Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Pigment Cell Melanoma Res ; 36(5): 399-406, 2023 09.
Article in English | MEDLINE | ID: mdl-37132544

ABSTRACT

The epigenetic repressor BMI1 plays an integral role in promoting the self-renewal and proliferation of many adult stem cell populations, and also tumor types, primarily through silencing the Cdkn2a locus, which encodes the tumor suppressors p16Ink4a and p19Arf . However, in cutaneous melanoma, BMI1 drives epithelial-mesenchymal transition programs, and thus metastasis, while having little impact on proliferation or primary tumor growth. This raised questions about the requirement and role for BMI1 in melanocyte stem cell (McSC) biology. Here, we demonstrate that murine melanocyte-specific Bmi1 deletion causes premature hair greying and gradual loss of melanocyte lineage cells. Depilation enhances this hair greying defect, accelerating depletion of McSCs in early hair cycles, suggesting that BMI1 acts to protect McSCs against stress. RNA-seq of McSCs, harvested before onset of detectable phenotypic defects, revealed that Bmi1 deletion derepresses p16Ink4a and p19Arf , as observed in many other stem cell contexts. Additionally, BMI1 loss downregulated the glutathione S-transferase enzymes, Gsta1 and Gsta2, which can suppress oxidative stress. Accordingly, treatment with the antioxidant N-acetyl cysteine (NAC) partially rescued melanocyte expansion. Together, our data establish a critical function for BMI1 in McSC maintenance that reflects a partial role for suppression of oxidative stress, and likely transcriptional repression of Cdkn2a.


Subject(s)
Melanoma , Skin Neoplasms , Mice , Animals , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Melanoma/metabolism , Proto-Oncogene Proteins , Skin Neoplasms/metabolism , Stem Cells/metabolism , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Pigmentation , Melanocytes/metabolism , Hair/metabolism
2.
Mol Biol Cell ; 32(20): ar1, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34260288

ABSTRACT

Multiciliated cells play critical roles in the airway, reproductive organs, and brain. Generation of multiple cilia requires both activation of a specialized transcriptional program and subsequent massive amplification of centrioles within the cytoplasm. The E2F4 transcription factor is required for both roles and consequently for multiciliogenesis. Here we establish that E2F4 associates with two distinct components of the centriole replication machinery, Deup1 and SAS6, targeting nonhomologous domains in these proteins. We map Deup1 and SAS6 binding to E2F4's N-terminus and show that this domain is sufficient to mediate E2F4's cytoplasmic role in multiciliogenesis. This sequence is highly conserved across the E2F family, but the ability to bind Deup1 and SAS6 is specific to E2F4 and E2F5, consistent with their shared roles in multiciliogenesis. By generating E2F4/E2F1 chimeras, we identify a six-residue motif that is critical for Deup1 and SAS6 binding. We propose that the ability of E2F4 and E2F5 to recruit Deup1 and/or SAS6, and enable centriole replication, contributes to their cytoplasmic roles in multiciliogenesis.


Subject(s)
Cell Cycle Proteins/metabolism , E2F4 Transcription Factor/metabolism , Microtubule-Associated Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Cell Communication/physiology , Cell Cycle/physiology , Centrioles/metabolism , Cilia/metabolism , Cytoplasm/metabolism , Cytosol/metabolism , HEK293 Cells , Humans , Protein Binding , Protein Domains
4.
Nat Commun ; 8: 15857, 2017 07 04.
Article in English | MEDLINE | ID: mdl-28675157

ABSTRACT

Abnormal development of multiciliated cells is a hallmark of a variety of human conditions associated with chronic airway diseases, hydrocephalus and infertility. Multiciliogenesis requires both activation of a specialized transcriptional program and assembly of cytoplasmic structures for large-scale centriole amplification that generates basal bodies. It remains unclear, however, what mechanism initiates formation of these multiprotein complexes in epithelial progenitors. Here we show that this is triggered by nucleocytoplasmic translocation of the transcription factor E2f4. After inducing a transcriptional program of centriole biogenesis, E2f4 forms apical cytoplasmic organizing centres for assembly and nucleation of deuterosomes. Using genetically altered mice and E2F4 mutant proteins we demonstrate that centriole amplification is crucially dependent on these organizing centres and that, without cytoplasmic E2f4, deuterosomes are not assembled, halting multiciliogenesis. Thus, E2f4 integrates nuclear and previously unsuspected cytoplasmic events of centriole amplification, providing new perspectives for the understanding of normal ciliogenesis, ciliopathies and cancer.


Subject(s)
Centrioles/metabolism , Cilia/metabolism , Cytoplasm/metabolism , E2F4 Transcription Factor/metabolism , Active Transport, Cell Nucleus , Animals , Autoantigens/metabolism , Basal Bodies/metabolism , Cell Cycle Proteins/metabolism , Cytoplasmic Granules/metabolism , Female , Humans , Male , Mice, Inbred C57BL , Transcription, Genetic
5.
Cell Cycle ; 15(2): 250-60, 2016.
Article in English | MEDLINE | ID: mdl-26825228

ABSTRACT

The E2F transcription factors are primarily implicated in the regulation of entry and exit from the cell cycle. However, in vivo studies have established additional roles for E2Fs during organ development and homeostasis. With the goal of addressing the intestinal requirements of E2f4 and E2f5, we crossed mice carrying Vil-cre, E2f4 conditional and E2f5 germline alleles. E2f4 deletion had no detectable effect on intestinal development. However, E2f4f/f;E2f5+/-;Vil-cre males, but not E2f4f/f;Vil-cre littermates, were unexpectedly sterile. This defect was not due to defective spermatogenesis. Instead, the seminiferous tubules and rete testes showed significant dilation, and spermatozoa accumulated aberrantly in the rete testis and efferent ducts. Our data show that these problems result from defective efferent ducts, a tissue whose primary function is to concentrate sperm through fluid absorption. First, Vil-cre expression, and consequent E2F4 loss, was specific to the efferent ducts and not other reproductive tract tissues. Second, the E2f4f/f;E2f5+/-;Vil-cre efferent ducts had completely lost multiciliated cells and greatly reduced levels of critical absorptive cell proteins: aquaporin1, a water channel protein, and clusterin, an endocytic marker. Collectively, the observed testis phenotypes suggest a fluid flux defect. Remarkably, we observed rete testis dilation prior to the normal time of seminiferous fluid production, arguing that the efferent duct defects promote excessive secretory activity within the reproductive tract. Finally, we also detect key aspects of these testis defects in E2f5-/- mice. Thus, we conclude that E2f4 and E2f5 display overlapping roles in controlling the normal development of the male reproductive system.


Subject(s)
E2F4 Transcription Factor/genetics , E2F5 Transcription Factor/genetics , Rete Testis/metabolism , Seminiferous Tubules/metabolism , Spermatozoa/metabolism , Animals , Aquaporin 1/genetics , Aquaporin 1/metabolism , Clusterin/genetics , Clusterin/metabolism , Crosses, Genetic , E2F4 Transcription Factor/metabolism , E2F5 Transcription Factor/metabolism , Female , Gene Expression Regulation, Developmental , Integrases/genetics , Integrases/metabolism , Male , Mice , Mice, Knockout , Rete Testis/growth & development , Rete Testis/ultrastructure , Seminiferous Tubules/growth & development , Seminiferous Tubules/ultrastructure , Signal Transduction , Spermatogenesis/genetics , Spermatozoa/cytology
6.
Genes Dev ; 29(17): 1875-89, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26314710

ABSTRACT

The retinoblastoma tumor suppressor (pRb) protein associates with chromatin and regulates gene expression. Numerous studies have identified Rb-dependent RNA signatures, but the proteomic effects of Rb loss are largely unexplored. We acutely ablated Rb in adult mice and conducted a quantitative analysis of RNA and proteomic changes in the colon and lungs, where Rb(KO) was sufficient or insufficient to induce ectopic proliferation, respectively. As expected, Rb(KO) caused similar increases in classic pRb/E2F-regulated transcripts in both tissues, but, unexpectedly, their protein products increased only in the colon, consistent with its increased proliferative index. Thus, these protein changes induced by Rb loss are coupled with proliferation but uncoupled from transcription. The proteomic changes in common between Rb(KO) tissues showed a striking decrease in proteins with mitochondrial functions. Accordingly, RB1 inactivation in human cells decreased both mitochondrial mass and oxidative phosphorylation (OXPHOS) function. RB(KO) cells showed decreased mitochondrial respiratory capacity and the accumulation of hypopolarized mitochondria. Additionally, RB/Rb loss altered mitochondrial pyruvate oxidation from (13)C-glucose through the TCA cycle in mouse tissues and cultured cells. Consequently, RB(KO) cells have an enhanced sensitivity to mitochondrial stress conditions. In summary, proteomic analyses provide a new perspective on Rb/RB1 mutation, highlighting the importance of pRb for mitochondrial function and suggesting vulnerabilities for treatment.


Subject(s)
Mitochondria/metabolism , Oxidative Phosphorylation , Retinoblastoma Protein/genetics , Animals , Cells, Cultured , Colon/physiopathology , Gene Expression Regulation , Gene Knockout Techniques , Humans , Lung/physiopathology , Mice , Mitochondria/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Proteomics , Retinoblastoma Protein/metabolism , Stress, Physiological/genetics , Transcriptome
7.
Nature ; 466(7310): 1110-4, 2010 Aug 26.
Article in English | MEDLINE | ID: mdl-20686481

ABSTRACT

Mutation of the retinoblastoma gene (RB1) tumour suppressor occurs in one-third of all human tumours and is particularly associated with retinoblastoma and osteosarcoma. Numerous functions have been ascribed to the product of the human RB1 gene, the retinoblastoma protein (pRb). The best known is pRb's ability to promote cell-cycle exit through inhibition of the E2F transcription factors and the transcriptional repression of genes encoding cell-cycle regulators. In addition, pRb has been shown in vitro to regulate several transcription factors that are master differentiation inducers. Depending on the differentiation factor and cellular context, pRb can either suppress or promote their transcriptional activity. For example, pRb binds to Runx2 and potentiates its ability to promote osteogenic differentiation in vitro. In contrast, pRb acts with E2F to suppress peroxisome proliferator-activated receptor gamma subunit (PPAR-gamma), the master activator of adipogenesis. Because osteoblasts and adipocytes can both arise from mesenchymal stem cells, these observations suggest that pRb might play a role in the choice between these two fates. However, so far, there is no evidence for this in vivo. Here we use mouse models to address this hypothesis in mesenchymal tissue development and tumorigenesis. Our data show that Rb status plays a key role in establishing fate choice between bone and brown adipose tissue in vivo.


Subject(s)
Adipose Tissue, Brown/cytology , Cell Differentiation , Cell Lineage , Osteoblasts/cytology , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/cytology , Adipose Tissue, White/metabolism , Animals , Cell Differentiation/genetics , Cell Line, Tumor , Core Binding Factor Alpha 1 Subunit/metabolism , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation , Lipoma/physiopathology , Mice , Mutation/genetics , PPAR gamma/metabolism , Sarcoma/physiopathology
8.
Genes Dev ; 24(2): 183-94, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20080954

ABSTRACT

Eukaryotes have numerous checkpoint pathways to protect genome fidelity during normal cell division and in response to DNA damage. Through a screen for G2/M checkpoint regulators in zebrafish, we identified ticrr (for TopBP1-interacting, checkpoint, and replication regulator), a previously uncharacterized gene that is required to prevent mitotic entry after treatment with ionizing radiation. Ticrr deficiency is embryonic-lethal in the absence of exogenous DNA damage because it is essential for normal cell cycle progression. Specifically, the loss of ticrr impairs DNA replication and disrupts the S/M checkpoint, leading to premature mitotic entry and mitotic catastrophe. We show that the human TICRR ortholog associates with TopBP1, a known checkpoint protein and a core component of the DNA replication preinitiation complex (pre-IC), and that the TICRR-TopBP1 interaction is stable without chromatin and requires BRCT motifs essential for TopBP1's replication and checkpoint functions. Most importantly, we find that ticrr deficiency disrupts chromatin binding of pre-IC, but not prereplication complex, components. Taken together, our data show that TICRR acts in association with TopBP1 and plays an essential role in pre-IC formation. It remains to be determined whether Ticrr represents the vertebrate ortholog of the yeast pre-IC component Sld3, or a hitherto unknown metazoan replication and checkpoint regulator.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , DNA Replication/genetics , Genes, cdc/physiology , Mitosis/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Animals , Chromatin/metabolism , DNA-Binding Proteins/metabolism , Embryo, Nonmammalian , Humans , Mutation/genetics , Phenotype , Zebrafish/genetics
9.
Proc Natl Acad Sci U S A ; 106(35): 14932-6, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19706423

ABSTRACT

The retinoblastoma tumor-suppressor protein, pRb, is a member of the pocket protein family that includes p107 and p130. These proteins have well-defined roles in regulating entry into and exit from the cell cycle and also have cell cycle-independent roles in facilitating differentiation. Here we investigate the overlap between pocket protein's function during embryonic development by using conditional mutant alleles to generate Rb;p107 double-mutant embryos (DKOs) that develop in the absence of placental defects. These DKOs die between e13.5 and e14.5, much earlier than either the conditional Rb or the germline p107 single mutants, which survive to birth or are largely viable, respectively. Analyses of the e13.5 DKOs shows that p107 mutation exacerbates the phenotypes resulting from pRb loss in the central nervous system and lens, but not in the peripheral nervous system. In addition, these embryos exhibit novel phenotypes, including increased proliferation of blood vessel endothelial cells, and heart defects, including double-outlet right ventricle (DORV). The DORV is caused, at least in part, by a defect in blood vessel endothelial cells and/or heart mesenchymal cells. These findings demonstrate novel, overlapping functions for pRb and p107 in numerous murine tissues.


Subject(s)
Embryo, Mammalian/metabolism , Heart Diseases/metabolism , Mutation , Retinoblastoma Protein/deficiency , Retinoblastoma-Like Protein p107/genetics , Animals , Apoptosis , Cell Proliferation , Central Nervous System/cytology , Central Nervous System/metabolism , Embryo, Mammalian/blood supply , Female , Gene Expression Regulation, Developmental , Heart Diseases/embryology , Heart Diseases/genetics , Heart Diseases/pathology , Lens, Crystalline/cytology , Lens, Crystalline/metabolism , Male , Mice , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Retinoblastoma-Like Protein p107/deficiency
10.
Dev Biol ; 332(1): 104-15, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19433082

ABSTRACT

The retinoblastoma gene, RB-1, was the first identified tumor suppressor. Rb(-/-) mice die in mid-gestation with defects in proliferation, differentiation and apoptosis. The activating E2F transcription factors, E2F1-3, contribute to these embryonic defects, indicating that they are key downstream targets of the retinoblastoma protein, pRB. E2F4 is the major pRB-associated E2F in vivo, yet its role in Rb(-/-) embryos is unknown. Here we establish that E2f4 deficiency reduced the lifespan of Rb(-/-) embryos by exacerbating the Rb mutant placental defect. We further show that this reflects the accumulation of trophectoderm-like cells in both Rb and Rb;E2f4 mutant placentas. Thus, Rb and E2f4 play cooperative roles in placental development. We used a conditional mouse model to allow Rb(-/-);E2f4(-/-) embryos to develop in the presence of Rb wild-type placentas. Under these conditions, Rb(-/-);E2f4(-/-) mutants survived to birth. These Rb(-/-);E2f4(-/-) embryos exhibited all of the defects characteristic of the Rb and E2f4 single mutants and had no novel defects. Taken together, our data show that pRB and E2F4 cooperate in placental development, but play largely non-overlapping roles in the development of many embryonic tissues.


Subject(s)
E2F4 Transcription Factor/metabolism , Extraembryonic Membranes/embryology , Extraembryonic Membranes/metabolism , Retinoblastoma Protein/metabolism , Anemia/embryology , Animals , Apoptosis , Biomarkers/metabolism , Cell Proliferation , E2F4 Transcription Factor/deficiency , Embryo Loss/metabolism , Embryo, Mammalian/abnormalities , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Embryonic Development , Erythrocytes/pathology , Extraembryonic Membranes/abnormalities , Extraembryonic Membranes/pathology , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Mice , Mice, Knockout , Mutation/genetics , Oligonucleotide Array Sequence Analysis , Phenotype , Placenta/metabolism , Placenta/pathology , Retinoblastoma Protein/deficiency , Survival Analysis
11.
Proc Natl Acad Sci U S A ; 105(33): 11851-6, 2008 Aug 19.
Article in English | MEDLINE | ID: mdl-18697945

ABSTRACT

Mutation of the RB-1 and p53 tumor suppressors is associated with the development of human osteosarcoma. With the goal of generating a mouse model of this disease, we used conditional and transgenic mouse strains to inactivate Rb and/or p53 specifically in osteoblast precursors. The resulting Rb;p53 double mutant (DKO) animals are viable but develop early onset osteosarcomas with complete penetrance. These tumors display many of the characteristics of human osteosarcomas, including being highly metastatic. We established cell lines from the DKO osteosarcomas to further investigate their properties. These immortalized cell lines are highly proliferative and they retain their tumorigenic potential, as judged by their ability to form metastatic tumors in immunocompromised mice. Moreover, they can be induced to differentiate and, depending on the inductive signal, will adopt either the osteogenic or adipogenic fate. Consistent with this multipotency, a significant portion of these tumor cells express Sca-1, a marker that is typically associated with stem cells/uncommitted progenitors. By assaying sorted cells in transplant assays, we demonstrate that the tumorigenicity of the osteosarcoma cell lines correlates with the presence of the Sca-1 marker. Finally, we show that loss of Rb and p53 in Sca-1-positive mesenchymal stem/progenitor cells is sufficient to yield transformed cells that can initiate osteosarcoma formation in vivo.


Subject(s)
Cell Lineage , Osteoblasts/cytology , Osteoblasts/metabolism , Osteosarcoma/metabolism , Osteosarcoma/pathology , Retinoblastoma Protein/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Antigens, Ly/metabolism , Biomarkers, Tumor/metabolism , Cell Differentiation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Disease Models, Animal , Genotype , Membrane Proteins/metabolism , Mice , Mice, Nude , Mice, Transgenic , Mutation/genetics , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Neoplasm Transplantation , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/metabolism , Osteosarcoma/genetics , Retinoblastoma Protein/genetics , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
12.
Dev Biol ; 305(2): 564-76, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17383628

ABSTRACT

The airway epithelium is comprised of specialized cell types that play key roles in protecting the lungs from environmental insults. The cellular composition of the murine respiratory epithelium is established during development and different cell types populate specific regions along the airway. Here we show that E2f4-deficiency leads to an absence of ciliated cells from the entire airway epithelium and the epithelium of the submucosal glands in the paranasal sinuses. This defect is particularly striking in the nasal epithelium of E2f4-/- mice where ciliated cells are replaced by columnar secretory cells that produce mucin-like substances. In addition, in the proximal lung, E2f4 loss causes a reduction in Clara cell marker expression indicating that Clara cell development is also affected. These defects arise during embryogenesis and, in the nasal epithelium, appear to be independent of any changes in cell proliferation, the principal process regulated by members of the E2f family of transcription factors. We therefore conclude that E2f4 is required to determine the appropriate development of the airway epithelium. Importantly, the combination of no ciliated cells and excess mucous cells can account for the chronic rhinitis and increased susceptibility to opportunistic infections that causes the postnatal lethality of E2f4 mutant mice.


Subject(s)
E2F4 Transcription Factor/physiology , Gene Expression Regulation, Developmental/physiology , Respiratory Mucosa/embryology , Animals , Cell Proliferation , Cilia/pathology , Cilia/ultrastructure , E2F4 Transcription Factor/deficiency , E2F4 Transcription Factor/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Respiratory Mucosa/pathology , Respiratory Mucosa/ultrastructure
13.
Genes Dev ; 20(22): 3117-29, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17085480

ABSTRACT

Checkpoint genes maintain genomic stability by arresting cells after DNA damage. Many of these genes also control cell cycle events in unperturbed cells. By conducting a screen for checkpoint genes in zebrafish, we found that dtl/cdt2 is an essential component of the early, radiation-induced G2/M checkpoint. We subsequently found that dtl/cdt2 is required for normal cell cycle control, primarily to prevent rereplication. Both the checkpoint and replication roles are conserved in human DTL. Our data indicate that the rereplication reflects a requirement for DTL in regulating CDT1, a protein required for prereplication complex formation. CDT1 is degraded in S phase to prevent rereplication, and following DNA damage to prevent origin firing. We show that DTL associates with the CUL4-DDB1 E3 ubiquitin ligase and is required for CDT1 down-regulation in unperturbed cells and following DNA damage. The cell cycle defects of Dtl-deficient zebrafish are suppressed by reducing Cdt1 levels. In contrast, the early G2/M checkpoint defect appears to be Cdt1-independent. Thus, DTL promotes genomic stability through two distinct mechanisms. First, it is an essential component of the CUL4-DDB1 complex that controls CDT1 levels, thereby preventing rereplication. Second, it is required for the early G2/M checkpoint.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/metabolism , G2 Phase/physiology , Mitosis/physiology , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Cullin Proteins/metabolism , DNA Damage , DNA-Binding Proteins/metabolism , Embryo, Nonmammalian/abnormalities , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/radiation effects , G2 Phase/radiation effects , Genetic Testing , HCT116 Cells , HeLa Cells , Humans , Mitosis/radiation effects , Models, Biological , Mutagenesis, Insertional , Mutation/genetics , Nuclear Proteins , Protein Binding/radiation effects , Radiation, Ionizing , Ubiquitin-Protein Ligases/metabolism , Zebrafish/embryology
14.
Development ; 130(10): 2275-87, 2003 May.
Article in English | MEDLINE | ID: mdl-12668639

ABSTRACT

Leptomeningeal glioneuronal heterotopias are a focal type of cortical dysplasia in which neural cells migrate aberrantly into superficial layers of the cerebral cortex and meninges. These heterotopias are frequently observed as microscopic abnormalities in the brains of individuals with central nervous system (CNS) malformations and epilepsy. Previous work has demonstrated that the function of Emx2, which encodes a homeodomain transcription factor, is essential for development of the cortical preplate, which gives rise to the marginal zone and subplate. However, transcriptional targets of EMX2 during CNS development are unknown. We report that leptomeningeal glioneuronal heterotopias form in Emx2(-/-) mice that are equivalent to human lesions. Additionally, we observed ectopic expression of Wnt1 in the embryonic roofplate organizer region and dorsal telencephalon. To determine the phenotypic consequences of such Wnt1 misexpression, we deleted a putative EMX2 DNA-binding site from the Wnt1 enhancer and used this to misexpress Wnt1 in the developing murine CNS. Heterotopias were detected in transgenic mice as early as 13.5 days postcoitum, consistent with a defect of preplate development during early phases of radial neuronal migration. Furthermore, we observed diffuse abnormalities of reelin- and calretinin-positive cell populations in the marginal zone and subplate similar to those observed in Emx2-null animals. Taken together, these findings indicate that EMX2 is a direct repressor of Wnt1 expression in the developing mammalian telencephalon. They further suggest that EMX2-Wnt1 interactions are essential for normal development of preplate derivatives in the mammalian cerebral cortex.


Subject(s)
Choristoma/pathology , Homeodomain Proteins/metabolism , Meninges/pathology , Neuroglia , Neurons , Proto-Oncogene Proteins/metabolism , Telencephalon/growth & development , Zebrafish Proteins , Animals , Binding Sites , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Humans , In Situ Hybridization , Mice , Mice, Transgenic , Proto-Oncogene Proteins/genetics , Reelin Protein , Transcription Factors , Transgenes , Wnt Proteins , Wnt1 Protein
15.
Proc Natl Acad Sci U S A ; 100(5): 2456-61, 2003 Mar 04.
Article in English | MEDLINE | ID: mdl-12604789

ABSTRACT

The E2F and pocket protein families are known to play an important role in the regulation of both cellular proliferation and terminal differentiation. In this study, we have used compound E2F and pocket protein mutant mouse embryonic fibroblasts to dissect the role of these proteins in adipogenesis. This analysis shows that loss of E2F4 allows cells to undergo spontaneous differentiation. The ability of E2F4 to prevent adipogenesis seems to be quite distinct from the known properties of E2F. First, it can be separated from any change in either E2F-responsive gene expression or cell cycle regulation. Second, it is a specific property of E2F4, and not other E2Fs, and it occurs independently of E2F4's ability to interact with pocket proteins. In addition, E2F4 loss does not override the differentiation defect resulting from pRB loss even though it completely suppresses the proliferation defect of Rb(-/-) mouse embryonic fibroblasts. This finding definitively separates the known, positive role of pRB in adipogenesis from its cell cycle function and shows that this pocket protein is required to act downstream of E2F4 in the differentiation process.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/physiology , Proteins , Transcription Factors/chemistry , Transcription Factors/physiology , Adipocytes/cytology , Adipocytes/metabolism , Animals , Azo Compounds/pharmacology , Blood Proteins/metabolism , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cell Cycle , Cell Differentiation , Cell Division , E2F4 Transcription Factor , Fibroblasts/metabolism , Genotype , Immunoblotting , Mice , Nuclear Proteins/metabolism , Protein Binding , Receptors, Cytoplasmic and Nuclear/metabolism , Retinoblastoma Protein/metabolism , Retinoblastoma-Like Protein p107 , Retinoblastoma-Like Protein p130 , Time Factors , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...