Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 21
Filtrer
1.
Blood ; 137(20): 2721-2735, 2021 05 20.
Article de Anglais | MEDLINE | ID: mdl-33824975

RÉSUMÉ

Selective targeting of BCL-2 with the BH3-mimetic venetoclax has been a transformative treatment for patients with various leukemias. TP-53 controls apoptosis upstream of where BCL-2 and its prosurvival relatives, such as MCL-1, act. Therefore, targeting these prosurvival proteins could trigger apoptosis across diverse blood cancers, irrespective of TP53 mutation status. Indeed, targeting BCL-2 has produced clinically relevant responses in blood cancers with aberrant TP-53. However, in our study, TP53-mutated or -deficient myeloid and lymphoid leukemias outcompeted isogenic controls with intact TP-53, unless sufficient concentrations of BH3-mimetics targeting BCL-2 or MCL-1 were applied. Strikingly, tumor cells with TP-53 dysfunction escaped and thrived over time if inhibition of BCL-2 or MCL-1 was sublethal, in part because of an increased threshold for BAX/BAK activation in these cells. Our study revealed the key role of TP-53 in shaping long-term responses to BH3-mimetic drugs and reconciled the disparate pattern of initial clinical response to venetoclax, followed by subsequent treatment failure among patients with TP53-mutant chronic lymphocytic leukemia or acute myeloid leukemia. In contrast to BH3-mimetics targeting just BCL-2 or MCL-1 at doses that are individually sublethal, a combined BH3-mimetic approach targeting both prosurvival proteins enhanced lethality and durably suppressed the leukemia burden, regardless of TP53 mutation status. Our findings highlight the importance of using sufficiently lethal treatment strategies to maximize outcomes of patients with TP53-mutant disease. In addition, our findings caution against use of sublethal BH3-mimetic drug regimens that may enhance the risk of disease progression driven by emergent TP53-mutant clones.


Sujet(s)
Antinéoplasiques/pharmacologie , Protéines régulatrices de l'apoptose/antagonistes et inhibiteurs , Apoptose/effets des médicaments et des substances chimiques , Composés hétérocycliques bicycliques/pharmacologie , Indolizine/pharmacologie , Isoquinoléines/pharmacologie , Leucémie chronique lymphocytaire à cellules B/traitement médicamenteux , Leucémie aigüe myéloïde/traitement médicamenteux , Morpholines/pharmacologie , Protéines tumorales/physiologie , Fragments peptidiques/antagonistes et inhibiteurs , Protéines proto-oncogènes/antagonistes et inhibiteurs , Sulfonamides/pharmacologie , Protéine p53 suppresseur de tumeur/physiologie , Animaux , Antinéoplasiques/administration et posologie , Antinéoplasiques/usage thérapeutique , Apoptose/physiologie , Protéines régulatrices de l'apoptose/physiologie , Composés hétérocycliques bicycliques/administration et posologie , Composés hétérocycliques bicycliques/usage thérapeutique , Systèmes CRISPR-Cas , Lignée cellulaire tumorale , Altération de l'ADN , Gènes p53 , Humains , Indolizine/usage thérapeutique , Sous-unité alpha du récepteur à l'interleukine-2/déficit , Isoquinoléines/usage thérapeutique , Leucémie aigüe myéloïde/anatomopathologie , Leucémie aigüe myéloïde/thérapie , Souris , Souris de lignée NOD , Souris knockout , Souris SCID , Morpholines/usage thérapeutique , Protéine Mcl-1/antagonistes et inhibiteurs , Protéines tumorales/antagonistes et inhibiteurs , Phosphorylation oxydative/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-bcl-2/antagonistes et inhibiteurs , Sulfonamides/administration et posologie , Sulfonamides/usage thérapeutique , Protéine p53 suppresseur de tumeur/déficit , Tests d'activité antitumorale sur modèle de xénogreffe
2.
Blood Adv ; 4(12): 2762-2767, 2020 06 23.
Article de Anglais | MEDLINE | ID: mdl-32569380

RÉSUMÉ

Improving survival outcomes in adult B-cell acute lymphoblastic leukemia (B-ALL) remains a clinical challenge. Relapsed disease has a poor prognosis despite the use of tyrosine kinase inhibitors (TKIs) for Philadelphia chromosome positive (Ph+ ALL) cases and immunotherapeutic approaches, including blinatumomab and chimeric antigen receptor T cells. Targeting aberrant cell survival pathways with selective small molecule BH3-mimetic inhibitors of BCL-2 (venetoclax, S55746), BCL-XL (A1331852), or MCL1 (S63845) is an emerging therapeutic option. We report that combined targeting of BCL-2 and MCL1 is synergistic in B-ALL in vitro. The combination demonstrated greater efficacy than standard chemotherapeutics and TKIs in primary samples from adult B-ALL with Ph+ ALL, Ph-like ALL, and other B-ALL. Moreover, combined BCL-2 or MCL1 inhibition with dasatinib showed potent killing in primary Ph+ B-ALL cases, but the BH3-mimetic combination appeared superior in vitro in a variety of Ph-like ALL samples. In PDX models, combined BCL-2 and MCL1 targeting eradicated ALL from Ph- and Ph+ B-ALL cases, although fatal tumor lysis was observed in some instances of high tumor burden. We conclude that a dual BH3-mimetic approach is highly effective in diverse models of high-risk human B-ALL and warrants assessment in clinical trials that incorporate tumor lysis precautions.


Sujet(s)
Leucémie-lymphome lymphoblastique à précurseurs B et T , Protéines proto-oncogènes c-bcl-2 , Adulte , Lymphocytes B , Lignée cellulaire tumorale , Humains , Protéine Mcl-1/génétique , Leucémie-lymphome lymphoblastique à précurseurs B et T/traitement médicamenteux , Protéines proto-oncogènes c-bcl-2/génétique
3.
Cells ; 9(1)2020 01 19.
Article de Anglais | MEDLINE | ID: mdl-31963783

RÉSUMÉ

Triple-negative breast cancer (TNBC) is an aggressive form of breast cancer with high risk of relapse and metastasis. TNBC is a heterogeneous disease comprising different molecular subtypes including those with mesenchymal features. The tyrosine kinase AXL is expressed in mesenchymal cells and plays a role in drug resistance, migration and metastasis. We confirm that AXL is more expressed in mesenchymal TNBC cells compared to luminal breast cancer cells, and that its invalidation impairs cell migration while having no or little effect on cell viability. Here, we found that AXL controls directed migration. We observed that AXL displays a polarized localization at the Golgi apparatus and the leading edge of migratory mesenchymal TNBC cells. AXL co-localizes with F-actin at the front of the cells. In migratory polarized cells, the specific AXL inhibitor R428 displaces AXL and F-actin from the leading edge to a lateral area localized between the front and the rear of the cells where both are enriched in protrusions. In addition, R428 treatment disrupts the polarized localization of the Golgi apparatus towards the leading edge in migratory cells. Immunohistochemical analysis of aggressive chemo-resistant TNBC samples obtained before treatment reveals inter- and intra-tumor heterogeneity of the percentage of AXL expressing tumor cells, and a preference of these cells to be in contact with the stroma. Taken together, our study demonstrates that AXL controls directed cell migration most likely by regulating cell polarity.


Sujet(s)
Mouvement cellulaire , Mésoderme/anatomopathologie , Protéines proto-oncogènes/métabolisme , Récepteurs à activité tyrosine kinase/métabolisme , Tumeurs du sein triple-négatives/enzymologie , Tumeurs du sein triple-négatives/anatomopathologie , Lignée cellulaire tumorale , Femelle , Appareil de Golgi/métabolisme , Humains , Polymérisation , Cellules stromales/anatomopathologie , Axl Receptor Tyrosine Kinase
5.
Nat Commun ; 10(1): 1812, 2019 04 18.
Article de Anglais | MEDLINE | ID: mdl-31000705

RÉSUMÉ

Non-small cell lung cancer (NSCLC) tumors harboring mutations in EGFR ultimately relapse to therapy with EGFR tyrosine kinase inhibitors (EGFR TKIs). Here, we show that resistant cells without the p.T790M or other acquired mutations are sensitive to the Aurora B (AURKB) inhibitors barasertib and S49076. Phospho-histone H3 (pH3), a major product of AURKB, is increased in most resistant cells and treatment with AURKB inhibitors reduces the levels of pH3, triggering G1/S arrest and polyploidy. Senescence is subsequently induced in cells with acquired mutations while, in their absence, polyploidy is followed by cell death. Finally, in NSCLC patients, pH3 levels are increased after progression on EGFR TKIs and high pH3 baseline correlates with shorter survival. Our results reveal that AURKB activation is associated with acquired resistance to EGFR TKIs, and that AURKB constitutes a potential target in NSCLC progressing to anti-EGFR therapy and not carrying resistance mutations.


Sujet(s)
Antinéoplasiques/pharmacologie , Aurora kinase B/métabolisme , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Tumeurs du poumon/traitement médicamenteux , Inhibiteurs de protéines kinases/pharmacologie , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Animaux , Antinéoplasiques/usage thérapeutique , Aurora kinase B/antagonistes et inhibiteurs , Aurora kinase B/génétique , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/mortalité , Carcinome pulmonaire non à petites cellules/anatomopathologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Évolution de la maladie , Résistance aux médicaments antinéoplasiques/génétique , Récepteurs ErbB/antagonistes et inhibiteurs , Récepteurs ErbB/génétique , Femelle , Points de contrôle de la phase G1 du cycle cellulaire/effets des médicaments et des substances chimiques , Histone/métabolisme , Humains , Tumeurs du poumon/génétique , Tumeurs du poumon/mortalité , Tumeurs du poumon/anatomopathologie , Mâle , Souris , Adulte d'âge moyen , Organophosphates/pharmacologie , Organophosphates/usage thérapeutique , Phosphorylation/effets des médicaments et des substances chimiques , Polyploïdie , Inhibiteurs de protéines kinases/usage thérapeutique , Quinazolines/pharmacologie , Quinazolines/usage thérapeutique , Petit ARN interférent/métabolisme , Analyse de survie , Résultat thérapeutique , Tests d'activité antitumorale sur modèle de xénogreffe
6.
Nat Commun ; 9(1): 1036, 2018 03 07.
Article de Anglais | MEDLINE | ID: mdl-29515130

RÉSUMÉ

In the original version of this Article, financial support and contributions in manuscript preparation were not fully acknowledged. The PDF and HTML versions of the Article have now been corrected to include the following:'M.P. and P.O. would like to thank Prof. Roderick Y.H. Lim for advice during manuscript preparation and for providing the laboratory and microscopy infrastructure.… [We also thank] the NanoteraProject, awarded to the PATLiSciII Consortium (M.P and P.O)…'.

7.
Nat Commun ; 8(1): 924, 2017 10 13.
Article de Anglais | MEDLINE | ID: mdl-29030636

RÉSUMÉ

At the stage of carcinoma in situ, the basement membrane (BM) segregates tumor cells from the stroma. This barrier must be breached to allow dissemination of the tumor cells to adjacent tissues. Cancer cells can perforate the BM using proteolysis; however, whether stromal cells play a role in this process remains unknown. Here we show that an abundant stromal cell population, cancer-associated fibroblasts (CAFs), promote cancer cell invasion through the BM. CAFs facilitate the breaching of the BM in a matrix metalloproteinase-independent manner. Instead, CAFs pull, stretch, and soften the BM leading to the formation of gaps through which cancer cells can migrate. By exerting contractile forces, CAFs alter the organization and the physical properties of the BM, making it permissive for cancer cell invasion. Blocking the ability of stromal cells to exert mechanical forces on the BM could therefore represent a new therapeutic strategy against aggressive tumors.Stromal cells play various roles in tumor establishment and metastasis. Here the authors, using an ex-vivo model, show that cancer-associated fibroblasts facilitate colon cancer cells invasion in a matrix metalloproteinase-independent manner, likely by pulling and stretching the basement membrane to form gaps.


Sujet(s)
Membrane basale , Fibroblastes associés au cancer/physiologie , Invasion tumorale , Cellules HCT116 , Cellules HT29 , Humains , Matrix metalloproteinases/métabolisme
8.
Curr Oncol Rep ; 19(3): 19, 2017 Mar.
Article de Anglais | MEDLINE | ID: mdl-28251492

RÉSUMÉ

A major challenge in anticancer treatment is the pre-existence or emergence of resistance to therapy. AXL and MER are two members of the TAM (TYRO3-AXL-MER) family of receptor tyrosine kinases, which, when activated, can regulate tumor cell survival, proliferation, migration and invasion, angiogenesis, and tumor-host interactions. An increasing body of evidence strongly suggests that these receptors play major roles in resistance to targeted therapies and conventional cytotoxic agents. Multiple resistance mechanisms exist, including the direct and indirect crosstalk of AXL and MER with other receptors and the activation of feedback loops regulating AXL and MER expression and activity. These mechanisms may be innate, adaptive, or acquired. A principal role of AXL appears to be in sustaining a mesenchymal phenotype, itself a major mechanism of resistance to diverse anticancer therapies. Both AXL and MER play a role in the repression of the innate immune response which may also limit response to treatment. Small molecule and antibody inhibitors of AXL and MER have recently been described, and some of these have already entered clinical trials. The optimal design of treatment strategies to maximize the clinical benefit of these AXL and MER targeting agents are discussed in relation to the different cancer types and the types of resistance encountered. One of the major challenges to successful development of these therapies will be the application of robust predictive biomarkers for clear-cut patient stratification.


Sujet(s)
Résistance aux médicaments antinéoplasiques/génétique , Tumeurs/traitement médicamenteux , Protéines proto-oncogènes/génétique , Récepteurs à activité tyrosine kinase/génétique , Apoptose/effets des médicaments et des substances chimiques , Survie cellulaire , Humains , Tumeurs/génétique , Protéines proto-oncogènes/antagonistes et inhibiteurs , Récepteurs à activité tyrosine kinase/antagonistes et inhibiteurs , Transduction du signal/effets des médicaments et des substances chimiques , Bibliothèques de petites molécules/usage thérapeutique , c-Mer Tyrosine kinase , Axl Receptor Tyrosine Kinase
9.
Oncotarget ; 7(23): 33542-56, 2016 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-27507190

RÉSUMÉ

Uveal melanoma (UM) is the most common cancer of the eye in adults. Many UM patients develop metastases for which no curative treatment has been identified. Novel therapeutic approaches are therefore urgently needed. UM is characterized by mutations in the genes GNAQ and GNA11 which activate the PKC pathway, leading to the use of PKC inhibitors as a rational strategy to treat UM tumors. Encouraging clinical activity has been noted in UM patients treated with PKC inhibitors. However, it is likely that curative treatment regimens will require a combination of targeted therapeutic agents. Employing a large panel of UM patient-derived xenograft models (PDXs), several PKC inhibitor-based combinations were tested in vivo using the PKC inhibitor AEB071. The most promising approaches were further investigated in vitro using our unique panel of UM cell lines. When combined with AEB071, the two agents CGM097 (p53-MDM2 inhibitor) and RAD001 (mTORC1 inhibitor) demonstrated greater activity than single agents, with tumor regression observed in several UM PDXs. Follow-up studies in UM cell lines on these two drug associations confirmed their combination activity and ability to induce cell death. While no effective treatment currently exists for metastatic uveal melanoma, we have discovered using our unique panel of preclinical models that combinations between PKC/mTOR inhibitors and PKC/p53-MDM2 inhibitors are two novel and very effective therapeutic approaches for this disease. Together, our study reveals that combining PKC and p53-MDM2 or mTORC1 inhibitors may provide significant clinical benefit for UM patients.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Mélanome/traitement médicamenteux , Tumeurs de l'uvée/traitement médicamenteux , Animaux , Lignée cellulaire tumorale , Antienzymes/pharmacologie , Évérolimus/pharmacologie , Humains , Isoquinoléines/pharmacologie , Complexe-1 cible mécanistique de la rapamycine/antagonistes et inhibiteurs , Souris , Pipérazines/pharmacologie , Protéine kinase C/antagonistes et inhibiteurs , Protéines proto-oncogènes c-mdm2/antagonistes et inhibiteurs , Pyrroles/pharmacologie , Quinazolines/pharmacologie , Protéine p53 suppresseur de tumeur/antagonistes et inhibiteurs , Tests d'activité antitumorale sur modèle de xénogreffe
11.
Elife ; 52016 05 16.
Article de Anglais | MEDLINE | ID: mdl-27183006

RÉSUMÉ

The TMPRSS2:ERG gene fusion is common in androgen receptor (AR) positive prostate cancers, yet its function remains poorly understood. From a screen for functionally relevant ERG interactors, we identify the arginine methyltransferase PRMT5. ERG recruits PRMT5 to AR-target genes, where PRMT5 methylates AR on arginine 761. This attenuates AR recruitment and transcription of genes expressed in differentiated prostate epithelium. The AR-inhibitory function of PRMT5 is restricted to TMPRSS2:ERG-positive prostate cancer cells. Mutation of this methylation site on AR results in a transcriptionally hyperactive AR, suggesting that the proliferative effects of ERG and PRMT5 are mediated through attenuating AR's ability to induce genes normally involved in lineage differentiation. This provides a rationale for targeting PRMT5 in TMPRSS2:ERG positive prostate cancers. Moreover, methylation of AR at arginine 761 highlights a mechanism for how the ERG oncogene may coax AR towards inducing proliferation versus differentiation.


Sujet(s)
Cellules épithéliales/métabolisme , Régulation de l'expression des gènes tumoraux , Protéines de fusion oncogènes/génétique , Protein-arginine N-methyltransferases/génétique , Récepteurs aux androgènes/génétique , Serine endopeptidases/génétique , Séquence nucléotidique , Différenciation cellulaire , Lignée cellulaire tumorale , Prolifération cellulaire , Cellules épithéliales/anatomopathologie , Humains , Mâle , Méthylation , Modèles moléculaires , Mutation , Protéines de fusion oncogènes/métabolisme , Prostate/métabolisme , Prostate/anatomopathologie , Liaison aux protéines , Motifs et domaines d'intéraction protéique , Multimérisation de protéines , Structure secondaire des protéines , Protein-arginine N-methyltransferases/antagonistes et inhibiteurs , Protein-arginine N-methyltransferases/métabolisme , Petit ARN interférent/génétique , Petit ARN interférent/métabolisme , Récepteurs aux androgènes/composition chimique , Récepteurs aux androgènes/métabolisme , Serine endopeptidases/métabolisme , Transduction du signal , Régulateur transcriptionnel ERG/génétique , Régulateur transcriptionnel ERG/métabolisme
12.
Oncotarget ; 7(17): 23633-46, 2016 Apr 26.
Article de Anglais | MEDLINE | ID: mdl-26988753

RÉSUMÉ

Uveal melanoma (UM) is the most frequent malignant ocular tumor in adults. While the primary tumor is efficiently treated by surgery and/or radiotherapy, about one third of UM patients develop metastases, for which no effective treatment is currently available. The PKC, MAPK and PI3K/AKT/mTOR signaling cascades have been shown to be associated with tumor growth. However, none of the compounds against those pathways results in tumor regression when used as single agents. To identify more effective therapeutic strategies for UM patients, we performed a combination screen using seven targeted agents inhibiting PKC, MEK, AKT, PI3K and mTOR in a panel of ten UM cell lines, representative of the UM disease. We identified a strong synergy between the mTOR inhibitor Everolimus and the PI3K inhibitor GDC0941. This combination resulted in an increase in apoptosis in several UM cell lines compared to monotherapies and enhanced the anti-tumor effect of each single agent in two patient-derived xenografts. Furthermore, we showed that the synergism between the two drugs was associated with the relief by GDC0491 of a reactivation of AKT induced by Everolimus. Altogether, our results highlight a novel and effective combination strategy, which could be beneficial for UM patients.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Synergie des médicaments , Inhibiteurs des phosphoinositide-3 kinases , Sérine-thréonine kinases TOR/antagonistes et inhibiteurs , Animaux , Apoptose/effets des médicaments et des substances chimiques , Marqueurs biologiques tumoraux , Cycle cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Évérolimus/administration et posologie , Humains , Indazoles/administration et posologie , Mélanome , Souris , Souris SCID , Transduction du signal , Sulfonamides/administration et posologie , Cellules cancéreuses en culture , Tumeurs de l'uvée , Tests d'activité antitumorale sur modèle de xénogreffe
14.
Br J Cancer ; 113(9): 1249-53, 2015 Nov 03.
Article de Anglais | MEDLINE | ID: mdl-26505679

RÉSUMÉ

The past few years have witnessed major advances in the understanding of the molecular landscape of uveal melanoma (UM). The discovery of a mutational background that is completely different from the one of skin melanoma has granted to UM a stand-alone status. The absence of effective therapy for metastatic disease offers now a chessboard for targeted therapy but at the same time urges preclinical science to develop accordingly, to guide the use of economical resources to the best profit of patients. This review describes the current knowledge on the biology of this disease and discusses the challenges that must be undertaken to translate this knowledge into real benefit for patients.


Sujet(s)
Mélanome/génétique , Tumeurs de l'uvée/génétique , Animaux , Humains , Mutation/génétique , Tumeurs cutanées ,
15.
Eur J Cell Biol ; 93(10-12): 388-95, 2014 Oct.
Article de Anglais | MEDLINE | ID: mdl-25269996

RÉSUMÉ

While absent from normal epithelia, an actin bundling protein, fascin, becomes expressed in invasive carcinoma of different origins. It is highly enriched at the tumors' invasive front suggesting that it could play a role in cancer invasion. Multiple studies have shown that fascin, through its role in formation of cellular protrusions such as filopodia and invadopodia, enhances cancer cell migration and invasion in vitro. However, the role of fascin in vivo remains unknown. We have generated a compound transgenic mouse model that allows expression of fascin in the intestinal epithelium in the Apc-mutated background. Conditional expression of fascin led to decrease in mice survival and increase in tumor burden compared to control animals. Induction of fascin expression in adult tumor-bearing animals accelerated tumor progression and led to formation of invasive adenocarcinoma. Altogether, our study shows that fascin can promote tumor progression in vivo, but also unravels an unexpected role of fascin in tumor initiation.


Sujet(s)
Adénocarcinome/métabolisme , Protéines de transport/génétique , Tumeurs colorectales/métabolisme , Protéines des microfilaments/génétique , Adénocarcinome/anatomopathologie , Animaux , Protéines de transport/métabolisme , Prolifération cellulaire/génétique , Tumeurs colorectales/anatomopathologie , Évolution de la maladie , Muqueuse intestinale/métabolisme , Muqueuse intestinale/anatomopathologie , Souris , Souris transgéniques , Protéines des microfilaments/métabolisme , Invasion tumorale , Métastase tumorale
16.
Cancer Res ; 74(12): 3294-305, 2014 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-24747911

RÉSUMÉ

Tankyrases (TNKS) play roles in Wnt signaling, telomere homeostasis, and mitosis, offering attractive targets for anticancer treatment. Using unbiased combination screening in a large panel of cancer cell lines, we have identified a strong synergy between TNKS and MEK inhibitors (MEKi) in KRAS-mutant cancer cells. Our study uncovers a novel function of TNKS in the relief of a feedback loop induced by MEK inhibition on FGFR2 signaling pathway. Moreover, dual inhibition of TNKS and MEK leads to more robust apoptosis and antitumor activity both in vitro and in vivo than effects observed by previously reported MEKi combinations. Altogether, our results show how a novel combination of TNKS and MEK inhibitors can be highly effective in targeting KRAS-mutant cancers by suppressing a newly discovered resistance mechanism.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Protéines proto-oncogènes/génétique , Récepteur FGFR2/métabolisme , Tankyrases/métabolisme , Protéines G ras/génétique , Acétamides/administration et posologie , Aminopyridines/administration et posologie , Dérivés de l'aniline/administration et posologie , Animaux , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Lignée cellulaire tumorale , Synergie des médicaments , Chlorhydrate d'erlotinib , Rétrocontrôle physiologique , Femelle , Humains , MAP Kinase Kinase Kinases/antagonistes et inhibiteurs , MAP Kinase Kinase Kinases/métabolisme , Souris , Souris nude , Morpholines/administration et posologie , Mutation , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Protéines proto-oncogènes p21(ras) , Pyrimidinones/administration et posologie , Quinazolines/administration et posologie , Récepteur FGFR2/antagonistes et inhibiteurs , Transduction du signal , Sulfonamides/administration et posologie , Tankyrases/antagonistes et inhibiteurs , Thiazoles/administration et posologie , Tests d'activité antitumorale sur modèle de xénogreffe
17.
Methods Mol Biol ; 1046: 133-44, 2013.
Article de Anglais | MEDLINE | ID: mdl-23868586

RÉSUMÉ

To escape the primary tumor and infiltrate stromal compartments, invasive cancer cells must traverse the basement membrane (BM). To break this dense matrix, cells develop finger-like protrusions, called invadopodia, at their ventral surface. Invadopodia secrete proteases to degrade the BM, and then elongate which allows the cell to invade the subjacent tissue. Here, we describe two complementary invasion assays. The native BM invasion assay, based on BM isolated from rat or mouse mesentery, is a physiologically significant approach for studying the stages of BM crossing at the cellular level. The Matrigel-based chemoinvasion assay is a powerful technique for studying invadopodia's molecular composition and organization at the subcellular level.


Sujet(s)
Membrane basale/cytologie , Dosage biologique/méthodes , Biologie moléculaire/méthodes , Tumeurs/génétique , Animaux , Membrane basale/métabolisme , Mouvement cellulaire , Collagène/composition chimique , Association médicamenteuse , Matrice extracellulaire/métabolisme , Matrice extracellulaire/anatomopathologie , Humains , Laminine/composition chimique , Souris , Invasion tumorale/génétique , Invasion tumorale/anatomopathologie , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Protéoglycanes/composition chimique , Rats
18.
Eur J Cell Biol ; 90(2-3): 93-9, 2011.
Article de Anglais | MEDLINE | ID: mdl-20609495

RÉSUMÉ

The basement membrane (BM) is a dense, tightly cross-linked matrix that acts as physiological barrier to maintain tissue homeostasis. Studies on Caenorhabditis elegans, leucocytes diapedesis and cancer cell invasion have shown that BM transmigration is a conserved three-stage process. Firstly, invadopodia-like protrusions form at the ventral surface of invasive cells; later, one protrusion elongates that lastly drives the infiltration of cells into the underlying compartment. This review illustrates the mechanism used by invasive cancer cells to cross the BM barrier by focusing on the role of key cytoskeleton components. We also describe currently available in vitro assays to study each step of the BM transmigration program.


Sujet(s)
Membrane basale/physiologie , Mouvement cellulaire/physiologie , Cytosquelette/physiologie , Animaux , Membrane basale/métabolisme , Cytosquelette/métabolisme , Humains , Microtubules/métabolisme
19.
J Cell Biol ; 189(3): 541-56, 2010 May 03.
Article de Anglais | MEDLINE | ID: mdl-20421424

RÉSUMÉ

Invasive cancer cells are believed to breach the basement membrane (BM) using specialized protrusions called invadopodia. We found that the crossing of a native BM is a three-stage process: invadopodia indeed form and perforate the BM, elongate into mature invadopodia, and then guide the cell toward the stromal compartment. We studied the remodeling of cytoskeleton networks during invadopodia formation and elongation using ultrastructural analysis, spatial distribution of molecular markers, and RNA interference silencing of protein expression. We show that formation of invadopodia requires only the actin cytoskeleton and filopodia- and lamellipodia-associated proteins. In contrast, elongation of invadopodia is mostly dependent on filopodial actin machinery. Moreover, intact microtubules and vimentin intermediate filament networks are required for further growth. We propose that invadopodia form by assembly of dendritic/diagonal and bundled actin networks and then mature by elongation of actin bundles, followed by the entry of microtubules and vimentin filaments. These findings provide a link between the epithelial to mesenchymal transition and BM transmigration.


Sujet(s)
Actines/métabolisme , Prolongements cytoplasmiques/ultrastructure , Microtubules/métabolisme , Vimentine/métabolisme , Animaux , Membrane basale/métabolisme , Mouvement cellulaire , Cellules cultivées , Humains , Interférence par ARN , Transfection
20.
Adv Cancer Res ; 100: 85-111, 2008.
Article de Anglais | MEDLINE | ID: mdl-18620093

RÉSUMÉ

In this review, the major signal transduction pathways that have been shown to play an important role in intestinal homeostasis are highlighted. Each of them, the Wnt, Notch, Hedgehog, and Bone Morphogenetic Protein, as well as growth-factor regulated Receptor Tyrosine Kinases are depicted with a special emphasis through their involvement in stem cell maintenance and their role in intestinal tumorigenesis. Finally, we discuss recent data on the final steps of tumor progression, notably the formation of distant metastases. This multistep process is highly complex and still far from being understood while being of major importance for the survival of patients with digestive cancer.


Sujet(s)
Muqueuse intestinale/embryologie , Tumeurs de l'intestin/génétique , Morphogenèse/génétique , Transduction du signal/génétique , Animaux , Protéines morphogénétiques osseuses/génétique , Protéines morphogénétiques osseuses/physiologie , Régulation de l'expression des gènes au cours du développement , Régulation de l'expression des gènes tumoraux , Protéines Hedgehog/génétique , Protéines Hedgehog/physiologie , Humains , Muqueuse intestinale/physiologie , Tumeurs de l'intestin/anatomopathologie , Modèles biologiques , Morphogenèse/physiologie , Métastase tumorale , Phosphohydrolase PTEN/génétique , Phosphohydrolase PTEN/physiologie , Récepteurs à activité tyrosine kinase/génétique , Récepteurs à activité tyrosine kinase/physiologie , Récepteurs Notch/génétique , Récepteurs Notch/physiologie , Protéines de type Wingless/génétique , Protéines de type Wingless/physiologie , bêta-Caténine/génétique , bêta-Caténine/physiologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE