Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Data Brief ; 21: 527-539, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30370323

ABSTRACT

Data included are related to the research article "Isolation of biofunctional bovine immunoglobulin G from milk- and colostral whey with mixed-mode chromatography at lab and pilot scale" (Heidebrecht et al., 2018) [1]. Data show individual bovine whey proteins in flow-through and elution fractions using different chromatographic resins as well as different binding and elution conditions. The relevant analytical methods for individual protein detection were SDS-PAGE and reversed phase- high performance liquid chromatography. The focus of the data is on the two mixed mode materials MEP HyperCel™ and Capto™-multimodal chromatography. Resins were used individually, in series and at different scale. Data provide information at which binding and elution conditions it is possible to isolate bovine IgG from milk and colostral whey and at which purity.

2.
J Chromatogr A ; 1562: 59-68, 2018 Aug 10.
Article in English | MEDLINE | ID: mdl-29843945

ABSTRACT

The aim of the present work was to develop a new scalable and cost-efficient process to isolate bovine immunoglobulin G from colostral whey with high purity and minimal loss of activity. The mixed mode material Mercapto-Ethyl-Pyridine-Hypercel™ was identified appropriate for direct capture of immunoglobulin G. The binding mechanism is primarily based on hydrophobic interactions at physiological conditions. As compared to immunoglobulin G, all other low molecular whey proteins such as α-Lactalbumin or ß-Lactoglobulin, except lactoperoxidase, are more hydrophilic and were therefore found in the flow-through fraction. In order to remove lactoperoxidase as an impurity the column was combined in series with a second mixed mode material (Capto™- with N-benzoyl-homocysteine as ligand) using the same binding conditions. At pH 7.5 the carboxyl group of this ligand is negatively charged and can hence bind the positively charged lactoperoxidase, whose isoelectric point is at pH 9.6. After sample application, the columns were eluted separately. By combining the two columns it was possible to obtain immunoglobulin G with a purity of >96.1% and yield of 65-80%. The process development was carried out using 1 mL columns and upscaling was performed in three steps up to a column volume of 8800 mL for the Hypercel™ column and 3000 mL for the Capto™- column. At this scale it is possible to obtain 130-150 g pure immunoglobulin G from 3 L colostrum within five hours, including the regeneration of both columns. Additionally, the impact of freeze-drying on the isolated immunoglobulin G was studied. The nativity of the freeze dried immunoglobulin was above 95%, which was proven by reversed phase liquid chromatography and validated by differential scanning calorimetry. The activity of immunoglobulin G was preserved over the isolation process and during drying as measured by enzyme-linked immunosorbent assay. In conclusion, by applying the proposed isolation process, it becomes feasible to obtain pure, active and stable imunnunoglobulin G at large scale.


Subject(s)
Chemistry Techniques, Analytical/methods , Chromatography , Colostrum/chemistry , Immunoglobulin G/isolation & purification , Milk/chemistry , Whey/chemistry , Animals , Cattle , Enzyme-Linked Immunosorbent Assay , Milk Proteins/analysis
3.
Mol Cell Proteomics ; 16(7): 1365-1376, 2017 07.
Article in English | MEDLINE | ID: mdl-28450419

ABSTRACT

Constitutively activating internal tandem duplication (ITD) alterations of the receptor tyrosine kinase FLT3 (Fms-like tyrosine kinase 3) are common in acute myeloid leukemia (AML) and classifies FLT3 as an attractive therapeutic target. So far, applications of FLT3 small molecule inhibitors have been investigated primarily in FLT3-ITD+ patients. Only recently, a prolonged event-free survival has been observed in AML patients who were treated with the multikinase inhibitor sorafenib in addition to standard therapy. Here, we studied the sorafenib effect on proliferation in a panel of 13 FLT3-ITD- and FLT3-ITD+ AML cell lines. Sorafenib IC50 values ranged from 0.001 to 5.6 µm, whereas FLT3-ITD+ cells (MOLM-13, MV4-11) were found to be more sensitive to sorafenib than FLT3-ITD- cells. However, we identified two FLT3-ITD- cell lines (MONO-MAC-1 and OCI-AML-2) which were also sorafenib sensitive. Phosphoproteome analyses revealed that the affected pathways differed in sorafenib sensitive FLT3-ITD- and FLT3-ITD+ cells. In MV4-11 cells sorafenib suppressed mTOR signaling by direct inhibition of FLT3. In MONO-MAC-1 cells sorafenib inhibited the MEK/ERK pathway. These data suggest that the FLT3 status in AML patients might not be the only factor predicting response to treatment with sorafenib.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , Mutation , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Phosphoproteins/drug effects , Protein Kinase Inhibitors/pharmacology , fms-Like Tyrosine Kinase 3/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Regulatory Networks/drug effects , Humans , Inhibitory Concentration 50 , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Niacinamide/pharmacology , Phosphoproteins/analysis , Proteomics/methods , Sorafenib
4.
Antiviral Res ; 124: 101-9, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26546752

ABSTRACT

Infection with human cytomegalovirus (HCMV) is a serious medical problem, particularly in immunocompromised individuals and neonates. The success of standard antiviral therapy is hampered by low drug compatibility and induction of viral resistance. A novel strategy is based on the exploitation of cell-directed signaling inhibitors. The broad antiinfective drug artesunate (ART) offers additional therapeutic options such as oral bioavailability and low levels of toxic side-effects. Here, novel ART-derived compounds including dimers and trimers were synthesized showing further improvements over the parental drug. Antiviral activity and mechanistic aspects were determined leading to the following statements: (i) ART exerts antiviral activity towards human and animal herpesviruses, (ii) no induction of ART-resistant HCMV mutants occurred in vitro, (iii) chemically modified derivatives of ART showed strongly enhanced anti-HCMV efficacy, (iv) NF-κB reporter constructs, upregulated during HCMV replication, could be partially blocked by ART treatment, (v) ART activity analyzed in stable reporter cell clones indicated an inhibition of stimulated NF-κB but not CREB pathway, (vi) solid-phase immobilized ART was able to bind to NF-κB RelA/p65, and (vii) peptides within NF-κB RelA/p65 represent candidates of ART binding as analyzed by in silico docking and mass spectrometry. These novel findings open new prospects for the future medical use of ART and ART-related drug candidates.


Subject(s)
Artemisinins/pharmacology , Cytomegalovirus/drug effects , Cytomegalovirus/metabolism , NF-kappa B/metabolism , Transcription Factor RelA/metabolism , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Artemisinins/chemistry , Artesunate , Cyclic AMP Response Element-Binding Protein/metabolism , Cytomegalovirus/genetics , Drug Resistance, Viral , HEK293 Cells , Herpesviridae/drug effects , Humans , Mutation , NF-kappa B/antagonists & inhibitors , Signal Transduction/drug effects , Transcriptional Activation , Up-Regulation
5.
Antiviral Res ; 99(2): 139-48, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23648710

ABSTRACT

Human cytomegalovirus infection can lead to life-threatening clinical manifestations particularly in the immunocompromised host. Current therapy options face severe limitations leading to a continued search for alternative drug candidates. Viral replication is dependent on a balanced interaction between viral and cellular proteins. Especially protein kinases are important regulators of virus-host interaction indicated by remarkable kinome alterations induced upon HCMV infection. Here we report a novel approach of kinome profiling with an outcome that suggests an important role of specific cellular protein kinases, such as AMPK, ABL2 and Aurora A. Inhibition of AMPK and ABL kinases showed a significant reduction, whereas inhibition of Aurora A kinase led to a slight activation of HCMV replication, as measured in a GFP reporter-based replication assay. Furthermore, analysis of the mode of antiviral action suggested a substantial benefit for the efficiency of viral replication at the immediate early (AMPK) or early-late (ABL) phases of HCMV gene expression. In contrast, inhibition of Aurora A kinase promoted an enhancement of viral early-late gene expression, suggesting a putative role of Aurora A signaling in host defense. Thus, the combined data provide new information on host cell kinases involved in viral replication and uncovered potential targets for future antiviral strategies.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Aurora Kinase A/metabolism , Cytomegalovirus Infections/enzymology , Cytomegalovirus/physiology , Proto-Oncogene Proteins c-abl/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Aurora Kinase A/antagonists & inhibitors , Benzamides/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Cytomegalovirus/genetics , Cytomegalovirus Infections/virology , Gene Expression Regulation, Viral/drug effects , Humans , Imatinib Mesylate , Intracellular Signaling Peptides and Proteins , Nucleic Acid Synthesis Inhibitors/pharmacology , Nucleic Acid Synthesis Inhibitors/therapeutic use , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Pyrimidines/pharmacology , Serine-Threonine Kinase 3 , Virus Replication/drug effects
6.
Eur J Cancer ; 49(6): 1491-500, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23245330

ABSTRACT

ST1926 is an atypical retinoid and a promising anti-tumour agent with selective apoptotic activity on the leukaemic blast. The anti-tumour activity of the compound has been associated with its capacity to induce DNA double stranded breaks. Target profiling by affinity chromatography coupled to mass spectrometry led to the identification of histone H2A.Z as a protein capable of binding ST1926 specifically. The result was confirmed by studies involving Surface Plasmon Resonance (SPR). This indicates that H2A.Z is a primary target of ST1926 and links the perturbations of the histone pathway observed by microarray analysis to the DNA damage and apoptotic responses caused by the atypical retinoid. Comparison of the whole-genome gene-expression profiles of the ST1926-sensitive NB4 and the ST1926-resistant NB4.437r cell lines demonstrated differential expression of numerous genes. Network analysis of the data indicated enrichment of the cellular pathways controlling cAMP (cyclic adenosine-monophosphate)-dependent signal transduction, proteasome-dependent protein degradation and nuclear histones in NB4.437r cells. Pharmacological inhibition of cAMP-dependent protein kinase A with H89 partially reverted resistance of NB4.437r cells to ST1926. Conversely, inhibition of the proteasome with MG132 or bortezomib blocked the apoptotic response afforded by ST1926 in the NB4 cell line. This last effect was associated with a dramatic reduction in the DNA damage caused by the atypical retinoid. The results corroborate the idea that DNA damage is an important determinant of ST1926 apoptotic activity. More importantly, they demonstrate a proactive role of the proteasome in the DNA damaging and ensuing apoptotic response observed upon the challenge of acute myeloid leukaemia cells with ST1926.


Subject(s)
Adamantane/analogs & derivatives , Apoptosis/drug effects , Cinnamates/pharmacology , Acute Disease , Adamantane/metabolism , Adamantane/pharmacology , Apoptosis/genetics , Blotting, Western , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Cinnamates/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Enzyme Inhibitors/pharmacology , Gene Expression Profiling , Gene Expression Regulation, Leukemic/drug effects , Histones/genetics , Histones/metabolism , Humans , Isoquinolines/pharmacology , Leukemia, Myeloid/genetics , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/pathology , Leupeptins/pharmacology , Oligonucleotide Array Sequence Analysis , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Signal Transduction/drug effects , Signal Transduction/genetics , Sulfonamides/pharmacology , Surface Plasmon Resonance
7.
Mol Cell Proteomics ; 11(9): 651-68, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22617229

ABSTRACT

Targeted drugs are less toxic than traditional chemotherapeutic therapies; however, the proportion of patients that benefit from these drugs is often smaller. A marker that confidently predicts patient response to a specific therapy would allow an individual therapy selection most likely to benefit the patient. Here, we used quantitative mass spectrometry to globally profile the basal phosphoproteome of a panel of non-small cell lung cancer cell lines. The effect of the kinase inhibitor dasatinib on cellular growth was tested against the same panel. From the phosphoproteome profiles, we identified 58 phosphorylation sites, which consistently differ between sensitive and resistant cell lines. Many of the corresponding proteins are involved in cell adhesion and cytoskeleton organization. We showed that a signature of only 12 phosphorylation sites is sufficient to accurately predict dasatinib sensitivity. Four of the phosphorylation sites belong to integrin ß4, a protein that mediates cell-matrix or cell-cell adhesion. The signature was validated in cross-validation and label switch experiments and in six independently profiled breast cancer cell lines. The study supports that the phosphorylation of integrin ß4, as well as eight further proteins comprising the signature, are candidate biomarkers for predicting response to dasatinib in solid tumors. Furthermore, our results show that identifying predictive phosphorylation signatures from global, quantitative phosphoproteomic data is possible and can open a new path to discovering molecular markers for response prediction.


Subject(s)
Breast Neoplasms/drug therapy , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Phosphoproteins/analysis , Pyrimidines/pharmacology , Thiazoles/pharmacology , Biomarkers, Tumor , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Adhesion , Cell Line, Tumor , Cell Proliferation/drug effects , Dasatinib , Drug Resistance, Neoplasm , Female , Gene Expression Profiling , Humans , Integrin beta4/chemistry , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mass Spectrometry , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Proteome/analysis
8.
Neoplasia ; 13(11): 1026-34, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22131878

ABSTRACT

A placebo-controlled phase 3 trial demonstrated that the epidermal growth factor receptor (EGFR) inhibitor erlotinib in combination with gemcitabine was especially efficient in a pancreatic ductal adenocarcinoma (PDAC) subgroup of patients developing skin toxicity. However, EGFR expression was not predictive for response, and markers to characterize an erlotinib-responding PDAC group are currently missing. In this work, we observed high erlotinib IC50 values in a panel of human and murine PDAC cell lines. Using EGFR small interfering RNA, we detected that the erlotinib response was marginally influenced by EGFR. To find novel EGFR targets, we used an unbiased chemical proteomics approach for target identification and quality-controlled target affinity determination combined with quantitative mass spectrometry based on stable isotope labeling by amino acids in cell culture. In contrast to gefitinib, we observed a broad target profile of erlotinib in PDAC cells by quantitative proteomics. Six protein kinases bind to erlotinib with similar or higher affinity (K(d) = 0.09-0.358 µM) than the EGFR (K(d) 0.434 µM). We provide evidence that one of the novel erlotinib targets, ARG, contributes in part to the erlotinib response in a PDAC cell line. Our data show that erlotinib is a multikinase inhibitor, which can act independent of EGFR in PDAC. These findings may help to monitor future erlotinib trials in the clinic.


Subject(s)
Adenocarcinoma/pathology , Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Animals , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Drug Evaluation, Preclinical , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Erlotinib Hydrochloride , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Quinazolines/administration & dosage , Quinazolines/adverse effects , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/pharmacology , Transfection , Tumor Cells, Cultured
9.
BMC Bioinformatics ; 11: 351, 2010 Jun 28.
Article in English | MEDLINE | ID: mdl-20584295

ABSTRACT

BACKGROUND: Various high throughput methods are available for detecting regulations at the level of transcription, translation or posttranslation (e.g. phosphorylation). Integrating these data with protein networks should make it possible to identify subnetworks that are significantly regulated. Furthermore, such integration can support identification of regulated entities from often noisy high throughput data. In particular, processing mass spectrometry-based phosphoproteomic data in this manner may expose signal transduction pathways and, in the case of experiments with drug-treated cells, reveal the drug's mode of action. RESULTS: Here, we introduce SubExtractor, an algorithm that combines phosphoproteomic data with protein network information from STRING to identify differentially regulated subnetworks and individual proteins. The method is based on a Bayesian probabilistic model combined with a genetic algorithm and rigorous significance testing. The Bayesian model accounts for information about both differential regulation and network topology. The method was tested with artificial data and subsequently applied to a comprehensive phosphoproteomics study investigating the mode of action of sorafenib, a small molecule kinase inhibitor. CONCLUSIONS: SubExtractor reliably identifies differentially regulated subnetworks from phosphoproteomic data by integrating protein networks. The method can also be applied to gene or protein expression data.


Subject(s)
Algorithms , Proteomics/methods , Bayes Theorem , Gene Expression Profiling , Models, Statistical , Phosphorylation , Signal Transduction
10.
Cancer Res ; 65(15): 6919-26, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-16061676

ABSTRACT

Knowledge about molecular drug action is critical for the development of protein kinase inhibitors for cancer therapy. Here, we establish a chemical proteomic approach to profile the anticancer drug SU6668, which was originally designed as a selective inhibitor of receptor tyrosine kinases involved in tumor vascularization. By employing immobilized SU6668 for the affinity capture of cellular drug targets in combination with mass spectrometry, we identified previously unknown targets of SU6668 including Aurora kinases and TANK-binding kinase 1. Importantly, a cell cycle block induced by SU6668 could be attributed to inhibition of Aurora kinase activity. Moreover, SU6668 potently suppressed antiviral and inflammatory responses by interfering with TANK-binding kinase 1-mediated signal transmission. These results show the potential of chemical proteomics to provide rationales for the development of potent kinase inhibitors, which combine rather unexpected biological modes of action by simultaneously targeting defined sets of both serine/threonine and tyrosine kinases involved in cancer progression.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Indoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrroles/pharmacology , Animals , Aurora Kinases , COS Cells , Cell Division/drug effects , Chlorocebus aethiops , HeLa Cells , Humans , Oxindoles , Propionates , Protein Serine-Threonine Kinases/antagonists & inhibitors , Transfection
11.
Cancer Res ; 65(2): 379-82, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15695376

ABSTRACT

Targeted inhibition of protein kinases with small molecule drugs has evolved into a viable approach for anticancer therapy. However, the true selectivity of these therapeutic agents has remained unclear. Here, we used a proteomic method to profile the cellular targets of the clinical epidermal growth factor receptor kinase inhibitor gefitinib. Our data suggest alternative cellular modes of action for gefitinib and provide rationales for the development of related drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Animals , COS Cells , Chlorocebus aethiops , ErbB Receptors/antagonists & inhibitors , Gefitinib , HeLa Cells , Humans , Proteomics/methods , Structure-Activity Relationship
12.
Mol Cell Proteomics ; 3(12): 1181-93, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15475568

ABSTRACT

Small molecule inhibitors belonging to the pyrido[2,3-d]pyrimidine class of compounds were developed as antagonists of protein tyrosine kinases implicated in cancer progression. Derivatives from this compound class are effective against most of the imatinib mesylate-resistant BCR-ABL mutants isolated from advanced chronic myeloid leukemia patients. Here, we established an efficient proteomics method employing an immobilized pyrido[2,3-d]pyrimidine ligand as an affinity probe and identified more than 30 human protein kinases affected by this class of compounds. Remarkably, in vitro kinase assays revealed that the serine/threonine kinases Rip-like interacting caspase-like apoptosis-regulatory protein kinase (RICK) and p38alpha were among the most potently inhibited kinase targets. Thus, pyrido[2,3-d]pyrimidines did not discriminate between tyrosine and serine/threonine kinases. Instead, we found that these inhibitors are quite selective for protein kinases possessing a conserved small amino acid residue such as threonine at a critical site of the ATP binding pocket. We further demonstrated inhibition of both p38 and RICK kinase activities in intact cells upon pyrido[2,3-d]pyrimidine inhibitor treatment. Moreover, the established functions of these two kinases as signal transducers of inflammatory responses could be correlated with a potent in vivo inhibition of cytokine production by a pyrido[2,3-d]pyrimidine compound. Thus, our data demonstrate the utility of proteomic methods employing immobilized kinase inhibitors for identifying new targets linked to previously unrecognized therapeutic applications.


Subject(s)
Antineoplastic Agents/pharmacology , Protein Kinase Inhibitors/pharmacology , Proteomics/methods , Pyridines/pharmacology , Pyrimidines/pharmacology , Adenosine Triphosphate/chemistry , Amino Acid Sequence , Animals , Antineoplastic Agents/chemistry , Apoptosis , Binding Sites , COS Cells , Cell Line, Tumor , Cells, Cultured , Chromatography, Liquid , Cytokines/metabolism , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Enzyme Inhibitors/chemistry , HeLa Cells , Humans , Inflammation , Inhibitory Concentration 50 , Ligands , Lipopolysaccharides/chemistry , Mass Spectrometry , Models, Chemical , Molecular Sequence Data , Mutagenesis, Site-Directed , Plasmids/metabolism , Polyethylene Glycols/chemistry , Protein Kinase Inhibitors/chemistry , Pyridines/chemistry , Pyrimidines/chemistry , Sequence Homology, Amino Acid , Signal Transduction , Time Factors
13.
Assay Drug Dev Technol ; 2(2): 215-24, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15165517

ABSTRACT

Small-molecule inhibitors of protein kinases constitute a novel class of drugs for therapeutic intervention in a variety of human diseases. Most of these agents target the relatively conserved ATP-binding site of protein kinases and have only been tested against a rather small subset of all human protein kinases. Therefore, the selectivity of protein kinase inhibitors has remained a widely underestimated, but highly important issue in drug development programs. In this review, we focus on the recent advancement of chemical proteomic methods to evaluate drug selectivity in an unbiased, comprehensive way. Efficient affinity purification procedures using immobilized kinase inhibitors combined with the sensitivity of mass spectrometry detection permit the mapping of drug targets on a proteome-wide scale. Data from this type of assessment can be used to set up tailor-made selectivity panels, which guide compound development in the context of the most relevant off-targets during lead optimization. In cases in which identified alternative targets are of validated clinical relevance, chemical proteomics provides the opportunity to repeatedly exploit a once established kinase inhibitor principle for additional target kinases and can thereby dramatically shorten the time toward highly selective, preclinical candidates. Moreover, the identification of alternative targets for preclinical or clinical drugs can provide new insights into their cellular modes of action, which might help to define those disease settings in which the most beneficial therapeutic effect is likely to occur.


Subject(s)
Enzyme Inhibitors/pharmacology , Protein Kinase Inhibitors , Proteomics/methods , Animals , Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/chemistry , Enzymes, Immobilized/chemistry , Humans , Protein Kinases/metabolism
14.
Mol Cell Proteomics ; 3(5): 490-500, 2004 May.
Article in English | MEDLINE | ID: mdl-14769951

ABSTRACT

Bisindolylmaleimide compounds such as GF109203X are potent inhibitors of protein kinase C (PKC) activity. Although bisindolylmaleimides are not entirely selective for PKC and are known to inhibit a few other protein kinases, these reagents have been extensively used to study the functional roles of PKC family enzymes in cellular signal transduction for more than a decade. Here, we establish a proteomics approach to gain further insights into the cellular effects of this compound class. Functional immobilization of suitable bisindolylmaleimide analogues in combination with the specific purification of cellular binding proteins by affinity chromatography led to the identification of several known and previously unknown enzyme targets. Subsequent in vitro binding and activity assays confirmed the protein kinases Ste20-related kinase and cyclin-dependent kinase 2 (CDK2) and the non-protein kinases adenosine kinase and quinone reductase type 2 as novel targets of bisindolylmaleimide inhibitors. As observed specifically for CDK2, minor chemical variation of the ligand by immobilizing the closely related bisindolylmaleimides III, VIII, and X dramatically affected target binding. These observed changes in affinity correlated with both the measured IC(50) values for in vitro CDK2 inhibition and results from molecular docking into the CDK2 crystal structure. Moreover, the conditions for affinity purification could be adapted in a way that immobilized bisindolylmaleimide III selectively interacted with either PKC alpha or ribosomal S6 protein kinase 1 only after activation of these kinases. Thus, we have established an efficient technique for the rapid identification of cellular bisindolylmaleimide targets and further demonstrate the comparative selectivity profiling of closely related kinase inhibitors within a cellular proteome.


Subject(s)
CDC2-CDC28 Kinases/metabolism , Indoles/pharmacology , Maleimides/pharmacology , Protein Kinase C/metabolism , Protein Kinases/metabolism , Proteomics , Animals , CDC2-CDC28 Kinases/antagonists & inhibitors , COS Cells , Cells, Cultured , Chlorocebus aethiops , Chromatography, Affinity , Cyclin-Dependent Kinase 2 , Enzyme Inhibitors/pharmacology , HeLa Cells , Humans , Models, Molecular , Protein Kinase C/antagonists & inhibitors , Protein Kinase C-alpha , Protein Kinase Inhibitors , Quinone Reductases/antagonists & inhibitors , Quinone Reductases/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors , Ribosomal Protein S6 Kinases, 90-kDa/metabolism
15.
Cell Cycle ; 3(4): 393-5, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14752275

ABSTRACT

Small molecule inhibitors of protein kinases have become highly popular tools in signal transduction research, despite the fact that rather limited data about their respective selectivities have been available. We established an efficient chemical proteomics method to characterize the cellular targets of the widely used inhibitor SB 203580, which was deemed to be rather specific for p38 kinase. Our results revealed several protein kinases as high affinity targets of SB 203580 and therefore imply a far more complicated cellular mode of action of this inhibitor than previously assumed. This raises the important question whether a lack of selectivity is inherent to many other "specific" inhibitors of protein kinases and warrants their evaluation employing experimental approaches adapted from our described proteomic technique.


Subject(s)
Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Pyridines/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Chromatography, Affinity , Drosophila , Humans , Phosphorylation , Proteome , Signal Transduction , Ultraviolet Rays , p38 Mitogen-Activated Protein Kinases
16.
Proc Natl Acad Sci U S A ; 100(26): 15434-9, 2003 Dec 23.
Article in English | MEDLINE | ID: mdl-14668439

ABSTRACT

Small molecule inhibitors of protein kinases are widely used in signal transduction research and are emerging as a major class of drugs. Although interpretation of biological results obtained with these reagents critically depends on their selectivity, efficient methods for proteome-wide assessment of kinase inhibitor selectivity have not yet been reported. Here, we address this important issue and describe a method for identifying targets of the widely used p38 kinase inhibitor SB 203580. Immobilization of a suitable SB 203580 analogue and thoroughly optimized biochemical conditions for affinity chromatography permitted the dramatic enrichment and identification of several previously unknown protein kinase targets of SB 203580. In vitro kinase assays showed that cyclin G-associated kinase (GAK) and CK1 were almost as potently inhibited as p38alpha whereas RICK [Rip-like interacting caspase-like apoptosis-regulatory protein (CLARP) kinase/Rip2/CARDIAK] was even more sensitive to inhibition by SB 203580. The cellular kinase activity of RICK, a known signal transducer of inflammatory responses, was already inhibited by submicromolar concentrations of SB 203580 in intact cells. Therefore, our results warrant a reevaluation of the vast amount of data obtained with SB 203580 and might have significant implications on the development of p38 inhibitors as antiinflammatory drugs. Based on the procedures described here, efficient affinity purification techniques can be developed for other protein kinase inhibitors, providing crucial information about their cellular modes of action.


Subject(s)
Enzyme Inhibitors/pharmacology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/chemistry , Protein Kinase Inhibitors , Proteomics , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , HeLa Cells , Humans , Mass Spectrometry , Mitogen-Activated Protein Kinases/isolation & purification , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Transfection , p38 Mitogen-Activated Protein Kinases
17.
Nucleic Acids Res ; 31(21): e128, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-14576328

ABSTRACT

Faced with the current wealth of genomic data, it is essential to have robust and reliable methods of converting DNA sequences into their functional gene products. We demonstrate here that when conditions are established that take advantage of the replication-associated virus amplification, the virus-induced shutdown of host protein synthesis as well as the activation of signalling pathways that normally occur during virus replication, adenovirus biology can be exploited to generate a potent kinase expression system. Residual virus in the protein production has always been a limitation for adenovirus systems and we describe a DNA intercalator/ultraviolet light treatment that eliminates residual adenovirus in protein preparations that has no deleterious effect on enzyme activity. The use of mammalian cells in combination with adenovirus generated a variety of active enzymes which could not be produced in Escherichia coli or baculovirus-infected insect cells. Thus, the utility of adenovirus-mediated enzyme expression as a versatile alternative to established protein production technologies is demonstrated.


Subject(s)
Adenoviridae/physiology , Mammals , Protein Kinases/biosynthesis , Protein Kinases/genetics , Virus Replication , Adenoviridae/drug effects , Adenoviridae/genetics , Adenoviridae/radiation effects , Animals , Baculoviridae/genetics , Baculoviridae/physiology , Cell Line , Chromatography, Affinity , Drug Contamination , Enzyme Activation , Genetic Engineering , Humans , Intercalating Agents/pharmacology , Mammals/virology , Multienzyme Complexes/genetics , Multienzyme Complexes/isolation & purification , Multienzyme Complexes/metabolism , Protein Kinases/isolation & purification , Protein Kinases/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Transfection , Ultraviolet Rays
18.
J Biol Chem ; 277(49): 47248-56, 2002 Dec 06.
Article in English | MEDLINE | ID: mdl-12374796

ABSTRACT

The N terminus of the human MUC2 mucin (amino acids 1-1397) has been expressed as a recombinant tagged protein in Chinese hamster ovary cells. The intracellular form was found to be an endoglycosidase H-sensitive monomer, whereas the secreted form was an oligomer that gave monomers upon disulfide bond reduction. The secreted MUC2 N terminus contained a trypsin-resistant core fragment. Edman sequencing and mass spectrometry of the peptides obtained localized this core fragment to the C-terminal end of the recombinant protein. This core retained its oligomeric nature with an apparent mass of approximately 240 kDa. Upon reduction, peptides of approximately 85 kDa were found, suggesting that the N terminus forms trimers. This interpretation was also supported by gel electrophoresis and gel filtration of the intact MUC2 N terminus. Electron microscopy revealed three globular domains each linked via an extended and flexible region to a central part in a trefoil-like manner. Immunostaining with gold-labeled antibodies localized the N-terminal end to the three globular structures, and the antibodies directed against the Myc and green fluorescent protein tags attached at the C terminus localized these to the stalk side of the central trefoil. The N terminus of the MUC2 mucin is thus assembled into trimers that contain proteolytically stable parts, suggesting that MUC2 can only be partly degraded by intestinal proteases and thus is able to maintain a mucin network protecting the intestine.


Subject(s)
Mucins/chemistry , Amino Acid Sequence , Animals , CHO Cells , Chromatography, Gel , Cricetinae , Dimerization , Disulfides , Electrophoresis, Polyacrylamide Gel , Genetic Vectors , Glycosylation , Hydrofluoric Acid/pharmacology , Immunoblotting , Microscopy, Electron , Molecular Sequence Data , Mucin-2 , Mucins/metabolism , Mutagenesis, Site-Directed , Peptides/chemistry , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Spectrometry, Mass, Electrospray Ionization , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Trypsin/pharmacology
19.
Plant Cell ; 14(5): 1147-60, 2002 May.
Article in English | MEDLINE | ID: mdl-12034903

ABSTRACT

The alga Volvox carteri is one of the simplest multicellular organisms, yet it has a surprisingly complex extracellular matrix (ECM), making Volvox suitable as a model system in which to study ECM self-assembly. Here, we analyze the primary structures and post-translational modifications of two main ECM components synthesized in response to sexual induction as well as wounding. These proteins are members of the pherophorin family with as yet unknown properties. They contain polyhydroxyproline spacers as long as 500 and 2750 residues. Even the highly purified proteins retain the capacity to self-assemble and cross-link, producing an insoluble fibrous network in an apparently autocatalytic reaction. This pherophorin-based network is located within the deep zone of the ECM. A molecular genetic search for additional members of the pherophorin family indicates that at least nine different pherophorin species can be expected to serve as precursors for ECM substructures. Therefore, the highly diversified members of the pherophorin family represent region-specific morphological building blocks for ECM assembly and cross-linking.


Subject(s)
Algal Proteins/genetics , Chlorophyta/genetics , Extracellular Matrix Proteins/genetics , Extracellular Matrix/metabolism , Glycoproteins/metabolism , Plant Proteins , Adaptation, Physiological , Algal Proteins/metabolism , Amino Acid Sequence , Chlorophyta/metabolism , DNA, Complementary/chemistry , DNA, Complementary/genetics , Extracellular Matrix Proteins/metabolism , Glycoproteins/genetics , Glycosylation , Molecular Sequence Data , Multigene Family/genetics , Peptides/metabolism , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Stress, Mechanical
SELECTION OF CITATIONS
SEARCH DETAIL
...