes, Cytotoxic/immunology
- Tetanus Toxin/genetics
- Tetanus Toxin/immunology
- Tumor Cells, Cultured
- Vaccination
- Vaccines, DNA/therapeutic use
Collapse
|
27
|
Gonda TJ, Leo P, Ramsay RG. Estrogen and MYB in breast cancer: potential for new therapies. Expert Opin Biol Ther 2008; 8:713-7. [PMID: 18476782 DOI: 10.1517/14712598.8.6.713] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
MYB is highly expressed in almost all estrogen receptor (ER)-positive breast tumours and is a direct target of estrogen/ER signalling. Our recent studies have shown that MYB is also required for the proliferation of ER-positive breast tumour cell lines, and have shed further light on the mechanism of ER regulation of MYB expression. Here we discuss the rationale for therapeutic targeting of MYB in breast cancer and consider a number of approaches to developing an anti-MYB therapeutic.
Collapse
Affiliation(s)
- Thomas J Gonda
- Professorial Research Fellow, Deputy Director and Head Cancer Biology Program, University of Queensland Diamantina Institute for Cancer, Immunology and Metabolic Medicine, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia.
| | | | | |
Collapse
|
28
|
Genome profiling of acute myelomonocytic leukemia: alteration of the MYB locus in MYST3-linked cases. Leukemia 2008; 23:85-94. [DOI: 10.1038/leu.2008.257] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
29
|
Barroga CF, Pham H, Kaushansky K. Thrombopoietin regulates c-Myb expression by modulating micro RNA 150 expression. Exp Hematol 2008; 36:1585-92. [PMID: 18814950 DOI: 10.1016/j.exphem.2008.07.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2007] [Revised: 07/15/2008] [Accepted: 07/17/2008] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Mice harboring c-Myb hypomorphic mutations display enhanced thrombopoiesis because of increased numbers of megakaryocytes and their progenitors. Thrombopoietin induces these same effects, which lead us to hypothesize that the hormone acts through modulation of c-Myb expression, as c-Myb levels falls during thrombopoietin-induced megakaryocyte (MK) maturation. Micro RNAs (miRs) downregulate gene expression by binding to the 3' untranslated region (UTR) of specific messenger RNAs (mRNAs); we noted that the 3'UTR of c-Myb contains four miR-150 binding sites. MATERIALS AND METHODS We used quantitative reverse transcriptase polymerase chain reaction, Western blotting, and reporter gene analyses to assess the response of c-Myb to thrombopoietin stimulation and to gain of and loss of miR-150 expression. RESULTS We found that thrombopoietin reduced c-Myb mRNA and protein levels within 7 hours in megakaryocytes and UT7/thrombopoietin (TPO) cells. Using a reporter gene containing the c-Myb 3'UTR region, including its four miR150 binding sites, we found that expression of miR150 reduced luciferase expression to 50% of baseline at 24 hours and to 25% at 48 hours in UT7/TPO cells. Quantitative polymerase chain reaction and Western blotting also revealed that miR-150 reduced endogenous c-Myb mRNA and protein to 50% in UT7/TPO cells, and to 65% in mature megakaryocytes. Converse experiments utilizing anti-miR150 increased luciferase activity twofold over control anti-miR. Finally, TPO increased miR150 expression 1.8-fold within 24 hours and 3.4-fold within 48 hours. CONCLUSIONS These findings establish that miR150 downmodulates c-Myb levels, and because TPO affects miR150 expression, our results indicate that, in addition to affecting MK progenitor cell growth, TPO downmodulates c-Myb expression through induction of miR-150.
Collapse
Affiliation(s)
- Charlene F Barroga
- Department of Medicine and Division of Hematology/Oncology, University of California, San Diego School of Medicine, San Diego, CA 92103-8811, USA
| | | | | |
Collapse
|
30
|
Huang YC, Su LH, Lee GA, Chiu PW, Cho CC, Wu JY, Sun CH. Regulation of cyst wall protein promoters by Myb2 in Giardia lamblia. J Biol Chem 2008; 283:31021-9. [PMID: 18768462 DOI: 10.1074/jbc.m805023200] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Myb family transcription factors are important in regulating cell proliferation, differentiation, and cell cycle progression. Giardia lamblia differentiates into infectious cysts to survive outside of the host. During encystation, genes encoding cyst wall proteins (CWPs) are coordinately induced. We have identified an encystation-induced Myb2 protein, which binds to the promoter regions of the cwp genes and myb2 itself in vitro. To elucidate the role of Myb2 in G. lamblia, we tested the hypothesis that Myb2 can activate encystation-induced genes. We found that overexpression of Myb2 resulted in an increase of expression of CWP1 at both protein and mRNA levels. Interestingly, the Myb2-overexpressing trophozoites had increased capability to differentiate into cysts. In cotransfection assays, Myb2 was able to transactivate the cwp promoters and its own promoter in vivo, suggesting that its gene can be positively autoregulated. Moreover, deletion of the N- or C-terminal domain resulted in a decrease of transactivation and autoregulation function of Myb2. We also found that the promoter of a newly identified encystation-induced gene, the giardial myeloid leukemia factor-like gene, has the Myb2 binding sites and that its mRNA levels were increased by Myb2 overexpression. Chromatin immunoprecipitation assays confirmed that Myb2 was bound to the promoters with its binding sites. Transfection of the myb2 antisense construct reduced the levels of the cwp1 transcripts and cyst formation. Our results suggest that Myb2 is a potent transactivator of the cwp genes and other endogenous genes and plays an important role in G. lamblia differentiation into cysts.
Collapse
Affiliation(s)
- Yu-Chang Huang
- Department of Parasitology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
The transcription factor MYB has a key role as a regulator of stem and progenitor cells in the bone marrow, colonic crypts and a neurogenic region of the adult brain. It is in these compartments that a deficit in MYB activity leads to severe or lethal phenotypes. As was predicted from its leukaemogenicity in several animal species, MYB has now been identified as an oncogene that is involved in some human leukaemias. Moreover, recent evidence has strengthened the case that MYB is activated in colon and breast cancer: a block to MYB expression is overcome by mutation of the regulatory machinery in the former disease and by oestrogen receptor-alpha (ERalpha) in the latter.
Collapse
Affiliation(s)
- Robert G Ramsay
- Peter MacCallum Cancer Centre, St Andrew's Place, Melbourne, Victoria 3002, Australia
| | | |
Collapse
|
32
|
Greig KT, Carotta S, Nutt SL. Critical roles for c-Myb in hematopoietic progenitor cells. Semin Immunol 2008; 20:247-56. [PMID: 18585056 DOI: 10.1016/j.smim.2008.05.003] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2007] [Accepted: 05/14/2008] [Indexed: 11/16/2022]
Abstract
While it has long been known that the transcription factor c-Myb is an essential regulator of hematopoiesis, its precise molecular targets have remained elusive. Cell line studies suggest that c-Myb promotes proliferation and at the same time inhibits differentiation, however the early lethality of c-Myb deficient embryos precluded analysis of its role in adult hematopoiesis. Here we review insights derived from recently developed mouse models of c-Myb deficiency that are viable as adults. These studies reveal a complex array of functions for c-Myb in multiple hematopoietic cell types that will redefine our understanding of this crucial transcription factor.
Collapse
Affiliation(s)
- Kylie T Greig
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3050, Australia.
| | | | | |
Collapse
|
33
|
Ramsay RG, Malaterre J. Insights into c-Myb functions through investigating colonic crypts. Blood Cells Mol Dis 2007; 39:287-91. [PMID: 17659914 DOI: 10.1016/j.bcmd.2007.05.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2007] [Accepted: 05/21/2007] [Indexed: 10/23/2022]
Abstract
c-Myb has been investigated in the context of the hematopoietic system where it has been shown to regulate progenitor cell expansion and differentiation of a number of lineages. The capacity to grow and expand specific blood cell lineages in vitro using well defined growth factors plus the vast range of cell surface lineage markers that identify different cell types has driven our understanding of the spectrum of roles that c-Myb plays in this tissue compartment. In addition, c-Myb is also an important transcription factor in non-hematopoietic tissues but the restricted spectrum of cell phenotyping reagents has hampered in-depth investigation. In the case of the colonic crypt the absence of phenotyping reagents of the quality employed in identifying blood cell lineages is partly compensated for by the spatial and temporal information that is inherent in the crypt structure. Using different tools to those used in the blood system we have gained insights in the multiple roles played by c-Myb in colon epithelial cells. These observations, when combined with the understanding of c-Myb action in blood cells, is providing a clearer view as to how c-Myb operates in normal cells and how this is subverted in diseases like cancer.
Collapse
Affiliation(s)
- Robert G Ramsay
- Peter MacCallum Cancer Centre, East Melbourne and Department of Pathology, The University of Melbourne, Parkville, Australia.
| | | |
Collapse
|
34
|
Kaspar P, Dvorák M. Involvement of phosphatidylserine externalization in the down-regulation of c-myb expression in differentiating C2C12 cells. Differentiation 2007; 76:245-52. [PMID: 17924964 DOI: 10.1111/j.1432-0436.2007.00222.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Analysis of c-myb gene down-regulation in differentiating C212 cells revealed that in proliferating cells, c-myb expression is high and ceases as the proliferation rate decreases. However, a low level of c-myb mRNA was detected in confluent non-proliferating differentiating cells for an extended period of time before it declined to an undetectable level. The time course of c-myb gene silencing in differentiating cells correlated with exposition of phosphatidylserine (PS) on the cell surface. Moreover, the interaction of exposed PS with exogenously added annexin V perturbed PS-mediated cell signaling and transiently up-regulated the declining c-myb expression. We, therefore, suggest that cell surface-exposed PS, which plays a role in the process of myotube formation, is also involved in the down-regulation of c-myb expression.
Collapse
Affiliation(s)
- Petr Kaspar
- Institute of Molecular Genetics, AS CR v.v.i., Vídenská 1083, Prague 4, CZ-14220, Czech Republic.
| | | |
Collapse
|
35
|
Malaterre J, Carpinelli M, Ernst M, Alexander W, Cooke M, Sutton S, Dworkin S, Heath JK, Frampton J, McArthur G, Clevers H, Hilton D, Mantamadiotis T, Ramsay RG. c-Myb is required for progenitor cell homeostasis in colonic crypts. Proc Natl Acad Sci U S A 2007; 104:3829-34. [PMID: 17360438 PMCID: PMC1820669 DOI: 10.1073/pnas.0610055104] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The colonic crypt is the functional unit of the colon mucosa with a central role in ion and water reabsorption. Under steady-state conditions, the distal colonic crypt harbors a single stem cell at its base that gives rise to highly proliferative progenitor cells that differentiate into columnar, goblet, and endocrine cells. The role of c-Myb in crypt homeostasis has not been elucidated. Here we have studied three genetically distinct hypomorphic c-myb mutant mouse strains, all of which show reduced colonic crypt size. The mutations target the key domains of the transcription factor: the DNA binding, transactivation, and negative regulatory domains. In vivo proliferation and cell cycle marker studies suggest that these mice have a progenitor cell proliferation defect mediated in part by reduced Cyclin E1 expression. To independently assess the extent to which c-myb is required for colonic crypt homeostasis we also generated a novel tissue-specific mouse model to allow the deletion of c-myb in adult colon, and using these mice we show that c-Myb is required for crypt integrity, normal differentiation, and steady-state proliferation.
Collapse
Affiliation(s)
- Jordane Malaterre
- *Peter MacCallum Cancer Centre and Pathology Department, University of Melbourne, Melbourne VIC 8006, Australia
| | - Marina Carpinelli
- The Walter and Eliza Hall Institute for Medical Research, Parkville VIC 3050, Australia
| | - Matthias Ernst
- Tumour Biology Branch, Ludwig Institute for Cancer Research, Parkville VIC 3050, Australia
| | - Warren Alexander
- The Walter and Eliza Hall Institute for Medical Research, Parkville VIC 3050, Australia
| | - Michael Cooke
- Genomics Institute, Institute for Biomedical Research, San Diego, CA 92121
| | - Susan Sutton
- Genomics Institute, Institute for Biomedical Research, San Diego, CA 92121
| | - Sebastian Dworkin
- *Peter MacCallum Cancer Centre and Pathology Department, University of Melbourne, Melbourne VIC 8006, Australia
| | - Joan K. Heath
- Tumour Biology Branch, Ludwig Institute for Cancer Research, Parkville VIC 3050, Australia
| | - Jon Frampton
- Medical School, Birmingham University, Edgbaston, Birmingham B15 2TT, United Kingdom; and
| | - Grant McArthur
- *Peter MacCallum Cancer Centre and Pathology Department, University of Melbourne, Melbourne VIC 8006, Australia
| | - Hans Clevers
- Hubrecht Laboratory, 3584 CT, Utrecht, The Netherlands
| | - Douglas Hilton
- The Walter and Eliza Hall Institute for Medical Research, Parkville VIC 3050, Australia
| | - Theo Mantamadiotis
- *Peter MacCallum Cancer Centre and Pathology Department, University of Melbourne, Melbourne VIC 8006, Australia
| | - Robert G. Ramsay
- *Peter MacCallum Cancer Centre and Pathology Department, University of Melbourne, Melbourne VIC 8006, Australia
- **To whom correspondence should be addressed. E-mail:
| |
Collapse
|
36
|
[ST1]: Proto‐oncogene c‐Myb regulates neural progenitor proliferation in adult brain. Int J Dev Neurosci 2006. [DOI: 10.1016/j.ijdevneu.2006.09.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|