Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Leukemia ; 31(7): 1658, 2017 07.
Article in English | MEDLINE | ID: mdl-28322226

ABSTRACT

This corrects the article DOI: 10.1038/leu.2014.119.

4.
Leukemia ; 29(12): 2382-9, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26172401

ABSTRACT

In acute myeloid leukemia (AML), about 25-30% of patients harbor a constitutively active receptor tyrosine kinase (RTK) FLT3 encoded by a FLT3 allele harboring internal tandem duplication (FLT3-ITD) mutation. The presence of FLT3-ITD correlates with poor prognosis in AML and it makes FLT3 an attractive therapeutic target in AML. Unfortunately, to date small-molecule inhibitors of FLT3 have resulted in only partial and transient clinical responses with residual leukemic blasts resistant to FLT3 inhibitors detected in blood or bone marrow. In this study, we investigated whether the RTK Axl is responsible for resistance of FLT3-ITD(+) AML cells to PKC412 and AC220, FLT3 inhibitors currently under clinical trials for FLT3-ITD(+) AML patients. Upon treatment with PKC412 or AC220, phosphorylation of Axl was significantly enhanced in the FLT3-ITD(+) MV4-11 AML cell line and in primary blasts from a FLT3-ITD(+) AML patient. Consistently, a PKC412-resistant AML cell line and PKC412-resistant primary blasts from FLT3-ITD(+) AML patients had significantly higher levels of constitutively phosphorylated Axl and total Axl when compared with a PKC412-sensitive AML cell line and PKC412-sensitive primary blasts from FLT3-ITD(+) AML patients. We also found that resistance of AML cells against the FLT3 inhibitor PKC412 and AC220 was substantially diminished by the inhibition of Axl via a small-molecule inhibitor TP-0903, a soluble receptor Axl fusion protein Axl-Fc or knockdown of Axl gene expression by shRNA. Collectively, our study suggests that Axl is required for resistance of FLT3-ITD(+) AML cells against the FLT3 inhibitor PKC412 and AC220, and that inhibition of Axl activation may overcome resistance to FLT3-targeted therapy in FLT3-ITD(+) AML.


Subject(s)
Leukemia, Myeloid, Acute/drug therapy , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Phosphorylation , Axl Receptor Tyrosine Kinase
5.
Placenta ; 35(11): 883-90, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25260566

ABSTRACT

INTRODUCTION: Preeclampsia (PE) and intrauterine growth restriction (IUGR) are two diseases that affect pregnant women and their unborn children. These diseases cause low birth weight, pre-term delivery, and neurological and cardiovascular disorders in babies. Combined they account for 20% of preterm deliveries. Pyruvate kinase M2 (PKM2) is a metabolism enzyme found in developing embryonic and cancer tissues. Our objective is to determine the expression of PKM2 in human PE and IUGR compared to normal pregnancies. Understanding expression of PKM2 in PE and IUGR could help us to better understand the mechanisms and find treatments for PE and IUGR. METHODS: Human placental tissues were obtained for PKM2 determination and analyzed by immunohistochemistry, Western blot, and a pyruvate assay. Placental samples were homogenized and cytoplasmic and nuclear proteins were extracted for Western blot analysis. RESULTS: Preeclampsia samples had elevated levels of p-PKM2, p-ERK, and ERK in the cytoplasm. Beta-catenin and lactose dehydrogenase (LDH) were also elevated in preeclampsia placenta samples. DISCUSSION AND CONCLUSION: We conclude that PKM2 is expressed in normal, PE and IUGR pregnancies. Also, that this expression is increased in the PE placenta at delivery. These results suggest placental metabolism through PKM2 could play a role in human preeclampsia.


Subject(s)
Carrier Proteins/metabolism , Fetal Growth Retardation/enzymology , Membrane Proteins/metabolism , Placenta/enzymology , Pre-Eclampsia/enzymology , Thyroid Hormones/metabolism , Adult , Case-Control Studies , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Lactate Dehydrogenases/metabolism , Phosphorylation , Pregnancy , Pyruvate Kinase/metabolism , beta Catenin/metabolism , Thyroid Hormone-Binding Proteins
7.
Leukemia ; 28(11): 2155-64, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24699304

ABSTRACT

The histone demethylase LSD1 (KDM1A) demethylates mono- and di-methylated (Me2) lysine (K) 4 on histone H3. High LSD1 expression blocks differentiation and confers a poor prognosis in acute myeloid leukemia (AML). Here, treatment with the novel LSD1 antagonist SP2509 attenuated the binding of LSD1 with the corepressor CoREST, increased the permissive H3K4Me3 mark on the target gene promoters, and increased the levels of p21, p27 and CCAAT/enhancer binding protein α in cultured AML cells. In addition, SP2509 treatment or LSD1 shRNA inhibited the colony growth of AML cells. SP2509 also induced morphological features of differentiation in the cultured and primary AML blasts. SP2509 induced more apoptosis of AML cells expressing mutant NPM1 than mixed-lineage leukemia fusion oncoproteins. Treatment with SP2509 alone significantly improved the survival of immune-depleted mice following tail-vein infusion and engraftment of cultured or primary human AML cells. Co-treatment with pan-HDAC inhibitor (HDI) panobinostat (PS) and SP2509 was synergistically lethal against cultured and primary AML blasts. Compared with each agent alone, co-treatment with SP2509 and PS significantly improved the survival of the mice engrafted with the human AML cells, without exhibiting any toxicity. Collectively, these findings show that the combination of LSD1 antagonist and pan-HDI is a promising therapy warranting further testing against AML.


Subject(s)
Enzyme Inhibitors/pharmacology , Histone Acetyltransferases/antagonists & inhibitors , Histone Demethylases/antagonists & inhibitors , Hydrazines/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Sulfonamides/pharmacology , Animals , Apoptosis/drug effects , Cells, Cultured , Co-Repressor Proteins/metabolism , Disease Models, Animal , Female , Histone Acetyltransferases/metabolism , Histone Demethylases/genetics , Histone Demethylases/metabolism , Histones/metabolism , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mice, Inbred NOD , Mice, SCID , Nerve Tissue Proteins/metabolism , Nucleophosmin , RNA, Small Interfering/genetics , Stem Cells/cytology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
Oncogene ; 32(42): 5089-100, 2013 Oct 17.
Article in English | MEDLINE | ID: mdl-23178492

ABSTRACT

Ewing sarcoma provides an important model for transcription-factor-mediated oncogenic transformation because of its reliance on the ETS-type fusion oncoprotein EWS/FLI. EWS/FLI functions as a transcriptional activator and transcriptional activation is required for its oncogenic activity. Here, we demonstrate that a previously less-well characterized transcriptional repressive function of the EWS/FLI fusion is also required for the transformed phenotype of Ewing sarcoma. Through comparison of EWS/FLI transcriptional profiling and genome-wide localization data, we define the complement of EWS/FLI direct downregulated target genes. We demonstrate that LOX is a previously undescribed EWS/FLI-repressed target that inhibits the transformed phenotype of Ewing sarcoma cells. Mechanistic studies demonstrate that the NuRD co-repressor complex interacts with EWS/FLI, and that its associated histone deacetylase and LSD1 activities contribute to the repressive function. Taken together, these data reveal a previously unknown molecular function for EWS/FLI, demonstrate a more highly coordinated oncogenic transcriptional hierarchy mediated by EWS/FLI than previously suspected, and implicate a new paradigm for therapeutic intervention aimed at controlling NuRD activity in Ewing sarcoma tumors.


Subject(s)
Bone Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Protein c-fli-1/genetics , RNA-Binding Protein EWS/genetics , Sarcoma, Ewing/genetics , Animals , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Co-Repressor Proteins/genetics , Co-Repressor Proteins/metabolism , Genes, Tumor Suppressor , Histone Deacetylases/metabolism , Histone Demethylases/genetics , Histone Demethylases/metabolism , Humans , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Mice, Nude , Oncogene Proteins, Fusion/metabolism , Protein Structure, Tertiary , Protein-Lysine 6-Oxidase/genetics , Protein-Lysine 6-Oxidase/metabolism , Proto-Oncogene Protein c-fli-1/metabolism , RNA-Binding Protein EWS/metabolism , Sarcoma, Ewing/pathology , Transcription, Genetic , Xenograft Model Antitumor Assays
10.
Br J Cancer ; 105(10): 1563-73, 2011 Nov 08.
Article in English | MEDLINE | ID: mdl-22015557

ABSTRACT

BACKGROUND: Upregulation of PIM kinase expression has been reported in many malignancies, suggesting that inhibition of PIM kinase activity may be an attractive therapeutic strategy. We hypothesised that inhibition of PIM kinase activity with SGI-1776, a novel small molecule inhibitor of PIM kinase activity, would reduce the viability of renal cell carcinoma (RCC) cells and enhance the activity of sunitinib. METHODS: Immunoblotting, qRT-PCR, and gene expression arrays were carried out to identify genes modulated by SGI-1776 treatment. The anticancer activity of SGI-1776 and sunitinib was determined by viability and apoptosis assays and in tumour xenografts in vivo. RESULTS: Treatment with SGI-1776 led to a decrease in phosphorylated and total c-Myc levels, which resulted in the modulation of c-Myc target genes. SGI-1776 in combination with sunitinib induced a further reduction in c-Myc levels, which was associated with enhanced anticancer activity. siRNA-mediated knockdown of c-Myc demonstrated that its expression has a key role in regulating the sensitivity to the combination of SGI-1776 and sunitinib. Importantly, the combination significantly reduced tumour burden in two RCC xenograft models compared with single-agent therapy and was very well tolerated. CONCLUSION: These data indicate that targeting PIM kinase signalling is a promising treatment strategy for RCC.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell/pathology , Imidazoles/pharmacology , Indoles/pharmacology , Kidney Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Pyridazines/pharmacology , Pyrroles/pharmacology , Animals , Carcinoma, Renal Cell/enzymology , Cell Line, Tumor , Female , Humans , Immunohistochemistry , Kidney Neoplasms/enzymology , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphorylation , Proto-Oncogene Proteins c-myc/metabolism , Real-Time Polymerase Chain Reaction , Sunitinib
11.
Oncogene ; 26(27): 3909-19, 2007 Jun 07.
Article in English | MEDLINE | ID: mdl-17325667

ABSTRACT

KIT or alpha-platelet-derived growth factor receptor (alpha-PDGFR) activating mutations are the pathogenic mechanisms that characterize gastrointestinal stromal tumors (GIST). Despite excellent responses to imatinib mesylate (IM), patients are relapsing. We developed an IM-resistant GIST cell line (GIST-R) from the IM-sensitive GIST882 cell line (GIST-S) by growing these cells in IM. Gene expression profiling (GEP) of GIST-S, GIST-R cells and two IM resistant GIST patients demonstrated that KIT is downregulated implying a major role in IM resistance. Instead, GIST-R cells have acquired IM resistance by overexpressing the oncogenic receptor tyrosine kinase - AXL - in a 'kinase switch'. Further, the two IM resistant GIST patients express AXL and not c-Kit, seen by immunohistochemistry (IHC). Real time reverse transcriptase-polymerase chain reaction and Western blotting of the GIST-S and GIST-R cells confirmed the switch from Kit to AXL. In GIST-R, AXL is tyrosine phosphorylated and its ligand growth-arrest-specific gene 6 is overexpressed implying autocrine activation. The kinase switch is associated with a morphological change from spindle to epithelioid. Molecular modeling of the kinase domain of mutant c-Kit (V654A) and AXL showed no binding to IM but efficient binding to MP470, a novel c-Kit/AXL kinase inhibitor. MP470 synergizes with docetaxel (taxotere) and is cytotoxic to GIST cells.


Subject(s)
Drug Resistance, Neoplasm/genetics , Gastrointestinal Stromal Tumors/genetics , Oncogene Proteins/genetics , Piperazines/pharmacology , Proto-Oncogene Proteins c-kit/genetics , Pyrimidines/pharmacology , Receptor Protein-Tyrosine Kinases/genetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Benzamides , Blotting, Western , Cell Line, Tumor , Cell Shape/drug effects , Cell Survival/drug effects , Docetaxel , Drug Synergism , Gastrointestinal Stromal Tumors/drug therapy , Gastrointestinal Stromal Tumors/pathology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Imatinib Mesylate , Models, Molecular , Mutation , Oligonucleotide Array Sequence Analysis , Oncogene Proteins/chemistry , Oncogene Proteins/metabolism , Phosphorylation/drug effects , Piperazines/therapeutic use , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Structure, Tertiary , Proto-Oncogene Proteins , Proto-Oncogene Proteins c-kit/chemistry , Proto-Oncogene Proteins c-kit/metabolism , Pyrimidines/chemistry , Pyrimidines/therapeutic use , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Protein-Tyrosine Kinases/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Taxoids/pharmacology , Thiourea , Axl Receptor Tyrosine Kinase
12.
Chem Biol ; 8(11): 1033-49, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11731295

ABSTRACT

BACKGROUND: Ecteinascidin 743 (Et 743), a natural product derived from a marine tunicate, is a potent antitumor agent presently in phase II clinical trials. Et 743 binds in the minor groove of DNA and alkylates N2 of guanine via a unique mechanism involving catalytic activation. The sequence selectivity of Et 743 is governed by different patterns of hydrogen-bonding to DNA, which results in differential reversibility of the covalent adducts. As determined by nuclear magnetic resonance spectroscopy, the preferred sequences 5'-PuGC and 5'-PyGG are stabilized by a hydrogen-bonding network, while the non-preferred sequences 5'-NG(A/T) are much less stabilized due to the lack of a key hydrogen bond to the GC base pair on the 3'-side of the alkylated guanine. RESULTS: Mammalian cell lines (XPB, XPD, XPF, XPG, and ERCC1) deficient in the nucleotide excision repair (NER) gene products show resistance to Et 743. The recognition and subsequent incision of Et 743-DNA adducts by the bacterial multisubunit endonuclease UvrABC were used to evaluate DNA repair-mediated toxicity as a rationale for the resistance of NER-defective cell lines and the antitumor activity of Et 743. The Et 743-DNA adducts are indeed recognized and incised by the UvrABC repair proteins; however, the pattern of incision indicated that the non-preferred, and less stable, sequences (i.e. 5'-NG(A/T)) modified with Et 743 are generally incised at a much higher efficiency than the preferred, more stable sequences (i.e. 5'-PuGC or 5'-PyGG). In addition, within the same Et 743 recognition sequence, the level of incision varies, indicating that flanking regions also contribute to the differential incision frequency. CONCLUSIONS: The inefficient repair incision by the UvrABC nuclease of Et 743-DNA adducts provides a basis for rationalizing the observed repair-dependent cytotoxicities of these DNA adducts, if other associated structural properties of Et 743-DNA adducts are taken into account. In particular, the wedge-shaped Et 743, which forces open the minor groove of DNA, introducing a major groove bend, and the extrahelical protrusion of the C-subunit of Et 743 provide unique characteristics alongside the hydrogen-bonding stabilization of a covalent DNA adduct, which we propose traps an intermediate in NER processing of Et 743-DNA adducts. This trapped intermediate protein-Et 743-DNA adduct complex can be considered analogous to a poisoned topoisomerase I- or topoisomerase II-DNA complex. In the absence of an intact NER nuclease complex, this toxic lesion is unable to form, and the Et 743-DNA adducts, although not repaired by the NER pathway, are less toxic to cells. Conversely, elevated levels of either of these nucleases should lead to enhanced Et 743 toxicity.


Subject(s)
Antineoplastic Agents, Alkylating/chemistry , Antineoplastic Agents, Alkylating/metabolism , DNA/metabolism , Dioxoles/chemistry , Drug Delivery Systems , Endodeoxyribonucleases/metabolism , Escherichia coli Proteins/metabolism , Isoquinolines/chemistry , Animals , Antineoplastic Agents, Alkylating/pharmacology , Base Sequence , Binding Sites , CHO Cells , Cell Survival/drug effects , Cricetinae , DNA/chemistry , DNA/genetics , DNA Adducts/chemistry , DNA Adducts/metabolism , DNA Repair , Dioxoles/metabolism , Dioxoles/pharmacology , Gene Targeting/methods , Humans , Isoquinolines/metabolism , Isoquinolines/pharmacology , Models, Molecular , Molecular Sequence Data , Mutagenesis, Insertional/methods , Tetrahydroisoquinolines , Trabectedin
13.
Oncogene ; 19(8): 1114-22, 2000 Feb 21.
Article in English | MEDLINE | ID: mdl-10713698

ABSTRACT

Several transgenic mouse tumor models were utilized to explore how specific genetic alterations affect the tumor cell response to chemotherapeutic agents in vivo. Specifically, MMTV-ras transgenic mice were interbred to p53 knock-out mice to create a model for assessing the role of p53 in chemotherapeutic responses. In addition, MMTV-ras tumors were compared to MMTV-myc and MMTV-ras/myc tumors. Mice of each genotype reproducibly develop mammary and/or salivary tumors, but tumor growth dynamics vary considerably between genotypes. MMTV-ras/p53-/- tumors exhibit higher S phase fractions than MMTV-ras/p53+/+ tumors, although both tumor types display very low apoptosis levels. In contrast, MMTV-myc tumors exhibit both high S phase fractions and spontaneous apoptosis levels. Tumor-bearing mice of each genotype were treated with either doxorubicin or paclitaxel, and effects on overall tumor growth, cell cycle distribution and apoptosis were evaluated. Surprisingly, neither agent efficiently induced apoptosis in any of the tumor models, including those with wildtype p53. Rather, tumor responses were mediated primarily by changes in cell cycle distribution. However, the spontaneous apoptosis levels did serve as a predictor of tumor growth response, in that only those tumors with high pretreatment apoptosis levels underwent significant regression following treatment with either agent.


Subject(s)
Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/genetics , Salivary Gland Neoplasms/drug therapy , Salivary Gland Neoplasms/genetics , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Division/drug effects , Cell Division/genetics , Doxorubicin/pharmacology , Female , Genes, ras , Mammary Tumor Virus, Mouse/genetics , Mice , Mice, Inbred Strains , Mice, Transgenic , Paclitaxel/pharmacology , Tumor Suppressor Protein p53/genetics
14.
Oncogene ; 19(56): 6632-41, 2000 Dec 27.
Article in English | MEDLINE | ID: mdl-11426649

ABSTRACT

The shortening of the telomeric DNA sequences at the ends of chromosomes is thought to play a critical role in regulating the lifespan of human cells. Since all dividing cells are subject to the loss of telomeric sequences, cells with long proliferative lifespans need mechanisms to maintain telomere integrity. It appears that the activation of the enzyme telomerase is the major mechanism by which these cells maintain their telomeres. The proposal that a critical step in the process of the malignant transformation of cells is the upregulation of expression of telomerase has made this enzyme a potentially useful prognostic and diagnostic marker for cancer, as well as a new target for therapeutic intervention for the treatment of patients with cancer. It is now clear that simply inhibiting telomerase may not result in the anticancer effects that were originally hypothesized. While telomerase may not be the universal target for cancer therapy, we certainly believe that targeting the telomere maintenance mechanisms will be important in future research aimed toward a successful strategy for curing cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Design , Tankyrases , Telomerase/antagonists & inhibitors , Telomere/drug effects , Animals , Anthracenes/pharmacology , Antineoplastic Agents/therapeutic use , Drug Delivery Systems , Glycoside Hydrolases/metabolism , Humans , Neoplasms/enzymology , Oligonucleotides, Antisense/therapeutic use , Perylene/analogs & derivatives , Piperidines/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , RNA , RNA, Long Noncoding , RNA, Untranslated/antagonists & inhibitors , Reverse Transcriptase Inhibitors/pharmacology , Telomerase/physiology , Telomere/chemistry , Telomere/physiology
15.
J Biol Chem ; 273(32): 20448-55, 1998 Aug 07.
Article in English | MEDLINE | ID: mdl-9685399

ABSTRACT

Neu (c-erbB2) is a proto-oncogene product that encodes an epidermal growth factor-like receptor tyrosine kinase. Amplification of wild-type c-Neu and mutational activation of Neu (Neu T) have been implicated in oncogenic transformation of cultured fibroblasts and mammary tumorigenesis in vivo. Here, we examine the relationship between Neu tyrosine kinase activity and caveolin-1 protein expression in vitro and in vivo. Recent studies have suggested that caveolins may function as negative regulators of signal transduction. Our current results show that mutational activation of c-Neu down-regulates caveolin-1 protein expression, but not caveolin-2, in cultured NIH 3T3 and Rat 1 cells. Conversely, recombinant overexpression of caveolin-1 blocks Neu-mediated signal transduction in vivo. These results suggest a reciprocal relationship between c-Neu tyrosine kinase activity and caveolin-1 protein expression. We next analyzed a variety of caveolin-1 deletion mutants to map this caveolin-1-dependent inhibitory activity to a given region of the caveolin-1 molecule. Results from this mutational analysis show that this functional in vivo inhibitory activity is contained within caveolin-1 residues 32-95. In accordance with these in vivo studies, a 20-amino acid peptide derived from this region (the caveolin-1 scaffolding domain) was sufficient to inhibit Neu autophosphorylation in an in vitro kinase assay. To further confirm or refute the relevance of our findings in vivo, we next examined the expression levels of caveolin-1 in mammary tumors derived from c-Neu transgenic mice. Our results indicate that dramatic reduction of caveolin-1 expression occurs in mammary tumors derived from c-Neu-expressing transgenic mice and other transgenic mice expressing downstream effectors of Neu-mediated signal transduction, such as Src and Ras. Taken together, our data suggest that a novel form of reciprocal negative regulation exists between c-Neu and caveolin-1.


Subject(s)
Caveolins , Gene Expression Regulation, Neoplastic/genetics , Membrane Proteins/physiology , Receptor, ErbB-2/physiology , Animals , Caveolin 1/pharmacology , Cell Line , Ceramides/pharmacology , DNA Mutational Analysis , Down-Regulation/physiology , Immunohistochemistry , Mice , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Peptide Fragments/pharmacology , Phosphorylation , Proto-Oncogene Mas , Receptor Protein-Tyrosine Kinases/physiology , Sequence Deletion/genetics , Signal Transduction/physiology , Transformation, Genetic/genetics
16.
Mol Cell Biol ; 18(1): 85-92, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9418856

ABSTRACT

The farnesyltransferase inhibitor L-744,832 selectively blocks the transformed phenotype of cultured cells expressing a mutated H-ras gene and induces dramatic regression of mammary and salivary carcinomas in mouse mammary tumor virus (MMTV)-v-Ha-ras transgenic mice. To better understand how the farnesyltransferase inhibitors might be used in the treatment of human tumors, we have further explored the mechanisms by which L-744,832 induces tumor regression in a variety of transgenic mouse tumor models. We assessed whether L-744,832 induces apoptosis or alterations in cell cycle distribution and found that the tumor regression in MMTV-v-Ha-ras mice could be attributed entirely to elevation of apoptosis levels. In contrast, treatment with doxorubicin, which induces apoptosis in many tumor types, had a minimal effect on apoptosis in these tumors and resulted in a less dramatic tumor response. To determine whether functional p53 is required for L-744,832-induced apoptosis and the resultant tumor regression, MMTV-v-Ha-ras mice were interbred with p53(-/-) mice. Tumors in ras/p53(-/-) mice treated with L-744,832 regressed as efficiently as MMTV-v-Ha-ras tumors, although this response was found to be mediated by both the induction of apoptosis and an increase in G1 with a corresponding decrease in the S-phase fraction. MMTV-v-Ha-ras mice were also interbred with MMTV-c-myc mice to determine whether ras/myc tumors, which possess high levels of spontaneous apoptosis, have the potential to regress through a further increase in apoptosis levels. The ras/myc tumors were found to respond nearly as efficiently to L-744,832 treatment as the MMTV-v-Ha-ras tumors, although no induction of apoptosis was observed. Rather, the tumor regression in the ras/myc mice was found to be mediated by a large reduction in the S-phase fraction. In contrast, treatment of transgenic mice harboring an activated MMTV-c-neu gene did not result in tumor regression. These results demonstrate that a farnesyltransferase inhibitor can induce regression of v-Ha-ras-bearing tumors by multiple mechanisms, including the activation of a suppressed apoptotic pathway, which is largely p53 independent, or by cell cycle alterations, depending upon the presence of various other oncogenic genetic alterations.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma/genetics , Enzyme Inhibitors/pharmacology , Mammary Neoplasms, Experimental/genetics , Methionine/analogs & derivatives , Salivary Gland Neoplasms/genetics , Animals , Antineoplastic Agents/therapeutic use , Carcinoma/drug therapy , Carcinoma/pathology , Cell Cycle/drug effects , Enzyme Inhibitors/therapeutic use , Farnesyltranstransferase , Female , Genes, ras , Humans , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Mammary Tumor Virus, Mouse , Methionine/pharmacology , Methionine/therapeutic use , Mice , Mice, Transgenic , Salivary Gland Neoplasms/drug therapy , Salivary Gland Neoplasms/pathology
17.
Proc Natl Acad Sci U S A ; 93(23): 12845-50, 1996 Nov 12.
Article in English | MEDLINE | ID: mdl-8917507

ABSTRACT

YY1 is a mammalian zinc-finger transcription factor with unusual structural and functional features. It has been implicated as a positive and a negative regulatory factor that binds to the CCATNTT consensus DNA element located in promoters of many cellular and viral genes. A mammalian cDNA that encodes a YY1-binding protein and possesses sequence homology with the yeast transcriptional factor RPD3 has been identified. A Gal4 DNA binding domain-mammalian RPD3 fusion protein strongly represses transcription from a promoter containing Gal4 binding sites. Association between YY1 and mammalian RPD3 requires a glycine-rich region on YY1. Mutations in this region abolish the interaction with mammalian RPD3 and eliminate transcriptional repression by YY1. These data suggest that YY1 negatively regulates transcription by tethering RPD3 to DNA as a cofactor and that this transcriptional mechanism is highly conserved from yeast to human.


Subject(s)
DNA-Binding Proteins/metabolism , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism , Transcription, Genetic , Amino Acid Sequence , Animals , DNA-Binding Proteins/biosynthesis , Erythroid-Specific DNA-Binding Factors , Fungal Proteins/biosynthesis , Glutathione Transferase/biosynthesis , Histone Deacetylases , Humans , Mammals , Mice , Molecular Sequence Data , Promoter Regions, Genetic , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Transcription Factors/biosynthesis , YY1 Transcription Factor , Zinc Fingers
SELECTION OF CITATIONS
SEARCH DETAIL
...