Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
EMBO Mol Med ; 15(11): e17683, 2023 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-37724723

RESUMEN

Spinal muscular atrophy (SMA) is a leading genetic cause of infant mortality. The advent of approved treatments for this devastating condition has significantly changed SMA patients' life expectancy and quality of life. Nevertheless, these are not without limitations, and research efforts are underway to develop new approaches for improved and long-lasting benefits for patients. Protein arginine methyltransferases (PRMTs) are emerging as druggable epigenetic targets, with several small-molecule PRMT inhibitors already in clinical trials. From a screen of epigenetic molecules, we have identified MS023, a potent and selective type I PRMT inhibitor able to promote SMN2 exon 7 inclusion in preclinical SMA models. Treatment of SMA mice with MS023 results in amelioration of the disease phenotype, with strong synergistic amplification of the positive effect when delivered in combination with the antisense oligonucleotide nusinersen. Moreover, transcriptomic analysis revealed that MS023 treatment has minimal off-target effects, and the added benefit is mainly due to targeting neuroinflammation. Our study warrants further clinical investigation of PRMT inhibition both as a stand-alone and add-on therapy for SMA.


Asunto(s)
Atrofia Muscular Espinal , Calidad de Vida , Animales , Humanos , Lactante , Ratones , Exones , Atrofia Muscular Espinal/tratamiento farmacológico , Atrofia Muscular Espinal/genética , Oligonucleótidos/farmacología , Oligonucleótidos/uso terapéutico , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Proteína 2 para la Supervivencia de la Neurona Motora/uso terapéutico
2.
J Biol Chem ; 297(1): 100821, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34029587

RESUMEN

Viral proteins are known to be methylated by host protein arginine methyltransferases (PRMTs) necessary for the viral life cycle, but it remains unknown whether severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) proteins are methylated. Herein, we show that PRMT1 methylates SARS-CoV-2 nucleocapsid (N) protein at residues R95 and R177 within RGG/RG motifs, preferred PRMT target sequences. We confirmed arginine methylation of N protein by immunoblotting viral proteins extracted from SARS-CoV-2 virions isolated from cell culture. Type I PRMT inhibitor (MS023) or substitution of R95 or R177 with lysine inhibited interaction of N protein with the 5'-UTR of SARS-CoV-2 genomic RNA, a property required for viral packaging. We also defined the N protein interactome in HEK293 cells, which identified PRMT1 and many of its RGG/RG substrates, including the known interacting protein G3BP1 as well as other components of stress granules (SGs), which are part of the host antiviral response. Methylation of R95 regulated the ability of N protein to suppress the formation of SGs, as R95K substitution or MS023 treatment blocked N-mediated suppression of SGs. Also, the coexpression of methylarginine reader Tudor domain-containing protein 3 quenched N protein-mediated suppression of SGs in a dose-dependent manner. Finally, pretreatment of VeroE6 cells with MS023 significantly reduced SARS-CoV-2 replication. Because type I PRMT inhibitors are already undergoing clinical trials for cancer treatment, inhibiting arginine methylation to target the later stages of the viral life cycle such as viral genome packaging and assembly of virions may represent an additional therapeutic application of these drugs.


Asunto(s)
Arginina/metabolismo , COVID-19/metabolismo , COVID-19/virología , Proteínas de la Nucleocápside/química , Proteínas de la Nucleocápside/metabolismo , ARN Viral/metabolismo , SARS-CoV-2/fisiología , Secuencias de Aminoácidos , COVID-19/genética , Gránulos Citoplasmáticos/genética , Gránulos Citoplasmáticos/metabolismo , Células HEK293 , Humanos , Metilación , Proteínas de la Nucleocápside/genética , Estabilidad del ARN , ARN Viral/química , ARN Viral/genética , SARS-CoV-2/química , SARS-CoV-2/genética , Replicación Viral
3.
Front Cell Neurosci ; 15: 633668, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33833668

RESUMEN

Hexanucleotide repeat expansion (G4C2n) mutations in the gene C9ORF72 account for approximately 30% of familial cases of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), as well as approximately 7% of sporadic cases of ALS. G4C2n mutations are known to result in the production of five species of dipeptide repeat proteins (DRPs) through non-canonical translation processes. Arginine-enriched dipeptide repeat proteins, glycine-arginine (polyGR), and proline-arginine (polyPR) have been demonstrated to be cytotoxic and deleterious in multiple experimental systems. Recently, we and others have implicated methylation of polyGR/polyPR arginine residues in disease processes related to G4C2n mutation-mediated neurodegeneration. We previously reported that inhibition of asymmetric dimethylation (ADMe) of arginine residues is protective in cell-based models of polyGR/polyPR cytotoxicity. These results are consistent with the idea that PRMT-mediated arginine methylation in the context of polyGR/polyPR exposure is harmful. However, it remains unclear why. Here we discuss the influence of arginine methylation on diverse cellular processes including liquid-liquid phase separation, chromatin remodeling, transcription, RNA processing, and RNA-binding protein localization, and we consider how methylation of polyGR/polyPR may disrupt processes essential for normal cellular function and survival.

4.
Curr Protein Pept Sci ; 21(7): 699-712, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32379587

RESUMEN

Protein arginine methyltransferase (PRMT) enzymes play a crucial role in RNA splicing, DNA damage repair, cell signaling, and differentiation. Arginine methylation is a prominent posttransitional modification of histones and various non-histone proteins that can either activate or repress gene expression. The aberrant expression of PRMTs has been linked to multiple abnormalities, notably cancer. Herein, we review a number of non-histone protein substrates for all nine members of human PRMTs and how PRMT-mediated non-histone arginine methylation modulates various diseases. Additionally, we highlight the most recent clinical studies for several PRMT inhibitors.


Asunto(s)
Arginina/metabolismo , Enfermedades Cardiovasculares/enzimología , Enfermedades Metabólicas/enzimología , Neoplasias/enzimología , Procesamiento Proteico-Postraduccional , Proteína-Arginina N-Metiltransferasas/metabolismo , Factores de Transcripción/metabolismo , Antineoplásicos/uso terapéutico , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/patología , Inhibidores Enzimáticos/uso terapéutico , Epigénesis Genética , Histonas/genética , Histonas/metabolismo , Humanos , Enfermedades Metabólicas/genética , Enfermedades Metabólicas/patología , Metilación , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/patología , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Proteína-Arginina N-Metiltransferasas/clasificación , Proteína-Arginina N-Metiltransferasas/genética , Empalme del ARN , Transducción de Señal , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/clasificación , Factores de Transcripción/genética
5.
Methods ; 175: 44-52, 2020 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-31794835

RESUMEN

The protein arginine methyltransferase family (PRMT) is known as being the catalytic driving force for arginine methylation. This specific type of post translational modification is extensively used in biological processes, and therefore is highly relevant in the pathology of a profusion of diseases. Since altered PRMT expression or deregulation has been shown to contribute to a vast range of those diseases including cancer, their study is of great interest. Although an increasing number of substrates are being discovered for each PRMT, large scale proteomic methods can be used to identify novel interactors/substrates, further elucidating the role that PRMTs perform in physiological or disease states. Here, we describe the use of affinity purification (AP) coupled with stable isotope labeling with amino acids in cell culture (SILAC) quantitative mass spectrometry (MS) to identify protein interactors and substrates of PRMTs. We also explore the possibility of exploiting the fact most PRMTs display lower dissociation rates with their hypomethylated substrates as a strategy to increase the proportion of substrates identified in AP/MS studies.


Asunto(s)
Cromatografía de Afinidad/métodos , Inhibidores Enzimáticos/química , Espectrometría de Masas/métodos , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Proteína-Arginina N-Metiltransferasas/química , Proteómica/métodos , Aminoácidos/metabolismo , Arginina/análisis , Arginina/química , Arginina/metabolismo , Expresión Génica , Histonas/química , Histonas/metabolismo , Humanos , Marcaje Isotópico , Células MCF-7 , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Unión Proteica/efectos de los fármacos , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Recombinantes
6.
Front Genet ; 10: 986, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31681423

RESUMEN

In the last 10 years, major advances have been made in the diagnosis and development of selective therapies for several blood cancers, including B-cell non-Hodgkin lymphoma (B-NHL), a heterogeneous group of malignancies arising from the mature B lymphocyte compartment. However, most of these entities remain incurable and current treatments are associated with variable efficacy, several adverse events, and frequent relapses. Thus, new diagnostic paradigms and novel therapeutic options are required to improve the prognosis of patients with B-NHL. With the recent deciphering of the mutational landscapes of B-cell disorders by high-throughput sequencing, it came out that different epigenetic deregulations might drive and/or promote B lymphomagenesis. Consistently, over the last decade, numerous epigenetic drugs (or epidrugs) have emerged in the clinical management of B-NHL patients. In this review, we will present an overview of the most relevant epidrugs tested and/or used so far for the treatment of different subtypes of B-NHL, from first-generation epigenetic therapies like histone acetyl transferases (HDACs) or DNA-methyl transferases (DNMTs) inhibitors to new agents showing selectivity for proteins that are mutated, translocated, and/or overexpressed in these diseases, including EZH2, BET, and PRMT. We will dissect the mechanisms of action of these epigenetic inhibitors, as well as the molecular processes underlying their lack of efficacy in refractory patients. This review will also provide a summary of the latest strategies being employed in preclinical and clinical settings, and will point out the most promising lines of investigation in the field.

7.
Artículo en Inglés | MEDLINE | ID: mdl-29685976

RESUMEN

DNA, RNA and histone methylation is implicated in various human diseases such as cancer or viral infections, playing a major role in cell process regulation, especially in modulation of gene expression. Here we developed a convergent synthetic pathway starting from a protected bromomethylcytosine derivative to synthesize transition state analogues of the DNA methyltransferases. This approach led to seven 5-methylcytosine-adenosine compounds that were, surprisingly, inactive against hDNMT1, hDNMT3Acat, TRDMT1 and other RNA human and viral methyltransferases. Interestingly, compound 4 and its derivative 2 showed an inhibitory activity against PRMT4 in the micromolar range. Crystal structures showed that compound 4 binds to the PRMT4 active site, displacing strongly the S-adenosyl-l-methionine cofactor, occupying its binding site, and interacting with the arginine substrate site through the cytosine moiety that probes the space filled by a substrate peptide methylation intermediate. Furthermore, the binding of the compounds induces important structural switches. These findings open new routes for the conception of new potent PRMT4 inhibitors based on the 5-methylcytosine-adenosine scaffold.This article is part of a discussion meeting issue 'Frontiers in epigenetic chemical biology'.


Asunto(s)
Dominio Catalítico , Metiltransferasas/síntesis química , Péptidos/metabolismo , Humanos
8.
Chemistry ; 22(39): 14022-14028, 2016 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-27515561

RESUMEN

Protein arginine N-methyl transferases (PRMTs) belong to a family of enzymes that modulate the epigenetic code through modifications of histones. In the present study, peptides emerging from a phage display screening were modified in the search for PRMT inhibitors through substitution with non-proteinogenic amino acids, N-alkylation of the peptide backbone, and incorporation of constrained dipeptide mimics. One of the modified peptides (23) showed an increased inhibitory activity towards several PRMTs in the low µm range and the conformational preference of this peptide was investigated and compared with the original hit using circular dichroism and NMR spectroscopy. Introducing two constrained tryptophan residue mimics (l-Aia) spaced by a single amino acid was found to induce a unique turn structure stabilized by a hydrogen bond and aromatic π-stacking interaction between the two l-Aia residues.


Asunto(s)
Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Peptidomiméticos/química , Peptidomiméticos/farmacología , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Alquilación , Secuencia de Aminoácidos , Técnicas de Visualización de Superficie Celular , Dipéptidos/síntesis química , Dipéptidos/química , Dipéptidos/farmacología , Inhibidores Enzimáticos/síntesis química , Humanos , Modelos Moleculares , Conformación Molecular , Peptidomiméticos/síntesis química , Proteína-Arginina N-Metiltransferasas/metabolismo
9.
J Neurosci ; 35(37): 12890-902, 2015 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-26377474

RESUMEN

Recent evidence suggests that histone modifications play a role in the behavioral effects of cocaine in rodent models. Histone arginine is known to be methylated by protein arginine N-methyltransferases (PRMTs). Evidence shows that PRMT1 contributes to >90% of cellular PRMT activity, which regulates histone H4 arginine 3 asymmetric dimethylation (H4R3me2a). Though histone arginine methylation represents a chemical modification that is relatively stable compared with other histone alterations, it is less well studied in the setting of addiction. Here, we demonstrate that repeated noncontingent cocaine injections increase PRMT1 activity in the nucleus accumbens (NAc) of C57BL/6 mice. We, subsequently, identify a selective inhibitor of PRMT1, SKLB-639, and show that systemic injections of the drug decrease cocaine-induced conditioned place preference to levels observed with genetic knockdown of PRMT1. NAc-specific downregulation of PRMT1 leads to hypomethylation of H4R3me2a, and hypoacetylation of histone H3 lysine 9 and 14. We also found that H4R3me2a is upregulated in NAc after repeated cocaine administration, and that H4R3me2a upregulation in turn controls the expression of Cdk5 and CaMKII. Additionally, the suppression of PRMT1 in NAc with lentiviral-short hairpin PMRT1 decreases levels of CaMKII and Cdk5 in the cocaine-treated group, demonstrating that PRMT1 affects the ability of cocaine to induce CaMKII and Cdk5 in NAc. Notably, increased H4R3me2a by repeated cocaine injections is relatively long-lived, as increased expression was observed for up to 7 d after the last cocaine injection. These results show the role of PRMT1 in the behavioral effects of cocaine. SIGNIFICANCE STATEMENT: This work demonstrated that repeated cocaine injections led to an increase of protein arginine N-methyltransferase (PRMT1) in nucleus accumbens (NAc). We then identified a selective inhibitor of PRMT1 (SKLB-639), which inhibited cocaine-induced conditioned place preference (CPP). Additionally, genetic downregulation of PRMT1 in NAc also attenuated cocaine-caused CPP and locomotion activity, which was associated with decreased expression of histone H4 arginine 3 asymmetric demethylation (H4R3me2a) and hypoacetylation of histone H3 lysine 9 and 14 (acH3K9/K14). This study also showed that H4R3me2a controlled transcriptions of Cdk5 and CaMKII, and that PRMT1 negatively affected the ability of cocaine to induce CaMKII and Cdk5 in NAc. Notably, increased H4R3me2a by repeated cocaine injection was relatively long-lived as increased expression was observed up to 7 d after withdrawal from cocaine. Together, this study suggests that PRMT1 inhibition may serve as a potential therapeutic strategy for cocaine addiction.


Asunto(s)
Amidinas/farmacología , Cocaína/farmacología , Histonas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Núcleo Accumbens/enzimología , Procesamiento Proteico-Postraduccional , Proteína-Arginina N-Metiltransferasas/fisiología , Pirimidinas/farmacología , Animales , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Metilación , Ratones , Modelos Moleculares , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/fisiología , Conformación Proteica , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Interferencia de ARN , ARN Interferente Pequeño/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA