Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 6 de 6
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Sci Rep ; 14(1): 13227, 2024 06 09.
Article de Anglais | MEDLINE | ID: mdl-38851782

RÉSUMÉ

There are hundreds of genes typically overexpressed in breast cancer cells and it's often assumed that their overexpression contributes to cancer progression. However, the precise proportion of these overexpressed genes contributing to tumorigenicity remains unclear. To address this gap, we undertook a comprehensive screening of a diverse set of seventy-two genes overexpressed in breast cancer. This systematic screening evaluated their potential for inducing malignant transformation and, concurrently, assessed their impact on breast cancer cell proliferation and viability. Select genes including ALDH3B1, CEACAM5, IL8, PYGO2, and WWTR1, exhibited pronounced activity in promoting tumor formation and establishing gene dependencies critical for tumorigenicity. Subsequent investigations revealed that CEACAM5 overexpression triggered the activation of signaling pathways involving ß-catenin, Cdk4, and mTOR. Additionally, it conferred a growth advantage independent of exogenous insulin in defined medium and facilitated spheroid expansion by inducing multiple layers of epithelial cells while preserving a hollow lumen. Furthermore, the silencing of CEACAM5 expression synergized with tamoxifen-induced growth inhibition in breast cancer cells. These findings underscore the potential of screening overexpressed genes for both oncogenic drivers and tumor dependencies to expand the repertoire of therapeutic targets for breast cancer treatment.


Sujet(s)
Tumeurs du sein , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , Humains , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Femelle , Prolifération cellulaire/génétique , Lignée cellulaire tumorale , Transduction du signal , Oncogènes , bêta-Caténine/métabolisme , bêta-Caténine/génétique , Tamoxifène/pharmacologie , Animaux , Molécules d'adhérence cellulaire/génétique , Molécules d'adhérence cellulaire/métabolisme , Sérine-thréonine kinases TOR/métabolisme , Sérine-thréonine kinases TOR/génétique , Kinase-4 cycline-dépendante/génétique , Kinase-4 cycline-dépendante/métabolisme , Transformation cellulaire néoplasique/génétique
2.
Semin Cancer Biol ; 54: 101-108, 2019 02.
Article de Anglais | MEDLINE | ID: mdl-29170065

RÉSUMÉ

Pancreatic cancer is considered among the most aggressive and the least curable of all human malignancies. It is usually characterized by multiple aberrations in tumor suppressor genes and oncogenes, most notably activating mutations in KRAS. This review examines the various attempts that have been made to inhibit Kras and its downstream signaling pathways in pancreatic cancer with an emphasis on challenges related to clinical trials. Attempts include preventing the localization of Ras protein to the plasma membrane, inhibiting downstream oncogenic signaling by targeting Kras effectors such as MEK1/2, Erk1/2 or Akt singly or in combination, and directly inhibiting Kras protein. Most clinical trials have focused on inhibiting downstream effector pathways and clinical benefit has been limited due to compensatory mechanisms and toxicity associated with small therapeutic windows. Additionally, genetic screens have been conducted to identify gene or genes that could provide therapeutic vulnerabilities in mutant KRAS cells and provide a way to target mutant Kras protein only. We also discuss how potentially transforming clinical trials have failed in the past and what new strategies are on-going in clinical trials for pancreas cancer. For long-term success in targeting Kras, future efforts should focus on combinatorial strategies to more effectively block Kras pathways at multiple points, and improve translational application of pre-clinical data to the clinic.


Sujet(s)
Tumeurs du pancréas/génétique , Tumeurs du pancréas/métabolisme , Protéines G ras/génétique , Protéines G ras/métabolisme , Animaux , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Essais cliniques comme sujet , Découverte de médicament , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Mitogen-Activated Protein Kinases/métabolisme , Thérapie moléculaire ciblée , Mutation , Protéines oncogènes/génétique , Protéines oncogènes/métabolisme , Oncogènes , Tumeurs du pancréas/traitement médicamenteux , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Transport des protéines , Protéines proto-oncogènes c-akt/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Protéines G ras/antagonistes et inhibiteurs
3.
Mol Biol Cell ; 21(2): 311-22, 2010 Jan 15.
Article de Anglais | MEDLINE | ID: mdl-19940019

RÉSUMÉ

In the canonical Wnt pathway, beta-catenin acts as a key coactivator that stimulates target gene expression through interaction with Tcf/Lef transcription factors. Its nuclear accumulation is the hallmark of active Wnt signaling and is frequently associated with cancers. Chibby (Cby) is an evolutionarily conserved molecule that represses beta-catenin-dependent gene activation. Although Cby, in conjunction with 14-3-3 chaperones, controls beta-catenin distribution, its molecular nature remains largely unclear. Here, we provide compelling evidence that Cby harbors bona fide nuclear localization signal (NLS) and nuclear export signal (NES) motifs, and constitutively shuttles between the nucleus and cytoplasm. Efficient nuclear export of Cby requires a cooperative action of the intrinsic NES, 14-3-3, and the CRM1 nuclear export receptor. Notably, 14-3-3 docking provokes Cby binding to CRM1 while inhibiting its interaction with the nuclear import receptor importin-alpha, thereby promoting cytoplasmic compartmentalization of Cby at steady state. Importantly, the NLS- and NES-dependent shuttling of Cby modulates the dynamic intracellular localization of beta-catenin. In support of our model, short hairpin RNA-mediated knockdown of endogenous Cby results in nuclear accumulation of beta-catenin. Taken together, these findings unravel the molecular basis through which a combinatorial action of Cby and 14-3-3 proteins controls the dynamic nuclear-cytoplasmic trafficking of beta-catenin.


Sujet(s)
Protéines de transport/métabolisme , Noyau de la cellule/métabolisme , Protéines nucléaires/métabolisme , Transduction du signal , bêta-Caténine/métabolisme , Protéines 14-3-3/métabolisme , Transport nucléaire actif , Séquence d'acides aminés , Animaux , Protéines de transport/composition chimique , Lignée cellulaire , Techniques de knock-down de gènes , Humains , Souris , Données de séquences moléculaires , Signaux d'export nucléaire , Signaux de localisation nucléaire/métabolisme , Protéines nucléaires/composition chimique , Liaison aux protéines , Fractions subcellulaires/métabolisme , Cariophérines alpha/métabolisme
4.
BMC Mol Biol ; 10: 41, 2009 May 12.
Article de Anglais | MEDLINE | ID: mdl-19435523

RÉSUMÉ

BACKGROUND: The Wnt/beta-catenin signaling pathway plays crucial roles in embryonic development and in maintenance of organs and tissues in adults. Chibby (Cby) is an evolutionarily conserved molecule that physically interacts with the key downstream coactivator beta-catenin and represses its transcriptional activation potential. Although Cby harbors a predicted coiled-coil motif in the C-terminal region, its molecular nature and functional importance remain largely unexplored. RESULTS: Here we report that Cby forms a stable complex with itself. Alanine substitutions of two or more of four critical leucine residues within the C-terminal heptad repeats completely eliminate the Cby-Cby interaction. The Cby oligomer predominantly exists as a homodimer. Furthermore, we found that dimerization-deficient Cby mutants still retain the ability to bind to beta-catenin and to repress beta-catenin-dependent gene activation. More importantly, Cby homodimerization is required for its efficient interaction with the nuclear import receptor importin-alpha and subsequent nuclear translocation. CONCLUSION: Our comprehensive mutational analysis of the Cby coiled-coil domain reveals that the four heptad leucine residues play an essential role in mediating Cby homodimerization. Although monomeric Cby is sufficient to bind to beta-catenin and block beta-catenin-mediated transcriptional activation, homodimer formation of Cby is indispensable for its efficient nuclear import.


Sujet(s)
Protéines de transport/composition chimique , Protéines de transport/métabolisme , Leucine/composition chimique , Protéines nucléaires/composition chimique , Protéines nucléaires/métabolisme , Transport nucléaire actif , Motifs d'acides aminés , Séquence d'acides aminés , Protéines de transport/génétique , Lignée cellulaire , Dimérisation , Humains , Leucine/génétique , Leucine/métabolisme , Données de séquences moléculaires , Protéines nucléaires/génétique , Liaison aux protéines , Transport des protéines , Alignement de séquences , Activation de la transcription , Cariophérines alpha/génétique , Cariophérines alpha/métabolisme , bêta-Caténine/génétique , bêta-Caténine/métabolisme
5.
J Cell Biol ; 181(7): 1141-54, 2008 Jun 30.
Article de Anglais | MEDLINE | ID: mdl-18573912

RÉSUMÉ

beta-Catenin functions in both cell-cell adhesion and as a transcriptional coactivator in the canonical Wnt pathway. Nuclear accumulation of beta-catenin is the hallmark of active Wnt signaling and is frequently observed in human cancers. Although beta-catenin shuttles in and out of the nucleus, the molecular mechanisms underlying its translocation remain poorly understood. Chibby (Cby) is an evolutionarily conserved molecule that inhibits beta-catenin-mediated transcriptional activation. Here, we identified 14-3-3epsilon and 14-3-3zeta as Cby-binding partners using affinity purification/mass spectrometry. 14-3-3 proteins specifically recognize serine 20 within the 14-3-3-binding motif of Cby when phosphorylated by Akt kinase. Notably, 14-3-3 binding results in sequestration of Cby into the cytoplasm. Moreover, Cby and 14-3-3 form a stable tripartite complex with beta-catenin, causing beta-catenin to partition into the cytoplasm. Our results therefore suggest a novel paradigm through which Cby acts in concert with 14-3-3 proteins to facilitate nuclear export of beta-catenin, thereby antagonizing beta-catenin signaling.


Sujet(s)
Protéines 14-3-3/métabolisme , Protéines de transport/métabolisme , Protéines nucléaires/métabolisme , Transduction du signal , bêta-Caténine/métabolisme , Motifs d'acides aminés , Séquence d'acides aminés , Animaux , Cellules COS , Protéines de transport/composition chimique , Noyau de la cellule/enzymologie , Chlorocebus aethiops , Humains , Modèles biologiques , Données de séquences moléculaires , Protéines nucléaires/composition chimique , Phosphorylation , Liaison aux protéines , Isoformes de protéines/métabolisme , Transport des protéines , Protéines proto-oncogènes c-akt/métabolisme , Fractions subcellulaires/métabolisme , Spécificité du substrat , Activation de la transcription
6.
Mol Cell Biol ; 27(12): 4347-54, 2007 Jun.
Article de Anglais | MEDLINE | ID: mdl-17403895

RÉSUMÉ

The canonical Wnt/beta-catenin signaling pathway plays diverse roles in embryonic development and disease. Activation of this pathway, likely by Wnt-10b, has been shown to inhibit adipogenesis in cultured 3T3-L1 preadipocytes and in mice. Here, we report that the beta-catenin antagonist Chibby (Cby) is required for adipocyte differentiation. Cby is expressed in adipose tissue in mice, and Cby protein levels increase during adipogenic differentiation of 3T3-L1 cells. Ectopic expression of Cby induces spontaneous differentiation of these cells into mature adipocytes to an extent similar to that of dominant-negative Tcf-4. In contrast, depletion of Cby by RNA interference potently blocks adipogenesis of 3T3-L1 and mouse embryonic stem cells. In support of this, embryonic fibroblasts obtained from Cby-deficient embryos display attenuated differentiation to the adipogenic lineage. Mechanistically, Cby promotes adipocyte differentiation, in part by inhibiting beta-catenin, since gain or loss of function of Cby influences beta-catenin signaling in 3T3-L1 cells. Our results therefore establish Cby as a novel proadipogenic factor required for adipocyte differentiation.


Sujet(s)
Adipocytes/physiologie , Protéines de transport/physiologie , Différenciation cellulaire , Protéines nucléaires/physiologie , bêta-Caténine/antagonistes et inhibiteurs , Cellules 3T3-L1 , Adipogenèse , Animaux , Protéines de transport/génétique , Lignée cellulaire , Cellules cultivées , Cellules souches embryonnaires/cytologie , Cellules souches embryonnaires/physiologie , Gènes rapporteurs , Histocytochimie , Humains , Luciferases/métabolisme , Souris , Protéines nucléaires/génétique , Interférence par ARN , Transduction du signal
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...