Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Zhongguo Zhong Yao Za Zhi ; 47(21): 5900-5907, 2022 Nov.
Artículo en Chino | MEDLINE | ID: mdl-36472009

RESUMEN

This study was designed to determine the inhibitory effect of astragaloside Ⅳ(AS-Ⅳ), a principal bioactive component extracted from the Chinese medicinal Astragali Radix, on the inflammatory response of vascular endothelial cells induced by angiotensin Ⅱ(Ang Ⅱ), the most major pathogenic factor for cardiovascular diseases, and to clarify the role of calcium(Ca~(2+))/phosphatidylinosi-tol-3-kinase(PI3K)/protein kinase B(Akt)/endothelial nitric oxide synthase(eNOS)/nitric oxide(NO) pathway in the process. To be specific, human umbilical vein endothelial cells(HUVECs) were cultured in the presence of AS-Ⅳ with or without the specific inhibitor of NO synthase(NG-monomethyl-L-arginine, L-NMMA), inhibitor of PI3K/Akt signaling pathway(LY294002), or Ca~(2+)-chelating agent(ethylene glycol tetraacetic acid, EGTA) prior to Ang Ⅱ stimulation. The inhibitory effect of AS-Ⅳ on Ang Ⅱ-induced inflammatory response and the involved mechanism was determined with enzyme-linked immunosorbent assay(ELISA), cell-based ELISA assay, Western blot, and monocyte adhesion assay which determined the fluorescently labeled human monocytic cell line(THP-1) adhered to Ang Ⅱ-stimulated endothelial cells. AS-Ⅳ increased the production of NO by HUVECs in a dose-and time-dependent manner(P<0.05) and raised the level of phosphorylated eNOS(P<0.05). The above AS-Ⅳ-induced changes were abolished by pretreatment with L-NMMA, LY294002, or EGTA. Compared with the control group, Ang Ⅱ obviously enhanced the production and release of cytokines(tumor necrosis factor-α, interleukin-6), chemokines(monocyte chemoattractant protein-1) and adhesion molecules(intercellular adhesion molecule-1, vascular cellular adhesion molecule-1), and the number of monocytes adhered to HUVECs(P<0.05), which were accompanied by the enhanced levels of phosphorylated inhibitor of nuclear factor-κBα protein and activities of nuclear factor-κB(NF-κB)(P<0.05). This study also demonstrated that Ang Ⅱ-induced inflammatory response was inhibited by pretreatment with AS-Ⅳ(P<0.05). In addition, the inhibitory effect of AS-Ⅳ was abrogated by pretreatment with L-NMMA, LY294002, or EGTA(P<0.05). This study provides a direct link between AS-Ⅳ and Ca~(2+)/PI3K/Akt/eNOS/NO pathway in AS-Ⅳ-mediated anti-inflammatory actions in endothelial cells exposed to Ang Ⅱ. The results indicate that AS-Ⅳ attenuates endothelial cell-mediated inflammatory response induced by Ang Ⅱ via the activation of Ca~(2+)/PI3K/Akt/eNOS/NO signaling pathway.


Asunto(s)
Angiotensina II , Proteínas Proto-Oncogénicas c-akt , Humanos , Angiotensina II/metabolismo , Angiotensina II/farmacología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , omega-N-Metilarginina/metabolismo , omega-N-Metilarginina/farmacología , Ácido Egtácico/metabolismo , Ácido Egtácico/farmacología , Células Endoteliales de la Vena Umbilical Humana , FN-kappa B/genética , FN-kappa B/metabolismo , Óxido Nítrico/metabolismo , Células Cultivadas
2.
World J Gastroenterol ; 28(29): 3854-3868, 2022 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-36157548

RESUMEN

BACKGROUND: The mechanisms underlying gastrointestinal (GI) dysmotility with ulcerative colitis (UC) have not been fully elucidated. The enteric nervous system (ENS) plays an essential role in the GI motility. As a vital neurotransmitter in the ENS, the gas neurotransmitter nitric oxide (NO) may impact the colonic motility. In this study, dextran sulfate sodium (DSS)-induced UC rat model was used for investigating the effects of NO by examining the effects of rate-limiting enzyme nitric oxide synthase (NOS) changes on the colonic motility as well as the role of the ENS in the colonic motility during UC. AIM: To reveal the relationship between the effects of NOS expression changes in NOS-containing nitrergic neurons and the colonic motility in a rat UC model. METHODS: Male rats (n = 8/each group) were randomly divided into a control (CG), a UC group (EG1), a UC + thrombin derived polypeptide 508 trifluoroacetic acid (TP508TFA; an NOS agonist) group (EG2), and a UC + NG-monomethyl-L-arginine monoacetate (L-NMMA; an NOS inhibitor) group (EG3). UC was induced by administering 5.5% DSS in drinking water without any other treatment (EG1), while the EG2 and EG3 were gavaged with TP508 TFA and L-NMMA, respectively. The disease activity index (DAI) and histological assessment were recorded for each group, whereas the changes in the proportion of colonic nitrergic neurons were counted using immunofluorescence histochemical staining, Western blot, and enzyme linked immunosorbent assay, respectively. In addition, the contractile tension changes in the circular and longitudinal muscles of the rat colon were investigated in vitro using an organ bath system. RESULTS: The proportion of NOS-positive neurons within the colonic myenteric plexus (MP), the relative expression of NOS, and the NOS concentration in serum and colonic tissues were significantly elevated in EG1, EG2, and EG3 compared with CG rats. In UC rats, stimulation with agonists and inhibitors led to variable degrees of increase or decrease for each indicator in the EG2 and EG3. When the rats in EGs developed UC, the mean contraction tension of the colonic smooth muscle detected in vitro was higher in the EG1, EG2, and EG3 than in the CG group. Compared with the EG1, the contraction amplitude and mean contraction tension of the circular and longitudinal muscles of the colon in the EG2 and EG3 were enhanced and attenuated, respectively. Thus, during UC, regulation of the expression of NOS within the MP improved the intestinal motility, thereby favoring the recovery of intestinal functions. CONCLUSION: In UC rats, an increased number of nitrergic neurons in the colonic MP leads to the attenuation of colonic motor function. To intervene NOS activity might modulate the function of nitrergic neurons in the colonic MP and prevent colonic motor dysfunction. These results might provide clues for a novel approach to alleviate diarrhea symptoms of UC patients.


Asunto(s)
Colitis Ulcerosa , Agua Potable , Neuronas Nitrérgicas , Animales , Masculino , Ratas , Colitis Ulcerosa/patología , Colon/patología , Sulfato de Dextran/toxicidad , Motilidad Gastrointestinal , Neuronas Nitrérgicas/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/metabolismo , omega-N-Metilarginina/metabolismo , omega-N-Metilarginina/farmacología , Trombina/metabolismo , Ácido Trifluoroacético/metabolismo , Ácido Trifluoroacético/farmacología
3.
PLoS One ; 17(9): e0274487, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36149900

RESUMEN

The endothelium maintains and regulates vascular homeostasis mainly by balancing interplay between vasorelaxation and vasoconstriction via regulating Nitric Oxide (NO) availability. Endothelial nitric oxide synthase (eNOS) is one of three NOS isoforms that catalyses the synthesis of NO to regulate endothelial function. However, eNOS's role in the regulation of endothelial function, such as cell proliferation and migration remain unclear. To gain a better understanding, we genetically knocked down eNOS in cultured endothelial cells using sieNOS and evaluated cell proliferation, migration and also tube forming potential in vitro. To our surprise, loss of eNOS significantly induced endothelial cell proliferation, which was associated with significant downregulation of both cell cycle inhibitor p21 and cell proliferation antigen Ki-67. Knockdown of eNOS induced cell migration but inhibited formation of tube-like structures in vitro. Mechanistically, loss of eNOS was associated with activation of MAPK/ERK and inhibition of PI3-K/AKT signaling pathway. On the contrary, pharmacologic inhibition of eNOS by inhibitors L-NAME or L-NMMA, inhibited cell proliferation. Genetic and pharmacologic inhibition of eNOS, both promoted endothelial cell migration but inhibited tube-forming potential. Our findings confirm that eNOS regulate endothelial function by inversely controlling endothelial cell proliferation and migration, and by directly regulating its tube-forming potential. Differential results obtained following pharmacologic versus genetic inhibition of eNOS indicates a more complex mechanism behind eNOS regulation and activity in endothelial cells, warranting further investigation.


Asunto(s)
Células Endoteliales , Óxido Nítrico Sintasa de Tipo III , Células Cultivadas , Células Endoteliales/metabolismo , Endotelio/metabolismo , Antígeno Ki-67/metabolismo , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , omega-N-Metilarginina/metabolismo
4.
Molecules ; 25(9)2020 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-32380665

RESUMEN

Quercus mongolica (QM)-a member of the Fagaceae family-has been used as traditional medicine in Korea, China and Mongolia as a treatment for inflammation of oral, genital or anal mucosa and for external inflammation of skin. To treat acne vulgaris (AV), we evaluated the inhibition of inflammatory cytokines (IL-6 and IL-8) of QM leaf extract (QML) and its main compound, pedunculagin (PD) in vitro and 5α-reductase inhibitory activity by western blotting. As results, QML and PD showed potent NO production inhibitory activity compared with the positive control (PC), NG-monomethyl-L-arginine (L-NMMA). QML and PD was also showed the decreases of IL-6 and IL-8 compared with the PC, EGCG and exhibited potent 5α-reductase type 1 inhibitory activities compared with the PC, dutasteride.


Asunto(s)
Inhibidores de 5-alfa-Reductasa/farmacología , Antiinflamatorios/farmacología , Quercus/química , Taninos/farmacología , Acné Vulgar/tratamiento farmacológico , Línea Celular , Colestenona 5 alfa-Reductasa/metabolismo , Regulación hacia Abajo , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Lipopolisacáridos/efectos adversos , Medicina Tradicional , Óxido Nítrico/metabolismo , Extractos Vegetales/farmacología , Hojas de la Planta/química , omega-N-Metilarginina/metabolismo
5.
J Biol Chem ; 295(51): 17441-17459, 2020 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-33453990

RESUMEN

Cancer cachexia is characterized by reductions in peripheral lean muscle mass. Prior studies have primarily focused on increased protein breakdown as the driver of cancer-associated muscle wasting. Therapeutic interventions targeting catabolic pathways have, however, largely failed to preserve muscle mass in cachexia, suggesting that other mechanisms might be involved. In pursuit of novel pathways, we used untargeted metabolomics to search for metabolite signatures that may be linked with muscle atrophy. We injected 7-week-old C57/BL6 mice with LLC1 tumor cells or vehicle. After 21 days, tumor-bearing mice exhibited reduced body and muscle mass and impaired grip strength compared with controls, which was accompanied by lower synthesis rates of mixed muscle protein and the myofibrillar and sarcoplasmic muscle fractions. Reductions in protein synthesis were accompanied by mitochondrial enlargement and reduced coupling efficiency in tumor-bearing mice. To generate mechanistic insights into impaired protein synthesis, we performed untargeted metabolomic analyses of plasma and muscle and found increased concentrations of two methylarginines, asymmetric dimethylarginine (ADMA) and NG-monomethyl-l-arginine, in tumor-bearing mice compared with control mice. Compared with healthy controls, human cancer patients were also found to have higher levels of ADMA in the skeletal muscle. Treatment of C2C12 myotubes with ADMA impaired protein synthesis and reduced mitochondrial protein quality. These results suggest that increased levels of ADMA and mitochondrial changes may contribute to impaired muscle protein synthesis in cancer cachexia and could point to novel therapeutic targets by which to mitigate cancer cachexia.


Asunto(s)
Caquexia/metabolismo , Proteínas Musculares/biosíntesis , Neoplasias/complicaciones , omega-N-Metilarginina/metabolismo , Animales , Arginina/análogos & derivados , Caquexia/etiología , Femenino , Xenoinjertos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias Musculares/metabolismo
6.
Cytokine ; 64(1): 62-70, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23953854

RESUMEN

In contrast to the well known immunostimulatory roles of IL-12, little has been known about its immunosuppressive roles. In the present study, IL-12-activated lymphocyte-mediated macrophage apoptosis was investigated by employing murine lymphocyte/macrophage cocultures. IL-12-activated lymphocytes and their culture supernatants induced an inducible nitric oxide synthase (iNOS)-mediated nitric oxide (NO) synthesis in macrophages. The NO synthesis was markedly inhibited by blocking antibodies to IFN-γ and TNF-α, suggesting the key role of these lymphocyte cytokines in mediating the NO synthesis. The endogenously produced NO inhibited macrophage proliferation, and induced apoptosis in concordance with the accumulation of p53, phosphatase and tensin homologue deleted on chromosome 10 (PTEN) and DR5, and the activation of caspase-3, processes that were inhibited by N(G)-monomethyl-l-arginine, aminoguanidine (NO synthase inhibitors) and 2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl 3-oxide (an NO scavenger). These results were further supported by the findings obtained from the experiments employing IFN-γ-knockout and iNOS-knockout mice. Our study demonstrated a novel, non-contact-dependent mechanism of macrophage suppression by IL-12-activated lymphocytes: induction of growth inhibition and apoptosis of macrophages due to endogenous NO synthesis induced by cytokines secreted from IL-12-activated lymphocytes.


Asunto(s)
Apoptosis/efectos de los fármacos , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD8-positivos/efectos de los fármacos , Interleucina-12/farmacología , Macrófagos/inmunología , Animales , Anticuerpos Bloqueadores/inmunología , Apoptosis/inmunología , Benzoatos/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Caspasa 3/metabolismo , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Guanidinas/metabolismo , Imidazoles/metabolismo , Interferón gamma/genética , Interferón gamma/inmunología , Interferón gamma/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico/biosíntesis , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fosfohidrolasa PTEN/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Transducción de Señal/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , omega-N-Metilarginina/metabolismo
7.
J Recept Signal Transduct Res ; 31(2): 168-72, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21385054

RESUMEN

Protein arginine methylation is a common post-translational modification in eukaryotes that is catalyzed by a family of the protein arginine methyltransferases (PRMTs). PRMTs are classified into three types: type I and type II add asymmetrically and symmetrically dimethyl groups to arginine, respectively, while type III adds solely monomethyl group to arginine. However, although the enzymatic activity of type I and type II PRMTs have been reported, the substrate specificity and the methylation activity of type III PRMTs still remains unknown. Here, we report the characterization of Caenorhabditis elegans PRMT-2 and PRMT-3, both of which are highly homologous to human PRMT7. We find that these two PRMTs can bind to S-adenosyl methionine (SAM), but only PRMT-3 has methyltransferase activity for histone H2A depending on its SAM-binding domain. Importantly, thin-layer chromatographic analysis demonstrates that PRMT-3 catalyzes the formation of monomethylated, but not dimethylated arginine. Our study thus identifies the first type III PRMT in C. elegans and provides a means to elucidate the physiological significance of arginine monomethylation in multicellular organisms.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/enzimología , Proteína-Arginina N-Metiltransferasas/metabolismo , Animales , Biocatálisis , Proteínas de Caenorhabditis elegans/genética , Histonas/metabolismo , Humanos , Metilación , Metiltransferasas/química , Filogenia , Unión Proteica , Proteína-Arginina N-Metiltransferasas/genética , S-Adenosilmetionina/metabolismo , Homología de Secuencia de Aminoácido , omega-N-Metilarginina/metabolismo
8.
Injury ; 39 Suppl 4: 40-6, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18804585

RESUMEN

SUMMARY: Cigarette smoking is hazardous to a range of human tissues. For instance, cigarette smoke inhalation has been proven to delay bone healing. This study analysed the effects of cigarette smoking on tibial vascular endothelium and blood flow using the bone-chamber model. The effects of smoking cessation and hyperbaric oxygen (HBO) on the damage caused by smoking were also compared. 54 adult New Zealand rabbits were divided into three groups. Group 1: control, Group 2: 1 week smoking, and Group 3: 6 weeks' smoking. This study on rabbits confirmed that both short-term and long-term cigarette smoking is dangerous to the bony vascular endothelium of the tibia. The vasodilatation caused by nitric oxide production was significantly attenuated in Group 2 and 3's tibia. Long-term smoking damaged the vascular endothelium more severely than short-term smoking (P<.01). Cessation of smoking effectively reduces the adverse effects of smoking when the cessation time equals the smoking time. HBO also effectively reduces the adverse effects of smoking.


Asunto(s)
Células Endoteliales/fisiología , Endotelio Vascular/fisiología , Oxigenoterapia Hiperbárica , Cese del Hábito de Fumar , Fumar/efectos adversos , Tibia/irrigación sanguínea , Acetilcolina/farmacología , Animales , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Endotelio Vascular/efectos de los fármacos , Femenino , Masculino , Modelos Animales , Conejos , Arterias Tibiales/efectos de los fármacos , Arterias Tibiales/fisiología , Factores de Tiempo , omega-N-Metilarginina/administración & dosificación , omega-N-Metilarginina/metabolismo
9.
J Allergy Clin Immunol ; 120(6): 1346-53, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17935768

RESUMEN

BACKGROUND: Nasal polyposis is characterized by impaired regulation of nasal tissue growth and is associated with chronic inflammation, sinus infections, and low levels of nitric oxide (NO). Based on its critical role in mediating cell growth and antimicrobial function, decrease of NO levels has been implicated in the pathogenesis of nasal polyposis. OBJECTIVE: We sought to evaluate mechanisms for the low NO level in polyposis, including factors regulating NO synthase (NOS) expression and activity and NO consumptive processes in nasal epithelial cells and nasal lavage fluid. METHODS: Eighteen patients with nasal polyposis and 8 healthy control subjects were studied. Nasal brushings, nasal lavage fluid, and nasal biopsy specimens were collected and analyzed. RESULTS: NO metabolite levels (nitrite and nitrate) in nasal lavage fluid from patients with polyps were less than those in control subjects, but activation of signal transduction and inducer of transcription 1, which regulates inducible NOS gene expression and protein expression, was present at higher levels in polyp than in healthy control tissue. Levels of arginine, methylarginine, and endogenous NOS inhibitors were similar between polyp and control tissue. In contrast, superoxide dismutase activity of polyp tissues was lower than that seen in control tissue and associated with increased nitrotyrosine, a biomarker of oxidant consumptive products of NO. CONCLUSION: Taken together, these data suggest that the nasal polyp environment is characterized by abnormalities in NO metabolism that might predispose to altered regulation of tissue growth and infection. CLINICAL IMPLICATIONS: Identification of NO metabolic abnormalities might lead to novel treatments for sinonasal polyposis targeted against the pathways identified within this study.


Asunto(s)
Pólipos Nasales/metabolismo , Transducción de Señal/inmunología , Adulto , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Masculino , Pólipos Nasales/enzimología , Pólipos Nasales/inmunología , Óxido Nítrico/genética , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/metabolismo , Oxidación-Reducción , Consumo de Oxígeno/efectos de los fármacos , Consumo de Oxígeno/inmunología , Polimorfismo de Nucleótido Simple , Factor de Transcripción STAT1/biosíntesis , Transducción de Señal/efectos de los fármacos , Especificidad por Sustrato , Superóxido Dismutasa/metabolismo , Tirosina/análogos & derivados , Tirosina/metabolismo , omega-N-Metilarginina/antagonistas & inhibidores , omega-N-Metilarginina/metabolismo
10.
J Mol Biol ; 369(4): 967-84, 2007 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-17475277

RESUMEN

Previously, we demonstrated that signal transducer and activator of transcription factor 1 (STAT1) plays an essential role in liver injury induced by lipopolysaccharide (LPS)/D-galactosamine (D-GalN); however, the underlying mechanism involved remains unclear. Here, we showed that LPS/D-GalN administration induced secretion of tumor necrosis factor alpha (TNF-alpha) and interferon gamma (IFN-gamma), which mediated apoptosis synergistically. Moreover, LPS/D-GalN-induced apoptosis was associated with increased inducible nitric oxide synthase (iNOS) and nitric oxide (NO) production, as well as elevated reactive oxygen species (ROS) production, which were all strongly inhibited by treatment with the antioxidant N-acetyl-L-cysteine (NAC) and an iNOS/NO inhibitor, L-NMMA. Although STAT1 activation and expression did not change significantly in TNF-alpha/IFN-gamma-cotreated cells compared with cells treated with IFN-gamma alone, the absence of STAT1 or interferon regulatory factor 1 (IRF-1) in genetic knockout mice strongly abrogated the observed effects of TNF-alpha/IFN-gamma on iNOS/NO induction, ROS production, loss of mitochondrial transmembrane potential (DeltaPsim), and apoptosis compared with STAT1(+/+) and IRF-1(+/+) mice. Additionally, the synergistic effects of TNF-alpha/IFN-gamma on iNOS/NO induction, ROS production, and apoptosis were significantly inhibited by overexpression of dominant negative STAT1 in contrast to overexpression of wild-type STAT1. In STAT1-deficient mice, nuclear factor kappaB (NF-kappaB) activation by TNF-alpha/IFN-gamma was attenuated and strongly inhibited by both NAC and L-NMMA. Moreover, the proteasome inhibitor, MG132, inhibited NF-kappaB activation and strongly inhibited iNOS/NO induction, ROS production, and loss of DeltaPsim induced by TNF-alpha/IFN-gamma, thereby inhibiting apoptosis. Interestingly, it appears peroxynitrite, which is produced by TNF-alpha/IFN-gamma, may interfere with STAT1 phosphorylation by inducing STAT1 nitration. Collectively, these findings demonstrate that TNF-alpha/IFN-gamma synergistically potentiates iNOS/NO induction, ROS production, and loss of DeltaPsim via STAT1 overexpression, playing an important role in promoting apoptosis and liver injury induced by LPS/D-GalN.


Asunto(s)
Galactosamina/farmacología , Hepatocitos , Factor 1 Regulador del Interferón/metabolismo , Lipopolisacáridos/farmacología , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción STAT1/metabolismo , Acetilcisteína/metabolismo , Animales , Apoptosis/fisiología , Células Cultivadas , Inhibidores Enzimáticos/metabolismo , Depuradores de Radicales Libres/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/fisiología , Humanos , Factor 1 Regulador del Interferón/genética , Interferón gamma/genética , Interferón gamma/metabolismo , Peroxidación de Lípido , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/efectos de los fármacos , FN-kappa B/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/metabolismo , Factor de Transcripción STAT1/genética , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , omega-N-Metilarginina/metabolismo
11.
Am J Respir Cell Mol Biol ; 36(6): 697-705, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17255557

RESUMEN

Hyperresponsiveness to bronchoconstrictor stimuli is a major pathophysiologic feature of asthma, but the molecular mechanisms behind this are not fully understood. The release of TNF-alpha and IL-1beta during the inflammatory process is believed to play an important role in airway hyperresponsiveness. We have previously demonstrated, using a murine in vitro model of chronic airway inflammation, that TNF-alpha up-regulated bradykinin B(1) and B(2) receptors in the airway smooth muscle. By using the same model, the present study was designed to investigate the effects of IL-1beta and its interaction with TNF-alpha on the expression of bradykinin B(1) and B(2) receptors in mouse tracheal smooth muscle. IL-1beta up-regulated bradykinin B(1) and B(2) receptor expression and increased contractile response to bradykinin B(1) and B(2) receptor agonists (des-Arg(9)-bradykinin and bradykinin, respectively) in the tracheal smooth muscle. Transcriptional inhibitor actinomycin D, c-Jun N-terminal kinase (JNK) inhibitors SP600125 and TAT-TI-JIP(153-163), but not extracellular signal-regulated kinase 1 and 2 (ERK 1/2) inhibitor PD98059, significantly attenuated this up-regulation, indicating that a transcriptional mechanism and intracellular JNK signal transduction pathway were involved. In addition, IL-1beta did not affect bradykinin B(1) and B(2) receptor mRNA stability. Remicade, an anti-TNF-alpha antibody, markedly suppressed IL-1beta-induced up-regulation of bradykinin B(1) and B(2) receptors, suggesting that TNF-alpha was involved in the up-regulation, which is further supported by the fact that IL-1beta enhanced TNF-alpha mRNA expression in the tracheae. Intracellular JNK pathway and TNF-alpha might provide key links between inflammatory mediators like IL-1beta and airway hyperresponsiveness to bradykinin.


Asunto(s)
Regulación de la Expresión Génica , Interleucina-1beta/metabolismo , Receptor de Bradiquinina B1/metabolismo , Receptor de Bradiquinina B2/metabolismo , Tráquea/metabolismo , Transcripción Genética , Animales , Antiinflamatorios/metabolismo , Anticuerpos Monoclonales , Asma/metabolismo , Bradiquinina/análogos & derivados , Bradiquinina/metabolismo , Hiperreactividad Bronquial/metabolismo , Inhibidores Enzimáticos/metabolismo , Humanos , Infliximab , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Contracción Muscular/fisiología , Músculo Liso/metabolismo , Receptor de Bradiquinina B1/genética , Receptor de Bradiquinina B2/genética , Transducción de Señal/fisiología , Tráquea/anatomía & histología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , omega-N-Metilarginina/metabolismo
12.
Eur J Neurosci ; 24(4): 1175-81, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16930443

RESUMEN

The aim of the present study was to determine the effect of pertussis toxin (PTX) on inflammatory hypernociception measured by the rat paw pressure test and to elucidate the mechanism involved in this effect. In this test, prostaglandin E(2) (PGE(2)) administered subcutaneously induces hypernociception via a mechanism associated with neuronal cAMP increase. Local intraplantar pre-treatment (30 min before), and post-treatment (5 min after) with PTX (600 ng/paw1, in 100 microL) reduced hypernociception induced by prostaglandin E(2) (100 ng/paw, in 100 microL, intraplantar). Furthermore, local intraplantar pre-treatment (30 min before) with PTX (600 ng/paw, in 100 microL) reduced hypernociception induced by DbcAMP, a stable analogue of cAMP (100 microg/paw, in 100 microL, intraplantar), which indicates that PTX may have an effect other than just G(i)/G(0) inhibition. PTX-induced analgesia was blocked by selective inhibitors of nitric oxide synthase (L-NMMA), guanylyl cyclase (ODQ), protein kinase G (KT5823) and ATP-sensitive K(+) channel (Kir6) blockers (glybenclamide and tolbutamide). In addition, PTX was shown to induce nitric oxide (NO) production in cultured neurons of the dorsal root ganglia. In conclusion, this study shows a peripheral antinociceptive effect of pertussis toxin, resulting from the activation of the arginine/NO/cGMP/PKG/ATP-sensitive K(+) channel pathway.


Asunto(s)
Analgésicos/metabolismo , Arginina/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Óxido Nítrico/metabolismo , Toxina del Pertussis/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Adenosina Trifosfato/metabolismo , Analgesia , Animales , Bucladesina/metabolismo , Carbazoles/metabolismo , Células Cultivadas , Dinoprostona/administración & dosificación , Dinoprostona/inmunología , Inhibidores Enzimáticos/metabolismo , Ganglios Espinales/citología , Gliburida/metabolismo , Indoles/metabolismo , Canales KATP , Masculino , Neuronas/citología , Neuronas/metabolismo , Oxadiazoles/metabolismo , Dolor/metabolismo , Dimensión del Dolor , Quinoxalinas/metabolismo , Ratas , Ratas Wistar , Transducción de Señal/fisiología , Tolbutamida/metabolismo , omega-N-Metilarginina/metabolismo
13.
Int J Mol Med ; 18(2): 355-63, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16820946

RESUMEN

To obtain a better understanding of the mechanisms involved in the up-regulation of the Fas apoptotic signaling cascade induced by P. aeruginosa type III secretion system (TTSS), human umbilical vein endothelial cells (HUVEC) were infected with P. aeruginosa PAO-1 or its TTSS-negative mutant PAO-1::exsA. PAO-1 was significantly more cytotoxic than the mutant and features of apoptosis (DNA fragmentation and annexin V reactivity) were more prominent in cultures infected with the wild-type bacteria. PAO-1 induced the up-regulation of Fas and the release of soluble FasL (sFasL) from infected cells but cell treatment with antagonist anti-Fas did not completely abrogate apoptosis suggesting that, besides the activated Fas-FasL pathway, other mechanisms are likely to be associated with the induction of apoptosis. LNMMA, a potent inhibitor of NO synthesis, completely inhibited apoptosis in both PAO-1 and PAO-1::exsA infected cultures. Moreover, PAO-1 was shown to up-regulate both the expression of iNOS and NO production by HUVEC. Treatment of cells with LNMMA completely inhibited cell expression of mFas. Based on these results we speculate that P. aeruginosa TTSS not only accounts for HUVEC higher expression of Fas and release of sFasL but also leads to overproduction of NO and to a NO-dependent up-regulation of the Fas-FasL proteins.


Asunto(s)
Proteínas Bacterianas/metabolismo , Células Endoteliales , Proteína Ligando Fas/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico/metabolismo , Pseudomonas aeruginosa/metabolismo , Receptor fas/metabolismo , Animales , Apoptosis/fisiología , Línea Celular , Células Endoteliales/citología , Células Endoteliales/metabolismo , Células Endoteliales/microbiología , Inhibidores Enzimáticos/metabolismo , Humanos , Pseudomonas aeruginosa/patogenicidad , Regulación hacia Arriba , omega-N-Metilarginina/metabolismo
14.
Biochemistry ; 45(17): 5618-30, 2006 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-16634643

RESUMEN

The enzyme dimethylargininase (also known as dimethylarginine dimethylaminohydrolase or DDAH; EC 3.5.3.18) catalyzes the hydrolysis of endogenous nitric oxide synthase inhibitors, N(omega)-methyl-l-arginine and N(omega),N(omega)-dimethyl-l-arginine. Understanding the mechanism and regulation of DDAH activity is important for developing ways to control nitric oxide production during angiogenesis and in many cases of vascular endothelial pathobiology. Several possible physiological regulation mechanisms of DDAH depend upon the presence of an active-site cysteine residue, Cys249 in Pseudomonas aeruginosa (Pa) DDAH, which is proposed to serve as a nucleophile in the catalytic mechanism. Through the use of pH-dependent ultraviolet and visible (UV-vis) difference spectroscopy and inactivation kinetics, the pK(a) of the active-site Cys249 in the resting enzyme was found to be unperturbed from pK(a) values of typical noncatalytic cysteine residues. In contrast, the pH dependence of k(cat) values indicates a much lower apparent pK(a) value. UV-vis difference spectroscopy between wild-type and C249S DDAH shows absorbance changes consistent with Cys249 deprotonation to the anionic thiolate upon binding positively charged ligands. The proton from Cys249 is lost either to the solvent or to an unidentified general base. A mutation of the active-site histidine residue, H162G, does not eliminate cysteine nucleophilicity, further arguing against a pre-formed ion pair with Cys249. Finally, UV-vis and X-ray absorption spectroscopy revealed that inhibitory metal ions can bind at these two active-site residues, Cys249 and His162, and also stabilize the anionic form of Cys249. These results support a proposed substrate-assisted mechanism for Pa DDAH in which ligand binding modulates the reactivity of the active-site cysteine.


Asunto(s)
Amidohidrolasas/metabolismo , Cisteína/metabolismo , Pseudomonas aeruginosa/enzimología , Amidohidrolasas/antagonistas & inhibidores , Sitios de Unión , Citrulina/análogos & derivados , Citrulina/metabolismo , Cobalto/metabolismo , Concentración de Iones de Hidrógeno , Yodoacetamida/farmacología , Cinética , Lisina/farmacología , Espectrometría de Masas , Modelos Químicos , Protones , Espectrofotometría Ultravioleta , Análisis Espectral , Tiourea/análogos & derivados , Tiourea/metabolismo , Rayos X , Zinc/farmacología , omega-N-Metilarginina/metabolismo
15.
Biochem J ; 395(3): 563-70, 2006 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-16426232

RESUMEN

The HSL7 (histone synthetic lethal 7) gene in the yeast Saccharomyces cerevisiae encodes a protein with close sequence similarity to the mammalian PRMT5 protein, a member of the class of protein arginine methyltransferases that catalyses the formation of omega-N(G)-monomethylarginine and symmetric omega-N(G),N'(G)-dimethylarginine residues in a number of methyl-accepting species. A full-length HSL7 construct was expressed as a FLAG-tagged protein in Saccharomyces cerevisiae. We found that FLAG-tagged Hsl7 effectively catalyses the transfer of methyl groups from S-adenosyl-[methyl-3H]-L-methionine to calf thymus histone H2A. When the acid-hydrolysed radiolabelled protein products were separated by high-resolution cation-exchange chromatography, we were able to detect one tritiated species that co-migrated with an omega-N(G)-monomethylarginine standard. No radioactivity was observed that co-migrated with either the asymmetric or symmetric dimethylated derivatives. In control experiments, no methylation of histone H2A was found with two mutant constructs of Hsl7. Surprisingly, FLAG-Hsl7 does not appear to effectively catalyse the in vitro methylation of a GST (glutathione S-transferase)-GAR [glycine- and arginine-rich human fibrillarin-(1-148) peptide] fusion protein or bovine brain myelin basic protein, both good methyl-accepting substrates for the human homologue PRMT5. Additionally, FLAG-Hsl7 demonstrates no activity on purified calf thymus histones H1, H2B, H3 or H4. GST-Rmt1, the GST-fusion protein of the major yeast protein arginine methyltransferase, was also found to methylate calf thymus histone H2A. Although we detected Rmt1-dependent arginine methylation in vivo in purified yeast histones H2A, H2B, H3 and H4, we found no evidence for Hsl7-dependent methylation of endogenous yeast histones. The physiological substrates of the Hsl7 enzyme remain to be identified.


Asunto(s)
Histonas/metabolismo , Proteínas Quinasas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimología , Timo , omega-N-Metilarginina/biosíntesis , Secuencia de Aminoácidos , Animales , Catálisis , Bovinos , Expresión Génica , Histonas/aislamiento & purificación , Péptidos y Proteínas de Señalización Intracelular , Metilación , Metiltransferasas/metabolismo , Datos de Secuencia Molecular , Proteínas Quinasas/química , Proteínas Quinasas/genética , Proteína-Arginina N-Metiltransferasas , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Alineación de Secuencia , Especificidad por Sustrato , omega-N-Metilarginina/metabolismo
16.
J Int Med Res ; 33(5): 545-54, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16222888

RESUMEN

Electromagnetic field (EMF) radiation has been found to induce arteriolar dilatation, but the mechanism of action remains largely unknown. This study investigated the effect of EMF radiation on the production of endothelin-1 (ET-1), a potent vasoconstrictor, by cultured endothelial cells. EMF radiation reduced ET-1 basal levels in human umbilical vein and microvascular endothelial cells, but failed to reduce ET-1 basal levels in bovine and human aortic endothelial cells. EMF radiation significantly inhibited thrombin-stimulated ET-1 production in all four endothelial cell types in a dose-dependent manner. EMF radiation significantly inhibited thrombin-induced endothelin-1 mRNA expression in all four cell types. The inhibitory effect of EMF radiation on ET-1 production was abolished by the nitric oxide synthase inhibitor NG-monomethyl-L-arginine (10(-3) mol/1). These results demonstrate that EMF radiation modulates ET-1 production in cultured vascular endothelial cells and the inhibitory effect of EMF radiation is, at least partly, mediated through a nitric oxide-related pathway.


Asunto(s)
Campos Electromagnéticos , Células Endoteliales/metabolismo , Células Endoteliales/efectos de la radiación , Endotelina-1/metabolismo , Trombina/metabolismo , Animales , Aorta/anatomía & histología , Bovinos , Células Cultivadas , Relación Dosis-Respuesta en la Radiación , Células Endoteliales/citología , Endotelina-1/genética , Endotelio Vascular/citología , Inhibidores Enzimáticos/metabolismo , Humanos , Óxido Nítrico/metabolismo , ARN Mensajero/metabolismo , Venas Umbilicales/anatomía & histología , omega-N-Metilarginina/metabolismo
17.
Proc Natl Acad Sci U S A ; 101(20): 7799-804, 2004 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-15136749

RESUMEN

ATP-sensitive potassium (K(ATP)) channels regulate insulin release, vascular tone, and neuronal excitability. Whether these channels are modulated by NO, a membrane-permeant messenger in various physiological and pathological processes, is not known. The possibility of NO signaling via K(ATP) channel modulation is of interest because both NO and K(ATP) have been implicated in physiological functions such as vasodilation and neuroprotection. In this report, we demonstrate a mechanism that leads to K(ATP) activation via NO/Ras/mitogen-activated protein kinase pathway. By monitoring K(ATP) single-channel activities from human embryonic kidney 293 cell-attached patches expressing sulfonylurea receptor 2B and Kir6.2, we found K(ATP) stimulation by NO donor Noc-18, a specific NO effect abolished by NO scavenger 2-phenyl-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide (PTIO) but not guanylyl cyclase inhibitor 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ). NO stimulation of K(ATP) is indirect and requires Ras and mitogen-activated protein kinase kinase activities. Blockade of Ras activation by pharmacological means or by coexpressing either a dominant-negative or an S-nitrosylation-site mutant Ras protein significantly abrogates the effects of NO. Inhibition of mitogen-activated protein kinase kinase abolishes the NO activation of K(ATP) but suppression of phosphatidylinositol 3-kinase does not. The NO precursor l-Arg also stimulates K(ATP) via endogenous NO synthase and the Ras signaling pathway. In addition, in rat hippocampal neurons, the protective effect of ischemic preconditioning induced by oxygen-glucose deprivation requires K(ATP) and NO synthase activity during preconditioning. Thus, neuroprotection caused by NO released during the short episode of sublethal ischemia may be mediated partly by K(ATP) stimulation.


Asunto(s)
Proteínas Quinasas Activadas por Mitógenos/metabolismo , Óxido Nítrico/metabolismo , Canales de Potasio/metabolismo , Proteínas ras/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Arginina/metabolismo , Humanos , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Ratas , omega-N-Metilarginina/metabolismo , Proteínas ras/antagonistas & inhibidores
18.
Med Sci Monit ; 9(7): CR297-303, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12883448

RESUMEN

BACKGROUND: Von Willebrand factor (vWF) has generally been regarded as a good predictor of vascular risk. However, no previous studies have examined its relationship with coronary calcification. The aim of this study was to determine whether vWF activity is higher in type 1 diabetic patients than controls; its relationship with cardiovascular risk factors; and to endothelial nitric oxide production and coronary artery calcification. MATERIAL/METHODS: Von Willebrand factor activity was measured in 181 type 1 diabetic patients and 188 controls. Coronary artery calcification was measured by Electron Beam Computed Tomography. Forearm blood flow was measured by venous plethysmography in response to intra-brachial infusion of bradykinin, glyceryl trinitrate, noradrenaline and NG-monomethyl-L-arginine (L-NMMA) in 149 subjects. RESULTS: Von Willebrand factor was significantly increased in diabetic patients compared to controls (median 100% vs 87%, p=0.001). Von Willebrand factor activity was significantly higher in diabetic patients with micro/macroalbuminuria than those with normoalbuminuria (109% vs 93%, p<0.001). Among diabetic subjects, being in the top quartile for vWF was associated with a lower response to L-NMMA (p=0.009). There was no association between vWF activity and coronary artery calcification in either the diabetic (p=0.9) or control group (p=0.8). CONCLUSIONS: Cardiovascular risk factors including albuminuria do not explain the high vWF activity in type 1 diabetic patients. There is some evidence that vWF correlates with endothelial nitric oxide production. The lack of correlation with coronary artery calcification indicates that vWF is not a useful marker of atheroma burden.


Asunto(s)
Calcinosis/metabolismo , Enfermedad de la Arteria Coronaria/metabolismo , Diabetes Mellitus Tipo 1/sangre , Endotelio Vascular/metabolismo , Óxido Nítrico/metabolismo , Factor de von Willebrand/metabolismo , Adulto , Albuminuria/metabolismo , Área Bajo la Curva , Bradiquinina/metabolismo , Bradiquinina/uso terapéutico , Calcinosis/patología , Enfermedad de la Arteria Coronaria/tratamiento farmacológico , Enfermedad de la Arteria Coronaria/patología , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/uso terapéutico , Femenino , Humanos , Masculino , Nitroglicerina/metabolismo , Nitroglicerina/uso terapéutico , Factores de Riesgo , Estadística como Asunto , Vasodilatadores/metabolismo , Vasodilatadores/uso terapéutico , omega-N-Metilarginina/metabolismo , omega-N-Metilarginina/uso terapéutico
19.
Cancer ; 97(5): 1358-63, 2003 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-12599246

RESUMEN

BACKGROUND: The winged helix/forkhead transcriptional factor FKHRL1 (FOXO3a) triggers apoptosis, but its mode of action is not well understood. ROCK kinase is an effector molecule in human breast carcinoma cell apoptosis, but its relation to FKHRL1 is unknown. Because the human breast carcinoma T47D cell line releases a great amount of nitric oxide (NO), I investigated signaling of FKHRL1/ROCK [corrected] kinase during NO suppression. METHODS: Expression of phosphorylated FKHRL1 in T47D cells was analyzed using Western blotting. Apoptosis was evaluated by flow cytometry. Transfection of FKHRL1-HA wild-type and mutant FKHRL1-HA T32A constructs were performed by lipofectamine plus reagent. Measurement of NO generation was performed by Griess reaction. RESULTS: Nitric oxide suppression promotes FKHRL1 thr-32-enhanced phosphorylation, which was significantly (P < 0.005) sensitive to Y-27632, a specific inhibitor of the ROCK kinase, but not to capase-3 inhibitor or wortnannin, a specific inhibitor of phosphoinositol-3-OH kinase (PI3K). Nitric oxide suppression by N-(G)-monomethyl-L-arginine, an inhibitor of NO synthase, causes a significant (P < 0.001) increase in the apoptosis of T47D cells. However, a significant decrease (P < 0.01) in NO generation and a significant (P < 0.01) increase in apoptosis were observed when FKHRL1-HA wild-type cells were transfected, which caused increased FKHRL1 thr-32 phosphorylation. CONCLUSIONS: This novel unknown phenomenon of breast carcinoma cell apoptosis was triggered by NO suppression, which promotes FKHRL1 thr-32-enhanced phosphorylation and initiates signaling of FKHRL1 to ROCK kinase as an effector molecule. This apoptotic signalling process is caspase-3 as well as PI3K/Akt independent.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias de la Mama/metabolismo , Proteínas de Unión al ADN/metabolismo , Óxido Nítrico/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Transcripción/metabolismo , Western Blotting , Neoplasias de la Mama/enzimología , Caspasa 3 , Caspasas/genética , Caspasas/metabolismo , Proteínas de Unión al ADN/genética , Femenino , Proteína Forkhead Box O1 , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead , Humanos , Péptidos y Proteínas de Señalización Intracelular , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal , Factores de Transcripción/genética , Células Tumorales Cultivadas , omega-N-Metilarginina/genética , omega-N-Metilarginina/metabolismo , Quinasas Asociadas a rho
20.
Sci STKE ; 2001(93): pl1, 2001 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-11752667

RESUMEN

Posttranslational modification of proteins allows cells to adapt and react quickly to their environment beyond the boundaries set forth by genetic code. Arginine methylation, a protein modification discovered almost 30 years ago, has recently experienced a renewed interest as several new arginine methyltransferases have been identified and numerous proteins were found to be regulated by methylation on arginine residues. Until recently, the detection of arginine methylation required the use of chromatography and mass-spectrometrical analysis. The following protocol provides guidelines for the straightforward identification of arginine-methylated proteins, made possible by the availability of novel, commercially available reagents.


Asunto(s)
Arginina/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas/metabolismo , Animales , Anticuerpos Monoclonales/metabolismo , Arginina/análogos & derivados , Arginina/química , Proteínas de Unión al ADN/química , Activación Enzimática , Humanos , Immunoblotting , Metilación , Ratones , Pruebas de Precipitina , Procesamiento Proteico-Postraduccional , Proteínas/química , Factor de Transcripción STAT1 , Transducción de Señal , Transactivadores/química , Células Tumorales Cultivadas , omega-N-Metilarginina/química , omega-N-Metilarginina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA