Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 12(1): 6967, 2021 11 29.
Article in English | MEDLINE | ID: mdl-34845227

ABSTRACT

Breast cancer is now globally the most frequent cancer and leading cause of women's death. Two thirds of breast cancers express the luminal estrogen receptor-positive (ERα + ) phenotype that is initially responsive to antihormonal therapies, but drug resistance emerges. A major barrier to the understanding of the ERα-pathway biology and therapeutic discoveries is the restricted repertoire of luminal ERα + breast cancer models. The ERα + phenotype is not stable in cultured cells for reasons not fully understood. We examine 400 patient-derived breast epithelial and breast cancer explant cultures (PDECs) grown in various three-dimensional matrix scaffolds, finding that ERα is primarily regulated by the matrix stiffness. Matrix stiffness upregulates the ERα signaling via stress-mediated p38 activation and H3K27me3-mediated epigenetic regulation. The finding that the matrix stiffness is a central cue to the ERα phenotype reveals a mechanobiological component in breast tissue hormonal signaling and enables the development of novel therapeutic interventions. Subject terms: ER-positive (ER + ), breast cancer, ex vivo model, preclinical model, PDEC, stiffness, p38 SAPK.


Subject(s)
Breast Neoplasms/genetics , Estrogen Receptor alpha/genetics , Mechanotransduction, Cellular/genetics , Transcriptome , p38 Mitogen-Activated Protein Kinases/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Case-Control Studies , Cell Line, Tumor , Cinnamates/pharmacology , Collagen/chemistry , Collagen/pharmacology , Drug Combinations , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Female , Fulvestrant/pharmacology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Histones/genetics , Histones/metabolism , Humans , Indazoles/pharmacology , Laminin/chemistry , Laminin/pharmacology , Mammary Glands, Human/drug effects , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Phenotype , Proteoglycans/chemistry , Proteoglycans/pharmacology , Tamoxifen/pharmacology , Tissue Culture Techniques , p38 Mitogen-Activated Protein Kinases/metabolism
2.
Sci Rep ; 7(1): 3414, 2017 06 13.
Article in English | MEDLINE | ID: mdl-28611383

ABSTRACT

Conformational activation of integrins is generally required for ligand binding and cellular signalling. However, we have previously reported that the nonactivated conformation of α2ß1 integrin can also bind to large ligands, such as human echovirus 1. In this study, we show that the interaction between the nonactivated integrin and a ligand resulted in the activation of focal adhesion kinase (FAK) in a protein kinase C dependent manner. A loss-of-function mutation, α2E336A, in the α2-integrin did not prevent the activation of FAK, nor did EDTA-mediated inactivation of the integrin. Full FAK activation was observed, since phosphorylation was not only confirmed in residue Y397, but also in residues Y576/7. Furthermore, initiation of downstream signaling by paxillin phosphorylation in residue Y118 was evident, even though this activation was transient by nature, probably due to the lack of talin involvement in FAK activation and the absence of vinculin in the adhesion complexes formed by the nonactivated integrins. Altogether these results indicate that the nonactivated integrins can induce cellular signaling, but the outcome of the signaling differs from conventional integrin signaling.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/metabolism , Integrin alpha2beta1/metabolism , Signal Transduction , HeLa Cells , Humans , Integrin alpha2beta1/chemistry , Protein Conformation
3.
Int J Biochem Cell Biol ; 78: 22-30, 2016 09.
Article in English | MEDLINE | ID: mdl-27373681

ABSTRACT

Phorbol diester PMA (phorbol 12-myristate 13-acetate) is a well-known promoter of tumor progression. PMA also regulates cell adhesion by several mechanisms including conformational activation of integrins and integrin clustering. Here, PMA was shown to induce lamellipodia formation and reorganization of the adhesion sites as well as actin and vimentin filaments independently of integrin preactivation. To further analyze the mechanism of PMA action, the protein composition in the α1ß1 integrin/collagen IV adhesion sites was analyzed by mass spectrometry and proteomics. In four independent experiments we observed the reduced recruitment of vimentin in relation to integrin α1 subunit. This was in full agreement with the fact that we also detected the retraction of vimentin from cell adhesions by confocal microscopy. Furthermore, the accumulation of kindlin-2 into cell adhesions was significantly increased after PMA treatment. Kindlin-2 siRNA inhibited cell spreading as well as the formation of actin fibrils and cell adhesions, but did not prevent the effect of PMA on lamellipodia formation. Thus, kindlin-2 recruitment was considered to be a consequence rather than the primary cause for the loss of connection between vimentin and the adhesion sites.


Subject(s)
Cell Adhesion/drug effects , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Vimentin/metabolism , Animals , Carcinogenesis/drug effects , Cell Line , Cricetinae , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Integrin alpha1beta1/metabolism , Protein Transport/drug effects , Pseudopodia/drug effects , Pseudopodia/metabolism
4.
Exp Cell Res ; 342(2): 113-24, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26934497

ABSTRACT

The ability of cells to migrate and form metastases is one of the fatal hallmarks of cancer that can be conquered only with better understanding of the molecules and regulatory mechanisms involved. The oncogenic PIM kinases have been shown to support cancer cell survival and motility, but the PIM-regulated pathways stimulating cell migration and invasion are less well characterized than those affecting cell survival. Here we have identified the glycogen synthase kinase 3ß (GSK3B) and the forkhead box P3 (FOXP3) transcription factor as direct PIM targets, whose tumour-suppressive effects in prostate cancer cells are inhibited by PIM-induced phosphorylation, resulting in increased cell migration. Targeting GSK3B is also essential for the observed PIM-enhanced expression of the prostaglandin-endoperoxide synthase 2 (PTGS2), which is an important regulator of both cell migration and adhesion. Accordingly, selective inhibition of PIM activity not only reduces cell migration, but also affects integrin-mediated cell adhesion. Taken together, these data provide novel mechanistic insights on how and why patients with metastatic prostate cancer may benefit from therapies targeting PIM kinases, and how such approaches may also be applicable to inflammatory conditions.


Subject(s)
Adenocarcinoma/enzymology , Cell Movement , Prostatic Neoplasms/enzymology , Proto-Oncogene Proteins c-pim-1/physiology , Adenocarcinoma/pathology , Amino Acid Sequence , Cell Line, Tumor , Forkhead Transcription Factors/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Male , Phosphorylation , Prostatic Neoplasms/pathology , Protein Processing, Post-Translational , Protein Transport , Signal Transduction
5.
J Biol Chem ; 287(53): 44694-702, 2012 Dec 28.
Article in English | MEDLINE | ID: mdl-23132859

ABSTRACT

The interaction between α2ß1 integrin (GPIa/IIa, VLA-2) and vascular collagen is one of the initiating events in thrombus formation. Here, we describe two structurally similar sulfonamide derivatives, BTT-3033 and BTT-3034, and show that, under static conditions, they have an almost identical effect on α2-expressing CHO cell adhesion to collagen I, but only BTT-3033 blocks platelet attachment under flow (90 dynes/cm(2)). Differential scanning fluorimetry showed that both molecules bind to the α2I domain of the recombinant α2 subunit. To further study integrin binding mechanism(s) of the two sulfonamides, we created an α2 Y285F mutant containing a substitution near the metal ion-dependent adhesion site motif in the α2I domain. The action of BTT-3033, unlike that of BTT-3034, was dependent on Tyr-285. In static conditions BTT-3034, but not BTT-3033, inhibited collagen binding by an α2 variant carrying a conformationally activating E318W mutation. Conversely, in under flow conditions (90 dynes/cm(2)) BTT-3033, but not BTT-3034, inhibited collagen binding by an α2 variant expressing E336A loss-of-function mutation. Thus, the binding sites for BTT-3033 and BTT-3034 are differentially available in distinct integrin conformations. Therefore, these sulfonamides can be used to study the biological role of different functional stages of α2ß1. Furthermore, only the inhibitor that recognized the non-activated conformation of α2ß1 integrin under shear stress conditions effectively blocked platelet adhesion, suggesting that the initial interaction between integrin and collagen takes place prior to receptor activation.


Subject(s)
Collagen Type I/metabolism , Integrin alpha2beta1/antagonists & inhibitors , Integrin alpha2beta1/metabolism , Platelet Membrane Glycoproteins/metabolism , Sulfonamides/metabolism , Animals , Blood Platelets/chemistry , Blood Platelets/cytology , Blood Platelets/metabolism , Cell Adhesion/drug effects , Cell Line , Humans , Integrin alpha2beta1/genetics , Mice , Mice, Inbred C57BL , Molecular Structure , Platelet Membrane Glycoproteins/genetics , Protein Binding/drug effects , Stress, Mechanical , Sulfonamides/pharmacology
6.
EMBO J ; 29(1): 196-208, 2010 Jan 06.
Article in English | MEDLINE | ID: mdl-19927126

ABSTRACT

Conformational activation increases the affinity of integrins to their ligands. On ligand binding, further changes in integrin conformation elicit cellular signalling. Unlike any of the natural ligands of alpha2beta1 integrin, human echovirus 1 (EV1) seemed to bind more avidly a 'closed' than an activated 'open' form of the alpha2I domain. Furthermore, a mutation E336A in the alpha2 subunit, which inactivated alpha2beta1 as a collagen receptor, enhanced alpha2beta1 binding to EV1. Thus, EV1 seems to recognize an inactive integrin, and not even the virus binding could trigger the conformational activation of alpha2beta1. This was supported by the fact that the integrin clustering by EV1 did not activate the p38 MAP kinase pathway, a signalling pathway that was shown to be dependent on E336-related conformational changes in alpha2beta1. Furthermore, the mutation E336A did neither prevent EV1 induced and alpha2beta1 mediated protein kinase C activation nor EV1 internalization. Thus, in its entry strategy EV1 seems to rely on the activation of signalling pathways that are dependent on alpha2beta1 clustering, but do not require the conformational regulation of the receptor.


Subject(s)
Enterovirus B, Human/physiology , Enterovirus B, Human/pathogenicity , Integrin alpha2beta1/metabolism , Amino Acid Substitution , Animals , Binding Sites , CHO Cells , Cell Line , Chlorocebus aethiops , Cricetinae , Cricetulus , Humans , In Vitro Techniques , Integrin alpha2beta1/chemistry , Integrin alpha2beta1/genetics , Models, Molecular , Mutagenesis, Site-Directed , Protein Conformation , Protein Structure, Tertiary , Receptors, Virus/chemistry , Receptors, Virus/genetics , Receptors, Virus/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...