Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Michael Dews is active.

Publication


Featured researches published by Michael Dews.


Nature Genetics | 2006

Augmentation of tumor angiogenesis by a Myc-activated microRNA cluster

Michael Dews; Asal Homayouni; Duonan Yu; Danielle A. Murphy; Cinzia Sevignani; Emma E. Furth; William M. F. Lee; Greg H. Enders; Joshua T. Mendell; Andrei Thomas-Tikhonenko

Human adenocarcinomas commonly harbor mutations in the KRAS and MYC proto-oncogenes and the TP53 tumor suppressor gene. All three genetic lesions are potentially pro-angiogenic, as they sustain production of vascular endothelial growth factor (VEGF). Yet Kras-transformed mouse colonocytes lacking p53 formed indolent, poorly vascularized tumors, whereas additional transduction with a Myc-encoding retrovirus promoted vigorous vascularization and growth. In addition, VEGF levels were unaffected by Myc, but enhanced neovascularization correlated with downregulation of anti-angiogenic thrombospondin-1 (Tsp1) and related proteins, such as connective tissue growth factor (CTGF). Both Tsp1 and CTGF are predicted targets for repression by the miR-17-92 microRNA cluster, which was upregulated in colonocytes coexpressing K-Ras and c-Myc. Indeed, miR-17-92 knockdown with antisense 2′-O-methyl oligoribonucleotides partly restored Tsp1 and CTGF expression; in addition, transduction of Ras-only cells with a miR-17-92–encoding retrovirus reduced Tsp1 and CTGF levels. Notably, miR-17-92–transduced cells formed larger, better-perfused tumors. These findings establish a role for microRNAs in non–cell-autonomous Myc-induced tumor phenotypes.


Molecular and Cellular Biology | 1999

Multiple Signaling Pathways of the Insulin-Like Growth Factor 1 Receptor in Protection from Apoptosis

Francesca Peruzzi; Marco Prisco; Michael Dews; Paolo Salomoni; Emanuela Grassilli; Gaetano Romano; Bruno Calabretta; Renato Baserga

ABSTRACT The type 1 insulin-like growth factor receptor (IGF-1R), activated by its ligands, protects several cell types from a variety of apoptotic injuries. The main signaling pathway for IGF-1R-mediated protection from apoptosis has been previously elucidated and rests on the activation of phosphatidylinositol 3-kinase, Akt/protein kinase B, and the phosphorylation and inactivation of BAD, a member of the Bcl-2 family of proteins. In 32D cells (a murine hemopoietic cell line devoid of insulin receptor substrate 1 [IRS-1]), the IGF-1R activates alternative pathways for protection from apoptosis induced by withdrawal of interleukin-3. One of these pathways leads to the activation of mitogen-activated protein kinase, while a third pathway results in the mitochondrial translocation of Raf and depends on the integrity of a group of serines in the C terminus of the receptor that are known to interact with 14.3.3 proteins. All three pathways, however, result in BAD phosphorylation. The presence of multiple antiapoptotic pathways may explain the remarkable efficacy of the IGF-1R in protecting cells from apoptosis.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Lin-28B transactivation is necessary for Myc-mediated let-7 repression and proliferation

Tsung Cheng Chang; Lauren R. Zeitels; Hun-Way Hwang; Raghu R. Chivukula; Michael Dews; Jason J. Jung; Ping Gao; Chi V. Dang; Michael Beer; Andrei Thomas-Tikhonenko; Joshua T. Mendell

Direct control of microRNA (miRNA) expression by oncogenic and tumor suppressor networks results in frequent dysregulation of miRNAs in cancer cells and contributes to tumorigenesis. We previously demonstrated that activation of the c-Myc oncogenic transcription factor (Myc) broadly influences miRNA expression and in particular leads to widespread miRNA down-regulation. miRNA transcripts repressed by Myc include several with potent tumor suppressor activity such as miR-15a/16–1, miR-34a, and let-7 family members. In this study, we have investigated mechanisms downstream of Myc that contribute to miRNA repression. Consistent with transcriptional down-regulation, Myc activity results in the decreased abundance of multiple miRNA primary transcripts. Surprisingly, however, primary transcripts encoding several let-7 miRNAs are not reduced in the high Myc state, suggesting a posttranscriptional mechanism of repression. The Lin-28 and Lin-28B RNA binding proteins were recently demonstrated to negatively regulate let-7 biogenesis. We now show that Myc induces Lin-28B expression in multiple human and mouse tumor models. Chromatin immunoprecipitation and reporter assays reveal direct association of Myc with the Lin-28B promoter resulting in transcriptional transactivation. Moreover, we document that activation of Lin-28B is necessary and sufficient for Myc-mediated let-7 repression. Accordingly, Lin-28B loss-of-function significantly impairs Myc-dependent cellular proliferation. These findings highlight an important role for Lin-28B in Myc-driven cellular phenotypes and uncover an orchestration of transcriptional and posttranscriptional mechanisms in Myc-mediated reprogramming of miRNA expression.


Molecular Cell | 2010

The miR-17-92 microRNA cluster regulates multiple components of the TGF-β pathway in neuroblastoma.

Pieter Mestdagh; Anna-Karin Boström; Francis Impens; Erik Fredlund; Gert Van Peer; Pasqualino De Antonellis; Kristoffer von Stedingk; Bart Ghesquière; Stefanie Schulte; Michael Dews; Andrei Thomas-Tikhonenko; Johannes H. Schulte; Massimo Zollo; Alexander Schramm; Kris Gevaert; Håkan Axelson; Franki Speleman; Jo Vandesompele

The miR-17-92 microRNA cluster is often activated in cancer cells, but the identity of its targets remains elusive. Using SILAC and quantitative mass spectrometry, we examined the effects of activation of the miR-17-92 cluster on global protein expression in neuroblastoma (NB) cells. Our results reveal cooperation between individual miR-17-92 miRNAs and implicate miR-17-92 in multiple hallmarks of cancer, including proliferation and cell adhesion. Most importantly, we show that miR-17-92 is a potent inhibitor of TGF-β signaling. By functioning both upstream and downstream of pSMAD2, miR-17-92 activation triggers downregulation of multiple key effectors along the TGF-β signaling cascade as well as direct inhibition of TGF-β-responsive genes.


Molecular and Cellular Biology | 2004

Direct repression of FLIP expression by c-myc is a major determinant of TRAIL sensitivity

M. Stacey Ricci; Zhaoyu Jin; Michael Dews; Duonan Yu; Andrei Thomas-Tikhonenko; David T. Dicker; Wafik S. El-Deiry

ABSTRACT Tumor necrosis factor alpha (TNF-α)-related apoptosis-inducing ligand (TRAIL) is a member of the TNF-α family of death receptor ligands and holds great therapeutic potential as a tumor cell-specific cytotoxic agent. Using a panel of established tumor cell lines and normal cells, we found a significant difference between the number of TRAIL-sensitive cells expressing high levels of c-myc and TRAIL-resistant cells expressing low levels of c-myc (P < 0.05, n = 19). We also found a direct linear correlation between c-myc levels and TRAIL sensitivity in TRAIL-sensitive cell lines (r = 0.94, n = 6). Overexpression of c-myc or activation of a myc-estrogen receptor (ER) fusion sensitized TRAIL-resistant cells to TRAIL. Conversely, small interfering RNA (siRNA)-mediated knockdown of c-myc significantly reduced both c-myc expression and TRAIL-induced apoptosis. The gene encoding the inhibitor of caspase activation, FLICE inhibitory protein (FLIP), appears to be a direct target of c-myc-mediated transcriptional repression. Overexpression of c-myc or activation of myc-estrogen receptor (ER) decreased FLIP levels both in cell culture and in mouse models of c-myc-induced tumorigenesis, while knocking down c-myc using siRNA increased FLIP expression. Chromatin immunoprecipitation and luciferase reporter analyses showed that c-myc binds and represses the human FLIP promoter. c-myc expression enhanced TRAIL-induced caspase 8 cleavage and FLIP cleavage at the death-inducing signaling complex. Combined siRNA-mediated knockdown of FLIP and c-myc resensitized cells to TRAIL. Therefore, c-myc down-regulation of FLIP expression provides a universal mechanism to explain the ability of c-myc to sensitize cells to death receptor stimuli. In addition, identification of c-myc as a major determinant of TRAIL sensitivity provides a potentially important screening tool for identification of TRAIL-sensitive tumors.


Cancer Research | 2010

The Myc–miR-17∼92 Axis Blunts TGFβ Signaling and Production of Multiple TGFβ-Dependent Antiangiogenic Factors

Michael Dews; Jamie L. Fox; Stacy Hultine; Prema Sundaram; Wenge Wang; Yingqiu Y. Liu; Emma E. Furth; Gregory H. Enders; Wafik S. El-Deiry; Janell M. Schelter; Michele A. Cleary; Andrei Thomas-Tikhonenko

c-Myc stimulates angiogenesis in tumors through mechanisms that remain incompletely understood. Recent work indicates that c-Myc upregulates the miR-17∼92 microRNA cluster and downregulates the angiogenesis inhibitor thrombospondin-1, along with other members of the thrombospondin type 1 repeat superfamily. Here, we show that downregulation of the thrombospondin type 1 repeat protein clusterin in cells overexpressing c-Myc and miR-17∼92 promotes angiogenesis and tumor growth. However, clusterin downregulation by miR-17∼92 is indirect. It occurs as a result of reduced transforming growth factor-β (TGFβ) signaling caused by targeting of several regulatory components in this signaling pathway. Specifically, miR-17-5p and miR-20 reduce the expression of the type II TGFβ receptor and miR-18 limits the expression of Smad4. Supporting these results, in human cancer cell lines, levels of the miR-17∼92 primary transcript MIR17HG negatively correlate with those of many TGFβ-induced genes that are not direct targets of miR-17∼92 (e.g., clusterin and angiopoietin-like 4). Furthermore, enforced expression of miR-17∼92 in MIR17HG(low) cell lines (e.g., glioblastoma) results in impaired gene activation by TGFβ. Together, our results define a pathway in which c-Myc activation of miR-17∼92 attenuates the TGFβ signaling pathway to shut down clusterin expression, thereby stimulating angiogenesis and tumor cell growth.


Endocrine | 1997

The IGF-I receptor and cancer

Renato Baserga; Mariana Resnicoff; Michael Dews

It has been known for a long time that the growth of tumors depends not only on the rate of cell proliferation, but also on the rate of cell death (I). An increase in the former and a decrease in the latter cumulatively result in an increase in cell number, which is the most relevant characteristic of tumors. From this perspective, the insulin-like growth factor 1 receptor (IGF-IR) is an important component of tumor growth (and a good potential target for therapeutic interventions), because it both stimulates cell proliferation and protects cells from cell death in general, and apoptosis in particular. Indeed, a functional impairment of the IGFIR actually achieves the dual goal of inhibiting cell proliferation and of inducing massive apoptosis. In this presentation, the authors will briefly summarize the functions of the IGF-IR related to growth, and then focus on some aspects of its targeting that are unusually interesting.


Journal of the National Cancer Institute | 2009

Clusterin, a haploinsufficient tumor suppressor gene in neuroblastomas.

Olesya Chayka; Daisy Corvetta; Michael Dews; Alessandro E. Caccamo; Izabela Piotrowska; Giorgia Santilli; Sian Gibson; Nj Sebire; Nourredine Himoudi; Michael D. Hogarty; John Anderson; Saverio Bettuzzi; Andrei Thomas-Tikhonenko; Arturo Sala

BACKGROUND Clusterin expression in various types of human cancers may be higher or lower than in normal tissue, and clusterin may promote or inhibit apoptosis, cell motility, and inflammation. We investigated the role of clusterin in tumor development in mouse models of neuroblastoma. METHODS We assessed expression of microRNAs in the miR-17-92 cluster by real-time reverse transcription-polymerase chain reaction in MYCN-transfected SH-SY5Y and SH-EP cells and inhibited expression by transfection with microRNA antisense oligonucleotides. Tumor development was studied in mice (n = 66) that were heterozygous or homozygous for the MYCN transgene and/or for the clusterin gene; these mice were from a cross between MYCN-transgenic mice, which develop neuroblastoma, and clusterin-knockout mice. Tumor growth and metastasis were studied in immunodeficient mice that were injected with human neuroblastoma cells that had enhanced (by clusterin transfection, four mice per group) or reduced (by clusterin short hairpin RNA [shRNA] transfection, eight mice per group) clusterin expression. All statistical tests were two-sided. RESULTS Clusterin expression increased when expression of MYCN-induced miR-17-92 microRNA cluster in SH-SY5Y neuroblastoma cells was inhibited by transfection with antisense oligonucleotides compared with scrambled oligonucleotides. Statistically significantly more neuroblastoma-bearing MYCN-transgenic mice were found in groups with zero or one clusterin allele than in those with two clusterin alleles (eg, 12 tumor-bearing mice in the zero-allele group vs three in the two-allele group, n = 22 mice per group; relative risk for neuroblastoma development = 4.85, 95% confidence interval [CI] = 1.69 to 14.00; P = .005). Five weeks after injection, fewer clusterin-overexpressing LA-N-5 human neuroblastoma cells than control cells were found in mouse liver or bone marrow, but statistically significantly more clusterin shRNA-transfected HTLA230 cells (3.27%, with decreased clusterin expression) than control-transfected cells (1.53%) were found in the bone marrow (difference = 1.74%, 95% CI = 0.24% to 3.24%, P = .026). CONCLUSIONS We report, to our knowledge, the first genetic evidence that clusterin is a tumor and metastasis suppressor gene.


Oncogene | 1997

Co-operation of Simian virus 40 T antigen and insulin receptor substrate-1 in protection from apoptosis induced by Interleukin-3 withdrawal

Fei Zhou-Li; Shi-Qiong Xu; Michael Dews; Renato Baserga

32D cells are interleukin-3 (IL-3) dependent murine hemopoietic cells, that undergo apoptosis after IL-3 withdrawal. An overexpressed insulin-like growth factor I receptor (IGF-IR) protects these cells from apoptosis induced by IL-3 withdrawal. When 32D cells are stably transfected with plasmids expressing either IRS-1 (a major substrate of the IGF-IR) or the Simian virus 40 large T antigen, singly, they still undergo apoptosis after IL-3 withdrawal, although IRS-1 offers partial protection. The cells, however, are fully protected when they are stably transfected with both IRS-1 and SV40 T antigen. Protection from apoptosis in these cells is characterized by the stabilization of the Stat1 and Stat5 protein levels, whose synthesis is inhibited when IL-3 is withdrawn.


Cancer Research | 2004

Myc-Transformed Epithelial Cells Down-Regulate Clusterin, Which Inhibits Their Growth in Vitro and Carcinogenesis in Vivo

Andrei Thomas-Tikhonenko; Isabelle Viard-Leveugle; Michael Dews; Philippe Wehrli; Cinzia Sevignani; Duonan Yu; Stacey Ricci; Wafik S. El-Deiry; Bruce J. Aronow; Gürkan Kaya; Jean-Hilaire Saurat; Lars E. French

Effective treatment of malignant carcinomas requires identification of proteins regulating epithelial cell proliferation. To this end, we compared gene expression profiles in murine colonocytes and their c-Myc-transformed counterparts, which possess enhanced proliferative potential. A surprisingly short list of deregulated genes included the cDNA for clusterin, an extracellular glycoprotein without a firmly established function. We had previously demonstrated that in organs such as skin, clusterin expression is restricted to differentiating but not proliferating cell layers, suggesting a possible negative role in cell division. Indeed, its transient overexpression in Myc-transduced colonocytes decreased cell accumulation. Furthermore, clusterin was down-regulated in rapidly dividing human keratinocytes infected with a Myc-encoding adenovirus. Its knockdown via antisense RNA in neoplastic epidermoid cells enhanced proliferation. Finally, recombinant human clusterin suppressed, in a dose-dependent manner, DNA replication in keratinocytes and other cells of epithelial origin. Thus, clusterin appears to be an inhibitor of epithelial cell proliferation in vitro. To determine whether it also affects neoplastic growth in vivo, we compared wild-type and clusterin-null mice with respect to their sensitivity to 7, 12-dimethylbenz(a)anthracene /12-Otetradecanoylphorbol-13-acetate (DMBA/TPA)-induced skin carcinogenesis. We observed that the mean number of papillomas/mouse was higher in clusterin-null animals. Moreover, these papillomas did not regress as readily as in wild-type mice and persisted beyond week 35. The rate of progression toward squamous cell carcinoma was not altered, although those developing in clusterin-null mice were on average better differentiated. These data suggest that clusterin not only suppresses epithelial cell proliferation in vitro but also interferes with the promotion stage of skin carcinogenesis.

Collaboration


Dive into the Michael Dews's collaboration.

Top Co-Authors

Avatar

Andrei Thomas-Tikhonenko

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Renato Baserga

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar

Duonan Yu

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Gaetano Romano

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar

Joshua T. Mendell

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Andrea Morrione

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar

Barbara Valentinis

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar

Elaine Y. Chung

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Jamie L. Fox

University of Pennsylvania

View shared research outputs
Researchain Logo
Decentralizing Knowledge