1
|
Schwartz I, Vunjak M, Budroni V, Cantoran García A, Mastrovito M, Soderholm A, Hinterndorfer M, de Almeida M, Hacker K, Wang J, Froussios K, Jude J, Decker T, Zuber J, Versteeg GA. SPOP targets the immune transcription factor IRF1 for proteasomal degradation. eLife 2023; 12:e89951. [PMID: 37622993 PMCID: PMC10491434 DOI: 10.7554/elife.89951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/16/2023] [Indexed: 08/26/2023] Open
Abstract
Adaptation of the functional proteome is essential to counter pathogens during infection, yet precisely timed degradation of these response proteins after pathogen clearance is likewise key to preventing autoimmunity. Interferon regulatory factor 1 (IRF1) plays an essential role as a transcription factor in driving the expression of immune response genes during infection. The striking difference in functional output with other IRFs is that IRF1 also drives the expression of various cell cycle inhibiting factors, making it an important tumor suppressor. Thus, it is critical to regulate the abundance of IRF1 to achieve a 'Goldilocks' zone in which there is sufficient IRF1 to prevent tumorigenesis, yet not too much which could drive excessive immune activation. Using genetic screening, we identified the E3 ligase receptor speckle type BTB/POZ protein (SPOP) to mediate IRF1 proteasomal turnover in human and mouse cells. We identified S/T-rich degrons in IRF1 required for its SPOP MATH domain-dependent turnover. In the absence of SPOP, elevated IRF1 protein levels functionally increased IRF1-dependent cellular responses, underpinning the biological significance of SPOP in curtailing IRF1 protein abundance.
Collapse
Affiliation(s)
- Irene Schwartz
- Department of Microbiology, Immunobiology and Genetics, Max Perutz Labs, University of ViennaViennaAustria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna BiocenterViennaAustria
| | - Milica Vunjak
- Department of Microbiology, Immunobiology and Genetics, Max Perutz Labs, University of ViennaViennaAustria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna BiocenterViennaAustria
| | - Valentina Budroni
- Department of Microbiology, Immunobiology and Genetics, Max Perutz Labs, University of ViennaViennaAustria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna BiocenterViennaAustria
| | - Adriana Cantoran García
- Department of Microbiology, Immunobiology and Genetics, Max Perutz Labs, University of ViennaViennaAustria
| | - Marialaura Mastrovito
- Department of Microbiology, Immunobiology and Genetics, Max Perutz Labs, University of ViennaViennaAustria
| | - Adrian Soderholm
- Department of Microbiology, Immunobiology and Genetics, Max Perutz Labs, University of ViennaViennaAustria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna BiocenterViennaAustria
| | - Matthias Hinterndorfer
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna BiocenterViennaAustria
- Research Institute of Molecular Pathology, Vienna BiocenterViennaAustria
| | - Melanie de Almeida
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna BiocenterViennaAustria
- Research Institute of Molecular Pathology, Vienna BiocenterViennaAustria
| | - Kathrin Hacker
- Department of Microbiology, Immunobiology and Genetics, Max Perutz Labs, University of ViennaViennaAustria
| | - Jingkui Wang
- Research Institute of Molecular Pathology, Vienna BiocenterViennaAustria
| | - Kimon Froussios
- Research Institute of Molecular Pathology, Vienna BiocenterViennaAustria
| | - Julian Jude
- Research Institute of Molecular Pathology, Vienna BiocenterViennaAustria
| | - Thomas Decker
- Department of Microbiology, Immunobiology and Genetics, Max Perutz Labs, University of ViennaViennaAustria
| | - Johannes Zuber
- Research Institute of Molecular Pathology, Vienna BiocenterViennaAustria
- Medical University of Vienna, Vienna BioCenterViennaAustria
| | - Gijs A Versteeg
- Department of Microbiology, Immunobiology and Genetics, Max Perutz Labs, University of ViennaViennaAustria
| |
Collapse
|
2
|
MYCN in Neuroblastoma: "Old Wine into New Wineskins". Diseases 2021; 9:diseases9040078. [PMID: 34842635 PMCID: PMC8628738 DOI: 10.3390/diseases9040078] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/20/2021] [Accepted: 10/27/2021] [Indexed: 12/12/2022] Open
Abstract
MYCN Proto-Oncogene, BHLH Transcription Factor (MYCN) has been one of the most studied genes in neuroblastoma. It is known for its oncogenetic mechanisms, as well as its role in the prognosis of the disease and it is considered one of the prominent targets for neuroblastoma therapy. In the present work, we attempted to review the literature, on the relation between MYCN and neuroblastoma from all possible mechanistic sites. We have searched the literature for the role of MYCN in neuroblastoma based on the following topics: the references of MYCN in the literature, the gene's anatomy, along with its transcripts, the protein's anatomy, the epigenetic mechanisms regulating MYCN expression and function, as well as MYCN amplification. MYCN plays a significant role in neuroblastoma biology. Its functions and properties range from the forming of G-quadraplexes, to the interaction with miRNAs, as well as the regulation of gene methylation and histone acetylation and deacetylation. Although MYCN is one of the most primary genes studied in neuroblastoma, there is still a lot to be learned. Our knowledge on the exact mechanisms of MYCN amplification, etiology and potential interventions is still limited. The knowledge on the molecular mechanisms of MYCN in neuroblastoma, could have potential prognostic and therapeutic advantages.
Collapse
|
3
|
Spaan CN, Smit WL, van Lidth de Jeude JF, Meijer BJ, Muncan V, van den Brink GR, Heijmans J. Expression of UPR effector proteins ATF6 and XBP1 reduce colorectal cancer cell proliferation and stemness by activating PERK signaling. Cell Death Dis 2019; 10:490. [PMID: 31227689 PMCID: PMC6588629 DOI: 10.1038/s41419-019-1729-4] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 05/18/2019] [Accepted: 05/20/2019] [Indexed: 12/15/2022]
Abstract
The unfolded protein response (UPR) acts through its downstream branches, PERK-eIF2α signaling, IRE1α-XBP1 signaling and ATF6 signaling. In the intestine, activation of the UPR through the kinase PERK results in differentiation of intestinal epithelial stem cells and colon cancer stem cells, whereas deletion of XBP1 results in increased stemness and adenomagenesis. How downstream activation of XBP1 and ATF6 influences intestinal stemness and proliferation remains largely unknown. We generated colorectal cancer cells (LS174T) that harbor doxycycline inducible expression of the active forms of either XBP1(s) or ATF61-373. Activation of either XBP1 or ATF6 resulted in reduced cellular proliferation and reduced expression of markers of intestinal epithelial stemness. Moreover, XBP1 and ATF6 activation reduced global protein synthesis and lowered the threshold for UPR activation. XBP1-mediated loss of stemness and proliferation resulted from crossactivation of PERK-eIF2α signaling and could be rescued by constitutive expression of eIF2α phosphatase GADD34. We thus find that enforced activation of XBP1 and ATF6 results in reduction of stemness and proliferation. We expose a novel interaction between XBP1 and PERK-eIF2α signaling.
Collapse
Affiliation(s)
- Claudia N Spaan
- Amsterdam UMC, University of Amsterdam, Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Meibergdreef 71, Amsterdam, The Netherlands
| | - Wouter L Smit
- Amsterdam UMC, University of Amsterdam, Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Meibergdreef 71, Amsterdam, The Netherlands
| | - Jooske F van Lidth de Jeude
- Amsterdam UMC, University of Amsterdam, Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Meibergdreef 71, Amsterdam, The Netherlands
| | - Bartolomeus J Meijer
- Amsterdam UMC, University of Amsterdam, Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Meibergdreef 71, Amsterdam, The Netherlands
| | - Vanesa Muncan
- Amsterdam UMC, University of Amsterdam, Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Meibergdreef 71, Amsterdam, The Netherlands
| | - Gijs R van den Brink
- Amsterdam UMC, University of Amsterdam, Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Meibergdreef 71, Amsterdam, The Netherlands
- Roche Innovation Center Basel, F. Hoffmann-La Roche AG, Basel, Switzerland
| | - Jarom Heijmans
- Amsterdam UMC, University of Amsterdam, Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Meibergdreef 71, Amsterdam, The Netherlands.
- Amsterdam UMC, University of Amsterdam, Department of Internal Medicine and Hematology, Meibergdreef 9, Amsterdam, The Netherlands.
| |
Collapse
|
4
|
Hacker K, Benke S, Agerer B, Scinicariello S, Budroni V, Versteeg GA. A repetitive acidic region contributes to the extremely rapid degradation of the cell-context essential protein TRIM52. Sci Rep 2019; 9:7901. [PMID: 31133683 PMCID: PMC6536530 DOI: 10.1038/s41598-019-44359-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 02/20/2019] [Indexed: 12/14/2022] Open
Abstract
Tripartite motif protein 52 (TRIM52) is a non-canonical TRIM family member harbouring the largest RING domain encoded in the human genome. In humans TRIM52 is conserved and has been under positive selection pressure, yet it has been lost in many non-primates. Competitive cell fitness assays demonstrated that TRIM52 ablation reduces cellular fitness in multiple different cell types. To better understand how this cell-essential factor is controlled, we investigated how expression of this non-canonical protein is regulated. Here, we show that TRIM52 mRNA is constitutively expressed from an intergenic region preceding the TRIM52 gene. Yet, TRIM52 protein is rapidly turned-over by the proteasome with a 3.5-minute half-life, one of the shortest in the human proteome. Consistent with this extremely rapid degradation rate, all three TRIM52 domains were identified to contribute to its instability. Intriguingly, a repetitive acidic loop in the RING domain was identified as one of the main destabilizing regions, which was unexpected given the prevailing notion that these sequences are poor proteasome substrates. This work indicates that the effect of such repetitive acidic regions on proteasomal degradation depends on the protein context, and it identifies TRIM52 as an attractive model protein to study what these contextual properties are.
Collapse
Affiliation(s)
- Kathrin Hacker
- Department of Microbiology, Immunobiology, and Genetics, Max F. Perutz Laboratories (MFPL), University of Vienna, Vienna Biocenter (VBC), Dr. Bohr-Gasse 9, 1030, Vienna, Austria
| | - Stefan Benke
- Department of Microbiology, Immunobiology, and Genetics, Max F. Perutz Laboratories (MFPL), University of Vienna, Vienna Biocenter (VBC), Dr. Bohr-Gasse 9, 1030, Vienna, Austria
| | - Benedikt Agerer
- Department of Microbiology, Immunobiology, and Genetics, Max F. Perutz Laboratories (MFPL), University of Vienna, Vienna Biocenter (VBC), Dr. Bohr-Gasse 9, 1030, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14 AKH BT25.3, 1090, Vienna, Austria
| | - Sara Scinicariello
- Department of Microbiology, Immunobiology, and Genetics, Max F. Perutz Laboratories (MFPL), University of Vienna, Vienna Biocenter (VBC), Dr. Bohr-Gasse 9, 1030, Vienna, Austria
| | - Valentina Budroni
- Department of Microbiology, Immunobiology, and Genetics, Max F. Perutz Laboratories (MFPL), University of Vienna, Vienna Biocenter (VBC), Dr. Bohr-Gasse 9, 1030, Vienna, Austria
| | - Gijs A Versteeg
- Department of Microbiology, Immunobiology, and Genetics, Max F. Perutz Laboratories (MFPL), University of Vienna, Vienna Biocenter (VBC), Dr. Bohr-Gasse 9, 1030, Vienna, Austria.
| |
Collapse
|
5
|
Song J, Lee JE. ASK1 modulates the expression of microRNA Let7A in microglia under high glucose in vitro condition. Front Cell Neurosci 2015; 9:198. [PMID: 26041997 PMCID: PMC4438231 DOI: 10.3389/fncel.2015.00198] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 05/07/2015] [Indexed: 12/26/2022] Open
Abstract
Hyperglycemia results in oxidative stress and leads to neuronal apoptosis in the brain. Diabetes studies show that microglia participate in the progression of neuropathogenesis through their involvement in inflammation in vivo and in vitro. In high-glucose-induced inflammation, apoptosis signal regulating kinase 1 (ASK1) triggers the release of apoptosis cytokines and apoptotic gene expression. MicroRNA-Let7A (miR-Let7A) is reported to be a regulator of inflammation. In the present study, we investigated whether miR-Let7A regulates the function of microglia by controlling ASK1 in response to high-glucose-induced oxidative stress. We performed reverse transcription (RT) polymerase chain reaction, Taqman assay, real-time polymerase chain reaction, and immunocytochemistry to confirm the alteration of microglia function. Our results show that miR-Let7A is associated with the activation of ASK1 and the expression of anti-inflammatory cytokine (interleukin (IL)-10) and Mycs (c-Myc and N-Myc). Thus, the relationship between Let-7A and ASK1 could be a novel target for enhancing the beneficial function of microglia in central nervous system (CNS) disorders.
Collapse
Affiliation(s)
- Juhyun Song
- Department of Anatomy, Yonsei University College of Medicine Seoul, South Korea
| | - Jong Eun Lee
- Department of Anatomy, Yonsei University College of Medicine Seoul, South Korea ; Brain Korea 21 Plus Project for Medical Sciences, Brain Research Institute, Yonsei University College of Medicine Seoul, South Korea
| |
Collapse
|
6
|
An Introspective Update on the Influence of miRNAs in Breast Carcinoma and Neuroblastoma Chemoresistance. GENETICS RESEARCH INTERNATIONAL 2014; 2014:743050. [PMID: 25548681 PMCID: PMC4273469 DOI: 10.1155/2014/743050] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 10/23/2014] [Accepted: 11/04/2014] [Indexed: 02/08/2023]
Abstract
Chemoresistance to conventional cytotoxic drugs may occur in any type of cancer and this can either be inherent or develop through time. Studies have linked this acquired resistance to the abnormal expression of microRNAs (miRNAs) that normally silence genes. At abnormal levels, miRNAs can either gain ability to silence tumour suppressor genes or else lose ability to silence oncogenes. miRNAs can also affect pathways that are involved in drug metabolism, such as drug efflux pumps, resulting in a resistant phenotype. The scope of this review is to provide an introspective analysis on the specific niches of breast carcinoma and neuroblastoma research.
Collapse
|
7
|
Rasmuson A, Segerström L, Nethander M, Finnman J, Elfman LHM, Javanmardi N, Nilsson S, Johnsen JI, Martinsson T, Kogner P. Tumor development, growth characteristics and spectrum of genetic aberrations in the TH-MYCN mouse model of neuroblastoma. PLoS One 2012; 7:e51297. [PMID: 23284678 PMCID: PMC3524187 DOI: 10.1371/journal.pone.0051297] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 11/01/2012] [Indexed: 01/24/2023] Open
Abstract
Background The TH-MYCN transgenic neuroblastoma model, with targeted MYCN expression to the developing neural crest, has been used to study neuroblastoma development and evaluate novel targeted tumor therapies. Methods We followed tumor development in 395 TH-MYCN (129X1/SvJ) mice (125 negative, 206 hemizygous and 64 homozygous mice) by abdominal palpations up to 40 weeks of age. DNA sequencing of MYCN in the original plasmid construct and mouse genomic DNA was done to verify the accuracy. Copy number analysis with Affymetrix® Mouse Diversity Genotyping Arrays was used to characterize acquired genetic aberrations. Results DNA sequencing confirmed presence of human MYCN cDNA in genomic TH-MYCN DNA corresponding to the original plasmid construct. Tumor incidence and growth correlated significantly to transgene status with event-free survival for hemizygous mice at 50%, and 0% for homozygous mice. Hemizygous mice developed tumors at 5.6–19 weeks (median 9.1) and homozygous mice at 4.0–6.9 weeks (5.4). The mean treatment window, time from palpable tumor to sacrifice, for hemizygous and homozygous mice was 15 and 5.2 days, respectively. Hemizygous mice developing tumors as early as homozygous mice had a longer treatment window. Age at tumor development did not influence treatment window for hemizygous mice, whereas treatment window in homozygous mice decreased significantly with increasing age. Seven out of 10 analysed tumors had a flat DNA profile with neither segmental nor numerical chromosomal aberrations. Only three tumors from hemizygous mice showed acquired genetic features with one or more numerical aberrations. Of these, one event corresponded to gain on the mouse equivalent of human chromosome 17. Conclusion Hemizygous and homozygous TH-MYCN mice have significantly different neuroblastoma incidence, tumor growth characteristics and treatment windows but overlap in age at tumor development making correct early genotyping essential to evaluate therapeutic interventions. Contrasting previous studies, our data show that TH-MYCN tumors have few genetic aberrations.
Collapse
Affiliation(s)
- Agnes Rasmuson
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- * E-mail: (AR); (PK)
| | - Lova Segerström
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Maria Nethander
- Genomics Core Facility, Gothenburg University, Gothenburg, Sweden
| | - Jennie Finnman
- Department of Clinical Genetics, Institute of Biomedicine, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Lotta H. M. Elfman
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Niloufar Javanmardi
- Department of Clinical Genetics, Institute of Biomedicine, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Staffan Nilsson
- Department of Mathematical Statistics, Chalmers University of Technology, Gothenburg, Sweden
| | - John Inge Johnsen
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Tommy Martinsson
- Department of Clinical Genetics, Institute of Biomedicine, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Per Kogner
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- * E-mail: (AR); (PK)
| |
Collapse
|
8
|
Sanosaka T, Namihira M, Asano H, Kohyama J, Aisaki K, Igarashi K, Kanno J, Nakashima K. Identification of genes that restrict astrocyte differentiation of midgestational neural precursor cells. Neuroscience 2008; 155:780-8. [PMID: 18640244 DOI: 10.1016/j.neuroscience.2008.06.039] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2008] [Revised: 05/22/2008] [Accepted: 06/13/2008] [Indexed: 01/21/2023]
Abstract
During development of the mammalian CNS, neurons and glial cells (astrocytes and oligodendrocytes) are generated from common neural precursor cells (NPCs). However, neurogenesis precedes gliogenesis, which normally commences at later stages of fetal telencephalic development. Astrocyte differentiation of mouse NPCs at embryonic day (E) 14.5 (relatively late gestation) is induced by activation of the transcription factor signal transducer and activator of transcription (STAT) 3, whereas at E11.5 (mid-gestation) NPCs do not differentiate into astrocytes even when stimulated by STAT3-activating cytokines such as leukemia inhibitory factor (LIF). This can be explained in part by the fact that astrocyte-specific gene promoters are highly methylated in NPCs at E11.5, but other mechanisms are also likely to play a role. We therefore sought to identify genes involved in the inhibition of astrocyte differentiation of NPCs at midgestation. We first examined gene expression profiles in E11.5 and E14.5 NPCs, using Affymetrix GeneChip analysis, applying the Percellome method to normalize gene expression level. We then conducted in situ hybridization analysis for selected genes found to be highly expressed in NPCs at midgestation. Among these genes, we found that N-myc and high mobility group AT-hook 2 (Hmga2) were highly expressed in the E11.5 but not the E14.5 ventricular zone of mouse brain, where NPCs reside. Transduction of N-myc and Hmga2 by retroviruses into E14.5 NPCs, which normally differentiate into astrocytes in response to LIF, resulted in suppression of astrocyte differentiation. However, sustained expression of N-myc and Hmga2 in E11.5 NPCs failed to maintain the hypermethylated status of an astrocyte-specific gene promoter. Taken together, our data suggest that astrocyte differentiation of NPCs is regulated not only by DNA methylation but also by genes whose expression is controlled spatio-temporally during brain development.
Collapse
Affiliation(s)
- T Sanosaka
- Laboratory of Molecular Neuroscience, Graduate School of Biological Sciences, Nara Institute of Science and Technology, 8916-5, Takayama, Ikoma, Nara 630-0101, Japan
| | | | | | | | | | | | | | | |
Collapse
|
9
|
|
10
|
Loeb-Hennard C, Kremmer E, Bally-Cuif L. Prominent transcription of zebrafish N-myc (nmyc1) in tectal and retinal growth zones during embryonic and early larval development. Gene Expr Patterns 2005; 5:341-7. [PMID: 15661639 DOI: 10.1016/j.modgep.2004.10.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2004] [Revised: 10/15/2004] [Accepted: 10/15/2004] [Indexed: 10/26/2022]
Abstract
Because of its oncogenic capacity and expression restricted to embryonic and newborn tissues, the N-myc proto-oncogene is suggested to play a key role in vertebrate organogenesis as well as in the control of cell proliferation and differentiation. To further approach the developmental function of N-myc, we cloned full-length zebrafish N-myc (nmyc1) and analyzed its expression in the embryo and early larva. nmyc1 transcription is initiated at the mid-blastula stage. At somitogenesis stages, its expression was detected in the retina, midbrain, posterior hindbrain and presumptive spinal cord. nmyc1 was also transcribed in the endoderm and its derivatives as well as in branchial arches. At later developmental stages, posterior neural expression of nmyc1 was switched off, but expression remained intense in the brain, mainly in the optic tectum, cerebellar plate and dorsal rhombomere 2. Comparison of nmyc1 transcription with proliferation zones using a M phase mitotic marker revealed that nmyc1 expression is specifically associated with mitosis in the optic tectum and the retina. This result contrasts with previous studies in other vertebrates where N-myc expression can persist in differentiating cells.
Collapse
Affiliation(s)
- Christine Loeb-Hennard
- GSF-Research Center for Environment and Health, Institute of Molecular Immunology, Marchioninistrasse 25, Munich 81377, Germany.
| | | | | |
Collapse
|
11
|
Smith KP, Byron M, O'Connell BC, Tam R, Schorl C, Guney I, Hall LL, Agrawal P, Sedivy JM, Lawrence JB. c-Myc localization within the nucleus: evidence for association with the PML nuclear body. J Cell Biochem 2005; 93:1282-96. [PMID: 15503302 DOI: 10.1002/jcb.20273] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Definitive localization of c-Myc within the nucleus is important to fully understand the regulation and function of this oncoprotein. Studies of c-Myc distribution, however, have produced conflicting results. To overcome technical challenges inherent in c-Myc cytology, we use here three methods to visualize c-Myc and in addition examine the impact of proteasome inhibition. EYFP or HA-tagged Myc was reintroduced by stable transfection into myc null diploid rat fibroblasts, replacing endogenous Myc with tagged Myc expressed at or near normal levels. This tagged Myc is shown to functionally replace the endogenous Myc by restoration of normal cell morphology and growth rate. We were able to confirm key findings using antibodies to the endogenous c-Myc and/or its partner, Max. Contrary to some published reports, by all three methods the c-Myc protein in rat fibroblasts distributes predominantly throughout the nucleus in a dispersed granular pattern, avoiding the nucleolus. Importantly, however, several findings provide evidence for an unanticipated relationship between c-Myc and PML nuclear bodies, which is enhanced under conditions of proteasome inhibition. Evidence of Max concentration within PML bodies is shown both with and without proteasome inhibition, strengthening the relationship between PML bodies and Myc/Max. Some accumulation of Myc and Max in nucleoli upon proteasome inhibition is also observed, although co-localization of ubiquitin was only seen with PML bodies. This work provides a comprehensive study of c-Myc distribution and also presents the first evidence of a relationship between turnover of this oncoprotein and PML nuclear bodies, known to break down in certain cancers.
Collapse
Affiliation(s)
- Kelly P Smith
- Department of Cell Biology, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, Massachusetts 01655, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Li H, Wu X. Histone deacetylase inhibitor, Trichostatin A, activates p21WAF1/CIP1 expression through downregulation of c-myc and release of the repression of c-myc from the promoter in human cervical cancer cells. Biochem Biophys Res Commun 2004; 324:860-7. [PMID: 15474507 DOI: 10.1016/j.bbrc.2004.09.130] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2004] [Indexed: 12/23/2022]
Abstract
Histone deacetylase (HDAC) inhibitors have shown promise in clinical cancer therapy and to consistently induce p21WAF1/CIP1 expression in a p53-independent manner and via increased acetylation of the chromatin at the Sp1 sites in the p21WAF1/CIP1 promoter region. However, the exact mechanism by which HDAC inhibitors induce p21WAF1/CIP1 remains unclear. In this study, we observed that Trichostatin A (TSA), a HDAC inhibitor, induced strikingly p21WAF1/CIP1 expression in human cervical cancer (HeLa) cells, and this induction correlated with downregulation of c-myc expression. Coincident with this observation, knock down of c-myc with a c-myc specific small interfering RNA dramatically induced expression of p21WAF1/CIP1 in these cancer cells. These data suggest that c-myc may play a critical role in repression of p21WAF1/CIP1 expression in HeLa cells. More importantly, using chromatin immunoprecipitation assay, we observed for the first time that c-myc bound to the endogenous p21WAF1/CIP1 promoter in untreated HeLa cells, but not in TSA-treated cells. Taken together, TSA induced c-myc downregulation and release from the endogenous p21WAF1/CIP1 promoter contributes, at least partially, to transcriptional activation of the p21WAF1/CIP1 in HeLa cells.
Collapse
Affiliation(s)
- Hui Li
- Institute of Medical Virology, Wuhan University School of Medicine, Wuhan, Hubei 430071, PR China.
| | | |
Collapse
|
13
|
Abstract
Neuroblastoma is the most frequently occurring solid tumour in children, with an incidence of 1.3 cases per 100000 children aged 0-14 years. Despite many advances during the past three decades, neuroblastoma has remained an enigmatic challenge to clinical and basic scientists. 20 years ago, the MYCN gene was found to be amplified in neuroblastomas, and research since then has focused on the search for other genetic markers. It has emerged that neuroblastoma cells, like cells of many other tumour types, often suffer from extensive, non-random genetic damage at multiple genetic loci. Elucidation of the exact molecular make-up of neuroblastomas will enable researchers to analyse how much specific markers, alone or in combination, can help to stratify disease in prospective studies; at present, stratification is based on age, stage, MYCN, and Shimada pathology. Neuroblastoma may be one of the first examples of the use of genetic tumour markers as a tool for defining tumour behaviour and to aid clinical staging.
Collapse
|
14
|
Kim SK, Srivastava M. Stability of Nucleolin protein as the basis for the differential expression of Nucleolin mRNA and protein during serum starvation. DNA Cell Biol 2003; 22:171-8. [PMID: 12804115 DOI: 10.1089/104454903321655792] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Nucleolin is a nucleolar phosphoprotein that plays a direct role in ribosome biogenesis. Our aim was to determine how its activity as a growth-promoting factor is coordinated with, if not regulated by, the cell cycle machinery. In serum starting and then rescuing these cells with serum, we found that the protein level did not drop in the same way that the mRNA level did. In addition, although the mRNA level rises during the immediate period during serum rescue, the protein level remained the same. We found that the protein level was maintained after serum starvation as a result of high stability. There was no selective enhanced translation of the remaining amount of Nucleolin mRNA. With regard to the constancy in protein level despite the rise in mRNA level during serum rescue, there is no concomitant degradation of newly synthesized or old protein and synthesis of new protein. Because Nucleolin has been documented to bind mRNA, APP mRNA being one among them, we propose a autoregulatory model in which Nucleolin regulates the translation of Nucleolin mRNA, such that during a period of excess protein, translation is inhibited through direct binding of Nucleolin protein to its mRNA.
Collapse
Affiliation(s)
- Steven K Kim
- Department of Anatomy, Physiology and Genetics, and Institute for Molecular Medicine, Uniformed Services University School of Medicine (USUHS), Bethesda, Maryland 20814, USA
| | | |
Collapse
|
15
|
Xie S, Lin H, Sun T, Arlinghaus RB. Jak2 is involved in c-Myc induction by Bcr-Abl. Oncogene 2002; 21:7137-46. [PMID: 12370803 DOI: 10.1038/sj.onc.1205942] [Citation(s) in RCA: 115] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2002] [Revised: 07/31/2002] [Accepted: 08/07/2002] [Indexed: 11/09/2022]
Abstract
We have previously shown that the Jak2 tyrosine kinase is activated in Bcr-Abl positive cell lines and blood cells from CML blast crisis patients by tyrosine phosphorylation. We are searching for downstream targets of Jak2 in Bcr-Abl positive cells. It is known that c-Myc expression is required for the oncogenic effects of Bcr-Abl, and that over-expression of c-Myc complements the transformation defect of the Bcr-Abl SH2 deletion mutant. Moreover, the Bcr-Abl SH2 deletion mutant and an Abl C-terminal deletion mutant are deficient in activating c-Myc expression. Since the Jak2 binds to the C-terminal domain of Bcr-Abl and optimal Jak2 activation requires the SH2 domain, we tested whether Jak2 was involved in c-Myc protein induction by Bcr-Abl. We treated the 32Dp210 Bcr-Abl cells with the Jak2 specific tyrosine kinase inhibitor, AG490, and found that this drug, like the Abl tyrosine kinase inhibitor STI-571, inhibited c-Myc protein induction by Bcr-Abl. Treatment of 32Dp210 Bcr-Abl cells with AG490 also inhibited c-MYC RNA expression. It is also known that c-Myc protein is a labile protein that is increased in amounts in response to various growth factors by a mechanism not involving new Myc protein formation. Treatment of 32Dp210 Bcr-Abl cells with both the proteasome inhibitor MG132 and AG490 blocked the reduction of the c-Myc protein observed by AG490 alone. An adaptor protein SH2-Bbeta is involved in the enhancement of the tyrosine kinase activity of Jak2 following ligand/receptor interaction. In this regard we showed that the Jak2/Bcr-Abl complex contains SH2-Bbeta. Expression of the SH2-Bbeta R555E mutant in 32Dp210 Bcr-Abl cells reduced c-Myc expression about 40% compared to a vector control. Interestingly, we found the reduction of the c-Myc protein in several clones of dominant-negative (DN) Jak2 expressing K562 cells correlated very well with the reduction of tumor growth of these cells in nude mice as compared to vector transfected K562 cells. Both STI-571 and AG490 also induced apoptosis in 32Dp210 cells. Of interest, IL-3 containing medium reversed the STI-571 induced apoptosis of 32Dp210 cells but did not reverse the induction of apoptosis by AG490, which strongly supports the specificity of the inhibitory effects of AG490 on the Jak2 tyrosine kinase. In summary, our findings indicate that Jak2 mediates the increase in c-Myc expression that is induced by Bcr-Abl. Our results indicate that activated Jak2 not only mediates an increase of c-MYC RNA expression but also interferes with proteasome-dependent degradation of c-Myc protein.
Collapse
Affiliation(s)
- Shanhai Xie
- Department of Molecular Pathology, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Blvd, Houston, Texas, TX 77030, USA
| | | | | | | |
Collapse
|
16
|
Abstract
During the last 30 years, investigation of the transcriptional and translational mechanisms of gene regulation has been a major focus of molecular cancer biology. More recently, it has become evident that cancer-related mutations and cancer-related therapies also can affect post-translational processing of cellular proteins and that control exerted at this level can be critical in defining both the cancer phenotype and the response to therapeutic intervention. One post-translational mechanism that is receiving considerable attention is degradation of intracellular proteins through the multicatalytic 26S proteasome. This follows growing recognition of the fact that protein degradation is a well-regulated and selective process that can differentially control intracellular protein expression levels. The proteasome is responsible for the degradation of all short-lived proteins and 70-90% of all long-lived proteins, thereby regulating signal transduction through pathways involving factors such as AP1 and NFKB, and processes such as cell cycle progression and arrest, DNA transcription, DNA repair/misrepair, angiogenesis, apoptosis/survival, growth and development, and inflammation and immunity, as well as muscle wasting (e.g. in cachexia and sepsis). In this review, we discuss the potential involvement of the proteasome in both cancer biology and cancer treatment.
Collapse
Affiliation(s)
- F Pajonk
- Department of Radiation Therapy, Radiological University Clinic, Hugstetter Str. 55, 79106 Freiburg i. Brsg., Germany.
| | | |
Collapse
|
17
|
Tani E, Kitagawa H, Ikemoto H, Matsumoto T. Proteasome inhibitors induce Fas-mediated apoptosis by c-Myc accumulation and subsequent induction of FasL message in human glioma cells. FEBS Lett 2001; 504:53-8. [PMID: 11522296 DOI: 10.1016/s0014-5793(01)02770-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Proteasome inhibitors were shown previously to induce mitochondria-independent and caspase-3-dependent apoptosis in human glioma cell lines by unknown mechanisms. Here, we showed that treatment with proteasome inhibitors, lactacystin or acetyl-leucinyl-leucinyl-norleucinal, led to elevation of the steady-state c-Myc protein but not c-myc mRNA, suggesting the accumulation of c-Myc protein by proteasome inhibitors. In addition, the marked association of c-Myc protein with ubiquitin by treatment with proteasome inhibitors indicated the involvement of proteasome in c-Myc proteolysis and the stabilization of c-Myc protein by proteasome inhibitors in vivo. The expression of Fas (also termed CD95 or APO-1) mRNA, if analyzed by reverse transcriptase polymerase chain reaction assay, was found to occur constitutively, and increased slightly by the treatment with proteasome inhibitors. In contrast, the expression of Fas ligand (FasL) mRNA was markedly induced temporarily before the activation of caspase-3 by the treatment. Agonistic anti-Fas antibody (CH11) induced apoptotic cell death, suggesting the presence of a functional Fas receptor. In addition, proteasome inhibitor-induced apoptosis was prevented by the addition of antagonistic anti-FasL antibody (4A5) or z-IETD.fmk, a potent inhibitor of caspase-8, indicating the involvement of the Fas receptor-ligand apoptotic signaling system in proteasome inhibitor-mediated apoptosis. Thus, it is suggested that proteasome inhibitors cause the accumulation of c-Myc protein which induces transiently FasL message to stimulate the Fas receptor-ligand apoptotic signaling pathway.
Collapse
Affiliation(s)
- E Tani
- Molecular Research Laboratory, Department of Neurosurgery, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, 663-8501, Hyogo, Japan.
| | | | | | | |
Collapse
|
18
|
Jackson J, Ramsay G, Sharkov NV, Lium E, Katzen AL. The role of transcriptional activation in the function of the Drosophila myb gene. Blood Cells Mol Dis 2001; 27:446-55. [PMID: 11500058 DOI: 10.1006/bcmd.2001.0403] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Vertebrate myb genes encode DNA-binding proteins that regulate transcription and have been implicated in regulation of cell proliferation, differentiation, and apoptosis. We have demonstrated that the single myb gene in Drosophila melanogaster, Dm myb, is required for the G(2)/M transition of the cell cycle and for suppression of endoreduplication. Recently, it has become apparent that the family of proteins containing Myb-related DNA-binding domains is much larger than originally believed and that the biochemical properties and functions of these proteins are diverse. We undertook studies to characterize the biochemical properties of the Drosophila Myb protein (DMyb). We now provide evidence that in addition to having homology with the vertebrate Myb proteins, the Drosophila Myb protein (DMyb) shares its biochemical properties. DMyb binds to a similar consensus sequence and activates transcription from a reporter construct regulated by vertebrate Myb proteins. We also show that DMyb proteins carrying mutations corresponding to previously isolated mutant alleles of Dm myb are less active as transcriptional activators than wild-type DMyb, indicating that a decrease in transcriptional activation ability is likely to cause the mutant phenotypes.
Collapse
Affiliation(s)
- J Jackson
- The G. W. Hooper Foundation, University of Illinois at Chicago, College of Medicine, Chicago, Illinois 60607-7170, USA
| | | | | | | | | |
Collapse
|
19
|
Noguchi K, Kokubu A, Kitanaka C, Ichijo H, Kuchino Y. ASK1-signaling promotes c-Myc protein stability during apoptosis. Biochem Biophys Res Commun 2001; 281:1313-20. [PMID: 11243879 DOI: 10.1006/bbrc.2001.4498] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We previously reported that JNK is involved in the regulation of c-Myc-mediated apoptosis triggered by UV irradiation and anticancer drug treatment. Here we show that ASK1 is an upstream regulator for c-Myc-mediated apoptosis triggered by UV, and we found a direct role for Ser-62 and Ser-71 in the regulation of protein stability and function of c-Myc. The ASK1-JNK pathway enhanced the protein stability of c-Myc through phosphorylation at Ser-62 and Ser-71, which was required for c-Myc-dependent apoptosis by ASK1-signaling. Interestingly, ASK1-signaling attenuated the degradation of ubiquitinated c-Myc without affecting the ubiquitination process. Together, these findings indicate that the ASK1-JNK pathway promotes the proapoptotic activity of c-Myc by modulating c-Myc protein stability through phosphorylation at Ser-62 and Ser-71.
Collapse
Affiliation(s)
- K Noguchi
- Department of Bioactive Molecules, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan.
| | | | | | | | | |
Collapse
|
20
|
Batsché E, Crémisi C. Opposite transcriptional activity between the wild type c-myc gene coding for c-Myc1 and c-Myc2 proteins and c-Myc1 and c-Myc2 separately. Oncogene 1999; 18:5662-71. [PMID: 10523846 DOI: 10.1038/sj.onc.1202927] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
E-cadherin expression was previously shown to be activated by RB and c-myc specifically in epithelial cells, through interaction with the AP-2 transcription factor. Here we show that only a wild type c-myc gene, coding for the two c-Myc proteins c-Myc2 and c-Myc1, was able to transactivate the E-cadherin promoter, in contrast to c-Myc2 or c-Myc1 alone which strongly repressed E-cadherin in both epithelial cells and fibroblasts. In addition, overexpression of c-myc2 or c-myc1 inhibited c-myc and RB-mediated activation in a dose-dependent manner, suggesting that the ratio of the two c-Myc proteins is essential for transactivation. We also showed by using several mutants of the E-cadherin promoter, that the AP-2 binding sites were the main target of c-myc2- and c-myc1-mediated repression. AP-2-mediated inhibition was cell-type specific, as was the activation. Nevertheless, when high amounts of c-myc2 and c-myc1 were used, a second c-myc-mediated repression was observed, possibly mediated by the Inr sequence of the E-cadherin promoter. However, this repression was independent of cell type. Our results suggest a new way to regulate c-myc transcriptional activity by interfering with the ratio of the two c-myc proteins, which has already been found to be disrupted in vivo in several tumor types.
Collapse
Affiliation(s)
- E Batsché
- CJF INSERM 94-02, Université René Descartes, 45 rue des Saints-Pères, 75270 Paris cedex 06, France
| | | |
Collapse
|
21
|
Abstract
Various experiments have demonstrated a collaborative action of Myc and Ras, both in normal cell growth control as well as during oncogenesis. We now show that Ras enhances the accumulation of Myc activity by stabilizing the Myc protein. Whereas Myc has a very short half-life when produced in the absence of mitogenic signals, due to degradation by the 26S proteasome, the half-life of Myc increases markedly in growth-stimulated cells. This stabilization is dependent on the Ras/Raf/MAPK pathway and is not augmented by proteasome inhibition, suggesting that Ras inhibits the proteasome-dependent degradation of Myc. We propose that one aspect of Myc-Ras collaboration is an ability of Ras to enhance the accumulation of transcriptionally active Myc protein.
Collapse
Affiliation(s)
- R Sears
- Department of Genetics, Howard Hughes Medical Institute, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | |
Collapse
|
22
|
Abstract
Gene amplification refers to a genomic change that results in an increased dosage of the gene(s) affected. Amplification represents one of the major molecular pathways through which the oncogenic potential of proto-oncogenes is activated during tumorigenesis. The architecture of amplified genomic structures is simple in some tumor types, involving in the vast majority of cases only one gene, such as MYCN in neuroblastomas. On the other hand, it can be complex and discontinuous, involving several syntenic co-amplified genes, such as in the 11q13 amplification in breast cancer, although in many of these cases there may be a single target gene. The presence of different nonsyntenic amplified genes raises the possibility that cells of certain tumors are susceptible to independent amplification events. In general, the amplified genes do not undergo additional damage by mutations. The data indicate that it is the enhanced level of a wild-type protein that contributes to tumorigenesis.
Collapse
Affiliation(s)
- M Schwab
- Cytogenetics Division, German Cancer Research Center, Heidelberg, Germany.
| |
Collapse
|
23
|
Ambros IM, Rumpler S, Luegmayr A, Hattinger CM, Strehl S, Kovar H, Gadner H, Ambros PF. Neuroblastoma cells can actively eliminate supernumerary MYCN gene copies by micronucleus formation--sign of tumour cell revertance? Eur J Cancer 1997; 33:2043-9. [PMID: 9516850 DOI: 10.1016/s0959-8049(97)00204-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Human neuroblastoma cell lines frequently exhibit MYCN amplification and many are characterised by the presence of morphologically distinct cell types. The neuronal cells (N-cells) and the so-called flat cells (F-cells) are thought to represent manifestations of different neural crest cell lineages and are considered to be the consequence of neuroblastoma cell pluripotency. In this study, various neuroblastoma cell lines were examined for micronuclei. In F-cells of neuroblastoma cell lines with extrachromosomally amplified MYCN, we observed the frequent occurrence of micronuclei. Using fluorescence in situ hybridisation (FISH) with a MYCN specific probe, we demonstrated that these micronuclei were packed with MYCN hybridisation signals. In addition, in a minor percentage of cells, MYCN signals occurred in clusters, adhered to the nuclear membrane and aggregated in nuclear protrusions. In F-cells, a substantial reduction or lack of amplified MYCN copies was observed. These observations let us conclude that extrachromosomally amplified genes can be actively eliminated from the nucleus resulting in a dramatic loss of amplified sequences in the F-cells. Moreover, reduction or loss of amplified sequences in F-cells was shown to be accompanied by downregulation of MYCN expression, by a decrease in proliferative activity and by upregulation of molecules of the major histocompatibility complex class I (MHC I). Interestingly, F-cells are not restricted to neuroblastoma cell cultures, but also occur in cell lines of other tissue origin. All F-cells share important biological features, interpreted as cell revertance, i.e. loss of the malignant phenotype and properties. This fact, together with the demonstration that neuroblastoma cells do not differentiate into Schwann cells in vivo [1] Ambros et al. NEJM 1996, 334, 1505-1511, do not support the hypothesis that F-cells represent Schwannian/glial differentiation in vitro. We therefore postulate that the elimination of amplified MYCN gene copies in cultivated neuroblastoma cells is in line with the phenomenon of tumour cell revertance.
Collapse
Affiliation(s)
- I M Ambros
- Children's Cancer Research Institute CCRI, St. Anna Kinderspital, Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Wakamatsu Y, Watanabe Y, Nakamura H, Kondoh H. Regulation of the neural crest cell fate by N-myc: promotion of ventral migration and neuronal differentiation. Development 1997; 124:1953-62. [PMID: 9169842 DOI: 10.1242/dev.124.10.1953] [Citation(s) in RCA: 119] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
During neural crest development in avian embryos, transcription factor N-myc is initially expressed in the entire cell population. The expression is then turned off in the period following colonization in ganglion and nerve cord areas except for the cells undergoing neuronal differentiation. This was also recapitulated in the culture of Japanese quail neural crest, and the cells expressing N-myc eventually coincided with those expressing neurofilaments. These findings suggested that N-myc is involved in regulation of neuronal differentiation in the neural crest cell population. In fact, transient overexpression of N-myc in the neural crest culture by transfection resulted in a remarkable promotion of neuronal differentiation. An experimental procedure was developed to examine the effect of exogenous N-myc expression in the neural crest cells in embryos. Neural crest cell clusters still attached to the neural tube were excised from Japanese quail embryos, transfected and grafted into chicken host embryos. Using this chimera technique, we were able to analyze the consequence of transient high N-myc during the early phase of neural crest migration. Two effects were demonstrated in the embryos: first, high N-myc expression provoked massive ventral migration of the neural crest population and, second, those cells that migrated to the ganglion-forming areas underwent neuronal differentiation with the cell type determined by the nature of the ganglion. Thus, N-myc is involved in regulation of the neural crest fate in two different aspects: ventral migration and neuronal differentiation.
Collapse
Affiliation(s)
- Y Wakamatsu
- Institute for Molecular and Cellular Biology, Osaka University, Suita, Japan
| | | | | | | |
Collapse
|
25
|
Ogata T. Fluid flow induces enhancement of the Egr-1 mRNA level in osteoblast-like cells: involvement of tyrosine kinase and serum. J Cell Physiol 1997; 170:27-34. [PMID: 9012782 DOI: 10.1002/(sici)1097-4652(199701)170:1<27::aid-jcp4>3.0.co;2-n] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
It is widely accepted that mechanical loading is necessary to construct the architecture of bone and to maintain bone mass. However, the mechanism of how bone cells respond to mechanical stimuli is not known. To clarify this, we stimulated osteoblast-like MC3T3E1 cells by mechanical shaking of the culture dishes and found that the level of the egr-1 gene, which is an early response gene induced by growth factors or serum and encodes a transcription factor, increased 15-45 min after the shaking, with a peak at 30 min. The egr-1 gene product increased 1 h after the shaking. The egr-1 gene elevation was not blocked by prior exposure to indomethacin, saralasin, Rp-cAMP, A23187, and colchicine, and it was blocked partially by cytochalasin D, H-7, and prolonged exposure to TPA. On the other hand, a prior incubation with cycloheximide, DRB, genistein, herbimycin A, and BAPTA/AM completely blocked the egr-1 gene level enhanced by shaking the culture dishes. Moreover, we found that in serum-deprived cells the egr-1 gene response to shaking was not induced. These results suggested that the egr-1 gene response is regulated at the transcriptional level and that it involves tyrosine kinase as well as labile or de novo protein and requires a particular level of intracellular calcium and serum.
Collapse
Affiliation(s)
- T Ogata
- Department of Molecular Pharmacology, Tokyo Medical and Dental University, Japan
| |
Collapse
|
26
|
Barr FG, Nauta LE, Davis RJ, Schäfer BW, Nycum LM, Biegel JA. In vivo amplification of the PAX3-FKHR and PAX7-FKHR fusion genes in alveolar rhabdomyosarcoma. Hum Mol Genet 1996; 5:15-21. [PMID: 8789435 DOI: 10.1093/hmg/5.1.15] [Citation(s) in RCA: 108] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
In the pediatric cancer alveolar rhabdomyosarcoma, characteristic t(2;13)(q35;q14) or variant t(1;13)(p36;q14) chromosomal translocations generate PAX3-FKHR or PAX7-FKHR fusion genes. Using fluorescence in situ hybridization, reverse transcriptase-polymerase chain reaction and quantitative Southern blot analyses, we demonstrate that these fusion genes are amplified in 20% of fusion-positive tumors. In particular, we found in vivo amplification of these fusions in one of 22 PAX3-FKHR-positive cases and five of seven PAX7-FKHR-positive cases. These findings indicate that translocation and amplification can occur sequentially in a cancer to alter both the structure and copy number of a gene and thereby activate oncogenic activity by complementary mechanisms.
Collapse
Affiliation(s)
- F G Barr
- Department of Pathology, University of Pennsylvania School of Medicine, Philadelphia 19104, USA
| | | | | | | | | | | |
Collapse
|
27
|
Judware R, Lechner R, Culp LA. Inverse expressions of the N-myc oncogene and beta 1 integrin in human neuroblastoma: relationships to disease progression in a nude mouse model system. Clin Exp Metastasis 1995; 13:123-33. [PMID: 7533687 DOI: 10.1007/bf00133617] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
A nude mouse model for human neuroblastoma has been developed to examine possible relationships between amplification/over-expression of the N-myc oncogene and altered regulation of expression of specific integrin subunits during tumor progression. Subcutaneous (ectopic) or intra-adrenal (orthotopic) injection of the neuroblastoma cell lines SK-N-SH or IMR-32 has generated a number of derivative tumor cell lines. Tumor cell lines derived from SK-N-SH cells (which do not express N-myc) or IMR-32 cells (which over-express N-myc) produce tumors at higher rates when re-injected into the subcutaneous space of nude mice. Moreover, cell lines derived from tumors initiated by IMR-32 cells exhibit shorter latent periods than do IMR-32 cells direct from tissue culture. With regard to integrin subunit expression, SK-N-SH and related cell lines express high levels of beta 1 integrin, which is associated with the alpha 2 and alpha 3 integrin subunits (predominantly alpha 3). IMR-32 cells display reduced beta 1 expression, and that which is produced is not associated with common alpha subunits. LaN1 cells, which express N-myc at even higher levels than do IMR-32 cells, express even less beta 1. Interestingly, the tumor-derived cell lines (especially those from tumors initiated in adrenal glands) also exhibit reduced integrin expression compared with the parental cell lines; this reduction is associated with the enhanced tumor take rate observed when the cells are re-injected into nude mice. Our results raise the possibility of a relationship between over-expression of N-myc and down-regulation of beta 1 integrin expression (possibly some alpha subunits also). In addition, the data suggest that human neuroblastoma-derived cell lines which exhibit reduced integrin expression display more aggressive tumor growth in nude mice.
Collapse
Affiliation(s)
- R Judware
- Department of Molecular Biology and Microbiology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106
| | | | | |
Collapse
|
28
|
Hong Y, Frame M, Coussens PM. A 14-kDa immediate-early phosphoprotein is specifically expressed in cells infected with oncogenic Marek's disease virus strains and their attenuated derivatives. Virology 1995; 206:695-700. [PMID: 7831828 DOI: 10.1016/s0042-6822(95)80091-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Previously, we reported two cDNAs derived from the Marek's disease virus (MDV) long internal repeat region. A 14-kDa polypeptide (p14) encoded by two small open reading frames (ORFs) from at least two distinct cDNAs is expressed in cells lytically infected with both oncogenic and attenuated MDV as well as in cells latently infected and transformed by MDV. In this study, we demonstrate that p14 is serotype 1 specific and highly phosphorylated. Given the degree of phosphorylation and lack of homology to known proteins, we propose the name pp14 for the polypeptide encoded by ORF1a and ORF1b. Further analysis reveals that pp14 is predominantly found in cytoplasmic fractions of MDV-infected cells and can be detected in the cytoplasm of MDV-infected cells by immunofluorescence with polyclonal antisera prepared against pp14-glutathione S-transferase fusion protein.
Collapse
Affiliation(s)
- Y Hong
- Department of Animal Science, Michigan State University, East Lansing 48824
| | | | | |
Collapse
|
29
|
Hann SR. Methionine deprivation regulates the translation of functionally-distinct c-Myc proteins. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1995; 375:107-16. [PMID: 7645422 DOI: 10.1007/978-1-4899-0949-7_10] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Numerous studies have demonstrated a critical role for the c-myc gene in the control of cellular growth. Alterations of the c-myc gene have been found associated with many different types of tumors in several species, including humans. The increased synthesis of one of the major forms of c-Myc protein, c-Myc 1, upon methionine deprivation provides a link between the regulation of oncogenes and the nutritional status of the cell. While deregulation or overexpression of the other major form, c-Myc 2, has been shown to cause tumorigenesis, the synthesis of c-Myc 1 protein is lost in many tumors. This suggests that the c-Myc 1 protein is necessary to keep the c-Myc 2 protein "in check" and prevent certain cells from becoming tumorigenic. Indeed, we have shown that overproduction of c-Myc 1 can inhibit cell growth. We have also shown that c-Myc 1 and 2 proteins have a differential molecular function in the regulation of transcription through a new binding site of Myc/Max heterodimers. We have also recently identified new translational forms of the c-Myc protein which we term delta-c-Myc. These proteins arise from translational initiation at downstream start sites which yield N-terminally-truncated c-Myc proteins. Since these proteins lack a significant portion of the transactivation domain of c-Myc, they behave as dominant-negative inhibitors of the full-length c-Myc 1 and 2 proteins. The synthesis of delta-c-Myc proteins is also regulated during cell growth and is repressed by methionine deprivation. Therefore, the synthesis of c-Myc 1 and delta-c-Myc proteins are reciprocally regulated by methionine availability. We have also found some tumor cell lines which synthesize high levels of the delta-c-Myc proteins. Taken together, our data suggest that c-Myc function is dependent on the levels of these different translational forms of c-Myc protein which are regulated by the nutritional status of the cell during growth. Numerous reports have demonstrated a fundamental and diverse role for the myc gene in cellular events, including proliferation, differentiation and apoptosis (Cole 1986; Spencer and Groudine 1991; Askew et al. 1991; Evan et al. 1992). This is dramatically illustrated by the frequent occurrence of a variety of tumors in many species having alterations of myc genes and the transduction of c-myc sequences by retroviruses (Spencer and Groudine 1991).4+ Eisenman 1990).(ABSTRACT TRUNCATED AT 400 WORDS)
Collapse
Affiliation(s)
- S R Hann
- Department of Cell Biology, Vanderbilt Univeristy, School of Medicine, Nashville, Tennessee 37232-2175, USA
| |
Collapse
|
30
|
Abstract
The oncogenic activation by amplification of the MYCN gene is frequently observed in human neuroblastomas and occasionally in other tumours with neuronal qualities. As a consequence of amplification, elevated levels of the mycN protein are expressed. mycN contains a C-terminal basic region (BR) that can bind to DNA, and a helix-loop-helix (HLH)-leucine zipper (Zip) domain, which is responsible for the physical interaction with another HLH-Zip protein, max. This principle structure is conserved among all members of the MYC gene family. The resulting dimers can bind to the DNA sequence CACGTG. The mycN protein, but not max, contains, near the N-terminus, a region conferring the ability to activate the transcription of genes. mycN/max heterodimers probably activate and max/max homodimers repress transcription of, as yet, unidentified target genes. In neuroblastoma cells, where mycN is deregulated, the balanced interaction of BR-HLH-Zip proteins is probably perturbed, and, therefore, genes controlled by mycN might be abnormally expressed and thereby alter normal cell growth with the consequence of tumorigenesis.
Collapse
Affiliation(s)
- A Wenzel
- Department of Cytogenetics, German Cancer Research Center, Heidelberg, Germany
| | | |
Collapse
|
31
|
Degradation of the tumor suppressor protein p53 by the ubiquitin-mediated proteolytic system requires a novel species of ubiquitin-carrier protein, E2. J Biol Chem 1994. [DOI: 10.1016/s0021-9258(17)36921-1] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
32
|
Abstract
N-myc expression in the mouse embryo was examined during the late cochlear organogenesis. Tissue distribution of N-myc expression was histologically analyzed by in situ hybridization of the transcript in the cochlea between 15 and 18 days of gestation. At 15 days of gestation, N-myc expression was found very conspicuous in nervous structure of the cochlea such as the auditory nerve and the spiral ganglion. Moreover, N-myc was also present in the Köllikers organ and in the epithelium surrounding the cochlear canal. A few days later, N-myc expression was still clearly present in the Köllikers organ but less so in nervous structures. This study shows that cochlear tissues derived from the otic placode present a significant level of N-myc transcript during late embryogenesis. N-myc expression seems to be related to cell differentiation in the inner ear.
Collapse
Affiliation(s)
- R Romand
- Laboratoire de Neurobiologie et Physiologie du développement, Université Blaise Pascal Clermont-II, Aubière, France
| | | | | |
Collapse
|
33
|
Imamura Y, Nakagawa T, Iguchi-Ariga SM, Ariga H. Transcriptional regulation of the N-myc gene: identification of positive regulatory element and its double- and single-stranded DNA binding proteins. BIOCHIMICA ET BIOPHYSICA ACTA 1993; 1216:273-85. [PMID: 8241268 DOI: 10.1016/0167-4781(93)90154-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The N-myc gene is amplified and overexpressed in neuroblastoma, retinoblastoma and small cell lung carcinoma, and is considered to be related to cell proliferation and/or differentiation. The transcriptional regulatory sequences of the c-myc gene have been already identified, while those of N-myc have remained obscure for a long time. In this report, we have identified several positive and negative transcriptional regulatory elements in the upstream region of the mouse N-myc gene. Among them, an activating sequence spanning -860 to -797 bp (63 bp) could be reduced to a functional core of 21 bp from -846 to -826. This sequence, termed N21 box, worked as a positive transcriptional element when linked directly upstream (but not downstream) of the putative N-myc promoter in HeLa, not in IMR32 cells. At least two proteins, of 42 kDa and 100 kDa, bound to the double-stranded N21 box, and were expressed in HeLa as well as in IMR32 cells. Moreover, the plus strand of N21 box could be specifically bound by a species of 42 kDa from either cell type and by a 37 kDa protein found only in HeLa cells. These proteins may be factors binding to positive transcriptional regulatory elements and may have a role in the regulation of N-myc expression.
Collapse
Affiliation(s)
- Y Imamura
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | | | | | | |
Collapse
|
34
|
Cheng JM, Hiemstra JL, Schneider SS, Naumova A, Cheung NK, Cohn SL, Diller L, Sapienza C, Brodeur GM. Preferential amplification of the paternal allele of the N-myc gene in human neuroblastomas. Nat Genet 1993; 4:191-4. [PMID: 8102299 DOI: 10.1038/ng0693-191] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Genomic imprinting plays a role in influencing the parental origin of genes involved in cancer-specific rearrangements. We have analysed 22 neuroblastomas with N-myc amplification to determine the parental origin of the amplified N-myc allele and the allele that is deleted from chromosome 1p. We analysed DNA from neuroblastoma patients and their parents, using four polymorphisms for 1p and three for the N-myc amplicon. We determined that the paternal allele of N-myc was preferentially amplified (12 out of 13 cases; P = 0.002). However, the paternal allele was lost from 1p in six out of ten cases, consistent with a random distribution (P > 0.2). These results suggest that parental imprinting influences which N-myc allele is amplified in neuroblastomas, but it does not appear to affect the 1p allele that is deleted in the cases that we have examined.
Collapse
Affiliation(s)
- J M Cheng
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri 63110
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Sawai S, Shimono A, Wakamatsu Y, Palmes C, Hanaoka K, Kondoh H. Defects of embryonic organogenesis resulting from targeted disruption of the N-myc gene in the mouse. Development 1993; 117:1445-55. [PMID: 8404543 DOI: 10.1242/dev.117.4.1445] [Citation(s) in RCA: 135] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The highest expression of the N-myc gene occurs during embryonic organogenesis in the mouse ontogeny, with the peak of expression around embryonic day 9.5. Homozygous N-myc-deficient mice, produced by germline transmission of a disrupted allele in ES cells, developed normally to day 10.5, indicating dispensability of N-myc expression in the earlier period, but later accumulated organogenic abnormalities and died around day 11.5. The most notable abnormalities were found in the limb bud, visceral organs (lung, stomach, liver and heart) and the central/peripheral nervous systems, and were highly correlated with the site of N-myc expression. The limb buds and the lungs excised from N-myc-deficient mutant embryos were placed in culture to allow their development to stages beyond the point of death of the embryos. Analyses indicated that the mutant limbs failed to develop distal structures and the development of bronchi from the trachea was defective in the lungs. The latter defect was largely corrected by addition of fetal calf serum to the culture medium, suggesting that an activity missing in the mutant lung was replenished by a component of the serum. The phenotype of N-myc-deficient mutant embryos indicated requirement of the N-myc function in many instances of tissue interactions in organogenesis and also in cell-autonomous regulation of tissue maturation.
Collapse
Affiliation(s)
- S Sawai
- Department of Molecular Biology, School of Science, Nagoya University, Japan
| | | | | | | | | | | |
Collapse
|
36
|
Cook RM, Miller YE, Bunn PA. Small cell lung cancer: etiology, biology, clinical features, staging, and treatment. Curr Probl Cancer 1993; 17:69-141. [PMID: 8395998 DOI: 10.1016/0147-0272(93)90010-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Lung cancer is the leading cause of cancer death in the United States. Small cell lung cancer (SCLC) accounts for 20% to 25% of all bronchogenic carcinoma and is associated with the poorest 5-year survival of all histologic types. SCLC differs in its etiologic, pathologic, biologic, and clinical features from non-SCLC, and these differences have translated to distinct approaches to its prevention and treatment. Compared with other histologic types of lung cancer, exposures to tobacco smoke, ionizing radiation, and chloromethyl ethers pose a substantially greater risk for development of SCLC. The histologic classification of SCLC has been revised to include three categories: (1) small cell carcinoma, (2) mixed small cell/large cell, and (3) combined small cell carcinoma. Ultrastructurally, SCLC displays a number of neuroendocrine features in common with pulmonary neuroendocrine cells, including dense core vesicles or neurosecretory granules. These dense core vesicles are associated with a variety of secretory products, cell surface antigens, and enzymes. The biology of SCLC is complex. The activation of a number of dominant proto-oncogenes and the inactivation of tumor suppressor genes in SCLC have been described. Dominant proto-oncogenes that have been found to be amplified or overexpressed in SCLC include the myc family, c-myb, c-kit, c-jun, and c-src. Altered expression of two tumor suppressor genes in SCLC, p53 and the retinoblastoma gene product, has been demonstrated. Cytogenetic and molecular evidence for chromosomal loss of 3p, 5q, 9p, 11p, 13q, and 17p in SCLC has intensified the search for other tumor suppressor genes with potential import in this malignancy. Bombesin/gastrin-releasing peptide, insulin-like growth factor I, and transferrin have been identified as autocrine growth factors in SCLC, with a number of other peptides under active investigation. Several mechanisms of drug resistance in SCLC have been described, including gene amplification, the recently described overexpression of multi-drug resistance-related protein (MRP), and the expression of P-glycoprotein. The classic SCLC staging system has been supplanted by a revised TNM staging system where limited disease and extensive disease are equivalent to the TNM stages I through III and stage IV, respectively. Therapeutically, recent strategies have attained small improvements in survival but significant reductions in the toxicities of chemotherapeutic regimens. Presently, the overall 5-year survival for SCLC is 5% to 10%, with limited disease associated with a significantly higher survival rate.(ABSTRACT TRUNCATED AT 400 WORDS)
Collapse
Affiliation(s)
- R M Cook
- Department of Medicine, University of Colorado Health Sciences Center, Denver
| | | | | |
Collapse
|
37
|
|
38
|
Hagiwara T, Nakaya K, Nakamura Y, Nakajima H, Nishimura S, Taya Y. Specific phosphorylation of the acidic central region of the N-myc protein by casein kinase II. EUROPEAN JOURNAL OF BIOCHEMISTRY 1992; 209:945-50. [PMID: 1425701 DOI: 10.1111/j.1432-1033.1992.tb17367.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The central region of the N-myc protein has a characteristic amino acid sequence EDTLSDSDDEDD, which is very similar to those of particular domains of adenovirus E1A, human papilloma virus E7, Simian virus 40 large T, c-myc and L-myc proteins. Domains of these three viral oncoproteins have recently been shown to be specific binding sites for the tumor-suppressor gene retinoblastoma protein. We have noted that the sequence of serine followed by a cluster of acidic amino acids is exactly the same as that of a typical substrate of casein kinase II (CKII). Therefore, we investigated whether these nuclear oncoproteins are phosphorylated by CKII. For this purpose, we fused the beta-galactosidase and N-myc genes including this domain and expressed it in Escherichia coli cells. Several mutant N-myc genes, containing single amino acid substitutions in this domain, were also used to produce fused proteins. Strong phosphorylation by CKII was detected with the fused protein of wild-type N-myc. However, no phosphorylation of beta-galactosidase itself was observed and the phosphorylations of fused mutant proteins were low. Another fused N-myc protein containing most of the C-terminal region downstream of this acidic region was not phosphorylated by CKII. Analysis of phosphorylation sites in synthetic peptides of this acidic region identified the major sites phosphorylated by CKII as Ser261 and Ser263. On two-dimensional tryptic mapping of phosphorylated N-myc proteins, major spots of in vitro-labeled and in-vivo-labeled N-myc proteins were detected in the same positions. These results suggest that two serine residues of the acidic central region of the N-myc protein are phosphorylated by CKII in vivo as well as in vitro. The functional significance of this acidic domain is discussed.
Collapse
Affiliation(s)
- T Hagiwara
- Laboratory of Biological Chemistry, School of Pharmaceutical Sciences, Showa University, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
39
|
Imamura Y, Iguchi-Ariga SM, Ariga H. The upstream region of the mouse N-myc gene: identification of an enhancer element that functions preferentially in neuroblastoma IMR32 cells. BIOCHIMICA ET BIOPHYSICA ACTA 1992; 1132:177-87. [PMID: 1327147 DOI: 10.1016/0167-4781(92)90009-o] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The various members of the myc gene family, including c-myc and N-myc, are supposed to play a role in the regulation of cell cycle and proliferation. Whereas c-myc is expressed nearly ubiquitously, the N-myc gene product is found mainly in actively proliferating neural tissues such as early development tissues or in retinoblastomas and neuroblastomas. In this report, the upstream region of mouse N-myc gene was ligated to pSVPCAT, which carries the simian virus 40 (SV40) promoter and bacterial chloramphenicol acetyltransferase (CAT) gene, and transcriptional activities were examined by CAT and S1 protection assays after transfection of the DNAs into human cervical carcinoma HeLa or neuroblastoma IMR32 cells. Several regulatory regions were identified: two promoting regions (-980 to -860 and -279 to +108) and an inhibiting one (-860 to -797). The region spanning positions -980 to -860 increased CAT expression independently of orientation and distance to the SV40 promoter, indicating that the element is a typical enhancer. Moreover, the expression levels from this enhancer were higher in IMR32 cells than in HeLa cells, indicating that action has, if not cell-type specificity, cell-type preference. These findings may provide useful bases for the understanding of the cell-type specific regulation of N-myc expression.
Collapse
Affiliation(s)
- Y Imamura
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | | | | |
Collapse
|
40
|
Affiliation(s)
- M Schwab
- German Cancer Research Center, Institute for Experimental Pathology, Heidelberg
| |
Collapse
|
41
|
Harris LL, Talian JC, Zelenka PS. Contrasting patterns of c-myc and N-myc expression in proliferating, quiescent, and differentiating cells of the embryonic chicken lens. Development 1992; 115:813-20. [PMID: 1339339 DOI: 10.1242/dev.115.3.813] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The present study uses the polymerase chain reaction and in situ hybridization to examine c-myc and N-myc mRNA in the embryonic chicken lens at 6, 10, 14 and 19 days of development and compares the pattern of expression obtained with the developmental pattern of cell proliferation and differentiation. In the central epithelium, c-myc mRNA levels were proportional to the percentage of proliferating cells throughout development. N-myc mRNA expression in this region was relatively low and showed no correlation with cell proliferation. The ratio of N-myc to c-myc mRNA increased markedly with the onset of epithelial cell elongation and terminal fiber cell differentiation, although both c-myc and N-myc mRNAs continued to be expressed in postmitotic, elongating cells of the equatorial epithelium and in terminally differentiating lens fiber cells. Thus, increased expression of N-myc, a gene whose protein product may compete with c-myc protein for dimerization partners, accompanies the dissociation of c-myc expression and cell proliferation during terminal differentiation of lens fiber cells.
Collapse
Affiliation(s)
- L L Harris
- Howard Hughes Medical Institute, N.I.H. Research Scholars Program, Bethesda, Maryland 20814
| | | | | |
Collapse
|
42
|
Amler LC, Shibasaki Y, Savelyeva L, Schwab M. Amplification of the N-myc gene in human neuroblastomas: tandemly repeated amplicons within homogeneously staining regions on different chromosomes with the retention of single copy gene at the resident site. Mutat Res 1992; 276:291-7. [PMID: 1374521 DOI: 10.1016/0165-1110(92)90015-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- L C Amler
- Institut für Experimentelle Pathologie, Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | | | | | | |
Collapse
|
43
|
Gazitt Y, He YJ, Erdos GW, Chang L, Ashktorab H, Cohen RJ. Development of a two color immunofluorescence stain and immunolocalization method for N-myc and c-myc oncoproteins with a newly generated mouse IgM anti N-myc antibody. J Immunol Methods 1992; 148:159-69. [PMID: 1564326 DOI: 10.1016/0022-1759(92)90169-t] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
A new mouse monoclonal antibody specific for N-myc oncoprotein was generated and used in combination with an anti-c-myc antibody to develop two color immunofluorescence staining and ultrastructural immunolocalization of N-myc and c-myc in well established (SK-N-SH; CHP 126) and in newly established neuroblastoma (NB) cell lines. Analysis and quantitation of c-myc and N-myc in dually stained cells was done by flow cytometry. Immunolocalization was done by staining with immunogold secondary antibodies and transmission electron microscopy. The results obtained from analysis of 13 newly established NB cell lines revealed, great heterogeneity in the expression of N-myc oncoprotein with 10/13 cell lines over expressing the protein. C-myc oncoprotein was also expressed in all cell lines, however, the level of expression was 4-10-fold lower than the N-myc oncoprotein. Localization studies of c-myc and N-myc oncoproteins on the level of light microscopy and electron microscopy revealed exclusive nuclear localization of c-myc whereas N-myc was localized to the nucleus and to the cytoplasm.
Collapse
Affiliation(s)
- Y Gazitt
- Department of Pediatrics Hematology Oncology, University of Florida Medical Center, Gainesville 32610
| | | | | | | | | | | |
Collapse
|
44
|
Affiliation(s)
- T P Mäkelä
- Department of Virology, University of Helsinki, Finland
| | | | | |
Collapse
|
45
|
Hasegawa T, Hara E, Takehana K, Nakada S, Oda K, Kawata M, Kimura H, Sekiya S. A transient decrease in N-myc expression and its biological role during differentiation of human embryonal carcinoma cells. Differentiation 1991; 47:107-17. [PMID: 1955105 DOI: 10.1111/j.1432-0436.1991.tb00228.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The human embryonal carcinoma (EC) cell line, NEC14 can be induced to morphologically differentiate by the addition of 10(-2) M N,N'-hexamethylene-bis-acetamide (HMBA) in vitro. The expression of several cellular oncogenes (c-onc) in NEC14 cells was examined after induction of differentiation by HMBA. The level of N-myc expression was the highest in undifferentiated cells but decreased transiently to less than 1/10 of the original level shortly after the induction of differentiation. To investigate the role of the transient decrease in N-myc level on NEC14 cell differentiation, a chimeric human N-myc gene in which transcription is initiated at the human beta-actin gene promoter was constructed and introduced into NEC14 cells. Several transformants expressing the exogenous N-myc gene constitutively were established. These transformants showed 10- to 70-fold increases in plating efficiency and shorter population doubling times as compared with the parental NEC14 cells. The transformants were hard to induce, spontaneously differentiated cells on the periphery of cell clusters in culture, unlike parental NEC14 cells, and took longer for HMBA-induced morphological differentiation. The populations of the cells expressing HLA and SSEA-1 antigens increased from 10%-20% to nearly 100% in NEC14 cells after the induction of differentiation, while the populations expressing these antigens increased only to 50%-60% in one of the transformants, S11. The transformants gained an increased tumorigenic potential in nude mice, and the tumors produced consisted exclusively of EC stem cells. These results suggest that the additional expression of the exogenous N-myc gene (increased about two-fold) confers the more transformed state on the cells.
Collapse
Affiliation(s)
- T Hasegawa
- Department of Applied Biological Science, Science University of Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Kato K, Kanamori A, Wakamatsu Y, Sawai S, Kondoh H. Tissue Distribution of N-myc Expression in the Early Organogenesis Period of the Mouse Embryo. (N-myc/mouse embryo/in situ hybridization/neural/crest/sclerotome). Dev Growth Differ 1991. [DOI: 10.1111/j.1440-169x.1991.00029.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
47
|
Two signals mediate nuclear localization of influenza virus (A/WSN/33) polymerase basic protein 2. J Virol 1991; 65:245-53. [PMID: 1985200 PMCID: PMC240511 DOI: 10.1128/jvi.65.1.245-253.1991] [Citation(s) in RCA: 97] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Polymerase basic protein 2 (PB2), a component of the influenza virus polymerase complex, when expressed alone from cloned cDNA in the absence of other influenza virus proteins, is transported into the nucleus. In this study, we have examined the nuclear translocation signal of PB2 by making deletions and mutations in the PB2 sequence. Our studies showed that two distant regions in the polypeptide sequence were involved in the nuclear translocation of PB2. In one region, four basic residues (K-736 R K R) played a critical role in the nuclear translocation of PB2, since the deletion or mutation of these residues rendered the protein totally cytoplasmic. However, seven residues (M K R K R N S) of this region, including the four basic residues, failed to translocate a cytoplasmic reporter protein into the nucleus, suggesting that these sequences were necessary but not sufficient for nuclear translocation. Deletion of another region (amino acids 449 to 495) resulted in a mutant protein which was cytoplasmic with a perinuclear distribution. This novel phenotype suggests that a perinuclear binding step was involved prior to translocation of PB2 across the nuclear pore and that a signal might be involved in perinuclear binding. Possible involvement of these two signal sequences in the nuclear localization of PB2 is discussed.
Collapse
|
48
|
DePinho RA, Schreiber-Agus N, Alt FW. myc family oncogenes in the development of normal and neoplastic cells. Adv Cancer Res 1991; 57:1-46. [PMID: 1950701 DOI: 10.1016/s0065-230x(08)60994-x] [Citation(s) in RCA: 189] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- R A DePinho
- Department of Microbiology, Albert Einstein College of Medicine, Bronx, New York 10461
| | | | | |
Collapse
|
49
|
Abstract
The myc oncoproteins are expressed in a wide range of normal adult and embryonic tissues. They are also found to be over-expressed in a variety of tumor types. All myc proteins are short-lived nuclear phosphoproteins thought to act as regulatory components of cell proliferation. The rapid induction of c-myc mRNA and protein following the addition of growth factors to quiescent cells, together with the short half-life of these molecules, suggests that they are sensitive and continuous indicators of external stimuli, consistent with a role in signal transduction. Furthermore, in untransformed cells, c-myc protein expression is tightly regulated, at least in part, by a mechanism of autoregulation. Deregulated expression of myc genes is a frequent observation in tumors and may lead to a cell becoming independent of one or more growth factors, with the concomitant potential for uncontrolled proliferation. Although the precise functions of the myc proteins are unknown, they all bear the hallmarks of multimeric DNA-binding proteins probably involved in the regulation of expression of specific genes.
Collapse
|
50
|
Cai M, Davis RW. Yeast centromere binding protein CBF1, of the helix-loop-helix protein family, is required for chromosome stability and methionine prototrophy. Cell 1990; 61:437-46. [PMID: 2185892 DOI: 10.1016/0092-8674(90)90525-j] [Citation(s) in RCA: 269] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The centromere and its binding proteins constitute the kinetochore structure of metaphase chromosomes, which is crucial for the high accuracy of the chromosome segregation process. Isolation and analysis of the gene encoding a centromere binding protein from the yeast S. cerevisiae, CBF1, are described in this paper. DNA sequence analysis of the CBF1 gene reveals homology with the transforming protein myc and a family of regulatory proteins known as the helix-loop-helix (HLH) proteins. Disruption of the CBF1 gene caused a decrease in the growth rate, an increase in the rate of chromosome loss/nondisjunction, and hypersensitivity to the antimitotic drug thiabendazole. Unexpectedly, the cbf1 null mutation concomitantly resulted in a methionine auxotrophic phenotype, which suggests that CBF1, like other HLH proteins in higher eukaryotic cells, participates in the regulation of gene expression.
Collapse
Affiliation(s)
- M Cai
- Department of Biochemistry, Stanford University School of Medicine, California 94305-5307
| | | |
Collapse
|