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










Database
Publication year range
1.
Mol Cell Oncol ; 8(4): 1927446, 2021.
Article in English | MEDLINE | ID: mdl-34616867

ABSTRACT

Conversion of peptidyl-arginine to peptidyl citrulline, known as citrullination, is a post-translational protein modification catalyzed by the PADI (Protein Arginine Deiminase) family of enzymes. PADI1 and PADI3 catalyze citrullination of arginine 106 in the glycolytic enzyme pyruvate kinase M2 modulating its allosteric regulation, glycolysis and cancer cell proliferation.

2.
Nat Commun ; 12(1): 1718, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33741961

ABSTRACT

Chromodomain helicase DNA binding protein 4 (CHD4) is an ATPase subunit of the Nucleosome Remodelling and Deacetylation (NuRD) complex that regulates gene expression. CHD4 is essential for growth of multiple patient derived melanoma xenografts and for breast cancer. Here we show that CHD4 regulates expression of PADI1 (Protein Arginine Deiminase 1) and PADI3 in multiple cancer cell types modulating citrullination of arginine residues of the allosterically-regulated glycolytic enzyme pyruvate kinase M2 (PKM2). Citrullination of PKM2 R106 reprogrammes cross-talk between PKM2 ligands lowering its sensitivity to the inhibitors Tryptophan, Alanine and Phenylalanine and promoting activation by Serine. Citrullination thus bypasses normal physiological regulation by low Serine levels to promote excessive glycolysis and reduced cell proliferation. We further show that PADI1 and PADI3 expression is up-regulated by hypoxia where PKM2 citrullination contributes to increased glycolysis. We provide insight as to how conversion of arginines to citrulline impacts key interactions within PKM2 that act in concert to reprogramme its activity as an additional mechanism regulating this important enzyme.


Subject(s)
Cell Proliferation/physiology , Citrullination/physiology , Glycolysis/physiology , Neoplasms/metabolism , Protein-Arginine Deiminase Type 1/metabolism , Protein-Arginine Deiminase Type 3/metabolism , Pyruvate Kinase/metabolism , Allosteric Regulation , Carrier Proteins/genetics , Carrier Proteins/metabolism , Humans , Melanoma , Membrane Proteins , Mi-2 Nucleosome Remodeling and Deacetylase Complex , Neoplasms/genetics , Protein-Arginine Deiminase Type 1/genetics , Protein-Arginine Deiminase Type 3/genetics , Thyroid Hormones , Up-Regulation , Thyroid Hormone-Binding Proteins
3.
Cell Death Differ ; 27(1): 29-43, 2020 01.
Article in English | MEDLINE | ID: mdl-31065107

ABSTRACT

Somatic oncogenic mutation of BRAF coupled with inactivation of PTEN constitute a frequent combination of genomic alterations driving the development of human melanoma. Mice genetically engineered to conditionally express oncogenic BrafV600E and inactivate Pten in melanocytes following tamoxifen treatment rapidly develop melanoma. While early-stage melanomas comprised melanin-pigmented Mitf and Dct-expressing cells, expression of these and other melanocyte identity genes was lost in later stage tumours that showed histological and molecular characteristics of de-differentiated neural crest type cells. Melanocyte identity genes displayed loss of active chromatin marks and RNA polymerase II and gain of heterochromatin marks, indicating epigenetic reprogramming during tumour progression. Nevertheless, late-stage tumour cells grown in culture re-expressed Mitf, and melanocyte markers and Mitf together with Sox10 coregulated a large number of genes essential for their growth. In this melanoma model, somatic inactivation that the catalytic Brg1 (Smarca4) subunit of the SWI/SNF complex and the scaffolding Bptf subunit of the NuRF complex delayed tumour formation and deregulated large and overlapping gene expression programs essential for normal tumour cell growth. Moreover, we show that Brg1 and Bptf coregulated many genes together with Mitf and Sox10. Together these transcription factors and chromatin remodelling complexes orchestrate essential gene expression programs in mouse melanoma cells.


Subject(s)
Antigens, Nuclear/physiology , DNA Helicases/physiology , Gene Expression Regulation, Neoplastic , Melanoma, Experimental/genetics , Nerve Tissue Proteins/physiology , Nuclear Proteins/physiology , Proto-Oncogene Proteins B-raf/genetics , Skin Neoplasms/genetics , Transcription Factors/physiology , Animals , Antigens, Nuclear/genetics , DNA Helicases/genetics , Disease Progression , Epigenesis, Genetic , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Mutation , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , PTEN Phosphohydrolase/genetics , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Transcription Factors/genetics , Tumor Cells, Cultured
4.
J Invest Dermatol ; 139(8): 1769-1778.e2, 2019 08.
Article in English | MEDLINE | ID: mdl-30776432

ABSTRACT

In melanoma, initiating oncogenic mutations in BRAF or NRAS are detected in premalignant lesions that accumulate additional mutations and genomic instability as the tumor evolves to the metastatic state. Here we investigate evolution of clonal composition and neoantigen landscape in an atypical melanoma displaying recurrent cutaneous lesions over a 6-year period without development of extracutaneous metastases. Whole exome sequencing of four cutaneous lesions taken during the 6-year period identified a collection of single nucleotide variants and small insertions and deletions shared among all tumors, along with progressive selection of subclones displaying fewer single nucleotide variants. Later tumors also displayed lower neoantigen burden compared to early tumors, suggesting that clonal evolution was driven, at least in part, by counter selection of subclones with high neoantigen burdens. Among the selected mutations are a missense mutation in MAP2K1 (F53Y) and an inversion on chromosome 7 generating a AKAP9-BRAF fusion. The mutant proteins cooperatively activate the MAPK signaling pathway confirming they are potential driver mutations of this tumor. We therefore describe the long-term genetic evolution of cutaneous metastatic melanoma characterized by an unexpected phenotypic stability and neoantigen-driven clonal selection.


Subject(s)
Antigens, Neoplasm/metabolism , Carcinogenesis/genetics , Melanoma/genetics , Neoplasm Recurrence, Local/genetics , Skin Neoplasms/genetics , A Kinase Anchor Proteins/genetics , Adult , Antigens, Neoplasm/immunology , Biopsy , Carcinogenesis/immunology , Clonal Evolution , Cytoskeletal Proteins/genetics , DNA Mutational Analysis , HEK293 Cells , Humans , MAP Kinase Kinase 1/genetics , Male , Melanoma/immunology , Melanoma/pathology , Mutation , Mutation, Missense , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/pathology , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Proteins B-raf/genetics , Skin/pathology , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Exome Sequencing
5.
Clin Cancer Res ; 23(22): 7097-7107, 2017 Nov 15.
Article in English | MEDLINE | ID: mdl-28855355

ABSTRACT

Purpose: Understanding tumor heterogeneity is an important challenge in current cancer research. Transcription and epigenetic profiling of cultured melanoma cells have defined at least two distinct cell phenotypes characterized by distinctive gene expression signatures associated with high or low/absent expression of microphthalmia-associated transcription factor (MITF). Nevertheless, heterogeneity of cell populations and gene expression in primary human tumors is much less well characterized.Experimental Design: We performed single-cell gene expression analyses on 472 cells isolated from needle biopsies of 5 primary human melanomas, 4 superficial spreading, and one acral melanoma. The expression of MITF-high and MITF-low signature genes was assessed and compared to investigate intra- and intertumoral heterogeneity and correlated gene expression profiles.Results: Single-cell gene expression analyses revealed varying degrees of intra- and intertumor heterogeneity conferred by the variable expression of distinct sets of genes in different tumors. Expression of MITF partially correlated with that of its known target genes, while SOX10 expression correlated best with PAX3 and ZEB2 Nevertheless, cells simultaneously expressing MITF-high and MITF-low signature genes were observed both by single-cell analyses and RNAscope.Conclusions: Single-cell analyses can be performed on limiting numbers of cells from primary human melanomas revealing their heterogeneity. Although tumors comprised variable proportions of cells with the MITF-high and MITF-low gene expression signatures characteristic of melanoma cultures, primary tumors also comprised cells expressing markers of both signatures defining a novel cell state in tumors in vivoClin Cancer Res; 23(22); 7097-107. ©2017 AACR.


Subject(s)
Gene Expression Regulation, Neoplastic , Genetic Heterogeneity , Melanoma/genetics , Melanoma/pathology , Microphthalmia-Associated Transcription Factor/genetics , Adult , Aged, 80 and over , Biomarkers, Tumor , Cell Line, Tumor , Female , Gene Expression Profiling , Humans , Male , Melanoma/metabolism , Microphthalmia-Associated Transcription Factor/metabolism , Mutation , Single-Cell Analysis
6.
Biol Aujourdhui ; 210(4): 283-295, 2016.
Article in French | MEDLINE | ID: mdl-28327285

ABSTRACT

Malignant melanoma is a highly aggressive cancer with a propensity for early metastasis. Melanocyte transformation results predominantly from oncogenic mutations in BRAF, NRAS or NF1 leading to constitutive activation of the MAP kinase pathway driving cell proliferation and second site mutations such as loss of CDKN1A, or PTEN or activating mutations in the beta-catenin pathway that allow escape from oncogene induced senescence. Nevertheless, irrespective of the nature of the driver mutations, melanoma cell physiology is strongly regulated by transcription factors and epigenetic mechanisms. MITF (Microphthalmia-associated Transcription Factor) and SOX10 are two major transcription factors that regulate both normal melanocyte and melanoma cell physiology. Using a combination of mouse genetics, biochemistry and high throughput genomics we have identified cofactors for MITF and addressed the mechanisms by which MITF, SOX10 and their cofactors regulate gene expression in melanocytes and melanoma.


Subject(s)
Epigenesis, Genetic/physiology , Melanoma/genetics , Skin Neoplasms/genetics , Animals , Cell Lineage/genetics , Chromatin Assembly and Disassembly/physiology , Gene Expression Regulation, Neoplastic , Humans , Melanocytes/metabolism , Melanocytes/physiology , Mice , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Protein Binding , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism
7.
PLoS Genet ; 11(10): e1005555, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26440048

ABSTRACT

MIcrophthalmia-associated Transcription Factor (MITF) regulates melanocyte and melanoma physiology. We show that MITF associates the NURF chromatin-remodelling factor in melanoma cells. ShRNA-mediated silencing of the NURF subunit BPTF revealed its essential role in several melanoma cell lines and in untransformed melanocytes in vitro. Comparative RNA-seq shows that MITF and BPTF co-regulate overlapping gene expression programs in cell lines in vitro. Somatic and specific inactivation of Bptf in developing murine melanoblasts in vivo shows that Bptf regulates their proliferation, migration and morphology. Once born, Bptf-mutant mice display premature greying where the second post-natal coat is white. This second coat is normally pigmented by differentiated melanocytes derived from the adult melanocyte stem cell (MSC) population that is stimulated to proliferate and differentiate at anagen. An MSC population is established and maintained throughout the life of the Bptf-mutant mice, but these MSCs are abnormal and at anagen, give rise to reduced numbers of transient amplifying cells (TACs) that do not express melanocyte markers and fail to differentiate into mature melanin producing melanocytes. MSCs display a transcriptionally repressed chromatin state and Bptf is essential for reactivation of the melanocyte gene expression program at anagen, the subsequent normal proliferation of TACs and their differentiation into mature melanocytes.


Subject(s)
Antigens, Nuclear/genetics , Chromatin Assembly and Disassembly/genetics , Melanoma/genetics , Mesenchymal Stem Cells , Microphthalmia-Associated Transcription Factor/genetics , Nerve Tissue Proteins/genetics , Transcription Factors/genetics , Animals , Cell Cycle/genetics , Cell Differentiation/genetics , Cell Division/genetics , Gene Expression Regulation, Developmental , Hair Follicle , Melanocytes/metabolism , Melanoma/pathology , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...