Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Front Cell Dev Biol ; 12: 1316048, 2024.
Article in English | MEDLINE | ID: mdl-38444828

ABSTRACT

Mutations in PRDM15 lead to a syndromic form of holoprosencephaly (HPE) known as the Galloway-Mowat syndrome (GAMOS). While a connection between PRDM15, a zinc finger transcription factor, and WNT/PCP signaling has been established, there is a critical need to delve deeper into their contributions to early development and GAMOS pathogenesis. We used the South African clawed frog Xenopus laevis as the vertebrate model organism and observed that prdm15 was enriched in the tissues and organs affected in GAMOS. Furthermore, we generated a morpholino oligonucleotide-mediated prdm15 knockdown model showing that the depletion of Prdm15 leads to abnormal eye, head, and brain development, effectively recapitulating the anterior neural features in GAMOS. An analysis of the underlying molecular basis revealed a reduced expression of key genes associated with eye, head, and brain development. Notably, this reduction could be rescued by the introduction of wnt4 RNA, particularly during the induction of the respective tissues. Mechanistically, our data demonstrate that Prdm15 acts upstream of both canonical and non-canonical Wnt4 signaling during anterior neural development. Our findings describe severe ocular and anterior neural abnormalities upon Prdm15 depletion and elucidate the role of Prdm15 in canonical and non-canonical Wnt4 signaling.

2.
bioRxiv ; 2023 Dec 10.
Article in English | MEDLINE | ID: mdl-38106050

ABSTRACT

Targeting cancer stem cells (CSCs) is crucial for effective cancer treatment 1 . However, the molecular mechanisms underlying resistance to LGR5 + CSCs depletion in colorectal cancer (CRC) 2,3 remain largely elusive. Here, we unveil the existence of a primitive cell state dubbed the oncofetal (OnF) state, which works in tandem with the LGR5 + stem cells (SCs) to fuel tumor evolution in CRC. OnF cells emerge early during intestinal tumorigenesis and exhibit features of lineage plasticity. Normally suppressed by the Retinoid X Receptor (RXR) in mature SCs, the OnF program is triggered by genetic deletion of the gatekeeper APC. We demonstrate that diminished RXR activity unlocks an epigenetic circuity governed by the cooperative action of YAP and AP1, leading to OnF reprogramming. This high-plasticity state is inherently resistant to conventional chemotherapies and its adoption by LGR5 + CSCs enables them to enter a drug-tolerant state. Furthermore, through phenotypic tracing and ablation experiments, we uncover a functional redundancy between the OnF and stem cell (SC) states and show that targeting both cellular states is essential for sustained tumor regression in vivo . Collectively, these findings establish a mechanistic foundation for developing effective combination therapies with enduring impact on CRC treatment.

3.
Mol Cell ; 83(23): 4255-4271.e9, 2023 Dec 07.
Article in English | MEDLINE | ID: mdl-37995687

ABSTRACT

Endogenous retroviruses (ERVs) are remnants of ancient parasitic infections and comprise sizable portions of most genomes. Although epigenetic mechanisms silence most ERVs by generating a repressive environment that prevents their expression (heterochromatin), little is known about mechanisms silencing ERVs residing in open regions of the genome (euchromatin). This is particularly important during embryonic development, where induction and repression of distinct classes of ERVs occur in short temporal windows. Here, we demonstrate that transcription-associated RNA degradation by the nuclear RNA exosome and Integrator is a regulatory mechanism that controls the productive transcription of most genes and many ERVs involved in preimplantation development. Disrupting nuclear RNA catabolism promotes dedifferentiation to a totipotent-like state characterized by defects in RNAPII elongation and decreased expression of long genes (gene-length asymmetry). Our results indicate that RNA catabolism is a core regulatory module of gene networks that safeguards RNAPII activity, ERV expression, cell identity, and developmental potency.


Subject(s)
Endogenous Retroviruses , Endogenous Retroviruses/genetics , RNA, Nuclear , Epigenesis, Genetic , Heterochromatin , Gene Expression
4.
Nat Cancer ; 4(8): 1157-1175, 2023 08.
Article in English | MEDLINE | ID: mdl-37537299

ABSTRACT

Hepatocellular carcinoma (HCC) is a leading cause of cancer-related deaths worldwide. ß-Catenin (CTNNB1)-mutated HCC represents 30% of cases of the disease with no precision therapeutics available. Using chemical libraries derived from clinical multi-kinase inhibitor (KI) scaffolds, we screened HCC organoids to identify WNTinib, a KI with exquisite selectivity in CTNNB1-mutated human and murine models, including patient samples. Multiomic and target engagement analyses, combined with rescue experiments and in vitro and in vivo efficacy studies, revealed that WNTinib is superior to clinical KIs and inhibits KIT/mitogen-activated protein kinase (MAPK) signaling at multiple nodes. Moreover, we demonstrate that reduced engagement on BRAF and p38α kinases by WNTinib relative to several multi-KIs is necessary to avoid compensatory feedback signaling-providing a durable and selective transcriptional repression of mutant ß-catenin/Wnt targets through nuclear translocation of the EZH2 transcriptional repressor. Our studies uncover a previously unknown mechanism to harness the KIT/MAPK/EZH2 pathway to potently and selectively antagonize CTNNB1-mutant HCC with an unprecedented wide therapeutic index.


Subject(s)
Antineoplastic Agents , Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Mice , Animals , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , beta Catenin/genetics , beta Catenin/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Transcription Factors/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use
5.
Curr Res Immunol ; 3: 215-221, 2022.
Article in English | MEDLINE | ID: mdl-36065205

ABSTRACT

As vaccine deployment improves the healthcare emergency status caused by the SARS-CoV-2 pandemic, we need reliable tools to evaluate the duration of protective immunity at a global scale. Seminal studies have demonstrated that while neutralizing antibodies can protect us from viral infection, T cell-mediated cellular immunity provides long-term protection from severe COVID-19, even in the case of emerging new variants of concern (VOC). Indeed, the emergence of VOCs, able to substantially escape antibodies generated by current vaccines, has made the analysis of correlates of humoral protection against infection obsolete. The focus should now shift towards immunological correlates of protection from disease based on quantification of cellular immunity. Despite this evidence, an assessment of T cell responses is still overlooked. This is largely due to technical challenges and lack of validated diagnostic tests. Here, we review the current state of the art of available tests to distinguish between SARS-CoV-2 antigen-specific Tcells and non-antigen specific T-cells. These assays range from the analysis of the T cell-receptor (TCR) diversity (i.e. Immunoseq and MHC tetramer staining) to the detection of functional T cell activation (i.e. ICS, AIM, Elispot, ELLA, dqTACT, etc.) either from purified Peripheral Blood Mononuclear Cells (PBMCs) or whole blood. We discuss advantages and disadvantages of each assay, proposing their ideal use for different scopes. Finally, we argue how it is paramount to deploy cheap, standardized, and scalable assays to measure T cell functionality to fill this critical diagnostic gap and manage these next years of the pandemic.

6.
Nat Biotechnol ; 40(11): 1680-1689, 2022 11.
Article in English | MEDLINE | ID: mdl-35697804

ABSTRACT

Fast, high-throughput methods for measuring the level and duration of protective immune responses to SARS-CoV-2 are needed to anticipate the risk of breakthrough infections. Here we report the development of two quantitative PCR assays for SARS-CoV-2-specific T cell activation. The assays are rapid, internally normalized and probe-based: qTACT requires RNA extraction and dqTACT avoids sample preparation steps. Both assays rely on the quantification of CXCL10 messenger RNA, a chemokine whose expression is strongly correlated with activation of antigen-specific T cells. On restimulation of whole-blood cells with SARS-CoV-2 viral antigens, viral-specific T cells secrete IFN-γ, which stimulates monocytes to produce CXCL10. CXCL10 mRNA can thus serve as a proxy to quantify cellular immunity. Our assays may allow large-scale monitoring of the magnitude and duration of functional T cell immunity to SARS-CoV-2, thus helping to prioritize revaccination strategies in vulnerable populations.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , SARS-CoV-2/genetics , Immunity, Cellular , Polymerase Chain Reaction , T-Lymphocytes
7.
FEBS J ; 289(5): 1256-1275, 2022 03.
Article in English | MEDLINE | ID: mdl-33774927

ABSTRACT

PRDF1 and RIZ1 homology domain containing (PRDMs) are a subfamily of Krüppel-like zinc finger proteins controlling key processes in metazoan development and in cancer. PRDMs exhibit unique dualities: (a) PR domain/ZNF arrays-their structure combines a SET-like domain known as a PR domain, typically found in methyltransferases, with a variable array of C2H2 zinc fingers (ZNF) characteristic of DNA-binding transcription factors; (b) transcriptional activators/repressors-their physiological function is context- and cell-dependent; mechanistically, some PRDMs have a PKMT activity and directly catalyze histone lysine methylation, while others are rather pseudomethyltransferases and act by recruiting transcriptional cofactors; (c) oncogenes/tumor suppressors-their pathological function depends on the specific PRDM isoform expressed during tumorigenesis. This duality is well known as the 'Yin and Yang' of PRDMs and involves a complex regulation of alternative splicing or alternative promoter usage, to generate full-length or PR-deficient isoforms with opposing functions in cancer. In conclusion, once their dualities are fully appreciated, PRDMs represent a promising class of targets in oncology by virtue of their widespread upregulation across multiple tumor types and their somatic dispensability, conferring a broad therapeutic window and limited toxic side effects. The recent discovery of a first-in-class compound able to inhibit PRDM9 activity has paved the way for the identification of further small molecular inhibitors able to counteract PRDM oncogenic activity.


Subject(s)
Epigenesis, Genetic , Protein Methyltransferases/metabolism , Amino Acid Sequence , Carcinogenesis , Crystallization , DNA/metabolism , Meiosis , Neoplasms/enzymology , Neoplasms/pathology , Oncogenes , Protein Binding , Protein Conformation , Protein Domains , Protein Methyltransferases/chemistry , Protein Methyltransferases/genetics , Sequence Alignment , Signal Transduction
8.
Curr Opin Pharmacol ; 59: 33-42, 2021 08.
Article in English | MEDLINE | ID: mdl-34052526

ABSTRACT

Protein arginine methylation is an abundant post-translational modification involved in the modulation of essential cellular processes ranging from transcription, post-transcriptional RNA metabolism, and propagation of signaling cascades to the regulation of the DNA damage response. Excitingly for the field, in the past few years there have been remarkable advances in the development of molecular tools and clinical compounds able to selectively and potently inhibit protein arginine methyltransferase (PRMT) functions. In this review, we first discuss how the somatic mutations that confer advantages to cancer cells are often associated with vulnerabilities that can be exploited by PRMTs' inhibition. In a second part, we discuss strategies to uncover synthetic lethal combinations between existing therapies and PRMT inhibitors.


Subject(s)
Neoplasms , Protein-Arginine N-Methyltransferases , Arginine/genetics , Arginine/metabolism , Humans , Methylation , Neoplasms/drug therapy , Protein Processing, Post-Translational , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism
9.
J Am Soc Nephrol ; 32(3): 580-596, 2021 03.
Article in English | MEDLINE | ID: mdl-33593823

ABSTRACT

BACKGROUND: Galloway-Mowat syndrome (GAMOS) is characterized by neurodevelopmental defects and a progressive nephropathy, which typically manifests as steroid-resistant nephrotic syndrome. The prognosis of GAMOS is poor, and the majority of children progress to renal failure. The discovery of monogenic causes of GAMOS has uncovered molecular pathways involved in the pathogenesis of disease. METHODS: Homozygosity mapping, whole-exome sequencing, and linkage analysis were used to identify mutations in four families with a GAMOS-like phenotype, and high-throughput PCR technology was applied to 91 individuals with GAMOS and 816 individuals with isolated nephrotic syndrome. In vitro and in vivo studies determined the functional significance of the mutations identified. RESULTS: Three biallelic variants of the transcriptional regulator PRDM15 were detected in six families with proteinuric kidney disease. Four families with a variant in the protein's zinc-finger (ZNF) domain have additional GAMOS-like features, including brain anomalies, cardiac defects, and skeletal defects. All variants destabilize the PRDM15 protein, and the ZNF variant additionally interferes with transcriptional activation. Morpholino oligonucleotide-mediated knockdown of Prdm15 in Xenopus embryos disrupted pronephric development. Human wild-type PRDM15 RNA rescued the disruption, but the three PRDM15 variants did not. Finally, CRISPR-mediated knockout of PRDM15 in human podocytes led to dysregulation of several renal developmental genes. CONCLUSIONS: Variants in PRDM15 can cause either isolated nephrotic syndrome or a GAMOS-type syndrome on an allelic basis. PRDM15 regulates multiple developmental kidney genes, and is likely to play an essential role in renal development in humans.


Subject(s)
DNA-Binding Proteins/genetics , Hernia, Hiatal/genetics , Microcephaly/genetics , Mutation, Missense , Nephrosis/genetics , Transcription Factors/genetics , Amino Acid Sequence , Amino Acid Substitution , Animals , Cell Line , Child, Preschool , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/deficiency , Female , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Gene Knockout Techniques , High-Throughput Nucleotide Sequencing , Humans , Infant , Infant, Newborn , Male , Models, Molecular , Nephrotic Syndrome/genetics , Podocytes/metabolism , Polymorphism, Single Nucleotide , Pronephros/embryology , Pronephros/metabolism , Protein Stability , Transcription Factors/chemistry , Transcription Factors/deficiency , Xenopus laevis/embryology , Xenopus laevis/genetics , Zinc Fingers/genetics
10.
Development ; 147(21)2020 11 05.
Article in English | MEDLINE | ID: mdl-33033118

ABSTRACT

Mitchell-Riley syndrome (MRS) is caused by recessive mutations in the regulatory factor X6 gene (RFX6) and is characterised by pancreatic hypoplasia and neonatal diabetes. To determine why individuals with MRS specifically lack pancreatic endocrine cells, we micro-CT imaged a 12-week-old foetus homozygous for the nonsense mutation RFX6 c.1129C>T, which revealed loss of the pancreas body and tail. From this foetus, we derived iPSCs and show that differentiation of these cells in vitro proceeds normally until generation of pancreatic endoderm, which is significantly reduced. We additionally generated an RFX6HA reporter allele by gene targeting in wild-type H9 cells to precisely define RFX6 expression and in parallel performed in situ hybridisation for RFX6 in the dorsal pancreatic bud of a Carnegie stage 14 human embryo. Both in vitro and in vivo, we find that RFX6 specifically labels a subset of PDX1-expressing pancreatic endoderm. In summary, RFX6 is essential for efficient differentiation of pancreatic endoderm, and its absence in individuals with MRS specifically impairs formation of endocrine cells of the pancreas head and tail.


Subject(s)
Cell Differentiation , Diabetes Mellitus/genetics , Diabetes Mellitus/pathology , Endoderm/embryology , Gallbladder Diseases/genetics , Gallbladder Diseases/pathology , Induced Pluripotent Stem Cells/pathology , Intestinal Atresia/genetics , Intestinal Atresia/pathology , Mutation/genetics , Pancreas/embryology , Regulatory Factor X Transcription Factors/genetics , Alleles , Base Sequence , Cell Differentiation/genetics , Chromatin/metabolism , Consanguinity , Diabetes Mellitus/diagnostic imaging , Embryo, Mammalian/metabolism , Embryonic Development , Family , Female , Gallbladder Diseases/diagnostic imaging , Genome, Human , Humans , Induced Pluripotent Stem Cells/metabolism , Intestinal Atresia/diagnostic imaging , Male , Pedigree , Transcription, Genetic , Transcriptome/genetics , X-Ray Microtomography
11.
Nat Commun ; 11(1): 3520, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32665551

ABSTRACT

PRDM (PRDI-BF1 and RIZ homology domain containing) family members are sequence-specific transcriptional regulators involved in cell identity and fate determination, often dysregulated in cancer. The PRDM15 gene is of particular interest, given its low expression in adult tissues and its overexpression in B-cell lymphomas. Despite its well characterized role in stem cell biology and during early development, the role of PRDM15 in cancer remains obscure. Herein, we demonstrate that while PRDM15 is largely dispensable for mouse adult somatic cell homeostasis in vivo, it plays a critical role in B-cell lymphomagenesis. Mechanistically, PRDM15 regulates a transcriptional program that sustains the activity of the PI3K/AKT/mTOR pathway and glycolysis in B-cell lymphomas. Abrogation of PRDM15 induces a metabolic crisis and selective death of lymphoma cells. Collectively, our data demonstrate that PRDM15 fuels the metabolic requirement of B-cell lymphomas and validate it as an attractive and previously unrecognized target in oncology.


Subject(s)
DNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Blotting, Western , Cell Survival/genetics , Cell Survival/physiology , Chromatin Immunoprecipitation , Computational Biology , DNA-Binding Proteins/genetics , Female , Flow Cytometry , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Humans , Lymphoma/genetics , Lymphoma/metabolism , Mice , Mice, SCID , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Random Allocation , Transcription Factors/genetics , Transcriptome/genetics
12.
Sci Adv ; 6(2): eaax9852, 2020 01.
Article in English | MEDLINE | ID: mdl-31950080

ABSTRACT

Holoprosencephaly (HPE) is a congenital forebrain defect often associated with embryonic lethality and lifelong disabilities. Currently, therapeutic and diagnostic options are limited by lack of knowledge of potential disease-causing mutations. We have identified a new mutation in the PRDM15 gene (C844Y) associated with a syndromic form of HPE in multiple families. We demonstrate that C844Y is a loss-of-function mutation impairing PRDM15 transcriptional activity. Genetic deletion of murine Prdm15 causes anterior/posterior (A/P) patterning defects and recapitulates the brain malformations observed in patients. Mechanistically, PRDM15 regulates the transcription of key effectors of the NOTCH and WNT/PCP pathways to preserve early midline structures in the developing embryo. Analysis of a large cohort of patients with HPE revealed potentially damaging mutations in several regulators of both pathways. Our findings uncover an unexpected link between NOTCH and WNT/PCP signaling and A/P patterning and set the stage for the identification of new HPE candidate genes.


Subject(s)
Cell Polarity , DNA-Binding Proteins/genetics , Holoprosencephaly/genetics , Loss of Function Mutation/genetics , Receptors, Notch/metabolism , Transcription Factors/genetics , Wnt Signaling Pathway , Animals , Body Patterning/genetics , Brain/abnormalities , Brain/embryology , Cell Polarity/genetics , Cohort Studies , Embryo, Mammalian/abnormalities , Embryo, Mammalian/metabolism , Female , Gene Deletion , Gene Expression Regulation, Developmental , Humans , Mice , Neural Plate/metabolism , Pregnancy , Transcription, Genetic , Zinc Fingers
13.
Cancer Cell ; 36(2): 194-209.e9, 2019 08 12.
Article in English | MEDLINE | ID: mdl-31408619

ABSTRACT

Cancer-associated mutations in genes encoding RNA splicing factors (SFs) commonly occur in leukemias, as well as in a variety of solid tumors, and confer dependence on wild-type splicing. These observations have led to clinical efforts to directly inhibit the spliceosome in patients with refractory leukemias. Here, we identify that inhibiting symmetric or asymmetric dimethylation of arginine, mediated by PRMT5 and type I protein arginine methyltransferases (PRMTs), respectively, reduces splicing fidelity and results in preferential killing of SF-mutant leukemias over wild-type counterparts. These data identify genetic subsets of cancer most likely to respond to PRMT inhibition, synergistic effects of combined PRMT5 and type I PRMT inhibition, and a mechanistic basis for the therapeutic efficacy of PRMT inhibition in cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Ethylenediamines/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Pyrroles/pharmacology , RNA Splicing/drug effects , RNA, Neoplasm/metabolism , Animals , Antineoplastic Agents/pharmacokinetics , Catalysis , Enzyme Inhibitors/pharmacokinetics , Ethylenediamines/pharmacokinetics , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , K562 Cells , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Pyrroles/pharmacokinetics , RNA, Neoplasm/genetics , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/metabolism , THP-1 Cells , Tumor Cells, Cultured , U937 Cells , Xenograft Model Antitumor Assays
14.
Development ; 146(19)2019 07 10.
Article in English | MEDLINE | ID: mdl-30846446

ABSTRACT

Global epigenetic reprogramming is vital to purge germ cell-specific epigenetic features to establish the totipotent state of the embryo. This process transpires to be carefully regulated and is not an undirected, radical erasure of parental epigenomes. The TRIM28 complex has been shown to be crucial in embryonic epigenetic reprogramming by regionally opposing DNA demethylation to preserve vital parental information to be inherited from germline to soma. Yet the DNA-binding factors guiding this complex to specific targets are largely unknown. Here, we uncover and characterize a novel, maternally expressed, TRIM28-interacting KRAB zinc-finger protein: ZFP708. It recruits the repressive TRIM28 complex to RMER19B retrotransposons to evoke regional heterochromatin formation. ZFP708 binding to these hitherto unknown TRIM28 targets is DNA methylation and H3K9me3 independent. ZFP708 mutant mice are viable and fertile, yet embryos fail to inherit and maintain DNA methylation at ZFP708 target sites. This can result in activation of RMER19B-adjacent genes, while ectopic expression of ZFP708 results in transcriptional repression. Finally, we describe the evolutionary conservation of ZFP708 in mice and rats, which is linked to the conserved presence of the targeted RMER19B retrotransposons in these species.


Subject(s)
Epigenetic Repression , Repressor Proteins/metabolism , Retroelements/genetics , Zinc Fingers , Animals , Base Sequence , Binding Sites/genetics , Blastocyst/metabolism , DNA Methylation/genetics , Embryo, Mammalian/metabolism , Evolution, Molecular , Mice , Mice, Knockout , Mouse Embryonic Stem Cells/metabolism , Protein Binding/genetics , Rats , Transcription, Genetic , Tripartite Motif-Containing Protein 28/metabolism
15.
Nat Genet ; 49(9): 1354-1363, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28740264

ABSTRACT

The transcriptional network acting downstream of LIF, WNT and MAPK-ERK to stabilize mouse embryonic stem cells (ESCs) in their naive state has been extensively characterized. However, the upstream factors regulating these three signaling pathways remain largely uncharted. PR-domain-containing proteins (PRDMs) are zinc-finger sequence-specific chromatin factors that have essential roles in embryonic development and cell fate decisions. Here we characterize the transcriptional regulator PRDM15, which acts independently of PRDM14 to regulate the naive state of mouse ESCs. Mechanistically, PRDM15 modulates WNT and MAPK-ERK signaling by directly promoting the expression of Rspo1 (R-spondin1) and Spry1 (Sprouty1). Consistent with these findings, CRISPR-Cas9-mediated disruption of PRDM15-binding sites in the Rspo1 and Spry1 promoters recapitulates PRDM15 depletion, both in terms of local chromatin organization and the transcriptional modulation of these genes. Collectively, our findings uncover an essential role for PRDM15 as a chromatin factor that modulates the transcription of upstream regulators of WNT and MAPK-ERK signaling to safeguard naive pluripotency.


Subject(s)
DNA-Binding Proteins/genetics , Embryonic Stem Cells/metabolism , Gene Expression Regulation , MAP Kinase Signaling System/genetics , Transcription Factors/genetics , Wnt Signaling Pathway/genetics , Animals , Blotting, Western , Cell Line , Cell Self Renewal/genetics , Cells, Cultured , Cellular Reprogramming/genetics , DNA-Binding Proteins/metabolism , Fluorescent Antibody Technique , Gene Expression Profiling/methods , Humans , Induced Pluripotent Stem Cells/metabolism , Mice, Knockout , Mice, Nude , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/metabolism
16.
Bull Cancer ; 104(4): 363-369, 2017 Apr.
Article in French | MEDLINE | ID: mdl-28216075

ABSTRACT

Triple-negative (TN) breast cancer are characterized by lack of estrogen receptor (OR) and progesterone receptor (PR) expression, and the absence of overexpression of human epidermal growth factor receptor 2 (HER2). It is a heterogeneous group of tumors with a more pejorative prognosis than other subtypes of breast cancer. Androgen receptors (AR) are nuclear receptors whose expression varies from 80 to 85% of primary breast cancers and 60 to 75% of metastatic cancers. Among the TN breast cancers, the luminal androgen receptor (LAR) subtype expresses AR more frequently, up to 53% of the cases. AR are associated with lower tumor size, histological grade, Ki67, and lymph node involvement. The results of recent clinical trials evaluating anti-androgen therapies in locally advanced or metastatic TN breast cancer are promising. Many new therapies are tested, including enzalutamide or abiraterone acetate, and numerous therapeutic combinations including PI3K/AKT/mTOR inhibitors or CDK inhibitors. These therapies would allow an alternative treatment of patients with TN breast cancer for which there is often a therapeutic impasse.


Subject(s)
Neoplasm Proteins/metabolism , Receptors, Androgen/metabolism , Triple Negative Breast Neoplasms/metabolism , Abiraterone Acetate/therapeutic use , Androgen Antagonists/therapeutic use , Antineoplastic Agents/therapeutic use , Benzamides , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Female , Humans , Ki-67 Antigen/metabolism , Lymphatic Metastasis , Nitriles , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/therapeutic use , Prognosis , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Receptors, Androgen/chemistry , Steroid 17-alpha-Hydroxylase/antagonists & inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Tumor Burden
17.
Curr Opin Genet Dev ; 36: 83-91, 2016 02.
Article in English | MEDLINE | ID: mdl-27153352

ABSTRACT

The PRDM family of proteins share a unique structure, with an N-terminal PR domain, which has a potential methyltransferase activity, followed by a distinct number of zinc fingers at the C-terminus, potentially mediating protein-protein, protein-RNA or protein-DNA interactions. Interestingly, despite no comprehensive functional data, all family members have been associated with deletions, mutations, epigenetic silencing or overexpression, in multiple cancer types. The intriguing observation is that different isoforms exist for almost all PRDM family members. These isoforms are not only differentially regulated, but play opposite roles in cancer, in what has been termed 'Yin and Yang' regulation, typical of this class of epigenetic regulators. Collectively, these findings set the stage for future intervention, by targeting directly their intrinsic catalytic activities, or indirectly, pathways that differentially regulate tumor suppressor/oncogenic isoform-expression.


Subject(s)
Neoplasms/genetics , Protein Interaction Maps/genetics , Repressor Proteins/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Multigene Family/genetics , Positive Regulatory Domain I-Binding Factor 1 , RNA-Binding Motifs/genetics
18.
Nucleic Acids Res ; 40(19): 9534-42, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22987071

ABSTRACT

p21 is a potent cyclin-dependent kinase inhibitor that plays a role in promoting G1 cell cycle arrest and cellular senescence. Consistent with this role, p21 is a downstream target of several tumour suppressors and oncogenes, and it is downregulated in the majority of tumours, including breast cancer. Here, we report that protein arginine methyltransferase 6 (PRMT6), a type I PRMT known to act as a transcriptional cofactor, directly represses the p21 promoter. PRMT6 knock-down (KD) results in a p21 derepression in breast cancer cells, which is p53-independent, and leads to cell cycle arrest, cellular senescence and reduced growth in soft agar assays and in severe combined immunodeficiency (SCID) mice for all the cancer lines examined. We finally show that bypassing the p21-mediated arrest rescues PRMT6 KD cells from senescence, and it restores their ability to grow on soft agar. We conclude that PRMT6 acts as an oncogene in breast cancer cells, promoting growth and preventing senescence, making it an attractive target for cancer therapy.


Subject(s)
Breast Neoplasms/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Gene Expression Regulation, Neoplastic , Nuclear Proteins/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Proliferation , Cellular Senescence , Female , Gene Knockdown Techniques , Humans , Mice , Mice, SCID , Nuclear Proteins/genetics , Phenotype , Promoter Regions, Genetic , Protein-Arginine N-Methyltransferases/genetics , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL