Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Cancer Res ; 81(4): 847-859, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33509944

ABSTRACT

Triple-negative breast cancers (TNBC) are resistant to standard-of-care chemotherapy and lack known targetable driver gene alterations. Identification of novel drivers could aid the discovery of new treatment strategies for this hard-to-treat patient population, yet studies using high-throughput and accurate models to define the functions of driver genes in TNBC to date have been limited. Here, we employed unbiased functional genomics screening of the 200 most frequently mutated genes in breast cancer, using spheroid cultures to model in vivo-like conditions, and identified the histone acetyltransferase CREBBP as a novel tumor suppressor in TNBC. CREBBP protein expression in patient tumor samples was absent in 8% of TNBCs and at a high frequency in other tumors, including squamous lung cancer, where CREBBP-inactivating mutations are common. In TNBC, CREBBP alterations were associated with higher genomic heterogeneity and poorer patient survival and resulted in upregulation and dependency on a FOXM1 proliferative program. Targeting FOXM1-driven proliferation indirectly with clinical CDK4/6 inhibitors (CDK4/6i) selectively impaired growth in spheroids, cell line xenografts, and patient-derived models from multiple tumor types with CREBBP mutations or loss of protein expression. In conclusion, we have identified CREBBP as a novel driver in aggressive TNBC and identified an associated genetic vulnerability in tumor cells with alterations in CREBBP and provide a preclinical rationale for assessing CREBBP alterations as a biomarker of CDK4/6i response in a new patient population. SIGNIFICANCE: This study demonstrates that CREBBP genomic alterations drive aggressive TNBC, lung cancer, and lymphomas and may be selectively treated with clinical CDK4/6 inhibitors.


Subject(s)
CREB-Binding Protein/physiology , Carcinogenesis/genetics , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Animals , CREB-Binding Protein/genetics , Cell Proliferation/genetics , Cells, Cultured , Drug Screening Assays, Antitumor/methods , Female , Genomics/methods , HCT116 Cells , HEK293 Cells , Humans , Mice , Mice, Inbred NOD , Mice, Nude , Molecular Targeted Therapy , Mutation , Neoplasm Invasiveness , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Xenograft Model Antitumor Assays
2.
Cancer Cell ; 37(1): 85-103.e9, 2020 01 13.
Article in English | MEDLINE | ID: mdl-31935375

ABSTRACT

Despite substantial clinical benefit of targeted and immune checkpoint blockade-based therapies in melanoma, resistance inevitably develops. We show cytoskeletal remodeling and changes in expression and activity of ROCK-myosin II pathway during acquisition of resistance to MAPK inhibitors. MAPK regulates myosin II activity, but after initial therapy response, drug-resistant clones restore myosin II activity to increase survival. High ROCK-myosin II activity correlates with aggressiveness, identifying targeted therapy- and immunotherapy-resistant melanomas. Survival of resistant cells is myosin II dependent, regardless of the therapy. ROCK-myosin II ablation specifically kills resistant cells via intrinsic lethal reactive oxygen species and unresolved DNA damage and limits extrinsic myeloid and lymphoid immunosuppression. Efficacy of targeted therapies and immunotherapies can be improved by combination with ROCK inhibitors.


Subject(s)
Cytoskeleton/metabolism , Drug Resistance, Neoplasm , Melanoma/metabolism , Myosin Type II/metabolism , Animals , B7-H1 Antigen/metabolism , Cell Cycle , Cell Line, Tumor , DNA Damage , Female , Humans , Immunotherapy , MAP Kinase Signaling System , Male , Melanoma/immunology , Melanoma/therapy , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Oxidative Stress , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Reactive Oxygen Species , T-Lymphocytes, Regulatory/immunology , Treatment Outcome , Tumor Microenvironment/immunology , rho-Associated Kinases/metabolism
3.
J Immunother Cancer ; 7(1): 101, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30982469

ABSTRACT

BACKGROUND: The T cell bispecific antibody cibisatamab (CEA-TCB) binds Carcino-Embryonic Antigen (CEA) on cancer cells and CD3 on T cells, which triggers T cell killing of cancer cell lines expressing moderate to high levels of CEA at the cell surface. Patient derived colorectal cancer organoids (PDOs) may more accurately represent patient tumors than established cell lines which potentially enables more detailed insights into mechanisms of cibisatamab resistance and sensitivity. METHODS: We established PDOs from multidrug-resistant metastatic CRCs. CEA expression of PDOs was determined by FACS and sensitivity to cibisatamab immunotherapy was assessed by co-culture of PDOs and allogeneic CD8 T cells. RESULTS: PDOs could be categorized into 3 groups based on CEA cell-surface expression: CEAhi (n = 3), CEAlo (n = 1) and CEAmixed PDOs (n = 4), that stably maintained populations of CEAhi and CEAlo cells, which has not previously been described in CRC cell lines. CEAhi PDOs were sensitive whereas CEAlo PDOs showed resistance to cibisatamab. PDOs with mixed expression showed low sensitivity to cibisatamab, suggesting that CEAlo cells maintain cancer cell growth. Culture of FACS-sorted CEAhi and CEAlo cells from PDOs with mixed CEA expression demonstrated high plasticity of CEA expression, contributing to resistance acquisition through CEA antigen loss. RNA-sequencing revealed increased WNT/ß-catenin pathway activity in CEAlo cells. Cell surface CEA expression was up-regulated by inhibitors of the WNT/ß-catenin pathway. CONCLUSIONS: Based on these preclinical findings, heterogeneity and plasticity of CEA expression appear to confer low cibisatamab sensitivity in PDOs, supporting further clinical evaluation of their predictive effect in CRC. Pharmacological inhibition of the WNT/ß-catenin pathway may be a rational combination to sensitize CRCs to cibisatamab. Our novel PDO and T cell co-culture immunotherapy models enable pre-clinical discovery of candidate biomarkers and combination therapies that may inform and accelerate the development of immuno-oncology agents in the clinic.


Subject(s)
Antibodies, Bispecific/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Carcinoembryonic Antigen/genetics , Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm/genetics , Antibodies, Bispecific/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , CD8-Positive T-Lymphocytes , Coculture Techniques , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Drug Screening Assays, Antitumor , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/genetics , Gene Expression Regulation, Neoplastic , Genetic Heterogeneity , Humans , Tissue Culture Techniques
4.
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
5.
Br J Cancer ; 117(1): 113-123, 2017 Jun 27.
Article in English | MEDLINE | ID: mdl-28535155

ABSTRACT

BACKGROUND: Elevated APOBEC3B expression in tumours correlates with a kataegic pattern of localised hypermutation. We assessed the cellular phenotypes associated with high-level APOBEC3B expression and the influence of p53 status on these phenotypes using an isogenic system. METHODS: We used RNA interference of p53 in cells with inducible APOBEC3B and assessed DNA damage response (DDR) biomarkers. The mutational effects of APOBEC3B were assessed using whole-genome sequencing. In vitro small-molecule inhibitor sensitivity profiling was used to identify candidate therapeutic vulnerabilities. RESULTS: Although APOBEC3B expression increased the incorporation of genomic uracil, invoked DDR biomarkers and caused cell cycle arrest, inactivation of p53 circumvented APOBEC3B-induced cell cycle arrest without reversing the increase in genomic uracil or DDR biomarkers. The continued expression of APOBEC3B in p53-defective cells not only caused a kataegic mutational signature but also caused hypersensitivity to small-molecule DDR inhibitors (ATR, CHEK1, CHEK2, PARP, WEE1 inhibitors) as well as cisplatin/ATR inhibitor and ATR/PARP inhibitor combinations. CONCLUSIONS: Although loss of p53 might allow tumour cells to tolerate elevated APOBEC3B expression, continued expression of this enzyme might impart a number of therapeutic vulnerabilities upon tumour cells.


Subject(s)
Cell Cycle Checkpoints/genetics , Cytidine Deaminase/genetics , DNA Damage/genetics , Gene Expression Regulation, Neoplastic , Minor Histocompatibility Antigens/genetics , Tumor Suppressor Protein p53/genetics , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Blotting, Western , CRISPR-Cas Systems , Cell Cycle Checkpoints/drug effects , Cell Cycle Proteins/antagonists & inhibitors , Cell Line , Checkpoint Kinase 1/antagonists & inhibitors , Checkpoint Kinase 2/antagonists & inhibitors , Cisplatin/pharmacology , Cytidine Deaminase/metabolism , DNA Damage/drug effects , G2 Phase Cell Cycle Checkpoints , Gene Knockout Techniques , HEK293 Cells , Humans , Minor Histocompatibility Antigens/metabolism , Mutation , Nuclear Proteins/antagonists & inhibitors , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , RNA Interference , Uracil/metabolism
6.
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
7.
Cold Spring Harb Protoc ; 2016(4): pdb.prot087387, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-27037070

ABSTRACT

Fluorescence-activated cell sorting (FACS) is a laser-based, biophysical technology that allows simultaneous multiparametric analysis. For the analysis of dying cells, fluorescently labeled Annexin V (Annexin V(FITC)) and propidium iodide (PI) are the most commonly used reagents. Instead of PI, 4',6-diamidino-2-phenylindole (DAPI) can also be used. DAPI is a fluorescent stain that binds strongly to A-T-rich regions in DNA. DAPI and PI only inefficiently pass through an intact cell membrane and, therefore, preferentially stain dead cells. DAPI can be combined with Annexin V(FITC)and the potentiometric fluorescent dye, tetramethylrhodamine methyl ester (TMRM), which measures mitochondrial permeability transition and mitochondrial membrane depolarization. TMRM is a cell-permeable fluorescent dye that is sequestered to active mitochondria, and hence labels live cells. On apoptosis or necroptosis the TMRM signal is lost. The advantage of using Annexin V(FITC)/DAPI/TMRM is that the entire cell population is labeled, and it is easy to distinguish living (TMRM + /Annexin V(FITC)-/DAPI-) from dying or dead cells (apoptosis: TMRM-/Annexin V(FITC)+ /DAPI-; necrosis: TMRM-/Annexin V(FITC)+ /DAPI+). This is important because cell debris (fluorescent negative particles) must be avoided to establish the correct parameters for the FACS analysis, otherwise incorrect statistical values will be obtained. To obtain information on the cell concentration or absolute cell counts in a sample, it is recommended to add an internal microsphere counting standard to the flow cytrometric sample. This protocol describes the FACS analysis of cell death in HT1080 and L929 cells, but it can be readily adapted to other cell types of interest.


Subject(s)
Apoptosis , Flow Cytometry/methods , Necrosis , Staining and Labeling/methods , Cell Membrane/physiology , Fluorescent Dyes/metabolism , Mitochondrial Membranes/physiology , Permeability
8.
Cold Spring Harb Protoc ; 2016(4): pdb.top070375, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-27037077

ABSTRACT

The processes by which cells die are as tightly regulated as those that govern cell growth and proliferation. Recent studies of the molecular pathways that regulate and execute cell death have uncovered a plethora of signaling cascades that lead to distinct modes of cell death, including "apoptosis," "necrosis," "autophagic cell death," and "mitotic catastrophe." Cells can readily switch from one form of death to another; therefore, it is vital to have the ability to monitor the form of death that cells are undergoing. A number of techniques are available that allow the detection of cell death and when combined with either knockdown approaches or inhibitors of specific signaling pathways, such as caspase or RIP kinase pathways, they allow the rapid dissection of divergent cell death pathways. However, techniques that reveal the end point of cell death cannot reconstruct the sequence of events that have led to death; therefore, they need to be complemented with methods that can distinguish all forms of cell death. Apoptotic cells frequently undergo secondary necrosis under in vitro culture conditions; therefore, novel methods relying on high-throughput time-lapse fluorescence video microscopy are necessary to provide temporal resolution to cell death events. Further, visualizing the assembly of multiprotein signaling hubs that can execute apoptosis or necroptosis helps to explore the underlying processes. Here we introduce a suite of techniques that reliably distinguish necrosis from apoptosis and secondary necrosis, and that enable investigation of signaling platforms capable of instructing apoptosis or necroptosis.


Subject(s)
Apoptosis , Cytological Techniques/methods , Necrosis , Signal Transduction , Microscopy, Fluorescence/methods , Microscopy, Video/methods , Time-Lapse Imaging/methods
9.
Cold Spring Harb Protoc ; 2016(3): pdb.prot087395, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26933245

ABSTRACT

The best approach to distinguish between necrosis and apoptosis is time-lapse video microscopy. This technique enables a biological process to be photographed at regular intervals over a period, which may last from a few hours to several days, and can be applied to cells in culture or in vivo. We have established two time-lapse microscopy methods based on different ways of calculating cell death: semiautomated and automated. In the semiautomated approach, cell death can be visualized by staining with combinations of Alexa Fluor 647-conjugated Annexin V and Sytox Green (SG), or Annexin V(FITC) and Propidium iodide (PI). The automated method is similar except that all cells are labeled with dyes. This allows faster quantification of data. To this end Cell Tracker Green is used to label all cells at time zero in combination with PI and Alexa Fluor 647-conjugated Annexin V. Necrotic cell death is accompanied by either simultaneous labeling with Annexin V and PI or SG (double-positive), or direct PI or SG staining. Additionally, necrotic cells display characteristic morphology, such as cytoplasmic swelling. In contrast to necrosis where membrane permeabilization is an early event, cells that die by apoptosis lose their membrane permeability relatively late. Therefore, the time between Annexin V staining and PI or SG uptake (double-positive) can be used to distinguish necrosis from apoptosis. This protocol describes the analysis of cell death by time-lapse imaging of HT1080 and L929 cells stained with these dyes, but it can be readily adapted to other cell types of interest.


Subject(s)
Cell Death , Microscopy, Video/methods , Time-Lapse Imaging/methods , Animals , Cell Line , Fibroblasts , Fluorescent Dyes/analysis , Humans , Mice , Staining and Labeling/methods
10.
Methods Mol Biol ; 1004: 17-29, 2013.
Article in English | MEDLINE | ID: mdl-23733566

ABSTRACT

The processes of dying are as tightly regulated as those of growth and proliferation. Recent work into the molecular pathways that regulate and execute cell death have uncovered a plethora of signalling cascades that lead to distinct modes of cell death, including "apoptosis," "necrosis," "autophagic cell death," and "mitotic catastrophe." Given that cells can readily switch from one form of death to another, it is vital to carefully monitor the form of death under investigation. Particularly, end-point techniques are intrinsically unsuitable for assessing apoptosis versus necrosis, as they cannot reconstruct the sequence of events that have led to cell death. Since apoptotic cells frequently undergo secondary necrosis under in vitro culture conditions, novel methods relying on high-throughput time-lapse fluorescence video microscopy have been developed. Here we describe the use of this technique to reliably distinguish necrosis from apoptosis and secondary necrosis.


Subject(s)
Necrosis/pathology , Time-Lapse Imaging/methods , Apoptosis , Cell Line , Gene Knockdown Techniques , Humans , Microscopy, Video , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Statistics as Topic
11.
PLoS One ; 8(4): e61520, 2013.
Article in English | MEDLINE | ID: mdl-23634208

ABSTRACT

Genetic perturbation screens have the potential to dissect a wide range of cellular phenotypes. Such screens have historically been difficult in diploid mammalian cells. The recent derivation of haploid embryonic stem cells provides an opportunity to cause loss of function mutants with a random mutagen in a mammalian cell with a normal genetic background. We describe an approach to genetic screens that exploits the highly active piggyBac transposon in haploid mammalian cells. As an example of haploid transposon (HTP) screening, we apply this approach to identifying determinants of cancer drug toxicity and resistance. In a screen for 6-thioguanine resistance we recovered components of the DNA mismatch repair pathway, a known requirement for toxicity. In a further screen for resistance to the clinical poly(ADP-ribose) polymerase (PARP) inhibitor olaparib we recovered multiple Parp1 mutants. Our results show that olaparib toxicity to normal cells is mediated predominantly via Parp1, and suggest that the clinical side effects of olaparib may be on target. The transposon mutant libraries are stable and can be readily reused to screen other drugs. The screening protocol described has several advantages over other methods such as RNA interference: it is rapid and low cost, and mutations can be easily reverted to establish causality.


Subject(s)
DNA Transposable Elements/drug effects , DNA Transposable Elements/genetics , Genetic Testing , Haploidy , Phthalazines/pharmacology , Piperazines/pharmacology , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Animals , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Humans , Mice , Mutation , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerase Inhibitors
12.
Cancer Cell ; 20(2): 229-45, 2011 Aug 16.
Article in English | MEDLINE | ID: mdl-21840487

ABSTRACT

Proinflammatory cytokines are frequently observed in the tumor microenvironment, and chronic inflammation is involved in cancer initiation and progression. We show that cytokine signaling through the receptor subunit GP130-IL6ST and the kinase JAK1 generates actomyosin contractility through Rho-kinase dependent signaling. This pathway generates contractile force in stromal fibroblasts to remodel the extracellular matrix to create tracks for collective migration of squamous carcinoma cells and provides the high levels of actomyosin contractility required for migration of individual melanoma cells in the rounded, "amoeboid" mode. Thus, cytokine signaling can generate actomyosin contractility in both stroma and tumor cells. Strikingly, actomyosin contractility itself positively modulates activity of the transcription factor STAT3 downstream of JAK1, demonstrating positive feedback within the signaling network.


Subject(s)
Actomyosin/metabolism , Janus Kinase 1/metabolism , Neoplasms/metabolism , Signal Transduction , Stromal Cells/metabolism , rho-Associated Kinases/metabolism , Cell Movement , Humans , Melanoma/metabolism , Melanoma/pathology , Neoplasms/pathology , STAT3 Transcription Factor/metabolism , Stromal Cells/pathology
13.
Mol Cell ; 43(3): 432-48, 2011 Aug 05.
Article in English | MEDLINE | ID: mdl-21737329

ABSTRACT

A better understanding of the mechanisms through which anticancer drugs exert their effects is essential to improve combination therapies. While studying how genotoxic stress kills cancer cells, we discovered a large ∼2MDa cell death-inducing platform, referred to as "Ripoptosome." It contains the core components RIP1, FADD, and caspase-8, and assembles in response to genotoxic stress-induced depletion of XIAP, cIAP1 and cIAP2. Importantly, it forms independently of TNF, CD95L/FASL, TRAIL, death-receptors, and mitochondrial pathways. It also forms upon Smac-mimetic (SM) treatment without involvement of autocrine TNF. Ripoptosome assembly requires RIP1's kinase activity and can stimulate caspase-8-mediated apoptosis as well as caspase-independent necrosis. It is negatively regulated by FLIP, cIAP1, cIAP2, and XIAP. Mechanistically, IAPs target components of this complex for ubiquitylation and inactivation. Moreover, we find that etoposide-stimulated Ripoptosome formation converts proinflammatory cytokines into prodeath signals. Together, our observations shed new light on fundamental mechanisms by which chemotherapeutics may kill cancer cells.


Subject(s)
Apoptosis/physiology , Caspase 8/physiology , DNA Damage , Fas-Associated Death Domain Protein/physiology , Inhibitor of Apoptosis Proteins/genetics , Nuclear Pore Complex Proteins/physiology , RNA-Binding Proteins/physiology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , CASP8 and FADD-Like Apoptosis Regulating Protein/physiology , Caspase 8/chemistry , Caspase 8/metabolism , Cell Line, Tumor , Enzyme Activation , Etoposide/pharmacology , Fas-Associated Death Domain Protein/chemistry , Fas-Associated Death Domain Protein/metabolism , Humans , Inhibitor of Apoptosis Proteins/physiology , Ligands , Mitochondria/metabolism , Nuclear Pore Complex Proteins/chemistry , Nuclear Pore Complex Proteins/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Signal Transduction
14.
Nat Cell Biol ; 13(7): 819-26, 2011 Jun 19.
Article in English | MEDLINE | ID: mdl-21685891

ABSTRACT

Individual tumour cells move in three-dimensional environments with either a rounded or an elongated 'mesenchymal' morphology. These two modes of movement are tightly regulated by Rho family GTPases: elongated movement requires activation of Rac1, whereas rounded/amoeboid movement engages specific Cdc42 and Rho signalling pathways. In siRNA screens targeting the genes encoding guanine nucleotide exchange factors (GEFs), we found that the Ras GEF RasGRF2 regulates conversion between elongated- and rounded-type movement. RasGRF2 suppresses rounded movement by inhibiting the activation of Cdc42 independently of its capacity to activate Ras. RasGRF2 and RasGRF1 directly bind to Cdc42, outcompeting Cdc42 GEFs, thereby preventing Cdc42 activation. By this mechanism, RasGRFs regulate other Cdc42-mediated cellular processes such as the formation of actin spikes, transformation and invasion in vitro and in vivo. These results demonstrate a role for RasGRF GEFs as negative regulators of Cdc42 activation.


Subject(s)
Cell Movement , Cell Shape , Cell Transformation, Neoplastic/metabolism , Cytoskeleton/metabolism , Melanoma/enzymology , cdc42 GTP-Binding Protein/metabolism , ras Guanine Nucleotide Exchange Factors/metabolism , Animals , COS Cells , Cell Transformation, Neoplastic/genetics , Chlorocebus aethiops , Down-Regulation , Enzyme Activation , HeLa Cells , Humans , Jurkat Cells , Melanoma/genetics , Melanoma/pathology , Mice , Microscopy, Video , Mutation , NIH 3T3 Cells , Neoplasm Invasiveness , Protein Binding , Proto-Oncogene Proteins c-vav/genetics , Proto-Oncogene Proteins c-vav/metabolism , RNA Interference , Recombinant Fusion Proteins/metabolism , Time Factors , Transfection , cdc42 GTP-Binding Protein/genetics , ras Guanine Nucleotide Exchange Factors/genetics , ras-GRF1/metabolism
15.
Biotechnol Lett ; 27(13): 919-26, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16091887

ABSTRACT

Multi-parameter flow cytometry was used to monitor the formation of promegapoietin (PMP) inclusion bodies during a high cell density Escherichia coli fed-batch fermentation process. Inclusion bodies were labelled with a primary antibody and then with a secondary fluorescent antibody. Using this method it was possible to detect PMP inclusion body formation with a high specificity and it was possible to monitor the increased accumulation of the protein with process time (6-48 mg PMP/g CDW) whilst highlighting population heterogeneity.


Subject(s)
Escherichia coli/metabolism , Flow Cytometry/methods , Inclusion Bodies/metabolism , Bioreactors/microbiology , Chromatography, High Pressure Liquid , Escherichia coli/growth & development , Fermentation , Fluorescent Dyes/chemistry , Humans , Inclusion Bodies/chemistry , Interleukin-3 , Microscopy, Fluorescence , Receptors, Interleukin-3/chemistry , Receptors, Interleukin-3/genetics , Receptors, Interleukin-3/metabolism , Recombinant Fusion Proteins , Recombinant Proteins/metabolism , Thrombopoietin/chemistry , Thrombopoietin/genetics , Thrombopoietin/metabolism
16.
Biotechnol Lett ; 26(19): 1533-9, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15604793

ABSTRACT

In Escherichia coli fermentation processes, a drastic drop in viable cell count as measured by the number of colony forming units per ml (c.f.u. ml(-1)) is often observed. This phenomenon was investigated in a process for the production of the recombinant fusion protein, promegapoietin (PMP). After induction, the number of c.f.u. ml(-1) dropped to approximately 10% of its maximum though the biomass concentration continued to increase. Flow cytometric analysis of viability and intracellular concentration of PMP showed that almost all cells were alive and contributed to the production. Thus, the drop in the number of c.f.u. ml(-1) probably reflects a loss of cell division capability rather than cell death.


Subject(s)
Colony Count, Microbial/methods , Escherichia coli/cytology , Escherichia coli/physiology , Mitosis/physiology , Protein Engineering/methods , Receptors, Interleukin-3/biosynthesis , Thrombopoietin/biosynthesis , Apoptosis/physiology , Bioreactors/microbiology , Cell Aggregation/physiology , Cell Survival/physiology , Fermentation/physiology , Flow Cytometry/methods , Interleukin-3 , Receptors, Interleukin-3/genetics , Recombinant Fusion Proteins/biosynthesis , Thrombopoietin/genetics
17.
Plant Mol Biol ; 52(2): 259-71, 2003 May.
Article in English | MEDLINE | ID: mdl-12856934

ABSTRACT

Mitochondrial precursor proteins synthesized in rabbit reticulocyte lysate (RRL) are readily imported into mitochondria, whereas the same precursors synthesized in wheat germ extract (WGE) fail to be imported. We have investigated factors that render import incompetence from WGE. A precursor that does not require addition of extramitochondrial ATP for import, the F(A)d ATP synthase subunit, is imported from WGE. Import of chimeric constructs between precursors of the F(A)d protein and alternative oxidase (AOX) with switched presequences revealed that the mature domain of the F(A)d precursor defines the import competence in WGE as only the construct containing the presequence of AOX and mature portion of F(A)d (pAOX-mF(A)d) could be imported. Import competence of F(A)d and pAOX-mF(A)d correlated with solubility of these precursors in WGE, however, solubilization of import-incompetent precursors with urea did not restore import competence. Addition of RRL to WGE-synthesized precursors did not stimulate import but addition of WGE to the RRL-synthesized precursors or to the over-expressed mitochondrial precursor derived from the F1beta ATP synthase precursor inhibited import into mitochondria. The dual-targeted glutathione reductase precursor synthesized in WGE was imported into chloroplasts, but not into mitochondria. Antibodies against the 14-3-3 guidance complex characterized for chloroplast targeting were able to immunoprecipitate all of the precursors tested except the F(A)d ATP synthase precursor. Our results point to the conclusion that the import incompetence of WGE-synthesized mitochondrial precursors is not presequence dependent and is a result of interaction of WGE inhibitory factors with the mature portion of precursor proteins.


Subject(s)
Mitochondrial Proteins/genetics , Protein Biosynthesis/genetics , Transcription, Genetic/genetics , 14-3-3 Proteins , Animals , Cell-Free System/metabolism , Chloroplasts/drug effects , Chloroplasts/metabolism , Glutathione Reductase/genetics , Glutathione Reductase/metabolism , HSP70 Heat-Shock Proteins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Plant Extracts/genetics , Plant Extracts/pharmacology , Precipitin Tests , Protein Binding , Protein Folding , Protein Precursors/chemistry , Protein Precursors/genetics , Protein Precursors/metabolism , Protein Transport/drug effects , Rabbits , Reticulocytes/metabolism , Solubility , Triticum/genetics , Tyrosine 3-Monooxygenase/metabolism
18.
Microb Cell Fact ; 2(1): 6, 2003 Jun 18.
Article in English | MEDLINE | ID: mdl-12871597

ABSTRACT

BACKGROUND: A temperature limited fed-batch (TLFB) technique is described and used for Pichia pastoris Mut+ strain cultures and compared with the traditional methanol limited fed-batch (MLFB) technique. A recombinant fusion protein composed of a cellulose-binding module (CBM) from Neocallimastix patriciarum cellulase 6A and lipase B from Candida antarctica (CALB), was produced and secreted by this strain. RESULTS: A protein concentration of about 1 g L-1 was produced in the MLFB process. However, this product was considerably degraded by protease(s). By applying the TLFB process, the yield was increased to 2 g L-1 full-length product and no proteolytic degradation was observed. Flow cytometry analysis showed that the percentage of dead cells increased rapidly during the initial methanol feed phase in the MLFB process and reached a maximum of about 12% after about 40-70 hours of methanol feeding. In the TLFB process, cell death rate was low and constant and reached 4% dead cells at the end of cultivation (about 150 hours methanol feeding time). The lower cell death rate in the TLFB correlated with a lower protease activity in the culture supernatant. The specific alcohol oxidase (AOX) activity in the TLFB process was 3.5 times higher than in the MLFB process. CONCLUSION: Three mechanisms that may contribute to the much higher accumulation of product in the TLFB process are: 1) reduced proteolysis due to lower temperature, 2) reduced proteolysis due to lower cell death and protease release to the medium, 3) increased synthesis rate due to higher AOX activity.

SELECTION OF CITATIONS
SEARCH DETAIL
...