1
|
Lok JB. CRISPR/Cas9 Mutagenesis and Expression of Dominant Mutant Transgenes as Functional Genomic Approaches in Parasitic Nematodes. Front Genet 2019; 10:656. [PMID: 31379923 PMCID: PMC6646703 DOI: 10.3389/fgene.2019.00656] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 06/21/2019] [Indexed: 12/13/2022] Open
Abstract
DNA transformation of parasitic nematodes enables novel approaches to validating predictions from genomic and transcriptomic studies of these important pathogens. Notably, proof of principle for CRISPR/Cas9 mutagenesis has been achieved in Strongyloides spp., allowing identification of molecules essential to the functions of sensory neurons that mediate behaviors comprising host finding, invasion, and location of predilection sites by parasitic nematodes. Likewise, CRISPR/Cas9 knockout of the developmental regulatory transcription factor Ss-daf-16 has validated its function in regulating morphogenesis of infective third-stage larvae in Strongyloides stercoralis. While encouraging, these studies underscore challenges that remain in achieving straightforward validation of essential intervention targets in parasitic nematodes. Chief among these is the likelihood that knockout of multifunctional regulators like Ss-DAF-16 or its downstream mediator, the nuclear receptor Ss-DAF-12, will produce phenotypes so complex as to defy interpretation and will render affected worms incapable of infecting their hosts, thus preventing establishment of stable mutant lines. Approaches to overcoming these impediments could involve refinements to current CRISPR/Cas9 methods in Strongyloides including regulatable Cas9 expression from integrated transgenes and CRISPR/Cas9 editing to ablate specific functional motifs in regulatory molecules without complete knockout. Another approach would express transgenes encoding regulatory molecules of interest with mutations designed to similarly ablate or degrade specific functional motifs such as the ligand binding domain of Ss-DAF-12 while preserving core functions such as DNA binding. Such mutant transgenes would be expected to exert a dominant interfering effect on their endogenous counterparts. Published reports validate the utility of such dominant-negative approaches in Strongyloides.
Collapse
Affiliation(s)
- James B Lok
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
2
|
Chimeric NANOG repressors inhibit glioblastoma growth in vivo in a context-dependent manner. Sci Rep 2019; 9:3891. [PMID: 30846719 PMCID: PMC6405761 DOI: 10.1038/s41598-019-39473-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 01/25/2019] [Indexed: 01/02/2023] Open
Abstract
Targeting stemness promises new therapeutic strategies against highly invasive tumors. While a number of approaches are being tested, inhibiting the core transcription regulatory network of cancer stem cells is an attractive yet challenging possibility. Here we have aimed to provide the proof of principle for a strategy, previously used in developmental studies, to directly repress the targets of a salient stemness and pluripotency factor: NANOG. In doing so we expected to inhibit the expression of so far unknown mediators of pro-tumorigenic NANOG function. We chose NANOG since previous work showed the essential requirement for NANOG activity for human glioblastoma (GBM) growth in orthotopic xenografts, and it is apparently absent from many adult human tissues thus likely minimizing unwanted effects on normal cells. NANOG repressor chimeras, which we name NANEPs, bear the DNA-binding specificity of NANOG through its homeodomain (HD), and this is linked to transposable human repressor domains. We show that in vitro and in vivo, NANEP5, our most active NANEP with a HES1 repressor domain, mimics knock-down (kd) of NANOG function in GBM cells. Competition orthotopic xenografts also reveal the effectiveness of NANEP5 in a brain tumor context, as well as the specificity of NANEP activity through the abrogation of its function via the introduction of specific mutations in the HD. The transcriptomes of cells expressing NANEP5 reveal multiple potential mediators of pro-tumorigenic NANEP/NANOG action including intercellular signaling components. The present results encourage further studies on the regulation of context-dependent NANEP abundance and function, and the development of NANEP-based anti-cancer therapies.
Collapse
|
3
|
Xenopus SOX5 enhances myogenic transcription indirectly through transrepression. Dev Biol 2018; 442:262-275. [PMID: 30071218 DOI: 10.1016/j.ydbio.2018.07.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 07/16/2018] [Accepted: 07/28/2018] [Indexed: 02/06/2023]
Abstract
In anamniotes, somite compartimentalization in the lateral somitic domain leads simultaneously to myotome and dermomyotome formation. In the myotome, Xenopus Sox5 is co-expressed with Myod1 in the course of myogenic differentiation. Here, we studied the function of Sox5 using a Myod1-induced myogenic transcription assay in pluripotent cells of animal caps. We found that Sox5 enhances myogenic transcription of muscle markers Des, Actc1, Ckm and MyhE3. The use of chimeric transactivating or transrepressive Sox5 proteins indicates that Sox5 acts as a transrepressor and indirectly stimulates myogenic transcription except for the slow muscle-specific genes Myh7L, Myh7S, Myl2 and Tnnc1. We showed that this role is shared by Sox6, which is structurally similar to Sox5, both belonging to the SoxD subfamily of transcription factors. Moreover, Sox5 can antagonize the inhibitory function of Meox2 on myogenic differentiation. Meox2 which is a dermomyotome marker, represses myogenic transcription in Myod-induced myogenic transcription assay and in Nodal5-induced mesoderm from animal cap assay. The inhibitory function of Meox2 and the pro-myogenic function of Sox5 were confirmed during Xenopus normal development by the use of translation-blocking oligomorpholinos and dexamethasone inducible chimeric Sox5 and Meox2 proteins. We have therefore identified a new function for SoxD proteins in muscle cells, which can indirectly enhance myogenic transcription through transrepression, in addition to the previously identified function as a direct repressor of slow muscle-specific genes.
Collapse
|
4
|
Hif-1α and Hif-2α regulate hemogenic endothelium and hematopoietic stem cell formation in zebrafish. Blood 2018; 131:963-973. [PMID: 29339404 DOI: 10.1182/blood-2017-07-797795] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 01/05/2018] [Indexed: 12/18/2022] Open
Abstract
During development, hematopoietic stem cells (HSCs) derive from specialized endothelial cells (ECs) called hemogenic endothelium (HE) via a process called endothelial-to-hematopoietic transition (EHT). Hypoxia-inducible factor-1α (HIF-1α) has been reported to positively modulate EHT in vivo, but current data indicate the existence of other regulators of this process. Here we show that in zebrafish, Hif-2α also positively modulates HSC formation. Specifically, HSC marker gene expression is strongly decreased in hif-1aa;hif-1ab (hif-1α) and in hif-2aa;hif-2ab (hif-2α) zebrafish mutants and morphants. Moreover, live imaging studies reveal a positive role for hif-1α and hif-2α in regulating HE specification. Knockdown of hif-2α in hif-1α mutants leads to a greater decrease in HSC formation, indicating that hif-1α and hif-2α have partially overlapping roles in EHT. Furthermore, hypoxic conditions, which strongly stimulate HSC formation in wild-type animals, have little effect in the combined absence of Hif-1α and Hif-2α function. In addition, we present evidence for Hif and Notch working in the same pathway upstream of EHT. Both notch1a and notch1b mutants display impaired EHT, which cannot be rescued by hypoxia. However, overexpression of the Notch intracellular domain in ECs is sufficient to rescue the hif-1α and hif-2α morphant EHT phenotype, suggesting that Notch signaling functions downstream of the Hif pathway during HSC formation. Altogether, our data provide genetic evidence that both Hif-1α and Hif-2α regulate EHT upstream of Notch signaling.
Collapse
|
5
|
Zhang J, Shu Y, Qu Y, Zhang L, Chu T, Zheng Y, Zhao H. C-myb Plays an Essential Role in the Protective Function of IGF-1 on Cytotoxicity Induced by Aβ 25-35 via the PI3K/Akt Pathway. J Mol Neurosci 2017; 63:412-418. [PMID: 29110181 DOI: 10.1007/s12031-017-0991-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 10/11/2017] [Indexed: 01/23/2023]
Abstract
There have been numerous reports about neurodegenerative diseases, including Alzheimer's disease. Nevertheless, the molecules responsible for the neurodegeneration in Alzheimer's disease are basically unknown. Recent findings indicate that the cellular myeloblastosis (c-myb) regulates neural progenitor cell proliferation. In the current study, the function of insulin-like growth factor-1 (IGF-1) against cell toxicity in SH-SY5Y cells induced by β-amyloid 25-35 (Aβ25-35) and its molecular mechanism were investigated. It was found that p25 protein production was raised by Aβ25-35 (25 μM), similar to the increased expression of μ-calpain. The results also showed that Aβ25-35 reduced c-myb, elevated tau hyper-phosphorylation, and induced death of SH-SY5Y cells. Loss of cell viability and apoptosis of SH-SY5Y cells induced by Aβ25-35 were attenuated by IGF-1 pretreatment in a dose-dependent manner. In addition, IGF-1 blocked μ-calpain expression, which was induced by Aβ25-35 and reduced p25 formation and tau hyper-phosphorylation. Moreover, the expression of c-myb in SH-SY5Y cells was increased by combining IGF-1 with Aβ25-35 or IGF-1 alone. The neuroprotective function of IGF-1 was attenuated in the SH-SY5Y cells, which were transfected with a c-myb small interfering RNA. Furthermore, LY294002, a specific PI3K inhibitor, reduced c-myb expression and abolished IGF-1's protective function in SH-SY5Y cell apoptosis induced by Aβ25-35. The facts above indicate that c-myb has a role in IGF-1-mediated protection from Aβ25-35-induced cytotoxicity via the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Jingyu Zhang
- Department of Neurology, Fourth Affiliated Hospital of Harbin Medical University, 37 Yiyuan Street, Nangang District, Harbin, Heilongjiang Province, 150001, China
| | - Yongwei Shu
- Department of Neurology, Fourth Affiliated Hospital of Harbin Medical University, 37 Yiyuan Street, Nangang District, Harbin, Heilongjiang Province, 150001, China
| | - Yang Qu
- Department of Neurology, Fourth Affiliated Hospital of Harbin Medical University, 37 Yiyuan Street, Nangang District, Harbin, Heilongjiang Province, 150001, China
| | - Lina Zhang
- Department of Neurology, Fourth Affiliated Hospital of Harbin Medical University, 37 Yiyuan Street, Nangang District, Harbin, Heilongjiang Province, 150001, China
| | - Tingting Chu
- Department of Neurology, Fourth Affiliated Hospital of Harbin Medical University, 37 Yiyuan Street, Nangang District, Harbin, Heilongjiang Province, 150001, China
| | - Yonghui Zheng
- Department of Neurology, Fourth Affiliated Hospital of Harbin Medical University, 37 Yiyuan Street, Nangang District, Harbin, Heilongjiang Province, 150001, China
| | - Hong Zhao
- Department of Neurology, Fourth Affiliated Hospital of Harbin Medical University, 37 Yiyuan Street, Nangang District, Harbin, Heilongjiang Province, 150001, China.
| |
Collapse
|
6
|
Maharry SE, Walker CJ, Liyanarachchi S, Mehta S, Patel M, Bainazar MA, Huang X, Lankenau MA, Hoag KW, Ranganathan P, Garzon R, Blachly JS, Guttridge DC, Bloomfield CD, de la Chapelle A, Eisfeld AK. Dissection of the Major Hematopoietic Quantitative Trait Locus in Chromosome 6q23.3 Identifies miR-3662 as a Player in Hematopoiesis and Acute Myeloid Leukemia. Cancer Discov 2016; 6:1036-51. [PMID: 27354268 DOI: 10.1158/2159-8290.cd-16-0023] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 06/23/2016] [Indexed: 12/14/2022]
Abstract
UNLABELLED Chromosomal aberrations and multiple genome-wide association studies (GWAS) have established a major hematopoietic quantitative trait locus in chromosome 6q23.3. The locus comprises an active enhancer region, in which some of the associated SNPs alter transcription factor binding. We now identify miR-3662 as a new functional driver contributing to the associated phenotypes. The GWAS SNPs are strongly associated with higher miR-3662 expression. Genome editing of rs66650371, a three-base-pair deletion, suggests a functional link between the SNP genotype and the abundance of miR-3662. Increasing miR-3662's abundance increases colony formation in hematopoietic progenitor cells, particularly the erythroid lineage. In contrast, miR-3662 is not expressed in acute myeloid leukemia cells, and its overexpression has potent antileukemic effects in vitro and in vivo Mechanistically, miR-3662 directly targets NF-κB-mediated transcription. Thus, miR-3662 is a new player of the hematopoietic 6q23.3 locus. SIGNIFICANCE The characterization of miR-3662 has identified a new actor in the prominent hematopoietic quantitative trait locus in chromosome 6q23.3. The mechanistic insights into miR-3662's function may reveal novel or only partially known pathways for normal and malignant hematopoietic cell proliferation. Cancer Discov; 6(9); 1036-51. ©2016 AACR.This article is highlighted in the In This Issue feature, p. 932.
Collapse
Affiliation(s)
- Sophia E Maharry
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | | | | | - Sujay Mehta
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Mitra Patel
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Maryam A Bainazar
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Xiaomeng Huang
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Malori A Lankenau
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Kevin W Hoag
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | | | - Ramiro Garzon
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - James S Blachly
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Denis C Guttridge
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | | | | | | |
Collapse
|
7
|
Abstract
The growth of axons is an intricately regulated process involving intracellular signaling cascades and gene transcription. We had previously shown that the stimulus-dependent transcription factor, serum response factor (SRF), plays a critical role in regulating axon growth in the mammalian brain. However, the molecular mechanisms underlying SRF-dependent axon growth remains unknown. Here we report that SRF is phosphorylated and activated by GSK-3 to promote axon outgrowth in mouse hippocampal neurons. GSK-3 binds to and directly phosphorylates SRF on a highly conserved serine residue. This serine phosphorylation is necessary for SRF activity and for its interaction with MKL-family cofactors, MKL1 and MKL2, but not with TCF-family cofactor, ELK-1. Axonal growth deficits caused by GSK-3 inhibition could be rescued by expression of a constitutively active SRF. The SRF target gene and actin-binding protein, vinculin, is sufficient to overcome the axonal growth deficits of SRF-deficient and GSK-3-inhibited neurons. Furthermore, short hairpin RNA-mediated knockdown of vinculin also attenuated axonal growth. Thus, our findings reveal a novel phosphorylation and activation of SRF by GSK-3 that is critical for SRF-dependent axon growth in mammalian central neurons.
Collapse
|
8
|
cMyb regulates hematopoietic stem/progenitor cell mobilization during zebrafish hematopoiesis. Blood 2011; 118:4093-101. [PMID: 21856868 DOI: 10.1182/blood-2011-03-342501] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The establishment of the HSC pool in vertebrates depends not only on the formation and the propagation of these stem cells but also on their proper trafficking among the defined hematopoietic organs. However, the physiologic mechanisms that regulate HSC mobilization remain elusive. Through analysis of the zebrafish cmyb mutant cmyb(hkz3), we show that the suppression of cMyb function abrogates larval and adult hematopoiesis, with concomitant accumulation of hematopoietic stem/progenitor cells (HSPCs) in their birthplace, the ventral wall of the dorsal aorta (VDA). Cell tracking and time-lapse recording reveal that the accumulation of HSPCs in cmyb(hkz3) mutants is caused by the impairment of HSPC egression from the VDA. Further analysis demonstrates that the HSPC migratory defects in cmyb(hkz3) mutants are at least partly because of adversely elevated levels of chemokine stromal cell-derived factor 1a (Sdf1a). Our study reveals that cMyb plays a hitherto unidentified role in dictating physiologic HSPC migration by modulating Sdf1a signaling.
Collapse
|
9
|
Marikawa Y, Tamashiro DAA, Fujita TC, Alarcon VB. Dual roles of Oct4 in the maintenance of mouse P19 embryonal carcinoma cells: as negative regulator of Wnt/β-catenin signaling and competence provider for Brachyury induction. Stem Cells Dev 2011; 20:621-33. [PMID: 21083502 DOI: 10.1089/scd.2010.0209] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Transcription factor Oct4 is expressed in pluripotent cell lineages during mouse development, namely, in inner cell mass (ICM), primitive ectoderm, and primordial germ cells. Functional studies have revealed that Oct4 is essential for the maintenance of pluripotency in inner cell mass and for the survival of primordial germ cells. However, the function of Oct4 in the primitive ectoderm has not been fully explored. In this study, we investigated the role of Oct4 in mouse P19 embryonal carcinoma (EC) cells, which exhibit molecular and developmental properties similar to the primitive ectoderm, as an in vitro model. Knockdown of Oct4 in P19 EC cells upregulated several early mesoderm-specific genes, such as Wnt3, Sp5, and Fgf8, by activating Wnt/β-catenin signaling. Overexpression of Oct4 was sufficient to suppress Wnt/β-catenin signaling through its action as a transcriptional activator. However, Brachyury, a key regulator of early mesoderm development and a known direct target of Wnt/β-catenin signaling, was unable to be upregulated in the absence of Oct4, even with additional activation of Wnt/β-catenin signaling. Microarray analysis revealed that Oct4 positively regulated the expression of Tdgf1, a critical component of Nodal signaling, which was required for the upregulation of Brachyury in response to Wnt/β-catenin signaling in P19 EC cells. We propose a model that Oct4 maintains pluripotency of P19 EC cells through 2 counteracting actions: one is to suppress mesoderm-inducing Wnt/β-catenin signaling, and the other is to provide competence to Brachyury gene to respond to Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Yusuke Marikawa
- Department of Anatomy, Biochemistry, and Physiology, Institute for Biogenesis Research, University of Hawaii John A. Burns School of Medicine, Honolulu, HI 96813, USA.
| | | | | | | |
Collapse
|
10
|
|
11
|
Conditional c-myb knockout in adult hematopoietic stem cells leads to loss of self-renewal due to impaired proliferation and accelerated differentiation. Proc Natl Acad Sci U S A 2009; 106:21689-94. [PMID: 19955420 DOI: 10.1073/pnas.0907623106] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Hematopoietic stem cells (HSCs) have a unique capacity to undergo self-renewal and multi-lineage differentiation to provide a lifetime supply of mature blood cells. By using conditional knockout technology, we disrupted the c-myb proto-oncogene specifically in adult bone marrow (BM) to demonstrate that this transcription factor is a regulator of self-renewal and multi-lineage differentiation of adult HSCs. Targeted disruption of the c-myb gene resulted in a critical depletion of the HSC pool. In addition, BM hematopoiesis in adult mice was impaired, resulting in profound reductions of various hematopoietic lineages including neutrophilic, monocytic, B lymphoid, erythroid, and, unexpectedly, megakaryocytic cells. Serial BM transplantation into lethally irradiated recipient mice indicated an essential role for c-myb in the self-renewal process. Furthermore, in vitro functional assays demonstrated that deletion of the c-myb gene leads to a slightly reduced proliferative capacity and an aberrant and accelerated differentiation of HSCs. In addition to long-term HSCs, functional studies also show that c-myb plays a critical role in short-term HSCs and multi-potential progenitors. Collectively, our data indicate a critical role for c-myb in adult BM hematopoiesis and in self-renewal and multi-lineage differentiation of adult HSCs.
Collapse
|
12
|
Functional mode of FoxD1/CBF2 for the establishment of temporal retinal specificity in the developing chick retina. Dev Biol 2009; 331:300-10. [PMID: 19450575 DOI: 10.1016/j.ydbio.2009.05.549] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2009] [Revised: 05/12/2009] [Accepted: 05/12/2009] [Indexed: 12/13/2022]
Abstract
Two winged-helix transcription factors, FoxG1 (previously called chick brain factor1, CBF1) and FoxD1 (chick brain factor2, CBF2), are expressed specifically in the nasal and temporal regions of the developing chick retina, respectively. We previously demonstrated that FoxG1 controls the expression of topographic molecules including FoxD1, and determines the regional specificity of the nasal retina. FoxD1 is known to prescribe temporal specificity, however, molecular mechanisms and downstream targets have not been elucidated. Here we addressed the genetic mechanisms for establishing temporal specificity in the developing retina using an in ovo electroporation technique. Fibroblast growth factor (Fgf) and Wnt first play pivotal roles in inducing the region-specific expression of FoxG1 and FoxD1 in the optic vesicle. Misexpression of FoxD1 represses the expression of FoxG1, GH6, SOHo1, and ephrin-A5, and induces that of EphA3 in the retina. GH6 and SOHo1 repress the expression of FoxD1. In contrast to the inhibitory effect of FoxG1 on bone morphogenic protein (BMP) signaling, FoxD1 does not alter the expression of BMP4 or BMP2. Studies with chimeric mutants of FoxD1 showed that FoxD1 acts as a transcription repressor in controlling its downstream targets in the retina. Taken together with previous findings, our data suggest that FoxG1 and FoxD1 are located at the top of the gene cascade for regional specification along the nasotemporal (anteroposterior) axis in the retina, and FoxD1 determines temporal specificity.
Collapse
|
13
|
Rogers CD, Harafuji N, Archer T, Cunningham DD, Casey ES. Xenopus Sox3 activates sox2 and geminin and indirectly represses Xvent2 expression to induce neural progenitor formation at the expense of non-neural ectodermal derivatives. Mech Dev 2008; 126:42-55. [PMID: 18992330 DOI: 10.1016/j.mod.2008.10.005] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2008] [Revised: 09/01/2008] [Accepted: 10/12/2008] [Indexed: 11/28/2022]
Abstract
The SRY-related, HMG box SoxB1 transcription factors are highly homologous, evolutionarily conserved proteins that are expressed in neuroepithelial cells throughout neural development. SoxB1 genes are down-regulated as cells exit the cell-cycle to differentiate and are considered functionally redundant in maintaining neural precursor populations. However, little is known about Sox3 function and its mode of action during primary neurogenesis. Using gain and loss-of-function studies, we analyzed Sox3 function in detail in Xenopus early neural development and compared it to that of Sox2. Through these studies we identified the first targets of a SoxB1 protein during primary neurogenesis. Sox3 functions as an activator to induce expression of the early neural genes, sox2 and geminin in the absence of protein synthesis and to indirectly inhibit the Bmp target Xvent2. As a result, Sox3 increases cell proliferation, delays neurogenesis and inhibits epidermal and neural crest formation to expand the neural plate. Our studies indicate that Sox3 and 2 have many similar functions in this process including the ability to activate expression of geminin in naïve ectodermal explants. However, there are some differences; Sox3 activates the expression of sox2, while Sox2 does not activate expression of sox3 and sox3 is uniquely expressed throughout the ectoderm prior to neural induction suggesting a role in neural competence. With morpholino-mediated knockdown of Sox3, we demonstrate that it is required for induction of neural tissue by BMP inhibition. Together these data indicate that Sox3 has multiple roles in early neural development including as a factor required for nogginmediated neural induction.
Collapse
Affiliation(s)
- Crystal D Rogers
- Department of Biology, Georgetown University, Washington, DC 20057, USA
| | | | | | | | | |
Collapse
|
14
|
The c-myb proto-oncogene and microRNA-15a comprise an active autoregulatory feedback loop in human hematopoietic cells. Blood 2008; 113:505-16. [PMID: 18818396 DOI: 10.1182/blood-2008-01-136218] [Citation(s) in RCA: 134] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The c-myb proto-oncogene encodes an obligate hematopoietic cell transcription factor important for lineage commitment, proliferation, and differentiation. Given its critical functions, c-Myb regulatory factors are of great interest but remain incompletely defined. Herein we show that c-Myb expression is subject to posttranscriptional regulation by microRNA (miRNA)-15a. Using a luciferase reporter assay, we found that miR-15a directly binds the 3'-UTR of c-myb mRNA. By transfecting K562 myeloid leukemia cells with a miR-15a mimic, functionality of binding was shown. The mimic decreased c-Myb expression, and blocked the cells in the G(1) phase of cell cycle. Exogenous expression of c-myb mRNA lacking the 3'-UTR partially rescued the miR-15a induced cell-cycle block. Of interest, the miR-15a promoter contained several potential c-Myb protein binding sites. Occupancy of one canonical c-Myb binding site was demonstrated by chromatin immunoprecipitation analysis and shown to be required for miR-15a expression in K562 cells. Finally, in studies using normal human CD34(+) cells, we showed that c-Myb and miR-15a expression were inversely correlated in cells undergoing erythroid differentiation, and that overexpression of miR-15a blocked both erythroid and myeloid colony formation in vitro. In aggregate, these findings suggest the presence of a c-Myb-miR-15a autoregulatory feedback loop of potential importance in human hematopoiesis.
Collapse
|
15
|
Tamashiro DAA, Alarcón VB, Marikawa Y. Ectopic expression of mouse Sry interferes with Wnt/beta-catenin signaling in mouse embryonal carcinoma cell lines. Biochim Biophys Acta Gen Subj 2008; 1780:1395-402. [PMID: 18675318 DOI: 10.1016/j.bbagen.2008.07.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2008] [Revised: 07/01/2008] [Accepted: 07/03/2008] [Indexed: 12/14/2022]
Abstract
In mammals, Sry is the master regulator of male sex determination, although how it functions is still unclear. By contrast, female sex determination depends on the action of Rspo1 and Wnt4, the regulators of Wnt/beta-catenin signaling. To seek a possible interaction between male and female sex determination mechanisms, we examined whether Sry affects Wnt/beta-catenin signaling. Using the TOPFLASH reporter system to measure Lef/Tcf-dependent transcriptional activity, we showed that ectopic expression of mouse Sry strongly suppressed Wnt/beta-catenin signaling in mouse embryonal carcinoma and human embryonic kidney cell lines. This inhibition occurred downstream of beta-catenin but upstream of Lef/Tcf, and depended on both the HMG-box and the C-terminal transcriptional activation domain. By contrast, TOPFLASH was not inhibited by human SRY, which apparently lacks a transcriptional activation domain. However, a fusion construct consisting of human SRY attached to the C-terminal domain of mouse Sry was able to inhibit TOPFLASH effectively. Furthermore, Sry constructs carrying point mutations equivalent to those in human sex reversal mutations were less effective in inhibiting Wnt/beta-catenin signaling. Also, we showed that the action of Sry as a transcriptional activator was both necessary and sufficient to inhibit Wnt/beta-catenin signaling, suggesting that the transcriptional targets of Sry are responsible for the inhibition of signaling. Sox9 is a potential transcriptional target of Sry, although quantitative RT-PCR analysis indicates that the expression of Sox9 was not up-regulated by the ectopic expression of mouse Sry in mouse embryonal carcinoma cells. While the present study demonstrates an impact of mouse Sry on Wnt/beta-catenin signaling at an in vitro level, it requires further investigations to assess whether such action also takes place in vivo to regulate male sex determination.
Collapse
Affiliation(s)
- Dana Ann A Tamashiro
- Department of Anatomy, Biochemistry and Physiology, Institute for Biogenesis Research, University of Hawaii School of Medicine, Honolulu, HI 96813, USA
| | | | | |
Collapse
|
16
|
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: 97] [Impact Index Per Article: 5.7] [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
|
17
|
Requirement of c-Myb for p210(BCR/ABL)-dependent transformation of hematopoietic progenitors and leukemogenesis. Blood 2008; 111:4771-9. [PMID: 18227349 DOI: 10.1182/blood-2007-08-105072] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The c-Myb gene encodes a transcription factor required for proliferation and survival of normal myeloid progenitors and leukemic blast cells. Targeting of c-Myb by antisense oligodeoxynucleotides has suggested that myeloid leukemia blasts (including chronic myelogenous leukemia [CML]-blast crisis cells) rely on c-Myb expression more than normal progenitors, but a genetic approach to assess the requirement of c-Myb by p210(BCR/ABL)-transformed hematopoietic progenitors has not been taken. We show here that loss of a c-Myb allele had modest effects (20%-28% decrease) on colony formation of nontransduced progenitors, while the effect on p210(BCR/ABL)-expressing Lin(-) Sca-1(+) and Lin(-) Sca-1(+)Kit(+) cells was more pronounced (50%-80% decrease). Using a model of CML-blast crisis, mice (n = 14) injected with p210(BCR/ABL)-transduced p53(-/-)c-Myb(w/w) marrow cells developed leukemia rapidly and had a median survival of 26 days, while only 67% of mice (n = 12) injected with p210(BCR/ABL)-transduced p53(-/-)c-Myb(w/d) marrow cells died of leukemia with a median survival of 96 days. p210(BCR/ABL)-transduced c-Myb(w/w) and c-Myb(w/d) marrow progenitors expressed similar levels of the c-Myb-regulated genes c-Myc and cyclin B1, while those of Bcl-2 were reduced. However, ectopic Bcl-2 expression did not enhance colony formation of p210(BCR/ABL)-transduced c-Myb(w/d) Lin(-)Sca-1(+)Kit(+) cells. Together, these studies support the requirement of c-Myb for p210(BCR/ABL)-dependent leukemogenesis.
Collapse
|
18
|
Castellanos A, Lang G, Frampton J, Weston K. Regulation of erythropoiesis by the neuronal transmembrane protein Lrfn2. Exp Hematol 2007; 35:724-34. [PMID: 17577922 DOI: 10.1016/j.exphem.2007.02.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE The transgenic mouse line MEnTCD2.5 expresses a dominant interfering Myb protein in a T-cell-specific fashion. When MEnTCD2.5 animals are crossed to a second line ubiquitously expressing Myc, they develop a rapid onset, fatal disease characterized by enlarged lymph nodes full of nonlymphoid cells. This study aimed to elucidate the reason for this anomalous non-T-cell phenotype. MATERIALS AND METHODS We studied the cells by morphological analysis, surface marker staining, mRNA expression studies and in vitro colony-forming assays. RESULTS Aberrant cells in MEnTCD2.5 lymph nodes are erythroblasts, and cooperation between MEnTCD2.5 and Myc causes severe erythroblastosis, but not erythroleukemia. MEnTCD2.5:Myc and MEnTCD2.5 animals have pronounced extramedullary erythropoiesis in their lymph nodes, and some increase in bone marrow-derived erythroid progenitors; no other MEnTCD2 transgenic line cooperates in this fashion with Myc, suggesting that the MEnTCD2.5 integration site, in intron 2 of the Lrfn2 gene, is of importance. To confirm this, in in vitro colony-forming assays, expression of wild-type Lrfn2 phenocopies the MEnTCD2.5 defect. Finally, Lrfn2 expression also causes the outgrowth of a bizarre cell type in colony-forming assays that stains positively for both early hematopoietic and fibroblast/fibrocyte surface markers. CONCLUSIONS The Lrfn2 protein, a transmembrane adhesion-type molecule, is able to subvert hematopoietic differentiation to increase erythropoiesis. In cooperation with Myc, this leads to erythroblastosis. Lrfn2 may also be involved in colony forming units-fibroblast regulation. As Lrfn2 expression is detectable in wild-type bone marrow, it likely plays a novel role during normal hematopoiesis.
Collapse
Affiliation(s)
- Andres Castellanos
- Institute of Cancer Research, CR-UK Centre for Cell and Molecular Biology, London, UK
| | | | | | | |
Collapse
|
19
|
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
|
20
|
Hooper J, Maurice D, Argent-Katwala MJG, Weston K. Myb proteins regulate expression of histone variant H2A.Z during thymocyte development. Immunology 2007; 123:282-9. [PMID: 17931383 DOI: 10.1111/j.1365-2567.2007.02697.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
The c-myb gene encodes a transcription factor required for the normal development of T cells in the thymus, and for subsequent peripheral T-cell activation and survival. However, the profile of genes known to be transcriptionally regulated by c-Myb in T cells does not adequately explain the pleiotrophic nature of the effects of c-Myb. We present here a detailed molecular characterization of the regulation of a novel target gene, the histone variant H2A.Z. We show that c-Myb is able to bind to and activate the H2A.Z promoter in T cells both in vitro and in vivo, and present evidence that perturbation of Myb activity during T-cell development results in reduced H2A.Z expression. As H2A.Z is absolutely required for the early stages of mammalian development, and plays essential roles in the regulation of chromatin structure in gene promoters in yeast, its regulation by c-Myb is likely to be of some importance during T-cell development.
Collapse
Affiliation(s)
- Joel Hooper
- Institute of Cancer Research, CRUK Centre for Cell and Molecular Biology, London, UK
| | | | | | | |
Collapse
|
21
|
Peng S, Lalani S, Leavenworth JW, Ho IC, Pauza ME. c-Maf interacts with c-Myb to down-regulate Bcl-2 expression and increase apoptosis in peripheral CD4 cells. Eur J Immunol 2007; 37:2868-80. [PMID: 17823980 DOI: 10.1002/eji.200636979] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The transcription factor c-Maf is critical for IL-4 production and the development of Th2 cells, which promote humoral immunity and protect against extracellular parasites. Yet, little else is known of c-Maf function in CD4 cells. Here, we identify a novel role for c-Maf in regulating susceptibility to apoptosis. Overexpression of c-Maf results in increased susceptibility of CD4 cells to apoptosis induced by multiple stimuli, including growth factor withdrawal, dexamethasone, irradiation, and TCR engagement. This effect is independent of Fas or p53; however, Bcl-2 expression is reduced in c-Maf Tg CD4 cells. Immunoprecipitation and Western blot analyses demonstrate that c-Maf-c-Myb complex formation is enhanced among T cells from c-Maf Tg mice compared to non-Tg littermates following TCR engagement. Unlike non-Tg T cells, c-Myb binding to the Bcl-2 promoter is not detectable in c-Maf Tg T cells by chromatin immunoprecipitation. In reporter assays, Bcl-2 promoter activity is reduced by c-Maf in a dose-dependent manner. Furthermore, transgene-mediated Bcl-2 expression corrects the apoptosis defect observed among c-Maf Tg CD4 cells. These data suggest that c-Maf can interact with c-Myb to reduce Bcl-2 expression, thereby limiting CD4 cell survival following TCR engagement.
Collapse
Affiliation(s)
- Siying Peng
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL 62794, USA
| | | | | | | | | |
Collapse
|
22
|
Maurice D, Hooper J, Lang G, Weston K. c-Myb regulates lineage choice in developing thymocytes via its target gene Gata3. EMBO J 2007; 26:3629-40. [PMID: 17641686 PMCID: PMC1949015 DOI: 10.1038/sj.emboj.7601801] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2006] [Accepted: 06/26/2007] [Indexed: 01/01/2023] Open
Abstract
During T-cell development, thymocytes with intermediate avidity for antigen-MHC complexes are positively selected and then differentiate into functional cytotoxic and helper T cells. This process is controlled by signalling from the T-cell receptor (TCR). Here, we show that the c-Myb transcription factor is a critical downstream regulator of positive selection, promoting the development of helper T cells and blocking the development of cytotoxic T cells. A gain-of-function c-Myb transgene stops development of cytotoxic T cells, instead causing accumulation of a precursor population. Conversely, loss of c-Myb in selecting cells results in significantly fewer helper T cells. In c-Myb-null thymocytes, Gata3, a critical inducer of T-helper cell fate, is not upregulated in response to T-cell receptor signaling, following selection. We show that Gata3 is a direct target of c-Myb, and propose that c-Myb is an important regulator of Gata3, required for transduction of the T-cell receptor signal for subsequent helper cell lineage differentiation.
Collapse
Affiliation(s)
- Diane Maurice
- Institute of Cancer Research, Cancer Research UK Centre for Cell and Molecular Biology, London, UK
| | - Joel Hooper
- Institute of Cancer Research, Cancer Research UK Centre for Cell and Molecular Biology, London, UK
| | - Georgina Lang
- Institute of Cancer Research, Cancer Research UK Centre for Cell and Molecular Biology, London, UK
| | - Kathleen Weston
- Institute of Cancer Research, Cancer Research UK Centre for Cell and Molecular Biology, London, UK
- Institute of Cancer Research, Cancer Research UK Centre for Cell and Molecular Biology, 237 Fulham Road, London SW3 6JB, UK. Tel.: +44 207 153 5253; Fax: +44 207 352 3299; E-mail:
| |
Collapse
|
23
|
Clappier E, Cuccuini W, Kalota A, Crinquette A, Cayuela JM, Dik WA, Langerak AW, Montpellier B, Nadel B, Walrafen P, Delattre O, Aurias A, Leblanc T, Dombret H, Gewirtz AM, Baruchel A, Sigaux F, Soulier J. The C-MYB locus is involved in chromosomal translocation and genomic duplications in human T-cell acute leukemia (T-ALL), the translocation defining a new T-ALL subtype in very young children. Blood 2007; 110:1251-61. [PMID: 17452517 DOI: 10.1182/blood-2006-12-064683] [Citation(s) in RCA: 206] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The C-Myb transcription factor is essential for hematopoiesis, including in the T-cell lineage. The C-Myb locus is a common site of retroviral insertional mutagenesis, however no recurrent genomic involvement has been reported in human malignancies. Here, we identified 2 types of genomic alterations involving the C-MYB locus at 6q23 in human T-cell acute leukemia (T-ALL). First, we found a reciprocal translocation, t(6;7)(q23;q34), that juxtaposed the TCRB and C-MYB loci (n = 6 cases). Second, a genome-wide copy-number analysis by array-based comparative genomic hybridization (array-CGH) identified short somatic duplications that include C-MYB (MYB(dup), n = 13 cases of 84 T-ALL, 15%). Expression analysis, including allele-specific approaches, showed stronger C-MYB expression in the MYB-rearranged cases compared with other T-ALLs, and a dramatically skewed C-MYB allele expression in the TCRB-MYB cases, which suggests that a translocation-driven deregulated expression may overcome a cellular attempt to down-regulate C-MYB. Strikingly, profiling of the T-ALLs by clinical, genomic, and large-scale gene expression analyses shows that the TCRB-MYB translocation defines a new T-ALL subtype associated with a very young age for T-cell leukemia (median, 2.2 years) and with a proliferation/mitosis expression signature. By contrast, the MYB(dup) alteration was associated with the previously defined T-ALL subtypes.
Collapse
Affiliation(s)
- Emmanuelle Clappier
- Genome Rearrangements and Cancer Group, Institut National de la Santé et de la Recherche Médicale U728 and Institut Universitaire d'Hématologie, Paris 7 University, Hôpital Saint-Louis, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Bussolari R, Candini O, Colomer D, Corradini F, Guerzoni C, Mariani SA, Cattelani S, Silvestri C, Pecorari L, Iacobucci I, Soverini S, Fasano T, Martinelli G, Cervantes F, Calabretta B. Coding sequence and intron–exon junctions of the c-myb gene are intact in the chronic phase and blast crisis stages of chronic myeloid leukemia patients. Leuk Res 2007; 31:163-7. [PMID: 16797705 DOI: 10.1016/j.leukres.2006.05.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2006] [Revised: 05/05/2006] [Accepted: 05/08/2006] [Indexed: 01/01/2023]
Abstract
The c-myb gene encodes a transcription factor required for proliferation, differentiation and survival of normal and leukemic hematopoietic cells. c-Myb has a longer half-life in BCR/ABL-expressing than in normal cells, a feature which depends, in part, on PI-3K/Akt-dependent regulation of proteins interacting with the leucine zipper/negative regulatory region of c-Myb. Thus, we asked whether the stability of c-Myb in leukemic cells might be enhanced by mutations interfering with its degradation. We analyzed the c-myb gene in 133 chronic myeloid leukemia (CML) patients in chronic phase and/or blast crisis by denaturing-high performance liquid chromatography (D-HPLC) and sequence analysis of PCR products corresponding to the entire coding sequence and each exon-intron boundary. No mutations were found. We found four single nucleotide polymorphisms (SNPs) and identified an alternatively spliced transcript lacking exon 5, but SNPs frequency and expression of the alternatively spliced transcript were identical in normal and CML cells. Thus, the enhanced stability of c-Myb in CML blast crisis cells and perhaps in other types of leukemia is not caused by a genetic mechanism.
Collapse
Affiliation(s)
- R Bussolari
- Department of Medical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Steiner AB, Engleka MJ, Lu Q, Piwarzyk EC, Yaklichkin S, Lefebvre JL, Walters JW, Pineda-Salgado L, Labosky PA, Kessler DS. FoxD3 regulation of Nodal in the Spemann organizer is essential for Xenopus dorsal mesoderm development. Development 2006; 133:4827-38. [PMID: 17092955 PMCID: PMC1676154 DOI: 10.1242/dev.02663] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Induction and patterning of the mesodermal germ layer is a key early step of vertebrate embryogenesis. We report that FoxD3 function in the Xenopus gastrula is essential for dorsal mesodermal development and for Nodal expression in the Spemann organizer. In embryos and explants, FoxD3 induced mesodermal genes, convergent extension movements and differentiation of axial tissues. Engrailed-FoxD3, but not VP16-FoxD3, was identical to native FoxD3 in mesoderm-inducing activity, indicating that FoxD3 functions as a transcriptional repressor to induce mesoderm. Antagonism of FoxD3 with VP16-FoxD3 or morpholino-knockdown of FoxD3 protein resulted in a complete block to axis formation, a loss of mesodermal gene expression, and an absence of axial mesoderm, indicating that transcriptional repression by FoxD3 is required for mesodermal development. FoxD3 induced mesoderm in a non-cell-autonomous manner, indicating a role for secreted inducing factors in the response to FoxD3. Consistent with this mechanism, FoxD3 was necessary and sufficient for the expression of multiple Nodal-related genes, and inhibitors of Nodal signaling blocked mesoderm induction by FoxD3. Therefore, FoxD3 is required for Nodal expression in the Spemann organizer and this function is essential for dorsal mesoderm formation.
Collapse
Affiliation(s)
- Aaron B. Steiner
- Department of Cell and Developmental Biology University of Pennsylvania School of Medicine 1110 BRB 2/3, 421 Curie Boulevard Philadelphia, PA 19104, USA Tel: 215-898-1478 Fax: 215-573-7601
| | - Mark J. Engleka
- Department of Cell and Developmental Biology University of Pennsylvania School of Medicine 1110 BRB 2/3, 421 Curie Boulevard Philadelphia, PA 19104, USA Tel: 215-898-1478 Fax: 215-573-7601
| | - Qun Lu
- Department of Cell and Developmental Biology University of Pennsylvania School of Medicine 1110 BRB 2/3, 421 Curie Boulevard Philadelphia, PA 19104, USA Tel: 215-898-1478 Fax: 215-573-7601
| | - Eileen C. Piwarzyk
- Department of Cell and Developmental Biology University of Pennsylvania School of Medicine 1110 BRB 2/3, 421 Curie Boulevard Philadelphia, PA 19104, USA Tel: 215-898-1478 Fax: 215-573-7601
| | - Sergey Yaklichkin
- Department of Cell and Developmental Biology University of Pennsylvania School of Medicine 1110 BRB 2/3, 421 Curie Boulevard Philadelphia, PA 19104, USA Tel: 215-898-1478 Fax: 215-573-7601
| | - Julie L. Lefebvre
- Department of Cell and Developmental Biology University of Pennsylvania School of Medicine 1110 BRB 2/3, 421 Curie Boulevard Philadelphia, PA 19104, USA Tel: 215-898-1478 Fax: 215-573-7601
| | - James W. Walters
- Department of Cell and Developmental Biology University of Pennsylvania School of Medicine 1110 BRB 2/3, 421 Curie Boulevard Philadelphia, PA 19104, USA Tel: 215-898-1478 Fax: 215-573-7601
| | - Liliam Pineda-Salgado
- Department of Cell and Developmental Biology University of Pennsylvania School of Medicine 1110 BRB 2/3, 421 Curie Boulevard Philadelphia, PA 19104, USA Tel: 215-898-1478 Fax: 215-573-7601
| | | | | |
Collapse
|
26
|
Hwang IK, Yoo KY, Cho BM, Hwang HS, Kim SM, Oh SM, Choi SK, Hwang DY, Won MH, Moon SM. The pattern of E2F1 and c-myb immunoreactivities in the CA1 region is different from those in the CA2/3 region of the gerbil hippocampus induced by transient ischemia. J Neurol Sci 2006; 247:192-201. [PMID: 16782130 DOI: 10.1016/j.jns.2006.05.048] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2005] [Revised: 05/02/2006] [Accepted: 05/04/2006] [Indexed: 11/26/2022]
Abstract
In this study, we examined transient ischemia-induced changes in transcription factor E2F1 and c-myb expressions in the gerbil hippocampus after 5 min of transient forebrain ischemia. E2F1 immunoreactivity significantly increased in the CA1 region 6-12 h after ischemia/reperfusion. c-myb immunoreactivity increased mainly in CA1 pyramidal cells with time by 12 h after ischemia. Thereafter, E2F1 and c-myb immunoreactivities significantly decreased compared to those in the 12 h post-ischemic group. Four days after ischemia/reperfusion, E2F1 and c-myb immunoreactivities were detected in non-pyramidal cells. Ten days after ischemia, c-myb immunoreactivity increased again: at this time, astrocytes as well as non-pyramidal cells showed E2F1 and c-myb immunoreactivities. In the CA2/3 region, E2F1 and c-myb immunoreactivities mainly changed in non-pyramidal cells, and 10 days after ischemia, c-myb immunoreactivity was not expressed in astrocytes. In conclusion, E2F1 and c-myb significantly alter in pyramidal cells and express in astrocytes in the gerbil hippocampal CA1 region after transient ischemia. These results indicate that E2F1 and c-myb in the CA1 region after ischemic damage may be associated with delayed neuronal death.
Collapse
Affiliation(s)
- In Koo Hwang
- Department of Anatomy, College of Medicine, Hallym University, Chunchon 200-702, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Chen J, Kremer CS, Bender TP. The carbonic anhydrase I locus contains a c-Myb target promoter and modulates differentiation of murine erythroleukemia cells. Oncogene 2006; 25:2758-72. [PMID: 16407849 DOI: 10.1038/sj.onc.1209295] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The Myb proto-oncogene encodes a transcription factor (c-Myb) that is essential for normal hematopoiesis and is thought to regulate hematopoietic cell proliferation and differentiation by regulating expression of specific target genes. We identify the mouse erythroid-specific carbonic anhydrase I promoter (CAIe) as a target of c-Myb activity and demonstrate that Myb activity is critical for carbonic anhydrase I (CAI) expression in C19 MEL cells. CAI expression is downregulated when MEL cells differentiate in response to MEnT or treatment with N, N-hexamethylene bisacetamide (HMBA). Coexpression of GATA-1 with c-Myb results in synergistic activation of transcription from the CAIe promoter and both transcription factors interact with the CAIe promoter in vivo. We identify a novel 20 bp sequence in the CAIe promoter that is sufficient to mediate synergistic activation of the CAIe promoter by c-Myb and GATA-1. c-Myb and GATA-1 interact with this DNA sequence suggesting that c-Myb and GATA-1 may be contained in a complex that interacts with this region of the CAIe promoter. Forced expression of CAI delayed HMBA-induced differentiation of MEL cells and maintained them in a proliferating state. These data strongly suggest that CAI is a c-Myb target and is involved in regulating MEL cell proliferation and differentiation.
Collapse
MESH Headings
- Acetamides/pharmacology
- Animals
- Antineoplastic Agents/pharmacology
- Base Sequence
- Carbonic Anhydrase I/genetics
- Carbonic Anhydrase I/metabolism
- Cell Differentiation
- Cell Proliferation/drug effects
- Chromatin Immunoprecipitation
- GATA1 Transcription Factor/metabolism
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Genes, Dominant
- Leukemia, Erythroblastic, Acute/enzymology
- Leukemia, Erythroblastic, Acute/genetics
- Leukemia, Erythroblastic, Acute/pathology
- Mice
- Molecular Sequence Data
- Promoter Regions, Genetic/genetics
- Proto-Oncogene Proteins c-myb/metabolism
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- J Chen
- Department of Molecular Physiology and Biological Physics, University of Virginia Health System, Charlottesville, VA 22908-0734, USA
| | | | | |
Collapse
|
28
|
Miyamoto T, Akashi K. Lineage promiscuous expression of transcription factors in normal hematopoiesis. Int J Hematol 2005; 81:361-7. [PMID: 16158815 DOI: 10.1532/ijh97.05003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Hematopoiesis has provided a valuable model for examining how genetic programs are established and executed in terms of cell fate decision. Identification of common myeloid and lymphoid progenitors allows us to directly assess the regulatory mechanisms of lineage commitment. Multiple markers of hematopoietic lineages are coexpressed in hematopoietic stem cells and progenitors, a phenomenon referred to as lineage priming. Promiscuous expression of several lineage-affiliated genes precedes lineage commitment but does not alter the biological potential of hematopoietic stem cells and multipotent progenitors. Promiscuous accessibility of multiple programs allows flexibility in cell fate commitment at the multipotent stages, indicating that transcriptional promiscuity can operate in stem cells and progenitors to control their transition from multipotency to single-lineage commitment.
Collapse
Affiliation(s)
- Toshihiro Miyamoto
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.
| | | |
Collapse
|
29
|
Sala A. B-MYB, a transcription factor implicated in regulating cell cycle, apoptosis and cancer. Eur J Cancer 2005; 41:2479-84. [PMID: 16198555 DOI: 10.1016/j.ejca.2005.08.004] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
B-MYB belongs to the MYB family of transcription factors that include A-MYB and c-MYB. While A-MYB and c-MYB are tissue-specific, B-MYB is broadly expressed in rapidly dividing cells of developing or adult mammals. B-MYBs liaisons with important players of the cell cycle and transcription machinery, such as E2F and retinoblastoma proteins, suggest that its essential function in stem cell formation and mammalian development could be related to its ability to directly or indirectly impinge on gene expression. Besides its role in the cell cycle, B-MYB has been shown to promote cell survival by activating antiapoptotic genes such as ApoJ/clusterin and BCL2. Here, we discuss how B-MYB could be implicated in tumourigenesis by regulating gene expression.
Collapse
Affiliation(s)
- Arturo Sala
- Molecular Haematology and Cancer Biology Unit, Institute of Child Health, WC1N 1EH London, UK.
| |
Collapse
|
30
|
Corradini F, Cesi V, Bartella V, Pani E, Bussolari R, Candini O, Calabretta B. Enhanced proliferative potential of hematopoietic cells expressing degradation-resistant c-Myb mutants. J Biol Chem 2005; 280:30254-62. [PMID: 15927960 DOI: 10.1074/jbc.m504703200] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The c-myb gene encodes a transcription factor required for proliferation, differentiation, and survival of hematopoietic cells. Expression of c-Myb is often increased in hematological malignancies, but the underlying mechanisms are poorly understood. We show here that c-Myb has a longer half-life (at least 2-fold) in BCR/ABL-expressing than in normal hematopoietic cells. Such enhanced stability was dependent on a phosphatidylinositol 3-kinase (PI-3K)/Akt/GSKIIIbeta pathway(s) as indicated by the suppression of c-Myb expression upon treatment with PI-3K inhibitors or co-expression with dominant negative Akt or constitutively active GSKIIIbeta. Moreover, inhibition of GSKIIIbeta by LiCl enhanced c-Myb expression in parental 32Dcl3 cells. Compared with wild type c-Myb, three mutants (delta(358-452), delta(389-418), and L389A/L396A c-Myb) of the leucine zipper domain had increased stability. However, only expression of delta(358-452) was not affected by inhibition of the PI-3K/Akt pathway and was not enhanced by a proteasome inhibitor, suggesting that leucine zipper-dependent and -independent mechanisms are involved in the regulation of c-Myb stability. Indeed, delta(389-418) carrying four lysine-to-alanine substitutions (delta(389-418) K387A/K428A/K442A/K445A) was as stable as delta(358-452) c-Myb. Compared with full-length c-Myb, constitutive expression of delta(358-452) and delta(389-418) c-Myb in Lin-Sca-1+ mouse marrow cells increased cytokine-dependent primary and secondary colony formation. In K562 cells, expression of delta(358-452), delta(389-418), and L389A/L396A c-Myb led to enhanced proliferation after STI571 treatment. Thus, enhanced stability of c-Myb by activation of PI-3K-dependent pathway(s) might contribute to the higher proliferative potential of BCR/ABL-expressing and, perhaps, other leukemic cells.
Collapse
Affiliation(s)
- Francesca Corradini
- Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson Medical College, Philadelphia, Pennsylvania 19107, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Lang G, White JR, Argent-Katwala MJG, Allinson CG, Weston K. Myb proteins regulate the expression of diverse target genes. Oncogene 2005; 24:1375-84. [PMID: 15608679 DOI: 10.1038/sj.onc.1208301] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Hematopoiesis, the process by which mature blood cells arise, is controlled by multiple transcription factors, which act in stage- and lineage-specific complexes. It is a major goal to elucidate the genes regulated by these transcription factors, in order to obtain a full understanding of the process and its malignant counterpart, leukemia. Myb family transcription factors play a central role in hematopoiesis. To identify new Myb family target genes, we have used an inducible dominant-negative protein for a subtraction cloning protocol in a model cell system (FDCP-Mix) with many characteristics of normal hematopoiesis. We present here a novel group of 29 validated Myb family target genes of diverse functions.
Collapse
Affiliation(s)
- Georgina Lang
- CRUK Centre for Cell and Molecular Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | | | | | | | | |
Collapse
|
32
|
Sandberg ML, Sutton SE, Pletcher MT, Wiltshire T, Tarantino LM, Hogenesch JB, Cooke MP. c-Myb and p300 regulate hematopoietic stem cell proliferation and differentiation. Dev Cell 2005; 8:153-66. [PMID: 15691758 DOI: 10.1016/j.devcel.2004.12.015] [Citation(s) in RCA: 221] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2004] [Revised: 11/18/2004] [Accepted: 12/16/2004] [Indexed: 02/04/2023]
Abstract
Precise control of hematopoietic stem cell (HSC) proliferation and differentiation is needed to maintain a lifetime supply of blood cells. Using genome-wide ENU mutagenesis and phenotypic screening, we have identified a mouse line that harbors a point mutation in the transactivation (TA) domain of the transcription factor c-Myb (M303V), which reduces c-Myb-dependent TA by disrupting its interaction with the transcriptional coactivator p300. The biological consequences of the c-Myb(M303V/M303V) mutation include thrombocytosis, megakaryocytosis, anemia, lymphopenia, and the absence of eosinophils. Detailed analysis of hematopoiesis in c-Myb(M303V/M303V) mice reveals distinct blocks in T cell, B cell, and red blood cell development, as well as a remarkable 10-fold increase in the number of HSCs. Cell cycle analyses show that twice as many HSCs from c-Myb(M303V/M303V) animals are actively cycling. Thus c-Myb, through interaction with p300, controls the proliferation and differentiation of hematopoietic stem and progenitor cells.
Collapse
Affiliation(s)
- Mark L Sandberg
- Genomics Institute of the Novartis Research Foundation, San Diego, California 92121, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Vickers ER, Kasza A, Kurnaz IA, Seifert A, Zeef LAH, O'donnell A, Hayes A, Sharrocks AD. Ternary complex factor-serum response factor complex-regulated gene activity is required for cellular proliferation and inhibition of apoptotic cell death. Mol Cell Biol 2005; 24:10340-51. [PMID: 15542842 PMCID: PMC529045 DOI: 10.1128/mcb.24.23.10340-10351.2004] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Members of the ternary complex factor (TCF) subfamily of the ETS-domain transcription factors are activated through phosphorylation by mitogen-activated protein kinases (MAPKs) in response to a variety of mitogenic and stress stimuli. The TCFs bind and activate serum response elements (SREs) in the promoters of target genes in a ternary complex with a second transcription factor, serum response factor (SRF). The association of TCFs with SREs within immediate-early gene promoters is suggestive of a role for the ternary TCF-SRF complex in promoting cell cycle entry and proliferation in response to mitogenic signaling. Here we have investigated the downstream gene regulatory and phenotypic effects of inhibiting the activity of genes regulated by TCFs by expressing a dominantly acting repressive form of the TCF, Elk-1. Inhibition of ternary complex activity leads to the downregulation of several immediate-early genes. Furthermore, blocking TCF-mediated gene expression leads to growth arrest and triggers apoptosis. By using mutant Elk-1 alleles, we demonstrated that these effects are via an SRF-dependent mechanism. The antiapoptotic gene Mcl-1 is identified as a key target for the TCF-SRF complex in this system. Thus, our data confirm a role for TCF-SRF-regulated gene activity in regulating proliferation and provide further evidence to indicate a role in protecting cells from apoptotic cell death.
Collapse
Affiliation(s)
- Elaine R Vickers
- Faculty of Life Sciences, University of Manchester, Michael Smith Building, Oxford Rd., Manchester M13 9PT, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Ataliotis P, Ivins S, Mohun TJ, Scambler PJ. XTbx1 is a transcriptional activator involved in head and pharyngeal arch development inXenopus laevis. Dev Dyn 2005; 232:979-91. [PMID: 15736267 DOI: 10.1002/dvdy.20276] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The development of pharyngeal arch derivatives in mouse and zebrafish embryos depends on the activity of the transcription factor Tbx1. We cloned the Xenopus laevis orthologue of Tbx1 (XTbx1) and show that the pattern of expression is similar to that in other vertebrate species. Zygotic transcripts are first detected shortly after the mid-blastula transition and are localized to the presumptive mesoderm at mid-gastrula stages. XTbx1 expression persists in the lateral plate mesoderm at neurula stages and is found in the pharyngeal arches and otic vesicles from early tail bud stages onward. We demonstrate that XTbx1 is a transcriptional activator and that this trans-activation requires the C-terminal region of the protein. A dominant interfering mutant of XTbx1 disrupts the development of Xenopus head structures and pharyngeal arch derivatives. Lineage labeling reveals a requirement for XTbx1 function in cells that contribute to the pharyngeal mesoderm and for fgf8 expression.
Collapse
Affiliation(s)
- Paris Ataliotis
- Molecular Medicine Unit, Institute of Child Health, London WC1N 1EH, United Kingdom.
| | | | | | | |
Collapse
|
35
|
Sheridan PJ, Lawrie A, Crossman DC, Holt CM, Newman CM. VP22-mediated intercellular transport correlates with enhanced biological activity of MybEngrailed but not (HSV-I) thymidine kinase fusion proteins in primary vascular cells following non-viral transfection. J Gene Med 2005; 7:375-85. [PMID: 15543525 DOI: 10.1002/jgm.679] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND The intercellular transport properties of the herpes simplex virus (HSV) protein VP22 have been harnessed to enhance the effectiveness of viral gene transfer. We investigated the intercellular transport and biological effects of VP22 fused with the dominant negative c-Myb chimera, MybEngrailed (MybEn) and HSV-I thymidine kinase (TK), in primary vascular smooth muscle cells (VSMC) following non-viral transfection. MATERIALS AND METHODS Porcine VSMC transfected with plasmids encoding MybEn, TK and their respective N- and C-terminal VP22 fusion proteins were assayed for the extent and distribution of transgene expression (by immunohistochemistry), culture growth and apoptosis. RESULTS The N-terminal MybEn fusion with VP22 (MybEnVP22) and both TK fusions, but not VP22MybEn, exhibited intercellular spread from primary transfected to up to 200 surrounding cells. pMybEnVP22-transfected cultures exhibited growth inhibition and apoptosis rates that were 10.6 +/- 3.6 and 3.2 +/- 1.0 fold higher than in pMybEn-transfected cultures; pVP22MybEn-transfected cultures showed no difference in these parameters. pTK-transfected cultures underwent 60-70% cell death in the presence of ganciclovir despite <2% primary transfection, which was not increased in cultures transfected with plasmids encoding VP22-TK fusions. CONCLUSIONS The close correlation between immunocytochemical and biological assays suggests that intercellular transport is crucial to the enhanced biological activity of the MybEnVP22 fusion. The "intrinsic" bystander activity of TK was 4-fold greater than was "engineered" by VP22 fusion, probably reflecting the abundance of gap junctions between VSMC. VP22 fusion may enhance the efficiency of non-viral gene delivery when combined with the appropriate therapeutic transgene, target tissue and transfection method.
Collapse
Affiliation(s)
- Paul J Sheridan
- Cardiovascular Research Unit, Division of Clinical Sciences (North), Clinical Sciences Centre, University of Sheffield, Northern General Hospital, Sheffield S5 7AU, UK.
| | | | | | | | | |
Collapse
|
36
|
Lieu YK, Kumar A, Pajerowski AG, Rogers TJ, Reddy EP. Requirement of c-myb in T cell development and in mature T cell function. Proc Natl Acad Sci U S A 2004; 101:14853-8. [PMID: 15466706 PMCID: PMC522009 DOI: 10.1073/pnas.0405338101] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Previous reports have suggested that the protooncogene c-myb participates in T cell development in the thymus and mature T cell proliferation. We have generated two T cell-specific c-myb knockout mouse models, myb/LckCre and myb/CD4Cre. We have demonstrated that c-myb is required for the development of thymocytes at the DN3 stage, for survival and proliferation of double-positive thymocytes, for differentiation of single-positive CD4 and CD8 T cells, and for the proliferative responses of mature T cells. In addition, our data show that c-myb is directly involved in the formation of double-positive CD4+CD8+CD25+, CD4+CD25+, and CD8+CD25+ T cells, developmental processes that may imply a role for c-myb in autoimmune dysfunction.
Collapse
Affiliation(s)
- Yen K Lieu
- Fels Institute for Cancer Research and Molecular Biology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | | | | | | | | |
Collapse
|
37
|
Shetzline SE, Rallapalli R, Dowd KJ, Zou S, Nakata Y, Swider CR, Kalota A, Choi JK, Gewirtz AM. Neuromedin U: a Myb-regulated autocrine growth factor for human myeloid leukemias. Blood 2004; 104:1833-40. [PMID: 15187020 DOI: 10.1182/blood-2003-10-3577] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The c-myb proto-oncogene has been implicated in leukemogenesis, but possible mechanisms remain ill defined. To gain further insight to this process, we used transcript profiling in K562 cells expressing a dominant-negative Myb (MERT) protein. A total of 105 potential Myb gene targets were identified. Neuromedin U (NmU), a peptide affecting calcium transport, underwent the greatest expression change ( approximately 5-fold decrease). To verify a linkage between c-myb and NmU, their mRNA levels were quantitated using real-time polymerase chain reaction in primary acute myeloid leukemia (AML) and acute lymphoid leukemia (ALL), as well as normal hematopoietic cells. We found that c-myb was elevated in AML and ALL samples, but NmU expression was increased only in AML cells. Significantly, only AML cells expressed the cognate receptor of NmU, NMU1R, suggesting the presence of a novel autocrine loop. We examined this possibility in detail. Exogenous NmU "rescued" growth suppression in K562-MERT cells and stimulated the growth of primary AML cells. Short interfering RNA "knockdown" of NmU in K562 cells arrested cell growth. Exposing Indo-1-labeled K562 cells to NmU induced an intracellular Ca(++) flux consistent with engagement of the NMU1R. Combined, these results suggest that NmU expression is related to Myb and that the NmU/NMU1R axis constitutes a previously unknown growth-promoting autocrine loop in myeloid leukemia cells.
Collapse
Affiliation(s)
- Susan E Shetzline
- Department of Internal Medicine, Division of Hematology/Oncology, University of Pennsylvania School of Medicine, 421 Curie Blvd, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Zhou N, Wilson KA, Andrews ME, Kauffman JS, Raff RA. Evolution of OTP-independent larval skeleton patterning in the direct-developing sea urchin, Heliocidaris erythrogramma. JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2004; 300:58-71. [PMID: 14984035 DOI: 10.1002/jez.b.46] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Heliocidaris erythrogramma is a direct-developing sea urchin that has evolved a modified ontogeny, a reduced larval skeleton, and accelerated development of the adult skeleton. The Orthopedia gene (Otp) encodes a homeodomain transcription factor crucial in patterning the larval skeleton of indirect-developing sea urchins. We compare the role of Otp in larvae of the indirect-developing sea urchin Heliocidaris tuberculata and its direct-developing congener H. erythrogramma. Otp is a single-copy gene with an identical protein sequence in these species. Expression of Otp is initiated by the late gastrula, initially in two cells of the oral ectoderm in H. tuberculata. These cells are restricted to oral ectoderm and exhibit left-right symmetry. There are about 266 copies of Otp mRNA per Otp- expressing cell in H. tuberculata. We tested OTP function in H. tuberculata and H. erythrogramma embryos by microinjection of Otp mRNA. Mis-expression of Otp mRNA in H. tuberculata radialized the embryos and caused defects during larval skeletogenesis. Mis-expression of Otp mRNA in H. erythrogramma embryos did not affect skeleton formation. This is consistent with the observation by in situ hybridization of no concentration of Otp transcript in any particular cells or region of the H. erythrogramma larva, and measurement of a level of less than one copy of endogenous Otp mRNA per cell in H. erythrogramma. OTP plays an important role in patterning the larval skeleton of H. tuberculata, but this role apparently has been lost in the evolution of the H. erythrogramma larva, and replaced by a new patterning mechanism.
Collapse
Affiliation(s)
- Na Zhou
- Department of Biology, Indiana University, Bloomington, Indiana 47405, USA
| | | | | | | | | |
Collapse
|
39
|
Sykes DB, Kamps MP. E2a/Pbx1 induces the rapid proliferation of stem cell factor-dependent murine pro-T cells that cause acute T-lymphoid or myeloid leukemias in mice. Mol Cell Biol 2004; 24:1256-69. [PMID: 14729970 PMCID: PMC321418 DOI: 10.1128/mcb.24.3.1256-1269.2004] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2003] [Revised: 05/27/2003] [Accepted: 10/07/2003] [Indexed: 11/20/2022] Open
Abstract
Oncoprotein E2a/Pbx1 is produced by the t(1;19) chromosomal translocation of human pre-B acute lymphoblastic leukemia. E2a/Pbx1 blocks differentiation of primary myeloid progenitors but, paradoxically, induces apoptosis in established pre-B-cell lines, and no transforming function of E2a/Pbx1 has been reported in cultured lymphoid progenitors. Here, we demonstrate that E2a/Pbx1 induces immortal proliferation of stem cell factor (SCF)-dependent pro-T thymocytes by a mechanism dependent upon both its transactivation and DNA-binding functions. E2a-Pbx1 cooperated with cytokines or activated signaling oncoproteins to induce cell division, as inactivation of conditional E2a/Pbx1 in either factor-dependent pro-T cells or pro-T cells made factor independent by expression of Bcr/Abl resulted in pro-T-cell quiescence, while reactivation of E2a/Pbx1 restored cell division. Infusion of E2a/Pbx1 pro-T cells in mice caused T lymphoblastic leukemia and, unexpectedly, acute myeloid leukemia. The acute lymphoblastic leukemia did not evidence further maturation, suggesting that E2a/Pbx1 establishes an early block in pro-T-cell development that cannot be overcome by marrow or thymic microenvironments. In an E2a/Pbx1 pro-T thymocyte clone that induced only pro-T acute lymphoblastic leukemia, coexpression of Bcr/Abl expanded its leukemic phenotype to include acute myeloid leukemia, suggesting that unique functions of cooperating signaling oncoproteins can influence the lymphoid versus myeloid character of E2a/Pbx1 leukemia and may cooperate with E2a/Pbx1 to dictate the pre-B-cell phenotype of human leukemia containing t(1;19).
Collapse
Affiliation(s)
- David B Sykes
- Department of Pathology, University of California-San Diego, La Jolla, California 92093-0612, USA
| | | |
Collapse
|
40
|
Yan RT, Wang SZ. Requirement of neuroD for photoreceptor formation in the chick retina. Invest Ophthalmol Vis Sci 2004; 45:48-58. [PMID: 14691153 PMCID: PMC1988846 DOI: 10.1167/iovs.03-0774] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE The genetic control of photoreceptor cell fate in the vertebrate retina is poorly understood. Published studies suggest that the genetic program underlying photoreceptor production involves neuroD, a proneural basic helix-loop-helix (bHLH) gene. The present study investigates whether neuroD is necessary for photoreceptor cell development, by using loss-of-function analyses. METHOD Engrailed-mediated active repression, antisense oligonucleotides, and small interfering RNA (siRNA) were used to attenuate neuroD expression and function in embryonic chick retina. The development of the retina was subsequently analyzed to determine whether these experimental manipulations would yield photoreceptor deficits in otherwise normal retina. RESULTS Chick embryos infected with retroviruses expressing an active repression construct, En-NeuroDDeltaC, exhibited severe photoreceptor deficits. The outer nuclear layer (ONL) of the retina was no longer a contiguous structure, but became fragmented with regions that contained fewer or no photoreceptor cells. Photoreceptor deficiency was evident even before the retina became laminated, suggesting that active repression of NeuroD may have affected photoreceptor genesis. No deficiency was observed in other types of retinal cells. Culturing retinal cells in the presence of siRNA against neuroD resulted in a more than 50% reduction in the number of photoreceptor cells and an increase in the number of chx10+ cells. Subjecting the developing retina to antisense oligonucleotides against neuroD yielded fewer photoreceptor cells both in vivo and in vitro. Consistent with these observations, anti-NeuroD antibody specifically labeled the nuclei of the ONL. CONCLUSIONS The data suggest a specific and an essential role of neuroD in photoreceptor formation in the chick retina.
Collapse
Affiliation(s)
- Run-Tao Yan
- Department of Ophthalmology, University of Alabama at Birmingham, School of Medicine, Birmingham, Alabama, USA
| | | |
Collapse
|
41
|
Ishii Y, Kawaguchi M, Takagawa K, Oya T, Nogami S, Tamura A, Miura Y, Ido A, Sakata N, Hashimoto-Tamaoki T, Kimura T, Saito T, Tamaoki T, Sasahara M. ATBF1-A protein, but not ATBF1-B, is preferentially expressed in developing rat brain. J Comp Neurol 2003; 465:57-71. [PMID: 12926016 DOI: 10.1002/cne.10807] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The ATBF1 gene encodes transcription factors containing four homeodomains and multiple zinc finger motifs. However, the gene products have yet to be identified and the role remains unknown in vivo. In this study, we raised an antiserum for ATBF1 and found high levels of expression of ATBF1 in developing rat brain. Western and Northern blot analyses detected a 400 kDa protein and 12.5 kb mRNA in developing rat brain, respectively; both corresponding to ATBF1-A but not the B isoform. The protein was highly expressed in the midbrain and diencephalon and mRNA was highly expressed in the brainstem, mostly in embryo and neonatal brain. Immunohistochemistry identified postmitotic neurons in the brainstem as the major site of ATBF1 expression, and the expression levels varied depending on age of and location in the brain. Expression was transient and weak in the precursor cells at early neurogenesis. ATBF1 decreased postnatally, but remained in mature neurons, including those expressing DOPA decarboxylase (DDC). High levels of ATBF1 were expressed in precursor cells in accordance with neurogenesis and were continued to the mature neurons in specific areas such as the inferior colliculus. Expression was not significant from precursor cells to mature neurons in the cerebral cortex and hippocampus. ATBF1 and its Drosophila homolog, Zfh-2, are known to regulate cell differentiation and proliferation via the interaction with either of the basic helix-loop-helix transcription factors, c-myb, or the DDC gene. Together with these reported functions the expression features detected here suggest that ATBF1 may participate in the regulation of neuronal cell maturation or region-specific central nervous system differentiation.
Collapse
Affiliation(s)
- Yoko Ishii
- Faculty of Medicine, Toyama Medical and Pharmaceutical University, Toyama 930-0194, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
RaxL is a paired-like homeobox gene involved in vertebrate eye morphogenesis. We examined RaxL protein expression patterns during chick retinal development in combination with ganglion cell markers including the RA4 antigen, cBrn-3, Islet-1 and neuronal type III beta-tubulin. Double-immunostaining demonstrated that downregulation of RaxL protein correlates with upregulation of ganglion cell markers in the ganglion cell layer (GCL). To explore this correlation in vivo, we performed gain- and loss-of-function experiments by electroporating retroviral vectors encoding wild-type and dominant-negative-RaxL into the optic vesicles of stage 10 chick embryos. Infection with virus expressing RaxL led to a 35% decrease in Islet-1-positive ganglion cells at E5.0 and a complete loss of ganglion cells at E15, with no effect on displaced amacrine cells in the GCL. When dominant-negative RaxL was expressed, the total number of cells in the GCL increased by approximately 40% at E5.0 but was reduced to 40% at E15, due to ectopic apoptosis in the GCL from E9 to E15. These results suggest that RaxL gives an inhibitory effect on ganglion cell development and that the loss of RaxL expression is required for maintenance of ganglion cells.
Collapse
Affiliation(s)
- Kiyo Sakagami
- Graduate School of Biological Sciences, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma 630-0101, Japan
| | | | | | | |
Collapse
|
43
|
Matre V, Høvring PI, Fjeldheim AK, Helgeland L, Orvain C, Andersson KB, Gautvik KM, Gabrielsen OS. The human neuroendocrine thyrotropin-releasing hormone receptor promoter is activated by the haematopoietic transcription factor c-Myb. Biochem J 2003; 372:851-9. [PMID: 12628004 PMCID: PMC1223435 DOI: 10.1042/bj20030057] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2003] [Revised: 03/03/2003] [Accepted: 03/10/2003] [Indexed: 01/14/2023]
Abstract
Thyrotropin-releasing hormone (TRH) receptor (TRHR) is a G-protein-coupled receptor playing a crucial role in the anterior pituitary where it controls the synthesis and secretion of thyroid-stimulating hormone and prolactin. Its widespread presence not only in the central nervous system, but also in peripheral tissues, including thymus, indicates other important, but unknown, functions. One hypothesis is that the neuropeptide TRH could play a role in the immune system. We report here that the human TRHR promoter contains 11 putative response elements for the haematopoietic transcription factor c-Myb and is highly Myb-responsive in transfection assays. Analysis of Myb binding to putative response elements revealed one preferred binding site in intron 1 of the receptor gene. Transfection studies of promoter deletions confirmed that this high-affinity element is necessary for efficient Myb-dependent transactivation of reporter plasmids in CV-1 cells. The Myb-dependent activation of the TRHR promoter was strongly suppressed by expression of a dominant negative Myb-Engrailed fusion. In line with these observations, reverse transcriptase PCR analysis of rat tissues showed that the TRHR gene is expressed both in thymocytes and bone marrow. Furthermore, specific, high-affinity TRH agonist binding to cell-surface receptors was demonstrated in thymocytes and a haematopoietic cell line. Our findings imply a novel functional link between the neuroendocrine and the immune systems at the level of promoter regulation.
Collapse
Affiliation(s)
- Vilborg Matre
- Department of Biochemistry, University of Oslo, P.O. Box 1041 Blindern, Norway.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Hiratsu K, Matsui K, Koyama T, Ohme-Takagi M. Dominant repression of target genes by chimeric repressors that include the EAR motif, a repression domain, in Arabidopsis. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2003; 34:733-9. [PMID: 12787253 DOI: 10.1046/j.1365-313x.2003.01759.x] [Citation(s) in RCA: 575] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
The redundancy of genes for plant transcription factors often interferes with efforts to identify the biologic functions of such factors. We show here that four different transcription factors fused to the EAR motif, a repression domain of only 12 amino acids, act as dominant repressors in transgenic Arabidopsis and suppress the expression of specific target genes, even in the presence of the redundant transcription factors, with resultant dominant loss-of-function phenotypes. Chimeric EIN3, CUC1, PAP1, and AtMYB23 repressors that included the EAR motif dominantly suppressed the expression of their target genes and caused insensitivity to ethylene, cup-shaped cotyledons, reduction in the accumulation of anthocyanin, and absence of trichomes, respectively. This chimeric repressor silencing technology (CRES-T), exploiting the EAR-motif repression domain, is simple and effective and can overcome genetic redundancy. Thus, it should be useful not only for the rapid analysis of the functions of redundant plant transcription factors but also for the manipulation of plant traits via the suppression of gene expression that is regulated by specific transcription factors.
Collapse
Affiliation(s)
- Keiichiro Hiratsu
- Gene Function Research Laboratory, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, Tsukuba 305-8566, Japan
| | | | | | | |
Collapse
|
45
|
Jin Z, Zhang J, Klar A, Chédotal A, Rao Y, Cepko CL, Bao ZZ. Irx4-mediated regulation of Slit1 expression contributes to the definition of early axonal paths inside the retina. Development 2003; 130:1037-48. [PMID: 12571096 DOI: 10.1242/dev.00326] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Although multiple axon guidance cues have been discovered in recent years, little is known about the mechanism by which the spatiotemporal expression patterns of the axon guidance cues are regulated in vertebrates. We report that a homeobox gene Irx4 is expressed in a pattern similar to that of Slit1 in the chicken retina. Overexpression of Irx4 led to specific downregulation of Slit1 expression, whereas inhibition of Irx4 activity by a dominant negative mutant led to induction of Slit1 expression, indicating that Irx4 is a crucial regulator of Slit1 expression in the retina. In addition, by examining axonal behavior in the retinas with overexpression of Irx4 and using several in vivo assays to test the effect of Slit1, we found that Slit1 acts positively to guide the retinal axons inside the optic fiber layer (OFL). We further show that the regulation of Slit1 expression by Irx4 is important for providing intermediate targets for retinal axons during their growth within the retina.
Collapse
Affiliation(s)
- Zhe Jin
- Department of Medicine, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA 01605, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
You XM, Mungrue IN, Kalair W, Afroze T, Ravi B, Sadi AM, Gros R, Husain M. Conditional expression of a dominant-negative c-Myb in vascular smooth muscle cells inhibits arterial remodeling after injury. Circ Res 2003; 92:314-21. [PMID: 12595344 DOI: 10.1161/01.res.0000056758.73215.5a] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Inhibiting activity of the c-Myb transcription factor attenuates G1 to S phase cell cycle transitions in vascular smooth muscle cells (SMCs) in vitro. To determine the effects of arterial SMC-specific expression of a dominant-negative c-Myb molecule (Myb-Engrailed) on vascular remodeling in vivo, we performed carotid artery wire-denudation in 2 independent lines of binary transgenic mice with SM22alpha promoter-defined Doxycycline-suppressible expression of Myb-Engrailed. Adult mice with arterial SMC-specific expression of Myb-Engrailed were overtly normal in appearance and did not display any changes in cardiovascular structure or physiology. However, bromodeoxyuridine-defined arterial SMC proliferation, neointima formation, medial hyperplasia, and arterial remodeling were markedly decreased in mice expressing arterial SMC-restricted Myb-Engrailed after arterial injury. These data suggest that c-Myb activity in arterial SMCs is not essential for arterial structure or function during development, but is involved in the proliferation of arterial SMCs as occurs in vascular pathology, and that the expression of a dominant-negative c-Myb can dramatically reduce adverse arterial remodeling in an in vivo model of restenosis. As such, this model represents a novel tissue-specific strategy for the potential gene therapy of diseases characterized by arterial SMC proliferation.
Collapse
MESH Headings
- Animals
- Bromodeoxyuridine
- Carotid Stenosis/pathology
- Carotid Stenosis/prevention & control
- Cell Division/genetics
- Disease Models, Animal
- Gene Expression/drug effects
- Genes, Dominant
- Homeodomain Proteins/genetics
- Mice
- Mice, Transgenic
- Microfilament Proteins/genetics
- Muscle Proteins/genetics
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Promoter Regions, Genetic
- Proto-Oncogene Proteins c-myb/biosynthesis
- Proto-Oncogene Proteins c-myb/genetics
- Proto-Oncogene Proteins c-myb/pharmacology
- Tetracycline/pharmacology
- Transcription Factors
- Tunica Intima/drug effects
- Tunica Intima/metabolism
- Tunica Intima/pathology
Collapse
Affiliation(s)
- Xiao-Mang You
- Division of Cellular and Molecular Biology, The Toronto General Hospital Research Institute, Ontario, Canada
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Andersson KB, Kowenz-Leutz E, Brendeford EM, Tygsett AHH, Leutz A, Gabrielsen OS. Phosphorylation-dependent down-regulation of c-Myb DNA binding is abrogated by a point mutation in the v-myb oncogene. J Biol Chem 2003; 278:3816-24. [PMID: 12456674 DOI: 10.1074/jbc.m209404200] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The viral Myb (v-Myb) oncoprotein of the avian myeloblastosis virus (AMV) is an activated form of the cellular transcription factor c-Myb causing acute monoblastic leukemia in chicken. Oncogenic v-Myb alterations include N- and C-terminal deletions as well as point mutations. Whereas truncations in Myb cause loss of various protein modifications, none of the point mutations in v-Myb has been directly linked to protein modifications. Here we show that the DNA-binding domain of c-Myb can be phosphorylated on serine 116 by the catalytic subunit of protein kinase A. Phosphorylation of Ser(116) differentially destabilizes a subtype of c-Myb-DNA complexes. The V117D mutation of the AMV v-Myb oncoprotein abolishes phosphorylation of the adjacent Ser(116) residue. Modification of Ser(116) was also detected in live cells in c-Myb, but not in AMV v-Myb. Phosphorylation-mimicking mutants of c-Myb failed to activate the resident mim-1 gene. Our data imply that protein kinase A or a kinase with similar specificity negatively regulates c-Myb function, including collaboration with C/EBP, and that the leukemogenic AMV v-Myb version evades inactivation by a point mutation that abolishes a phosphoacceptor consensus site. This suggests a novel link between Myb, a signal transduction pathway, cooperativity with C/EBP, and a point mutation in the myb oncogene.
Collapse
|
48
|
Alexandre C, Vincent JP. Requirements for transcriptional repression and activation by Engrailed in Drosophila embryos. Development 2003; 130:729-39. [PMID: 12506003 DOI: 10.1242/dev.00286] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Genetic analysis shows that Engrailed (En), a homeodomain-containing transcription factor, has both negative and positive targets. Negative regulation is expected from a factor that has a well-defined repressor domain but activation is harder to comprehend. We used VP16En, a form of En that had its repressor domain replaced by the activation domain of VP16, to show that En activates targets using two parallel routes, by repressing a repressor and by being a bona fide activator. We identified the intermediate repressor activity as being encoded by sloppy paired 1 and 2 and showed that bona fide activation is dramatically enhanced by Wingless signaling. Thus, En is a bifunctional transcription factor and the recruitment of additional cofactors presumably specifies which function prevails on an individual promoter. Extradenticle (Exd) is a cofactor thought to be required for activation by Hox proteins. However, in thoracic segments, Exd is required for repression (as well as activation) by En. This is consistent with in vitro results showing that Exd is involved in recognition of positive and negative targets. Moreover, we provide genetic evidence that, in abdominal segments, Ubx and Abd-A, two homeotic proteins not previously thought to participate in the segmentation cascade, are also involved in the repression of target genes by En. We suggest that, like Exd, Ubx and Abd-A could help En recognize target genes or activate the expression of factors that do so.
Collapse
Affiliation(s)
- Cyrille Alexandre
- National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, UK.
| | | |
Collapse
|
49
|
Rushton JJ, Davis LM, Lei W, Mo X, Leutz A, Ness SA. Distinct changes in gene expression induced by A-Myb, B-Myb and c-Myb proteins. Oncogene 2003; 22:308-13. [PMID: 12527900 DOI: 10.1038/sj.onc.1206131] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The c-Myb, A-Myb and B-Myb transcription factors have nearly identical DNA-binding domains, activate the same reporter gene constructs in animal cells, but have different biological roles. The Myb proteins are often coexpressed in the same cells, raising questions about whether they activate similar or distinct gene expression profiles, and whether they cooperate or compete in regulating the same promoters. Here, recombinant adenoviruses were used to express each protein in human mammary cells, and then microarray assays were used to assess global changes in gene expression. Each Myb protein induced a unique and specific set of changes, displaying activities far more complex than revealed by standard reporter gene assays. These results have important implications for the roles of various Myb proteins in normal and transformed human cells, for regulatory pathways that might modify their activities and for the importance of acquired mutations that may qualitatively alter their functions in tumors.
Collapse
Affiliation(s)
- John J Rushton
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, USA
| | | | | | | | | | | |
Collapse
|
50
|
Abstract
Xenopus myogenesis is characterized by specific features, different from those of mammalian and avian systems both at the cellular level and in gene expression patterns. During early embryogenesis, after the initial molecular signals inducing mesoderm, the myogenic determination factors XMyoD and XMyf-5 are activated in presomitic mesoderm in response to mesoderm-inducing factors. After these first inductions of the myogenic program, forming muscles in Xenopus can have different destinies, some of these resulting in cell death before adulthood. In particular, it is quite characteristic of this species that, during metamorphosis, the primary myotomal myofibers completely die and are progressively replaced by secondary "adult" multinucleated myofibers. This feature offers the unique opportunity to totally separate the molecular analysis of these two distinct types of myogenesis. The aim of this review is to summarize our knowledge on the cellular and molecular events as well as the epigenetic regulations involved in the construction of Xenopus muscles during development.
Collapse
Affiliation(s)
- Christophe Chanoine
- Laboratoire de Biologie du Développement et de la Différenciation Musculaire (LNRS UMR 7060 CNRS), Paris, France.
| | | |
Collapse
|