Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Mol Cell Biochem ; 478(5): 1031-1044, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36214894

RESUMEN

Protein arginine methyltransferase 5 (PRMT5) is overexpressed in lung carcinoma, which promotes tumor cell proliferation, survival, migration and invasion. Compound Kushen injection (CKI) is a mixture of natural compounds extracted from Kushen and Baituling, which are mainly used to stop in cancer pain and bleeding. Here we found that cell viability and colony formation were inhibited after the incubation of AMI-1. Meanwhile, AMI-1 suppressed cell migration, enhanced apoptosis, induced cell cycle arrest, inhibited PRMT5 expression and histone H3R8 and H4R3 symmetric di-methylation (H3R8me2s and H4R3me2s) accumulation, down-regulated the expression of eukaryotic translation initiation factor 4E (eIF4E) in lung carcinoma cells. Moreover, AMI-1 suppressed tumor growth, decreased H3R8me2s and H4R3me2s accumulation, down-regulated eIF4E expression and increased p53 expression in lung carcinoma xenografts of BALB/c nude mice. Of note, combined and CKI markedly enhanced the anticancer efficacy CKI in lung carcinoma. The above findings demonstrated that AMI-1 has established antineoplastic activity and this role may be associated with affecting the function of eIF4E via inhibiting PRMT5 activity or protein levels in lung carcinoma. This study highlights evidence of novel selective anticancer activity of AMI-1 in combination with CKI in lung carcinoma.


Asunto(s)
Antineoplásicos , Carcinoma , Neoplasias Pulmonares , Animales , Humanos , Ratones , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular , Regulación hacia Abajo , Factor 4E Eucariótico de Iniciación/metabolismo , Pulmón/patología , Neoplasias Pulmonares/patología , Ratones Desnudos , Proteína-Arginina N-Metiltransferasas/genética
2.
Toxicol Appl Pharmacol ; 435: 115829, 2022 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-34919946

RESUMEN

Type I co-activator-associated arginine methyltransferase 1 (CARM1) and type II protein arginine methyltransferase 5 (PRMT5) are highly expressed in multiple cancers including liver cancer and their overexpression contributes to poor prognosis, thus making them promising therapeutic targets. Here, we evaluated anti-tumor activity of ribavirin in hepatocellular carcinoma (HCC). We found that ribavirin significantly inhibited the proliferation of HCC cells in a time- and dose-dependent manner. Furthermore, ribavirin suppressed the growth of subcutaneous and orthotopic xenograft of HCC in mice, decreased vascular endothelial growth factor (VEGF) and peritoneal permeability to reduce ascites production, and prolonged the survival of mice in HCC ascites tumor models. Mechanistically, ribavirin potently down-regulated global protein expression of CARM1 and PRMT5, and concurrently decreased accumulation of H3R17me2a and H3R8me2s/H4R3me2s. However, ribavirin did not affect the activity and mRNA levels of both CARM1 and PRMT5 in vivo and in vitro HCC cells. In addition, our ChIP results shown that ribavirin inhibited CARM1 which in turn decreased the H3R17me2a, binds to the eukaryotic translation initiation factor 4E (eIF4E) and VEGF promoter region, and reduced the relative mRNA expression level of eIF4E and VEGF in HCC cells. Our findings suggested a potential therapeutic strategy for patients with HCC through inhibition of the abnormal activation/expression of both CARM1 and PRMT5.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Ascitis/tratamiento farmacológico , Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Ribavirina/farmacología , Animales , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Factor 4E Eucariótico de Iniciación/biosíntesis , Factor 4E Eucariótico de Iniciación/genética , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Ratones , Proteína-Arginina N-Metiltransferasas/genética , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/genética , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Toxicol Appl Pharmacol ; 415: 115450, 2021 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-33577917

RESUMEN

Eukaryotic translation initiation factor 4E (eIF4E) and protein arginine methyltransferase 5 (PRMT5) are frequently overexpressed in colorectal cancer (CRC) tissues and associated with poor prognosis. Ribavirin, the only clinically approved drug known to target eIF4E, is an anti-viral molecule currently used in hepatitis C therapy. The potential of ribavirin to treat CRC remains largely unknown. Ribavirin treatment in CRC cell lines drastically inhibited cell proliferation and colony formation, induced S phase arrest and reduced cyclin D1, cyclin A/E and proliferating cell nuclear antigen (PCNA) levels in vitro, and suppressed tumorigenesis in mouse model of colitis-associated CRC. Mechanistically, ribavirin treatment significantly reduced PRMT5 and eIF4E protein levels and the accumulation of symmetric dimethylation of histone 3 at arginine 8 (H3R8me2s) and that of histone 4 at arginine 3 (H4R3me2s). Importantly, inhibition of PRMT5 by ribavirin resulted in promoted H3R8 methylation in eIF4E promoter region. Our results demonstrate the anti-cancer efficacy of ribavirin in CRC and suggest that the anti-cancer efficacy of ribavirin may be mediated by downregulating PRMT5 levels but not its enzymatic activity.


Asunto(s)
Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/tratamiento farmacológico , Metilación de ADN/efectos de los fármacos , Factor 4E Eucariótico de Iniciación/metabolismo , Histonas/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Ribavirina/farmacología , Animales , Neoplasias Asociadas a Colitis/tratamiento farmacológico , Neoplasias Asociadas a Colitis/enzimología , Neoplasias Asociadas a Colitis/genética , Neoplasias Asociadas a Colitis/patología , Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Regulación hacia Abajo , Factor 4E Eucariótico de Iniciación/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Humanos , Ratones Endogámicos ICR , Regiones Promotoras Genéticas , Proteína-Arginina N-Metiltransferasas/genética , Puntos de Control de la Fase S del Ciclo Celular/efectos de los fármacos , Transducción de Señal , Carga Tumoral/efectos de los fármacos
4.
Pharmazie ; 75(4): 147-150, 2020 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-32295691

RESUMEN

Ginsenoside Rd (GS-Rd), isolated from the Chinese traditional herbal medicine Panax ginseng, is used for the treatment of cardiovascular diseases, inflammation, different body pains, and trauma. Caspase-3 and Caspase-9 belong to cysteine aspartic acid specific protease (Caspase) family that plays an important role in apoptosis progression of cancers. In the present study, we investigated the anti-tumor effect of GS-Rd by MTT assay, colony formation assessment, flow cytometry, and Western blotting. Our results revealed that ginsenoside Rd significantly inhibits human gastric cancer (GC) growth and cell proliferation. Flow cytometer analysis showed that the GS-Rd could significantly induce apoptosis and arrest the G0/G1 phase in GC cells. Further, GS-Rd was found to increase the ratio of Bax/Bcl-2 and the expression of Caspase-3 and Caspase-9, respectively, and to decrease the expression of Cyclin D1. Taken together, our study suggests that GS-Rd significantly inhibits GC cell proliferation, induces cell apoptosis through increase the expression of Caspase-3, Caspase-9, and the ratio of Bax/Bcl-2. GS-Rd also induces cell cycle arrest at G0/G1 phase by down-regulation Cyclin D1. Thus, GS-Rd could serve as a lead to develop novel therapeutic agents to against human gastric cancer.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Caspasa 3/genética , Caspasa 9/genética , Ginsenósidos/uso terapéutico , Neoplasias Gástricas/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Caspasa 3/biosíntesis , Caspasa 9/biosíntesis , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ensayo de Tumor de Célula Madre , Regulación hacia Arriba/efectos de los fármacos , Proteína X Asociada a bcl-2/metabolismo
5.
Pharmazie ; 73(5): 269-273, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29724292

RESUMEN

Protein arginine methyltransferase 5 (PRMT5), a type II PRMT, is highly expressed in several types of tumors including cervical cancer. Arginine methyltransferase inhibitor 1 (AMI-1) inhibits solid tumors by targeting PRMT5. However, the effect of AMI-1 on cervical cancer is still unknown. In this study, we provided the first evidence that AMI-1 reduced cervical cancer cell proliferation, colony formation and promoted cell apoptosis in vitro. Suppression of tumorigenicity was also confirmed in vivo. Mechanistic studies revealed that AMI-1 significantly reduced PRMT5 level in cells and mice xenografts model of cervical cancer. These results suggest that AMI-1 inhibits cervical cancer by type II PRMT5.


Asunto(s)
Antineoplásicos/farmacología , Naftalenosulfonatos/farmacología , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Urea/análogos & derivados , Neoplasias del Cuello Uterino/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Factor 4E Eucariótico de Iniciación/metabolismo , Femenino , Células HeLa , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteína-Arginina N-Metiltransferasas/metabolismo , Urea/farmacología , Neoplasias del Cuello Uterino/enzimología , Neoplasias del Cuello Uterino/patología , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Toxicol Appl Pharmacol ; 336: 1-7, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-28987382

RESUMEN

Arginine methylation is carried out by protein arginine methyltransferase (PRMTs) family. Arginine methyltransferase inhibitor 1 (AMI-1) is mainly used to inhibit type I PRMT activity in vitro. However, the effects of AMI-1 on type II PRMT5 activity and gastric cancer (GC) remain unclear. In this study, we provided the first evidence that AMI-1 significantly inhibited GC cell proliferation and migration while induced GC cell apoptosis, and reduced the expression of PRMT5, eukaryotic translation initiation factor 4E (eIF4E), symmetric dimethylation of histone 3 (H3R8me2s) and histone 4 (H4R3me2s). In addition, AMI-1 inhibited tumor growth, downregulated eIF4E, H4R3me2s and H3R8me2s expression in mice xenografts model of GC. Collectively, our results suggest that AMI-1 inhibits GC by downregulating eIF4E and targeting type II PRMT5.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Factor 4E Eucariótico de Iniciación/metabolismo , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Represoras/antagonistas & inhibidores , Neoplasias Gástricas/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Femenino , Histonas/metabolismo , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas Represoras/metabolismo , Transducción de Señal/efectos de los fármacos , Neoplasias Gástricas/enzimología , Neoplasias Gástricas/patología , Factores de Tiempo , Carga Tumoral/efectos de los fármacos
7.
J Neurosci ; 35(37): 12890-902, 2015 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-26377474

RESUMEN

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


Asunto(s)
Amidinas/farmacología , Cocaína/farmacología , Histonas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Núcleo Accumbens/enzimología , Procesamiento Proteico-Postraduccional , Proteína-Arginina N-Metiltransferasas/fisiología , Pirimidinas/farmacología , Animales , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Metilación , Ratones , Modelos Moleculares , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/fisiología , Conformación Proteica , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Interferencia de ARN , ARN Interferente Pequeño/farmacología
8.
Neurobiol Dis ; 82: 54-65, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26054440

RESUMEN

Studies have showed that prenatal cocaine exposure (PCOC) can impair cognitive function and social behavior of the offspring; however, the mechanism underlying such effect is poorly understood. Insulin-like growth factor II (Igf-II), an imprinted gene, has a critical role in memory consolidation and enhancement. We hypothesized that epigenetic regulation of hippocampal Igf-II may attribute to the cognitive deficits of PCOC offspring. We used Morris water maze and open-field task to test the cognitive function in PCOC offspring. The epigenetic alteration involved in hippocampal Igf-II expression deficit in PCOC offspring was studied by determining Igf-II methylation status, DNA methyltransferases (DNMT) expressions and L-methionine level. Moreover, IGF-II rescue experiments were performed and the downstream signalings were investigated in PCOC offspring. In behavioral tests, we observed impaired spatial learning and memory and increased anxiety in PCOC offspring; moreover, hippocampal IGF-II mRNA and protein expressions were significantly decreased. Hippocampal methylation of cytosine-phospho-guanine (CpG) dinucleotides in differentially methylated region (DMR) 2 of Igf-II was elevated in PCOC offspring, which may be driven by the upregulation of L-methionine and DNA methyltransferase (DNMT) 1. Importantly, intra-hippocampal injection of recombinant IGF-II reactivated the repressed calcium calmodulin kinase II α (CaMKIIα) and reversed cognitive deficits in PCOC offspring. Collectively, our findings suggest that cocaine exposure during pregnancy impairs cognitive function of offspring through epigenetic modification of Igf-II gene. Enhancing IGF-II signaling may represent a novel therapeutical strategy for cocaine-induced cognitive impairment.


Asunto(s)
Cocaína/farmacología , Cognición/efectos de los fármacos , Epigénesis Genética , Factor II del Crecimiento Similar a la Insulina/genética , Efectos Tardíos de la Exposición Prenatal/genética , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Cognición/fisiología , Metilación de ADN/efectos de los fármacos , Femenino , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Factor II del Crecimiento Similar a la Insulina/metabolismo , Factor II del Crecimiento Similar a la Insulina/farmacología , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Ratones , Fosforilación , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Efectos Tardíos de la Exposición Prenatal/psicología
9.
J Transl Med ; 13: 349, 2015 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-26541651

RESUMEN

BACKGROUND: Protein arginine methyltransferase 5 (PRMT5), a type II PRMT, is highly expressed in some tumors, but its role in hepatocellular carcinoma (HCC) is still unknown. METHODS: PRMT5 level in HCC specimens was determined by immunohistochemical staining and the association with clinicopathologic features was evaluated. PRMT5 was inhibited by AMI-1 (a small molecule inhibitor of PRMTs) or small interference RNA (siRNA). The proliferation of HCC cells was tested by Cell Counting Kit-8, cell migration was evaluated by Transwell assay and cell cycle and apoptosis were analyzed by flow cytometry. The effect of AMI-1 on HCC in vivo was examined by mouse xenograft model. RESULTS: PRMT5 expression was markedly upregulated in HCC tissues, and correlated inversely with overall patient survival. Knockdown of PRMT5 significantly reduced the proliferation of HCC cells, but did not affect the growth of normal liver cells. Furthermore, ß-catenin was identified as a target of PRMT5. Silencing PRMT5 significantly down-regulated the expression of ß-catenin and the downstream effector Cyclin D1 in HCC cells. AMI-1 strongly inhibited HCC growth in vivo, increased the ratio of Bax/Bcl-2, and led to apoptosis and loss of migratory activity in several HCC cells. Meanwhile, AMI-1 decreased the expression levels of symmetric dimethylation of H4 (H4R3me2s), a histone mark of PRMT5. CONCLUSIONS: PRMT5 plays an important role in HCC. PRMT5 may be a promising target for HCC therapy.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias Hepáticas/metabolismo , Naftalenosulfonatos/farmacología , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Urea/análogos & derivados , beta Catenina/metabolismo , Animales , Apoptosis , Ciclo Celular , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Regulación hacia Abajo , Femenino , Citometría de Flujo , Silenciador del Gen , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos BALB C , Trasplante de Neoplasias , Proteína-Arginina N-Metiltransferasas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Ratas , Urea/farmacología
10.
BMC Neurosci ; 15: 32, 2014 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-24558969

RESUMEN

BACKGROUND: Nicotine is rapidly absorbed from cigarette smoke and therefore induces a number of chronic illnesses with the widespread use of tobacco products. Studies have shown a few cerebral metabolites modified by nicotine; however, endogenous metabolic profiling in brain has not been well explored. RESULTS: H NMR-based on metabonomics was applied to investigate the endogenous metabolic profiling of brain hippocampus, nucleus acumens (NAc), prefrontal cortex (PFC) and striatum. We found that nicotine significantly increased CPP in mice, and some specific cerebral metabolites differentially changed in nicotine-treated mice. These modified metabolites included glutamate, acetylcholine, tryptamine, glucose, lactate, creatine, 3-hydroxybutyrate and nicotinamide-adenine dinucleotide (NAD), which was closely associated with neurotransmitter and energy source. Additionally, glutathione and taurine in hippocampus and striatum, phosphocholine in PFC and glycerol in NAc were significantly modified by nicotine, implying the dysregulation of anti-oxidative stress response and membrane metabolism. CONCLUSIONS: Nicotine induces significant metabonomic alterations in brain, which are involved in neurotransmitter disturbance, energy metabolism dysregulation, anti-oxidation and membrane function disruptions, as well as amino acid metabolism imbalance. These findings provide a new insight into rewarding effects of nicotine and the underlying mechanism.


Asunto(s)
Encéfalo/metabolismo , Espectroscopía de Resonancia Magnética/métodos , Metaboloma/fisiología , Neurotransmisores/metabolismo , Nicotina/farmacología , Animales , Encéfalo/efectos de los fármacos , Masculino , Metaboloma/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Agonistas Nicotínicos/farmacología , Protones
11.
Chem Biol Interact ; 370: 110305, 2023 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-36529159

RESUMEN

Ribavirin is a common antiviral drug, especially for patients with hepatitis C. Our recent studies demonstrated that ribavirin showed anti-tumor activity in colorectal cancer and hepatocellular carcinoma, but its effects on lung cancer remains unclear. This study aimed to evaluate the anti-tumor activity of ribavirin against lung cancer and elucidate the underlying mechanism. We established orthotopic mouse model of lung cancer (LLC and GLC-82) and employed an ultra-high-performance liquid chromatography quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF/MS)-based metabolomics approach. We found that ribavirin significantly inhibited the proliferation and colony formation of lung cancer cells. Tumor sizes of orthotopic lung cancer in ribavirin-treated groups were also significantly lower than those in control groups. Metabolomics analysis revealed that ribavirin mainly affected 5 metabolic pathways in orthotopic lung tumor models, taurine and hypotaurine metabolism, nicotinate and nicotinamide metabolism, linoleic acid metabolism, arginine biosynthesis and arachidonic acid metabolism. Furthermore, we identified 5 upregulated metabolites including ß-nicotinamide adenine dinucleotide (NAD+), nicotinamide (NAM), taurine, ornithine and citrulline, and 7 downregulated metabolites including 1-methylnicotinamide (MNAM), S-adenosyl-l-homocysteine (SAH), N1-Methyl-2-pyridone-5-carboxamide (2PY), homocysteine (Hcy), linoleic acid, arachidonic acid (AA) and argininosuccinic acid in ribavirin-treated groups. These results provide new insight into the anti-tumor mechanism of ribavirin for lung cancer.


Asunto(s)
Neoplasias Pulmonares , Ribavirina , Ratones , Animales , Ribavirina/farmacología , Ribavirina/uso terapéutico , Ácido Araquidónico , Ácido Linoleico , Metabolómica/métodos , Neoplasias Pulmonares/tratamiento farmacológico , Cromatografía Líquida de Alta Presión/métodos , Niacinamida , Taurina , Biomarcadores
12.
J Neurochem ; 123(5): 790-9, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22957495

RESUMEN

Cocaine dependence involves in the brain's reward circuit as well as nucleus accumbens (NAc), a key region of the mesolimbic dopamine pathway. Many studies have documented altered expression of genes and identified transcription factor networks and epigenetic processes that are fundamental to cocaine addiction. However, all these investigations have focused on mRNA of encoding genes, which may not always reflect the involvement of long non-coding RNAs (lncRNAs), which has been implied in a broad range of biological processes and complex diseases including brain development and neuropathological process. To explore the potential involvement of lncRNAs in drug addiction, which is viewed as a form of aberrant neuroplasticity, we used a custom-designed microarray to examine the expression profiles of mRNAs and lncRNAs in brain NAc of cocaine-conditioned mice and identified 764 mRNAs, and 603 lncRNAs were differentially expressed. Candidate lncRNAs were identified for further genomic context characterization as sense-overlap, antisense-overlap, intergenic, bidirection, and ultra-conserved region encoding lncRNAs. We found that 410 candidate lncRNAs which have been reported to act in cis or trans to their targeted loci, providing 48 pair mRNA-lncRNAs. These results suggest that the modification of mRNAs expression by cocaine may be associated with the actions of lncRNAs. Taken together, our results show that cocaine can cause the genome-wide alterations of lncRNAs expressed in NAc, and some of these modified RNA transcripts may to play a role in cocaine-induced neural plasticity and addiction.


Asunto(s)
Trastornos Relacionados con Cocaína/genética , Núcleo Accumbens , ARN no Traducido/análisis , Transcriptoma , Animales , Condicionamiento Operante , Masculino , Ratones , Ratones Endogámicos C57BL , ARN no Traducido/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
13.
Anticancer Drugs ; 23(6): 590-6, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22241170

RESUMEN

The aim of this study was to establish xenograft models of tumor in mice bladder and evaluate the antitumor efficacy of ampelopsin sodium (Amp-Na). A total of 2×10 human bladder carcinoma EJ cells and murine sarcoma 180 cells were instilled into the bladder of BALB/c nu/nu mice and Swiss mice after preconditioning to establish the tumor model. Mice bearing orthotopic tumors were treated with Amp-Na by intravenous, intraperitoneal, or intravesical instillation. In addition, the pharmacokinetics property of Amp-Na was investigated in normal BALB/c mice. Our results showed that Amp-Na was excreted mainly through the urine, where it existed at a high concentration. Amp-Na significantly inhibited the proliferation of EJ and sarcoma 180 cells both in vivo and in vitro and this can be at least partially attributed to the cell cycle arrest induced by Amp-Na. This study suggests that the use of Amp-Na is an attractive chemotherapeutic modality for bladder cancer patients.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Flavonoides/farmacología , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Administración Intravesical , Animales , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/farmacocinética , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Flavonoides/administración & dosificación , Flavonoides/farmacocinética , Humanos , Inyecciones Intraperitoneales , Inyecciones Intravenosas , Ratones , Ratones Endogámicos BALB C , Sarcoma/tratamiento farmacológico , Sarcoma/patología , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Basic Clin Pharmacol Toxicol ; 131(1): 18-33, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35470570

RESUMEN

Protein arginine methyltransferases 1 and 5 (PRMT1 and PRMT5) are frequently overexpressed in diverse types of cancers and correlate with poor prognosis, thus making these enzymes potential therapeutic targets. The aim of this study was to assess and elucidate the anti-tumour effect and epigenetic regulatory mechanism of ribavirin in soft tissue sarcomas (STS). We showed that ribavirin inhibited growth and metastasis and prolonged survival in animals bearing STS cells by downregulating the mRNA and protein levels of PRMT1/PRMT5 and attenuating the accumulation of asymmetric and symmetric di-methylation of arginine (ADMA and SDMA). Furthermore, ribavirin lowered the permeability of the peritoneum in KM mice bearing S180 ascites via decreasing the level of vascular endothelial growth factor (VEGF). Ribavirin was a potent inhibitor of cell proliferation and metastasis in STS cells through downregulation of both type I PRMT1 and type II PRMT5. Ribavirin could be used to enhance the efficacy of doxorubicin in STS allograft tumour models.


Asunto(s)
Proteína-Arginina N-Metiltransferasas , Sarcoma , Animales , Arginina , Proliferación Celular , Ratones , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Ribavirina/farmacología , Ribavirina/uso terapéutico , Sarcoma/tratamiento farmacológico , Factor A de Crecimiento Endotelial Vascular
15.
Regul Toxicol Pharmacol ; 60(1): 73-8, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21338646

RESUMEN

Previously studies have shown that tumor-homing peptide NGR enhances the therapeutic efficacy of human interferon α2a (IFNα2a) against tumors. Here we investigated in vivo anti-tumor effect of recombinant human IFNα2a-NGR (rhIFNα2a-NGR) against human lung adenocarcinoma cell line SPC-A-1, A549 and murine Lewis lung carcinoma (LLC) subcutaneously xenografted tumors and further assessed the immunogenicity of rhIFNα2a-NGR in Sprague Dawley (SD) rats and rhesus monkeys. We found that rhIFNα2a-NGR significantly inhibited the growth of SPC-A-1, A549 and LLC cells-xenografted tumors in a dose-dependent manner. Although the antibodies to rhIFNα were detected in the serum of SD rats and rhesus monkeys treated with rhIFNα2a-NGR, these antibodies did not cause obvious pathological consequence. Taken together, these data demonstrate that rhIFNα2a-NGR has obvious anti-tumor efficacy in vivo, perhaps due to the tumor-homing peptide NGR. Thus rhIFNα2a-NGR represents a promising novel drug for effective treatment of cancer.


Asunto(s)
Antineoplásicos/farmacología , Interferón-alfa/genética , Neoplasias Pulmonares/tratamiento farmacológico , Oligopéptidos/genética , Receptores Inmunológicos/genética , Proteínas Recombinantes de Fusión/farmacología , Animales , Antineoplásicos/inmunología , Línea Celular Tumoral , Humanos , Interferón alfa-2 , Interferón-alfa/inmunología , Neoplasias Pulmonares/patología , Macaca mulatta , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Oligopéptidos/inmunología , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Pharmazie ; 66(9): 690-3, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22026125

RESUMEN

The compound rh-IFNalpha-2a-NGR can inhibit tumor angiogenesis and could be used for targeted therapy. In the present study, double antibody sandwich ELISA analysis was used to determine the concentration of rh-IFNalpha-2a-NGR in serum after intramuscular administration of various dosages to mice, rats and monkeys. The results showed that the pharmacokinetic properties of rh-IFNalpha2a-NGR after i.m. administration to mice, rats and monkeys were consistent with a one-compartment open model. The main pharmacokinetic parameters in mice (9.36 microg/kg), rats (4.68 microg/kg) and monkeys (2.34 microg/kg) after i.m. rh-IFNalpha2a-NGR were as follows: T(peak) was 0.49, 1.65 and 3.60h, C(max) was 3030.20, 654.49 and 268.13 ng/L, t1/2 was 0.39, 4.52 and 2.70 h, and AUC(0-infinity)) was 4197.65, 5784.58 and 2622.06 ng/L x h, respectively. Also, mice, rats and monkeys had their own distinct metabolic characteristics. These data would provide references for further clinical pharmacokinetic study of rh-IFNalpha2a-NGR.


Asunto(s)
Inhibidores de la Angiogénesis/farmacocinética , Interferón-alfa/farmacocinética , Inhibidores de la Angiogénesis/administración & dosificación , Animales , Área Bajo la Curva , Calibración , Ensayo de Inmunoadsorción Enzimática , Haplorrinos , Inyecciones Intramusculares , Interferón-alfa/administración & dosificación , Ratones , Ratas , Reproducibilidad de los Resultados
17.
Clin Chim Acta ; 510: 291-297, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32707041

RESUMEN

BACKGROUND: Protein arginine methyltransferase 5 (PRMT5) belongs to a large family of protein arginine methyltransferases (PRMTs) that play essential role in gene transcription and regulate tumorigenesis. However, the role of PRMT5 in the regulation of cancer cell metabolism remains unclear. METHODS: Cell metabolomic analysis was performed on SW480 cells transfected with small interfering RNA (siRNA) specifically targeting PRMT5, followed by metabolomic pathway analysis. RESULTS: PRMT5 was overexpressed in colorectal cancer (CRC) tissues, and downregulation of PRMT5 suppressed CRC cell proliferation and the levels of PRMT5 and symmetric dimethylation of histone H3 (H3R8me2s). In addition, we found distinct differences in metabolite classification and function in PRMT5 knockdown SW480 cells compared to control SW480 cells. PRMT5 knockdown increased the levels of amino acids and carbohydrates, particularly related to the arginine metabolism such as glutamate, glutamine (Gln), proline, creatine, creatinine and phosphocreatine (PCr). CONCLUSIONS: These findings revealed a key role for PRMT5 as a regulator of CRC cell metabolism to mediate arginine methylation in CRC cells.


Asunto(s)
Neoplasias Colorrectales , Proteína-Arginina N-Metiltransferasas , Proliferación Celular , Neoplasias Colorrectales/genética , Histonas , Humanos , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Espectroscopía de Protones por Resonancia Magnética
18.
Oncol Lett ; 18(1): 189-196, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31289488

RESUMEN

Previous studies have demonstrated that ampelopsin (AMP), a type of flavonoid isolated from the stems and leaves of Ampelopsis grossedentata, exhibits anti-cancer activity in various types of cancer. Conversion of AMP into its sodium salt (AMP-Na) conferred enhanced solubility and stability to it. The present study aimed to evaluate the anti-cancer activity of AMP-Na in human lung adenocarcinoma cell lines and to investigate its mechanisms of action. Cell proliferation and viability were assessed by MTT and colony formation assays, and cell migration was determined using a scratch wound healing assay. The cell cycle distribution, apoptosis rate and tubulin immunofluorescence intensity were analyzed using flow cytometry, the cell ultra-microstructure was examined using transmission electron microscopy and the accumulation of tubulin was determined using laser confocal microscopy. The results demonstrated that AMP-Na significantly inhibited the proliferation, clonogenicity and migration of human lung adenocarcinoma cells. Furthermore, AMP-Na induced SPC-A-1 cell apoptosis, and promoted tubulin polymerization. The results suggested that the underlying mechanisms of AMP-Na may involve targeting of microtubules and tubulin polymerization to subsequently disrupt mitosis and induce cell cycle arrest at the S-phase.

19.
Oncol Lett ; 16(2): 2161-2166, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30008914

RESUMEN

Protein arginine methyltransferases (PRMTs) are a class of epigenetic modified enzymes that are overexpressed in a various types of cancer and serve pivotal functions in malignant transformation. Arginine methyltransferase inhibitor-1 (AMI-1) is a symmetrical sulfonated urea that inhibits the activity of type I PRMT in vitro. However, previous studies demonstrated that AMI-1 may also inhibit the activity of type II PRMT5 in vitro. To the best of our knowledge, the present study provides the first evidence that AMI-1 may significantly inhibit the viability of mouse sarcoma 180 (S180) and human osteosarcoma U2OS cells. Additionally, the results demonstrated that AMI-1 downregulated the activities of PRMT5, the symmetric dimethylation of histone 4 and histone 3 (a PRMT5-specific epigenetic mark) in a mouse xenograft model of S180 and induced apoptosis in S180 cells. Taken together, the results suggest that AMI-1 may exhibit antitumor effects against sarcoma cells by targeting PRMT5.

20.
Exp Neurol ; 307: 52-61, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29753648

RESUMEN

Nicotinamide phosphoribosyltransferase (NAMPT), a rate-limiting enzyme in nicotinamide adenine dinucleotide (NAD) biosynthesis in mammals, converts nicotinamide into nicotinamide mononucleotide (NMN). NMN is subsequently converted to NAD, a component that is critical for cell energy metabolism and survival. Sirtuin 1 (SIRT1), an NAD-dependent histone deacetylase, plays an important role in mediating memory and synaptic plasticity. Here, we found that NAMPT was significantly upregulated in the ventral tegmental area (VTA) of cocaine-conditioned mice. Intraperitoneal or intra-VTA injection of FK866, a specific inhibitor of NAMPT, significantly attenuated cocaine reward. However, such effects were clearly repressed by intra-VTA expression of NAMPT or supplementation with NMN. Using 1H-nuclear magnetic resonance metabolomic analysis, we found that the content of NAD and NMN were increased in the VTA of cocaine-conditioned mice; moreover, the expression of SIRT1 was also upregulated. Interestingly, the inhibitory effect of FK866 on cocaine reward was significantly weakened in Sirt1 midbrain conditional knockout mice. Our results suggest that NAMPT-mediated NAD biosynthesis may modify cocaine behavioral effects through SIRT1. Moreover, our findings reveal that the interplay between NAD biosynthesis and SIRT1 regulation may comprise a novel regulatory pathway that responds to chronic cocaine stimuli.


Asunto(s)
Cocaína/farmacología , Citocinas/biosíntesis , Nicotinamida Fosforribosiltransferasa/biosíntesis , Recompensa , Sirtuina 1/biosíntesis , Animales , Condicionamiento Psicológico/efectos de los fármacos , Condicionamiento Psicológico/fisiología , Inhibidores de Captación de Dopamina/farmacología , Locomoción/efectos de los fármacos , Locomoción/fisiología , Espectroscopía de Resonancia Magnética/métodos , Mesencéfalo/efectos de los fármacos , Mesencéfalo/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Área Tegmental Ventral/efectos de los fármacos , Área Tegmental Ventral/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA