Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
EMBO Rep ; 23(12): e54978, 2022 12 06.
Article in English | MEDLINE | ID: mdl-36321428

ABSTRACT

Mitochondrial Ca2+ ions are crucial regulators of bioenergetics and cell death pathways. Mitochondrial Ca2+ content and cytosolic Ca2+ homeostasis strictly depend on Ca2+ transporters. In recent decades, the major players responsible for mitochondrial Ca2+ uptake and release have been identified, except the mitochondrial Ca2+ /H+ exchanger (CHE). Originally identified as the mitochondrial K+ /H+ exchanger, LETM1 was also considered as a candidate for the mitochondrial CHE. Defining the mitochondrial interactome of LETM1, we identify TMBIM5/MICS1, the only mitochondrial member of the TMBIM family, and validate the physical interaction of TMBIM5 and LETM1. Cell-based and cell-free biochemical assays demonstrate the absence or greatly reduced Na+ -independent mitochondrial Ca2+ release in TMBIM5 knockout or pH-sensing site mutants, respectively, and pH-dependent Ca2+ transport by recombinant TMBIM5. Taken together, we demonstrate that TMBIM5, but not LETM1, is the long-sought mitochondrial CHE, involved in setting and regulating the mitochondrial proton gradient. This finding provides the final piece of the puzzle of mitochondrial Ca2+ transporters and opens the door to exploring its importance in health and disease, and to developing drugs modulating Ca2+ exchange.


Subject(s)
Antiporters , Protons , Antiporters/genetics
2.
Nat Struct Mol Biol ; 28(2): 190-201, 2021 02.
Article in English | MEDLINE | ID: mdl-33479542

ABSTRACT

NUP98 fusion proteins cause leukemia via unknown molecular mechanisms. All NUP98 fusion proteins share an intrinsically disordered region (IDR) in the NUP98 N terminus, featuring repeats of phenylalanine-glycine (FG), and C-terminal fusion partners often function in gene control. We investigated whether mechanisms of oncogenic transformation by NUP98 fusion proteins are hardwired in their protein interactomes. Affinity purification coupled to mass spectrometry (MS) and confocal imaging of five NUP98 fusion proteins expressed in human leukemia cells revealed that shared interactors were enriched for proteins involved in biomolecular condensation and that they colocalized with NUP98 fusion proteins in nuclear puncta. We developed biotinylated isoxazole-mediated condensome MS (biCon-MS) to show that NUP98 fusion proteins alter the global composition of biomolecular condensates. An artificial FG-repeat-containing fusion protein phenocopied the nuclear localization patterns of NUP98 fusion proteins and their capability to drive oncogenic gene expression programs. Thus, we propose that IDR-containing fusion proteins combine biomolecular condensation with transcriptional control to induce cancer.


Subject(s)
Cell Nucleus/metabolism , Homeodomain Proteins , Leukemia , Nuclear Pore Complex Proteins , Oncogene Proteins, Fusion , Animals , Gene Expression , Gene Expression Regulation, Leukemic , HEK293 Cells , HL-60 Cells , Homeodomain Proteins/chemistry , Homeodomain Proteins/physiology , Humans , Leukemia/metabolism , Leukemia/pathology , Mice , NIH 3T3 Cells , Nuclear Pore Complex Proteins/chemistry , Nuclear Pore Complex Proteins/physiology , Oncogene Proteins, Fusion/chemistry , Oncogene Proteins, Fusion/physiology
3.
Nat Genet ; 51(9): 1399-1410, 2019 09.
Article in English | MEDLINE | ID: mdl-31427792

ABSTRACT

Aberrations in genes coding for subunits of the BRG1/BRM associated factor (BAF) chromatin remodeling complexes are highly abundant in human cancers. Currently, it is not understood how these mostly loss-of-function mutations contribute to cancer development and how they can be targeted therapeutically. The cancer-type-specific occurrence patterns of certain subunit mutations suggest subunit-specific effects on BAF complex function, possibly by the formation of aberrant residual complexes. Here, we systematically characterize the effects of individual subunit loss on complex composition, chromatin accessibility and gene expression in a panel of knockout cell lines deficient for 22 BAF subunits. We observe strong, specific and sometimes discordant alterations dependent on the targeted subunit and show that these explain intracomplex codependencies, including the synthetic lethal interactions SMARCA4-ARID2, SMARCA4-ACTB and SMARCC1-SMARCC2. These data provide insights into the role of different BAF subcomplexes in genome-wide chromatin organization and suggest approaches to therapeutically target BAF-mutant cancers.


Subject(s)
Chromatin Assembly and Disassembly/genetics , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Mutation , Neoplasms/pathology , Nuclear Proteins/metabolism , Transcription Factors/metabolism , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Humans , Neoplasms/genetics , Neoplasms/metabolism , Nuclear Proteins/genetics , Transcription Factors/genetics , Transcriptome
4.
Nat Genet ; 51(6): 990-998, 2019 06.
Article in English | MEDLINE | ID: mdl-31133746

ABSTRACT

The histone acetyl reader bromodomain-containing protein 4 (BRD4) is an important regulator of chromatin structure and transcription, yet factors modulating its activity have remained elusive. Here we describe two complementary screens for genetic and physical interactors of BRD4, which converge on the folate pathway enzyme MTHFD1 (methylenetetrahydrofolate dehydrogenase, cyclohydrolase and formyltetrahydrofolate synthetase 1). We show that a fraction of MTHFD1 resides in the nucleus, where it is recruited to distinct genomic loci by direct interaction with BRD4. Inhibition of either BRD4 or MTHFD1 results in similar changes in nuclear metabolite composition and gene expression; pharmacological inhibitors of the two pathways synergize to impair cancer cell viability in vitro and in vivo. Our finding that MTHFD1 and other metabolic enzymes are chromatin associated suggests a direct role for nuclear metabolism in the control of gene expression.


Subject(s)
Folic Acid/metabolism , Gene Expression Regulation , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Minor Histocompatibility Antigens/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Cell Cycle Proteins , Cell Line, Tumor , Cell Nucleus/metabolism , Chromatin/genetics , Gene Knockout Techniques , Humans , Loss of Function Mutation , Protein Binding , Protein Interaction Mapping , Protein Interaction Maps , Protein Transport , Signal Transduction , Transcription, Genetic
5.
Cancer Cell ; 35(1): 125-139.e9, 2019 01 14.
Article in English | MEDLINE | ID: mdl-30645971

ABSTRACT

The marsupial Tasmanian devil (Sarcophilus harrisii) faces extinction due to transmissible devil facial tumor disease (DFTD). To unveil the molecular underpinnings of this transmissible cancer, we combined pharmacological screens with an integrated systems-biology characterization. Sensitivity to inhibitors of ERBB tyrosine kinases correlated with their overexpression. Proteomic and DNA methylation analyses revealed tumor-specific signatures linked to the evolutionary conserved oncogenic STAT3. ERBB inhibition blocked phosphorylation of STAT3 and arrested cancer cells. Pharmacological blockade of ERBB or STAT3 prevented tumor growth in xenograft models and restored MHC class I expression. This link between the hyperactive ERBB-STAT3 axis and major histocompatibility complex class I-mediated tumor immunosurveillance provides mechanistic insights into horizontal transmissibility and puts forward a dual chemo-immunotherapeutic strategy to save Tasmanian devils from DFTD. VIDEO ABSTRACT.


Subject(s)
ErbB Receptors/metabolism , Facial Neoplasms/drug therapy , Facial Neoplasms/veterinary , Proteomics/methods , STAT3 Transcription Factor/metabolism , Small Molecule Libraries/administration & dosage , Animals , DNA Methylation , Drug Screening Assays, Antitumor , Facial Neoplasms/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Histocompatibility Antigens Class I/metabolism , Marsupialia , Mice , Phosphorylation , Signal Transduction , Small Molecule Libraries/pharmacology , Xenograft Model Antitumor Assays
6.
Science ; 362(6419): 1171-1177, 2018 12 07.
Article in English | MEDLINE | ID: mdl-30442766

ABSTRACT

In genetic screens aimed at understanding drug resistance mechanisms in chronic myeloid leukemia cells, inactivation of the cullin 3 adapter protein-encoding leucine zipper-like transcription regulator 1 (LZTR1) gene led to enhanced mitogen-activated protein kinase (MAPK) pathway activity and reduced sensitivity to tyrosine kinase inhibitors. Knockdown of the Drosophila LZTR1 ortholog CG3711 resulted in a Ras-dependent gain-of-function phenotype. Endogenous human LZTR1 associates with the main RAS isoforms. Inactivation of LZTR1 led to decreased ubiquitination and enhanced plasma membrane localization of endogenous KRAS (V-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog). We propose that LZTR1 acts as a conserved regulator of RAS ubiquitination and MAPK pathway activation. Because LZTR1 disease mutations failed to revert loss-of-function phenotypes, our findings provide a molecular rationale for LZTR1 involvement in a variety of inherited and acquired human disorders.


Subject(s)
Proto-Oncogene Proteins p21(ras)/metabolism , Transcription Factors/physiology , Ubiquitination , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Drosophila melanogaster , Drug Resistance, Neoplasm/genetics , Fusion Proteins, bcr-abl/antagonists & inhibitors , Gain of Function Mutation , Gene Knockdown Techniques , Humans , Imidazoles/pharmacology , Imidazoles/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/epidemiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Loss of Function Mutation , MAP Kinase Signaling System/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyridazines/pharmacology , Pyridazines/therapeutic use , Signal Transduction , Transcription Factors/genetics , Ubiquitination/genetics
7.
J Cell Biol ; 217(6): 2205-2221, 2018 06 04.
Article in English | MEDLINE | ID: mdl-29650776

ABSTRACT

Lymphatic endothelial cells (LECs) release extracellular chemokines to guide the migration of dendritic cells. In this study, we report that LECs also release basolateral exosome-rich endothelial vesicles (EEVs) that are secreted in greater numbers in the presence of inflammatory cytokines and accumulate in the perivascular stroma of small lymphatic vessels in human chronic inflammatory diseases. Proteomic analyses of EEV fractions identified >1,700 cargo proteins and revealed a dominant motility-promoting protein signature. In vitro and ex vivo EEV fractions augmented cellular protrusion formation in a CX3CL1/fractalkine-dependent fashion and enhanced the directional migratory response of human dendritic cells along guidance cues. We conclude that perilymphatic LEC exosomes enhance exploratory behavior and thus promote directional migration of CX3CR1-expressing cells in complex tissue environments.


Subject(s)
Cell Movement , Dendritic Cells/cytology , Dendritic Cells/metabolism , Exosomes/metabolism , Lymphatic Vessels/metabolism , Animals , Cell Line, Tumor , Cell Surface Extensions/metabolism , Cellular Microenvironment , Chemokine CX3CL1/metabolism , Collagen/metabolism , Cues , Endothelial Cells/metabolism , Endothelial Cells/ultrastructure , Exosomes/ultrastructure , Humans , Inflammation/pathology , Kidney/metabolism , Kidney/pathology , Male , Mice , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proteomics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
8.
ACS Chem Biol ; 13(3): 553-560, 2018 03 16.
Article in English | MEDLINE | ID: mdl-29356495

ABSTRACT

Protein degradation is an emerging therapeutic strategy with a unique molecular pharmacology that enables the disruption of all functions associated with a target. This is particularly relevant for proteins depending on molecular scaffolding, such as transcription factors or receptor tyrosine kinases (RTKs). To address tractability of multiple RTKs for chemical degradation by the E3 ligase CUL4-RBX1-DDB1-CRBN (CRL4CRBN), we synthesized a series of phthalimide degraders based on the promiscuous kinase inhibitors sunitinib and PHA665752. While both series failed to induce degradation of their consensus targets, individual molecules displayed pronounced efficacy in leukemia cell lines. Orthogonal target identification supported by molecular docking led us to identify the translation termination factor G1 to S phase transition 1 (GSPT1) as a converging off-target, resulting from inadvertent E3 ligase modulation. This research highlights the importance of monitoring degradation events that are independent of the respective targeting ligand as a unique feature of small-molecule degraders.


Subject(s)
Peptide Chain Termination, Translational , Peptide Termination Factors , Proteolysis , Cell Line, Tumor , Humans , Molecular Docking Simulation , Phthalimides/chemistry , Protein Kinase Inhibitors/chemistry , Ubiquitin-Protein Ligases/metabolism
9.
J Am Soc Nephrol ; 29(1): 268-282, 2018 01.
Article in English | MEDLINE | ID: mdl-29046343

ABSTRACT

Cardiovascular disease (CVD) is the leading cause of increased mortality in patients with CKD and is further aggravated by peritoneal dialysis (PD). Children are devoid of preexisting CVD and provide unique insight into specific uremia- and PD-induced pathomechanisms of CVD. We obtained peritoneal specimens from children with stage 5 CKD at time of PD catheter insertion (CKD5 group), children with established PD (PD group), and age-matched nonuremic controls (n=6/group). We microdissected omental arterioles from tissue layers not directly exposed to PD fluid and used adjacent sections of four arterioles per patient for transcriptomic and proteomic analyses. Findings were validated in omental and parietal arterioles from independent pediatric control (n=5), CKD5 (n=15), and PD (n=15) cohorts. Transcriptomic analysis revealed differential gene expression in control versus CKD5 arterioles and in CKD5 versus PD arterioles. Gene ontology analyses revealed activation of metabolic processes in CKD5 arterioles and of inflammatory, immunologic, and stress-response cascades in PD arterioles. PD arterioles exhibited particular upregulation of the complement system and respective regulatory pathways, with concordant findings at the proteomic level. In the validation cohorts, PD specimens had the highest abundance of omental and parietal arteriolar C1q, C3d, terminal complement complex, and phosphorylated SMAD2/3, a downstream effector of TGF-ß Furthermore, in the PD parietal arterioles, C1q and terminal complement complex abundance correlated with the level of dialytic glucose exposure, abundance of phosphorylated SMAD2/3, and degree of vasculopathy. We conclude that PD fluids activate arteriolar complement and TGF-ß signaling, which quantitatively correlate with the severity of arteriolar vasculopathy.


Subject(s)
Arterioles/metabolism , Complement Activation , Complement System Proteins/metabolism , Kidney Failure, Chronic/therapy , Peritoneal Dialysis/adverse effects , Vascular Diseases/metabolism , Adolescent , Case-Control Studies , Child , Child, Preschool , Complement C1q/metabolism , Complement C3d/metabolism , Complement Membrane Attack Complex/metabolism , Female , Gene Ontology , Humans , Infant , Infant, Newborn , Kidney Failure, Chronic/complications , Male , Omentum/blood supply , Phosphorylation , Proteome , Severity of Illness Index , Signal Transduction , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transcriptome , Transforming Growth Factor beta/metabolism , Uremia/etiology , Vascular Diseases/etiology , Vascular Endothelial Growth Factor A/metabolism
10.
Mol Cell Proteomics ; 17(3): 516-532, 2018 03.
Article in English | MEDLINE | ID: mdl-29208752

ABSTRACT

Peritoneal dialysis (PD) is a modality of renal replacement therapy in which the high volumes of available PD effluent (PDE) represents a rich source of biomarkers for monitoring disease and therapy. Although this information could help guide the management of PD patients, little is known about the potential of PDE to define pathomechanism-associated molecular signatures in PD.We therefore subjected PDE to a high-performance multiplex proteomic analysis after depletion of highly-abundant plasma proteins and enrichment of low-abundance proteins. A combination of label-free and isobaric labeling strategies was applied to PDE samples from PD patients (n = 20) treated in an open-label, randomized, two-period, cross-over clinical trial with standard PD fluid or with a novel PD fluid supplemented with alanyl-glutamine (AlaGln).With this workflow we identified 2506 unique proteins in the PDE proteome, greatly increasing coverage beyond the 171 previously-reported proteins. The proteins identified range from high abundance plasma proteins to low abundance cellular proteins, and are linked to larger numbers of biological processes and pathways, some of which are novel for PDE. Interestingly, proteins linked to membrane remodeling and fibrosis are overrepresented in PDE compared with plasma, whereas the proteins underrepresented in PDE suggest decreases in host defense, immune-competence and response to stress. Treatment with AlaGln-supplemented PD fluid is associated with reduced activity of membrane injury-associated mechanisms and with restoration of biological processes involved in stress responses and host defense.Our study represents the first application of the PDE proteome in a randomized controlled prospective clinical trial of PD. This novel proteomic workflow allowed detection of low abundance biomarkers to define pathomechanism-associated molecular signatures in PD and their alterations by a novel therapeutic intervention.


Subject(s)
Dipeptides/pharmacology , Peritoneal Dialysis , Proteome , Blood Proteins/metabolism , Cross-Over Studies , Female , Humans , Male
11.
Cell ; 168(1-2): 86-100.e15, 2017 Jan 12.
Article in English | MEDLINE | ID: mdl-27916275

ABSTRACT

Type 1 diabetes is characterized by the destruction of pancreatic ß cells, and generating new insulin-producing cells from other cell types is a major aim of regenerative medicine. One promising approach is transdifferentiation of developmentally related pancreatic cell types, including glucagon-producing α cells. In a genetic model, loss of the master regulatory transcription factor Arx is sufficient to induce the conversion of α cells to functional ß-like cells. Here, we identify artemisinins as small molecules that functionally repress Arx by causing its translocation to the cytoplasm. We show that the protein gephyrin is the mammalian target of these antimalarial drugs and that the mechanism of action of these molecules depends on the enhancement of GABAA receptor signaling. Our results in zebrafish, rodents, and primary human pancreatic islets identify gephyrin as a druggable target for the regeneration of pancreatic ß cell mass from α cells.


Subject(s)
Artemisinins/pharmacology , Diabetes Mellitus, Type 1/drug therapy , Disease Models, Animal , Receptors, GABA-A/metabolism , Signal Transduction , Animals , Artemether , Artemisinins/administration & dosage , Carrier Proteins/metabolism , Cell Transdifferentiation/drug effects , Cells, Cultured , Diabetes Mellitus/drug therapy , Diabetes Mellitus, Type 1/pathology , Gene Expression Profiling , Homeodomain Proteins/metabolism , Humans , Insulin/genetics , Insulin/metabolism , Islets of Langerhans/drug effects , Membrane Proteins/metabolism , Mice , Protein Stability/drug effects , Rats , Single-Cell Analysis , Transcription Factors/metabolism , Zebrafish , gamma-Aminobutyric Acid/metabolism
13.
PLoS One ; 10(11): e0142162, 2015.
Article in English | MEDLINE | ID: mdl-26539827

ABSTRACT

We performed a pilot proteogenomic study to compare lung adenocarcinoma to lung squamous cell carcinoma using quantitative proteomics (6-plex TMT) combined with a customized Affymetrix GeneChip. Using MaxQuant software, we identified 51,001 unique peptides that mapped to 7,241 unique proteins and from these identified 6,373 genes with matching protein expression for further analysis. We found a minor correlation between gene expression and protein expression; both datasets were able to independently recapitulate known differences between the adenocarcinoma and squamous cell carcinoma subtypes. We found 565 proteins and 629 genes to be differentially expressed between adenocarcinoma and squamous cell carcinoma, with 113 of these consistently differentially expressed at both the gene and protein levels. We then compared our results to published adenocarcinoma versus squamous cell carcinoma proteomic data that we also processed with MaxQuant. We selected two proteins consistently overexpressed in squamous cell carcinoma in all studies, MCT1 (SLC16A1) and GLUT1 (SLC2A1), for further investigation. We found differential expression of these same proteins at the gene level in our study as well as in other public gene expression datasets. These findings combined with survival analysis of public datasets suggest that MCT1 and GLUT1 may be potential prognostic markers in adenocarcinoma and druggable targets in squamous cell carcinoma. Data are available via ProteomeXchange with identifier PXD002622.


Subject(s)
Adenocarcinoma/genetics , Biomarkers, Tumor/genetics , Glucose Transporter Type 1/genetics , Lung Neoplasms/genetics , Monocarboxylic Acid Transporters/genetics , Symporters/genetics , Adenocarcinoma/diagnosis , Adenocarcinoma of Lung , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/genetics , Female , Gene Expression/genetics , Humans , Lung Neoplasms/diagnosis , Male , Pilot Projects , Prognosis , Proteomics/methods , Survival Analysis
14.
Mol Cancer Ther ; 13(11): 2751-62, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25189542

ABSTRACT

A growing number of gene mutations, which are recognized as cancer drivers, can be successfully targeted with drugs. The redundant and dynamic nature of oncogenic signaling networks and complex interactions between cancer cells and the microenvironment, however, can cause drug resistance. While these challenges can be addressed by developing drug combinations or polypharmacology drugs, this benefits greatly from a detailed understanding of the proteome-wide target profiles. Using mass spectrometry-based chemical proteomics, we report the comprehensive characterization of the drug-protein interaction networks for the multikinase inhibitors dasatinib and sunitinib in primary lung cancer tissue specimens derived from patients. We observed in excess of 100 protein kinase targets plus various protein complexes involving, for instance, AMPK, TBK1 (sunitinib), and ILK (dasatinib). Importantly, comparison with lung cancer cell lines and mouse xenografts thereof showed that most targets were shared between cell lines and tissues. Several targets, however, were only present in tumor tissues. In xenografts, most of these proteins were of mouse origin suggesting that they originate from the tumor microenvironment. Furthermore, intersection with subsequent global phosphoproteomic analysis identified several activated signaling pathways. These included MAPK, immune, and integrin signaling, which were affected by these drugs in both cancer cells and the microenvironment. Thus, the combination of chemical and phosphoproteomics can generate a systems view of proteins, complexes, and signaling pathways that are simultaneously engaged by multitargeted drugs in cancer cells and the tumor microenvironment. This may allow for the design of novel anticancer therapies that concurrently target multiple tumor compartments.


Subject(s)
Lung Neoplasms/drug therapy , Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Cell Line, Tumor , Female , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Mice , Models, Molecular , Molecular Targeted Therapy , Protein Kinase Inhibitors/pharmacology , Proteomics , Signal Transduction , Tumor Microenvironment , Xenograft Model Antitumor Assays
15.
J Proteome Res ; 13(6): 2830-45, 2014 Jun 06.
Article in English | MEDLINE | ID: mdl-24803318

ABSTRACT

Melanoma, the deadliest form of skin cancer, is highly immunogenic and frequently infiltrated with immune cells including B cells. The role of tumor-infiltrating B cells (TIBCs) in melanoma is as yet unresolved, possibly due to technical challenges in obtaining TIBCs in sufficient quantity for extensive studies and due to the limited life span of B cells in vitro. A comprehensive workflow has thus been developed for successful isolation and proteomic analysis of a low number of TIBCs from fresh, human melanoma tissue. In addition, we generated in vitro-proliferating TIBC cultures using simultaneous stimulation with Epstein-Barr virus (EBV) and the TLR9 ligand CpG-oligodesoxynucleotide (CpG ODN). The FASP method and iTRAQ labeling were utilized to obtain a comparative, semiquantitative proteome to assess EBV-induced changes in TIBCs. By using as few as 100 000 B cells (∼5 µg protein)/sample for our proteomic study, a total number of 6507 proteins were identified. EBV-induced changes in TIBCs are similar to those already reported for peripheral B cells and largely involve changes in cell cycle proliferation, apoptosis, and interferon response, while most of the proteins were not significantly altered. This study provides an essential, further step toward detailed characterization of TIBCs including functional in vitro analysis.


Subject(s)
B-Lymphocytes/metabolism , Herpesvirus 4, Human/physiology , Melanoma/immunology , Proteome/metabolism , B-Lymphocytes/virology , Cell Proliferation , Cell Separation , Gene Expression , Gene Expression Regulation, Neoplastic , Gene Ontology , Humans , Melanoma/pathology , Melanoma/secondary , Molecular Sequence Annotation , Proteome/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Cells, Cultured
16.
J Proteome Res ; 13(2): 1147-55, 2014 Feb 07.
Article in English | MEDLINE | ID: mdl-24400740

ABSTRACT

Affinity purification coupled to 1-D gel-free liquid chromatography mass spectrometry (LC-MS) is a well-established and widespread approach for the analyses of noncovalently interacting protein complexes. In this study, two proteins conjugated to a streptavidin-binding peptide and hemagglutinin double tag were expressed in the respective Flp-In HEK293 cell lines: green fluorescent protein (SH-GFP) and TANK binding kinase 1 (SH-TBK1_MOUSE). Fluorescent anti-HA immunoblots revealed that the expression level of SH-GFP was ∼50% lower than that of SH-TBK1_MOUSE. Subsequently, the input material was normalized to obtain a similar quantity of purified SH-tagged proteins. Optimization of the release of protein complexes from the anti-HA-agarose with different eluting agents was then assessed. With respect to the total number of protein groups identified in the purified complexes, elution with 2% SDS surpassed both 100 mM glycine and 100 mM formic acid. Relative quantitation of the purified protein complexes using TMT 6-plex reagents confirmed the higher efficiency of the 2% SDS elution followed by filter-aided sample preparation (FASP). The data presented in this study provide a new application of FASP to quantitative MS analysis of affinity-purified protein complexes. We have termed the approach abFASP-MS, or affinity-based filter-aided sample preparation mass spectrometry.


Subject(s)
Proteins/analysis , Tandem Mass Spectrometry/methods , Blotting, Western , Chromatography, Affinity , Chromatography, Reverse-Phase , Electrophoresis, Polyacrylamide Gel , Proteins/chemistry , Trypsin/chemistry
17.
PLoS One ; 8(10): e77155, 2013.
Article in English | MEDLINE | ID: mdl-24130846

ABSTRACT

Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ ALL) is in part driven by the tyrosine kinase bcr-abl, but imatinib does not produce long-term remission. Therefore, second-generation ABL inhibitors are currently in clinical investigation. Considering different target specificities and the pronounced genetic heterogeneity of Ph+ ALL, which contributes to the aggressiveness of the disease, drug candidates should be evaluated with regard to their effects on the entire Ph+ ALL-specific signaling network. Here, we applied an integrated experimental and computational approach that allowed us to estimate the differential impact of the bcr-abl inhibitors nilotinib, dasatinib, Bosutinib and Bafetinib. First, we determined drug-protein interactions in Ph+ ALL cell lines by chemical proteomics. We then mapped those interactions along with known genetic lesions onto public protein-protein interactions. Computation of global scores through correlation of target affinity, network topology, and distance to disease-relevant nodes assigned the highest impact to dasatinib, which was subsequently confirmed by proliferation assays. In future, combination of patient-specific genomic information with detailed drug target knowledge and network-based computational analysis should allow for an accurate and individualized prediction of therapy.


Subject(s)
Fusion Proteins, bcr-abl/antagonists & inhibitors , Models, Biological , Philadelphia Chromosome , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Protein Kinase Inhibitors/pharmacology , Proteomics , Systems Biology , Cell Proliferation/drug effects , Humans , Molecular Targeted Therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Interaction Maps/drug effects , Protein Kinase Inhibitors/therapeutic use
18.
Nat Methods ; 10(10): 965-71, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24161985

ABSTRACT

Knockout collections are invaluable tools for studying model organisms such as yeast. However, there are no large-scale knockout collections of human cells. Using gene-trap mutagenesis in near-haploid human cells, we established a platform to generate and isolate individual 'gene-trapped cells' and used it to prepare a collection of human cell lines carrying single gene-trap insertions. In most cases, the insertion can be reversed. This growing library covers 3,396 genes, one-third of the expressed genome, is DNA-barcoded and allows systematic screens for a wide variety of cellular phenotypes. We examined cellular responses to TNF-α, TGF-ß, IFN-γ and TNF-related apoptosis-inducing ligand (TRAIL), to illustrate the value of this unique collection of isogenic human cell lines.


Subject(s)
Gene Library , Haploidy , Mutagenesis, Insertional/methods , Reverse Genetics/methods , Cell Line, Tumor , Genome, Human , Humans , Molecular Sequence Data
19.
J Proteome Res ; 12(9): 4005-17, 2013 Sep 06.
Article in English | MEDLINE | ID: mdl-23901793

ABSTRACT

While targeted therapy based on the idea of attenuating the activity of a preselected, therapeutically relevant protein has become one of the major trends in modern cancer therapy, no truly specific targeted drug has been developed and most clinical agents have displayed a degree of polypharmacology. Therefore, the specificity of anticancer therapeutics has emerged as a highly important but severely underestimated issue. Chemical proteomics is a powerful technique combining postgenomic drug-affinity chromatography with high-end mass spectrometry analysis and bioinformatic data processing to assemble a target profile of a desired therapeutic molecule. Due to high demands on the starting material, however, chemical proteomic studies have been mostly limited to cancer cell lines. Herein, we report a down-scaling of the technique to enable the analysis of very low abundance samples, as those obtained from needle biopsies. By a systematic investigation of several important parameters in pull-downs with the multikinase inhibitor bosutinib, the standard experimental protocol was optimized to 100 µg protein input. At this level, more than 30 well-known targets were detected per single pull-down replicate with high reproducibility. Moreover, as presented by the comprehensive target profile obtained from miniaturized pull-downs with another clinical drug, dasatinib, the optimized protocol seems to be extendable to other drugs of interest. Sixty distinct human and murine targets were finally identified for bosutinib and dasatinib in chemical proteomic experiments utilizing core needle biopsy samples from xenotransplants derived from patient tumor tissue. Altogether, the developed methodology proves robust and generic and holds many promises for the field of personalized health care.


Subject(s)
Aniline Compounds/chemistry , Carcinoma, Non-Small-Cell Lung/enzymology , Lung Neoplasms/enzymology , Nitriles/chemistry , Protein-Tyrosine Kinases/isolation & purification , Pyrimidines/chemistry , Quinolines/chemistry , Thiazoles/chemistry , Animals , Biopsy , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Chromatography, Affinity , Dasatinib , Humans , K562 Cells , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Molecular Sequence Annotation , Molecular Targeted Therapy , Neoplasm Transplantation , Protein Interaction Maps , Protein Kinase Inhibitors/chemistry , Proteomics
20.
Exp Eye Res ; 108: 48-58, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23201028

ABSTRACT

The purpose of the study was to perform a detailed, quantitative protein analysis of the aqueous (AF) and vitreous fluids (VF) from human eyes with idiopathic epiretinal membranes (iERM). The complementary approaches of quantitative liquid chromatography mass spectrometry (LCMS) and multiplex protein assays were utilised to reveal the protein composition in ocular fluids of this retinal disorder. In a prospective clinical trial, AF and VF was collected during surgery from twenty four eyes corresponding to twenty four patients with iERM. VF and AF from eight patients were labelled with the 4-plex iTRAQ reagent and analysed by LCMS. Each iTRAQ 4-plex experiment consisted of the AH and VH from two patients. A total of 323 proteins were identified in the AF and VF from eyes with iERM. Grouping the proteins according to involvement in biological processes, showed that the majority were involved in the classical and alternative pathway of complement activation (n = 27), proteolysis (n = 26) and cell adhesion (n = 28). iTRAQ relative quantitation revealed minimal variation in the protein content between both ocular compartments with only 3.96% of the identified proteins significantly, differentially-expressed. Eight proteins were expressed at a higher level in the VF compared to the AF; and 4 proteins were expressed at a lower level in the VF compared to the AF. For the multiplex bead assays, 29 growth factors and cytokines were assessed in the AF and VF from 16 additional patients with iERM. The protein profile was shown to be similar between VF and AF for the majority of factors except PDGF-AA. This factor was expressed at a higher level in the VF compared to the AF. The data presented in this study revealed that the majority of the proteins identified by LCMS and multiplex bead assays were present in both ocular compartments in similar quantities. This study is a first step, 'discovery phase' towards revealing and quantitating the protein content in the aqueous and vitreous fluid in human eyes with iERM.


Subject(s)
Aqueous Humor/chemistry , Epiretinal Membrane/metabolism , Eye Proteins/analysis , Proteomics , Vitreous Body/chemistry , Aged , Aged, 80 and over , Austria , Biomarkers/analysis , Chromatography, High Pressure Liquid , Chromatography, Reverse-Phase , Databases, Protein , Epiretinal Membrane/surgery , Humans , Middle Aged , Peptide Mapping , Prospective Studies , Proteomics/methods , Spectrometry, Mass, Electrospray Ionization
SELECTION OF CITATIONS
SEARCH DETAIL
...