Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Cureus ; 12(5): e7931, 2020 May 02.
Article in English | MEDLINE | ID: mdl-32499975

ABSTRACT

Background As Radiation Oncology (RO) is a field with limited exposure in undergraduate medical education curricula, the information sources used to form students' perception of the field can have a substantial impact on whether students decide to pursue experiences in RO. Furthermore, the effects of a single elective experience in RO can strongly influence career decisions as it may serve as the only experience for students to gain an understanding of RO as a specialty. This study analyzes which information sources students use and most strongly value when forming their perception of RO both before and after participating in the program, while also analyzing changes in the perception of various speciality-related factors associated with RO. Methods To address underrepresented specialties, the Pre-clerkship Residency Exploration Program (PREP) was developed to provide students exposure to RO and 13 other specialties through half-day clinical rotations, simulations, skills sessions, and panel discussions. A total of 37 participants completed both "Pre-program" and "Post-program" surveys to evaluate which information sources they use and value most when forming their perception of RO, and student perception of career factors associated with RO was assessed. Results Students reported that Pre-program information sources of RO were based on Lectures (35 students, 94.6%) and Preceptors (18 students, 48.7%). Post-program responses indicated that the greatest sources of information used were from Preceptors (36 students, 97.3%) and Residents (34 students, 91.9%), with the greatest increase being found in interactions with Residents for gaining specialty information (78% increase). Students most highly valued Preceptors, Residents, and Lectures as information sources when forming their perception of RO. Pre-program, students had the greatest positive perception of RO with respect to Income Potential (mean: 3.76/5.00 ± 0.87), Intellectual Challenge (mean: 3.90/5.00 ± 0.94), and Research Opportunities (mean: 3.86/5.00 ± 0.83) while most negatively assessing the factors of Flexibility (mean: 2.69/5.00 ± 0.93) and Level of Stress (mean: 2.93/5.00 ± 0.94). Conclusions Student perception of a medical specialty is a factor that may influence student elective choice and career decisions. Through participating in PREP, significant positive increases were found in students' perception of RO in the areas of Flexibility, Patient Population, Competitiveness of the Specialty, Quality of the Working Environment, and Levels of Stress. This study highlights which information sources students value the most when forming their perception of RO and the impact a single elective experience has on improving student perception of the field. RO-based programs and lectures can be better designed using this information to introduce students to this specialty.

2.
Sci Rep ; 9(1): 201, 2019 01 17.
Article in English | MEDLINE | ID: mdl-30655555

ABSTRACT

Inhibition of the PARP superfamily tankyrase enzymes suppresses Wnt/ß-catenin signalling in tumour cells. Here, we describe here a novel, drug-like small molecule inhibitor of tankyrase MSC2504877 that inhibits the growth of APC mutant colorectal tumour cells. Parallel siRNA and drug sensitivity screens showed that the clinical CDK4/6 inhibitor palbociclib, causes enhanced sensitivity to MSC2504877. This tankyrase inhibitor-CDK4/6 inhibitor combinatorial effect is not limited to palbociclib and MSC2504877 and is elicited with other CDK4/6 inhibitors and toolbox tankyrase inhibitors. The addition of MSC2504877 to palbociclib enhances G1 cell cycle arrest and cellular senescence in tumour cells. MSC2504877 exposure suppresses the upregulation of Cyclin D2 and Cyclin E2 caused by palbociclib and enhances the suppression of phospho-Rb, providing a mechanistic explanation for these effects. The combination of MSC2504877 and palbociclib was also effective in suppressing the cellular hyperproliferative phenotype seen in Apc defective intestinal stem cells in vivo. However, the presence of an oncogenic Kras p.G12D mutation in mice reversed the effects of the MSC2504877/palbociclib combination, suggesting one molecular route that could lead to drug resistance.


Subject(s)
Colorectal Neoplasms/drug therapy , Enzyme Inhibitors/pharmacology , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Tankyrases/antagonists & inhibitors , Animals , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cellular Senescence/drug effects , Colorectal Neoplasms/pathology , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Drug Resistance, Neoplasm , Drug Synergism , Enzyme Inhibitors/therapeutic use , Humans , Mice , Piperazines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyridines/therapeutic use
4.
Sci Rep ; 8(1): 10614, 2018 Jul 13.
Article in English | MEDLINE | ID: mdl-30006631

ABSTRACT

Osteosarcoma (OS) is an aggressive sarcoma, where novel treatment approaches are required. Genomic studies suggest that a subset of OS, including OS tumour cell lines (TCLs), exhibit genomic loss of heterozygosity (LOH) patterns reminiscent of BRCA1 or BRCA2 mutant tumours. This raises the possibility that PARP inhibitors (PARPi), used to treat BRCA1/2 mutant cancers, could be used to target OS. Using high-throughput drug sensitivity screening we generated chemosensitivity profiles for 79 small molecule inhibitors, including three clinical PARPi. Drug screening was performed in 88 tumour cell lines, including 18 OS TCLs. This identified known sensitivity effects in OS TCLs, such as sensitivity to FGFR inhibitors. When compared to BRCA1/2 mutant TCLs, OS TCLs, with the exception of LM7, were PARPi resistant, including those with previously determined BRCAness LoH profiles. Post-screen validation experiments confirmed PARPi sensitivity in LM7 cells as well as a defect in the ability to form nuclear RAD51 foci in response to DNA damage. LM7 provides one OS model for the study of PARPi sensitivity through a potential defect in RAD51-mediated DNA repair. The drug sensitivity dataset we generated in 88 TCLs could also serve as a resource for the study of drug sensitivity effects in OS.


Subject(s)
BRCA1 Protein/genetics , BRCA2 Protein/genetics , Drug Resistance, Neoplasm/genetics , Osteosarcoma/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , CRISPR-Cas Systems/genetics , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/genetics , Cell Nucleus/metabolism , DNA Damage/drug effects , DNA Damage/genetics , DNA Repair/drug effects , DNA Repair/genetics , Datasets as Topic , Drug Screening Assays, Antitumor/methods , High-Throughput Screening Assays/methods , Humans , Mutagenesis , Mutation , Osteosarcoma/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Rad51 Recombinase/metabolism
5.
Nat Commun ; 9(1): 1849, 2018 05 10.
Article in English | MEDLINE | ID: mdl-29748565

ABSTRACT

Although PARP inhibitors (PARPi) target homologous recombination defective tumours, drug resistance frequently emerges, often via poorly understood mechanisms. Here, using genome-wide and high-density CRISPR-Cas9 "tag-mutate-enrich" mutagenesis screens, we identify close to full-length mutant forms of PARP1 that cause in vitro and in vivo PARPi resistance. Mutations both within and outside of the PARP1 DNA-binding zinc-finger domains cause PARPi resistance and alter PARP1 trapping, as does a PARP1 mutation found in a clinical case of PARPi resistance. This reinforces the importance of trapped PARP1 as a cytotoxic DNA lesion and suggests that PARP1 intramolecular interactions might influence PARPi-mediated cytotoxicity. PARP1 mutations are also tolerated in cells with a pathogenic BRCA1 mutation where they result in distinct sensitivities to chemotherapeutic drugs compared to other mechanisms of PARPi resistance (BRCA1 reversion, 53BP1, REV7 (MAD2L2) mutation), suggesting that the underlying mechanism of PARPi resistance that emerges could influence the success of subsequent therapies.


Subject(s)
Drug Resistance, Neoplasm/genetics , Neoplasms/drug therapy , Poly (ADP-Ribose) Polymerase-1/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Aged , Animals , BRCA1 Protein/genetics , CRISPR-Cas Systems , Cell Line, Tumor , DNA Mutational Analysis/methods , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Mouse Embryonic Stem Cells , Mutagenesis , Neoplasms/genetics , Neoplasms/pathology , Phthalazines/pharmacology , Phthalazines/therapeutic use , Point Mutation , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Precision Medicine/methods , Whole Genome Sequencing/methods , Xenograft Model Antitumor Assays , Zinc Fingers/genetics
6.
Cancer Discov ; 8(4): 498-515, 2018 04.
Article in English | MEDLINE | ID: mdl-29610289

ABSTRACT

The cell adhesion glycoprotein E-cadherin (CDH1) is commonly inactivated in breast tumors. Precision medicine approaches that exploit this characteristic are not available. Using perturbation screens in breast tumor cells with CRISPR/Cas9-engineered CDH1 mutations, we identified synthetic lethality between E-cadherin deficiency and inhibition of the tyrosine kinase ROS1. Data from large-scale genetic screens in molecularly diverse breast tumor cell lines established that the E-cadherin/ROS1 synthetic lethality was not only robust in the face of considerable molecular heterogeneity but was also elicited with clinical ROS1 inhibitors, including foretinib and crizotinib. ROS1 inhibitors induced mitotic abnormalities and multinucleation in E-cadherin-defective cells, phenotypes associated with a defect in cytokinesis and aberrant p120 catenin phosphorylation and localization. In vivo, ROS1 inhibitors produced profound antitumor effects in multiple models of E-cadherin-defective breast cancer. These data therefore provide the preclinical rationale for assessing ROS1 inhibitors, such as the licensed drug crizotinib, in appropriately stratified patients.Significance: E-cadherin defects are common in breast cancer but are currently not targeted with a precision medicine approach. Our preclinical data indicate that licensed ROS1 inhibitors, including crizotinib, should be repurposed to target E-cadherin-defective breast cancers, thus providing the rationale for the assessment of these agents in molecularly stratified phase II clinical trials. Cancer Discov; 8(4); 498-515. ©2018 AACR.This article is highlighted in the In This Issue feature, p. 371.


Subject(s)
Breast Neoplasms/drug therapy , Cadherins/deficiency , Crizotinib/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Anilides/pharmacology , Anilides/therapeutic use , Animals , Antigens, CD/genetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/metabolism , Cadherins/genetics , Cell Line, Tumor , Crizotinib/therapeutic use , Female , Humans , Mice , Mutation , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Quinolines/pharmacology , Quinolines/therapeutic use
7.
Mol Cancer Ther ; 16(9): 2022-2034, 2017 09.
Article in English | MEDLINE | ID: mdl-28619759

ABSTRACT

Although PARP inhibitors target BRCA1- or BRCA2-mutant tumor cells, drug resistance is a problem. PARP inhibitor resistance is sometimes associated with the presence of secondary or "revertant" mutations in BRCA1 or BRCA2 Whether secondary mutant tumor cells are selected for in a Darwinian fashion by treatment is unclear. Furthermore, how PARP inhibitor resistance might be therapeutically targeted is also poorly understood. Using CRISPR mutagenesis, we generated isogenic tumor cell models with secondary BRCA1 or BRCA2 mutations. Using these in heterogeneous in vitro culture or in vivo xenograft experiments in which the clonal composition of tumor cell populations in response to therapy was monitored, we established that PARP inhibitor or platinum salt exposure selects for secondary mutant clones in a Darwinian fashion, with the periodicity of PARP inhibitor administration and the pretreatment frequency of secondary mutant tumor cells influencing the eventual clonal composition of the tumor cell population. In xenograft studies, the presence of secondary mutant cells in tumors impaired the therapeutic effect of a clinical PARP inhibitor. However, we found that both PARP inhibitor-sensitive and PARP inhibitor-resistant BRCA2 mutant tumor cells were sensitive to AZD-1775, a WEE1 kinase inhibitor. In mice carrying heterogeneous tumors, AZD-1775 delivered a greater therapeutic benefit than olaparib treatment. This suggests that despite the restoration of some BRCA1 or BRCA2 gene function in "revertant" tumor cells, vulnerabilities still exist that could be therapeutically exploited. Mol Cancer Ther; 16(9); 2022-34. ©2017 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Drug Resistance, Neoplasm/genetics , Mutation , Animals , Cell Cycle/drug effects , Cell Cycle Proteins/antagonists & inhibitors , Cell Line, Tumor , DNA Mutational Analysis , Disease Models, Animal , Female , Gene Knockdown Techniques , Humans , Mice , Nuclear Proteins/antagonists & inhibitors , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Pyrimidinones , Selection, Genetic , Xenograft Model Antitumor Assays
8.
Sci Data ; 4: 170020, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28248920

ABSTRACT

We describe a screen for cellular response to drugs that makes use of haploid embryonic stem cells. We generated ten libraries of mutants with piggyBac gene trap transposon integrations, totalling approximately 100,000 mutant clones. Random barcode sequences were inserted into the transposon vector to allow the number of cells bearing each insertion to be measured by amplifying and sequencing the barcodes. These barcodes were associated with their integration sites by inverse PCR. We exposed these libraries to commonly used cancer drugs and profiled changes in barcode abundance by Ion Torrent sequencing in order to identify mutations that conferred sensitivity. Drugs tested included conventional chemotherapeutics as well as targeted inhibitors of topoisomerases, poly(ADP-ribose) polymerase (PARP), Hsp90 and WEE1.


Subject(s)
DNA Transposable Elements , Mouse Embryonic Stem Cells , Neoplasms , Animals , Antineoplastic Agents/pharmacology , Genome-Wide Association Study , Haploidy , Mice , Neoplasms/drug therapy , Neoplasms/genetics
9.
Nat Commun ; 7: 13837, 2016 12 13.
Article in English | MEDLINE | ID: mdl-27958275

ABSTRACT

Identifying genetic biomarkers of synthetic lethal drug sensitivity effects provides one approach to the development of targeted cancer therapies. Mutations in ARID1A represent one of the most common molecular alterations in human cancer, but therapeutic approaches that target these defects are not yet clinically available. We demonstrate that defects in ARID1A sensitize tumour cells to clinical inhibitors of the DNA damage checkpoint kinase, ATR, both in vitro and in vivo. Mechanistically, ARID1A deficiency results in topoisomerase 2A and cell cycle defects, which cause an increased reliance on ATR checkpoint activity. In ARID1A mutant tumour cells, inhibition of ATR triggers premature mitotic entry, genomic instability and apoptosis. The data presented here provide the pre-clinical and mechanistic rationale for assessing ARID1A defects as a biomarker of single-agent ATR inhibitor response and represents a novel synthetic lethal approach to targeting tumour cells.


Subject(s)
Cell Cycle Checkpoints/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics , Antineoplastic Agents/pharmacology , Apoptosis , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , DNA-Binding Proteins , Humans , RNA Interference
10.
Mol Cancer Ther ; 15(7): 1472-84, 2016 07.
Article in English | MEDLINE | ID: mdl-27364904

ABSTRACT

New targeted approaches to ovarian clear cell carcinomas (OCCC) are needed, given the limited treatment options in this disease and the poor response to standard chemotherapy. Using a series of high-throughput cell-based drug screens in OCCC tumor cell models, we have identified a synthetic lethal (SL) interaction between the kinase inhibitor dasatinib and a key driver in OCCC, ARID1A mutation. Imposing ARID1A deficiency upon a variety of human or mouse cells induced dasatinib sensitivity, both in vitro and in vivo, suggesting that this is a robust synthetic lethal interaction. The sensitivity of ARID1A-deficient cells to dasatinib was associated with G1-S cell-cycle arrest and was dependent upon both p21 and Rb. Using focused siRNA screens and kinase profiling, we showed that ARID1A-mutant OCCC tumor cells are addicted to the dasatinib target YES1. This suggests that dasatinib merits investigation for the treatment of patients with ARID1A-mutant OCCC. Mol Cancer Ther; 15(7); 1472-84. ©2016 AACR.


Subject(s)
Adenocarcinoma, Clear Cell/genetics , Antineoplastic Agents/pharmacology , Dasatinib/pharmacology , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Ovarian Neoplasms/genetics , Protein Kinase Inhibitors/pharmacology , Synthetic Lethal Mutations , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Adenocarcinoma, Clear Cell/drug therapy , Adenocarcinoma, Clear Cell/pathology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , DNA-Binding Proteins , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Drug Screening Assays, Antitumor , Female , Gene Expression Profiling , Humans , Mice , Molecular Targeted Therapy , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Xenograft Model Antitumor Assays
11.
Cell Rep ; 14(10): 2490-501, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26947069

ABSTRACT

One approach to identifying cancer-specific vulnerabilities and therapeutic targets is to profile genetic dependencies in cancer cell lines. Here, we describe data from a series of siRNA screens that identify the kinase genetic dependencies in 117 cancer cell lines from ten cancer types. By integrating the siRNA screen data with molecular profiling data, including exome sequencing data, we show how vulnerabilities/genetic dependencies that are associated with mutations in specific cancer driver genes can be identified. By integrating additional data sets into this analysis, including protein-protein interaction data, we also demonstrate that the genetic dependencies associated with many cancer driver genes form dense connections on functional interaction networks. We demonstrate the utility of this resource by using it to predict the drug sensitivity of genetically or histologically defined subsets of tumor cell lines, including an increased sensitivity of osteosarcoma cell lines to FGFR inhibitors and SMAD4 mutant tumor cells to mitotic inhibitors.


Subject(s)
Neoplasms/enzymology , Neoplasms/genetics , Protein Kinases/metabolism , Cell Line, Tumor , Gene Expression Profiling , Humans , Mutation , Neoplasms/pathology , Protein Kinases/chemistry , Protein Kinases/genetics , RNA Interference , RNA, Small Interfering/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Smad4 Protein/antagonists & inhibitors , Smad4 Protein/genetics , Smad4 Protein/metabolism
12.
Cancer Res ; 74(1): 287-97, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24240700

ABSTRACT

Small-molecule inhibitors of PARP1/2, such as olaparib, have been proposed to serve as a synthetic lethal therapy for cancers that harbor BRCA1 or BRCA2 mutations. Indeed, in clinical trials, PARP1/2 inhibitors elicit sustained antitumor responses in patients with germline BRCA gene mutations. In hypothesizing that additional genetic determinants might direct use of these drugs, we conducted a genome-wide synthetic lethal screen for candidate olaparib sensitivity genes. In support of this hypothesis, the set of identified genes included known determinants of olaparib sensitivity, such as BRCA1, RAD51, and Fanconi's anemia susceptibility genes. In addition, the set included genes implicated in established networks of DNA repair, DNA cohesion, and chromatin remodeling, none of which were known previously to confer sensitivity to PARP1/2 inhibition. Notably, integration of the list of candidate sensitivity genes with data from tumor DNA sequencing studies identified CDK12 deficiency as a clinically relevant biomarker of PARP1/2 inhibitor sensitivity. In models of high-grade serous ovarian cancer (HGS-OVCa), CDK12 attenuation was sufficient to confer sensitivity to PARP1/2 inhibition, suppression of DNA repair via homologous recombination, and reduced expression of BRCA1. As one of only nine genes known to be significantly mutated in HGS-OVCa, CDK12 has properties that should confirm interest in its use as a biomarker, particularly in ongoing clinical trials of PARP1/2 inhibitors and other agents that trigger replication fork arrest.


Subject(s)
Ovarian Neoplasms/genetics , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Cystadenocarcinoma, Serous/drug therapy , Cystadenocarcinoma, Serous/enzymology , Cystadenocarcinoma, Serous/genetics , Enzyme Inhibitors/pharmacology , Female , Gene Expression Profiling , Genome, Human , Genome-Wide Association Study , Germ-Line Mutation , Humans , Immunohistochemistry , Mice , Mice, Nude , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/enzymology , Phthalazines/pharmacology , Piperazines/pharmacology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Random Allocation , Survival Analysis , Xenograft Model Antitumor Assays
13.
Cancer Discov ; 1(3): 260-73, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21984977

ABSTRACT

UNLABELLED: The design of targeted therapeutic strategies for cancer has largely been driven by the identification of tumor-specific genetic changes. However, the large number of genetic alterations present in tumor cells means that it is difficult to discriminate between genes that are critical for maintaining the disease state and those that are merely coincidental. Even when critical genes can be identified, directly targeting these is often challenging, meaning that alternative strategies such as exploiting synthetic lethality may be beneficial. To address these issues, we have carried out a functional genetic screen in >30 commonly used models of breast cancer to identify genes critical to the growth of specific breast cancer subtypes. In particular, we describe potential new therapeutic targets for PTEN-mutated cancers and for estrogen receptor-positive breast cancers. We also show that large-scale functional profiling allows the classification of breast cancers into subgroups distinct from established subtypes. SIGNIFICANCE: Despite the wealth of molecular profiling data that describe breast tumors and breast tumor cell models, our understanding of the fundamental genetic dependencies in this disease is relatively poor. Using high-throughput RNA interference screening of a series of pharmacologically tractable genes, we have generated comprehensive functional viability profiles for a wide panel of commonly used breast tumor cell models. Analysis of these profiles identifies a series of novel genetic dependencies, including that of PTEN-null breast tumor cells upon mitotic checkpoint kinases, and provides a framework upon which additional dependencies and candidate therapeutic targets may be identified.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Gene Expression Profiling/trends , Genetic Testing/methods , Humans , M Phase Cell Cycle Checkpoints/genetics , Mutation , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Protein Kinases/genetics , Protein Kinases/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism
14.
Proteomics ; 9(18): 4284-97, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19725078

ABSTRACT

Parkin is an ubiquitin-protein ligase (E3), mutations of which cause juvenile onset - autosomal recessive Parkinson's disease, and result in reduced enzymic activity. In contrast, increased levels are protective against mitochondrial dysfunction and neurodegeneration, the mechanism of which is largely unknown. In this study, 2-DE and MS proteomic techniques were utilised to investigate the effects of increased Parkin levels on protein expression in whole cell lysates using in an inducible Parkin expression system in HEK293 cells, and also to isolate potential interactants of Parkin using tandem affinity purification and MS. Nine proteins were significantly differentially expressed (+/-2-fold change; p<0.05) using 2-DE analysis. MS revealed the identity of these proteins to be ACAT2, HNRNPK, HSPD1, PGK1, PRDX6, VCL, VIM, TPI1, and IMPDH2. The first seven of these were reduced in expression. Western blot analysis confirmed the reduction in one of these proteins (HNRNPK), and that its levels were dependent on 26S proteasomal activity. Tandem affinity purification/MS revealed 14 potential interactants of Parkin; CKB, DBT, HSPD1, HSPA9, LRPPRC, NDUFS2, PRDX6, SLC25A5, TPI1, UCHL1, UQCRC1, VCL, YWHAZ, YWHAE. Nine of these are directly involved in mitochondrial energy metabolism and glycolysis; four were also identified in the 2-DE study (HSP60, PRDX6, TPI1, and VCL). This study provides further evidence for a role for Parkin in regulating mitochondrial activity within cells.


Subject(s)
Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Proteomics/methods , Ubiquitin-Protein Ligases/metabolism , Cell Line , Chaperonin 60/metabolism , Electrophoresis, Gel, Two-Dimensional , Heterogeneous-Nuclear Ribonucleoprotein K , Humans , Mass Spectrometry , Protein Interaction Mapping , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Reproducibility of Results , Ribonucleoproteins/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/biosynthesis , Ubiquitin-Protein Ligases/genetics
15.
Clin Cancer Res ; 14(6): 1775-81, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18347179

ABSTRACT

PURPOSE: Conventional renal cell carcinoma (RCC) has a variable natural history, and determining individual prognosis is important to guide management. We have examined the prognostic significance of a large number of hematologic and biochemical variables, as well as traditional tumor-related factors in patients with RCC. EXPERIMENTAL DESIGN: Patients undergoing nephrectomy for newly diagnosed RCC between September 1998 and March 2005 were invited to participate. Clinical, pathologic, and laboratory data were recorded in each case, and immunophenotyping was carried out on a subset of patients. A planned subset analysis of patients presenting with N0M0 disease was done. RESULTS: Two hundred twelve patients with RCC formed the study population. In addition to tumor-related factors, multivariate analyses revealed preoperative serum sodium concentration to be independently and significantly associated with overall survival and disease-free survival when considered as both a continuous variable and when dichotomized to above and below the median value [139 mmol/L; reference range 135-145 mmol/L, hazard ratio 0.44, 95% confidence interval (95% CI) 0.22-0.88, P = 0.014]. Five-year overall survival estimates for patients above and below the median serum sodium were 67.6% (95% CI 54.2-80.9) and 44.3% (95% CI 32.8-55.8), respectively. These findings persisted in the N0M0 subgroup analysis. CONCLUSIONS: We have confirmed the prognostic value of traditional tumor-related factors but, to our knowledge, these are the first data to show that low preoperative sodium concentration may be an important factor associated with reduced survival in patients with RCC, suggesting that serum sodium should be considered with established prognostic variables in modeling survival in RCC.


Subject(s)
Carcinoma, Renal Cell/diagnosis , Carcinoma, Renal Cell/mortality , Kidney Neoplasms/diagnosis , Kidney Neoplasms/mortality , Sodium/blood , Adult , Aged , Aged, 80 and over , Algorithms , Carcinoma, Renal Cell/blood , Carcinoma, Renal Cell/surgery , Cross-Sectional Studies , Female , Humans , Kidney Neoplasms/blood , Kidney Neoplasms/surgery , Male , Middle Aged , Models, Theoretical , Multivariate Analysis , Prognosis , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...