Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Pigment Cell Melanoma Res ; 28(3): 307-17, 2015 May.
Article in English | MEDLINE | ID: mdl-25728708

ABSTRACT

No effective targeted therapy is currently available for NRAS mutant melanoma. Experimental MEK inhibition is rather toxic and has only limited efficacy in clinical trials. At least in part, this is caused by the emergence of drug resistance, which is commonly seen for single agent treatment and shortens clinical responses. Therefore, there is a dire need to identify effective companion drug targets for NRAS mutant melanoma. Here, we show that at concentrations where single drugs had little effect, ROCK inhibitors GSK269962A or Fasudil, in combination with either MEK inhibitor GSK1120212 (Trametinib) or ERK inhibitor SCH772984 cooperatively caused proliferation inhibition and cell death in vitro. Simultaneous inhibition of MEK and ROCK caused induction of BimEL , PARP, and Puma, and hence apoptosis. In vivo, MEK and ROCK inhibition suppressed growth of established tumors. Our findings warrant clinical investigation of the effectiveness of combinatorial targeting of MAPK/ERK and ROCK in NRAS mutant melanoma.


Subject(s)
Apoptosis/drug effects , GTP Phosphohydrolases/genetics , Melanoma/pathology , Membrane Proteins/genetics , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mutation/genetics , rho-Associated Kinases/antagonists & inhibitors , Cell Line, Tumor , Humans , Melanoma/enzymology , Mitogen-Activated Protein Kinase Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Proteome/metabolism , rho-Associated Kinases/metabolism
2.
EMBO J ; 30(9): 1753-65, 2011 May 04.
Article in English | MEDLINE | ID: mdl-21448135

ABSTRACT

Oncogene-induced cellular senescence (OIS) is an increasingly recognized tumour suppressor mechanism that confines the outgrowth of neoplastic cells in vivo. It relies on a complex signalling network, but only few components have been identified so far. Gene-expression profiling revealed a >100-fold increase in the levels of the transcription factor and putative tumour suppressor gene TGFß-stimulated clone 22 (TSC22D1) in BRAF(E600)-induced senescence, in both human fibroblasts and melanocytes. Only the short TSC22D1 transcript was upregulated, whereas the abundance of the large protein variant was suppressed by proteasomal degradation. The TSC22D1 protein variants, in complex with their dimerization partner TSC22 homologue gene 1 (THG1), exerted opposing functions, as selective depletion of the short form, or conversely, overexpression of the large variant, resulted in abrogation of OIS. This was accompanied by the suppression of several inflammatory factors and p15(INK4B), with TSC22D1 acting as a critical effector of C/EBPß. Our results demonstrate that the differential regulation of antagonistic TSC22D1 variants is required for the establishment of OIS and suggest distinct contributions of TSC22 family members to the progression of BRAF(E600)-driven neoplasia.


Subject(s)
Cellular Senescence/physiology , Gene Expression Regulation, Neoplastic/physiology , Melanocytes/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Repressor Proteins/metabolism , Signal Transduction/physiology , Tumor Suppressor Proteins/metabolism , DNA Primers/genetics , Gene Expression Profiling , Humans , Immunoprecipitation , Microarray Analysis , Plasmids/genetics , RNA, Small Interfering/genetics , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction
3.
Nat Cell Biol ; 12(5): 492-9, 2010 May.
Article in English | MEDLINE | ID: mdl-20364141

ABSTRACT

Chromosomal instability (CIN) is a major hallmark of human cancer and might contribute to tumorigenesis. Genes required for the normal progression of mitosis represent potential CIN genes and, as such, are important tumour suppressors. The Chk2 kinase and its downstream targets p53 and Brca1 are tumour suppressors that have been functionally linked to the DNA damage response pathway. Here, we report a function of Chk2, independent of p53 and DNA damage, that is required for proper progression of mitosis, and for the maintenance of chromosomal stability in human somatic cells. Depletion of Chk2 or abrogation of its kinase activity causes abnormal mitotic spindle assembly associated with a delay in mitosis, which promotes the generation of lagging chromosomes, chromosome missegregation and CIN, while still allowing survival and growth. Furthermore, we have identified Brca1 as a mitotic target of the Chk2 kinase in the absence of DNA damage. Accordingly, loss of BRCA1 or its Chk2-mediated phosphorylation leads to spindle formation defects and CIN. Thus, the CHK2-BRCA1 tumour suppressor pathway is required for chromosomal stability, which might contribute to their tumour suppressor function.


Subject(s)
BRCA1 Protein/physiology , Chromosomal Instability , Protein Serine-Threonine Kinases/physiology , Tumor Suppressor Proteins/metabolism , BRCA1 Protein/metabolism , Cell Line , Checkpoint Kinase 2 , Humans , Mitosis , Phosphorylation , Protein Serine-Threonine Kinases/deficiency , Signal Transduction , Spindle Apparatus/metabolism , Spindle Apparatus/pathology
4.
Cancer Res ; 69(9): 3874-83, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19366805

ABSTRACT

The mitotic spindle checkpoint represents a signal transduction pathway that prevents the onset of anaphase until all chromosomes are properly aligned on a metaphase plate. Partial inactivation of this checkpoint allows premature separation of sister chromatids and results in aneuploidy, which might contribute to tumorigenesis. Unlike other cell cycle checkpoints, the spindle checkpoint is essential for cell viability, giving rise to the idea that the spindle checkpoint itself might represent a valuable target for anticancer therapy. We used a cell-based screen and identified the indolocarbazole compound Gö6976 as a pharmacologic inhibitor of the spindle checkpoint. Gö6976 potently overrides a spindle checkpoint-mediated mitotic arrest by abrogating the phosphorylation and kinetochore localization of several spindle checkpoint proteins. We identified the Aurora-A and Aurora-B kinases, which have been previously implicated in proper mitotic progression and spindle checkpoint function, as targets for Gö6976. Accordingly, Gö6976 treatment causes severe mitotic abnormalities and chromosome alignment defects, which are not properly detected by the drug-inactivated spindle checkpoint. This results in an aberrant progression of mitosis, leading to apoptosis in various human cancer cell lines, including spindle checkpoint-compromised cancer cells. Thus, our work describes a novel and promising strategy for anticancer treatment that targets the mitotic spindle checkpoint.


Subject(s)
Carbazoles/pharmacology , Colonic Neoplasms/drug therapy , Enzyme Inhibitors/pharmacology , Spindle Apparatus/drug effects , Apoptosis/drug effects , Aurora Kinase B , Aurora Kinases , Chromosome Aberrations , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Drug Synergism , HCT116 Cells , Humans , Mitosis/drug effects , Nocodazole/pharmacology , Paclitaxel/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism
5.
Cancer Res ; 67(1): 339-45, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-17210716

ABSTRACT

The novel concept of anticancer treatment termed "G(2) checkpoint abrogation" aims to target p53-deficient tumor cells and is currently explored in clinical trials. The anticancer drug UCN-01 is used to abrogate a DNA damage-induced G(2) cell cycle arrest leading to mitotic entry and subsequent cell death, which is poorly defined as "mitotic cell death" or "mitotic catastrophe." We show here that UCN-01 treatment results in a mitotic arrest that requires an active mitotic spindle checkpoint, involving the function of Mad2, Bub1, BubR1, Mps1, Aurora B, and survivin. During the mitotic arrest, hallmark parameters of the mitochondria-associated apoptosis pathway become activated. Interestingly, this apoptotic response requires the spindle checkpoint protein Mad2, suggesting a proapoptotic function for Mad2. However, although survivin and Aurora B are also required for the mitotic arrest, both proteins are part of an antiapoptotic pathway that restrains the UCN-01-induced apoptosis by promoting hyperphosphorylation of Bcl-2 and by inhibiting the activation of Bax. Consequently, inhibition of the antiapoptotic pathway by genetic ablation of survivin or by pharmacologic inhibitors of Aurora B or cyclin-dependent kinase 1 lead to a significant enhancement of apoptosis and therefore act synergistically with UCN-01. Thus, by defining the mechanism of cell death on G(2) checkpoint abrogation we show a highly improved strategy for an anticancer treatment by the combined use of UCN-01 with abrogators of the survivin/Aurora B-dependent antiapoptotic pathway that retains the selectivity for p53-defective cancer cells.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , G2 Phase/drug effects , Mitosis/drug effects , Staurosporine/analogs & derivatives , Apoptosis/physiology , Aurora Kinase B , Aurora Kinases , CDC2 Protein Kinase/antagonists & inhibitors , CDC2 Protein Kinase/metabolism , Calcium-Binding Proteins/physiology , Cell Cycle Proteins/physiology , Doxorubicin/administration & dosage , Drug Synergism , G2 Phase/physiology , HeLa Cells , Humans , Inhibitor of Apoptosis Proteins , Mad2 Proteins , Microtubule-Associated Proteins/physiology , Mitosis/physiology , Neoplasm Proteins/physiology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Repressor Proteins/physiology , Spindle Apparatus/drug effects , Spindle Apparatus/physiology , Staurosporine/administration & dosage , Staurosporine/pharmacology , Survivin
6.
Oncogene ; 24(26): 4301-10, 2005 Jun 16.
Article in English | MEDLINE | ID: mdl-15782113

ABSTRACT

The mitotic spindle assembly checkpoint ensures proper chromosome segregation during mitosis by inhibiting the onset of anaphase until all kinetochores are attached to the mitotic spindle and tension across the kinetochores is generated. Here, we report that the stable partial downregulation of the spindle checkpoint gene MAD1, which is observed in human cancer, leads to a functional inactivation of the spindle checkpoint resulting in gross aneuploidy. Interestingly, although Mad1 is thought to act as a kinetochore based activator of Mad2 during checkpoint activation, we show that normal levels of Mad2, but not of Mad1, are required for preventing premature sister chromatid separation and for maintaining the timing of an undisturbed mitosis, suggesting a Mad1 independent function of Mad2 that operates independent of its checkpoint function. Most significantly, a partial repression of either MAD1 or MAD2 confers resistance to nocodazole, a drug that inhibits microtubule attachment. In contrast, sensitivity to clinically relevant drugs like taxol or monastrol that inhibit the generation of tension across kinetochores is not modulated by partial downregulation of MAD1, suggesting a functional bifurcation of spindle checkpoint dependent apoptotic pathways.


Subject(s)
Aneuploidy , Carcinoma/genetics , Carcinoma/pathology , Cell Cycle Proteins/biosynthesis , Cell Cycle/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Mitosis , Nuclear Proteins/biosynthesis , Spindle Apparatus , Apoptosis , Cell Cycle Proteins/genetics , Down-Regulation , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Genomic Instability , Humans , Kinetochores , Nuclear Proteins/genetics
7.
J Biol Chem ; 280(6): 4025-8, 2005 Feb 11.
Article in English | MEDLINE | ID: mdl-15611124

ABSTRACT

A novel strategy in cancer therapy is the induction of mitotic cell death by the pharmacological abrogation of cell cycle checkpoints. UCN-01 is such a compound that overrides the G2 cell cycle arrest induced by DNA damage and forces cells into a deleterious mitosis. The molecular pathways leading to mitotic cell death are largely unknown although recent evidence indicates that mitotic cell death represents a special case of apoptosis. Here, we demonstrate that the mitotic spindle checkpoint is activated upon chemotherapeutic treatment with topoisomerase II poisons and UCN-01. Cells that are forced to enter mitosis in the presence of topoisomerase inhibition arrest transiently in a prometaphase like state. By using a novel pharmacological inhibitor of the spindle checkpoint and spindle checkpoint-deficient cells we show that the spindle checkpoint function is required for the mitotic arrest and, most importantly, for efficient induction of mitotic cell death. Thus, our results demonstrate that the mitotic spindle checkpoint is an important determinant for the outcome of a chemotherapy based on the induction of mitotic cell death. Its frequent inactivation in human cancer might contribute to the observed resistance of tumor cells to these chemotherapeutic drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Spindle Apparatus , Staurosporine/analogs & derivatives , Topoisomerase Inhibitors , Apoptosis , Blotting, Western , Cell Death , Cell Line, Tumor , Cell Separation , DNA Damage , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Flow Cytometry , G2 Phase , Humans , Mitosis , Nocodazole/pharmacology , Protein Kinase Inhibitors/pharmacology , Signal Transduction , Staurosporine/pharmacology , Time Factors , Transgenes
8.
Oncogene ; 23(41): 6845-53, 2004 Sep 09.
Article in English | MEDLINE | ID: mdl-15286707

ABSTRACT

Treatment of cells with microtubule inhibitors results in activation of the mitotic spindle assembly checkpoint, leading to mitotic arrest before anaphase. Upon prolonged treatment, however, cells can adapt and exit mitosis aberrantly, resulting in the occurrence of tetraploid cells in G1. Those cells subsequently arrest in postmitotic G1 due to the activation of a p53-dependent G1 checkpoint. Failure of the G1 checkpoint leads to endoreduplication and further polyploidization. Using HCT116 and isogenic p53-deficient or spindle checkpoint compromised derivatives, we show here that not only p53 but also a functional spindle assembly checkpoint is required for postmitotic G1 checkpoint function. During transient mitotic arrest, p53 stabilization and activation is triggered by a pathway independent of ATM/ATR, Chk1 and Chk2. We further show that a prolonged spindle checkpoint-mediated mitotic arrest is required for proper postmitotic G1 checkpoint function. In addition, we demonstrate that polyploid cells are inhibited to re-enter mitosis by an additional checkpoint acting in G2. Thus, during a normal cell cycle, polyploidization and subsequent aneuploidization is prevented by the function of the mitotic spindle checkpoint, a p53-dependent G1 checkpoint and an additional G2 checkpoint.


Subject(s)
Polyploidy , Spindle Apparatus/physiology , Tumor Suppressor Protein p53/physiology , Cells, Cultured , DNA Damage , G1 Phase , G2 Phase , Humans , Mitosis
SELECTION OF CITATIONS
SEARCH DETAIL
...