Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Métodos Terapéuticos y Terapias MTCI
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Environ Toxicol ; 39(5): 3225-3237, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38357781

RESUMEN

Lung cancer (LC) is the most prevalent cancer type, with a high mortality rate worldwide. The current treatment options for LC have not been particularly successful in improving patient outcomes. Yifei Sanjie (YFSJ), a well-applicated traditional Chinese medicine formula, is widely used to treat pulmonary diseases, especially LC, yet little is known about its molecular mechanisms. This study was conducted to explore the molecular mechanism by which YFSJ ameliorated LC progression. The A549, NCI-H1975, and Calu-3 cells were treated with the YFSJ formula and observed for colony number, apoptosis, migration, and invasion properties recorded via corresponding assays. The PRMT6-YBX1-CDC25A axis was tested and verified through luciferase reporter, RNA immunoprecipitation, and chromatin immunoprecipitation assays and rescue experiments. Our results demonstrated that YFSJ ameliorated LC cell malignant behaviors by increasing apoptosis and suppressing proliferation, migration, and invasion processes. We also noticed that the xenograft mouse model treated with YFSJ significantly reduced tumor growth compared with the control untreated group in vivo. Mechanistically, it was found that YFSJ suppressed the expression of PRMT6, YBX1, and CDC25A, while the knockdown of these proteins significantly inhibited colony growth, migration, and invasion, and boosted apoptosis in LC cells. In summary, our results suggest that YFSJ alleviates LC progression via the PRMT6-YBX1-CDC25A axis, confirming its efficacy in clinical use. The findings of our study provide a new regulatory network for LC growth and metastasis, which could shed new insights into pulmonary medical research.


Asunto(s)
Neoplasias Pulmonares , MicroARNs , Humanos , Animales , Ratones , Neoplasias Pulmonares/patología , Proliferación Celular/genética , Movimiento Celular/genética , Pulmón/patología , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Proteína 1 de Unión a la Caja Y/genética , Proteína 1 de Unión a la Caja Y/metabolismo , Proteínas Nucleares/genética , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteína-Arginina N-Metiltransferasas/uso terapéutico , Fosfatasas cdc25/genética , Fosfatasas cdc25/metabolismo
2.
J Proteome Res ; 23(3): 1014-1027, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38272855

RESUMEN

Protein arginine methylations are important post-translational modifications (PTMs) in eukaryotes, regulating many biological processes. However, traditional collision-based mass spectrometry methods inevitably cause neutral losses of methylarginines, preventing the deep mining of biologically important sites. Herein we developed an optimized mass spectrometry workflow based on electron-transfer dissociation (ETD) with supplemental activation for proteomic profiling of arginine methylation in human cells. Using symmetric dimethylarginine (sDMA) as an example, we show that the ETD-based optimized workflow significantly improved the identification and site localization of sDMA. Quantitative proteomics identified 138 novel sDMA sites as potential PRMT5 substrates in HeLa cells. Further biochemical studies on SERBP1, a newly identified PRMT5 substrate, confirmed the coexistence of sDMA and asymmetric dimethylarginine in the central RGG/RG motif, and loss of either methylation caused increased the recruitment of SERBP1 to stress granules under oxidative stress. Overall, our optimized workflow not only enabled the identification and localization of extensive, nonoverlapping sDMA sites in human cells but also revealed novel PRMT5 substrates whose sDMA may play potentially important biological functions.


Asunto(s)
Arginina , Proteómica , Humanos , Células HeLa , Arginina/metabolismo , Procesamiento Proteico-Postraduccional , Metilación , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo
3.
Thorax ; 78(4): 383-393, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-35354645

RESUMEN

BACKGROUND: One hallmark of sepsis is the reduced number of lymphocytes, termed lymphopenia, that occurs from decreased lymphocyte proliferation or increased cell death contributing to immune suppression. Histone modification enzymes regulate immunity by their epigenetic and non-epigenetic functions; however, the role of these enzymes in lymphopenia remains elusive. METHODS: We used molecular biological approaches to investigate the high expression and function of a chromatin modulator protein arginine N-methyltransferase 4 (PRMT4)/coactivator-associated arginine methyltransferase 1 in human samples from septic patients and cellular and animal septic models. RESULTS: We identified that PRMT4 is elevated systemically in septic patients and experimental sepsis. Gram-negative bacteria and their derived endotoxin lipopolysaccharide (LPS) increased PRMT4 in B and T lymphocytes and THP-1 monocytes. Single-cell RNA sequencing results indicate an increase of PRMT4 gene expression in activated T lymphocytes. Augmented PRMT4 is crucial for inducing lymphocyte apoptosis but not monocyte THP-1 cells. Ectopic expression of PRMT4 protein caused substantial lymphocyte death via caspase 3-mediated cell death signalling, and knockout of PRMT4 abolished LPS-mediated lymphocyte death. PRMT4 inhibition with a small molecule compound attenuated lymphocyte death in complementary models of sepsis. CONCLUSIONS: These findings demonstrate a previously uncharacterised role of a key chromatin modulator in lymphocyte survival that may shed light on devising therapeutic modalities to lessen the severity of septic immunosuppression.


Asunto(s)
Linfopenia , Proteína-Arginina N-Metiltransferasas , Sepsis , Animales , Humanos , Arginina/genética , Caspasa 3/genética , Caspasa 3/inmunología , Cromatina , Lipopolisacáridos/farmacología , Linfopenia/etiología , Linfopenia/genética , Linfopenia/inmunología , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Sepsis/complicaciones , Sepsis/genética , Sepsis/inmunología
4.
Biochim Biophys Acta Mol Cell Res ; 1868(10): 119083, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34147561

RESUMEN

Airway wall remodeling, a main pathology of asthma was linked to vitamin-D deficiency and protein arginine methyltransferase-1 (PRMT1) expression in sub-epithelial cell layers. Calcitriol reduced remodeling in asthma model, but its mode of action is unclear. This study assessed the effect of calcitriol on PRMT1-dependent fibroblast remodeling in human lung fibroblasts, and allergen-induced asthma in E3-rats. Fibroblasts were activated with thymic stromal lymphopoietin (TLSP); asthma was induced by ovalbumin inhalation in rats. The airway structure was assessed by immunohistology. Protein expression in fibroblasts and activation of the mitogen activated protein kinases were detected by Western-blotting. Transcription factor activation was determined by luciferase reporter assay. PRMT1 action was blocked by siRNA and PRMT-inhibition. Ovalbumin upregulated the expression of TSLP, PRMT1, matrix metallopro-teinase-1 (MMP1), interleukin-25, and collagen type-I in sub-epithelial fibroblasts. In isolated fibroblasts, TSLP induced the same proteins, which were blocked by inhibition of Erk1/2 and p38. TLSP induced PRMT1 through activation of signal transducer and activator of transcription-3. PRMT1 inhibition reduced collagen type-I expression and suppressed MMP1. In fibroblasts, calcitriol supplementation over 12 days prevented TSLP-induced remodeling by blocking the PRMT1 levels. Interestingly, short-term calcitriol treatment had no such effect. The data support the beneficial role of calcitriol in asthma therapy.


Asunto(s)
Colágeno Tipo I/biosíntesis , Citocinas/metabolismo , Fibroblastos/metabolismo , Pulmón/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Represoras/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Calcitriol/farmacología , Línea Celular , Fibroblastos/efectos de los fármacos , Humanos , Pulmón/efectos de los fármacos , Ratas
5.
Cell Death Differ ; 27(9): 2697-2709, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32447347

RESUMEN

Senescence is accompanied with histones level alteration; however, the roles and the mechanisms of histone reduction in cellular senescence are largely unknown. Protein arginine methyltransferase 1 (PRMT1) is the major enzyme that generates monomethyl and asymmetrical dimethyl arginine. Here we showed that abrogation of PRMT1-mediated senescence was accompanied with decreasing histone H4 level. Consistently, under multiple classic senescence models, H4 decreasing was also been found prior to the other 3 core histones. Noticeably, asymmetric demethylation of histone H4 at arginine 3 (H4R3me2as), catalyzed by PRMT1, was decreased prior to histone H4. In addition, we showed that the PRMT1-mediated H4R3me2as maintained H4 stability. Reduction of H4R3me2as level increased the interaction between proteasome activator PA200 and histone H4, which catalyzes the poly-ubiquitin-independent degradation of H4. Moreover, H4 degradation promoted nucleosome decomposition, resulting in increased senescence-associated genes transcription. Significantly, H4 was restored by 3 well-informed anti-aging drugs (metformin, rapamycin, and resveratrol) much earlier than other senescence markers detected under H2O2-induced senescence. Thus, we uncovered a novel function of H4R3me2as in modulation of cellular senescence via regulating H4 stability. This finding also points to the value of histone H4 as a senescence indicator and a potential anti-aging drug screening marker.


Asunto(s)
Arginina/metabolismo , Biomarcadores/metabolismo , Senescencia Celular , Histonas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Apoptosis/genética , Ciclo Celular/genética , Línea Celular , Evaluación Preclínica de Medicamentos , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Metilación , Modelos Biológicos , Proteínas Nucleares/metabolismo , Estabilidad Proteica , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Represoras/metabolismo , Envejecimiento de la Piel
6.
Int J Mol Sci ; 21(8)2020 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-32316567

RESUMEN

As obesity promotes ectopic fat accumulation in skeletal muscle, resulting in impaired skeletal muscle and mitochondria function, it is associated with skeletal muscle loss and dysfunction. This study investigated whether Chrysanthemi zawadskii var. latilobum (CZH) protected mice against obesity-induced skeletal muscle atrophy and the underlying molecular mechanisms. High-fat diet (HFD)-induced obese mice were orally administered either distilled water, low-dose CZH (125 mg/kg), or high-dose CZH (250 mg/kg) for 8 w. CZH reduced obesity-induced increases in inflammatory cytokines levels and skeletal muscle atrophy, which is induced by expression of atrophic genes such as muscle RING-finger protein 1 and muscle atrophy F-box. CZH also improved muscle function according to treadmill running results and increased the muscle fiber size in skeletal muscle. Furthermore, CZH upregulated mRNA and protein levels of protein arginine methyltransferases (PRMT)1 and PRMT7, which subsequently attenuated mitochondrial dysfunction in the skeletal muscle of obese mice. We also observed that CZH significantly decreased PRMT6 mRNA and protein expression, which resulted in decreased muscle atrophy. These results suggest that CZH ameliorated obesity-induced skeletal muscle atrophy in mice via regulation of PRMTs in skeletal muscle.


Asunto(s)
Chrysanthemum/química , Dieta Alta en Grasa/efectos adversos , Músculo Esquelético/patología , Atrofia Muscular/tratamiento farmacológico , Obesidad/complicaciones , Extractos Vegetales/administración & dosificación , Proteína-Arginina N-Metiltransferasas/metabolismo , Administración Oral , Animales , Citocinas/metabolismo , Masculino , Ratones , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Atrofia Muscular/etiología , Atrofia Muscular/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Obesidad/inducido químicamente , Obesidad/metabolismo , Extractos Vegetales/química , Extractos Vegetales/farmacología , Proteína-Arginina N-Metiltransferasas/genética , Regulación hacia Arriba/efectos de los fármacos
7.
J Cell Sci ; 132(11)2019 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-31064814

RESUMEN

The use of proximity-dependent biotinylation assays coupled to mass spectrometry (PDB-MS) has changed the field of protein-protein interaction studies. However, despite the recurrent and successful use of BioID-based protein-protein interactions screening in mammalian cells, the implementation of PDB-MS in yeast has not been effective. Here, we report a simple and rapid approach in yeast to effectively screen for proximal and interacting proteins in their natural cellular environment by using TurboID, a recently described version of the BirA biotin ligase. Using the protein arginine methyltransferase Rmt3 and the RNA exosome subunits, Rrp6 and Dis3, the application of PDB-MS in yeast by using TurboID was able to recover protein-protein interactions previously identified using other biochemical approaches and provided new complementary information for a given protein bait. The development of a rapid and effective PDB assay that can systematically analyze protein-protein interactions in living yeast cells opens the way for large-scale proteomics studies in this powerful model organism.


Asunto(s)
Biotinilación/métodos , Mapeo de Interacción de Proteínas/métodos , Mapas de Interacción de Proteínas/fisiología , Saccharomyces cerevisiae/metabolismo , Schizosaccharomyces/metabolismo , Ligasas de Carbono-Nitrógeno/metabolismo , Complejo Multienzimático de Ribonucleasas del Exosoma/metabolismo , Espectrometría de Masas/métodos , Mapas de Interacción de Proteínas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteómica/métodos , Ribonucleasas/metabolismo , Saccharomyces cerevisiae/genética , Schizosaccharomyces/genética , Proteínas de Schizosaccharomyces pombe/metabolismo
8.
Osteoarthritis Cartilage ; 27(6): 932-944, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30858101

RESUMEN

OBJECTIVES: Selenium (Se) plays pivotal roles in maintaining optimal health. Nevertheless, how Se influences the metabolism of extracellular matrix (ECM) in cartilage remains unclear. The aim of the present study was to observe protein dimethylation by certain Se-sensitive PRMT and to elucidate its effects on the key transcriptional factor in cartilage. METHODS: We observed the expression of selenoproteins and markers of ECM metabolism in chondrocytes and articular cartilage of the rats under Se-deficiency by RT-qPCR, immunoblotting and immunohistochemistry. Then, we analyzed the expression of total dimethylated protein by using specific antibody under different Se statuses. After Se sensitive PRMT was identified, we used siRNA or PRMT inhibitor or stably overexpressing vector to intervene in the PRMT expression and identified the key transcriptional factor. Co-immunoprecipitation was applied to verify the interaction between PRMT and the key transcriptional factor. Finally, we measured the half-life time of the key transcriptional factor by immunoblotting after cycloheximide treatment. RESULTS: In chondrocytes and cartilage of the rats with Se deficiency, we found an aberrant metabolism manifesting decreased expression of Col2a1 and increased expression of Mmp-3. Then, we identified that PRMT5 was the unique type II PRMT, sensitive to Se status. PRMT5 upregulation led to the increased COL2A1 expression but decreased MMP-3 expression in chondrocytes. Furthermore, we revealed that PRMT5 improved SOX9 stability by dimethylating the protein, which contributed to maintain the matrix metabolic homeostasis of the chondrocytes. CONCLUSIONS: Se-sensitive PRMT5 increases the half-life of SOX9 protein via PTM and helps to maintain ECM homeostasis of the articular cartilage.


Asunto(s)
Cartílago Articular/metabolismo , Condrocitos/metabolismo , Matriz Extracelular/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Factor de Transcripción SOX9/metabolismo , Selenio/metabolismo , Animales , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Homeostasis , Metaloproteinasa 3 de la Matriz/genética , Metaloproteinasa 3 de la Matriz/metabolismo , Metilación , Procesamiento Proteico-Postraduccional , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Proteína-Arginina N-Metiltransferasas/genética , ARN Interferente Pequeño , Ratas , Selenio/deficiencia , Regulación hacia Arriba
9.
Sci Rep ; 9(1): 1638, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30733583

RESUMEN

Photodynamic therapy (PDT) is a cancer treatment that make use of the cancer-specific accumulation of porphyrins. We have reported that mitochondrial reactive oxygen species (mitROS) upregulate uptake transporter of porphyrins, heme carrier protein-1 (HCP-1). The accumulation of cancer-specific porphyrins was increased by mitROS production, thereby the cancer-specific PDT cytotoxicity was enhanced. Thus we investigated whether mitROS production by hyperthermia can enhanced the cytotoxicity of PDT or not. In this study, 1 h of hyperthermia at 42 °C increased the mitROS production, and both the accumulation of cancer-specific porphyrins and the PDT cytotoxicity increased. Moreover, the authors treated cells with N-acetyl-L-cysteine (NAC) to examine the effect of mitROS. NAC inhibited the increasing ROS production after hyperthermia to restrain the post-treatment increase of cancer-specific porphyrins accumulation. Moreover, the increase of ROS production in cancer cells after hyperthermia upregulated HCP-1 expression and downregulated ABCG2 expression. These regulation were inhibited by NAC. These results suggest that hyperthermia treatment increased mitROS production, which involved HpD accumulation and enhanced PDT effects in cancer cells. The mechanism of this phenomenon was most likely to be due to both the upregulation of HCP-1 and the downregulation of ABCG2 by mitROS.


Asunto(s)
Hipertermia Inducida/métodos , Fotoquimioterapia/métodos , Transportador de Folato Acoplado a Protón/metabolismo , Neoplasias Gástricas/terapia , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Acetilcisteína/farmacología , Animales , Línea Celular , Línea Celular Tumoral , Doxorrubicina/farmacología , Mitocondrias/metabolismo , Mitocondrias/patología , Proteína-Arginina N-Metiltransferasas/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología
10.
J Chem Inf Model ; 59(1): 522-534, 2019 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-30607947

RESUMEN

CARM1 (coactivator-associated arginine methyltransferase 1), also known as PRMT4 (protein arginine N-methyltransferase 4), belongs to the protein arginine methyltransferase (PRMT) family, which has emerged as a potential anticancer drug target. To discover new CARM1 inhibitors, we performed virtual screening against the substrate-binding site in CARM1. Structure-based pharmacophore models, which were generated according to three druggable subpockets embedding critical residues for ligand binding, were applied for virtual screening. The importance of the solvent-exposed substrate-binding cavity was highlighted due to significant hydrophobicity. Aided by molecular docking, 15 compounds structurally distinct from known CARM1 inhibitors were selected to evaluate their inhibitory effects on CARM1 methyltransferase activity, which resulted in seven compounds exhibiting micromolar inhibition, with selectivity over other members in the PRMT protein family. Moreover, three of them exhibited potent antiproliferation activities in breast cancer cells. Particularly, compound NO.2 exhibited potent activity both in vitro and in cultured cells, which will serve as a leading hit for developing CARM1 inhibitors with improved efficacy. The virtual screening strategy in this study will be applicable for the discovery of substrate-competitive inhibitors targeting other members in the PRMT protein family.


Asunto(s)
Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Simulación del Acoplamiento Molecular , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Sitios de Unión , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/toxicidad , Humanos , Ligandos , Células MCF-7 , Conformación Proteica , Proteína-Arginina N-Metiltransferasas/química , Proteína-Arginina N-Metiltransferasas/metabolismo , Interfaz Usuario-Computador
11.
Autophagy ; 15(6): 1069-1081, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30653406

RESUMEN

Protein arginine methyltransferases (PRMTs) have emerged as important regulators of skeletal muscle metabolism and regeneration. However, the direct roles of the various PRMTs during skeletal muscle remodeling remain unclear. Using skeletal muscle-specific prmt1 knockout mice, we examined the function and downstream targets of PRMT1 in muscle homeostasis. We found that muscle-specific PRMT1 deficiency led to muscle atrophy. PRMT1-deficient muscles exhibited enhanced expression of a macroautophagic/autophagic marker LC3-II, FOXO3 and muscle-specific ubiquitin ligases, TRIM63/MURF-1 and FBXO32, likely contributing to muscle atrophy. The mechanistic study reveals that PRMT1 regulates FOXO3 through PRMT6 modulation. In the absence of PRMT1, increased PRMT6 specifically methylates FOXO3 at arginine 188 and 249, leading to its activation. Finally, we demonstrate that PRMT1 deficiency triggers FOXO3 hyperactivation, which is abrogated by PRMT6 depletion. Taken together, PRMT1 is a key regulator for the PRMT6-FOXO3 axis in the control of autophagy and protein degradation underlying muscle maintenance. Abbreviations: Ad-RNAi: adenovirus-delivered small interfering RNA; AKT: thymoma viral proto-oncogene; AMPK: AMP-activated protein kinase; Baf A1: bafilomycin A1; CSA: cross-sectional area; EDL: extensor digitorum longus; FBXO32: F-box protein 32; FOXO: forkhead box O; GAS: gatrocnemieus; HDAC: histone deacetylase; IGF: insulin-like growth factor; LAMP: lysosomal-associated membrane protein; MAP1LC3B/LC3B: microtubule-associated protein 1 light chain 3 beta; mKO: Mice with skeletal muscle-specific deletion of Prmt1; MTOR: mechanistic target of rapamycin kinase; MYH: myosin heavy chain; MYL1/MLC1f: myosin, light polypeptide 1; PRMT: protein arginine N-methyltransferase; sgRNA: single guide RNA; SQSTM1: sequestosome 1; SOL: soleus; TA: tibialis anterior; TRIM63/MURF-1: tripartite motif-containing 63; YY1: YY1 transcription factor.


Asunto(s)
Autofagia/genética , Proteína Forkhead Box O3/metabolismo , Músculo Esquelético/metabolismo , Atrofia Muscular/metabolismo , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Animales , Proteína Forkhead Box O3/química , Proteína Forkhead Box O3/genética , Células HEK293 , Histona Desacetilasa 2/metabolismo , Histona Desacetilasas/metabolismo , Humanos , Metilación , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/patología , Fosforilación , Proto-Oncogenes Mas , Transducción de Señal/genética , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Factor de Transcripción YY1/metabolismo
12.
Bioorg Med Chem Lett ; 28(9): 1476-1483, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29628326

RESUMEN

As one of the most promising anticancer target in protein arginine methyltransferase (PRMT) family, PRMT5 has been drawing more and more attentions, and many efforts have been devoted to develop its inhibitors. In this study, three PRMT5 inhibitors (9, 16, and 23) with novel scaffolds were identified by performing pharmacophore- and docking-based virtual screening combined with in vitro radiometric-based scintillation proximity assay (SPA). Substructure search based on the scaffold of the most active 9 afforded 26 additional analogues, and SPA results indicated that two analogues (9-1 and 9-2) showed increased PRMT5 inhibitory activity compared with the parental compound. Resynthesis of 9, 9-1, and 9-2 confirmed their PRMT5 enzymatic inhibition activity. In addition, compound 9-1 displayed selectivity against PRMT5 over other key homological members (PRMT1 and CARM1 (PRMT4)). While the structure-activity relationship (SAR) of this series of compounds was discussed to provide clues for further structure optimization, the probable binding modes of active compounds were also probed by molecular docking and molecular dynamics simulations. Finally, the antiproliferative effect of 9-1 on MV4-11 leukemia cell line was confirmed and its impact on regulating the target gene of PRMT5 was also validated. The hit compounds identified in this work have provided more novel scaffolds for future hit-to-lead optimization of small-molecule PRMT5 inhibitors.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Modelos Moleculares , Estructura Molecular , Proteína-Arginina N-Metiltransferasas/metabolismo , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad
13.
J Chem Inf Model ; 58(5): 1066-1073, 2018 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-29672052

RESUMEN

Protein arginine methyltransferase 5 (PRMT5), a type II PRMT enzyme, is reported as an important therapeutic target in leukemia and lymphoma. In the present study, based on the combination of virtual screening and biochemical validations, we discovered a series of small-molecule inhibitors targeting PRMT5. Among those, DC_Y134 exhibited the most potent activity with IC50 value of 1.7 µM and displayed good selectivity against other methyltransferases. Further treatment with DC_Y134 inhibited the proliferation of several hematological malignancy cell lines by causing cell cycle arrest and apoptosis. Western blot assays indicated that DC_Y134 reduced the cellular symmetrically dimethylated levels. In addition, we analyzed the binding mode of DC_Y134 through molecular docking, which revealed that DC_Y134 occupies the binding site of substrate arginine and explained the selectivity of this inhibitor. Taken together, compound DC_Y134 could be used to elucidate the biological roles of PRMT5 and serve as a lead compound for treatment of hematologic malignancies.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Humanos , Simulación del Acoplamiento Molecular , Conformación Proteica , Proteína-Arginina N-Metiltransferasas/química , Proteína-Arginina N-Metiltransferasas/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Relación Estructura-Actividad , Interfaz Usuario-Computador
14.
J Exp Bot ; 69(3): 579-588, 2018 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-29253181

RESUMEN

Proline plays a crucial role in the drought stress response in plants. However, there are still gaps in our knowledge about the molecular mechanisms that regulate proline metabolism under drought stress. Here, we report that the histone methylase encoded by CAU1, which is genetically upstream of P5CS1 (encoding the proline biosynthetic enzyme Δ1-pyrroline-5-carboxylate synthetase 1), plays a crucial role in proline-mediated drought tolerance. We determined that the transcript level of CAU1 decreased while that of ANAC055 (encoding a transcription factor) increased in wild-type Arabidopsis under drought stress. Further analyses showed that CAU1 bound to the promoter of ANAC055 and suppressed its expression via H4R3sme2-type histone methylation in the promoter region. Thus, under drought stress, a decreased level of CAU1 led to an increased transcript level of ANAC055, which induced the expression of P5CS1 and increased proline level independently of CAS. Drought tolerance and the level of proline were found to be decreased in the cau1 anac055 double-mutant, while proline supplementation restored drought sensitivity in the anac055 mutant. Our results reveal the details of a novel pathway leading to drought tolerance mediated by CAU1.


Asunto(s)
Proteínas de Arabidopsis/genética , Arabidopsis/fisiología , Sequías , Prolina/metabolismo , Proteína-Arginina N-Metiltransferasas/genética , Factores de Transcripción/genética , Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Regulación de la Expresión Génica de las Plantas , Glutamato-5-Semialdehído Deshidrogenasa/genética , Glutamato-5-Semialdehído Deshidrogenasa/metabolismo , Metilación , Complejos Multienzimáticos/genética , Complejos Multienzimáticos/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Factores de Transcripción/metabolismo
15.
Org Biomol Chem ; 15(17): 3648-3661, 2017 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-28397890

RESUMEN

Protein arginine methyltransferase 5 (PRMT5) is a type II PRMT enzyme critical for diverse cellular processes and different types of cancers. Many efforts have been made to discover novel scaffold PRMT5 inhibitors. Herein, we report the discovery of DC_P33 as a hit compound of PRMT5 inhibitor, identified by molecular docking based virtual screening and 3H-labeled radioactive methylation assays. Structure-activity relationship (SAR) analysis was performed on the analogs of DC_P33 and then structural modifications were done to improve its activity. Among the derivatives, the compound DC_C01 displayed an IC50 value of 2.8 µM, and good selectivity toward PRMT1, EZH2 and DNMT3A. Moreover, DC_C01 exhibited anti-proliferation activities against Z-138, Maver-1, and Jeko-1 cancer cells with EC50 values of 12 µM, 12 µM, and 10.5 µM, respectively. Taken together, these results contribute to the development of specific inhibitors against PRMT5 and cancer therapy.


Asunto(s)
Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Simulación del Acoplamiento Molecular , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Proteína-Arginina N-Metiltransferasas/metabolismo , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/metabolismo , Humanos , Concentración 50 Inhibidora , Conformación Proteica , Proteína-Arginina N-Metiltransferasas/química , Relación Estructura-Actividad , Interfaz Usuario-Computador
16.
J Biol Chem ; 292(17): 6882-6894, 2017 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-28330868

RESUMEN

Arginine methylation is a common posttranslational modification that has been shown to regulate both gene expression and extranuclear signaling events. We recently reported defects in protein arginine methyltransferase 1 (PRMT1) activity and arginine methylation in the livers of cirrhosis patients with a history of recurrent infections. To examine the role of PRMT1 in innate immune responses in vivo, we created a cell type-specific knock-out mouse model. We showed that myeloid-specific PRMT1 knock-out mice demonstrate higher proinflammatory cytokine production and a lower survival rate after cecal ligation and puncture. We found that this defect is because of defective peroxisome proliferator-activated receptor γ (PPARγ)-dependent M2 macrophage differentiation. PPARγ is one of the key transcription factors regulating macrophage polarization toward a more anti-inflammatory and pro-resolving phenotype. We found that PRMT1 knock-out macrophages failed to up-regulate PPARγ expression in response to IL4 treatment resulting in 4-fold lower PPARγ expression in knock-out cells than in wild-type cells. Detailed study of the mechanism revealed that PRMT1 regulates PPARγ gene expression through histone H4R3me2a methylation at the PPARγ promoter. Supplementing with PPARγ agonists rosiglitazone and GW1929 was sufficient to restore M2 differentiation in vivo and in vitro and abrogated the difference in survival between wild-type and PRMT1 knock-out mice. Taken together these data suggest that PRMT1-dependent regulation of macrophage PPARγ expression contributes to the infection susceptibility in PRMT1 knock-out mice.


Asunto(s)
Inmunidad Innata , Macrófagos/metabolismo , PPAR gamma/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Represoras/metabolismo , Animales , Anticuerpos/química , Arginina/química , Diferenciación Celular , Citocinas/metabolismo , Femenino , Histonas/metabolismo , Humanos , Interleucina-4/farmacología , Lipopolisacáridos , Cirrosis Hepática/metabolismo , Macrófagos/citología , Macrófagos Peritoneales/citología , Masculino , Metilación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/citología , Procesamiento Proteico-Postraduccional , Sepsis/metabolismo
17.
Mol Cell ; 64(4): 673-687, 2016 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-27840030

RESUMEN

Distinctive from their normal counterparts, cancer cells exhibit unique metabolic dependencies on glutamine to fuel anabolic processes. Specifically, pancreatic ductal adenocarcinoma (PDAC) cells rely on an unconventional metabolic pathway catalyzed by aspartate aminotransferase, malate dehydrogenase 1 (MDH1), and malic enzyme 1 to rewire glutamine metabolism and support nicotinamide adenine dinucleotide phosphate (NADPH) production. Here, we report that methylation on arginine 248 (R248) negatively regulates MDH1. Protein arginine methyltransferase 4 (PRMT4/CARM1) methylates and inhibits MDH1 by disrupting its dimerization. Knockdown of MDH1 represses mitochondria respiration and inhibits glutamine metabolism, which sensitizes PDAC cells to oxidative stress and suppresses cell proliferation. Meanwhile, re-expression of wild-type MDH1, but not its methylation-mimetic mutant, protects cells from oxidative injury and restores cell growth and clonogenic activity. Importantly, MDH1 is hypomethylated at R248 in clinical PDAC samples. Our study reveals that arginine methylation of MDH1 by CARM1 regulates cellular redox homeostasis and suppresses glutamine metabolism of pancreatic cancer.


Asunto(s)
Carcinoma Ductal Pancreático/genética , Regulación Neoplásica de la Expresión Génica , Glutamina/metabolismo , Malato-Deshidrogenasa (NADP+)/genética , Neoplasias Pancreáticas/genética , Proteína-Arginina N-Metiltransferasas/genética , Arginina/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Proliferación Celular , Células HEK293 , Humanos , Malato-Deshidrogenasa (NADP+)/antagonistas & inhibidores , Malato-Deshidrogenasa (NADP+)/metabolismo , Metilación , Mitocondrias/genética , Mitocondrias/metabolismo , Mitocondrias/patología , Modelos Moleculares , NADP/biosíntesis , Oxidación-Reducción , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Multimerización de Proteína , Estructura Secundaria de Proteína , Proteína-Arginina N-Metiltransferasas/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal
18.
Biomed Res Int ; 2016: 7086390, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27872854

RESUMEN

Overexpression of coactivator associated arginine methyltransferase 1 (CARM1), a protein arginine N-methyltransferase (PRMT) family enzyme, is associated with various diseases including cancers. Consequently, the development of small-molecule inhibitors targeting PRMTs has significant value for both research and therapeutic purposes. In this study, together with structure-based virtual screening with biochemical assays, two compounds DC_C11 and DC_C66 were identified as novel inhibitors of CARM1. Cellular studies revealed that the two inhibitors are cell membrane permeable and effectively blocked proliferation of cancer cells including HELA, K562, and MCF7. We further predicted the binding mode of these inhibitors through molecular docking analysis, which indicated that the inhibitors competitively occupied the binding site of the substrate and destroyed the protein-protein interactions between CARM1 and its substrates. Overall, this study has shed light on the development of small-molecule CARM1 inhibitors with novel scaffolds.


Asunto(s)
Bioensayo/métodos , Evaluación Preclínica de Medicamentos/métodos , Inhibidores Enzimáticos/farmacología , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Inhibidores Enzimáticos/análisis , Inhibidores Enzimáticos/química , Humanos , Proteína-Arginina N-Metiltransferasas/metabolismo , Termodinámica
19.
Cell Death Dis ; 7: e2180, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-27054335

RESUMEN

Cancer is as much an epigenetic disease as it is a genetic disease, and epigenetic alterations in cancer often serve as potent surrogates for genetic mutations. Because the epigenetic factors involved in the DNA damage response are regulated by multiple elements, therapies to target specific components of the epigenetic machinery can be inefficient. In contrast, therapies aimed at inhibiting the methionine cycle can indirectly inhibit both DNA and protein methylation, and the wide variety of genes and pathways that are affected by these methylations make this global strategy very attractive. In the present study, we propose an adjuvant therapy that targets the epigenetics of the DNA damage response in breast cancer cells and that results in efficient apoptosis and a reduction in distant metastases in vivo. We observed that a combined therapy designed to uncouple adenosine metabolism using dipyridamole in the presence of a new synthetic antifolate, 3-O-(3,4,5-trimethoxybenzoyl)-(-)-catechin, simultaneously and efficiently blocked both the folic cycle and the methionine cycle in breast cancer cells and sensitized these cells to radiotherapy. The treatment impeded the recruitment of 53BP1 and BRCA1 to the chromatin regions flanking DNA double-strand breaks and thereby avoided the DNA damage responses in breast cancer cells that were exposed to ionizing radiation. In addition, this hypomethylating therapy was also efficient in reducing the self-renewal capability of breast cancer-initiating cells and induced reversion of mesenchymal phenotypes in breast cancer cells.


Asunto(s)
Reparación del ADN , Epigénesis Genética , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Proteína BRCA1/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Neoplasias de la Mama/radioterapia , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de la radiación , Línea Celular Tumoral , Cromatina/metabolismo , Roturas del ADN de Doble Cadena/efectos de los fármacos , Roturas del ADN de Doble Cadena/efectos de la radiación , Reparación del ADN/efectos de los fármacos , Reparación del ADN/efectos de la radiación , Dipiridamol/metabolismo , Femenino , Antagonistas del Ácido Fólico/farmacología , Histonas/metabolismo , Humanos , Células MCF-7 , Metilación/efectos de los fármacos , Metilación/efectos de la radiación , Ratones , Ratones Endogámicos BALB C , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo
20.
Gastric Cancer ; 19(3): 789-97, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26472729

RESUMEN

BACKGROUND: Metastatic and refractory gastric cancer (GC) are associated with a poor prognosis; therefore, the identification of prognostic factors and chemosensitivity markers is extremely important. Protein arginine methyltransferase 1 (PRMT1) may play a role in chemosensitivity/apoptosis induction via activation of the tumor suppressor forkhead box O1 (FOXO1). The purpose of this study was to clarify the expression of and relationship between PRMT1 and FOXO1 to evaluate the applicability of PRMT1 as a prognostic marker and a therapeutic tool in GC. METHODS: We investigated the clinical and functional significance of PRMT1 and FOXO1 in 195 clinical GC samples using immunohistochemistry. We performed suppression analysis of PRMT1 using small interfering RNA to determine the biological roles of PRMT1 in chemosensitivity. RESULTS: PRMT1 and FOXO1 in GC samples were predominantly expressed in the nucleus. Patients with lower PRMT1 expression (n = 131) had suppressed nuclear accumulation of FOXO1, higher recurrence after adjuvant chemotherapy, and poorer prognosis than those with higher PRMT1 expression (n = 64). PRMT1 downregulation in GC cells by RNA interference inhibited cisplatin and 5-fluorouracil sensitivity. The expression of phosphorylated FOXO1 and phosphorylated BCL-2 antagonist of cell death was upregulated in PRMT1 small interfering RNA groups. CONCLUSION: Our data suggest that the evaluation of PRMT1 expression in GC is a useful predictor of poor prognosis and recurrence after adjuvant chemotherapy. Moreover, these data suggest that PRMT1 is a promising therapeutic tool for overcoming refractory GC.


Asunto(s)
Adenocarcinoma/secundario , Biomarcadores de Tumor/metabolismo , Carcinoma de Células en Anillo de Sello/secundario , Fluorouracilo/uso terapéutico , Proteína Forkhead Box O1/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Represoras/metabolismo , Neoplasias Gástricas/patología , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Antimetabolitos Antineoplásicos/uso terapéutico , Apoptosis , Western Blotting , Carcinoma de Células en Anillo de Sello/tratamiento farmacológico , Carcinoma de Células en Anillo de Sello/metabolismo , Proliferación Celular , Resistencia a Antineoplásicos/genética , Femenino , Estudios de Seguimiento , Humanos , Técnicas para Inmunoenzimas , Hibridación Fluorescente in Situ , Metástasis Linfática , Masculino , Persona de Mediana Edad , Invasividad Neoplásica , Metástasis de la Neoplasia , Estadificación de Neoplasias , Pronóstico , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Proteína-Arginina N-Metiltransferasas/genética , ARN Interferente Pequeño/genética , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/metabolismo , Tasa de Supervivencia , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA