Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Mol Sci ; 22(11)2021 Jun 03.
Article in English | MEDLINE | ID: mdl-34204950

ABSTRACT

The dysregulation of autophagy is important in the development of many cancers, including thyroid cancer, where V600EBRAF is a main oncogene. Here, we analyse the effect of V600EBRAF inhibition on autophagy, the mechanisms involved in this regulation and the role of autophagy in cell survival of thyroid cancer cells. We reveal that the inhibition of V600EBRAF activity with its specific inhibitor PLX4720 or the depletion of its expression by siRNA induces autophagy in thyroid tumour cells. We show that V600EBRAF downregulation increases LKB1-AMPK signalling and decreases mTOR activity through a MEK/ERK-dependent mechanism. Moreover, we demonstrate that PLX4720 activates ULK1 and increases autophagy through the activation of the AMPK-ULK1 pathway, but not by the inhibition of mTOR. In addition, we find that autophagy blockade decreases cell viability and sensitize thyroid cancer cells to V600EBRAF inhibition by PLX4720 treatment. Finally, we generate a thyroid xenograft model to demonstrate that autophagy inhibition synergistically enhances the anti-proliferative and pro-apoptotic effects of V600EBRAF inhibition in vivo. Collectively, we uncover a new role of AMPK in mediating the induction of cytoprotective autophagy by V600EBRAF inhibition. In addition, these data establish a rationale for designing an integrated therapy targeting V600EBRAF and the LKB1-AMPK-ULK1-autophagy axis for the treatment of V600EBRAF-positive thyroid tumours.


Subject(s)
Autophagy-Related Protein-1 Homolog/genetics , Intracellular Signaling Peptides and Proteins/genetics , Protein Kinases/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins B-raf/genetics , Thyroid Neoplasms/genetics , AMP-Activated Protein Kinase Kinases , Apoptosis/drug effects , Autophagy/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Indoles/pharmacology , Mutation/genetics , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Sulfonamides/pharmacology , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/pathology
2.
Mol Carcinog ; 55(11): 1639-1654, 2016 Nov.
Article in English | MEDLINE | ID: mdl-26392228

ABSTRACT

The epithelial-mesenchymal transition (EMT) is a crucial process in tumour progression, by which epithelial cells acquire a mesenchymal phenotype, increasing its motility and the ability to invade distant sites. Here, we describe the molecular mechanisms by which V600E BRAF, TGFß and the Src/FAK complex cooperatively regulate EMT induction and cell motility of anaplastic thyroid cancer cells. Analysis of EMT marker levels reveals a positive correlation between TGFß and Snail expression, with a concomitant downregulation of E-cadherin, accompanied by an increase of cell migration and invasion. Furthermore, we show that V600E BRAF depletion by siRNA or inhibition of its activity by treatment with its inhibitor PLX4720 reverses the TGFß-mediated effects on Snail, E-cadherin, migration and invasion. Moreover, V600E BRAF induces TGFß secretion through a MEK/ERK-dependent mechanism. In addition, TGFß activates the Src/FAK complex, which in turn regulates the expression of Snail and E-cadherin as well as cell migration. The inhibition of Src with the inhibitor SU6656 or abrogation of FAK expression with a specific siRNA reverses the TGFß-induced effects. Interestingly, we demonstrate that activation of the Src/FAK complex by TGFß is independent of V600E BRAF signalling, since inhibition of this oncogene does not affect its phosphorylation. Our data strongly suggest that TGFß induces EMT and aggressiveness of thyroid cancer cells by parallel mechanisms involving both the V600E BRAF/MEK/ERK and Src/FAK pathways independently. Thus, we describe novel functions for Src/FAK in mediating the EMT program and aggressiveness regulated by TGFß, establishing the inhibition of these proteins as a possible effective approach in preventing tumour progression of V600E BRAF-expressing thyroid tumours. © 2015 Wiley Periodicals, Inc.


Subject(s)
Focal Adhesion Kinase 1/metabolism , Proto-Oncogene Proteins B-raf/genetics , Thyroid Neoplasms/metabolism , Transforming Growth Factor beta/metabolism , src-Family Kinases/metabolism , Cell Line, Tumor , Cell Movement , Epithelial-Mesenchymal Transition , Humans , Indoles/pharmacology , MAP Kinase Signaling System , Mutation , Neoplasm Invasiveness , Proto-Oncogene Proteins B-raf/metabolism , Sulfonamides/pharmacology
3.
Mol Cell Biochem ; 411(1-2): 253-60, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26472731

ABSTRACT

Somatostatin (SST) is one of the main regulators of thyroid function. It acts by binding to its receptors, which lead to the dissociation of G proteins into Gαi and Gßγ subunits. However, much less is known about the function of Gßγ in thyroid cells. Here, we studied the role of SST and Gßγ dimers released upon SST stimulation on the Ras-ERK1/2 pathway in FTRL-5 thyroid cells. We demonstrate that SST activates Ras through Gi proteins, since SST-induced Ras activation is inhibited by pertussis toxin. Moreover, the specific sequestration of Gßγ dimers decreases Ras-GTP and phosphorylated ERK1/2 levels, and overexpression of Gßγ increases ERK1/2 phosphorylation induced by SST, indicating that Gßγ dimers released after SST treatment mediate activation of Ras and ERK1/2. On the other hand, SST treatment does not modify the expression of the thyroid differentiation marker sodium/iodide symporter (NIS) through ERK1/2 activation. However, SST increases AKT activation and the inhibition of the Src/PI3K/AKT pathway increases NIS levels in SST-treated cells. Thus, we conclude that, in thyroid cells, signalling from SST receptors to ERK1/2 involves a Gßγ-mediated signal acting on a Ras-dependent pathway. Moreover, we demonstrate that SST might regulates NIS expression through a Src/PI3K/AKT-dependent mechanism, but not through ERK1/2 signalling, showing the main role of this hormone in thyroid function.


Subject(s)
GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , MAP Kinase Signaling System , Somatostatin/administration & dosage , Thyroid Gland/drug effects , ras Proteins/metabolism , Cell Line , Humans , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Thyroid Gland/cytology , Thyroid Gland/metabolism
4.
Cancer Lett ; 335(1): 232-41, 2013 Jul 10.
Article in English | MEDLINE | ID: mdl-23435375

ABSTRACT

BRAF is a main oncogene in human thyroid cancer. Here, we show that BRAF depletion by siRNA or inhibition of its activity by treatment with BRAF inhibitor PLX4720 decreases migration and invasion in thyroid cancer cells expressing oncogenic (V600E)BRAF through a MEK/ERK-dependent mechanism, since treatment with the MEK inhibitor U0126 exerts the same effect. Moreover, over-expression of (V600E)BRAF increases migration and invasion of wild-type BRAF thyroid cells. Using the same strategies, we demonstrate that these effects are mediated by upregulation of the transcriptional repressor Snail with a concomitant decrease of its target E-cadherin, both hallmarks of EMT. These results reveal a novel (V600E)BRAF-induced mechanism in thyroid tumours progression and provides a rationale for using the PLX4720 inhibitor to target (V600E)BRAF signalling to effectively control progression of thyroid cancer.


Subject(s)
Cadherins/metabolism , Carcinoma/metabolism , Indoles/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Sulfonamides/pharmacology , Thyroid Neoplasms/metabolism , Transcription Factors/metabolism , Antigens, CD , Butadienes/pharmacology , Cadherins/genetics , Carcinoma/pathology , Carcinoma, Papillary , Cell Line, Tumor , Cell Movement , Gene Expression , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/metabolism , Mutation, Missense , Neoplasm Invasiveness , Nitriles/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/metabolism , RNA, Small Interfering/genetics , Snail Family Transcription Factors , Thyroid Cancer, Papillary , Thyroid Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...