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.
J Biol Chem ; 296: 100163, 2021.
Article in English | MEDLINE | ID: mdl-33288675

ABSTRACT

Uveal melanoma is the most common eye cancer in adults and is clinically and genetically distinct from skin cutaneous melanoma. In a subset of cases, the oncogenic driver is an activating mutation in CYSLTR2, the gene encoding the G protein-coupled receptor cysteinyl-leukotriene receptor 2 (CysLTR2). The mutant CYSLTR2 encodes for the CysLTR2-L129Q receptor, with the substitution of Leu to Gln at position 129 (3.43). The ability of CysLTR2-L129Q to cause malignant transformation has been hypothesized to result from constitutive activity, but how the receptor could escape desensitization is unknown. Here, we characterize the functional properties of CysLTR2-L129Q. We show that CysLTR2-L129Q is a constitutively active mutant that strongly drives Gq/11 signaling pathways. However, CysLTR2-L129Q only poorly recruits ß-arrestin. Using a modified Slack-Hall operational model, we quantified the constitutive activity for both pathways and conclude that CysLTR2-L129Q displays profound signaling bias for Gq/11 signaling pathways while escaping ß-arrestin-mediated downregulation. CYSLTR2 is the first known example of a G protein-coupled receptor driver oncogene that encodes a highly biased constitutively active mutant receptor. These results provide new insights into the mechanism of CysLTR2-L129Q oncoprotein signaling and suggest CYSLTR2 as a promising potential therapeutic target in uveal melanoma.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Gene Expression Regulation, Neoplastic , Receptors, Leukotriene/genetics , Signal Transduction/genetics , beta-Arrestin 2/genetics , Amino Acid Substitution , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Glutamine/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Kinetics , Lysine/metabolism , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Models, Biological , Mutation , Protein Binding , Receptors, Leukotriene/metabolism , Receptors, Vasopressin/genetics , Receptors, Vasopressin/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Uveal Neoplasms/genetics , Uveal Neoplasms/metabolism , Uveal Neoplasms/pathology , beta-Arrestin 2/metabolism
2.
Clin Cancer Res ; 27(5): 1476-1490, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33229459

ABSTRACT

PURPOSE: All uveal melanoma and a fraction of other melanoma subtypes are driven by activation of the G-protein alpha-q (Gαq) pathway. Targeting these melanomas has proven difficult despite advances in the molecular understanding of key driver signaling pathways in the disease pathogenesis. Inhibitors of Gαq have shown promising preclinical results, but their therapeutic activity in distinct Gαq mutational contexts and in vivo have remained elusive. EXPERIMENTAL DESIGN: We used an isogenic melanocytic cellular system to systematically examine hotspot mutations in GNAQ (e.g., G48V, R183Q, Q209L) and CYSLTR2 (L129Q) found in human uveal melanoma. This cellular system and human uveal melanoma cell lines were used in vitro and in in vivo xenograft studies to assess the efficacy of Gαq inhibition as a single agent and in combination with MEK inhibition. RESULTS: We demonstrate that the Gαq inhibitor YM-254890 inhibited downstream signaling and in vitro growth in all mutants. In vivo, YM-254890 slowed tumor growth but did not cause regression in human uveal melanoma xenografts. Through comprehensive transcriptome analysis, we observed that YM-254890 caused inhibition of the MAPK signaling with evidence of rebound by 24 hours and combination treatment of YM-254890 and a MEK inhibitor led to sustained MAPK inhibition. We further demonstrated that the combination caused synergistic growth inhibition in vitro and tumor shrinkage in vivo. CONCLUSIONS: These data suggest that the combination of Gαq and MEK inhibition provides a promising therapeutic strategy and improved therapeutic window of broadly targeting Gαq in uveal melanoma.See related commentary by Neelature Sriramareddy and Smalley, p. 1217.


Subject(s)
Melanoma , Uveal Neoplasms , Cell Line, Tumor , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Humans , Melanoma/drug therapy , Melanoma/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Mutation , Uveal Neoplasms/drug therapy , Uveal Neoplasms/genetics
4.
Nature ; 574(7778): 432-436, 2019 10.
Article in English | MEDLINE | ID: mdl-31597964

ABSTRACT

SF3B1 is the most commonly mutated RNA splicing factor in cancer1-4, but the mechanisms by which SF3B1 mutations promote malignancy are poorly understood. Here we integrated pan-cancer splicing analyses with a positive-enrichment CRISPR screen to prioritize splicing alterations that promote tumorigenesis. We report that diverse SF3B1 mutations converge on repression of BRD9, which is a core component of the recently described non-canonical BAF chromatin-remodelling complex that also contains GLTSCR1 and GLTSCR1L5-7. Mutant SF3B1 recognizes an aberrant, deep intronic branchpoint within BRD9 and thereby induces the inclusion of a poison exon that is derived from an endogenous retroviral element and subsequent degradation of BRD9 mRNA. Depletion of BRD9 causes the loss of non-canonical BAF at CTCF-associated loci and promotes melanomagenesis. BRD9 is a potent tumour suppressor in uveal melanoma, such that correcting mis-splicing of BRD9 in SF3B1-mutant cells using antisense oligonucleotides or CRISPR-directed mutagenesis suppresses tumour growth. Our results implicate the disruption of non-canonical BAF in the diverse cancer types that carry SF3B1 mutations and suggest a mechanism-based therapeutic approach for treating these malignancies.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Neoplasms/genetics , RNA Splicing , Spliceosomes/metabolism , Animals , Cell Line, Tumor , Chromatin Assembly and Disassembly/genetics , Chromosomal Proteins, Non-Histone/genetics , Humans , Mice , Neoplasm Transplantation , Neoplasms/pathology , Phosphoproteins/metabolism , RNA Splicing Factors/metabolism , Spliceosomes/genetics , Transcription Factors/metabolism
5.
Cell Rep ; 22(9): 2455-2468, 2018 02 27.
Article in English | MEDLINE | ID: mdl-29490280

ABSTRACT

Uveal melanoma (UM) is characterized by mutually exclusive activating mutations in GNAQ, GNA11, CYSLTR2, and PLCB4, four genes in a linear pathway to activation of PLCß in almost all tumors and loss of BAP1 in the aggressive subset. We generated mice with melanocyte-specific expression of GNA11Q209L with and without homozygous Bap1 loss. The GNA11Q209L mice recapitulated human Gq-associated melanomas, and they developed pigmented neoplastic lesions from melanocytes of the skin and non-cutaneous organs, including the eye and leptomeninges, as well as at atypical sites, including the lymph nodes and lungs. The addition of Bap1 loss increased tumor proliferation and cutaneous melanoma size. Integrative transcriptome analysis of human and murine melanomas identified RasGRP3 to be specifically expressed in GNAQ/GNA11-driven melanomas. In human UM cell lines and murine models, RasGRP3 is specifically required for GNAQ/GNA11-driven Ras activation and tumorigenesis. This implicates RasGRP3 as a critical node and a potential target in UM.


Subject(s)
GTP-Binding Protein alpha Subunits/metabolism , Melanocytes/metabolism , Melanoma/metabolism , Melanoma/pathology , Signal Transduction , Uveal Neoplasms/metabolism , Uveal Neoplasms/pathology , ras Guanine Nucleotide Exchange Factors/metabolism , Animals , Cell Line, Tumor , Cell Lineage/drug effects , Cell Proliferation/drug effects , Central Nervous System Neoplasms/pathology , Disease Models, Animal , Female , Humans , Male , Melanocytes/drug effects , Melanocytes/pathology , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Invasiveness , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Skin Neoplasms/pathology , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism
6.
J Pathol ; 242(1): 102-112, 2017 05.
Article in English | MEDLINE | ID: mdl-28188619

ABSTRACT

Chromosomal rearrangements encoding oncogenic fusion proteins are found in a wide variety of malignancies. The use of programmable nucleases to generate specific double-strand breaks in endogenous loci, followed by non-homologous end joining DNA repair, has allowed several of these translocations to be generated as constitutively expressed fusion genes within a cell population. Here, we describe a novel approach that combines CRISPR-Cas9 technology with homology-directed repair to engineer, capture, and modulate the expression of chromosomal translocation products in a human cell line. We have applied this approach to the genetic modelling of t(11;22)(q24;q12) and t(11;22)(p13;q12), translocation products of the EWSR1 gene and its 3' fusion partners FLI1 and WT1, present in Ewing's sarcoma and desmoplastic small round cell tumour, respectively. Our innovative approach allows for temporal control of the expression of engineered endogenous chromosomal rearrangements, and provides a means to generate models to study tumours driven by fusion genes. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Desmoplastic Small Round Cell Tumor/genetics , Recombinational DNA Repair/genetics , Sarcoma, Ewing/genetics , Translocation, Genetic , Artificial Gene Fusion/methods , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 22/genetics , DNA, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Oncogene Proteins, Fusion/genetics , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...