Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Transl Oncol ; 10(3): 304-310, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28325666

ABSTRACT

PURPOSE: Although targeting angiogenesis with tyrosine kinase inhibitors (TKIs) has become standard of care in the treatment of clear cell renal cell carcinoma (RCC), resistance mechanism are not fully understood, and there is a need to develop new therapeutic options overcoming them. METHODS AND MATERIALS: To develop a preclinical model that predicts clinical activity of novel agents in 19 RCC patients, we established patient-derived cell (PDC) and xenograft (PDX) models derived from malignant effusions or surgical specimen. RESULTS: Successful PDCs, defined as cells that maintained growth following two passages, were established in 5 of 15 malignant effusions and 1 of 4 surgical specimens. One PDC, clinically refractory to TKIs, was implanted and engrafted in mice, resulting in a comparable histology to the primary tumor. The PDC-PDX model also showed similar genomic features when tested using targeted sequencing of cancer-related genes. When we examined the drug effects of the PDX model, the tumor cells showed resistance to TKIs and everolimus in vitro. CONCLUSION: The results suggest that the PDC-PDX preclinical model we developed using malignant effusions can be a useful preclinical model to interrogate sensitivity to targeted agents based on genomic alterations.

2.
J Nat Prod ; 79(6): 1576-85, 2016 06 24.
Article in English | MEDLINE | ID: mdl-27300079

ABSTRACT

M-Phase Phosphoprotein 1 (MPP1), a microtubule plus end directed kinesin, is required for the completion of cytokinesis. Previous studies have shown that MPP1 is upregulated in various types of bladder cancer. This article describes inhibitor screening leading to the identification of a new class of natural product inhibitors of MPP1. Two compounds with structural similarity, norlobaridone (1) and physodic acid (2), were found to inhibit MPP1. Physodic acid is not competitive with ATP, indicating the presence of an allosteric inhibitor-binding pocket. Initial drug-like property screening indicates that physodic acid is more soluble than norlobaridone and has more favorable lipophilicity. However, both suffer from high clearance in human microsomal stability assays mediated by the lability of the lactone ring as well as hydroxylation of the alkyl chains as shown by metabolite identification studies. In cell-based assays physodic acid is a weak inhibitor with EC50 values of about 30 µM in a range of tumor cell lines. The two depsidones identified and characterized here could be used for future improvement of their activity against MPP1 and will be useful chemical probes for studying this unique molecular motor in more depth.


Subject(s)
Depsides/isolation & purification , Dibenzoxepins/isolation & purification , Kinesins/antagonists & inhibitors , Lactones/isolation & purification , Lichens/chemistry , Algorithms , Antineoplastic Combined Chemotherapy Protocols , Cytokinesis/drug effects , Depsides/chemistry , Depsides/pharmacology , Dibenzoxepins/chemistry , Dibenzoxepins/pharmacology , Humans , Kinesins/drug effects , Kinesins/metabolism , Lactones/chemistry , Lactones/pharmacology , Melphalan , Microtubules/drug effects , Microtubules/metabolism , Molecular Structure , Prednisone , Procarbazine
3.
Transl Oncol ; 9(3): 197-202, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27267837

ABSTRACT

BACKGROUND: Although pazopanib treatment has become the standard chemotherapy in salvage setting for metastatic sarcoma patients, most patients progress after pazopanib treatment in 4 to 6 months. After failure to pazopanib, patients have limited options for treatment. Therefore, subsequent therapy in patients who failed to pazopanib is urgently needed and the use of patient derived cells or patient derived tumors for accompanying testing with various pharmacological inhibitors could offer additional treatment options for these patients. METHODS: Patient derived tumor cells were collected from ascites at the time of progression to pazopanib and a 13-drug panel was tested for drug sensitivity. We confirmed the results using in vitro cell viability assay and immunoblot assay. We also performed the genomic profiling of PDX model. RESULTS: The growth of patient derived tumor cells was significantly reduced by exposure to 1.0 µM AZD2014 compared with control (control versus AZD2014, mean growth = 100.0% vs 16.04%, difference = 83.96%, 95% CI = 70.01% to 97.92%, P = .0435). Similarly, 1.0 µM BEZ235 profoundly inhibited tumor cell growth in vitro when compared to control (control versus BEZ235, mean growth = 100.0% vs 7.308%, difference = 92.69%, 95% CI = 78.87% to 106.5%, P < .0001). Despite the presence of CDK4 amplification in the patient-derived tumor cells, LEE011 did not considerably inhibit cell proliferation when compared with control (control vs LEE011, mean growth = 100.0% vs 80.23%, difference = 19.77%, 95% CI = 1.828% to 37.72%, P = .0377). The immunoblot analysis showed that BEZ235 treatment decreased pAKT, pmTOR and pERK whereas AZD2014 decreased only pmTOR. CONCLUSION: Taken together, upregulation of mTOR/AKT pathway in sarcoma patient derived cells was considerably inhibited by the treatment of AZD2014 and BEZ235 with downregulation of AKT pathway (greater extent for BEZ235). These molecules may be considered as treatment option in STS patient who have failed to pazopanib in the context of clinical trials.

4.
Oncotarget ; 6(36): 38469-86, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26540348

ABSTRACT

The actin and microtubule cytoskeletons are critically important for cancer cell proliferation, and drugs that target microtubules are widely-used cancer therapies. However, their utility is compromised by toxicities due to dose and exposure. To overcome these issues, we characterized how inhibition of the actin and microtubule cytoskeleton regulatory LIM kinases could be used in drug combinations to increase efficacy. A previously-described LIMK inhibitor (LIMKi) induced dose-dependent microtubule alterations that resulted in significant mitotic defects, and increased the cytotoxic potency of microtubule polymerization inhibitors. By combining LIMKi with 366 compounds from the GSK Published Kinase Inhibitor Set, effective combinations were identified with kinase inhibitors including EGFR, p38 and Raf. These findings encouraged a drug discovery effort that led to development of CRT0105446 and CRT0105950, which potently block LIMK1 and LIMK2 activity in vitro, and inhibit cofilin phosphorylation and increase αTubulin acetylation in cells. CRT0105446 and CRT0105950 were screened against 656 cancer cell lines, and rhabdomyosarcoma, neuroblastoma and kidney cancer cells were identified as significantly sensitive to both LIMK inhibitors. These large-scale screens have identified effective LIMK inhibitor drug combinations and sensitive cancer types. In addition, the LIMK inhibitory compounds CRT0105446 and CRT0105950 will enable further development of LIMK-targeted cancer therapy.


Subject(s)
Lim Kinases/antagonists & inhibitors , Mitosis/drug effects , Neoplasms/drug therapy , Neoplasms/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , MCF-7 Cells , Microtubules/metabolism , Mitosis/physiology , Neoplasms/enzymology , Neuroblastoma/drug therapy , Neuroblastoma/enzymology , Neuroblastoma/pathology
5.
Oncotarget ; 6(28): 25619-30, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26296973

ABSTRACT

BACKGROUND: In this study, we established patient-derived tumor cell (PDC) models using tissues collected from patients with metastatic cancer and assessed whether these models could be used as a tool for genome-based cancer treatment. METHODS: PDCs were isolated and cultured from malignant effusions including ascites and pleural fluid. Pathological examination, immunohistochemical analysis, and genomic profiling were performed to compare the histological and genomic features of primary tumors, PDCs. An exploratory gene expression profiling assay was performed to further characterize PDCs. RESULTS: From January 2012 to May 2013, 176 samples from patients with metastatic cancer were collected. PDC models were successfully established in 130 (73.6%) samples. The median time from specimen collection to passage 1 (P1) was 3 weeks (range, 0.5-4 weeks), while that from P1 to P2 was 2.5 weeks (range, 0.5-5 weeks). Sixteen paired samples of genomic alterations were highly concordant between each primary tumor and progeny PDCs, with an average variant allele frequency (VAF) correlation of 0.878. We compared genomic profiles of the primary tumor (P0), P1 cells, P2 cells, and patient-derived xenografts (PDXs) derived from P2 cells and found that three samples (P0, P1, and P2 cells) were highly correlated (0.99-1.00). Moreover, PDXs showed more than 100 variants, with correlations of only 0.6-0.8 for the other samples. Drug responses of PDCs were reflective of the clinical response to targeted agents in selected patient PDC lines. CONCLUSION(S): Our results provided evidence that our PDC model was a promising model for preclinical experiments and closely resembled the patient tumor genome and clinical response.


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Tumor/genetics , Gene Expression Profiling , Genome, Human , Neoplasms/drug therapy , Neoplasms/genetics , Precision Medicine , Adult , Aged , Aged, 80 and over , Animals , Ascitic Fluid/pathology , Female , Gene Expression Profiling/methods , Gene Frequency , Genetic Predisposition to Disease , Humans , Male , Mice, Nude , Middle Aged , Molecular Targeted Therapy , Neoplasm Metastasis , Neoplasms/pathology , Patient Selection , Phenotype , Pleural Effusion, Malignant/pathology , Predictive Value of Tests , Primary Cell Culture , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Young Adult
6.
Mol Cell Biol ; 33(22): 4526-37, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24043311

ABSTRACT

In certain Ras mutant cell lines, the inhibition of extracellular signal-regulated kinase (ERK) signaling increases RhoA activity and inhibits cell motility, which was attributed to a decrease in Fra-1 levels. Here we report a Fra-1-independent augmentation of RhoA signaling during short-term inhibition of ERK signaling. Using mass spectrometry-based proteomics, we identified guanine exchange factor H1 (GEF-H1) as mediating this effect. ERK binds to the Rho exchange factor GEF-H1 and phosphorylates it on S959, causing inhibition of GEF-H1 activity and a consequent decrease in RhoA activity. Knockdown experiments and expression of a nonphosphorylatable S959A GEF-H1 mutant showed that this site is crucial in regulating cell motility and invasiveness. Thus, we identified GEF-H1 as a critical ERK effector that regulates motility, cell morphology, and invasiveness.


Subject(s)
Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , rhoA GTP-Binding Protein/metabolism , Amino Acid Sequence , Animals , Cell Line, Tumor , Cell Movement , HEK293 Cells , Humans , Molecular Sequence Data , Mutation , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Phosphorylation , Proto-Oncogene Proteins c-fos/metabolism , RNA Interference , Rats , Rho Guanine Nucleotide Exchange Factors/chemistry , Rho Guanine Nucleotide Exchange Factors/genetics , Signal Transduction
7.
J Med Chem ; 56(5): 1878-93, 2013 Mar 14.
Article in English | MEDLINE | ID: mdl-23394180

ABSTRACT

The mitotic kinesin Eg5 is critical for the assembly of the mitotic spindle and is a promising chemotherapy target. Previously, we identified S-trityl-L-cysteine as a selective inhibitor of Eg5 and developed triphenylbutanamine analogues with improved potency, favorable drug-like properties, but moderate in vivo activity. We report here their further optimization to produce extremely potent inhibitors of Eg5 (K(i)(app) < 10 nM) with broad-spectrum activity against cancer cell lines comparable to the Phase II drug candidates ispinesib and SB-743921. They have good oral bioavailability and pharmacokinetics and induced complete tumor regression in nude mice explanted with lung cancer patient xenografts. Furthermore, they display fewer liabilities with CYP-metabolizing enzymes and hERG compared with ispinesib and SB-743921, which is important given the likely application of Eg5 inhibitors in combination therapies. We present the case for this preclinical series to be investigated in single and combination chemotherapies, especially targeting hematological malignancies.


Subject(s)
Antineoplastic Agents/pharmacology , Butylamines/pharmacology , Cysteine/analogs & derivatives , Kinesins/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Benzamides/pharmacology , Butylamines/chemistry , Cell Line, Tumor , Chromones/pharmacology , Cysteine/chemistry , Cysteine/pharmacology , Lung Neoplasms/drug therapy , Mice , Mice, Nude , Neoplasm Transplantation , Quinazolines/pharmacology , Structure-Activity Relationship , Transplantation, Heterologous
8.
J Am Chem Soc ; 135(6): 2263-72, 2013 Feb 13.
Article in English | MEDLINE | ID: mdl-23305346

ABSTRACT

Human kinesin Eg5 is a target for drug development in cancer chemotherapy with compounds in phase II clinical trials. These agents bind to a well-characterized allosteric pocket involving the loop L5 region, a structural element in kinesin-5 family members thought to provide inhibitor specificity. Using X-ray crystallography, kinetic, and biophysical methods, we have identified and characterized a distinct allosteric pocket in Eg5 able to bind inhibitors with nanomolar K(d). This pocket is formed by key structural elements thought to be pivotal for force generation in kinesins and may represent a novel site for therapeutic intervention in this increasingly well-validated drug target.


Subject(s)
Benzimidazoles/pharmacology , Enzyme Inhibitors/pharmacology , Kinesins/antagonists & inhibitors , Benzimidazoles/chemistry , Binding Sites/drug effects , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Humans , Kinesins/chemistry , Kinesins/metabolism , Kinetics , Models, Molecular , Molecular Structure , Structure-Activity Relationship
9.
Nat Rev Cancer ; 12(8): 527-39, 2012 Jul 24.
Article in English | MEDLINE | ID: mdl-22825217

ABSTRACT

Kinesins are a family of molecular motors that travel unidirectionally along microtubule tracks to fulfil their many roles in intracellular transport or cell division. Over the past few years kinesins that are involved in mitosis have emerged as potential targets for cancer drug development. Several compounds that inhibit two mitotic kinesins (EG5 (also known as KIF11) and centromere-associated protein E (CENPE)) have entered Phase I and II clinical trials either as monotherapies or in combination with other drugs. Additional mitotic kinesins are currently being validated as drug targets, raising the possibility that the range of kinesin-based drug targets may expand in the future.


Subject(s)
Kinesins/antagonists & inhibitors , Kinesins/metabolism , Mitosis/drug effects , Neoplasms/drug therapy , Neoplasms/enzymology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Drug Evaluation, Preclinical , Humans
10.
Eur J Med Chem ; 54: 483-98, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22749640

ABSTRACT

S-Trityl L-cysteine (STLC) is an inhibitor of the mitotic kinesin Eg5 with potential as an antimitotic chemotherapeutic agent. We previously reported the crystal structure of the ligand-protein complex, and now for the first time, have quantified the interactions using a molecular dynamics based approach. Based on these data, we have explored the SAR of the trityl head group using the methylene shuffle strategy to expand the occupation of one of the hydrophobic pockets. The most potent compounds exhibit strong (<100 nM) inhibition of Eg5 in the basal ATPase assay and inhibit growth in a variety of tumour-derived cell lines.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cysteine/analogs & derivatives , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Kinesins/antagonists & inhibitors , Antineoplastic Agents/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cysteine/chemistry , Cysteine/metabolism , Cysteine/pharmacology , Drug Resistance, Multiple/drug effects , Enzyme Inhibitors/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Kinesins/chemistry , Kinesins/metabolism , Molecular Dynamics Simulation , Protein Conformation , Structure-Activity Relationship , Thermodynamics
11.
J Med Chem ; 55(4): 1511-25, 2012 Feb 23.
Article in English | MEDLINE | ID: mdl-22248262

ABSTRACT

The human mitotic kinesin Eg5 represents a novel mitotic spindle target for cancer chemotherapy. We previously identified S-trityl-l-cysteine (STLC) and related analogues as selective potent inhibitors of Eg5. We herein report on the development of a series of 4,4,4-triphenylbutan-1-amine inhibitors derived from the STLC scaffold. This new generation systematically improves on potency: the most potent C-trityl analogues exhibit K(i)(app) ≤ 10 nM and GI(50) ≈ 50 nM, comparable to results from the phase II clinical benchmark ispinesib. Crystallographic studies reveal that they adopt the same overall binding configuration as S-trityl analogues at an allosteric site formed by loop L5 of Eg5. Evaluation of their druglike properties reveals favorable profiles for future development and, in the clinical candidate ispinesib, moderate hERG and CYP inhibition. One triphenylbutanamine analogue and ispinesib possess very good bioavailability (51% and 45%, respectively), with the former showing in vivo antitumor growth activity in nude mice xenograft studies.


Subject(s)
Antineoplastic Agents/chemical synthesis , Benzene Derivatives/chemical synthesis , Butylamines/chemical synthesis , Kinesins/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Benzene Derivatives/pharmacokinetics , Benzene Derivatives/pharmacology , Biological Availability , Butylamines/pharmacokinetics , Butylamines/pharmacology , Cell Line, Tumor , Drug Screening Assays, Antitumor , Female , Humans , Mice , Mice, Nude , Models, Molecular , Molecular Structure , Neoplasm Transplantation , Protein Binding , Protein Conformation , Quinazolines/pharmacology , Stereoisomerism , Structure-Activity Relationship , Transplantation, Heterologous
12.
Cancer Res ; 71(4): 1334-43, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21303975

ABSTRACT

Raf kinase inhibitory protein (RKIP) is a physiologic inhibitor of c-RAF kinase and nuclear factor κB signaling that represses tumor invasion and metastasis. Glycogen synthase kinase-3ß (GSK3ß) suppresses tumor progression by downregulating multiple oncogenic pathways including Wnt signaling and cyclin D1 activation. Here, we show that RKIP binds GSK3 proteins and maintains GSK3ß protein levels and its active form. Depletion of RKIP augments oxidative stress-mediated activation of the p38 mitogen activated protein kinase, which, in turn, inactivates GSK3ß by phosphorylating it at the inhibitory T390 residue. This pathway de-represses GSK3ß inhibition of oncogenic substrates causing stabilization of cyclin D, which induces cell-cycle progression and ß-catenin, SNAIL, and SLUG, which promote epithelial to mesenchymal transition. RKIP levels in human colorectal cancer positively correlate with GSK3ß expression. These findings reveal the RKIP/GSK3 axis as both a potential therapeutic target and a prognosis-based predictor of cancer progression.


Subject(s)
Glycogen Synthase Kinase 3/physiology , Phosphatidylethanolamine Binding Protein/physiology , Animals , Carcinoma/genetics , Carcinoma/metabolism , Carcinoma/pathology , Cells, Cultured , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Disease Progression , Enzyme Stability , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Mice , Mice, Knockout , Oxidative Stress/genetics , Oxidative Stress/physiology , Phosphatidylethanolamine Binding Protein/genetics , Phosphatidylethanolamine Binding Protein/metabolism , Phosphorylation , Protein Binding/physiology , Signal Transduction/genetics , Up-Regulation/physiology
13.
Mol Biosyst ; 7(3): 928-41, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21180766

ABSTRACT

RKIP-1 is a metastasis suppressor that is frequently downregulated in aggressive cancers. However, the consequences of RKIP loss in primary or immortalized cells have not yet been explored. Using HEK-293 RKIP depleted (termed HEK-499) and Flp-In T-Rex-293 RKIP inducible cell lines combined with whole transcriptome analysis, we show that RKIP-1 silencing accelerates DNA synthesis and G1/S transition entry by inducing the expression of cdc6, MCM 2, 4, 6, 7, cdc45L, cyclin D2, cyclin E2, cyclin D1, SKP2 and the downregulation of p21(cip1). Moreover, RKIP depletion accelerates the time from nuclear envelop breakdown (NEB) to anaphase markedly, while the upregulation of RKIP shortened the NEB to anaphase time. We show that RKIP depletion induces the expression of NEK6, a molecule known to enhance G2/M transition, and down-regulates G2/M checkpoint molecules like Aurora B, cyclin G1 and sertuin that slow the G2/M transition time. These subtle changes in the kinetics of the cell cycle culminate in a higher proliferation rate of HEK-499 compared to control cells. Finally, we show that RKIP depletion enhances cellular motility by inducing the expression/stabilization of ß-catenin, vimentin, MET and PAK1. Overall, our data suggest that modulation of the cell cycle checkpoints and motility by RKIP may be fundamental to its metastasis suppressive function in cancer and that RKIP role in a cell is more intricate and diverse than previously thought.


Subject(s)
Cell Cycle , Cell Movement , Phosphatidylethanolamine Binding Protein/metabolism , Apoptosis , Cell Proliferation , Cells, Cultured , Gene Silencing , Humans , Kinetics , Phosphatidylethanolamine Binding Protein/genetics
15.
Mol Biosyst ; 6(11): 2174-91, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20717620

ABSTRACT

The investigation of the structure and dynamics of signal transduction systems through data-based mathematical models in ordinary differential equations or other paradigms has proven to be a successful approach in recent times. Extending this concept, we here analysed the use of kinetic models based on power-law terms with non-integer kinetic orders in the validation of hypotheses concerning regulatory structures in signalling systems. We integrated pre-existent biological knowledge, hypotheses and experimental quantitative data into a power-law model to validate the existence of certain regulatory loops in the Ras/Raf-1/MEK/ERK pathway, a MAPK pathway involved in the transduction of mitogenic and differentiation signals. Towards this end, samples of a human mammary epithelial cell line (MCF-10A) were used to obtain time-series data, characterising the behaviour of the system after epidermal growth factor stimulation in different scenarios of expression for the critical players of the system regarding the investigated loops (e.g., the inhibitory protein RKIP). The mathematical model was calibrated using a computational procedure that included: analysis of structural identifiability, global ranking of parameters to detect the most sensitivity ones towards the experimental setup, model calibration using global optimization methods to find the parameter values that better fit the data, and practical identifiability analysis to estimate the confidence in the estimated values for the parameters. The obtained model was used to perform computational simulations concerning the role of the investigated regulatory loops in the time response of the signalling pathway. Our findings suggest that the special regularity in the structure of the power-law terms make them suitable for a data-based validation of regulatory loops in signalling pathways. The model-based analysis performed identified RKIP as an actual inhibitor of the activation of the ERK pathway, but also suggested the existence of an intense feedback-loop control of the pathway by the activated ERK that maybe responsible for the damped oscillations we saw in the fraction of activated MEK both in the experiments and simulations. In addition, the model analysis suggested that phosphorylation/deactivation of RKIP during the transient stimulation may have a significant effect on the signalling peaks of both MEK and ERK. This later result suggests that dynamic modulation of signal inhibitors during stimulation may be a regulatory mechanism in ERK signalling and other pathways.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Feedback, Physiological , MAP Kinase Signaling System , Models, Biological , Calibration , Cell Line , Enzyme Activation , Humans , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphatidylethanolamine Binding Protein/metabolism , Phosphorylation
16.
Cancer Res ; 70(17): 6715-24, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20736375

ABSTRACT

Epithelial-mesenchymal transition (EMT) is a key event in the generation of invasive tumor cells. A hallmark of EMT is the repression of E-cadherin expression, which is regulated by various signal transduction pathways including extracellular signal-regulated kinase (ERK) and Wnt. These pathways are highly interconnected via multiple coupled feedback loops (CFL). As the function of such coupled feedback regulations is difficult to analyze experimentally, we used a systems biology approach where computational models were designed to predict biological effects that result from the complex interplay of CFLs. Using epidermal growth factor (EGF) and Wnt as input and E-cadherin transcriptional regulation as output, we established an ordinary differential equation model of the ERK and Wnt signaling network containing six feedback links and used extensive computer simulations to analyze the effects of these feedback links in isolation and different combinations. The results show that the feedbacks can generate a rich dynamic behavior leading to various dose-response patterns and have a decisive role in determining network responses to EGF and Wnt. In particular, we made two important findings: first, that coupled positive feedback loops composed of phosphorylation of Raf kinase inhibitor RKIP by ERK and transcriptional repression of RKIP by Snail have an essential role in causing a switch-like behavior of E-cadherin expression; and second, that RKIP expression inhibits EMT progression by preventing E-cadherin suppression. Taken together, our findings provide us with a system-level understanding of how RKIP can regulate EMT progression and may explain why RKIP is downregulated in so many metastatic cancer cells.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Models, Biological , Neoplasms/metabolism , Neoplasms/pathology , Wnt Proteins/metabolism , Animals , Cadherins/biosynthesis , Epidermal Growth Factor/metabolism , Epithelial Cells/pathology , Feedback, Physiological , Humans , MAP Kinase Signaling System , Mesoderm/pathology , Phosphatidylethanolamine Binding Protein/metabolism , Snail Family Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
17.
J Med Chem ; 53(15): 5676-83, 2010 Aug 12.
Article in English | MEDLINE | ID: mdl-20597485

ABSTRACT

Human kinesin Eg5, which plays an essential role in mitosis by establishing the bipolar spindle, has proven to be an interesting drug target for the development of cancer chemotherapeutics. Here, we report the crystal structures of the Eg5 motor domain complexed with enastron, dimethylenastron, and fluorastrol. By comparing these structures to that of monastrol and mon-97, we identified the main reasons for increased potency of these new inhibitors, namely the better fit of the ligand to the allosteric binding site and the addition of fluorine atoms. We also noticed preferential binding of the S-enantiomer of enastron and dimethylenastron to Eg5, while the R-enantiomer of fluorastrol binds preferentially to Eg5. In addition, we performed a multidrug resistance (MDR) study in cell lines overexpressing P-glycoprotein (Pgp). We showed that one of these inhibitors may have the potential to overcome susceptibility to this efflux pump and hence overcome common resistance associated with tubulin-targeting drugs.


Subject(s)
Antimitotic Agents/chemistry , Kinesins/antagonists & inhibitors , Pyrimidines/chemistry , Quinazolines/chemistry , Thiones/chemistry , ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Antimitotic Agents/chemical synthesis , Antimitotic Agents/pharmacology , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallography, X-Ray , Drug Resistance, Multiple , Humans , Kinesins/chemistry , Models, Molecular , Protein Conformation , Protein Structure, Tertiary , Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , Quinazolines/chemical synthesis , Quinazolines/pharmacology , Stereoisomerism , Structure-Activity Relationship , Thiones/chemical synthesis , Thiones/pharmacology
18.
EMBO Rep ; 10(3): 278-84, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19197339

ABSTRACT

The RAF-MEK-ERK pathway regulates both myoblast proliferation and differentiation; however, it is unclear how these events are coordinated. Here, we show that human phosphatidylethanolamine-binding protein 4 (PEBP4), a RAF kinase inhibitory protein (RKIP) family protein expressed preferentially in muscle, regulates the activity of the ERK pathway and myoblast differentiation by acting as a scaffold protein. In contrast to RKIP, which disrupts the RAF1-MEK interaction, PEBP4 forms ternary complexes with RAF1 and MEK, and can scaffold this interaction. PEBP4 expression is induced during the differentiation of primary human myoblasts. Consistent with the properties of a scaffold, PEBP4 enhances the RAF1-MEK interaction and the activation of MEK at low expression levels, whereas it inhibits these parameters at higher expression levels. Downregulation of PEBP4 by short hairpin RNA in human myoblasts increases MEK signalling and inhibits differentiation; by contrast, PEBP4 overexpression enhances differentiation. Thus, PEBP4 participates in the control of muscle cell differentiation by modulating the activity of MEK and ERK.


Subject(s)
Cell Differentiation/physiology , MAP Kinase Signaling System/physiology , Myoblasts/physiology , Phosphatidylethanolamine Binding Protein/metabolism , Animals , COS Cells , Chlorocebus aethiops , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Molecular Sequence Data , Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , Myoblasts/cytology , Phosphatidylethanolamine Binding Protein/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/metabolism , RNA Interference
19.
J Cell Sci ; 122(Pt 3): 425-35, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-19158341

ABSTRACT

The Ras-Raf-MEK-ERK pathway (or ERK pathway) is an important signal transduction system involved in the control of cell proliferation, survival and differentiation. However, the dynamic regulation of the pathway by positive- and negative-feedback mechanisms, in particular the functional role of Raf kinase inhibitor protein (RKIP) are still incompletely understood. RKIP is a physiological endogenous inhibitor of MEK phosphorylation by Raf kinases, but also participates in a positive-feedback loop in which ERK can inactivate RKIP. The aim of this study was to elucidate the hidden dynamics of these feedback mechanisms and to identify the functional role of RKIP through combined efforts of biochemical experiments and in silico simulations based on an experimentally validated mathematical model. We show that the negative-feedback loop from ERK to SOS plays a crucial role in generating an oscillatory behavior of ERK activity. The positive-feedback loop in which ERK functionally inactivates RKIP also enhances the oscillatory activation pattern of ERK. However, RKIP itself has an important role in inducing a switch-like behavior of MEK activity. When overexpressed, RKIP also causes delayed and reduced responses of ERK. Thus, positive- and negative-feedback loops and RKIP work together to shape the response pattern and dynamical characteristics of the ERK pathway.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Kinase Kinases/metabolism , Phosphatidylethanolamine Binding Protein/metabolism , Signal Transduction , raf Kinases/metabolism , ras Proteins/metabolism , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Feedback, Physiological , Models, Theoretical , Phosphorylation/physiology
20.
Cell Signal ; 20(5): 935-41, 2008 May.
Article in English | MEDLINE | ID: mdl-18294816

ABSTRACT

The Raf-MEK-ERK pathway regulates many fundamental biological processes, and its activity is finely tuned at multiple levels. The Raf kinase inhibitory protein (RKIP) is a widely expressed negative modulator of the Raf-MEK-ERK signaling pathway. We have previously shown that RKIP inhibits the phosphorylation of MEK by Raf-1 through interfering with the formation of a kinase-substrate complex by direct binding to both Raf-1 and MEK. Here, we show that the evolutionarily conserved ligand-binding pocket of RKIP is required for its inhibitory activity towards the Raf-1 kinase mediated activation of MEK. Single amino acid substitutions of two of the conserved residues form the base and the wall of the pocket confers a loss-of-function phenotype on RKIP. Loss-of-function RKIP mutants still appear to bind to Raf-1. However the stability of the complexes formed between mutants and the N-region Raf-1 phosphopeptide were drastically reduced. Our results therefore suggest that the RKIP conserved pocket may constitute a novel phosphoamino-acid binding motif and is absolutely required for RKIP function.


Subject(s)
Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphatidylethanolamine Binding Protein/chemistry , Phosphatidylethanolamine Binding Protein/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Amino Acid Substitution , Animals , Binding Sites/genetics , COS Cells , Chlorocebus aethiops , Conserved Sequence , Humans , MAP Kinase Signaling System , Models, Molecular , Mutagenesis, Site-Directed , Phosphatidylethanolamine Binding Protein/genetics , Phosphorylation , Protein Conformation , Proto-Oncogene Proteins c-raf/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...