Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Pol J Pathol ; 75(2): 115-125, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39166520

RESUMEN

Precancerous cervical lesions are metaplastic alterations of epithelial cells of the cervix, eventually developing into cervical cancer. Despite primary and secondary prevention, the burden of cervical cancer remains high globally. Protein arginine methyltransferases (PRMT) represent post-translational modifications that interact with multiple signalling pathways, playing a role in epithelial-mesenchymal transition. In complex with desmoglein-2 (DSG2), a cell adhesion protein, both participate in the progression of dysplastic changes with potential malignant development. The presented study was performed on archival paraffin-embedded blocks from adult women. The studied samples were categorised into low-grade and high-grade intraepithelial lesions. Immunohistochemical analysis was used to observe subcellular localisation, immunoreaction intensity, and percentage of PRMT5- and DSG2-expressing cells, followed by statistical analysis. Preliminary results identified statistically significant differences between the expression and subcellular localisation of proteins in question in low-grade and high-grade squamous intraepithelial lesions. The primary goal of the presented study is to perceive the involvement of PRMT5 and DSG2 in the initiation and progression of cervical lesions. Our observations indicate the potential of the assessed proteins as prognostic markers. However, further studies of PRMT5 and DSG2 are required to provide greater insight into cervical carcinogenesis.


Asunto(s)
Biomarcadores de Tumor , Desmogleína 2 , Lesiones Precancerosas , Proteína-Arginina N-Metiltransferasas , Neoplasias del Cuello Uterino , Humanos , Femenino , Neoplasias del Cuello Uterino/patología , Neoplasias del Cuello Uterino/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteína-Arginina N-Metiltransferasas/análisis , Desmogleína 2/metabolismo , Desmogleína 2/análisis , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/metabolismo , Lesiones Precancerosas/patología , Lesiones Precancerosas/metabolismo , Adulto , Pronóstico , Persona de Mediana Edad , Inmunohistoquímica , Displasia del Cuello del Útero/patología , Displasia del Cuello del Útero/metabolismo
2.
Biochem Biophys Res Commun ; 530(2): 389-395, 2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32532423

RESUMEN

Recent studies have revealed that protein arginine methyltransferases (PRMTs) are responsible for diverse neurodegenerative diseases. However, their pathophysiological role in dopaminergic neuronal death in Parkinson's disease (PD) has not been evaluated. In this study, we demonstrated that 1-Methyl-4-phenylpyridinium iodide (MPP+), rotenone and paraquat, which cause dopaminergic neuronal cell death, increased PRMT1 expression in dopaminergic cell line. Dopaminergic neuronal cell death was increased by PRMT1 overexpression. MPP+-induced cell death was attenuated by PRMT1 knockdown. Poly (ADP-ribose) polymerase-1 (PARP1) expression and activity, poly-ADP-ribosylation (PARylation), were elevated by MPP+. Moreover, we found that PRMT1 positively regulates nuclear translocation of apoptosis-inducing factor (AIF). Elevated PRMT1 expression was observed in the substantia nigra pars compacta of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-injected mice. Furthermore, MPTP-induced dopaminergic neuronal death was reduced in PRMT1 haploinsufficient (prmt1+/-) mice. These data suggest that PRMT1 is implicated in PARP1/AIF-mediated dopaminergic neuronal cell death, which might be involved in the pathology of PD. Therefore, our results propose PRMT1 as a new target to develop a potential treatment of PD.


Asunto(s)
Neuronas Dopaminérgicas/patología , Enfermedad de Parkinson/patología , Proteína-Arginina N-Metiltransferasas/metabolismo , Animales , Muerte Celular , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/metabolismo , Humanos , Masculino , Ratones , Enfermedad de Parkinson/metabolismo , Proteína-Arginina N-Metiltransferasas/análisis
3.
Bioorg Med Chem ; 27(1): 224-229, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30529151

RESUMEN

Protein arginine methyltransferases (PRMTs) are a family of mammalian enzymes catalyzing the symmetric dimethylation (Type I), asymmetric dimethylation (Type II), or monomethylation (Type III) of arginine residues within proteins. This family is composed of 11 isozymes, however the vast majority of asymmetric and symmetric dimethylation in mammals is completed by either PRMT1 or PRMT5, respectively. In recent years, a number of chemical probes targeting this family of enzymes have been developed, but the majority of these probes lack isozyme specificity. Herein, we report the development of a chemical probe, based on a non-natural peptide sequence, which specifically labels PRMT1 over PRMT5 with high selectivity and sensitivity.


Asunto(s)
Isoenzimas/química , Sondas Moleculares/química , Péptidos/química , Proteína-Arginina N-Metiltransferasas/química , Secuencia de Aminoácidos , Pruebas de Enzimas , Isoenzimas/análisis , Cinética , Límite de Detección , Metilación , Sondas Moleculares/síntesis química , Péptidos/síntesis química , Proteína-Arginina N-Metiltransferasas/análisis , Especificidad por Sustrato
4.
Anal Biochem ; 547: 7-13, 2018 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-29410016

RESUMEN

The protein arginine N-methyltransferase 6 (PRMT6) is overexpressed in a variety of different cancer types and plays a role in human immunodeficiency virus (HIV) infections. Furthermore, the PRMT6 activity might also influence the pathogenesis of neurodegenerative, inflammatory, and cardiovascular diseases, whereby it becomes an interesting target for drug development. Previously reported activity assays for PRMT6 activity are either expensive, time-consuming or use radioactive substrates. To overcome these challenges, we developed a coupled fluorescence-based activity assay using recombinant PRMT6 expressed in E. coli. In the first step of the assay, the fluorogenic substrate Nα-Benzoyl-L-arginine-7-amido-4-methylcoumarin (Bz-Arg-AMC) is methylated by PRMT6, while in a second step the remaining un-methylated substrate is cleaved by trypsin, producing the fluorescent 7-amino-4-methylcoumarin.


Asunto(s)
Proteínas Nucleares/análisis , Péptidos/química , Proteína-Arginina N-Metiltransferasas/análisis , Fluorescencia , Humanos , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteína-Arginina N-Metiltransferasas/química , Proteína-Arginina N-Metiltransferasas/genética , Proteínas Recombinantes/análisis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética
5.
Pathol Int ; 68(6): 359-366, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29603824

RESUMEN

Protein arginine methyltransferases (PRMT) 5, a member of type II arginine methyltransferases, catalyzes the symmetrical dimethylation of arginine residues on histone and non-histone substrates. Although the overexpression of PRMT5 has been reported in various cancers, its role in oral squamous cell carcinoma (OSCC) has not been elucidated. In the present study, we immunohistochemically examined the expression of PRMT5 in surgically resected oral epithelial dysplasia (OED, n = 8), oral intraepithelial neoplasia (OIN)/carcinoma in situ (CIS) (n = 11) and OSCC (n = 52) with or without contiguous OED lesions. In the normal epithelium, PRMT5 was weakly expressed in the cytoplasm of basal layer cells. In OED, OIN/CIS, and OSCC, its expression consistently and uniformly increased in the cytoplasm of dysplastic and cancer cells. Moreover, nuclear and cytoplasmic localization was detected in the invasive front of cancer cells, particularly in cases showing poor differentiation or aggressive invasion patterns. The concomitant nuclear and cytoplasmic expression of PRMT5 correlated with the loss of E-cadherin and cytokeratin 17, and the upregulation of vimentin, features that are both indicative of epithelial-to-mesenchymal transition. PRMT5 may play a role from early oncogenesis through to the progression of OSCC, particularly in the aggressive mode of stromal invasion.


Asunto(s)
Biomarcadores de Tumor/análisis , Carcinoma de Células Escamosas/patología , Transición Epitelial-Mesenquimal/fisiología , Neoplasias de Cabeza y Cuello/patología , Neoplasias de la Boca/patología , Proteína-Arginina N-Metiltransferasas/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Células Escamosas/enzimología , Progresión de la Enfermedad , Femenino , Neoplasias de Cabeza y Cuello/enzimología , Humanos , Masculino , Persona de Mediana Edad , Neoplasias de la Boca/enzimología , Invasividad Neoplásica/patología , Proteína-Arginina N-Metiltransferasas/análisis , Carcinoma de Células Escamosas de Cabeza y Cuello
6.
Tumour Biol ; 39(4): 1010428317695917, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28381188

RESUMEN

Protein arginine methyltransferase 5 is one of the type II protein arginine methyltransferase family members that can symmetrically dimethylate arginine residues on target proteins in both the cytoplasm and the nucleus. Protein arginine methyltransferase 5 was reported to be an oncoprotein that participates in tumor progression through both epigenetic silencing and organelle biogenesis. So far, it has been implicated in various cancers, but its expression pattern in breast cancer has not been elucidated thoroughly. We analyzed the protein arginine methyltransferase 5 expression patterns in several breast cancer samples and tissue arrays to better characterize its contribution to breast cancer. Primary breast tumors showed increased protein arginine methyltransferase 5 expression compared with adjacent normal tissues in both the fresh tissue samples and tissue arrays. Also, there was a tendency that metastatic lymph nodes demonstrated enhanced protein arginine methyltransferase 5 expression compared to primary sites. Moreover, we found a significant correlation between protein arginine methyltransferase 5 and Ki-67, with higher Ki-67 and protein arginine methyltransferase 5 expressions in primary breast tumors compared with normal breast tissues. Moreover, the Cancer Genome Atlas cohort analysis revealed that high protein arginine methyltransferase 5 messenger RNA expression was associated with an unfavorable prognosis in human epidermal growth factor receptor 2 (HER-2) positive and triple negative breast cancer patients. Finally, the roles and mechanisms of protein arginine methyltransferase 5 in the proliferation, cell cycle progression, and apoptosis of MDA-MB-231 cells were assessed using protein arginine methyltransferase 5 and shPRMT5 transfection. In conclusion, we proposed that protein arginine methyltransferase 5 is an independent prognostic biomarker for breast cancer, and targeting protein arginine methyltransferase 5 might be a promising strategy for breast cancer treatment.


Asunto(s)
Neoplasias de la Mama/mortalidad , Proteína-Arginina N-Metiltransferasas/fisiología , Apoptosis , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Femenino , Humanos , Pronóstico , Proteína-Arginina N-Metiltransferasas/análisis
7.
Tumour Biol ; 35(8): 8225-33, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24850177

RESUMEN

Epigenetic processes play a critical role in melanoma development. However, little is known about proteins responsible for epigenetic transformations in melanoma cells. The processes in the peritumoral skin within the excision margin are almost unstudied. We studied the changes in expression of 112 proteins involved in epigenetic regulation of gene expression in the human cutaneous melanoma and its peritumoral zone using "The Proteomic Antibody Microarrays" (GRAA2, Sigma-Aldrich). Dimethylated histone H3 at lysines 4 and 9 as well as proteins involved in the regulation of transcription (histone deacetylases HDAC-1 and HDAC-11, DNA methyl-binding protein Kaiso), cell cycle control (protein kinases Aurora-В and PKR, chromosome protein CENP-E , and phosphorylated and acetylated histone H3), DNA repair (phosphorylated histone H2AX), and nuclear protein import (importins α3 and α5/7) were over-expressed in the melanoma tissue as compared with normal skin. At the same time, HDAC-10 and proliferating cell nuclear antigen PCNA were downregulated. In the peritumoral skin, at the excision margin (1-2 cm from the melanoma edge), we observed similar changes in expression of these proteins and, additionally, over-expression of arginine methyltransferases PRMT5 and NAD-dependent histone deacetylase SIR2. Histone methyltransferase G9a and metastasis-associated protein 2 were downregulated. Therefore, epigenetic regulation that requires histone modifications and expression of some regulatory proteins is of importance for melanoma development and propagation. The observed changes in the peritumoral skin may indicate the epigenetic pre-tuning in this zone possibly involved in malignant transformation. These results can be potentially useful for melanoma diagnostics and targeted therapy.


Asunto(s)
Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Melanoma/genética , Análisis por Matrices de Proteínas/métodos , Neoplasias Cutáneas/genética , Piel/metabolismo , Proliferación Celular , Histona Desacetilasas/análisis , Humanos , Carioferinas/análisis , Melanoma/metabolismo , Melanoma/patología , Antígeno Nuclear de Célula en Proliferación/análisis , Proteína-Arginina N-Metiltransferasas/análisis , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología
8.
Chin Med Sci J ; 27(1): 1-6, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22734206

RESUMEN

OBJECTIVE: To screen the asymmetric dimethyl arginines (ADMA)-containing proteins which could combine with protein arginine methyltransferase 1 (PRMT1). METHODS: Western blot was adopted to identify the expression of PRMT1 and the proteins with ADMA in glioma cell lines and normal brain tissues, and then to detect the changes of ADMA level after knock-down of PRMT1 with RNAi transfection in U87MG cells. Co-Immunoprecipitation (Co-IP), western blot, and sliver staining were employed to screen the candidate binding proteins of PRMT1. Then liquid chromatography-tandem mass spectrometry (LC-MS/MS) was used to identify the binding proteins of PRMT1. RESULTS: The expression of PRMT1 and some levels of ADMA were higher in glioma cell lines than in normal brain tissues. After knocking down PRMT1, some ADMA levels were found declined. After screening the binding proteins of PRMT1 with Co-IP and LC-MS/MS, 26 candidate binding proteins were identified. Among them, 6 candidate proteins had higher ions scores (> 38) and bioinformation analysis predicted that SEC23-IP, ANKHD1-EIF4EBP3 protein, and 1-phosphatidylinositol-3-phosphate 5-kinase isoform 2 had possible methylated aginine sites. CONCLUSIONS: The high expression of PRMT1 in glioma may induce the change of ADMA levels. Altogether 26 candidate proteins were identified, which contain ADMA and specifically bind with PRMT1.


Asunto(s)
Arginina/análogos & derivados , Glioma/química , Proteína-Arginina N-Metiltransferasas/análisis , Proteínas Represoras/análisis , Arginina/análisis , Línea Celular Tumoral , Cromatografía Liquida , Humanos , Inmunoprecipitación , Proteína-Arginina N-Metiltransferasas/fisiología , Proteínas Represoras/fisiología , Especificidad por Sustrato , Espectrometría de Masas en Tándem
9.
Tumour Biol ; 32(3): 575-82, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21229402

RESUMEN

Methylation of arginine residues has been implicated in many cellular activities like mRNA splicing, transcription regulation, signal transduction and protein-protein interactions. Protein arginine methyltransferases are the enzymes responsible for this modification in living cells. The most commonly used methyltransferase in man is protein arginine methyltransferase 1 (PRMT1). Since methylation processes appear to interfere in the emergence of several diseases, including cancer, we investigated the localisation of the protein in cancer tissue and, for the first time, the relation that possibly exists between the expression of PRMT1 gene and breast cancer progression. We used tumour specimens from 62 breast cancer patients and semi-quantitative RT-PCR to determine the expression of PRMT1 gene and was found to be associated with patient's age (p = 0.002), menopausal status (p = 0.006), tumour grade (p = 0.03), and progesterone receptor status (p = 0.001). Survival curves revealed that PRMT1-v1 status-low expression relates to longer disease-free survival (DFS; p = 0.036). To the contrary, PRMT1-v2 status is not associated neither with the clinical or pathological parameters nor with DFS (p = 0.31). PRMT1-v3 was not statistically significantly expressed in breast cancer tissue. Selected cancer and normal breast samples were stained for PRMT1. In both normal and cancerous breast tissues, staining was in the cytoplasm and only in rare cases the cell nucleus appeared stained. Present results show a potential use for this gene as a marker of unfavourable prognosis for breast cancer patients.


Asunto(s)
Neoplasias de la Mama/genética , Regulación Neoplásica de la Expresión Génica , Proteína-Arginina N-Metiltransferasas/genética , Proteínas Represoras/genética , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Femenino , Humanos , Persona de Mediana Edad , Proteína-Arginina N-Metiltransferasas/análisis , Proteína-Arginina N-Metiltransferasas/fisiología , Proteínas Represoras/análisis , Proteínas Represoras/fisiología
10.
J Vasc Res ; 44(5): 391-402, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17551258

RESUMEN

Previous investigations have demonstrated that angiotensin (Ang) II induces inflammatory reactions and asymmetric dimethylarginine (ADMA), an endogenous NOS inhibitor, might be a novel inflammatory factor. Endothelial cell activation was induced by incubation with Ang II or ADMA. Incubation with Ang II (10(-6) M) for 24 h elevated the levels of ADMA and decreased the levels of nitrite/nitrate concomitantly with a significant increase in the expression of protein arginine methyltransferase and a decrease in the activity of dimethylarginine dimethylaminohydrolase (DDAH). Exposure to Ang II (10(-6) M for 24 h) also enhanced intracellular ROS elaboration and the levels of tumor necrosis factor (TNF)-alpha and interleukin (IL)-8, upregulated chemokine receptor CXCR2 mRNA expression, increased adhesion of endothelial cells to monocytes and induced a significant increase in the activity of nuclear factor (NF)-kappaB, which was attenuated by pretreatment with the Ang II receptor blocker losartan (1, 3 and 10 muM). Exogenous ADMA (30 microM) also increased ROS generation and the levels of TNF-alpha and IL-8, decreased the levels of nitrite/nitrate, upregulated CXCR2 gene expression, increased endothelial cell binding with monocytes and activated the NF-kappaB pathway, which was inhibited by pretreatment with losartan or L-arginine. These data suggest that ADMA is a potential proinflammatory factor and may be involved in the inflammatory reaction induced by Ang II.


Asunto(s)
Angiotensina II/toxicidad , Arginina/análogos & derivados , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/citología , Vasculitis/inducido químicamente , Amidohidrolasas/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Arginina/análisis , Arginina/farmacología , Arginina/fisiología , Adhesión Celular/efectos de los fármacos , Células Cultivadas/efectos de los fármacos , Medios de Cultivo Condicionados/química , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Interleucina-8/biosíntesis , Losartán/farmacología , Monocitos/citología , FN-kappa B/metabolismo , Nitratos/análisis , Óxido Nítrico/metabolismo , Nitritos/análisis , Proteína-Arginina N-Metiltransferasas/análisis , ARN Mensajero/biosíntesis , Especies Reactivas de Oxígeno , Receptores de Interleucina-8B/biosíntesis , Receptores de Interleucina-8B/genética , Proteínas Represoras/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Necrosis Tumoral alfa/biosíntesis , Venas Umbilicales , Vasodilatación/efectos de los fármacos , Vasodilatación/fisiología
11.
Sci Rep ; 7(1): 1072, 2017 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-28432361

RESUMEN

Advancement in science has tended to improve treatment of fatal diseases such as cancer. A major concern in the area is the spread of cancerous cells, technically refered to as metastasis into other organs beyond the primary organ. Treatment in such a stage of cancer is extremely difficult and usually palliative only. In this study, we focus on finding gene-gene network modules which are functionally similar in nature in the case of breast cancer. These modules extracted during the disease progression stages are analyzed using p-value and their associated pathways. We also explore interesting patterns associated with the causal genes, viz., SCGB1D2, MET, CYP1B1 and MMP9 in terms of expression similarity and pathway contexts. We analyze the genes involved in both the stages- non metastasis and metastatsis and change in their expression values, their associated pathways and roles as the disease progresses from one stage to another. We discover three additional pathways viz., Glycerophospholipid metablism, h-Efp pathway and CARM1 and Regulation of Estrogen Receptor, which can be related to the metastasis phase of breast cancer. These new pathways can be further explored to identify their relevance during the progression of the disease.


Asunto(s)
Biomarcadores de Tumor/análisis , Neoplasias de la Mama/patología , Neoplasias de la Mama/secundario , Redes Reguladoras de Genes , Neoplasias de la Mama/diagnóstico , Progresión de la Enfermedad , Femenino , Glicerofosfolípidos/metabolismo , Humanos , Proteína-Arginina N-Metiltransferasas/análisis , Receptores de Estrógenos/análisis , Factores de Transcripción/análisis , Proteínas de Motivos Tripartitos/análisis , Ubiquitina-Proteína Ligasas/análisis
12.
Cancer Res ; 41(4): 1253-6, 1981 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-6783293

RESUMEN

The possible role of alterations of histone methylation by ethionine in the mechanism of ethionine carcinogenesis was studied. In regenerating rat liver, histone synthesis was inhibited by only 20 to 30% with large doses of ethionine (0.75 to 1.0 mg/g body weight). The effect of ethionine on the in vivo methylation of histones was studied by giving 0.5 mg ethionine and [methyl-3H]methionine per g body weight. In vivo methylation of lysine was inhibited by 50%, whereas the arginine methylation was inhibited by 89%. The cellular localization of the methyltransferases and S-adenosyl-L-ethionine may be related to this differential effect. Utilizing an in vitro assay for protein-lysine and protein-arginine methyltransferases, we have demonstrated that the methyl-deficient histones are transported to the nucleus and with time lose their ability to accept methyl groups in vitro.


Asunto(s)
Etionina/farmacología , Histonas/metabolismo , Hígado/metabolismo , Animales , Arginina/metabolismo , Núcleo Celular/análisis , N-Metiltransferasa de Histona-Lisina/análisis , Lisina/metabolismo , Masculino , Metionina/farmacología , Metilación , Proteína-Arginina N-Metiltransferasas/análisis , Ratas
13.
Mol Oncol ; 10(10): 1497-1515, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27717710

RESUMEN

Histone methyltransferases (HMTs) catalyze the methylation of lysine and arginine residues on histone tails and non-histone targets. These important post-translational modifications are exquisitely regulated and affect chromatin compaction and transcriptional programs leading to diverse biological outcomes. There is accumulating evidence that genetic alterations of several HMTs impinge on oncogenic or tumor-suppressor functions and influence both cancer initiation and progression. HMTs therefore represent an opportunity for therapeutic targeting in those patients with tumors in which HMTs are dysregulated, to reverse the histone marks and transcriptional programs associated with aggressive tumor behavior. In this review, we describe the known histone methyltransferases and their emerging roles in breast cancer tumorigenesis.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Mama/patología , Regulación Neoplásica de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/genética , Proteína-Arginina N-Metiltransferasas/genética , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Mama/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Epigénesis Genética/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Código de Histonas/efectos de los fármacos , N-Metiltransferasa de Histona-Lisina/análisis , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Terapia Molecular Dirigida , Proteína-Arginina N-Metiltransferasas/análisis , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Proteína-Arginina N-Metiltransferasas/metabolismo , Activación Transcripcional/efectos de los fármacos
14.
Mol Cell Biol ; 35(12): 2203-14, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25870105

RESUMEN

Alternative splicing contributes greatly to the diversification of mammalian proteomes, but the molecular basis for the evolutionary emergence of splice variants remains poorly understood. We have recently found a novel class of splicing regulatory elements between the polypyrimidine tract (Py) and 3' AG (REPA) at intron ends in many human genes, including the multifunctional PRMT5 (for protein arginine methyltransferase 5) gene. The PRMT5 element is comprised of two G tracts that arise in most mammals and accompany significant exon skipping in human transcripts. The G tracts inhibit splicing by recruiting heterogeneous nuclear ribonucleoprotein (hnRNP) H and F (H/F) to reduce U2AF65 binding to the Py, causing exon skipping. The resulting novel shorter variant PRMT5S exhibits a histone H4R3 methylation effect similar to that seen with the original longer PRMT5L isoform but exhibits a distinct localization and preferential control of critical genes for cell cycle arrest at interphase in comparison to PRMT5L. This report thus provides a molecular mechanism for the evolutionary emergence of a novel splice variant with an opposite function in a fundamental cell process. The presence of REPA elements in a large group of genes implies their wider impact on different cellular processes for increased protein diversity in humans.


Asunto(s)
Empalme Alternativo , Ciclo Celular , Evolución Molecular , Proteína-Arginina N-Metiltransferasas/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Puntos de Control del Ciclo Celular , Exones , Células HeLa , Humanos , Datos de Secuencia Molecular , Isoformas de Proteínas/análisis , Isoformas de Proteínas/genética , Proteína-Arginina N-Metiltransferasas/análisis
15.
Assay Drug Dev Technol ; 13(4): 235-40, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-26065559

RESUMEN

Conformational remodeling of chromatin in cells is known to alter gene expression. The histone code hypothesis postulates that multiple modifications present on histone tails can regulate gene expression both through direct effects on chromatin compaction as well as through recruitment of unique complexes that signal specific downstream functions. Histone methylation is an important component of the histone code, and the dysregulation of histone methylation in disease makes methyltransferases and demethylases viable targets for drug discovery. We developed a biochemical assay platform, which takes advantage of the fact that protein methyltransferases (PMTs) all utilize the cofactor S-Adenosyl-L-methionine (SAM) as the methyl donor. The platform utilizes the High-throughput Mass Spectrometry (MS) technology to measure SAM and the S-Adenosyl-L-homocysteine product in a label-free manner. The platform has all the advantages of a label-free system coupled with the benefit of substrate agnostic measurements making it an ideal setup for PMT biochemical studies and drug discovery. In addition, MS is ideally suited for detecting multiple modification events within the same substrate. The ability to adjust the detection to monitor the methyl acceptor product allows for real-time measurements of multiple product species simultaneously, a distinct advantage over other commonly used assay formats.


Asunto(s)
Pruebas de Enzimas/métodos , Ensayos Analíticos de Alto Rendimiento , Espectrometría de Masas , Proteína-Arginina N-Metiltransferasas/análisis , Radiometría/métodos , Sitios de Unión , Humanos , Proteína-Arginina N-Metiltransferasas/metabolismo
16.
Life Sci ; 65(8): 737-45, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10466739

RESUMEN

Enzymatic methylation of endogenous proteins in several cancer cell lines was investigated to understand a possible relationship between protein-arginine methylation and cellular proliferation. Cytosolic extracts prepared from several cancer cells (HeLa, HCT-48, A549, and HepG2) and incubated with S-adenosyl-L-[methyl-3H]methionine revealed an intensely [methyl-3H]-labeled 20-kDa polypeptide. On the other hand, cytosolic extracts prepared from normal colon cells did not show any methylation of the 20-kDa protein under identical conditions. To identify nature of the 20-kDa polypeptide, purified histones were methylated with HCT-48 cytosolic extracts and analyzed by SDS-PAGE. However, none of the histones comigrated with the methylated 20-kDa polypeptide, indicating that it is unlikely to be any of the histone subclasses. The [methyl-3H]group in the 20-kDa polypeptide was stable at pH 10-11 (37 degrees C for 30 min) and methylation was not stimulated by GTPgammaS (4 mM), thus the reaction is neither carboxyl methylesterification on isoaspartyl residues, nor on C-terminal farnesylated cysteine. The present study together with the previous identification of N(G)-methylated arginine residues in the HCT-48 cytosol fraction suggests that this novel endogenous 20-kDa arginine-methylation is a cellular proliferation-related posttranslational modification reaction.


Asunto(s)
Proteínas de Neoplasias/química , Proteína-Arginina N-Metiltransferasas/análisis , Células Tumorales Cultivadas/química , Animales , Extractos Celulares/química , Transformación Celular Neoplásica/química , Células HeLa/química , Humanos , Metilación , Tritio , Células Tumorales Cultivadas/citología
17.
Biomed Res Int ; 2014: 627434, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25243167

RESUMEN

We investigated the effects of hepatic ischemia/reperfusion (I/R) injury on asymmetric dimethylarginine (ADMA, a nitric oxide synthase inhibitor), protein methyltransferase (PRMT) and dimethylarginine dimethylaminohydrolase (DDAH) (involved, resp., in ADMA synthesis and degradation), and the cationic transporter (CAT). Male Wistar rats were subjected to 30 or 60 min hepatic ischemia followed by 60 min reperfusion. ADMA levels in serum and bile were determined. Tissue ADMA, DDAH activity, DDAH-1 and CAT-2 protein, DDAH-1 and PRMT-1 mRNA expression, GSH/GSSG, ROS production, and lipid peroxidation were detected. ADMA was found in bile. I/R increased serum and bile ADMA levels while an intracellular decrease was detected after 60 min ischemia. Decreased DDAH activity, mRNA, and protein expression were observed at the end of reperfusion. No significant difference was observed in GSH/GSSG, ROS, lipid peroxidation, and CAT-2; a decrease in PRMT-1 mRNA expression was found after I/R. Liver is responsible for the biliary excretion of ADMA, as documented here for the first time, and I/R injury is associated with an oxidative stress-independent alteration in DDAH activity. These data are a step forward in the understanding of the pathways that regulate serum, tissue, and biliary levels of ADMA in which DDAH enzyme plays a crucial role.


Asunto(s)
Amidohidrolasas/metabolismo , Arginina/análogos & derivados , Bilis/fisiología , Hígado/lesiones , Daño por Reperfusión/metabolismo , Amidohidrolasas/análisis , Amidohidrolasas/genética , Animales , Arginina/análisis , Arginina/metabolismo , Bilis/química , Transportador de Aminoácidos Catiônicos 2/análisis , Transportador de Aminoácidos Catiônicos 2/metabolismo , Modelos Animales de Enfermedad , Hígado/química , Hígado/metabolismo , Masculino , Especificidad de Órganos , Estrés Oxidativo , Proteína-Arginina N-Metiltransferasas/análisis , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Ratas , Ratas Wistar
18.
Diagn Pathol ; 8: 129, 2013 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-23915145

RESUMEN

BACKGROUND: Coactivator-associated arginine methyltransferase 1 (CARM1) belongs to the protein arginine methyltransferase family. CARM1 has been reported to be associated with high grade tumors in breast cancer. It still remains unknown the expression pattern of CARM1 in breast cancer and its relationships with clinicopathological characteristics and molecular subtypes. METHODS: Two hundred forty-seven invasive breast cancer cases were collected and prepared for tissue array. There were thirty-seven tumors with benign glandular epithelium adjacent to the tumors among these cases. Molecular subtype and CARM1 expression were investigated using immunohistochemistry. RESULTS: Cell staining was observed in the cytoplasm and/or nucleus. Staining for CARM1 was significantly stronger in adenocarcinoma compared with adjacent benign epithelium. There is a significant correlation between CARM1 overexpression with young age, high grade, estrogen receptor (ER) and progesterone receptor (PR) negative, increased p53 expression, and high Ki-67 index. Our study demonstrated CARM1 overexpression was associated with an increase in the protein expression of HER2. Furthermore, our data indicated CARM1-overexpression rate were remarkably higher in HER2 subtype (69.6%), luminal B subtype (59.6%) and TN subtype (57.1%) compared with luminal A subtype (41.3%). CONCLUSIONS: CARM1 expression was increased in invasive breast cancer. CARM1 overexpression was associated with poorly characterized clinicopathologic parameters and HER2 overexpression. There were significant differences between different molecular subtypes in their relationship to CARM1 overexpression. Our results support the value of using CARM1 in prognostic stratification of breast cancer patients and its potential therapeutic implications in targeting treatment. VIRTUAL SLIDES: The virtual slide(s) for this article can be found here: http://www.diagnosticpathology.diagnomx.eu/vs/4116338491022965.


Asunto(s)
Adenocarcinoma/enzimología , Biomarcadores de Tumor/análisis , Neoplasias de la Mama/enzimología , Proteína-Arginina N-Metiltransferasas/análisis , Adenocarcinoma/clasificación , Adenocarcinoma/patología , Adulto , Factores de Edad , Neoplasias de la Mama/clasificación , Neoplasias de la Mama/patología , Femenino , Humanos , Inmunohistoquímica , Antígeno Ki-67/análisis , Clasificación del Tumor , Invasividad Neoplásica , Estadificación de Neoplasias , Receptor ErbB-2/análisis , Receptores de Estrógenos/análisis , Receptores de Progesterona/análisis , Factores de Riesgo , Análisis de Matrices Tisulares , Proteína p53 Supresora de Tumor/análisis , Regulación hacia Arriba
19.
J Biomol Screen ; 17(2): 237-44, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21821785

RESUMEN

Methylation of arginine residues, catalyzed by protein arginine methyltransferases (PRMTs), is one important protein posttranslational modification involved in epigenetic regulation of gene expression. A fast and effective assay for PRMT can provide valuable information for dissecting the biological functions of PRMTs, as well as for screening small-molecule inhibitors of arginine methylation. Currently, among the methods used for PRMT activity measurement, many contain laborious separation procedures, which restrict the applications of these assays for high-throughput screening (HTS) in drug discovery. The authors report here a mix-and-measure method to measure PRMT activity based on the principle of scintillation proximity assay (SPA). In this assay, (3)H-AdoMet was used as methyl donor, and biotin-modified histone H4 peptide served as a methylation substrate. Following the methylation reaction catalyzed by PRMTs, streptavidin-coated SPA beads were added to the reaction solution, and SPA signals were detected by a MicroBeta scintillation counter. No separation step is needed, which simplifies the assay procedure and greatly enhances the assay speed. Particularly, the miniaturization and robustness suggest that this method is suited for HTS of PRMT inhibitors.


Asunto(s)
Arginina/metabolismo , Ensayos Analíticos de Alto Rendimiento/métodos , Proteína-Arginina N-Metiltransferasas/análisis , Conteo por Cintilación/métodos , Metilación , Microesferas , Estreptavidina/química
20.
Electrophoresis ; 29(11): 2381-90, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18449859

RESUMEN

Embryonic stem cells (ESCs) and embryonic germ cells (EGCs) provide exciting models for understanding the underlying mechanisms that make a cell pluripotent. Indeed, such understanding would enable dedifferentiation and reprogrammation of any cell type from a patient needing a cell therapy treatment. Proteome analysis has emerged as an important technology for deciphering these biological processes and thereby ESC and EGC proteomes are increasingly studied. Nevertheless, their nuclear proteomes have only been poorly investigated up to now. In order to investigate signaling pathways potentially involved in pluripotency, proteomic analyses have been performed on mouse ESC and EGC nuclear proteins. Nuclei from ESCs and EGCs at undifferentiated stage were purified by subcellular fractionation. After 2-D separation, a subtractive strategy (subtracting culture environment contaminating spots) was applied and a comparison of ESC, (8.5 day post coïtum (dpc))-EGC and (11.5 dpc)-EGC specific nuclear proteomes was performed. A total of 33 ESC, 53 (8.5 dpc)-EGC, and 36 (11.5 dpc)-EGC spots were identified by MALDI-TOF-MS and/or nano-LC-MS/MS. This approach led to the identification of two isoforms (with and without N-terminal acetylation) of a known pluripotency marker, namely developmental pluripotency associated 5 (DPPA5), which has never been identified before in 2-D gel-MS studies of ESCs and EGCs. Furthermore, we demonstrated the efficiency of our subtracting strategy, in association with a nuclear subfractionation by the identification of a new protein (protein arginine N-methyltransferase 7; PRMT7) behaving as proteins involved in pluripotency.


Asunto(s)
Núcleo Celular/química , Células Madre Embrionarias/química , Células Germinativas/química , Proteoma/análisis , Proteómica/métodos , Animales , Biomarcadores/análisis , Ratones , Proteína-Arginina N-Metiltransferasas/análisis
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda