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










Publication year range
1.
J Med Chem ; 67(11): 8569-8584, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38758695

ABSTRACT

The trafficking chaperone PDE6D (or PDEδ) was proposed as a surrogate target for K-Ras, leading to the development of a series of inhibitors that block its prenyl binding pocket. These inhibitors suffered from low solubility and suspected off-target effects, preventing their clinical development. Here, we developed a highly soluble, low nanomolar PDE6D inhibitor (PDE6Di), Deltaflexin3, which has the lowest off-target activity as compared to three prominent reference compounds. Deltaflexin3 reduces Ras signaling and selectively decreases the growth of KRAS mutant and PDE6D-dependent cancer cells. We further show that PKG2-mediated phosphorylation of Ser181 lowers K-Ras binding to PDE6D. Thus, Deltaflexin3 combines with the approved PKG2 activator Sildenafil to more potently inhibit PDE6D/K-Ras binding, cancer cell proliferation, and microtumor growth. As observed previously, inhibition of Ras trafficking, signaling, and cancer cell proliferation remained overall modest. Our results suggest reevaluating PDE6D as a K-Ras surrogate target in cancer.


Subject(s)
Cell Proliferation , Cyclic Nucleotide Phosphodiesterases, Type 6 , Proto-Oncogene Proteins p21(ras) , Sildenafil Citrate , Humans , Cyclic Nucleotide Phosphodiesterases, Type 6/antagonists & inhibitors , Cyclic Nucleotide Phosphodiesterases, Type 6/metabolism , Sildenafil Citrate/pharmacology , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/metabolism , Cell Proliferation/drug effects , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Mutation , Animals , Structure-Activity Relationship , Phosphodiesterase Inhibitors/pharmacology , Phosphodiesterase Inhibitors/chemistry , Phosphodiesterase Inhibitors/chemical synthesis
2.
Biochem Soc Trans ; 51(1): 447-456, 2023 02 27.
Article in English | MEDLINE | ID: mdl-36688434

ABSTRACT

RAS drug development has made enormous strides in the past ten years, with the first direct KRAS inhibitor being approved in 2021. However, despite the clinical success of covalent KRAS-G12C inhibitors, we are immediately confronted with resistances as commonly found with targeted drugs. Previously believed to be undruggable due to its lack of obvious druggable pockets, a couple of new approaches to hit this much feared oncogene have now been carved out. We here concisely review these approaches to directly target four druggable sites of RAS from various angles. Our analysis focuses on the lessons learnt during the development of allele-specific covalent and non-covalent RAS inhibitors, the potential of macromolecular binders to facilitate the discovery and validation of targetable sites on RAS and finally an outlook on a future that may engage more small molecule binders to become drugs. We foresee that the latter could happen mainly in two ways: First, non-covalent small molecule inhibitors may be derived from the development of covalent binders. Second, reversible small molecule binders could be utilized for novel targeting modalities, such as degraders of RAS. Provided that degraders eliminate RAS by recruiting differentially expressed E3-ligases, this approach could enable unprecedented tissue- or developmental stage-specific destruction of RAS with potential advantages for on-target toxicity. We conclude that novel creative ideas continue to be important to exterminate RAS in cancer and other RAS pathway-driven diseases, such as RASopathies.


Subject(s)
Antineoplastic Agents , Neoplasms , Humans , Proto-Oncogene Proteins p21(ras) , Antineoplastic Agents/therapeutic use , Oncogenes , Neoplasms/genetics , Genes, ras , Mutation
3.
Pharmaceuticals (Basel) ; 15(2)2022 Jan 31.
Article in English | MEDLINE | ID: mdl-35215292

ABSTRACT

The multi-target effects of natural products allow us to fight complex diseases like cancer on multiple fronts. Unlike docking techniques, network-based approaches such as genome-scale metabolic modelling can capture multi-target effects. However, the incompleteness of natural product target information reduces the prediction accuracy of in silico gene knockout strategies. Here, we present a drug selection workflow based on context-specific genome-scale metabolic models, built from the expression data of cancer cells treated with natural products, to predict cell viability. The workflow comprises four steps: first, in silico single-drug and drug combination predictions; second, the assessment of the effects of natural products on cancer metabolism via the computation of a dissimilarity score between the treated and control models; third, the identification of natural products with similar effects to the approved drugs; and fourth, the identification of drugs with the predicted effects in pathways of interest, such as the androgen and estrogen pathway. Out of the initial 101 natural products, nine candidates were tested in a 2D cell viability assay. Bruceine D, emodin, and scutellarein showed a dose-dependent inhibition of MCF-7 and Hs 578T cell proliferation with IC50 values between 0.7 to 65 µM, depending on the drug and cell line. Bruceine D, extracted from Brucea javanica seeds, showed the highest potency.

4.
Cell Commun Signal ; 19(1): 22, 2021 02 22.
Article in English | MEDLINE | ID: mdl-33618712

ABSTRACT

BACKGROUND: Metastasis is the predominant cause for cancer morbidity and mortality accounting for approximatively 90% of cancer deaths. The actin-bundling protein L-plastin has been proposed as a metastatic marker and phosphorylation on its residue Ser5 is known to increase its actin-bundling activity. We recently showed that activation of the ERK/MAPK signalling pathway leads to L-plastin Ser5 phosphorylation and that the downstream kinases RSK1 and RSK2 are able to directly phosphorylate Ser5. Here we investigate the involvement of the PI3K pathway in L-plastin Ser5 phosphorylation and the functional effect of this phosphorylation event in breast cancer cells. METHODS: To unravel the signal transduction network upstream of L-plastin Ser5 phosphorylation, we performed computational modelling based on immunoblot analysis data, followed by experimental validation through inhibition/overexpression studies and in vitro kinase assays. To assess the functional impact of L-plastin expression/Ser5 phosphorylation in breast cancer cells, we either silenced L-plastin in cell lines initially expressing endogenous L-plastin or neoexpressed L-plastin wild type and phosphovariants in cell lines devoid of endogenous L-plastin. The established cell lines were used for cell biology experiments and confocal microscopy analysis. RESULTS: Our modelling approach revealed that, in addition to the ERK/MAPK pathway and depending on the cellular context, the PI3K pathway contributes to L-plastin Ser5 phosphorylation through its downstream kinase SGK3. The results of the transwell invasion/migration assays showed that shRNA-mediated knockdown of L-plastin in BT-20 or HCC38 cells significantly reduced cell invasion, whereas stable expression of the phosphomimetic L-plastin Ser5Glu variant led to increased migration and invasion of BT-549 and MDA-MB-231 cells. Finally, confocal image analysis combined with zymography experiments and gelatin degradation assays provided evidence that L-plastin Ser5 phosphorylation promotes L-plastin recruitment to invadopodia, MMP-9 activity and concomitant extracellular matrix degradation. CONCLUSION: Altogether, our results demonstrate that L-plastin Ser5 phosphorylation increases breast cancer cell invasiveness. Being a downstream molecule of both ERK/MAPK and PI3K/SGK pathways, L-plastin is proposed here as a potential target for therapeutic approaches that are aimed at blocking dysregulated signalling outcome of both pathways and, thus, at impairing cancer cell invasion and metastasis formation. Video abstract.


Subject(s)
Breast Neoplasms/metabolism , Membrane Glycoproteins/metabolism , Microfilament Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinases/metabolism , Breast Neoplasms/pathology , Cell Line , Cell Movement , Female , Humans , Membrane Glycoproteins/genetics , Microfilament Proteins/genetics , Models, Biological , Neoplasm Invasiveness , Phosphorylation , Serine/metabolism
5.
Int Rev Cell Mol Biol ; 355: 109-154, 2020.
Article in English | MEDLINE | ID: mdl-32859369

ABSTRACT

The dynamic organization of the actin cytoskeleton into bundles and networks is orchestrated by a large variety of actin-binding proteins. Among them, the actin-bundling protein L-plastin is normally expressed in hematopoietic cells, where it is involved in the immune response. However, L-plastin is also often ectopically expressed in malignant cancer cells of non-hematopoietic origin and is even considered as a marker for cancer progression. Post-translational modification modulates L-plastin activity. In particular, L-plastin Ser5 phosphorylation has been shown to be important for the immune response in leukocytes as well as for invasion and metastasis formation of carcinoma cells. This chapter discusses the physiological and pathological role of L-plastin with a special focus on the importance of L-plastin Ser5 phosphorylation for the protein functions. The potential use of Ser5 phosphorylated L-plastin as a biomarker and/or therapeutic target will be evoked.


Subject(s)
Immunity , Microfilament Proteins/metabolism , Neoplasms/metabolism , Actin Cytoskeleton/metabolism , Humans , Microfilament Proteins/immunology , Phosphorylation , Protein Processing, Post-Translational , Signal Transduction
6.
FASEB J ; 31(11): 5019-5035, 2017 11.
Article in English | MEDLINE | ID: mdl-28768720

ABSTRACT

Within the kidney, angiotensin II (AngII) targets different cell types in the vasculature, tubuli, and glomeruli. An important part of the renal filtration barrier is composed of podocytes with their actin-rich foot processes. In this study, we used stable isotope labeling with amino acids in cell culture coupled to mass spectrometry to characterize relative changes in the phosphoproteome of human podocytes in response to short-term treatment with AngII. In 4 replicates, we identified a total of 17,956 peptides that were traceable to 2081 distinct proteins. Bioinformatic analyses revealed that among the increasingly phosphorylated peptides are predominantly peptides that are related to actin filaments, cytoskeleton, lamellipodia, mammalian target of rapamycin, and MAPK signaling. Among others, this screening approach highlighted the increased phosphorylation of actin-bundling protein, l-plastin (LCP1). AngII-dependent phosphorylation of LCP1 in cultured podocytes was mediated by the kinases ERK, p90 ribosomal S6 kinase, PKA, or PKC. LCP1 phosphorylation increased filopodia formation. In addition, treatment with AngII led to LCP1 redistribution to the cell margins, membrane ruffling, and formation of lamellipodia. Our data highlight the importance of AngII-triggered actin cytoskeleton-associated signal transduction in podocytes.-Schenk, L. K., Möller-Kerutt, A., Klosowski, R., Wolters, D., Schaffner-Reckinger, E., Weide, T., Pavenstädt, H., Vollenbröker, B. Angiotensin II regulates phosphorylation of actin-associated proteins in human podocytes.


Subject(s)
Angiotensin II/pharmacology , MAP Kinase Signaling System , Microfilament Proteins/metabolism , Podocytes/metabolism , Angiotensin II/genetics , Angiotensin II/metabolism , Cell Line, Transformed , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Microfilament Proteins/genetics , Phosphorylation/drug effects , Phosphorylation/genetics , Protein Kinase C/genetics , Protein Kinase C/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Ribosomal Protein S6 Kinases, 90-kDa/metabolism
7.
PLoS One ; 12(5): e0177879, 2017.
Article in English | MEDLINE | ID: mdl-28542564

ABSTRACT

The focal adhesion protein testin is a modular scaffold and tumour suppressor that consists of an N-terminal cysteine rich (CR) domain, a PET domain of unknown function and three C-terminal LIM domains. Testin has been proposed to have an open and a closed conformation based on the observation that its N-terminal half and C-terminal half directly interact. Here we extend the testin conformational model by demonstrating that testin can also form an antiparallel homodimer. In support of this extended model we determined that the testin region (amino acids 52-233) harbouring the PET domain interacts with the C-terminal LIM1-2 domains in vitro and in cells, and assign a critical role to tyrosine 288 in this interaction.


Subject(s)
Cytoskeletal Proteins/chemistry , LIM Domain Proteins/chemistry , Protein Multimerization , Amino Acid Sequence , Cytoskeletal Proteins/metabolism , Humans , LIM Domain Proteins/metabolism , Protein Domains , RNA-Binding Proteins
8.
J Proteome Res ; 16(5): 2054-2071, 2017 05 05.
Article in English | MEDLINE | ID: mdl-28378594

ABSTRACT

The multimodular nature of many eukaryotic proteins underlies their temporal or spatial engagement in a range of protein cocomplexes. Using the multimodule protein testin (TES), we here report a proteomics approach to increase insight in cocomplex diversity. The LIM-domain containing and tumor suppressor protein TES is present at different actin cytoskeleton adhesion structures in cells and influences cell migration, adhesion and spreading. TES module accessibility has been proposed to vary due to conformational switching and variants of TES lacking specific domains target to different subcellular locations. By applying iMixPro AP-MS ("intelligent Mixing of Proteomes"-affinity purification-mass spectrometry) to a set of tagged-TES modular variants, we identified proteins residing in module-specific cocomplexes. The obtained distinct module-specific interactomes combine to a global TES interactome that becomes more extensive and richer in information. Applying pathway analysis to the module interactomes revealed expected actin-related canonical pathways and also less expected pathways. We validated two new TES cocomplex partners: TGFB1I1 and a short form of the glucocorticoid receptor. TES and TGFB1I1 are shown to oppositely affect cell spreading providing biological validity for their copresence in complexes since they act in similar processes.


Subject(s)
Cytoskeletal Proteins/metabolism , LIM Domain Proteins/metabolism , Actins/metabolism , Focal Adhesions/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Space/metabolism , Protein Multimerization , Proteomics/methods , RNA-Binding Proteins
9.
FASEB J ; 30(3): 1218-33, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26631483

ABSTRACT

Deregulated cell migration and invasion are hallmarks of metastatic cancer cells. Phosphorylation on residue Ser5 of the actin-bundling protein L-plastin activates L-plastin and has been reported to be crucial for invasion and metastasis. Here, we investigate signal transduction leading to L-plastin Ser5 phosphorylation using 4 human breast cancer cell lines. Whole-genome microarray analysis comparing cell lines with different invasive capacities and corresponding variations in L-plastin Ser5 phosphorylation level revealed that genes of the ERK/MAPK pathway are differentially expressed. It is noteworthy that in vitro kinase assays showed that ERK/MAPK pathway downstream ribosomal protein S6 kinases α-1 (RSK1) and α-3 (RSK2) are able to directly phosphorylate L-plastin on Ser5. Small interfering RNA- or short hairpin RNA-mediated knockdown and activation/inhibition studies followed by immunoblot analysis and computational modeling confirmed that ribosomal S6 kinase (RSK) is an essential activator of L-plastin. Migration and invasion assays showed that RSK knockdown led to a decrease of up to 30% of migration and invasion of MDA-MB-435S cells. Although the presence of L-plastin was not necessary for migration/invasion of these cells, immunofluorescence assays illustrated RSK-dependent recruitment of Ser5-phosphorylated L-plastin to migratory structures. Altogether, we provide evidence that the ERK/MAPK pathway is involved in L-plastin Ser5 phosphorylation in breast cancer cells with RSK1 and RSK2 kinases able to directly phosphorylate L-plastin residue Ser5.


Subject(s)
Breast Neoplasms/metabolism , MAP Kinase Signaling System/physiology , Actins/metabolism , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/physiology , Female , Humans , MCF-7 Cells , Membrane Glycoproteins/metabolism , Microfilament Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation/physiology , Ribosomes/metabolism , Serine/metabolism , Small-Conductance Calcium-Activated Potassium Channels/metabolism
10.
PLoS One ; 10(10): e0140511, 2015.
Article in English | MEDLINE | ID: mdl-26509500

ABSTRACT

Focal adhesions are integrin-based structures that link the actin cytoskeleton and the extracellular matrix. They play an important role in various cellular functions such as cell signaling, cell motility and cell shape. To ensure and fine tune these different cellular functions, adhesions are regulated by a large number of proteins. The LIM domain protein zyxin localizes to focal adhesions where it participates in the regulation of the actin cytoskeleton. Because of its interactions with a variety of binding partners, zyxin has been proposed to act as a molecular scaffold. Here, we studied the interaction of zyxin with such a partner: Tes. Similar to zyxin, Tes harbors three highly conserved LIM domains of which the LIM1 domain directly interacts with zyxin. Using different zyxin variants in pull-down assays and ectopic recruitment experiments, we identified the Tes binding site in zyxin and showed that four highly conserved amino acids are crucial for its interaction with Tes. Based upon these findings, we used a zyxin mutant defective in Tes-binding to assess the functional consequences of abrogating the zyxin-Tes interaction in focal adhesions. Performing fluorescence recovery after photobleaching, we showed that zyxin recruits Tes to focal adhesions and modulates its turnover in these structures. However, we also provide evidence for zyxin-independent localization of Tes to focal adhesions. Zyxin increases focal adhesion numbers and reduces focal adhesion lifetimes, but does so independent of Tes. Quantitative analysis showed that the loss of interaction between zyxin and Tes affects the process of cell spreading. We conclude that zyxin influences focal adhesion dynamics, that it recruits Tes and that this interaction is functional in regulating cell spreading.


Subject(s)
DNA-Binding Proteins/metabolism , Protein Interaction Mapping , Zyxin/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cell Count , Cell Movement , Conserved Sequence , Cytoskeletal Proteins , Focal Adhesions/metabolism , Humans , Kinetics , Mice , Mitochondria/metabolism , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , RNA-Binding Proteins , Structure-Activity Relationship , Zyxin/chemistry
11.
PLoS One ; 10(9): e0134952, 2015.
Article in English | MEDLINE | ID: mdl-26332040

ABSTRACT

Agonist-stimulated platelet activation triggers conformational changes of integrin αIIbß3, allowing fibrinogen binding and platelet aggregation. We have previously shown that an octapeptide, p1YMESRADR8, corresponding to amino acids 313-320 of the ß-ribbon extending from the ß-propeller domain of αIIb, acts as a potent inhibitor of platelet aggregation. Here we have performed in silico modelling analysis of the interaction of this peptide with αIIbß3 in its bent and closed (not swing-out) conformation and show that the peptide is able to act as a substitute for the ß-ribbon by forming a clasp restraining the ß3 hybrid and ßI domains in a closed conformation. The involvement of species-specific residues of the ß3 hybrid domain (E356 and K384) and the ß1 domain (E297) as well as an intrapeptide bond (pE315-pR317) were confirmed as important for this interaction by mutagenesis studies of αIIbß3 expressed in CHO cells and native or substituted peptide inhibitory studies on platelet functions. Furthermore, NMR data corroborate the above results. Our findings provide insight into the important functional role of the αIIb ß-ribbon in preventing integrin αIIbß3 head piece opening, and highlight a potential new therapeutic approach to prevent integrin ligand binding.


Subject(s)
Integrin alpha2/metabolism , Platelet Aggregation/physiology , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Membrane Glycoprotein IIb/metabolism , Fibrinogen/metabolism , Humans , Platelet Activation , Protein Binding
12.
PLoS One ; 5(2): e9210, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20169155

ABSTRACT

BACKGROUND: Initially detected in leukocytes and cancer cells derived from solid tissues, L-plastin/fimbrin belongs to a large family of actin crosslinkers and is considered as a marker for many cancers. Phosphorylation of L-plastin on residue Ser5 increases its F-actin binding activity and is required for L-plastin-mediated cell invasion. METHODOLOGY/PRINCIPAL FINDINGS: To study the kinetics of L-plastin and the impact of L-plastin Ser5 phosphorylation on L-plastin dynamics and actin turn-over in live cells, simian Vero cells were transfected with GFP-coupled WT-L-plastin, Ser5 substitution variants (S5/A, S5/E) or actin and analyzed by fluorescence recovery after photobleaching (FRAP). FRAP data were explored by mathematical modeling to estimate steady-state reaction parameters. We demonstrate that in Vero cell focal adhesions L-plastin undergoes rapid cycles of association/dissociation following a two-binding-state model. Phosphorylation of L-plastin increased its association rates by two-fold, whereas dissociation rates were unaffected. Importantly, L-plastin affected actin turn-over by decreasing the actin dissociation rate by four-fold, increasing thereby the amount of F-actin in the focal adhesions, all these effects being promoted by Ser5 phosphorylation. In MCF-7 breast carcinoma cells, phorbol 12-myristate 13-acetate (PMA) treatment induced L-plastin translocation to de novo actin polymerization sites in ruffling membranes and spike-like structures and highly increased its Ser5 phosphorylation. Both inhibition studies and siRNA knock-down of PKC isozymes pointed to the involvement of the novel PKC-delta isozyme in the PMA-elicited signaling pathway leading to L-plastin Ser5 phosphorylation. Furthermore, the L-plastin contribution to actin dynamics regulation was substantiated by its association with a protein complex comprising cortactin, which is known to be involved in this process. CONCLUSIONS/SIGNIFICANCE: Altogether these findings quantitatively demonstrate for the first time that L-plastin contributes to the fine-tuning of actin turn-over, an activity which is regulated by Ser5 phosphorylation promoting its high affinity binding to the cytoskeleton. In carcinoma cells, PKC-delta signaling pathways appear to link L-plastin phosphorylation to actin polymerization and invasion.


Subject(s)
Actins/metabolism , Cytoskeleton/metabolism , Membrane Glycoproteins/metabolism , Microfilament Proteins/metabolism , Algorithms , Amino Acid Substitution , Animals , Cell Line, Tumor , Chlorocebus aethiops , Cortactin/metabolism , Fluorescence Recovery After Photobleaching , Focal Adhesions/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Kinetics , Membrane Glycoproteins/genetics , Microfilament Proteins/genetics , Models, Biological , Phosphorylation/drug effects , Protein Binding , Protein Kinase C-delta/genetics , Protein Kinase C-delta/metabolism , Protein Transport/drug effects , RNA Interference , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Serine/genetics , Serine/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Transfection , Vero Cells
13.
J Cell Mol Med ; 14(6A): 1264-75, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19799649

ABSTRACT

We used a tumour necrosis factor (TNF)-alpha resistant breast adenocarcinoma MCF-7 cell line to investigate the involvement of the actin cytoskeleton in the mechanism of cell resistance to this cytokine. We found that TNF resistance correlates with the loss of cell epithelial properties and the gain of a mesenchymal phenotype, reminiscent of an epithelial-to-mesenchymal transition (EMT). Morphological changes were associated with a profound reorganization of the actin cytoskeleton and with a change in the repertoire of expressed actin cytoskeleton genes and EMT markers, as revealed by DNA microarray-based expression profiling. L-plastin, an F-actin cross-linking and stabilizing protein, was identified as one of the most significantly up-regulated genes in TNF-resistant cells. Knockdown of L-plastin in these cells revealed its crucial role in conferring TNF resistance. Importantly, overexpression of wild-type L-plastin in TNF-sensitive MCF-7 cells was sufficient to protect them against TNF-mediated cell death. Furthermore, we found that this effect is dependent on serine-5 phosphorylation of L-plastin and that non-conventional protein kinase C isoforms and the ceramide pathway may regulate its phosphorylation state. The protective role of L-plastin was not restricted to TNF-alpha resistant MCF-7 cells because a correlation between the expression of L-plastin and the resistance to TNF-alpha was observed in other breast cancer cell lines. Together, our study discloses a novel unexpected role of the actin bundling protein L-plastin as a cell protective protein against TNF-cytotoxicity.


Subject(s)
Actin Cytoskeleton/metabolism , Breast Neoplasms/metabolism , Cross-Linking Reagents/metabolism , Drug Resistance, Neoplasm/drug effects , Membrane Glycoproteins/metabolism , Microfilament Proteins/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Actins/metabolism , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Death/drug effects , Cell Line, Tumor , Ceramides/metabolism , Cytoskeleton/drug effects , Cytoskeleton/genetics , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Genes, Neoplasm/genetics , Humans , Mesoderm/drug effects , Mesoderm/pathology , Phenotype , Phosphorylation/drug effects , Protein Kinase C-delta/metabolism , Sphingomyelins/metabolism
14.
J Biol Chem ; 283(35): 24212-23, 2008 Aug 29.
Article in English | MEDLINE | ID: mdl-18577523

ABSTRACT

Talin establishes a major link between integrins and actin filaments and contains two distinct integrin binding sites: one, IBS1, located in the talin head domain and involved in integrin activation and a second, IBS2, that maps to helix 50 of the talin rod domain and is essential for linking integrin beta subunits to the cytoskeleton ( Moes, M., Rodius, S., Coleman, S. J., Monkley, S. J., Goormaghtigh, E., Tremuth, L., Kox, C., van der Holst, P. P., Critchley, D. R., and Kieffer, N. (2007) J. Biol. Chem. 282, 17280-17288 ). Through the combined approach of mutational analysis of the beta3 integrin cytoplasmic tail and the talin rod IBS2 site, SPR binding studies, as well as site-specific antibody inhibition experiments, we provide evidence that the integrin beta3-talin rod interaction relies on a helix-helix association between alpha-helix 50 of the talin rod domain and the membrane-proximal alpha-helix of the beta3 integrin cytoplasmic tail. Moreover, charge complementarity between the highly conserved talin rod IBS2 lysine residues and integrin beta3 glutamic acid residues is necessary for this interaction. Our results support a model in which talin IBS2 binds to the same face of the beta3 subunit cytoplasmic helix as the integrin alphaIIb cytoplasmic tail helix, suggesting that IBS2 can only interact with the beta3 subunit following integrin activation.


Subject(s)
Integrin beta3/metabolism , Models, Molecular , Talin/metabolism , Actin Cytoskeleton/chemistry , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Humans , Integrin beta3/chemistry , Integrin beta3/genetics , Peptide Mapping/methods , Platelet Membrane Glycoprotein IIb/chemistry , Platelet Membrane Glycoprotein IIb/genetics , Platelet Membrane Glycoprotein IIb/metabolism , Protein Binding/physiology , Protein Structure, Secondary/physiology , Protein Structure, Tertiary/physiology , Talin/chemistry , Talin/genetics
15.
Blood ; 111(7): 3407-14, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18065693

ABSTRACT

We report a 3-generation pedigree with 5 individuals affected with a dominantly inherited macrothrombocytopenia. All 5 carry 2 nonsynonymous mutations resulting in a D723H mutation in the beta3 integrin and a P53L mutation in glycoprotein (GP) Ibalpha. We show that GPIbalpha-L53 is phenotypically silent, being also present in 3 unaffected pedigree members and in 7 of 1639 healthy controls. The beta3-H723 causes constitutive, albeit partial, activation of the alphaIIbbeta3 complex by disruption of the highly conserved cytoplasmic salt bridge with arginine 995 in the alphaIIb integrin as evidenced by increased PAC-1 but not fibrinogen binding to the patients' resting platelets. This was confirmed in CHO alphaIIbbeta3-H723 transfectants, which also exhibited increased PAC-1 binding, increased adhesion to von Willebrand factor (VWF) in static conditions and to fibrinogen under shear stress. Crucially, we show that in the presence of fibrinogen, alphaIIbbeta3-H723, but not wild-type alphaIIbbeta3, generates a signal that leads to the formation of proplatelet-like protrusions in transfected CHO cells. Abnormal proplatelet formation was confirmed in the propositus's CD34+ stem cell-derived megakaryocytes. We conclude that the constitutive activation of the alphaIIbbeta3-H723 receptor causes abnormal proplatelet formation, leading to incorrect sizing of platelets and the thrombocytopenia observed in the pedigree.


Subject(s)
Blood Platelets/metabolism , Integrin beta3/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Polymorphism, Single Nucleotide , Thrombocytopenia/genetics , Thrombocytopenia/metabolism , Adult , Animals , Blood Platelets/pathology , CHO Cells , Cricetinae , Cricetulus , Female , Fibrinogen/genetics , Fibrinogen/metabolism , Gene Expression , Humans , Integrin beta3/metabolism , Male , Megakaryocytes/metabolism , Megakaryocytes/pathology , Mutation, Missense , Pedigree , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Platelet Glycoprotein GPIb-IX Complex/genetics , Platelet Glycoprotein GPIb-IX Complex/metabolism , Protein Binding/genetics , Thrombocytopenia/pathology , Transfection , von Willebrand Factor/genetics , von Willebrand Factor/metabolism
16.
Eur J Cell Biol ; 85(3-4): 249-54, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16546569

ABSTRACT

Some RGD-type integrins rely on a synergistic site in addition to the canonical RGD site for ligand binding. However, the precise involvement of each of these recognition sites during cell adhesion is still unclear. Here we review recent investigations on integrin alphaIIbbeta3-mediated cell adhesion to immobilized fibrinogen providing evidence that the fibrinogen synergy gamma(400-411) sequence by itself promotes cell attachment by initiating alphaIIbbeta3 clustering and recruitment of intracellular proteins to focal complexes, while the RGD motif subsequently acts as a molecular switch on the beta3 subunit to induce a conformational change necessary for RhoA activation and full cell spreading.


Subject(s)
Fibrinogen/chemistry , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , rhoA GTP-Binding Protein/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , CHO Cells , Cell Adhesion , Cricetinae , Fibrinogen/genetics , Fibrinogen/metabolism , Humans , Ligands , Models, Biological , Molecular Sequence Data , Oligopeptides/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Protein Structure, Tertiary , rhoA GTP-Binding Protein/genetics
17.
J Biol Chem ; 280(39): 33610-9, 2005 Sep 30.
Article in English | MEDLINE | ID: mdl-15955823

ABSTRACT

A number of RGD-type integrins rely on a synergistic site in addition to the canonical RGD site for ligand binding and signaling, although it is still unclear whether these two recognition sites function independently, synergistically, or competitively. Experimental evidence has suggested that fibrinogen binding to the RGD-type integrin alphaIIbbeta3 occurs exclusively through the synergistic gamma(400-411) sequence, thus questioning the functional role of the RGD recognition site. Here we have investigated the respective role of the fibrinogen gamma(400-411) sequence and the RGD motif in the molecular events leading to ligand-induced alphaIIbbeta3-dependent Chinese hamster ovary (CHO) cell or platelet spreading, by using intact fibrinogen and well characterized plasmin-generated fibrinogen fragments containing either the RGD motif (fragment C) or the gamma(400-411) sequence (fragment D), and CHO cells expressing resting wild type (alphaIIbbeta3wt), constitutively active (alphaIIbbeta3T562N), or non-functional (alphaIIbbeta3D119Y) receptors. Our data provide evidence that the gamma(400-411) site by itself is able to initiate alphaIIbbeta3 clustering and recruitment of intracellular proteins to early focal complexes, mediating cell attachment, FAK phosphorylation, and Rac1 activation, while the RGD motif subsequently acts as a molecular switch on the beta3 subunit to trigger cell spreading. More importantly, we show that the premier functional role of the RGD site is not to reinforce cell attachment but, rather, to imprint a conformational change on the beta3 subunit leading to maximal RhoA activation and actin cytoskeleton organization in CHO cells as well as in platelets. Finally, alphaIIbbeta3-dependent RhoA stimulation and cell spreading, but not cell attachment, are Src-dependent and phosphoinositide 3-kinase-independent and are inhibited by the Src antagonist PP2.


Subject(s)
Fibrinogen/metabolism , Platelet Activation , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , rhoA GTP-Binding Protein/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Blood Platelets/metabolism , CHO Cells , Cell Adhesion , Cricetinae , Cricetulus , Enzyme Activation , Fibrinogen/chemistry , Fibrinolysin/metabolism , Flow Cytometry , Fluorescein-5-isothiocyanate , Fluorescent Dyes , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Kinetics , Ligands , Microscopy, Fluorescence , Mutation , Phalloidine/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Protein Conformation , Rhodamines , Transfection
18.
Thromb Haemost ; 91(4): 743-54, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15045136

ABSTRACT

Recombinant HPA-1a antibodies with Fc, mutated to remove destructive effector functions, have been developed as a potential therapy for fetomaternal alloimmune thrombocytopenia (FMAIT), via blockade of binding of human HPA-1a polyclonal antibodies to fetal HPA-1a1b platelets. We have assessed the effect of the IgG1 HPA-1a antibody B2G1 and two mutated derivatives in various functional assays in resting and agonist-stimulated platelets of the three HPA-1 genotypes. With HPA-1a1b platelets (fetal genotype), the antibodies did not activate signalling or CD62P expression in resting platelets, did not change in vitro bleeding time (IVBT), and did not inhibit platelet adhesion to collagen in flowing blood. Adhesion of HPA-1a1b platelets to fibrinogen was reduced by 20%, and aggregation induced by ADP by 50%, but collagen-related peptide (CRP-XL)-induced aggregation was normal. With HPA-1a1a platelets, aggregation to both ADP and CRP-XL was inhibited, with total blockade of adhesion to fibrinogen and of IVBT responses. Interestingly, a monovalent antibody fragment with identical specificity had no inhibitory effect on aggregation. In static adhesion assays using human alphaIIbbeta3 or alphaVbeta3 transfectants of HPA-1a (Leu(33)) phenotype, attachment to fibrinogen of the latter but not of the former was completely blocked by the HPA-1a antibodies. These observations are best explained by antibody-mediated blockade of the RGD binding site on beta3 by a mechanism of steric hindrance. As the effect on platelet function is modest with HPA-1a1b (fetal type) platelets, the mutated HPA-1a antibodies described here could be developed further for FMAIT therapy.


Subject(s)
Antibodies/pharmacology , Antigens, Human Platelet/immunology , Platelet Activation/immunology , Polymorphism, Single Nucleotide/immunology , Antibodies/genetics , Antibodies/immunology , Collagen/metabolism , Female , Fibrinogen/metabolism , Humans , Immunoglobulin G/genetics , Immunoglobulin G/pharmacology , Integrin alphaVbeta3/metabolism , Integrin beta3/immunology , Leucine , Platelet Function Tests , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Pregnancy , Protein Engineering , Recombinant Proteins
19.
Article in English | MEDLINE | ID: mdl-15544009

ABSTRACT

Integrins are major cell adhesion receptors which assume two important functions: first they act as anchoring molecules allowing firm cellular attachment to the extracellular matrix, and second they work as signalling receptors being able to transduce signals in both directions (outside-in and inside-out) through the plasma membrane. Their biological importance is determined by their involvement in many physiological phenomena. Furthermore, their implication in various diseases and their accessibility to drugs make them interesting therapeutic targets.


Subject(s)
Integrin beta3 , Biomarkers , Cardiovascular Diseases/therapy , Humans , Integrin beta3/chemistry , Integrin beta3/physiology , Platelet Aggregation , Thrombasthenia/therapy
20.
Blood ; 99(5): 1833-9, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11861302

ABSTRACT

A single nucleotide polymorphism (SNP) at position 196 in the beta 3 integrin causes a Leu33Pro substitution in the mature protein. Alloimmunization against the beta 3Leu33 form (human platelet antigen [HPA]-1a, Pl(A1), Zw(a)) in patients who are beta 3Pro33 homozygous (HPA-1b1b, Pl(A2A2), Zw(bb)) causes neonatal alloimmune thrombocytopenia, posttransfusion purpura, or refractoriness to platelet transfusion. Studies with recombinant proteins have demonstrated that amino acids 1 to 66 and 288 to 490 of the beta 3 integrin contribute to HPA-1a epitope formation. In determining the HPA-1a status of more than 6000 donors, we identified a donor with an HPA-1a(weak) phenotype and an HPA-1a1b genotype. The platelets from this donor had normal levels of surface alpha IIb beta 3 but reacted only weakly with monoclonal and polyclonal anti-HPA-1a by whole blood enzyme-linked immunosorbent assay (ELISA), flow cytometry, and sandwich ELISA. We reasoned that an alteration in the primary nucleotide sequence of the beta 3Leu33 allele of this donor was disrupting the HPA-1a epitope. In agreement with this hypothesis, sequencing platelet RNA-derived alpha IIb and beta 3 cDNA identified a novel G/A SNP at position 376 of the beta 3 integrin that encodes for an Arg93Gln replacement in the beta 3Leu33 allele. Coexpression of the beta 3Leu33Gln93 encoding cDNA in Chinese hamster ovary cells with human alpha IIb cDNA showed that the surface-expressed alpha IIb beta 3 reacted normally with beta 3 integrin-specific monoclonal antibodies but only weakly with monoclonal anti-HPA-1a. Our results show that an Arg93Gln mutation in the beta 3Leu33 encoding allele disrupts the HPA-1a epitope, suggesting that Arg93 contributes to the formation of the HPA-1a B-cell epitope.


Subject(s)
Antigens, CD/genetics , Antigens, Human Platelet/genetics , Platelet Membrane Glycoproteins/genetics , Polymorphism, Single Nucleotide/genetics , Amino Acid Substitution , Animals , Antibodies, Monoclonal/immunology , Antigen-Antibody Reactions , Antigens, Human Platelet/immunology , CHO Cells , Cricetinae , Epitopes, B-Lymphocyte/genetics , Epitopes, B-Lymphocyte/immunology , Genotype , Humans , Integrin beta3 , Mass Screening , Phenotype , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Sequence Analysis, DNA , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...