Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Hepatol ; 75(2): 351-362, 2021 08.
Article in English | MEDLINE | ID: mdl-33741397

ABSTRACT

BACKGROUND & AIMS: About 15% of intrahepatic cholangiocarcinomas (iCCAs) express fibroblast growth factor receptor 2 (FGFR2) fusion proteins (FFs), usually alongside mutational inactivation of TP53, CDKN2A or BAP1. In FFs, FGFR2 residues 1-768 fuse to sequences encoded by a diverse array of partner genes (>60) causing oncogenic FF activation. While FGFR-specific tyrosine kinase inhibitors (F-TKI) provide clinical benefit in FF+ iCCA, responses are partial and/or limited by resistance mechanisms, such as the V565F substitution in the FGFR2 gatekeeper residue. Improving on FF targeting in iCCA therefore remains a critical unmet need. Herein, we aimed to generate a murine model of FF-driven iCCA and use this to uncover actionable FF-associated dependencies. METHODS: Four iCCA FFs carrying different fusion sequences were expressed in Tp53-/- mouse liver organoids. Tumorigenic properties of genetically modified liver organoids were assessed by transplantation into immuno-deficient mice. Cellular models derived from neoplastic lesions were exploited for pre-clinical studies. RESULTS: Transplantation of FF-expressing liver organoids yielded tumors diagnosed as CCA based on histological, phenotypic and transcriptomic analyses. The penetrance of this tumorigenic phenotype was influenced by FF identity. Tumor organoids and 2D cell lines derived from CCA lesions were addicted to FF signaling via Ras-Erk, regardless of FF identity or V565F mutation. Dual blockade of FF and the Ras-Erk pathway by concomitant pharmacological inhibition of FFs and Mek1/2 provided greater therapeutic efficacy than single agent F-TKI in vitro and in vivo. CONCLUSIONS: FF-driven iCCA pathogenesis was successfully modeled on a Tp53-/- murine background, revealing biological heterogeneity among structurally different FFs. Double blockade of FF-ERK signaling deserves consideration for precision-based approaches against human FF+ iCCA. LAY SUMMARY: Intrahepatic cholangiocarcinoma (iCCA) is a rare cancer that is difficult to treat. A subtype of iCCA is caused by genomic alterations that generate oncogenic drivers known as FGFR2 fusions. Patients with FGFR2 fusions respond to FGFR inhibitors, but clinical responses are often of modest duration. We used animal and cellular models to show that FGFR2 fusions require the activity of a downstream effector named Mek1/2. We found that dual blockade of FGFR2 fusions and Mek1/2 was more effective than isolated inhibition of FGFR2 fusions, pointing to the potential clinical utility of dual FGFR2-MEK1/2 blockade in patients with iCCA.


Subject(s)
Cholangiocarcinoma/etiology , Receptor, Fibroblast Growth Factor, Type 2/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 2/genetics , Tumor Suppressor Protein p53/drug effects , Analysis of Variance , Animals , Cell Line/metabolism , Cholangiocarcinoma/genetics , Disease Models, Animal , Mice , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Signal Transduction/drug effects
2.
Neurobiol Aging ; 84: 239.e9-239.e14, 2019 12.
Article in English | MEDLINE | ID: mdl-31000212

ABSTRACT

Variants in tank-binding kinase 1 (TBK1) are responsible for a significant proportion of amyotrophic lateral sclerosis (ALS) cases. In the present study, we analyzed variants in TBK1 extracted by targeted sequencing of 32 genes in a group of 406 Italian patients with ALS. We identified 7 different TBK1 variants in 7 sporadic cases, resulting in a frequency of 1.7%. Three patients had missense variants (p.R357Q, p.R358H, and p.R724C), one patient had a small deletion (p.E618del), and 3 had truncating variants (p.Y482*, p.R229*, and p.N681*). Notably, we found that 4 patients had an additional variant in ALS-related genes: 2 in OPTN and 2 in the 3'UTR region of FUS. By studying an independent group of 7 TBK1-mutated patients previously reported, we found another variant in the 3'UTR region of FUS in one patient. The presence of a second variant in TBK1 variant carriers is an interesting finding that needs to be investigated in larger cohorts of patients. These findings suggest that TBK1 belongs to the category of genes conferring a significantly increased risk but not sufficient to cause disease.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Genetic Variation , Protein Serine-Threonine Kinases/genetics
3.
Hepatology ; 69(1): 131-142, 2019 01.
Article in English | MEDLINE | ID: mdl-30067876

ABSTRACT

About 15% of intrahepatic cholangiocarcinomas (ICCs) express constitutively active fibroblast growth factor receptor 2 (FGFR2) fusion proteins (FFs) generated by chromosomal translocations. FFs have been nominated as oncogenic drivers because administration of FGFR tyrosine kinase inhibitors (F-TKIs) can elicit meaningful objective clinical responses in patients carrying FF-positive ICC. Thus, optimization of FF targeting is a pressing clinical need. Herein, we report that three different FFs, previously isolated from ICC samples, are heat shock protein 90 (HSP90) clients and undergo rapid degradation upon HSP90 pharmacological blockade by the clinically advanced HSP90 inhibitor ganetespib. Combining catalytic suppression by the F-TKI BGJ398 with HSP90 blockade by ganetespib suppressed FGFR2-TACC3 (transforming acidic coiled-coil containing protein 3) signaling in cultured cells more effectively than either BGJ398 or ganetespib in isolation. The BGJ398 + ganetespib combo was also superior to single agents when tested in mice carrying subcutaneous tumors generated by transplantation of FGFR2-TACC3 NIH3T3 transformants. Of note, FF mutants known to enforce clinical resistance to BGJ398 in ICC patients retained full sensitivity to ganetespib in cultured cells. Conclusion: Our data provide a proof of principle that upfront treatment with the BGJ398 + ganetespib combo improves therapeutic targeting of FGFR2 fusions in an experimental setting, which may be relevant to precision medicine approaches to FF-driven ICC.


Subject(s)
Bile Duct Neoplasms/drug therapy , Cholangiocarcinoma/drug therapy , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/metabolism , Phenylurea Compounds/administration & dosage , Pyrimidines/administration & dosage , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Triazoles/administration & dosage , Animals , Cells, Cultured , Drug Combinations , Female , Humans , Mice
5.
Semin Cell Dev Biol ; 50: 115-24, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26456277

ABSTRACT

The ErbB signaling network instructs the execution of key cellular programs, such as cell survival, proliferation and motility, through the generation of robust signals of defined strength and duration. In contrast, unabated ErbB signaling disrupts tissue homeostasis and leads to cell transformation. Cells oppose the threat inherent in excessive ErbB activity through several mechanisms of negative feedback regulation. Inducible feedback inhibitors (IFIs) are expressed in the context of transcriptional responses triggered by ErbB signaling, thus being uniquely suited to regulate ErbB activity during the execution of complex cellular programs. This review focuses on MIG6, an IFI that restrains ErbB signaling by mediating ErbB kinase suppression and receptor down-regulation. We will review key issues in MIG6 function, regulation and tumor suppressor activity. Subsequently, the role for MIG6 loss in the pathogenesis of tumors driven by ErbB oncogenes as well as in the generation of cellular addiction to ErbB signaling will be discussed. We will conclude by analyzing feedback inhibition by MIG6 in the context of therapies directed against ErbB and non-ErbB oncogenes.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , ErbB Receptors/metabolism , Feedback, Physiological , Molecular Targeted Therapy , Oncogenes , Tumor Suppressor Proteins/metabolism , Animals , Humans
6.
Mol Cell Endocrinol ; 394(1-2): 105-14, 2014 Aug 25.
Article in English | MEDLINE | ID: mdl-25025809

ABSTRACT

GDNF is a Sertoli-cell-derived factor that controls the balance between self-renewal and differentiation of the spermatogonial stem cells. Although research in recent years has concentrated on the impact of GDNF on target germ cells rather little attention has been paid to the molecular control of GDNF expression in Sertoli cells. Here, we aimed to characterize the promoter region of the mouse gdnf gene active in Sertoli cells. We identified the transcriptional start sites and analyzed the promoter activity of the 5'-flanking regions. By in-silico analysis of evolutionarily conserved DNA sequences we identified several putative transcription factor-binding regions. Deletion analysis showed the involvement of the three CRE sites for basal and cAMP-induced expression of gdnf in murine Sertoli cells. These results provide the basis for future studies to analyze how hormonal or paracrine signals modulate the transcriptional activity of gdnf in Sertoli cells.


Subject(s)
5' Flanking Region , Glial Cell Line-Derived Neurotrophic Factor/genetics , Promoter Regions, Genetic , Sertoli Cells/metabolism , Transcription, Genetic , Animals , Base Sequence , Binding Sites , Cell Differentiation , Conserved Sequence , Cyclic AMP/metabolism , Gene Expression Regulation, Developmental , Glial Cell Line-Derived Neurotrophic Factor/chemistry , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Primary Cell Culture , Seminiferous Tubules/cytology , Seminiferous Tubules/metabolism , Sertoli Cells/cytology , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Initiation Site
7.
PLoS One ; 8(4): e59431, 2013.
Article in English | MEDLINE | ID: mdl-23613711

ABSTRACT

In mammals, the biological activity of the stem/progenitor compartment sustains production of mature gametes through spermatogenesis. Spermatogonial stem cells and their progeny belong to the class of undifferentiated spermatogonia, a germ cell population found on the basal membrane of the seminiferous tubules. A large body of evidence has demonstrated that glial cell line-derived neurotrophic factor (GDNF), a Sertoli-derived factor, is essential for in vivo and in vitro stem cell self-renewal. However, the mechanisms underlying this activity are not completely understood. In this study, we show that GDNF induces dose-dependent directional migration of freshly selected undifferentiated spermatogonia, as well as germline stem cells in culture, using a Boyden chamber assay. GDNF-induced migration is dependent on the expression of the GDNF co-receptor GFRA1, as shown by migration assays performed on parental and GFRA1-transduced GC-1 spermatogonial cell lines. We found that the actin regulatory protein vasodilator-stimulated phosphoprotein (VASP) is specifically expressed in undifferentiated spermatogonia. VASP belongs to the ENA/VASP family of proteins implicated in actin-dependent processes, such as fibroblast migration, axon guidance, and cell adhesion. In intact seminiferous tubules and germline stem cell cultures, GDNF treatment up-regulates VASP in a dose-dependent fashion. These data identify a novel role for the niche-derived factor GDNF, and they suggest that GDNF may impinge on the stem/progenitor compartment, affecting the actin cytoskeleton and cell migration.


Subject(s)
Cell Movement/drug effects , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Spermatogonia/cytology , Stem Cell Niche , Stem Cells/cytology , Stem Cells/drug effects , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Actins/metabolism , Animals , Cell Adhesion Molecules/metabolism , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Phosphoproteins/metabolism , Rats , Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...