101
|
Ye J, He J, Li Q, Feng Y, Bai X, Chen X, Zhao Y, Hu X, Yu Z, Li N. Generation of c-Myc transgenic pigs for autosomal dominant polycystic kidney disease. Transgenic Res 2013; 22:1231-9. [PMID: 23543409 DOI: 10.1007/s11248-013-9707-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 03/22/2013] [Indexed: 01/28/2023]
Abstract
After several decades of research, autosomal dominant polycystic kidney disease (ADPKD) is still incurable and imposes enormous physical, psychological, and economic burdens on patients and their families. Murine models of ADPKD represent invaluable tools for studying this disease. These murine forms of ADPKD can arise spontaneously, or they can be induced via chemical or genetic manipulations. Although these models have improved our understanding of the etiology and pathogenesis of ADPKD, they have not led to effective treatment strategies. The mini-pig represents an effective biomedical model for studying human diseases, as the pig's human-like physiological processes help to understand disease mechanisms and to develop novel therapies. Here, we tried to generate a transgenic model of ADPKD in pigs by overexpressing c-Myc in kidney tissue. Western-blot analysis showed that c-Myc was overexpressed in the kidney, brain, heart, and liver of transgenic pigs. Immunohistochemical staining of kidney tissue showed that exogenous c-Myc predominantly localized to renal tubules. Slightly elevated blood urea nitrogen levels were observed in transgenic pigs 1 month after birth, but no obvious abnormalities were detected after that time. In the future, we plan to subject this model to renal injury in an effort to promote ADPKD progression.
Collapse
Affiliation(s)
- Jianhua Ye
- State Key Laboratory for Agrobiotechnology, China Agricultural University, Beijing, 100193, China,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
102
|
Shi JW, Liu W, Zhang TT, Wang SC, Lin XL, Li J, Jia JS, Sheng HF, Yao ZF, Zhao WT, Zhao ZL, Xie RY, Yang S, Gao F, Fan QR, Zhang MY, Yue M, Yuan J, Gu WW, Yao KT, Xiao D. The enforced expression of c-Myc in pig fibroblasts triggers mesenchymal-epithelial transition (MET) via F-actin reorganization and RhoA/Rock pathway inactivation. Cell Cycle 2013; 12:1119-27. [PMID: 23466707 DOI: 10.4161/cc.24164] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
In previous studies from other labs it has been well demonstrated that the ectopic expression of c-Myc in mammary epithelial cells can induce epithelial-mesenchymal transition (EMT), whereas in our pilot experiment, epithelial-like morphological changes were unexpectedly observed in c-Myc-expressing pig fibroblasts [i.e., porcine embryonic fibroblasts (PEFs) and porcine dermal fibroblasts (PDFs)] and pig mesenchymal stem cells, suggesting that the same c-Myc gene is entitled to trigger EMT in epithelial cells and mesenchymal-epithelial transition (MET) in fibroblasts. This prompted us to characterize the existence of a MET in c-Myc-expressing PEFs and PDFs at the molecular level. qRT-PCR, immunofluorescence and western blot analysis illustrated that epithelial-like morphological changes were accompanied by the increased expression of epithelial markers [such as cell adhesion proteins (E-cadherin, α-catenin and Bves), tight junction protein occludin and cytokeratins (Krt8 and Krt18)], the reduced expression of mesenchymal markers [vimentin, fibronectin 1 (FN1), snail1, collagen family of proteins (COL1A1, COL5A2) and matrix metalloproteinase (MMP) family (MMP12 and MMP14)] and the decreased cell motility and increased cell adhesion in c-Myc-expressing PEFs and PDFs. Furthermore, the ectopic expression of c-Myc in pig fibroblasts disrupted the stress fiber network, suppressed the formation of filopodia and lamellipodia, and resulted in RhoA/Rock pathway inactivation, which finally participates in epithelial-like morphological conversion. Taken together, these findings demonstrate, for the first time, that the enforced expression of c-Myc in fibroblasts can trigger MET, to which cytoskeleton depolymerization and RhoA/Rock pathway inactivation contribute.
Collapse
Affiliation(s)
- Jun-Wen Shi
- Cancer Research Institute, Southern Medical University, Guangzhou, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
103
|
Coker WJ, Jeter A, Schade H, Kang Y. Plasma cell disorders in HIV-infected patients: epidemiology and molecular mechanisms. Biomark Res 2013; 1:8. [PMID: 24252328 PMCID: PMC4177611 DOI: 10.1186/2050-7771-1-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Accepted: 01/04/2013] [Indexed: 12/19/2022] Open
Abstract
Highly active antiretroviral therapy (HAART) has significantly improved the outcome and survival of human immunodeficiency virus (HIV)-infected patients. Subsequently, long-term morbidities including cancer have become of major public health and clinical interest for this patient population. Plasma cell disorders occur at higher incidence in HIV-infected patients; however, the molecular mechanisms driving the plasma cell disease process and the optimal management for these patients remain to be defined. This article provides an up-to-date review of the characteristics and management of HIV-infected patients with plasma cell disorders. We first present 3 cases of plasma cell disorders in HIV-infected patients, ranging from polyclonal hypergammaglobulinemia to symptomatic multiple myeloma. We then discuss the epidemiology, clinical presentation, and management of each of these plasma cell disorders, with an emphasis on the molecular events underlying the progression of plasma cell diseases from monoclonal gammopathy to symptomatic multiple myeloma. We propose a three-step hypothesis for the development of multiple myeloma. Finally, we discuss the use of high dose chemotherapy and autologous hematopoietic stem cell transplantation in the treatment of HIV-infected patients with multiple myeloma. Our review includes the care of HIV-infected patients with plasma cell disorders in the current era of HAART and novel agents available for the treatment of multiple myeloma.
Collapse
Affiliation(s)
- Woodrow J Coker
- Division of Hematology and Oncology, Department of Medicine, Medical University of South Carolina, 86 Jonathan Lucas Street, Hollings Cancer Center, Room# HO307, Charleston, SC, 29425, USA.
| | | | | | | |
Collapse
|
104
|
Abstract
In this issue of Blood, Holien et al report that MYC addiction is responsible for rapid death of myeloma cell lines and primary myeloma tumor cells treated with a specific MYC inhibitor.
Collapse
|
105
|
Gururaj AE, Gibson L, Panchabhai S, Bai M, Manyam G, Lu Y, Latha K, Rojas ML, Hwang Y, Liang S, Bogler O. Access to the nucleus and functional association with c-Myc is required for the full oncogenic potential of ΔEGFR/EGFRvIII. J Biol Chem 2012; 288:3428-38. [PMID: 23250739 DOI: 10.1074/jbc.m112.399352] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
ΔEGFR is a potent glioblastoma oncogene which has been studied primarily as a plasma membrane kinase. Using intracranial xenograft studies in mice, we show that blocking ΔEGFR access to the nucleus attenuates its tumorigenicity and, conversely, that promoting nuclear accumulation enhances this, providing the first in vivo evidence that the nuclear actions of ΔEGFR contribute strongly to its oncogenic function. Nuclear actions of ΔEGFR include regulation of gene expression by participation in chromatin-bound complexes, and genome-wide mapping of these sequences by chromatin immunoprecipitation and massively parallel sequencing identified 2294 peaks. Bioinformatic analysis showed enrichment of the E-box motif in the dataset, and c-Myc and ΔEGFR were corecruited to the promoters of and transcriptionally activated a subset of nuclear ΔEGFR chromatin targets. Knockdown of c-Myc decreased the expression of these targets and diminished ΔEGFR-stimulated anchorage-independent colony formation. We conclude that transcriptional regulation of target genes by association with gene regulatory chromatin in cooperation with c-Myc by nuclear ΔEGFR makes a unique contribution to its oncogenicity and propose that this venue provides new targets for therapeutic intervention.
Collapse
Affiliation(s)
- Anupama E Gururaj
- Department of Neurosurgery, University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
106
|
Moumen M, Chiche A, Deugnier MA, Petit V, Gandarillas A, Glukhova MA, Faraldo MM. The proto-oncogene Myc is essential for mammary stem cell function. Stem Cells 2012; 30:1246-54. [PMID: 22438054 DOI: 10.1002/stem.1090] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The mammary epithelium comprises two major cell lineages: basal and luminal. Basal cells (BCs) isolated from the mammary epithelium and transplanted into the mouse mammary fat pad cleared from the endogenous epithelium regenerate the mammary gland, strongly suggesting that the basal epithelial compartment harbors a long-lived cell population with multipotent stem cell potential. The luminal cell layer is devoid of the regenerative potential, but it contains cells with clonogenic capacity, the luminal progenitors. Mammary BCs and luminal progenitors express high levels of the transcription factor Myc. Here, we show that deletion of Myc from mammary basal epithelial cells led to impaired stem cell self-renewal as evaluated by limiting dilution and serial transplantation assays. Luminal progenitor population was significantly diminished in mutant epithelium suggesting control by the BC layer. Colony formation assay performed with isolated BCs showed that clonogenic capacity was abolished by Myc deletion. Moreover, transplanted BCs depleted of Myc failed to produce epithelial outgrowths. Stimulation with ovarian hormones estrogen (E) and progesterone (P) partially rescued the repopulation capacity of Myc-depleted BCs; however, the Myc-deficient mammary epithelium developed in response to E/P treatment lacked stem and progenitor cells. This study provides the first evidence that in the mammary gland, Myc has an essential nonredundant function in the maintenance of the self-renewing multipotent stem cell population responsible for the regenerative capacity of the mammary epithelium and is required downstream from ovarian hormones, for the control of mammary stem and progenitor cell functions.
Collapse
Affiliation(s)
- Mejdi Moumen
- Institut Curie, Centre de Recherche, Paris, France
| | | | | | | | | | | | | |
Collapse
|
107
|
The MYC-associated protein CDCA7 is phosphorylated by AKT to regulate MYC-dependent apoptosis and transformation. Mol Cell Biol 2012; 33:498-513. [PMID: 23166294 DOI: 10.1128/mcb.00276-12] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cell division control protein A7 (CDCA7) is a recently identified target of MYC-dependent transcriptional regulation. We have discovered that CDCA7 associates with MYC and that this association is modulated in a phosphorylation-dependent manner. The prosurvival kinase AKT phosphorylates CDCA7 at threonine 163, promoting binding to 14-3-3, dissociation from MYC, and sequestration to the cytoplasm. Upon serum withdrawal, induction of CDCA7 expression in the presence of MYC sensitized cells to apoptosis, whereas CDCA7 knockdown reduced MYC-dependent apoptosis. The transformation of fibroblasts by MYC was reduced by coexpression of CDCA7, while the non-MYC-interacting protein Δ(156-187)-CDCA7 largely inhibited MYC-induced transformation. These studies provide insight into a new mechanism by which AKT signaling to CDCA7 could alter MYC-dependent growth and transformation, contributing to tumorigenesis.
Collapse
|
108
|
Hannan KM, Sanij E, Rothblum LI, Hannan RD, Pearson RB. Dysregulation of RNA polymerase I transcription during disease. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2012; 1829:342-60. [PMID: 23153826 DOI: 10.1016/j.bbagrm.2012.10.014] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Revised: 10/30/2012] [Accepted: 10/31/2012] [Indexed: 12/13/2022]
Abstract
Transcription of the ribosomal RNA genes by the dedicated RNA polymerase I enzyme and subsequent processing of the ribosomal RNA are fundamental control steps in the synthesis of functional ribosomes. Dysregulation of Pol I transcription and ribosome biogenesis is linked to the etiology of a broad range of human diseases. Diseases caused by loss of function mutations in the molecular constituents of the ribosome, or factors intimately associated with RNA polymerase I transcription and processing are collectively termed ribosomopathies. Ribosomopathies are generally rare and treatment options are extremely limited tending to be more palliative than curative. Other more common diseases are associated with profound changes in cellular growth such as cardiac hypertrophy, atrophy or cancer. In contrast to ribosomopathies, altered RNA polymerase I transcriptional activity in these diseases largely results from dysregulated upstream oncogenic pathways or by direct modulation by oncogenes or tumor suppressors at the level of the RNA polymerase I transcription apparatus itself. Ribosomopathies associated with mutations in ribosomal proteins and ribosomal RNA processing or assembly factors have been covered by recent excellent reviews. In contrast, here we review our current knowledge of human diseases specifically associated with dysregulation of RNA polymerase I transcription and its associated regulatory apparatus, including some cases where this dysregulation is directly causative in disease. We will also provide insight into and discussion of possible therapeutic approaches to treat patients with dysregulated RNA polymerase I transcription. This article is part of a Special Issue entitled: Transcription by Odd Pols.
Collapse
Affiliation(s)
- K M Hannan
- Oncogenic Signalling and Growth Control Program, Peter MacCallum Cancer Centre, Locked Bag 1, A'Beckett St, Melbourne, Victoria 8006, Australia
| | | | | | | | | |
Collapse
|
109
|
A critical role for Mnt in Myc-driven T-cell proliferation and oncogenesis. Proc Natl Acad Sci U S A 2012; 109:19685-90. [PMID: 23150551 DOI: 10.1073/pnas.1206406109] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Mnt (Max's next tango) is a Max-interacting transcriptional repressor that can antagonize both the proproliferative and proapoptotic functions of Myc in vitro. To ascertain the physiologically relevant functions of Mnt and to help define the relationship between Mnt and Myc in vivo, we generated a series of mouse strains in which Mnt was deleted in T cells in the absence of endogenous c-Myc or in the presence of ectopic c-Myc. We found that apoptosis caused by loss of Mnt did not require Myc but that ectopic Myc expression dramatically decreased the survival of both Mnt-deficient T cells in vivo and Mnt-deficient MEFs in vitro. Consequently, Myc-driven proliferative expansion of T cells in vitro and thymoma formation in vivo were prevented by the absence of Mnt. Consistent with T-cell models, mouse embryo fibroblasts (MEFs) lacking Mnt were refractory to oncogenic transformation by Myc. Tumor suppression caused by loss of Mnt was linked to increased apoptosis mediated by reactive oxygen species (ROS). Thus, although theoretically and experimentally a Myc antagonist, the dominant physiological role of Mnt appears to be suppression of apoptosis. Our results redefine the physiological relationship between Mnt and Myc and requirements for Myc-driven oncogenesis.
Collapse
|
110
|
Sung CK, Yim H, Gu H, Li D, Andrews E, Duraisamy S, Li C, Drapkin R, Benjamin T. The polyoma virus large T binding protein p150 is a transcriptional repressor of c-MYC. PLoS One 2012; 7:e46486. [PMID: 23029531 PMCID: PMC3460914 DOI: 10.1371/journal.pone.0046486] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Accepted: 09/02/2012] [Indexed: 11/18/2022] Open
Abstract
p150, product of the SALL2 gene, is a binding partner of the polyoma virus large T antigen and a putative tumor suppressor. p150 binds to the nuclease hypersensitive element of the c-MYC promoter and represses c-MYC transcription. Overexpression of p150 in human ovarian surface epithelial cells leads to decreased expression, and downregulation to increased expression, of c-MYC. c-MYC is repressed upon restoration of p150 to ovarian carcinoma cells. Induction of apoptosis by etoposide results in recruitment of p150 to the c-MYC promoter and to repression of c-MYC. Analysis of data in The Cancer Genome Atlas shows negative correlations between SALL2 and c-MYC expression in four common solid tumor types.
Collapse
Affiliation(s)
- Chang Kyoo Sung
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Hyungshin Yim
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Hongcang Gu
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Dawei Li
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Erik Andrews
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Sekhar Duraisamy
- Dana Farber Cancer Institute, Department of Medical Oncology, Center for Molecular Oncologic Pathology, Boston, Massachusetts, United States of America
| | - Cheng Li
- Dana Farber Cancer Institute, Department of Biostatistics and Computational Biology, Boston, Massachusetts, United States of America
| | - Ronny Drapkin
- Dana Farber Cancer Institute, Department of Medical Oncology, Center for Molecular Oncologic Pathology, Boston, Massachusetts, United States of America
- Brigham and Women's Hospital, Department of Pathology, Boston, Massachusetts, United States of America
| | - Thomas Benjamin
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
111
|
Berry T, Luther W, Bhatnagar N, Jamin Y, Poon E, Sanda T, Pei D, Sharma B, Vetharoy WR, Hallsworth A, Ahmad Z, Barker K, Moreau L, Webber H, Wang W, Liu Q, Perez-Atayde A, Rodig S, Cheung NK, Raynaud F, Hallberg B, Robinson SP, Gray NS, Pearson AD, Eccles SA, Chesler L, George RE. The ALK(F1174L) mutation potentiates the oncogenic activity of MYCN in neuroblastoma. Cancer Cell 2012; 22:117-30. [PMID: 22789543 PMCID: PMC3417812 DOI: 10.1016/j.ccr.2012.06.001] [Citation(s) in RCA: 237] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2012] [Revised: 03/18/2012] [Accepted: 06/05/2012] [Indexed: 01/03/2023]
Abstract
The ALK(F1174L) mutation is associated with intrinsic and acquired resistance to crizotinib and cosegregates with MYCN in neuroblastoma. In this study, we generated a mouse model overexpressing ALK(F1174L) in the neural crest. Compared to ALK(F1174L) and MYCN alone, co-expression of these two oncogenes led to the development of neuroblastomas with earlier onset, higher penetrance, and enhanced lethality. ALK(F1174L)/MYCN tumors exhibited increased MYCN dosage due to ALK(F1174L)-induced activation of the PI3K/AKT/mTOR and MAPK pathways, coupled with suppression of MYCN pro-apoptotic effects. Combined treatment with the ATP-competitive mTOR inhibitor Torin2 overcame the resistance of ALK(F1174L)/MYCN tumors to crizotinib. Our findings demonstrate a pathogenic role for ALK(F1174L) in neuroblastomas overexpressing MYCN and suggest a strategy for improving targeted therapy for ALK-positive neuroblastoma.
Collapse
Affiliation(s)
- Teeara Berry
- Divisions of Clinical Studies & Cancer Therapeutics, The Institute of Cancer Research, Sutton, Surrey, UK
| | - William Luther
- Department of Pediatric Hematology & Oncology, Dana-Farber Cancer Institute and Children’s Hospital Boston, Harvard Medical School, Boston, MA, USA
| | - Namrata Bhatnagar
- Department of Pediatric Hematology & Oncology, Dana-Farber Cancer Institute and Children’s Hospital Boston, Harvard Medical School, Boston, MA, USA
| | - Yann Jamin
- Division of Radiotherapy & Imaging, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Evon Poon
- Divisions of Clinical Studies & Cancer Therapeutics, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Takaomi Sanda
- Department of Pediatric Hematology & Oncology, Dana-Farber Cancer Institute and Children’s Hospital Boston, Harvard Medical School, Boston, MA, USA
| | - Desheng Pei
- Department of Pediatric Hematology & Oncology, Dana-Farber Cancer Institute and Children’s Hospital Boston, Harvard Medical School, Boston, MA, USA
| | - Bandana Sharma
- Department of Pediatric Hematology & Oncology, Dana-Farber Cancer Institute and Children’s Hospital Boston, Harvard Medical School, Boston, MA, USA
| | - Winston R. Vetharoy
- Divisions of Clinical Studies & Cancer Therapeutics, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Albert Hallsworth
- Divisions of Clinical Studies & Cancer Therapeutics, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Zai Ahmad
- Divisions of Clinical Studies & Cancer Therapeutics, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Karen Barker
- Divisions of Clinical Studies & Cancer Therapeutics, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Lisa Moreau
- Department of Pediatric Hematology & Oncology, Dana-Farber Cancer Institute and Children’s Hospital Boston, Harvard Medical School, Boston, MA, USA
| | - Hannah Webber
- Divisions of Clinical Studies & Cancer Therapeutics, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Wenchao Wang
- Department of Pediatric Hematology & Oncology, Dana-Farber Cancer Institute and Children’s Hospital Boston, Harvard Medical School, Boston, MA, USA
| | - Qingsong Liu
- Departments of Cancer Biology, Dana Farber Cancer Institute and Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | | | - Scott Rodig
- Department of Pathology, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Nai-Kong Cheung
- Department of Pediatrics, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Florence Raynaud
- Divisions of Clinical Studies & Cancer Therapeutics, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Bengt Hallberg
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Simon P. Robinson
- Division of Radiotherapy & Imaging, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Nathanael S. Gray
- Departments of Cancer Biology, Dana Farber Cancer Institute and Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Andrew D.J. Pearson
- Divisions of Clinical Studies & Cancer Therapeutics, The Institute of Cancer Research, Sutton, Surrey, UK
- The Children and Young People’s Unit, The Royal Marsden NHS Trust, Sutton, Surrey, UK
| | - Suzanne A. Eccles
- Divisions of Clinical Studies & Cancer Therapeutics, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Louis Chesler
- Divisions of Clinical Studies & Cancer Therapeutics, The Institute of Cancer Research, Sutton, Surrey, UK
- The Children and Young People’s Unit, The Royal Marsden NHS Trust, Sutton, Surrey, UK
| | - Rani E. George
- Department of Pediatric Hematology & Oncology, Dana-Farber Cancer Institute and Children’s Hospital Boston, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
112
|
N-Myc and GCN5 regulate significantly overlapping transcriptional programs in neural stem cells. PLoS One 2012; 7:e39456. [PMID: 22745758 PMCID: PMC3383708 DOI: 10.1371/journal.pone.0039456] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Accepted: 05/21/2012] [Indexed: 12/11/2022] Open
Abstract
Here we examine the functions of the Myc cofactor and histone acetyltransferase, GCN5/KAT2A, in neural stem and precursor cells (NSC) using a conditional knockout approach driven by nestin-cre. Mice with GCN5-deficient NSC exhibit a 25% reduction in brain mass with a microcephaly phenotype similar to that observed in nestin-cre driven knockouts of c- or N-myc. In addition, the loss of GCN5 inhibits precursor cell proliferation and reduces their populations in vivo, as does loss of N-myc. Gene expression analysis indicates that about one-sixth of genes whose expression is affected by loss of GCN5 are also affected in the same manner by loss of N-myc. These findings strongly support the notion that GCN5 protein is a key N-Myc transcriptional cofactor in NSC, but are also consistent with recruitment of GCN5 by other transcription factors and the use by N-Myc of other histone acetyltransferases. Putative N-Myc/GCN5 coregulated transcriptional pathways include cell metabolism, cell cycle, chromatin, and neuron projection morphogenesis genes. GCN5 is also required for maintenance of histone acetylation both at its putative specific target genes and at Myc targets. Thus, we have defined an important role for GCN5 in NSC and provided evidence that GCN5 is an important Myc transcriptional cofactor in vivo.
Collapse
|
113
|
Rosiglitazone suppresses glioma cell growth and cell cycle by blocking the transforming growth factor-beta mediated pathway. Neurochem Res 2012; 37:2076-84. [PMID: 22707243 DOI: 10.1007/s11064-012-0828-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Revised: 05/10/2012] [Accepted: 06/08/2012] [Indexed: 12/25/2022]
Abstract
Glioma is one of the most malignant tumors in the central nervous system. As a peroxisome proliferator-activated receptor γ (PPAR-γ) activator, the thiazolidinediones (TZDs) induce growth arrest and cell death in a broad spectrum of tumor cells. In this study, we investigated the role of rosiglitazone in glioma cells. We found that rosiglitazone, a member of TZDs, suppresses growth of human glioma cell lines U87 and U251. Rosiglitazone also induces cell cycle arrest and apoptosis, which may be the mechanism of its anti-proliferation effect. Next, we found that rosiglitazone suppresses the expression of TGF-beta and its receptor TGF-betaR2, and suppresses phosphorylation of Smad3. Rosiglitazone also inhibits formation of the Smad3/Smad4 complex. Furthermore, Rosiglitazone affects the expression of Smad3/Smad4 associated regulators of gene expression, including p21 and c-Myc. These results suggest that rosiglitazone suppresses growth and cell cycle of human glioma cells by blocking the TGF-beta mediated pathway.
Collapse
|
114
|
Nagata T, Shimada Y, Sekine S, Hori R, Matsui K, Okumura T, Sawada S, Fukuoka J, Tsukada K. Prognostic significance of NANOG and KLF4 for breast cancer. Breast Cancer 2012; 21:96-101. [PMID: 22528804 DOI: 10.1007/s12282-012-0357-y] [Citation(s) in RCA: 97] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Accepted: 03/07/2012] [Indexed: 12/13/2022]
Abstract
BACKGROUND Some of the induced pluripotent stem cell (iPS cell)-inducing factors have been reported to be expressed in breast cancer. The aim of the present study was to examine the relationship between the expression of iPS cell-inducing factors and the prognosis of breast cancer patients. METHODS In 100 breast cancer patients, the expression of c-MYC, KLF4, NANOG, OCT4, and SOX2 was determined by immunohistochemistry using a tissue microarray analysis. RESULTS Patients with strong expression of NANOG had significantly lower disease-free survival (DFS) and overall survival rates than those with weak expression of NANOG (P = 0.004 and 0.033, respectively). In contrast, patients with strong expression of KLF4 had better DFS (P = 0.014). CONCLUSIONS Strong expression of NANOG is an indicator of a poor prognosis for breast cancer patients, whereas KLF4 is a favorable prognostic indicator. Our results suggest that NANOG stimulates the growth and metastasis of breast cancer cells, whereas KLF4 inhibits these processes.
Collapse
Affiliation(s)
- Takuya Nagata
- Department of Surgery and Science, Graduate School of Research Into Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, Toyama, Japan,
| | | | | | | | | | | | | | | | | |
Collapse
|
115
|
Catuogno S, Cerchia L, Romano G, Pognonec P, Condorelli G, de Franciscis V. miR-34c may protect lung cancer cells from paclitaxel-induced apoptosis. Oncogene 2012; 32:341-51. [PMID: 22370637 DOI: 10.1038/onc.2012.51] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
MicroRNAs (miRNAs) constitute a class of small non-coding RNAs that negatively regulate the expression of their target genes. They are involved in many biological processes, including cell proliferation, apoptosis and differentiation, and are considered as promising new therapeutic targets for cancer. However, the identity of miRNAs involved in apoptosis and their respective targets remain largely unknown. Given the elevated complexity of miRNA regulation of gene expression, we performed a functional screening as an alternative strategy to identify those miRNAs that in lung cancer cells may interfere with the apoptotic process. To this aim, we generated a derivative of the non-small cell lung carcinoma A549 cell line in which caspase-8, a critical upstream initiator of apoptosis, can be activated by administration of the small dimerizer drug AP20187. We found a number of miRNAs that may rescue cell viability from caspase-8 activation. They included miRNAs already described as oncogenic such as miR-17, miR-135 and miR-520, but also some miRNAs such as miR-124-1 and miR-34c for which a tumor-suppressive role has instead been described or expected. Among them, miR-34c-5p markedly increased resistance to paclitaxel-induced apoptosis. We demonstrate that Bmf (Bcl-2-modifying factor) is a target of miR-34c-5p, and that its silencing, together with that of c-myc, a known target of miR-34c-5p, contributes to resistance to apoptosis induced by paclitaxel through p53 downregulation.
Collapse
Affiliation(s)
- S Catuogno
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale del CNR 'G Salvatore', Naples, Italy
| | | | | | | | | | | |
Collapse
|
116
|
Montagne M, Beaudoin N, Fortin D, Lavoie CL, Klinck R, Lavigne P. The Max b-HLH-LZ can transduce into cells and inhibit c-Myc transcriptional activities. PLoS One 2012; 7:e32172. [PMID: 22384171 PMCID: PMC3284561 DOI: 10.1371/journal.pone.0032172] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Accepted: 01/22/2012] [Indexed: 12/23/2022] Open
Abstract
The inhibition of the functions of c-Myc (endogenous and oncogenic) was recently shown to provide a spectacular therapeutic index in cancer mouse models, with complete tumor regression and minimal side-effects in normal tissues. This was achieved by the systemic and conditional expression of omomyc, the cDNA of a designed mutant of the b-HLH-LZ of c-Myc named Omomyc. The overall mode of action of Omomyc consists in the sequestration of Max and the concomitant competition of the Omomyc/Max complex with the endogenous c-Myc/Max heterodimer. This leads to the inhibition of the transactivation of Myc target genes involved in proliferation and metabolism. While this body of work has provided extraordinary insights to guide the future development of new cancer therapies that target c-Myc, Omomyc itself is not a therapeutic agent. In this context, we sought to exploit the use of a b-HLH-LZ to inhibit c-Myc in a cancer cell line in a more direct fashion. We demonstrate that the b-HLH-LZ domain of Max (Max*) behaves as a bona fide protein transduction domain (PTD) that can efficiently transduce across cellular membrane via through endocytosis and translocate to the nucleus. In addition, we show that the treatment of HeLa cells with Max* leads to a reduction of metabolism and proliferation rate. Accordingly, we observe a decrease of the population of HeLa cells in S phase, an accumulation in G1/G0 and the induction of apoptosis. In agreement with these phenotypic changes, we show by q-RT-PCR that the treatment of HeLa cells with Max* leads to the activation of the transcription c-Myc repressed genes as well as the repression of the expression of c-Myc activated genes. In addition to the novel discovery that the Max b-HLH-LZ is a PTD, our findings open up new avenues and strategies for the direct inhibition of c-Myc with b-HLH-LZ analogs.
Collapse
Affiliation(s)
- Martin Montagne
- Département de Pharmacologie, Faculté de médicine et des sciences de la santé, Université de Sherbrooke, Québec, Canada
| | - Nicolas Beaudoin
- Département de Chirurgie, Faculté de médicine et des sciences de la santé, Université de Sherbrooke, Québec, Canada
| | - David Fortin
- Département de Chirurgie, Faculté de médicine et des sciences de la santé, Université de Sherbrooke, Québec, Canada
| | - Christine L. Lavoie
- Département de Pharmacologie, Faculté de médicine et des sciences de la santé, Université de Sherbrooke, Québec, Canada
| | - Roscoe Klinck
- Département de Microbiologie et Infectiologie et Laboratoire de Génomique Fonctionnelle de l'Université de Sherbrooke, Québec, Canada
| | - Pierre Lavigne
- Département de Pharmacologie, Faculté de médicine et des sciences de la santé, Université de Sherbrooke, Québec, Canada
- * E-mail:
| |
Collapse
|
117
|
Hou JM, Krebs MG, Lancashire L, Sloane R, Backen A, Swain RK, Priest LJ, Greystoke A, Zhou C, Morris K, Ward T, Blackhall FH, Dive C. Clinical Significance and Molecular Characteristics of Circulating Tumor Cells and Circulating Tumor Microemboli in Patients With Small-Cell Lung Cancer. J Clin Oncol 2012; 30:525-32. [DOI: 10.1200/jco.2010.33.3716] [Citation(s) in RCA: 626] [Impact Index Per Article: 48.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Purpose Circulating tumor cells (CTCs) may have utility as surrogate biomarkers and “virtual” biopsies. We report the clinical significance and molecular characteristics of CTCs and CTC clusters, termed circulating tumor microemboli (CTM), detected in patients with small-cell lung cancer (SCLC) undergoing standard treatment. Patients and Methods Serial blood samples from 97 patients receiving chemotherapy were analyzed using EpCam-based immunomagnetic detection and a filtration-based technique. Proliferation status (Ki67) and apoptotic morphology were examined. Associations of CTC and CTM number with clinical factors and prognosis were determined. Results CTCs were present in 85% of patients (77 of 97 patients) and were abundant (mean ± standard deviation = 1,589 ± 5,565). CTM and apoptotic CTCs were correlated with total CTC number and were detected in 32% and 57% of patients, respectively. Pretreatment CTCs, change in CTC number after one cycle of chemotherapy, CTM, and apoptotic CTCs were independent prognostic factors. Overall survival was 5.4 months for patients with ≥ 50 CTCs/7.5 mL of blood and 11.5 months (P < .0001) for patients with less than 50 CTCs/7.5 mL of blood before chemotherapy (hazard ratio = 2.45; 95% CI, 1.39 to 4.30; P = .002). Subpopulations of apoptotic and of proliferating solitary CTCs were detected, whereas neither were observed within cell clusters (CTM), implicating both protection from anoikis and relative resistance to cytotoxic drugs for cells within CTM. Conclusion Both baseline CTC number and change in CTC number after one cycle of chemotherapy are independent prognostic factors for SCLC. Molecular comparison of CTCs to cells in CTM may provide novel insights into SCLC biology.
Collapse
Affiliation(s)
- Jian-Mei Hou
- All authors, Clinical and Experimental Pharmacology Group, Paterson Institute for Cancer Research, University of Manchester, Manchester Cancer Research Centre, Manchester; Matthew G. Krebs and Fiona H. Blackhall, the Christie National Health Service Foundation Trust, Manchester, United Kingdom
| | - Matthew G. Krebs
- All authors, Clinical and Experimental Pharmacology Group, Paterson Institute for Cancer Research, University of Manchester, Manchester Cancer Research Centre, Manchester; Matthew G. Krebs and Fiona H. Blackhall, the Christie National Health Service Foundation Trust, Manchester, United Kingdom
| | - Lee Lancashire
- All authors, Clinical and Experimental Pharmacology Group, Paterson Institute for Cancer Research, University of Manchester, Manchester Cancer Research Centre, Manchester; Matthew G. Krebs and Fiona H. Blackhall, the Christie National Health Service Foundation Trust, Manchester, United Kingdom
| | - Robert Sloane
- All authors, Clinical and Experimental Pharmacology Group, Paterson Institute for Cancer Research, University of Manchester, Manchester Cancer Research Centre, Manchester; Matthew G. Krebs and Fiona H. Blackhall, the Christie National Health Service Foundation Trust, Manchester, United Kingdom
| | - Alison Backen
- All authors, Clinical and Experimental Pharmacology Group, Paterson Institute for Cancer Research, University of Manchester, Manchester Cancer Research Centre, Manchester; Matthew G. Krebs and Fiona H. Blackhall, the Christie National Health Service Foundation Trust, Manchester, United Kingdom
| | - Rajeeb K. Swain
- All authors, Clinical and Experimental Pharmacology Group, Paterson Institute for Cancer Research, University of Manchester, Manchester Cancer Research Centre, Manchester; Matthew G. Krebs and Fiona H. Blackhall, the Christie National Health Service Foundation Trust, Manchester, United Kingdom
| | - Lynsey J.C. Priest
- All authors, Clinical and Experimental Pharmacology Group, Paterson Institute for Cancer Research, University of Manchester, Manchester Cancer Research Centre, Manchester; Matthew G. Krebs and Fiona H. Blackhall, the Christie National Health Service Foundation Trust, Manchester, United Kingdom
| | - Alastair Greystoke
- All authors, Clinical and Experimental Pharmacology Group, Paterson Institute for Cancer Research, University of Manchester, Manchester Cancer Research Centre, Manchester; Matthew G. Krebs and Fiona H. Blackhall, the Christie National Health Service Foundation Trust, Manchester, United Kingdom
| | - Cong Zhou
- All authors, Clinical and Experimental Pharmacology Group, Paterson Institute for Cancer Research, University of Manchester, Manchester Cancer Research Centre, Manchester; Matthew G. Krebs and Fiona H. Blackhall, the Christie National Health Service Foundation Trust, Manchester, United Kingdom
| | - Karen Morris
- All authors, Clinical and Experimental Pharmacology Group, Paterson Institute for Cancer Research, University of Manchester, Manchester Cancer Research Centre, Manchester; Matthew G. Krebs and Fiona H. Blackhall, the Christie National Health Service Foundation Trust, Manchester, United Kingdom
| | - Tim Ward
- All authors, Clinical and Experimental Pharmacology Group, Paterson Institute for Cancer Research, University of Manchester, Manchester Cancer Research Centre, Manchester; Matthew G. Krebs and Fiona H. Blackhall, the Christie National Health Service Foundation Trust, Manchester, United Kingdom
| | - Fiona H. Blackhall
- All authors, Clinical and Experimental Pharmacology Group, Paterson Institute for Cancer Research, University of Manchester, Manchester Cancer Research Centre, Manchester; Matthew G. Krebs and Fiona H. Blackhall, the Christie National Health Service Foundation Trust, Manchester, United Kingdom
| | - Caroline Dive
- All authors, Clinical and Experimental Pharmacology Group, Paterson Institute for Cancer Research, University of Manchester, Manchester Cancer Research Centre, Manchester; Matthew G. Krebs and Fiona H. Blackhall, the Christie National Health Service Foundation Trust, Manchester, United Kingdom
| |
Collapse
|
118
|
Golomb L, Bublik DR, Wilder S, Nevo R, Kiss V, Grabusic K, Oren M. Importin 7 and exportin 1 link c-Myc and p53 to regulation of ribosomal biogenesis. Mol Cell 2012; 45:222-32. [PMID: 22284678 PMCID: PMC3270374 DOI: 10.1016/j.molcel.2011.11.022] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Revised: 07/13/2011] [Accepted: 11/04/2011] [Indexed: 11/22/2022]
Abstract
Members of the β-karyopherin family mediate nuclear import of ribosomal proteins and export of ribosomal subunits, both required for ribosome biogenesis. We report that transcription of the β-karyopherin genes importin 7 (IPO7) and exportin 1 (XPO1), and several additional nuclear import receptors, is regulated positively by c-Myc and negatively by p53. Partial IPO7 depletion triggers p53 activation and p53-dependent growth arrest. Activation of p53 by IPO7 knockdown has distinct features of ribosomal biogenesis stress, with increased binding of Mdm2 to ribosomal proteins L5 and L11 (RPL5 and RPL11). Furthermore, p53 activation is dependent on RPL5 and RPL11. Of note, IPO7 and XPO1 are frequently overexpressed in cancer. Altogether, we propose that c-Myc and p53 counter each other in the regulation of elements within the nuclear transport machinery, thereby exerting opposing effects on the rate of ribosome biogenesis. Perturbation of this balance may play a significant role in promoting cancer.
Collapse
Affiliation(s)
- Lior Golomb
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Debora Rosa Bublik
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Sylvia Wilder
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Reinat Nevo
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Vladimir Kiss
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Kristina Grabusic
- Department of Molecular medicine and Biotechnology, University of Rijeka, School of Medicine, Rijeka 51000, Croatia
| | - Moshe Oren
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
119
|
Jonassen JA, SanAgustin J, Baker SP, Pazour GJ. Disruption of IFT complex A causes cystic kidneys without mitotic spindle misorientation. J Am Soc Nephrol 2012; 23:641-51. [PMID: 22282595 DOI: 10.1681/asn.2011080829] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Intraflagellar transport (IFT) complexes A and B build and maintain primary cilia. In the mouse, kidney-specific or hypomorphic mutant alleles of IFT complex B genes cause polycystic kidneys, but the influence of IFT complex A proteins on renal development is not well understood. In the present study, we found that HoxB7-Cre-driven deletion of the complex A gene Ift140 from collecting ducts disrupted, but did not completely prevent, cilia assembly. Mutant kidneys developed collecting duct cysts by postnatal day 5, with rapid cystic expansion and renal dysfunction by day 15 and little remaining parenchymal tissue by day 20. In contrast to many models of polycystic kidney disease, precystic Ift140-deleted collecting ducts showed normal centrosomal positioning and no misorientation of the mitotic spindle axis, suggesting that disruption of oriented cell division is not a prerequisite to cyst formation in these kidneys. Precystic collecting ducts had an increased mitotic index, suggesting that cell proliferation may drive cyst expansion even with normal orientation of the mitotic spindle. In addition, we observed significant increases in expression of canonical Wnt pathway genes and mediators of Hedgehog and tissue fibrosis in highly cystic, but not precystic, kidneys. Taken together, these studies indicate that loss of Ift140 causes pronounced renal cystic disease and suggest that abnormalities in several different pathways may influence cyst progression.
Collapse
Affiliation(s)
- Julie A Jonassen
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | | | | |
Collapse
|
120
|
Kessler JD, Kahle KT, Sun T, Meerbrey KL, Schlabach MR, Schmitt EM, Skinner SO, Xu Q, Li MZ, Hartman ZC, Rao M, Yu P, Dominguez-Vidana R, Liang AC, Solimini NL, Bernardi RJ, Yu B, Hsu T, Golding I, Luo J, Osborne CK, Creighton CJ, Hilsenbeck SG, Schiff R, Shaw CA, Elledge SJ, Westbrook TF. A SUMOylation-dependent transcriptional subprogram is required for Myc-driven tumorigenesis. Science 2012; 335:348-53. [PMID: 22157079 PMCID: PMC4059214 DOI: 10.1126/science.1212728] [Citation(s) in RCA: 344] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Myc is an oncogenic transcription factor frequently dysregulated in human cancer. To identify pathways supporting the Myc oncogenic program, we used a genome-wide RNA interference screen to search for Myc-synthetic lethal genes and uncovered a role for the SUMO-activating enzyme (SAE1/2). Loss of SAE1/2 enzymatic activity drives synthetic lethality with Myc. Inactivation of SAE2 leads to mitotic catastrophe and cell death upon Myc hyperactivation. Mechanistically, SAE2 inhibition switches a transcriptional subprogram of Myc from activated to repressed. A subset of these SUMOylation-dependent Myc switchers (SMS genes) is required for mitotic spindle function and to support the Myc oncogenic program. SAE2 is required for growth of Myc-dependent tumors in mice, and gene expression analyses of Myc-high human breast cancers suggest that low SAE1 and SAE2 abundance in the tumors correlates with longer metastasis-free survival of the patients. Thus, inhibition of SUMOylation may merit investigation as a possible therapy for Myc-driven human cancers.
Collapse
MESH Headings
- Animals
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/mortality
- Breast Neoplasms/pathology
- Cell Cycle
- Cell Line, Tumor
- Cell Transformation, Neoplastic
- Female
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Genes, myc
- Humans
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/mortality
- Mammary Neoplasms, Experimental/pathology
- Mice
- Mice, Nude
- Mitosis
- Neoplasm Transplantation
- Proto-Oncogene Proteins c-myc/metabolism
- RNA Interference
- RNA, Small Interfering
- Spindle Apparatus/physiology
- Sumoylation
- Transcription, Genetic
- Transplantation, Heterologous
- Ubiquitin-Activating Enzymes/antagonists & inhibitors
- Ubiquitin-Activating Enzymes/genetics
- Ubiquitin-Activating Enzymes/metabolism
Collapse
Affiliation(s)
- Jessica D. Kessler
- Verna & Marrs McLean Dept. of Biochemistry & Molecular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
- Dept. of Molecular & Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
| | - Kristopher T. Kahle
- Howard Hughes Medical Institute, Dept. of Genetics, Harvard Medical School, Division of Genetics, Brigham & Women’s Hospital, Boston, MA02115
- Dept. of Neurosurgery, Massachusetts General Hospital, Boston, MA 02115
| | - Tingting Sun
- Verna & Marrs McLean Dept. of Biochemistry & Molecular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
| | - Kristen L. Meerbrey
- Verna & Marrs McLean Dept. of Biochemistry & Molecular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
- Dept. of Molecular & Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
| | - Michael R. Schlabach
- Howard Hughes Medical Institute, Dept. of Genetics, Harvard Medical School, Division of Genetics, Brigham & Women’s Hospital, Boston, MA02115
| | - Earlene M. Schmitt
- Verna & Marrs McLean Dept. of Biochemistry & Molecular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
- Dept. of Molecular & Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
| | - Samuel O. Skinner
- Verna & Marrs McLean Dept. of Biochemistry & Molecular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
- Dept. of Physics, University of Illinois, Urbana, IL61801
| | - Qikai Xu
- Howard Hughes Medical Institute, Dept. of Genetics, Harvard Medical School, Division of Genetics, Brigham & Women’s Hospital, Boston, MA02115
| | - Mamie Z. Li
- Howard Hughes Medical Institute, Dept. of Genetics, Harvard Medical School, Division of Genetics, Brigham & Women’s Hospital, Boston, MA02115
| | - Zachary C. Hartman
- Dept. of Clinical Cancer Prevention, M.D. Anderson Cancer Center, Houston, TX 77030
| | - Mitchell Rao
- Dept. of Molecular & Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
| | - Peng Yu
- Dept. of Molecular & Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
| | - Rocio Dominguez-Vidana
- Verna & Marrs McLean Dept. of Biochemistry & Molecular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
- Dept. of Molecular & Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
| | - Anthony C. Liang
- Howard Hughes Medical Institute, Dept. of Genetics, Harvard Medical School, Division of Genetics, Brigham & Women’s Hospital, Boston, MA02115
| | - Nicole L. Solimini
- Howard Hughes Medical Institute, Dept. of Genetics, Harvard Medical School, Division of Genetics, Brigham & Women’s Hospital, Boston, MA02115
| | - Ronald J. Bernardi
- Dept. of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
| | - Bing Yu
- Medical Oncology Branch, National Cancer Institute, Center Drive, Bethesda MD 20892
| | - Tiffany Hsu
- Verna & Marrs McLean Dept. of Biochemistry & Molecular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
- Dept. of Molecular & Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
| | - Ido Golding
- Verna & Marrs McLean Dept. of Biochemistry & Molecular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
- Dept. of Physics, University of Illinois, Urbana, IL61801
| | - Ji Luo
- Medical Oncology Branch, National Cancer Institute, Center Drive, Bethesda MD 20892
| | - C. Kent Osborne
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
- The Lester & Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
- Dept. of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
- Dept. of Molecular & Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
| | - Chad J. Creighton
- Division of Biostatistics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
| | - Susan G. Hilsenbeck
- Division of Biostatistics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
- The Lester & Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
| | - Rachel Schiff
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
- The Lester & Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
- Dept. of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
- Dept. of Molecular & Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
| | - Chad A. Shaw
- Dept. of Molecular & Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
| | - Stephen J. Elledge
- Howard Hughes Medical Institute, Dept. of Genetics, Harvard Medical School, Division of Genetics, Brigham & Women’s Hospital, Boston, MA02115
| | - Thomas F. Westbrook
- Verna & Marrs McLean Dept. of Biochemistry & Molecular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
- Dept. of Molecular & Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
- Dept. of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
| |
Collapse
|
121
|
Archbold HC, Yang YX, Chen L, Cadigan KM. How do they do Wnt they do?: regulation of transcription by the Wnt/β-catenin pathway. Acta Physiol (Oxf) 2012; 204:74-109. [PMID: 21624092 DOI: 10.1111/j.1748-1716.2011.02293.x] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Wnt/β-catenin signalling is known to play many roles in metazoan development and tissue homeostasis. Misregulation of the pathway has also been linked to many human diseases. In this review, specific aspects of the pathway's involvement in these processes are discussed, with an emphasis on how Wnt/β-catenin signalling regulates gene expression in a cell and temporally specific manner. The T-cell factor (TCF) family of transcription factors, which mediate a large portion of Wnt/β-catenin signalling, will be discussed in detail. Invertebrates contain a single TCF gene that contains two DNA-binding domains, the high mobility group (HMG) domain and the C-clamp, which increases the specificity of DNA binding. In vertebrates, the situation is more complex, with four TCF genes producing many isoforms that contain the HMG domain, but only some of which possess a C-clamp. Vertebrate TCFs have been reported to act in concert with many other transcription factors, which may explain how they obtain sufficient specificity for specific DNA sequences, as well as how they achieve a wide diversity of transcriptional outputs in different cells.
Collapse
Affiliation(s)
- H C Archbold
- Program in Cell and Molecular Biology, University of Michigan, Ann Arbor, 48109-1048, USA
| | | | | | | |
Collapse
|
122
|
Liu Y, Yin B, Zhang C, Zhou L, Fan J. Hsa-let-7a functions as a tumor suppressor in renal cell carcinoma cell lines by targeting c-myc. Biochem Biophys Res Commun 2012; 417:371-5. [DOI: 10.1016/j.bbrc.2011.11.119] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Accepted: 11/22/2011] [Indexed: 12/12/2022]
|
123
|
Azevedo JL, Feldman RA. Tinkering with transcription factors uncovers plasticity of somatic cells. Genes Cancer 2011; 1:1089-99. [PMID: 21779433 DOI: 10.1177/1947601911401908] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The advent of induced pluripotent stem cells (iPSCs) has brought the goal of using patient-derived cells for tissue repair closer to reality. However, the mechanisms involved in reprogramming to a pluripotent state are still not clear. It is understood that reprogramming to pluripotency involves epigenetic remodeling and the reactivation of "core" pluripotency factors. However, little is known about the mechanisms involved in overcoming senescence while avoiding oncogenesis, the maintenance of self-renewal, and the regulation of the balance between pluripotency and differentiation. Here, we review recent advances in reprogramming technology and what is currently known about the mechanism of reprogramming to pluripotency. Work with patient-derived iPSCs is already providing new insights into the cellular and molecular mechanisms involved in human disease. Further advances in reprogramming technology should result in efficient methods to reprogram patient-derived cells into iPSCs for use in regenerative medicine.
Collapse
Affiliation(s)
- Judi L Azevedo
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | | |
Collapse
|
124
|
Abstract
The c-Myc oncogene encodes a multifunctional transcription factor that directs the expression of genes required for cell growth and proliferation. Consistent with its potent growth-promoting properties, cells have evolved numerous mechanisms that limit the expression and activity of Myc. One of the most prominent of these mechanisms is proteolysis, which destroys Myc within minutes of its synthesis. The rapid and controlled destruction of Myc keeps its levels low and precisely tied to processes that regulate Myc production. In this review, we discuss how Myc protein stability is regulated and the influence of Myc proteolysis on its function. We describe what is known about how Myc is destroyed by ubiquitin (Ub)-mediated proteolysis, attempt to rationalize the role of different Ub-protein ligases and deubiquitylating enzymes (dUbs) in the regulation of Myc stability, and detail how these processes go awry in cancer. Finally, we discuss how our understanding of Myc regulation by the ubiquitin-proteasome system (UPS) can expose strategies for therapeutic intervention in human malignancies.
Collapse
Affiliation(s)
- Lance R Thomas
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | |
Collapse
|
125
|
Abstract
Impaired apoptosis is a cancer hallmark, and some types of lymphomas and other cancers harbor mutations that directly affect key cell death regulators, such as Bcl-2 family members. However, because the majority of tumors seem to lack such mutations, we are examining the hypothesis that tumorigenesis can be sustained at least initially by the normal expression of specific endogenous pro-survival Bcl-2 family members. We previously demonstrated that the lymphomagenesis in Εμ-myc transgenic mice, which constitutively overexpress the c-Myc oncoprotein in B-lymphoid cells and develop pre-B and B-cell lymphomas, does not require endogenous Bcl-2. In striking contrast, we report here that loss in these mice of its close relative Bcl-x(L) attenuated the pre-neoplastic expansion of pro-B and pre-B cells otherwise driven by c-Myc overexpression, sensitized these cells to apoptosis and ablated lymphoma formation. Remarkably, even loss of a single bcl-x allele delayed the lymphomagenesis. These findings identify Bcl-x(L) as a prerequisite for the emergence of c-Myc-driven pre-B/B lymphoma and suggest that BH3 mimetic drugs may provide a prophylactic strategy for c-Myc-driven tumors.
Collapse
|
126
|
Chesler L, Weiss WA. Genetically engineered murine models--contribution to our understanding of the genetics, molecular pathology and therapeutic targeting of neuroblastoma. Semin Cancer Biol 2011; 21:245-55. [PMID: 21958944 PMCID: PMC3504935 DOI: 10.1016/j.semcancer.2011.09.011] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Accepted: 09/14/2011] [Indexed: 01/01/2023]
Abstract
Genetically engineered mouse models (GEMM) have made major contributions to a molecular understanding of several adult cancers and these results are increasingly being translated into the pre-clinical setting where GEMM will very likely make a major impact on the development of targeted therapeutics in the near future. The relationship of pediatric cancers to altered developmental programs, and their genetic simplicity relative to adult cancers provides unique opportunities for the application of new advances in GEMM technology. In neuroblastoma the well-characterized TH-MYCN GEMM is increasingly used for a variety of molecular-genetic, developmental and pre-clinical therapeutics applications. We discuss: the present and historical application of GEMM to neuroblastoma research, future opportunities, and relevant targets suitable for new GEMM strategies in neuroblastoma. We review the potential of these models to contribute both to an understanding of the developmental nature of neuroblastoma and to improved therapy for this disease.
Collapse
Affiliation(s)
- Louis Chesler
- Division of Clinical Studies and Cancer Therapeutics, The Institute of Cancer Research & The Royal Marsden NHS Trust, Sutton, Surrey SM2 5NG, United Kingdom.
| | | |
Collapse
|
127
|
Robson SC, Ward L, Brown H, Turner H, Hunter E, Pelengaris S, Khan M. Deciphering c-MYC-regulated genes in two distinct tissues. BMC Genomics 2011; 12:476. [PMID: 21961992 PMCID: PMC3206520 DOI: 10.1186/1471-2164-12-476] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2011] [Accepted: 09/30/2011] [Indexed: 12/31/2022] Open
Abstract
Background The transcription factor MYC is a critical regulator of diverse cellular processes, including both replication and apoptosis. Differences in MYC-regulated gene expression responsible for such opposing outcomes in vivo remain obscure. To address this we have examined time-dependent changes in global gene expression in two transgenic mouse models in which MYC activation, in either skin suprabasal keratinocytes or pancreatic islet β-cells, promotes tissue expansion or involution, respectively. Results Consistent with observed phenotypes, expression of cell cycle genes is increased in both models (albeit enriched in β-cells), as are those involved in cell growth and metabolism, while expression of genes involved in cell differentiation is down-regulated. However, in β-cells, which unlike suprabasal keratinocytes undergo prominent apoptosis from 24 hours, there is up-regulation of genes associated with DNA-damage response and intrinsic apoptotic pathways, including Atr, Arf, Bax and Cycs. In striking contrast, this is not the case for suprabasal keratinocytes, where pro-apoptotic genes such as Noxa are down-regulated and key anti-apoptotic pathways (such as Igf1-Akt) and those promoting angiogenesis are up-regulated. Moreover, dramatic up-regulation of steroid hormone-regulated Kallikrein serine protease family members in suprabasal keratinocytes alone could further enhance local Igf1 actions, such as through proteolysis of Igf1 binding proteins. Conclusions Activation of MYC causes cell growth, loss of differentiation and cell cycle entry in both β-cells and suprabasal keratinocytes in vivo. Apoptosis, which is confined to β-cells, may involve a combination of a DNA-damage response and downstream activation of pro-apoptotic signalling pathways, including Cdc2a and p19Arf/p53, and downstream targets. Conversely, avoidance of apoptosis in suprabasal keratinocytes may result primarily from the activation of key anti-apoptotic signalling pathways, particularly Igf1-Akt, and induction of an angiogenic response, though intrinsic resistance to induction of p19Arf by MYC in suprabasal keratinocytes may contribute.
Collapse
Affiliation(s)
- Samuel C Robson
- Wellcome Trust/Cancer Research UK Gurdon Institute, Cambridge, CB2 1QN, UK
| | | | | | | | | | | | | |
Collapse
|
128
|
Smith RW, Mothersill C, Hinton T, Seymour CB. Exposure to low level chronic radiation leads to adaptation to a subsequent acute X-ray dose and communication of modified acute X-ray induced bystander signals in medaka (Japanese rice fish, Oryzias latipes). Int J Radiat Biol 2011; 87:1011-22. [DOI: 10.3109/09553002.2011.587861] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Richard W. Smith
- Medical Physics and Applied Radiation Sciences, McMaster University,
Hamilton, Ontario, Canada
| | - Carmel Mothersill
- Medical Physics and Applied Radiation Sciences, McMaster University,
Hamilton, Ontario, Canada
| | - Thomas Hinton
- Laboratoire de Radioécologie et d'Ecotoxicologie IRSN/DEI/SECRE (Bâtiment 159) Institut de Radioprotection et de Sûreté Nucléaire (IRSN) Centre de Cadarache Saint Paul-lez-Durance, France
| | - Colin B. Seymour
- Medical Physics and Applied Radiation Sciences, McMaster University,
Hamilton, Ontario, Canada
| |
Collapse
|
129
|
Höglund A, Nilsson LM, Muralidharan SV, Hasvold LA, Merta P, Rudelius M, Nikolova V, Keller U, Nilsson JA. Therapeutic implications for the induced levels of Chk1 in Myc-expressing cancer cells. Clin Cancer Res 2011; 17:7067-79. [PMID: 21933891 DOI: 10.1158/1078-0432.ccr-11-1198] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The transcription factor c-Myc (or "Myc") is a master regulator of pathways driving cell growth and proliferation. MYC is deregulated in many human cancers, making its downstream target genes attractive candidates for drug development. We report the unexpected finding that B-cell lymphomas from mice and patients exhibit a striking correlation between high levels of Myc and checkpoint kinase 1 (Chk1). EXPERIMENTAL DESIGN By in vitro cell biology studies as well as preclinical studies using a genetically engineered mouse model, we evaluated the role of Chk1 in Myc-overexpressing cells. RESULTS We show that Myc indirectly induces Chek1 transcript and protein expression, independently of DNA damage response proteins such as ATM and p53. Importantly, we show that inhibition of Chk1, by either RNA interference or a novel highly selective small molecule inhibitor, results in caspase-dependent apoptosis that affects Myc-overexpressing cells in both in vitro and in vivo mouse models of B-cell lymphoma. CONCLUSION Our data suggest that Chk1 inhibitors should be further evaluated as potential drugs against Myc-driven malignancies such as certain B-cell lymphoma/leukemia, neuroblastoma, and some breast and lung cancers.
Collapse
Affiliation(s)
- Andreas Höglund
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
130
|
Karslioglu E, Kleinberger JW, Salim FG, Cox AE, Takane KK, Scott DK, Stewart AF. cMyc is a principal upstream driver of beta-cell proliferation in rat insulinoma cell lines and is an effective mediator of human beta-cell replication. Mol Endocrinol 2011; 25:1760-72. [PMID: 21885567 DOI: 10.1210/me.2011-1074] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Adult human β-cells replicate slowly. Also, despite the abundance of rodent β-cell lines, there are no human β-cell lines for diabetes research or therapy. Prior studies in four commonly studied rodent β-cell lines revealed that all four lines displayed an unusual, but strongly reproducible, cell cycle signature: an increase in seven G(1)/S molecules, i.e. cyclins A, D3, and E, and cdk1, -2, -4, and -6. Here, we explore the upstream mechanism(s) that drive these cell cycle changes. Using biochemical, pharmacological and molecular approaches, we surveyed potential upstream mitogenic signaling pathways in Ins 1 and RIN cells. We used both underexpression and overexpression to assess effects on rat and human β-cell proliferation, survival and cell cycle control. Our results indicate that cMyc is: 1) uniquely up-regulated among other candidates; 2) principally responsible for the increase in the seven G(1)/S molecules; and, 3) largely responsible for proliferation in rat β-cell lines. Importantly, cMyc expression in β-cell lines, although some 5- to 7-fold higher than normal rat β-cells, is far below the levels (75- to 150-fold) previously associated with β-cell death and dedifferentiation. Notably, modest overexpression of cMyc is able to drive proliferation without cell death in normal rat and human β-cells. We conclude that cMyc is an important driver of replication in the two most commonly employed rat β-cell lines. These studies reverse the current paradigm in which cMyc overexpression is inevitably associated with β-cell death and dedifferentiation. The cMyc pathway provides potential approaches, targets, and tools for driving and sustaining human β-cell replication.
Collapse
Affiliation(s)
- Esra Karslioglu
- Division of Endocrinology, the University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15260, USA
| | | | | | | | | | | | | |
Collapse
|
131
|
Aparicio A, Den RB, Knudsen KE. Time to stratify? The retinoblastoma protein in castrate-resistant prostate cancer. Nat Rev Urol 2011; 8:562-8. [PMID: 21811228 DOI: 10.1038/nrurol.2011.107] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
It is generally held that the retinoblastoma (RB) tumor suppressor functions in multiple tissues to protect against tumor development. However, preclinical studies and analysis of tumor samples of early disease did not support an important role of RB loss in the origin of prostate cancer. By contrast, recent observations in the clinical setting and subsequent modeling of RB function indicate that the tumor suppressor has specialized roles in controlling androgen receptor expression in prostate cancer, and primarily functions to prevent progression to the castration-resistant stage of disease. Furthermore, preclinical models have now shown that loss of RB expression or functional activity decreases the effectiveness of hormone therapy, yet seems to increase sensitivity to a subset of chemotherapeutic agents. Here, the current state of knowledge regarding the implications of RB loss for prostate cancer progression will be reviewed, and potential opportunities for developing RB as a metric to predict therapeutic response will be considered.
Collapse
Affiliation(s)
- Ana Aparicio
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | | | | |
Collapse
|
132
|
Abstract
Transforming growth factor (TGF)-β antagonizes mitogenic Ras signaling during epithelial regeneration, but TGF-β and Ras act synergistically in driving tumor progression. Insights into these apparently contradictory effects have come from recent detailed analyses of the TGF-β signaling process. Here, we summarize the different modes of TGF-β/Ras signaling in normal epithelium and neoplasms and show how perturbation of TGF-β signaling by Ras may contribute to a shift from tumor-suppressive to protumorigenic TGF-β activity during tumor progression. Smad proteins, which convey signals from TGF-β receptors to the nucleus, have intermediate linker regions between conserved Mad homology (MH) 1 and MH2 domains. TGF-β Type I receptor and Ras-associated kinases differentially phosphorylate Smad2 and Smad3 to create C-terminally (C), linker (L) or dually (L/C) phosphorylated (p) isoforms. In epithelial homeostasis, TGF-β-mediated pSmad3C signaling opposes proliferative responses induced by mitogenic signals. During carcinogenesis, activation of cytoplasmic Ras-associated kinases including mitogen-activated protein kinase confers a selective advantage on benign tumors by shifting Smad3 signaling from a tumor-suppressive pSmad3C to an oncogenic pSmad3L pathway, leading to carcinoma in situ. Finally, at the edges of advanced carcinomas invading adjacent tissues, nuclear Ras-associated kinases such as cyclin-dependent kinases, together with cytoplasmic kinases, alter TGF-β signals to more invasive and proliferative pSmad2L/C and pSmad3L/C signaling. Taken together, TGF-β signaling specificity arises from spatiotemporal dynamics of Smad phosphoisoforms. Based on these findings, we have reason to hope that pharmacologic inhibition of linker phosphorylation might suppress progression to human advanced carcinomas by switching from protumorigenic to tumor-suppressive TGF-β signaling.
Collapse
Affiliation(s)
- Koichi Matsuzaki
- Department of Gastroenterology and Hepatology, Kansai Medical University, 10-15 Fumizonocho, Moriguchi, Osaka 570-8506, Japan.
| |
Collapse
|
133
|
Duensing S, Darr S, Cuevas R, Melquiot N, Brickner AG, Duensing A, Münger K. Tripeptidyl Peptidase II Is Required for c-MYC-Induced Centriole Overduplication and a Novel Therapeutic Target in c-MYC-Associated Neoplasms. Genes Cancer 2011; 1:883-92. [PMID: 21647238 DOI: 10.1177/1947601910389605] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2010] [Revised: 10/12/2010] [Accepted: 10/12/2010] [Indexed: 12/20/2022] Open
Abstract
Centrosome aberrations are frequently detected in c-MYC-associated human malignancies. Here, we show that c-MYC-induced centrosome and centriole overduplication critically depend on the protease tripeptidyl peptidase II (TPPII). We found that TPPII localizes to centrosomes and that overexpression of TPPII, similar to c-MYC, can disrupt centriole duplication control and cause centriole multiplication, a process during which maternal centrioles nucleate the formation of more than a single daughter centriole. We report that inactivation of TPPII using chemical inhibitors or siRNA-mediated protein knockdown effectively reduced c-MYC-induced centriole overduplication. Remarkably, the potent and selective TPPII inhibitor butabindide not only potently suppressed centriole aberrations but also caused significant cell death and growth suppression in aggressive human Burkitt lymphoma cells with c-MYC overexpression. Taken together, these results highlight the role of TPPII in c-MYC-induced centriole overduplication and encourage further studies to explore TPPII as a novel antineoplastic drug target.
Collapse
Affiliation(s)
- Stefan Duensing
- Cancer Virology Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | | | | | | | | | | | | |
Collapse
|
134
|
NAKAMURA IKUO, ROBERTS LEWISR. Myc, Max, and Mnt: molecular mechanisms of enhancement of cholangiocarcinogenesis by cholestasis. Gastroenterology 2011; 141:32-4. [PMID: 21620848 PMCID: PMC4040264 DOI: 10.1053/j.gastro.2011.05.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
|
135
|
Mora-Jensen H, Jendholm J, Fossum A, Porse B, Borregaard N, Theilgaard-Mönch K. Technical Advance: Immunophenotypical characterization of human neutrophil differentiation. J Leukoc Biol 2011; 90:629-34. [DOI: 10.1189/jlb.0311123] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
136
|
Shani T, Primov-Fever A, Wolf M, Shalmon B, Amarglio N, Trakhtenbrot L, Hirshberg A. Noninvasive detection of aneuploid cells in laryngeal epithelial precursor lesions. Cancer Cytopathol 2011; 119:235-46. [DOI: 10.1002/cncy.20157] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2010] [Revised: 03/11/2011] [Accepted: 03/17/2011] [Indexed: 01/22/2023]
|
137
|
Posttranscriptional deregulation of MYC via PTEN constitutes a major alternative pathway of MYC activation in T-cell acute lymphoblastic leukemia. Blood 2011; 117:6650-9. [PMID: 21527520 DOI: 10.1182/blood-2011-02-336842] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Cumulative evidence indicates that MYC, one of the major downstream effectors of NOTCH1, is a critical component of T-cell acute lymphoblastic leukemia (T-ALL) oncogenesis and a potential candidate for targeted therapy. However, MYC is a complex oncogene, involving both fine protein dosage and cell-context dependency, and detailed understanding of MYC-mediated oncogenesis in T-ALL is still lacking. To better understand how MYC is interspersed in the complex T-ALL oncogenic networks, we performed a thorough molecular and biochemical analysis of MYC activation in a comprehensive collection of primary adult and pediatric patient samples. We find that MYC expression is highly variable, and that high MYC expression levels can be generated in a large number of cases in absence of NOTCH1/FBXW7 mutations, suggesting the occurrence of multiple activation pathways in addition to NOTCH1. Furthermore, we show that posttranscriptional deregulation of MYC constitutes a major alternative pathway of MYC activation in T-ALL, operating partly via the PI3K/AKT axis through down-regulation of PTEN, and that NOTCH1(m) might play a dual transcriptional and posttranscriptional role in this process. Altogether, our data lend further support to the significance of therapeutic targeting of MYC and/or the PTEN/AKT pathways, both in GSI-resistant and identified NOTCH1-independent/MYC-mediated T-ALL patients.
Collapse
|
138
|
Wang C, Tai Y, Lisanti MP, Liao DJ. c-Myc induction of programmed cell death may contribute to carcinogenesis: a perspective inspired by several concepts of chemical carcinogenesis. Cancer Biol Ther 2011; 11:615-26. [PMID: 21278493 DOI: 10.4161/cbt.11.7.14688] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The c-Myc protein, encoded by c-myc gene, in its wild-type form can induce tumors with a high frequency and can induce massive programmed cell death (PCD) in most transgenic mouse models, with greater efficiency than other oncogenes. Evidence also indicates that c-Myc can cause proliferative inhibition, i.e. mitoinhibition. The c-Myc-induced PCD and mitoinhibition, which may be attributable to its inhibition of cyclin D1 and induction of p53, may impose a pressure of compensatory proliferation, i.e. regeneration, onto the initiated cells (cancer progenitor cells) that occur sporadically and are resistant to the mitoinhibition. The initiated cells can thus proliferate robustly and progress to a malignancy. This hypothetical thinking, i.e. the concurrent PCD and mitoinhibition induced by c-Myc can promote carcinogenesis, predicts that an optimal balance is achieved between cell death and ensuing regeneration during oncogenic transformation by c-Myc, which can better promote carcinogenesis. In this perspective, we summarize accumulating evidence and challenge the current model that oncoprotein induces carcinogenesis by promoting cellular proliferation and/or inhibiting PCD. Inspired by c-myc oncogene, we surmise that many tumor-suppressive or growth-inhibitory genes may also be able to promote carcinogenesis in a similar way, i.e. by inducing PCD and/or mitoinhibition of normal cells to create a need for compensatory proliferation that drives a robust replication of initiating cells.
Collapse
Affiliation(s)
- Chenguang Wang
- Department of Stem Cell and Regenerative Medicine, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | | | | | | |
Collapse
|
139
|
Kelly PN, Strasser A. The role of Bcl-2 and its pro-survival relatives in tumourigenesis and cancer therapy. Cell Death Differ 2011; 18:1414-24. [PMID: 21415859 DOI: 10.1038/cdd.2011.17] [Citation(s) in RCA: 351] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Tumour development requires a combination of defects that allow nascent neoplastic cells to become self-sufficient for cell proliferation and insensitive to signals that normally restrain cell growth. Among the latter, evasion of programmed cell death (apoptosis) has proven to be critical for the development and sustained growth of many, perhaps all, cancers. Apoptotic cell death is regulated by complex interactions between pro-survival members and two subgroups of pro-apoptotic members of the B-cell lymphoma-2 (Bcl-2) protein family. In this invited review article, we reminisce on the discovery of Bcl-2, the first regulator of cell death identified, we discuss the mechanisms that control apoptotic cell death, focussing on how defects in this process promote the development and sustained growth of tumours and also affect their responses to anticancer therapeutics and, finally, we describe how current knowledge of the regulatory networks of apoptosis is exploited to develop novel approaches for cancer therapy.
Collapse
Affiliation(s)
- P N Kelly
- Molecular Genetics of Cancer Division, Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3050, Australia
| | | |
Collapse
|
140
|
Boutz DR, Collins PJ, Suresh U, Lu M, Ramírez CM, Fernández-Hernando C, Huang Y, Abreu RDS, Le SY, Shapiro BA, Liu AM, Luk JM, Aldred SF, Trinklein ND, Marcotte EM, Penalva LOF. Two-tiered approach identifies a network of cancer and liver disease-related genes regulated by miR-122. J Biol Chem 2011; 286:18066-78. [PMID: 21402708 DOI: 10.1074/jbc.m110.196451] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
MicroRNAs function as important regulators of gene expression and are commonly linked to development, differentiation, and diseases such as cancer. To better understand their roles in various biological processes, identification of genes targeted by microRNAs is necessary. Although prediction tools have significantly helped with this task, experimental approaches are ultimately required for extensive target search and validation. We employed two independent yet complementary high throughput approaches to map a large set of mRNAs regulated by miR-122, a liver-specific microRNA implicated in regulation of fatty acid and cholesterol metabolism, hepatitis C infection, and hepatocellular carcinoma. The combination of luciferase reporter-based screening and shotgun proteomics resulted in the identification of 260 proteins significantly down-regulated in response to miR-122 in at least one method, 113 of which contain predicted miR-122 target sites. These proteins are enriched for functions associated with the cell cycle, differentiation, proliferation, and apoptosis. Among these miR-122-sensitive proteins, we identified a large group with strong connections to liver metabolism, diseases, and hepatocellular carcinoma. Additional analyses, including examination of consensus binding motifs for both miR-122 and target sequences, provide further insight into miR-122 function.
Collapse
Affiliation(s)
- Daniel R Boutz
- Center for Systems and Synthetic Biology, Institute for Cellular and Molecular Biology, University of Texas, Austin, Texas 78712, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
141
|
Cell Cycle Deregulation in Ewing's Sarcoma Pathogenesis. Sarcoma 2010; 2011:598704. [PMID: 21052502 PMCID: PMC2968116 DOI: 10.1155/2011/598704] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Accepted: 10/07/2010] [Indexed: 12/18/2022] Open
Abstract
Ewing's sarcoma is a highly aggressive pediatric tumor of bone that usually contains the characteristic chromosomal translocation t(11;22)(q24;q12). This translocation encodes the oncogenic fusion protein EWS/FLI, which acts as an aberrant transcription factor to deregulate target genes necessary for oncogenesis. One key feature of oncogenic transformation is dysregulation of cell cycle control. It is therefore likely that EWS/FLI and other cooperating mutations in Ewing's sarcoma modulate the cell cycle to facilitate tumorigenesis. This paper will summarize current published data associated with deregulation of the cell cycle in Ewing's sarcoma and highlight important questions that remain to be answered.
Collapse
|