Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Sci Signal ; 12(565)2019 01 22.
Article in English | MEDLINE | ID: mdl-30670633

ABSTRACT

Drug-induced compensatory signaling and subsequent rewiring of the signaling pathways that support cell proliferation and survival promote the development of acquired drug resistance in tumors. Here, we sought to analyze the adaptive kinase response in cancer cells after distinct treatment with agents targeting human epidermal growth factor receptor 2 (HER2), specifically those that induce either only temporary cell cycle arrest or, alternatively, apoptosis in HER2-overexpressing cancers. We compared trastuzumab, ARRY380, the combination thereof, and a biparatopic, HER2-targeted designed ankyrin repeat protein (DARPin; specifically, 6L1G) and quantified the phosphoproteome by isobaric tagging using tandem mass tag liquid chromatography/tandem mass spectrometry (TMT LC-MS/MS). We found a specific signature of persistently phosphorylated tyrosine peptides after the nonapoptotic treatments, which we used to distinguish between different treatment-induced cancer cell fates. Next, we analyzed the activation of serine/threonine and tyrosine kinases after treatment using a bait peptide chip array and predicted the corresponding active kinases. Through a combined system-wide analysis, we identified a common adaptive kinase response program that involved the activation of focal adhesion kinase 1 (FAK1), protein kinase C-δ (PRKCD), and Ephrin (EPH) family receptors. These findings reveal potential targets to prevent adaptive resistance to HER2-targeted therapies.


Subject(s)
Breast Neoplasms/metabolism , Protein Kinases/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Signal Transduction/drug effects , Small Molecule Libraries/pharmacology , Trastuzumab/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Breast Neoplasms/pathology , Cell Line, Tumor , Chromatography, Liquid , Drug Resistance, Neoplasm/drug effects , Female , Humans , Phosphoproteins/metabolism , Protein Kinase Inhibitors/pharmacology , Proteome/metabolism , Receptor, ErbB-2/metabolism , Tandem Mass Spectrometry
2.
Nat Commun ; 7: 11672, 2016 06 03.
Article in English | MEDLINE | ID: mdl-27255951

ABSTRACT

Compensatory mechanisms, such as relief of AKT-ErbB3-negative feedback, are known to desensitize ErbB2-dependent tumours to targeted therapy. Here we describe an adaptation mechanism leading to reactivation of the PI3K/AKT pathway during trastuzumab treatment, which occurs independently of ErbB3 re-phosphorylation. This signalling bypass of phospho-ErbB3 operates in ErbB2-overexpressing cells via RAS-PI3K crosstalk and is attributable to active ErbB2 homodimers. As demonstrated by dual blockade of ErbB2/RAS and ErbB3 by means of pharmacological inhibition, RNA interference or by specific protein binders obstructing the RAS-p110α interaction, both routes must be blocked to prevent reactivation of the PI3K/AKT pathway. Applying these general principles, we developed biparatopic designed ankyrin repeat proteins (DARPins) trapping ErbB2 in a dimerization-incompetent state, which entail pan-ErbB inhibition and a permanent OFF state in the oncogenic signalling, thereby triggering extensive apoptosis in ErbB2-addicted tumours. Thus, these novel insights into mechanisms underlying network robustness provide a guide for overcoming adaptation response to ErbB2/ErbB3-targeted therapy.


Subject(s)
Breast Neoplasms/metabolism , Class I Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism , ras Proteins/metabolism , Ankyrin Repeat , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Apoptosis , Cell Line, Tumor , Cell Proliferation , Humans , Protein Interaction Maps , RNA Interference , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-3/antagonists & inhibitors , Signal Transduction , Trastuzumab/pharmacology , ras Proteins/antagonists & inhibitors
3.
Structure ; 21(11): 1979-91, 2013 Nov 05.
Article in English | MEDLINE | ID: mdl-24095059

ABSTRACT

Human epidermal growth factor receptor-2 (HER2) is a receptor tyrosine kinase directly linked to the growth of malignancies from various origins and a validated target for monoclonal antibodies and kinase inhibitors. Utilizing a new approach with designed ankyrin repeat proteins (DARPins) as alternative binders, we show that binding of two DARPins connected by a short linker, one targeting extracellular subdomain I and the other subdomain IV, causes much stronger cytotoxic effects on the HER2-addicted breast cancer cell line BT474, surpassing the therapeutic antibody trastuzumab. We determined crystal structures of these DARPins in complex with the respective subdomains. Detailed models of the full-length receptor, constrained by its rigid domain structures and its membrane anchoring, explain how the bispecific DARPins connect two membrane-bound HER2 molecules, distorting them such that they cannot form signaling-competent dimers with any EGFR family member, preventing any kinase dimerization, and thus leading to a complete loss of signaling.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Receptor, ErbB-2/metabolism , Recombinant Fusion Proteins/pharmacology , Animals , Ankyrin Repeat , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents, Hormonal/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Crystallography, X-Ray , Epitope Mapping , Humans , Models, Molecular , Protein Binding , Protein Structure, Quaternary , Protein Structure, Secondary , Protein Structure, Tertiary , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/chemistry , Recombinant Fusion Proteins/chemistry , Sf9 Cells , Signal Transduction , Spodoptera , Trastuzumab
4.
Methods Enzymol ; 503: 101-34, 2012.
Article in English | MEDLINE | ID: mdl-22230567

ABSTRACT

Designed ankyrin repeat proteins (DARPins) have been developed into a robust and versatile scaffold for binding proteins. High-affinity binders are routinely selected by ribosome display and phage display. DARPins have entered clinical trials and have found numerous uses in research, due to their high stability and robust folding, allowing many new molecular formats. We summarize the DARPin properties and highlight some biomedical applications. Protocols are given for labeling with dyes and polyethylene glycol, for quantitatively measuring binding to cell surface receptors by kinetics and thermodynamics, and for exploiting new engineering opportunities from using "click chemistry" with nonnatural amino acids.


Subject(s)
Ankyrin Repeat , Carrier Proteins/chemistry , Drug Design , Protein Engineering/methods , Proteins/therapeutic use , Affinity Labels/chemistry , Amino Acid Sequence , Animals , Biomedical Research , Click Chemistry , Escherichia coli/chemistry , Fluorescent Dyes/chemistry , Genetic Vectors/chemistry , Humans , Molecular Sequence Data , Peptide Library , Polyethylene Glycols/chemistry , Protein Binding , Protein Stability , Proteins/chemistry , Receptors, Cell Surface/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Ribosomes/chemistry , Thermodynamics , Titrimetry , Viruses/chemistry
5.
Cancer Res ; 70(4): 1595-605, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20124480

ABSTRACT

Slow-clearing, tumor-targeting proteins such as monoclonal antibodies typically exhibit high tumor accumulation but low tissue contrast, whereas intermediate-sized proteins such as scFvs show faster clearance but only moderate tumor accumulation. For both, tumor targeting does not seem to improve further above an optimal affinity. We show here that with very small high-affinity proteins such as designed ankyrin repeat proteins (DARPins), these limits can be overcome. We have systematically investigated the influence of molecular mass and affinity on tumor accumulation with DARPins with specificity for HER2 in SK-OV-3.ip nude mouse xenografts. DARPins with a mass of 14.5 kDa and affinities between 270 nmol/L and 90 pmol/L showed a strong correlation of tumor accumulation with affinity to HER2, with the highest affinity DARPin reaching 8% ID/g after 24 hours and 6.5% ID/g after 48 hours (tumor-to-blood ratio >60). Tumor autoradiographs showed good penetration throughout the tumor mass. Genetic fusion of two DARPins (30 kDa) resulted in significantly lower tumor accumulation, similar to values observed for scFvs, whereas valency had no influence on accumulation. PEGylation of the DARPins increased the circulation half-life, leading to higher tumor accumulation (13.4% ID/g after 24 hours) but lower tumor-to-blood ratios. Affinity was less important for tumor uptake of the PEGylated constructs. We conclude that two regimes exist for delivering high levels of drug to a tumor: small proteins with very high affinity, such as unmodified DARPins, and large proteins with extended half-life, such as PEGylated DARPins, in which the importance of affinity is less pronounced.


Subject(s)
Ankyrin Repeat , Drug Delivery Systems/methods , Neoplasms/drug therapy , Proteins/administration & dosage , Proteins/chemical synthesis , Animals , Ankyrin Repeat/physiology , Antibody Affinity , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Drug Design , Female , Humans , Immunoglobulin Fc Fragments/administration & dosage , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/therapeutic use , Mice , Mice, Nude , Molecular Weight , Protein Engineering/methods , Proteins/chemistry , Proteins/therapeutic use , Substrate Specificity/physiology , Treatment Outcome , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Mol Cell Biol ; 25(24): 11019-29, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16314523

ABSTRACT

NDR protein kinases are involved in the regulation of cell cycle progression and morphology. NDR1/NDR2 protein kinase is activated by phosphorylation on the activation loop phosphorylation site Ser281/Ser282 and the hydrophobic motif phosphorylation site Thr444/Thr442. Autophosphorylation of NDR is responsible for phosphorylation on Ser281/Ser282, whereas Thr444/Thr442 is targeted by an upstream kinase. Here we show that MST3, a mammalian Ste20-like protein kinase, is able to phosphorylate NDR protein kinase at Thr444/Thr442. In vitro, MST3 selectively phosphorylated Thr442 of NDR2, resulting in a 10-fold stimulation of NDR activity. MOB1A (Mps one binder 1A) protein further increased the activity, leading to a fully active kinase. In vivo, Thr442 phosphorylation after okadaic acid stimulation was potently inhibited by MST3KR, a kinase-dead mutant of MST3. Knockdown of MST3 using short hairpin constructs abolished Thr442 hydrophobic motif phosphorylation of NDR in HEK293F cells. We conclude that activation of NDR is a multistep process involving phosphorylation of the hydrophobic motif site Thr444/2 by MST3, autophosphorylation of Ser281/2, and binding of MOB1A.


Subject(s)
Carrier Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Motifs , Animals , COS Cells , Catalytic Domain , Chlorocebus aethiops , Cytoplasm/chemistry , Enzyme Activation , Humans , Hydrophobic and Hydrophilic Interactions , Intracellular Signaling Peptides and Proteins , Okadaic Acid/pharmacology , Phosphorylation , Protein Serine-Threonine Kinases/analysis , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/genetics , Threonine/metabolism
7.
J Biol Chem ; 279(34): 35510-7, 2004 Aug 20.
Article in English | MEDLINE | ID: mdl-15169778

ABSTRACT

The tumor suppressor p53 is commonly inhibited under conditions in which the phosphatidylinositide 3'-OH kinase/protein kinase B (PKB)Akt pathway is activated. Intracellular levels of p53 are controlled by the E3 ubiquitin ligase Mdm2. Here we show that PKB inhibits Mdm2 self-ubiquitination via phosphorylation of Mdm2 on Ser(166) and Ser(188). Stimulation of human embryonic kidney 293 cells with insulin-like growth factor-1 increased Mdm2 phosphorylation on Ser(166) and Ser(188) in a phosphatidylinositide 3'-OH kinase-dependent manner, and the treatment of both human embryonic kidney 293 and COS-1 cells with phosphatidylinositide 3'-OH kinase inhibitor LY-294002 led to proteasome-mediated Mdm2 degradation. Introduction of a constitutively active form of PKB together with Mdm2 into cells induced phosphorylation of Mdm2 at Ser(166) and Ser(188) and stabilized Mdm2 protein. Moreover, mouse embryonic fibroblasts lacking PKBalpha displayed reduced Mdm2 protein levels with a concomitant increase of p53 and p21(Cip1), resulting in strongly elevated apoptosis after UV irradiation. In addition, activation of PKB correlated with Mdm2 phosphorylation and stability in a variety of human tumor cells. These findings suggest that PKB plays a critical role in controlling of the Mdm2.p53 signaling pathway by regulating Mdm2 stability.


Subject(s)
Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Cell Line , Enzyme Stability , Humans , Mice , Mice, Knockout , Nuclear Proteins/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-mdm2 , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Ubiquitins
8.
J Biol Chem ; 279(34): 35228-35, 2004 Aug 20.
Article in English | MEDLINE | ID: mdl-15197186

ABSTRACT

NDR (nuclear Dbf2-related) kinase belongs to a family of kinases that is highly conserved throughout the eukaryotic world. We showed previously that NDR is regulated by phosphorylation and by the Ca(2+)-binding protein, S100B. The budding yeast relatives of Homo sapiens NDR, Cbk1, and Dbf2, were shown to interact with Mob2 (Mps one binder 2) and Mob1, respectively. This interaction is required for the activity and biological function of these kinases. In this study, we show that hMOB1, the closest relative of yeast Mob1 and Mob2, stimulates NDR kinase activity and interacts with NDR both in vivo and in vitro. The point mutations of highly conserved residues within the N-terminal domain of NDR reduced NDR kinase activity as well as human MOB1 binding. A novel feature of NDR kinases is an insert within the catalytic domain between subdomains VII and VIII. The amino acid sequence within this insert shows a high basic amino acid content in all of the kinases of the NDR family known to interact with MOB proteins. We show that this sequence is autoinhibitory, and our data indicate that the binding of human MOB1 to the N-terminal domain of NDR induces the release of this autoinhibition.


Subject(s)
Carrier Proteins/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Animals , Binding Sites/genetics , COS Cells , Carrier Proteins/genetics , Catalytic Domain , Cell Cycle Proteins/genetics , Chlorocebus aethiops , Enzyme Activation , Humans , Molecular Sequence Data , Phosphoproteins/genetics , Point Mutation , Protein Binding , Protein Serine-Threonine Kinases/chemistry , Protein Structure, Tertiary , Saccharomyces cerevisiae Proteins/genetics , Structure-Activity Relationship
9.
J Biol Chem ; 279(22): 23806-12, 2004 May 28.
Article in English | MEDLINE | ID: mdl-15037617

ABSTRACT

Nuclear Dbf2-related (NDR) protein kinases are a family of AGC group kinases that are involved in the regulation of cell division and cell morphology. We describe the cloning and characterization of the human and mouse NDR2, a second mammalian isoform of NDR protein kinase. NDR1 and NDR2 share 86% amino acid identity and are highly conserved between human and mouse. However, they differ in expression pattern; mouse Ndr1 is expressed mainly in spleen, lung and thymus, whereas mouse Ndr2 shows highest expression in the gastrointestinal tract. NDR2 is potently activated in cells following treatment with the protein phosphatase 2A inhibitor okadaic acid, which also results in phosphorylation on the activation segment residue Ser-282 and the hydrophobic motif residue Thr-442. We show that Ser-282 becomes autophosphorylated in vivo, whereas Thr-442 is targeted by an upstream kinase. This phosphorylation can be mimicked by replacing the hydrophobic motif of NDR2 with a PRK2-derived sequence, resulting in a constitutively active kinase. Similar to NDR1, the autophosphorylation of NDR2 protein kinase was stimulated in vitro by S100B, an EF-hand Ca(2+)-binding protein of the S100 family, suggesting that the two isoforms are regulated by the same mechanisms. Further we show a predominant cytoplasmic localization of ectopically expressed NDR2.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Proteins/metabolism , S100 Proteins/metabolism , Amino Acid Sequence , Animals , Gene Expression Regulation, Enzymologic , Humans , Mice , Molecular Sequence Data , Nerve Growth Factors , Nodal Signaling Ligands , Phosphorylation , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases/genetics , Proteins/genetics , S100 Calcium Binding Protein beta Subunit , Sequence Alignment
10.
FEBS Lett ; 546(1): 73-80, 2003 Jul 03.
Article in English | MEDLINE | ID: mdl-12829239

ABSTRACT

The nuclear Dbf2-related (NDR) family of protein serine/threonine kinases comprises mammalian NDR and large tumor suppressor (LATS) kinases, their orthologs from Drosophila melanogaster and Caenorhabditis elegans, and a number of related kinases from yeast and plants. The members of this family were independently implicated in various aspects of the control of cell division and morphogenesis. They are crucial regulators of the actin and tubulin cytoskeletal organization during polarized growth and cytokinesis in yeast. Furthermore, they are key players in control of proliferation and morphology of many cell types in D. melanogaster and C. elegans. In mammalians, the LATS kinase is a tumor suppressor, negatively regulating the cyclin-dependent kinase CDK1, cell proliferation rate, and modulating cell survival.


Subject(s)
Cell Cycle , Morphogenesis , Protein Serine-Threonine Kinases/metabolism , Amino Acid Sequence , Animals , Cell Polarity , Conserved Sequence , Genes, Tumor Suppressor , Humans , Models, Biological , Molecular Sequence Data , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Sequence Homology, Amino Acid
11.
J Biol Chem ; 278(9): 6710-8, 2003 Feb 28.
Article in English | MEDLINE | ID: mdl-12493777

ABSTRACT

NDR1 (nuclear Dbf2-related) is a serine/threonine protein kinase belonging to subfamily of kinases implicated in the regulation of cell division and morphology. Previously, we demonstrated that the activity of NDR1 is controlled by phosphorylation of two regulatory residues, Ser-281 and Thr-444. Moreover, we found that NDR1 becomes activated through a direct interaction with EF-hand Ca(2+)-binding proteins of the S100 family. In this work, we characterize this regulatory mechanism in detail. We found that NDR1 autophosphorylates in vitro predominantly on Ser-281 and to a lesser extent on Thr-74 and Thr-444. All of these residues proved to be crucial also for NDR1 activity in vivo; however, in contrast to Ser-281 and Thr-444, Thr-74 seems to be involved only in binding to S100B rather than directly regulating NDR1 activity per se. When we added Ca(2+)/S100B, we observed an increased autophosphorylation on Ser-281 and Thr-444, resulting in stimulation of NDR1 activity in vitro. Using phosphospecific antibodies, we found that Ser-281 also becomes autophosphorylated in vivo, whereas Thr-444 is targeted predominantly by an as yet unidentified upstream kinase. Significantly, the Ca(2+)-chelating agent BAPTA-AM suppressed the activity and phosphorylation of NDR1 on both Ser-281 and Thr-444, and specifically, these effects were reversed when we added the sarcoplasmic-endoplasmic reticulum Ca(2+) ATPase pump inhibitor thapsigargin.


Subject(s)
Calcium/metabolism , Drosophila Proteins , Protein Serine-Threonine Kinases/metabolism , Amino Acid Sequence , Animals , Blotting, Western , COS Cells , Cytoplasm/metabolism , Gene Expression Regulation , Glutathione Transferase/metabolism , Humans , Mass Spectrometry , Models, Biological , Molecular Sequence Data , Nerve Growth Factors/metabolism , Phosphorylation , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , S100 Calcium Binding Protein beta Subunit , S100 Proteins/metabolism , Sequence Homology, Amino Acid , Serine/chemistry , Serine/metabolism , Threonine/metabolism , Time Factors , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...