Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Cell ; 171(6): 1301-1315.e14, 2017 Nov 30.
Article in English | MEDLINE | ID: mdl-29195074

ABSTRACT

The two oncogenes KRas and Myc cooperate to drive tumorigenesis, but the mechanism underlying this remains unclear. In a mouse lung model of KRasG12D-driven adenomas, we find that co-activation of Myc drives the immediate transition to highly proliferative and invasive adenocarcinomas marked by highly inflammatory, angiogenic, and immune-suppressed stroma. We identify epithelial-derived signaling molecules CCL9 and IL-23 as the principal instructing signals for stromal reprogramming. CCL9 mediates recruitment of macrophages, angiogenesis, and PD-L1-dependent expulsion of T and B cells. IL-23 orchestrates exclusion of adaptive T and B cells and innate immune NK cells. Co-blockade of both CCL9 and IL-23 abrogates Myc-induced tumor progression. Subsequent deactivation of Myc in established adenocarcinomas triggers immediate reversal of all stromal changes and tumor regression, which are independent of CD4+CD8+ T cells but substantially dependent on returning NK cells. We show that Myc extensively programs an immune suppressive stroma that is obligatory for tumor progression.


Subject(s)
Adenocarcinoma/immunology , Adenoma/immunology , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenoma/genetics , Adenoma/pathology , Animals , Carcinogenesis , Chemokines, CC/immunology , Disease Models, Animal , Female , Inflammation/immunology , Inflammation/metabolism , Interleukin-23/immunology , Lung Neoplasms/pathology , Macrophage Inflammatory Proteins/immunology , Macrophages/immunology , Male , Mice , Tumor Microenvironment
2.
Genes Dev ; 27(5): 504-13, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23475959

ABSTRACT

The principal reason for failure of targeted cancer therapies is the emergence of resistant clones that regenerate the tumor. Therapeutic efficacy therefore depends on not only how effectively a drug inhibits its target, but also the innate or adaptive functional redundancy of that target and its attendant pathway. In this regard, the Myc transcription factors are intriguing therapeutic targets because they serve the unique and irreplaceable role of coordinating expression of the many diverse genes that, together, are required for somatic cell proliferation. Furthermore, Myc expression is deregulated in most-perhaps all-cancers, underscoring its irreplaceable role in proliferation. We previously showed in a preclinical mouse model of non-small-cell lung cancer that systemic Myc inhibition using the dominant-negative Myc mutant Omomyc exerts a dramatic therapeutic impact, triggering rapid regression of tumors with only mild and fully reversible side effects. Using protracted episodic expression of Omomyc, we now demonstrate that metronomic Myc inhibition not only contains Ras-driven lung tumors indefinitely, but also leads to their progressive eradication. Hence, Myc does indeed serve a unique and nondegenerate role in lung tumor maintenance that cannot be complemented by any adaptive mechanism, even in the most aggressive p53-deficient tumors. These data endorse Myc as a compelling cancer drug target.


Subject(s)
Lung Neoplasms/metabolism , Peptide Fragments/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Animals , Animals, Genetically Modified , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Peptide Fragments/therapeutic use , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/pharmacology , Proto-Oncogene Proteins c-myc/therapeutic use , Survival Analysis , Tumor Suppressor Protein p53/metabolism
3.
Genes Dev ; 25(9): 907-16, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21478273

ABSTRACT

The ubiquitous deregulation of Myc in human cancers makes it an intriguing therapeutic target, a notion supported by recent studies in Ras-driven lung tumors showing that inhibiting endogenous Myc triggers ubiquitous tumor regression. However, neither the therapeutic mechanism nor the applicability of Myc inhibition to other tumor types driven by other oncogenic mechanisms is established. Here, we show that inhibition of endogenous Myc also triggers ubiquitous regression of tumors in a simian virus 40 (SV40)-driven pancreatic islet tumor model. Such regression is presaged by collapse of the tumor microenvironment and involution of tumor vasculature. Hence, in addition to its diverse intracellular roles, endogenous Myc serves an essential and nonredundant role in coupling diverse intracellular oncogenic pathways to the tumor microenvironment, further bolstering its credentials as a pharmacological target.


Subject(s)
Proto-Oncogene Proteins c-myc/metabolism , Tumor Microenvironment/physiology , Adenoma, Islet Cell , Animals , Antineoplastic Agents/pharmacology , Apoptosis/physiology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Proliferation/drug effects , Disease Models, Animal , Doxycycline/pharmacology , GTPase-Activating Proteins/metabolism , Gene Expression Regulation, Neoplastic , Mice , Neuroendocrine Tumors/blood supply , Neuroendocrine Tumors/pathology , Neuroendocrine Tumors/physiopathology , Protein Binding , Proto-Oncogene Proteins c-myc/genetics , Repressor Proteins/metabolism , Simian virus 40/physiology
4.
Nature ; 455(7213): 679-83, 2008 Oct 02.
Article in English | MEDLINE | ID: mdl-18716624

ABSTRACT

Myc is a pleiotropic basic helix-loop-helix leucine zipper transcription factor that coordinates expression of the diverse intracellular and extracellular programs that together are necessary for growth and expansion of somatic cells. In principle, this makes inhibition of Myc an attractive pharmacological approach for treating diverse types of cancer. However, enthusiasm has been muted by lack of direct evidence that Myc inhibition would be therapeutically efficacious, concerns that it would induce serious side effects by inhibiting proliferation of normal tissues, and practical difficulties in designing Myc inhibitory drugs. We have modelled genetically both the therapeutic impact and the side effects of systemic Myc inhibition in a preclinical mouse model of Ras-induced lung adenocarcinoma by reversible, systemic expression of a dominant-interfering Myc mutant. We show that Myc inhibition triggers rapid regression of incipient and established lung tumours, defining an unexpected role for endogenous Myc function in the maintenance of Ras-dependent tumours in vivo. Systemic Myc inhibition also exerts profound effects on normal regenerating tissues. However, these effects are well tolerated over extended periods and rapidly and completely reversible. Our data demonstrate the feasibility of targeting Myc, a common downstream conduit for many oncogenic signals, as an effective, efficient and tumour-specific cancer therapy.


Subject(s)
Genetic Therapy , Lung Neoplasms/therapy , Models, Biological , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/genetics , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma/therapy , Animals , Gastrointestinal Tract/cytology , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/pathology , Genes, Dominant/genetics , Genes, ras , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mice , Mutation/genetics , Oncogene Protein p21(ras)/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Skin/cytology , Skin/metabolism , Skin/pathology , Testis/cytology , Testis/metabolism , Testis/pathology , Transgenes/genetics
5.
Cancer Discov ; 13(11): 2339-2355, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37682219

ABSTRACT

The protein phosphatase SHP2/PTPN11 has been reported to be a key modulator of proliferative pathways in a wide range of malignancies. Intriguingly, SHP2 has also been described as a critical regulator of the tumor microenvironment. Based on this evidence SHP2 is considered a multifaceted target in cancer, spurring the notion that the development of direct inhibitors of SHP2 would provide the twofold benefit of tumor intrinsic and extrinsic inhibition. In this review, we will discuss the role of SHP2 in cancer and the tumor microenvironment, and the clinical strategies in which SHP2 inhibitors are leveraged as combination agents to improve therapeutic response. SIGNIFICANCE: The SHP2 phosphatase functions as a pleiotropic factor, and its inhibition not only hinders tumor growth but also reshapes the tumor microenvironment. Although their single-agent activity may be limited, SHP2 inhibitors hold the potential of being key combination agents to enhance the depth and the durability of tumor response to therapy.


Subject(s)
Neoplasms , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Tumor Microenvironment
6.
Nat Commun ; 14(1): 5195, 2023 09 06.
Article in English | MEDLINE | ID: mdl-37673892

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy in need of new therapeutic options. Using unbiased analyses of super-enhancers (SEs) as sentinels of core genes involved in cell-specific function, here we uncover a druggable SE-mediated RNA-binding protein (RBP) cascade that supports PDAC growth through enhanced mRNA translation. This cascade is driven by a SE associated with the RBP heterogeneous nuclear ribonucleoprotein F, which stabilizes protein arginine methyltransferase 1 (PRMT1) to, in turn, control the translational mediator ubiquitin-associated protein 2-like. All three of these genes and the regulatory SE are essential for PDAC growth and coordinately regulated by the Myc oncogene. In line with this, modulation of the RBP network by PRMT1 inhibition reveals a unique vulnerability in Myc-high PDAC patient organoids and markedly reduces tumor growth in male mice. Our study highlights a functional link between epigenetic regulation and mRNA translation and identifies components that comprise unexpected therapeutic targets for PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Male , Animals , Mice , RNA , Epigenesis, Genetic , Regulatory Sequences, Nucleic Acid , Pancreatic Neoplasms/genetics , Carcinoma, Pancreatic Ductal/genetics , Methyltransferases , RNA-Binding Proteins/genetics
7.
Nat Cancer ; 4(6): 812-828, 2023 06.
Article in English | MEDLINE | ID: mdl-37277530

ABSTRACT

The Hippo pathway is a key growth control pathway that is conserved across species. The downstream effectors of the Hippo pathway, YAP (Yes-associated protein) and TAZ (transcriptional coactivator with PDZ-binding motif), are frequently activated in cancers to drive proliferation and survival. Based on the premise that sustained interactions between YAP/TAZ and TEADs (transcriptional enhanced associate domain) are central to their transcriptional activities, we discovered a potent small-molecule inhibitor (SMI), GNE-7883, that allosterically blocks the interactions between YAP/TAZ and all human TEAD paralogs through binding to the TEAD lipid pocket. GNE-7883 effectively reduces chromatin accessibility specifically at TEAD motifs, suppresses cell proliferation in a variety of cell line models and achieves strong antitumor efficacy in vivo. Furthermore, we uncovered that GNE-7883 effectively overcomes both intrinsic and acquired resistance to KRAS (Kirsten rat sarcoma viral oncogene homolog) G12C inhibitors in diverse preclinical models through the inhibition of YAP/TAZ activation. Taken together, this work demonstrates the activities of TEAD SMIs in YAP/TAZ-dependent cancers and highlights their potential broad applications in precision oncology and therapy resistance.


Subject(s)
Neoplasms , Proto-Oncogene Proteins p21(ras) , Humans , Proto-Oncogene Proteins p21(ras)/genetics , Precision Medicine , Transcription Factors/metabolism , Signal Transduction
8.
Nat Commun ; 13(1): 6782, 2022 11 09.
Article in English | MEDLINE | ID: mdl-36351945

ABSTRACT

Germ-line hypomorphism of the pleiotropic transcription factor Myc in mice, either through Myc gene haploinsufficiency or deletion of Myc enhancers, delays onset of various cancers while mice remain viable and exhibit only relatively mild pathologies. Using a genetically engineered mouse model in which Myc expression may be systemically and reversibly hypomorphed at will, we asked whether this resistance to tumour progression is also emplaced when Myc hypomorphism is acutely imposed in adult mice. Indeed, adult Myc hypomorphism profoundly blocked KRasG12D-driven lung and pancreatic cancers, arresting their evolution at the early transition from indolent pre-tumour to invasive cancer. We show that such arrest is due to the incapacity of hypomorphic levels of Myc to drive release of signals that instruct the microenvironmental remodelling necessary to support invasive cancer. The cancer protection afforded by long-term adult imposition of Myc hypomorphism is accompanied by only mild collateral side effects, principally in haematopoiesis, but even these are circumvented if Myc hypomorphism is imposed metronomically whereas potent cancer protection is retained.


Subject(s)
Genes, ras , Pancreatic Neoplasms , Mice , Animals , Transcription Factors/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Cell Line, Tumor
9.
Carcinogenesis ; 32(6): 829-35, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21362628

ABSTRACT

In human pancreatic cancers, promoter CpG island hypermethylation is observed in both benign and malignant tumors. It is thought that silencing of key growth-controlling genes by promoter hypermethylation may play a role in pancreatic oncogenesis. We have shown previously that sufficient levels of DNA methyltransferase (Dnmt) 1 expression are required for the development of murine intestinal tumors. Here, we report the results of a large-scale triple cross (progeny n = 761) between Apc(Min/+), Trp53(-/-) and Dnmt1 hypomorphic mice to investigate the role of Dnmt levels in the Apc(Min/+), Trp53(-/-) mouse models of acinar cell pancreatic cancer. Mutations of both APC and TP53 are observed in human pancreatic cancer. We found that tumor burden, but not tumor size, is significantly reduced with decreasing Dnmt1 levels, suggesting that DNA methylation is involved in pancreatic tumorigenesis in this mouse model. Detailed analyses showed that the reduction in tumor burden is the result of a decrease in both early- and late-stage lesions. We observed decreased levels of DNA methylation at candidate genes in the normal pancreas of Dnmt1 hypomorphic mice. Some of these genes showed increased methylation associated with tumorigenesis, suggesting that the tumor-suppressive effects of Dnmt1 hypomorphic alleles may be mediated in part through reduced promoter hypermethylation. Our work is the first in vivo study to show the effects of reduced Dnmt levels on pancreatic tumor development.


Subject(s)
Carcinoma, Acinar Cell/prevention & control , DNA (Cytosine-5-)-Methyltransferases/physiology , DNA Methylation , Genes, APC/physiology , Pancreatic Neoplasms/prevention & control , Tumor Suppressor Protein p53/physiology , Animals , Carcinoma, Acinar Cell/genetics , Carcinoma, Acinar Cell/pathology , CpG Islands , DNA (Cytosine-5-)-Methyltransferase 1 , DNA, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Silencing , Immunoenzyme Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Polymerase Chain Reaction , Promoter Regions, Genetic
10.
Cancer Discov ; 10(4): 588-607, 2020 04.
Article in English | MEDLINE | ID: mdl-31941709

ABSTRACT

The signature features of pancreatic ductal adenocarcinoma (PDAC) are its fibroinflammatory stroma, poor immune activity, and dismal prognosis. We show that acute activation of Myc in indolent pancreatic intraepithelial neoplasm (PanIN) epithelial cells in vivo is, alone, sufficient to trigger immediate release of instructive signals that together coordinate changes in multiple stromal and immune-cell types and drive transition to pancreatic adenocarcinomas that share all the characteristic stromal features of their spontaneous human counterpart. We also demonstrate that this Myc-driven PDAC switch is completely and immediately reversible: Myc deactivation/inhibition triggers meticulous disassembly of advanced PDAC tumor and stroma and concomitant death of tumor cells. Hence, both the formation and deconstruction of the complex PDAC phenotype are continuously dependent on a single, reversible Myc switch. SIGNIFICANCE: We show that Myc activation in indolent Kras G12D-induced PanIN epithelium acts as an immediate pleiotropic switch, triggering tissue-specific signals that instruct all the diverse signature stromal features of spontaneous human PDAC. Subsequent Myc deactivation or inhibition immediately triggers a program that coordinately disassembles PDAC back to PanIN.See related commentary by English and Sears, p. 495.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins c-myc/genetics , Animals , Carcinoma, Pancreatic Ductal/pathology , Genes, myc , Humans , Mice , Pancreatic Neoplasms/pathology , Phenotype , Prognosis , Proto-Oncogene Proteins p21(ras)/genetics
11.
Clin Cancer Res ; 23(7): 1647-1655, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28373363

ABSTRACT

The "hallmarks" of pancreatic ductal adenocarcinoma (PDAC) include proliferative, invasive, and metastatic tumor cells and an associated dense desmoplasia comprised of fibroblasts, pancreatic stellate cells, extracellular matrix, and immune cells. The oncogenically activated pancreatic epithelium and its associated stroma are obligatorily interdependent, with the resulting inflammatory and immunosuppressive microenvironment contributing greatly to the evolution and maintenance of PDAC. The peculiar pancreas-specific tumor phenotype is a consequence of oncogenes hacking the resident pancreas regenerative program, a tissue-specific repair mechanism regulated by discrete super enhancer networks. Defined as genomic regions containing clusters of multiple enhancers, super enhancers play pivotal roles in cell/tissue specification, identity, and maintenance. Hence, interfering with such super enhancer-driven repair networks should exert a disproportionately disruptive effect on tumor versus normal pancreatic tissue. Novel drugs that directly or indirectly inhibit processes regulating epigenetic status and integrity, including those driven by histone deacetylases, histone methyltransferase and hydroxylases, DNA methyltransferases, various metabolic enzymes, and bromodomain and extraterminal motif proteins, have shown the feasibility of disrupting super enhancer-dependent transcription in treating multiple tumor types, including PDAC. The idea that pancreatic adenocarcinomas rely on embedded super enhancer transcriptional mechanisms suggests a vulnerability that can be potentially targeted as novel therapies for this intractable disease. Clin Cancer Res; 23(7); 1647-55. ©2017 AACRSee all articles in this CCR Focus section, "Pancreatic Cancer: Challenge and Inspiration."


Subject(s)
Adenocarcinoma/genetics , Carcinoma, Pancreatic Ductal/genetics , Enhancer Elements, Genetic , Tumor Microenvironment/genetics , Adenocarcinoma/pathology , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Humans , Molecular Targeted Therapy , Pancreas/pathology , Pancreatic Stellate Cells/pathology
12.
Cancer Res ; 75(8): 1675-81, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25878147

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is characterized by a dense stromal fibroinflammatory reaction that is a major obstacle to effective therapy. The desmoplastic stroma comprises many inflammatory cells, in particular mast cells as key components of the PDAC microenvironment, and such infiltration correlates with poor patient outcome. Indeed, it has been hypothesized that stromal ablation is critical to improve clinical response in patients with PDAC. Ibrutinib is a clinically approved Bruton's tyrosine kinase inhibitor that inhibits mast cells and tumor progression in a mouse model of ß-cell tumorigenesis. Here, we show that ibrutinib is highly effective at limiting the growth of PDAC in both transgenic mouse and patient-derived xenograft models of the disease. In these various experimental settings, ibrutinib effectively diminished fibrosis, extended survival, and improved the response to clinical standard-of-care therapy. Our results offer a preclinical rationale to immediately evaluate the clinical efficacy of ibrutinib in patients with PDAC.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/therapeutic use , Pancreatic Neoplasms/drug therapy , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Adenine/analogs & derivatives , Adenocarcinoma/pathology , Animals , Antineoplastic Agents/pharmacology , Female , Fibrosis/prevention & control , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Pancreatic Neoplasms/pathology , Piperidines , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Tumor Cells, Cultured , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
13.
Nat Commun ; 5: 4632, 2014 Aug 18.
Article in English | MEDLINE | ID: mdl-25130259

ABSTRACT

Gliomas are the most common primary tumours affecting the adult central nervous system and respond poorly to standard therapy. Myc is causally implicated in most human tumours and the majority of glioblastomas have elevated Myc levels. Using the Myc dominant negative Omomyc, we previously showed that Myc inhibition is a promising strategy for cancer therapy. Here, we preclinically validate Myc inhibition as a therapeutic strategy in mouse and human glioma, using a mouse model of spontaneous multifocal invasive astrocytoma and its derived neuroprogenitors, human glioblastoma cell lines, and patient-derived tumours both in vitro and in orthotopic xenografts. Across all these experimental models we find that Myc inhibition reduces proliferation, increases apoptosis and remarkably, elicits the formation of multinucleated cells that then arrest or die by mitotic catastrophe, revealing a new role for Myc in the proficient division of glioma cells.


Subject(s)
Astrocytoma/pathology , Brain Neoplasms/pathology , Glioblastoma/pathology , Glioma/pathology , Mitosis/physiology , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Animals , Apoptosis/physiology , Astrocytoma/physiopathology , Astrocytoma/therapy , Brain Neoplasms/physiopathology , Brain Neoplasms/therapy , Cell Line, Tumor , Cell Proliferation/physiology , Disease Models, Animal , Glioblastoma/physiopathology , Glioblastoma/therapy , Glioma/physiopathology , Glioma/therapy , Heterografts , Humans , Mice , Mice, Transgenic , Protein Tyrosine Phosphatase, Non-Receptor Type 1/physiology , Proto-Oncogene Proteins c-myc/physiology , Ubiquitin-Activating Enzymes/physiology
14.
Methods Mol Biol ; 1012: 1-6, 2013.
Article in English | MEDLINE | ID: mdl-24006054

ABSTRACT

Myc is a transcriptional coordinator of a wide range of intracellular and extracellular processes required for cell proliferation. These processes are tightly regulated in physiological conditions but hijacked when Myc is oncogenically activated. In fact, aberrantly elevated and/or deregulated activity of Myc is associated with the majority of human cancers. Several switchable mouse transgenic models have been developed and provided insights on the role of Myc in maintaining multiple aspects of the tumor phenotype, indicating that Myc inhibition would constitute an effective and broadly applicable anticancer therapeutic strategy. This issue of "The Myc gene: Methods and Protocols" provides a rich collection of techniques developed or routinely used by Myc investigators and serves as an invaluable resource for exploring the pleiotropic and still puzzling Myc biological functions.


Subject(s)
Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Animals , Humans
15.
Elife ; 2: e00822, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24137534

ABSTRACT

mir-17-92, a potent polycistronic oncomir, encodes six mature miRNAs with complex modes of interactions. In the Eµ-myc Burkitt's lymphoma model, mir-17-92 exhibits potent oncogenic activity by repressing c-Myc-induced apoptosis, primarily through its miR-19 components. Surprisingly, mir-17-92 also encodes the miR-92 component that negatively regulates its oncogenic cooperation with c-Myc. This miR-92 effect is, at least in part, mediated by its direct repression of Fbw7, which promotes the proteosomal degradation of c-Myc. Thus, overexpressing miR-92 leads to aberrant c-Myc increase, imposing a strong coupling between excessive proliferation and p53-dependent apoptosis. Interestingly, miR-92 antagonizes the oncogenic miR-19 miRNAs; and such functional interaction coordinates proliferation and apoptosis during c-Myc-induced oncogenesis. This miR-19:miR-92 antagonism is disrupted in B-lymphoma cells that favor a greater increase of miR-19 over miR-92. Altogether, we suggest a new paradigm whereby the unique gene structure of a polycistronic oncomir confers an intricate balance between oncogene and tumor suppressor crosstalk. DOI:http://dx.doi.org/10.7554/eLife.00822.001.


Subject(s)
Apoptosis/physiology , MicroRNAs/physiology , Oncogenes , Animals , Apoptosis/genetics , Cells, Cultured , Mice
16.
Oncotarget ; 2(12): 1307-13, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22202195

ABSTRACT

The biological programs of vertebrates exhibit a remarkable degree of functional degeneracy, adaptive compensation and robustness, to preserve homeostasis and generate reproducible phenotypic outputs irrespective of variations in signal strength, noise and quality. Cancers are difficult to treat not only because they are so mechanistically diverse but also because they adapt or evolve in response to any pharmacological elective pressure we impose upon them. Hence, an ideal cancer drug target would exert a function both necessary for cancer cell survival and functionally non-redundant, rendering it impossible for tumor cells to compensate for, or evolve independence from, the inhibitory effect of any drug aimed at that target. In this review, we discuss the unique, non-degenerate and highly pleiotropic role played by Myc in coordinating, engaging and maintaining the diverse intracellular and extracellular programs required for cell proliferation in vivo. These properties make Myc a compelling candidate cancer drug target, at least in principle: an assertion recently reinforced by new in vivo genetic data.


Subject(s)
Neoplasms/drug therapy , Neoplasms/pathology , Proto-Oncogene Proteins c-myc/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Mice , Neoplasms/genetics , Proto-Oncogene Proteins c-myc/genetics , Signal Transduction , Tumor Microenvironment
17.
J Biol ; 8(8): 77, 2009 Sep 28.
Article in English | MEDLINE | ID: mdl-19804609

ABSTRACT

Data recently published in BMC Biology provide insights into the normal physiological function of c-myc in the development and regeneration of the mammary gland and indicate a key role in epithelial cell proliferation, elaboration of ductal alveoli, and the biosynthetic capacity and milk production of the mature organ.


Subject(s)
Mammary Glands, Human/physiology , Proto-Oncogene Proteins c-myc/physiology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Proliferation , Epithelial Cells/cytology , Female , Humans , Lactation , Mammary Glands, Human/embryology , Neoplasms/drug therapy , Neoplasms/genetics , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Regeneration
18.
Gastroenterology ; 133(1): 207-18, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17631143

ABSTRACT

BACKGROUND & AIMS: Two genes (MAT1A and MAT2A) encode for methionine adenosyltransferase, an essential enzyme responsible for S-adenosylmethionine (SAMe) biosynthesis. MAT1A is expressed in liver, whereas MAT2A is widely distributed. In liver, increased MAT2A expression is associated with growth, while SAMe inhibits MAT2A expression and growth. The role of MAT2A in colon cancer in unknown. The aims of this study were to examine whether MAT2A expression and SAMe and its metabolite methylthioadenosine (MTA) can modulate growth of colon cancer cells. METHODS: Studies were conducted using resected colon cancer specimens, polyps from Min mice, and human colon cancer cell lines RKO and HT-29. MAT2A expression was measured by real-time polymerase chain reaction and cell growth by the 3-(4,5-dimethylthiazolyl-2-yl)-2,5-diphenyltetrazolium bromide assay. RESULTS: In 12 of 13 patients and all 9 polyps from Min mice, the MAT2A messenger RNA levels were 200%-340% of levels in adjacent normal tissues, respectively. Epidermal growth factor, insulin-like growth factor 1, and leptin increased growth and up-regulated MAT2A expression and MAT2A promoter activity in RKO and HT-29 cells. SAMe and MTA lowered the baseline expression of MAT2A and blocked the growth factor-mediated increase in MAT2A expression and growth in colon cancer cell lines. Importantly, the mitogenic effect of these growth factors was inhibited if MAT2A induction was prevented by RNA interference. SAMe and MTA supplementation in drinking water increased intestinal SAMe levels and lowered MAT2A expression. CONCLUSIONS: Similar to the liver, up-regulation of MAT2A also provides a growth advantage and SAMe and MTA can block mitogenic signaling in colon cancer cells.


Subject(s)
Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Methionine Adenosyltransferase/genetics , Methionine Adenosyltransferase/metabolism , S-Adenosylmethionine/metabolism , Adenosine/analogs & derivatives , Aged , Animals , Cell Death , Cell Division , Drug Interactions , Epidermal Growth Factor/pharmacology , Female , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , HT29 Cells , Humans , Insulin-Like Growth Factor I/pharmacology , Intestinal Polyps/genetics , Intestinal Polyps/metabolism , Leptin/pharmacology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Mitogens/pharmacology , Polyamines/metabolism , Promoter Regions, Genetic/physiology , S-Adenosylmethionine/pharmacology
19.
Cancer Res ; 66(17): 8430-8, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16951153

ABSTRACT

Colorectal cancer, one of the most common human malignancies in the Western world, is often subdivided based on tumor location in either the distal or proximal colon. Several mouse models have been developed to study human colorectal cancer, but few display this clear distinction between the two colonic locations. By crossing Apc(Min/+) and Smad3 mutant mice, we showed that combined activation of the Wnt pathway and attenuation of the transforming growth factor-beta (TGF-beta) pathway causes high multiplicity and rapid onset of invasive tumorigenesis almost exclusively in the distal colon, closely mimicking the familial adenomatous polyposis (FAP) disease and consisting with distinct colorectal cancer etiologies based on tumor location. Transcriptional profiling revealed higher expression of several TGF-beta activators in the normal distal mucosa than in proximal mucosa, suggesting a stronger reliance on TGF-beta-mediated growth control in the distal than in the proximal colon. Apc(Min/+)Smad3(-/-) mice provide an alternative model to Apc(Min/+) mice to study FAP and distal sporadic colorectal cancer. This model will be useful in dissecting mechanistic and etiologic differences between proximal and distal colonic cancer, whereas the confinement of tumorigenesis to the distal colon offers unique advantages in monitoring tumor progression by in vivo imaging.


Subject(s)
Colonic Neoplasms/genetics , Colorectal Neoplasms/genetics , Genes, APC , Smad3 Protein/deficiency , Adenomatous Polyposis Coli/genetics , Animals , Biopsy , Colonic Neoplasms/pathology , Colorectal Neoplasms/pathology , DNA/genetics , DNA/isolation & purification , Gene Expression Profiling , Humans , Intestinal Mucosa/pathology , Mice , Mice, Knockout , Mice, Transgenic , Neoplasm Invasiveness , Polymerase Chain Reaction , Rectal Neoplasms/genetics , Rectal Neoplasms/pathology , Smad3 Protein/genetics , Transcription, Genetic , Transforming Growth Factor beta/physiology
SELECTION OF CITATIONS
SEARCH DETAIL